WO2020051363A1 - Targeted il-12 treatments and methods to stimulate hank and nk92mi cells - Google Patents

Targeted il-12 treatments and methods to stimulate hank and nk92mi cells Download PDF

Info

Publication number
WO2020051363A1
WO2020051363A1 PCT/US2019/049797 US2019049797W WO2020051363A1 WO 2020051363 A1 WO2020051363 A1 WO 2020051363A1 US 2019049797 W US2019049797 W US 2019049797W WO 2020051363 A1 WO2020051363 A1 WO 2020051363A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
chimeric protein
cancer
seq
cells
Prior art date
Application number
PCT/US2019/049797
Other languages
French (fr)
Inventor
Kayvan Niazi
Original Assignee
Nantbio, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nantbio, Inc. filed Critical Nantbio, Inc.
Priority to EP19856745.5A priority Critical patent/EP3847242A4/en
Priority to US17/274,127 priority patent/US20220289805A1/en
Priority to CN201980058286.XA priority patent/CN112996904A/en
Publication of WO2020051363A1 publication Critical patent/WO2020051363A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5434IL-12
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464436Cytokines
    • A61K39/46444Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • 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]
    • C12N2501/2312Interleukin-12 (IL-12)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • the field of the invention is cancer treatments and methods using natural killer cells and immune stimulatory cytokines, and especially activated NK cells and IL-12.
  • NK92 derivatives such as activated NK cells (aNK cells), genetically modified NK cells with high affinity CD 16 receptors (haNK cells), or chimeric antigen receptors (taNK cells) were used. While such cell-based treatments are conceptually attractive, the tumor microenvironment and other patient- specific factors will often reduce their cytotoxic activity, and various attempts have been made to modulate cytotoxicity in NK92 cells.
  • Interleukin-2 IL-2
  • interleukin- 12 IL-12
  • cytokines elicit strong antitumor effects by stimulating unmodified immune cells, including T cells and natural killer (NK) cells.
  • T cells T cells and natural killer cells.
  • NK natural killer cells
  • IFN-g interferon-g
  • IL-12 is a stronger inducer of IFN-g from unmodified NK cells and activated T cells.
  • IFN-g mRNA has increased stability in NK cells co-stimulated with IL-2 and IL-12 (see e.g., Molecular and Cellular Biology, Mar. 2002, p. 1742-53).
  • the IL-2 and IL-12 concentrations used in vitro may not necessarily reflect achievable or even desirable levels in vivo. Indeed, IL-2 systemic administration of IL-2 is associated with relatively high toxicity and capillary leak syndrome, while several deaths have been attributed to systemic administration of IL-12.
  • the response of primary NK cells to various cytokines is not necessarily the same as the response of genetically modified NK92 cells.
  • compositions and methods are known to activate NK cells, there remains a need for compositions and methods to treat cancer using NK cells, especially where the NK cells are stimulated with IL-12 in a clinically safe manner.
  • NK cells that express and intracellularly retain IL-2, in which the NK cells are stimulated with a chimeric protein that comprises a cancer cell targeting portion and an IL-12 portion.
  • a chimeric protein that comprises a cancer cell targeting portion and an IL-12 portion.
  • such chimeric protein has substantially reduced systemic toxicity and moreover beneficially induces IFN-g secretion in a targeted manner.
  • such chimeric proteins significantly enhance IFN-g secretion in NK cells that express and intracellularly retain IL-2 as compared to native NK cells.
  • the inventor contemplates a method of inducing IFNy secretion in an NK cell that includes one step of providing an NK cell, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2; and another step of contacting the NK cell with a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion to induce interferon gamma (IFNy) secretion by the NK cell.
  • IFNy interferon gamma
  • the IL-2 sensitized NK cell is an NK cell that was previously and constitutively exposed to IL-2 or an NK cell was that constitutively exposed to at least 100 IU/mL IL-2.
  • the genetically modified NK cell may also be an aNK cell, an autologous NK cell, or an NK cell derived in vitro from a precursor cell, while in still further embodiments the genetically modified NK cell constitutively expresses and intracellularly retains IL-2 (e.g., haNK cell or an NK92MI cell).
  • the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IL- 12b chain, but homodimers of IL-l2a chain and other heterodimers are also deemed suitable.
  • the cancer cell targeting portion comprises an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion.
  • the cancer cell targeting portion targets a necrosis marker (e.g., DNA, RNA, histone protein, etc.) or a cancer associated antigen or a cancer neoepitope to so afford site specificity with respect to IL-12 action.
  • suitable cancer cell targeting portions may comprise a hu5l-4 antibody or portion thereof.
  • the step of contacting the NK cell with the chimeric protein is performed ex vivo and the NK cell is a haNK cell, or that the step of contacting the NK cell with the chimeric protein is performed in vivo and wherein the NK cell is a haNK cell or NK92MI cell.
  • the inventor also contemplates a method of treating cancer that includes a step of administering an NK cell to a patient, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2; and a further step of administering a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion to thereby induce interferon gamma (IFNy) secretion by the NK cell.
  • IFNy interferon gamma
  • the NK cell may be a genetically modified NK cell that constitutively expresses and intracellularly retains IL-2 (e.g., haNK cell or NK92MI cell), and/or that the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IL-12 chain.
  • IL-2 e.g., haNK cell or NK92MI cell
  • the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IL-12 chain.
  • the cancer cell targeting portion may comprise an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion, and it is generally preferred that the cancer cell targeting portion targets a necrosis marker (e.g., DNA, RNA, histone protein, etc.) or a cancer associated antigen or a cancer neoepitope.
  • a necrosis marker e.g., DNA, RNA, histone protein, etc.
  • the cancer cell targeting portion may comprise a hu5l-4 antibody or portion thereof.
  • the chimeric protein is administered at least 12 hours prior to administration of the NK cell, or that the chimeric protein is administered at least 12 hours after administration of the NK cell, or that the chimeric protein is contemporaneously administered with the NK cell.
  • the chimeric protein is bound to the NK cell.
  • the inventor also contemplates a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion that targets a necrosis marker (e.g., DNA, RNA, histone protein, etc.), and/or that the cancer cell targeting portion comprises a hu5l-4 antibody or portion thereof.
  • the cancer cell targeting portion includes an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion.
  • the IL-12 portion may be covalently coupled to the cancer cell targeting portion, and/or the IL-12 portion is covalently coupled to the cancer cell targeting portion via a peptide linker.
  • the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IL- 12b chain, and/or the cancer cell targeting portion comprises a hu5l-4 antibody or portion thereof.
  • the chimeric protein may be non-co valently bound to the CD 16 receptor of an NK cell.
  • the inventor also contemplates a pharmaceutical composition comprising the chimeric protein contemplated herein.
  • Such pharmaceutical composition may further comprise an NK cell as described above.
  • the inventor also contemplates a kit comprising (a) an NK cell, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2; and (b) a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion.
  • the same considerations as provided above apply.
  • the inventor therefore also contemplates use of a chimeric protein and an NK cell in the treatment of cancer, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2, and wherein the chimeric protein comprises an IL-12 portion and a cancer cell targeting portion that induces interferon gamma (IFNy) secretion by the NK cell.
  • the NK cells and/or the chimeric protein the same considerations as noted above apply.
  • Fig.1 shows exemplary graphs indicating that the tested NK cells require constitutive IL-2 expression/exposure for significant IL-12 mediated IFN-g secretion.
  • Fig.2 is an exemplary graph showing the biological activity of IL-12 and selected chimeric proteins comprising an IL-12 portion with respect to IL-12 mediated IFN-g secretion.
  • Fig.3 is an exemplary graph showing binding of selected chimeric proteins comprising an IL-12 portion linked to antigens exposed by cell death.
  • Fig.4 shows sequences for exemplary chimeric proteins comprising an IL-12 portion.
  • chimeric molecules comprising an IL- 12 portion can be used in an effective and targeted manner to induce IFNy secretion in IL-2 sensitized NK cells or in genetically modified NK cells that constitutively express IL-2 to thereby promote cytotoxic NK cell activity in a target tissue.
  • the chimeric molecules comprising the IL-12 portion help avoid toxicity otherwise encountered with systemically administered IL-12.
  • the chimeric molecule also includes a cancer targeting moiety (e.g., targeting one or more necrosis markers), induction of IFNy secretion by the NK cells can be rendered specific to the site of the cancer, and particularly to the hypoxic and/or necrotic tumor core.
  • contemplated compositions and methods will increase antigen presentation and MHC-I/II expression of immune competent cells (and especially CD4+ and/or CD8+ T cells) in a tumor, and bias the immune response in the tumor towards a Thl response.
  • use of the chimeric molecules presented herein not only avoids toxicity otherwise encountered with systemically administered IL-12, but also affords concentrated therapeutically effective quantities of IL-12 at the targeted tumor site.
  • necrosis markers in the context presented herein will advantageously allow for a tumor antigen independent antigen spread in the tumor microenvironment as the chimeric molecule will indiscriminately target necrotic tissue rather than cancer or tumor associated antigens.
  • the chimeric molecules presented herein retain the biological activity of the IL-12 heterodimer as is discussed in more detail below.
  • NK cells will fail to respond to IL-12, even where propagated with IL-2 supplied into the culture medium.
  • the inventor has also discovered that when IL-2 is supplied to the same cells in a constitutive or continuous manner, the cells are sensitive to IL-12 signaling.
  • the NK cells treated in this manner produce significant quantities of IFNy and have increased cytotoxicity, presumably due to increased intracellular concentrations and/or less fluctuations in IL-2 quantities.
  • the terms‘constitutive exposure’ or‘constitutively exposing’ in conjunction with IL- 2 as used herein means that biologically active IL-2 is supplied or produced in a continuous (or semi-continuous) manner such that variations in biologically active IL-2 concentration at or in the cell (or IL-2 stimulation) will vary by no more than 20% over 48 hours. Therefore, in some embodiments, variations in biologically active IL-2 concentration at or in the cell (or IL-2 stimulation) will vary by no more than 15% over 48 hours, or no more than 10% over 48 hours, or no more than 7% over 48 hours, or no more than 5% over 48 hours, or no more than 1% over 48 hours.
  • the terms “constitutive expression” or “constitutively expressing” in conjunction with IL-2 as used herein refers to (recombinant) expression of IL- 2 within a cell in a continuous fashion from a constitutive promoter (/. ⁇ ? ., without the need for induction). Most typically, such expression of IL-2 within a cell will be achieved by use of an endoplasmic retention sequence that is fused to the IL-2 to reduce or even entirely avoid secretion form the cell expressing such modified IL-2.
  • constitutive exposure to IL-2 is hypothesized to resemble more closely the exposure of NK cells to IL-2 in a natural setting, leading to durable sensitivity to IL-12 and IFNy secretion triggered by exposure to IL-12.
  • intermittent exposure of NK-92 cells to IL-2— as is the case in customary NK-92 cell culture, where culture media are renewed every two to three days— may lead to inactivation and/or degradation of biologically active IL-2, possibly due to binding by serum proteins or proteolysis by serum proteases of the culture media.
  • customary NK-92 cell culture conditions promote intermittent or‘pulsed’ IL-2 signaling which supports growth of NK-92 cells but fails to support cell signaling to render the NK-92 cells sensitive to IL-12 and IL-12 dependent IFNy secretion as well as increased (as compared to non-IL-l2 stimulated cells) expression of NKG2D.
  • chimeric proteins it is generally contemplated that all chimeric proteins that include at least a biologically active IL-12 portion are suitable. Moreover, it is generally preferred that the chimeric protein also includes a portion that extends serum half life time, that reduces systemic toxicity, and/or that allows target specific delivery and/or accumulation of IL-12 at a desired site. Consequently, with regard to the non- IL-12 portion of the chimeric protein, suitable non-IL-l2 portions include various serum proteins and portions thereof, and particularly albumin, lactoferrin, and various globulins. Therefore, one preferred non-IL-l2 portion comprises an antibody or fragment thereof.
  • the non-IL-l2 portion may be a heavy chain of an antibody, a light chain of an antibody (or portion thereof), or a scFv.
  • any binding specificity for such antibody or fragment thereof is generally deemed suitable, especially preferred antibodies will specifically bind to an antigen of a tumor cell.
