[go: up one dir, main page]
More Web Proxy on the site http://driver.im/

WO2023200929A1 - Tumor-infiltrating lymphocyte (til) compositions and uses thereof - Google Patents

Tumor-infiltrating lymphocyte (til) compositions and uses thereof Download PDF

Info

Publication number
WO2023200929A1
WO2023200929A1 PCT/US2023/018458 US2023018458W WO2023200929A1 WO 2023200929 A1 WO2023200929 A1 WO 2023200929A1 US 2023018458 W US2023018458 W US 2023018458W WO 2023200929 A1 WO2023200929 A1 WO 2023200929A1
Authority
WO
WIPO (PCT)
Prior art keywords
compounds comprise
tils
therapeutic population
mutant
methods
Prior art date
Application number
PCT/US2023/018458
Other languages
French (fr)
Inventor
Jinzhou YUAN
Xingliang ZHOU
Ruben Rodriguez
Eric GSCHWENG
Gray KUEBERUWA
Milena KALAITSIDOU
Original Assignee
Instil Bio, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Instil Bio, Inc. filed Critical Instil Bio, Inc.
Publication of WO2023200929A1 publication Critical patent/WO2023200929A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0638Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4636Immune checkpoint inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4637Other peptides or polypeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/57Skin; melanoma
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen

Definitions

  • TIL TUMOR-INFILTRATING LYMPHOCYTE
  • TILs Tumor infiltrating lymphocytes
  • TILs Tumor infiltrating lymphocytes
  • TILs Prior to excision, TILs in tumor tissue may be already differentiated and reaching exhaustion as a result of chronic tumor antigen stimulation.
  • TILs are massively expanded in vitro, which leads to further differentiation and exhaustion.
  • viability and recovery of a desired product from tissue may be affected by the conditions during tissue collection, disaggregation, and harvesting of cells, and expansion of cells.
  • TIL preparations and methods of manufacture that provide improved functionality.
  • the invention relates to rejuvenation of TILs.
  • Tumor-reactive T cells acquired from tumor microenvironments may already be exhausted or losing their functionality on their way to exhaustion.
  • TILs prepared and selected in vitro for activity such as high levels of IFN-y and efficient targeting of target cells may be less effective at causing regression of targeted tumors when adoptively transferred to a patient than TILs at earlier other stages of differentiation.
  • the invention provides methods and compositions for producing TIL populations exhibiting improved persistence and functionality.
  • the TILs can be considered “rejuvenated,” i.e., demonstrating high levels of in vitro and in vivo activity and less subject to exhaustion.
  • the TILs can be considered “prodigious,” i.e., exhibiting prolonged high levels of in vivo activity when administered to a subject.
  • the invention provides TIL compositions having improved persistence and functionality.
  • the invention provides methods and agents for generating such improved TIL compositions and methods of treatment that employ the improved TIL compositions.
  • T cells are typically divided into discreet subsets based on definitions that reflect their roles in immunity.
  • Features of T cell immune responses include clonal expansion, contraction, exhaustion, and memory formation. Expanded T cell populations are not homogeneous but consist of short lived effector cells and a smaller population of memory precursors.
  • the invention provides improved TIL populations by promoting expansion and persistence and reducing exhaustion of tumor-reactive T cells.
  • TILs tumor infiltrating lymphocytes
  • Some such methods comprise treating a first population of TILs with one or more compounds to improve T-cell fitness.
  • Some such methods comprise treating the first population of TILs with the one or more compounds multiple times.
  • the one or more compounds comprise on or more (any combination thereof) or all of the following: (1) a FAS/FASLG inhibitory agent; (2) a TGF ⁇ /TGF ⁇ R1 inhibitory agent; (3) an IRF7 inhibitory agent; (4) a POLR3A inhibitory agent; (5) an ETV7 inhibitory agent; (6) an ETV3 inhibitory agent; (7) an ASH2L inhibitory agent; (8) a PML inhibitory agent; (9) a STAT2 inhibitory agent; (10) a SPI1 inhibitory agent; (11) an IRF9 inhibitory agent; (12) a STAT1 inhibitory agent; (13) an IRF4 inhibitory agent; (14) a JDP2 inhibitory agent; (15) a ZNF337 inhibitory agent; (16) an ETV2 inhibitory agent; (17) an ETV3L inhibitory agent; (18) a SOX18 inhibitory agent; (19) a CEBPG inhibitory agent; (20) a CREB3L4 inhibitory agent; (21)
  • the one or more compounds comprise on or more or all of the following: (i) a FAS/FASLG inhibitory agent; (ii) a TGF ⁇ /TGF ⁇ R1 inhibitory agent; (iii) an IRF7 inhibitory agent; and (iv) a POLR3A inhibitory agent.
  • the treating decreases expression or activity of FAS or FASLG.
  • the treating transiently decreases expression or activity of FAS or FASLG.
  • the treating permanently decreases expression or activity of FAS or FASLG.
  • the one or more compounds comprise a DNA encoding a dominant negative FAS mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative FAS mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative FAS mutant.
