WO2025038745A1 - Compositions and methods for activating immune cells - Google Patents
Compositions and methods for activating immune cells Download PDFInfo
- Publication number
- WO2025038745A1 WO2025038745A1 PCT/US2024/042306 US2024042306W WO2025038745A1 WO 2025038745 A1 WO2025038745 A1 WO 2025038745A1 US 2024042306 W US2024042306 W US 2024042306W WO 2025038745 A1 WO2025038745 A1 WO 2025038745A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- activator
- cells
- factors
- ifn
- receptor
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0639—Dendritic cells, e.g. Langherhans cells in the epidermis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/10—Cellular immunotherapy characterised by the cell type used
- A61K40/19—Dendritic cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/20—Cellular immunotherapy characterised by the effect or the function of the cells
- A61K40/24—Antigen-presenting cells [APC]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/40—Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
- A61K40/41—Vertebrate antigens
- A61K40/42—Cancer antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/40—Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
- A61K40/41—Vertebrate antigens
- A61K40/42—Cancer antigens
- A61K40/4201—Neoantigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/46—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the cancer treated
- A61K2239/48—Blood cells, e.g. leukemia or lymphoma
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/22—Colony stimulating factors (G-CSF, GM-CSF)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2304—Interleukin-4 (IL-4)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2306—Interleukin-6 (IL-6)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2307—Interleukin-7 (IL-7)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2309—Interleukin-9 (IL-9)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/231—Interleukin-10 (IL-10)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2312—Interleukin-12 (IL-12)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2315—Interleukin-15 (IL-15)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2319—Interleukin-19 (IL-19)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/232—Interleukin-20 (IL-20)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2321—Interleukin-21 (IL-21)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2322—Interleukin-22 (IL-22)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2323—Interleukin-23 (IL-23)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2324—Interleukin-24 (IL-24)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/24—Interferons [IFN]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/25—Tumour necrosing factors [TNF]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/999—Small molecules not provided for elsewhere
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2506/00—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
- C12N2506/30—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from cancer cells, e.g. reversion of tumour cells
Definitions
- the present invention relates to compositions and methods for: a) promoting the differentiation and/or maturation of antigen presenting cells (APCs) derived from cancer cells (such as malignant myelogenous cells or B lymphoblasts/lymphocytic cells from cancer patients), and b) activating immune cells (such as T cells).
- APCs antigen presenting cells
- Hematologic malignancies also known as blood cancer
- diseases including acute and chronic lymphoblastic leukemia (ALL and CLL), acute and chronic myelogenous leukemia (AML and CML), myelodysplastic syndrome (MDS), Hodgkin’s and non-Hodgkin’s lymphoma, multiple myeloma (MM), etc.
- NHL non-Hodgkin’s lymphoma
- NHL is the most common disease (> 40%), followed by leukemia (31%), and then MM (18%).
- B cell-lineage malignancies such as B-ALL, CLL, NHL, and MM occupy > 50% of all cases, with the rest being chiefly myelogenous leukemia (AML and CML).
- AML and CML myelogenous leukemia
- malignant cells generally manifest in peripheral blood (leukemia), bone marrow (leukemia and MM), lymph nodes (lymphoma), and spleen, as well as disseminated areas such as the brain, spinal cord, and peritoneal cavity.
- the treatment of hematological malignancies mainly includes chemotherapy, radiotherapy, and hematopoietic stem cell transplantation (HSCT).
- CAR-T chimeric antigen receptor T
- the present application in one aspect provides a method of producing a population of antigen presenting cells (“APCs”) presenting an antigen associated with a hematological cancer (“HC-APC”), comprising: contacting hematological cancer cells obtained from an individual having the hematological cancer with one or more survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF ⁇ receptor (TNFR) activator, 3) an interleukin 4 receptor (IL-4R) activator, and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, thereby producing the population of HC-APCs.
- APCs antigen presenting cells
- HC-APC hematological cancer
- S/D/M factors maturation factors
- the present application in another aspect provides a method of producing activated immune cells, comprising: a) producing a population of antigen presenting cells (“APCs”) presenting an antigen associated with a hematological cancer (“HC-APC”) according to the method described above, and b) contacting the HC-APCs with immune cells, thereby producing activated immune cells.
- APCs antigen presenting cells
- HC-APC hematological cancer
- the present application in another aspect provides a method of producing activated immune cells, comprising contacting immune cells with a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), thereby generating activated immune cells; wherein the HC-APCs are derived from hematological cancer cells after having been contacted with one or more survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF ⁇ receptor (TNFR) activator, 3) an interleukin 4 (IL-4R) activator, and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator.
- S/D/M factors survival, differentiation, and/or maturation factors
- the STAT3 activator is selected from the group consisting of: a small molecule STAT3 activator, an IL- 10, an IL-10 family member, an IL-10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, and a small molecule activator of IL-10R.
- the STAT3 activator is selected from the group consisting of: an IL-10 family cytokine, an IL-12 family cytokine, an IL-6 family cytokine, IL-7 family cytokine, IL-9 family cytokine, IL-15 ny-2770598 Attorney Docket No.24516-20006.40 family cytokine, IL-21 family cytokine, and G-CSF.
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-12, IL-23, IL-6, colivelin TFA, Garcinone D, G-CSF, IL-7, IL-9, IL-15, and IL-21. In some embodiments, the STAT3 activator is selected from the group consisting of: IL-10, IL-22, IL- 19, IL-20, IL-24, IL-12, IL-23, Colivelin TFA, and Garcinone D.
- the one or more of S/D/M factors comprise an interleukin 4 receptor (IL-4R) activator.
- the IL-4R activator is selected from the group consisting of IL-4, an IL-4R agonist antibody, and a small molecule activator of IL-4.
- the IL-4R activator is IL-4.
- the one or more of S/D/M factors comprise a TNFR activator.
- the TNFR activator is selected from the group consisting of TNF ⁇ , a TNFR agonist antibody, and a small molecule activator of TNFR.
- the TNFR activator is TNF ⁇ .
- the one or more of S/D/M factors comprise an IFNGR activator.
- the IFNGR activator is selected from the group consisting of IFN ⁇ , an IFNGR agonist antibody, and a small molecule activator of IFNGR.
- the IFNGR activator is IFN ⁇ .
- the one or more of S/D/M factors are present in a single composition.
- the one or more of S/D/M factors comprise two or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator.
- the one or more of S/D/M factors comprise three or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator.
- the one or more of S/D/M factors comprise: 1) a STAT3 activator, 2) a TNFR activator, and 3) an IFNGR activator.
- the one or more of S/D/M factors comprise an IL-10 family cytokine, TNF ⁇ , and IFN ⁇ .
- the one or more of S/D/M factors comprise: 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator.
- the one ny-2770598 Attorney Docket No.24516-20006.40 or more of S/D/M factors comprise an IL-10 family cytokine, TNF ⁇ , IL-4, and IFN ⁇ .
- IL-10 family cytokines include IL-10, IL-22, IL-19, IL-24, IL-20, or IL-26.
- the one or more of the S/D/M factors further comprise an IL-6 receptor (IL-6R) activator and/or a GM- CSF receptor (GM-CSFR) activator.
- IL-6R IL-6 receptor
- GM-CSFR GM- CSF receptor
- the IL-6R activator is selected from the group consisting of IL-6, an IL-6R agonist antibody, and a small molecule activator of IL-6R.
- the GM-CSFR activator is selected from the group consisting of GM-CSF, a GM-CSFR agonist antibody, and a small molecule activator of GM- CSF.
- the IL-6R activator is IL-6, and the GM-CSFR activator is GM- CSF.
- the method further comprises contacting the HC-APCs with one or more refinement factors selected from the group consisting of type-I interferon, IFN ⁇ , TNF ⁇ , a TLR ligand, CD40L or a CD40-ligating antibody, an anti-PD-L1 antibody, and TPI-1.
- the type-I interferon comprises IFN ⁇ and/or IFN ⁇ .
- the TLR ligand is R848, poly IC, CpG, or LPS.
- the TLR ligand comprises R848 and poly IC.
- the one or more refinement factors comprise IFN ⁇ , IFN ⁇ , and TNF ⁇ . In some embodiments, the one or more refinement factors further comprise at least two agents selected from the group consisting of poly IC, CpG, CD40L, R848, and an anti-PD-L1 antibody. In some embodiments, the one or more refinement factors further comprises a SHP-1 inhibitor. In some embodiments, the SHP-1 inhibitor is TPI-1. In some embodiments, the HC-APCs are cultured with the one or more refinement factors for about 1-4 days. [0020] In some embodiments according to any of the methods described above, the method further comprises administering into the individual immune cells activated by contacting the immune cells with the population of HC-APCs.
- the method further comprises administering into a different individual than the individual having the hematological cancer immune cells activated by contacting the immune cells with the population of HC-APCs.
- the immune cells and the hematological ny-2770598 Attorney Docket No.24516-20006.40 cancer cells are from the same individual.
- the immune cells and the hematological cancer cells are from different individuals.
- the immune cells are administered to the individual from whom they are obtained.
- the immune cells are administered to a different individual than the individual from whom they are obtained.
- the immune cells are selected from the group consisting of peripheral blood mononuclear cells (“PBMCs”), cells derived from bone marrow biopsy, cells derived from lymphoma biopsy, tumor infiltrating T cells (TIL), and T cells.
- PBMCs peripheral blood mononuclear cells
- TIL tumor infiltrating T cells
- the immune cells are T cells.
- the T cells are CD8 T cells and/or CD4 T cells.
- the hematological cancer cells are obtained from PBMCs, enriched leukemia cells, a bone marrow biopsy, or a lymphoma biopsy.
- the hematological cancer cells are cultured with the one or more of S/D/M factors for at least 2 days.
- the immune cells have been enriched prior to contacting the HC-APCs.
- the hematological cancer cells have been enriched prior to being cultured with the one or more of S/D/M factors.
- the hematological cancer is a myeloid leukemia.
- the hematological cancer is acute myeloid leukemia (AML) or chronic myeloid leukemia (CML).
- the hematological cancer cells are any of M0-M5 myeloblasts.
- the hematological cancer cells have been enriched prior to being cultured with the one or more of S/D/M factors in the presence of an anti-CD11b antibody.
- the hematological cancer is a B-cell lymphoma or B-cell leukemia.
- the B- cell lymphoma or B-cell leukemia is selected from the group consisting of B-cell acute lymphoblastic leukemia (B-ALL), chronic lymphocytic leukemia (CLL), and non-Hodgkin’s lymphoma (NHL).
- B-ALL B-cell acute lymphoblastic leukemia
- CLL chronic lymphocytic leukemia
- NHL non-Hodgkin’s lymphoma
- the hematological cancer cells have been enriched ny-2770598 Attorney Docket No.24516-20006.40 prior to being cultured with the one or more of S/D/M factors in the presence of an anti-CD19 antibody or an anti-CD20 antibody.
- the method further comprises expanding the immune cells by contacting the immune cells with the HC- APCs for at least two rounds, at least three rounds, or at least four rounds.
- the present application in another aspect provides a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”) obtained by any of the methods described above.
- the present application in another aspect provides a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APCs”), wherein the HC-APCs are derived from hematological cancer cells, and wherein the HC-APCs express increased levels of MHC-1, MHC-II, CD40, CD80 and/or CD86 compared to the hematological cancer cells prior to the treatment.
- the HC-APCs comprise one or more exogenous antigens.
- the hematological cancer cells are primary cells from an individual having the hematological cancer.
- the hematological cancer is a myeloid leukemia.
- the hematological cancer is acute myeloid leukemia (AML) or chronic myeloid leukemia (CML).
- the hematological cancer cells are any of M0-M5 myeloblasts.
- the hematological cancer is a B-cell lymphoma or B-cell leukemia.
- the B-cell lymphoma or B-cell leukemia is selected from the group consisting of B-cell acute lymphoblastic leukemia (B-ALL), chronic lymphocytic leukemia (CLL), and non-Hodgkin’s lymphoma (NHL).
- the present application in another aspect provides a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APCs”), prepared by: a) obtaining hematological cancer cells from an individual having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF ⁇ receptor (TNFR) activator, 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, and 3) an IL-4 receptor (IL- 4R) activator, thereby producing the population of HC-APCs.
- S/D/M factors survival, differentiation, and/or maturation factors
- the present application in another aspect provides a population of activated immune cells obtained by any of the methods described above. ny-2770598 Attorney Docket No.24516-20006.40 [0033] The present application in another aspect provides a method of treating a hematological cancer in an individual, comprising administering to the individual an effective amount of any of the activated immune cells described above.
- the present application in another aspect provides a method of treating a hematological cancer in an individual, comprising: a) contacting hematological cancer cells and/or monocytes with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF ⁇ receptor (TNFR) activator, 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, and 4) an IL-4 receptor (IL-4R) activator, thereby producing a population of antigen presenting cells (“APCs”), b) contacting the APCs with a population of immune cells isolated from the individual, thereby producing activated immune cells, and c) administering the activated immune cells into the individual.
- S/D/M factors survival, differentiation, and/or maturation factors
- APCs antigen presenting cells
- the APCs are further loaded with one or more exogenous antigen.
- the hematological cancer cells and/or monocytes are obtained from the individual.
- the hematological cancer cells and monocytes are comprised in a mixture when cultured with the one or more of S/D/M factors.
- FIG.1 depicts ⁇ APC derived from cancer monocytes (cMO or cMo) or hematological cancer cells for the activation of cancer antigen-specific T cells.
- FIG.2 depicts the platform process for generating cancer antigen-specific T cells (e.g., NeoT) to treat hematological cancer(s).
- cancer antigen-specific T cells e.g., NeoT
- FIG.3 depicts the production process of ⁇ APC from cancer patient monocytes (cMo) and various hematological cancer cells.
- FIG.4 shows a table that summarizes results from representative studies that differentiate ⁇ APC from cMO or cancer cells from various sources.
- FIGs.5A-5C show the phenotype of B-ALL cells from Patient #1 before and after differentiation into ⁇ APC.
- FIG.5A provides the FACS analysis of PBMCs to quantify immune cell lineage percentages.
- FIG.5B displays histogram results from FACS analysis of the B-ALL cell phenotype before treatment with a hematological cancer cell activator (i.e., HC-activator) and optionally APC refinement reagent, and after differentiation into ⁇ APCs by treatment with HC-activator and optionally APC refinement reagent.
- Phenotypic markers ny-2770598 Attorney Docket No.24516-20006.40 of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86.
- FIG.5C shows a representative field using light microscopy for B-ALL cells before and after treatment with HC-activator and optionally APC refinement reagent.
- FIGs.6A-6B show the phenotype of B-ALL cells from Patient #2 before and after differentiation into ⁇ APC.
- FIG.6A provides the FACS analysis of PBMCs to quantify immune cell lineage percentages.
- FIG.6B displays histogram results from FACS analysis of the B-ALL cell phenotype before treatment with HC-activator and optionally APC refinement reagent, and after differentiation into ⁇ APCs by treatment with HC-activator and optionally APC refinement reagent.
- Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86.
- FIGs.7A-7B show the phenotype of B-ALL cells from Patient #3 before and after differentiation into ⁇ APC.
- FIG.7A provides the FACS analysis of PBMCs to quantify immune cell lineage percentages.
- FIG.7B displays histogram results from FACS analysis of the B-ALL cell phenotype before treatment with HC-activator and optionally APC refinement reagent, and after differentiation into ⁇ APCs by treatment with HC-activator and optionally APC refinement reagent.
- Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86.
- FIGs.8A-8B show the phenotype of CLL cells from Patient #4 before and after differentiation into ⁇ APC.
- FIG.8A provides the FACS analysis of PBMCs to quantify immune cell lineage percentages.
- FIG.8B displays histogram results from FACS analysis of the CLL cell phenotype before treatment with HC-activator and optionally APC refinement reagent, and after differentiation into ⁇ APCs by treatment with HC-activator and optionally APC refinement reagent.
- Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86.
- FIGs.9A-9B show the phenotype of non-Hodgkin’s lymphoma (NHL) cells from Patient #5 before and after differentiation into ⁇ APC.
- FIG.9A provides the FACS analysis of dissociated tumor cells to quantify immune cell lineage percentages.
- FIG.9B displays histogram results from FACS analysis of the NHL cell phenotype before treatment with HC- ny-2770598 Attorney Docket No.24516-20006.40 activator and optionally APC refinement reagent, and after differentiation into ⁇ APCs by treatment with HC-activator and optionally APC refinement reagent.
- Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86.
- PBMC peripheral blood mononuclear cells
- cMO cancer monocyte
- SSC side scatter
- DTC dissociated tumor cells.
- FIGs.10A-10B show the phenotype of multiple myeloma (MM) cells from Patient #6 before and after differentiation into ⁇ APC.
- FIG.10A provides the FACS analysis of PBMCs and bone marrow cells (BM) to quantify immune cell lineage percentages.
- FIG.10B displays histogram results from FACS analysis of the MM cell phenotype before treatment with HC- activator and optionally APC refinement reagent, and after differentiation into ⁇ APCs by treatment with HC-activator and optionally APC refinement reagent.
- Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86.
- PBMC peripheral blood mononuclear cells
- BM bone marrow cells
- cMO cancer monocyte
- SSC side scatter.
- FIGs.11A-11E depict the in vitro functional activity of ⁇ APC(B-ALL) to prime and activate NeoT cells.
- FIG.11A provides representative plots and a graphical representation of the percent of various cell populations found within the harvested PBMC or BM sample taken from a human B-ALL cancer patient as compared to a healthy donor.
- FIG.11B provides a schematic overview of the experimental design for preparing ⁇ APC(B-ALL) and co-culturing with NeoT isolated from the same patient for further downstream analyses (e.g., in vivo PDX cancer model and in vitro cancer cell killing assay).
- FIG.11C displays histogram results from FACS analysis of the B-ALL cell phenotype before treatment with HC-activator and optionally APC refinement reagent (“malignant B”), and after differentiation into ⁇ APCs by treatment with HC-activator and optionally APC refinement reagent (ALL- ⁇ APC).
- Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86.
- FIG.11D shows still images from a time lapse experiment wherein NeoT cells that were pre-emptively loaded with CFSE prior to co- culture were incubated with B-ALL cells in vitro for up to 10 days, and images of the cell interactions were taken using fluorescence microscopy and light microscopy.
- FIG.11E provides the quantification of NeoT cell expansion (T cell number over time) after two rounds of co-culturing with the ⁇ APC(B-ALL) cells: from day 0 to day 10 (round 1) and from day 10 to day 24 (round 2). Further provided are FACS plots of expanded effector CD4+ and CD8+ T cells as analyzed for CD45RA and CCR7 effector markers.
- FIGs.12A-12C depict in vitro cancer cell killing by NeoT cells that were primed using ⁇ APC(B-ALL) cells.
- FIG.12A provides representative plots and a graphical representation of the percent of B-ALL cells found within the harvested BM sample taken from a human B-ALL cancer patient before and after incubation with either non-specifically activated T cells (using anti-CD3 and anti-CD28 antibodies) or NeoT(B-ALL) cells at a 1:1 ratio of BM:T cells.
- FIG.12B shows the resulting IFN ⁇ production using ELISpot from BM cells alone, BM cells co-cultured 1:1 with non-specifically activated T cells, and BM cells co- cultured 1:1 with NeoT(B-ALL) activated cells (representative field image: left panel; graphical representation of the average IFN ⁇ levels: right panel).
- FIG.12C provides still images of time-lapse videos of the NeoT(B-ALL) cells homing to and killing B-ALL cells in vitro.
- BM bone marrow
- Non-specific T anti-CD3/anti-CD28 antibody-activated T cells
- h hour(s).
- FIGs.13A-13F show a B-ALL PDX murine model to assess the efficacy of NeoT(B- ALL) to clear tumors in vivo.
- FIG.13A provides a schematic overview of the PDX model experimental design from B-ALL engraftment into NSG mice, then adoptive transfer of NeoT(B-ALL) cells as a dose escalation series, and then analyzing the mice for markers of tumor clearance.
- FIG.13B shows the percent of human B-ALL cells present in murine PBMC at days 0, 6, and 14 after intravenous ACT injection compared to non-specific T cell- injected control mice.
- FIG.13C shows the percent of human B-ALL cells over time post- ACT in peripheral white blood cell samples and overall survival curves for each group tested.
- FIG.13D depicts the percent of NeoT(B-ALL) cells present in peripheral white blood cell samples over time post-ACT.
- FIG.13E demonstrates the level of markers of NeoT(B-ALL) cell activation using markers 4-1BB and CD25 to demonstrate cell activity.
- FIG.13F provides a graphical representation of the mean serum level of pro-inflammatory cytokines two days after intravenous ACT injection of NeoT(B-ALL) cells: IFN ⁇ , TNF ⁇ , IL-4, IL-6, CCL2, CCL20, and CXCL8.
- PDX patient-derived xenograft
- i.v. intravenous
- D day(s)
- ACT adoptive cellular transfer
- NeoT(B-ALL) ⁇ APC(B-ALL)-activated NeoT cells
- CR complete response
- BM bone marrow
- PBMC peripheral blood mononuclear cells
- hMHC I human major histocompatibility complex 1
- hCD19 human CD19
- Non-specific T anti- ny-2770598 Attorney Docket No.24516-20006.40
- CD3/anti-CD28 antibody-activated T cells 2x, two times or twice
- WBC white blood cells
- MFI mean fluorescence intensity.
- FIGs.14A-14B show FACS analyses of cancer cells, cancer monocytes, B cells, CD4+ T cells, and CD8+ T cells in AML patients (FIG.14A) and healthy donors (FIG. 14B).
- FIGs.15A-15B show the capacity of ⁇ APC(AML) cells to present antigens before vs. after treatment with HC-activator and optionally APC refinement reagent.
- FIG.15A shows a summary of AML studies.
- FIG.15B shows histogram plots of phenotypic markers of antigen presentation on AML cells before treatment with HC-activator and optionally APC refinement reagent, and on ⁇ APC(AML) cells after treatment with HC-activator and optionally APC refinement reagent: MHC-I, MHC-II, CD80, CD86, and CD40, wherein the increased level of each marker is indicative of increased antigen presentation capacity of the cells.
- AML cells were isolated from human patients with different stages of AML.
- FIGs.16A-16B show in vitro analyses of NeoT(AML) functional activity.
- FIG.16A provides a schematic overview of the in vitro experimental design for co-culturing ⁇ APC(AML) cells with PBLs or TILs isolated from the donor cancer patient and expanded in culture.
- FIG.16B shows light microscopy field images of NeoT(AML) cells engaging with ⁇ APC(AML) cells in vitro and clonally expanding over 72hrs, including FACS analysis of expanded cells to quantify the percentage of CD8+ and CD4+ clonally expanded T cells.
- PBL peripheral blood lymphocytes
- TIL tumor infiltrating lymphocytes
- NeoT(AML) ⁇ APC(AML)-activated NeoT cells
- Diff. differentiated.
- FIGs.17A-17C demonstrate the in vitro tumor killing activity of NeoT(AML) cells.
- FIG.17A shows the percentage of CD8 cells (representative of NeoT(AML) cells) to CFSE- loaded AML cells at 0hr and 6hr co-cultured at a 1:1 ratio.
- FIG.17B displays the number of AML cells in vitro over time at 0hr, 3hr, 6hr, 12hr, and 24hr incubations at 1:1 and 3:1 ratios of NeoT(AML) cells or Non-specific T cells to AML cells.
- FIG.17C shows FACS analysis of markers of CD8+ T cell activation in Non-specific T cells and in NeoT(AML) cells co- ny-2770598 Attorney Docket No.24516-20006.40 cultured with AML cells: IFN ⁇ , CD107a, 4-1BB, and CD25.
- FIGs.18A-18C show the successful generation of an AML PDX murine model for downstream use in the assessment of NeoT(AML) cell efficacy to clear tumors in vivo.
- FIG. 18A provides a schematic overview of the PDX model experimental design from AML engraftment into NSG mice over three generations (F0, F1, F2, and F3), to adoptive transfer of NeoT(B-ALL) cells as a dose escalation series for downstream analysis of the markers of tumor clearance.
- FIG.18B depicts the level of human AML cell engraftment in PBMC, BM, spleen, and extramedullary sites to confirm engraftment and disease progression across successive generations.
- FIG.18C shows the locations of extramedullary sites wherein the AML cells infiltrated, including lymph nodes, spleen, and peritoneal cavity.
- PBMC peripheral blood mononuclear cells
- BM bone marrow
- PDX patient-derived xenograft
- m mouse
- h human
- ACT adoptive cellular transfer
- NeoT(AML) ⁇ APC(AML)-activated NeoT cells
- LN lymph node(s).
- FIGs.19A-19E show the assessment of NeoT(AML) cell efficacy at clearing tumors in vivo using an AML PDX murine model.
- FIG.19A provides a schematic overview of the AML cell engraftment into F3 generation NSG mice followed by intravenous ACT of NeoT(AML) cells in a dose-escalation series.
- FIG. 19B provides the overall survival curve up to 180 days for the AML-engrafted mice provided no treatment, non-specific T cells, or NeoT(AML) cells.
- FIG.19C depicts the percentage of cells in the PBMC that were human AML cells up to 40 days post-ACT in cohort #1, and FIG.19D depicts the same for cohort #2.
- Cohort #1 included mice that were given ACT upon AML engraftment in the bone marrow, and cohort #2 included mice that were given ACT after AML cells had engrafted and then expanded from the bone marrow to become detectable in the PBMC. When mice reached 40% AML cells, the mice were then humanely euthanized.
- FIG.19E shows FACS analysis of the percentage of CD8+ T cells and AML cells in PBMC of mice of cohort #2 over time, including showing AML progression and regression as well as NeoT(AML) cell persistence.
- FIGs.20A-20F show the impact of TNF ⁇ neutralization on systemic inflammation and cytokine release syndrome (CRS).
- FIG.20A depicts the percentage of cells in the PBMC that were human AML cells up to 35 days post-ACT in PDX mice receiving no treatment, NeoT(AML) cells alone, or prophylactic anti-TNF ⁇ neutralizing antibody and NeoT(AML) cells.
- FIG.20B displays the overall survival curve over 150 days post-ACT of PDX mice receiving no treatment, NeoT(AML) cells alone, or prophylactic anti-TNF ⁇ neutralizing antibody and NeoT(AML) cells.
- FIG.20C provides the mean serum human cytokine levels produced by NeoT(AML) cells when the mice were either not treated or treated prophylactically with 100 ⁇ g anti-TNF ⁇ neutralizing antibody: IL-10, IL-6, IFN ⁇ , TNF ⁇ , IL- 4, and IL-2.
- FIG.20D provides the mean serum murine cytokine and chemokine levels when the mice were either not treated or treated prophylactically with 100 ⁇ g anti-TNF ⁇ neutralizing antibody: CCL2, CCL5, CXCL1, CXCL10, IL-12, TNF ⁇ , IL-6, IL-10, IL-1 ⁇ , IFN ⁇ , IFN ⁇ , and IFN ⁇ .
- FIG.20E shows the level of NeoT(AML) cell activation and expansion/proliferation after co-culturing with ⁇ APC(AML) cells for up to 10 days either without treatment or treated with anti-TNF ⁇ neutralizing antibody.
- FIG.20F shows the in vitro tumor cell killing activity of AML cells by NeoT(AML) cells co-cultured with ⁇ APC(AML) cells at 3:1 (left panel) or 1:1 (right panel) for up to 10 days either without treatment or treated with anti-TNF ⁇ neutralizing antibody, as represented by the number of AML cells over time.
- NeoT autologous cancer neoantigen-specific T cells
- ACT adoptive cell therapy
- Cancer neoantigens include genetic mutation-derived mutant peptide antigens, virus infection-derived foreign antigens (e.g., HTLV-1 infection-associated T- ALL), and aberrantly expressed peptide antigens.
- virus infection-derived foreign antigens e.g., HTLV-1 infection-associated T- ALL
- aberrantly expressed peptide antigens e.g., HTLV-1 infection-associated T- ALL
- cMO- ⁇ APCs monocyte-derived dendritic cells and macrophages that are defective under cancer conditions and possess robust capacities to phagocytose cancer antigens and to present immunogenic antigen(s) in order to activate NeoT cells.
- cMO- ⁇ APC are successfully differentiated from cMO for all tested cancer types.
- cMO- ⁇ APCs Following phagocytosis of cancer antigens, cMO- ⁇ APCs in all tested cases exhibited an exceptional ability to present immunogenic antigen(s) that induced the activation and expansion of cancer-killing NeoT cells.
- the cMO- ⁇ APC – NeoT cell system is a powerful platform for developing an immune-cell therapy against all cancers. See e.g., PCT/US2023/017002, which is incorporated herein by its entirety.
- the HC-activators are capable of promoting the survival and differentiation of not only cMO but also the differentiation of malignant myelogenous and B cell-lineage malignant leukocytes to become APCs, e.g., CLL- ⁇ APC cells.
- the HC-activator treatment induced hematological cancer cells to differentiate into cancer-specific- ⁇ APCs, for example, from acute and chronic myelogenous leukemia cells (i.e., CLL- ⁇ APC, CML- ⁇ APC), acute and chronic B lymphoblastic/ lymphocytic leukemia cells (i.e., ALL- ⁇ APC and CLL- ⁇ APC), and B lineage NHL lymphoma cells (i.e., NHL- ⁇ APC).
- acute and chronic myelogenous leukemia cells i.e., CLL- ⁇ APC, CML- ⁇ APC
- acute and chronic B lymphoblastic/ lymphocytic leukemia cells i.e., ALL- ⁇ APC and CLL- ⁇ APC
- B lineage NHL lymphoma cells i.e., NHL- ⁇ APC
- CLL- ⁇ APCs derived from malignant leukocytes directly presented their inherent neoantigens associated with genetic mutations, without the need to obtain cancer antigens (FIG.1).
- HC- ⁇ APCs hematological cancer cell-derived APCs
- NeoT cell production platform utilizes ⁇ APCs derived from malignant leukocytes to activate and expand NeoT cells from peripheral blood (PBL) and/or tumor- infiltrating lymphocytes (TILs). See, e.g., FIG.2.
- PBL peripheral blood
- TILs tumor- infiltrating lymphocytes
- the present application provides novel compositions and agents that convert cancer cells (e.g., B-ALL, CLL, NHL, CML, or AML cells) from cancer patients (e.g., patients having a hematological cancer) into powerful antigen presenting cells (hereinafter referred to as “HC-APCs”) specifically presenting cancer antigens.
- HC-APCs powerful antigen presenting cells
- These APCs can in turn be used to activate immune cells (e.g., T cells), rendering them highly effective therapeutic agents for cancer treatment.
- the present application provides methods of generating HC-APCs from hematological cancer cells (such as B-ALL cells, AML cells), use of the HC-APCs to activate immune cells, and use of the activated immune cells in treating cancer.
- Hematological cancer cells refer to cancer cells obtained from a hematological cancer.
- the hematological cancer is a myeloid leukemia, a B-cell leukemia, or a B-cell lymphoma.
- the hematological cancer is selected from the group consisting of AML, B-ALL, CML, CLL, and NHL.
- the present application in one aspect provides methods of producing a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), comprising: a) obtaining hematological cancer cells from an individual having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF ⁇ receptor (TNFR) activator, 3) an IL-4 receptor (IL-4R) activator, and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, thereby producing the population of HC-APCs.
- S/D/M factors survival, differentiation, and/or maturation factors
- the present application in another aspect provides a population of HC-APCs, such as HC-APCs generated by some of the above methods and use of the HC-APCs for cancer treatment.
- the present application in another aspect provides methods of producing activated immune cells, comprising: a) producing a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”) according to methods described herein, and b) contacting the HC-APCs with immune cells, thereby producing activated ny-2770598 Attorney Docket No.24516-20006.40 immune cells.
- HC-APC hematological cancer
- the present application in another aspect provides methods of producing activated immune cells, comprising contacting immune cells with a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), thereby generating activated immune cells; wherein the HC-APCs are derived from hematological cancer cells from an individual after having been contacted with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF ⁇ receptor (TNFR) activator, 3) an IL-4 receptor (IL-4R) activator, and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator.
- HC-APC hematological cancer
- S/D/M factors maturation factors
- the present application in another aspect provides methods of treating a hematological cancer in an individual, comprising administering activated immune cells produced by methods described herein. In some embodiments, the methods further comprise administering a TNF ⁇ inhibitor. In some embodiments, the TNF ⁇ inhibitor is an anti-TNF ⁇ antibody.
- TNF ⁇ inhibitor is an anti-TNF ⁇ antibody.
- An individual includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate. In some embodiments, the individual is human. In some embodiments, an individual suffers from a disease, such as cancer. In some embodiments, the individual is in need of treatment.
- “Monocytes,” “cancer cells,” “AML cells,” “B-ALL cells,” “immune cells,” “activated immune cells,” and “cells”, as used herein, are understood to refer not only to the monocytes, cancer cells, AML cells, B-ALL cells, immune cells, activated immune cells, or cells when obtained, but to the progeny or potential progeny of such cells.
- “High level” or “higher level,” “low level” or “lower level,” when referring to expression of a surface molecule in a population of cells refers to how the average expression level of the particular surface molecule on the population of cells compares to the average level of the surface molecule on a reference cell population.
- the reference cell population refers to a corresponding cell population derived from a healthy donor.
- a high level of a particular molecule is defined when the expression level of the molecule on the recited cell population is at least about 20% (such as about any of 20%, 30%, 40%, 50%, or more) higher than that on the reference cell population.
- a low level of a particular molecule is defined when the expression level of the molecule on the recited cell population is at least about 20% (such as about any of 20%, 30%, 40%, 50%, or less) lower than that on the reference cell population.
- a reference is obtained from a non-diseased or non-treated sample of an individual. In some examples, a reference is obtained from one or more healthy individuals who are not the individual or patient.
- the term "antigen" is a substance that induces an immune response. Such a substance can include, for example, a toxin, a chemical, a bacterium, a virus, a protein or peptide fragment thereof, a polysaccharide, a lipid, an allergen, a nucleic acid, etc.
- neoantigen is an antigen that has at least one alteration that makes it distinct from the corresponding wild-type parental antigen, e.g., via mutation in a tumor cell or post-translational modification specific to a tumor cell.
- a neoantigen can include a polypeptide sequence.
- a mutation that results in a neoantigen can include a frameshift or non-frameshift insertion or deletion (“indel”), missense or nonsense substitution, splice site alteration, genomic rearrangement or gene fusion, or any genomic or expression alteration giving rise to a neoORF (i.e., a new open reading frame).
- a mutation can also include a splice variant.
- Post-translational modifications specific to a tumor cell can include aberrant phosphorylation.
- Post-translational modifications specific to a tumor cell can ny-2770598 Attorney Docket No.24516-20006.40 also include a proteasome-generated spliced antigen. See Liepe et al., A large fraction of HLA class I ligands are proteasome-generated spliced peptides; Science.2016 Oct 21;354(6310):354-358.
- tumor neoantigen or “cancer neoantigen” is a neoantigen present in a subject's tumor cell or tumor tissue but not in the subject's corresponding normal cell or normal tissue.
- peptide refers to a polymer of amino acids of no more than 100 amino acids (including fragments of a protein), which may be linear or branched, comprise modified amino acids, and/or be interrupted by non-amino acids. The term also encompasses an amino acid polymer that has been modified naturally or by intervention, including, for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification.
- polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids, etc.
- the peptides described herein may be naturally occurring, obtained or derived from a natural source (e.g., blood) or synthesized (e.g., chemically synthesized or synthesized by recombinant DNA techniques).
- epitopope is the specific portion of an antigen typically bound by an antibody or T-cell receptor.
- immunoogenic is the ability to elicit an immune response, e.g., via T-cells, B cells, or both.
- HLA binding affinity or “MHC binding affinity” means affinity of binding between a specific antigen and a specific MHC allele.
- HLA type is the complement of HLA gene alleles.
- activated T cells refer to a population of monoclonal (e.g., encoding the same TCR) or polyclonal (e.g., with clones encoding different TCRs) T cells that have T cell receptors that recognize at least one tumor antigen peptide.
- Activated T cells may contain one or more subtypes of T cells, including, but not limited to, cytotoxic T cells (e.g., CD8 T cells), helper T cells (e.g., CD4 T cells), natural killer T cells, ⁇ T cells, regulatory T cells, and memory T cells.
- cytotoxic T cells e.g., CD8 T cells
- helper T cells e.g., CD4 T cells
- natural killer T cells e.g., ⁇ T cells
- regulatory T cells e.g., regulatory T cells
- memory T cells e.g., Activated T cells
- beneficial or desired ny-2770598 Attorney Docket No.24516-20006.40 clinical results include, but are not limited to, one or more of the following: decreasing one or more symptoms resulting from the disease; diminishing the extent of the disease; stabilizing the disease (e.g., preventing or delaying the worsening of the disease); preventing or delaying the spread (e.g., metastasis) of the disease; preventing or delaying the occurrence or recurrence of the disease; delay or slowing the progression of the disease; ameliorating the disease state; providing a remission (whether partial or total) of the disease; decreasing the dose of one or more other medications required to treat the disease; delaying the progression of the disease; increasing the quality of life; and/or prolonging survival.
- treatment is a reduction of pathological consequence(s) of cancer.
- the methods of the invention contemplate any one or more of these aspects of treatment.
- “delaying” the development of cancer means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease.
- a method that “delays” development of cancer is a method that reduces probability of disease development in a given time frame and/or reduces the extent of the disease in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies using a statistically significant number of individuals.
- Cancer development can be detectable using standard methods, including, but not limited to, computerized axial tomography (CAT Scan), Magnetic Resonance Imaging (MRI), abdominal ultrasound, clotting tests, arteriography, or biopsy. Development may also refer to cancer progression that may be initially undetectable and includes occurrence, recurrence, and onset.
- the term “simultaneous administration,” as used herein, means that a first therapy and a second therapy in a combination therapy are administered with a time separation of no more than about 15 minutes, such as no more than about any of 10, 5, or 1 minutes.
- the first and second therapies may be contained in the same composition (e.g., a composition comprising both a first therapy and a second therapy) or in separate compositions (e.g., a first therapy is contained in one composition and a second therapy is contained in another composition).
- the term “sequential administration” means that the first therapy and second therapy in a combination therapy are administered with a time separation of more than about 15 minutes, such as more than about any of 20, 30, 40, 50, 60, or more minutes. Either ny-2770598 Attorney Docket No.24516-20006.40 the first therapy or the second therapy may be administered first.
- the first and second therapies are contained in separate compositions, which may be contained in the same or different packages or kits.
- the term “concurrent administration” means that the administration of the first therapy and of the second therapy in a combination therapy overlap with each other.
- pharmaceutically acceptable or “pharmacologically compatible” is meant a material that is not biologically or otherwise undesirable, e.g., the material may be incorporated into a pharmaceutical composition administered to an individual without causing any significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained.
- Pharmaceutically acceptable carriers or excipients have preferably met the required standards of toxicological and manufacturing testing and/or are included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug Administration.
- embodiments of the application described herein include “consisting of” and/or “consisting essentially of” embodiments.
- references to “about” a value or parameter herein includes (and describes) variations that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X”.
- reference to “not” a value or parameter generally means and describes “other than” a value or parameter. For example, the method is not used to treat cancer of type X means the method is used to treat cancer of types other than X.
- the term “about X-Y” used herein has the same meaning as “about X to about Y.”
- the present application in one aspect provides methods of producing potent antigen presenting cells that are derived from cancer cells (e.g., hematological cancer cells from an individual having the hematological cancer). It was found that these cancer cells (e.g., primary cancer cells), once obtained in an in vitro setting, normally could not survive more than 2-5 days.
- cancer cells e.g., hematological cancer cells from an individual having the hematological cancer. It was found that these cancer cells (e.g., primary cancer cells), once obtained in an in vitro setting, normally could not survive more than 2-5 days.
- the methods described herein have been proven to consistently keep the primary cancer cells obtained from various donors with various hematological cancers (e.g., B-ALL, AML, CML, CLL, and NHL) alive for one or more weeks and effectively promote their differentiation into potent antigen presenting cells which present their endogenous cancer-specific antigens on the cell surface.
- hematological cancers e.g., B-ALL, AML, CML, CLL, and NHL
- the present application provides various methods of stimulating a population of hematological cancer cells (e.g., AML cells, CLL cells, CML cells, NHL cells, or B-ALL cells) from an individual to produce a population of APCs (“HC-APCs”).
- the methods involve obtaining hematological cancer cells from an individual and contacting the hematological cancer cells with one or more survival, differentiation, and/or maturation factors (“S/D/M factors”).
- S/D/M factors survival, differentiation, and/or maturation factors
- Exemplary one or more S/D/M factors are discussed in Section “Survival, differentiation, and/or maturation factors (‘S/D/M factors’)” below.
- the hematological cancer cells can be obtained from PBMC, enriched leukemia cells, a bone marrow biopsy, or a lymphoma biopsy from the individual having the hematological cancer.
- the cells are freshly obtained from the individual.
- the cells have been subjected to freeze-thaw procedures. Without being bound by theory, freeze-thaw procedures are for preservation of the cells.
- the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors.
- the hematological cancer cells are B cell malignancy cancer cells.
- B cell malignancy cancer cells are cancer cells from a B-cell lymphoma or a B cell leukemia.
- the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors in the presence of an anti-CD19 antibody, an anti- CD20 antibody, or an anti-CD22 antibody.
- the hematological cancer cells are cancer cells from a myeloid leukemia, and the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors in the presence of an anti-CD11b antibody.
- the hematological cancer cells are comprised in a mixture of cells when brought into contact with the one or more S/D/M factors.
- the hematological cancer cells are comprised in a PBMC or bone marrow sample obtained from the individual when being contacted with the one or more S/D/M factors.
- the hematological cancer cells constitute at least about any of 50%, 60%, 70%, 80%, or 90% of the mixture of the cells when contacting with the one or more S/D/M factors.
- the mixture of cells also comprises monocytes.
- the monocytes are cancer monocytes.
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual having the hematological cancer, and b) contacting the hematological cancer cells with one or more survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF ⁇ receptor (TNFR) activator, 3) an interleukin-4 receptor (IL-4R) activator, and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, thereby producing the population of HC-APCs.
- the individual is a human patient.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml.
- the one or more of S/D/M factors is comprised in a T cell culture medium (e.g., exemplary T cell culture illustrated in the examples).
- the one or more of S/D/M factors is comprised in a CD4 T cell culture medium obtained from a culture of primary CD4 T cells.
- the culture of primary CD4 T cells is at 2 or 3 days of culture.
- the T cell medium ny-2770598 Attorney Docket No.24516-20006.40 has a low level of IL-2 or does not constitute IL-2.
- the T cell culture medium is further supplemented with a STAT3 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a STAT3 activator e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1.
- S/D/M factors survival, differentiation, and/or maturation factors
- the one or more of S/D/M factors comprise: 1) a STAT3 activator, 2) a TNF ⁇ receptor (TNFR) activator, 3) an IL-4 receptor (IL-4R) activator, and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, thereby obtaining a population of HC-APCs.
- the one or more of S/D/M factors are present in a single composition. In some embodiments, at least one of the one or more of S/D/M factors is provided separately from one of the other S/D/M factors in the one or more of S/D/M factors. In some embodiments, the one or more of the S/D/M factors further comprises a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF). In some embodiments, the one or more of the S/D/M factors further comprises an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- GM-CSFR GM-CSF receptor
- IL-6R IL-6 receptor
- the cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors.
- the one or more of S/D/M factors are comprised in a composition derived from a medium (e.g., supernatant) derived from a culture of T cells after being treated with anti-CD3 and anti-CD28 antibodies.
- the T cells are CD4 T cells.
- the T cells are CD8 T cells.
- the T cells are isolated from PBMC of the same individual or a different individual and have not been previously treated with anti-CD3 and/or anti-CD28 antibodies prior to the treatment.
- the T cells are isolated from PBMC of the same individual or a different individual and have been previously treated with anti-CD3 and/or anti-CD28 antibodies prior to the treatment.
- the medium is derived from the culture after the T cells are treated with anti-CD3 and anti-CD28 antibodies for about 1-3 days, optionally for about 2 days.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L.
- the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml.
- the T cell culture is further supplemented with a STAT3 ny-2770598 Attorney Docket No.24516-20006.40 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 3) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs.
- S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL
- a method of producing a population of HC-APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, and 3) TNF ⁇ , wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs.
- S/D/M factors survival, differentiation, and/or maturation factors
- the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml).
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the IL-4 is a human IL-4 or a human recombinant IL-4.
- the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml).
- the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L.
- the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L.
- the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml.
- the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs.
- S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10,
- a method of producing a population of HC-APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, and 3) IFN ⁇ , wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs.
- S/D/M factors survival, differentiation, and/or maturation factors
- the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4.
- the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml).
- the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L.
- the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L.
- the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml.
- the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), wherein at least IL-4R activator is provided separately from the STAT3 activator or IFNGR activator, thereby obtaining a population of HC-APCs.
- STAT3 activator e.g., IL-10, e.g.,
- a method of producing a population of HC-APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, and 3) IFN ⁇ , wherein at least IL-4 is provided separately from the IL-10 or IFN ⁇ , thereby obtaining a population of HC-APCs.
- S/D/M factors survival, differentiation, and/or maturation factors
- the IL-4R activator or IL-4 is provided after the STAT3 activator or IL-10 is provided. In some embodiments, the IL-4R activator or IL-4 is provided after the IFNGR activator or IFN ⁇ is provided. In some embodiments, the STAT3 activator or IL-10 and the IFNGR activator or IFN ⁇ are provided simultaneously. In some embodiments, the STAT3 activator or IL-10 and the IFNGR activator or IFN ⁇ are provided sequentially. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10.
- the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml).
- the IL-4 is a human IL-4 or a human recombinant IL-4.
- the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml).
- the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of ny-2770598 Attorney Docket No.24516-20006.40 at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml.
- the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a STAT3 activator e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1.
- S/D/M factors survival, differentiation, and/or maturation factors
- a method of producing a population of HC-APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) TNF ⁇ , and 3) IFN ⁇ , wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs.
- S/D/M factors survival, differentiation, and/or maturation factors
- the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of ny-2770598 Attorney Docket No.24516-20006.40 at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L.
- the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L.
- the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml.
- the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), wherein at least STAT3 activator is provided separately from the TNFR activator or IFNGR activator, thereby obtaining a population of HC-APCs.
- STAT3 activator e.g., IL-10, e.g.,
- a method of producing a population of HC-APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) TNF ⁇ , and 3) IFN ⁇ , wherein at least IL-10 is provided separately from the TNF ⁇ or IFN ⁇ , thereby obtaining a population of HC-APCs.
- S/D/M factors survival, differentiation, and/or maturation factors
- the STAT3 activator or IL-10 is provided before the TNFR activator or TNF ⁇ is provided. In some embodiments, the STAT3 activator or IL-10 is provided before the IFNGR activator or IFN ⁇ is provided. In some embodiments, the TNFR activator or TNF ⁇ and the IFNGR activator or IFN ⁇ are provided simultaneously. In some embodiments, the TNFR activator or TNF ⁇ and the IFNGR activator or IFN ⁇ are provided sequentially. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10.
- the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml).
- the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about ny-2770598 Attorney Docket No.24516-20006.40 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L.
- the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a STAT3 activator e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1.
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of STAT3 activator (e.g.,
- a method of producing a population of HC-APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, 3) TNF ⁇ , and 4) IFN ⁇ , wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs.
- S/D/M factors survival, differentiation, and/or maturation factors
- the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, ny-2770598 Attorney Docket No.24516-20006.40 optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the IL-4 is a human IL-4 or a human recombinant IL-4.
- the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml).
- the hematological cancer cells are cultured for about 1-3 days (e.g., 2- 3 days) in the presence of at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L.
- the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L.
- the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml.
- the T cell culture is further supplemented with a STAT3 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), wherein at least one of the one or more of S/D/M factors is provided separately from at least one of the other S/
- S/D/M factors comprise: 1)
- a method of producing a population of HC-APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, 3) TNF ⁇ , and 4) IFN ⁇ , wherein at least one of the one or more of S/D/M factors is provided separately from at least one of the other ny-2770598 Attorney Docket No.24516-20006.40 S/D/M factors in the one or more of S/D/M factors, thereby obtaining a population of HC- APCs.
- S/D/M factors survival, differentiation, and/or maturation factors
- the STAT3 activator or IL-10 is provided before at least one of the other factors (e.g., the IL-4R activator or IL-4) is provided. In some embodiments, the IL- 4R activator or IL-4 is provided after at least one of the other factors (e.g., the STAT3 activator or IL-10, the IFNGR activator or IFN ⁇ , or the TNFGR activator or TNF ⁇ ) is provided. In some embodiments, the STAT3 activator or IL-10, the TNFR activator or TNF ⁇ , and the IFNGR activator or TNF ⁇ are provided simultaneously.
- the STAT3 activator or IL-10, the TNFR activator or TNF ⁇ , and the IFNGR activator or IFN ⁇ are provided sequentially.
- the IL-4R activator or IL-4, the TNFR activator or TNF ⁇ , and the IFNGR activator or IFN ⁇ are provided simultaneously.
- the IL-4R activator or IL-4, the TNFR activator or TNF ⁇ , and the IFNGR activator or IFN ⁇ are provided sequentially.
- the IL-10 is a human IL- 10 or a human recombinant IL-10.
- the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml).
- the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ . In some embodiments, the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4.
- the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml).
- the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L.
- the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a STAT3 activator e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1.
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 5) one or both of a GM
- STAT3 activator e.g., IL-10, e.g., IL-12
- a method of producing a population of HC-APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, 3) TNF ⁇ , 4) IFN ⁇ , and 5) one or both of GM-CSF and IL-6, wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs.
- S/D/M factors survival, differentiation, and/or maturation factors
- the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6.
- the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 ny-2770598 Attorney Docket No.24516-20006.40 pg/ml).
- the GM-CSF is a human GM-CSF or a human recombinant GM-CSF.
- the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml).
- the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L.
- the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a STAT3 activator e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1.
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 5) one or both of a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSFR) activator (e.g.
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, 3) TNF ⁇ , 4) IFN ⁇ , and 5) one or both of GM-CSF and IL-6, wherein at least one of the one or more of S/D/M factors is provided separately from at least one of the other S/D/M factors in the one or more of S/D/M factors, thereby obtaining a population of HC-APCs.
- S/D/M factors survival, differentiation, and/or maturation factors
- the STAT3 activator or IL-10, and/or the GM-CSFR activator or GM- ny-2770598 Attorney Docket No.24516-20006.40 CSF is provided before at least one of the other factors (e.g., the IL-4R activator or IL-4, e.g., the IL-6R activator or IL-6) is provided.
- the IL-4R activator or IL-4, and/or the IL-6R activator or IL-6 is provided after at least one of the other factors (e.g., the STAT3 activator or IL-10, e.g., the GM-CSFR activator or GM-CSF) is provided.
- the STAT3 activator or IL-10, the TNFR activator or TNF ⁇ , and the IFNGR activator or IFN ⁇ are provided simultaneously. In some embodiments, the STAT3 activator or IL-10, the TNFR activator or TNF ⁇ , and the IFNGR activator or IFN ⁇ are provided sequentially. In some embodiments, the IL-4R activator or IL-4, the TNFR activator or TNF ⁇ , and the IFNGR activator or IFN ⁇ are provided simultaneously. In some embodiments, the IL-4R activator or IL-4, the TNFR activator or TNF ⁇ , and the IFNGR activator or IFN ⁇ are provided sequentially.
- the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6.
- the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml).
- the GM-CSF is a human GM-CSF or a human recombinant GM-CSF.
- the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml).
- the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L.
- the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a STAT3 activator e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1.
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10 (e.g., a human IL-10 or a human recombinant IL-10), 2) IL-4 (e.g., a human IL-4 or a human recombinant IL-4), 3) TNF ⁇ (e.g., a human TNF ⁇ or a human recombinant TNF ⁇ ), 4) IFN ⁇ (e.g., a human IFN ⁇ or a human recombinant IFN ⁇ ), 5) GM-CSF (e.g., a human GM-CSF (e.g., a human
- the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in a medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6.
- the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 ny-2770598 Attorney Docket No.24516-20006.40 pg/ml).
- the GM-CSF is a human GM-CSF or a human recombinant GM-CSF.
- the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml).
- the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L.
- the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml.
- the T cell culture is further supplemented with an IL-10R activator (e.g., a STAT3 activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a method of producing a population of HC- APCs comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10 (e.g., a human IL-10 or a human recombinant IL-10), 2) IL-4 (e.g., a human IL-4 or a human recombinant IL-4), 3) TNF ⁇ (e.g., a human TNF ⁇ or a human recombinant TNF ⁇ ), 4) IFN ⁇ (e.g., a human IFN ⁇ or a human recombinant IFN ⁇ ), 5) GM-CSF (e.g., a human GM-CSF (e.g., a human
- IL-10 and/or GM-CSF is provided before at least one of the other factors (e.g., IL-4 or IL-6) is provided. In some embodiments, IL-4 and/or IL-6 is provided after at least one of the other factors (e.g., IL-10 or GM-CSF) is provided. In some embodiments, IL-10, TNF ⁇ , and IFN ⁇ are provided simultaneously. In some embodiments, IL-10, TNF ⁇ , and the IFN ⁇ are provided sequentially. In some embodiments, IL-4, TNF ⁇ , and the IFN ⁇ are provided simultaneously. In some embodiments, IL-4, TNF ⁇ , and the IFN ⁇ are provided sequentially.
- the IL-10 is a human IL-10 or a human recombinant IL-10.
- the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, ny-2770598 Attorney Docket No.24516-20006.40 further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml).
- the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6.
- the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml).
- the GM-CSF is a human GM-CSF or a human recombinant GM-CSF.
- the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml).
- the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L.
- the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a STAT3 activator e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1.
- a method of stimulating a population of cancer cells comprising contacting the population of cancer cells with a medium derived from a culture (e.g., a supernatant) of T cells after being treated with anti-CD3 and anti-CD28 antibodies, wherein the medium comprises a STAT3 activator (e.g., IL-10).
- a culture e.g., a supernatant
- the medium comprises a STAT3 activator (e.g., IL-10).
- the ny-2770598 Attorney Docket No.24516-20006.40 medium further comprises an IL-4R activator (e.g., IL-4), an IFNGR activator (e.g., IFN ⁇ ), a TNFR activator (e.g., TNF ⁇ ).
- the medium further comprises a GM- CSFR activator (e.g., GM-CSF) and/or an IL-6R activator (e.g., IL-6).
- the T cells are isolated from PBMC of the same individual or a different individual and have not been previously treated with anti-CD3 and/or anti-CD28 antibodies prior to the treatment.
- the medium is derived from the culture after the T cells are treated with anti-CD3 and anti-CD28 antibodies for about 1-3 days, optionally for about 2 days.
- the cancer cells are cultured for about 2-3 days in the presence of the medium derived from the culture of T cells.
- the IL-10 is a human IL- 10 or a human recombinant IL-10.
- the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml).
- the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ . In some embodiments, the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the IL-4 is a human IL-4 or a human recombinant IL-4.
- the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml).
- the IL-6 is a human IL-6 or a human recombinant IL-6. In some embodiments, the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml). In some embodiments, the GM-CSF is a human GM-CSF or a human recombinant GM-CSF.
- the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml).
- the cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors.
- the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L.
- the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 ny-2770598 Attorney Docket No.24516-20006.40 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1).
- a STAT3 activator e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1.
- the level of IL-10R on the hematological cancer cells before contacting the S/D/M factors is at least about any of 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-10R on corresponding cells from a reference individual (e.g., a healthy individual).
- the level of IL-4R on the cancer cells before contacting the S/D/M factors is at least about any of 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-4R on corresponding cells from a reference individual (e.g., a healthy individual).
- the method further comprises contacting the population of cancer cells (e.g., AML cells, CLL cells, CML cells, NHL cells, or B-ALL cells) with a one or more of refinement factors selected from the group consisting of type-I interferon, IFN ⁇ , TNF ⁇ , a TLR ligand, CD40L or a CD40-ligating antibody, an anti-PD-L1 antibody, and TPI-1, optionally wherein the type-I interferon comprises IFN ⁇ and/or IFN ⁇ , and optionally wherein the TLR ligand is R848, poly IC, CpG, or LPS.
- a one or more of refinement factors selected from the group consisting of type-I interferon, IFN ⁇ , TNF ⁇ , a TLR ligand, CD40L or a CD40-ligating antibody, an anti-PD-L1 antibody, and TPI-1, optionally wherein the type-I interferon comprises IFN ⁇ and/or IFN ⁇ , and optionally wherein the TLR lig
- the one or more of refinement factors is provided after the plurality of cancer cells are contacted with the one or more of S/D/M factors or the medium derived from the culture of T cells, thereby producing the population of APCs, and wherein the population of APCs is cultured for about 1-5 days in the presence of the one or more of the refinement factors, optionally wherein the population of APCs is cultured for about one day.
- the one or more of refinement factors are provided when: a) at least about 50% of the cancer cells differentiate into APCs and survive, b) at least about 30% of the population of APCs exhibit a dendritic cell morphology, and/or c) the population of APCs express: i) a high level of one or more molecules selected from the group consisting of MHC I, MHC II, CD80, CD86, and/or CD40, and/or ii) a low level of SIRP ⁇ .
- the method further comprises contacting the population of cancer cells with one or more of refinement factors comprising IFN ⁇ , IFN ⁇ , and TNF ⁇ .
- the one or more of refinement factors comprise IFN ⁇ , IFN ⁇ , TNF ⁇ , R848, poly IC, and CpG.
- the one or more of refinement factors comprise IFN ⁇ , IFN ⁇ , TNF ⁇ , R848, poly IC, CpG, CD40L, and an anti-PD-L1 antibody.
- the one or more of refinement factors comprise IFN ⁇ , IFN ⁇ , TNF ⁇ , R848, poly IC, CpG, CD40L, TPI-1, and an anti-PD-L1 antibody.
- survival, differentiation, and/or maturation factors (“S/D/M factors”) [0118]
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises one or more agents selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an interleukin-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- IL-4R interleukin-4 receptor
- IFN ⁇ interferon ⁇
- IFNGR interferon ⁇
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- IL-4R IL-4 receptor
- IFN ⁇ interferon ⁇
- IFN ⁇ interferon ⁇ receptor
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises one or more agents selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL- 4), and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- IL-4R IL-4 receptor
- IFN ⁇
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- IL-4R IL-4 receptor
- IFN ⁇ interferon ⁇
- IFN ⁇ interferon ⁇ receptor
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 6) an IL-6 receptor (IL-6R) activator (e.g., IL-6R) activator (e.g.
- the STAT3 activator is selected ny-2770598 Attorney Docket No.24516-20006.40 from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises one or more agents selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL- 4), 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- IL-4R IL-4 receptor
- IFN ⁇ interferon ⁇
- GM-CSFR GM-CSF receptor
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- IL-4R IL-4 receptor
- IFN ⁇ interferon ⁇
- GM-CSFR GM-CSF receptor
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises one or more agents selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL- 4), 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 6) an IL-6 receptor (IL
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 6) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- IL-4R IL-4 receptor
- IFN ⁇ interferon ⁇
- GM-CSFR GM-CSF
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) IFN ⁇ .
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein ny-2770598 Attorney Docket No.24516-20006.40 comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) TNF ⁇ .
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) IL-6.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) IL- 4.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) GM-CSF.
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 2) TNF ⁇ .
- IFN ⁇ interferon ⁇
- TNF ⁇ TNF ⁇
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 2) IL-6.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 2) IL-4.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 2) GM-CSF.
- IFN ⁇ interferon ⁇
- IFNGR interferon ⁇ receptor
- GM-CSF GM-CSF
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 2) IFN ⁇ .
- TNFR TNF ⁇ receptor
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 2) IL-6.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 2) IL-4.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 2) GM-CSF.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a GM-CSF receptor (GM-CSFR) ny-2770598 Attorney Docket No.24516-20006.40 activator (e.g., GM-CSF), and 2) IFN ⁇ .
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 2) IL-6.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 2) IL-4.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 2) TNF ⁇ .
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 2) IFN ⁇ .
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 2) IL-6.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 2) GM-CSF.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 3) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF).
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- GM-CSFR GM-CSF receptor
- IFN ⁇ interferon ⁇ receptor
- ny-2770598 Attorney Docket No.24516-20006.40 the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL- 24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 2) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- TNFR TNF ⁇ receptor
- GM-CSFR GM-CSF receptor
- IFNGR interferon ⁇
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4).
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- TNFR TNF ⁇ receptor
- IL-4R IL-4 receptor
- IFN ⁇ interferon ⁇ receptor
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) ny-2770598 Attorney Docket No.24516-20006.40 activator (e.g., IFN ⁇ ).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- IL-4R IL-4 receptor
- IFN ⁇ interferon ⁇ receptor
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF).
- TNFR TNF ⁇ receptor
- IL-4R an IL-4 receptor
- GM-CSFR GM-CSF receptor
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 4) a GM-CSF receptor (GM- CSFR) activator (e.g., GM-CSF).
- TNFR TNF ⁇ receptor
- IFN ⁇ interferon ⁇
- GM- CSFR GM-CSF receptor
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) a GM- ny-2770598 Attorney Docket No.24516-20006.40 CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- GM-CSFR CSF
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 4) a GM-CSF receptor (GM- CSFR) activator (e.g., GM-CSF).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- an IL-4 receptor (IL-4R) activator e.g., IL-4
- IFN ⁇ inter
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- an IL-4 receptor (IL-4R) activator e.g., IL-4
- IFN ⁇ interferon ⁇
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a GM- CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- an IL-4 receptor (IL-4R) activator e.g., IL-4
- GM-CSFR GM- CSF receptor
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 3) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- IFN ⁇ interferon ⁇
- GM-CSFR GM-CSF receptor
- IL-6R IL-6 receptor
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. ny-2770598 Attorney Docket No.24516-20006.40 [0146]
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 4) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF).
- TNF ⁇ receptor e.g., TNF ⁇
- IL-4R IL-4 receptor
- IFN ⁇ interferon ⁇
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL- 6).
- TNF ⁇ receptor e.g., TNF ⁇
- IL-4R IL-4 receptor
- IFN ⁇ interferon ⁇
- IFNGR interferon ⁇
- IL-6R IL-6 receptor activator
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a GM-CSF receptor (GM- CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- TNF ⁇ receptor e.g., TNF ⁇
- IL-4R IL-4 receptor
- GM- CSFR GM-CSF receptor
- IL-6R IL-6 receptor
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 2) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 3) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- TNF ⁇ receptor e.g., TNF ⁇
- IFN ⁇ interferon ⁇
- GM-CSFR GM-CSF receptor
- IL-6R IL-6 receptor
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 2) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 3) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- IL-4R IL-4 receptor
- IFN ⁇ interferon ⁇
- GM-CSFR GM-CSF receptor
- IL-6R IL-6 receptor
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- IL-4R IL-4 receptor
- IFN ⁇ interferon ⁇
- GM-CSFR GM-CSF receptor
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL- 19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. ny-2770598 Attorney Docket No.24516-20006.40 [0152]
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), and 5) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- TNFR TNF ⁇ receptor
- IL-4R
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 4) a GM-CSF receptor (GM- CSFR) activator (e.g., GM-CSF), and 5) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- the STAT3 activator is selected from the group consisting of IL-10, IL- 22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 4) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 5) an IL-6 receptor (IL-6R) activator (e.g., IL-6).
- TNF ⁇ receptor TNFR
- IL-4R IL-4 receptor
- IFNGR interferon ⁇
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10), and 2) IL-12.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) poly:IC.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) CpG.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) R848.
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. ny-2770598 Attorney Docket No.24516-20006.40 [0156]
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- a STAT3 activator e.g., IL-10, e.g., any of those listed in Table 1
- TNFR TNF ⁇ receptor
- IFNGR interferon ⁇
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6), and 5) a GM-CSF receptor (GM-CSF) activator (e.g., GM-CSF).
- a STAT3 activator e.g., IL-10, e.g., any
- the STAT3 activator is selected from the group consisting of IL-10, IL- 22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an IL-22, 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6), and 5) a GM-CSF receptor (GM- CSF) activator (e.g., GM-CSF).