  • suitable antibodies may bind to a tumor associated antigen, a tumor specific antigen, or a patient and tumor specific neoantigen (neoepitope). Therefore, the non- IL-12 portion is most preferably also used to deliver and retain the IL-12 at the site of a tumor to concentrate the biological effect of the IL-12 to the target site.
  • all sources of antibodies are deemed suitable, including human antibodies and all isoforms, non-human mammalian forms, humanized forms, and synthetic antibodies.
  • IL-12 is a heterodimer and that the IL-12 portion in the chimeric protein may be a single chain form of IL-12 (/. ⁇ ? ., scIL- 12) or a single subunit (/. ⁇ ? ., IL-l2a subunit (p35) and IL- 12b subunit (p40)) or active portion thereof.
  • the source of the IL-12 it is generally preferred that the source be human IL-12.
  • various other non-human (preferably mammalian) IL-12 forms are also expressly contemplated herein.
  • splice variants and mutants are also appropriate, so long as such forms exhibit at least some biological activity (e.g., induce IFNy secretion in haNK cells as described herein).
  • Suitable IL-12 protein sequences include human sequences, which may form a disulfide linked heterodimer formed from IL-l2a subunit (p35) and IL- 12b subunit (p40) or which may be covalently linked together via a flexible peptide linker as described in more detail below.
  • IL-12 sequences include publically available protein sequence (e.g., P29460 and P29459 at UniProt), and all natural and synthetic variants thereof, including truncated forms, splice variants, and mutant forms.
  • DNA and RNA sequences encoding IL-12 are known in the art and suitable sequences include those at Genbank (accession number AC010370 for genomic sequence and M65271 for mRNA of IL- 12a subunit; accession number AF512686 for genomic sequence and M65290 for mRNA of KM2b subunit).
  • Genbank accession number AC010370 for genomic sequence and M65271 for mRNA of IL- 12a subunit
  • accession number AF512686 for genomic sequence and M65290 for mRNA of KM2b subunit.
  • other non-human, preferably mammalian sequences are also deemed suitable for use herein.
  • Heterodimers of IL-l2a subunit and IL- 12b subunit may be formed by non-covalent association, or via a flexible peptide linker such as a G4S linker to form a single chain IL-12 molecule.
  • the chimeric proteins may also include addressing or leader sequences, linkers, tags, and/or flag for quantification and/or simplified purification. The proper choice will depend, at least in part, on the particular choice of production system, as is well known in the art.
  • a chimeric protein with the IL-12 portion and the non-IL-l2 portion may vary considerably, and will include covalent coupling of the two portions heavy chain as well as non-covalent coupling.
  • a necrosis targeting antibody hu5l-4 was used as a fusion partner with a single chain IL-12 (scIL-l2) as is described in more detail below.
  • NHS-IL12 is a chimeric construct that includes two molecules of IL-12 that are fused to a tumor necrosis-targeting human IgGl (NHS76).
  • antibody-cytokine conjugates may be prepared by chemical conjugation using cleavable (e.g., via disulfide bond or hydrazone, or proteolytic site, etc.) or non-cleavable linkers (e.g., via maleimide-modified PEG).
  • the conjugation may also be done using recombinant cloning in which the N- or C-terminus of the heavy or light chain (or fragment thereof) is modified to also encode in frame a linker portion and IL-12 as is also shown in more detail below.
  • chimeric recombinant proteins can be prepared that have an antibody portion that preferably binds to a component of a tumor cell, a linker, and an IL-12 portion.
  • exemplary antibody-drug conjugates with IL-12 retained significant activity as is shown in more detail below.
  • One exemplary fusion protein may therefore include a chimeric protein having a leader peptide portion, a murine scIL-l2 portion in normal type font (G 4 S linker in bold type), and a hu5l-4 antibody heavy chain portion as shown below in SEQ ID NO:l, in which the italicized sequence denotes the leader peptide portion, and in which the underlined sequence denotes the hu5l-4 antibody heavy chain portion.
  • Another exemplary fusion protein may include a leader peptide portion, a human scIL-l2 portion in normal type font (G 4 S linker in bold type), and a hu5l-4 antibody heavy chain portion as shown below in SEQ ID NO:2, in which the italicized sequence denotes the leader peptide portion, and in which the underlined sequence denotes the hu5 l-4 antibody heavy chain portion.
  • Still another exemplary protein includes a leader peptide portion and a hu5l-4 antibody light chain portion as shown below in SEQ ID NOG, in which the italicized sequence denotes the leader peptide portion, and in which the underlined sequence denotes the hu5l-4 antibody light chain portion.
  • NK cells it is generally contemplated that preferred NK cells are IL-2 sensitized NK cells or genetically modified NK cells that constitutively expresses IL-2.
  • IL-2 sensitized cells are cells that were subjected to constitutive exposure with IL-2 as noted above.
  • constitutive exposure can be done by continuous exposure and/or replenishment of IL-2 to the culture medium as described herein
  • constitutive exposure of NK/NK92 cells and their derivatives to IL-2 is more preferably achieved by intracellular expression of recombinant IL-2 in the respective cells.
  • intracellular expression is driven from a constitutively active promoter to achieve constant expression levels, and is expressed in a form that is not secreted (/. ⁇ ? ., lacks export signal sequence, and may include an endoplasmic or cytoplasmic retention sequence).
  • Such intracellular expression is believed to provide the same functional impact to the cell as constitutive exposure to externally provided IL-2.
  • NK92 cells or derivatives were genetically engineered to express and intracellularly retain IL-2
  • the cells were unexpectedly sensitized to IL-12 stimulation as measured by IFNy secretion and/or increased expression of NKG2D.
  • recombinant expression and intracellular retention of IL-2 can be done in numerous manners, and all of such methods are deemed suitable for use herein (see e.g., Oncotarget 2016 Dec 27; 7(52): 86359-86373 or Exp Hematol. 2005 Feb;33(2): 159-64).
  • recombinant cells can be administered to a patient in vivo without the need to administer to the same patient IL-2.
  • modified cells will be sensitized to IL-12 to secrete IFNy upon IL-12 stimulation.
  • sensitization by constitutive exposure (external or internal) to IL-2 provided substantial quantities of IFNy that is thought to provide a therapeutic effect in the context of concomitant immune therapy, particularly as NKG2D expression in such stimulated cells was also significantly increased.
  • genetically engineered NK cells may be NK-92 derivatives modified to express the high-affinity Fey receptor (CD16).
  • Sequences for high-affinity variants of the Fey receptor are well known in the art (see e.g., Blood 2009 113:3716-3725), and all manners of generating and expression are deemed suitable for use herein. Expression of such receptor is believed to allow specific targeting of tumor cells using antibodies that are specific to a patient’s tumor cells (e.g., neoepitopes), a particular tumor type (e.g., her2neu, PSA, PSMA, etc.), or that are associated with cancer (e.g., CEA-CAM).
  • tumor cells e.g., neoepitopes
  • a particular tumor type e.g., her2neu, PSA, PSMA, etc.
  • cancer e.g., CEA-CAM
  • such antibodies are commercially available and can be used in conjunction with the cells (e.g., bound to the Fey receptor), and such modified cells may also be commercially obtained from NantKwest as haNK cells (‘high-affinity natural killer cells).
  • haNK cells ‘high-affinity natural killer cells).
  • NK92MI cells have been reported (see e.g., Hum Gene Ther. 1999 May 20;l0(8):l359-73 or US 8034332) to express and retain IL-2 intracellularly and are commercially available from ATCC.
  • NK cells are deemed suitable for use herein so long as such cells are constitutively exposed to IL-2 or intracellularly express (and retain) IL-2 as described above.
  • suitable alternative NK cells include autologous NK cells from a patient (e.g., isolated from whole blood, or cultivated from precursor or stem cells using methods known in the art), and various allogenic NK cells.
  • the NK cells are genetically engineered to achieve one or more desirable traits, and particularly include NK-92 cells and derivatives thereof.
  • suitable genetically engineered NK cell include NK-92 derivatives that are modified to have reduced or abolished expression of at least one killer cell immunoglobulin-like receptor (KIR), which will render such cells constitutively activated (via lack of or reduced inhibition).
  • KIR killer cell immunoglobulin-like receptor
  • suitable NK cells may have one or more modified KIR that are mutated such as to reduce or abolish interaction with MHC class I molecules.
  • one or more KIRs may also be deleted or expression may be suppressed (e.g., via miRNA, siRNA, etc.).
  • Most typically, more than one KIR will be mutated, deleted, or silenced, and especially contemplated KIR include those with two or three domains, with short or long cytoplasmic tail.
  • modified, silenced, or deleted KIRs will include KIR2DL1, KIR2DL2, KIR2DL3, KIR2DL4, KIR2DL5A, KIR2DL5B, KIR2DS1, KIR2DS2, KIR2DS3, KIR2DS4, KIR2DS5, KIR3DL1, KIR3DL2, KIR3DL3, and/or KIR3DS1.
  • modified cells may be prepared using protocols well known in the art. Alternatively, such cells may also be commercially obtained from NantKwest (see URL www.nantkwest.com) as aNK cells (‘activated natural killer cells).
  • NK-92 cells exhibit an unusual receptor expression profile, expressing a relatively large number of activating (e.g., NKp30, NKp46, 2B4, NKGD, CD28) receptors. Conversely, NK-92 cells also express few inhibitory receptors (e.g., NKGA/B, low levels of KIR2DL4, ILT-2), and lack most of the killer inhibitory receptors (KIRs) clonally expressed on normal NK cells.
  • activating e.g., NKp30, NKp46, 2B4, NKGD, CD28
  • NK-92 cells also express few inhibitory receptors (e.g., NKGA/B, low levels of KIR2DL4, ILT-2), and lack most of the killer inhibitory receptors (KIRs) clonally expressed on normal NK cells.
  • KIRs killer inhibitory receptors
  • NK-92 expresses relatively high levels of molecules involved in the perforin-granzyme cytolytic pathway as well as additional cytotoxic effector molecules including tumor necrosis factor (TNF)-superfamily members FasL, TRAIL, TWEAK, TNF- alpha, indicating the ability to kill via alternative mechanisms.
  • TNF tumor necrosis factor
  • NK-92 cells also express other molecules implicated immune effector cell regulation (CD80, CD86, CD40L, TRANCE) whose relevance in NK killing is unclear.
  • NK-92 cells and derivatives thereof can be sensitized to IL-12 and exhibit an IL-12 dependent IFNy secretion phenotype by constitutively exposing the cells to IL-2.
  • constitutive exposure can be performed by continuous or semi-continuous addition of IL-2 to a culture medium to thereby maintain the concentration of biologically active IL-2 substantially constant. Therefore, it is contemplated that the concentration of biologically active IL-2 in the medium varies over 48 hours by no more than 15%, or by no more than 10%, or by no more than 7%, or by no more than 5%, or by no more than 3%.
  • Biological activity of IL-2 can be quantified using known procedures, e.g., using a CTLL-2 cell proliferation assay (J Immunol Methods. 2009 Aug 31; 348(1-2): 83-94).
  • Continuous or semi-continuous addition of IL-2 can be done in numerous manners, including use of a peristaltic pump or metered injector. Alternatively, and in less preferred aspects, continuous or semi-continuous addition of IL-2 can be done by media renewal in a frequent fashion (e.g., every two hours, every four hours, every eight hours, etc.). Where multiple or continuous additions are not preferred, the inventor also contemplates that the constitutive exposure can also be achieved using formulations that release IL-2 in a relatively slow manner.
  • delayed release of IL-2 can be done by pegylation of IL-2 as is known from NKTR- 214 (Nektar Therapeutics; 455 Mission Bay Blvd South; San Francisco, CA 94158).
  • pegylated IL-2 is believed to be a prodrug form of biologically active IL-2 that releases PEG chains over time to produce biologically active IL-2 ( PLoS One. 2017 Jul 5;l2(7):e0l7943l).
  • such pegylated IL-2 can be systemically administered and as such allows for constitutive exposure of NK/NK92 cells and their derivatives to IL-2 in vivo while at the same time systemic side effects of IL-2 are reduced, or even entirely avoided.
  • Continuous exposure advantageously is performed for at least 6 hours, or at least 12 hours, or at least 24 hours, or at least 36 hours, or at least 48 hours.
  • antibody conjugated IL-2 as such conjugates have shown increased stability, presumably due to decreased binding to serum proteins and decreased proteolysis by serum proteases.
  • antibody-drug conjugates will advantageously be administrable to a patient in vivo.
  • delivery of IL- 2 and with that activation of NK cells is possible with high specificity and selectivity as far as location is concerned.
  • such antibody-drug conjugates may target tumor markers that are patient and tumor specific (/. ⁇ ?
  • the antibody portion in such antibody-drug conjugates may be a full IgG antibody, or any suitable fragment thereof (e.g., scFv, Fab, Fab’, F(ab’)2, etc.).
  • IL-2 constitutive exposure of NK/NK92 cells and their derivatives to IL-2 (and modified forms of IL-2) will be at a concentration of between about 10-50 IU/mL, or between about 50-150 IU/mL, or between about 150-300 IU/mL, or between about 300-500 IU/mL, or between about 500- 1000 IU/mL, or even higher (as determined by CTLL-2 proliferation assay). Moreover, it is generally contemplated that the IL-2 concentration remains substantially constant over at least a limited period of time.