  • the dominant negative FAS mutant comprises a mutated FADD binding site, optionally wherein the dominant negative FAS mutant is FAS_D244V.
  • the dominant negative FAS mutant comprises a deleted DD domain, optionally wherein the dominant negative FAS mutant is FAS_del230-314.
  • the one or more compounds comprise an anti-FAS or anti-FASLG antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to a FAS or FASLG messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding FAS or FASLG.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FAS or FASLG.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule FAS/FASLG inhibitor.
  • the treating decreases expression or activity of TGF ⁇ 1 or TGF ⁇ R1.
  • the treating transiently decreases expression or activity of TGF ⁇ 1 or TGF ⁇ R1.
  • the treating permanently decreases expression or activity of TGF ⁇ 1 or TGF ⁇ R1.
  • the one or more compounds comprise a DNA encoding a dominant negative TGF ⁇ R1 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative TGF ⁇ R1 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative TGF ⁇ R1 mutant.
  • the one or more compounds comprise an anti-TGF ⁇ R1 or TGF ⁇ 1 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to a TGF ⁇ R1 or TGF ⁇ 1 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding TGF ⁇ R1 or TGF ⁇ 1.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding TGF ⁇ R1 or TGF ⁇ 1.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule TGF ⁇ R1 inhibitor, optionally wherein the small molecule 1 inhibitor is SB431542.
  • the treating decreases expression or activity of IRF7.
  • the treating transiently decreases expression or activity of IRF7.
  • the treating permanently decreases expression or activity of IRF7.
  • the one or more compounds comprise a DNA encoding a dominant negative IRF7 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative IRF7 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative IRF7 mutant.
  • the one or more compounds comprise an anti-IRF7 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF7 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF7.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF7.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule IRF7 inhibitor.
  • the treating decreases expression or activity of POLR3A.
  • the treating transiently decreases expression or activity of POLR3A.
  • the treating permanently decreases expression or activity of POLR3A.
  • the one or more compounds comprise a DNA encoding a dominant negative POLR3A mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative POLR3A mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative POLR3A mutant.
  • the one or more compounds comprise an anti-POLR3A antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to a POLR3A messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding POLR3A.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding POLR3A.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule POLR3A inhibitor.
  • the treating decreases expression or activity of ETV7.
  • the treating transiently decreases expression or activity of ETV7.
  • the treating permanently decreases expression or activity of ETV7.
  • the one or more compounds comprise a DNA encoding a dominant negative ETV7 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ETV7 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative ETV7 mutant.
  • the one or more compounds comprise an anti-ETV7 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV7 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV7.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV7.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule ETV7 inhibitor.
  • the treating decreases expression or activity of ETV3.
  • the treating permanently decreases expression or activity of ETV3.
  • the one or more compounds comprise a DNA encoding a dominant negative ETV3 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ETV3 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative ETV3 mutant.
  • the one or more compounds comprise an anti-ETV3 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV3 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV3.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule ETV3 inhibitor. [0021] In some such methods, the treating decreases expression or activity of ASH2L.
  • the treating transiently decreases expression or activity of ASH2L. In some such methods, the treating permanently decreases expression or activity of ASH2L.
  • the one or more compounds comprise a DNA encoding a dominant negative ASH2L mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative ASH2L mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative ASH2L mutant. In some such methods, the one or more compounds comprise an anti-ASH2L antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ASH2L messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ASH2L.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ASH2L.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule ASH2L inhibitor.
  • the treating decreases expression or activity of PML.
  • the treating transiently decreases expression or activity of PML.
  • the treating permanently decreases expression or activity of PML.
  • the one or more compounds comprise a DNA encoding a dominant negative PML mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative PML mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative PML mutant.
  • the one or more compounds comprise an anti-PML antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an PML messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding PML.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding PML.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule PML inhibitor.
  • the treating decreases expression or activity of STAT2.
  • the treating permanently decreases expression or activity of STAT2.
  • the one or more compounds comprise a DNA encoding a dominant negative STAT2 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative STAT2 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative STAT2 mutant.
  • the one or more compounds comprise an anti-STAT2 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an STAT2 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding STAT2.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT2.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule STAT2 inhibitor. [0027] In some such methods, the treating decreases expression or activity of SPI1.
  • the treating transiently decreases expression or activity of SPI1. In some such methods, the treating permanently decreases expression or activity of SPI1.
  • the one or more compounds comprise a DNA encoding a dominant negative SPI1 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative SPI1 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative SPI1 mutant. In some such methods, the one or more compounds comprise an anti-SPI1 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an SPI1 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding SPI1.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SPI1.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule SPI1 inhibitor.
  • the treating decreases expression or activity of IRF9.
  • the one or more compounds comprise a DNA encoding a dominant negative IRF9 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative IRF9 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative IRF9 mutant.
  • the one or more compounds comprise an anti-IRF9 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF9 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF9.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF9.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule IRF9 inhibitor.
  • the treating decreases expression or activity of STAT1.
  • the treating transiently decreases expression or activity of STAT1.
  • the treating permanently decreases expression or activity of STAT1.