- TNF ⁇ receptor e.g., TNF ⁇
- IFN ⁇ interferon ⁇
- IFNGR interferon ⁇
- IL-6R IL-6 receptor
- GM- CSF
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 2) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 3) an IL-6 receptor (IL-6R) activator (e.g., IL-6), and 4) a GM-CSF receptor (GM-CSF) activator (e.g., GM-CSF).
- TNF ⁇ receptor e.g., TNF ⁇
- IFN ⁇ interferon ⁇
- IFNGR interferon ⁇
- IL-6R IL-6 receptor
- GM-CSF GM-CSF
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6), 5) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 6) a GM-CSF receptor (GM-CSF) activator (e.g., GM-CSF).
- a STAT3 activator e.g., IL-10, e.g., any of those listed in Table 1
- a TNF ⁇ receptor (TNFR) activator e.g., TNF ⁇
- IFN ⁇ inter
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. ny-2770598 Attorney Docket No.24516-20006.40 [0161]
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described above are provided in a composition comprising a serum (e.g., human serum).
- the serum comprises human serum, fetal bovine serum, or murine serum, etc.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein are provided in a composition comprising a serum (e.g., human serum).
- a serum e.g., human serum.
- the one or more S/D/M factors is selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an interleukin-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- IL-4R interleukin-4 receptor
- IFN ⁇ interferon ⁇
- IFNGR interferon ⁇
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein are provided in a composition comprising a serum (e.g., human serum), wherein the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) combined in serum described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- a STAT3 activator e.g., IL-10 or any of those listed in Table 1
- TNFR TNF ⁇ receptor
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL- 19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein are provided in a composition comprising a serum (e.g., human serum), wherein the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) combined in serum described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-r), and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇
- a STAT3 activator e
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein are provided in a composition comprising a serum (e.g., human serum).
- the one or more S/D/M factors is selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an interleukin-4 receptor (IL-4R) ny-2770598 Attorney Docket No.24516-20006.40 activator (e.g., IL-4), 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 6) an IL-6 receptor (IL-6R) activator (e.g., IFN ⁇ ), IFN ⁇ , GM-
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone [0166]
- the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein are provided in a composition comprising a serum (e.g., human serum), wherein the one or more S/D/M factors comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), 3) an interleukin-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), 5) a GM-CSF receptor (GM-CSFR) activator (e
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D.
- IL-10 receptor (IL-10R) activator and IL-10 [0167] “IL-10 receptor (IL-10R) activator” described herein refers to a molecule that activates the IL-10 receptor-mediated signaling pathway. IL-10R includes both IL-10R1 and IL-10R2.
- Interleukin 10 also known as human cytokine synthesis inhibitory factor (CSIF) is an anti-inflammatory cytokine. In humans, interleukin 10 is encoded by the IL10 gene.
- IL-10 is encoded by the IL10 gene, which is located on chromosome 1 and comprises 5 exons, and is primarily produced by monocytes and, to a lesser extent, lymphocytes, namely type-II T helper cells (TH2), mast cells, CD4+CD25+Foxp3+ regulatory T cells, and in a certain subset of activated T cells and B cells.
- IL-10 can be produced by monocytes upon PD-1 triggering in these cells.
- ny-2770598 Attorney Docket No.24516-20006.40
- IL-10 is a cytokine with multiple, pleiotropic, effects in immunoregulation and inflammation.
- IL-10 can block NF- ⁇ B activity and is involved in the regulation of the JAK-STAT signaling pathway. [0171] IL-10 was initially reported to suppress cytokine secretion, antigen presentation, and CD4+ T cell activation.
- IL-10 as referred to herein comprises any constructs that have a component of IL-10 (e.g., a naturally occurring IL-10, e.g., a recombinant IL-10). These include and are not limited to natural IL-10 (e.g., various isoforms of human IL-10), synthetic or recombinant IL- 10, and fusion proteins having an IL-10 component.
- a component of IL-10 e.g., a naturally occurring IL-10, e.g., a recombinant IL-10.
- the IL-10R activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety.
- An Fc portion is a portion of the fragment crystallizable region (Fc region), which is the tail region of an antibody that can interact with cell surface receptors (e.g., Fc receptors and some proteins of the complement system).
- a PEG moiety is a polyethylene glycol polymer chain that can be attached, e.g., to a molecule such as an antibody and can extend the duration of time in which said molecule circulates before being degraded or reduced by renal clearance.
- the IL-10R activator is selected from the group consisting of: an IL-10 (e.g., a pegylated IL-10, e.g., pegilodecakin or AM0010), an IL-10 family member (e.g., IL-19, IL-20, IL-22, IL-24, IL-26, IL-28, IL-7, IL-9, IL-15, and IL-21), an IL-10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, a small molecule activator of IL-10R, and an activator of the IL-10R-downstream STAT3 (e.g., IL-12, e.g., Long noncoding RNA (LncRNA) PVT1, NEAT1, FEZF1-AS1, UICC).
- an IL-10 e.g., a pegylated IL-10, e.g., pegilodecakin or AM00
- an IL-10 family member e
- the IL-10R activator is IL-10.
- the IL-10 is a human IL- 10 or a human recombinant IL-10.
- the IL-10 (e.g., a human IL-10) is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml (e.g., about 20 ng/ml).
- the IL-10 (e.g., a human IL-10) is present in the medium at a concentration of about 2 ng/ml to about 200 ng/ml (e.g., about 10 ny-2770598 Attorney Docket No.24516-20006.40 ng/ml to about 200ng/ml, e.g., about 10 ng/ml to about 100 ng/ml, e.g., about 20 ng/ml to about 100 ng/ml).
- STAT3 activator and STAT3 [0175]
- the IL-10R activator is a STAT3 activator.
- STAT3 activator refers to a molecule that activates STAT3 signaling, e.g., the STAT3 nuclear localization and transcription factor activity.
- STAT3 is a transcription factor that resides in the cytoplasm in its inactive, unphosphorylated form and translocates to the nucleus upon its activation via phosphorylation, e.g., Tyr705 phosphorylation, and subsequent dimerization.
- the activated STAT3 dimer binds to the IFN ⁇ –activated sequence (GAS) in target promoters and thereby activates transcription of target genes.
- GAS IFN ⁇ –activated sequence
- tyrosine kinases have been described as intracellular activators of STAT3 activity (e.g., JAK1, JAK2, EGFR, Src, and ERK). Further mechanisms of activation include: i) STAT3 phosphorylation at Ser727 by protein kinase C (PKC), mitogen-activated protein kinases (MAPKs), and CDK5; and ii) STAT3 acetylation on Lys685 by histone acetyltransferase, which can enhance STAT3 dimer stability.
- PKC protein kinase C
- MAPKs mitogen-activated protein kinases
- CDK5 CDK5
- STAT3 acetylation on Lys685 by histone acetyltransferase which can enhance STAT3 dimer stability. See, e.g., Rébé et al., JAKSTAT 2013;2(1):e23010.
- STAT3 is expressed in most cell types under specific conditions, and generally is described to be involved with biological processes such as: cell proliferation, differentiation, apoptosis, angiogenesis, metastasis, inflammation, and immunity. In immune cells, STAT3 has been described in contradictory terms. For example, STAT3 has been described as promoting the differentiation of macrophages toward the M2 phenotype and the absence of functional dendritic cells (see, e.g., Rébé et al., 2013). STAT3 has also been described to promote the gp130-mediated maintenance of the pluripotential state of proliferating embryonic stem cells and for the gp130-induced macrophage differentiation of M1 cells.
- STAT3 activation is rapid and transient under normal biological conditions and mediated by many extracellular stimuli, including cytokines (IL-6, IL-10, IFNs, TNF ⁇ , LIF, OSM, etc.) and growth factors (e.g., EGF, G-CSF, GM-CSF, VEGF, HGF, GH, and Her2/Neu).
- cytokines IL-6, IL-10, IFNs, TNF ⁇ , LIF, OSM, etc.
- growth factors e.g., EGF, G-CSF, GM-CSF, VEGF, HGF, GH, and Her2/Neu.
- Active oncogenic proteins such as Src (e.g., v-Src) and Ras, as well as chemical carcinogens and other molecules also can activate STAT3.
- Src e.g., v-Src
- Ras Ras
- chemical carcinogens and other molecules also can activate STAT3.
- STAT3 regulated genes ny-2770598 Attorney Docket No.24516-20006.40 induced by STAT3 activity in turn activate the same STAT3 pathways and thereby keep a stable feedforward loop.
- the STAT3 activator comprises a cytokine selected from the group consisting of: IL-6, IL-10, IL-11, IL-12, IL-19, IL-20, IL-22, IL-23, IL-24, IL-26, IL-27, IL-7, IL-9, IL-15, IL-21, IFN ⁇ , IFN ⁇ , IFN ⁇ , TNF ⁇ , leukemia inhibitory factor (LIF), oncostatin M (OSM), biologically active derivatives thereof, and any combination thereof.
- a cytokine selected from the group consisting of: IL-6, IL-10, IL-11, IL-12, IL-19, IL-20, IL-22, IL-23, IL-24, IL-26, IL-27, IL-7, IL-9, IL-15, IL-21, IFN ⁇ , IFN ⁇ , IFN ⁇ , TNF ⁇ , leukemia inhibitory factor (LIF), oncostatin M (OSM), biologically active derivatives thereof, and any
- the STAT3 activator comprises a growth factor selected from the group consisting of: EGF, FGF, IGF, G-CSF, GM-CSF, VEGF, HGF, GF, Her2/Neu, biologically active derivatives thereof, and any combination thereof.
- the STAT3 activator comprises a JAK activator, such as an enzyme that phosphorylates JAK (e.g., JAK2).
- the STAT3 activator comprises a hormone (e.g., leptin).
- the STAT3 activator comprises a chaperone protein (e.g., HSP90, HSP70, HSP27, HSP110, HOP).
- STAT3 activity can be positively regulated by the signaling pathways of IL-10 and IL-10 family members, including IL-19, IL-20, IL-22, IL-24, and IL-26. Further, IL-12 and affiliated family members (e.g., IL-23) can activate STAT3 activity, at least in part by promoting IL-10/IL-10R production and autocrine signaling in the cancer cell-derived (e.g., B-ALL or AML) ⁇ APC cells. IL-6 has also been described as an activator of the STAT3 pathway.
- the STAT3 activator is selected from the group consisting of: IL-6, IL-10, IL-12, IL-19, IL-20, IL-22, IL-23, IL-24, IL-26, IL-7, IL-9, IL-15, IL-21, biologically active derivatives thereof, and any combination thereof.
- the STAT3 activator is an IL-10R activator, such as any described herein or known in the art.
- the IL-10R activator is an activator of the IL-10R-downstream STAT3.
- STAT3 activators can include, but are not limited to, any of: a small molecule, a nucleic acid (e.g., an siRNA, an shRNA, an antisense RNA, a microRNA), a nucleic acid base inhibitor (e.g., a circular RNA inhibitor), a nucleic acid editing system (e.g., CRISPR, ZFN, or TALENS systems), a decoy oligonucleotide, a peptide agent, a protein agent (e.g., an antibody agent that targets IL-10R; e.g., a protein agent that targets STAT3 phosphorylation and/or prevents STAT3 dephosphorylation), a protein stabilizing agent (e.g., a STAT3- stabilizing agent such as a chaperone protein, for example HSP90, HSP70, HSP27, HSP110, and/or HOP), a protein degrading or destabilizing agent (e.g., a phosphatase
- the STAT3 activator comprises a cancer cell STAT3 activator.
- cancer cell STAT3 activators can include any of: PVT1, NEAT1, FEZF1-AS1, UICC, MALAT1, XIST, miR-30d, CD109, CD146, CD24, CDK7, SOX, Smad6, Smad7, TRIM24, TRIM27, TRIM59, ADAM12, USP22, BMX AKR1C1, PRMT1, PBX1, HSP110, RanBP6, RAC1-GTP, PA28 ⁇ , E6, and FABP5.
- the STAT3 activator comprises a small molecule selected from the group consisting of: Colivelin, Colivelin TFA, Garcinone D, Butyzamide, Eflepedocokin alfa, Broussonin E, derivatives thereof, and any combinations thereof.
- the STAT3 activator comprises Colivelin, Colivelin TFA, and/or Garcinone D.
- the STAT3 activator comprises an inhibitor or antagonist of a molecule or compound that inactivates STAT3 (e.g., reduces phosphorylated STAT3 levels) or that reduces total STAT3 levels (e.g., via proteasomal degradation and/or transcriptional suppression) in a cell of interest, e.g., in a myeloid cell such as an AML-derived ⁇ APC cell.
- molecules or compounds that can inactivate STAT3 include, but are not limited to: ⁇ -elemene, selective serotonin-reuptake inhibitors (SSRIs, e.g., fluoxetine), minecoside, Luteolin (3,4,5,7-tetrahydroxyflavone), SHP-1, SHP-2, PTP1B, PTPRM, eEF2 kinase, PKM2, curcumin, cucurbitacin, honokiol, guggulsterone, resveratrol, berbamine, flavopiridol, JAK inhibitors/inactivated JAK (e.g., JAK2), low molecular weight-DSP2, PIAS3, etc.
- SSRIs selective serotonin-reuptake inhibitors
- fluoxetine selective serotonin-reuptake inhibitors
- minecoside Luteolin (3,4,5,7-tetrahydroxyflavone)
- Luteolin 3,4,5,7-tetrahydroxyflavon
- molecules or compounds that can reduce total STAT3 levels include, but are not limited to: PDLIM2, COP1, calcineurin, SOCS proteins, Rubulavirus (e.g., Mumps virus, e.g., the Mumps viral V protein, for example the V-dependent degradation complex VDC or V/DDB1/Cullin degradation complex), TSM-1, KYM-003, KTX-201, SD-36, AUY922, 17- DMAG, etc.
- PDLIM2 e.g., COP1, calcineurin
- SOCS proteins e.g., Rubulavirus (e.g., Mumps virus, e.g., the Mumps viral V protein, for example the V-dependent degradation complex VDC or V/DDB1/Cullin degradation complex), TSM-1, KYM-003, KTX-201, SD-36, AUY922, 17- DMAG, etc.
- the inhibitor of a molecule or compound that inactivates STAT3 or that reduces total STAT3 levels in a cell of interest competitively bind to STAT3 to prevent inactivation (e.g., DDIAS) or protein degradation (e.g., chaperones, such as HSP90).
- inactivation e.g., DDIAS
- protein degradation e.g., chaperones, such as HSP90.
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-12, IL-23, IL-6, colivelin TFA, Garcinone D, G-CSF, IL-7, IL-9, IL-15, and IL-21.
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-12, IL-23, IL-6, colivelin TFA, and Garcinone D.
- the STAT3 activator is selected from the group consisting of IL-10, IL-12, Colivelin TFA, and Garcinone D.
- TNF ⁇ receptor (TNFR) activator and TNF ⁇ [0189] “TNF ⁇ receptor (TNFR) activator” as described herein refers to a molecule that activates the TNFR-mediated signaling pathway. TNFR as described herein refers to either TNFR1 or TNFR2.
- Tumor necrosis factor ⁇ (TNF, cachexin, or cachectin; often called tumor necrosis factor alpha or TNF ⁇ ) is an adipokine and a cytokine.
- TNF ⁇ is a member of the TNF ⁇ superfamily, which consists of various transmembrane proteins with a homologous TNF ⁇ domain.
- TNF ⁇ can bind two receptors, TNFR1 (TNF ⁇ receptor type 1; CD120a; p55/60) and TNFR2 (TNF ⁇ receptor type 2; CD120b; p75/80).
- TNFR1 is 55-kDa and TNFR2 is 75-kDa.
- TNFR1 is expressed in most tissues, and can be fully activated by both the membrane-bound and soluble trimeric forms of TNF, whereas TNFR2 is found typically in cells of the immune system, and responds to the membrane-bound form of the TNF ⁇ homotrimer.
- TNF ⁇ was thought to be produced primarily by macrophages, but it is produced also by a broad variety of cell types including lymphoid cells, mast cells, endothelial cells, cardiac myocytes, adipose tissue, fibroblasts, and neurons. Large amounts of TNF ⁇ are released in response to lipopolysaccharide, other bacterial products, and interleukin-1 (IL-1). In the skin, mast cells appear to be the predominant source of pre-formed TNF ⁇ , which can be released upon inflammatory stimulus (e.g., LPS).
- inflammatory stimulus e.g., LPS
- TNF ⁇ as referred to herein comprises any constructs that have a component of TNF ⁇ (e.g., a naturally occurring TNF ⁇ , e.g., a recombinant TNF ⁇ ). These include and are not ny-2770598 Attorney Docket No.24516-20006.40 limited to natural TNF ⁇ (e.g., various isoforms of human TNF ⁇ ), synthetic or recombinant TNF ⁇ , and fusion proteins having a TNF ⁇ component.
- the TNFR activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety.
- the one or more of S/D/M factors comprise a TNFR activator, optionally wherein the TNFR activator is selected from the group consisting of TNF ⁇ , a TNFR agonist antibody, and a small molecule activator of TNFR.
- the TNFR activator is TNF ⁇ .
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ (e.g., a human TNF ⁇ ) is present in the medium at a concentration of at least about 0.2 ng/ml, optionally at least about 0.5 ng/ml (e.g., at least about 1 ng/ml, about 2 ng/ml, or about 3 ng/ml).
- the TNF ⁇ (e.g., a human TNF ⁇ ) is present in the medium at a concentration of about 0.2 ng/ml to about 30 ng/ml (e.g., about 0.5 ng/ml to about 10 ng/ml, e.g., about 1 ng/ml to about 5 ng/ml, e.g., about 2 ng/ml to about 4 ng/ml).
- Interferon gamma is a dimerized soluble cytokine that is the only member of the type II class of interferons.
- IFN ⁇ or type II interferon, is a cytokine that is critical for innate and adaptive immunity against viral, some bacterial, and protozoan infections.
- IFN ⁇ is an important activator of macrophages and inducer of major histocompatibility complex class II molecule expression.
- Aberrant IFN ⁇ expression is associated with a number of autoinflammatory and autoimmune diseases. The importance of IFN ⁇ in the immune system stems in part from its ability to inhibit viral replication directly, and most importantly from its immunostimulatory and immunomodulatory effects.
- IFN ⁇ is produced predominantly by natural killer cells (NK) and natural killer T cells (NKT) as part of the innate immune response, and by CD4 Th1 and CD8 cytotoxic T lymphocyte (CTL) effector T cells once antigen-specific immunity develops as part of the adaptive immune response.
- IFN ⁇ is also produced by non-cytotoxic innate lymphoid cells (ILC), a family of immune cells first discovered in the early 2010s.
- IFN ⁇ as referred to herein comprises any constructs that have a component of IFN ⁇ (e.g., a naturally occurring IFN ⁇ , e.g., a recombinant IFN ⁇ ).
- the IFNGR activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety.
- the one or more of S/D/M factors comprises an IFNGR activator, optionally wherein the IFNGR activator is selected from the group consisting of IFN ⁇ , an IFNGR agonist antibody, and a small molecule activator of IFNGR.
- the IFNGR activator is IFN ⁇ .
- the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ e.g., a human IFN ⁇
- the IFN ⁇ is present in the medium at a concentration of at least about 1 ng/ml, optionally at least about 5 ng/ml (e.g., at least about 10 ng/ml, about 20 ng/ml, or about 50 ng/ml).
- the IFN ⁇ (e.g., a human IFN ⁇ ) is present in the medium at a concentration of about 1 ng/ml to about 500 ng/ml (e.g., about 5 ng/ml to about 200 ng/ml, e.g., about 10 ng/ml to about 100 ng/ml, e.g., about 40 ng/ml to about 60 ng/ml, e.g., about 50 ng/ml).
- the one or more of S/D/M factors comprises two or more agents selected from the group consisting of an IL-4R activator, a TNFR activator, and an IFNGR activator as described herein.
- the one or more of S/D/M factors comprises IL-10, IL-4, TNF ⁇ , and IFN ⁇ .
- IL-4 receptor (IL-4R) activator and IL-4 [0203] “IL-4 receptor (IL-4R) activator” as described herein refers to a molecule that activates the IL-4 receptor-mediated signaling pathway.
- Interleukin 4 (IL-4) is a cytokine that induces differentiation of naive helper T cells (Th0 cells) to Th2 cells. Upon activation by IL-4, Th2 cells subsequently produce additional IL-4 in a positive feedback loop. IL-4 is produced primarily by mast cells, Th2 cells, eosinophils, and basophils.
- IL-4 is closely related to and has functions similar to IL-13.
- Interleukin 4 has many biological roles, including the stimulation of activated B cell and T cell proliferation, and the differentiation of B cells into plasma cells. It is a key regulator in humoral and adaptive immunity.
- IL-4 induces B cell class switching to IgE, and up-regulates MHC class II production.
- IL-4 decreases the production of IL-12 by Th1 cells, macrophages, IFN ⁇ , and dendritic cells.
- IL-4 signaling determines the levels of CD20 on the surface of ny-2770598 Attorney Docket No.24516-20006.40 normal and malignant B lymphocytes via activation of transcription factor STAT6. Overproduction of IL-4 is associated with allergies.
- IL-4 as referred to herein comprises any constructs that have a component of IL-4 (e.g., a naturally occurring IL-4, e.g., a recombinant IL-4). These include and are not limited to natural IL-4 (e.g., various isoforms of human IL-4), synthetic or recombinant IL-4, and fusion proteins having an IL-4 component.
- the IL-4R activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety.
- the one or more of S/D/M factors comprises an IL-4R activator, optionally wherein the IL-4R activator is selected from the group consisting of IL- 4, IL-13, an IL-4R agonist antibody, and a small molecule activator of IL-4R.
- the IL-4R activator is IL-4.
- the IL-4 is a human IL-4 or a human recombinant IL-4.
- the IL-4 (e.g., a human IL-4) is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml (e.g., at least about 30 pg/ml, 50 pg/ml, 75 pg/ml, 100 pg/ml, 125 pg/ml, or 150 pg/ml).
- the IL-4 (e.g., a human IL-4) is present in the medium at a concentration of about 15 pg/ml to about 1.5 ng/ml (e.g., about 30 pg/ml to about 1 ng/ml, e.g., about 100 pg/ml to about 1 ng/ml, e.g., about 100 pg/ml to about 1 ng/ml).
- the IL-4R activator is IL-13 (such as a human IL-13 or a human recombinant IL-13).
- the IL-13 is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 60 pg/ml, further optionally about 60 pg/ml to about 2 ng/ml (e.g., about 100 pg/ml to about 2 ng/ml).
- GM-CSF receptor (GM-CSFR) activator and GM-CSF [0209] “GM-CSF receptor (GM-CSFR) activator” as described herein refers to a molecule that activates the GM-CSFR-mediated signaling pathway.
- Granulocyte-macrophage colony-stimulating factor also known as colony-stimulating factor 2 (CSF2)
- CSF2 colony-stimulating factor 2
- GM-CSF stimulates stem cells to produce granulocytes (neutrophils, eosinophils, and basophils) and monocytes.
- Monocytes exit the circulation and migrate into tissue, whereupon they mature into macrophages and dendritic cells.
- GM-CSF As referred to herein comprises any constructs that have a component of GM-CSF (e.g., a naturally occurring GM-CSF, e.g., a recombinant GM-CSF).
- the GM-CSFR activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety.
- the one or more of the S/D/M factors further comprises a GM- CSF receptor (GM-CSFR) activator.
- the GM-CSFR activator is selected from the group consisting of GM-CSF, a GM-CSFR agonist antibody, and a small molecule activator of GM-CSFR.
- the GM-CSFR activator is GM- CSF.
- the GM-CSF is a human GM-CSF or a human recombinant GM-CSF.
- the GM-CSF (e.g., a human GM-CSF) is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml (e.g., at least about 100 pg/ml, about 150 pg/ml, about 200 pg/ml, or about 300 pg/ml).
- the GM-CSF (e.g., a human GM-CSF) is present in the medium at a concentration of about 30 pg/ml to about 3 ng/ml (e.g., about 50 pg/ml to about 1 ng/ml, e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 200 pg/ml to about 400 pg/ml, e.g., about 300 pg/ml).
- a concentration of about 30 pg/ml to about 3 ng/ml e.g., about 50 pg/ml to about 1 ng/ml, e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 200 pg/ml to about 400 pg/ml, e.g., about 300 pg/ml.
- IL-6 receptor (IL-6R) activator and IL-6
- IL-6 receptor (IL-6R) activator refers to a molecule that activates the IL-6 receptor-mediated signaling pathway.
- Interleukin 6 (IL-6) is an interleukin that acts as both a pro-inflammatory cytokine and an anti-inflammatory myokine. In the immune system, IL-6 is secreted by macrophages in response to specific microbial molecules, referred to as pathogen-associated molecular patterns (PAMPs). These PAMPs bind to an important group of detection molecules of the innate immune system, called pattern recognition receptors (PRRs), including Toll-like receptors (TLRs).
- PRRs pattern recognition receptors
- TLRs Toll-like receptors
- IL-6 as referred to herein comprises any constructs that have a component of IL-6 (e.g., a naturally occurring IL-6, e.g., a recombinant IL-6).
- the IL-6R activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety.
- the one or more of the S/D/M factors further comprise an IL-6 receptor (IL-6R) activator, optionally wherein the IL-6R activator is selected from the group consisting of IL-6, an IL-6R agonist antibody, and a small molecule activator of IL-6R.
- IL-6R IL-6 receptor
- the IL-6R activator is IL-6.
- the IL-6 is a human IL-6 or a human recombinant IL-6.
- the IL-6 (e.g., a human IL-6) is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml (e.g., at least about 10 pg/ml, about 15 pg/ml, about 20 pg/ml, or about 25 pg/ml).
- the IL-6 (e.g., a human IL-6) is present in the medium at a concentration of about 1 pg/ml to about 300 pg/ml (e.g., about 5 pg/ml to about 100 pg/ml, e.g., about 10 pg/ml to about 50 pg/ml, e.g., about 20 pg/ml to about 40 pg/ml, e.g., about 30 pg/ml).
- the one or more of S/D/M factors comprises IL-10, IL-4, TNF ⁇ , IFN ⁇ , GM-CSF, and IL-6.
- the one or more of S/D/M factors described herein is present in a single composition.
- the one or more of S/D/M factors described herein further comprises one or more cytokines selected from the group consisting of IL-2, IL-17, (e.g., IL- 17A), and/or M-CSF.
- at least one of the one or more of S/D/M factors is provided separately from other S/D/M factors in the one or more of S/D/M factors.
- the refinement factors are not required but optional to the methods discussed herein of promoting survival and/or differentiation of the monocytes ny-2770598 Attorney Docket No.24516-20006.40 (e.g., cancer monocytes) or the hematological cancer cells into potent antigen presenting cells (APCs) but can further refine the APCs to express even higher levels of surface markers (e.g., MHC-I, MHC-II, CD80, CD86, CD40) that indicate a more potent antigen presentation machinery.
- surface markers e.g., MHC-I, MHC-II, CD80, CD86, CD40
- the methods described above further comprise contacting the population of hematological cancer cells (e.g., AML cells, CLL cells, CML cells, NHL cells, or B-ALL cells) with one or more of refinement factors after the one or more of cancer cells are contacted with the one or more of S/D/M factors or the medium derived from the culture of T cells.
- the refinement factors are selected from the group consisting of type-I interferon (such as IFN ⁇ and/or IFN ⁇ ), IFN ⁇ , TNF ⁇ , a TLR ligand (such as R848, poly IC, CpG, or LPS), CD40L or a CD40-ligating antibody, an anti-PD-L1 antibody, and TPI-1.
- a method of refining a population of APCs derived from monocytes comprising contacting the population of APCs with a) IFN ⁇ , b) IFN ⁇ , and c) TNF ⁇ .
- a method of refining a population of APCs derived from monocytes comprising contacting the population of APCs with a) IFN ⁇ , b) IFN ⁇ , c) TNF ⁇ , d) poly IC, and e) CpG.
- a method of refining a population of APCs derived from monocytes comprising contacting the population of APCs with a) IFN ⁇ , b) IFN ⁇ , c) TNF ⁇ , d) poly IC, and e) R848.
- a method of refining a population of APCs derived from monocytes comprising contacting the population of APCs with a) IFN ⁇ , b) IFN ⁇ , c) TNF ⁇ , d) poly IC, e) CD40L, and f) anti-PD-L1 antibody.
- monocytes e.g., cancer monocytes
- hematological cancer cells comprising contacting the population of APCs with a) IFN ⁇ , b) IFN ⁇ , c) TNF ⁇ , d) poly IC, e) CD40L, and f) anti-PD-L1 antibody.
- a method of refining a population of APCs derived from monocytes comprising contacting the population of APCs with a) IFN ⁇ , b) IFN ⁇ , c) TNF ⁇ , d) poly IC, e) CD40L, f) anti-PD-L1 antibody, and g) a SHP-1 inhibitor (e.g., TPI-1).
- monocytes e.g., cancer monocytes
- hematological cancer cells comprising contacting the population of APCs with a) IFN ⁇ , b) IFN ⁇ , c) TNF ⁇ , d) poly IC, e) CD40L, f) anti-PD-L1 antibody, and g) a SHP-1 inhibitor (e.g., TPI-1).
- a method of refining a population of APCs derived from monocytes comprising contacting the population of APCs with a) IFN ⁇ , b) IFN ⁇ , c) R848, d) poly IC, and e) a SHP- 1 inhibitor (e.g., TPI-1).
- monocytes e.g., cancer monocytes
- hematological cancer cells comprising contacting the population of APCs with a) IFN ⁇ , b) IFN ⁇ , c) R848, d) poly IC, and e) a SHP- 1 inhibitor (e.g., TPI-1).
- ny-2770598 Attorney Docket No.24516-20006.40
- a method of refining a population of APCs derived from monocytes comprising contacting the population of APCs with a) IFN ⁇ , b) R848, c) poly IC, and d) a SHP-1 inhibitor (e.g., TPI-1).
- a method of refining a population of APCs derived from monocytes comprising contacting the population of APCs with a) IFN ⁇ , b) IFN ⁇ , c) poly IC, d) CpG, e) CD40L, f) anti-PD-L1 antibody, g) a SHP-1 inhibitor (e.g., TPI-1), and h) TNF ⁇ .
- monocytes e.g., cancer monocytes
- hematological cancer cells comprising contacting the population of APCs with a) IFN ⁇ , b) IFN ⁇ , c) poly IC, d) CpG, e) CD40L, f) anti-PD-L1 antibody, g) a SHP-1 inhibitor (e.g., TPI-1), and h) TNF ⁇ .
- a SHP-1 inhibitor e.g., TPI-1
- a method of refining a population of APCs derived from monocytes comprising contacting the population of APCs with a) R848, and b) poly IC.
- the one or more of refinement factors is provided immediately after the plurality of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B- ALL cells) is contacted with the one or more of S/D/M factors or the medium derived from the culture of T cells.
- the one or more of refinement factors is provided within about one day after the plurality of cancer cells is contacted with the one or more of S/D/M factors or the medium derived from the culture of T cells.
- the plurality of the cancer cells e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells
- the plurality of the cancer cells is cultured for about 1-5 days (e.g., for about one, two, three, four or five days) in the presence of the one or more of refinement factors.
- the one or more of refinement factors is provided when at least about 50% (e.g., about 50%, 60%, 70%, 80%, or 99%) of the cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) survive after the plurality of cancer cells is contacted with the one or more of S/D/M factors or the medium derived from the culture of T cells.
- the one or more of refinement factors is provided when at least about 10%, 20%, 30%, 40% or 50% of the cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) exhibit a dendritic cell morphology.
- the one or more of refinement factors is provided when cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) express a high level of one or more molecules selected from the group consisting of MHC I, MHC II, CD80, CD86, and/or CD40.
- the one or more of refinement factors is provided ny-2770598 Attorney Docket No.24516-20006.40 when cancer cells express a higher (e.g., at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%) level of one or more molecules selected from the group consisting of MHC-I, MHC-II, CD80, CD86, and/or CD40 than corresponding cells obtained from the same individual and cultured with GM-CSF and M-CSF (e.g., at a concentration routinely used in the field for such methods).
- a higher e.g., at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%
- the one or more of refinement factors described herein can be used independently from the methods described above for priming APCs (e.g., APCs obtained from a human).
- the one or more of refinement factors comprises Poly IC or R848.
- the one or more of refinement factors comprise Poly IC and R848.
- the one or more of refinement factors comprise IFN ⁇ , IFN ⁇ , and TNF ⁇ .
- the one or more of refinement factors comprise IFN ⁇ , IFN ⁇ , TNF ⁇ , poly IC, and CpG.
- the one or more of refinement factors are comprised in a medium comprising serum.
- the one or more of refinement factors comprise IFN ⁇ , IFN ⁇ , TNF ⁇ , poly IC, CpG, CD40L, and an anti-PD-L1 antibody.
- the one or more of refinement factors comprise IFN ⁇ , IFN ⁇ , TNF ⁇ , poly IC, CpG, CD40L, an anti-PD-L1 antibody, and TPI-1.
- the r one or more of efinement factors are comprised in a medium comprising serum (e.g., human serum).
- the concentration of IFN ⁇ in the one or more of refinement factors is about 1ng/ml to about 50 ng/ml (e.g., about 5 ng/ml to about 20 ng/ml, e.g., about 10 ng/ml).
- the concentration of IFN ⁇ in the one or more of refinement factors is about 5 ng/ml to about 500 ng/ml (e.g., about 10 ng/ml to about 250 ng/ml, e.g., about 20 ng/ml to about 100 ng/ml, e.g., about 50 ng/ml).
- the concentration of TNF ⁇ in the one or more of refinement factors is about 1ng/ml to about 50 ng/ml (e.g., about 5 ng/ml to about 20 ng/ml, e.g., about 10 ng/ml).
- the concentration of poly IC in the one or more of refinement factors is about 0.1 ⁇ g/ml to about 10 ⁇ g/ml (e.g., about 0.2 ⁇ g/ml to about 5 ⁇ g/ml, e.g., about 0.5 ⁇ g/ml to about 2.5 ⁇ g/ml, e.g., about 1 ⁇ g/ml).
- the concentration of CpG in the one or more of refinement factors is about 0.1 ⁇ g/ml to about 10 ⁇ g/ml (e.g., about 0.2 ⁇ g/ml to about 5 ⁇ g/ml, e.g., about 0.5 ⁇ g/ml to about 2.5 ⁇ g/ml, e.g., about 1 ⁇ g/ml).
- the concentration of CD40L in the one or more of refinement factors is about 1 ⁇ g/ml to about 100 ⁇ g/ml (e.g., about 2 ⁇ g/ml to about 50 ⁇ g/ml, e.g., about 5 ⁇ g/ml to about 20 ⁇ g/ml, e.g., about 10 ⁇ g/ml).
- the concentration of the anti-PD-L1 antibody in the one or more of refinement factors is about 1 ⁇ g/ml to about 200 ⁇ g/ml (e.g., about 5 ⁇ g/ml to about 100 ⁇ g/ml, e.g., about 10 ⁇ g/ml to about 50 ⁇ g/ml, e.g., about 20 ⁇ g/ml).
- the concentration of TPI-1 in the one or more of refinement factors is about 0.1 ⁇ g/ml to about 10 ⁇ g/ml (e.g., about 0.2 ⁇ g/ml to about 5 ⁇ g/ml, e.g., about 0.5 ⁇ g/ml to about 2.5 ⁇ g/ml, e.g., about 1 ⁇ g/ml).
- the concentration of R848 in the one or more of refinement factors is about 0.1 ⁇ g/ml to about 10 ⁇ g/ml (e.g., about 0.2 ⁇ g/ml to about 5 ⁇ g/ml, e.g., about 0.5 ⁇ g/ml to about 2.5 ⁇ g/ml, e.g., about 1 ⁇ g/ml).
- Methods for promoting survival of hematological cancer cells e.g., hematological cancer cells from an individual
- HC-APCs e.g., HC- APCs.
- the cancer cells are obtained (e.g., freshly isolated) from an individual (e.g., a human). It was observed that the hematological cancer cells obtained from an individual (e.g., PBMC of an individual) generally could not survive more than 1-2 days. The methods described herein promote the survival of these cells while they differentiate into APCs and/or promote the survival of these HC-APCs.
- the cancer cells obtained from the individual express a lower level of IL-10 receptor (“IL-10R”), IL-4 receptor (“IL-4R”), IL-6 receptor (“IL-6R”), M-CSF receptor (“GM-CSFR”), and/or M-CSF receptor (“GM-CSFR”) as compared to those corresponding cells obtained from a reference individual (e.g., a healthy individual).
- IL-10R IL-10 receptor
- IL-4R IL-4 receptor
- IL-6 receptor IL-6 receptor
- GM-CSFR M-CSF receptor
- GM-CSFR M-CSF receptor
- the present application provides a method of promoting the survival of a population of hematological cancer cells (e.g., hematological cancer cells from an individual) and/or HC-APCs from an individual in an in vitro culture, comprising cultivating the population of cancer cells in a medium having an IL-10R activator, optionally wherein the IL-10R activator is selected from the group consisting of: an IL-10 (e.g., a pegylated IL-10, e.g., pegilodecakin or AM0010), an IL-10 family member (e.g., IL-19, IL- 20, IL-22, IL-24, IL-26, IL-28), an IL-10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, a small molecule activator of IL-10R, and an activator of the IL- 10R-downstream
- an IL-10 e.g., a pegylated IL-10, e.g
- the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml).
- the culture further comprises a TNF ⁇ receptor (TNFR) activator, and/or an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, optionally wherein the TNFR activator is selected from the group consisting of TNF ⁇ , a TNFR agonist antibody, and a small molecule activator of TNFR, optionally wherein the IFNGR activator is selected from the group consisting of IFN ⁇ , an IFNGR agonist antibody, and a small molecule activator of IFNGR, and further optionally wherein the culture comprises TNF ⁇ and/or IFN ⁇ .
- TNFR TNF ⁇ receptor
- IFN ⁇ interferon ⁇ receptor
- IFNGR interferon ⁇ receptor
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the hematological cancer is a myeloid leukemia, a B-cell lymphoma or B-cell leukemia.
- the hematological cancer cells comprise AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells.
- the present application provides a method of promoting the survival of a population of cancer cell-derived APCs from an individual in an in vitro culture, comprising cultivating the population of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) in a medium having a TNF ⁇ receptor (TNFR) activator, and/or an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, optionally wherein the TNFR activator is selected from the group consisting of TNF ⁇ , a TNFR agonist antibody, and a small molecule activator of TNFR, optionally wherein the IFNGR activator is selected from the group ny-2770598 Attorney Docket No.24516-20006.40 consisting of IFN ⁇ , an IFNGR agonist antibody, and a small molecule activator
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the hematological cancer is a myeloid leukemia, a B-cell lymphoma or B-cell leukemia.
- the hematological cancer cells comprise AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells.
- the present application provides a method of promoting the survival of a population of cancer cell-derived APCs from an individual in an in vitro culture, comprising cultivating the population of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) in a medium having IL-10, TNF ⁇ , and IFN ⁇ .
- the IL-10 is a human IL-10 or a human recombinant IL-10.
- the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml).
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the hematological cancer is a myeloid leukemia, a B-cell lymphoma or B-cell leukemia.
- the hematological cancer cells comprise AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells.
- the culture further comprises a GM-CSF receptor (GM-CSFR) activator.
- GM-CSFR GM-CSF receptor
- the GM-CSFR activator is selected from the group consisting of GM-CSF, a GM-CSFR agonist antibody, and a small molecule activator of GM- CSFR. In some embodiments, the GM-CSFR activator is GM-CSF. In some embodiments, the GM-CSF is a human GM-CSF or a human recombinant GM-CSF.
- the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml).
- ny-2770598 Attorney Docket No.24516-20006.40
- the culture further comprises an IL-6 receptor (IL-6R) activator.
- IL-6R IL-6 receptor
- the IL-6R activator is selected from the group consisting of IL-6, an IL-6R agonist antibody, and a small molecule activator of IL-6R.
- the IL-6R activator is IL-6.
- the IL-6 is a human IL-6 or a human recombinant IL-6.
- the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml).
- the present application provides a method of promoting the survival of a population of cancer cell-derived APCs from an individual in an in vitro culture, comprising cultivating the population of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) in a medium derived from a culture of T cells after being treated with anti-CD3 and anti-CD28 antibodies, wherein the medium comprises an activator of IL- 10R.
- the T cells are CD4 T cells.
- the T cells are CD8 T cells.
- the T cells are isolated from PBMC of the same individual or a different individual and have not been previously treated with anti-CD3 and/or anti-CD28 antibodies prior to the treatment. In some embodiments, the T cells are isolated from PBMC of the same individual or a different individual and have been previously treated with anti-CD3 and/or anti-CD28 antibodies prior to the treatment. In some embodiments, the medium is derived from the culture after the T cells are treated with anti-CD3 and anti-CD28 antibodies for about 1-3 days, optionally for about 2 days. In some embodiments, the individual has a cancer (e.g., a hematological cancer). [0263]
- the anti-CD3/CD28 treatment for T cells described herein are techniques well known in the field for activating T cells.
- the present application further provides a method of increasing expression of IL-10 receptor (IL-10R) in a population of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) from an individual having cancer, comprising contacting the population of cancer cells with one or more agents selected from the group consisting of: an IL-10R activator, a TNFR activator, and an IFNGR activator.
- IL-10R IL-10 receptor
- the IL- 10R activator is selected from the group consisting of: an IL-10 (e.g., a pegylated IL-10, e.g., pegilodecakin or AM0010), an IL-10 family member (e.g., IL-19, IL-20, IL-22, IL-24, IL-26, IL-28), an IL-10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, a small molecule activator of IL-10R, and an activator of the IL-10R-downstream STAT3 (e.g., IL-12, e.g., Long noncoding RNA (LncRNA) PVT1, NEAT1, FEZF1-AS1, UICC).
- an IL-10 e.g., a pegylated IL-10, e.g., pegilodecakin or AM00
- an IL-10 family member e.g., IL-19, IL-20, IL-22,
- the IL-10R activator is IL-10.
- the IL-10 is a human IL- 10 or a human recombinant IL-10.
- the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml).
- the TNFR activator is selected from the group consisting of TNF ⁇ , a TNFR agonist antibody, and a small molecule activator of TNFR.
- the IFNGR activator is selected from the group consisting of IFN ⁇ , an IFNGR agonist antibody, and a small molecule activator of IFNGR.
- the IFNGR activator comprises TNF ⁇ .
- the IFNGR activator comprises IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the individual has a cancer (e.g., a hematological cancer).
- the cancer cells e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells
- the population of cancer cells express a lower level (at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45% or 50% lower) of IL-10R prior to contacting with the IL-10R activator as compared to corresponding cells obtained from a reference individual (e.g., a healthy individual).
- a reference individual e.g., a healthy individual.
- the present application provides a method of promoting the differentiation of a population of hematological cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) from an individual (e.g., a cancer patient) to antigen presenting cells (“APCs”) in an in vitro culture, comprising cultivating the population of hematological cancer cells in a medium having one or more molecules selected from the group consisting of an IL-4 receptor (IL-4R) activator (e.g., IL-4), a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ), optionally wherein the cancer cells have been contacted with a medium that further comprises an IL-10 receptor (IL-10R) activator (e.g., IL-10R) activator (e.g., IL-10R) activator (e.g., IL-10R) activator
- a method of promoting the differentiation of a population of hematological cancer cells from ny-2770598 Attorney Docket No.24516-20006.40 an individual (e.g., a cancer patient) to antigen presenting cells (“APCs”) in an in vitro culture comprising cultivating the population of cancer cells in a medium having an IL-4 receptor (IL-4R) activator (e.g., IL-4), a TNF ⁇ receptor (TNFR) activator (e.g., TNF ⁇ ), and an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., IFN ⁇ ).
- IL-4R IL-4 receptor
- TNF ⁇ receptor TNF ⁇ receptor
- IFN ⁇ interferon ⁇ receptor
- the IL- 4R activator is selected from the group consisting of IL-4, IL-13, an IL-4R agonist antibody, and a small molecule activator of IL-4R.
- the IL-4R activator is IL-4.
- the IL-4 is a human IL-4 or a human recombinant IL-4.
- the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml).
- the TNFR activator is selected from the group consisting of TNF ⁇ , a TNFR agonist antibody, and a small molecule activator of TNFR.
- the TNFR activator is TNF ⁇ .
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the IFNGR activator is selected from the group consisting of IFN ⁇ , an IFNGR agonist antibody, and a small molecule activator of IFNGR.
- the IFNGR activator is IFN ⁇ .
- the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- the culture further comprises an IL-6 receptor (IL-6R) activator.
- IL-6R IL-6 receptor
- the IL- 6R activator is selected from the group consisting of IL-6, an IL-6R agonist antibody, and a small molecule activator of IL-6R.
- the IL-6R activator is IL-6.
- the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml).
- a method of promoting the differentiation of a population of cancer cells comprising cultivating the population of cancer cells in a medium having IL-4, TNF ⁇ , and IFN ⁇ .
- a population of cancer cells e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells
- APCs antigen presenting cells
- the IL-4 is a human IL-4 or a human recombinant IL-4.
- the IL-4 is present in the medium at a concentration of at least ny-2770598 Attorney Docket No.24516-20006.40 about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml).
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ .
- the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml).
- a method of promoting the differentiation of a population of cancer cells comprising cultivating the population of cancer cells in a medium having IL-4, IL-6, TNF ⁇ , and IFN ⁇ .
- the IL-4 is a human IL-4 or a human recombinant IL-4.
- the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml).
- the TNF ⁇ is a human TNF ⁇ or a human recombinant TNF ⁇ .
- the TNF ⁇ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml).
- the IFN ⁇ is a human IFN ⁇ or a human recombinant IFN ⁇ . In some embodiments, the IFN ⁇ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6.
- the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml).
- the culture further comprises a GM-CSF receptor (GM-CSFR) activator.
- the GM-CSFR activator is selected from the group consisting of GM-CSF, a GM-CSFR agonist antibody, and a small molecule activator of GM- CSFR.
- the GM-CSFR activator is GM-CSF.
- the GM-CSF is a human GM-CSF or a human recombinant GM-CSF.
- the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, ny-2770598 Attorney Docket No.24516-20006.40 optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml).
- the culture further comprises a STAT3 activator, such as an IL-10 receptor (IL-10R) activator.
- the IL-10R activator is selected from the group consisting of: an IL-10 (e.g., a pegylated IL-10, e.g., pegilodecakin or AM0010), an IL-10 family member (e.g., IL-19, IL-20, IL-22, IL-24, IL-26, IL-28), an IL- 10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, a small molecule activator of IL-10R, and an activator of the IL-10R-downstream STAT3 (e.g., IL-12, e.g.
- an IL-10R-downstream STAT3 e.g., IL-12, e.g.
- the IL-10R activator is IL-10.
- the IL-10 is a human IL- 10 or a human recombinant IL-10.
- the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml).
- the hematological cancer cells obtained from the individual express a lower (e.g., at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45% or 50% lower) level of IL-10R or IL-4 receptor (“IL-4R”) as compared to those corresponding cells obtained from a reference individual (e.g., a healthy individual).
- the hematological cancer cells e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells
- the methods described herein further comprise loading one or more additional antigens (e.g., tumor-associated antigen peptides, neoantigen peptides) on the HC-APCs.
- the antigen comprises shared tumor-associated antigens.
- the shared tumor-associated antigens comprise self-antigens (e.g., abnormally expressed self-antigens).
- the shared tumor-associated antigens comprise non-self-antigens of viral origins (e.g., antigens from LMP1/2 associated with nasopharyngeal carcinoma and lymphoma, e.g., antigens from E6 and E7 proteins of high-risk human papillomavirus (HPV), e.g., antigens from retrovirus Tax protein found in adult T cell leukemia).
- the shared tumor-associated antigens comprise ny-2770598 Attorney Docket No.24516-20006.40 mutation-caused neoantigens shared in different types of cancer (e.g., neoantigens associated with p53 mutations or KRAS mutations).
- Tumor-associated antigen peptides [0276] Various approaches are available to identify tumor-associated antigen peptides. [0277] One approach often employed to identify the peptides recognized by such CTL is expression cloning, which consists of isolating the peptide-encoding gene by transfecting a library of tumoral cDNA and testing the transfected cells for their ability to activate the CTL clone. Fragments of the identified gene can then be transfected to define the region encoding the antigenic peptide, and finally candidate peptides bearing adequate HLA-binding motifs are tested for their ability to sensitize target cells to lysis by the CTL. This approach was successfully used to identify a large number of antigenic peptides.
- tumor-associated antigenic peptides are often identified using the “reverse immunology” approach, which consists in selecting peptides with adequate HLA-binding motifs inside a protein of interest, such as proteins encoded by mutated oncogenes or genes that are either selectively expressed or overexpressed by tumors.
- Candidate peptides are synthesized and tested for HLA binding in vitro.
- the most efficient binders are pulsed onto antigen-presenting cells, which are used to stimulate T lymphocytes in vitro, in order to derive CTL lines or clones that recognize peptide-pulsed target cells.
- a drawback of this approach is that the identified peptides might not be processed efficiently by tumors.
- a third approach to antigen identification is based on the elution of antigenic peptides from MHC class I molecules immunopurified from the surface of tumor cells.
- Neoantigen peptides [0281] Various methods are available to detect and screen neoantigens. Sandwich immunoassays in the miniaturized system could successfully identify tumor antigens in serum samples extracted from patients. See e.g., Pollard et al., Proteomics Clin. Appl.1934– 952 (2007); Yang et al., Biosens. Bioelectron.40385–392 (2013).
- Another tool named Serologic Proteome analysis (SERPA) or 2-D western blots consists of the isoelectric focusing (IEF) gel run in the first dimension and SDS-PAGE gel run in the second dimension.
- SERPA separates the proteins in the gel by their isoelectric point (IP) and molecular mass and then transfers the proteins from the gel to a carrier membrane to screen antibodies. Finally, the antigenic protein spots can be identified by MS. See e.g., Tjalsma et al., Proteomics Clin. Appl.2167–180 (2008). This approach has been used to identify antigens in different tumor types. Serological analysis of recombinant cDNA expression libraries (SEREX), which combines serological analysis with antigen cloning techniques, is a widely used technique to explore tumors’ antigen repertoire.
- SEEEX Serological analysis of recombinant cDNA expression libraries
- SEREX first construct a cDNA library from cancer cell lines or fresh tumor samples, then screen the cDNA library with autologous sera of cancer patients, and finally sequence the immune-reactive clones.
- SEREX have identified a variety of tumor antigens including CTAs, differentiation antigens, mutational antigens, splice-variant antigens, and overexpressed antigens. See e.g., Chen et al., Proc. Natl. Acad. Sci. U.S.A.941914–1918 (1997).
- MAPPing Multiple Affinity Protein Profiling
- nanoplasmonic biosensors have also been developed to identify tumor antigens. See e.g., Lee et al., Biosens.
- the one or more neoantigenic peptides described herein are obtained from a neoantigenic database (such as any of the neoantigenic databases described herein).
- a neoantigenic database such as any of the neoantigenic databases described herein.
- Tan et al. constructed a manually curated database (“dbPepNeo”) for human tumor neoantigen peptides based upon the four criteria as described below: (i) peptides were isolated from human tumor tissues or cell lines, (ii) peptides contained non- synonymous mutations in amino acid sequence, (iii) peptides can be bound by HLA-I molecules, and (iv) peptides can induce CD8+ T cell responses.
- dbPepNeo manually curated database
- the one or more neoantigenic peptides are obtained from analyzing the biological information of the individual (such as a patient who had a cancer).
- the one or more neoantigenic peptides are obtained from a computational analysis of a cancer patient’s tumor genome. See e.g., Roudko et al. Front Immunol.2020; 11: 27. In some embodiments, the one or more neoantigenic peptides are obtained from a computational analysis of a cancer patient’s transcriptome. See e.g., Caushi et al., Nature.2021 Aug;596(7870):126-132. In some embodiments, the one or more neoantigenic peptides are obtained from a computational analysis of a cancer patient’s proteome. See e.g., Wen et al.
- the one or more neoantigenic peptides are selected based upon patient data.
- the patient data are derived from data from a group of patients having a particular type of cancer (e.g., any of the cancers described here).
- the patient data are derived from data from a group of patients having any cancer.
- the group of patients are from the same sex (e.g., male or female).
- the group of patients are from the same ethnicity.
- the group of patients bear one or more biomarkers (e.g., an aberration in a particular gene, e.g., KRAS, e.g., PTEN).
- the one or more neoantigenic peptides are derived from any polypeptide known to or have been found to contain a tumor-specific mutation. Suitable polypeptides from which the one or more neoantigenic peptides can be derived can be found for example in various databases available in the field (e.g., COSMIC database). These databases curate comprehensive information on somatic mutations in human cancer.
- the one or more neoantigenic peptides contains a tumor-specific mutation.
- the tumor-specific mutation is a driver mutation for a particular cancer type.
- the tumor-associated peptides e.g., neoantigen peptides
- the one or more neoantigenic peptides are obtained by exome high throughput sequencing and prescreened with epitope prediction algorithms.
- the one or more neoantigenic peptides are selected based upon its binding affinity to an MHC molecule (e.g., an MHC-I molecule and/or an MHC-II molecule).
- the one or more neoantigenic peptides has a binding affinity that is less than 5000 nM (e.g., less than 500 nM, less than 250 nM, less than 100 nM or less than 50 nM) (IC50) to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity of about 500 nM to 5000 nM (IC50) to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity that is less than 500 nM (IC50) to an MHC molecule.
- the one or more neoantigenic peptides has a binding affinity of about 250 nM to 500 nM IC 50 to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity that is less than 250 nM (IC 50 ) to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity that is less than 100 nM (IC 50 ) to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity of about 50 nM to 500 nM IC50 to an MHC molecule.
- the one or more neoantigenic peptides has a binding affinity that is less than 50 nM (IC50) to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity of about 1 nM to 50 nM IC50 to an MHC molecule.
- a plurality of tumor-associated peptides are prepared from a surgical resection of tumor tissue or a biopsy extract thereof.
- a plurality of tumor-associated peptides are prepared from a mixture of tumor cells or extract thereof isolated from tumor tissue or a biopsy extract thereof.
- a plurality of tumor-associated peptides are prepared from a mixture of isolated tumor-associated peptides (e.g., neoantigen peptides).
- the tumor tissue or cell described above is a fresh tumor tissue or cell. In some embodiments, the tumor tissue or cell is obtained from a frozen sample.
- the tumor tissue or cell has been subjected to an induction of immunogenic cell death (e.g., freeze-thaw to lyse tumor cells, high dose UV irradiation, X- ray radiation).
- immunogenic cell death e.g., freeze-thaw to lyse tumor cells, high dose UV irradiation, X- ray radiation.
- the tumor tissue or cell has been subjected to a radiation treatment.
- the present application provides APCs produced by any of the methods described here.
- Hematological Cancer described in this section can be a hematological cancer type or kind and derived from cells of the myeloid or the lymphoid lineage. All the cancer types discussed in Section V are similarly applicable here.
- Hematological cancer cells include cancerous cells of the immune system or in blood- forming tissue, such as the bone marrow. Non-limiting examples include lymphoma, myeloma, and leukemia. Lymphomas derive from the lymph system (e.g., lymphocytes) and include Hodgkin’s and non-Hodgkin’s lymphomas (NHL).
- Myelomas derive from plasma cells, which terminally differentiate from activated B cells.
- Leukemias derive from blood cells and bone marrow, such as from lymphocytes (e.g., B cell acute lymphocytic leukemia (B-ALL)) or myeloid cells (e.g., acute myeloid leukemia (AML) or chronic myeloid leukemia (CML)) and can be chronic or acute.
- lymphocytes e.g., B cell acute lymphocytic leukemia (B-ALL)
- myeloid cells e.g., acute myeloid leukemia (AML) or chronic myeloid leukemia (CML)
- the hematological cancer is a lymphoma.
- the hematological cancer is a non-Hodgkin’s lymphoma.
- the hematological cancer is a Burkitt’s lymphoma, an anaplastic large cell lymphoma, or a splenic marginal zone lymphoma. [0298] In some embodiments, the hematological cancer is a leukemia.
- the hematological cancer is an acute lymphocytic leukemia (ALL), a chronic lymphocytic leukemia (CLL), an acute myelogenous leukemia (AML), an acute megakaryoblastic leukemia (AMKL), a chronic idiopathic myelofibrosis (MF), a chronic myelogenous leukemia (CML), a B-cell prolymphocytic leukemia (B-PLL), a chronic neutrophilic leukemia, a hairy cell leukemia (HCL), or an aggressive NK-cell leukemia.
- ALL acute lymphocytic leukemia
- CLL chronic lymphocytic leukemia
- AML acute myelogenous leukemia
- AKL acute megakaryoblastic leukemia
- MF chronic idiopathic myelofibrosis
- CML chronic myelogenous leukemia
- B-PLL B-cell prolymphocytic leukemia
- the hematological cancer is an advanced cancer. In some embodiments, the hematological cancer is a late-stage cancer. In some embodiments, the hematological cancer is stage II, III, or IV. In some embodiments, the cancer is malignant.
- hematological cancers described herein include, but are not limited to, acute lymphoblastic leukemia, chronic myelogenous leukemia, Hodgkin’s lymphoma, non- Hodgkin’s lymphoma, leukemia, B-cell chronic lymphocytic leukemia, acute myeloid leukemia, AIDS-related cancers (e.g., AIDS-related lymphoma), central nervous system lymphoma, chronic myeloproliferative disorders, leukemia, lymphoid neoplasm (e.g., lymphoma), myelodysplastic syndromes, myelodysplastic/myeloproliferative diseases, lymphoma, primary central nervous system lymphoma (microglioma), and post-transplant lymphoproliferative disorder (PTLD).
- acute lymphoblastic leukemia chronic myelogenous leukemia
- Hodgkin’s lymphoma non- Hodgkin’s lymphoma
- the hematological cancer is a virus-infection-related cancer.
- the hematological cancer is human T-lymphotrophic virus (HTLV-1)- related cancer (e.g., adult T cell leukemia or lymphoma).
- the hematological cancer is Epstein-Barr virus (EBV)-related cancer (e.g., Burkitt lymphoma, Hodgkin’s lymphoma, or non-Hodgkin’s lymphoma).
- EBV Epstein-Barr virus
- the hematological cancer is hepatitis C virus-related cancer (e.g., non-Hodgkin’s lymphoma).
- the hematological cancer is refractory to one or more of irradiation therapy, chemotherapy, or immunotherapy (e.g., checkpoint blockade).
- the hematological cancer is B-ALL, CLL, NHL, or AML.
- HC-APCs are derived from AML cells of any of M0, M1, M2, M3, M4, or M5 subtypes.
- B cell acute lymphoblastic leukemia cells B-ALL
- the methods described herein convert a plurality of B cell acute lymphoblastic leukemia (B-ALL) cells into HC-APCs.
- the plurality of B-ALL cells is obtained from the peripheral blood of the individual (e.g., human cancer patient).
- Various markers of B-ALL cells have been identified and described, including at different stages of development. For example, early pre B-ALL has been found to present with the phenotype of TdT + CD19 + CD10-. Common B-ALL displays CD19 + CD10 + CALLA + .
- Pre B-ALL cells are CD10 +/- CD19 + HLA-DR + cytoplasmic IgM + .
- Mature B-ALL cells express CD10 + CD19 + CD20 + CD22 + surface IgM + . Additional markers of the B-ALL cell lineages are known in the art.
- the B-ALL cells express CD19 and one or more of the markers selected from the group consisting of CD10, CD21, CD22, CD23, CD24, CD79a, CALLA, TdT, HLA-DR, and cytoplasmic IgM at the time when they are obtained from the peripheral blood.
- Methods of obtaining B-ALL cells from peripheral blood is well known in the art, for example by seeding peripheral blood cells onto an anti-CD19 antibody-coated plate.
- B-ALL cells can be separated by positive selection with (e.g.) anti-CD19 antibody and one or more antibodies targeting markers selected from the group consisting of CD10, CD21, CD22, CD23, CD24, CD79a, CALLA, TdT, HLA-DR, and cytoplasmic IgM, or by negative selection using all antibodies against other cells using magnetic-activated cell sorting (MACS).
- Further methods of B-ALL cell selection include surface marker staining and cell sorting by fluorescence activated cell sorting (FACS).
- the B-ALL cells obtained from the individual express a lower level of IL-10 receptor (“IL-10R”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a reference individual (e.g., a healthy individual).
- IL-10R IL-10 receptor
- the level of IL-10R on the B-ALL cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-10R on corresponding cells from the reference individual (e.g., a healthy individual).
- the B-ALL cells obtained from the individual express a lower level of IL-4 receptor (“IL-4R”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual.
- IL-4R IL-4 receptor
- the level of IL-4R on the B-ALL cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-4R on corresponding cells from the reference individual (e.g., a healthy individual).
- the B-ALL cells obtained from the individual express a lower level of IL-6 receptor (“IL-6R”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual.
- IL-6R IL-6 receptor
- the level of IL-6R on the B-ALL cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-6R on corresponding cells from the reference individual (e.g., a healthy individual).
- the B-ALL cells obtained from the individual express a lower level of M-CSF receptor (“M-CSFR”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual.
- M-CSFR M-CSF receptor
- the level of M-CSFR on the B-ALL cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, or 65% lower than the level of M-CSFR on corresponding cells from the reference individual (e.g., a healthy individual).
- the B-ALL cells obtained from the individual express a lower level of GM-CSF receptor (“GM-CSFR”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual.
- GM-CSFR GM-CSF receptor
- the level of GM-CSFR on the B-ALL cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, or 65% lower than the level of GM-CSFR on corresponding cells from the reference individual (e.g., a healthy individual).
- Acute myeloid leukemia cells [0312] The methods described herein convert a plurality of acute myeloid leukemia (AML) cells into HC-APCs.
- the plurality of AML cells is obtained from the peripheral blood of the individual (e.g., human cancer patient).
- LSCs leukemia stem cells
- the eight subtypes of AML are described in the Hematological Cancer section below. For example, early studies of human AML cases found that up to 96% of human AML cases expressed CD13 and/or CD33. Similarly, CD14 was identified in 82- 90% of M4 and M5 AML cases.
- Leukemia stem cells that differentiate into AML cells have been found to present with the surface marker profile of CD34 + CD38-CD123 + TIM3 + and can include additional markers.
- CD33, CD123, CLL1, TIM3 and CD244 were ubiquitously expressed on AML bulk cells at initial diagnosis and relapse, and CD33/TIM3 and CLL1/TIM3 have been shown to be highly positive in AML compared with normal hematopoiesis and non-hematopoietic tissues (Haubner et al., Leukemia 2019, 33:64-74). Further markers of the LSC and/or AML cell lineage are known in the art. [0314] Specifically, the eight subtypes, i.e., M0-M7 subtypes, are described as below.
- M0 subtype is also called acute dedifferentiated leukemia, acute minimally differentiated leukemia, acute myeloid leukemia with minimal differentiation, or acute myelogenous leukemia with minimal differentiation.
- AML-M0 is a rare subtype in which blasts fail to show morphologic differentiation by light microscopy, and conventional cytochemical stains and myeloid markers are negative. These blasts are CD45 dim and express the primitive hematopoietic markers CD34, CD38 and HLA Dr. One or more of the following pan-myeloid markers are expressed: CD13, CD33, and/or CD117.
- AML-M0 constitutes approximately 5% of AML cases.
- M1 subtype is also called acute myeloblastic leukemia without maturation.
- AML-M1 is defined and characterized by a high percentage of blasts in the bone marrow without significant evidence of myeloid maturation. Blasts constitute >90% of the nonerythroid cells. The M1 blasts express at least two of the following myeloid antigens: CD13, CD33, CD117, MPO, and/or HLA-DR. CD34 is often positive. There is generally no expression of the monocytoid markers CD11b or CD14. AML-M1 constitutes 10% of AML cases.
- M2 subtype is also called acute myeloblastic leukemia with maturation.
- AML-M2 Multiple genomic abnormalities are associated with AML-M2, e.g., formation of a fusion protein, AML1-ETO or RUNX1-RUNX1T1 due to a translocation of chromosome 8 to chromosome 21 or t(8;21) as well as translocation between chromosome 6p23 and chromosome 9q34 causing the formation of a fusion oncogene made of DEK (6p23) and CAN/NUP214 (9q34).