  • the concentration of the biologically active IL-2 fluctuates less than 25%, or less than 20%, or less than 15%, or less than 10%, or less than 7%, or less than 5%, or less than 3% as measured in % change of IU/mL over a period of 72 hours, or over a period of 60 hours, or over a period of 48 hours, or over a period of 36 hours, or over a period of 24 hours, or over a period of 18 hours.
  • suitable concentration of the biologically active IL-2 will be maintained throughout the entire cell culture between 50-70 IU/mL, or between 70- 100 IU/mL, or between 100-120 IU/mL, or between 120-150 IU/mL, or between 150-200 IU/mL, or between 200-230 IU/mL, or between 230-250 IU/mL, or between 250-280 IU/mL, or higher.
  • the genetically engineered NK cell may also be genetically engineered to express a chimeric T-cell receptor.
  • the chimeric T-cell receptor will have a scFv portion or other ectodomain with binding specificity against a tumor associated antigen, a tumor specific antigen, and a cancer neoepitope.
  • genetically engineering an NK cell to express such chimeric T-cell receptor there are numerous manners of genetically engineering an NK cell to express such chimeric T-cell receptor, and all manners are deemed suitable for use herein.
  • such cells may also be commercially obtained from NantKwest as taNK cells (‘target-activated natural killer cells’).
  • cancer associated antigens include CEA, MUC-l, CYPB1, etc.
  • cancer specific antigens include PSA, Her-2, PSA, brachyury, etc.
  • neoepitopes may be identified from a patient tumor in a first step by whole genome analysis of a tumor biopsy (or lymph biopsy or biopsy of a metastatic site) and matched normal tissue (/. ⁇ ? ., non-diseased tissue from the same patient) via synchronous comparison of the so obtained omics information.
  • the identified neoepitopes can then be further filtered for a match to the patient’s HLA type to increase likelihood of antigen presentation of the neoepitope. Most preferably, such matching can be done in silico.
  • all NK cells contemplated herein may also be genetically modified to express non- secreted IL-2 (e.g., retained in the ER compartment).
  • thusly stimulated NK cells as described herein may be used in a pharmaceutical composition or kit, typically formulated as a sterile injectable composition with between 10 4 -10 h cells, and more typically 10 5 -10 9 cells per dosage unit. Where desirable, these cells may be irradiated at a suitable radiation dosage to prevent further propagation after administration.
  • a pharmaceutical composition or kit typically formulated as a sterile injectable composition with between 10 4 -10 h cells, and more typically 10 5 -10 9 cells per dosage unit.
  • these cells may be irradiated at a suitable radiation dosage to prevent further propagation after administration.
  • alternative formulations are also deemed suitable for use herein, and all known routes and modes of administration are contemplated herein.
  • the term“administering” a pharmaceutical composition or drug refers to both direct and indirect administration of the pharmaceutical composition or drug, wherein direct administration of the pharmaceutical composition or drug is typically performed by a health care professional (e.g., physician, nurse, etc.), and wherein indirect administration includes a step of providing or making available the pharmaceutical composition or drug to the health care professional for direct administration (e.g., via injection into the tumor, infusion, oral delivery, topical delivery, etc.).
  • contemplated pharmaceutical compositions and kits may include a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion as described herein.
  • the inventor especially contemplates a method of treating cancer in which constitutively stimulated natural killer cells and chimeric protein contemplated herein are co administered.
  • such administration may follow various regimens.
  • the NK cell includes a high-affinity CD16 receptor (e.g., haNK cells)
  • the chimeric protein may be coupled to the NK cell via the receptor.
  • the NK cell is an autologous NK cell
  • the NK cells may be stimulated by constitutive exposure to IL-2 as described herein and then be separately from the chimeric protein (co-) administered to the patient.
  • the chimeric protein may be administered first, and after 12-24 hours or more, followed by administration of the stimulated NK cells.
  • the chimeric protein may be administered after treatment with the NK cells.
  • treatments may also include one or more steps that augment or trigger NKG2D ligand expression and presentation.
  • steps include low dose chemotherapy and/or low dose radiation therapy, typically performed at dosages that are equal or less than 50%, equal or less than 30%, equal or less than 20%, or equal or less than 10% of the maximum tolerated dose.
  • low dose treatment will preferably be performed in a metronomic fashion, for example, on alternating days, or every third day, or once weekly for several weeks, etc.
  • Embodiment 1 A method of inducing IFNy secretion in an NK cell, comprising: contacting the NK cell with a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion to induce interferon gamma (IFNy) secretion by the NK cell, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2.
  • IFNy interferon gamma
  • Embodiment 2 The method of Embodiment 1, wherein the IL-2 sensitized NK cell is an NK cell that was previously and constitutively exposed to IL-2, preferably at least 100 IU/mL IL-2.
  • Embodiment 3 The method of Embodiment 1 or 2, wherein any one or more of the following is true:
  • the genetically modified NK cell is an aNK cell, an autologous NK cell, or an NK cell derived in vitro from a precursor cell;
  • the genetically modified NK cell constitutively expresses and intracellularly retains IL-2;
  • the genetically modified NK cell is a haNK cell or an NK92MI cell
  • the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IE-12b chain.
  • Embodiment 4 The method of any one of the previous Embodiments, wherein the cancer cell targeting portion targets a necrosis marker or a cancer associated antigen or a cancer neoepitope, and wherein the necrosis marker is preferably a DNA, an RNA, or a histone protein, and wherein the cancer cell targeting portion preferably comprises hu5l-4 antibody or a portion thereof.
  • Embodiment 5 The method of any one of the previous Embodiments, wherein the chimeric protein comprises SEQ ID NO:l or SEQ ID NO:2, and optionally SEQ ID NO:3.
  • Embodiment 6 The method of any one of the previous Embodiments, wherein any one or more of the following is true:
  • the step of contacting the NK cell with the chimeric protein is performed ex vivo, and the NK cell is a haNK cell; and • the step of contacting the NK cell with the chimeric protein is performed in vivo, and the NK cell is a haNK cell or NK92MI cell.
  • Embodiment 7 A composition for use in treating cancer in a subject in need thereof, the composition comprising and NK cell and a chimeric protein,
  • NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2;
  • the chimeric protein comprises an IL-12 portion and a cancer cell targeting portion to thereby induce interferon gamma (IFNy) secretion from the NK cell.
  • IFNy interferon gamma
  • Embodiment 8 The composition of Embodiment 7, wherein the NK cell is a genetically modified NK cell that constitutively expresses and intracellularly retains IL-2, and wherein the NK cell is preferably a haNK cell or an NK92MI cell.
  • Embodiment 9 The composition of Embodiment 7 or 8, wherein any one or more of the following is true:
  • the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IE-12b chain;
  • the cancer cell targeting portion comprises an IgG antibody portion, a Fab portion, a F(ab’) 2 portion, a Fab 2 portion, or an scFv portion;
  • the cancer cell targeting portion targets a necrosis marker or a cancer associated antigen or a cancer neoepitope
  • the necrosis marker is a DNA, an RNA, or a histone protein
  • the cancer cell targeting portion comprises a hu5l-4 antibody or portion thereof
  • the chimeric protein comprises SEQ ID NO: l or SEQ ID NO:2, and optionally SEQ ID NO:3;
  • Embodiment 10 A chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion that targets a necrosis marker, and optionally wherein any one or more of the following is true:
  • the necrosis marker is a DNA, an RNA, or a histone protein
  • the cancer cell targeting portion comprises a hu51-4 antibody or portion thereof
  • the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IE-12b chain; • the chimeric protein comprises SEQ ID NO:l and optionally SEQ ID NO:3;
  • the chimeric protein comprises SEQ ID NO:2 and optionally SEQ ID NO:3 and
  • the cancer cell targeting portion comprises an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fat3 ⁇ 4 portion, or an scFv portion.
  • Embodiment 11 The chimeric protein of Embodiment 10, wherein the IL-12 portion is covalently coupled to the cancer cell targeting portion, and optionally wherein the IL-12 portion is covalently coupled to the cancer cell targeting portion via a peptide linker.
  • Embodiment 12 The chimeric protein of Embodiment 10 or 11, wherein the chimeric protein is bound to CD 16 of an NK cell.
  • Embodiment 13 A pharmaceutical composition comprising the chimeric protein of any one of Embodiments 10-12.
  • Embodiment 14 The pharmaceutical composition of Embodiment 13, wherein the composition further comprises an NK cell, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2, and optionally wherein any one or more of the following is true:
  • the NK cell is a haNK cell
  • the NK cell is an NK92MI cell
  • Embodiment 15 A chimeric antibody comprising a chimeric heavy chain that includes a scIL-l2 portion and antigen binding portion that binds a cell death-exposed antigen, and optionally wherein any one or more of the following is true:
  • the scIL-l2 comprises an IL-l2a subunit linked by a peptide linker to an IE-12b subunit;
  • the antibody comprises SEQ ID NO:l and optionally SEQ ID NO:3;
  • the antibody comprises SEQ ID NO:2 and optionally SEQ ID NO:3.
  • NK92 derivatives were exposed to IL-12.
  • the left panel of Fig.l comparatively depicts the data for aNK cells without constitutive IL-2 exposure and haNK cells with constitutive IL-2 exposure, while the right panel depicts NK92 derivatives that included stable integration of an IL-2 expression cassette (not expressing the high affinity CD16 variant).
  • the modified NK cells were not only responsive to IL-12 signaling to secrete IFNy, but secreted unexpected high quantities of IFNy (peaking near 200 ng/mL).
  • NK-92 cells were previously transfected with human IL-2 cDNA in a retroviral MFG-hIL-2 vector by particle-mediated gene transfer. The transfection was stable.
  • NK-92 and this derivative cell line NK-92MI had the following characteristics: surface marker positive for CD2, CD7, CDlla, CD28, CD45, CD54 and CD56 bright; surface marker negative for CD1, CD3, CD4, CD5, CD8, CD10, CD14, CD16, CD19, CD20, CD23, CD34 and HLA-DR.
  • Cells were grown following standard protocol. IFNy was measured using conventional ELISA methods well known in the art.
  • haNK cells (/. ⁇ ? ., aNK cell derivatives expressing high- affinity CD 16 and intracellularly retained IL-2) were cultured overnight in the presence of human recombinant IL-12 (rHuIL-l2), and selected human or murine recombinant IL-l2-Ab chimeric constructs (HuIL-l2/Hu5l-4 and MuIL-l2/Hu5l-4) having the sequence of SEQ ID NO: l and SEQ ID NO:2 or SEQ ID NO:l and SEQ ID NO:3.
  • Cells were cultured by seeding 2.5xl0 5 cells into a 24 well plate, X-Vivo 10 containing 5% human serum.
  • the cell culture supernatants were then collected and human IEN-g measured by ELISA.
  • constitutive IL-2 exposure via intracellular expression of IL-2 rendered haNK cells sensitive to IL-12 signaling as can be readily taken from the graphs in Fig.2.
  • exposure to IL-12 in various forms resulted in significant IFNy secretion for both murine and human IL-12.
  • human IL-12 produced somewhat stronger IFNy secretion in the haNK cells than murine IL-12.
  • the numbers expressing quantities of ingredients, properties such as concentration, reaction conditions, and so forth, used to describe and claim certain embodiments of the invention are to be understood as being modified in some instances by the term“about.” Accordingly, in some embodiments, the numerical parameters set forth in the written description and attached claims are approximations that can vary depending upon the desired properties sought to be obtained by a particular embodiment. In some embodiments, the numerical parameters should be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of some embodiments of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as practicable.
  • the phrase“at least one of A and B” is intended to refer to ‘A’ and/or ⁇ ’, regardless of the nature of ‘A’ and ⁇ ’.
  • ‘A’ may be single distinct species, while in other embodiments‘A’ may represent a single species within a genus that is denoted‘A’.
  • ⁇ ’ may be single distinct species, while in other embodiments ⁇ ’ may represent a single species within a genus that is denoted ⁇ ’ .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Mycology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Compositions and methods for NK cell based treatments, and particularly NK cells that express and intracellularly retain IL-2, are presented in which the NK cells are stimulated with a chimeric protein that has a cancer cell targeting portion and an IL-12 portion. Beneficially, such chimeric protein has substantially reduced systemic toxicity and induces IFN-γ secretion in a targeted manner. Moreover, chimeric proteins contemplated herein also significantly enhanced IFN-γ secretion in NK cells that express and intracellularly retain IL-2 as compared to native NK cells. Preferred chimeric proteins comprise SEQ ID NO:1 or SEQ ID NO:2, and SEQ ID NO:3.