  • the one or more compounds comprise a DNA encoding a dominant negative STAT1 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative STAT1 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative STAT1 mutant.
  • the one or more compounds comprise an anti-STAT1 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an STAT1 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding STAT1.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT1.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule STAT1 inhibitor. [0033] In some such methods, the treating decreases expression or activity of IRF4.
  • the treating transiently decreases expression or activity of IRF4. In some such methods, the treating permanently decreases expression or activity of IRF4.
  • the one or more compounds comprise a DNA encoding a dominant negative IRF4 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative IRF4 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative IRF4 mutant. In some such methods, the one or more compounds comprise an anti-IRF4 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF4 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF4.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF4.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule IRF4 inhibitor.
  • the treating decreases expression or activity of JDP2.
  • the one or more compounds comprise a DNA encoding a dominant negative JDP2 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative JDP2 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative JDP2 mutant.
  • the one or more compounds comprise an anti-JDP2 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an JDP2 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding JDP2.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding JDP2.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule JDP2 inhibitor.
  • the treating decreases expression or activity of ZNF337.
  • the treating transiently decreases expression or activity of ZNF337.
  • the treating permanently decreases expression or activity of ZNF337.
  • the one or more compounds comprise a DNA encoding a dominant negative ZNF337 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ZNF337 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative ZNF337 mutant.
  • the one or more compounds comprise an anti-ZNF337 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ZNF337 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ZNF337.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF337.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule ZNF337 inhibitor. [0039] In some such methods, the treating decreases expression or activity of ETV2.
  • the treating transiently decreases expression or activity of ETV2. In some such methods, the treating permanently decreases expression or activity of ETV2.
  • the one or more compounds comprise a DNA encoding a dominant negative ETV2 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative ETV2 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative ETV2 mutant. In some such methods, the one or more compounds comprise an anti-ETV2 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV2 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV2.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV2.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule ETV2 inhibitor.
  • the treating decreases expression or activity of ETV3L.
  • the treating transiently decreases expression or activity of ETV3L.
  • the treating permanently decreases expression or activity of ETV3L.
  • the one or more compounds comprise a DNA encoding a dominant negative ETV3L mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ETV3L mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative ETV3L mutant.
  • the one or more compounds comprise an anti-ETV3L antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV3L messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV3L.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3L.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule ETV3L inhibitor.
  • the treating decreases expression or activity of SOX18.
  • the treating transiently decreases expression or activity of SOX18.
  • the treating permanently decreases expression or activity of SOX18.
  • the one or more compounds comprise a DNA encoding a dominant negative SOX18 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative SOX18 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative SOX18 mutant.
  • the one or more compounds comprise an anti-SOX18 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an SOX18 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding SOX18.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SOX18.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule SOX18 inhibitor. [0045] In some such methods, the treating decreases expression or activity of CEBPG.
  • the treating transiently decreases expression or activity of CEBPG. In some such methods, the treating permanently decreases expression or activity of CEBPG.
  • the one or more compounds comprise a DNA encoding a dominant negative CEBPG mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative CEBPG mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative CEBPG mutant. In some such methods, the one or more compounds comprise an anti-CEBPG antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an CEBPG messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CEBPG.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPG.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule CEBPG inhibitor.
  • the treating decreases expression or activity of CREB3L4.
  • the treating transiently decreases expression or activity of CREB3L4.
  • the treating permanently decreases expression or activity of CREB3L4.
  • the one or more compounds comprise a DNA encoding a dominant negative CREB3L4 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative CREB3L4 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative CREB3L4 mutant. In some such methods, the one or more compounds comprise an anti-CREB3L4 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an CREB3L4 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CREB3L4.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CREB3L4.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule CREB3L4 inhibitor.
  • the treating decreases expression or activity of CEBPB.
  • the treating transiently decreases expression or activity of CEBPB.
  • the treating permanently decreases expression or activity of CEBPB.
  • the one or more compounds comprise a DNA encoding a dominant negative CEBPB mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative CEBPB mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative CEBPB mutant.
  • the one or more compounds comprise an anti-CEBPB antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an CEBPB messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CEBPB.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPB.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule CEBPB inhibitor. [0051] In some such methods, the treating decreases expression or activity of FOXD1.
  • the treating transiently decreases expression or activity of FOXD1. In some such methods, the treating permanently decreases expression or activity of FOXD1.
  • the one or more compounds comprise a DNA encoding a dominant negative FOXD1 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative FOXD1 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative FOXD1 mutant. In some such methods, the one or more compounds comprise an anti-FOXD1 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an FOXD1 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding FOXD1.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FOXD1.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule FOXD1 inhibitor.
  • the treating decreases expression or activity of EOMES.
  • the treating transiently decreases expression or activity of EOMES.
  • the treating permanently decreases expression or activity of EOMES.
  • the one or more compounds comprise a DNA encoding a dominant negative EOMES mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative EOMES mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative EOMES mutant.
  • the one or more compounds comprise an anti-EOMES antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an EOMES messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding EOMES.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding EOMES.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule EOMES inhibitor.