- AML-M2 is defined and characterized by the presence of > 20% blasts in the bone marrow or blood and evidence of maturation to more mature neutrophils. (>10% neutrophils are at different stages of maturation). Monocytes comprise ⁇ 20% of bone marrow cells.
- M2 blasts must express at least two of the following myeloid antigens: CD13, CD33, CD15, CD117, MPO and/or HLA-DR. MPO must be > 3% on blasts. These blasts are generally negative for the monocytic markers CD14 and CD11b.
- AML-M2 constitutes approximately 30-45% of AML cases. AML-M1 and AML-M2 are initially stratified by morphology (e.g., immature cells ⁇ 10% is indicative of AML-M1, and immature cells > 10% is indicative of AML-M2). [0318] M3 subtype is also called acute promyelocytic leukemia or acute progranulocytic leukemia (APL).
- APL/AML-M3 is characterized by proliferation of malignant promyelocytes with mature myeloid immunophenotype and the translocation t(15;17)(q22;q11), which causes the fusion of retinoic acid receptor-alpha (RARalpha) gene on chromosome 17 and the gene PML on chromosome 15.
- RARalpha retinoic acid receptor-alpha
- M3 morphologic variants include: 1) the typical hypergranular form, 2) the microgranular variant, and 3) the basophilic variant. See, e.g., Suci ⁇ et al., J Hematother Stem Cell Res.2002, 11(6):941-950.
- the blasts are positive for the myeloid markers CD33 and CD117.
- the blasts are generally positive for the markers CD13 and CD34 in the hypogranular variant.
- the blasts are characteristically negative HLA Dr, unlike in other AML subtypes.
- AML-M3 constitutes approximately 5-8% of AML cases in adults.
- M4 subtype is also called acute myelomonocytic leukemia (AMML).
- AML-M4 is defined as an acute leukemia with differentiation along both myeloid and monocytic lines. Myeloblasts and monocytes and promonocytes represent > 20%, but monocytes and promonocytes represent ⁇ 80% of the total nucleated cells in the marrow differential. Both myeloblasts and monoblasts are present. There are two types of AML-M4: AML-M4 and AML-M4e variant, which has an increased eosinophil count. Translocation t(8:16)(p11;p13) may be associated with AMML. The myeloblasts generally express the markers CD13, CD33, CD34, CD117, HLA Dr.
- M5 subtype is also called acute monocytic leukemia (AMoL), which is defined as greater than 20% blasts in the bone marrow, of which greater than 80% are of the monocytic lineage.
- a further subclassification is made depending on whether the monocytic cells are predominantly monoblasts (>80%) (acute monoblastic leukemia) or a mixture of monoblasts and promonocytes ( ⁇ 80% blasts).
- Translocation t(8:16)(p11;p13) may be associated with AMoL.
- Blasts characteristically express the markers CD4, CD11b, CD11c, CD13, CD33, CD45, CD56, CD64, and HLA Dr.
- a subset of these cells may also express CD2, CD7, CD10, CD16, CD23, lysosome, and CD117.
- CD34 is predominantly negative.
- AML-M5 constitutes approximately 5-8% of AML cases.
- M6 subtype is also called acute erythroblastic leukemia or erythroleukemia, wherein the cancer cells are either an erythroid/myeloid leukemic cell mix or pure erythroid leukemic cells and is a rare manifestation of AML. More than 30-50% of the nucleated marrow cells are abnormal nucleated red blood cells. Erythroleukemia is defined by > 50% erythroid precursors and > 20% myeloblasts. Pure erythroid leukemia is defined by >80% erythroid precursors. Erythroblasts are positive for Glycophorin A (GPHA) and CD71.
- GPHA Glycophorin A
- the erythroid ny-2770598 Attorney Docket No.24516-20006.40 component lacks MPO, CD34, CD45, and pan myeloid markers. The myeloblasts often express CD117 and CD43.
- AML-M6 constitutes approximately 5-6% of AML cases.
- M7 subtype is also called acute megakaryocytic leukemia (AMKL).
- AMKL is defined as an AML with >20% blasts, of which 50% or more are of the megakaryocyte lineage.
- AML-M7 blasts often resemble lymphoblasts, although AML-M7 leukemia may be accompanied by atypical megakaryocytes.
- Megakaryoblasts expresses of one or more platelet glycoprotein CD41 (glycoprotein IIb/IIIa) and/or CD61 (glycoprotein IIIa).
- Myeloid markers CD13 and CD33 may be positive; CD36 is typically positive.
- AML-M7 constitutes approximately 1.2% of adult AML cases and approximately 3-10% of pediatric AML cases.
- the AML cells express CD13, CD14, and/or CD33 at the time when the AML cells are obtained from the peripheral blood. Methods of obtaining AML cells from peripheral blood is well known in the art.
- AML leukocytes e.g., human AML leukocytes
- MSCs bone-marrow mesenchymal stromal cells
- lymphocytes are non-adherent cells and can therefore be separated from the AML cells.
- AML leukocytes e.g., human AML leukocytes
- AML cells can also be separated by positive selection with (e.g.) anti-CD11b antibody, anti-CD13 antibody, anti-CD14 antibody, and/or anti-CD33 antibody or negative selection using all antibodies against other cells using magnetic-activated cell sorting (MACS). Yet further methods include surface marker staining and cell sorting by fluorescence activated cell sorting (FACS).
- the AML cells obtained from the individual e.g., human cancer patient
- IL-10R IL-10 receptor
- the level of IL-10R on the AML cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-10R on corresponding cells from the reference individual (e.g., a healthy individual).
- the AML cells obtained from the individual e.g., human cancer patient
- IL-4R IL-4 receptor
- the level of IL-4R on the AML cells from the ny-2770598 Attorney Docket No.24516-20006.40 individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-4R on corresponding cells from the reference individual (e.g., a healthy individual).
- the AML cells obtained from the individual express a lower level of IL-6 receptor (“IL-6R”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual.
- IL-6R IL-6 receptor
- the level of IL-6R on the AML cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-6R on corresponding cells from the reference individual (e.g., a healthy individual).
- the AML cells obtained from the individual express a lower level of M-CSF receptor (“M-CSFR”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual.
- M-CSFR M-CSF receptor
- the level of M-CSFR on the AML cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, or 65% lower than the level of M-CSFR on corresponding cells from the reference individual (e.g., a healthy individual).
- the AML cells obtained from the individual express a lower level of GM-CSF receptor (“GM-CSFR”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual.
- GM-CSFR GM-CSF receptor
- the level of GM-CSFR on the AML cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, or 65% lower than the level of GM-CSFR on corresponding cells from the reference individual (e.g., a healthy individual).
- Chronic lymphocytic leukemia (CLL) [0329] Chronic lymphocytic leukemia (CLL) is one of the most frequent types of leukemia. It typically occurs in elderly patients and has a highly variable clinical course. Leukemic transformation is initiated by specific genomic alterations that interfere with the regulation of proliferation and of apoptosis in clonal B-cells.
- the diagnosis is established by blood counts, blood smears, and immunophenotyping of circulating B-lymphocytes, which identify a clonal B-cell population carrying the CD5 antigen as well as typical B-cell markers.
- the clinical staging systems provide prognostic information by using the results of physical examination and blood counts.
- Various biological and genetic markers provide ny-2770598 Attorney Docket No.24516-20006.40 additional prognostic information. Deletions of the short arm of chromosome 17 (del[17p]) and/or mutations of the TP53 gene predict resistance to chemoimmunotherapy and a shorter time to progression with most targeted therapies.
- the CLL international prognostic index integrates genetic, biological, and clinical variables to identify distinct risk groups of patients with CLL. [0331] Only patients with active or symptomatic disease or with advanced Binet or Rai stages require therapy. When treatment is indicated, several therapeutic options exist: a combination of the B-cell lymphoma 2 (BCL2) inhibitor venetoclax with obinutuzumab, monotherapy with inhibitors of Bruton tyrosine kinase (BTK) such as ibrutinib and acalabrutinib, or chemoimmunotherapy. At relapse, the initial treatment may be repeated, if the treatment-free interval exceeds 3 ⁇ years. If the disease relapses earlier, therapy should be changed using an alternative regimen.
- BCL2 B-cell lymphoma 2
- BTK Bruton tyrosine kinase
- NHL Non-Hodgkin's lymphoma
- NHL is the sixth most common cause of cancer-related death in the USA after prostate cancer, breast cancer, lung cancer, colorectal cancer, and bladder cancer.
- Oropharyngeal lymphomas are the second most common malignant disease in the oral region after squamous cell carcinoma. See e.g., J Family Med Prim Care.2020 Apr; 9(4): 1834–1840.
- NHL can be divided into two prognostic groups: the indolent lymphomas and the aggressive lymphomas.
- Indolent NHL types have a relatively good prognosis, with a median survival as long as 20 years, but they usually are not curable in advanced clinical stages.
- Early-stage (stage I and stage II) indolent NHL can be effectively treated with radiation therapy alone.
- Most of the indolent types are nodular (or follicular) in morphology.
- the ny-2770598 Attorney Docket No.24516-20006.40 aggressive type of NHL has a shorter natural history, but a significant number of these patients can be cured with intensive combination chemotherapy regimens. [0335]
- the overall survival rate at 5 years is over 60%. More than 50% of patients with aggressive NHL can be cured. Most relapses occur in the first 2 years after therapy.
- indolent NHL is responsive to immunotherapy, radiation therapy, and chemotherapy, a continuous rate of relapse is usually seen in advanced stages. However, patients can often be re-treated with considerable success if the disease histology remains low grade. Patients who present with or convert to aggressive forms of NHL may have sustained complete remissions with combination chemotherapy regimens or aggressive consolidation with marrow or stem cell support.
- the methods described herein further comprise assessing IL- 10R expression level of the cells, such as cancer-specific monocytes, hematological cancer cells such as B-ALL cells, CLL cells, NHL cells, or AML cells (e.g., prior to contacting the cancer-specific monocytes, B-ALL cells, or AML cells with one or more cytokines, such as IL-10, IFN ⁇ , and/or TNF ⁇ ).
- Chronic Myeloid Leukemia (CML) [0338] Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm with an incidence of 1-2 cases per 100,000 adults. It accounts for approximately 15% of newly diagnosed cases of leukemia in adults.
- BCR-ABL1 Central to the pathogenesis of CML is the fusion of the Abelson murine leukemia (ABL1) gene on chromosome 9 with the breakpoint cluster region (BCR) gene on chromosome 22. This results in expression of an oncoprotein termed BCR-ABL1.
- BCR-ABL1 is a constitutively active tyrosine kinase that promotes growth and replication through downstream signaling pathways such as RAS, RAF, JUN kinase, MYC and STAT. This influences leukemogenesis by creating a cytokine-independent cell cycle with aberrant apoptotic signals in response to cytokine withdrawal.
- CML chronic myeloma
- CP accelerated phase
- BP blast phase
- Rare manifestations include bleeding (associated with a low platelet count and/or platelet dysfunction), thrombosis (associated with thrombocytosis and/or marked leukocytosis), gouty arthritis (from elevated uric acid levels), priapism (usually with marked leukocytosis or thrombocytosis), retinal hemorrhages, and upper gastrointestinal ulceration and bleeding (from elevated histamine levels due to basophilia).
- Leukostatic symptoms due to leukemic cells sludging in the pulmonary or cerebral vessels, are uncommon in CP despite white blood cell (WBC) counts exceeding 100 ⁇ 10 9 /L.
- Splenomegaly is the most consistent physical sign detected in 20-40% of cases. Hepatomegaly is less common (less than 10%). Lymphadenopathy and infiltration of skin or other tissues are rare. When present, they favor Ph-negative CML or AP or BP of CML. Headaches, bone pain, arthralgias, pain from splenic infarction, and fever are more frequent with CML transformation. Most patients evolve into AP prior to BP, but 20% transition into BP without AP warning signals.
- CML-AP might be insidious or present with worsening anemia, splenomegaly, and organ infiltration
- CML-BP presents as an acute leukemia (myeloid in 60%, lymphoid in 30%, megakaryocytic or undifferentiated in 10%) with worsening constitutional symptoms, bleeding, fever, and infections. See e.g., Am J Hematol.2020; 95: 691–709.
- CML mostly involves similar types of cancer cells (mainly myeloid or lymphoid), it is expected that the methods described herein are also effective for CML.
- the individual has a hematological cancer.
- the individual has an advanced hematological cancer. In some embodiments, the individual has a late-stage cancer. In some embodiments, the individual has a cancer that is stage II, III, or IV. In some embodiments, the individual has metastases. In some embodiments, the cancer is malignant. [0344] In some embodiments, the individual has a myeloid leukemia, a B-cell lymphoma, or a B cell leukemia. [0345] In some embodiments, the individual has a B-ALL, CLL, AML, CML, or NHL. [0346] In some embodiments, the individual has a B-ALL, CLL, AML, or NHL.
- the individual is a female. In some embodiments, the individual is a male. [0348] In some embodiments, the individual is a human. In some embodiments, the individual is a human who is at least about any of 50, 55, 60, 65, 70, or 75 years old. In some embodiments, the individual is less than about 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 years old.
- the individual e.g., an individual with a hematological cancer
- the individual has a PBMC or BM sample that comprises at least about 20%, 30%, 40%, 50%, 60%, 70%, or 80% of the cells being the hematological cancer cells (e.g., B-ALL cells, CLL cells, NHL cells, CML cells, AML cells).
- the individual has a PBMC or bone marrow (BM) sample that a) comprises no more than about 5%, 4%, 3%, 2% or 1% of the cells being monocytes (e.g., cancer monocytes), and b) comprises at least about 20%, 30%, 40%, 50%, 60%, 70%, or 80% of the cells being hematological cancer cells (e.g., B-ALL cells, CLL cells, NHL cells, CML cells, AML cells).
- monocytes e.g., cancer monocytes
- b comprises at least about 20%, 30%, 40%, 50%, 60%, 70%, or 80% of the cells being hematological cancer cells (e.g., B-ALL cells, CLL cells, NHL cells, CML cells, AML cells).
- APCs Antigen presenting cells
- the present application provides HC-APCs such as those prepared according to any of the methods described above with unique properties that distinguish them from naturally occurring APCs or APCs generated in vitro by currently known methods.
- the exemplified HC-APCs described herein have distinct surface markers from the hematological cancer cells from which they are derived.
- the HC-APCs described herein express a high level of MHC-I, MHC-II, CD80, CD86, and CD40.
- the HC-APCs also display a clear shift in morphology to a larger cell diameter with increased cell spreading, which is indicative of the ⁇ APC phenotype.
- the HC-APCs described herein comprise one or more exogenous antigen (e.g., an antigenic peptide, a tumor-associated peptide, or a neoantigenic peptide).
- exogenous antigen e.g., an antigenic peptide, a tumor-associated peptide, or a neoantigenic peptide.
- “+/high” described herein refers to a positive expression of a certain surface molecule, or a high expression of a certain surface molecule.
- a high expression refers to the scenario that the cell (e.g., HC-APCs) expresses a higher level of the surface ny-2770598 Attorney Docket No.24516-20006.40 molecule than a reference cell population (e.g., corresponding cells such as myeloid cells or B cells in a healthy individual).
- a higher level refers to an expression level that is at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 2-fold, 5-fold, 10-fold, or 100-fold higher level of expression.
- an increased expression of a certain molecule refers to an expression level that is at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 2-fold, 5-fold, 10-fold, or 100-fold higher level of expression.
- a decreased expression of a certain molecule refers to an expression level that is at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, or 80% less.
- the HC-APCs express a high level of one or more (e.g., two, three, four, five, six, seven, or eight) antigen presentation molecule, wherein the antigen presentation molecule is selected from the group consisting of: MHC-I, MHC-II, CD86, CD80, OX40L, ICAML, ICOSL, and CD40, optionally wherein the HC-APCs are produced from hematological cancer cells (e.g., AML cells, CLL cells, NHL cells, CML cells, or B- ALL cells isolated from an animal, such as a human cancer patient) in a cell culture.
- hematological cancer cells e.g., AML cells, CLL cells, NHL cells, CML cells, or B- ALL cells isolated from an animal, such as a human cancer patient
- the HC-APCs express a high level of MHC-I, MHC-II, CD86, CD80, CD40, and/or OX40L. [0357] In some embodiments, the HC-APCs express a high level of MHC-I, MHC-II, CD86, CD80, CD40, and/or OX40L. [0358] In some embodiments, at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the HC-APCs have a dendritic cell morphology. See e.g., FIG.5C.
- the HC-APCs comprise one or more tumor-associated antigen peptides or tumor-specific antigen peptides, e.g., neoantigen peptides, such as any of those present on the hematological cancer cell (e.g., AML cells, CLL cells, NHL cells, CML cells, or B-ALL cells) at time of collection from a cancer patient.
- the HC-APCs comprise one or more virus-associated antigen peptide.
- the HC-APCs are capable of promoting proliferation of immune cells (e.g., T cells, e.g., CD4 T cells and/or CD8 T cells) upon incubation with the immune cells.
- the HC-APCs promote proliferation of T cells for at least about any of 5-fold, 10-fold, 15-fold, or 20-fold in a cell culture comprising IL-2, IL-7, ny-2770598 Attorney Docket No.24516-20006.40 and IL-15.
- the incubation is no longer than about 24 hours, 22 hours, 20 hours, or 18 hours.
- a composition e.g., a culture
- the HC-APCs present one or more disease-associated peptides (e.g., tumor-associated antigen peptides and/or tumor-specific antigen peptides) to immune cells.
- disease-associated peptides e.g., tumor-associated antigen peptides and/or tumor-specific antigen peptides
- the present application also provides methods of activating a population of immune cells.
- the methods comprise co-culturing the population of immune cells with the population of the HC-APCs described herein, wherein the HC-APCs present endogenous cancer specific antigens, and optionally one or more antigen peptides, (e.g., tumor peptides, e.g., tumor-associated peptides and/or tumor-specific peptides, e.g., neoantigen peptides).
- antigen peptides e.g., tumor peptides, e.g., tumor-associated peptides and/or tumor-specific peptides, e.g., neoantigen peptides.
- the HC-APCs produced according to the methods describe herein possess high potency and can effectively engage antigen-specific immune cells (e.g., T cells), activate and promote the expansion of these immune cells.
- activated immune cells can effectively kill the cancer cells both in in vitro setting (e.g., at a ratio of T cells vs. cancer cell 1:1 or 3:1) and in vivo. See e.g., FIG.17B.
- these activated immune cells are activated T cells.
- the activated T cells comprise a higher percentage of CD8+ T cells than the percentage of CD8+ T cells prior to the incubation with HC-APCs or a new round of expansion.
- the percentage of the CD8+ T cells in the activated T cells is at least about 20%, 40%, 60%, or 80% higher than the percentage of the CD8+ T cells in the T cells prior to the incubation with HC-APCs or a new round of expansion.
- the percentage of the effector memory T cells (i.e., CD45RA-CCR7- CD8 T cells) in the activated T cells is at least about 10%, 15%, 20%, or 25% higher than the percentage of the effector memory T cells in the T cells prior to the incubation with HC-APCs or a new round of expansion. See e.g., FIG.11E.
- the activated T cells express a higher level of CD107a, 4-1BB and/or CD25. See e.g., FIG.17C.
- a method of activating a population of immune cells e.g., T cells, e.g., TIL cells
- a population of immune cells obtained from an individual (e.g., a cancer patient)
- the HC-APCs are derived from B-ALL, CLL, NHL, CML cells, or AML cells obtained from the same individual.
- the ratio of HC-APCs and the immune cells (e.g., T cells, e.g., TIL cells) during co-culturing is about 10:1 to about 1:10 (e.g., about 5:1 to about 1:5, about 2:1 to about 1:2, about 1:1).
- the HC-APCs and the immune cells are co-cultured for at least about 4-8 hours.
- IL-2, IL-7, and/or IL-15 are supplemented to the co-culture (e.g., are supplemented about at least about 4-8 hours after the HC-APCs and the immune cells are first put into co-culture), e.g., for at least about 6 days, e.g., about 6-14 days for each round of expansion.
- the activated immune cells comprise at least 5-, 10-, or 20-fold (e.g., 50-100-fold) more cells than the immune cells prior to the co-culture.
- the activated immune cells are subject to the activation via co-culture with the HC-APCs described herein for at least two, three, four, or five rounds.
- the activated immune cells do not exhibit an exhausted phenotype (e.g., senescence) after two, three, or four consecutive rounds (e.g., about 6-14 days each round, e.g., about 6-10 days each round) of activation that involves the co-culture described herein.
- the co-culture does not involve use of an anti-CD3 antibody and/or an anti-CD28 antibody at least some of the rounds (e.g., anti-CD3 and anti-CD28 antibodies are only used in the first round but not in one or more later rounds). See e.g., FIG.16A.
- the HC-APCs present with one or more endogenous tumor- associated peptides (i.e., one or more tumor-associated or tumor-specific peptides of the isolated AML or B-ALL cells that are differentiated into the HC-APC as described herein).
- one or more endogenous tumor- associated peptides i.e., one or more tumor-associated or tumor-specific peptides of the isolated AML or B-ALL cells that are differentiated into the HC-APC as described herein.
- the method further comprises contacting the HC-APCs with a composition comprising one or more exogenous antigen such as a plurality of tumor- associated peptides (e.g., neoantigen peptides) that are unique from the endogenous neoantigen peptide(s) presented by the cancer cell (e.g., AML cell, NHL cell, CML cells, CLL cell, or B-ALL cell) that is differentiated into an HC-APC.
- a composition comprising one or more exogenous antigen such as a plurality of tumor- associated peptides (e.g., neoantigen peptides) that are unique from the endogenous neoantigen peptide(s) presented by the cancer cell (e.g., AML cell, NHL cell, CML cells, CLL cell, or B-ALL cell) that is differentiated into an HC-APC.
- a cancer cell e.g., AML cell, NHL cell, CML cells
- the HC-APCs are allowed to be in contact with the composition comprising a plurality of tumor-associated peptides (e.g., neoantigen peptides) ny-2770598 Attorney Docket No.24516-20006.40 for about 4 to about 24 hours.
- the HC-APCs have been pre-incubated with the composition comprising a plurality of tumor-associated peptides (e.g., neoantigen peptides).
- the composition comprising a plurality of tumor-associated peptides is a surgical resection of tumor tissue or a biopsy extract thereof.
- the composition comprising a plurality of tumor-associated peptides is a mixture of tumor cells or extract thereof isolated from tumor tissue or biopsy.
- the composition comprising a plurality of tumor-associated peptides is a mixture of isolated tumor- associated peptides (e.g., neoantigen peptides).
- the tumor tissue or cell is a fresh tumor tissue or cell. In some embodiments, the tumor tissue or cell is obtained from a frozen sample. In some embodiments, the tumor tissue or cells have been subjected to an apoptosis induction. In some embodiments, the tumor tissue or cells have been subjected to a radiation treatment. [0370] In some embodiment, the population of immune cells and the HC-APCs are derived from the same individual. [0371] In some embodiments, the population of immune cells and the HC-APCs are not derived from the same individual. [0372] In some embodiments, the HC-APCs are further loaded with one or more exogenous antigens (see e.g., descriptions below).
- the HC-APCs are not loaded with one or more exogenous antigens.
- the present application also provides activated immune cells (e.g., T cells) produced by any of the methods described here.
- T cells activated immune cells
- Tumor-associated Peptides Loading [0375] In some embodiments, the methods described herein further comprise contacting HC- APCs with a plurality of tumor-associated peptides (e.g., neoantigen peptides).
- the plurality of tumor-associated peptides have more than about any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 22, 25, 30, 40, or 50 tumor-associated peptides (e.g., neoantigen peptides).
- the APCs are allowed to be in contact with the composition comprising a ny-2770598 Attorney Docket No.24516-20006.40 plurality of tumor-associated peptides (e.g., neoantigen peptides) for about 4 to about 24 hours.
- the HC-APCs have been pre-incubated with the composition comprising a plurality of tumor-associated peptides (e.g., neoantigen peptides) prior to be used in the methods of activating immune cells described herein.
- An exemplary embodiment of the contacting of a population of HC-APCs with a plurality of tumor-associated peptides (e.g., neoantigen peptides) comprises pulsing the plurality of tumor-associated peptides (e.g., neoantigen peptides) into the population of HC- APCs.
- pulsing refers to a process of mixing cells, such as HC-APCs, with a solution containing tumor-associated peptides (e.g., neoantigen peptides), and optionally subsequently removing the tumor-associated peptides (e.g., neoantigen peptides) from the mixture.
- tumor-associated peptides e.g., neoantigen peptides
- the population of HC-APCs may be contacted with a plurality of tumor- associated peptides (e.g., neoantigen peptides) for seconds, minutes, or hours, such as about any of 30 seconds, 1 minute, 5 minutes, 10 minutes, 15 minutes, 20 minutes, 30 minutes, 1 hour, 5 hours, 10 hours, 12 hours, 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, one week, 10 days, or more.
- the concentration of each neoantigen peptide used in the contacting step may be about any of 0.1, 0.5, 1, 2, 3, 5, or 10 ⁇ g/mL.
- the concentration of the tumor-associated peptides is about 0.1-200 ⁇ g/mL, including for example about any of 0.1-0.5, 0.5-1, 1-10, 10-50, 50-100, 100-150, or 150-200 ⁇ g/mL.
- the population of HC-APCs is contacted with the plurality of tumor-associated peptides (e.g., neoantigen peptides) in the presence of a composition that facilitates the uptake of the plurality of tumor-associated peptides (e.g., neoantigen peptides) by the HC-APCs.
- compounds, materials, or compositions may be included in a solution of the plurality of tumor-associated peptides (e.g., neoantigen peptides) to facilitate peptide uptake by the HC-APCs.
- Compounds, materials, or compositions that facilitate the uptake of the plurality of tumor-associated peptides (e.g., neoantigen peptides) by the HC-APCs include, but are not limited to, lipid molecules and peptides with multiple positively charged amino acids.
- more than about any of 50%, 60%, 70%, 80%, 90%, or 95% of the tumor-associated peptides are uptaken by the population of HC-APCs. In some embodiments, more than about any of 50%, 60%, 70%, 80%, 90%, or 95% of the HCAPCs in the population uptake at least one exogenous tumor antigen peptide.
- ny-2770598 Attorney Docket No.24516-20006.40 Immune cells [0379]
- the immune cells described herein can be any type of immune cells that interact with HC-APCs and can be activated by HC-APCs, and then exert their desired functions. Exemplary immune cells include T cells.
- T cells or T lymphocytes, play a central role in cell-mediated immunity.
- Each clone of activated T cells express a distinct T-cell receptor (TCR) on the surface, which is responsible for recognizing antigens bound to MHC molecules on APCs (such as HC-APCs) and on target cells (such as cancer cells).
- T cells are subdivided into several types, each expressing a unique combination of surface proteins and each having a distinct function.
- Cytotoxic T cells participate in the immune response to and destruction of tumor cells and other infected cells, such as virus-infected cells.
- TC cells function by recognizing a class I MHC presented antigen on an APC (such as a HC-APC) or any target cell. Stimulation of the TCR, along with a co-stimulator (for example CD28 on the T cell binding to B7 on the APC, or stimulation by a helper T cell), results in activation of the TC cell. The activated TC cell can then proliferate and release cytotoxins, thereby destroying the APC, or a target cell (such as a cancer cell). Mature TC cells generally express surface proteins CD3 and CD8. Cytotoxic T cells belong to CD3+CD8+ T cells.
- Helper T cells are T cells that help the activity of other immune cells by releasing T cell cytokines, which can regulate or suppress immune responses, induce cytotoxic T cells, and maximize cell killing activities of macrophages.
- TH cells function by recognizing a class II MHC presented antigen on an APC (such as a HC-APC).
- Mature TH cells express the surface proteins CD3 and CD4.
- Helper T cells belong to CD3+CD4+ T cells.
- Regulatory T cells (TREG cells) generally modulate the immune system by promoting tolerance for self-antigens, thereby limiting autoimmune activity. In cancer immunotherapy, T REG contributes to escape of the cancer cells from the immune response.
- T REG cells generally express CD3, CD4, CD7, CD25, CTLA4, GITR, GARP, FOXP3, and/or LAP.
- CD4+CD25+Foxp3+ T cells are one class of T REG cells.
- Memory T cells Tm are T cells that have previously encountered and responded to their specific antigens, or T cells that differentiated from activated T cells. Although tumor specific Tms constitutes a small proportion of the total T cell amount, they serve critical functions in surveillance of tumor cells during a person’s entire lifespan. If tumor specific ny-2770598 Attorney Docket No.24516-20006.40 Tms encounter tumor cells expressing their specific tumor antigens, the Tms are immediately activated and clonally expanded.
- an antigen for T cells is a protein molecule or a linear fragment of a protein molecule that can be recognized by a T-cell receptor (TCR) to elicit specific T cell response.
- TCR T-cell receptor
- the antigen can be derived from a foreign source such as a virally encoded protein, or an endogenous source such as a protein expressed on the cell surface.
- the minimal fragment of an antigen that is directly involved in interaction with a particular TCR is known as an epitope.
- an antigen peptide or antigen fragment is processed into an epitope by an APC (such as a dendritic cell or HC-APC), and then bound in an extended conformation inside a Major Histocompatibility (MHC) molecule to form an MHC-peptide complex on the surface of an APC (such as a dendritic cell or HC-APC).
- APC such as a dendritic cell or HC-APC
- MHC molecules are also known as human leukocyte antigens (HLA).
- the MHC provides an enlarged binding surface for strong association between TCR and epitope, while a combination of unique amino acid residues within the epitope ensures specificity of interaction between TCR and the epitope.
- the human MHC molecules are classified into two types—MHC class I and MHC class II—based on their structural features, especially the length of epitopes bound inside the corresponding MHC complexes.
- MHC-I epitopes are epitopes bound to and represented by an MHC class I molecule.
- MHC-II epitopes are epitopes bound to and represented by an MHC class II molecule.
- MHC-I epitopes are typically about 8 to about 11 amino acids long, whereas MHC-II epitopes are about 13 to about 17 amino acids long.
- the immune cells are selected from the group consisting of PBMC, tumor infiltrating T cells (TIL), and T cells (e.g., CD4 T cells and/or CD8 T cells).
- TIL tumor infiltrating T cells
- TIL tumor infiltrating T cells
- TIL tumor infiltrating T cells
- the immune cells are CD4 T cells and/or CD8 T cells.
- the immune cells and the HC-APCs are derived from the same individual. In some embodiments, the immune cells and the HC-APCs are derived from different individuals.
- Methods described herein comprise co-culturing a population of immune cells (e.g., T cells) with a population of HC-APCs described herein that present with a plurality of one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides).
- the co-culturing was carried out for at least 24 hours. In some embodiments, the co-culturing was carried out for at least about 1-5 days (e.g., about 1-3 days).
- the population of immune cells (e.g., T cells) and the population of HC-APCs that present with the plurality of one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) are co-cultured for about any of 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, or 30 days.
- the population of immune cells (e.g., T cells) is co-cultured with the population of HC-APCs that present with the plurality of one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) for about 14 to about 21 days.
- the population of immune cells (e.g., T cells) is co-cultured with the population of HC-APCs that present with the plurality of one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) for about 14 days.
- the population of immune cells used in any embodiment of the methods described herein may be derived from a variety of sources. A convenient source of the immune cells is from the PBMCs of the human peripheral blood.
- the population of T cells may be isolated from the PBMCs, or alternatively, a population of PBMCs enriched with T cells (such as by addition of T cell specific antibodies and cytokines) can be used in the co-culture.
- the population of T cells used in the co- culture is obtained from the non-adherent fraction of peripheral blood mononuclear cells (PBMCs).
- the PBMCs are obtained by density gradient centrifugation of a sample of peripheral blood.
- the population of activated T cells is prepared by obtaining a population of non-adherent PBMCs, and co-culturing the population of non-adherent PBMCs with a population of HC-APCs that present with a plurality of one or ny-2770598 Attorney Docket No.24516-20006.40 more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) (such as in the presence of at least one cytokine (such as IL-2) and an anti-CD3 antibody).
- tumor-associated and/or tumor-specific peptides e.g., neoantigen peptides
- the co-culture may further include cytokines and other compounds to facilitate activation, maturation, and/or proliferation of the T cells, as well as to prime T cells for later differentiation into e.g., memory T cells.
- cytokines that may be used in this step include, but are not limited to, IL-7, IL-15, IL-21, and the like. Certain cytokines may help suppress the percentage of TREG in the population of activated T cells in the co-culture.
- a high dose (such as about any of 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1200, or 1500 U/ml) of a cytokine (such as IL-2) is used to co-culture the population of T cells and the population of dendritic cells loaded with the plurality of tumor antigen peptides to obtain a population of activated T cells with a low percentage of T REG cells.
- the methods of activating immune cells comprise co-culture of the immune cells (e.g., the T cells) and HC-APC populations for more than one round (e.g., two, three, or four rounds). In some embodiments, each round takes about 6-8 days.
- the first, second, third, and/or fourth round do not involve the addition of an anti-CD3 antibody and/or an anti-CD28 antibody.
- the immune cells e.g., the T cells
- the immune cells e.g., T cells
- each round takes about 5-10 days or 6-8 days.
- the number of the immune cells (e.g., T cells) after three or four rounds of co-culture reaches about 10 10 .
- the methods of activating immune cells described herein further comprise expanding the population of immune cells following the co-culturing step.
- expanding the population of immune cells comprises contacting the immune cells with a cytokine selected from the group consisting of IL-2, IL-7, and IL-15, optionally for about 2 to about 10 days.
- the co-culture is in the presence of an anti-CD3 antibody and a plurality of cytokines, such as IL-2, IL-7, IL-15, IL- 21, or any combination thereof.
- the present application also provides populations of activated immune cells obtained by the methods described in this section. ny-2770598 Attorney Docket No.24516-20006.40 V.
- the present application in another aspect provides methods of treating a disease or condition (e.g., a cancer) in an individual (e.g., a patient), comprising contacting hematological cancer cells and/or monocytes with one or more of survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) an IL-10 receptor (IL-10R) activator.2) a TNF ⁇ receptor (TNFR) activator, 3) a GM-CSF receptor (GM-CSFR) activator, and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator (e.g., any of the one or more S/D/M factors described in the Section named “Survival, differentiation, and/or maturation factors (‘S/D/M factors’)”), thereby producing a population of antigen presenting cells (“APCs”), optionally wherein the APCs are further loaded with one or more exogenous antigen,
- APCs antigen
- the hematological cancer cells and/or monocytes are obtained from the individual, optionally wherein the hematological cancer cells and monocytes are comprised in a mixture when contacting with the one or more S/D/M factors. See e.g., FIG.2.
- the method further comprises administering an effective amount of a TNF ⁇ inhibitor (e.g., an anti-TNF ⁇ antibody) into the individual.
- the TNF ⁇ inhibitor is administered to the individual prior to (e.g., at least 1, 2 or 3 days prior to) the administration of the activated immune cells.
- the method comprises: a) obtaining monocytes and/or hematological cancer cells from the individual (e.g., via obtaining a PBMC or BM sample from the individual), b) contacting monocytes and/or hematological cancer cells with one or more of survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) an IL-10 receptor (IL-10R) activator, 2) a TNF ⁇ receptor (TNFR) activator, 3) a GM-CSF receptor (GM-CSFR) activator, and 4) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, thereby producing a population of APCs, c) optionally further loading the APCs with one or more exogenous antigens, d) contacting the APCs with a population of immune cells (e.g., CD4+ and/or CD8+ T cells from the same individual), thereby producing activated immune
- S/D/M factors
- the hematological cancer cells are cancer cells of a myeloid leukemia or a B ny-2770598 Attorney Docket No.24516-20006.40 cell malignancy (e.g., a B cell lymphoma or a B cell leukemia).
- the hematological cancer cells are cancer cells of AML cells (e.g., M0-M5 myeloblasts), CML cells, B-ALL cells, CLL cells, or NHL cells.
- the method comprises contacting the APCs with the CD4 and CD8 T cells separately.
- the method comprises contacting the APCs with CD4 and CD8 T cells together.
- the method further comprises expanding the immune cells (e.g., CD4 and/or CD8 T cells) for at least one, two, three, or four rounds.
- the method further comprises obtaining at least about 10 8 , 5x10 8 , 10 9 , 5x10 9 , or 10 10 antigen-specific T cells (e.g., Neo-T cells) after several rounds of expansion.
- the method further comprises administering at least about 10 8 , 5x10 8 , 10 9 , 5x10 9 , or 10 10 antigen-specific T cells (e.g., Neo-T cells) into the individual.
- the one or more S/DM factors comprise two, three, or more agents selected from the group consisting of: 1) an IL- 10R activator, 2) a TNFR activator, 3) a GM-CSFR activator, and 4) an IFNGR activator.
- the one or more of S/D/M factors comprises: 1) an IL-10R activator, 2) a TNFR activator, and 3) an IFNGR activator.
- the one or more of S/D/M factors comprises: 1) an IL-10R activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) a GM-CSFR activator.
- the one or more of S/D/M factors comprises: 1) an IL-10R activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-6R activator.
- the one or more of S/D/M factors comprises: 1) an IL-10R activator (e.g., IL-10 or any of the IL-10R activator listed in Table 1), 2) a TNFR activator (e.g., TNF ⁇ ), 3) an IFNGR activator (e.g., IFN ⁇ ), 4) an IL-6R activator (e.g., IL-6), 5) a GM-CSFR activator (e.g., GM-CSF), and 6) an IL-4R activator (e.g., IL-4).
- an IL-10R activator e.g., IL-10 or any of the IL-10R activator listed in Table 1
- a TNFR activator e.g., TNF ⁇
- IFNGR activator
- the method further comprises, prior to contacting the HC- APCs with immune cells, contacting the HC-APCs with one or more of refinement factors selected from the group consisting of type-I interferon, IFN ⁇ , TNF ⁇ , a TLR ligand, CD40L or a CD40-ligating antibody, an anti-PD-L1 antibody, and TPI-1, optionally wherein the type-I interferon comprises IFN ⁇ and/or IFN ⁇ , and optionally wherein the TLR ligand is poly IC, CpG, or LPS.
- the immune cells and hematological cancer cells are from the same individual.
- the immune cells and hematological cancer cells are from different individuals. In some embodiments, the immune cells are administered to the individual from whom they are obtained. In some embodiments, the immune cells are administered to a different individual from whom they are obtained. In some embodiments, ny-2770598 Attorney Docket No.24516-20006.40 the immune cells are selected from the group consisting of PBMC, cells derived from bone marrow biopsy, cells derived from lymphoma biopsy, tumor infiltrating T cells (TILs), and T cells, optionally wherein the immune cells are T cells, optionally wherein the T cells are CD8 T cells and/or CD4 T cells.
- TILs tumor infiltrating T cells
- the hematological cancer cells are obtained from PBMC, enriched leukemia cells, a bone marrow biopsy, or a lymphoma biopsy. In some embodiments, the hematological cancer cells are cultured with the one or more of S/D/M factors for at least 2 days. In some embodiments, the HC-APCs are cultured with the refinement factors for about 1-4 days. In some embodiments, the immune cells have been enriched prior to contacting with the HC-APCs. In some embodiments, the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors.
- the hematological cancer cells are in a mixture (e.g., PBMC sample, e.g., a mixture comprising monocytes) when contacted with the S/D/M factors.
- the hematological cancer is a myeloid leukemia, optionally wherein the hematological cancer is acute myeloid leukemia (AML), optionally wherein the hematological cancer cells are any of M0-M5 myeloblasts.
- the hematological cancer cells are CML cells.
- the hematological cancer cells have been enriched prior to the contacting with the one or more of S/D/M factors in the presence of an anti-CD11b antibody.
- the hematological cancer is a B- cell lymphoma or B-cell leukemia.
- the B-cell lymphoma or B-cell leukemia is selected from the group consisting of B-cell acute lymphoblastic leukemia B- ALL), chronic lymphocytic leukemia (CLL), and non-Hodgkin’s lymphoma (NHL).
- the hematological cancer cells have been enriched prior to the contacting with the one or more of S/D/M factors in the presence of an anti-CD19 or anti-CD20 antibody.
- the present application also provides methods of treating a disease or condition (e.g., a cancer) in a patient, comprising administering to the patient a population of activated immune cells obtained by the methods described above.
- a disease or condition e.g., a cancer, e.g., a cancer associated with a hematological cancer, e.g., a hematological cancer
- a population of activated immune cells such as any of those described in Section IV.
- the cancer associated with a hematological cancer is a secondary cancer that is derived from a ny-2770598 Attorney Docket No.24516-20006.40 hematological cancer. In some embodiments, the cancer associated with a hematological cancer is a primary cancer from which the hematological cancer is derived from.
- a method of treating a cancer comprising administering to the patient a population of activated immune cells, wherein the activated immune cells are activated by hematological cancer antigen presenting cells (“HC-APCs”), wherein the HC-APCs are derived from hematological cancer cells (e.g., B-ALL, CLL, or NHL CML, or AML cells) obtained from an individual (e.g., a cancer patient), wherein the HC-APCs express a high level of one or more antigen presentation molecules, wherein the one or more antigen presentation molecules is selected from the group consisting of: MHC-I, MHC-II, CD86, CD80, and CD40.
- HC-APCs hematological cancer antigen presenting cells
- the method further comprises administering a second therapy that induces immunogenic cell death (e.g., radiotherapy).
- the method comprises administering the activated immune cells and a radiotherapy concurrently, simultaneously, or subsequently.
- the APCs comprise one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B- ALL, CLL, CML, NHL, or AML cells) from which the HC-APCs were derived.
- the HC-APCs have not been preloaded with a disease- or condition-associated antigen (e.g., a tumor antigen) prior to the administration. In some embodiments, the HC- APCs have been preloaded with a disease- or condition-associated antigen (e.g., tumor antigen or a virus antigen) prior to the administration.
- a disease- or condition-associated antigen e.g., tumor antigen or a virus antigen
- a method of treating a cancer comprising administering to the patient a population of activated immune cells, wherein the activated immune cells are activated by HC-APCs, wherein the HC-APCs are derived from hematological cancer cells (e.g., B-ALL, CLL, NHL, CML, or AML cells) obtained from an individual (e.g., a cancer patient), wherein the HC-APCs are obtained by contacting a population of hematological cancer cells (e.g., B-ALL, CLL, NHL, CML, or AML cells) obtained from an individual with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) an IL-10 receptor (IL-10R) activator
- the method further comprises administering a second therapy that induced immunogenic cell death (e.g., radiotherapy).
- the method comprises administering the activated immune cells and a radiotherapy concurrently, simultaneously, or subsequently.
- the HC- APCs comprise one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B-ALL, CLL, NHL, CML, or AML cells) from which the HC-APCs were derived.
- the HC-APCs have not been preloaded with a disease- or condition- associated antigen (e.g., a tumor antigen) prior to the administration. In some embodiments, the HC-APCs have been preloaded with a disease- or condition-associated antigen (e.g., tumor antigen or a virus antigen) prior to the administration.
- a disease- or condition-associated antigen e.g., tumor antigen or a virus antigen
- a method of treating a cancer comprising administering to the patient a population of activated immune cells, wherein the activated immune cells are activated by HC-APCs, wherein the HC-APCs are derived from hematological cancer cells (e.g., B-ALL, CLL, NHL, CML, or AML cells) obtained from an individual (e.g., a cancer patient), wherein the HC-APCs: a) express a high level of one or more antigen presentation molecules, wherein the one or more antigen presentation molecules is selected from the group consisting of: MHC-I, MHC-II, CD86, CD80, OX40L, ICAML, ICOSL, and CD40, and/or b) a low level of an inhibitory signaling molecule, wherein the inhibitory signaling
- the hematological cancer cells exhibit a lower expression level of M-CSFR, GM-CSFR, IL-6R, IL-10R, and/or IL-4R (e.g., at least about 10%, 20%, 30%, 40%, 50%, or 60% lower) at the time when they are obtained from the individual as compared to the corresponding cells obtained from a reference individual (e.g., a healthy individual).
- a reference individual e.g., a healthy individual.
- a method of treating a cancer comprising administering to the patient a population of activated immune cells, wherein the immune cells ny-2770598 Attorney Docket No.24516-20006.40 have been subject to co-culture with a population of HC-APCs, wherein the HC-APCs are produced after being contacted with an IL-10 receptor activator (IL-10R activator) and one or more of IFN ⁇ receptor activator (IFNR activator), TNF ⁇ receptor activator (TNFR activator), and IL-4 receptor activator (IL-4R activator), and wherein the HC-APCs comprise one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e
- a method of treating a hematological cancer in a patient comprising administering to the patient a population of activated immune cells (e.g., T cells), wherein the immune cells have been subject to co-culture with a population of HC-APCs, wherein the HC-APCs are produced after being contacted with IL-10 and one or more of IFN ⁇ , TNF ⁇ , and IL-4, and wherein the HC-APCs comprise one or more tumor- associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B-ALL, CLL, NHL, CML, or AML cells) from which the HC-APCs were derived.
- activated immune cells e.g., T cells
- the HC-APCs are produced after being contacted with IL-10 and one or more of IFN ⁇ , TNF ⁇ , and IL-4
- the immune cells are T cells.
- the immune cells are T cells (e.g., CD3 T cells, e.g., CD4 T cells, e.g., CD8 T cells, e.g., both CD4 and CD8 T cells, e.g., TILs) obtained from the peripheral blood of the patient.
- the immune cells are T cells (e.g., CD3 T cells, e.g., CD4 T cells, e.g., CD8 T cells, e.g., both CD4 and CD8 T cells, e.g., TILs) obtained from the peripheral blood of an individual different from the patient (optionally with a matching HLA type).
- the HC-APCs and the activated immune cells are derived from the same individual. In some embodiments, the HC-APCs and the activated immune cells are derived from different individuals (optionally with a matching HLA type). In some embodiments, the HC-APCs are produced after being contacted with IL-10, IFN ⁇ , TNF ⁇ , and IL-4. In some embodiments, the HC-APCs are produced after being contacted with IL-10, IFN ⁇ , TNF ⁇ , GM-CSF, IL-6, and IL-4. In some embodiments, the HC-APCs are produced after being contacted with one or more of the refinement factors described in Section II.
- the activated immune cells are administered intratumorally, intraperitoneally, or intravenously. In some embodiments, the activated immune cells are administered at about 10 7 to about 10 9 cells per dose.
- the methods of treatment described herein further comprise treating the patient with chemotherapy, radiation therapy, or an immune checkpoint inhibitor. In some embodiments, the method comprises ny-2770598 Attorney Docket No.24516-20006.40 treating the patient with irradiation. In some embodiments, the site of irradiation is different from the site of the cancer to be treated.
- a method of treating a virus-related cancer in a patient comprising administering to the patient a population of activated T cells, wherein the T cells have been subject to co-culture with a population of HC-APCs, wherein the HC- APCs are produced after being contacted with IL-10 and one or more of IFN ⁇ , TNF ⁇ , and IL- 4, and wherein the HC-APCs comprise one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B-ALL, CLL, NHL, CML, or AML cells) from which the HC-APCs were derived, wherein virus antigen-reactive T cells have been removed from the activated T cell population prior to the administration.
- tumor-associated and/or tumor-specific peptides e.g., neoantigen peptides
- the HC- APCs are derived from the patient.
- the activated T cells are derived from the patient.
- the HC-APCs and the activated T cells are both derived from the patient.
- the activated immune cells are administered intratumorally, intraperitoneally, or intravenously.
- the activated immune cells are administered at about 10 7 to about 10 9 cells per dose.
- the methods of treatment described herein further comprise treating the patient with chemotherapy, radiation therapy, or an immune checkpoint inhibitor.
- the method comprises treating the patient with irradiation.
- the site of irradiation is different from the site of the cancer to be treated.
- a method of treating a cancer comprising administering to the patient a population of activated T cells, wherein the T cells have been subject to co-culture with a population of HC-APCs, wherein the HC- APCs are produced after being contacted with IL-10 and one or more of IFN ⁇ , TNF ⁇ , and IL- 4, and wherein the HC-APCs comprise one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B-ALL, CLL, NHL, CML, or AML cells) from which the HC-APCs were derived, wherein virus antigen-reactive T cells have been removed from the hematological cancer cells (e.g., the B-ALL, CLL, NHL, CML, or AML cells) from which the HC-APCs were derived, wherein virus antigen-reactive T cells have
- the HC- APCs are derived from the patient.
- the activated T cells are derived from the patient.
- the HC-APCs and the activated T cells are both derived from the patient.
- the activated immune cells are administered ny-2770598 Attorney Docket No.24516-20006.40 intratumorally, intraperitoneally, or intravenously.
- the activated immune cells are administered at about 10 7 to about 10 9 cells per dose.
- the methods of treatment described herein further comprise treating the patient with chemotherapy, radiation therapy, or an immune checkpoint inhibitor.
- the method comprises treating the patient with irradiation.
- the site of irradiation is different from the site of the cancer to be treated.
- the patient has a hematological cancer.
- the patient has an advanced cancer.
- the patient has a late-stage cancer.
- the patient has a cancer that is stage II, III, or IV.
- the patient has metastases.
- the patient is a terminally ill patient.
- the patient is a female.
- the patient is a male.
- the patient is a human.
- the patient is at least about any of 50, 55, 60, 65, 70, or 75 years old. In some embodiments, the patient is less than about any of 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 years old.
- the individual e.g., an individual with a hematological cancer
- monocytes e.g., cancer monocytes
- the individual has a PBMC or bone marrow (BM) sample that: a) comprises no more than about any of 5%, 4%, 3%, 2% or 1% of the cells being monocytes (e.g., cancer monocytes), and b) comprises at least about any of 20%, 30%, 40%, 50%, 60%, 70%, or 80% of the cells being hematological cancer cells (e.g., B-ALL cells, CLL cells, NHL cells, CML cells, or AML cells).
- monocytes e.g., cancer monocytes
- b comprises at least about any of 20%, 30%, 40%, 50%, 60%, 70%, or 80% of the cells being hematological cancer cells (e.g., B-ALL cells, CLL cells, NHL cells, CML cells, or AML cells).
- the methods of treatment that involve immune cells (such as T cells) activated by APCs produced by various methods described herein are applicable to ny-2770598 Attorney Docket No.24516-20006.40 various types of cancer, including a non-hematological cancer in an individual who had, has, or is at risk of developing a hematological cancer.
- the non- hematological cancer is associated with the hematological cancer.
- the cancer is a hematologic cancer.
- the cancer is an advanced cancer.
- the cancer is a late-stage cancer.
- the cancer is stage II, III, or IV.
- the cancer is malignant.
- the cancer has been subjected to and/or failed one or more prior therapy (e.g., an immune checkpoint blockage therapy (e.g., an PD-1 antibody), a chemotherapy, a surgery, a cell therapy (e.g., an allogenic NK cell infusion therapy)).
- an immune checkpoint blockage therapy e.g., an PD-1 antibody
- a chemotherapy e.g., an PD-1 antibody
- a surgery e.g., an allogenic NK cell infusion therapy
- a cell therapy e.g., an allogenic NK cell infusion therapy
- cancers described herein include, but are not limited to, acute lymphoblastic leukemia, chronic myelogenous leukemia, Hodgkin’s lymphoma, non- Hodgkin’s lymphoma, leukemia, B-cell chronic lymphocytic leukemia, acute myeloid leukemia, AIDS-related cancers (e.g., AIDS-related lymphoma), central nervous system lymphoma, chronic myeloproliferative disorders, lymphoid neoplasm (e.g., lymphoma), myelodysplastic syndromes, myelodysplastic/myeloproliferative diseases, lymphoma, primary central nervous system lymphoma (microglioma), and post-transplant lymphoproliferative disorder (PTLD).
- acute lymphoblastic leukemia chronic myelogenous leukemia
- Hodgkin’s lymphoma non- Hodgkin’s lymphoma
- leukemia B-cell chronic lymph
- the cancer is a virus-infection-related cancer.
- the cancer is human T-lymphotrophic virus (HTLV-1)-related cancer (e.g., adult T cell leukemia or lymphoma).
- HTLV-1 human T-lymphotrophic virus
- the cancer is Epstein-Barr virus (EBV)-related cancer (e.g., Burkitt lymphoma, Hodgkin’s and non-Hodgkin’s lymphoma).
- EBV Epstein-Barr virus
- the cancer is hepatitis C virus-related cancer (e.g., non-Hodgkin’s lymphoma).
- the activated immune cells can be administered at any desired dosage, which in some aspects includes a desired dose or number of cells or cell type(s).
- the dosage of cells in some embodiments is based on a total number of cells (or number per kg body weight) and/or a desired ratio of the individual populations.
- the dosage of cells is ny-2770598 Attorney Docket No.24516-20006.40 based on a desired total number (or number per kg of body weight) of cells in the individual populations or of individual cell types.
- the activated immune cells are administered to the subject at a range of about one million to about 100 billion cells and/or that amount of cells per kilogram of body weight, such as, e.g., 1 million to about 50 billion cells (e.g., about 5 million cells, about 25 million cells, about 500 million cells, about 1 billion cells, about 5 billion cells, about 20 billion cells, about 30 billion cells, about 40 billion cells, or a range defined by any two of the foregoing values), such as about 10 million to about 100 billion cells (e.g., about 20 million cells, about 30 million cells, about 40 million cells, about 60 million cells, about 70 million cells, about 80 million cells, about 90 million cells, about 10 billion cells, about 25 billion cells, about 50 billion cells, about 75 billion cells, about 90 billion cells, or a range defined by any two of the foregoing values), and in some cases about 100 million to about 100 billion cells and/or that amount of cells per kilogram of body weight, such as, e.g., 1 million to about 50 billion cells (e.g., about 5 million
- the dose includes fewer than about 1 ⁇ 10 9 total activated immune cells, e.g., in the range of about 1 ⁇ 10 6 to 5 ⁇ 10 8 such cells, such as 2 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1 ⁇ 10 8 , 5 ⁇ 10 8 or 1 ⁇ 10 9 total such cells, or the range between any two of the foregoing values.
- the activated immune cells are administered at about 10 7 to about 10 9 cells per dose.
- the method of treatment comprises administration of a dose comprising a number of cells from about 1 ⁇ 10 6 to 1 ⁇ 10 9 (e.g., 10 6 to 10 7 , 10 7 to 10 8 , or 10 8 to 10 9 ) total activated immune cells (e.g., total CD3 T cells, both CD4 and CD8 T cells, CD4 T cells only, CD8 T cells only, or TILs).
- the dose of the activated immune cells is administered to the subject as a single dose or is administered only one time within a period of two weeks, one month, three months, six months, 1 year, or more.
- the dose of total activated immune cells is within a range of between at or about 10 4 and at or about 10 9 cells/kilograms (kg) body weight, such as between 10 5 and 10 6 cells / kg body weight, for example, at or about 1 ⁇ 10 5 cells/kg, 1.5 ⁇ 10 5 cells/kg, 2 ⁇ 10 5 cells/kg, or 1 ⁇ 10 6 cells/kg body weight.
- the activated immune cells are administered at, or within a certain range of error of, between at or about 10 4 and at or about 10 9 T cells/kilograms (kg) body weight, such as between 10 5 and 10 6 T cells / kg body weight, for example, at or about 1 ⁇ 10 5 T cells/kg, 1.5 ⁇ 10 5 T cells/kg, 2 ⁇ 10 5 T cells/kg, or 1 ⁇ 10 6 T cells/kg body weight.
- the activated immune cells are administered at or within a certain range of error of between at or about 10 4 and at or about 10 9 CD4 + and/or CD8 + cells/kilograms (kg) body weight, such as between 10 5 and 10 6 CD4 + and/or CD8 + cells / kg body weight, for example, at or about 1 ⁇ 10 5 CD4 + and/or CD8 + cells/kg, 1.5 ⁇ 10 5 CD4 + and/or CD8 + cells/kg, 2 ⁇ 10 5 CD4 + and/or CD8 + cells/kg, or 1 ⁇ 10 6 CD4 + and/or CD8 + cells/kg body weight.
- the activated immune cells are administered at or within a certain range of error of, greater than, and/or at least about 1 ⁇ 10 6 , about 2.5 ⁇ 10 6 , about 5 ⁇ 10 6 , about 7.5 ⁇ 10 6 , or about 9 ⁇ 10 6 CD4 + cells, and/or at least about 1 ⁇ 10 6 , about 2.5 ⁇ 10 6 , about 5 ⁇ 10 6 , about 7.5 ⁇ 10 6 , or about 9 ⁇ 10 6 CD8+ cells, and/or at least about 1 ⁇ 10 6 , about 2.5 ⁇ 10 6 , about 5 ⁇ 10 6 , about 7.5 ⁇ 10 6 , or about 9 ⁇ 10 6 T cells.
- the activated immune cells are administered at or within a certain range of error of between about 10 8 and 10 12 or between about 10 10 and 10 11 T cells, between about 10 8 and 10 12 or between about 10 10 and 10 11 CD4 + cells, and/or between about 10 8 and 10 12 or between about 10 10 and 10 11 CD8 + cells.
- the appropriate dosage may depend on the type of disease to be treated, the type of cells or recombinant receptors, the severity and course of the disease, whether the activated immune cells are administered for preventive or therapeutic purposes, previous therapy, the subject's clinical history and response to the activated immune cells, and the discretion of the attending physician.
- compositions and cells are in some embodiments suitably administered to the subject at one time or over a series of treatments.
- the APCs described herein are administered to the subject at a range of about 5000 to about 10,000 cells/mm 3 per tumor mass.
- the size of the dose is determined based on one or more criteria such as response of the subject to prior treatment, e.g., chemotherapy, disease burden in the subject, such as tumor load, bulk, size, or degree, extent, or type of metastasis, stage, and/or likelihood or incidence of the subject developing toxic outcomes, e.g., CRS, macrophage activation syndrome, tumor lysis syndrome, neurotoxicity, and/or a host immune response against the activated immune cells being administered.
- the size of the dose is determined by the burden of the disease or condition in the subject.
- the number of cells administered in the dose is determined based on the tumor burden that is present in the subject immediately prior to administration of the initiation of the dose of cells.
- the size of the first and/or subsequent dose is inversely correlated with disease burden.
- the subject is administered a low number of cells.
- the subject is administered a larger number of cells.
- the activated immune cells and/or a second agent described herein can be administered by any suitable means, for example, by bolus infusion, injection (e.g., intravenous or subcutaneous injections, intraocular injection, periocular injection, subretinal injection, intravitreal injection, trans-septal injection, subscleral injection, intrachoroidal injection, intracameral injection, subconjectval injection, subconjuntival injection, sub-Tenon's injection, retrobulbar injection, peribulbar injection), or posterior juxtascleral delivery.
- injection e.g., intravenous or subcutaneous injections, intraocular injection, periocular injection, subretinal injection, intravitreal injection, trans-septal injection, subscleral injection, intrachoroidal injection, intracameral injection, subconjectval injection, subconjuntival injection, sub-Tenon's injection, retrobulbar injection, peribulbar injection
- injection e.g., intravenous or subcutaneous injections,
- activated immune cells and/or a second agent described herein are administered by parenteral, intrapulmonary, intranasal, and, if desired for local treatment, intralesional administration.
- Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
- a given dose is administered by a single bolus administration of the activated immune cells.
- the dose is administered by multiple bolus administrations of the activated immune cells, for example, over a period of no more than 3 days, or by continuous infusion administration of the activated immune cells.
- the activated immune cells are administered intratumorally, intraperitoneally, or intravenously. ny-2770598 Attorney Docket No.24516-20006.40 Combination therapy [0441] In some embodiments, the activated immune cells are administered as part of a combination treatment, such as simultaneously with, concurrently with, or sequentially with, another therapeutic intervention (i.e., a second therapy), such as an antibody or engineered cell or receptor or agent, such as a cytotoxic or therapeutic agent. In some embodiments, the activated immune cells are administered prior to another therapeutic intervention. In some embodiments, the activated immune cells are administered after another therapeutic intervention.
- a combination treatment such as simultaneously with, concurrently with, or sequentially with, another therapeutic intervention (i.e., a second therapy), such as an antibody or engineered cell or receptor or agent, such as a cytotoxic or therapeutic agent.
- a second therapy such as an antibody or engineered cell or receptor or agent, such as a cytotoxic or therapeutic agent.
- the activated immune cells are administered prior to
- the activated immune cells in some embodiments are co-administered with one or more additional therapeutic agents or in connection with another therapeutic intervention, simultaneously, concurrently, or sequentially in any order.
- the activated immune cells are co-administered with another therapy sufficiently close in time such that the cell populations enhance the effect of one or more additional therapeutic agents, or vice versa.
- the activated immune cells are administered prior to the one or more additional therapeutic agents.
- the activated immune cells are administered after the one or more additional therapeutic agents.
- the one or more additional agents to be administered includes a cytokine, such as IL-2, for example, to enhance persistence of the activated immune cells.
- the methods comprise administration of a chemotherapeutic agent.
- the second therapy comprises a TNF ⁇ inhibitor (e.g., an anti- TNF ⁇ antibody).
- the TNF ⁇ inhibitor is administered to the individual prior to (e.g., at least 1, 2 or 3 days prior to) the administration of the activated immune cells.
- the TNF ⁇ inhibitor is selected from the group consisting of: a small molecule inhibitor, a neutralizing antibody, a TNF ⁇ receptor blockade antibody, a soluble TNF ⁇ receptor, a TNF ⁇ -targeting short interfering RNA (siRNA), a chemical inhibitor of TNF ⁇ mRNA stability, an inhibitor of TNF ⁇ converting enzyme (TACE), and derivatives thereof.
- the TNF ⁇ inhibitor is an anti-TNF ⁇ neutralizing antibody.
- the TNF ⁇ inhibitor is an anti-TNF ⁇ receptor blockade antibody.
- the anti-TNF ⁇ antibody is a monoclonal antibody.
- the anti-TNF ⁇ antibody is a chimeric, humanized, and/or fully human antibody.
- Suitable antibodies for use in the methods provided herein include, but are not limited to, Remicade® (Infliximab (Centocor)), and those antibodies described, for example, ny-2770598 Attorney Docket No.24516-20006.40 in U.S.
- Other suitable anti-TNF ⁇ antibodies for use in the methods provided herein are, by way of non-limiting example, Humira (Adalimumab (Abbott Laboratories, Esai)) as described in U.S.
- the TNF ⁇ inhibitor is a fusion protein.
- Suitable fusion proteins for use in the methods provided herein include, but are not limited to, Enbrel (Etanercept (Amgen)) and other fusion proteins or fragments thereof described in U.S. Patent No. 5,712,155, PCT Publication No. WO 91/03553, and related patents and applications, the contents of which are hereby incorporated by reference in their entirety.
- the anti-TNF ⁇ antagonist is a modified antibody antagonist or a non-antibody-based antagonist.
- Such antagonists include advanced antibody therapeutics, such as antibody fragments including, but not limited to, CimziaTM (Certolizumab pegol, CDP870 (Enzon)), bispecific antibodies, Nanobodies® such as ABX 0402 (Ablynx), immunotoxins, and radiolabeled therapeutics; peptide therapeutics; gene therapies, particularly intrabodies; oligonucleotide therapeutics such as aptamer therapeutics, antisense therapeutics, interfering RNA therapeutics; and small molecules such as LMP-420 (LeukoMed) as described in EP Patent No.0767793, and related patents and applications, the contents of which are hereby incorporated by reference in their entirety.
- advanced antibody therapeutics such as antibody fragments including, but not limited to, CimziaTM (Certolizumab pegol, CDP870 (Enzon)), bispecific antibodies, Nanobodies® such as ABX 0402 (Ablynx), immunotoxins, and radiolabeled therapeutics; peptide therapeutics;
- the TNF ⁇ inhibitor is an antibody, such as infliximab, adalimumab, etanercept, golimumab, and certolizumab.
- the TNF ⁇ inhibitor is administered systemically.
- the TNF ⁇ inhibitor is administered concurrently with the immune cells described herein. 111 ny-2770598 Attorney Docket No.24516-20006.40
- the individual does not develop cytokine release syndrome or pro-inflammatory organ damage.
- administration of the TNF ⁇ inhibitor does not compromise or weakly compromises tumor clearance.
- the second therapy comprises a chemotherapy, radiation therapy, or an immune checkpoint inhibitor.