Description

TARGETED IL-12 TREATMENTS AND METHODS TO STIMULATE haNK AND
NK92mi CELLS
[0001] This application claims priority to our US provisional patent application with the serial number 62/728510, which was filed September 7, 2018, which is incorporated by reference herein.
Field of the Invention
[0002] The field of the invention is cancer treatments and methods using natural killer cells and immune stimulatory cytokines, and especially activated NK cells and IL-12.
Background of the Invention
[0003] The background description includes information that may be useful in understanding the present invention. It is not an admission that any of the information provided herein is prior art or relevant to the presently claimed invention, or that any publication specifically or implicitly referenced is prior art.
[0004] All publications and patent applications herein are incorporated by reference to the same extent as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. Where a definition or use of a term in an incorporated reference is inconsistent or contrary to the definition of that term provided herein, the definition of that term provided herein applies and the definition of that term in the reference does not apply.
[0005] More recently, cell-based cancer treatments with genetically modified NK cells have gained attention due to positive treatment outcomes, particularly where NK92 derivatives such as activated NK cells (aNK cells), genetically modified NK cells with high affinity CD 16 receptors (haNK cells), or chimeric antigen receptors (taNK cells) were used. While such cell-based treatments are conceptually attractive, the tumor microenvironment and other patient- specific factors will often reduce their cytotoxic activity, and various attempts have been made to modulate cytotoxicity in NK92 cells.
[0006] Interleukin-2 (IL-2) and interleukin- 12 (IL-12) cytokines elicit strong antitumor effects by stimulating unmodified immune cells, including T cells and natural killer (NK) cells. Although either cytokine stimulates the proliferation of T cells, the production of interferon-g (IFN-g) by NK cells, and ultimately the cytolytic activity, the magnitude, and the spectrum of stimulatory effects by IL-2 and IL-12 are different (see e.g., J. Leukoc. Biol. 58: 225-233; 1995). For example, while IL-2 is a stronger stimulator of proliferation and cytolytic activity, IL-12 is a stronger inducer of IFN-g from unmodified NK cells and activated T cells. IFN-g mRNA has increased stability in NK cells co-stimulated with IL-2 and IL-12 (see e.g., Molecular and Cellular Biology, Mar. 2002, p. 1742-53). However, the IL-2 and IL-12 concentrations used in vitro may not necessarily reflect achievable or even desirable levels in vivo. Indeed, IL-2 systemic administration of IL-2 is associated with relatively high toxicity and capillary leak syndrome, while several deaths have been attributed to systemic administration of IL-12. In addition, the response of primary NK cells to various cytokines is not necessarily the same as the response of genetically modified NK92 cells.
[0007] Therefore, even though various compositions and methods are known to activate NK cells, there remains a need for compositions and methods to treat cancer using NK cells, especially where the NK cells are stimulated with IL-12 in a clinically safe manner.
Summary of the Invention
[0008] Disclosed herein are various compositions and methods of NK cell based treatments. In particular, the present disclosure describes NK cells that express and intracellularly retain IL-2, in which the NK cells are stimulated with a chimeric protein that comprises a cancer cell targeting portion and an IL-12 portion. Advantageously, such chimeric protein has substantially reduced systemic toxicity and moreover beneficially induces IFN-g secretion in a targeted manner. Unexpectedly, such chimeric proteins significantly enhance IFN-g secretion in NK cells that express and intracellularly retain IL-2 as compared to native NK cells.
[0009] In one aspect of the inventive subject matter, the inventor contemplates a method of inducing IFNy secretion in an NK cell that includes one step of providing an NK cell, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2; and another step of contacting the NK cell with a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion to induce interferon gamma (IFNy) secretion by the NK cell. [0010] In some embodiments, the IL-2 sensitized NK cell is an NK cell that was previously and constitutively exposed to IL-2 or an NK cell was that constitutively exposed to at least 100 IU/mL IL-2. In other embodiments, the genetically modified NK cell may also be an aNK cell, an autologous NK cell, or an NK cell derived in vitro from a precursor cell, while in still further embodiments the genetically modified NK cell constitutively expresses and intracellularly retains IL-2 (e.g., haNK cell or an NK92MI cell).
[0011] It is further contemplated that the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IL- 12b chain, but homodimers of IL-l2a chain and other heterodimers are also deemed suitable. In some embodiments, the cancer cell targeting portion comprises an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion. Most typically, the cancer cell targeting portion targets a necrosis marker (e.g., DNA, RNA, histone protein, etc.) or a cancer associated antigen or a cancer neoepitope to so afford site specificity with respect to IL-12 action. For example, suitable cancer cell targeting portions may comprise a hu5l-4 antibody or portion thereof.
[0012] While not limiting to the inventive subject matter, it is contemplated that the step of contacting the NK cell with the chimeric protein is performed ex vivo and the NK cell is a haNK cell, or that the step of contacting the NK cell with the chimeric protein is performed in vivo and wherein the NK cell is a haNK cell or NK92MI cell.
[0013] Therefore, and viewed from a different perspective, the inventor also contemplates a method of treating cancer that includes a step of administering an NK cell to a patient, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2; and a further step of administering a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion to thereby induce interferon gamma (IFNy) secretion by the NK cell.
[0014] As noted above, it is contemplated that the NK cell may be a genetically modified NK cell that constitutively expresses and intracellularly retains IL-2 (e.g., haNK cell or NK92MI cell), and/or that the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IL-12 chain. In other examples, the cancer cell targeting portion may comprise an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion, and it is generally preferred that the cancer cell targeting portion targets a necrosis marker (e.g., DNA, RNA, histone protein, etc.) or a cancer associated antigen or a cancer neoepitope. For example, the cancer cell targeting portion may comprise a hu5l-4 antibody or portion thereof.
[0015] In further embodiments it is contemplated that the chimeric protein is administered at least 12 hours prior to administration of the NK cell, or that the chimeric protein is administered at least 12 hours after administration of the NK cell, or that the chimeric protein is contemporaneously administered with the NK cell. Among other options, it is also contemplated that the chimeric protein is bound to the NK cell.
[0016] In yet another aspect of the inventive subject matter, the inventor also contemplates a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion that targets a necrosis marker (e.g., DNA, RNA, histone protein, etc.), and/or that the cancer cell targeting portion comprises a hu5l-4 antibody or portion thereof. In other embodiments, the cancer cell targeting portion includes an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion.
[0017] For example, the IL-12 portion may be covalently coupled to the cancer cell targeting portion, and/or the IL-12 portion is covalently coupled to the cancer cell targeting portion via a peptide linker. Typically, but not necessarily, the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IL- 12b chain, and/or the cancer cell targeting portion comprises a hu5l-4 antibody or portion thereof. Where desirable, the chimeric protein may be non-co valently bound to the CD 16 receptor of an NK cell.
[0018] Consequently, the inventor also contemplates a pharmaceutical composition comprising the chimeric protein contemplated herein. Such pharmaceutical composition may further comprise an NK cell as described above. Thus, the inventor also contemplates a kit comprising (a) an NK cell, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2; and (b) a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion. With respect to the NK cell and the chimeric protein, the same considerations as provided above apply.
[0019] Viewed from a different perspective, the inventor therefore also contemplates use of a chimeric protein and an NK cell in the treatment of cancer, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2, and wherein the chimeric protein comprises an IL-12 portion and a cancer cell targeting portion that induces interferon gamma (IFNy) secretion by the NK cell. With respect to the NK cells and/or the chimeric protein, the same considerations as noted above apply.
[0020] Various objects, features, aspects and advantages of the inventive subject matter will become more apparent from the following detailed description of preferred embodiments, along with the accompanying drawing figures in which like numerals represent like components.
Brief Description of the Drawings
[0021] Fig.1 shows exemplary graphs indicating that the tested NK cells require constitutive IL-2 expression/exposure for significant IL-12 mediated IFN-g secretion.
[0022] Fig.2 is an exemplary graph showing the biological activity of IL-12 and selected chimeric proteins comprising an IL-12 portion with respect to IL-12 mediated IFN-g secretion.
[0023] Fig.3 is an exemplary graph showing binding of selected chimeric proteins comprising an IL-12 portion linked to antigens exposed by cell death.
[0024] Fig.4 shows sequences for exemplary chimeric proteins comprising an IL-12 portion.
Detailed Description
[0025] The inventor has now discovered that selected chimeric molecules comprising an IL- 12 portion can be used in an effective and targeted manner to induce IFNy secretion in IL-2 sensitized NK cells or in genetically modified NK cells that constitutively express IL-2 to thereby promote cytotoxic NK cell activity in a target tissue. Advantageously, the chimeric molecules comprising the IL-12 portion help avoid toxicity otherwise encountered with systemically administered IL-12. Moreover, as the chimeric molecule also includes a cancer targeting moiety (e.g., targeting one or more necrosis markers), induction of IFNy secretion by the NK cells can be rendered specific to the site of the cancer, and particularly to the hypoxic and/or necrotic tumor core. Therefore, due to the target specific IFNy secretion, contemplated compositions and methods will increase antigen presentation and MHC-I/II expression of immune competent cells (and especially CD4+ and/or CD8+ T cells) in a tumor, and bias the immune response in the tumor towards a Thl response. [0026] Advantageously, use of the chimeric molecules presented herein not only avoids toxicity otherwise encountered with systemically administered IL-12, but also affords concentrated therapeutically effective quantities of IL-12 at the targeted tumor site. In addition, it should be appreciated that use of necrosis markers in the context presented herein will advantageously allow for a tumor antigen independent antigen spread in the tumor microenvironment as the chimeric molecule will indiscriminately target necrotic tissue rather than cancer or tumor associated antigens. In still further beneficial aspects, the chimeric molecules presented herein retain the biological activity of the IL-12 heterodimer as is discussed in more detail below.
[0027] In addition, it should be particularly recognized that various NK cells will fail to respond to IL-12, even where propagated with IL-2 supplied into the culture medium. Notably, however, the inventor has also discovered that when IL-2 is supplied to the same cells in a constitutive or continuous manner, the cells are sensitive to IL-12 signaling. The NK cells treated in this manner produce significant quantities of IFNy and have increased cytotoxicity, presumably due to increased intracellular concentrations and/or less fluctuations in IL-2 quantities.