  • the treating decreases expression or activity of ZNF683.
  • the treating permanently decreases expression or activity of ZNF683.
  • the one or more compounds comprise a DNA encoding a dominant negative ZNF683 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ZNF683 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more compounds comprise a messenger RNA encoding a dominant negative ZNF683 mutant.
  • the one or more compounds comprise an anti-ZNF683 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ZNF683 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ZNF683.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF683.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more compounds comprise a small molecule ZNF683 inhibitor.
  • the TILs originate from a subject.
  • the TILs are from a tumor biopsy, a lymph node, or ascites.
  • the tumor is from a bladder cancer, a breast cancer, a cancer caused by human papilloma virus, a cervical cancer, a head and neck cancer, a lung cancer, a melanoma, an ovarian cancer, a non-small-cell lung cancer (NSCLC), a renal cancer, or a renal cell carcinoma.
  • the tumor biopsy is from a melanoma.
  • the method further comprises: (i) obtaining a refined tumor product by cryopreserving a resected tumor and disaggregating the cryopreserved tumor, disaggregating a resected tumor and cryopreserving the disaggregated tumor, cryopreserving a resected tumor and processing the tumor into multiple tumor fragments, or processing a resected tumor into multiple tumor fragments and cryopreserving the tumor fragments; and (ii) performing a first expansion by culturing the refined resected tumor product in a cell culture medium comprising IL-2 to produce the first population of TILs, optionally wherein the first population of TILs is treated with the one or more compounds during or subsequent to the first expansion.
  • the cryopreserving comprises: (1) cooling under conditions whereby heat release to, into, around or in an environment including cells, as media crystalizes, is minimized or avoided; (2) continuous cooling, from disaggregation temperature to about - 80°C; (3) continuous cooling at a rate of about -2°C / min; (4) continuous cooling, from disaggregation temperature to about -80°C, at a rate of about -2°C / min; or (5) continuous cooling, from disaggregation temperature to about -80°C, or from disaggregation temperature to -80°C at a rate of about -2°C / min, wherein disaggregation temperature comprises a normal body temperature for an animal from which the tumor was resected, or room temperature or 20°C or 25°C , or normal human body temperature approximately 35°C or 36°C or 36.1°C to approximately 37°C or 37.1°C or 37.2°C or 37.3°C or below about 38.3°C.
  • the disaggregating comprises physical disaggregation, enzymatic disaggregation, or physical and enzymatic disaggregation.
  • a single cell suspension is obtained from the refined resected tumor product and used in step (ii), or wherein the refined resected tumor product from step (i) comprises a single cell suspension.
  • the first expansion in step (ii) is performed for about two weeks.
  • the culturing in step (ii) includes adding IL-7, IL-12, IL-15, IL- 18, IL-21, or a combination thereof.
  • the method further comprises: (iii) performing a second expansion by culturing the first population of TILs in a cell culture medium comprising IL-2, optionally wherein the first population of TILs is treated with the one or more compounds prior to, during, or subsequent to the second expansion.
  • the expanding in step (iii) comprises culturing the first population of TILs with IL-2, OKT-3, and antigen presenting cells (APCs).
  • the expanding in step (iii) is performed for about two weeks.
  • the culturing in step (iii) includes adding IL-7, IL-12, IL-15, IL- 18, IL-21, or a combination thereof.
  • the method further comprises harvesting and/or cryopreserving the therapeutic population of TILs.
  • isolated therapeutic populations of TILs obtained by or obtainable by any of the above methods.
  • isolated therapeutic populations of TILs comprising one or more (any combination thereof) exogenous compounds to improve T-cell fitness.
  • the one or more exogenous compounds comprise one or more or all of the following: (1) a FAS/FASLG inhibitory agent; (2) a TGF ⁇ /TGF ⁇ R1 inhibitory agent; (3) an IRF7 inhibitory agent; (4) a POLR3A inhibitory agent; (5) an ETV7 inhibitory agent; (6) an ETV3 inhibitory agent; (7) an ASH2L inhibitory agent; (8) a PML inhibitory agent; (9) a STAT2 inhibitory agent; (10) a SPI1 inhibitory agent; (11) an IRF9 inhibitory agent; (12) a STAT1 inhibitory agent; (13) an IRF4 inhibitory agent; (14) a JDP2 inhibitory agent; (15) a ZNF337 inhibitory agent; (16) an ETV2 inhibitory agent; (17) an ETV3L inhibitory agent; (18) a SOX18 inhibitory agent; (19) a CEBPG inhibitory agent; (20) a CREB3L4 inhibitory agent; (21) a CEBPB inhibitor
  • isolated therapeutic populations of TILs comprising one or more exogenous compounds to improve T-cell fitness.
  • the one or more exogenous compounds comprise one or more or all of the following: (i) a FAS/FASLG inhibitory agent; (ii) a TGF ⁇ /TGF ⁇ R1 inhibitory agent; (iii) an IRF7 inhibitory agent; and (iv) a POLR3A inhibitory agent.
  • the one or more exogenous compounds decrease expression or activity of FAS or FASLG.