- the second therapy is gene therapy (e.g., mRNA-based gene therapy).
- the second therapy comprises administration of a cancer vaccine (such as mRNA-based cancer vaccine or DNA- based cancer vaccine).
- the second therapy comprises administration of an oncolytic virus.
- the activated immune cells are administered prior to the administration of the second therapy.
- the activated immune cells are administered in a neoadjuvant setting.
- the second therapy comprises treating the patient with irradiation.
- the site of irradiation is different from the site of the cancer to be treated.
- a method of treating an individual having cancer comprising administering to the individual an effective amount of an immune cell activated by any of the methods described herein, wherein the individual is treated with a radiation therapy, and wherein the site of the irradiation is different from the site of the cancer to be treated.
- the radiation therapy is selected from the group consisting of external-beam radiation therapy, internal radiation therapy (brachytherapy), intraoperative radiation therapy (IORT), systemic radiation therapy, radioimmunotherapy, and administration of radiosensitizers and radioprotectors.
- the radiation therapy is external-beam radiation therapy, optionally comprising three-dimensional conformal radiation therapy (3D-RT), intensity modulated radiation therapy (IMRT), photon beam therapy, image-guided radiation therapy (IGRT), and sterotactic radiation therapy (SRT).
- the radiation therapy is brachytherapy, optionally comprising interstitial brachytherapy, intracavitary brachytherapy, intraluminal radiation therapy, and radioactively tagged molecules given intravenously.
- a method of producing a population of antigen presenting cells presenting an antigen associated with a hematological cancer comprising: ny-2770598 Attorney Docket No.24516-20006.40 contacting the hematological cancer cells obtained from an individual having the hematological cancer cells with one or more survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of 1) a STAT3 activator 2) a TNF ⁇ receptor (TNFR) activator, 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, and 4) and an interleukin-4 receptor (IL-4R) activator, thereby producing the population of HC-APCs.
- S/D/M factors survival, differentiation and/or maturation factors
- Embodiment 2 A method of producing activated immune cells, comprising: a) producing a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”) according to method of embodiment 1, and b) contacting the HC-APCs with immune cells, thereby producing activated immune cells.
- HC-APC hematological cancer
- a method of producing activated immune cells comprising contacting immune cells with a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), thereby generating activated immune cells; wherein the HC-APCs are derived from hematological cancer cells after having been contacted with one or more survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of 1) a STAT3 activator, 2) a TNF ⁇ receptor (TNFR) activator, 3) and an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, and 4) an interleukin-4 receptor (IL-4R) activator.
- S/D/M factors survival, differentiation and/or maturation factors
- the STAT3 activator is selected from the group consisting of: an IL-10, an IL-10 family member, an IL-10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, a small molecule activator of IL-10R, and a small molecule activator of STAT3, optionally wherein the activator of STAT3 is selected from an IL-10 family cytokine, an IL-12 family cytokine, an IL-6 family cytokine, a small molecule STAT3 activator, and G-CSF .
- the activator of STAT3 is selected from an IL-10 family cytokine, an IL-12 family cytokine, an IL-6 family cytokine, a small molecule STAT3 activator, and G-CSF .
- the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL20, IL-24, IL12, IL-23, IL-6, colivelin TFA, Garcinone D, and G-CSF, optionally wherein the IL-10R activator is IL-10, IL-22, IL-19, IL-20, IL-24, IL-12, IL-23, Colivelin TFA, or Garcinone D.
- the one or more of S/D/M factors comprise an interleukin-4 receptor (IL-4R) activator, optionally ny-2770598 Attorney Docket No.24516-20006.40 wherein the IL-4R activator is selected from the group consisting of IL-4, an IL-4R agonist antibody, and a small molecule activator of IL-4R.
- IL-4R activator is selected from the group consisting of IL-4, an IL-4R agonist antibody, and a small molecule activator of IL-4R.
- the one or more of S/D/M factors comprise a TNFR activator, optionally wherein the TNFR activator is selected from the group consisting of TNF ⁇ , a TNFR agonist antibody, and a small molecule activator of TNFR.
- Embodiment 9. The method of embodiment 8, wherein the TNFR activator is TNF ⁇ .
- Embodiment 10. The method of any one of embodiments 1-9, wherein the one or more of S/D/M factors comprise an IFNGR activator, optionally wherein the IFNGR activator is selected from the group consisting of IFN ⁇ , an IFNGR agonist antibody, and a small molecule activator of IFNGR.
- Embodiment 11 The method of embodiment 10, wherein the IFNGR activator is IFN ⁇ .
- Embodiment 12 The method of any one of embodiments 1-11, wherein the one or more of S/D/M factors are present in a single composition.
- Embodiment 13 The method of any one of embodiments 1-12, wherein the one or more of S/D/M factors comprise two or more agents selected from the group consisting of 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator.
- Embodiment 14 Embodiment 14.
- Embodiment 15 The method of any one of embodiments 1-13, wherein the one or more of S/D/M factors comprise three or more agents selected from the group consisting of 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator.
- the one or more of S/D/M factors comprise three or more agents selected from the group consisting of 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator.
- the one or more of S/D/M factors comprise 1) a STAT3 activator, 2) a TNFR activator, and 3) an IFNGR activator, optionally wherein the one or more of S/D/M factors comprise an IL-10 family cytokine (e.g., IL-10, IL-22, IL-19, IL-24, IL-20, IL-26), TNF ⁇ , and IFN ⁇ .
- an IL-10 family cytokine e.g., IL-10, IL-22, IL-19, IL-24, IL-20, IL-26
- TNF ⁇ e.g., TNF ⁇ , and IFN ⁇ .
- the one or more of S/D/M factors comprise 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, ny-2770598 Attorney Docket No.24516-20006.40 and 4) an IL-4R activator, optionally wherein the one or more of S/D/M factors comprise an IL-10 family cytokine (e.g., IL-10, IL-22, IL-19, IL-24, IL-20, IL-26), TNF ⁇ , IL-4, and IFN ⁇ .
- IL-10 family cytokine e.g., IL-10, IL-22, IL-19, IL-24, IL-20, IL-26
- TNF ⁇ IL-4
- IFN ⁇ IFN ⁇
- the one or more of the S/D/M factors further comprise an IL-6 receptor (IL-6R) activator and/or a GM-CSF receptor (GM-CSFR) activator, optionally wherein the IL-6R activator is selected from the group consisting of IL-6, an IL-6R agonist antibody, and a small molecule activator of IL-6R, and optionally wherein the GM-CSFR activator is selected from the group consisting of GM-CSF, a GM-CSFR agonist antibody, and a small molecule activator of GM- CSFR.
- IL-6R IL-6 receptor
- GM-CSFR GM-CSF receptor
- Embodiment 19 The method of any one of embodiments 1-18, wherein the hematological cancer cells are comprised in a mixture comprising monocytes from the individual. [0474] Embodiment 20.
- the method further comprises, prior to contacting the HC-APCs with immune cells, contacting the HC-APCs with one or more of refinement factors selected from the group consisting of type-I interferon, IFN ⁇ , TNF ⁇ , a TLR ligand, CD40L or a CD40-ligating antibody, an anti- PD-L1 antibody, and TPI-1, optionally wherein the type-I interferon comprises IFN ⁇ and/or IFN ⁇ , optionally wherein the refinement factors is selected from the group consisting of R848, poly IC, CpG, or LPS, and further optionally wherein the refinement factors comprise R848 and poly IC. [0475] Embodiment 21.
- Embodiment 22 The method of embodiment 19, wherein the refinement factors further comprise at least two agents selected from the group consisting of poly IC, CpG, CD40L, R848, and an anti-PD-L1 antibody, optionally wherein the refinement factors comprise a SHP-1 inhibitor (e.g., TPI-1).
- Embodiment 23 The method of any one of embodiments 2-22, wherein the method further comprises administering the activated immune cells into the individual. ny-2770598 Attorney Docket No.24516-20006.40 [0478] Embodiment 24.
- Embodiment 25 The method of any one of embodiments 2-24, wherein the immune cells and hematological cancer cells are from the same individual.
- Embodiment 26 The method of any one of embodiments 2-24, wherein the immune cells and hematological cancer cells are from the different individuals.
- Embodiment 27 The method of any one of embodiments 23-26, wherein the immune cells are administered to the individual where they are obtained from.
- Embodiment 28 The method of any one of embodiments 23-26, wherein the immune cells are administered to a different individual from the individual where they are obtained from.
- Embodiment 29 The method of any one of embodiments 2-28, the immune cells are selected from the group consisting of PBMC, cells derived from bone marrow biopsy, cells derived from lymphoma biopsy, tumor infiltrating T cells (TIL), and T cells, optionally wherein the immune cells are T cells, optionally wherein the T cells are CD8 T cells and/or CD4 T cells.
- Embodiment 30 The method of any one of embodiments 1-29, wherein the hematological cancer cells are obtained from PBMC, enriched leukemia cells, a bone marrow biopsy, or a lymphoma biopsy.
- Embodiment 31 Embodiment 31.
- Embodiment 32 The method of any one of embodiments 20-31, wherein the HC-APCs are cultured with the refinement factors for about 1-4 days.
- Embodiment 33 The method of any one of embodiments 2-32, wherein the immune cells have been enriched prior to contacting with the HC-APCs.
- Embodiment 34 The method of any one of embodiments 1-33, wherein the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors.
- Embodiment 35 The method of any one of embodiments 1-34, wherein the hematological cancer is a myeloid leukemia, optionally the hematological cancer is acute myeloid leukemia (AML) or chronic myeloid leukemia (CML), optionally wherein the hematological cancer cells are any of M0-M5 myeloblasts.
- Embodiment 36 The method of embodiment 35, wherein the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors in the presence of an anti-CD11b antibody.
- Embodiment 37 Embodiment 37.
- Embodiment 38 The method of embodiment 37, wherein the B-cell lymphoma or B-cell leukemia is selected from the group consisting of B-cell acute lymphoblastic leukemia B-ALL), chronic lymphocytic leukemia (CLL), and non-Hodgkin’s lymphoma (NHL).
- Embodiment 39 The method of embodiment 37 or embodiment 38, wherein the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors in the presence of an anti-CD19 or anti-CD20 antibody.
- Embodiment 40 The method of any one of embodiments 2-39, wherein the method further comprises expanding the immune cells by contacting immune cells with the HC-APCs for at least two, three or four rounds.
- Embodiment 41 A population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”) obtained by the method of any one of embodiments 1, 4-22, 30-31 and 33-39.
- Embodiment 42 Embodiment 42.
- a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), wherein the APCs are derived from hematological cancer cells, and wherein the APCs express increased levels of MHC-1, MHC- II, CD40, CD80, and/or CD86 than the hematological cancer cells prior to the treatment, optionally wherein the HC-APCs comprise one or more exogenous antigen.
- Embodiment 43 The population of HC-APC of embodiment 42, wherein the hematological cancer cells are primary cells from an individual having the hematological cancer.
- ny-2770598 Attorney Docket No.24516-20006.40
- hematological cancer is a myeloid leukemia
- the hematological cancer is acute myeloid leukemia (AML) or chronic myeloid leukemia (CML), optionally wherein the hematological cancer cells are any of M0-M5 myeloblasts.
- AML acute myeloid leukemia
- CML chronic myeloid leukemia
- HC-APC of embodiment 42 or embodiment 43 wherein the hematological cancer is a B-cell lymphoma or B-cell leukemia, optionally wherein the B-cell lymphoma or B-cell leukemia is selected from the group consisting of B- cell acute lymphoblastic leukemia B-ALL), chronic lymphocytic leukemia (CLL), and non- Hodgkin’s lymphoma (NHL).
- B-ALL B- cell acute lymphoblastic leukemia
- CLL chronic lymphocytic leukemia
- NHL non- Hodgkin’s lymphoma
- a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), prepared by a) obtaining hematological cancer cells from an individual having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of 1) a STAT3 activator 2) a TNF ⁇ receptor (TNFR) activator, 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, and 4) and an interleukin-4 receptor (IL-4R) activator, thereby producing the population of HC-APCs.
- S/D/M factors survival, differentiation and/or maturation factors
- Embodiment 48 A method of treating a hematological cancer in an individual, comprising administering to the individual an effective amount of activated immune cells of embodiment 47.
- Embodiment 49 A method of treating a hematological cancer in an individual, comprising administering to the individual an effective amount of activated immune cells of embodiment 47.
- a method of treating a hematological cancer in an individual comprising: a) contacting hematological cancer cells and/or monocytes with one or more of survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of 1) a STAT3 activator 2) a TNF ⁇ receptor (TNFR) activator, 3) an interferon ⁇ (IFN ⁇ ) receptor (IFNGR) activator, and 4) an interleukin-4 receptor (IL-4R) activator and, thereby producing a population of antigen ny-2770598 Attorney Docket No.24516-20006.40 presenting cells (“APCs”), optionally wherein the APCs are further loaded with one or more exogenous antigen, b) contacting the APCs with a population of immune cells from the individual, thereby producing activated immune cells, and c) administering activated immune cells into the individual.
- S/D/M factors survival, differentiation and/or maturation factors
- Embodiment 50 The method of embodiment 49, wherein the hematological cancer cells and/or monocytes are obtained from the individual, optionally wherein the hematological cancer cells and monocytes are comprised in a mixture when contacting with the one or more S/D/M factors.
- NeoT Multi-targeting neoantigen-specific T cells
- cMO- ⁇ APC Peripheral monocytes
- AML/CML- ⁇ APC myeloid leukemia cells
- ALL/CLL- ⁇ APC B-lineage leukemia cells
- NHL- ⁇ APC lymphoma cells
- FIG.4 shows a table that summarizes results from representative studies that consistently evidenced that the methods discussed herein successfully make ⁇ APC from either cMo or cancer cells of various sources.
- Step-1 Collection of Starting Materials All cancer patients were subjected to a round of mononuclear cell apheresis to obtain PBMC, from which peripheral monocytes (cMO), malignant leukemia cells (e.g., B-ALL, ny-2770598 Attorney Docket No.24516-20006.40 CLL), and peripheral blood lymphocytes (PBL) were further separated. cMO and malignant leukemia cells were used to generate ⁇ APC, as well as for separation of PBL that are used to produce NeoT. [0508] Bone marrow aspiration and bone marrow biopsy: Leukemia of all forms originate from bone marrow where greater numbers of malignant cells and the cancer microenvironment reside.
- Bone marrow aspiration (liquid) and biopsies (solid part) hence provide malignant cell materials for preparing cancer neoantigens/antigens and also TILs from which NeoT are derived. Multi-point aspiration and biopsies that comprise diversity of cancer neoantigens/antigens are preferred.
- Lymphoma biopsies For NHL, biopsies at the enlarged lymphomas were taken to obtain malignant cell materials and TILs.
- ⁇ APC are differentiated APC produced from peripheral monocytes of cancer patients (cMO) or healthy donors (MO), or myelogenous or B cell-lineage malignant cells using an HC-activator reagent.
- Nomenclature Dependent on the source from which the cells are derived, ⁇ APC are further designated as: [0512] Common - require uptake of cancer antigens for activation of NeoT.
- cMO- ⁇ APC produced from cancer patient peripheral monocytes (cMO).
- cMO- ⁇ APC are common ⁇ APC and can be used to activate autologous NeoT from patients of all types of cancer.
- MO- ⁇ APC produced from healthy donor peripheral monocytes (MO). MO- ⁇ APC also common ⁇ APC. Upon uptake of antigens, MO- ⁇ APC can activate antigen-specific T cells.
- AML- ⁇ APC produced from AML myelogenous cancer cells.
- CML- ⁇ APC produced from CML myelogenous cancer cells.
- ALL- ⁇ APC produced from B-ALL malignant B lymphoblastic cells.
- NeoT polyclonal multitargeting cancer neoantigen-specific CD4 and CD8 T cells produced by ⁇ APC-mediated antigen presentation that selectively activates NeoT, resulting in NeoT expansion. NeoT detect and kill malignant cells through TCRs recognizing neoantigens displayed by MHC molecules.
- Cancer neoantigens include gene mutation- resultant novel antigens (major category), virus infection-resultant non-self-antigens, as well as self-antigens expressed aberrantly that breach immune tolerance.
- all ⁇ APC are produced following a similar scheme: a minimum 1-2 days treatment of the source cells with HC- activator, optionally followed by 6-24h treatment with the APC refinement agent.
- HC-activator contains selective cytokines that support the source cell (e.g., cancer monocytes, e.g., hematological cancer cells) survival and differentiation into antigen- presenting cells ( ⁇ APC).
- the product ⁇ APC post HC-activator treatment are professional APCs that are highly phagocytic and feature a proinflammatory phenotype proficient for immunogenic antigen presentation and activation of NeoT.
- Six-component HC-activator [0524] The six-component HC-activator contains: a) a STAT3 activator, b) IFN ⁇ , c) TNF ⁇ , d) IL-4, e) GM-CSF, and f) IL-6.
- STAT3 activators are listed in Table 1 below, and the following STAT3 activators have been tested, and demonstrated comparable effects: IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. It was found that the six-component HC-activator unanimously achieved at least about 80%-90% survival and differentiation of ⁇ APC derived from hematological cancer cells from various cancer patients having different types of hematological cancers such as those discussed in the Examples below.
- Four-component HC-activator [0525] The four-component HC-activator does not contain IL-6 and GM-CSF.
- STAT3 activator It has: a) a STAT3 activator, b) IFN ⁇ , c) TNF ⁇ , and d) IL-4.
- Exemplary STAT3 activators are listed in Table 1 below.
- the four-component HC-activator is expected to achieve comparable effects as the six-component HC-activator.
- ny-2770598 Attorney Docket No.24516-20006.40
- Three-component HC-activator [0526]
- the three-component HC-activator has: a) a STAT3 activator, b) IFN ⁇ , and c) TNF ⁇ .
- Exemplary STAT3 activators are listed in Table 1 below.
- the three-component HC-activator is expected to achieve at least 80% of the effectiveness of the 6-component or 4-component HC-activator, including keeping the hematological cancer cells alive for one or more weeks and promoting the differentiation of these cells to ⁇ APCs.
- the six-component HC-activator can be prepared by having a cell culture medium (e.g., RPMI 1640 with heat-inactivated human serum) supplemented with a stock solution (10x), which contains human IL-10 and IFN ⁇ , TNF ⁇ , IL-4, GM-CSF, and IL-6.
- a cell culture medium e.g., RPMI 1640 with heat-inactivated human serum
- a stock solution (10x) which contains human IL-10 and IFN ⁇ , TNF ⁇ , IL-4, GM-CSF, and IL-6.
- the HC-activator includes about IL-10 (or other IL-10 family or IL-12 family cytokines in lieu of IL-10), IFN ⁇ , TNF ⁇ , IL-4, GM-CSF, and IL-6.
- concentrations of the exemplary STAT3 activators are shown in Table 1. ny-2770598 Attorney Docket No.24516-20006.40 [0528] It was also found that T cell medium prepared as follows can achieve comparable results due to the presence of the key ingredients in the T cell medium.
- Modified Act-T medium The TCR-activated T cells are prepared by treating peripheral blood lymphocytes (PBL) cultured with anti-human CD3 antibody and anti-human CD28 antibody to induce T cell activation.
- Cell culture medium is collected on day 2 (48h) or day 3 (72h), designated as IL-2 high Act-T medium or IL-2 low Act-T medium, respectively.
- Both IL-2 high and IL-2 low Act-T medium contain high levels of IFN ⁇ (> 50ng/ml) and TNF ⁇ (> 3ng/ml), and IL-4 (> 20pg/ml), GM-CSF (> 300pg/ml) and IL-6 (>1ng/ml).
- the IL-2 low Act-T medium was further mixed with a cell culture medium (e.g., RPMI 1640 with heat-inactivated human AB serum) and further supplemented with: a) recombinant human IL-10 to form HC-activator, b) another IL-10 family cytokine to form HC-activator, c) recombinant human IL-12 to form HC-activator, d) Colivelin TFA to form HC-activator, or e) Garcinone D to form HC-activator.
- a cell culture medium e.g., RPMI 1640 with heat-inactivated human AB serum
- IL-2 high Act-T medium with IL-2 depletion can be used to prepare the HC-activator series.
- IL-2 high HC-activator medium is a natural T cell growth medium.
- Optional APC Refinement Agent is a reagent complex that can be used optionally to further refine the ⁇ APC phenotype.
- HC-activator-optional APC refinement agent refers to the treatment of HC-activator alone or combined treatment of HC- activator and the optional APC refinement agent.
- the optional APC refinement agent treatment can further refine ⁇ APC but is not required for generation of potent ⁇ APC.
- the optional APC refinement agent comprises cytokines (choices of IFN ⁇ , IFN ⁇ , and TNF ⁇ ), TLR ligands (choices of Poly IC, CpG, and R848), and/or TPI-1.
- the optional APC refinement agent includes R848 and Poly IC. Step-3 Loading cMO- ⁇ APC With Tumor Antigens [0534] Only cMO- ⁇ APC are required to ‘feed’ with cancer antigens in order for antigen presentation and activation of NeoT.
- cancer antigen materials such as freeze-thaw cancer ny-2770598 Attorney Docket No.24516-20006.40 biopsy debris or synthetic neoantigen peptides, for a minimum of 4h (4-16h) to enable cMO- ⁇ APC phagocytosis of cancer materials, processing antigens, and loading antigens onto MHC molecules.
- cancer antigen materials such as freeze-thaw cancer ny-2770598 Attorney Docket No.24516-20006.40 biopsy debris or synthetic neoantigen peptides, for a minimum of 4h (4-16h) to enable cMO- ⁇ APC phagocytosis of cancer materials, processing antigens, and loading antigens onto MHC molecules.
- cancer antigen materials such as freeze-thaw cancer ny-2770598 Attorney Docket No.24516-20006.40 biopsy debris or synthetic neoantigen peptides, for a minimum of 4h (4-16h) to enable cMO- ⁇ AP
- enriched cancer cells were treated with 30Gy X-ray radiation and followed by a cycle of freeze-thawing to produce cell debris.
- ⁇ APC produced from malignant cells i.e., AML- ⁇ APC, CML- ⁇ APC, ALL- ⁇ APC CLL- ⁇ APC and NHL- ⁇ APC, directly present their inherent mutation-associated neoantigens to cognate T cells, resulting in NeoT activation. These ⁇ APC do NOT require uptake of additional cancer antigens.
- Step-4 ⁇ APC antigen presentation activating NeoT from PBL and/or TIL
- Antigen-loaded cMO- ⁇ APC, or prepared hematological malignant cell ⁇ APC (i.e., AML- ⁇ APC, CML- ⁇ APC, ALL- ⁇ APC, CLL- ⁇ APC and NHL- ⁇ APC) cultures in the optional APC refinement agent were exposed to peripheral blood lymphocytes (PBL) at a ratio of 1:100-500 of ⁇ APC: PBL.
- PBL peripheral blood lymphocytes
- the co-culture is gently tilted to enhance cognate engagement of ⁇ APC and cancer antigen specific T cells (NeoT).
- NeoT can also be expanded from TILs.
- fresh tumor biopsies or surgically resected tumor tissues were dissociated into single cells, followed by positive isolation of TILs using anti-CD3-conjugated beads.
- cancer antigen- loaded cMO- ⁇ APC, or prepared hematological malignant cell ⁇ APC i.e.
- AML- ⁇ APC, CML- ⁇ APC, ALL- ⁇ APC CLL- ⁇ APC or NHL- ⁇ APC) were co-cultured with isolated TILs at the ratio of ⁇ APC: TIL is 1:10-100.
- the NeoT cells were cultured for the next 6-10 days with IL- 2 supplementation (also IL-7 and IL-15), refreshed medium, and cell splitting to support NeoT activation and expansion.
- IL- 2 supplementation also IL-7 and IL-15
- PBL and TILs can be combined to co-culture with antigen-loaded cMO- ⁇ APC or hematological malignant cell ⁇ APC for NeoT selection, activation, and expansion.
- Step-4 is the 1 st round of antigen-specific NeoT activation and expansion, which generally harvest ⁇ 10 7-8 NeoT in ⁇ 10 days.
- additional rounds of antigen-specific NeoT expansion can be performed following the same procedure of Step-4. Each round further expands NeoT approximately 50-100 fold. After the 3 rd expansion, the NeoT number reaches > 10 10 .
- the final NeoT cells were washed and suspended in cold Plasma- Lyte A, ready for intravenous (i.v.) administration to the same patient for autologous adoptive cell therapy (ACT).
- the 2 nd and 3 rd rounds of large expansion together take 12-20 days.
- the 2 nd and 3 rd rounds of NeoT expansion can be done by antibody ligation of CD3 and CD28, a step that leads to T cell activation and proliferation. As this method is less optimal, it is only used when ⁇ APC or cancer antigen materials are insufficient for large-scale NeoT expansion.
- HC-activator and optional APC refinement agent differentiate B-lineage malignant leukocytes, along with cMO, into CLL- ⁇ APC for activating NeoT
- B-lineage malignancies include B-ALL, CLL, MM and NHL, together occupying > 50% of all cases of hematological cancer.
- PBMC samples were analyzed from patients of B-ALL (5 samples), CLL (1 sample), MM (2 samples, and NHL (1 sample) for the presence of cancer cells, cMO, and CD4 and CD8 T cells.
- Matched BM samples for B-ALL (# BAL1) and MM, and dissociated lymphoma tumor cells (DTC) from NHL were also obtained for analyses.
- Both cMO and malignant B lymphocytic cells from these samples were treated with the HC-activator (2d) and the optional APC refinement agent (1d) for ⁇ APC differentiation.
- the final cell products were analyzed for the antigen presentation phenotype by cell surface expression of MHC-I, MHC-II, CD80, CD86, and CD40.
- cMO were responsive to the HC-activator (2d) and optionally APC refinement agent (1d) treatment and differentiated into cMO- ⁇ APC.
- HC-activator (2d) and optionally APC refinement agent (1d) treatment were all depleted of cMO in PBMC and BM, and thus were unable to produce sufficient cMO- ⁇ APC for antigen presentation to activate NeoT.
- FIG.4 summarizes the representative studies. Representative sample analysis details are presented in FIGs.5A-9B.
- FIGs.5A-7B show studies of three cases of B-ALL patients (Karnelian #: BAL3, 4 & 5).
- FIGs.5A-5C show that the HC-activator (optionally also with the optional APC refinement agent) differentiate malignant B lymphoblasts from B-ALL into ALL- ⁇ APC.
- FIG.5A shows analyses of PBMC from a patient with newly diagnosed B-ALL (Karnelian # BAL3).
- FIG. 5B shows that the HC-activator (optionally also with the optional APC refinement agent) for treating malignant B lymphoblasts induced increased expression of antigen presentation molecules, including MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86, indicating cell differentiation into professional APC, termed ALL- ⁇ APC.
- FIG.5C shows representative microscopic images of malignant B lymphoblasts prior to and after HC-activator treatment (optionally with additional APC refinement agent treatment).
- FIGs.6A-6B show that HC-activator treatment (optionally with additional APC refinement agent treatment) differentiated malignant B lymphoblasts from B-ALL into ALL- ⁇ APC.
- FIG.6A shows analyses of PBMC from a newly diagnosed B-ALL patient (Karnelian ny-2770598 Attorney Docket No.24516-20006.40 # BAL4). Malignant B lymphoblasts (B-ALL cells) occupied 86.8% of total PBMC, and monocytes (cMO) were diminished (0.91%). CD4 and CD8 T cells were present.
- FIG.6B shows that HC-activator treatment (optionally with additional APC refinement agent treatment) for malignant B lymphoblasts induced their differentiation into ALL- ⁇ APC featured with increased expression of antigen presentation molecules MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86.
- FIGs.7A-7B show that HC-activator treatment (optionally with additional APC refinement agent treatment) differentiated malignant B lymphoblasts from B-ALL into ALL- ⁇ APC.
- FIG.7A shows the analyses of PBMC from a newly diagnosed B-ALL patient (Karnelian # BAL5).
- FIG.7B shows that HC-activator treatment (optionally with additional APC refinement agent treatment) of B lymphoblasts induced their differentiation into ALL- ⁇ APC that featured increased expression of antigen presentation molecules MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86.
- FIGs.8A-8B show that HC-activator treatment (optionally with additional APC refinement agent treatment) differentiated peripheral monocytes (cMO) and malignant B lymphocytic cells from CLL into cMO- ⁇ APC and CLL- ⁇ APC.
- FIG.8A shows that analyses of PBMC from a stage III CLL patient (Karnelian # CLL1) revealed that malignant B lymphocytic cells (CLL cells) occupied 4.7% of the total PBMC. Monocytes (cMO, 11.2%) and CD4 and CD8 T cells were present.
- FIG.8B shows that HC-activator treatment (optionally with additional APC refinement agent treatment) for PBMC induced differentiation of both cMO and malignant B lymphocytic cells into ⁇ APC, cMO- ⁇ APC, and CLL- ⁇ APC, respectively.
- Both cMO- ⁇ APC and CLL- ⁇ APC featured increased expression of antigen presentation molecules MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86.
- ny-2770598 Attorney Docket No.24516-20006.40 Non-Hodgkin’s lymphoma (NHL) (FIGs.9A-9B) [0557]
- DTC Dissociated tumor cells from the tumor of non-Hodgkin’s lymphoma (NHL) were obtained from Discovery Life Science.
- FIGs.9A-9B show that HC-activator treatment differentiated malignant B lymphocytes from non-Hodgkin’s lymphoma (NHL) into NHL- ⁇ APC.
- FIG.9A shows the analyses of dissociated tumor cells (DTC) from lymphoma of an NHL patient. Malignant B lymphocytes (NHL cells) occupied 65.8% of the total tumor cells.
- FIG.9B shows that HC-activator treated DTC induced differentiation of malignant B lymphocytes (NHL cells) into NHL- ⁇ APC, which featured increased expression of antigen presentation molecules MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86.
- Multiple myeloma (MM) (FIGs.10A-10B)
- PBMC and BM samples from patients of MM were analyzed by flow cytometry. MM cancer cells were found primarily in BM, whereas PBMC contained monocytes (cMO), CD4 lymphocytes, and CD8 lymphocytes.
- cMO monocytes
- MM cancer cells being B-lineage cells
- treating these cells with HC-activator (2d) and optional APC refinement agent (1d) failed to induce their differentiation into phenotypic antigen presenting cells.
- treating cMo from MM patients induced differentiation of cMO- ⁇ APC that featured increased expression of MHC-I, MHC-II, CD80, CD86, and CD40.
- cMO- ⁇ APC were used for developing NeoT therapy. Given that cMO were not malignant cells, cMO- ⁇ APC were required to phagocytose MM antigens prior to being used for antigen presentation and activation of NeoT.
- FIGs.10A-10B show that HC-activator differentiated cMO but not malignant plasma cells of MM into ⁇ APC.
- FIG.10A shows that the analyses of PBMC and BM samples from an MM patient revealed the presence of malignant plasma cells (MM), as well as CD4 and CD8 T cells, in both PBMC and BM. There were cMO and CD4 and CD8 T cells in PBMC.
- FIG.10B shows that HC-activator treated PBMC (optionally additionally with optional APC refinement agent treatment) induced differentiation of cMO into ⁇ APC.
- cMO- ⁇ APC featured increased expression of antigen presentation molecules MHC-I and MHC-II and co- ny-2770598 Attorney Docket No.24516-20006.40 stimulatory molecules CD40, CD80, and CD86. However, the same treatment failed to induce differentiation of malignant plasma cells (MM) from PBMC or BM into ⁇ APC.
- MM malignant plasma cells
- B-ALL- ⁇ APC activate NeoT B-ALL for autologous cell therapy for B-ALL [0562] PBMC and BM samples from a newly diagnosed B-ALL patient were used to prepare ALL- ⁇ APC, which were used to “call” (selection and activation) NeoT B-ALL from PBL and TILs (in this case: T cells in BM).
- NeoT B-ALL were tested in vitro for cytotoxicity against B- ALL cells and in vivo by adoptive cell therapy (ACT) to PDXs (i.e., patient-derived xenografts) of B-ALL cancer established in immunocompromised NSG mice.
- FIGs.11A-13F show details of the study.
- FIG.11A shows analyses of PBMC and BM mononuclear cells (BMMC) from a patient with newly diagnosed B-ALL (top panel) and PBMC from a healthy donor (bottom panel). Malignant B lymphoblasts (B-ALL cells) occupied 67.3% of total cells in PBMC and 81.8% of BMMC.
- BMMC BM mononuclear cells
- FIGs.11B-11E show the HC-activator treatment (optionally additionally with optional APC refinement agent treatment) differentiated malignant B lymphoblasts into ALL- ⁇ APC.
- FIG.11B shows the experimental scheme.
- PBMC and BM mononuclear cells (BMMC) from B-ALL were separated by CD3 selection into CD3 + T cell population (PBL and TILs) and CD3- cells that were primarily malignant B lymphoblasts (> 80%).
- the malignant B lymphoblasts were treated with HC-activator and optionally also with the optional APC refinement agent to differentiate ALL- ⁇ APC, which were then used for antigen presentation and activation of NeoT that were derived from PBL and TILs. NeoT were tested in vitro and in vivo for B-ALL cancer cell elimination.
- FIG.11C shows analyses of malignant B lymphoblasts prior to, and after, the HC-activator and optional APC refinement agent treatment and differentiation of the cells into ALL- ⁇ APC.
- ALL- ⁇ APC featured increased expression of MHC-I, MHC-II, CD80, CD86, and CD40.
- FIG.11D shows that co- incubation of ALL- ⁇ APC and PBL or TILs led to ALL- ⁇ APC antigen specific engagement with cognate NeoT (light gray), resulting in NeoT activation and expansion (right).
- FIG.11E shows that two rounds of ALL- ⁇ APC-mediated antigen-specific activation of NeoT led to large expansion of NeoT cells, within which effector memory T cells (TEM) occupied high frequencies.
- TEM effector memory T cells
- Malignant B lymphoblasts in BM prior to and after incubation with T cells were analyzed by flow cytometry.
- Data show that NeoT B-ALL mediated potent killing towards malignant B lymphoblasts.
- FIG.12B shows the results of IFN ⁇ ELISpot indicating NeoT B-ALL released IFN ⁇ during activation and killing of malignant B lymphoblasts.
- FIG.12C shows still-frames from time-lapse video clips of the NeoT B-ALL killing of malignant B lymphoblasts.
- FIGs.13A-13F show in vivo testing of NeoT B-ALL by adoptive cell therapy (ACT) into patient-derived xenograft (PDX) models of B-ALL.
- ACT adoptive cell therapy
- PDX patient-derived xenograft
- FIG.13A the BM and PBMC mixture from the B-ALL patient was engrafted into NSG mice to establish PDX models.
- NeoT B-ALL ACT was administered (i.v.) in a dose-escalation manner. Control experiments were done by giving non-specific T cells of the same number.
- NeoT B-ALL showed dose-dependent effects on cancer elimination.
- FIG.13D shows the persistence of NeoT B-ALL as detected in peripheral blood of recipient mice post- ACT.
- FIG.13E shows that NeoT B-ALL displayed activation characteristics with increased 4- 1BB and CD25 expression in recipient mice, indicating cytotoxic activities against B-ALL malignant cells.
- FIG.13F shows that NeoT B-ALL cytotoxicity in recipient mice was associated with cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- AML- ⁇ APC activate NeoT AML for autologous cell therapy for AML A.
- Producing AML- ⁇ APC from malignant myelogenous leukocytes [0569] PBMC samples from patients suffering different stages of acute myelogenous leukemia (AML) were obtained (Discovery Life Sciences). These samples were analyzed for the presence of malignant myelogenous leukocytes (AML cells), cMO, and T cells (FIGs. 14A-15A). In all samples, malignant myelogenous leukocytes occupied the majority of PBMC cells.
- NeoT AML Malignant myelogenous leukocytes were treated by HC-activator and optional APC refinement agent to differentiate into AML- ⁇ APC, which were then used to activate NeoT ny-2770598 Attorney Docket No.24516-20006.40 AML. After testing in vitro for the NeoT AML killing of cancer cells, NeoT AML were used to treat AML established in NSG mice (i.e., an AML PDX model) through adoptive cell therapy (ACT). See FIGs.14A-16B. [0571] FIGs.14A-14B show that malignant myeloblasts were either CD45- or CD45+, dependent on the stage of AML. AML patients were associated with reduction of CD8 T cells in PBMC.
- FIGs.15A-15B show analyses of cell compositions in PBMC obtained from AML patients and compared to healthy donors.
- FIG.15A is a table that summarizes representative AML studies.
- FIG.15B shows the differentiation of AML malignant myeloblasts into ⁇ APC, termed AML- ⁇ APC.
- PBMC were passed through anti-CD14-coated beads to collect cMo (CD14+), followed by positive selection with anti-CD33-coated beads to enrich AML malignant myeloblasts (CD33+CD14-).
- cMo CD14+
- anti-CD33-coated beads to enrich AML malignant myeloblasts
- bead-absorbed cMo and malignant myeloblasts were released from their respective beads and collected separately.
- T cells (PBL) in PBMC were collected by positive selection with anti-CD3 beads, and these T cells were used for antigen-specific activation of NeoT.
- cMo and malignant myeloblasts were treated with HC-activator (2d) and optional APC refinement agent (1d) to induce differentiation into ⁇ APC.
- FIG.15B shows malignant myeloblasts following HC- activator/optional APC refinement agent treatment differentiating into phenotypic antigen presenting cells, termed AML- ⁇ APC, which displayed increased expression of MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86.
- AML- ⁇ APC phenotypic antigen presenting cells
- AML- ⁇ APC activate NeoT AML for ACT to treat AML
- Malignant myelogenous leukocytes-derived AML- ⁇ APC were used to activate NeoT AML from PBL and/or TILs (in this case: T cells in BM). Expanded NeoT AML were tested in vitro for killing of AML malignant cells and in vivo by adoptive cell therapy (ACT) for treating AML PDXs established in NSG mice.
- ACT adoptive cell therapy
- FIGs.16A-16B illustrates that AML- ⁇ APC mediated antigen presentation to activate NeoT AML , leading to NeoT AML clonal expansion.
- FIG.16A depicts the experimental procedure. AML- ⁇ APC were differentiated from malignant myeloblasts by the HC-activator ny-2770598 Attorney Docket No.24516-20006.40 (optionally further with the optional APC refinement reagent) treatment. PBL comprising CD4 and CD8 T cells were added into differentiated AML- ⁇ APC cultures.
- FIG.16B shows images of NeoT engaging with AML- ⁇ APC. Rapid clonal expansion in 72h and increases in CD4 and CD8 T cells are also shown (right panel).
- FIGs.17A-17C show the results of an in vitro assay of NeoT AML killing towards AML malignant cells.
- FIG.17A shows that NeoT AML were incubated with AML malignant myeloblasts at 1:1 ratio for 6 hours, resulting in rapid killing that reduced malignant myeloblasts from 45.0% to 3.41% in the co-culture.
- FIG.17B shows NeoT AML killing efficiency measured by time course and varied ratios of T cells to AML malignant myeloblasts. Parallel experiments were performed using non-specific T cells (control).
- FIG.17C shows that NeoT AML targeting malignant myeloblasts were associated with increased expression of CD8 T cell activation markers: IFN ⁇ , CD107a, 4-1BB, and CD25.
- the control non-specific T cells which exhibited no/minimal killing towards malignant myeloblasts, did not show increased activation markers during co-incubation with malignant myeloblasts.
- ACT NeoT AML adoptive cell therapy
- FIG.18A illustrates the experimental steps for establishing AML PDXs for the subsequent testing of NeoT AML by ACT.
- Malignant myeloblasts (1 ⁇ 10 6 per mouse) from AML patients were implanted into NSG mice via i.v. injection.
- AML cells were detectable in the PBMC of xenografted mice (F1), mice were euthanized and BM single cells and PBMC were harvested, combined, and engrafted (i.v., 5 ⁇ 10 6 per mouse) into the next generation recipient mice (2nd generation, F2).
- the 3 rd generation of a cohort of at least 10 mice were generated for testing NeoT AML by ACT, as well as other treatments.
- FIG.18B showed the detection of AML malignant myeloblasts in PBMC, bone marrow, spleen, and extramedullary tissues of xenografted mice.
- FIG.18C showed that the F2 generation of AML PDXs displayed extramedullary disease.
- ny-2770598 Attorney Docket No.24516-20006.40
- FIGs.19A-19E illustrated that NeoT AML adoptive cell therapy (ACT) effectively treated AML patient-derived xenografts (PDXs).
- FIG.19A shows the experimental design: AML PDXs of the 3 rd (F3) generation were generated by i.v.
- AML diseases were generally established in F3 recipients in 2-3 weeks, manifesting malignancies in BM (> 30% cells of BM being malignant myeloblasts) and then in PBMC. BM and/or PBMC were sampled to confirm AML establishment prior to ACT with NeoT AML .
- Two cohorts were established: cohort #1 with ACT started on day 14 when BM manifested AML; cohort #2 with ACT started on day 26 when AML from BM spread to PBMC. Two rounds of ACT treatments were given with either NeoT AML or non-specific T cells (ctl.).
- FIG.19B shows overall survival of PDX mice without or with treatment of NeoT AML .
- FIG.19C shows results of cohort #1 wherein mice manifested BM disease (d14 and d18) and were treated with NeoT AML in a dose-escalation manner.
- FIG. 19D shows the results of cohort #2 wherein mice with late-stage AML manifesting in both BM and PBMC (d26) were treated with NeoT AML .
- FIG.19E shows sample PBMC analyses in cohort #2 prior to and after NeoT AML treatment. AML manifestation in PBMC was diminished after the 2 nd round of NeoT AML ACT, and NeoT AML cells displayed persistence in the circulation after AML elimination. D.
- FIGs.20A-20E show that prophylactic anti-TNF ⁇ neutralizing antibody administration ameliorated CRS without compromising NeoT AML ACT efficacy against AML.
- FIG.20A-20B shows that prophylactic anti-TNF ⁇ neutralizing antibody administration did not compromise NeoT AML ACT efficacy towards AML.
- AML PDXs (F3) with PBMC manifestation were treated with 5 ⁇ 10 6 NeoT AML without or with prophylactic administration (3 hours earlier) of anti-TNF ⁇ neutralizing Ab.
- FIG.20C shows that prophylactic anti-TNF ⁇ neutralizing antibody administration did not inhibit NeoT AML activation and production of IFN ⁇ associated with targeting malignant myeloblasts in vivo by ny-2770598 Attorney Docket No.24516-20006.40 ACT.
- NeoT AML producing human cytokines were detected in the plasma of recipient PDX mice 24 hours post NeoT AML ACT.
- FIGs.20D-20E show that anti-TNF ⁇ neutralization did not damage AML- ⁇ APC-mediated antigen presentation that activated NeoT AML (FIG.20D), nor did it affect NeoT AML killing of AML malignant cells (FIG.20E). ny-2770598
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Biomedical Technology (AREA)
- Chemical & Material Sciences (AREA)
- Hematology (AREA)
- Wood Science & Technology (AREA)
- Zoology (AREA)
- Genetics & Genomics (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Immunology (AREA)
- Biotechnology (AREA)
- Oncology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Cell Biology (AREA)
- Pharmacology & Pharmacy (AREA)
- General Chemical & Material Sciences (AREA)
- Microbiology (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
The present application provides compositions and methods for producing antigen presenting cells (APCs) from cancer cells (e.g., hematological cancer cells) that involve one or more agents selected from a STAT3 activator (e.g., IL-10), IFNγ receptor activator (e.g., IFNγ ), TNFα receptor activator (e.g., TNFα), IL-4 receptor activator (e.g., IL-4), GM-CSF receptor activator (e.g., GM-CSF), and/or IL-6 receptor activator (e.g., IL-6). APCs produced accordingly are also provided, as well as methods of activating immune cells (e.g., T cells) via co-culturing with APCs. The activated immune cell compositions and the methods of treatments that involve the activated immune cells are also provided.
Description
Attorney Docket No.24516-20006.40 COMPOSITIONS AND METHODS FOR ACTIVATING IMMUNE CELLS CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of, and priority to U.S. Provisional Application No.63/519,974, filed August 16, 2023, the content of which is incorporated herein by reference in its entirety. FIELD OF THE INVENTION [0002] The present invention relates to compositions and methods for: a) promoting the differentiation and/or maturation of antigen presenting cells (APCs) derived from cancer cells (such as malignant myelogenous cells or B lymphoblasts/lymphocytic cells from cancer patients), and b) activating immune cells (such as T cells). BACKGROUND OF THE INVENTION [0003] Hematologic malignancies, also known as blood cancer, are the fifth most common cancer group, with diseases including acute and chronic lymphoblastic leukemia (ALL and CLL), acute and chronic myelogenous leukemia (AML and CML), myelodysplastic syndrome (MDS), Hodgkin’s and non-Hodgkin’s lymphoma, multiple myeloma (MM), etc. Among different types of blood cancer, non-Hodgkin’s lymphoma (NHL) is the most common disease (> 40%), followed by leukemia (31%), and then MM (18%). Characterization of cell types shows that B cell-lineage malignancies such as B-ALL, CLL, NHL, and MM occupy > 50% of all cases, with the rest being chiefly myelogenous leukemia (AML and CML). Depending on where the cancer originates, malignant cells generally manifest in peripheral blood (leukemia), bone marrow (leukemia and MM), lymph nodes (lymphoma), and spleen, as well as disseminated areas such as the brain, spinal cord, and peritoneal cavity. [0004] Traditionally, the treatment of hematological malignancies mainly includes chemotherapy, radiotherapy, and hematopoietic stem cell transplantation (HSCT). However, with advances in tumor immunology, immune targeted therapies such as monoclonal antibodies, bispecific antibodies, antibody-drug conjugates, and chimeric antigen receptor T (CAR-T) cell therapies have opened a new avenue for the treatment of these malignancies. In particular, CAR-T cell therapy has revolutionized the treatment of hematological malignancies and achieved unprecedented responses in recent years. However, the rapid commercialization of CAR-T cell therapy poses a significant challenge for its management, ny-2770598
Attorney Docket No.24516-20006.40 such as managing the toxicities associated with CAR-T therapy and patient relapse after receiving CAR T-cell therapy. See e.g., Front Immunol.2022; 13: 927153. [0005] The disclosures of all publications, patents, patent applications and published patent applications referred to herein are hereby incorporated herein by reference in their entirety. BRIEF SUMMARY OF THE INVENTION [0006] The present application in one aspect provides a method of producing a population of antigen presenting cells (“APCs”) presenting an antigen associated with a hematological cancer (“HC-APC”), comprising: contacting hematological cancer cells obtained from an individual having the hematological cancer with one or more survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an interleukin 4 receptor (IL-4R) activator, and 4) an interferon ^ (IFN^) receptor (IFNGR) activator, thereby producing the population of HC-APCs. [0007] The present application in another aspect provides a method of producing activated immune cells, comprising: a) producing a population of antigen presenting cells (“APCs”) presenting an antigen associated with a hematological cancer (“HC-APC”) according to the method described above, and b) contacting the HC-APCs with immune cells, thereby producing activated immune cells. [0008] The present application in another aspect provides a method of producing activated immune cells, comprising contacting immune cells with a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), thereby generating activated immune cells; wherein the HC-APCs are derived from hematological cancer cells after having been contacted with one or more survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an interleukin 4 (IL-4R) activator, and 4) an interferon ^ (IFN^) receptor (IFNGR) activator. [0009] In some embodiments according to any of the methods described above, the STAT3 activator is selected from the group consisting of: a small molecule STAT3 activator, an IL- 10, an IL-10 family member, an IL-10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, and a small molecule activator of IL-10R. In some embodiments, the STAT3 activator is selected from the group consisting of: an IL-10 family cytokine, an IL-12 family cytokine, an IL-6 family cytokine, IL-7 family cytokine, IL-9 family cytokine, IL-15 ny-2770598
Attorney Docket No.24516-20006.40 family cytokine, IL-21 family cytokine, and G-CSF. In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-12, IL-23, IL-6, colivelin TFA, Garcinone D, G-CSF, IL-7, IL-9, IL-15, and IL-21. In some embodiments, the STAT3 activator is selected from the group consisting of: IL-10, IL-22, IL- 19, IL-20, IL-24, IL-12, IL-23, Colivelin TFA, and Garcinone D. [0010] In some embodiments according to any of the methods described above, the one or more of S/D/M factors comprise an interleukin 4 receptor (IL-4R) activator. In some embodiments, the IL-4R activator is selected from the group consisting of IL-4, an IL-4R agonist antibody, and a small molecule activator of IL-4. In some embodiments, the IL-4R activator is IL-4. [0011] In some embodiments according to any of the methods described above, the one or more of S/D/M factors comprise a TNFR activator. In some embodiments, the TNFR activator is selected from the group consisting of TNF^, a TNFR agonist antibody, and a small molecule activator of TNFR. In some embodiments, the TNFR activator is TNF^. [0012] In some embodiments according to any of the methods described above, the one or more of S/D/M factors comprise an IFNGR activator. In some embodiments, the IFNGR activator is selected from the group consisting of IFN^, an IFNGR agonist antibody, and a small molecule activator of IFNGR. In some embodiments, the IFNGR activator is IFN^. [0013] In some embodiments according to any of the methods described above, the one or more of S/D/M factors are present in a single composition. [0014] In some embodiments according to any of the methods described above, the one or more of S/D/M factors comprise two or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator. [0015] In some embodiments according to any of the methods described above, the one or more of S/D/M factors comprise three or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator. [0016] In some embodiments, the one or more of S/D/M factors comprise: 1) a STAT3 activator, 2) a TNFR activator, and 3) an IFNGR activator. In some embodiments, the one or more of S/D/M factors comprise an IL-10 family cytokine, TNF^, and IFN^. In some embodiments, the one or more of S/D/M factors comprise: 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator. In some embodiments, the one ny-2770598
Attorney Docket No.24516-20006.40 or more of S/D/M factors comprise an IL-10 family cytokine, TNF^, IL-4, and IFN^. Exemplary IL-10 family cytokines include IL-10, IL-22, IL-19, IL-24, IL-20, or IL-26. [0017] In some embodiments according to any of the methods described above, the one or more of the S/D/M factors further comprise an IL-6 receptor (IL-6R) activator and/or a GM- CSF receptor (GM-CSFR) activator. In some embodiments, the IL-6R activator is selected from the group consisting of IL-6, an IL-6R agonist antibody, and a small molecule activator of IL-6R. In some embodiments, the GM-CSFR activator is selected from the group consisting of GM-CSF, a GM-CSFR agonist antibody, and a small molecule activator of GM- CSF. In some embodiments, the IL-6R activator is IL-6, and the GM-CSFR activator is GM- CSF. [0018] In some embodiments according to any of the methods described above, the hematological cancer cells are comprised in a cell mixture comprising monocytes from the individual. [0019] In some embodiments according to any of the methods described above, prior to contacting the HC-APCs with immune cells, the method further comprises contacting the HC-APCs with one or more refinement factors selected from the group consisting of type-I interferon, IFN^, TNF^, a TLR ligand, CD40L or a CD40-ligating antibody, an anti-PD-L1 antibody, and TPI-1. In some embodiments, the type-I interferon comprises IFN^ and/or IFN^. In some embodiments, the TLR ligand is R848, poly IC, CpG, or LPS. In some embodiments, the TLR ligand comprises R848 and poly IC. In some embodiments, the one or more refinement factors comprise IFN^, IFN^, and TNF^. In some embodiments, the one or more refinement factors further comprise at least two agents selected from the group consisting of poly IC, CpG, CD40L, R848, and an anti-PD-L1 antibody. In some embodiments, the one or more refinement factors further comprises a SHP-1 inhibitor. In some embodiments, the SHP-1 inhibitor is TPI-1. In some embodiments, the HC-APCs are cultured with the one or more refinement factors for about 1-4 days. [0020] In some embodiments according to any of the methods described above, the method further comprises administering into the individual immune cells activated by contacting the immune cells with the population of HC-APCs. In some embodiments, the method further comprises administering into a different individual than the individual having the hematological cancer immune cells activated by contacting the immune cells with the population of HC-APCs. In some embodiments, the immune cells and the hematological ny-2770598
Attorney Docket No.24516-20006.40 cancer cells are from the same individual. In some embodiments, the immune cells and the hematological cancer cells are from different individuals. In some embodiments, the immune cells are administered to the individual from whom they are obtained. In some embodiments, the immune cells are administered to a different individual than the individual from whom they are obtained. [0021] In some embodiments according to any of the methods described above, the immune cells are selected from the group consisting of peripheral blood mononuclear cells (“PBMCs”), cells derived from bone marrow biopsy, cells derived from lymphoma biopsy, tumor infiltrating T cells (TIL), and T cells. In some embodiments, the immune cells are T cells. In some embodiments, the T cells are CD8 T cells and/or CD4 T cells. [0022] In some embodiments according to any of the methods described above, the hematological cancer cells are obtained from PBMCs, enriched leukemia cells, a bone marrow biopsy, or a lymphoma biopsy. [0023] In some embodiments according to any of the methods described above, the hematological cancer cells are cultured with the one or more of S/D/M factors for at least 2 days. [0024] In some embodiments according to any of the methods described above, the immune cells have been enriched prior to contacting the HC-APCs. [0025] In some embodiments according to any of the methods described above, the hematological cancer cells have been enriched prior to being cultured with the one or more of S/D/M factors. [0026] In some embodiments according to any of the methods described above, the hematological cancer is a myeloid leukemia. In some embodiments, the hematological cancer is acute myeloid leukemia (AML) or chronic myeloid leukemia (CML). In some embodiments, the hematological cancer cells are any of M0-M5 myeloblasts. In some embodiments, the hematological cancer cells have been enriched prior to being cultured with the one or more of S/D/M factors in the presence of an anti-CD11b antibody. [0027] In some embodiments according to any of the methods described above, the hematological cancer is a B-cell lymphoma or B-cell leukemia. In some embodiments, the B- cell lymphoma or B-cell leukemia is selected from the group consisting of B-cell acute lymphoblastic leukemia (B-ALL), chronic lymphocytic leukemia (CLL), and non-Hodgkin’s lymphoma (NHL). In some embodiments, the hematological cancer cells have been enriched ny-2770598
Attorney Docket No.24516-20006.40 prior to being cultured with the one or more of S/D/M factors in the presence of an anti-CD19 antibody or an anti-CD20 antibody. [0028] In some embodiments according to any of the methods described above, the method further comprises expanding the immune cells by contacting the immune cells with the HC- APCs for at least two rounds, at least three rounds, or at least four rounds. [0029] The present application in another aspect provides a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”) obtained by any of the methods described above. [0030] The present application in another aspect provides a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APCs”), wherein the HC-APCs are derived from hematological cancer cells, and wherein the HC-APCs express increased levels of MHC-1, MHC-II, CD40, CD80 and/or CD86 compared to the hematological cancer cells prior to the treatment. In some embodiments, the HC-APCs comprise one or more exogenous antigens. In some embodiments, the hematological cancer cells are primary cells from an individual having the hematological cancer. In some embodiments, the hematological cancer is a myeloid leukemia. In some embodiments, the hematological cancer is acute myeloid leukemia (AML) or chronic myeloid leukemia (CML). In some embodiments, the hematological cancer cells are any of M0-M5 myeloblasts. In some embodiments, the hematological cancer is a B-cell lymphoma or B-cell leukemia. In some embodiments, the B-cell lymphoma or B-cell leukemia is selected from the group consisting of B-cell acute lymphoblastic leukemia (B-ALL), chronic lymphocytic leukemia (CLL), and non-Hodgkin’s lymphoma (NHL). [0031] The present application in another aspect provides a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APCs”), prepared by: a) obtaining hematological cancer cells from an individual having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an interferon ^ (IFN^) receptor (IFNGR) activator, and 3) an IL-4 receptor (IL- 4R) activator, thereby producing the population of HC-APCs. [0032] The present application in another aspect provides a population of activated immune cells obtained by any of the methods described above. ny-2770598
Attorney Docket No.24516-20006.40 [0033] The present application in another aspect provides a method of treating a hematological cancer in an individual, comprising administering to the individual an effective amount of any of the activated immune cells described above. [0034] The present application in another aspect provides a method of treating a hematological cancer in an individual, comprising: a) contacting hematological cancer cells and/or monocytes with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an interferon ^ (IFN^) receptor (IFNGR) activator, and 4) an IL-4 receptor (IL-4R) activator, thereby producing a population of antigen presenting cells (“APCs”), b) contacting the APCs with a population of immune cells isolated from the individual, thereby producing activated immune cells, and c) administering the activated immune cells into the individual. In some embodiments, the APCs are further loaded with one or more exogenous antigen. In some embodiments, the hematological cancer cells and/or monocytes are obtained from the individual. In some embodiments, the hematological cancer cells and monocytes are comprised in a mixture when cultured with the one or more of S/D/M factors. BRIEF DESCRIPTION OF THE DRAWINGS [0035] FIG.1 depicts ^APC derived from cancer monocytes (cMO or cMo) or hematological cancer cells for the activation of cancer antigen-specific T cells. [0036] FIG.2 depicts the platform process for generating cancer antigen-specific T cells (e.g., NeoT) to treat hematological cancer(s). [0037] FIG.3 depicts the production process of ^APC from cancer patient monocytes (cMo) and various hematological cancer cells. [0038] FIG.4 shows a table that summarizes results from representative studies that differentiate ^APC from cMO or cancer cells from various sources. [0039] FIGs.5A-5C show the phenotype of B-ALL cells from Patient #1 before and after differentiation into ^APC. FIG.5A provides the FACS analysis of PBMCs to quantify immune cell lineage percentages. FIG.5B displays histogram results from FACS analysis of the B-ALL cell phenotype before treatment with a hematological cancer cell activator (i.e., HC-activator) and optionally APC refinement reagent, and after differentiation into ^APCs by treatment with HC-activator and optionally APC refinement reagent. Phenotypic markers ny-2770598
Attorney Docket No.24516-20006.40 of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86. FIG.5C shows a representative field using light microscopy for B-ALL cells before and after treatment with HC-activator and optionally APC refinement reagent. PBMC, peripheral blood mononuclear cells; cMO, cancer monocyte; SSC, side scatter. [0040] FIGs.6A-6B show the phenotype of B-ALL cells from Patient #2 before and after differentiation into ^APC. FIG.6A provides the FACS analysis of PBMCs to quantify immune cell lineage percentages. FIG.6B displays histogram results from FACS analysis of the B-ALL cell phenotype before treatment with HC-activator and optionally APC refinement reagent, and after differentiation into ^APCs by treatment with HC-activator and optionally APC refinement reagent. Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86. PBMC, peripheral blood mononuclear cells; cMO, cancer monocyte; SSC, side scatter. [0041] FIGs.7A-7B show the phenotype of B-ALL cells from Patient #3 before and after differentiation into ^APC. FIG.7A provides the FACS analysis of PBMCs to quantify immune cell lineage percentages. FIG.7B displays histogram results from FACS analysis of the B-ALL cell phenotype before treatment with HC-activator and optionally APC refinement reagent, and after differentiation into ^APCs by treatment with HC-activator and optionally APC refinement reagent. Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86. PBMC, peripheral blood mononuclear cells; cMO, cancer monocyte; SSC, side scatter. [0042] FIGs.8A-8B show the phenotype of CLL cells from Patient #4 before and after differentiation into ^APC. FIG.8A provides the FACS analysis of PBMCs to quantify immune cell lineage percentages. FIG.8B displays histogram results from FACS analysis of the CLL cell phenotype before treatment with HC-activator and optionally APC refinement reagent, and after differentiation into ^APCs by treatment with HC-activator and optionally APC refinement reagent. Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86. PBMC, peripheral blood mononuclear cells; cMO, cancer monocyte; SSC, side scatter. [0043] FIGs.9A-9B show the phenotype of non-Hodgkin’s lymphoma (NHL) cells from Patient #5 before and after differentiation into ^APC. FIG.9A provides the FACS analysis of dissociated tumor cells to quantify immune cell lineage percentages. FIG.9B displays histogram results from FACS analysis of the NHL cell phenotype before treatment with HC- ny-2770598
Attorney Docket No.24516-20006.40 activator and optionally APC refinement reagent, and after differentiation into ^APCs by treatment with HC-activator and optionally APC refinement reagent. Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86. PBMC, peripheral blood mononuclear cells; cMO, cancer monocyte; SSC, side scatter; DTC, dissociated tumor cells. [0044] FIGs.10A-10B show the phenotype of multiple myeloma (MM) cells from Patient #6 before and after differentiation into ^APC. FIG.10A provides the FACS analysis of PBMCs and bone marrow cells (BM) to quantify immune cell lineage percentages. FIG.10B displays histogram results from FACS analysis of the MM cell phenotype before treatment with HC- activator and optionally APC refinement reagent, and after differentiation into ^APCs by treatment with HC-activator and optionally APC refinement reagent. Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86. PBMC, peripheral blood mononuclear cells; BM, bone marrow cells; cMO, cancer monocyte; SSC, side scatter. [0045] FIGs.11A-11E depict the in vitro functional activity of ^APC(B-ALL) to prime and activate NeoT cells. FIG.11A provides representative plots and a graphical representation of the percent of various cell populations found within the harvested PBMC or BM sample taken from a human B-ALL cancer patient as compared to a healthy donor. FIG.11B provides a schematic overview of the experimental design for preparing ^APC(B-ALL) and co-culturing with NeoT isolated from the same patient for further downstream analyses (e.g., in vivo PDX cancer model and in vitro cancer cell killing assay). FIG.11C displays histogram results from FACS analysis of the B-ALL cell phenotype before treatment with HC-activator and optionally APC refinement reagent (“malignant B”), and after differentiation into ^APCs by treatment with HC-activator and optionally APC refinement reagent (ALL-^APC). Phenotypic markers of cells with antigen presentation capabilities include MHC-I, MHC-II, CD40, CD80, and CD86. FIG.11D shows still images from a time lapse experiment wherein NeoT cells that were pre-emptively loaded with CFSE prior to co- culture were incubated with B-ALL cells in vitro for up to 10 days, and images of the cell interactions were taken using fluorescence microscopy and light microscopy. FIG.11E provides the quantification of NeoT cell expansion (T cell number over time) after two rounds of co-culturing with the ^APC(B-ALL) cells: from day 0 to day 10 (round 1) and from day 10 to day 24 (round 2). Further provided are FACS plots of expanded effector CD4+ and CD8+ T cells as analyzed for CD45RA and CCR7 effector markers. PBL, peripheral blood ny-2770598
Attorney Docket No.24516-20006.40 lymphocytes; TIL, tumor infiltrating lymphocytes; NeoT(B-ALL), ^APC(B-ALL)-activated NeoT cells; PDX, patient-derived xenograft; ctl, control; CFSE, CarboxyFluoroscein Succinimidyl Ester. [0046] FIGs.12A-12C depict in vitro cancer cell killing by NeoT cells that were primed using ^APC(B-ALL) cells. FIG.12A provides representative plots and a graphical representation of the percent of B-ALL cells found within the harvested BM sample taken from a human B-ALL cancer patient before and after incubation with either non-specifically activated T cells (using anti-CD3 and anti-CD28 antibodies) or NeoT(B-ALL) cells at a 1:1 ratio of BM:T cells. FIG.12B shows the resulting IFN^ production using ELISpot from BM cells alone, BM cells co-cultured 1:1 with non-specifically activated T cells, and BM cells co- cultured 1:1 with NeoT(B-ALL) activated cells (representative field image: left panel; graphical representation of the average IFN^ levels: right panel). FIG.12C provides still images of time-lapse videos of the NeoT(B-ALL) cells homing to and killing B-ALL cells in vitro. BM, bone marrow; NeoT(B-ALL), ^APC(B-ALL)-activated NeoT cells; Non-specific T, anti-CD3/anti-CD28 antibody-activated T cells; h, hour(s). [0047] FIGs.13A-13F show a B-ALL PDX murine model to assess the efficacy of NeoT(B- ALL) to clear tumors in vivo. FIG.13A provides a schematic overview of the PDX model experimental design from B-ALL engraftment into NSG mice, then adoptive transfer of NeoT(B-ALL) cells as a dose escalation series, and then analyzing the mice for markers of tumor clearance. FIG.13B shows the percent of human B-ALL cells present in murine PBMC at days 0, 6, and 14 after intravenous ACT injection compared to non-specific T cell- injected control mice. FIG.13C shows the percent of human B-ALL cells over time post- ACT in peripheral white blood cell samples and overall survival curves for each group tested. FIG.13D depicts the percent of NeoT(B-ALL) cells present in peripheral white blood cell samples over time post-ACT. FIG.13E demonstrates the level of markers of NeoT(B-ALL) cell activation using markers 4-1BB and CD25 to demonstrate cell activity. FIG.13F provides a graphical representation of the mean serum level of pro-inflammatory cytokines two days after intravenous ACT injection of NeoT(B-ALL) cells: IFN^, TNF^, IL-4, IL-6, CCL2, CCL20, and CXCL8. PDX, patient-derived xenograft; i.v., intravenous; D, day(s); ACT, adoptive cellular transfer; NeoT(B-ALL), ^APC(B-ALL)-activated NeoT cells; CR, complete response; BM, bone marrow; PBMC, peripheral blood mononuclear cells; hMHC I, human major histocompatibility complex 1; hCD19, human CD19; Non-specific T, anti- ny-2770598
Attorney Docket No.24516-20006.40 CD3/anti-CD28 antibody-activated T cells; 2x, two times or twice; WBC, white blood cells; MFI, mean fluorescence intensity. [0048] FIGs.14A-14B show FACS analyses of cancer cells, cancer monocytes, B cells, CD4+ T cells, and CD8+ T cells in AML patients (FIG.14A) and healthy donors (FIG. 14B). M5, stage M5 AML; M2, stage M2 AML; nd, not defined/determined; M4, stage M4 AML; PBMC, peripheral blood mononuclear cell(s); AML, acute myeloid leukemia; cMo, cancer monocyte. [0049] FIGs.15A-15B show the capacity of ^APC(AML) cells to present antigens before vs. after treatment with HC-activator and optionally APC refinement reagent. FIG.15A shows a summary of AML studies. FIG.15B shows histogram plots of phenotypic markers of antigen presentation on AML cells before treatment with HC-activator and optionally APC refinement reagent, and on ^APC(AML) cells after treatment with HC-activator and optionally APC refinement reagent: MHC-I, MHC-II, CD80, CD86, and CD40, wherein the increased level of each marker is indicative of increased antigen presentation capacity of the cells. AML cells were isolated from human patients with different stages of AML. M5, stage M5 AML; M2, stage M2 AML; nd, not defined/determined; M4, stage M4 AML; PBMC, peripheral blood mononuclear cell(s); AML, acute myeloid leukemia; cMo, cancer monocyte; ctl, control. [0050] FIGs.16A-16B show in vitro analyses of NeoT(AML) functional activity. FIG.16A provides a schematic overview of the in vitro experimental design for co-culturing ^APC(AML) cells with PBLs or TILs isolated from the donor cancer patient and expanded in culture. FIG.16B shows light microscopy field images of NeoT(AML) cells engaging with ^APC(AML) cells in vitro and clonally expanding over 72hrs, including FACS analysis of expanded cells to quantify the percentage of CD8+ and CD4+ clonally expanded T cells. PBL, peripheral blood lymphocytes; TIL, tumor infiltrating lymphocytes; NeoT(AML), ^APC(AML)-activated NeoT cells; Diff., differentiated. [0051] FIGs.17A-17C demonstrate the in vitro tumor killing activity of NeoT(AML) cells. FIG.17A shows the percentage of CD8 cells (representative of NeoT(AML) cells) to CFSE- loaded AML cells at 0hr and 6hr co-cultured at a 1:1 ratio. FIG.17B displays the number of AML cells in vitro over time at 0hr, 3hr, 6hr, 12hr, and 24hr incubations at 1:1 and 3:1 ratios of NeoT(AML) cells or Non-specific T cells to AML cells. FIG.17C shows FACS analysis of markers of CD8+ T cell activation in Non-specific T cells and in NeoT(AML) cells co- ny-2770598
Attorney Docket No.24516-20006.40 cultured with AML cells: IFN^, CD107a, 4-1BB, and CD25. NeoT(AML), ^APC(AML)- activated NeoT cells; Non-specific T, anti-CD3/anti-CD28 antibody-activated T cells; h or hr, hour; CFSE, CarboxyFluoroscein Succinimidyl Ester. [0052] FIGs.18A-18C show the successful generation of an AML PDX murine model for downstream use in the assessment of NeoT(AML) cell efficacy to clear tumors in vivo. FIG. 18A provides a schematic overview of the PDX model experimental design from AML engraftment into NSG mice over three generations (F0, F1, F2, and F3), to adoptive transfer of NeoT(B-ALL) cells as a dose escalation series for downstream analysis of the markers of tumor clearance. FIG.18B depicts the level of human AML cell engraftment in PBMC, BM, spleen, and extramedullary sites to confirm engraftment and disease progression across successive generations. FIG.18C shows the locations of extramedullary sites wherein the AML cells infiltrated, including lymph nodes, spleen, and peritoneal cavity. PBMC, peripheral blood mononuclear cells; BM, bone marrow; PDX, patient-derived xenograft; m, mouse; h, human; i.v., intravenous; ACT, adoptive cellular transfer; NeoT(AML), ^APC(AML)-activated NeoT cells; LN, lymph node(s). [0053] FIGs.19A-19E show the assessment of NeoT(AML) cell efficacy at clearing tumors in vivo using an AML PDX murine model. FIG.19A provides a schematic overview of the AML cell engraftment into F3 generation NSG mice followed by intravenous ACT of NeoT(AML) cells in a dose-escalation series. FIG. 19B provides the overall survival curve up to 180 days for the AML-engrafted mice provided no treatment, non-specific T cells, or NeoT(AML) cells. FIG.19C depicts the percentage of cells in the PBMC that were human AML cells up to 40 days post-ACT in cohort #1, and FIG.19D depicts the same for cohort #2. Cohort #1 included mice that were given ACT upon AML engraftment in the bone marrow, and cohort #2 included mice that were given ACT after AML cells had engrafted and then expanded from the bone marrow to become detectable in the PBMC. When mice reached 40% AML cells, the mice were then humanely euthanized. FIG.19E shows FACS analysis of the percentage of CD8+ T cells and AML cells in PBMC of mice of cohort #2 over time, including showing AML progression and regression as well as NeoT(AML) cell persistence. D, day(s); w, week(s); PBMC, peripheral blood mononuclear cells; BM, bone marrow; i.v., intravenous; ACT, adoptive cellular transfer; NeoT(AML), ^APC(AML)- activated NeoT cells; PDX, patient-derived xenograft; hMHC I, human major histocompatibility complex 1; hCD19, human CD19. ny-2770598