[0028] The terms‘constitutive exposure’ or‘constitutively exposing’ in conjunction with IL- 2 as used herein means that biologically active IL-2 is supplied or produced in a continuous (or semi-continuous) manner such that variations in biologically active IL-2 concentration at or in the cell (or IL-2 stimulation) will vary by no more than 20% over 48 hours. Therefore, in some embodiments, variations in biologically active IL-2 concentration at or in the cell (or IL-2 stimulation) will vary by no more than 15% over 48 hours, or no more than 10% over 48 hours, or no more than 7% over 48 hours, or no more than 5% over 48 hours, or no more than 1% over 48 hours. Likewise, the terms “constitutive expression” or “constitutively expressing” in conjunction with IL-2 as used herein refers to (recombinant) expression of IL- 2 within a cell in a continuous fashion from a constitutive promoter (/.<?., without the need for induction). Most typically, such expression of IL-2 within a cell will be achieved by use of an endoplasmic retention sequence that is fused to the IL-2 to reduce or even entirely avoid secretion form the cell expressing such modified IL-2.
[0029] While not limiting to the inventive subject matter and without being bound by theory, constitutive exposure to IL-2 is hypothesized to resemble more closely the exposure of NK cells to IL-2 in a natural setting, leading to durable sensitivity to IL-12 and IFNy secretion triggered by exposure to IL-12. In contrast, intermittent exposure of NK-92 cells to IL-2— as is the case in customary NK-92 cell culture, where culture media are renewed every two to three days— may lead to inactivation and/or degradation of biologically active IL-2, possibly due to binding by serum proteins or proteolysis by serum proteases of the culture media. Consequently, and without wishing to be bound by any theory or hypothesis, the inventor believes that customary NK-92 cell culture conditions promote intermittent or‘pulsed’ IL-2 signaling which supports growth of NK-92 cells but fails to support cell signaling to render the NK-92 cells sensitive to IL-12 and IL-12 dependent IFNy secretion as well as increased (as compared to non-IL-l2 stimulated cells) expression of NKG2D.
[0030] With respect to contemplated chimeric proteins it is generally contemplated that all chimeric proteins that include at least a biologically active IL-12 portion are suitable. Moreover, it is generally preferred that the chimeric protein also includes a portion that extends serum half life time, that reduces systemic toxicity, and/or that allows target specific delivery and/or accumulation of IL-12 at a desired site. Consequently, with regard to the non- IL-12 portion of the chimeric protein, suitable non-IL-l2 portions include various serum proteins and portions thereof, and particularly albumin, lactoferrin, and various globulins. Therefore, one preferred non-IL-l2 portion comprises an antibody or fragment thereof. As will be readily appreciated, use of antibodies or fragments thereof will beneficially allow targeting while also extending the stability and persistence of the chimeric protein in blood. For example, the non-IL-l2 portion may be a heavy chain of an antibody, a light chain of an antibody (or portion thereof), or a scFv.
[0031] While any binding specificity for such antibody or fragment thereof is generally deemed suitable, especially preferred antibodies will specifically bind to an antigen of a tumor cell. For example, suitable antibodies may bind to a tumor associated antigen, a tumor specific antigen, or a patient and tumor specific neoantigen (neoepitope). Therefore, the non- IL-12 portion is most preferably also used to deliver and retain the IL-12 at the site of a tumor to concentrate the biological effect of the IL-12 to the target site. Furthermore, it should be recognized that all sources of antibodies are deemed suitable, including human antibodies and all isoforms, non-human mammalian forms, humanized forms, and synthetic antibodies. Where the non-IL-l2 portion is not an antibody, tumor selectivity or preference may nevertheless be achieved via albumin that is preferentially taken up into tumor cells from the neovasculature via gp60 mediated transcytosis. [0032] With respect to the IL-12 portion, it should be noted that IL-12 is a heterodimer and that the IL-12 portion in the chimeric protein may be a single chain form of IL-12 (/.<?., scIL- 12) or a single subunit (/.<?., IL-l2a subunit (p35) and IL- 12b subunit (p40)) or active portion thereof. With respect to the source of the IL-12, it is generally preferred that the source be human IL-12. However, various other non-human (preferably mammalian) IL-12 forms are also expressly contemplated herein. Moreover, it should be appreciated that splice variants and mutants are also appropriate, so long as such forms exhibit at least some biological activity (e.g., induce IFNy secretion in haNK cells as described herein).
[0033] Suitable IL-12 protein sequences include human sequences, which may form a disulfide linked heterodimer formed from IL-l2a subunit (p35) and IL- 12b subunit (p40) or which may be covalently linked together via a flexible peptide linker as described in more detail below. Among other suitable sequences, IL-12 sequences include publically available protein sequence (e.g., P29460 and P29459 at UniProt), and all natural and synthetic variants thereof, including truncated forms, splice variants, and mutant forms. Likewise, DNA and RNA sequences encoding IL-12 are known in the art and suitable sequences include those at Genbank (accession number AC010370 for genomic sequence and M65271 for mRNA of IL- 12a subunit; accession number AF512686 for genomic sequence and M65290 for mRNA of KM2b subunit). Moreover, it should be appreciated that other non-human, preferably mammalian sequences, are also deemed suitable for use herein.
[0034] Heterodimers of IL-l2a subunit and IL- 12b subunit may be formed by non-covalent association, or via a flexible peptide linker such as a G4S linker to form a single chain IL-12 molecule. Furthermore, the chimeric proteins may also include addressing or leader sequences, linkers, tags, and/or flag for quantification and/or simplified purification. The proper choice will depend, at least in part, on the particular choice of production system, as is well known in the art.
[0035] It is still further contemplated that the manner of production of a chimeric protein with the IL-12 portion and the non-IL-l2 portion may vary considerably, and will include covalent coupling of the two portions heavy chain as well as non-covalent coupling. For example, a necrosis targeting antibody hu5l-4 was used as a fusion partner with a single chain IL-12 (scIL-l2) as is described in more detail below. Likewise, NHS-IL12 is a chimeric construct that includes two molecules of IL-12 that are fused to a tumor necrosis-targeting human IgGl (NHS76). The addition of the human IgGl moiety resulted in a longer plasma half-life of NHS-IL12 than recombinant IL-12, and a selective targeting to murine tumors in vivo. Data from both in vitro assays using human PBMCs and in vivo primate studies showed that IFNy production by immune cells was attenuated following treatment with the immunocytokine, suggesting an improved toxicity profile than seen with recombinant IL-12 alone. NHS-IL12 was also superior to recombinant IL-12 when evaluated as an anti-tumor agent in three murine tumor models (see e.g., Oncotarget 2014 Apr 15 ;5(7): 1869-84).
[0036] As will be readily appreciated, such antibody-cytokine conjugates may be prepared by chemical conjugation using cleavable (e.g., via disulfide bond or hydrazone, or proteolytic site, etc.) or non-cleavable linkers (e.g., via maleimide-modified PEG). Alternatively, the conjugation may also be done using recombinant cloning in which the N- or C-terminus of the heavy or light chain (or fragment thereof) is modified to also encode in frame a linker portion and IL-12 as is also shown in more detail below. Thus, chimeric recombinant proteins can be prepared that have an antibody portion that preferably binds to a component of a tumor cell, a linker, and an IL-12 portion. Notably, exemplary antibody-drug conjugates with IL-12 retained significant activity as is shown in more detail below.
[0037] One exemplary fusion protein may therefore include a chimeric protein having a leader peptide portion, a murine scIL-l2 portion in normal type font (G4S linker in bold type), and a hu5l-4 antibody heavy chain portion as shown below in SEQ ID NO:l, in which the italicized sequence denotes the leader peptide portion, and in which the underlined sequence denotes the hu5l-4 antibody heavy chain portion.
[0038] MCtfeeLWSWFSLVFLASPLMWELEKDVYVVEVDWTPDAPGETVNLTCDTPE
EDDITWTSDQRHGVIGSGKTLTITVKEFLDAGQYTCHKGGETLSHSHLLLHKKENGI
WSTEILKNFKNKTFLKCEAPNYSGRFTCSWLVQRNMDLKFNIKSSSSSPDSRAVTCG
MASFSAEKVTFDQRDYEKYSVSCQEDVTCPTAEETFPIEFAFEARQQNKYENYSTSF
FIRDIIKPDPPKNLQMKPLKNSQVEVSWEYPDSWSTPHSYFSLKFFVRIQRKKEKMKE
TEEGCNQKGAFLVEKTSTEVQCKGGNVCVQAQDRYYNSSCSKWACVPCRVRSGGG
GSGGGGSGGGGSRVIPVSGPARCLSQSRNLLKTTDDMVKTAREKLKHYSCTAEDID
HKDITRDQTSTLKTCLPLELHKNESCLATRETSSTTRGSCLPPQKTSLMMTLCLGSIYE
DLKMYQTEFQAINAALQNHNHQQIILDKGMLVAIDELMQSLNHNGETLRQKPPVGE
ADPYRVKMKLCILLHAFSTRVVTINRVMGYLSSALEOVOLVOSGAEVKKPGASVKV
SCKASGYTFTRYWMHWVROAPGOGLEWIGAIYPGNSDTSYNOKFKGKATITADTST
NTAYMELSSLRSEDTAVYYCARGEEIGSRRWFAYWGOGTLVTVSSASTKGPSVFPL APSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSV
VTVPSSSLGTOTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLF
PPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNST
YRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRD
ELTKNOVSLTCLVKGFYPSDIAVEWESNGOPENNYKTTPPVLDSDGSFFLYSKLTVD
KSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK. rSEO ID NO: l]
[0039] Another exemplary fusion protein may include a leader peptide portion, a human scIL-l2 portion in normal type font (G4S linker in bold type), and a hu5l-4 antibody heavy chain portion as shown below in SEQ ID NO:2, in which the italicized sequence denotes the leader peptide portion, and in which the underlined sequence denotes the hu5 l-4 antibody heavy chain portion.
[0040] MCtfeeLWSWFSLVF ASPLIWELKKDVYVVELDWYPDAPGEMVVLTCDTPEE
DGITWTLDQSSEVLGSGKTLTIQVKEFGDAGQYTCHKGGEVLSHSLLLLHKKEDGIW
STDILKDQKEPKNKTFLRCEAKNYSGRFTCWWLTTISTDLTFSVKSSRGSSDPQGVTC
GAATLS AERVRGDNKEYEY S VECQEDS ACPAAEESLPIEVMVD AVHKLKYENYTS S
FFIRDIIKPDPPKNLQLKPLKNSRQVEV S WEYPDTWSTPHS YFSLTFC V QV QGKS KRE
KKDRVFTDKTSATVICRKNASISVRAQDRYYSSSWSEWASVPCSGGGGSGGGGSG
GGGSRNLPVATPDPGMFPCLHHSQNLLRAVSNMLQKARQTLEFYPCTSEEIDHEDIT
KDKTSTVEACLPLELTKNESCLNSRETSFITNGSCLASRKTSFMMALCLSSIYEDLKM
YQVEFKTMNAKLLMDPKRQIFLDQNMLAVIDELMQALNFNSETVPQKSSLEEPDFY
KTKIKLCILLHAFRIRAVTIDRVMSYLNASLEOVOLVOSGAEVKKPGASVKVSCKAS
GYTFTRYWMHWVROAPGOGLEWIGAIYPGNSDTSYNOKFKGKATITADTSTNTAY
MEFSSFRSEDTAVYYCARGEEIGSRRWFAYWGOGTFVTVSSASTKGPSVFPFAPSSK
STS GGT A ALGCL VKD YFPEPVT V S WN S G ALTS G VHTFPA VLO S S GLY SLSS V VT VPS
SSLGTOTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPK
DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEOYNSTYRVVS
VLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKN
O VS LTCLVKGFYPSDI A VEWESN GOPENN YKTTPPVLD SDGSFFLY S KLT VD KSRW O
QGNVFSCSVMHEALHNHYTQKSLSLSPGK. [SEQ ID NO:2]
[0041] Still another exemplary protein includes a leader peptide portion and a hu5l-4 antibody light chain portion as shown below in SEQ ID NOG, in which the italicized sequence denotes the leader peptide portion, and in which the underlined sequence denotes the hu5l-4 antibody light chain portion.
[0042] MDFOVOIFSFLLISASVAMSRGEWLEOSFAELSLSFGERAELSCRAROSlSNYUi WYQQKPGQAPRLLIYYASQSISGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQSNS WPLTFGOGTKVEIKRTVAAPSVFIFPPSDEOLKSGTASVVCLLNNFYPREAKVOWKV DNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTK SFNRGEC. [SEQ ID NO: 3]
[0043] With respect to NK cells, it is generally contemplated that preferred NK cells are IL-2 sensitized NK cells or genetically modified NK cells that constitutively expresses IL-2. In this context, it should be noted that IL-2 sensitized cells are cells that were subjected to constitutive exposure with IL-2 as noted above.