  • the one or more exogenous compounds transiently decrease expression or activity of FAS or FASLG.
  • the one or more exogenous compounds permanently decrease expression or activity of FAS or FASLG.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative FAS mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative FAS mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative FAS mutant.
  • the dominant negative FAS mutant comprises a mutated FADD binding site, optionally wherein the dominant negative FAS mutant is FAS_D244V.
  • the dominant negative FAS mutant comprises a deleted DD domain, optionally wherein the dominant negative FAS mutant is FAS_del230-314.
  • the one or more exogenous compounds comprise an anti-FAS or anti-FASLG antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to a FAS or FASLG messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding FAS or FASLG.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FAS or FASLG.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule FAS/FASLG inhibitor. [0068] In some such populations, the one or more exogenous compounds decrease expression or activity of TGF ⁇ 1 or TGF ⁇ R1.
  • the one or more exogenous compounds transiently decrease expression or activity of TGF ⁇ 1 or TGF ⁇ R1. In some such populations, the one or more exogenous compounds comprise permanently decrease expression or activity of TGF ⁇ 1 or TGF ⁇ R1.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative TGF ⁇ R1 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative TGF ⁇ R1 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative TGF ⁇ R1 mutant.
  • the one or more exogenous compounds comprise an anti-TGF ⁇ R or anti-TGF ⁇ 1 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to a TGF ⁇ 1 or TGF ⁇ R1 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding TGF ⁇ 1 or TGF ⁇ R1.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding TGF ⁇ R.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule TGF ⁇ R1 inhibitor, optionally wherein the small molecule TGF ⁇ R1 inhibitor is SB431542. [0070]
  • the one or more exogenous compounds decrease expression or activity of IRF7.
  • the one or more exogenous compounds transiently decrease expression or activity of IRF7.
  • the one or more exogenous compounds permanently decrease expression or activity of IRF7.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative IRF7 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative IRF7 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative IRF7 mutant.
  • the one or more exogenous compounds comprise an anti-IRF7 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF7 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF7.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF7.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule IRF7 inhibitor.
  • the one or more exogenous compounds decrease expression or activity of POLR3A.
  • the one or more exogenous compounds comprise transiently decrease expression or activity of POLR3A.
  • the one or more exogenous compounds permanently decrease expression or activity of POLR3A.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative POLR3A mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative POLR3A mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative POLR3A mutant.
  • the one or more exogenous compounds comprise an anti-POLR3A antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to a POLR3A messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding POLR3A.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding POLR3A.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule POLR3A inhibitor. [0074] In some such populations, the one or more exogenous compounds decrease expression or activity of ETV7. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ETV7. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ETV7.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative ETV7 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ETV7 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ETV7 mutant.
  • the one or more exogenous compounds comprise an anti-ETV7 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV7 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV7.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV7.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule ETV7 inhibitor.
  • the one or more exogenous compounds decrease expression or activity of ETV3.
  • the one or more exogenous compounds transiently decrease expression or activity of ETV3.
  • the one or more exogenous compounds permanently decrease expression or activity of ETV3.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative ETV3 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ETV3 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ETV3 mutant. In some such populations, the one or more exogenous compounds comprise an anti-ETV3 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV3 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV3.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule ETV3 inhibitor. [0078] In some such populations, the one or more exogenous compounds decrease expression or activity of ASH2L. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ASH2L. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ASH2L.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative ASH2L mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ASH2L mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ASH2L mutant.
  • the one or more exogenous compounds comprise an anti-ASH2L antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ASH2L messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ASH2L.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ASH2L.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule ASH2L inhibitor.
  • the one or more exogenous compounds decrease expression or activity of PML.
  • the one or more exogenous compounds transiently decrease expression or activity of PML.
  • the one or more exogenous compounds permanently decrease expression or activity of PML.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative PML mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative PML mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative PML mutant.
  • the one or more exogenous compounds comprise an anti-PML antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an PML messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding PML.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding PML.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule PML inhibitor. [0082] In some such populations, the one or more exogenous compounds decrease expression or activity of STAT2. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of STAT2. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of STAT2.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative STAT2 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative STAT2 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative STAT2 mutant.
  • the one or more exogenous compounds comprise an anti-STAT2 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an STAT2 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding STAT2.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT2.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule STAT2 inhibitor.
  • the one or more exogenous compounds decrease expression or activity of SPI1.
  • the one or more exogenous compounds transiently decrease expression or activity of SPI1.
  • the one or more exogenous compounds permanently decrease expression or activity of SPI1.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative SPI1 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative SPI1 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative SPI1 mutant. In some such populations, the one or more exogenous compounds comprise an anti-SPI1 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an SPI1 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding SPI1.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SPI1.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule SPI1 inhibitor. [0086] In some such populations, the one or more exogenous compounds decrease expression or activity of IRF9. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of IRF9. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of IRF9.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative IRF9 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative IRF9 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative IRF9 mutant.