Attorney Docket No.24516-20006.40 [0054] FIGs.20A-20F show the impact of TNF^ neutralization on systemic inflammation and cytokine release syndrome (CRS). FIG.20A depicts the percentage of cells in the PBMC that were human AML cells up to 35 days post-ACT in PDX mice receiving no treatment, NeoT(AML) cells alone, or prophylactic anti-TNF^ neutralizing antibody and NeoT(AML) cells. FIG.20B displays the overall survival curve over 150 days post-ACT of PDX mice receiving no treatment, NeoT(AML) cells alone, or prophylactic anti-TNF^ neutralizing antibody and NeoT(AML) cells. FIG.20C provides the mean serum human cytokine levels produced by NeoT(AML) cells when the mice were either not treated or treated prophylactically with 100µg anti-TNF^ neutralizing antibody: IL-10, IL-6, IFN^, TNF^, IL- 4, and IL-2. FIG.20D provides the mean serum murine cytokine and chemokine levels when the mice were either not treated or treated prophylactically with 100µg anti-TNF^ neutralizing antibody: CCL2, CCL5, CXCL1, CXCL10, IL-12, TNF^, IL-6, IL-10, IL-1^, IFN^, IFN^, and IFN^. FIG.20E shows the level of NeoT(AML) cell activation and expansion/proliferation after co-culturing with ^APC(AML) cells for up to 10 days either without treatment or treated with anti-TNF^ neutralizing antibody. FIG.20F shows the in vitro tumor cell killing activity of AML cells by NeoT(AML) cells co-cultured with ^APC(AML) cells at 3:1 (left panel) or 1:1 (right panel) for up to 10 days either without treatment or treated with anti-TNF^ neutralizing antibody, as represented by the number of AML cells over time. D, day(s); h, hour(s); PBMC, peripheral blood mononuclear cells; i.p., intraperitoneal; i.v., intravenous; ACT, adoptive cellular transfer; NeoT(AML), ^APC(AML)-activated NeoT cells; anti-TNF^, anti-TNF^ neutralizing antibody. DETAILED DESCRIPTION OF THE INVENTION [0055] This application discloses using hematological cancer cell-derived APCs (e.g., CLL- ^APC) and/or monocyte-derived APC (cMo-^APC) to produce autologous cancer neoantigen-specific T cells (NeoT) for adoptive cell therapy (ACT) treating all forms of hematological malignancies, including leukemia, lymphoma, and myeloma. [0056] NeoT are polyclonal CD4 and CD8 T cells that detect malignant cells through the TCR by recognizing neoantigens displayed in MHC molecules and thereby mediating malignant cell-killing. Cancer neoantigens include genetic mutation-derived mutant peptide antigens, virus infection-derived foreign antigens (e.g., HTLV-1 infection-associated T- ALL), and aberrantly expressed peptide antigens. Despite the fact that NeoT cells exist in cancer patients, their numbers are minute, and their functions are suppressed. This is because ny-2770598
Attorney Docket No.24516-20006.40 active cancer conditions are associated with strong immunosuppression that inhibits antigen presentation required for NeoT cell activation and expansion, as well as NeoT cell anticancer activities, thereby minimizing anticancer immunity and supporting cancer progression. [0057] Through vigorous laboratory research, it was discovered that unique reagent combinations, including HC-activators and optional APC refinement agents, could induce the differentiation of monocytes from cancer patients (cMO) into highly effective, professional antigen-presenting cells (APC). These APCs, termed cMO-^APCs, are dissimilar to monocyte-derived dendritic cells and macrophages that are defective under cancer conditions and possess robust capacities to phagocytose cancer antigens and to present immunogenic antigen(s) in order to activate NeoT cells. In the studies described herein, cMO-^APC are successfully differentiated from cMO for all tested cancer types. Following phagocytosis of cancer antigens, cMO-^APCs in all tested cases exhibited an exceptional ability to present immunogenic antigen(s) that induced the activation and expansion of cancer-killing NeoT cells. Thus, the cMO-^APC – NeoT cell system is a powerful platform for developing an immune-cell therapy against all cancers. See e.g., PCT/US2023/017002, which is incorporated herein by its entirety. [0058] Moreover, in hematological malignancies, it was found that the HC-activators are capable of promoting the survival and differentiation of not only cMO but also the differentiation of malignant myelogenous and B cell-lineage malignant leukocytes to become APCs, e.g., CLL-^APC cells. In particular, the HC-activator treatment induced hematological cancer cells to differentiate into cancer-specific-^APCs, for example, from acute and chronic myelogenous leukemia cells (i.e., CLL-^APC, CML-^APC), acute and chronic B lymphoblastic/ lymphocytic leukemia cells (i.e., ALL-^APC and CLL-^APC), and B lineage NHL lymphoma cells (i.e., NHL-^APC). Different from cMO-^APCs, which require the phagocytosis of cancer antigens for antigen presentation to T cells, CLL-^APCs derived from malignant leukocytes directly presented their inherent neoantigens associated with genetic mutations, without the need to obtain cancer antigens (FIG.1). [0059] Using hematological cancer cell-derived APCs (HC-^APCs) is also advantageous when patients display diminished levels of cMOs, as observed in patients with many hematological malignancies. Based on these findings, an ex vivo patient-oriented NeoT cell production platform has been developed that utilizes ^APCs derived from malignant leukocytes to activate and expand NeoT cells from peripheral blood (PBL) and/or tumor- infiltrating lymphocytes (TILs). See, e.g., FIG.2. This technology platform for the first time ny-2770598
Attorney Docket No.24516-20006.40 enables large production of NeoT cells for adoptive cell therapy to treat all types of hematological cancers. [0060] Accordingly, the present application provides novel compositions and agents that convert cancer cells (e.g., B-ALL, CLL, NHL, CML, or AML cells) from cancer patients (e.g., patients having a hematological cancer) into powerful antigen presenting cells (hereinafter referred to as “HC-APCs”) specifically presenting cancer antigens. These APCs can in turn be used to activate immune cells (e.g., T cells), rendering them highly effective therapeutic agents for cancer treatment. [0061] Thus, the present application provides methods of generating HC-APCs from hematological cancer cells (such as B-ALL cells, AML cells), use of the HC-APCs to activate immune cells, and use of the activated immune cells in treating cancer. Hematological cancer cells refer to cancer cells obtained from a hematological cancer. In some embodiments, the hematological cancer is a myeloid leukemia, a B-cell leukemia, or a B-cell lymphoma. In some embodiments, the hematological cancer is selected from the group consisting of AML, B-ALL, CML, CLL, and NHL. [0062] Accordingly, the present application in one aspect provides methods of producing a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), comprising: a) obtaining hematological cancer cells from an individual having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an IL-4 receptor (IL-4R) activator, and 4) an interferon ^ (IFN^) receptor (IFNGR) activator, thereby producing the population of HC-APCs. The S/D/M factors need not perform the same function. [0063] The present application in another aspect provides a population of HC-APCs, such as HC-APCs generated by some of the above methods and use of the HC-APCs for cancer treatment. [0064] The present application in another aspect provides methods of producing activated immune cells, comprising: a) producing a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”) according to methods described herein, and b) contacting the HC-APCs with immune cells, thereby producing activated ny-2770598
Attorney Docket No.24516-20006.40 immune cells. A population of activated immune cells obtained with said methods, and methods of treating cancer by administering the activated immune cells are also provided. [0065] The present application in another aspect provides methods of producing activated immune cells, comprising contacting immune cells with a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), thereby generating activated immune cells; wherein the HC-APCs are derived from hematological cancer cells from an individual after having been contacted with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an IL-4 receptor (IL-4R) activator, and 4) an interferon ^ (IFN^) receptor (IFNGR) activator. [0066] The present application in another aspect provides methods of treating a hematological cancer in an individual, comprising administering activated immune cells produced by methods described herein. In some embodiments, the methods further comprise administering a TNF^ inhibitor. In some embodiments, the TNF^ inhibitor is an anti-TNF^ antibody. I. Definitions [0067] In general, terms used in the claims and the specification are intended to be construed as having the plain meaning understood by a person of ordinary skill in the art. Certain terms are defined below to provide additional clarity. In case of conflict between the plain meaning and the provided definitions, the provided definitions are to be used. [0068] The term “individual,” “subject,” or “patient” is used synonymously herein to describe a mammal, including humans. An individual includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate. In some embodiments, the individual is human. In some embodiments, an individual suffers from a disease, such as cancer. In some embodiments, the individual is in need of treatment. [0069] “Monocytes,” “cancer cells,” “AML cells,” “B-ALL cells,” “immune cells,” “activated immune cells,” and “cells”, as used herein, are understood to refer not only to the monocytes, cancer cells, AML cells, B-ALL cells, immune cells, activated immune cells, or cells when obtained, but to the progeny or potential progeny of such cells. Because certain modifications may occur in succeeding generations due to e.g., environmental influences, ny-2770598
Attorney Docket No.24516-20006.40 such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein. [0070] “High level” or “higher level,” “low level” or “lower level,” when referring to expression of a surface molecule in a population of cells (e.g., monocytes or APCs) refers to how the average expression level of the particular surface molecule on the population of cells compares to the average level of the surface molecule on a reference cell population. Unless particularly stated otherwise, the reference cell population refers to a corresponding cell population derived from a healthy donor. In some cases, a high level of a particular molecule is defined when the expression level of the molecule on the recited cell population is at least about 20% (such as about any of 20%, 30%, 40%, 50%, or more) higher than that on the reference cell population. In some cases, a low level of a particular molecule is defined when the expression level of the molecule on the recited cell population is at least about 20% (such as about any of 20%, 30%, 40%, 50%, or less) lower than that on the reference cell population. [0071] A “reference”, as used herein, refers to any sample, standard, or level that is used for comparison purposes. A reference may be obtained from a healthy and/or non-diseased sample. In some examples, a reference may be obtained from an untreated sample. In some examples, a reference is obtained from a non-diseased or non-treated sample of an individual. In some examples, a reference is obtained from one or more healthy individuals who are not the individual or patient. [0072] As used herein, the term "antigen" is a substance that induces an immune response. Such a substance can include, for example, a toxin, a chemical, a bacterium, a virus, a protein or peptide fragment thereof, a polysaccharide, a lipid, an allergen, a nucleic acid, etc. [0073] As used herein, the term "neoantigen" is an antigen that has at least one alteration that makes it distinct from the corresponding wild-type parental antigen, e.g., via mutation in a tumor cell or post-translational modification specific to a tumor cell. A neoantigen can include a polypeptide sequence. A mutation that results in a neoantigen can include a frameshift or non-frameshift insertion or deletion (“indel”), missense or nonsense substitution, splice site alteration, genomic rearrangement or gene fusion, or any genomic or expression alteration giving rise to a neoORF (i.e., a new open reading frame). A mutation can also include a splice variant. Post-translational modifications specific to a tumor cell can include aberrant phosphorylation. Post-translational modifications specific to a tumor cell can ny-2770598
Attorney Docket No.24516-20006.40 also include a proteasome-generated spliced antigen. See Liepe et al., A large fraction of HLA class I ligands are proteasome-generated spliced peptides; Science.2016 Oct 21;354(6310):354-358. [0074] As used herein, the term "tumor neoantigen" or “cancer neoantigen” is a neoantigen present in a subject's tumor cell or tumor tissue but not in the subject's corresponding normal cell or normal tissue. [0075] The term "peptide" refers to a polymer of amino acids of no more than 100 amino acids (including fragments of a protein), which may be linear or branched, comprise modified amino acids, and/or be interrupted by non-amino acids. The term also encompasses an amino acid polymer that has been modified naturally or by intervention, including, for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification. Also included within this term are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art. The peptides described herein may be naturally occurring,
obtained or derived from a natural source (e.g., blood) or synthesized (e.g., chemically synthesized or synthesized by recombinant DNA techniques). [0076] As used herein, the term "epitope" is the specific portion of an antigen typically bound by an antibody or T-cell receptor. [0077] As used herein, the term "immunogenic" is the ability to elicit an immune response, e.g., via T-cells, B cells, or both. [0078] As used herein, the term "HLA binding affinity" or "MHC binding affinity" means affinity of binding between a specific antigen and a specific MHC allele. [0079] As used herein, the term "HLA type" is the complement of HLA gene alleles. [0080] As used herein, “activated T cells” refer to a population of monoclonal (e.g., encoding the same TCR) or polyclonal (e.g., with clones encoding different TCRs) T cells that have T cell receptors that recognize at least one tumor antigen peptide. Activated T cells may contain one or more subtypes of T cells, including, but not limited to, cytotoxic T cells (e.g., CD8 T cells), helper T cells (e.g., CD4 T cells), natural killer T cells, ^^ T cells, regulatory T cells, and memory T cells. [0081] As used herein, "treatment" or "treating" is an approach for obtaining beneficial or desired results including clinical results. For purposes of this invention, beneficial or desired ny-2770598
Attorney Docket No.24516-20006.40 clinical results include, but are not limited to, one or more of the following: decreasing one or more symptoms resulting from the disease; diminishing the extent of the disease; stabilizing the disease (e.g., preventing or delaying the worsening of the disease); preventing or delaying the spread (e.g., metastasis) of the disease; preventing or delaying the occurrence or recurrence of the disease; delay or slowing the progression of the disease; ameliorating the disease state; providing a remission (whether partial or total) of the disease; decreasing the dose of one or more other medications required to treat the disease; delaying the progression of the disease; increasing the quality of life; and/or prolonging survival. Also encompassed by "treatment" is a reduction of pathological consequence(s) of cancer. The methods of the invention contemplate any one or more of these aspects of treatment. [0082] As used herein, “delaying” the development of cancer means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease. A method that “delays” development of cancer is a method that reduces probability of disease development in a given time frame and/or reduces the extent of the disease in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies using a statistically significant number of individuals. Cancer development can be detectable using standard methods, including, but not limited to, computerized axial tomography (CAT Scan), Magnetic Resonance Imaging (MRI), abdominal ultrasound, clotting tests, arteriography, or biopsy. Development may also refer to cancer progression that may be initially undetectable and includes occurrence, recurrence, and onset. [0083] The term “simultaneous administration,” as used herein, means that a first therapy and a second therapy in a combination therapy are administered with a time separation of no more than about 15 minutes, such as no more than about any of 10, 5, or 1 minutes. When the first and second therapies are administered simultaneously, the first and second therapies may be contained in the same composition (e.g., a composition comprising both a first therapy and a second therapy) or in separate compositions (e.g., a first therapy is contained in one composition and a second therapy is contained in another composition). [0084] As used herein, the term “sequential administration” means that the first therapy and second therapy in a combination therapy are administered with a time separation of more than about 15 minutes, such as more than about any of 20, 30, 40, 50, 60, or more minutes. Either ny-2770598
Attorney Docket No.24516-20006.40 the first therapy or the second therapy may be administered first. The first and second therapies are contained in separate compositions, which may be contained in the same or different packages or kits. [0085] As used herein, the term “concurrent administration” means that the administration of the first therapy and of the second therapy in a combination therapy overlap with each other. [0086] As used herein, by “pharmaceutically acceptable” or “pharmacologically compatible” is meant a material that is not biologically or otherwise undesirable, e.g., the material may be incorporated into a pharmaceutical composition administered to an individual without causing any significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained. Pharmaceutically acceptable carriers or excipients have preferably met the required standards of toxicological and manufacturing testing and/or are included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug Administration. [0087] It is understood that embodiments of the application described herein include “consisting of” and/or “consisting essentially of” embodiments. [0088] Reference to “about” a value or parameter herein includes (and describes) variations that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X”. [0089] As used herein, reference to “not” a value or parameter generally means and describes “other than” a value or parameter. For example, the method is not used to treat cancer of type X means the method is used to treat cancer of types other than X. [0090] The term “about X-Y” used herein has the same meaning as “about X to about Y.” [0091] It should be noted that, as used in the specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. [0092] Any terms not directly defined herein shall be understood to have the meanings commonly associated with them as understood within the art of the invention. Certain terms are discussed herein to provide additional guidance to the practitioner in describing the compositions, devices, methods, and the like of aspects of the invention, and how to make or use them. It will be appreciated that the same thing may be said in more than one way. Consequently, alternative language and synonyms may be used for any one or more of the ny-2770598
Attorney Docket No.24516-20006.40 terms discussed herein. No significance is to be placed upon whether or not a term is elaborated upon or discussed herein. Some synonyms or substitutable methods, materials, and the like are provided. Recital of one or a few synonyms or equivalents does not exclude use of other synonyms or equivalents, unless it is explicitly stated. Use of examples, including examples of terms, is for illustrative purposes only and does not limit the scope and meaning of the aspects of the invention herein. II. Method of stimulating a population of cancer cells to generate APCs [0093] The present application in one aspect provides methods of producing potent antigen presenting cells that are derived from cancer cells (e.g., hematological cancer cells from an individual having the hematological cancer). It was found that these cancer cells (e.g., primary cancer cells), once obtained in an in vitro setting, normally could not survive more than 2-5 days. The methods described herein have been proven to consistently keep the primary cancer cells obtained from various donors with various hematological cancers (e.g., B-ALL, AML, CML, CLL, and NHL) alive for one or more weeks and effectively promote their differentiation into potent antigen presenting cells which present their endogenous cancer-specific antigens on the cell surface. These methods provide feasible and promising clinical application, especially in view of their consistent success with various hematological cancers and patients, and the capacity of activating and expanding T cells in large scale. [0094] The present application provides various methods of stimulating a population of hematological cancer cells (e.g., AML cells, CLL cells, CML cells, NHL cells, or B-ALL cells) from an individual to produce a population of APCs (“HC-APCs”). The methods involve obtaining hematological cancer cells from an individual and contacting the hematological cancer cells with one or more survival, differentiation, and/or maturation factors (“S/D/M factors”). Exemplary one or more S/D/M factors are discussed in Section “Survival, differentiation, and/or maturation factors (‘S/D/M factors’)” below. [0095] The hematological cancer cells can be obtained from PBMC, enriched leukemia cells, a bone marrow biopsy, or a lymphoma biopsy from the individual having the hematological cancer. In some embodiments, the cells are freshly obtained from the individual. In some embodiments, the cells have been subjected to freeze-thaw procedures. Without being bound by theory, freeze-thaw procedures are for preservation of the cells. In some embodiments, the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors. In some embodiments, the hematological cancer cells are B cell malignancy cancer cells. For ny-2770598
Attorney Docket No.24516-20006.40 example, B cell malignancy cancer cells are cancer cells from a B-cell lymphoma or a B cell leukemia. In some embodiments, the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors in the presence of an anti-CD19 antibody, an anti- CD20 antibody, or an anti-CD22 antibody. In some embodiments, the hematological cancer cells are cancer cells from a myeloid leukemia, and the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors in the presence of an anti-CD11b antibody. [0096] In some embodiments, the hematological cancer cells are comprised in a mixture of cells when brought into contact with the one or more S/D/M factors. In some embodiments, the hematological cancer cells are comprised in a PBMC or bone marrow sample obtained from the individual when being contacted with the one or more S/D/M factors. In some embodiments, the hematological cancer cells constitute at least about any of 50%, 60%, 70%, 80%, or 90% of the mixture of the cells when contacting with the one or more S/D/M factors. In some embodiments, the mixture of cells also comprises monocytes. In some embodiments, the monocytes are cancer monocytes. [0097] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual having the hematological cancer, and b) contacting the hematological cancer cells with one or more survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an interleukin-4 receptor (IL-4R) activator, and 4) an interferon ^ (IFN^) receptor (IFNGR) activator, thereby producing the population of HC-APCs. In some embodiments, the individual is a human patient. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. [0098] In some embodiments, the one or more of S/D/M factors is comprised in a T cell culture medium (e.g., exemplary T cell culture illustrated in the examples). In some embodiments, the one or more of S/D/M factors is comprised in a CD4 T cell culture medium obtained from a culture of primary CD4 T cells. In some embodiments, the culture of primary CD4 T cells is at 2 or 3 days of culture. In some embodiments, the T cell medium ny-2770598
Attorney Docket No.24516-20006.40 has a low level of IL-2 or does not constitute IL-2. In some embodiments, the T cell culture medium is further supplemented with a STAT3 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0099] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an IL-4 receptor (IL-4R) activator, and 4) an interferon ^ (IFN^) receptor (IFNGR) activator, thereby obtaining a population of HC-APCs. In some embodiments, the one or more of S/D/M factors are present in a single composition. In some embodiments, at least one of the one or more of S/D/M factors is provided separately from one of the other S/D/M factors in the one or more of S/D/M factors. In some embodiments, the one or more of the S/D/M factors further comprises a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF). In some embodiments, the one or more of the S/D/M factors further comprises an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors are comprised in a composition derived from a medium (e.g., supernatant) derived from a culture of T cells after being treated with anti-CD3 and anti-CD28 antibodies. In some embodiments, the T cells are CD4 T cells. In some embodiments, the T cells are CD8 T cells. In some embodiments, the T cells are isolated from PBMC of the same individual or a different individual and have not been previously treated with anti-CD3 and/or anti-CD28 antibodies prior to the treatment. In some embodiments, the T cells are isolated from PBMC of the same individual or a different individual and have been previously treated with anti-CD3 and/or anti-CD28 antibodies prior to the treatment. In some embodiments, the medium is derived from the culture after the T cells are treated with anti-CD3 and anti-CD28 antibodies for about 1-3 days, optionally for about 2 days. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 ny-2770598
Attorney Docket No.24516-20006.40 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0100] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 3) a TNF^ receptor (TNFR) activator (e.g., TNF^), wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs. In some embodiments, there is provided a method of producing a population of HC-APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, and 3) TNF^, wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some ny-2770598
Attorney Docket No.24516-20006.40 embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0101] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs. In some embodiments, there is provided a method of producing a population of HC-APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, and 3) IFN^, wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some ny-2770598
Attorney Docket No.24516-20006.40 embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0102] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), wherein at least IL-4R activator is provided separately from the STAT3 activator or IFNGR activator, thereby obtaining a population of HC-APCs. In some embodiments, there is provided a method of producing a population of HC-APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, and 3) IFN^, wherein at least IL-4 is provided separately from the IL-10 or IFN^, thereby obtaining a population of HC-APCs. In some embodiments, the IL-4R activator or IL-4 is provided after the STAT3 activator or IL-10 is provided. In some embodiments, the IL-4R activator or IL-4 is provided after the IFNGR activator or IFN^ is provided. In some embodiments, the STAT3 activator or IL-10 and the IFNGR activator or IFN^ are provided simultaneously. In some embodiments, the STAT3 activator or IL-10 and the IFNGR activator or IFN^ are provided sequentially. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of ny-2770598
Attorney Docket No.24516-20006.40 at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0103] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs. In some embodiments, there is provided a method of producing a population of HC-APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) TNF^, and 3) IFN^, wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of ny-2770598
Attorney Docket No.24516-20006.40 at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0104] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), wherein at least STAT3 activator is provided separately from the TNFR activator or IFNGR activator, thereby obtaining a population of HC-APCs. In some embodiments, there is provided a method of producing a population of HC-APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) TNF^, and 3) IFN^, wherein at least IL-10 is provided separately from the TNF^ or IFN^, thereby obtaining a population of HC-APCs. In some embodiments, the STAT3 activator or IL-10 is provided before the TNFR activator or TNF^ is provided. In some embodiments, the STAT3 activator or IL-10 is provided before the IFNGR activator or IFN^ is provided. In some embodiments, the TNFR activator or TNF^ and the IFNGR activator or IFN^ are provided simultaneously. In some embodiments, the TNFR activator or TNF^ and the IFNGR activator or IFN^ are provided sequentially. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about ny-2770598
Attorney Docket No.24516-20006.40 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0105] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs. In some embodiments, there is provided a method of producing a population of HC-APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, 3) TNF^, and 4) IFN^, wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, ny-2770598
Attorney Docket No.24516-20006.40 optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the hematological cancer cells are cultured for about 1-3 days (e.g., 2- 3 days) in the presence of at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0106] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), wherein at least one of the one or more of S/D/M factors is provided separately from at least one of the other S/D/M factors in the one or more of S/D/M factors, thereby obtaining a population of HC-APCs. In some embodiments, there is provided a method of producing a population of HC-APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, 3) TNF^, and 4) IFN^, wherein at least one of the one or more of S/D/M factors is provided separately from at least one of the other ny-2770598
Attorney Docket No.24516-20006.40 S/D/M factors in the one or more of S/D/M factors, thereby obtaining a population of HC- APCs. In some embodiments, the STAT3 activator or IL-10 is provided before at least one of the other factors (e.g., the IL-4R activator or IL-4) is provided. In some embodiments, the IL- 4R activator or IL-4 is provided after at least one of the other factors (e.g., the STAT3 activator or IL-10, the IFNGR activator or IFN^, or the TNFGR activator or TNF^) is provided. In some embodiments, the STAT3 activator or IL-10, the TNFR activator or TNF^, and the IFNGR activator or TNF^ are provided simultaneously. In some embodiments, the STAT3 activator or IL-10, the TNFR activator or TNF^, and the IFNGR activator or IFN^ are provided sequentially. In some embodiments, the IL-4R activator or IL-4, the TNFR activator or TNF^, and the IFNGR activator or IFN^ are provided simultaneously. In some embodiments, the IL-4R activator or IL-4, the TNFR activator or TNF^, and the IFNGR activator or IFN^ are provided sequentially. In some embodiments, the IL-10 is a human IL- 10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). ny-2770598
Attorney Docket No.24516-20006.40 [0107] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a TNF^ receptor (TNFR) activator (e.g., TNF^), 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 5) one or both of a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF) and an IL-6 receptor (IL-6R) activator (e.g., IL-6), wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs. In some embodiments, there is provided a method of producing a population of HC-APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, 3) TNF^, 4) IFN^, and 5) one or both of GM-CSF and IL-6, wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6. In some embodiments, the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 ny-2770598
Attorney Docket No.24516-20006.40 pg/ml). In some embodiments, the GM-CSF is a human GM-CSF or a human recombinant GM-CSF. In some embodiments, the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml). In some embodiments, the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0108] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator (e.g., IL-10, e.g., IL-12), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a TNF^ receptor (TNFR) activator (e.g., TNF^), 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 5) one or both of a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF) and an IL-6 receptor (IL-6R) activator (e.g., IL-6), wherein at least one of the one or more of S/D/M factors is provided separately from at least one of the other S/D/M factors in the one or more of S/D/M factors, thereby obtaining a population of HC- APCs. In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10, 2) IL-4, 3) TNF^, 4) IFN^, and 5) one or both of GM-CSF and IL-6, wherein at least one of the one or more of S/D/M factors is provided separately from at least one of the other S/D/M factors in the one or more of S/D/M factors, thereby obtaining a population of HC-APCs. In some embodiments, the STAT3 activator or IL-10, and/or the GM-CSFR activator or GM- ny-2770598
Attorney Docket No.24516-20006.40 CSF is provided before at least one of the other factors (e.g., the IL-4R activator or IL-4, e.g., the IL-6R activator or IL-6) is provided. In some embodiments, the IL-4R activator or IL-4, and/or the IL-6R activator or IL-6 is provided after at least one of the other factors (e.g., the STAT3 activator or IL-10, e.g., the GM-CSFR activator or GM-CSF) is provided. In some embodiments, the STAT3 activator or IL-10, the TNFR activator or TNF^, and the IFNGR activator or IFN^ are provided simultaneously. In some embodiments, the STAT3 activator or IL-10, the TNFR activator or TNF^, and the IFNGR activator or IFN^ are provided sequentially. In some embodiments, the IL-4R activator or IL-4, the TNFR activator or TNF^, and the IFNGR activator or IFN^ are provided simultaneously. In some embodiments, the IL-4R activator or IL-4, the TNFR activator or TNF^, and the IFNGR activator or IFN^ are provided sequentially. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6. In some embodiments, the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml). In some embodiments, the GM-CSF is a human GM-CSF or a human recombinant GM-CSF. In some embodiments, the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml). In some embodiments, the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some ny-2770598
Attorney Docket No.24516-20006.40 embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0109] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10 (e.g., a human IL-10 or a human recombinant IL-10), 2) IL-4 (e.g., a human IL-4 or a human recombinant IL-4), 3) TNF^ (e.g., a human TNF^ or a human recombinant TNF^), 4) IFN^ (e.g., a human IFN^ or a human recombinant IFN^), 5) GM-CSF (e.g., a human GM-CSF or a human recombinant GM-CSF), and 6) IL-6 (e.g., a human IL-6 or a human recombinant IL- 6), wherein the one or more of S/D/M factors are present in a single composition, thereby obtaining a population of HC-APCs. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in a medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6. In some embodiments, the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 ny-2770598
Attorney Docket No.24516-20006.40 pg/ml). In some embodiments, the GM-CSF is a human GM-CSF or a human recombinant GM-CSF. In some embodiments, the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml). In some embodiments, the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with an IL-10R activator (e.g., a STAT3 activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0110] In some embodiments, there is provided a method of producing a population of HC- APCs, comprising: a) obtaining hematological cancer cells from an individual (e.g., a human patient) having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) IL-10 (e.g., a human IL-10 or a human recombinant IL-10), 2) IL-4 (e.g., a human IL-4 or a human recombinant IL-4), 3) TNF^ (e.g., a human TNF^ or a human recombinant TNF^), 4) IFN^ (e.g., a human IFN^ or a human recombinant IFN^), 5) GM-CSF (e.g., a human GM-CSF or a human recombinant GM-CSF), and 6) IL-6 (e.g., a human IL-6 or a human recombinant IL- 6), wherein at least one of the one or more of S/D/M factors is provided separately from at least one of the other S/D/M factors in the one or more of S/D/M factors, thereby obtaining a population of HC-APCs. In some embodiments, IL-10 and/or GM-CSF is provided before at least one of the other factors (e.g., IL-4 or IL-6) is provided. In some embodiments, IL-4 and/or IL-6 is provided after at least one of the other factors (e.g., IL-10 or GM-CSF) is provided. In some embodiments, IL-10, TNF^, and IFN^ are provided simultaneously. In some embodiments, IL-10, TNF^, and the IFN^ are provided sequentially. In some embodiments, IL-4, TNF^, and the IFN^ are provided simultaneously. In some embodiments, IL-4, TNF^, and the IFN^ are provided sequentially. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, ny-2770598
Attorney Docket No.24516-20006.40 further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6. In some embodiments, the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml). In some embodiments, the GM-CSF is a human GM-CSF or a human recombinant GM-CSF. In some embodiments, the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml). In some embodiments, the hematological cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL-10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0111] In some embodiments, there is provided a method of stimulating a population of cancer cells (e.g., AML cells, CLL cells, CML cells, NHL cells, or B-ALL cells) from an individual having a cancer to produce a population of antigen presenting cells (“APCs”), comprising contacting the population of cancer cells with a medium derived from a culture (e.g., a supernatant) of T cells after being treated with anti-CD3 and anti-CD28 antibodies, wherein the medium comprises a STAT3 activator (e.g., IL-10). In some embodiments, the ny-2770598
Attorney Docket No.24516-20006.40 medium further comprises an IL-4R activator (e.g., IL-4), an IFNGR activator (e.g., IFN^), a TNFR activator (e.g., TNF^). In some embodiments, the medium further comprises a GM- CSFR activator (e.g., GM-CSF) and/or an IL-6R activator (e.g., IL-6). In some embodiments, the T cells are isolated from PBMC of the same individual or a different individual and have not been previously treated with anti-CD3 and/or anti-CD28 antibodies prior to the treatment. In some embodiments, the medium is derived from the culture after the T cells are treated with anti-CD3 and anti-CD28 antibodies for about 1-3 days, optionally for about 2 days. In some embodiments, the cancer cells are cultured for about 2-3 days in the presence of the medium derived from the culture of T cells. In some embodiments, the IL-10 is a human IL- 10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6. In some embodiments, the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml). In some embodiments, the GM-CSF is a human GM-CSF or a human recombinant GM-CSF. In some embodiments, the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml). In some embodiments, the cancer cells are cultured for about 1-3 days (e.g., 2-3 days) in the presence of at least one of the S/D/M factors. In some embodiments, the one or more of S/D/M factors does not comprise any one or more of IL-3, IL-7, and/or FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise any of IL-3, IL-7, and FLT3L. In some embodiments, the one or more of S/D/M factors does not comprise IL-2 or has a level of IL-2 ny-2770598
Attorney Docket No.24516-20006.40 that is lower than 3000 pg/ml, 2000 pg/ml, 1000 pg/ml, 500 pg/ml, or 300 pg/ml. In some embodiments, the T cell culture is further supplemented with a STAT3 activator (e.g., an IL- 10R activator, e.g., any of the exemplary IL-10R activators disclosed in Table 1). [0112] In some embodiments, the level of IL-10R on the hematological cancer cells before contacting the S/D/M factors is at least about any of 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-10R on corresponding cells from a reference individual (e.g., a healthy individual). In some embodiments, the level of IL-4R on the cancer cells before contacting the S/D/M factors is at least about any of 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-4R on corresponding cells from a reference individual (e.g., a healthy individual). [0113] In some embodiments according to any of the embodiments described above, the method further comprises contacting the population of cancer cells (e.g., AML cells, CLL cells, CML cells, NHL cells, or B-ALL cells) with a one or more of refinement factors selected from the group consisting of type-I interferon, IFN^, TNF^, a TLR ligand, CD40L or a CD40-ligating antibody, an anti-PD-L1 antibody, and TPI-1, optionally wherein the type-I interferon comprises IFN^ and/or IFN^, and optionally wherein the TLR ligand is R848, poly IC, CpG, or LPS. In some embodiments, the one or more of refinement factors is provided after the plurality of cancer cells are contacted with the one or more of S/D/M factors or the medium derived from the culture of T cells, thereby producing the population of APCs, and wherein the population of APCs is cultured for about 1-5 days in the presence of the one or more of the refinement factors, optionally wherein the population of APCs is cultured for about one day. In some embodiments, the one or more of refinement factors are provided when: a) at least about 50% of the cancer cells differentiate into APCs and survive, b) at least about 30% of the population of APCs exhibit a dendritic cell morphology, and/or c) the population of APCs express: i) a high level of one or more molecules selected from the group consisting of MHC I, MHC II, CD80, CD86, and/or CD40, and/or ii) a low level of SIRP^. [0114] In some embodiments, the method further comprises contacting the population of cancer cells with one or more of refinement factors comprising IFN^, IFN^, and TNF^. [0115] In some embodiments, the one or more of refinement factors comprise IFN^, IFN^, TNF^, R848, poly IC, and CpG. [0116] In some embodiments, the one or more of refinement factors comprise IFN^, IFN^, TNF^, R848, poly IC, CpG, CD40L, and an anti-PD-L1 antibody. ny-2770598
Attorney Docket No.24516-20006.40 [0117] In some embodiments, the one or more of refinement factors comprise IFN^, IFN^, TNF^, R848, poly IC, CpG, CD40L, TPI-1, and an anti-PD-L1 antibody. Survival, differentiation, and/or maturation factors (“S/D/M factors”) [0118] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises one or more agents selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an interleukin-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0119] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises one or more agents selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL- 4), and 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0120] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 6) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected ny-2770598
Attorney Docket No.24516-20006.40 from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0121] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises one or more agents selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL- 4), 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF). In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0122] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises one or more agents selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL- 4), 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 6) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 6) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0123] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) IFN^. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein ny-2770598
Attorney Docket No.24516-20006.40 comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) TNF^. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) IL-6. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) IL- 4. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) GM-CSF. In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0124] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 2) TNF^. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 2) IL-6. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 2) IL-4. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 2) GM-CSF. [0125] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 2) IFN^. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 2) IL-6. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 2) IL-4. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 2) GM-CSF. [0126] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a GM-CSF receptor (GM-CSFR) ny-2770598
Attorney Docket No.24516-20006.40 activator (e.g., GM-CSF), and 2) IFN^. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 2) IL-6. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 2) IL-4. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 2) TNF^. [0127] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 2) IFN^. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 2) IL-6. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 2) GM-CSF. [0128] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 3) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0129] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0130] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, ny-2770598
Attorney Docket No.24516-20006.40 the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL- 24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0131] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), 2) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). [0132] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0133] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0134] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0135] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). [0136] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ^ (IFN^) receptor (IFNGR) ny-2770598
Attorney Docket No.24516-20006.40 activator (e.g., IFN^). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0137] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0138] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0139] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 4) a GM-CSF receptor (GM- CSFR) activator (e.g., GM-CSF). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0140] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0141] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) a GM- ny-2770598
Attorney Docket No.24516-20006.40 CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0142] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 4) a GM-CSF receptor (GM- CSFR) activator (e.g., GM-CSF). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0143] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0144] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a GM- CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0145] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 3) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. ny-2770598
Attorney Docket No.24516-20006.40 [0146] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 4) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF). [0147] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL- 6). [0148] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) a GM-CSF receptor (GM- CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6). [0149] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), 2) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 3) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6). [0150] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 2) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 3) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6). [0151] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL- 19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. ny-2770598
Attorney Docket No.24516-20006.40 [0152] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), and 5) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0153] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 4) a GM-CSF receptor (GM- CSFR) activator (e.g., GM-CSF), and 5) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL- 22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0154] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), 2) an IL-4 receptor (IL-4R) activator (e.g., IL-4), 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 4) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 5) an IL-6 receptor (IL-6R) activator (e.g., IL-6). [0155] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10), and 2) IL-12. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) poly:IC. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) CpG. In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), and 2) R848. In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. ny-2770598
Attorney Docket No.24516-20006.40 [0156] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0157] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6), and 5) a GM-CSF receptor (GM-CSF) activator (e.g., GM-CSF). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL- 22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0158] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) an IL-22, 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6), and 5) a GM-CSF receptor (GM- CSF) activator (e.g., GM-CSF). [0159] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a TNF^ receptor (TNFR) activator (e.g., TNF^), 2) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 3) an IL-6 receptor (IL-6R) activator (e.g., IL-6), and 4) a GM-CSF receptor (GM-CSF) activator (e.g., GM-CSF). [0160] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein comprises: 1) a STAT3 activator (e.g., IL-10, e.g., any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 4) an IL-6 receptor (IL-6R) activator (e.g., IL-6), 5) an IL-4 receptor (IL-4R) activator (e.g., IL-4), and 6) a GM-CSF receptor (GM-CSF) activator (e.g., GM-CSF). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. ny-2770598
Attorney Docket No.24516-20006.40 [0161] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described above are provided in a composition comprising a serum (e.g., human serum). In some embodiments, the serum comprises human serum, fetal bovine serum, or murine serum, etc. [0162] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein are provided in a composition comprising a serum (e.g., human serum). In some embodiments, the one or more S/D/M factors is selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an interleukin-4 receptor (IL-4R) activator (e.g., IL-4), and 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0163] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein are provided in a composition comprising a serum (e.g., human serum), wherein the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) combined in serum described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), and 3) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL- 19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. [0164] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein are provided in a composition comprising a serum (e.g., human serum), wherein the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) combined in serum described herein comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an IL-4 receptor (IL-4R) activator (e.g., IL-r), and 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). [0165] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein are provided in a composition comprising a serum (e.g., human serum). In some embodiments, the one or more S/D/M factors is selected from the group consisting of: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an interleukin-4 receptor (IL-4R) ny-2770598
Attorney Docket No.24516-20006.40 activator (e.g., IL-4), 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 6) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone [0166] In some embodiments, the one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) described herein are provided in a composition comprising a serum (e.g., human serum), wherein the one or more S/D/M factors comprises: 1) a STAT3 activator (e.g., IL-10 or any of those listed in Table 1), 2) a TNF^ receptor (TNFR) activator (e.g., TNF^), 3) an interleukin-4 receptor (IL-4R) activator (e.g., IL-4), 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), 5) a GM-CSF receptor (GM-CSFR) activator (e.g., GM-CSF), and 6) an IL-6 receptor (IL-6R) activator (e.g., IL-6). In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. IL-10 receptor (IL-10R) activator and IL-10 [0167] “IL-10 receptor (IL-10R) activator” described herein refers to a molecule that activates the IL-10 receptor-mediated signaling pathway. IL-10R includes both IL-10R1 and IL-10R2. [0168] Interleukin 10 (IL-10), also known as human cytokine synthesis inhibitory factor (CSIF), is an anti-inflammatory cytokine. In humans, interleukin 10 is encoded by the IL10 gene. IL-10 signals through a receptor complex consisting of two IL-10 receptor-1 and two IL-10 receptor-2 proteins. Consequently, the functional receptor consists of four IL-10 receptor molecules. IL-10 binding induces STAT3 signaling via the phosphorylation of the cytoplasmic tails of IL-10 receptor 1 and IL-10 receptor 2 by JAK1 and Tyk2, respectively. See e.g., Saraiva, M., O'Garra, A. The regulation of IL-10 production by immune cells. Nat Rev Immunol 10, 170–181 (2010). [0169] In humans, IL-10 is encoded by the IL10 gene, which is located on chromosome 1 and comprises 5 exons, and is primarily produced by monocytes and, to a lesser extent, lymphocytes, namely type-II T helper cells (TH2), mast cells, CD4+CD25+Foxp3+ regulatory T cells, and in a certain subset of activated T cells and B cells. IL-10 can be produced by monocytes upon PD-1 triggering in these cells. ny-2770598
Attorney Docket No.24516-20006.40 [0170] IL-10 is a cytokine with multiple, pleiotropic, effects in immunoregulation and inflammation. It downregulates the expression of Th1 cytokines, MHC class II antigens, and co-stimulatory molecules on macrophages. It also enhances B cell survival, proliferation, and antibody production. IL-10 can block NF-^B activity and is involved in the regulation of the JAK-STAT signaling pathway. [0171] IL-10 was initially reported to suppress cytokine secretion, antigen presentation, and CD4+ T cell activation. Further investigation has shown that IL-10 predominantly inhibits lipopolysaccharide (LPS) and bacterial product-mediated induction of the pro-inflammatory cytokines TNF^, IL-1^, IL-12, and IFN^ secretion from Toll-Like Receptor (TLR)-triggered myeloid lineage cells. [0172] IL-10 as referred to herein comprises any constructs that have a component of IL-10 (e.g., a naturally occurring IL-10, e.g., a recombinant IL-10). These include and are not limited to natural IL-10 (e.g., various isoforms of human IL-10), synthetic or recombinant IL- 10, and fusion proteins having an IL-10 component. [0173] In some embodiments, the IL-10R activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety. An Fc portion is a portion of the fragment crystallizable region (Fc region), which is the tail region of an antibody that can interact with cell surface receptors (e.g., Fc receptors and some proteins of the complement system). A PEG moiety is a polyethylene glycol polymer chain that can be attached, e.g., to a molecule such as an antibody and can extend the duration of time in which said molecule circulates before being degraded or reduced by renal clearance. [0174] In some embodiments, the IL-10R activator is selected from the group consisting of: an IL-10 (e.g., a pegylated IL-10, e.g., pegilodecakin or AM0010), an IL-10 family member (e.g., IL-19, IL-20, IL-22, IL-24, IL-26, IL-28, IL-7, IL-9, IL-15, and IL-21), an IL-10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, a small molecule activator of IL-10R, and an activator of the IL-10R-downstream STAT3 (e.g., IL-12, e.g., Long noncoding RNA (LncRNA) PVT1, NEAT1, FEZF1-AS1, UICC). In some embodiments, the IL-10R activator is IL-10. In some embodiments, the IL-10 is a human IL- 10 or a human recombinant IL-10. In some embodiments, the IL-10 (e.g., a human IL-10) is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IL-10 (e.g., a human IL-10) is present in the medium at a concentration of about 2 ng/ml to about 200 ng/ml (e.g., about 10 ny-2770598
Attorney Docket No.24516-20006.40 ng/ml to about 200ng/ml, e.g., about 10 ng/ml to about 100 ng/ml, e.g., about 20 ng/ml to about 100 ng/ml). STAT3 activator and STAT3 [0175] In some embodiments, the IL-10R activator is a STAT3 activator. “STAT3 activator” as described herein refers to a molecule that activates STAT3 signaling, e.g., the STAT3 nuclear localization and transcription factor activity. [0176] STAT3 is a transcription factor that resides in the cytoplasm in its inactive, unphosphorylated form and translocates to the nucleus upon its activation via phosphorylation, e.g., Tyr705 phosphorylation, and subsequent dimerization. Upon entering the nucleus, the activated STAT3 dimer binds to the IFN^–activated sequence (GAS) in target promoters and thereby activates transcription of target genes. Multiple tyrosine kinases have been described as intracellular activators of STAT3 activity (e.g., JAK1, JAK2, EGFR, Src, and ERK). Further mechanisms of activation include: i) STAT3 phosphorylation at Ser727 by protein kinase C (PKC), mitogen-activated protein kinases (MAPKs), and CDK5; and ii) STAT3 acetylation on Lys685 by histone acetyltransferase, which can enhance STAT3 dimer stability. See, e.g., Rébé et al., JAKSTAT 2013;2(1):e23010. [0177] STAT3 is expressed in most cell types under specific conditions, and generally is described to be involved with biological processes such as: cell proliferation, differentiation, apoptosis, angiogenesis, metastasis, inflammation, and immunity. In immune cells, STAT3 has been described in contradictory terms. For example, STAT3 has been described as promoting the differentiation of macrophages toward the M2 phenotype and the absence of functional dendritic cells (see, e.g., Rébé et al., 2013). STAT3 has also been described to promote the gp130-mediated maintenance of the pluripotential state of proliferating embryonic stem cells and for the gp130-induced macrophage differentiation of M1 cells. Both c-myc and pim have been identified as target genes of STAT3 and together can compensate for STAT3 in cell survival and cell-cycle transition (see, e.g., Hirano et al. Oncogene 2000;19:2548-2556). [0178] STAT3 activation is rapid and transient under normal biological conditions and mediated by many extracellular stimuli, including cytokines (IL-6, IL-10, IFNs, TNF^, LIF, OSM, etc.) and growth factors (e.g., EGF, G-CSF, GM-CSF, VEGF, HGF, GH, and Her2/Neu). Active oncogenic proteins, such as Src (e.g., v-Src) and Ras, as well as chemical carcinogens and other molecules also can activate STAT3. Indeed, many regulated genes ny-2770598
Attorney Docket No.24516-20006.40 induced by STAT3 activity in turn activate the same STAT3 pathways and thereby keep a stable feedforward loop. In some embodiments, the STAT3 activator comprises a cytokine selected from the group consisting of: IL-6, IL-10, IL-11, IL-12, IL-19, IL-20, IL-22, IL-23, IL-24, IL-26, IL-27, IL-7, IL-9, IL-15, IL-21, IFN^, IFN^, IFN^, TNF^, leukemia inhibitory factor (LIF), oncostatin M (OSM), biologically active derivatives thereof, and any combination thereof. In some embodiments, the STAT3 activator comprises a growth factor selected from the group consisting of: EGF, FGF, IGF, G-CSF, GM-CSF, VEGF, HGF, GF, Her2/Neu, biologically active derivatives thereof, and any combination thereof. In some embodiments, the STAT3 activator comprises a JAK activator, such as an enzyme that phosphorylates JAK (e.g., JAK2). In some embodiments, the STAT3 activator comprises a hormone (e.g., leptin). In some embodiments, the STAT3 activator comprises a chaperone protein (e.g., HSP90, HSP70, HSP27, HSP110, HOP). [0179] STAT3 activity can be positively regulated by the signaling pathways of IL-10 and IL-10 family members, including IL-19, IL-20, IL-22, IL-24, and IL-26. Further, IL-12 and affiliated family members (e.g., IL-23) can activate STAT3 activity, at least in part by promoting IL-10/IL-10R production and autocrine signaling in the cancer cell-derived (e.g., B-ALL or AML) ^APC cells. IL-6 has also been described as an activator of the STAT3 pathway. In some embodiments, the STAT3 activator is selected from the group consisting of: IL-6, IL-10, IL-12, IL-19, IL-20, IL-22, IL-23, IL-24, IL-26, IL-7, IL-9, IL-15, IL-21, biologically active derivatives thereof, and any combination thereof. In some embodiments, the STAT3 activator is an IL-10R activator, such as any described herein or known in the art. [0180] In some embodiments, the IL-10R activator is an activator of the IL-10R-downstream STAT3. STAT3 activators can include, but are not limited to, any of: a small molecule, a nucleic acid (e.g., an siRNA, an shRNA, an antisense RNA, a microRNA), a nucleic acid base inhibitor (e.g., a circular RNA inhibitor), a nucleic acid editing system (e.g., CRISPR, ZFN, or TALENS systems), a decoy oligonucleotide, a peptide agent, a protein agent (e.g., an antibody agent that targets IL-10R; e.g., a protein agent that targets STAT3 phosphorylation and/or prevents STAT3 dephosphorylation), a protein stabilizing agent (e.g., a STAT3- stabilizing agent such as a chaperone protein, for example HSP90, HSP70, HSP27, HSP110, and/or HOP), a protein degrading or destabilizing agent (e.g., a phosphatase degrading or destabilizing agent such as a phosphatase-targeting PROTAC, LYTAC, molecular glue, AbTAC, CMATAC, etc.), a protein modified with an unnatural amino acid, a viral agent ny-2770598
Attorney Docket No.24516-20006.40 (e.g., Kaposi sarcoma herpesvirus (KSHV)), derivatives thereof, and any combination thereof. [0181] In some embodiments, the STAT3 activator comprises a cancer cell STAT3 activator. These cancer cell STAT3 activators can include any of: PVT1, NEAT1, FEZF1-AS1, UICC, MALAT1, XIST, miR-30d, CD109, CD146, CD24, CDK7, SOX, Smad6, Smad7, TRIM24, TRIM27, TRIM59, ADAM12, USP22, BMX AKR1C1, PRMT1, PBX1, HSP110, RanBP6, RAC1-GTP, PA28^, E6, and FABP5. [0182] In some embodiments, the STAT3 activator comprises a small molecule selected from the group consisting of: Colivelin, Colivelin TFA, Garcinone D, Butyzamide, Eflepedocokin alfa, Broussonin E, derivatives thereof, and any combinations thereof. In some embodiments, the STAT3 activator comprises Colivelin, Colivelin TFA, and/or Garcinone D. [0183] In some embodiments, the STAT3 activator comprises an inhibitor or antagonist of a molecule or compound that inactivates STAT3 (e.g., reduces phosphorylated STAT3 levels) or that reduces total STAT3 levels (e.g., via proteasomal degradation and/or transcriptional suppression) in a cell of interest, e.g., in a myeloid cell such as an AML-derived ^APC cell. For example, molecules or compounds that can inactivate STAT3 include, but are not limited to: ^-elemene, selective serotonin-reuptake inhibitors (SSRIs, e.g., fluoxetine), minecoside, Luteolin (3,4,5,7-tetrahydroxyflavone), SHP-1, SHP-2, PTP1B, PTPRM, eEF2 kinase, PKM2, curcumin, cucurbitacin, honokiol, guggulsterone, resveratrol, berbamine, flavopiridol, JAK inhibitors/inactivated JAK (e.g., JAK2), low molecular weight-DSP2, PIAS3, etc. In some embodiments, molecules or compounds that can reduce total STAT3 levels (e.g., via proteasomal degradation and/or transcriptional suppression) include, but are not limited to: PDLIM2, COP1, calcineurin, SOCS proteins, Rubulavirus (e.g., Mumps virus, e.g., the Mumps viral V protein, for example the V-dependent degradation complex VDC or V/DDB1/Cullin degradation complex), TSM-1, KYM-003, KTX-201, SD-36, AUY922, 17- DMAG, etc. In some embodiments, the inhibitor of a molecule or compound that inactivates STAT3 or that reduces total STAT3 levels in a cell of interest competitively bind to STAT3 to prevent inactivation (e.g., DDIAS) or protein degradation (e.g., chaperones, such as HSP90). [0184] See, e.g., Kim et al. Oncol Lett.2022;23(3):94; Zheng et al. Exp Mol Med. 2018;50(9):1-14; Liao et al. Anticancer Res.2022;42(8):3807-3814; Xiao et al. Cell ny-2770598
Attorney Docket No.24516-20006.40 Commun Signal.2020;18(1):25; Jego et al. Cancers (Basel) 2020;12(1):21; Liu et al. Cell Death Dis.2014;5(6):e1293; and Yang et al. Cytokine Growth Factor Rev. 2019;46:10-22. [0185] The amount of exemplified STAT3 activator can be seen in e.g., Table 1. [0186] In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-12, IL-23, IL-6, colivelin TFA, Garcinone D, G-CSF, IL-7, IL-9, IL-15, and IL-21. [0187] In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL-20, IL-24, IL-12, IL-23, IL-6, colivelin TFA, and Garcinone D. [0188] In some embodiments, the STAT3 activator is selected from the group consisting of IL-10, IL-12, Colivelin TFA, and Garcinone D. TNF^ receptor (TNFR) activator and TNF^ [0189] “TNF^ receptor (TNFR) activator” as described herein refers to a molecule that activates the TNFR-mediated signaling pathway. TNFR as described herein refers to either TNFR1 or TNFR2. [0190] Tumor necrosis factor ^ (TNF, cachexin, or cachectin; often called tumor necrosis factor alpha or TNF^) is an adipokine and a cytokine. TNF^ is a member of the TNF^ superfamily, which consists of various transmembrane proteins with a homologous TNF^ domain. [0191] TNF^ can bind two receptors, TNFR1 (TNF^ receptor type 1; CD120a; p55/60) and TNFR2 (TNF^ receptor type 2; CD120b; p75/80). TNFR1 is 55-kDa and TNFR2 is 75-kDa. TNFR1 is expressed in most tissues, and can be fully activated by both the membrane-bound and soluble trimeric forms of TNF, whereas TNFR2 is found typically in cells of the immune system, and responds to the membrane-bound form of the TNF^ homotrimer. [0192] TNF^ was thought to be produced primarily by macrophages, but it is produced also by a broad variety of cell types including lymphoid cells, mast cells, endothelial cells, cardiac myocytes, adipose tissue, fibroblasts, and neurons. Large amounts of TNF^ are released in response to lipopolysaccharide, other bacterial products, and interleukin-1 (IL-1). In the skin, mast cells appear to be the predominant source of pre-formed TNF^, which can be released upon inflammatory stimulus (e.g., LPS). [0193] TNF^ as referred to herein comprises any constructs that have a component of TNF^ (e.g., a naturally occurring TNF^, e.g., a recombinant TNF^). These include and are not ny-2770598
Attorney Docket No.24516-20006.40 limited to natural TNF^ (e.g., various isoforms of human TNF^), synthetic or recombinant TNF^, and fusion proteins having a TNF^ component. [0194] In some embodiments, the TNFR activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety. [0195] In some embodiments, the one or more of S/D/M factors comprise a TNFR activator, optionally wherein the TNFR activator is selected from the group consisting of TNF^, a TNFR agonist antibody, and a small molecule activator of TNFR. In some embodiments, the TNFR activator is TNF^. In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ (e.g., a human TNF^) is present in the medium at a concentration of at least about 0.2 ng/ml, optionally at least about 0.5 ng/ml (e.g., at least about 1 ng/ml, about 2 ng/ml, or about 3 ng/ml). In some embodiments, the TNF^ (e.g., a human TNF^) is present in the medium at a concentration of about 0.2 ng/ml to about 30 ng/ml (e.g., about 0.5 ng/ml to about 10 ng/ml, e.g., about 1 ng/ml to about 5 ng/ml, e.g., about 2 ng/ml to about 4 ng/ml). IFN^ receptor (IFNGR) activator and IFN^ [0196] “IFN^ receptor (INFGR) activator” as described herein refers to a molecule that activates the INFGR-mediated signaling pathway. [0197] Interferon gamma (IFN^) is a dimerized soluble cytokine that is the only member of the type II class of interferons. IFN^, or type II interferon, is a cytokine that is critical for innate and adaptive immunity against viral, some bacterial, and protozoan infections. IFN^ is an important activator of macrophages and inducer of major histocompatibility complex class II molecule expression. Aberrant IFN^ expression is associated with a number of autoinflammatory and autoimmune diseases. The importance of IFN^ in the immune system stems in part from its ability to inhibit viral replication directly, and most importantly from its immunostimulatory and immunomodulatory effects. IFN^ is produced predominantly by natural killer cells (NK) and natural killer T cells (NKT) as part of the innate immune response, and by CD4 Th1 and CD8 cytotoxic T lymphocyte (CTL) effector T cells once antigen-specific immunity develops as part of the adaptive immune response. IFN^ is also produced by non-cytotoxic innate lymphoid cells (ILC), a family of immune cells first discovered in the early 2010s. [0198] IFN^ as referred to herein comprises any constructs that have a component of IFN^ (e.g., a naturally occurring IFN^, e.g., a recombinant IFN^). These include but are not limited ny-2770598
Attorney Docket No.24516-20006.40 to natural IFN^ (e.g., various isoforms of human IFN^), synthetic or recombinant IFN^, and fusion proteins having an IFN^ component. [0199] In some embodiments, the IFNGR activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety. [0200] In some embodiments, the one or more of S/D/M factors comprises an IFNGR activator, optionally wherein the IFNGR activator is selected from the group consisting of IFN^, an IFNGR agonist antibody, and a small molecule activator of IFNGR. In some embodiments, the IFNGR activator is IFN^. In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ (e.g., a human IFN^) is present in the medium at a concentration of at least about 1 ng/ml, optionally at least about 5 ng/ml (e.g., at least about 10 ng/ml, about 20 ng/ml, or about 50 ng/ml). In some embodiments, the IFN^ (e.g., a human IFN^) is present in the medium at a concentration of about 1 ng/ml to about 500 ng/ml (e.g., about 5 ng/ml to about 200 ng/ml, e.g., about 10 ng/ml to about 100 ng/ml, e.g., about 40 ng/ml to about 60 ng/ml, e.g., about 50 ng/ml). [0201] In some embodiments, the one or more of S/D/M factors comprises two or more agents selected from the group consisting of an IL-4R activator, a TNFR activator, and an IFNGR activator as described herein. [0202] In some embodiments, the one or more of S/D/M factors comprises IL-10, IL-4, TNF^, and IFN^. IL-4 receptor (IL-4R) activator and IL-4 [0203] “IL-4 receptor (IL-4R) activator” as described herein refers to a molecule that activates the IL-4 receptor-mediated signaling pathway. [0204] Interleukin 4 (IL-4) is a cytokine that induces differentiation of naive helper T cells (Th0 cells) to Th2 cells. Upon activation by IL-4, Th2 cells subsequently produce additional IL-4 in a positive feedback loop. IL-4 is produced primarily by mast cells, Th2 cells, eosinophils, and basophils. IL-4 is closely related to and has functions similar to IL-13. Interleukin 4 has many biological roles, including the stimulation of activated B cell and T cell proliferation, and the differentiation of B cells into plasma cells. It is a key regulator in humoral and adaptive immunity. IL-4 induces B cell class switching to IgE, and up-regulates MHC class II production. IL-4 decreases the production of IL-12 by Th1 cells, macrophages, IFN^, and dendritic cells. IL-4 signaling determines the levels of CD20 on the surface of ny-2770598
Attorney Docket No.24516-20006.40 normal and malignant B lymphocytes via activation of transcription factor STAT6. Overproduction of IL-4 is associated with allergies. [0205] IL-4 as referred to herein comprises any constructs that have a component of IL-4 (e.g., a naturally occurring IL-4, e.g., a recombinant IL-4). These include and are not limited to natural IL-4 (e.g., various isoforms of human IL-4), synthetic or recombinant IL-4, and fusion proteins having an IL-4 component. [0206] In some embodiments, the IL-4R activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety. [0207] In some embodiments, the one or more of S/D/M factors comprises an IL-4R activator, optionally wherein the IL-4R activator is selected from the group consisting of IL- 4, IL-13, an IL-4R agonist antibody, and a small molecule activator of IL-4R. In some embodiments, the IL-4R activator is IL-4. In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 (e.g., a human IL-4) is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml (e.g., at least about 30 pg/ml, 50 pg/ml, 75 pg/ml, 100 pg/ml, 125 pg/ml, or 150 pg/ml). In some embodiments, the IL-4 (e.g., a human IL-4) is present in the medium at a concentration of about 15 pg/ml to about 1.5 ng/ml (e.g., about 30 pg/ml to about 1 ng/ml, e.g., about 100 pg/ml to about 1 ng/ml, e.g., about 100 pg/ml to about 1 ng/ml). [0208] In some embodiments, the IL-4R activator is IL-13 (such as a human IL-13 or a human recombinant IL-13). In some embodiments, the IL-13 is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 60 pg/ml, further optionally about 60 pg/ml to about 2 ng/ml (e.g., about 100 pg/ml to about 2 ng/ml). GM-CSF receptor (GM-CSFR) activator and GM-CSF [0209] “GM-CSF receptor (GM-CSFR) activator” as described herein refers to a molecule that activates the GM-CSFR-mediated signaling pathway. [0210] Granulocyte-macrophage colony-stimulating factor (GM-CSF), also known as colony-stimulating factor 2 (CSF2), is a monomeric glycoprotein secreted by macrophages, T cells, mast cells, natural killer cells, endothelial cells, and fibroblasts that functions as a cytokine. GM-CSF stimulates stem cells to produce granulocytes (neutrophils, eosinophils, and basophils) and monocytes. Monocytes exit the circulation and migrate into tissue, whereupon they mature into macrophages and dendritic cells. It is part of the immune/inflammatory cascade, by which activation of a small number of macrophages can ny-2770598