[0044] While constitutive exposure can be done by continuous exposure and/or replenishment of IL-2 to the culture medium as described herein, constitutive exposure of NK/NK92 cells and their derivatives to IL-2 is more preferably achieved by intracellular expression of recombinant IL-2 in the respective cells. Typically, intracellular expression is driven from a constitutively active promoter to achieve constant expression levels, and is expressed in a form that is not secreted (/.<?., lacks export signal sequence, and may include an endoplasmic or cytoplasmic retention sequence). Such intracellular expression is believed to provide the same functional impact to the cell as constitutive exposure to externally provided IL-2. Indeed, as is shown in more detail below, the inventor discovered that where NK92 cells or derivatives were genetically engineered to express and intracellularly retain IL-2, the cells were unexpectedly sensitized to IL-12 stimulation as measured by IFNy secretion and/or increased expression of NKG2D. As will be readily appreciated, recombinant expression and intracellular retention of IL-2 can be done in numerous manners, and all of such methods are deemed suitable for use herein (see e.g., Oncotarget 2016 Dec 27; 7(52): 86359-86373 or Exp Hematol. 2005 Feb;33(2): 159-64). Among other benefits, it should be noted that such recombinant cells can be administered to a patient in vivo without the need to administer to the same patient IL-2. Such modified cells will be sensitized to IL-12 to secrete IFNy upon IL-12 stimulation. Indeed, sensitization by constitutive exposure (external or internal) to IL-2 provided substantial quantities of IFNy that is thought to provide a therapeutic effect in the context of concomitant immune therapy, particularly as NKG2D expression in such stimulated cells was also significantly increased. [0045] For example, genetically engineered NK cells may be NK-92 derivatives modified to express the high-affinity Fey receptor (CD16). Sequences for high-affinity variants of the Fey receptor are well known in the art (see e.g., Blood 2009 113:3716-3725), and all manners of generating and expression are deemed suitable for use herein. Expression of such receptor is believed to allow specific targeting of tumor cells using antibodies that are specific to a patient’s tumor cells (e.g., neoepitopes), a particular tumor type (e.g., her2neu, PSA, PSMA, etc.), or that are associated with cancer (e.g., CEA-CAM). Advantageously, such antibodies are commercially available and can be used in conjunction with the cells (e.g., bound to the Fey receptor), and such modified cells may also be commercially obtained from NantKwest as haNK cells (‘high-affinity natural killer cells). Alternatively, NK92MI cells have been reported (see e.g., Hum Gene Ther. 1999 May 20;l0(8):l359-73 or US 8034332) to express and retain IL-2 intracellularly and are commercially available from ATCC.
[0046] In still further contemplated aspects, and with respect to NK cells it is contemplated that all NK cells are deemed suitable for use herein so long as such cells are constitutively exposed to IL-2 or intracellularly express (and retain) IL-2 as described above. Thus suitable alternative NK cells include autologous NK cells from a patient (e.g., isolated from whole blood, or cultivated from precursor or stem cells using methods known in the art), and various allogenic NK cells. However, in preferred aspects of the inventive subject matter, the NK cells are genetically engineered to achieve one or more desirable traits, and particularly include NK-92 cells and derivatives thereof. For example, suitable genetically engineered NK cell include NK-92 derivatives that are modified to have reduced or abolished expression of at least one killer cell immunoglobulin-like receptor (KIR), which will render such cells constitutively activated (via lack of or reduced inhibition).
[0047] Therefore, suitable NK cells may have one or more modified KIR that are mutated such as to reduce or abolish interaction with MHC class I molecules. Of course, it should be noted that one or more KIRs may also be deleted or expression may be suppressed (e.g., via miRNA, siRNA, etc.). Most typically, more than one KIR will be mutated, deleted, or silenced, and especially contemplated KIR include those with two or three domains, with short or long cytoplasmic tail. Viewed from a different perspective, modified, silenced, or deleted KIRs will include KIR2DL1, KIR2DL2, KIR2DL3, KIR2DL4, KIR2DL5A, KIR2DL5B, KIR2DS1, KIR2DS2, KIR2DS3, KIR2DS4, KIR2DS5, KIR3DL1, KIR3DL2, KIR3DL3, and/or KIR3DS1. Such modified cells may be prepared using protocols well known in the art. Alternatively, such cells may also be commercially obtained from NantKwest (see URL www.nantkwest.com) as aNK cells (‘activated natural killer cells).
[0048] NK-92 cells exhibit an unusual receptor expression profile, expressing a relatively large number of activating (e.g., NKp30, NKp46, 2B4, NKGD, CD28) receptors. Conversely, NK-92 cells also express few inhibitory receptors (e.g., NKGA/B, low levels of KIR2DL4, ILT-2), and lack most of the killer inhibitory receptors (KIRs) clonally expressed on normal NK cells. In addition, NK-92 expresses relatively high levels of molecules involved in the perforin-granzyme cytolytic pathway as well as additional cytotoxic effector molecules including tumor necrosis factor (TNF)-superfamily members FasL, TRAIL, TWEAK, TNF- alpha, indicating the ability to kill via alternative mechanisms. Moreover, NK-92 cells also express other molecules implicated immune effector cell regulation (CD80, CD86, CD40L, TRANCE) whose relevance in NK killing is unclear.
[0049] Therefore, the inventor contemplates that NK-92 cells and derivatives thereof can be sensitized to IL-12 and exhibit an IL-12 dependent IFNy secretion phenotype by constitutively exposing the cells to IL-2. For example, in one embodiment constitutive exposure can be performed by continuous or semi-continuous addition of IL-2 to a culture medium to thereby maintain the concentration of biologically active IL-2 substantially constant. Therefore, it is contemplated that the concentration of biologically active IL-2 in the medium varies over 48 hours by no more than 15%, or by no more than 10%, or by no more than 7%, or by no more than 5%, or by no more than 3%. Biological activity of IL-2 can be quantified using known procedures, e.g., using a CTLL-2 cell proliferation assay (J Immunol Methods. 2009 Aug 31; 348(1-2): 83-94).
[0050] Continuous or semi-continuous addition of IL-2 can be done in numerous manners, including use of a peristaltic pump or metered injector. Alternatively, and in less preferred aspects, continuous or semi-continuous addition of IL-2 can be done by media renewal in a frequent fashion (e.g., every two hours, every four hours, every eight hours, etc.). Where multiple or continuous additions are not preferred, the inventor also contemplates that the constitutive exposure can also be achieved using formulations that release IL-2 in a relatively slow manner. For example, delayed release of IL-2 (or increased stability against protein binding and/or protease digest) can be done by pegylation of IL-2 as is known from NKTR- 214 (Nektar Therapeutics; 455 Mission Bay Blvd South; San Francisco, CA 94158). Here, pegylated IL-2 is believed to be a prodrug form of biologically active IL-2 that releases PEG chains over time to produce biologically active IL-2 ( PLoS One. 2017 Jul 5;l2(7):e0l7943l). Advantageously, such pegylated IL-2 can be systemically administered and as such allows for constitutive exposure of NK/NK92 cells and their derivatives to IL-2 in vivo while at the same time systemic side effects of IL-2 are reduced, or even entirely avoided. Continuous exposure advantageously is performed for at least 6 hours, or at least 12 hours, or at least 24 hours, or at least 36 hours, or at least 48 hours.
[0051] Alternatively, or additionally, it should be noted that constitutive exposure can also be achieved using antibody conjugated IL-2 as such conjugates have shown increased stability, presumably due to decreased binding to serum proteins and decreased proteolysis by serum proteases. Once more, such antibody-drug conjugates will advantageously be administrable to a patient in vivo. In this embodiment, however (and in contrast to NKTR-214), delivery of IL- 2 and with that activation of NK cells, is possible with high specificity and selectivity as far as location is concerned. For example, such antibody-drug conjugates may target tumor markers that are patient and tumor specific (/.<?., tumor neoepitopes), cancer associated, cancer specific, or specific to necrotic tissue commonly found in a tumor microenvironment. Of course, it should be appreciated that the antibody portion in such antibody-drug conjugates may be a full IgG antibody, or any suitable fragment thereof (e.g., scFv, Fab, Fab’, F(ab’)2, etc.).
[0052] Regardless of the particular form of IL-2 it is generally contemplated that constitutive exposure of NK/NK92 cells and their derivatives to IL-2 (and modified forms of IL-2) will be at a concentration of between about 10-50 IU/mL, or between about 50-150 IU/mL, or between about 150-300 IU/mL, or between about 300-500 IU/mL, or between about 500- 1000 IU/mL, or even higher (as determined by CTLL-2 proliferation assay). Moreover, it is generally contemplated that the IL-2 concentration remains substantially constant over at least a limited period of time. For example, it is typically preferred that the concentration of the biologically active IL-2 fluctuates less than 25%, or less than 20%, or less than 15%, or less than 10%, or less than 7%, or less than 5%, or less than 3% as measured in % change of IU/mL over a period of 72 hours, or over a period of 60 hours, or over a period of 48 hours, or over a period of 36 hours, or over a period of 24 hours, or over a period of 18 hours. Thus, viewed from a different perspective, suitable concentration of the biologically active IL-2 will be maintained throughout the entire cell culture between 50-70 IU/mL, or between 70- 100 IU/mL, or between 100-120 IU/mL, or between 120-150 IU/mL, or between 150-200 IU/mL, or between 200-230 IU/mL, or between 230-250 IU/mL, or between 250-280 IU/mL, or higher.
[0053] In yet a further aspect of the inventive subject matter, the genetically engineered NK cell may also be genetically engineered to express a chimeric T-cell receptor. In especially preferred aspects, the chimeric T-cell receptor will have a scFv portion or other ectodomain with binding specificity against a tumor associated antigen, a tumor specific antigen, and a cancer neoepitope. As noted before, there are numerous manners of genetically engineering an NK cell to express such chimeric T-cell receptor, and all manners are deemed suitable for use herein. Alternatively, such cells may also be commercially obtained from NantKwest as taNK cells (‘target-activated natural killer cells’).
[0054] Where the cells are engineered to have affinity towards a cancer associated antigen or antibody with specificity towards a cancer associated antigen (e.g., via expression of a CAR), it is contemplated that all known cancer associated antigens are considered appropriate for use. For example, cancer associated antigens include CEA, MUC-l, CYPB1, etc. Likewise, where the cells are engineered to have affinity towards a cancer specific antigen or antibody with specificity towards a cancer specific antigen, it is contemplated that all known cancer specific antigens are considered appropriate for use. For example, cancer specific antigens include PSA, Her-2, PSA, brachyury, etc. Where the cells are engineered to have affinity towards a cancer neoepitope or antibody with specificity towards a cancer neoepitope, it is contemplated that all known manners of identifying neoepitopes will lead to suitable targets. For example, neoepitopes may be identified from a patient tumor in a first step by whole genome analysis of a tumor biopsy (or lymph biopsy or biopsy of a metastatic site) and matched normal tissue (/.<?., non-diseased tissue from the same patient) via synchronous comparison of the so obtained omics information. The identified neoepitopes can then be further filtered for a match to the patient’s HLA type to increase likelihood of antigen presentation of the neoepitope. Most preferably, such matching can be done in silico. In addition, all NK cells contemplated herein may also be genetically modified to express non- secreted IL-2 (e.g., retained in the ER compartment).
[0055] Therefore, in yet another aspect of the inventive subject matter, thusly stimulated NK cells as described herein may be used in a pharmaceutical composition or kit, typically formulated as a sterile injectable composition with between 104-10h cells, and more typically 105-109 cells per dosage unit. Where desirable, these cells may be irradiated at a suitable radiation dosage to prevent further propagation after administration. However, alternative formulations are also deemed suitable for use herein, and all known routes and modes of administration are contemplated herein. As used herein, the term“administering” a pharmaceutical composition or drug refers to both direct and indirect administration of the pharmaceutical composition or drug, wherein direct administration of the pharmaceutical composition or drug is typically performed by a health care professional (e.g., physician, nurse, etc.), and wherein indirect administration includes a step of providing or making available the pharmaceutical composition or drug to the health care professional for direct administration (e.g., via injection into the tumor, infusion, oral delivery, topical delivery, etc.). Additionally, or alternatively, contemplated pharmaceutical compositions and kits may include a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion as described herein.