  • the one or more exogenous compounds comprise an anti-IRF9 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF9 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF9.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF9.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule IRF9 inhibitor.
  • the one or more exogenous compounds decrease expression or activity of STAT1.
  • the one or more exogenous compounds transiently decrease expression or activity of STAT1.
  • the one or more exogenous compounds permanently decrease expression or activity of STAT1.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative STAT1 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative STAT1 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative STAT1 mutant. In some such populations, the one or more exogenous compounds comprise an anti-STAT1 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an STAT1 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding STAT1.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT1.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule STAT1 inhibitor. [0090] In some such populations, the one or more exogenous compounds decrease expression or activity of IRF4. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of IRF4. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of IRF4.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative IRF4 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative IRF4 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative IRF4 mutant.
  • the one or more exogenous compounds comprise an anti-IRF4 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF4 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF4.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF4.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule IRF4 inhibitor.
  • the one or more exogenous compounds decrease expression or activity of JDP2.
  • the one or more exogenous compounds transiently decrease expression or activity of JDP2.
  • the one or more exogenous compounds permanently decrease expression or activity of JDP2.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative JDP2 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative JDP2 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative JDP2 mutant. In some such populations, the one or more exogenous compounds comprise an anti-JDP2 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an JDP2 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding JDP2.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding JDP2.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule JDP2 inhibitor. [0094] In some such populations, the one or more exogenous compounds decrease expression or activity of ZNF337. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ZNF337. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ZNF337.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative ZNF337 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ZNF337 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ZNF337 mutant.
  • the one or more exogenous compounds comprise an anti-ZNF337 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ZNF337 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ZNF337.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF337.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule ZNF337 inhibitor.
  • the one or more exogenous compounds decrease expression or activity of ETV2.
  • the one or more exogenous compounds transiently decrease expression or activity of ETV2.
  • the one or more exogenous compounds permanently decrease expression or activity of ETV2.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative ETV2 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ETV2 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ETV2 mutant. In some such populations, the one or more exogenous compounds comprise an anti-ETV2 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV2 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV2.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV2.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule ETV2 inhibitor. [0098] In some such populations, the one or more exogenous compounds decrease expression or activity of ETV3L. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ETV3L. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ETV3L.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative ETV3L mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ETV3L mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ETV3L mutant.
  • the one or more exogenous compounds comprise an anti-ETV3L antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV3L messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV3L.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3L.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule ETV3L inhibitor.
  • the one or more exogenous compounds decrease expression or activity of SOX18.
  • the one or more exogenous compounds transiently decrease expression or activity of SOX18.
  • the one or more exogenous compounds permanently decrease expression or activity of SOX18.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative SOX18 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative SOX18 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative SOX18 mutant. In some such populations, the one or more exogenous compounds comprise an anti-SOX18 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an SOX18 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding SOX18.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SOX18.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule SOX18 inhibitor. [00102] In some such populations, the one or more exogenous compounds decrease expression or activity of CEBPG. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of CEBPG. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of CEBPG.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative CEBPG mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative CEBPG mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative CEBPG mutant.
  • the one or more exogenous compounds comprise an anti-CEBPG antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an CEBPG messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CEBPG.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPG.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule CEBPG inhibitor.
  • the one or more exogenous compounds decrease expression or activity of CREB3L4.
  • the one or more exogenous compounds transiently decrease expression or activity of CREB3L4.
  • the one or more exogenous compounds permanently decrease expression or activity of CREB3L4.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative CREB3L4 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative CREB3L4 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative CREB3L4 mutant.
  • the one or more exogenous compounds comprise an anti-CREB3L4 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an CREB3L4 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CREB3L4.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CREB3L4.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule CREB3L4 inhibitor.
  • the one or more exogenous compounds decrease expression or activity of CEBPB.
  • the one or more exogenous compounds transiently decrease expression or activity of CEBPB. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of CEBPB.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative CEBPB mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative CEBPB mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative CEBPB mutant.
  • the one or more exogenous compounds comprise an anti-CEBPB antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an CEBPB messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CEBPB.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPB.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule CEBPB inhibitor. [00108] In some such populations, the one or more exogenous compounds decrease expression or activity of FOXD1. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of FOXD1. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of FOXD1.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative FOXD1 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative FOXD1 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative FOXD1 mutant.
  • the one or more exogenous compounds comprise an anti-FOXD1 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an FOXD1 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding FOXD1.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FOXD1.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule FOXD1 inhibitor.
  • the one or more exogenous compounds decrease expression or activity of EOMES.
  • the one or more exogenous compounds transiently decrease expression or activity of EOMES.
  • the one or more exogenous compounds permanently decrease expression or activity of EOMES.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative EOMES mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative EOMES mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative EOMES mutant. In some such populations, the one or more exogenous compounds comprise an anti-EOMES antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an EOMES messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding EOMES.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding EOMES.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule EOMES inhibitor. [00112] In some such populations, the one or more exogenous compounds decrease expression or activity of ZNF683. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ZNF683. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ZNF683.