Attorney Docket No.24516-20006.40 rapidly lead to an increase in their numbers, a process crucial for fighting infection. GM-CSF also has some effects on mature cells of the immune system. These include, for example, enhancing neutrophil migration and causing an alteration of the receptors expressed on the cells’ surface. [0211] GM-CSF as referred to herein comprises any constructs that have a component of GM-CSF (e.g., a naturally occurring GM-CSF, e.g., a recombinant GM-CSF). These include and are not limited to natural GM-CSF (e.g., various isoforms of human GM-CSF), synthetic or recombinant GM-CSF, and fusion proteins having a GM-CSF component. [0212] In some embodiments, the GM-CSFR activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety. [0213] In some embodiments, the one or more of the S/D/M factors further comprises a GM- CSF receptor (GM-CSFR) activator. In some embodiments, the GM-CSFR activator is selected from the group consisting of GM-CSF, a GM-CSFR agonist antibody, and a small molecule activator of GM-CSFR. In some embodiments, the GM-CSFR activator is GM- CSF. In some embodiments, the GM-CSF is a human GM-CSF or a human recombinant GM-CSF. In some embodiments, the GM-CSF (e.g., a human GM-CSF) is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml (e.g., at least about 100 pg/ml, about 150 pg/ml, about 200 pg/ml, or about 300 pg/ml). In some embodiments, the GM-CSF (e.g., a human GM-CSF) is present in the medium at a concentration of about 30 pg/ml to about 3 ng/ml (e.g., about 50 pg/ml to about 1 ng/ml, e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 200 pg/ml to about 400 pg/ml, e.g., about 300 pg/ml). IL-6 receptor (IL-6R) activator and IL-6 [0214] “IL-6 receptor (IL-6R) activator” as described herein refers to a molecule that activates the IL-6 receptor-mediated signaling pathway. [0215] Interleukin 6 (IL-6) is an interleukin that acts as both a pro-inflammatory cytokine and an anti-inflammatory myokine. In the immune system, IL-6 is secreted by macrophages in response to specific microbial molecules, referred to as pathogen-associated molecular patterns (PAMPs). These PAMPs bind to an important group of detection molecules of the innate immune system, called pattern recognition receptors (PRRs), including Toll-like receptors (TLRs). These are present on the cell surface and intracellular compartments and induce intracellular signaling cascades that give rise to inflammatory cytokine production. IL- ny-2770598
Attorney Docket No.24516-20006.40 6 is an important mediator of fever and of the acute phase response. IL-6 is responsible for stimulating acute phase protein synthesis, as well as the production of neutrophils in the bone marrow. It supports the growth of B cells and is antagonistic to regulatory T cells. [0216] IL-6 as referred to herein comprises any constructs that have a component of IL-6 (e.g., a naturally occurring IL-6, e.g., a recombinant IL-6). These include and are not limited to natural IL-6 (e.g., various isoforms of human IL-6), synthetic or recombinant IL-6, and fusion proteins having an IL-6 component. [0217] In some embodiments, the IL-6R activator further comprises a moiety that enhances stability or half-life, including for example an Fc portion or a PEG moiety. [0218] In some embodiments, the one or more of the S/D/M factors further comprise an IL-6 receptor (IL-6R) activator, optionally wherein the IL-6R activator is selected from the group consisting of IL-6, an IL-6R agonist antibody, and a small molecule activator of IL-6R. In some embodiments, the IL-6R activator is IL-6. In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6. In some embodiments, the IL-6 (e.g., a human IL-6) is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml (e.g., at least about 10 pg/ml, about 15 pg/ml, about 20 pg/ml, or about 25 pg/ml). In some embodiments, the IL-6 (e.g., a human IL-6) is present in the medium at a concentration of about 1 pg/ml to about 300 pg/ml (e.g., about 5 pg/ml to about 100 pg/ml, e.g., about 10 pg/ml to about 50 pg/ml, e.g., about 20 pg/ml to about 40 pg/ml, e.g., about 30 pg/ml). [0219] In some embodiments, the one or more of S/D/M factors comprises IL-10, IL-4, TNF^, IFN^, GM-CSF, and IL-6. [0220] In some embodiments, the one or more of S/D/M factors described herein is present in a single composition. [0221] In some embodiments, the one or more of S/D/M factors described herein further comprises one or more cytokines selected from the group consisting of IL-2, IL-17, (e.g., IL- 17A), and/or M-CSF. [0222] In some embodiments, at least one of the one or more of S/D/M factors (e.g., IL-10) is provided separately from other S/D/M factors in the one or more of S/D/M factors. APC Refinement factors [0223] As discussed in the examples, the refinement factors are not required but optional to the methods discussed herein of promoting survival and/or differentiation of the monocytes ny-2770598
Attorney Docket No.24516-20006.40 (e.g., cancer monocytes) or the hematological cancer cells into potent antigen presenting cells (APCs) but can further refine the APCs to express even higher levels of surface markers (e.g., MHC-I, MHC-II, CD80, CD86, CD40) that indicate a more potent antigen presentation machinery. [0224] In some embodiments, the methods described above further comprise contacting the population of hematological cancer cells (e.g., AML cells, CLL cells, CML cells, NHL cells, or B-ALL cells) with one or more of refinement factors after the one or more of cancer cells are contacted with the one or more of S/D/M factors or the medium derived from the culture of T cells. The refinement factors are selected from the group consisting of type-I interferon (such as IFN^ and/or IFN^), IFN^, TNF^, a TLR ligand (such as R848, poly IC, CpG, or LPS), CD40L or a CD40-ligating antibody, an anti-PD-L1 antibody, and TPI-1. [0225] In some embodiments, there is provided a method of refining a population of APCs derived from monocytes (e.g., cancer monocytes), or hematological cancer cells, comprising contacting the population of APCs with a) IFN^, b) IFN^, and c) TNF^. [0226] In some embodiments, there is provided a method of refining a population of APCs derived from monocytes (e.g., cancer monocytes), or hematological cancer cells, comprising contacting the population of APCs with a) IFN^, b) IFN^, c) TNF^, d) poly IC, and e) CpG. [0227] In some embodiments, there is provided a method of refining a population of APCs derived from monocytes (e.g., cancer monocytes), or hematological cancer cells, comprising contacting the population of APCs with a) IFN^, b) IFN^, c) TNF^, d) poly IC, and e) R848. [0228] In some embodiments, there is provided a method of refining a population of APCs derived from monocytes (e.g., cancer monocytes), or hematological cancer cells, comprising contacting the population of APCs with a) IFN^, b) IFN^, c) TNF^, d) poly IC, e) CD40L, and f) anti-PD-L1 antibody. [0229] In some embodiments, there is provided a method of refining a population of APCs derived from monocytes (e.g., cancer monocytes), or hematological cancer cells, comprising contacting the population of APCs with a) IFN^, b) IFN^, c) TNF^, d) poly IC, e) CD40L, f) anti-PD-L1 antibody, and g) a SHP-1 inhibitor (e.g., TPI-1). [0230] In some embodiments, there is provided a method of refining a population of APCs derived from monocytes (e.g., cancer monocytes), or hematological cancer cells, comprising contacting the population of APCs with a) IFN^, b) IFN^, c) R848, d) poly IC, and e) a SHP- 1 inhibitor (e.g., TPI-1). ny-2770598
Attorney Docket No.24516-20006.40 [0231] In some embodiments, there is provided a method of refining a population of APCs derived from monocytes (e.g., cancer monocytes), or hematological cancer cells, comprising contacting the population of APCs with a) IFN^, b) R848, c) poly IC, and d) a SHP-1 inhibitor (e.g., TPI-1). [0232] In some embodiments, there is provided a method of refining a population of APCs derived from monocytes (e.g., cancer monocytes), or hematological cancer cells, comprising contacting the population of APCs with a) IFN^, b) IFN^, c) poly IC, d) CpG, e) CD40L, f) anti-PD-L1 antibody, g) a SHP-1 inhibitor (e.g., TPI-1), and h) TNF^. [0233] In some embodiments, there is provided a method of refining a population of APCs derived from monocytes (e.g., cancer monocytes), or hematological cancer cells, comprising contacting the population of APCs with a) R848, and b) poly IC. [0234] In some embodiments, the one or more of refinement factors is provided immediately after the plurality of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B- ALL cells) is contacted with the one or more of S/D/M factors or the medium derived from the culture of T cells. In some embodiments, the one or more of refinement factors is provided within about one day after the plurality of cancer cells is contacted with the one or more of S/D/M factors or the medium derived from the culture of T cells. [0235] In some embodiments, the plurality of the cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) is cultured for about 1-5 days (e.g., for about one, two, three, four or five days) in the presence of the one or more of refinement factors. [0236] In some embodiments, the one or more of refinement factors is provided when at least about 50% (e.g., about 50%, 60%, 70%, 80%, or 99%) of the cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) survive after the plurality of cancer cells is contacted with the one or more of S/D/M factors or the medium derived from the culture of T cells. [0237] In some embodiments, the one or more of refinement factors is provided when at least about 10%, 20%, 30%, 40% or 50% of the cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) exhibit a dendritic cell morphology. [0238] In some embodiments, the one or more of refinement factors is provided when cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) express a high level of one or more molecules selected from the group consisting of MHC I, MHC II, CD80, CD86, and/or CD40. In some embodiments, the one or more of refinement factors is provided ny-2770598
Attorney Docket No.24516-20006.40 when cancer cells express a higher (e.g., at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%) level of one or more molecules selected from the group consisting of MHC-I, MHC-II, CD80, CD86, and/or CD40 than corresponding cells obtained from the same individual and cultured with GM-CSF and M-CSF (e.g., at a concentration routinely used in the field for such methods). [0239] In some embodiments, the one or more of refinement factors described herein can be used independently from the methods described above for priming APCs (e.g., APCs obtained from a human). [0240] In some embodiments, the one or more of refinement factors comprises Poly IC or R848. In some embodiments, the one or more of refinement factors comprise Poly IC and R848. [0241] In some embodiments, the one or more of refinement factors comprise IFN^, IFN^, and TNF^. [0242] In some embodiments, the one or more of refinement factors comprise IFN^, IFN^, TNF^, poly IC, and CpG. In some embodiments, the one or more of refinement factors are comprised in a medium comprising serum. [0243] In some embodiments, the one or more of refinement factors comprise IFN^, IFN^, TNF^, poly IC, CpG, CD40L, and an anti-PD-L1 antibody. [0244] In some embodiments, the one or more of refinement factors comprise IFN^, IFN^, TNF^, poly IC, CpG, CD40L, an anti-PD-L1 antibody, and TPI-1. [0245] In some embodiments, the r one or more of efinement factors are comprised in a medium comprising serum (e.g., human serum). [0246] In some embodiments, the concentration of IFN^ in the one or more of refinement factors is about 1ng/ml to about 50 ng/ml (e.g., about 5 ng/ml to about 20 ng/ml, e.g., about 10 ng/ml). [0247] In some embodiments, the concentration of IFN^ in the one or more of refinement factors is about 5 ng/ml to about 500 ng/ml (e.g., about 10 ng/ml to about 250 ng/ml, e.g., about 20 ng/ml to about 100 ng/ml, e.g., about 50 ng/ml). [0248] In some embodiments, the concentration of TNF^ in the one or more of refinement factors is about 1ng/ml to about 50 ng/ml (e.g., about 5 ng/ml to about 20 ng/ml, e.g., about 10 ng/ml). ny-2770598
Attorney Docket No.24516-20006.40 [0249] In some embodiments, the concentration of poly IC in the one or more of refinement factors is about 0.1µg/ml to about 10 µg/ml (e.g., about 0.2 µg/ml to about 5 µg/ml, e.g., about 0.5 µg/ml to about 2.5 µg/ml, e.g., about 1 µg/ml). [0250] In some embodiments, the concentration of CpG in the one or more of refinement factors is about 0.1µg/ml to about 10 µg/ml (e.g., about 0.2 µg/ml to about 5 µg/ml, e.g., about 0.5 µg/ml to about 2.5 µg/ml, e.g., about 1 µg/ml). [0251] In some embodiments, the concentration of CD40L in the one or more of refinement factors is about 1µg/ml to about 100 µg/ml (e.g., about 2 µg/ml to about 50 µg/ml, e.g., about 5 µg/ml to about 20 µg/ml, e.g., about 10 µg/ml). [0252] In some embodiments, the concentration of the anti-PD-L1 antibody in the one or more of refinement factors is about 1µg/ml to about 200 µg/ml (e.g., about 5 µg/ml to about 100 µg/ml, e.g., about 10 µg/ml to about 50 µg/ml, e.g., about 20 µg/ml). [0253] In some embodiments, the concentration of TPI-1 in the one or more of refinement factors is about 0.1µg/ml to about 10 µg/ml (e.g., about 0.2 µg/ml to about 5 µg/ml, e.g., about 0.5 µg/ml to about 2.5 µg/ml, e.g., about 1 µg/ml). [0254] In some embodiments, the concentration of R848 in the one or more of refinement factors is about 0.1µg/ml to about 10 µg/ml (e.g., about 0.2 µg/ml to about 5 µg/ml, e.g., about 0.5 µg/ml to about 2.5 µg/ml, e.g., about 1 µg/ml). Methods for promoting survival of hematological cancer cells (e.g., hematological cancer cells from an individual) and/or HC-APCs [0255] The present application provides various methods for promoting survival of hematological cancer cells (e.g., hematological cancer cells from an individual) and/or HC- APCs. In some embodiments, the cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) are obtained (e.g., freshly isolated) from an individual (e.g., a human). It was observed that the hematological cancer cells obtained from an individual (e.g., PBMC of an individual) generally could not survive more than 1-2 days. The methods described herein promote the survival of these cells while they differentiate into APCs and/or promote the survival of these HC-APCs. [0256] In some embodiments, the cancer cells obtained from the individual express a lower level of IL-10 receptor (“IL-10R”), IL-4 receptor (“IL-4R”), IL-6 receptor (“IL-6R”), M-CSF receptor (“GM-CSFR”), and/or M-CSF receptor (“GM-CSFR”) as compared to those corresponding cells obtained from a reference individual (e.g., a healthy individual). ny-2770598
Attorney Docket No.24516-20006.40 [0257] In some embodiments, the present application provides a method of promoting the survival of a population of hematological cancer cells (e.g., hematological cancer cells from an individual) and/or HC-APCs from an individual in an in vitro culture, comprising cultivating the population of cancer cells in a medium having an IL-10R activator, optionally wherein the IL-10R activator is selected from the group consisting of: an IL-10 (e.g., a pegylated IL-10, e.g., pegilodecakin or AM0010), an IL-10 family member (e.g., IL-19, IL- 20, IL-22, IL-24, IL-26, IL-28), an IL-10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, a small molecule activator of IL-10R, and an activator of the IL- 10R-downstream STAT3 (e.g., IL-12, e.g., Long noncoding RNA (LncRNA) PVT1, NEAT1, FEZF1-AS1, UICC). In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the culture further comprises a TNF^ receptor (TNFR) activator, and/or an interferon ^ (IFN^) receptor (IFNGR) activator, optionally wherein the TNFR activator is selected from the group consisting of TNF^, a TNFR agonist antibody, and a small molecule activator of TNFR, optionally wherein the IFNGR activator is selected from the group consisting of IFN^, an IFNGR agonist antibody, and a small molecule activator of IFNGR, and further optionally wherein the culture comprises TNF^ and/or IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the hematological cancer is a myeloid leukemia, a B-cell lymphoma or B-cell leukemia. In some embodiments, the hematological cancer cells comprise AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells. [0258] In some embodiments, the present application provides a method of promoting the survival of a population of cancer cell-derived APCs from an individual in an in vitro culture, comprising cultivating the population of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) in a medium having a TNF^ receptor (TNFR) activator, and/or an interferon ^ (IFN^) receptor (IFNGR) activator, optionally wherein the TNFR activator is selected from the group consisting of TNF^, a TNFR agonist antibody, and a small molecule activator of TNFR, optionally wherein the IFNGR activator is selected from the group ny-2770598
Attorney Docket No.24516-20006.40 consisting of IFN^, an IFNGR agonist antibody, and a small molecule activator of IFNGR, and further optionally the culture comprises TNF^ and/or IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the hematological cancer is a myeloid leukemia, a B-cell lymphoma or B-cell leukemia. In some embodiments, the hematological cancer cells comprise AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells. [0259] In some embodiments, the present application provides a method of promoting the survival of a population of cancer cell-derived APCs from an individual in an in vitro culture, comprising cultivating the population of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) in a medium having IL-10, TNF^, and IFN^. In some embodiments, the IL-10 is a human IL-10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the hematological cancer is a myeloid leukemia, a B-cell lymphoma or B-cell leukemia. In some embodiments, the hematological cancer cells comprise AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells. [0260] In some embodiments, the culture further comprises a GM-CSF receptor (GM-CSFR) activator. In some embodiments, the GM-CSFR activator is selected from the group consisting of GM-CSF, a GM-CSFR agonist antibody, and a small molecule activator of GM- CSFR. In some embodiments, the GM-CSFR activator is GM-CSF. In some embodiments, the GM-CSF is a human GM-CSF or a human recombinant GM-CSF. In some embodiments, the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml). ny-2770598
Attorney Docket No.24516-20006.40 [0261] In some embodiments, the culture further comprises an IL-6 receptor (IL-6R) activator. In some embodiments, the IL-6R activator is selected from the group consisting of IL-6, an IL-6R agonist antibody, and a small molecule activator of IL-6R. In some embodiments, the IL-6R activator is IL-6. In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6. In some embodiments, the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml). [0262] In some embodiments, the present application provides a method of promoting the survival of a population of cancer cell-derived APCs from an individual in an in vitro culture, comprising cultivating the population of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) in a medium derived from a culture of T cells after being treated with anti-CD3 and anti-CD28 antibodies, wherein the medium comprises an activator of IL- 10R. In some embodiments, the T cells are CD4 T cells. In some embodiments, the T cells are CD8 T cells. In some embodiments, the T cells are isolated from PBMC of the same individual or a different individual and have not been previously treated with anti-CD3 and/or anti-CD28 antibodies prior to the treatment. In some embodiments, the T cells are isolated from PBMC of the same individual or a different individual and have been previously treated with anti-CD3 and/or anti-CD28 antibodies prior to the treatment. In some embodiments, the medium is derived from the culture after the T cells are treated with anti-CD3 and anti-CD28 antibodies for about 1-3 days, optionally for about 2 days. In some embodiments, the individual has a cancer (e.g., a hematological cancer). [0263] The anti-CD3/CD28 treatment for T cells described herein are techniques well known in the field for activating T cells. [0264] The present application further provides a method of increasing expression of IL-10 receptor (IL-10R) in a population of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) from an individual having cancer, comprising contacting the population of cancer cells with one or more agents selected from the group consisting of: an IL-10R activator, a TNFR activator, and an IFNGR activator. In some embodiments, the IL- 10R activator is selected from the group consisting of: an IL-10 (e.g., a pegylated IL-10, e.g., pegilodecakin or AM0010), an IL-10 family member (e.g., IL-19, IL-20, IL-22, IL-24, IL-26, IL-28), an IL-10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, a small molecule activator of IL-10R, and an activator of the IL-10R-downstream STAT3 (e.g., IL-12, e.g., Long noncoding RNA (LncRNA) PVT1, NEAT1, FEZF1-AS1, UICC). In some ny-2770598
Attorney Docket No.24516-20006.40 embodiments, the IL-10R activator is IL-10. In some embodiments, the IL-10 is a human IL- 10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). In some embodiments, the TNFR activator is selected from the group consisting of TNF^, a TNFR agonist antibody, and a small molecule activator of TNFR. In some embodiments, the IFNGR activator is selected from the group consisting of IFN^, an IFNGR agonist antibody, and a small molecule activator of IFNGR. In some embodiments, the IFNGR activator comprises TNF^. In some embodiments, the IFNGR activator comprises IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the individual has a cancer (e.g., a hematological cancer). [0265] In some embodiments, the cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) are cultured for at least about 2 days (e.g., about 2-3 days). [0266] In some embodiments, the population of cancer cells express a lower level (at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45% or 50% lower) of IL-10R prior to contacting with the IL-10R activator as compared to corresponding cells obtained from a reference individual (e.g., a healthy individual). Methods for promoting the differentiation of a population of hematological cancer cells to antigen presenting cells (“APCs”) [0267] The present application provides a method of promoting the differentiation of a population of hematological cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) from an individual (e.g., a cancer patient) to antigen presenting cells (“APCs”) in an in vitro culture, comprising cultivating the population of hematological cancer cells in a medium having one or more molecules selected from the group consisting of an IL-4 receptor (IL-4R) activator (e.g., IL-4), a TNF^ receptor (TNFR) activator (e.g., TNF^), and an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^), optionally wherein the cancer cells have been contacted with a medium that further comprises an IL-10 receptor (IL-10R) activator (e.g., IL-10, e.g., IL-12). In some embodiments, there is provided a method of promoting the differentiation of a population of hematological cancer cells from ny-2770598
Attorney Docket No.24516-20006.40 an individual (e.g., a cancer patient) to antigen presenting cells (“APCs”) in an in vitro culture, comprising cultivating the population of cancer cells in a medium having an IL-4 receptor (IL-4R) activator (e.g., IL-4), a TNF^ receptor (TNFR) activator (e.g., TNF^), and an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., IFN^). In some embodiments, the IL- 4R activator is selected from the group consisting of IL-4, IL-13, an IL-4R agonist antibody, and a small molecule activator of IL-4R. In some embodiments, the IL-4R activator is IL-4. In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the TNFR activator is selected from the group consisting of TNF^, a TNFR agonist antibody, and a small molecule activator of TNFR. In some embodiments, the TNFR activator is TNF^. In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IFNGR activator is selected from the group consisting of IFN^, an IFNGR agonist antibody, and a small molecule activator of IFNGR. In some embodiments, the IFNGR activator is IFN^. In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the culture further comprises an IL-6 receptor (IL-6R) activator. In some embodiments, the IL- 6R activator is selected from the group consisting of IL-6, an IL-6R agonist antibody, and a small molecule activator of IL-6R. In some embodiments, the IL-6R activator is IL-6. In some embodiments, the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml). [0268] In some embodiments, there is provided a method of promoting the differentiation of a population of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) from an individual (e.g., a cancer patient) to antigen presenting cells (“APCs”) in an in vitro culture, comprising cultivating the population of cancer cells in a medium having IL-4, TNF^, and IFN^. In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least ny-2770598
Attorney Docket No.24516-20006.40 about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). [0269] In some embodiments, there is provided a method of promoting the differentiation of a population of cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) from an individual (e.g., a cancer patient) to antigen presenting cells (“APCs”) in an in vitro culture, comprising cultivating the population of cancer cells in a medium having IL-4, IL-6, TNF^, and IFN^. In some embodiments, the IL-4 is a human IL-4 or a human recombinant IL-4. In some embodiments, the IL-4 is present in the medium at a concentration of at least about 15 pg/ml, optionally at least about 30 pg/ml, further optionally about 30 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 1 ng/ml). In some embodiments, the TNF^ is a human TNF^ or a human recombinant TNF^. In some embodiments, the TNF^ is present in the medium at a concentration of at least about 0.5 ng/ml, optionally at least about 1 ng/ml, further optionally about 0.5 ng/ml to about 30 ng/ml (e.g., about 1-10 ng/ml). In some embodiments, the IFN^ is a human IFN^ or a human recombinant IFN^. In some embodiments, the IFN^ is present in the medium at a concentration of at least about 5 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 50-100 ng/ml). In some embodiments, the IL-6 is a human IL-6 or a human recombinant IL-6. In some embodiments, the IL-6 is present in the medium at a concentration of at least about 1 pg/ml, optionally at least about 5 pg/ml, further optionally about 5 pg/ml to about 100 pg/ml (e.g., about 10-50 pg/ml, e.g., about 30 pg/ml). [0270] In some embodiments, the culture further comprises a GM-CSF receptor (GM-CSFR) activator. In some embodiments, the GM-CSFR activator is selected from the group consisting of GM-CSF, a GM-CSFR agonist antibody, and a small molecule activator of GM- CSFR. In some embodiments, the GM-CSFR activator is GM-CSF. In some embodiments, the GM-CSF is a human GM-CSF or a human recombinant GM-CSF. In some embodiments, the GM-CSF is present in the medium at a concentration of at least about 30 pg/ml, ny-2770598
Attorney Docket No.24516-20006.40 optionally at least about 50 pg/ml, further optionally about 100 pg/ml to about 1 ng/ml (e.g., about 100 pg/ml to about 500 pg/ml, e.g., about 300 pg/ml). [0271] In some embodiments, the culture further comprises a STAT3 activator, such as an IL-10 receptor (IL-10R) activator. In some embodiments, the IL-10R activator is selected from the group consisting of: an IL-10 (e.g., a pegylated IL-10, e.g., pegilodecakin or AM0010), an IL-10 family member (e.g., IL-19, IL-20, IL-22, IL-24, IL-26, IL-28), an IL- 10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, a small molecule activator of IL-10R, and an activator of the IL-10R-downstream STAT3 (e.g., IL-12, e.g. Long noncoding RNA (LncRNA) PVT1, NEAT1, FEZF1-AS1, UICC). In some embodiments, the IL-10R activator is IL-10. In some embodiments, the IL-10 is a human IL- 10 or a human recombinant IL-10. In some embodiments, the IL-10 is present in the medium at a concentration of at least about 2 ng/ml, optionally at least about 10 ng/ml, further optionally about 10 ng/ml to about 200 ng/ml (e.g., about 20 ng/ml). [0272] In some embodiments, the hematological cancer cells obtained from the individual express a lower (e.g., at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45% or 50% lower) level of IL-10R or IL-4 receptor (“IL-4R”) as compared to those corresponding cells obtained from a reference individual (e.g., a healthy individual). [0273] In some embodiments, the hematological cancer cells (e.g., AML cells, CML cells, CLL cells, NHL cells, or B-ALL cells) are cultured for at least about 2 days (e.g., about 2-3 days). Loading HC-APCs with additional antigens [0274] In some embodiments, the methods described herein further comprise loading one or more additional antigens (e.g., tumor-associated antigen peptides, neoantigen peptides) on the HC-APCs. [0275] In some embodiments, the antigen comprises shared tumor-associated antigens. In some embodiments, the shared tumor-associated antigens comprise self-antigens (e.g., abnormally expressed self-antigens). In some embodiments, the shared tumor-associated antigens comprise non-self-antigens of viral origins (e.g., antigens from LMP1/2 associated with nasopharyngeal carcinoma and lymphoma, e.g., antigens from E6 and E7 proteins of high-risk human papillomavirus (HPV), e.g., antigens from retrovirus Tax protein found in adult T cell leukemia). In some embodiments, the shared tumor-associated antigens comprise ny-2770598
Attorney Docket No.24516-20006.40 mutation-caused neoantigens shared in different types of cancer (e.g., neoantigens associated with p53 mutations or KRAS mutations). Tumor-associated antigen peptides [0276] Various approaches are available to identify tumor-associated antigen peptides. [0277] One approach often employed to identify the peptides recognized by such CTL is expression cloning, which consists of isolating the peptide-encoding gene by transfecting a library of tumoral cDNA and testing the transfected cells for their ability to activate the CTL clone. Fragments of the identified gene can then be transfected to define the region encoding the antigenic peptide, and finally candidate peptides bearing adequate HLA-binding motifs are tested for their ability to sensitize target cells to lysis by the CTL. This approach was successfully used to identify a large number of antigenic peptides. [0278] Nowadays, tumor-associated antigenic peptides are often identified using the “reverse immunology” approach, which consists in selecting peptides with adequate HLA-binding motifs inside a protein of interest, such as proteins encoded by mutated oncogenes or genes that are either selectively expressed or overexpressed by tumors. Candidate peptides are synthesized and tested for HLA binding in vitro. The most efficient binders are pulsed onto antigen-presenting cells, which are used to stimulate T lymphocytes in vitro, in order to derive CTL lines or clones that recognize peptide-pulsed target cells. A drawback of this approach is that the identified peptides might not be processed efficiently by tumors. It is therefore essential to verify that the CTL do recognize tumor cells that naturally express the peptide-encoding gene. Additionally, one should test transfectants that express normal levels of the gene or cells where expression of the gene has been knocked down using siRNAs or shRNAs. [0279] A third approach to antigen identification is based on the elution of antigenic peptides from MHC class I molecules immunopurified from the surface of tumor cells. The direct identification by mass spectrometry of the sequence of the eluted peptides is technically demanding but proved useful to identify or to confirm the relevance of peptides that have undergone posttranslational modifications such as serine/threonine phosphorylation, glycosylation-dependent asparagine deamidation, or peptide splicing. [0280] A large number of antigenic peptides recognized by antitumor CTL have been identified using these various approaches. These antigens are conveniently classified according to the expression pattern of the parent gene. A regularly updated database of those ny-2770598
Attorney Docket No.24516-20006.40 antigenic peptides effectively presented by tumor cells can be found on the http://www.cancerimmunity.org/ website. See Vigneron, Biomed Res Int.2015; 2015: 948501. Neoantigen peptides [0281] Various methods are available to detect and screen neoantigens. Sandwich immunoassays in the miniaturized system could successfully identify tumor antigens in serum samples extracted from patients. See e.g., Pollard et al., Proteomics Clin. Appl.1934– 952 (2007); Yang et al., Biosens. Bioelectron.40385–392 (2013). Another tool named Serologic Proteome analysis (SERPA) or 2-D western blots, consists of the isoelectric focusing (IEF) gel run in the first dimension and SDS-PAGE gel run in the second dimension. SERPA separates the proteins in the gel by their isoelectric point (IP) and molecular mass and then transfers the proteins from the gel to a carrier membrane to screen antibodies. Finally, the antigenic protein spots can be identified by MS. See e.g., Tjalsma et al., Proteomics Clin. Appl.2167–180 (2008). This approach has been used to identify antigens in different tumor types. Serological analysis of recombinant cDNA expression libraries (SEREX), which combines serological analysis with antigen cloning techniques, is a widely used technique to explore tumors’ antigen repertoire. SEREX first construct a cDNA library from cancer cell lines or fresh tumor samples, then screen the cDNA library with autologous sera of cancer patients, and finally sequence the immune-reactive clones. SEREX have identified a variety of tumor antigens including CTAs, differentiation antigens, mutational antigens, splice-variant antigens, and overexpressed antigens. See e.g., Chen et al., Proc. Natl. Acad. Sci. U.S.A.941914–1918 (1997). Furthermore, other methods such as Multiple Affinity Protein Profiling (MAPPing) and nanoplasmonic biosensors have also been developed to identify tumor antigens. See e.g., Lee et al., Biosens. Bioelectron.74341–346 (2015). [0282] In some embodiments, the one or more neoantigenic peptides described herein are obtained from a neoantigenic database (such as any of the neoantigenic databases described herein). For example, Tan et al. constructed a manually curated database (“dbPepNeo”) for human tumor neoantigen peptides based upon the four criteria as described below: (i) peptides were isolated from human tumor tissues or cell lines, (ii) peptides contained non- synonymous mutations in amino acid sequence, (iii) peptides can be bound by HLA-I molecules, and (iv) peptides can induce CD8+ T cell responses. See Tan et al., Database (Oxford).2020 Jan 1;2020:baaa004. Xia et al. constructed another database, NEPdb, which ny-2770598
Attorney Docket No.24516-20006.40 provides pan-cancer level predicted HLA-I neoepitopes derived from 16,745 shared cancer somatic mutations, using state-of-the-art predictors. See Xia et al., Front Immunol.2021; 12: 644637. Wu et al. developed a comprehensive tumor-specific neoantigen database (TSNAdb v1.0), based on pan-cancer immunogenomic analyses of somatic mutation data and human leukocyte antigen (HLA) allele information for 16 tumor types with 7748 tumor samples from The Cancer Genome Atlas (TCGA) and The Cancer Immunome Atlas (TCIA). See Wu et al., Genomics Proteomics Bioinformatics.2018 Aug;16(4):276-282. [0283] In some embodiments, the one or more neoantigenic peptides are obtained from analyzing the biological information of the individual (such as a patient who had a cancer). In some embodiments, the one or more neoantigenic peptides are obtained from a computational analysis of a cancer patient’s tumor genome. See e.g., Roudko et al. Front Immunol.2020; 11: 27. In some embodiments, the one or more neoantigenic peptides are obtained from a computational analysis of a cancer patient’s transcriptome. See e.g., Caushi et al., Nature.2021 Aug;596(7870):126-132. In some embodiments, the one or more neoantigenic peptides are obtained from a computational analysis of a cancer patient’s proteome. See e.g., Wen et al. Nat Commun.2020 Apr 9;11(1):1759. [0284] In some embodiments, the one or more neoantigenic peptides are selected based upon patient data. In some embodiments, the patient data are derived from data from a group of patients having a particular type of cancer (e.g., any of the cancers described here). In some embodiments, the patient data are derived from data from a group of patients having any cancer. In some embodiments, the group of patients are from the same sex (e.g., male or female). In some embodiments, the group of patients are from the same ethnicity. In some embodiments, the group of patients bear one or more biomarkers (e.g., an aberration in a particular gene, e.g., KRAS, e.g., PTEN). [0285] In some embodiments, the one or more neoantigenic peptides are derived from any polypeptide known to or have been found to contain a tumor-specific mutation. Suitable polypeptides from which the one or more neoantigenic peptides can be derived can be found for example in various databases available in the field (e.g., COSMIC database). These databases curate comprehensive information on somatic mutations in human cancer. In some embodiments, the one or more neoantigenic peptides contains a tumor-specific mutation. In some embodiments, the tumor-specific mutation is a driver mutation for a particular cancer type. ny-2770598
Attorney Docket No.24516-20006.40 [0286] In some embodiments, the tumor-associated peptides (e.g., neoantigen peptides) are synthetic peptides. In some embodiments, the one or more neoantigenic peptides are obtained by exome high throughput sequencing and prescreened with epitope prediction algorithms. [0287] In some embodiments, the one or more neoantigenic peptides are selected based upon its binding affinity to an MHC molecule (e.g., an MHC-I molecule and/or an MHC-II molecule). In some embodiments, the one or more neoantigenic peptides has a binding affinity that is less than 5000 nM (e.g., less than 500 nM, less than 250 nM, less than 100 nM or less than 50 nM) (IC50) to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity of about 500 nM to 5000 nM (IC50) to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity that is less than 500 nM (IC50) to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity of about 250 nM to 500 nM IC50 to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity that is less than 250 nM (IC50) to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity that is less than 100 nM (IC50) to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity of about 50 nM to 500 nM IC50 to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity that is less than 50 nM (IC50) to an MHC molecule. In some embodiments, the one or more neoantigenic peptides has a binding affinity of about 1 nM to 50 nM IC50 to an MHC molecule. [0288] In some embodiments, a plurality of tumor-associated peptides (e.g., neoantigen peptides) are prepared from a surgical resection of tumor tissue or a biopsy extract thereof. [0289] In some embodiments, a plurality of tumor-associated peptides (e.g., neoantigen peptides) are prepared from a mixture of tumor cells or extract thereof isolated from tumor tissue or a biopsy extract thereof. [0290] In some embodiments, a plurality of tumor-associated peptides (e.g., neoantigen peptides) are prepared from a mixture of isolated tumor-associated peptides (e.g., neoantigen peptides). [0291] In some embodiments, the tumor tissue or cell described above is a fresh tumor tissue or cell. In some embodiments, the tumor tissue or cell is obtained from a frozen sample. ny-2770598
Attorney Docket No.24516-20006.40 [0292] In some embodiments, the tumor tissue or cell has been subjected to an induction of immunogenic cell death (e.g., freeze-thaw to lyse tumor cells, high dose UV irradiation, X- ray radiation). [0293] In some embodiments, the tumor tissue or cell has been subjected to a radiation treatment. [0294] The present application provides APCs produced by any of the methods described here. Hematological Cancer [0295] Cancer described in this section (e.g., in the context of HC-APCs derived from cancer cells isolated from a cancer patient) can be a hematological cancer type or kind and derived from cells of the myeloid or the lymphoid lineage. All the cancer types discussed in Section V are similarly applicable here. [0296] Hematological cancer cells include cancerous cells of the immune system or in blood- forming tissue, such as the bone marrow. Non-limiting examples include lymphoma, myeloma, and leukemia. Lymphomas derive from the lymph system (e.g., lymphocytes) and include Hodgkin’s and non-Hodgkin’s lymphomas (NHL). Myelomas derive from plasma cells, which terminally differentiate from activated B cells. Leukemias derive from blood cells and bone marrow, such as from lymphocytes (e.g., B cell acute lymphocytic leukemia (B-ALL)) or myeloid cells (e.g., acute myeloid leukemia (AML) or chronic myeloid leukemia (CML)) and can be chronic or acute. [0297] In some embodiments, the hematological cancer is a lymphoma. In some embodiments, the hematological cancer is a non-Hodgkin’s lymphoma. In some embodiments, the hematological cancer is a Burkitt’s lymphoma, an anaplastic large cell lymphoma, or a splenic marginal zone lymphoma. [0298] In some embodiments, the hematological cancer is a leukemia. In some embodiments, the hematological cancer is an acute lymphocytic leukemia (ALL), a chronic lymphocytic leukemia (CLL), an acute myelogenous leukemia (AML), an acute megakaryoblastic leukemia (AMKL), a chronic idiopathic myelofibrosis (MF), a chronic myelogenous leukemia (CML), a B-cell prolymphocytic leukemia (B-PLL), a chronic neutrophilic leukemia, a hairy cell leukemia (HCL), or an aggressive NK-cell leukemia. ny-2770598
Attorney Docket No.24516-20006.40 [0299] In some embodiments, the hematological cancer is an advanced cancer. In some embodiments, the hematological cancer is a late-stage cancer. In some embodiments, the hematological cancer is stage II, III, or IV. In some embodiments, the cancer is malignant. [0300] Examples of hematological cancers described herein include, but are not limited to, acute lymphoblastic leukemia, chronic myelogenous leukemia, Hodgkin’s lymphoma, non- Hodgkin’s lymphoma, leukemia, B-cell chronic lymphocytic leukemia, acute myeloid leukemia, AIDS-related cancers (e.g., AIDS-related lymphoma), central nervous system lymphoma, chronic myeloproliferative disorders, leukemia, lymphoid neoplasm (e.g., lymphoma), myelodysplastic syndromes, myelodysplastic/myeloproliferative diseases, lymphoma, primary central nervous system lymphoma (microglioma), and post-transplant lymphoproliferative disorder (PTLD). [0301] In some embodiments, the hematological cancer is a virus-infection-related cancer. In some embodiments, the hematological cancer is human T-lymphotrophic virus (HTLV-1)- related cancer (e.g., adult T cell leukemia or lymphoma). In some embodiments, the hematological cancer is Epstein-Barr virus (EBV)-related cancer (e.g., Burkitt lymphoma, Hodgkin’s lymphoma, or non-Hodgkin’s lymphoma). In some embodiments, the hematological cancer is hepatitis C virus-related cancer (e.g., non-Hodgkin’s lymphoma). [0302] In some embodiments, the hematological cancer is refractory to one or more of irradiation therapy, chemotherapy, or immunotherapy (e.g., checkpoint blockade). [0303] In some embodiments, the hematological cancer is B-ALL, CLL, NHL, or AML. In some embodiments, HC-APCs are derived from AML cells of any of M0, M1, M2, M3, M4, or M5 subtypes. B cell acute lymphoblastic leukemia cells (B-ALL) [0304] The methods described herein convert a plurality of B cell acute lymphoblastic leukemia (B-ALL) cells into HC-APCs. In some embodiments, the plurality of B-ALL cells is obtained from the peripheral blood of the individual (e.g., human cancer patient). [0305] Various markers of B-ALL cells have been identified and described, including at different stages of development. For example, early pre B-ALL has been found to present with the phenotype of TdT+CD19+CD10-. Common B-ALL displays CD19+CD10+CALLA+. Pre B-ALL cells are CD10+/-CD19+HLA-DR+cytoplasmic IgM+. Mature B-ALL cells express CD10+CD19+CD20+CD22+surface IgM+. Additional markers of the B-ALL cell lineages are known in the art. ny-2770598
Attorney Docket No.24516-20006.40 [0306] In some embodiments, the B-ALL cells express CD19 and one or more of the markers selected from the group consisting of CD10, CD21, CD22, CD23, CD24, CD79a, CALLA, TdT, HLA-DR, and cytoplasmic IgM at the time when they are obtained from the peripheral blood. Methods of obtaining B-ALL cells from peripheral blood is well known in the art, for example by seeding peripheral blood cells onto an anti-CD19 antibody-coated plate. B-ALL cells can be separated by positive selection with (e.g.) anti-CD19 antibody and one or more antibodies targeting markers selected from the group consisting of CD10, CD21, CD22, CD23, CD24, CD79a, CALLA, TdT, HLA-DR, and cytoplasmic IgM, or by negative selection using all antibodies against other cells using magnetic-activated cell sorting (MACS). Further methods of B-ALL cell selection include surface marker staining and cell sorting by fluorescence activated cell sorting (FACS). [0307] The B-ALL cells obtained from the individual (e.g., human cancer patient) in some embodiments express a lower level of IL-10 receptor (“IL-10R”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a reference individual (e.g., a healthy individual). In some embodiments, the level of IL-10R on the B-ALL cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-10R on corresponding cells from the reference individual (e.g., a healthy individual). [0308] In some embodiments, the B-ALL cells obtained from the individual (e.g., human cancer patient) express a lower level of IL-4 receptor (“IL-4R”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual. In some embodiments, the level of IL-4R on the B-ALL cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-4R on corresponding cells from the reference individual (e.g., a healthy individual). [0309] In some embodiments, the B-ALL cells obtained from the individual (e.g., human cancer patient) express a lower level of IL-6 receptor (“IL-6R”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual. In some embodiments, the level of IL-6R on the B-ALL cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-6R on corresponding cells from the reference individual (e.g., a healthy individual). ny-2770598
Attorney Docket No.24516-20006.40 [0310] In some embodiments, the B-ALL cells obtained from the individual (e.g., human cancer patient) express a lower level of M-CSF receptor (“M-CSFR”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual. In some embodiments, the level of M-CSFR on the B-ALL cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, or 65% lower than the level of M-CSFR on corresponding cells from the reference individual (e.g., a healthy individual). [0311] In some embodiments, the B-ALL cells obtained from the individual (e.g., human cancer patient) express a lower level of GM-CSF receptor (“GM-CSFR”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual. In some embodiments, the level of GM-CSFR on the B-ALL cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, or 65% lower than the level of GM-CSFR on corresponding cells from the reference individual (e.g., a healthy individual). Acute myeloid leukemia cells [0312] The methods described herein convert a plurality of acute myeloid leukemia (AML) cells into HC-APCs. In some embodiments, the plurality of AML cells is obtained from the peripheral blood of the individual (e.g., human cancer patient). [0313] Various markers of AML cells and of leukemia stem cells (LSCs) have been identified and described. The eight subtypes of AML are described in the Hematological Cancer section below. For example, early studies of human AML cases found that up to 96% of human AML cases expressed CD13 and/or CD33. Similarly, CD14 was identified in 82- 90% of M4 and M5 AML cases. (See, e.g., Kumar et al., Med J Armed Forces India 1995, 51(3):165-169.) Leukemia stem cells that differentiate into AML cells have been found to present with the surface marker profile of CD34+CD38-CD123+TIM3+ and can include additional markers. (See, e.g., Ding et al., Stem Cell Investig.2017, 4:19.) CD33, CD123, CLL1, TIM3 and CD244 were ubiquitously expressed on AML bulk cells at initial diagnosis and relapse, and CD33/TIM3 and CLL1/TIM3 have been shown to be highly positive in AML compared with normal hematopoiesis and non-hematopoietic tissues (Haubner et al., Leukemia 2019, 33:64-74). Further markers of the LSC and/or AML cell lineage are known in the art. [0314] Specifically, the eight subtypes, i.e., M0-M7 subtypes, are described as below. ny-2770598
Attorney Docket No.24516-20006.40 [0315] M0 subtype is also called acute dedifferentiated leukemia, acute minimally differentiated leukemia, acute myeloid leukemia with minimal differentiation, or acute myelogenous leukemia with minimal differentiation. AML-M0 is a rare subtype in which blasts fail to show morphologic differentiation by light microscopy, and conventional cytochemical stains and myeloid markers are negative. These blasts are CD45 dim and express the primitive hematopoietic markers CD34, CD38 and HLA Dr. One or more of the following pan-myeloid markers are expressed: CD13, CD33, and/or CD117. AML-M0 constitutes approximately 5% of AML cases. [0316] M1 subtype is also called acute myeloblastic leukemia without maturation. AML-M1 is defined and characterized by a high percentage of blasts in the bone marrow without significant evidence of myeloid maturation. Blasts constitute >90% of the nonerythroid cells. The M1 blasts express at least two of the following myeloid antigens: CD13, CD33, CD117, MPO, and/or HLA-DR. CD34 is often positive. There is generally no expression of the monocytoid markers CD11b or CD14. AML-M1 constitutes 10% of AML cases. [0317] M2 subtype is also called acute myeloblastic leukemia with maturation. Multiple genomic abnormalities are associated with AML-M2, e.g., formation of a fusion protein, AML1-ETO or RUNX1-RUNX1T1 due to a translocation of chromosome 8 to chromosome 21 or t(8;21) as well as translocation between chromosome 6p23 and chromosome 9q34 causing the formation of a fusion oncogene made of DEK (6p23) and CAN/NUP214 (9q34). AML-M2 is defined and characterized by the presence of > 20% blasts in the bone marrow or blood and evidence of maturation to more mature neutrophils. (>10% neutrophils are at different stages of maturation). Monocytes comprise <20% of bone marrow cells. M2 blasts must express at least two of the following myeloid antigens: CD13, CD33, CD15, CD117, MPO and/or HLA-DR. MPO must be > 3% on blasts. These blasts are generally negative for the monocytic markers CD14 and CD11b. AML-M2 constitutes approximately 30-45% of AML cases. AML-M1 and AML-M2 are initially stratified by morphology (e.g., immature cells < 10% is indicative of AML-M1, and immature cells > 10% is indicative of AML-M2). [0318] M3 subtype is also called acute promyelocytic leukemia or acute progranulocytic leukemia (APL). APL/AML-M3 is characterized by proliferation of malignant promyelocytes with mature myeloid immunophenotype and the translocation t(15;17)(q22;q11), which causes the fusion of retinoic acid receptor-alpha (RARalpha) gene on chromosome 17 and the gene PML on chromosome 15. Four other gene rearrangements have been described in APL fusing RAR^ to promyelocytic leukemia zinc finger (PLZF), nucleophosmin (NPM), nuclear ny-2770598
Attorney Docket No.24516-20006.40 matrix associated (NUMA), or signal transducer and activator of transcription 5b (STAT5B) genes. Three M3 morphologic variants include: 1) the typical hypergranular form, 2) the microgranular variant, and 3) the basophilic variant. See, e.g., Suci^ et al., J Hematother Stem Cell Res.2002, 11(6):941-950. The blasts are positive for the myeloid markers CD33 and CD117. The blasts are generally positive for the markers CD13 and CD34 in the hypogranular variant. The blasts are characteristically negative HLA Dr, unlike in other AML subtypes. AML-M3 constitutes approximately 5-8% of AML cases in adults. [0319] M4 subtype is also called acute myelomonocytic leukemia (AMML). AML-M4 is defined as an acute leukemia with differentiation along both myeloid and monocytic lines. Myeloblasts and monocytes and promonocytes represent > 20%, but monocytes and promonocytes represent < 80% of the total nucleated cells in the marrow differential. Both myeloblasts and monoblasts are present. There are two types of AML-M4: AML-M4 and AML-M4e variant, which has an increased eosinophil count. Translocation t(8:16)(p11;p13) may be associated with AMML. The myeloblasts generally express the markers CD13, CD33, CD34, CD117, HLA Dr. The monocytoid cells (monoblasts and promonocytes) generally express the markers CD11b, CD11c, CD13, CD14, CD33, CD64, and HLA Dr. AML-M4 constitutes approximately 15-25% of AML cases. [0320] M5 subtype is also called acute monocytic leukemia (AMoL), which is defined as greater than 20% blasts in the bone marrow, of which greater than 80% are of the monocytic lineage. A further subclassification (M5a versus M5b) is made depending on whether the monocytic cells are predominantly monoblasts (>80%) (acute monoblastic leukemia) or a mixture of monoblasts and promonocytes (<80% blasts). There is a strong association with AMoL and deletions and translocations involving chromosome 11 band 23. Translocation t(8:16)(p11;p13) may be associated with AMoL. Blasts characteristically express the markers CD4, CD11b, CD11c, CD13, CD33, CD45, CD56, CD64, and HLA Dr. A subset of these cells may also express CD2, CD7, CD10, CD16, CD23, lysosome, and CD117. CD34 is predominantly negative. AML-M5 constitutes approximately 5-8% of AML cases. [0321] M6 subtype is also called acute erythroblastic leukemia or erythroleukemia, wherein the cancer cells are either an erythroid/myeloid leukemic cell mix or pure erythroid leukemic cells and is a rare manifestation of AML. More than 30-50% of the nucleated marrow cells are abnormal nucleated red blood cells. Erythroleukemia is defined by > 50% erythroid precursors and > 20% myeloblasts. Pure erythroid leukemia is defined by >80% erythroid precursors. Erythroblasts are positive for Glycophorin A (GPHA) and CD71. The erythroid ny-2770598
Attorney Docket No.24516-20006.40 component lacks MPO, CD34, CD45, and pan myeloid markers. The myeloblasts often express CD117 and CD43. AML-M6 constitutes approximately 5-6% of AML cases. [0322] M7 subtype is also called acute megakaryocytic leukemia (AMKL). AMKL is defined as an AML with >20% blasts, of which 50% or more are of the megakaryocyte lineage. AML-M7 blasts often resemble lymphoblasts, although AML-M7 leukemia may be accompanied by atypical megakaryocytes. Megakaryoblasts expresses of one or more platelet glycoprotein CD41 (glycoprotein IIb/IIIa) and/or CD61 (glycoprotein IIIa). Myeloid markers CD13 and CD33 may be positive; CD36 is typically positive. AML-M7 constitutes approximately 1.2% of adult AML cases and approximately 3-10% of pediatric AML cases. [0323] In some embodiments, the AML cells express CD13, CD14, and/or CD33 at the time when the AML cells are obtained from the peripheral blood. Methods of obtaining AML cells from peripheral blood is well known in the art. For example, AML leukocytes (e.g., human AML leukocytes) can be seeded onto a confluent layer of bone-marrow mesenchymal stromal cells (MSCs, such as human MSCs), to which the AML cells adhere, whereas lymphocytes are non-adherent cells and can therefore be separated from the AML cells. For example, AML leukocytes (e.g., human AML leukocytes) can be seeded on anti-CD11b antibody- coated plates. AML cells can also be separated by positive selection with (e.g.) anti-CD11b antibody, anti-CD13 antibody, anti-CD14 antibody, and/or anti-CD33 antibody or negative selection using all antibodies against other cells using magnetic-activated cell sorting (MACS). Yet further methods include surface marker staining and cell sorting by fluorescence activated cell sorting (FACS). [0324] The AML cells obtained from the individual (e.g., human cancer patient) in some embodiments express a lower level of IL-10 receptor (“IL-10R”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a reference individual (e.g., a healthy individual). In some embodiments, the level of IL-10R on the AML cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-10R on corresponding cells from the reference individual (e.g., a healthy individual). [0325] In some embodiments, the AML cells obtained from the individual (e.g., human cancer patient) express a lower level of IL-4 receptor (“IL-4R”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual. In some embodiments, the level of IL-4R on the AML cells from the ny-2770598
Attorney Docket No.24516-20006.40 individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-4R on corresponding cells from the reference individual (e.g., a healthy individual). [0326] In some embodiments, the AML cells obtained from the individual express a lower level of IL-6 receptor (“IL-6R”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual. In some embodiments, the level of IL-6R on the AML cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% lower than the level of IL-6R on corresponding cells from the reference individual (e.g., a healthy individual). [0327] In some embodiments, the AML cells obtained from the individual express a lower level of M-CSF receptor (“M-CSFR”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual. In some embodiments, the level of M-CSFR on the AML cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, or 65% lower than the level of M-CSFR on corresponding cells from the reference individual (e.g., a healthy individual). [0328] In some embodiments, the AML cells obtained from the individual express a lower level of GM-CSF receptor (“GM-CSFR”) prior to contacting with one or more cytokines described above as compared to corresponding cells obtained from a healthy individual. In some embodiments, the level of GM-CSFR on the AML cells from the individual is at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, or 65% lower than the level of GM-CSFR on corresponding cells from the reference individual (e.g., a healthy individual). Chronic lymphocytic leukemia (CLL) [0329] Chronic lymphocytic leukemia (CLL) is one of the most frequent types of leukemia. It typically occurs in elderly patients and has a highly variable clinical course. Leukemic transformation is initiated by specific genomic alterations that interfere with the regulation of proliferation and of apoptosis in clonal B-cells. The diagnosis is established by blood counts, blood smears, and immunophenotyping of circulating B-lymphocytes, which identify a clonal B-cell population carrying the CD5 antigen as well as typical B-cell markers. [0330] The clinical staging systems provide prognostic information by using the results of physical examination and blood counts. Various biological and genetic markers provide ny-2770598
Attorney Docket No.24516-20006.40 additional prognostic information. Deletions of the short arm of chromosome 17 (del[17p]) and/or mutations of the TP53 gene predict resistance to chemoimmunotherapy and a shorter time to progression with most targeted therapies. The CLL international prognostic index integrates genetic, biological, and clinical variables to identify distinct risk groups of patients with CLL. [0331] Only patients with active or symptomatic disease or with advanced Binet or Rai stages require therapy. When treatment is indicated, several therapeutic options exist: a combination of the B-cell lymphoma 2 (BCL2) inhibitor venetoclax with obinutuzumab, monotherapy with inhibitors of Bruton tyrosine kinase (BTK) such as ibrutinib and acalabrutinib, or chemoimmunotherapy. At relapse, the initial treatment may be repeated, if the treatment-free interval exceeds 3^years. If the disease relapses earlier, therapy should be changed using an alternative regimen. Patients with a del(17p) or TP53 mutation are usually resistant to chemotherapy and should, therefore, be treated with targeted agents. See e.g., Am J Hematol.2021 Dec 1;96(12):1679-1705. Non-Hodgkin's lymphoma (NHL) [0332] NHL (Non-Hodgkin’s Lymphomas) are a heterogenous group of lymphoproliferative malignancies that are much less predictable than Hodgkin's lymphomas and have a far greater predilection to disseminate to extranodal locations. Nearly 25% of NHL cases arise in extranodal locations and most of them are seen involving both nodal and extranodal sites. [0333] The most common NHL subtypes by far in developed countries are diffuse large B- cell lymphoma (about 30%) and follicular lymphoma (about 20%). All other NHL subtypes have a frequency of less than 10%. NHL is the sixth most common cause of cancer-related death in the USA after prostate cancer, breast cancer, lung cancer, colorectal cancer, and bladder cancer. Oropharyngeal lymphomas are the second most common malignant disease in the oral region after squamous cell carcinoma. See e.g., J Family Med Prim Care.2020 Apr; 9(4): 1834–1840. [0334] NHL can be divided into two prognostic groups: the indolent lymphomas and the aggressive lymphomas. Indolent NHL types have a relatively good prognosis, with a median survival as long as 20 years, but they usually are not curable in advanced clinical stages. Early-stage (stage I and stage II) indolent NHL can be effectively treated with radiation therapy alone. Most of the indolent types are nodular (or follicular) in morphology. The ny-2770598
Attorney Docket No.24516-20006.40 aggressive type of NHL has a shorter natural history, but a significant number of these patients can be cured with intensive combination chemotherapy regimens. [0335] In general, with modern treatment of patients with NHL, the overall survival rate at 5 years is over 60%. More than 50% of patients with aggressive NHL can be cured. Most relapses occur in the first 2 years after therapy. The risk of late relapse is higher in patients who manifest both indolent and aggressive histologies. [0336] While indolent NHL is responsive to immunotherapy, radiation therapy, and chemotherapy, a continuous rate of relapse is usually seen in advanced stages. However, patients can often be re-treated with considerable success if the disease histology remains low grade. Patients who present with or convert to aggressive forms of NHL may have sustained complete remissions with combination chemotherapy regimens or aggressive consolidation with marrow or stem cell support. [0337] In some embodiments, the methods described herein further comprise assessing IL- 10R expression level of the cells, such as cancer-specific monocytes, hematological cancer cells such as B-ALL cells, CLL cells, NHL cells, or AML cells (e.g., prior to contacting the cancer-specific monocytes, B-ALL cells, or AML cells with one or more cytokines, such as IL-10, IFN^, and/or TNF^). Chronic Myeloid Leukemia (CML) [0338] Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm with an incidence of 1-2 cases per 100,000 adults. It accounts for approximately 15% of newly diagnosed cases of leukemia in adults. [0339] Central to the pathogenesis of CML is the fusion of the Abelson murine leukemia (ABL1) gene on chromosome 9 with the breakpoint cluster region (BCR) gene on chromosome 22. This results in expression of an oncoprotein termed BCR-ABL1. Thus, BCR-ABL1 is a constitutively active tyrosine kinase that promotes growth and replication through downstream signaling pathways such as RAS, RAF, JUN kinase, MYC and STAT. This influences leukemogenesis by creating a cytokine-independent cell cycle with aberrant apoptotic signals in response to cytokine withdrawal. [0340] About 50% of patients diagnosed with CML in the United States are asymptomatic. The diagnosis of CML often occurs during a routine physical examination or blood tests. CML can be classified into three phases: CP, accelerated phase (AP), and blast phase (BP). Most (90%-95%) patients present in CML-CP. Common signs and symptoms of CML-CP, ny-2770598
Attorney Docket No.24516-20006.40 when present, result from anemia and splenomegaly. These include fatigue, weight loss, malaise, easy satiety, and left upper quadrant fullness or pain. Rare manifestations include bleeding (associated with a low platelet count and/or platelet dysfunction), thrombosis (associated with thrombocytosis and/or marked leukocytosis), gouty arthritis (from elevated uric acid levels), priapism (usually with marked leukocytosis or thrombocytosis), retinal hemorrhages, and upper gastrointestinal ulceration and bleeding (from elevated histamine levels due to basophilia). Leukostatic symptoms (dyspnea, drowsiness, loss of coordination, confusion) due to leukemic cells sludging in the pulmonary or cerebral vessels, are uncommon in CP despite white blood cell (WBC) counts exceeding 100^×^109/L. Splenomegaly is the most consistent physical sign detected in 20-40% of cases. Hepatomegaly is less common (less than 10%). Lymphadenopathy and infiltration of skin or other tissues are rare. When present, they favor Ph-negative CML or AP or BP of CML. Headaches, bone pain, arthralgias, pain from splenic infarction, and fever are more frequent with CML transformation. Most patients evolve into AP prior to BP, but 20% transition into BP without AP warning signals. Therefore, CML-AP might be insidious or present with worsening anemia, splenomegaly, and organ infiltration; CML-BP presents as an acute leukemia (myeloid in 60%, lymphoid in 30%, megakaryocytic or undifferentiated in 10%) with worsening constitutional symptoms, bleeding, fever, and infections. See e.g., Am J Hematol.2020; 95: 691–709. [0341] Because CML mostly involves similar types of cancer cells (mainly myeloid or lymphoid), it is expected that the methods described herein are also effective for CML. Individuals [0342] In some embodiments, the individual has a hematological cancer. [0343] In some embodiments, the individual has an advanced hematological cancer. In some embodiments, the individual has a late-stage cancer. In some embodiments, the individual has a cancer that is stage II, III, or IV. In some embodiments, the individual has metastases. In some embodiments, the cancer is malignant. [0344] In some embodiments, the individual has a myeloid leukemia, a B-cell lymphoma, or a B cell leukemia. [0345] In some embodiments, the individual has a B-ALL, CLL, AML, CML, or NHL. [0346] In some embodiments, the individual has a B-ALL, CLL, AML, or NHL. ny-2770598
Attorney Docket No.24516-20006.40 [0347] In some embodiments, the individual is a female. In some embodiments, the individual is a male. [0348] In some embodiments, the individual is a human. In some embodiments, the individual is a human who is at least about any of 50, 55, 60, 65, 70, or 75 years old. In some embodiments, the individual is less than about 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 years old. [0349] In some embodiments, the individual (e.g., an individual with a hematological cancer) has a PBMC or bone marrow (BM) sample that comprises no more than about 5%, 4%, 3%, 2%, or 1% of the cells being monocytes (e.g., cancer monocytes). [0350] In some embodiments, the individual has a PBMC or BM sample that comprises at least about 20%, 30%, 40%, 50%, 60%, 70%, or 80% of the cells being the hematological cancer cells (e.g., B-ALL cells, CLL cells, NHL cells, CML cells, AML cells). [0351] In some embodiments, the individual has a PBMC or bone marrow (BM) sample that a) comprises no more than about 5%, 4%, 3%, 2% or 1% of the cells being monocytes (e.g., cancer monocytes), and b) comprises at least about 20%, 30%, 40%, 50%, 60%, 70%, or 80% of the cells being hematological cancer cells (e.g., B-ALL cells, CLL cells, NHL cells, CML cells, AML cells). III. Antigen presenting cells (APCs), compositions, and cultures. [0352] The present application provides HC-APCs such as those prepared according to any of the methods described above with unique properties that distinguish them from naturally occurring APCs or APCs generated in vitro by currently known methods. [0353] The exemplified HC-APCs described herein have distinct surface markers from the hematological cancer cells from which they are derived. For example, the HC-APCs described herein express a high level of MHC-I, MHC-II, CD80, CD86, and CD40. In some embodiments, the HC-APCs also display a clear shift in morphology to a larger cell diameter with increased cell spreading, which is indicative of the ^APC phenotype. [0354] In some embodiments, the HC-APCs described herein comprise one or more exogenous antigen (e.g., an antigenic peptide, a tumor-associated peptide, or a neoantigenic peptide). [0355] “+/high” described herein refers to a positive expression of a certain surface molecule, or a high expression of a certain surface molecule. In some embodiments, a high expression refers to the scenario that the cell (e.g., HC-APCs) expresses a higher level of the surface ny-2770598
Attorney Docket No.24516-20006.40 molecule than a reference cell population (e.g., corresponding cells such as myeloid cells or B cells in a healthy individual). In some embodiments, a higher level refers to an expression level that is at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 2-fold, 5-fold, 10-fold, or 100-fold higher level of expression. In some embodiments, an increased expression of a certain molecule refers to an expression level that is at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 2-fold, 5-fold, 10-fold, or 100-fold higher level of expression. In some embodiments, a decreased expression of a certain molecule refers to an expression level that is at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, or 80% less. [0356] In some embodiments, the HC-APCs express a high level of one or more (e.g., two, three, four, five, six, seven, or eight) antigen presentation molecule, wherein the antigen presentation molecule is selected from the group consisting of: MHC-I, MHC-II, CD86, CD80, OX40L, ICAML, ICOSL, and CD40, optionally wherein the HC-APCs are produced from hematological cancer cells (e.g., AML cells, CLL cells, NHL cells, CML cells, or B- ALL cells isolated from an animal, such as a human cancer patient) in a cell culture. In some embodiments, the HC-APCs express a high level of MHC-I, MHC-II, CD86, CD80, CD40, and/or OX40L. [0357] In some embodiments, the HC-APCs express a high level of MHC-I, MHC-II, CD86, CD80, CD40, and/or OX40L. [0358] In some embodiments, at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the HC-APCs have a dendritic cell morphology. See e.g., FIG.5C. [0359] In some embodiments, the HC-APCs comprise one or more tumor-associated antigen peptides or tumor-specific antigen peptides, e.g., neoantigen peptides, such as any of those present on the hematological cancer cell (e.g., AML cells, CLL cells, NHL cells, CML cells, or B-ALL cells) at time of collection from a cancer patient. [0360] In some embodiments, the HC-APCs comprise one or more virus-associated antigen peptide. [0361] In some embodiments, the HC-APCs are capable of promoting proliferation of immune cells (e.g., T cells, e.g., CD4 T cells and/or CD8 T cells) upon incubation with the immune cells. In some embodiments, the HC-APCs promote proliferation of T cells for at least about any of 5-fold, 10-fold, 15-fold, or 20-fold in a cell culture comprising IL-2, IL-7, ny-2770598
Attorney Docket No.24516-20006.40 and IL-15. In some embodiments, the incubation is no longer than about 24 hours, 22 hours, 20 hours, or 18 hours. [0362] In some embodiments, there is provided a composition (e.g., a culture) comprising the HC-APCs described herein. In some embodiments, the HC-APCs present one or more disease-associated peptides (e.g., tumor-associated antigen peptides and/or tumor-specific antigen peptides) to immune cells. IV. Methods of activating immune cells and activated immune cell compositions [0363] The present application also provides methods of activating a population of immune cells. In some embodiments, the methods comprise co-culturing the population of immune cells with the population of the HC-APCs described herein, wherein the HC-APCs present endogenous cancer specific antigens, and optionally one or more antigen peptides, (e.g., tumor peptides, e.g., tumor-associated peptides and/or tumor-specific peptides, e.g., neoantigen peptides). [0364] The HC-APCs produced according to the methods describe herein possess high potency and can effectively engage antigen-specific immune cells (e.g., T cells), activate and promote the expansion of these immune cells. These activated immune cells (e.g., T cells) can effectively kill the cancer cells both in in vitro setting (e.g., at a ratio of T cells vs. cancer cell 1:1 or 3:1) and in vivo. See e.g., FIG.17B. In some embodiments, these activated immune cells are activated T cells. In some embodiments, the activated T cells comprise a higher percentage of CD8+ T cells than the percentage of CD8+ T cells prior to the incubation with HC-APCs or a new round of expansion. In some embodiments, the percentage of the CD8+ T cells in the activated T cells is at least about 20%, 40%, 60%, or 80% higher than the percentage of the CD8+ T cells in the T cells prior to the incubation with HC-APCs or a new round of expansion. In some embodiments, the percentage of the effector memory T cells (i.e., CD45RA-CCR7- CD8 T cells) in the activated T cells is at least about 10%, 15%, 20%, or 25% higher than the percentage of the effector memory T cells in the T cells prior to the incubation with HC-APCs or a new round of expansion. See e.g., FIG.11E. In some embodiments, the activated T cells express a higher level of CD107a, 4-1BB and/or CD25. See e.g., FIG.17C. [0365] In some embodiments, there is provided a method of activating a population of immune cells (e.g., T cells, e.g., TIL cells) obtained from an individual (e.g., a cancer patient), comprising co-culturing the population of immune cells with the population of the ny-2770598