[0056] Therefore, the inventor especially contemplates a method of treating cancer in which constitutively stimulated natural killer cells and chimeric protein contemplated herein are co administered. As will be readily appreciated, such administration may follow various regimens. For example, where the NK cell includes a high-affinity CD16 receptor (e.g., haNK cells), the chimeric protein may be coupled to the NK cell via the receptor. On the other hand, for example, where the NK cell is an autologous NK cell, the NK cells may be stimulated by constitutive exposure to IL-2 as described herein and then be separately from the chimeric protein (co-) administered to the patient. Thus, the chimeric protein may be administered first, and after 12-24 hours or more, followed by administration of the stimulated NK cells. On the other hand, where a patient has already received a transfusion with NK cells that constitutively express IL-2 (e.g., haNK cells, NK92MI cells), the chimeric protein may be administered after treatment with the NK cells.
[0057] Furthermore, as contemplated methods are also thought to increase NKD2D surface expression on various cells in addition to IFNy secretion, and especially immune competent cells such as NK cells, CD8+, and CD4+ T-cells, it is further contemplated that treatments may also include one or more steps that augment or trigger NKG2D ligand expression and presentation. For example, such steps include low dose chemotherapy and/or low dose radiation therapy, typically performed at dosages that are equal or less than 50%, equal or less than 30%, equal or less than 20%, or equal or less than 10% of the maximum tolerated dose. Moreover, such low dose treatment will preferably be performed in a metronomic fashion, for example, on alternating days, or every third day, or once weekly for several weeks, etc.
Further Embodiments
[0058] Embodiment 1. A method of inducing IFNy secretion in an NK cell, comprising: contacting the NK cell with a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion to induce interferon gamma (IFNy) secretion by the NK cell, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2.
[0059] Embodiment 2. The method of Embodiment 1, wherein the IL-2 sensitized NK cell is an NK cell that was previously and constitutively exposed to IL-2, preferably at least 100 IU/mL IL-2.
[0060] Embodiment 3. The method of Embodiment 1 or 2, wherein any one or more of the following is true:
• the genetically modified NK cell is an aNK cell, an autologous NK cell, or an NK cell derived in vitro from a precursor cell;
• the genetically modified NK cell constitutively expresses and intracellularly retains IL-2;
• the genetically modified NK cell is a haNK cell or an NK92MI cell; and
• the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IE-12b chain.
[0061] Embodiment 4. The method of any one of the previous Embodiments, wherein the cancer cell targeting portion targets a necrosis marker or a cancer associated antigen or a cancer neoepitope, and wherein the necrosis marker is preferably a DNA, an RNA, or a histone protein, and wherein the cancer cell targeting portion preferably comprises hu5l-4 antibody or a portion thereof.
[0062] Embodiment 5. The method of any one of the previous Embodiments, wherein the chimeric protein comprises SEQ ID NO:l or SEQ ID NO:2, and optionally SEQ ID NO:3.
[0063] Embodiment 6. The method of any one of the previous Embodiments, wherein any one or more of the following is true:
• the step of contacting the NK cell with the chimeric protein is performed ex vivo, and the NK cell is a haNK cell; and • the step of contacting the NK cell with the chimeric protein is performed in vivo, and the NK cell is a haNK cell or NK92MI cell.
[0064] Embodiment 7. A composition for use in treating cancer in a subject in need thereof, the composition comprising and NK cell and a chimeric protein,
wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2; and
wherein the chimeric protein comprises an IL-12 portion and a cancer cell targeting portion to thereby induce interferon gamma (IFNy) secretion from the NK cell.
[0065] Embodiment 8. The composition of Embodiment 7, wherein the NK cell is a genetically modified NK cell that constitutively expresses and intracellularly retains IL-2, and wherein the NK cell is preferably a haNK cell or an NK92MI cell.
[0066] Embodiment 9. The composition of Embodiment 7 or 8, wherein any one or more of the following is true:
• the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IE-12b chain;
• the cancer cell targeting portion comprises an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion;
• the cancer cell targeting portion targets a necrosis marker or a cancer associated antigen or a cancer neoepitope;
• the necrosis marker is a DNA, an RNA, or a histone protein;
• the cancer cell targeting portion comprises a hu5l-4 antibody or portion thereof;
• the chimeric protein comprises SEQ ID NO: l or SEQ ID NO:2, and optionally SEQ ID NO:3; and
• the chimeric protein is bound to the NK cell.
[0067] Embodiment 10. A chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion that targets a necrosis marker, and optionally wherein any one or more of the following is true:
• the necrosis marker is a DNA, an RNA, or a histone protein;
• the cancer cell targeting portion comprises a hu51-4 antibody or portion thereof;
• the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IE-12b chain; • the chimeric protein comprises SEQ ID NO:l and optionally SEQ ID NO:3;
• the chimeric protein comprises SEQ ID NO:2 and optionally SEQ ID NO:3 and
• the cancer cell targeting portion comprises an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fat¾ portion, or an scFv portion.
[0068] Embodiment 11. The chimeric protein of Embodiment 10, wherein the IL-12 portion is covalently coupled to the cancer cell targeting portion, and optionally wherein the IL-12 portion is covalently coupled to the cancer cell targeting portion via a peptide linker.
[0069] Embodiment 12. The chimeric protein of Embodiment 10 or 11, wherein the chimeric protein is bound to CD 16 of an NK cell.
[0070] Embodiment 13. A pharmaceutical composition comprising the chimeric protein of any one of Embodiments 10-12.
[0071] Embodiment 14. The pharmaceutical composition of Embodiment 13, wherein the composition further comprises an NK cell, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2, and optionally wherein any one or more of the following is true:
• the NK cell is a haNK cell;
• the NK cell is an NK92MI cell; and
• the chimeric protein is bound to CD 16 of an NK cell.
[0072] Embodiment 15. A chimeric antibody comprising a chimeric heavy chain that includes a scIL-l2 portion and antigen binding portion that binds a cell death-exposed antigen, and optionally wherein any one or more of the following is true:
• the scIL-l2 comprises an IL-l2a subunit linked by a peptide linker to an IE-12b subunit;
• the antibody comprises SEQ ID NO:l and optionally SEQ ID NO:3; and
• the antibody comprises SEQ ID NO:2 and optionally SEQ ID NO:3.
Examples
[0073] To investigate the influence of constitutive exposure of NK cells to IL-2, three NK92 derivatives, aNK cells, haNK cells (commercially available from NantKwest), and NK92MI cells (commercially available from ATCC) were exposed to IL-12. The left panel of Fig.l comparatively depicts the data for aNK cells without constitutive IL-2 exposure and haNK cells with constitutive IL-2 exposure, while the right panel depicts NK92 derivatives that included stable integration of an IL-2 expression cassette (not expressing the high affinity CD16 variant). As can be taken from these results, the modified NK cells were not only responsive to IL-12 signaling to secrete IFNy, but secreted unexpected high quantities of IFNy (peaking near 200 ng/mL).
[0074] More particularly, NK-92 cells were previously transfected with human IL-2 cDNA in a retroviral MFG-hIL-2 vector by particle-mediated gene transfer. The transfection was stable. NK-92 and this derivative cell line NK-92MI had the following characteristics: surface marker positive for CD2, CD7, CDlla, CD28, CD45, CD54 and CD56 bright; surface marker negative for CD1, CD3, CD4, CD5, CD8, CD10, CD14, CD16, CD19, CD20, CD23, CD34 and HLA-DR. Cells were grown following standard protocol. IFNy was measured using conventional ELISA methods well known in the art.
[0075] In a further set of experiments, haNK cells (/.<?., aNK cell derivatives expressing high- affinity CD 16 and intracellularly retained IL-2) were cultured overnight in the presence of human recombinant IL-12 (rHuIL-l2), and selected human or murine recombinant IL-l2-Ab chimeric constructs (HuIL-l2/Hu5l-4 and MuIL-l2/Hu5l-4) having the sequence of SEQ ID NO: l and SEQ ID NO:2 or SEQ ID NO:l and SEQ ID NO:3. Cells were cultured by seeding 2.5xl05 cells into a 24 well plate, X-Vivo 10 containing 5% human serum. The cell culture supernatants were then collected and human IEN-g measured by ELISA. Remarkably, constitutive IL-2 exposure via intracellular expression of IL-2 rendered haNK cells sensitive to IL-12 signaling as can be readily taken from the graphs in Fig.2. Indeed, exposure to IL-12 in various forms resulted in significant IFNy secretion for both murine and human IL-12. As expected, human IL-12 produced somewhat stronger IFNy secretion in the haNK cells than murine IL-12.
[0076] Binding specificity of chimeric proteins HuIL-l2/Hu5l-4 and MuIL-l2/Hu5l-4 towards necrotic cells was evaluated using dead Raji cancer cells (human Burkitt’s lymphoma cells) and exemplary results are depicted in Fig.3. Results are shown for negative control (NC), positive control (PC), and binding of HuIL-l2/Hu5l-4 and MuIL-l2/Hu5l-4 as indicated. As can be readily seen from the graph, both HuIL-l2/Hu5l-4 and MuIL-l2/Hu5l- 4 avidly bound to the target antigens in the dead cells. Exemplary sequences for HuIL- !2/Hu5l-4 and MuIL-l2/Hu5l-4 are also depicted in Fig.4. [0077] In some embodiments, the numbers expressing quantities of ingredients, properties such as concentration, reaction conditions, and so forth, used to describe and claim certain embodiments of the invention are to be understood as being modified in some instances by the term“about.” Accordingly, in some embodiments, the numerical parameters set forth in the written description and attached claims are approximations that can vary depending upon the desired properties sought to be obtained by a particular embodiment. In some embodiments, the numerical parameters should be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of some embodiments of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as practicable. The numerical values presented in some embodiments of the invention may contain certain errors necessarily resulting from the standard deviation found in their respective testing measurements. Unless the context dictates the contrary, all ranges set forth herein should be interpreted as being inclusive of their endpoints, and open-ended ranges should be interpreted to include commercially practical values. Similarly, all lists of values should be considered as inclusive of intermediate values unless the context indicates the contrary.
[0078] As used in the description herein and throughout the claims that follow, the meaning of“a,”“an,” and“the” includes plural reference unless the context clearly dictates otherwise. Also, as used in the description herein, the meaning of“in” includes“in” and“on” unless the context clearly dictates otherwise. Moreover, and unless the context dictates otherwise, the term "coupled to" is intended to include both direct coupling (in which two elements that are coupled to each other contact each other) and indirect coupling (in which at least one additional element is located between the two elements). Therefore, the terms "coupled to" and "coupled with" are used synonymously.
[0079] Moreover, as used herein, the phrase“at least one of A and B” is intended to refer to ‘A’ and/or Έ’, regardless of the nature of ‘A’ and Έ’. For example, in some embodiments, ‘A’ may be single distinct species, while in other embodiments‘A’ may represent a single species within a genus that is denoted‘A’. Likewise, in some embodiments, Έ’ may be single distinct species, while in other embodiments Έ’ may represent a single species within a genus that is denoted Έ’ . [0080] It should be apparent to those skilled in the art that many more modifications besides those already described are possible without departing from the inventive concepts herein. The inventive subject matter, therefore, is not to be restricted except in the scope of the appended claims. Moreover, in interpreting both the specification and the claims, all terms should be interpreted in the broadest possible manner consistent with the context. In particular, the terms“comprises” and“comprising” should be interpreted as referring to elements, components, or steps in a non-exclusive manner, indicating that the referenced elements, components, or steps may be present, or utilized, or combined with other elements, components, or steps that are not expressly referenced. Where the specification claims refers to at least one of something selected from the group consisting of A, B, C .... and N, the text should be interpreted as requiring only one element from the group, not A plus N, or B plus N, etc.