  • the one or more exogenous compounds comprise a DNA encoding a dominant negative ZNF683 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ZNF683 mutant is in a viral vector.
  • the viral vector is a lentiviral vector.
  • the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ZNF683 mutant.
  • the one or more exogenous compounds comprise an anti-ZNF683 antigen-binding protein.
  • the antigen-binding protein comprises an antibody.
  • the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ZNF683 messenger RNA.
  • the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
  • the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ZNF683.
  • the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF683.
  • the Cas protein is a Cas9 protein or a Cas12a protein.
  • the one or more exogenous compounds comprise a small molecule ZNF683 inhibitor.
  • the TILs originate from a subject.
  • the TILs are from a tumor biopsy, a lymph node, or ascites.
  • the tumor is from a bladder cancer, a breast cancer, a cancer caused by human papilloma virus, a cervical cancer, a head and neck cancer, a lung cancer, a melanoma, an ovarian cancer, a non-small-cell lung cancer (NSCLC), a renal cancer, or a renal cell carcinoma.
  • the tumor biopsy is from a melanoma.
  • the population comprises about 5x10 9 to about 5x10 10 TILs.
  • pharmaceutical formulations comprising a pharmaceutically acceptable excipient and any of the above isolated therapeutic population of TILs.
  • cryopreserved bags or an intravenous infusion bags, containers, or vessels containing contents comprising any of the above isolated therapeutic population of TILs.
  • methods of treating a cancer in a subject comprising administering any of the above isolated therapeutic population of TILs or the above pharmaceutical formulation to the subject.
  • a cancer in a subject comprising: (a) preparing a therapeutic population of TILs according to any of the above methods; and (b) administering a therapeutic amount of the therapeutic population of TILs to the subject with the cancer.
  • the TILs are autologous or allogeneic.
  • the cancer is a bladder cancer, a breast cancer, a cancer caused by human papilloma virus, a cervical cancer, a head and neck cancer, a head and neck squamous cell carcinoma (HNSCC), a lung cancer, a melanoma, an ovarian cancer, a non-small-cell lung cancer (NSCLC), a renal cancer, or a renal cell carcinoma.
  • the cancer is a melanoma.
  • the subject is a human.
  • the subject is a non-human mammal.
  • the non-human mammal is a primate, a rodent, a rat, a mouse, a domesticated mammal, a domesticated cat, a domesticated dog, a domesticated horse, a guinea pig, a laboratory animal, or a companion animal.
  • the subject is an adult or individual having secondary sexual characteristics.
  • the subject is not an adult or not individual having secondary sexual characteristics, or is a child or is a not physically mature mammal.
  • the administering is performed more than once.
  • the administering is performed more than once over a course of time, wherein: (1) the course of time is a week and the administering is twice, thrice, four times or five times in the week; (2) the course of time is a month and the administering is twice, thrice of four times in a month; (3) the course of time is three, six, nine, or twelve months and the administering is performed once monthly or once weekly.
  • the administering is intravenous administration.
  • FIGS 2A-2F Cell expansion curves of PBMCs under different stimulation schemes. Total viable cell counts of PBMC from two different donors ( Figures 2A, 2B, 2C for D2040, Figures 2D, 2E, 2F for D2740) were measured and plotted over time in “Repeat Stim” group ( Figures 2A, 2D), “R1 Rest group” ( Figures 2B, 2E), and “R2 Rest” group ( Figures 2C, 2F). Comparisons between different stimulation methods are shown in each plot.
  • T cells in “Repeat Stim” group were counted, washed, diluted to 1x10 6 cells/mL, and repeatedly stimulated every 3 days for a total of 10 rounds in 34 days.
  • T cells in “Round 1 Rest” group were counted, washed, and diluted to 1x10 6 cells/mL every 3 days without stimulation.
  • a fraction of T cells was stained and analyzed by flow cytometry for surface tCD34 expression along with markers of activation, exhaustion, and differentiation phenotypes.
  • Figure 5 T cell expansion curves under repeat stimulation. Total viable cell count was measured and plotted over time in LOF candidate groups.
  • pIB1123 is empty vector that served as a control. Candidates that enable enhanced proliferation, as a T cell expansion curve above pIB1123 control curve, were selected as candidate and highlighted below figure legend in each panel. [00128] Figure 6. T cell expansion curves under resting conditions. Total viable cell count was measured and plotted over time in LOF candidate groups. pIB1123 is empty vector that served as a control. Candidates that enable enhanced proliferation, as a T cell expansion curve above pIB1123 control curve, were selected as candidate and highlighted below figure legend in each panel. [00129] Figures 7A-7B. tCD34+ cells were enriched in T cells transduced positive hits under repeat stimulation.
  • FIG. 8A, 8D Surface expression of 4-1BB ( Figures 8A, 8D), PD-1 ( Figures 8B, 8E), and OX40 ( Figures 8C, 8F) were measured on tCD34+CD4+ ( Figures 8A, 8B, 8C) and tCD34+CD8+ ( Figures 8D, 8E, 8F) populations by flow cytometry and plotted over time.
  • Figure 9A-9F T cell exhaustion markers in tCD34+ T cells from positive hit groups under repeat stimulation.