Attorney Docket No.24516-20006.40 HC-APCs (e.g., the HC-APCs described herein), thereby producing a population of activated immune cells, wherein the APCs present one or more tumor peptides and are thereby able to prime and activate the population of immune cells. In some embodiments, the HC-APCs are derived from B-ALL, CLL, NHL, CML cells, or AML cells obtained from the same individual. In some embodiments, the ratio of HC-APCs and the immune cells (e.g., T cells, e.g., TIL cells) during co-culturing is about 10:1 to about 1:10 (e.g., about 5:1 to about 1:5, about 2:1 to about 1:2, about 1:1). In some embodiments, the HC-APCs and the immune cells are co-cultured for at least about 4-8 hours. In some embodiments, IL-2, IL-7, and/or IL-15 are supplemented to the co-culture (e.g., are supplemented about at least about 4-8 hours after the HC-APCs and the immune cells are first put into co-culture), e.g., for at least about 6 days, e.g., about 6-14 days for each round of expansion. In some embodiments, the activated immune cells comprise at least 5-, 10-, or 20-fold (e.g., 50-100-fold) more cells than the immune cells prior to the co-culture. In some embodiments, the activated immune cells are subject to the activation via co-culture with the HC-APCs described herein for at least two, three, four, or five rounds. In some embodiments, the activated immune cells do not exhibit an exhausted phenotype (e.g., senescence) after two, three, or four consecutive rounds (e.g., about 6-14 days each round, e.g., about 6-10 days each round) of activation that involves the co-culture described herein. In some embodiments, the co-culture does not involve use of an anti-CD3 antibody and/or an anti-CD28 antibody at least some of the rounds (e.g., anti-CD3 and anti-CD28 antibodies are only used in the first round but not in one or more later rounds). See e.g., FIG.16A. [0366] In some embodiments, the HC-APCs present with one or more endogenous tumor- associated peptides (i.e., one or more tumor-associated or tumor-specific peptides of the isolated AML or B-ALL cells that are differentiated into the HC-APC as described herein). [0367] In some embodiments, the method further comprises contacting the HC-APCs with a composition comprising one or more exogenous antigen such as a plurality of tumor- associated peptides (e.g., neoantigen peptides) that are unique from the endogenous neoantigen peptide(s) presented by the cancer cell (e.g., AML cell, NHL cell, CML cells, CLL cell, or B-ALL cell) that is differentiated into an HC-APC. Exemplary exogenous antigens or methods of identifying of selecting these antigens are discussed in Section II under “loading HC-APCs with additional antigens.” [0368] In some embodiments, the HC-APCs are allowed to be in contact with the composition comprising a plurality of tumor-associated peptides (e.g., neoantigen peptides) ny-2770598
Attorney Docket No.24516-20006.40 for about 4 to about 24 hours. In some embodiments, the HC-APCs have been pre-incubated with the composition comprising a plurality of tumor-associated peptides (e.g., neoantigen peptides). [0369] In some embodiments, the composition comprising a plurality of tumor-associated peptides (e.g., neoantigen peptides) is a surgical resection of tumor tissue or a biopsy extract thereof. In some embodiments, the composition comprising a plurality of tumor-associated peptides (e.g., neoantigen peptides) is a mixture of tumor cells or extract thereof isolated from tumor tissue or biopsy. In some embodiments, the composition comprising a plurality of tumor-associated peptides (e.g., neoantigen peptides) is a mixture of isolated tumor- associated peptides (e.g., neoantigen peptides). In some embodiments, the tumor tissue or cell is a fresh tumor tissue or cell. In some embodiments, the tumor tissue or cell is obtained from a frozen sample. In some embodiments, the tumor tissue or cells have been subjected to an apoptosis induction. In some embodiments, the tumor tissue or cells have been subjected to a radiation treatment. [0370] In some embodiment, the population of immune cells and the HC-APCs are derived from the same individual. [0371] In some embodiments, the population of immune cells and the HC-APCs are not derived from the same individual. [0372] In some embodiments, the HC-APCs are further loaded with one or more exogenous antigens (see e.g., descriptions below). [0373] In some embodiments, the HC-APCs are not loaded with one or more exogenous antigens. [0374] The present application also provides activated immune cells (e.g., T cells) produced by any of the methods described here. Tumor-associated Peptides Loading [0375] In some embodiments, the methods described herein further comprise contacting HC- APCs with a plurality of tumor-associated peptides (e.g., neoantigen peptides). In some embodiments, the plurality of tumor-associated peptides (e.g., neoantigen peptides) have more than about any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 22, 25, 30, 40, or 50 tumor-associated peptides (e.g., neoantigen peptides). In some embodiments, the APCs are allowed to be in contact with the composition comprising a ny-2770598
Attorney Docket No.24516-20006.40 plurality of tumor-associated peptides (e.g., neoantigen peptides) for about 4 to about 24 hours. [0376] In some embodiments, the HC-APCs have been pre-incubated with the composition comprising a plurality of tumor-associated peptides (e.g., neoantigen peptides) prior to be used in the methods of activating immune cells described herein. [0377] An exemplary embodiment of the contacting of a population of HC-APCs with a plurality of tumor-associated peptides (e.g., neoantigen peptides) comprises pulsing the plurality of tumor-associated peptides (e.g., neoantigen peptides) into the population of HC- APCs. As known in the art, pulsing refers to a process of mixing cells, such as HC-APCs, with a solution containing tumor-associated peptides (e.g., neoantigen peptides), and optionally subsequently removing the tumor-associated peptides (e.g., neoantigen peptides) from the mixture. The population of HC-APCs may be contacted with a plurality of tumor- associated peptides (e.g., neoantigen peptides) for seconds, minutes, or hours, such as about any of 30 seconds, 1 minute, 5 minutes, 10 minutes, 15 minutes, 20 minutes, 30 minutes, 1 hour, 5 hours, 10 hours, 12 hours, 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, one week, 10 days, or more. The concentration of each neoantigen peptide used in the contacting step may be about any of 0.1, 0.5, 1, 2, 3, 5, or 10 ^g/mL. In some embodiments, the concentration of the tumor-associated peptides (e.g., neoantigen peptides) is about 0.1-200 ^g/mL, including for example about any of 0.1-0.5, 0.5-1, 1-10, 10-50, 50-100, 100-150, or 150-200 ^g/mL. [0378] In some embodiments, the population of HC-APCs is contacted with the plurality of tumor-associated peptides (e.g., neoantigen peptides) in the presence of a composition that facilitates the uptake of the plurality of tumor-associated peptides (e.g., neoantigen peptides) by the HC-APCs. In some embodiments, compounds, materials, or compositions may be included in a solution of the plurality of tumor-associated peptides (e.g., neoantigen peptides) to facilitate peptide uptake by the HC-APCs. Compounds, materials, or compositions that facilitate the uptake of the plurality of tumor-associated peptides (e.g., neoantigen peptides) by the HC-APCs include, but are not limited to, lipid molecules and peptides with multiple positively charged amino acids. In some embodiments, more than about any of 50%, 60%, 70%, 80%, 90%, or 95% of the tumor-associated peptides (e.g., neoantigen peptides) are uptaken by the population of HC-APCs. In some embodiments, more than about any of 50%, 60%, 70%, 80%, 90%, or 95% of the HCAPCs in the population uptake at least one exogenous tumor antigen peptide. ny-2770598
Attorney Docket No.24516-20006.40 Immune cells [0379] The immune cells described herein can be any type of immune cells that interact with HC-APCs and can be activated by HC-APCs, and then exert their desired functions. Exemplary immune cells include T cells. [0380] T cells, or T lymphocytes, play a central role in cell-mediated immunity. Each clone of activated T cells express a distinct T-cell receptor (TCR) on the surface, which is responsible for recognizing antigens bound to MHC molecules on APCs (such as HC-APCs) and on target cells (such as cancer cells). T cells are subdivided into several types, each expressing a unique combination of surface proteins and each having a distinct function. [0381] Cytotoxic T cells (TC) participate in the immune response to and destruction of tumor cells and other infected cells, such as virus-infected cells. Generally, TC cells function by recognizing a class I MHC presented antigen on an APC (such as a HC-APC) or any target cell. Stimulation of the TCR, along with a co-stimulator (for example CD28 on the T cell binding to B7 on the APC, or stimulation by a helper T cell), results in activation of the TC cell. The activated TC cell can then proliferate and release cytotoxins, thereby destroying the APC, or a target cell (such as a cancer cell). Mature TC cells generally express surface proteins CD3 and CD8. Cytotoxic T cells belong to CD3+CD8+ T cells. [0382] Helper T cells (TH) are T cells that help the activity of other immune cells by releasing T cell cytokines, which can regulate or suppress immune responses, induce cytotoxic T cells, and maximize cell killing activities of macrophages. Generally, TH cells function by recognizing a class II MHC presented antigen on an APC (such as a HC-APC). Mature TH cells express the surface proteins CD3 and CD4. Helper T cells belong to CD3+CD4+ T cells. [0383] Regulatory T cells (TREG cells) generally modulate the immune system by promoting tolerance for self-antigens, thereby limiting autoimmune activity. In cancer immunotherapy, TREG contributes to escape of the cancer cells from the immune response. TREG cells generally express CD3, CD4, CD7, CD25, CTLA4, GITR, GARP, FOXP3, and/or LAP. CD4+CD25+Foxp3+ T cells are one class of TREG cells. [0384] Memory T cells (Tm) are T cells that have previously encountered and responded to their specific antigens, or T cells that differentiated from activated T cells. Although tumor specific Tms constitutes a small proportion of the total T cell amount, they serve critical functions in surveillance of tumor cells during a person’s entire lifespan. If tumor specific ny-2770598
Attorney Docket No.24516-20006.40 Tms encounter tumor cells expressing their specific tumor antigens, the Tms are immediately activated and clonally expanded. The activated and expanded T cells differentiate into effector T cells to kill tumor cells with high efficiency. Memory T cells are important for establishing and maintaining long-term tumor antigen specific responses of T cells. [0385] Typically, an antigen for T cells is a protein molecule or a linear fragment of a protein molecule that can be recognized by a T-cell receptor (TCR) to elicit specific T cell response. The antigen can be derived from a foreign source such as a virally encoded protein, or an endogenous source such as a protein expressed on the cell surface. The minimal fragment of an antigen that is directly involved in interaction with a particular TCR is known as an epitope. Multiple epitopes can exist in a single antigen, wherein each epitope is recognized by a distinct TCR encoded by a particular clone of T cells. [0386] In order to be recognized by a TCR, an antigen peptide or antigen fragment is processed into an epitope by an APC (such as a dendritic cell or HC-APC), and then bound in an extended conformation inside a Major Histocompatibility (MHC) molecule to form an MHC-peptide complex on the surface of an APC (such as a dendritic cell or HC-APC). MHC molecules are also known as human leukocyte antigens (HLA). The MHC provides an enlarged binding surface for strong association between TCR and epitope, while a combination of unique amino acid residues within the epitope ensures specificity of interaction between TCR and the epitope. The human MHC molecules are classified into two types—MHC class I and MHC class II—based on their structural features, especially the length of epitopes bound inside the corresponding MHC complexes. MHC-I epitopes are epitopes bound to and represented by an MHC class I molecule. MHC-II epitopes are epitopes bound to and represented by an MHC class II molecule. MHC-I epitopes are typically about 8 to about 11 amino acids long, whereas MHC-II epitopes are about 13 to about 17 amino acids long. Due to genetic polymorphism, various subtypes exist for both MHC class I and MHC class II molecules among the human population. T cell response to a specific antigen peptide presented by an MHC class I or MHC class II molecule on an APC is known as MHC-restricted T cell response. [0387] In some embodiments, the immune cells are selected from the group consisting of PBMC, tumor infiltrating T cells (TIL), and T cells (e.g., CD4 T cells and/or CD8 T cells). [0388] In some embodiments, the immune cells are PBMC. [0389] In some embodiments, the immune cells are tumor infiltrating T cells (TIL). ny-2770598
Attorney Docket No.24516-20006.40 [0390] In some embodiments, the immune cells are CD4 T cells and/or CD8 T cells. [0391] In some embodiments, the immune cells and the HC-APCs are derived from the same individual. In some embodiments, the immune cells and the HC-APCs are derived from different individuals. Preparation of activated immune cells (e.g., activated T cells) [0392] Methods described herein comprise co-culturing a population of immune cells (e.g., T cells) with a population of HC-APCs described herein that present with a plurality of one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides). [0393] In some embodiments, the co-culturing was carried out for at least 24 hours. In some embodiments, the co-culturing was carried out for at least about 1-5 days (e.g., about 1-3 days). In some embodiments, the population of immune cells (e.g., T cells) and the population of HC-APCs that present with the plurality of one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) are co-cultured for about any of 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, or 30 days. In some embodiments, the population of immune cells (e.g., T cells) is co-cultured with the population of HC-APCs that present with the plurality of one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) for about 14 to about 21 days. In some embodiments, the population of immune cells (e.g., T cells) is co-cultured with the population of HC-APCs that present with the plurality of one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) for about 14 days. [0394] The population of immune cells (e.g., T cells) used in any embodiment of the methods described herein may be derived from a variety of sources. A convenient source of the immune cells is from the PBMCs of the human peripheral blood. For example, the population of T cells may be isolated from the PBMCs, or alternatively, a population of PBMCs enriched with T cells (such as by addition of T cell specific antibodies and cytokines) can be used in the co-culture. In some embodiments, the population of T cells used in the co- culture is obtained from the non-adherent fraction of peripheral blood mononuclear cells (PBMCs). In some embodiments, the PBMCs are obtained by density gradient centrifugation of a sample of peripheral blood. In some embodiments, the population of activated T cells is prepared by obtaining a population of non-adherent PBMCs, and co-culturing the population of non-adherent PBMCs with a population of HC-APCs that present with a plurality of one or ny-2770598
Attorney Docket No.24516-20006.40 more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) (such as in the presence of at least one cytokine (such as IL-2) and an anti-CD3 antibody). [0395] The co-culture may further include cytokines and other compounds to facilitate activation, maturation, and/or proliferation of the T cells, as well as to prime T cells for later differentiation into e.g., memory T cells. Exemplary cytokines that may be used in this step include, but are not limited to, IL-7, IL-15, IL-21, and the like. Certain cytokines may help suppress the percentage of TREG in the population of activated T cells in the co-culture. For example, in some embodiments, a high dose (such as about any of 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1200, or 1500 U/ml) of a cytokine (such as IL-2) is used to co-culture the population of T cells and the population of dendritic cells loaded with the plurality of tumor antigen peptides to obtain a population of activated T cells with a low percentage of TREG cells. [0396] In some embodiments, the methods of activating immune cells comprise co-culture of the immune cells (e.g., the T cells) and HC-APC populations for more than one round (e.g., two, three, or four rounds). In some embodiments, each round takes about 6-8 days. In some embodiments, the first, second, third, and/or fourth round do not involve the addition of an anti-CD3 antibody and/or an anti-CD28 antibody. In some embodiment, the immune cells (e.g., the T cells) show non-exhaustive feature after two, three, or four rounds of co-culture. In some embodiments, the immune cells (e.g., T cells) are capable of expanding about 10- 200-fold (e.g., at least about 10-fold, such as about 50-fold) after each round of culture. In some embodiments, each round takes about 5-10 days or 6-8 days. In some embodiments, the number of the immune cells (e.g., T cells) after three or four rounds of co-culture reaches about 1010. [0397] In some embodiments, the methods of activating immune cells described herein further comprise expanding the population of immune cells following the co-culturing step. In some embodiments, expanding the population of immune cells comprises contacting the immune cells with a cytokine selected from the group consisting of IL-2, IL-7, and IL-15, optionally for about 2 to about 10 days. In some embodiments, the co-culture is in the presence of an anti-CD3 antibody and a plurality of cytokines, such as IL-2, IL-7, IL-15, IL- 21, or any combination thereof. [0398] The present application also provides populations of activated immune cells obtained by the methods described in this section. ny-2770598
Attorney Docket No.24516-20006.40 V. Methods of treatment [0399] The present application in another aspect provides methods of treating a disease or condition (e.g., a cancer) in an individual (e.g., a patient), comprising contacting hematological cancer cells and/or monocytes with one or more of survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) an IL-10 receptor (IL-10R) activator.2) a TNF^ receptor (TNFR) activator, 3) a GM-CSF receptor (GM-CSFR) activator, and 4) an interferon ^ (IFN^) receptor (IFNGR) activator (e.g., any of the one or more S/D/M factors described in the Section named “Survival, differentiation, and/or maturation factors (‘S/D/M factors’)”), thereby producing a population of antigen presenting cells (“APCs”), optionally wherein the APCs are further loaded with one or more exogenous antigen, b) contacting the APCs with a population of immune cells (e.g., immune cells from the same individual), thereby producing activated immune cells, and c) administering activated immune cells into the individual. In some embodiments, the hematological cancer cells and/or monocytes are obtained from the individual, optionally wherein the hematological cancer cells and monocytes are comprised in a mixture when contacting with the one or more S/D/M factors. See e.g., FIG.2. In some embodiments, the method further comprises administering an effective amount of a TNF^ inhibitor (e.g., an anti-TNF^ antibody) into the individual. In some embodiments, the TNF^ inhibitor is administered to the individual prior to (e.g., at least 1, 2 or 3 days prior to) the administration of the activated immune cells. [0400] In some embodiments, the method comprises: a) obtaining monocytes and/or hematological cancer cells from the individual (e.g., via obtaining a PBMC or BM sample from the individual), b) contacting monocytes and/or hematological cancer cells with one or more of survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) an IL-10 receptor (IL-10R) activator, 2) a TNF^ receptor (TNFR) activator, 3) a GM-CSF receptor (GM-CSFR) activator, and 4) an interferon ^ (IFN^) receptor (IFNGR) activator, thereby producing a population of APCs, c) optionally further loading the APCs with one or more exogenous antigens, d) contacting the APCs with a population of immune cells (e.g., CD4+ and/or CD8+ T cells from the same individual), thereby producing activated immune cells, and e) administering the activated immune cells to the individual, optionally wherein a TNF^ inhibitor is administered to the individual (e.g., prior to the administration of the activated immune cells). In some embodiments, the hematological cancer cells are cancer cells of a myeloid leukemia or a B ny-2770598
Attorney Docket No.24516-20006.40 cell malignancy (e.g., a B cell lymphoma or a B cell leukemia). In some embodiments, the hematological cancer cells are cancer cells of AML cells (e.g., M0-M5 myeloblasts), CML cells, B-ALL cells, CLL cells, or NHL cells. In some embodiments, the method comprises contacting the APCs with the CD4 and CD8 T cells separately. In some embodiments, the method comprises contacting the APCs with CD4 and CD8 T cells together. In some embodiments, the method further comprises expanding the immune cells (e.g., CD4 and/or CD8 T cells) for at least one, two, three, or four rounds. In some embodiments, the method further comprises obtaining at least about 108, 5x108, 109, 5x109, or 1010 antigen-specific T cells (e.g., Neo-T cells) after several rounds of expansion. In some embodiments, the method further comprises administering at least about 108, 5x108, 109, 5x109, or 1010 antigen-specific T cells (e.g., Neo-T cells) into the individual. In some embodiments, the one or more S/DM factors comprise two, three, or more agents selected from the group consisting of: 1) an IL- 10R activator, 2) a TNFR activator, 3) a GM-CSFR activator, and 4) an IFNGR activator. In some embodiments, the one or more of S/D/M factors comprises: 1) an IL-10R activator, 2) a TNFR activator, and 3) an IFNGR activator. In some embodiments, the one or more of S/D/M factors comprises: 1) an IL-10R activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) a GM-CSFR activator. In some embodiments, the one or more of S/D/M factors comprises: 1) an IL-10R activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-6R activator. In some embodiments, the one or more of S/D/M factors comprises: 1) an IL-10R activator (e.g., IL-10 or any of the IL-10R activator listed in Table 1), 2) a TNFR activator (e.g., TNF^), 3) an IFNGR activator (e.g., IFN^), 4) an IL-6R activator (e.g., IL-6), 5) a GM-CSFR activator (e.g., GM-CSF), and 6) an IL-4R activator (e.g., IL-4). [0401] In some embodiments, the method further comprises, prior to contacting the HC- APCs with immune cells, contacting the HC-APCs with one or more of refinement factors selected from the group consisting of type-I interferon, IFN^, TNF^, a TLR ligand, CD40L or a CD40-ligating antibody, an anti-PD-L1 antibody, and TPI-1, optionally wherein the type-I interferon comprises IFN^ and/or IFN^, and optionally wherein the TLR ligand is poly IC, CpG, or LPS. [0402] In some embodiments, the immune cells and hematological cancer cells are from the same individual. In some embodiments, the immune cells and hematological cancer cells are from different individuals. In some embodiments, the immune cells are administered to the individual from whom they are obtained. In some embodiments, the immune cells are administered to a different individual from whom they are obtained. In some embodiments, ny-2770598
Attorney Docket No.24516-20006.40 the immune cells are selected from the group consisting of PBMC, cells derived from bone marrow biopsy, cells derived from lymphoma biopsy, tumor infiltrating T cells (TILs), and T cells, optionally wherein the immune cells are T cells, optionally wherein the T cells are CD8 T cells and/or CD4 T cells. In some embodiments, the hematological cancer cells are obtained from PBMC, enriched leukemia cells, a bone marrow biopsy, or a lymphoma biopsy. In some embodiments, the hematological cancer cells are cultured with the one or more of S/D/M factors for at least 2 days. In some embodiments, the HC-APCs are cultured with the refinement factors for about 1-4 days. In some embodiments, the immune cells have been enriched prior to contacting with the HC-APCs. In some embodiments, the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors. In some embodiments, the hematological cancer cells are in a mixture (e.g., PBMC sample, e.g., a mixture comprising monocytes) when contacted with the S/D/M factors. In some embodiments, the hematological cancer is a myeloid leukemia, optionally wherein the hematological cancer is acute myeloid leukemia (AML), optionally wherein the hematological cancer cells are any of M0-M5 myeloblasts. In some embodiments, the hematological cancer cells are CML cells. In some embodiments, the hematological cancer cells have been enriched prior to the contacting with the one or more of S/D/M factors in the presence of an anti-CD11b antibody. In some embodiments, the hematological cancer is a B- cell lymphoma or B-cell leukemia. In some embodiments, the B-cell lymphoma or B-cell leukemia is selected from the group consisting of B-cell acute lymphoblastic leukemia B- ALL), chronic lymphocytic leukemia (CLL), and non-Hodgkin’s lymphoma (NHL). In some embodiments, the hematological cancer cells have been enriched prior to the contacting with the one or more of S/D/M factors in the presence of an anti-CD19 or anti-CD20 antibody. [0403] The present application also provides methods of treating a disease or condition (e.g., a cancer) in a patient, comprising administering to the patient a population of activated immune cells obtained by the methods described above. [0404] In some embodiments, there is provided a method of treating a disease or condition (e.g., a cancer, e.g., a cancer associated with a hematological cancer, e.g., a hematological cancer) in a patient, comprising administering to the patient a population of activated immune cells (such as any of those described in Section IV). In some embodiments, the cancer associated with a hematological cancer is a secondary cancer that is derived from a ny-2770598
Attorney Docket No.24516-20006.40 hematological cancer. In some embodiments, the cancer associated with a hematological cancer is a primary cancer from which the hematological cancer is derived from. [0405] In some embodiments, there is provided a method of treating a cancer (e.g., a cancer associated with a hematological cancer, e.g., a hematological cancer) in a patient, comprising administering to the patient a population of activated immune cells, wherein the activated immune cells are activated by hematological cancer antigen presenting cells (“HC-APCs”), wherein the HC-APCs are derived from hematological cancer cells (e.g., B-ALL, CLL, or NHL CML, or AML cells) obtained from an individual (e.g., a cancer patient), wherein the HC-APCs express a high level of one or more antigen presentation molecules, wherein the one or more antigen presentation molecules is selected from the group consisting of: MHC-I, MHC-II, CD86, CD80, and CD40. In some embodiments, the method further comprises administering a second therapy that induces immunogenic cell death (e.g., radiotherapy). In some embodiments, the method comprises administering the activated immune cells and a radiotherapy concurrently, simultaneously, or subsequently. In some embodiments, the APCs comprise one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B- ALL, CLL, CML, NHL, or AML cells) from which the HC-APCs were derived. In some embodiments, the HC-APCs have not been preloaded with a disease- or condition-associated antigen (e.g., a tumor antigen) prior to the administration. In some embodiments, the HC- APCs have been preloaded with a disease- or condition-associated antigen (e.g., tumor antigen or a virus antigen) prior to the administration. [0406] In some embodiments, there is provided a method of treating a cancer (e.g., a cancer associated with a hematological cancer, e.g., a hematological cancer) in a patient, comprising administering to the patient a population of activated immune cells, wherein the activated immune cells are activated by HC-APCs, wherein the HC-APCs are derived from hematological cancer cells (e.g., B-ALL, CLL, NHL, CML, or AML cells) obtained from an individual (e.g., a cancer patient), wherein the HC-APCs are obtained by contacting a population of hematological cancer cells (e.g., B-ALL, CLL, NHL, CML, or AML cells) obtained from an individual with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) separately or simultaneously, wherein the one or more of S/D/M factors comprise: 1) an IL-10 receptor (IL-10R) activator, and 2) one or more agents selected from the group consisting of: an IL-4 receptor (IL-4R) activator, a TNF^ receptor (TNFR) activator, and an interferon ^ (IFN^) receptor (IFNGR) activator, thereby obtaining a ny-2770598
Attorney Docket No.24516-20006.40 population of HC-APCs. In some embodiments, the method further comprises administering a second therapy that induced immunogenic cell death (e.g., radiotherapy). In some embodiments, the method comprises administering the activated immune cells and a radiotherapy concurrently, simultaneously, or subsequently. In some embodiments, the HC- APCs comprise one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B-ALL, CLL, NHL, CML, or AML cells) from which the HC-APCs were derived. In some embodiments, the HC-APCs have not been preloaded with a disease- or condition- associated antigen (e.g., a tumor antigen) prior to the administration. In some embodiments, the HC-APCs have been preloaded with a disease- or condition-associated antigen (e.g., tumor antigen or a virus antigen) prior to the administration. [0407] In some embodiments, there is provided a method of treating a cancer (e.g., a cancer associated with a hematological cancer, e.g., a hematological cancer) in a patient, comprising administering to the patient a population of activated immune cells, wherein the activated immune cells are activated by HC-APCs, wherein the HC-APCs are derived from hematological cancer cells (e.g., B-ALL, CLL, NHL, CML, or AML cells) obtained from an individual (e.g., a cancer patient), wherein the HC-APCs: a) express a high level of one or more antigen presentation molecules, wherein the one or more antigen presentation molecules is selected from the group consisting of: MHC-I, MHC-II, CD86, CD80, OX40L, ICAML, ICOSL, and CD40, and/or b) a low level of an inhibitory signaling molecule, wherein the inhibitory signaling molecule is selected from the group consisting of: TGF^R, SIRP^, LILRB (LILRB1 and/or LILRB2), and Siglec-10, wherein the HC-APCs comprise one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B-ALL, CLL, NHL, CML, or AML cells) from which the HC-APCs were derived. In some embodiments, the hematological cancer cells (e.g., B-ALL, CLL, NHL, CML, or AML cells) exhibit a lower expression level of M-CSFR, GM-CSFR, IL-6R, IL-10R, and/or IL-4R (e.g., at least about 10%, 20%, 30%, 40%, 50%, or 60% lower) at the time when they are obtained from the individual as compared to the corresponding cells obtained from a reference individual (e.g., a healthy individual). [0408] In some embodiments, there is provided a method of treating a cancer (e.g., a cancer associated with a hematological cancer, e.g., a hematological cancer) in a patient, comprising administering to the patient a population of activated immune cells, wherein the immune cells ny-2770598
Attorney Docket No.24516-20006.40 have been subject to co-culture with a population of HC-APCs, wherein the HC-APCs are produced after being contacted with an IL-10 receptor activator (IL-10R activator) and one or more of IFN^ receptor activator (IFNR activator), TNF^ receptor activator (TNFR activator), and IL-4 receptor activator (IL-4R activator), and wherein the HC-APCs comprise one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B-ALL, CLL, NHL, CML, or AML cells) from which the HC-APCs were derived. [0409] In some embodiments, there is provided a method of treating a hematological cancer in a patient, comprising administering to the patient a population of activated immune cells (e.g., T cells), wherein the immune cells have been subject to co-culture with a population of HC-APCs, wherein the HC-APCs are produced after being contacted with IL-10 and one or more of IFN^, TNF^, and IL-4, and wherein the HC-APCs comprise one or more tumor- associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B-ALL, CLL, NHL, CML, or AML cells) from which the HC-APCs were derived. In some embodiments, the immune cells are T cells. In some embodiments, the immune cells are T cells (e.g., CD3 T cells, e.g., CD4 T cells, e.g., CD8 T cells, e.g., both CD4 and CD8 T cells, e.g., TILs) obtained from the peripheral blood of the patient. In some embodiments, the immune cells are T cells (e.g., CD3 T cells, e.g., CD4 T cells, e.g., CD8 T cells, e.g., both CD4 and CD8 T cells, e.g., TILs) obtained from the peripheral blood of an individual different from the patient (optionally with a matching HLA type). In some embodiments, the HC-APCs and the activated immune cells are derived from the same individual. In some embodiments, the HC-APCs and the activated immune cells are derived from different individuals (optionally with a matching HLA type). In some embodiments, the HC-APCs are produced after being contacted with IL-10, IFN^, TNF^, and IL-4. In some embodiments, the HC-APCs are produced after being contacted with IL-10, IFN^, TNF^, GM-CSF, IL-6, and IL-4. In some embodiments, the HC-APCs are produced after being contacted with one or more of the refinement factors described in Section II. In some embodiments, the activated immune cells are administered intratumorally, intraperitoneally, or intravenously. In some embodiments, the activated immune cells are administered at about 107 to about 109 cells per dose. In some embodiments, the methods of treatment described herein further comprise treating the patient with chemotherapy, radiation therapy, or an immune checkpoint inhibitor. In some embodiments, the method comprises ny-2770598
Attorney Docket No.24516-20006.40 treating the patient with irradiation. In some embodiments, the site of irradiation is different from the site of the cancer to be treated. [0410] In some embodiments, there is provided a method of treating a virus-related cancer in a patient, comprising administering to the patient a population of activated T cells, wherein the T cells have been subject to co-culture with a population of HC-APCs, wherein the HC- APCs are produced after being contacted with IL-10 and one or more of IFN^, TNF^, and IL- 4, and wherein the HC-APCs comprise one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B-ALL, CLL, NHL, CML, or AML cells) from which the HC-APCs were derived, wherein virus antigen-reactive T cells have been removed from the activated T cell population prior to the administration. In some embodiments, the HC- APCs are derived from the patient. In some embodiments, the activated T cells are derived from the patient. In some embodiments, the HC-APCs and the activated T cells are both derived from the patient. In some embodiments, the activated immune cells are administered intratumorally, intraperitoneally, or intravenously. In some embodiments, the activated immune cells are administered at about 107 to about 109 cells per dose. In some embodiments, the methods of treatment described herein further comprise treating the patient with chemotherapy, radiation therapy, or an immune checkpoint inhibitor. In some embodiments, the method comprises treating the patient with irradiation. In some embodiments, the site of irradiation is different from the site of the cancer to be treated. [0411] In some embodiments, there is provided a method of treating a cancer (e.g., a cancer associated with a hematological cancer, e.g., a hematological cancer) associated with a virus in a patient, comprising administering to the patient a population of activated T cells, wherein the T cells have been subject to co-culture with a population of HC-APCs, wherein the HC- APCs are produced after being contacted with IL-10 and one or more of IFN^, TNF^, and IL- 4, and wherein the HC-APCs comprise one or more tumor-associated and/or tumor-specific peptides (e.g., neoantigen peptides) that are endogenous and/or exogenous to the hematological cancer cells (e.g., the B-ALL, CLL, NHL, CML, or AML cells) from which the HC-APCs were derived, wherein virus antigen-reactive T cells have been removed from the activated T cell population prior to the administration. In some embodiments, the HC- APCs are derived from the patient. In some embodiments, the activated T cells are derived from the patient. In some embodiments, the HC-APCs and the activated T cells are both derived from the patient. In some embodiments, the activated immune cells are administered ny-2770598
Attorney Docket No.24516-20006.40 intratumorally, intraperitoneally, or intravenously. In some embodiments, the activated immune cells are administered at about 107 to about 109 cells per dose. In some embodiments, the methods of treatment described herein further comprise treating the patient with chemotherapy, radiation therapy, or an immune checkpoint inhibitor. In some embodiments, the method comprises treating the patient with irradiation. In some embodiments, the site of irradiation is different from the site of the cancer to be treated. Patient [0412] In some embodiments, the patient has a hematological cancer. [0413] In some embodiments, the patient has an advanced cancer. In some embodiments, the patient has a late-stage cancer. In some embodiments, the patient has a cancer that is stage II, III, or IV. In some embodiments, the patient has metastases. In some embodiments, the patient is a terminally ill patient. [0414] In some embodiments, the patient is a female. In some embodiments, the patient is a male. [0415] In some embodiments, the patient is a human. In some embodiments, the patient is at least about any of 50, 55, 60, 65, 70, or 75 years old. In some embodiments, the patient is less than about any of 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 years old. [0416] In some embodiments, the individual (e.g., an individual with a hematological cancer) has a PBMC or bone marrow (BM) sample that comprises no more than about any of 5%, 4%, 3%, 2% or 1% of the cells being monocytes (e.g., cancer monocytes). [0417] In some embodiments, the individual has a PBMC or BM sample that comprises at least about any of 20%, 30%, 40%, 50%, 60%, 70%, or 80% of the cells being hematological cancer cells (e.g., B-ALL cells, CLL cells, NHL cells, CML cells, or AML cells). [0418] In some embodiments, the individual has a PBMC or bone marrow (BM) sample that: a) comprises no more than about any of 5%, 4%, 3%, 2% or 1% of the cells being monocytes (e.g., cancer monocytes), and b) comprises at least about any of 20%, 30%, 40%, 50%, 60%, 70%, or 80% of the cells being hematological cancer cells (e.g., B-ALL cells, CLL cells, NHL cells, CML cells, or AML cells). Cancer [0419] As discussed above, the methods of treatment that involve immune cells (such as T cells) activated by APCs produced by various methods described herein are applicable to ny-2770598
Attorney Docket No.24516-20006.40 various types of cancer, including a non-hematological cancer in an individual who had, has, or is at risk of developing a hematological cancer. In some embodiments, the non- hematological cancer is associated with the hematological cancer. [0420] In some embodiments, the cancer is a hematologic cancer. [0421] In some embodiments, the cancer is an advanced cancer. In some embodiments, the cancer is a late-stage cancer. In some embodiments, the cancer is stage II, III, or IV. In some embodiments, the cancer is malignant. [0422] In some embodiments, the cancer has been subjected to and/or failed one or more prior therapy (e.g., an immune checkpoint blockage therapy (e.g., an PD-1 antibody), a chemotherapy, a surgery, a cell therapy (e.g., an allogenic NK cell infusion therapy)). [0423] In some embodiments, the cancer is a recurrent or refractory cancer. [0424] Examples of cancers described herein include, but are not limited to, acute lymphoblastic leukemia, chronic myelogenous leukemia, Hodgkin’s lymphoma, non- Hodgkin’s lymphoma, leukemia, B-cell chronic lymphocytic leukemia, acute myeloid leukemia, AIDS-related cancers (e.g., AIDS-related lymphoma), central nervous system lymphoma, chronic myeloproliferative disorders, lymphoid neoplasm (e.g., lymphoma), myelodysplastic syndromes, myelodysplastic/myeloproliferative diseases, lymphoma, primary central nervous system lymphoma (microglioma), and post-transplant lymphoproliferative disorder (PTLD). [0425] In some embodiments, the cancer is a virus-infection-related cancer. In some embodiments, the cancer is human T-lymphotrophic virus (HTLV-1)-related cancer (e.g., adult T cell leukemia or lymphoma). In some embodiments, the cancer is Epstein-Barr virus (EBV)-related cancer (e.g., Burkitt lymphoma, Hodgkin’s and non-Hodgkin’s lymphoma). In some embodiments, the cancer is hepatitis C virus-related cancer (e.g., non-Hodgkin’s lymphoma). Dosing and Method of Administering the activated immune cells (e.g., T cells) [0426] The activated immune cells can be administered at any desired dosage, which in some aspects includes a desired dose or number of cells or cell type(s). Thus, the dosage of cells in some embodiments is based on a total number of cells (or number per kg body weight) and/or a desired ratio of the individual populations. In some embodiments, the dosage of cells is ny-2770598
Attorney Docket No.24516-20006.40 based on a desired total number (or number per kg of body weight) of cells in the individual populations or of individual cell types. [0427] In certain embodiments, the activated immune cells (e.g., T cells, e.g., CD4 and/or CD8 T cells, e.g., TILs), are administered to the subject at a range of about one million to about 100 billion cells and/or that amount of cells per kilogram of body weight, such as, e.g., 1 million to about 50 billion cells (e.g., about 5 million cells, about 25 million cells, about 500 million cells, about 1 billion cells, about 5 billion cells, about 20 billion cells, about 30 billion cells, about 40 billion cells, or a range defined by any two of the foregoing values), such as about 10 million to about 100 billion cells (e.g., about 20 million cells, about 30 million cells, about 40 million cells, about 60 million cells, about 70 million cells, about 80 million cells, about 90 million cells, about 10 billion cells, about 25 billion cells, about 50 billion cells, about 75 billion cells, about 90 billion cells, or a range defined by any two of the foregoing values), and in some cases about 100 million cells to about 50 billion cells (e.g., about 120 million cells, about 250 million cells, about 350 million cells, about 450 million cells, about 650 million cells, about 800 million cells, about 900 million cells, about 3 billion cells, about 30 billion cells, about 45 billion cells) or any value in between these ranges and/or per kilogram of body weight. Dosages may vary depending on attributes particular to the disease or disorder and/or patient and/or other treatments. [0428] In some embodiments, for example, where the subject is a human, the dose includes fewer than about 1×109 total activated immune cells, e.g., in the range of about 1×106 to 5×108 such cells, such as 2×106, 5×106, 1×107, 5×107, 1×108, 5×108 or 1×109 total such cells, or the range between any two of the foregoing values. [0429] In some embodiments, the activated immune cells are administered at about 107 to about 109 cells per dose. [0430] In some embodiments, the method of treatment comprises administration of a dose comprising a number of cells from about 1×106 to 1×109 (e.g., 106 to 107, 107 to 108, or 108 to 109) total activated immune cells (e.g., total CD3 T cells, both CD4 and CD8 T cells, CD4 T cells only, CD8 T cells only, or TILs). [0431] In some embodiments, the dose of the activated immune cells is administered to the subject as a single dose or is administered only one time within a period of two weeks, one month, three months, six months, 1 year, or more. ny-2770598
Attorney Docket No.24516-20006.40 [0432] In some embodiments, the dose of total activated immune cells is within a range of between at or about 104 and at or about 109 cells/kilograms (kg) body weight, such as between 105 and 106 cells / kg body weight, for example, at or about 1×105 cells/kg, 1.5×105 cells/kg, 2×105 cells/kg, or 1×106 cells/kg body weight. For example, in some embodiments, the activated immune cells are administered at, or within a certain range of error of, between at or about 104 and at or about 109 T cells/kilograms (kg) body weight, such as between 105 and 106 T cells / kg body weight, for example, at or about 1×105 T cells/kg, 1.5×105 T cells/kg, 2×105 T cells/kg, or 1×106 T cells/kg body weight. [0433] In some embodiments, the activated immune cells are administered at or within a certain range of error of between at or about 104 and at or about 109 CD4+ and/or CD8+ cells/kilograms (kg) body weight, such as between 105 and 106 CD4+ and/or CD8+cells / kg body weight, for example, at or about 1×105 CD4+ and/or CD8+ cells/kg, 1.5×105 CD4+ and/or CD8+ cells/kg, 2×105 CD4+ and/or CD8+ cells/kg, or 1×106 CD4+ and/or CD8+ cells/kg body weight. [0434] In some embodiments, the activated immune cells are administered at or within a certain range of error of, greater than, and/or at least about 1×106, about 2.5×106, about 5×106, about 7.5×106, or about 9×106 CD4+ cells, and/or at least about 1×106, about 2.5×106, about 5×106, about 7.5×106, or about 9×106 CD8+ cells, and/or at least about 1×106, about 2.5×106, about 5×106, about 7.5×106, or about 9×106 T cells. In some embodiments, the activated immune cells are administered at or within a certain range of error of between about 108 and 1012 or between about 1010 and 1011 T cells, between about 108 and 1012 or between about 1010 and 1011 CD4+ cells, and/or between about 108 and 1012 or between about 1010 and 1011 CD8+ cells. [0435] For the prevention or treatment of disease, the appropriate dosage may depend on the type of disease to be treated, the type of cells or recombinant receptors, the severity and course of the disease, whether the activated immune cells are administered for preventive or therapeutic purposes, previous therapy, the subject's clinical history and response to the activated immune cells, and the discretion of the attending physician. The compositions and cells are in some embodiments suitably administered to the subject at one time or over a series of treatments. [0436] In some embodiments, the APCs described herein are administered to the subject at a range of about 5000 to about 10,000 cells/mm3 per tumor mass. ny-2770598
Attorney Docket No.24516-20006.40 [0437] In some aspects, the size of the dose is determined based on one or more criteria such as response of the subject to prior treatment, e.g., chemotherapy, disease burden in the subject, such as tumor load, bulk, size, or degree, extent, or type of metastasis, stage, and/or likelihood or incidence of the subject developing toxic outcomes, e.g., CRS, macrophage activation syndrome, tumor lysis syndrome, neurotoxicity, and/or a host immune response against the activated immune cells being administered. [0438] In some aspects, the size of the dose is determined by the burden of the disease or condition in the subject. For example, in some aspects, the number of cells administered in the dose is determined based on the tumor burden that is present in the subject immediately prior to administration of the initiation of the dose of cells. In some embodiments, the size of the first and/or subsequent dose is inversely correlated with disease burden. In some aspects, as in the context of a large disease burden, the subject is administered a low number of cells. In other embodiments, as in the context of a lower disease burden, the subject is administered a larger number of cells. [0439] The activated immune cells and/or a second agent described herein (e.g., a TNF^ inhibitor) can be administered by any suitable means, for example, by bolus infusion, injection (e.g., intravenous or subcutaneous injections, intraocular injection, periocular injection, subretinal injection, intravitreal injection, trans-septal injection, subscleral injection, intrachoroidal injection, intracameral injection, subconjectval injection, subconjuntival injection, sub-Tenon's injection, retrobulbar injection, peribulbar injection), or posterior juxtascleral delivery. In some embodiments, activated immune cells and/or a second agent described herein (e.g., a TNF^ inhibitor) are administered by parenteral, intrapulmonary, intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. In some embodiments, a given dose is administered by a single bolus administration of the activated immune cells. In some embodiments, the dose is administered by multiple bolus administrations of the activated immune cells, for example, over a period of no more than 3 days, or by continuous infusion administration of the activated immune cells. [0440] In some embodiments, the activated immune cells are administered intratumorally, intraperitoneally, or intravenously. ny-2770598
Attorney Docket No.24516-20006.40 Combination therapy [0441] In some embodiments, the activated immune cells are administered as part of a combination treatment, such as simultaneously with, concurrently with, or sequentially with, another therapeutic intervention (i.e., a second therapy), such as an antibody or engineered cell or receptor or agent, such as a cytotoxic or therapeutic agent. In some embodiments, the activated immune cells are administered prior to another therapeutic intervention. In some embodiments, the activated immune cells are administered after another therapeutic intervention. The activated immune cells in some embodiments are co-administered with one or more additional therapeutic agents or in connection with another therapeutic intervention, simultaneously, concurrently, or sequentially in any order. In some contexts, the activated immune cells are co-administered with another therapy sufficiently close in time such that the cell populations enhance the effect of one or more additional therapeutic agents, or vice versa. In some embodiments, the activated immune cells are administered prior to the one or more additional therapeutic agents. In some embodiments, the activated immune cells are administered after the one or more additional therapeutic agents. In some embodiments, the one or more additional agents to be administered includes a cytokine, such as IL-2, for example, to enhance persistence of the activated immune cells. In some embodiments, the methods comprise administration of a chemotherapeutic agent. [0442] In some embodiments, the second therapy comprises a TNF^ inhibitor (e.g., an anti- TNF^ antibody). In some embodiments, the TNF^ inhibitor is administered to the individual prior to (e.g., at least 1, 2 or 3 days prior to) the administration of the activated immune cells. [0443] In some embodiments, the TNF^ inhibitor is selected from the group consisting of: a small molecule inhibitor, a neutralizing antibody, a TNF^ receptor blockade antibody, a soluble TNF^ receptor, a TNF^-targeting short interfering RNA (siRNA), a chemical inhibitor of TNF^ mRNA stability, an inhibitor of TNF^ converting enzyme (TACE), and derivatives thereof. In some embodiments, the TNF^ inhibitor is an anti-TNF^ neutralizing antibody. In some embodiments, the TNF^ inhibitor is an anti-TNF^ receptor blockade antibody. In some embodiments, the anti-TNF^ antibody is a monoclonal antibody. In some embodiments, the anti-TNF^ antibody is a chimeric, humanized, and/or fully human antibody. [0444] Suitable antibodies for use in the methods provided herein include, but are not limited to, Remicade® (Infliximab (Centocor)), and those antibodies described, for example, ny-2770598
Attorney Docket No.24516-20006.40 in U.S. Patent No.6,835,823; 6,790,444; 6,284,471; 6,277,969; 5,919,452; 5,698,195; 5,656,272; and 5,223,395 and in EP Patent No.0610201, the contents of each of which are hereby incorporated by reference in their entirety, or antibodies that bind to the same epitope as Remicade®. Other suitable anti-TNF^ antibodies for use in the methods provided herein are, by way of non-limiting example, Humira (Adalimumab (Abbott Laboratories, Esai)) as described in U.S. Patent No.6,090,382; 6,258,562; or 6,509,015 and related patents and applications, the contents of which are hereby incorporated by reference in their entirety; Simponi™ (Golimimab, CNTO 148 (Centocor)) as described in PCT Publication No. WO 02/12502 and related patents and applications, the contents of which are hereby incorporated by reference in their entirety; ART621 (Arana Therapeutics), SSS 07 (Epitopmics and 3SBio) or antibodies that bind to the same epitope as Humira, Simponi, ART621, or SSS 07. [0445] In some embodiments, the TNF^ inhibitor is a fusion protein. Suitable fusion proteins for use in the methods provided herein include, but are not limited to, Enbrel (Etanercept (Amgen)) and other fusion proteins or fragments thereof described in U.S. Patent No. 5,712,155, PCT Publication No. WO 91/03553, and related patents and applications, the contents of which are hereby incorporated by reference in their entirety. [0446] In some embodiments, the anti-TNF^ antagonist is a modified antibody antagonist or a non-antibody-based antagonist. Such antagonists include advanced antibody therapeutics, such as antibody fragments including, but not limited to, Cimzia™ (Certolizumab pegol, CDP870 (Enzon)), bispecific antibodies, Nanobodies® such as ABX 0402 (Ablynx), immunotoxins, and radiolabeled therapeutics; peptide therapeutics; gene therapies, particularly intrabodies; oligonucleotide therapeutics such as aptamer therapeutics, antisense therapeutics, interfering RNA therapeutics; and small molecules such as LMP-420 (LeukoMed) as described in EP Patent No.0767793, and related patents and applications, the contents of which are hereby incorporated by reference in their entirety. [0447] In some embodiments, the TNF^ inhibitor is an antibody, such as infliximab, adalimumab, etanercept, golimumab, and certolizumab. [0448] In some embodiments, the TNF^ inhibitor is administered systemically. [0449] In some embodiments, the TNF^ inhibitor is administered concurrently with the immune cells described herein. 111 ny-2770598
Attorney Docket No.24516-20006.40 [0450] In some embodiments, the individual does not develop cytokine release syndrome or pro-inflammatory organ damage. In some embodiments, administration of the TNF^ inhibitor does not compromise or weakly compromises tumor clearance. [0451] In some embodiments, the second therapy comprises a chemotherapy, radiation therapy, or an immune checkpoint inhibitor. In some embodiments, the second therapy is gene therapy (e.g., mRNA-based gene therapy). In some embodiments, the second therapy comprises administration of a cancer vaccine (such as mRNA-based cancer vaccine or DNA- based cancer vaccine). In some embodiments, the second therapy comprises administration of an oncolytic virus. In some embodiments, the activated immune cells are administered prior to the administration of the second therapy. In some embodiments, the activated immune cells are administered in a neoadjuvant setting. [0452] In some embodiments, the second therapy comprises treating the patient with irradiation. In some embodiments, the site of irradiation is different from the site of the cancer to be treated. [0453] Thus, for example, in some embodiments, there is provided a method of treating an individual having cancer, comprising administering to the individual an effective amount of an immune cell activated by any of the methods described herein, wherein the individual is treated with a radiation therapy, and wherein the site of the irradiation is different from the site of the cancer to be treated. [0454] In some embodiments, the radiation therapy is selected from the group consisting of external-beam radiation therapy, internal radiation therapy (brachytherapy), intraoperative radiation therapy (IORT), systemic radiation therapy, radioimmunotherapy, and administration of radiosensitizers and radioprotectors. In some embodiments, the radiation therapy is external-beam radiation therapy, optionally comprising three-dimensional conformal radiation therapy (3D-RT), intensity modulated radiation therapy (IMRT), photon beam therapy, image-guided radiation therapy (IGRT), and sterotactic radiation therapy (SRT). In some embodiments, the radiation therapy is brachytherapy, optionally comprising interstitial brachytherapy, intracavitary brachytherapy, intraluminal radiation therapy, and radioactively tagged molecules given intravenously. EMBODIMENTS [0455] Embodiment 1. A method of producing a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), comprising: ny-2770598
Attorney Docket No.24516-20006.40 contacting the hematological cancer cells obtained from an individual having the hematological cancer cells with one or more survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of 1) a STAT3 activator 2) a TNF^ receptor (TNFR) activator, 3) an interferon ^ (IFN^) receptor (IFNGR) activator, and 4) and an interleukin-4 receptor (IL-4R) activator, thereby producing the population of HC-APCs. [0456] Embodiment 2. A method of producing activated immune cells, comprising: a) producing a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”) according to method of embodiment 1, and b) contacting the HC-APCs with immune cells, thereby producing activated immune cells. [0457] Embodiment 3. A method of producing activated immune cells, comprising contacting immune cells with a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), thereby generating activated immune cells; wherein the HC-APCs are derived from hematological cancer cells after having been contacted with one or more survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) and an interferon ^ (IFN^) receptor (IFNGR) activator, and 4) an interleukin-4 receptor (IL-4R) activator. [0458] Embodiment 4. The method of any one of embodiments 1-3, wherein the STAT3 activator is selected from the group consisting of: an IL-10, an IL-10 family member, an IL-10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, a small molecule activator of IL-10R, and a small molecule activator of STAT3, optionally wherein the activator of STAT3 is selected from an IL-10 family cytokine, an IL-12 family cytokine, an IL-6 family cytokine, a small molecule STAT3 activator, and G-CSF . [0459] Embodiment 5. The method of embodiment 4, wherein the STAT3 activator is selected from the group consisting of IL-10, IL-22, IL-19, IL20, IL-24, IL12, IL-23, IL-6, colivelin TFA, Garcinone D, and G-CSF, optionally wherein the IL-10R activator is IL-10, IL-22, IL-19, IL-20, IL-24, IL-12, IL-23, Colivelin TFA, or Garcinone D. [0460] Embodiment 6. The method of any one of embodiments 1-5, wherein the one or more of S/D/M factors comprise an interleukin-4 receptor (IL-4R) activator, optionally ny-2770598
Attorney Docket No.24516-20006.40 wherein the IL-4R activator is selected from the group consisting of IL-4, an IL-4R agonist antibody, and a small molecule activator of IL-4R. [0461] Embodiment 7. The method of embodiment 6, wherein the IL-4R activator is IL-4. [0462] Embodiment 8. The method of any one of embodiments 1-7, wherein the one or more of S/D/M factors comprise a TNFR activator, optionally wherein the TNFR activator is selected from the group consisting of TNF^, a TNFR agonist antibody, and a small molecule activator of TNFR. [0463] Embodiment 9. The method of embodiment 8, wherein the TNFR activator is TNF^. [0464] Embodiment 10. The method of any one of embodiments 1-9, wherein the one or more of S/D/M factors comprise an IFNGR activator, optionally wherein the IFNGR activator is selected from the group consisting of IFN^, an IFNGR agonist antibody, and a small molecule activator of IFNGR. [0465] Embodiment 11. The method of embodiment 10, wherein the IFNGR activator is IFN^. [0466] Embodiment 12. The method of any one of embodiments 1-11, wherein the one or more of S/D/M factors are present in a single composition. [0467] Embodiment 13. The method of any one of embodiments 1-12, wherein the one or more of S/D/M factors comprise two or more agents selected from the group consisting of 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator. [0468] Embodiment 14. The method of any one of embodiments 1-13, wherein the one or more of S/D/M factors comprise three or more agents selected from the group consisting of 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator. [0469] Embodiment 15. The method of embodiment 14, wherein the one or more of S/D/M factors comprise 1) a STAT3 activator, 2) a TNFR activator, and 3) an IFNGR activator, optionally wherein the one or more of S/D/M factors comprise an IL-10 family cytokine (e.g., IL-10, IL-22, IL-19, IL-24, IL-20, IL-26), TNF^, and IFN^. [0470] Embodiment 16. The method of embodiment 15, wherein the one or more of S/D/M factors comprise 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, ny-2770598
Attorney Docket No.24516-20006.40 and 4) an IL-4R activator, optionally wherein the one or more of S/D/M factors comprise an IL-10 family cytokine (e.g., IL-10, IL-22, IL-19, IL-24, IL-20, IL-26), TNF^, IL-4, and IFN^. [0471] Embodiment 17. The method of any one of embodiments 1-16, wherein the one or more of the S/D/M factors further comprise an IL-6 receptor (IL-6R) activator and/or a GM-CSF receptor (GM-CSFR) activator, optionally wherein the IL-6R activator is selected from the group consisting of IL-6, an IL-6R agonist antibody, and a small molecule activator of IL-6R, and optionally wherein the GM-CSFR activator is selected from the group consisting of GM-CSF, a GM-CSFR agonist antibody, and a small molecule activator of GM- CSFR. [0472] Embodiment 18. The method of embodiment 17, wherein the IL-6R activator is IL-6, and wherein the GM-CSFR activator is GM-CSF. [0473] Embodiment 19. The method of any one of embodiments 1-18, wherein the hematological cancer cells are comprised in a mixture comprising monocytes from the individual. [0474] Embodiment 20. The method of any one of embodiments 1-19, wherein the method further comprises, prior to contacting the HC-APCs with immune cells, contacting the HC-APCs with one or more of refinement factors selected from the group consisting of type-I interferon, IFN^, TNF^, a TLR ligand, CD40L or a CD40-ligating antibody, an anti- PD-L1 antibody, and TPI-1, optionally wherein the type-I interferon comprises IFN^ and/or IFN^, optionally wherein the refinement factors is selected from the group consisting of R848, poly IC, CpG, or LPS, and further optionally wherein the refinement factors comprise R848 and poly IC. [0475] Embodiment 21. The method of embodiment 18, wherein the refinement factors comprise IFN^, IFN^, and TNF^. [0476] Embodiment 22. The method of embodiment 19, wherein the refinement factors further comprise at least two agents selected from the group consisting of poly IC, CpG, CD40L, R848, and an anti-PD-L1 antibody, optionally wherein the refinement factors comprise a SHP-1 inhibitor (e.g., TPI-1). [0477] Embodiment 23. The method of any one of embodiments 2-22, wherein the method further comprises administering the activated immune cells into the individual. ny-2770598
Attorney Docket No.24516-20006.40 [0478] Embodiment 24. The method of any one of embodiments 2-23, wherein the method further comprises administering the activated immune cells into a different individual. [0479] Embodiment 25. The method of any one of embodiments 2-24, wherein the immune cells and hematological cancer cells are from the same individual. [0480] Embodiment 26. The method of any one of embodiments 2-24, wherein the immune cells and hematological cancer cells are from the different individuals. [0481] Embodiment 27. The method of any one of embodiments 23-26, wherein the immune cells are administered to the individual where they are obtained from. [0482] Embodiment 28. The method of any one of embodiments 23-26, wherein the immune cells are administered to a different individual from the individual where they are obtained from. [0483] Embodiment 29. The method of any one of embodiments 2-28, the immune cells are selected from the group consisting of PBMC, cells derived from bone marrow biopsy, cells derived from lymphoma biopsy, tumor infiltrating T cells (TIL), and T cells, optionally wherein the immune cells are T cells, optionally wherein the T cells are CD8 T cells and/or CD4 T cells. [0484] Embodiment 30. The method of any one of embodiments 1-29, wherein the hematological cancer cells are obtained from PBMC, enriched leukemia cells, a bone marrow biopsy, or a lymphoma biopsy. [0485] Embodiment 31. The method of any one of embodiments 1-30, wherein the hematological cancer cells are cultured with the one or more of S/D/M factors for at least 2 days. [0486] Embodiment 32. The method of any one of embodiments 20-31, wherein the HC-APCs are cultured with the refinement factors for about 1-4 days. [0487] Embodiment 33. The method of any one of embodiments 2-32, wherein the immune cells have been enriched prior to contacting with the HC-APCs. [0488] Embodiment 34. The method of any one of embodiments 1-33, wherein the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors. ny-2770598
Attorney Docket No.24516-20006.40 [0489] Embodiment 35. The method of any one of embodiments 1-34, wherein the hematological cancer is a myeloid leukemia, optionally the hematological cancer is acute myeloid leukemia (AML) or chronic myeloid leukemia (CML), optionally wherein the hematological cancer cells are any of M0-M5 myeloblasts. [0490] Embodiment 36. The method of embodiment 35, wherein the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors in the presence of an anti-CD11b antibody. [0491] Embodiment 37. The method of any one of embodiments 1-33, wherein the hematological cancer is a B-cell lymphoma or B-cell leukemia. [0492] Embodiment 38. The method of embodiment 37, wherein the B-cell lymphoma or B-cell leukemia is selected from the group consisting of B-cell acute lymphoblastic leukemia B-ALL), chronic lymphocytic leukemia (CLL), and non-Hodgkin’s lymphoma (NHL). [0493] Embodiment 39. The method of embodiment 37 or embodiment 38, wherein the hematological cancer cells have been enriched prior to the contacting with the S/D/M factors in the presence of an anti-CD19 or anti-CD20 antibody. [0494] Embodiment 40. The method of any one of embodiments 2-39, wherein the method further comprises expanding the immune cells by contacting immune cells with the HC-APCs for at least two, three or four rounds. [0495] Embodiment 41. A population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”) obtained by the method of any one of embodiments 1, 4-22, 30-31 and 33-39. [0496] Embodiment 42. A population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), wherein the APCs are derived from hematological cancer cells, and wherein the APCs express increased levels of MHC-1, MHC- II, CD40, CD80, and/or CD86 than the hematological cancer cells prior to the treatment, optionally wherein the HC-APCs comprise one or more exogenous antigen. [0497] Embodiment 43. The population of HC-APC of embodiment 42, wherein the hematological cancer cells are primary cells from an individual having the hematological cancer. ny-2770598
Attorney Docket No.24516-20006.40 [0498] Embodiment 44. The population of HC-APC of embodiment 42 or embodiment 43, wherein the hematological cancer is a myeloid leukemia, optionally the hematological cancer is acute myeloid leukemia (AML) or chronic myeloid leukemia (CML), optionally wherein the hematological cancer cells are any of M0-M5 myeloblasts. [0499] Embodiment 45. The population of HC-APC of embodiment 42 or embodiment 43, wherein the hematological cancer is a B-cell lymphoma or B-cell leukemia, optionally wherein the B-cell lymphoma or B-cell leukemia is selected from the group consisting of B- cell acute lymphoblastic leukemia B-ALL), chronic lymphocytic leukemia (CLL), and non- Hodgkin’s lymphoma (NHL). [0500] Embodiment 46. A population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), prepared by a) obtaining hematological cancer cells from an individual having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of 1) a STAT3 activator 2) a TNF^ receptor (TNFR) activator, 3) an interferon ^ (IFN^) receptor (IFNGR) activator, and 4) and an interleukin-4 receptor (IL-4R) activator, thereby producing the population of HC-APCs. [0501] Embodiment 47. A population of activated immune cells obtained by the method of any one of embodiments 2-40. [0502] Embodiment 48. A method of treating a hematological cancer in an individual, comprising administering to the individual an effective amount of activated immune cells of embodiment 47. [0503] Embodiment 49. A method of treating a hematological cancer in an individual, comprising: a) contacting hematological cancer cells and/or monocytes with one or more of survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of 1) a STAT3 activator 2) a TNF^ receptor (TNFR) activator, 3) an interferon ^ (IFN^) receptor (IFNGR) activator, and 4) an interleukin-4 receptor (IL-4R) activator and, thereby producing a population of antigen ny-2770598
Attorney Docket No.24516-20006.40 presenting cells (“APCs”), optionally wherein the APCs are further loaded with one or more exogenous antigen, b) contacting the APCs with a population of immune cells from the individual, thereby producing activated immune cells, and c) administering activated immune cells into the individual. [0504] Embodiment 50. The method of embodiment 49, wherein the hematological cancer cells and/or monocytes are obtained from the individual, optionally wherein the hematological cancer cells and monocytes are comprised in a mixture when contacting with the one or more S/D/M factors. EXAMPLES [0505] The examples below are intended to be purely exemplary of the invention and should therefore not be considered to limit the invention in any way. The following examples and detailed description are offered by way of illustration and not by way of limitation. Example 1. Multi-targeting neoantigen-specific T cells (NeoT) for hematological malignancies [0506] The process is summarized in FIGs.1-3. Peripheral monocytes (cMO) are a common source for producing ^APC, termed cMO-^APC, which require uptake of cancer antigens in order for antigen presentation and activation of cancer-specific T cells (NeoT). Differently, ^APC differentiated from myeloid leukemia cells (AML/CML-^APC), B-lineage leukemia cells (ALL/CLL-^APC), and lymphoma cells (NHL-^APC) directly present inherent, mutation-associated neoantigens to activate NeoT. Irrespective of source cells from which ^APC are differentiated, the production of all ^APC involves a 2-day treatment with the HC- activator, optionally followed by 6-24hr treatment with an APC refinement agent. The quality control of ^APC final product involves flow cytometry assays testing cell surface expression of immunogenic antigen presentation machinery including MHC-I, MHC-II, CD80, CD86, CD40 and OX40L. See FIG.3. FIG.4 shows a table that summarizes results from representative studies that consistently evidenced that the methods discussed herein successfully make ^APC from either cMo or cancer cells of various sources. Step-1 Collection of Starting Materials [0507] All cancer patients were subjected to a round of mononuclear cell apheresis to obtain PBMC, from which peripheral monocytes (cMO), malignant leukemia cells (e.g., B-ALL, ny-2770598
Attorney Docket No.24516-20006.40 CLL), and peripheral blood lymphocytes (PBL) were further separated. cMO and malignant leukemia cells were used to generate ^APC, as well as for separation of PBL that are used to produce NeoT. [0508] Bone marrow aspiration and bone marrow biopsy: Leukemia of all forms originate from bone marrow where greater numbers of malignant cells and the cancer microenvironment reside. Bone marrow aspiration (liquid) and biopsies (solid part) hence provide malignant cell materials for preparing cancer neoantigens/antigens and also TILs from which NeoT are derived. Multi-point aspiration and biopsies that comprise diversity of cancer neoantigens/antigens are preferred. [0509] Lymphoma biopsies: For NHL, biopsies at the enlarged lymphomas were taken to obtain malignant cell materials and TILs. Step-2 Generating ^APC from cMO and malignant cells [0510] ^APC are differentiated APC produced from peripheral monocytes of cancer patients (cMO) or healthy donors (MO), or myelogenous or B cell-lineage malignant cells using an HC-activator reagent. [0511] Nomenclature: Dependent on the source from which the cells are derived, ^APC are further designated as: [0512] Common - require uptake of cancer antigens for activation of NeoT.