Claims

CLAIMS What is claimed is:
1. A method of inducing IFNy secretion in an NK cell, comprising:
contacting the NK cell with a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion to induce interferon gamma (IFNy) secretion by the NK cell;
wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2.
2. The method of claim 1 wherein the IL-2 sensitized NK cell is an NK cell that was previously and constitutively exposed to IL-2.
3. The method of claim 1 wherein the IL-2 sensitized NK cell was constitutively exposed to at least 100 IU/mL IL-2.
4. The method of claim 1 wherein the genetically modified NK cell is an aNK cell, an autologous NK cell, or an NK cell derived in vitro from a precursor cell.
5. The method of claim 1 wherein the genetically modified NK cell constitutively expresses and intracellularly retains IL-2.
6. The method of claim 1 wherein the genetically modified NK cell is a haNK cell or an NK92MI cell.
7. The method of claim 1 wherein the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an I L- 12b chain.
8. The method of claim 1 wherein the cancer cell targeting portion comprises an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion.
9. The method of claim 1 wherein the cancer cell targeting portion targets a necrosis marker or a cancer associated antigen or a cancer neoepitope.
10. The method of claim 9 wherein the necrosis marker is a DNA, an RNA, or a histone protein.
11. The method of claim 1 wherein the cancer cell targeting portion comprises a hu5l-4 antibody or portion thereof.
12. The method of claim 1 wherein the chimeric protein comprises SEQ ID NO:l or SEQ ID NO:2, and optionally SEQ ID NO:3.
13. The method of claim 1 wherein the step of contacting the NK cell with the chimeric protein is performed ex vivo and wherein the NK cell is a haNK cell, or wherein the step of contacting the NK cell with the chimeric protein is performed in vivo and wherein the NK cell is a haNK cell or NK92MI cell.
14. A method of treating cancer, comprising:
administering an NK cell to a patient, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2; and administering a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion to thereby induce interferon gamma (IFNy) secretion by the NK cell.
15. The method of claim 14 wherein the NK cell is a genetically modified NK cell that constitutively expresses and intracellularly retains IL-2.
16. The method of claim 15 wherein the NK cell is a haNK cell or an NK92MI cell.
17. The method of claim 14 wherein the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IE-12b chain.
18. The method of claim 14 wherein the cancer cell targeting portion comprises an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion.
19. The method of claim 14 wherein the cancer cell targeting portion targets a necrosis marker or a cancer associated antigen or a cancer neoepitope.
20. The method of claim 19 wherein the necrosis marker is a DNA, an RNA, or a histone protein.
21. The method of claim 14 wherein the cancer cell targeting portion comprises a hu5l-4 antibody or portion thereof.
22. The method of claim 14 wherein the chimeric protein comprises SEQ ID NO:l or SEQ ID NO:2, and optionally SEQ ID NO:3.
23. The method of claim 14 wherein the chimeric protein is administered at least 12 hours prior to administration of the NK cell, or wherein the chimeric protein is administered at least 12 hours after administration of the NK cell.
24. The method of claim 14 wherein the chimeric protein is contemporaneously administered with the NK cell.
25. The method of claim 24 wherein the chimeric protein is bound to the NK cell.
26. A chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion that targets a necrosis marker.
27. The chimeric protein of claim 26 wherein the necrosis marker is a DNA, an RNA, or a histone protein.
28. The chimeric protein of claim 26 wherein the cancer cell targeting portion comprises a hu5l-4 antibody or portion thereof.
29. The chimeric protein of claim 26 wherein the cancer cell targeting portion comprises an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion.
30. The chimeric protein of claim 26 wherein the IL-12 portion is covalently coupled to the cancer cell targeting portion.
31. The chimeric protein of claim 30 wherein the IL-12 portion is covalently coupled to the cancer cell targeting portion via a peptide linker.
32. The chimeric protein of claim 26 wherein the IL-12 portion of the chimeric protein comprises an IL-l2a chain and an IE-12b chain.
33. The chimeric protein of claim 26 comprising SEQ ID NO:l or SEQ ID NO:2, and optionally SEQ ID NO:3.
34. The chimeric protein of claim 26 bound to CD16 of an NK cell.
35. A pharmaceutical composition comprising the chimeric protein of any one of claims 26- 34.
36. The pharmaceutical composition of claim 35 further comprising an NK cell, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2.
37. The pharmaceutical composition of claim 35 wherein the NK cell is a genetically modified NK cell that constitutively expresses and intracellularly retains IL-2.
38. The pharmaceutical composition of claim 37 wherein the NK cell is a haNK cell or an NK92MI cell.
39. The pharmaceutical composition of claim 37 wherein the chimeric protein is bound to CD16 of an NK cell.
40. A kit comprising:
an NK cell, wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2; and
a chimeric protein that comprises an IL-12 portion and a cancer cell targeting portion.
41. The kit of claim 40 wherein the IL-2 sensitized NK cell is an NK cell that was previously and constitutively exposed to IL-2.
42. The kit of claim 40 wherein the genetically modified NK cell is an aNK cell, an autologous NK cell, or an NK cell derived in vitro from a precursor cell.
43. The kit of claim 40 wherein the genetically modified NK cell constitutively expresses and intracellularly retains IL-2.
44. The kit of claim 40 wherein the genetically modified NK cell is a haNK cell or an NK92MI cell.
45. The kit of claim 40 wherein the IL-12 portion of the chimeric protein comprises an IL- 12a chain and an IL-12 chain.
46. The kit of claim 40 wherein the cancer cell targeting portion comprises an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion.
47. The kit of claim 40 wherein the cancer cell targeting portion targets a necrosis marker or a cancer associated antigen or a cancer neoepitope.
48. The kit of claim 47 wherein the necrosis marker is a DNA, an RNA, or a histone protein.
49. The kit of claim 40 wherein the cancer cell targeting portion comprises a hu5l-4 antibody or portion thereof.
50. The kit of claim 40 wherein the chimeric protein is formulated for transfusion and/or injection, and wherein the NK cell is formulated for transfusion.
51. A chimeric antibody comprising a chimeric heavy chain that includes a scIL-l2 portion and antigen binding portion that binds a cell death-exposed antigen.
52. The chimeric antibody of claim 51 wherein the scIL-l2 comprises an IL-l2a subunit linked by a peptide linker to an IL-12 subunit.
53. The chimeric antibody of claim 51 comprising SEQ ID NO:l or SEQ ID NO:2, and optionally SEQ ID NO:3.
54. The chimeric antibody of claim 51 comprising SEQ ID NO:l and SEQ ID NO:3.
55. The chimeric antibody of claim 51 comprising SEQ ID NO:2 and SEQ ID NO:3.
56. Use of a chimeric protein and an NK cell in the treatment of cancer,
wherein the NK cell is an IL-2 sensitized NK cell or a genetically modified NK cell that constitutively expresses IL-2; and
wherein the chimeric protein comprises an IL-12 portion and a cancer cell targeting portion that induces interferon gamma (IFNy) secretion by the NK cell.
57. The use of claim 56 wherein the NK cell is a genetically modified NK cell that constitutively expresses and intracellularly retains IL-2.
58. The use of claim 56 wherein the NK cell is a haNK cell or an NK92MI cell.
59. The use of claim 56 wherein the IL-12 portion of the chimeric protein comprises an IL- 12a chain and an IE-12b chain.
60. The use of claim 56 wherein the cancer cell targeting portion comprises an IgG antibody portion, a Fab portion, a F(ab’)2 portion, a Fab2 portion, or an scFv portion.
61. The use of claim 56 wherein the cancer cell targeting portion targets a necrosis marker or a cancer associated antigen or a cancer neoepitope.
62. The method of claim 61 wherein the necrosis marker is a DNA, an RNA, or a histone protein.
63. The use of claim 56 wherein the cancer cell targeting portion comprises a hu5l-4 antibody or portion thereof.
64. The use of claim 56 wherein the chimeric protein comprises SEQ ID NO: l or SEQ ID NO:2, and optionally SEQ ID NO:3.
65. The use of claim 56 wherein the chimeric protein is administered at least 12 hours prior to administration of the NK cell, or wherein the chimeric protein is administered at least 12 hours after administration of the NK cell.
66. The use of claim 56 wherein the chimeric protein is contemporaneously administered with the NK cell.
67. The use of claim 66 wherein the chimeric protein is bound to the NK cell.
PCT/US2019/049797 2018-09-07 2019-09-05 Targeted il-12 treatments and methods to stimulate hank and nk92mi cells WO2020051363A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP19856745.5A EP3847242A4 (en) 2018-09-07 2019-09-05 Targeted il-12 treatments and methods to stimulate hank and nk92mi cells
US17/274,127 US20220289805A1 (en) 2018-09-07 2019-09-05 Targeted IL-12 Treatments and Methods to Stimulate haNK and NK92mi Cells
CN201980058286.XA CN112996904A (en) 2018-09-07 2019-09-05 Targeted IL-12 therapy and methods for stimulating HANK and NK92MI cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862728510P 2018-09-07 2018-09-07
US62/728,510 2018-09-07

Publications (1)

Publication Number Publication Date
WO2020051363A1 true WO2020051363A1 (en) 2020-03-12

Family

ID=69722892

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/049797 WO2020051363A1 (en) 2018-09-07 2019-09-05 Targeted il-12 treatments and methods to stimulate hank and nk92mi cells

Country Status (4)

Country Link
US (1) US20220289805A1 (en)
EP (1) EP3847242A4 (en)
CN (1) CN112996904A (en)
WO (1) WO2020051363A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8034332B2 (en) 1997-04-30 2011-10-11 Conkwest, Inc. Interleukin-secreting natural killer cell lines and methods of use
US8945571B2 (en) * 2009-08-17 2015-02-03 Roche GlyeArt AG Targeted immunoconjugates
WO2016160602A2 (en) * 2015-03-27 2016-10-06 Nantkwest, Inc. Genetically modified nk-92 cells and monoclonal antibodies for the treatment of cancer
US20170166620A1 (en) * 2014-02-19 2017-06-15 Merck Patent Gmbh Cancer-targeted il-12 immunotherapy
CN108250303A (en) * 2016-12-19 2018-07-06 南京金斯瑞生物科技有限公司 Single domain antibody fusion protein and its application
US20180200366A1 (en) * 2016-10-21 2018-07-19 Altor Bioscience Corporation Multimeric il-15-based molecules

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1954073B (en) * 2004-03-19 2013-01-16 默克专利股份有限公司 Modified Bouganin proteins, cytotoxins and methods and uses thereof
EP3339320A4 (en) * 2015-08-17 2019-01-23 Seoul National University R&DB Foundation Chimeric antibody receptor to which anti-cotinine antibody is linked, and use thereof
GB201521389D0 (en) * 2015-12-03 2016-01-20 Ucb Biopharma Sprl Method
CN110785435B (en) * 2017-03-06 2024-01-23 艾尔特生物科技公司 IL-15-based fusions with IL-12 and IL-18
CA3058144A1 (en) * 2017-03-27 2018-10-04 Nantcell, Inc. Ank and il-12 compositions and methods
US10688182B2 (en) * 2018-03-29 2020-06-23 Cho Pharma Usa, Inc. Monoclonal antibodies that bind to SSEA4 and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8034332B2 (en) 1997-04-30 2011-10-11 Conkwest, Inc. Interleukin-secreting natural killer cell lines and methods of use
US8945571B2 (en) * 2009-08-17 2015-02-03 Roche GlyeArt AG Targeted immunoconjugates
US20170166620A1 (en) * 2014-02-19 2017-06-15 Merck Patent Gmbh Cancer-targeted il-12 immunotherapy
WO2016160602A2 (en) * 2015-03-27 2016-10-06 Nantkwest, Inc. Genetically modified nk-92 cells and monoclonal antibodies for the treatment of cancer
US20180200366A1 (en) * 2016-10-21 2018-07-19 Altor Bioscience Corporation Multimeric il-15-based molecules
CN108250303A (en) * 2016-12-19 2018-07-06 南京金斯瑞生物科技有限公司 Single domain antibody fusion protein and its application

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
"Genbank", Database accession no. AF512686
"UniProt", Database accession no. P29459
BLOOD, vol. 113, 2009, pages 3716 - 3725
ECKERT FRANZISKA ET AL., CANCER IMMUNOLOGY IMMUNOTHERAPY, vol. 65, 2016, pages 1003 - 1013
EXP HEMATOL., vol. 33, no. 2, February 2005 (2005-02-01), pages 159 - 64
FALLON J ET AL., ONCOTARGET, vol. 5, 2014, pages 1869 - 1884
GILLIES S ET AL., J. IMMUNOLOGY, vol. 160, 1998, pages 6195 - 6203
HALIN C ET AL., NATURE BIOTECH., vol. 20, 2002, pages 264 - 269
HUM GENE THER, vol. 10, no. 8, 20 May 1999 (1999-05-20), pages 1359 - 73
J IMMUNOLMETHODS, vol. 348, no. 1-2, 31 August 2009 (2009-08-31), pages 83 - 94
J. LEUKOC. BIOL., vol. 58, 1995, pages 225 - 233
MOLECULAR AND CELLULAR BIOLOGY, March 2002 (2002-03-01), pages 1742 - 53
ONCOTARGET, vol. 5, no. 7, 15 April 2014 (2014-04-15), pages 1869 - 84
ONCOTARGET, vol. 7, no. 52, 27 December 2016 (2016-12-27), pages 86359 - 86373
PASCHE N ET AL., CLIN, CANCER RES., vol. 18, 2012, pages 4092 - 4103
PLOS ONE, vol. 12, no. 7, 5 July 2017 (2017-07-05), pages e0179431
See also references of EP3847242A4
VALEDKARIMI, Z. ET AL.: "Antibody-cytokine fusion proteins for improving efficacy and safety of cancer therapy", BIOMEDICINE & PHARMACOTHERAPY, vol. 95, 2017, pages 731 - 742, XP085226466, DOI: 10.1016/j.biopha.2017.07.160 *
VANDEVEER AMANDA J ET AL., IMMUNOLOGY, 15 July 2016 (2016-07-15), pages 1480 - 1480

Also Published As

Publication number Publication date
US20220289805A1 (en) 2022-09-15
EP3847242A1 (en) 2021-07-14
EP3847242A4 (en) 2022-06-08
CN112996904A (en) 2021-06-18

Similar Documents

Publication Publication Date Title
AU2018244221B2 (en) ANK and IL-12 compositions and methods
Overwijk et al. Engineering IL-2 to give new life to T cell immunotherapy
US10894816B2 (en) Use of IL-15 to increase thymic output and to treat lymphopenia
Sim et al. The IL-2 cytokine family in cancer immunotherapy
US11230698B2 (en) Cytokine receptor genes and the use thereof to enhance therapy
Kaiga et al. Systemic administration of IL-23 induces potent antitumor immunity primarily mediated through Th1-type response in association with the endogenously expressed IL-12
CN110396133B (en) Fusion protein type prodrug with interleukin 12 as active component
Tomala et al. IL-2/anti-IL-2 mAb immunocomplexes: A renascence of IL-2 in cancer immunotherapy?
US20230172982A1 (en) Elimination of BCMA-positive malignancies by CAR expressing NK cells
Pieper et al. Radiation augments the local anti-tumor effect of in situ vaccine with CpG-oligodeoxynucleotides and anti-OX40 in immunologically cold tumor models
Gu et al. Dectin-1 controls TSLP-induced Th2 response by regulating STAT3, STAT6, and p50-RelB activities in dendritic cells
WO2022086988A1 (en) Multi-functional and multi-valent interleukin-tgf-beta receptor fusion polypeptides
US20220289805A1 (en) Targeted IL-12 Treatments and Methods to Stimulate haNK and NK92mi Cells
US11407814B2 (en) Multi-functional and multi-valent interleukin-TGF-beta receptor fusion polypeptides
TW201938196A (en) Fusion protein extensions
WO2023088246A1 (en) Membrane surface protein containing gpi anchor region
Guimarães RNA-Modified T Cells as a Tool to Deliver Immunomodulatory Agents to Malignant Brain Tumors
NZ736967B2 (en) Use of il-15 to increase thymic output and to treat lymphopenia

Legal Events

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

Ref document number: 19856745

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019856745

Country of ref document: EP

Effective date: 20210407