[0513] cMO-^APC: produced from cancer patient peripheral monocytes (cMO). cMO-^APC are common ^APC and can be used to activate autologous NeoT from patients of all types of cancer. cMO-^APC require uptake of cancer antigens in order for antigen presentation and activation of NeoT. [0514] MO-^APC: produced from healthy donor peripheral monocytes (MO). MO-^APC also common ^APC. Upon uptake of antigens, MO-^APC can activate antigen-specific T cells. [0515] Cancer type-specific ^APC – direct activation of NeoT by presenting inherent cancer neoantigens. [0516] AML-^APC: produced from AML myelogenous cancer cells. [0517] CML-^APC: produced from CML myelogenous cancer cells. [0518] ALL-^APC: produced from B-ALL malignant B lymphoblastic cells. ny-2770598
Attorney Docket No.24516-20006.40 [0519] CLL-^APC: produced from CLL malignant B lymphocytes. [0520] NHL-^APC: produced from B-lineage NHL lymphoma cells. [0521] NeoT: polyclonal multitargeting cancer neoantigen-specific CD4 and CD8 T cells produced by ^APC-mediated antigen presentation that selectively activates NeoT, resulting in NeoT expansion. NeoT detect and kill malignant cells through TCRs recognizing neoantigens displayed by MHC molecules. Cancer neoantigens include gene mutation- resultant novel antigens (major category), virus infection-resultant non-self-antigens, as well as self-antigens expressed aberrantly that breach immune tolerance. [0522] Irrespective of the source from which ^APC are produced, all ^APC are produced following a similar scheme: a minimum 1-2 days treatment of the source cells with HC- activator, optionally followed by 6-24h treatment with the APC refinement agent. HC-activator [0523] The HC-activator contains selective cytokines that support the source cell (e.g., cancer monocytes, e.g., hematological cancer cells) survival and differentiation into antigen- presenting cells (^APC). The product ^APC post HC-activator treatment are professional APCs that are highly phagocytic and feature a proinflammatory phenotype proficient for immunogenic antigen presentation and activation of NeoT. Six-component HC-activator [0524] The six-component HC-activator contains: a) a STAT3 activator, b) IFN^, c) TNF^, d) IL-4, e) GM-CSF, and f) IL-6. Exemplary STAT3 activators are listed in Table 1 below, and the following STAT3 activators have been tested, and demonstrated comparable effects: IL-10, IL-22, IL-19, IL-20, IL-24, IL-26, IL-12, colivelin TFA, and Garcinone D. It was found that the six-component HC-activator unanimously achieved at least about 80%-90% survival and differentiation of ^APC derived from hematological cancer cells from various cancer patients having different types of hematological cancers such as those discussed in the Examples below. Four-component HC-activator [0525] The four-component HC-activator does not contain IL-6 and GM-CSF. It has: a) a STAT3 activator, b) IFN^, c) TNF^, and d) IL-4. Exemplary STAT3 activators are listed in Table 1 below. The four-component HC-activator is expected to achieve comparable effects as the six-component HC-activator. ny-2770598
Attorney Docket No.24516-20006.40 Three-component HC-activator [0526] The three-component HC-activator has: a) a STAT3 activator, b) IFN^, and c) TNF^. Exemplary STAT3 activators are listed in Table 1 below. The three-component HC-activator is expected to achieve at least 80% of the effectiveness of the 6-component or 4-component HC-activator, including keeping the hematological cancer cells alive for one or more weeks and promoting the differentiation of these cells to ^APCs. Table 1. STAT3 activators that can be used in the HC-activator.
[0527] The six-component HC-activator can be prepared by having a cell culture medium (e.g., RPMI 1640 with heat-inactivated human serum) supplemented with a stock solution (10x), which contains human IL-10 and IFN^, TNF^, IL-4, GM-CSF, and IL-6. Alternatively, recombinant human IL-10 in the stock was replaced with another IL-10 family cytokine, recombinant human IL-12 family, Colivelin TFA, or Garcinone D to form the stock. According, the HC-activator includes about IL-10 (or other IL-10 family or IL-12 family cytokines in lieu of IL-10), IFN^, TNF^, IL-4, GM-CSF, and IL-6. The concentrations of the exemplary STAT3 activators are shown in Table 1. ny-2770598
Attorney Docket No.24516-20006.40 [0528] It was also found that T cell medium prepared as follows can achieve comparable results due to the presence of the key ingredients in the T cell medium. [0529] Modified Act-T medium: The TCR-activated T cells are prepared by treating peripheral blood lymphocytes (PBL) cultured with anti-human CD3 antibody and anti-human CD28 antibody to induce T cell activation. Cell culture medium is collected on day 2 (48h) or day 3 (72h), designated as IL-2high Act-T medium or IL-2low Act-T medium, respectively. Both IL-2high and IL-2low Act-T medium contain high levels of IFN^ (> 50ng/ml) and TNF^ (> 3ng/ml), and IL-4 (> 20pg/ml), GM-CSF (> 300pg/ml) and IL-6 (>1ng/ml). [0530] To prepare the HC-activator, the IL-2low Act-T medium was further mixed with a cell culture medium (e.g., RPMI 1640 with heat-inactivated human AB serum) and further supplemented with: a) recombinant human IL-10 to form HC-activator, b) another IL-10 family cytokine to form HC-activator, c) recombinant human IL-12 to form HC-activator, d) Colivelin TFA to form HC-activator, or e) Garcinone D to form HC-activator. [0531] Alternatively, IL-2high Act-T medium with IL-2 depletion (using anti-IL-2-conjugated beads) can be used to prepare the HC-activator series. Note: IL-2high HC-activator medium is a natural T cell growth medium. Optional APC Refinement Agent [0532] The optional APC refinement agent is a reagent complex that can be used optionally to further refine the ^APC phenotype. For convenience, HC-activator-optional APC refinement agent (or HC-activator/optional APC refinement agent, or similar expressions in this application) refers to the treatment of HC-activator alone or combined treatment of HC- activator and the optional APC refinement agent. It was found that the optional APC refinement agent treatment can further refine ^APC but is not required for generation of potent ^APC. [0533] The optional APC refinement agent comprises cytokines (choices of IFN^, IFN^, and TNF^), TLR ligands (choices of Poly IC, CpG, and R848), and/or TPI-1. For the data included herein, the optional APC refinement agent includes R848 and Poly IC. Step-3 Loading cMO-^APC With Tumor Antigens [0534] Only cMO-^APC are required to ‘feed’ with cancer antigens in order for antigen presentation and activation of NeoT. For this, cMO-^APCs in culture with the optional APC refinement agent were incubated with cancer antigen materials, such as freeze-thaw cancer ny-2770598
Attorney Docket No.24516-20006.40 biopsy debris or synthetic neoantigen peptides, for a minimum of 4h (4-16h) to enable cMO- ^APC phagocytosis of cancer materials, processing antigens, and loading antigens onto MHC molecules. [0535] For hematological malignancies, cancer materials were prepared from enriched leukemia cells, bone marrow biopsies and lymphoma biopsies following a cycle of freeze- thaw treatment. Alternatively, enriched cancer cells were treated with 30Gy X-ray radiation and followed by a cycle of freeze-thawing to produce cell debris. [0536] Different from cMO-^APC, ^APC produced from malignant cells, i.e., AML-^APC, CML-^APC, ALL-^APC CLL-^APC and NHL-^APC, directly present their inherent mutation-associated neoantigens to cognate T cells, resulting in NeoT activation. These ^APC do NOT require uptake of additional cancer antigens. Step-4 ^APC antigen presentation activating NeoT from PBL and/or TIL [0537] Antigen-loaded cMO-^APC, or prepared hematological malignant cell ^APC (i.e., AML-^APC, CML-^APC, ALL-^APC, CLL-^APC and NHL-^APC) cultures in the optional APC refinement agent were exposed to peripheral blood lymphocytes (PBL) at a ratio of 1:100-500 of ^APC: PBL. The co-culture is gently tilted to enhance cognate engagement of ^APC and cancer antigen specific T cells (NeoT). The ^APC-NeoT co-culture was then maintained for 6-10 days with supplementation of IL-2 (also IL-7 and IL-15), refreshed medium, and cell splitting to support NeoT activation and expansion. [0538] NeoT can also be expanded from TILs. To obtain TILs, fresh tumor biopsies or surgically resected tumor tissues were dissociated into single cells, followed by positive isolation of TILs using anti-CD3-conjugated beads. For expansion of NeoT, cancer antigen- loaded cMO-^APC, or prepared hematological malignant cell ^APC (i.e. AML-^APC, CML- ^APC, ALL-^APC CLL-^APC or NHL-^APC) were co-cultured with isolated TILs at the ratio of ^APC: TIL is 1:10-100. The NeoT cells were cultured for the next 6-10 days with IL- 2 supplementation (also IL-7 and IL-15), refreshed medium, and cell splitting to support NeoT activation and expansion. [0539] Alternatively, PBL and TILs can be combined to co-culture with antigen-loaded cMO-^APC or hematological malignant cell ^APC for NeoT selection, activation, and expansion. ny-2770598
Attorney Docket No.24516-20006.40 Step-5 Large expansion of NeoT for adoptive cell therapy (ACT) [0540] Step-4 is the 1st round of antigen-specific NeoT activation and expansion, which generally harvest ~107-8 NeoT in ~10 days. [0541] To obtain sufficient NeoT for adoptive cell therapy, additional rounds of antigen- specific NeoT expansion can be performed following the same procedure of Step-4. Each round further expands NeoT approximately 50-100 fold. After the 3rd expansion, the NeoT number reaches > 1010. The final NeoT cells were washed and suspended in cold Plasma- Lyte A, ready for intravenous (i.v.) administration to the same patient for autologous adoptive cell therapy (ACT). The 2nd and 3rd rounds of large expansion together take 12-20 days. [0542] Alternatively, the 2nd and 3rd rounds of NeoT expansion can be done by antibody ligation of CD3 and CD28, a step that leads to T cell activation and proliferation. As this method is less optimal, it is only used when ^APC or cancer antigen materials are insufficient for large-scale NeoT expansion. Example 2. HC-activator and optional APC refinement agent differentiate B-lineage malignant leukocytes, along with cMO, into CLL-^APC for activating NeoT [0543] B-lineage malignancies include B-ALL, CLL, MM and NHL, together occupying > 50% of all cases of hematological cancer. [0544] Multiple PBMC samples were analyzed from patients of B-ALL (5 samples), CLL (1 sample), MM (2 samples, and NHL (1 sample) for the presence of cancer cells, cMO, and CD4 and CD8 T cells. Matched BM samples for B-ALL (# BAL1) and MM, and dissociated lymphoma tumor cells (DTC) from NHL were also obtained for analyses. Both cMO and malignant B lymphocytic cells from these samples were treated with the HC-activator (2d) and the optional APC refinement agent (1d) for ^APC differentiation. The final cell products were analyzed for the antigen presentation phenotype by cell surface expression of MHC-I, MHC-II, CD80, CD86, and CD40. [0545] Consistent with previous findings, cMO were responsive to the HC-activator (2d) and optionally APC refinement agent (1d) treatment and differentiated into cMO-^APC. However, five newly diagnosed B-ALL patients were all depleted of cMO in PBMC and BM, and thus were unable to produce sufficient cMO-^APC for antigen presentation to activate NeoT. [0546] It was found that malignant B lymphoblasts/lymphocytic cells from B-ALL and CLL, and B lymphocytes from NHL responded to the HC-activator and optionally APC refinement ny-2770598
Attorney Docket No.24516-20006.40 agent treatment, differentiating into ALL-^APC, CLL-^APC, and NHL-^APC. On the other hand, malignant plasma cells from MM were non-responsive and were unable to differentiate MM-^APC. FIG.4 summarizes the representative studies. Representative sample analysis details are presented in FIGs.5A-9B. B-ALL (FIGs.5A-7B) [0547] PBMC and BM samples from five newly diagnosed B-ALL patients were all depleted of cMO, and thus were unable to provide sufficient cMO-^APC. However, all samples were associated with high frequencies of malignant B lymphoblasts, which were responsive to the HC-activator and the optional APC refinement agent treatment, and therefore differentiated into ALL-^APC. [0548] FIGs.5A-7B show studies of three cases of B-ALL patients (Karnelian #: BAL3, 4 & 5). The HC-activator-optional APC refinement agent treatment induced malignant B lymphoblasts phenotypic changes into ALL-^APC, which demonstrated an immunogenic antigen presentation machinery featured with high expression of MHC-I, MHC-II, CD80, CD86, and CD40 on the cell surface. ALL-^APC also demonstrated a spread-cell morphology that was different from malignant B lymphoblasts. [0549] FIGs.5A-5C show that the HC-activator (optionally also with the optional APC refinement agent) differentiate malignant B lymphoblasts from B-ALL into ALL-^APC. FIG.5A shows analyses of PBMC from a patient with newly diagnosed B-ALL (Karnelian # BAL3). Malignant B lymphoblasts (B-ALL cells) occupied 76% of total PBMC, while monocytes (cMO) were diminished (0.38%). Both CD4 and CD8 T cells were present. FIG. 5B shows that the HC-activator (optionally also with the optional APC refinement agent) for treating malignant B lymphoblasts induced increased expression of antigen presentation molecules, including MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86, indicating cell differentiation into professional APC, termed ALL-^APC. [0550] FIG.5C shows representative microscopic images of malignant B lymphoblasts prior to and after HC-activator treatment (optionally with additional APC refinement agent treatment). Differentiation of ALL-^APC was associated with cell morphology changes. [0551] FIGs.6A-6B show that HC-activator treatment (optionally with additional APC refinement agent treatment) differentiated malignant B lymphoblasts from B-ALL into ALL- ^APC. FIG.6A shows analyses of PBMC from a newly diagnosed B-ALL patient (Karnelian ny-2770598
Attorney Docket No.24516-20006.40 # BAL4). Malignant B lymphoblasts (B-ALL cells) occupied 86.8% of total PBMC, and monocytes (cMO) were diminished (0.91%). CD4 and CD8 T cells were present. [0552] FIG.6B shows that HC-activator treatment (optionally with additional APC refinement agent treatment) for malignant B lymphoblasts induced their differentiation into ALL-^APC featured with increased expression of antigen presentation molecules MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86. [0553] FIGs.7A-7B show that HC-activator treatment (optionally with additional APC refinement agent treatment) differentiated malignant B lymphoblasts from B-ALL into ALL- ^APC. FIG.7A shows the analyses of PBMC from a newly diagnosed B-ALL patient (Karnelian # BAL5). Malignant B lymphoblasts (B-ALL cells) occupied 86.8% of the total PBMC, and monocytes (cMO) were diminished (0.91%). Frequencies of CD4 and CD8 T cells were substantially reduced. [0554] FIG.7B shows that HC-activator treatment (optionally with additional APC refinement agent treatment) of B lymphoblasts induced their differentiation into ALL-^APC that featured increased expression of antigen presentation molecules MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86. CLL (FIGs.8A-8B) [0555] The HC-activator/optional APC refinement agent treatment induced differentiation of both cMO and malignant B lymphocytic cells into cMO-^APC and CLL-^APC. PBMC samples of CLL were obtained from Discovery Life Science (Karnelian #: CLL1) of a patient that was clinically confirmed to have the disease. [0556] FIGs.8A-8B show that HC-activator treatment (optionally with additional APC refinement agent treatment) differentiated peripheral monocytes (cMO) and malignant B lymphocytic cells from CLL into cMO-^APC and CLL-^APC. FIG.8A shows that analyses of PBMC from a stage III CLL patient (Karnelian # CLL1) revealed that malignant B lymphocytic cells (CLL cells) occupied 4.7% of the total PBMC. Monocytes (cMO, 11.2%) and CD4 and CD8 T cells were present. FIG.8B shows that HC-activator treatment (optionally with additional APC refinement agent treatment) for PBMC induced differentiation of both cMO and malignant B lymphocytic cells into ^APC, cMO-^APC, and CLL-^APC, respectively. Both cMO-^APC and CLL-^APC featured increased expression of antigen presentation molecules MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86. ny-2770598
Attorney Docket No.24516-20006.40 Non-Hodgkin’s lymphoma (NHL) (FIGs.9A-9B) [0557] Dissociated tumor cells (DTC) from the tumor of non-Hodgkin’s lymphoma (NHL) were obtained from Discovery Life Science. Treating DTC with HC-activator (2d) (and optionally with APC refinement agent (1d)) differentiated malignant B lymphocytes (NHL cells) into professional antigen presenting cells, termed NHL-^APC, which featured increased expression of MHC-I, MHC-II, CD80, CD86, and CD40. [0558] FIGs.9A-9B show that HC-activator treatment differentiated malignant B lymphocytes from non-Hodgkin’s lymphoma (NHL) into NHL-^APC. FIG.9A shows the analyses of dissociated tumor cells (DTC) from lymphoma of an NHL patient. Malignant B lymphocytes (NHL cells) occupied 65.8% of the total tumor cells. CD4 and CD8 T cells were detected in DTC. [0559] FIG.9B shows that HC-activator treated DTC induced differentiation of malignant B lymphocytes (NHL cells) into NHL-^APC, which featured increased expression of antigen presentation molecules MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86. Multiple myeloma (MM) (FIGs.10A-10B) [0560] PBMC and BM samples from patients of MM were analyzed by flow cytometry. MM cancer cells were found primarily in BM, whereas PBMC contained monocytes (cMO), CD4 lymphocytes, and CD8 lymphocytes. Despite MM cancer cells being B-lineage cells, treating these cells with HC-activator (2d) and optional APC refinement agent (1d) failed to induce their differentiation into phenotypic antigen presenting cells. However, treating cMo from MM patients induced differentiation of cMO-^APC that featured increased expression of MHC-I, MHC-II, CD80, CD86, and CD40. Thus, for developing NeoT therapy, cMO-^APC were used. Given that cMO were not malignant cells, cMO-^APC were required to phagocytose MM antigens prior to being used for antigen presentation and activation of NeoT. [0561] FIGs.10A-10B show that HC-activator differentiated cMO but not malignant plasma cells of MM into ^APC. FIG.10A shows that the analyses of PBMC and BM samples from an MM patient revealed the presence of malignant plasma cells (MM), as well as CD4 and CD8 T cells, in both PBMC and BM. There were cMO and CD4 and CD8 T cells in PBMC. FIG.10B shows that HC-activator treated PBMC (optionally additionally with optional APC refinement agent treatment) induced differentiation of cMO into ^APC. cMO-^APC featured increased expression of antigen presentation molecules MHC-I and MHC-II and co- ny-2770598
Attorney Docket No.24516-20006.40 stimulatory molecules CD40, CD80, and CD86. However, the same treatment failed to induce differentiation of malignant plasma cells (MM) from PBMC or BM into ^APC. Example 3. B-ALL-^APC activate NeoT B-ALL for autologous cell therapy for B-ALL [0562] PBMC and BM samples from a newly diagnosed B-ALL patient were used to prepare ALL-^APC, which were used to “call” (selection and activation) NeoT B-ALL from PBL and TILs (in this case: T cells in BM). NeoT B-ALL were tested in vitro for cytotoxicity against B- ALL cells and in vivo by adoptive cell therapy (ACT) to PDXs (i.e., patient-derived xenografts) of B-ALL cancer established in immunocompromised NSG mice. [0563] FIGs.11A-13F show details of the study. [0564] FIG.11A shows analyses of PBMC and BM mononuclear cells (BMMC) from a patient with newly diagnosed B-ALL (top panel) and PBMC from a healthy donor (bottom panel). Malignant B lymphoblasts (B-ALL cells) occupied 67.3% of total cells in PBMC and 81.8% of BMMC. Monocytes (cMO) were diminished in PBMC and BM, whereas CD4 and CD8 T cells were present with reduced frequencies. A sample of PBMC from a healthy donor was analyzed in parallel as comparison. [0565] FIGs.11B-11E show the HC-activator treatment (optionally additionally with optional APC refinement agent treatment) differentiated malignant B lymphoblasts into ALL- ^APC. FIG.11B shows the experimental scheme. PBMC and BM mononuclear cells (BMMC) from B-ALL were separated by CD3 selection into CD3+ T cell population (PBL and TILs) and CD3- cells that were primarily malignant B lymphoblasts (> 80%). The malignant B lymphoblasts were treated with HC-activator and optionally also with the optional APC refinement agent to differentiate ALL-^APC, which were then used for antigen presentation and activation of NeoT that were derived from PBL and TILs. NeoT were tested in vitro and in vivo for B-ALL cancer cell elimination. FIG.11C shows analyses of malignant B lymphoblasts prior to, and after, the HC-activator and optional APC refinement agent treatment and differentiation of the cells into ALL-^APC. ALL-^APC featured increased expression of MHC-I, MHC-II, CD80, CD86, and CD40. FIG.11D shows that co- incubation of ALL-^APC and PBL or TILs led to ALL-^APC antigen specific engagement with cognate NeoT (light gray), resulting in NeoT activation and expansion (right). FIG.11E shows that two rounds of ALL-^APC-mediated antigen-specific activation of NeoT led to large expansion of NeoT cells, within which effector memory T cells (TEM) occupied high frequencies. ny-2770598
Attorney Docket No.24516-20006.40 [0566] FIGs.12A-12C show in vitro NeoTB-ALL killing of malignant B lymphoblasts. FIG. 12A shows the results of the specific lysis of malignant B lymphoblasts in BM samples harvested from the patient that were incubated with either non-specifically expanded T cells (by CD3/CD28 ligation) or NeoTB-ALL at the ratio of BM cell:T cell =1:1 for 18 hours. Malignant B lymphoblasts in BM prior to and after incubation with T cells were analyzed by flow cytometry. Data show that NeoTB-ALL mediated potent killing towards malignant B lymphoblasts. FIG.12B shows the results of IFN^ ELISpot indicating NeoTB-ALL released IFN^ during activation and killing of malignant B lymphoblasts. FIG.12C shows still-frames from time-lapse video clips of the NeoTB-ALL killing of malignant B lymphoblasts. [0567] FIGs.13A-13F show in vivo testing of NeoTB-ALL by adoptive cell therapy (ACT) into patient-derived xenograft (PDX) models of B-ALL. As shown in FIG.13A, the BM and PBMC mixture from the B-ALL patient was engrafted into NSG mice to establish PDX models. After B-ALL was stabilized (3rd generation PDX), NeoTB-ALL ACT was administered (i.v.) in a dose-escalation manner. Control experiments were done by giving non-specific T cells of the same number. As shown in FIG.13B, two doses of 5×106 NeoTB-ALL given on day 0 and day 4 led to the elimination of B-ALL malignant cells in PDX mice. FIG.13C demonstrates that NeoTB-ALL showed dose-dependent effects on cancer elimination. FIG.13D shows the persistence of NeoTB-ALL as detected in peripheral blood of recipient mice post- ACT. [0568] FIG.13E shows that NeoTB-ALL displayed activation characteristics with increased 4- 1BB and CD25 expression in recipient mice, indicating cytotoxic activities against B-ALL malignant cells. FIG.13F shows that NeoTB-ALL cytotoxicity in recipient mice was associated with cytokine release syndrome (CRS). Example 4. AML-^APC activate NeoTAML for autologous cell therapy for AML A. Producing AML-^APC from malignant myelogenous leukocytes [0569] PBMC samples from patients suffering different stages of acute myelogenous leukemia (AML) were obtained (Discovery Life Sciences). These samples were analyzed for the presence of malignant myelogenous leukocytes (AML cells), cMO, and T cells (FIGs. 14A-15A). In all samples, malignant myelogenous leukocytes occupied the majority of PBMC cells. [0570] Malignant myelogenous leukocytes were treated by HC-activator and optional APC refinement agent to differentiate into AML-^APC, which were then used to activate NeoT ny-2770598
Attorney Docket No.24516-20006.40 AML. After testing in vitro for the NeoT AML killing of cancer cells, NeoT AML were used to treat AML established in NSG mice (i.e., an AML PDX model) through adoptive cell therapy (ACT). See FIGs.14A-16B. [0571] FIGs.14A-14B show that malignant myeloblasts were either CD45- or CD45+, dependent on the stage of AML. AML patients were associated with reduction of CD8 T cells in PBMC. [0572] FIGs.15A-15B show analyses of cell compositions in PBMC obtained from AML patients and compared to healthy donors. FIG.15A is a table that summarizes representative AML studies. [0573] FIG.15B shows the differentiation of AML malignant myeloblasts into ^APC, termed AML-^APC. PBMC were passed through anti-CD14-coated beads to collect cMo (CD14+), followed by positive selection with anti-CD33-coated beads to enrich AML malignant myeloblasts (CD33+CD14-). Prior to use, bead-absorbed cMo and malignant myeloblasts were released from their respective beads and collected separately. T cells (PBL) in PBMC were collected by positive selection with anti-CD3 beads, and these T cells were used for antigen-specific activation of NeoT. Following cell collection, cMo and malignant myeloblasts were treated with HC-activator (2d) and optional APC refinement agent (1d) to induce differentiation into ^APC. FIG.15B shows malignant myeloblasts following HC- activator/optional APC refinement agent treatment differentiating into phenotypic antigen presenting cells, termed AML-^APC, which displayed increased expression of MHC-I and MHC-II and co-stimulatory molecules CD40, CD80, and CD86. B. AML-^APC activate NeoTAML for ACT to treat AML [0574] Malignant myelogenous leukocytes-derived AML-^APC were used to activate NeoTAML from PBL and/or TILs (in this case: T cells in BM). Expanded NeoTAML were tested in vitro for killing of AML malignant cells and in vivo by adoptive cell therapy (ACT) for treating AML PDXs established in NSG mice. [0575] See FIGs.16A-17C: AML-^APC antigen presentation activated NeoTAML and in vitro testing of NeoTAML showed killing of AML malignant cells. [0576] FIGs.16A-16B illustrates that AML-^APC mediated antigen presentation to activate NeoTAML, leading to NeoTAML clonal expansion. FIG.16A depicts the experimental procedure. AML-^APC were differentiated from malignant myeloblasts by the HC-activator ny-2770598
Attorney Docket No.24516-20006.40 (optionally further with the optional APC refinement reagent) treatment. PBL comprising CD4 and CD8 T cells were added into differentiated AML-^APC cultures. [0577] FIG.16B shows images of NeoT engaging with AML-^APC. Rapid clonal expansion in 72h and increases in CD4 and CD8 T cells are also shown (right panel). [0578] FIGs.17A-17C show the results of an in vitro assay of NeoTAML killing towards AML malignant cells. FIG.17A shows that NeoTAML were incubated with AML malignant myeloblasts at 1:1 ratio for 6 hours, resulting in rapid killing that reduced malignant myeloblasts from 45.0% to 3.41% in the co-culture. [0579] FIG.17B shows NeoTAML killing efficiency measured by time course and varied ratios of T cells to AML malignant myeloblasts. Parallel experiments were performed using non-specific T cells (control). [0580] FIG.17C shows that NeoTAML targeting malignant myeloblasts were associated with increased expression of CD8 T cell activation markers: IFN^, CD107a, 4-1BB, and CD25. The control non-specific T cells, which exhibited no/minimal killing towards malignant myeloblasts, did not show increased activation markers during co-incubation with malignant myeloblasts. C. NeoTAML adoptive cell therapy (ACT) treating AML [0581] See FIGs.18A-19E: AML PDX models were established and tested in vivo for NeoTAML killing of AML following ACT. [0582] FIGs.18A-18C established AML patient-derived xenograft (PDX) models in immunocompromised NSG mice. FIG.18A illustrates the experimental steps for establishing AML PDXs for the subsequent testing of NeoTAML by ACT. Malignant myeloblasts (1×106 per mouse) from AML patients were implanted into NSG mice via i.v. injection. Once AML cells were detectable in the PBMC of xenografted mice (F1), mice were euthanized and BM single cells and PBMC were harvested, combined, and engrafted (i.v., 5×106 per mouse) into the next generation recipient mice (2nd generation, F2). Following the same procedure, the 3rd generation of a cohort of at least 10 mice were generated for testing NeoTAML by ACT, as well as other treatments. [0583] FIG.18B showed the detection of AML malignant myeloblasts in PBMC, bone marrow, spleen, and extramedullary tissues of xenografted mice. FIG.18C showed that the F2 generation of AML PDXs displayed extramedullary disease. ny-2770598
Attorney Docket No.24516-20006.40 [0584] FIGs.19A-19E illustrated that NeoTAML adoptive cell therapy (ACT) effectively treated AML patient-derived xenografts (PDXs). FIG.19A shows the experimental design: AML PDXs of the 3rd (F3) generation were generated by i.v. engraftment of malignant myeloblasts (5×106 per mouse) harvested from the 2nd generation (F2) PDX mice. AML diseases were generally established in F3 recipients in 2-3 weeks, manifesting malignancies in BM (> 30% cells of BM being malignant myeloblasts) and then in PBMC. BM and/or PBMC were sampled to confirm AML establishment prior to ACT with NeoTAML. Two cohorts were established: cohort #1 with ACT started on day 14 when BM manifested AML; cohort #2 with ACT started on day 26 when AML from BM spread to PBMC. Two rounds of ACT treatments were given with either NeoTAML or non-specific T cells (ctl.). FIG.19B shows overall survival of PDX mice without or with treatment of NeoTAML. FIG.19C shows results of cohort #1 wherein mice manifested BM disease (d14 and d18) and were treated with NeoTAML in a dose-escalation manner. FIG. 19D shows the results of cohort #2 wherein mice with late-stage AML manifesting in both BM and PBMC (d26) were treated with NeoTAML. FIG.19E shows sample PBMC analyses in cohort #2 prior to and after NeoTAML treatment. AML manifestation in PBMC was diminished after the 2nd round of NeoTAML ACT, and NeoTAML cells displayed persistence in the circulation after AML elimination. D. Prophylactic administration with anti-TNF^ neutralization ameliorates CRS without compromising NeoTAML ACT efficacy [0585] Anti-TNF^ neutralization did not interfere with antigen presentation or NeoT activation and expansion, nor did it affect NeoT killing of targeted cancer cells. Prophylactic administration of anti-TNF^ neutralization did not reduce the potency of NeoT once encountering targeted cancer cells, nor did it compromise NeoT therapeutic efficacy. Anti- TNF^ neutralization ameliorated systemic proinflammatory response and CRS subsequent to NeoT activation (see FIGs.20A-20E). [0586] FIGs.20A-20E show that prophylactic anti-TNF^ neutralizing antibody administration ameliorated CRS without compromising NeoTAML ACT efficacy against AML. FIG.20A-20B shows that prophylactic anti-TNF^ neutralizing antibody administration did not compromise NeoTAML ACT efficacy towards AML. AML PDXs (F3) with PBMC manifestation were treated with 5×106 NeoTAML without or with prophylactic administration (3 hours earlier) of anti-TNF^ neutralizing Ab. FIG.20C shows that prophylactic anti-TNF^ neutralizing antibody administration did not inhibit NeoTAML activation and production of IFN^ associated with targeting malignant myeloblasts in vivo by ny-2770598
Attorney Docket No.24516-20006.40 ACT. NeoTAML producing human cytokines were detected in the plasma of recipient PDX mice 24 hours post NeoTAML ACT. Meanwhile, measuring murine cytokines revealed that anti-TNF^ substantially reduced proinflammatory cytokines IL-6, CXCL1, CCL2, CXCL10, and other cytokines that cause CRS symptoms. FIGs.20D-20E show that anti-TNF^ neutralization did not damage AML-^APC-mediated antigen presentation that activated NeoTAML (FIG.20D), nor did it affect NeoTAML killing of AML malignant cells (FIG.20E). ny-2770598
Claims
Attorney Docket No.24516-20006.40 CLAIMS 1. A method of producing a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), comprising: contacting hematological cancer cells obtained from an individual having the hematological cancer with one or more survival, differentiation and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an interferon ^ (IFN^) receptor (IFNGR) activator, and 4) an interleukin-4 receptor (IL-4R) activator, thereby producing the population of HC-APCs. 2. A method of producing activated immune cells, comprising: a) producing a population of HC-APCs according to the method of claim 1, and b) contacting the HC-APCs with immune cells, thereby producing activated immune cells. 3. A method of producing activated immune cells, comprising contacting immune cells with a population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APC”), thereby generating activated immune cells; wherein the HC-APCs are derived from hematological cancer cells after having been contacted with one or more survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an interferon ^ (IFN^) receptor (IFNGR) activator, and 4) an interleukin-4 receptor (IL-4R) activator. 4. The method of any one of claims 1-3, wherein the STAT3 activator is selected from the group consisting of: an IL-10, an IL-10 family member, an IL-10R agonist antibody, an IL-10 family cytokine receptor agonist antibody, a small molecule activator of IL-10R, and a small molecule activator of STAT3; optionally wherein the STAT3 activator is selected from the group consisting of: an IL-10 family cytokine, an IL-12 family cytokine, an IL-6 family cytokine, a small molecule STAT3 activator, and G-CSF. 5. The method of claim 4, wherein the STAT3 activator is selected from the group consisting of: IL-10, IL-22, IL-19, IL20, IL-24, IL-12, IL-23, IL-6, colivelin TFA, Garcinone ny-2770598
Attorney Docket No.24516-20006.40 D, G-CSF, IL-7, IL-9, IL-15, and IL-21; optionally wherein the STAT3 activator is selected from the group consisting of: IL-10, IL-22, IL-19, IL-20, IL-24, IL-12, IL-23, Colivelin TFA, and Garcinone D. 6. The method of any one of claims 1-5, wherein the one or more of S/D/M factors comprise an interleukin-4 receptor (IL-4R) activator; optionally wherein the IL-4R activator is selected from the group consisting of IL-4, an IL-4R agonist antibody, and a small molecule activator of IL-4R. The method of claim 6, wherein the IL-4R activator is IL-4. 8. The method of any one of claims 1-7, wherein the one or more of S/D/M factors comprise a TNFR activator; optionally wherein the TNFR activator is selected from the group consisting of TNF^, a TNFR agonist antibody, and a small molecule activator of TNFR. 9. The method of claim 8, wherein the TNFR activator is TNF^. 10. The method of any one of claims 1-9, wherein the one or more of S/D/M factors comprise an IFNGR activator; optionally wherein the IFNGR activator is selected from the group consisting of IFN^, an IFNGR agonist antibody, and a small molecule activator of IFNGR. 11. The method of claim 10, wherein the IFNGR activator is IFN^. 12. The method of any one of claims 1-11, wherein the one or more of S/D/M factors are present in a single composition. 13. The method of any one of claims 1-12, wherein the one or more of S/D/M factors comprise two or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator. ny-2770598
Attorney Docket No.24516-20006.40 14. The method of any one of claims 1-13, wherein the one or more of S/D/M factors comprise three or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator. 15. The method of claim 14, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator, 2) a TNFR activator, and 3) an IFNGR activator; optionally wherein the one or more of S/D/M factors comprise an IL-10 family cytokine, TNF^, and IFN^. 16. The method of claim 15, wherein the one or more of S/D/M factors comprise: 1) a STAT3 activator, 2) a TNFR activator, 3) an IFNGR activator, and 4) an IL-4R activator; optionally wherein the one or more of S/D/M factors comprise an IL-10 family cytokine, TNF^, IL-4, and IFN^. 17. The method of any one of claims 1-16, wherein the one or more of S/D/M factors further comprises an IL-6 receptor (IL-6R) activator and/or a GM-CSF receptor (GM-CSFR) activator; optionally wherein the IL-6R activator is selected from the group consisting of IL- 6, an IL-6R agonist antibody, and a small molecule activator of IL-6R; and optionally wherein the GM-CSFR activator is selected from the group consisting of GM-CSF, a GM- CSFR agonist antibody, and a small molecule activator of GM-CSFR. 18. The method of claim 17, wherein the IL-6R activator is IL-6, and wherein the GM- CSFR activator is GM-CSF. 19. The method of any one of claims 1-18, wherein the hematological cancer cells are comprised in a cell mixture comprising monocytes from the individual. 20. The method of any one of claims 1-19, wherein prior to contacting the HC-APCs with immune cells, the method further comprises contacting the HC-APCs with one or more refinement factors selected from the group consisting of type-I interferon, IFN^, TNF^, a TLR ligand, CD40L or a CD40-ligating antibody, an anti-PD-L1 antibody, and TPI-1; optionally wherein the type-I interferon comprises IFN^ and/or IFN^; optionally wherein the TLR ligand is selected from the group consisting of R848, poly IC, CpG, or LPS; and further optionally wherein the TLR ligand comprises R848 and poly IC. ny-2770598
Attorney Docket No.24516-20006.40 21. The method of claim 18, wherein the one or more refinement factors comprise IFN^, IFN^, and TNF^. 22. The method of claim 19, wherein the one or more refinement factors further comprise at least two agents selected from the group consisting of poly IC, CpG, CD40L, R848, and an anti-PD-L1 antibody; optionally wherein the one or more refinement factors further comprises a SHP-1 inhibitor; further optionally wherein the SHP-1 inhibitor is TPI-1. 23. The method of any one of claims 2-22, wherein the method further comprises administering the activated immune cells into the individual. 24. The method of any one of claims 2-22, wherein the method further comprises administering the activated immune cells into a different individual than the individual having the hematological cancer. 25. The method of any one of claims 2-24, wherein the immune cells and the hematological cancer cells are from the same individual. 26. The method of any one of claims 2-24, wherein the immune cells and the hematological cancer cells are from different individuals. The method of any one of claims 23-26, wherein the immune cells are administered to the individual from whom they are obtained. 28. The method of any one of claims 23-26, wherein the immune cells are administered to a different individual than the individual from whom they are obtained. 29. The method of any one of claims 2-28, wherein the immune cells are selected from the group consisting of peripheral blood mononuclear cells (“PBMCs”), cells derived from bone marrow biopsy, cells derived from lymphoma biopsy, tumor infiltrating T cells (TILs), and T cells; optionally wherein the immune cells are T cells; further optionally wherein the T cells are CD8 T cells and/or CD4 T cells. ny-2770598
Attorney Docket No.24516-20006.40 30. The method of any one of claims 1-29, wherein the hematological cancer cells are obtained from PBMCs, enriched leukemia cells, a bone marrow biopsy, or a lymphoma biopsy. 31. The method of any one of claims 1-30, wherein the hematological cancer cells are cultured with the one or more of S/D/M factors for at least 2 days. 32. The method of any one of claims 20-31, wherein the HC-APCs are cultured with the refinement factors for about 1-4 days. 33. The method of any one of claims 2-32, wherein the immune cells have been enriched prior to contacting the HC-APCs. 34. The method of any one of claims 1-33, wherein the hematological cancer cells have been enriched prior to being cultured with the one or more of S/D/M factors. 35. The method of any one of claims 1-34, wherein the hematological cancer is a myeloid leukemia; optionally the hematological cancer is acute myeloid leukemia (AML) or chronic myeloid leukemia (CML); further optionally wherein the hematological cancer cells are any of M0-M5 myeloblasts. 36. The method of claim 35, wherein the hematological cancer cells have been enriched prior to being cultured with the one or more of S/D/M factors in the presence of an anti- CD11b antibody. 37. The method of any one of claims 1-34, wherein the hematological cancer is a B-cell lymphoma or B-cell leukemia. 38. The method of claim 37, wherein the B-cell lymphoma or B-cell leukemia is selected from the group consisting of B-cell acute lymphoblastic leukemia B-ALL), chronic lymphocytic leukemia (CLL), and non-Hodgkin’s lymphoma (NHL). ny-2770598
Attorney Docket No.24516-20006.40 39. The method of claim 37 or claim 38, wherein the hematological cancer cells have been enriched prior to being cultured with the one or more of S/D/M factors in the presence of an anti-CD19 antibody or an anti-CD20 antibody. 40. The method of any one of claims 2-39, wherein the method further comprises expanding the immune cells by contacting the immune cells with the HC-APCs for at least two rounds, at least three rounds, or at least four rounds. 41. A population of HC-APCs obtained by the method of any one of claims 1, 4-22, and 30-39. 42. A population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APCs”), wherein the HC-APCs are derived from hematological cancer cells, and wherein the HC-APCs express increased levels of MHC-1, MHC-II, CD40, CD80, and/or CD86 compared to the hematological cancer cells prior to the treatment; optionally wherein the HC-APCs comprise one or more exogenous antigens. 43. The population of HC-APCs of claim 42, wherein the hematological cancer cells are primary cells from an individual having the hematological cancer. 44. The population of HC-APCs of claim 42 or claim 43, wherein the hematological cancer is a myeloid leukemia; optionally the hematological cancer is acute myeloid leukemia (AML) or chronic myeloid leukemia (CML); further optionally wherein the hematological cancer cells are any of M0-M5 myeloblasts. 45. The population of HC-APCs of claim 42 or claim 43, wherein the hematological cancer is a B-cell lymphoma or B-cell leukemia; optionally wherein the B-cell lymphoma or B-cell leukemia is selected from the group consisting of B-cell acute lymphoblastic leukemia B-ALL), chronic lymphocytic leukemia (CLL), and non-Hodgkin’s lymphoma (NHL). 46. A population of antigen presenting cells presenting an antigen associated with a hematological cancer (“HC-APCs”), prepared by: ny-2770598
Attorney Docket No.24516-20006.40 a) obtaining hematological cancer cells from an individual having the hematological cancer, and b) contacting the hematological cancer cells with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an interferon ^ (IFN^) receptor (IFNGR) activator, and 4) an interleukin-4 receptor (IL-4R) activator, thereby producing the population of HC-APCs. 47. A population of activated immune cells obtained by the method of any one of claims 2-40. 48. A method of treating a hematological cancer in an individual, comprising administering to the individual an effective amount of activated immune cells of claim 47. 49. A method of treating a hematological cancer in an individual, comprising: a) contacting hematological cancer cells and/or monocytes with one or more of survival, differentiation, and/or maturation factors (“S/D/M factors”) comprising one or more agents selected from the group consisting of: 1) a STAT3 activator, 2) a TNF^ receptor (TNFR) activator, 3) an interferon ^ (IFN^) receptor (IFNGR) activator, and 4) an interleukin-4 receptor (IL-4R) activator, thereby producing a population of antigen presenting cells (“APCs”), optionally wherein the APCs are further loaded with one or more exogenous antigens, b) contacting the APCs with a population of immune cells isolated from the individual, thereby producing activated immune cells, and c) administering the activated immune cells into the individual. 50. The method of claim 49, wherein the hematological cancer cells and/or monocytes are obtained from the individual; optionally wherein the hematological cancer cells and monocytes are comprised in a mixture when cultured with the one or more of S/D/M factors. ny-2770598
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363519974P | 2023-08-16 | 2023-08-16 | |
| US63/519,974 | 2023-08-16 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025038745A1 true WO2025038745A1 (en) | 2025-02-20 |
Family
ID=92632960
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2024/042306 Pending WO2025038745A1 (en) | 2023-08-16 | 2024-08-14 | Compositions and methods for activating immune cells |
Country Status (3)
| Country | Link |
|---|---|
| AR (1) | AR133555A1 (en) |
| TW (1) | TW202513587A (en) |
| WO (1) | WO2025038745A1 (en) |
Citations (18)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO1991003553A1 (en) | 1989-09-05 | 1991-03-21 | Immunex Corporation | TUMOR NECROSIS FACTOR-α AND -β RECEPTORS |
| US5223395A (en) | 1988-12-01 | 1993-06-29 | Centocor, Inc. | Immunometric assays for tumor necrosis factor-alpha and methods for preventing the loss of biological activity of tumor necrosis factor-alpha in biological samples |
| EP0610201A1 (en) | 1991-03-18 | 1994-08-17 | New York University | Monoclonal and chimeric antibodies specific for human tumor necrosis factor |
| EP0767793A1 (en) | 1994-06-22 | 1997-04-16 | Macronex, Inc. | N-substituted-(dihydroxyboryl)alkyl purine, indole and pyrimidine derivatives, useful as inhibitors of inflammatory cytokines |
| US5656272A (en) | 1991-03-18 | 1997-08-12 | New York University Medical Center | Methods of treating TNF-α-mediated Crohn's disease using chimeric anti-TNF antibodies |
| US5698195A (en) | 1991-03-18 | 1997-12-16 | New York University Medical Center | Methods of treating rheumatoid arthritis using chimeric anti-TNF antibodies |
| US5712155A (en) | 1989-09-05 | 1998-01-27 | Immunex Corporation | DNA encoding tumor necrosis factor-α and -β receptors |
| US5919452A (en) | 1991-03-18 | 1999-07-06 | New York University | Methods of treating TNFα-mediated disease using chimeric anti-TNF antibodies |
| US6090382A (en) | 1996-02-09 | 2000-07-18 | Basf Aktiengesellschaft | Human antibodies that bind human TNFα |
| WO2000044880A1 (en) * | 1999-01-29 | 2000-08-03 | Institut National De La Sante Et De La Recherche Medicale (I.N.S.E.R.M.) | Means for inducing or increasing the immunogenicity of myeloid acute leukemia cells |
| US6258562B1 (en) | 1996-02-09 | 2001-07-10 | Basf Aktiengesellschaft | Human antibodies that bind human TNFα |
| US6277969B1 (en) | 1991-03-18 | 2001-08-21 | New York University | Anti-TNF antibodies and peptides of human tumor necrosis factor |
| US6284471B1 (en) | 1991-03-18 | 2001-09-04 | New York University Medical Center | Anti-TNFa antibodies and assays employing anti-TNFa antibodies |
| WO2002012502A2 (en) | 2000-08-07 | 2002-02-14 | Centocor, Inc. | Anti-tnf antibodies, compositions, methods and uses |
| US20160045548A1 (en) * | 2012-07-05 | 2016-02-18 | Fullhope Biomedical Co., Ltd | Method for treating cancers with dendritic killer cells and pharmaceutical composition comprising the same |
| WO2016036319A1 (en) * | 2014-09-04 | 2016-03-10 | Agency For Science, Technology And Research | A method to up-regulate cancer stem cell markers for the generation of antigen specific cytotoxic effector t cells |
| WO2019183924A1 (en) * | 2018-03-30 | 2019-10-03 | Syz Cell Therapy Co. | Improved multiple antigen specific cell therapy methods |
| WO2019196087A1 (en) * | 2018-04-13 | 2019-10-17 | Syz Cell Therapy Co. | Methods of cancer treatment using tumor antigen-specific t cells |
-
2024
- 2024-08-14 WO PCT/US2024/042306 patent/WO2025038745A1/en active Pending
- 2024-08-15 TW TW113130733A patent/TW202513587A/en unknown
- 2024-08-15 AR ARP240102161A patent/AR133555A1/en unknown
Patent Citations (21)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US5223395A (en) | 1988-12-01 | 1993-06-29 | Centocor, Inc. | Immunometric assays for tumor necrosis factor-alpha and methods for preventing the loss of biological activity of tumor necrosis factor-alpha in biological samples |
| US5712155A (en) | 1989-09-05 | 1998-01-27 | Immunex Corporation | DNA encoding tumor necrosis factor-α and -β receptors |
| WO1991003553A1 (en) | 1989-09-05 | 1991-03-21 | Immunex Corporation | TUMOR NECROSIS FACTOR-α AND -β RECEPTORS |
| US6790444B2 (en) | 1991-03-18 | 2004-09-14 | New York University Medical Center | Anti-TNF antibodies and peptides of human necrosis factor |
| US6835823B2 (en) | 1991-03-18 | 2004-12-28 | New York University | Anti-TNF antibodies and peptides of human tumor necrosis factor |
| US5698195A (en) | 1991-03-18 | 1997-12-16 | New York University Medical Center | Methods of treating rheumatoid arthritis using chimeric anti-TNF antibodies |
| US5656272A (en) | 1991-03-18 | 1997-08-12 | New York University Medical Center | Methods of treating TNF-α-mediated Crohn's disease using chimeric anti-TNF antibodies |
| US5919452A (en) | 1991-03-18 | 1999-07-06 | New York University | Methods of treating TNFα-mediated disease using chimeric anti-TNF antibodies |
| EP0610201A1 (en) | 1991-03-18 | 1994-08-17 | New York University | Monoclonal and chimeric antibodies specific for human tumor necrosis factor |
| US6284471B1 (en) | 1991-03-18 | 2001-09-04 | New York University Medical Center | Anti-TNFa antibodies and assays employing anti-TNFa antibodies |
| US6277969B1 (en) | 1991-03-18 | 2001-08-21 | New York University | Anti-TNF antibodies and peptides of human tumor necrosis factor |
| EP0767793A1 (en) | 1994-06-22 | 1997-04-16 | Macronex, Inc. | N-substituted-(dihydroxyboryl)alkyl purine, indole and pyrimidine derivatives, useful as inhibitors of inflammatory cytokines |
| US6258562B1 (en) | 1996-02-09 | 2001-07-10 | Basf Aktiengesellschaft | Human antibodies that bind human TNFα |
| US6509015B1 (en) | 1996-02-09 | 2003-01-21 | Basf Aktiengesellschaft | Human antibodies that bind human TNFa |
| US6090382A (en) | 1996-02-09 | 2000-07-18 | Basf Aktiengesellschaft | Human antibodies that bind human TNFα |
| WO2000044880A1 (en) * | 1999-01-29 | 2000-08-03 | Institut National De La Sante Et De La Recherche Medicale (I.N.S.E.R.M.) | Means for inducing or increasing the immunogenicity of myeloid acute leukemia cells |
| WO2002012502A2 (en) | 2000-08-07 | 2002-02-14 | Centocor, Inc. | Anti-tnf antibodies, compositions, methods and uses |
| US20160045548A1 (en) * | 2012-07-05 | 2016-02-18 | Fullhope Biomedical Co., Ltd | Method for treating cancers with dendritic killer cells and pharmaceutical composition comprising the same |
| WO2016036319A1 (en) * | 2014-09-04 | 2016-03-10 | Agency For Science, Technology And Research | A method to up-regulate cancer stem cell markers for the generation of antigen specific cytotoxic effector t cells |
| WO2019183924A1 (en) * | 2018-03-30 | 2019-10-03 | Syz Cell Therapy Co. | Improved multiple antigen specific cell therapy methods |
| WO2019196087A1 (en) * | 2018-04-13 | 2019-10-17 | Syz Cell Therapy Co. | Methods of cancer treatment using tumor antigen-specific t cells |
Non-Patent Citations (33)
| Title |
|---|
| AM J HEMATOL., vol. 95, 2020, pages 691 - 709 |
| AM J HEMATOL., vol. 96, no. 12, 1 December 2021 (2021-12-01), pages 1679 - 1705 |
| BIAN Z ET AL: "Next-generation immunotherapy using patient-derived APC", JOURNAL FOR IMMUNOTHERAPY OF CANCER, vol. 10, no. Suppl. 2, 969, 7 November 2022 (2022-11-07), 37th SITC Annual Meeting; Boston, MA, USA; 8-10 November 2022, pages A1011, XP093208730, DOI: 10.1136/jitc-2022-SITC2022.0969 * |
| BIAN Z ET AL: "Next-generation pan-cancer immunotherapy with patient-derived APC", CANCER RESEARCH, vol. 83, no. 7_Suppl., 6438, 4 April 2023 (2023-04-04), AACR Annual Meeting; Orlando, FL, USA; 14-19 April 2023, pages 6438, XP093208726, ISSN: 1538-7445, DOI: 10.1158/1538-7445.AM2023-6438 * |
| BIAN Z ET AL: "Next-generation pan-cancer immunotherapy with patient-derived APC", JOURNAL FOR IMMUNOTHERAPY OF CANCER, vol. 11, no. Suppl. 1, 405, 2 November 2023 (2023-11-02), 38th SITC Annual Meeting; San Diego, CA, USA; 1-5 November 2023, pages A454, XP093208722, ISSN: 2051-1426, DOI: 10.1136/jitc-2023-SITC2023.0405 * |
| CAUSHI ET AL., NATURE, vol. 596, no. 7870, August 2021 (2021-08-01), pages 126 - 132 |
| CHARBONNIER A ET AL: "Human acute myeloblastic leukemia cells differentiatein vitro into mature dendritic cells and induce the differentiation of cytotoxic T cells against autologous leukemias", EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 29, no. 8, August 1999 (1999-08-01), pages 2567 - 2578, XP093210933, ISSN: 0014-2980, DOI: 10.1002/(SICI)1521-4141(199908)29:08<2567::AID-IMMU2567>3.0.CO;2-S * |
| CHEN ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 94, 1997, pages 1914 - 1918 |
| DING ET AL., STEM CELL INVESTIG., vol. 4, 2017, pages 19 |
| HAUBNER ET AL., LEUKEMIA, vol. 33, 2019, pages 64 - 74 |
| HIRANO ET AL., ONCOGENE, vol. 19, 2000, pages 2548 - 2556 |
| J FAMILY MED PRIM CARE., vol. 9, no. 4, April 2020 (2020-04-01), pages 1834 - 1840 |
| JEGO ET AL., CANCERS (BASEL, vol. 12, no. 1, 2020, pages 21 |
| KIM ET AL., ONCOL LETT., vol. 23, no. 3, 2022, pages 94 |
| KUMAR ET AL., MED J ARMED FORCES INDIA, vol. 51, no. 3, 1995, pages 165 - 169 |
| LEE ET AL., BIOSENS. BIOELECTRON., vol. 74, 2015, pages 341 - 346 |
| LIAO ET AL., ANTICANCER RES., vol. 42, no. 8, 2022, pages 3807 - 3814 |
| LIEPE ET AL.: "A large fraction of HLA class I ligands are proteasome-generated spliced peptides", SCIENCE, vol. 354, no. 6310, 21 October 2016 (2016-10-21), pages 354 - 358, XP055925896, DOI: 10.1126/science.aaf4384 |
| LIU ET AL., CELL DEATH DIS., vol. 5, no. 6, 2014, pages 1293 |
| POLLARD ET AL., PROTEOMICS CLIN. APPL., vol. 1, 2007, pages 934 - 952 |
| ROUDKO ET AL., FRONT IMMUNOL., vol. 11, 2020, pages 27 |
| SARAIVA, MO'GARRA, A.: "The regulation of IL-10 production by immune cells", NAT REV IMMUNOL, vol. 10, 2010, pages 170 - 181, XP055962471, DOI: 10.1038/nri2711 |
| SUCIC ET AL., J HEMATOTHER STEM CELL RES., vol. 11, no. 6, 2002, pages 941 - 950 |
| TAN ET AL., DATABASE (OXFORD, 1 January 2020 (2020-01-01) |
| TJALSMA ET AL., PROTEOMICS CLIN. APPL., vol. 2, 2008, pages 167 - 180 |
| VIGNERON, BIOMED REV INT., vol. 2015, 2015, pages 948501 |
| WEN ET AL., NAT COMMUN., vol. 11, no. 1, 9 April 2020 (2020-04-09), pages 1759 |
| WU ET AL., GENOMICS PROTEOMICS BIOINFORMATICS., vol. 16, no. 4, August 2018 (2018-08-01), pages 276 - 282 |
| XIA ET AL., FRONT IMMUNOL., vol. 12, 2021, pages 644637 |
| XIAO ET AL., CELL COMMUN SIGNAL., vol. 18, no. 1, 2020, pages 25 |
| YANG ET AL., BIOSENS. BIOELECTRON., vol. 40, 2013, pages 385 - 392 |
| YANG ET AL., CYTOKINE GROWTH FACTOR REV., vol. 46, 2019, pages 10 - 22 |
| ZHENG ET AL., EXP MOL MED., vol. 50, no. 9, 2018, pages 1 - 14 |
Also Published As
| Publication number | Publication date |
|---|---|
| TW202513587A (en) | 2025-04-01 |
| AR133555A1 (en) | 2025-10-08 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| ES3042082T3 (en) | Methods of making chimeric antigen receptor-expressing cells | |
| KR102809909B1 (en) | Method for isolating T cells having antigen specificity for p53 cancer-specific mutations | |
| CN103635573B (en) | The lymphocytic series, its composition and the production method that comprise gamma delta T cells | |
| KR102441940B1 (en) | Proliferation of Lymphocytes for Active Cellular Immune Therapy Using Cytokine Compositions | |
| EP3752599A1 (en) | Modified pluripotent stem cells and methods of making and use | |
| US20240050570A1 (en) | T Cell Modification | |
| US9657269B2 (en) | Regulatory B cells (tBREGS) and their use | |
| CN117802050A (en) | Modified cell populations and compositions | |
| US20250205337A1 (en) | Compositions and methods for activating immune cells | |
| US20240263136A1 (en) | Methods of preparing an isolated population of dendritic cells and methods of treating cancer using same | |
| US20240293452A1 (en) | Compositions and methods of enhancing anti-tumor response using hybrid neutrophils | |
| Wu et al. | CXCR5-transduced bone marrow-derived dendritic cells traffic to B cell zones of lymph nodes and modify antigen-specific immune responses | |
| EP4136216A1 (en) | Improved t cell manufacturing process | |
| JP2024540102A (en) | How to generate cells | |
| EP3830125A1 (en) | Chimeric antigen receptor therapy t cell expansion kinetics and uses thereof | |
| WO2020112815A1 (en) | Anti-lmp2 tcr-t cell therapy for the treatment of ebv-associated cancers | |
| WO2025038745A1 (en) | Compositions and methods for activating immune cells | |
| CN112972491B (en) | Cell composition for treating or preventing cancer and method for producing same | |
| WO2019243888A9 (en) | Compositions and methods for making engineered t cells | |
| WO2024054863A1 (en) | T-cell receptor that targets egfr mutation and methods of using the same | |
| WO2025122569A1 (en) | T cell receptor specific to kmt2a::aff1 neoantigen and use thereof in adoptive immunotherapy | |
| Stadler | Dissecting ALK-specific CD4 T Cell Responses for ALK-positive Anaplastic Large Cell Lymphoma Immunotherapy | |
| CN115927200A (en) | Dendritic cell vaccine targeting receptor HER2 | |
| WO2024206155A1 (en) | Utilizing t cells derived from tumor draining lymph nodes for chimeric antigen receptor (car) t cell therapy for the treatment of cancer | |
| HK1236418A1 (en) | Expansion of lymphocytes with a cytokine composition for active cellular immunotherapy |
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: 24765283 Country of ref document: EP Kind code of ref document: A1 |