  • FIG. 10A-10B T cell phenotypic subsets in tCD34+ T cells from positive hit groups under repeat stimulation. Surface expression of CCR7 and CD45RA were measured on tCD34+CD4+ ( Figure 10A) and tCD34+CD8+ ( Figure 10B) populations by flow cytometry.
  • FIG. 12A-12B Cell expansion curves of TILs from renal cell carcinoma during Outgrowth (Figure 12A) and REP (Figure 12B). Total viable cell count from groups transduced with different candidates were plotted over time for comparison.
  • Figures 13A-13D Fold expansion of TILs from renal cell carcinoma during Outgrowth ( Figure 13A, 13C) and REP ( Figure 13B, 13D). Total viable cell count from groups transduced with different candidates were plotted over time for comparison.
  • Figures 14A-14C Figures 14A-14C.
  • FIG. 14C Fold change of tCD34+ viable count from day 13 to day 25 was calculated as [(tCD34+ viable count on day 25) – (tCD34+ viable count on day13)] / (tCD34+ viable count on day13) * 100%. Any relative tCD34+% enrichment in groups 3-7 greater than that in group 2 was selected as positive hits.
  • Figures 15A-15B Relative abundance of CD4+ and CD8+ cells in CD3+ populations by the end of Outgrowth (day 13, Figure 15A) and REP (day 25, Figure 15B). Surface expression of CD4 and CD8 were measured by flow cytometry and compared across different groups.
  • Figures 16A-16B Relative abundance of CD4+ and CD8+ cells in CD3+ populations by the end of Outgrowth (day 13, Figure 15A) and REP (day 25, Figure 15B). Surface expression of CD4 and CD8 were measured by flow cytometry and compared across different groups.
  • FIG. 17A-17B TIM-3+% in CD4+ ( Figure 17A) and CD8+ ( Figure 17B) populations by the end of Outgrowth (day 13) and REP (day 25). Surface expression of TIM-3 were measured by flow cytometry and compared across different groups.
  • Figures 18A-18B LAG-3+% in CD4+ ( Figure 18A) and CD8+ ( Figure 18B) populations by the end of Outgrowth (day 13) and REP (day 25).
  • FIG. 19A-19B CD127+% in CD4+ ( Figure 19A) and CD8+ ( Figure 19B) populations by the end of Outgrowth (day 13) and REP (day 25). Surface expression of CD127 were measured by flow cytometry and compared across different groups.
  • Figures 20A-20B CD27+% in CD4+ ( Figure 20A) and CD8+ ( Figure 20B) populations by the end of Outgrowth (day 13) and REP (day 25). Surface expression of CD27 were measured by flow cytometry and compared across different groups.
  • Figures 21A-21D Figures 21A-21D.
  • Surface expression of CCR7 and CD45RA were measured by flow cytometry. Relative abundance of Tn/Tscm (CCR7+CD45RA+), Tcm (CCR7+CD45RA-), Tem (CCR7-CD45RA-), and Temra (CCR7-CD45RA+) populations were compared across different groups.
  • FIG. 22A Unsupervised clustering (Figure 22A) of the gene expression profile of individual cells identified cell subpopulations with distinct transcriptional profiles (Figure 22B) previously undescribed in TIL products. UMAP, uniform manifold approximation and projection.
  • Figures 23A-23C Frequency of C7 (MX1+OAS1+; Figure 23A), C9 (BBC3+CHAC1+; Figure 23B), or C7 and C9 (Figure 23C) T-cell subpopulations in TIL product samples. Low abundance of C7 TIL or C9 TIL subpopulations is associated with response ( Figures 23A and 23B). Low abundance of the combined C7 and C9 TIL subpopulations is associated with response ( Figure 23C). [00146] Figure 24.
  • anti-CD3 antibody refers to an antibody or variant thereof, e.g., a monoclonal antibody and including human, humanized, chimeric, murine or mammalian antibodies which are directed against the CD3 receptor in the T cell antigen receptor of mature human T cells.
  • Anti-CD3 antibodies include OKT-3, also known as muromonab.
  • Anti-CD3 antibodies also include the UHCT1 clone, also known as T3 and CD3.epsilon.
  • Other anti-CD3 antibodies include, for example, otelixizumab, teplizumab, and visilizumab.
  • secondary TILs or genetically modified cytotoxic lymphocytes described herein may be administered at a dosage of 10 4 to 10 11 cells/kg body weight (e.g., 10 5 to 10 6 , 10 5 to 10 10 , 10 5 to 10 11 , 10 6 to 10 10 , 10 6 to 10 11 , 10 7 to 10 11 , 10 7 to 10 10 , 10 8 to 10 11 , 10 8 to 10 10 , 10 9 to 10 11 , or 10 9 to 10 10 cells/kg body weight), including all integer values within those ranges.
  • Tumor infiltrating lymphocytes (including in some cases, genetically modified cytotoxic lymphocytes) compositions may also be administered multiple times at these dosages.
  • the tumor infiltrating lymphocytes can be administered by using infusion techniques that are commonly known in immunotherapy (see, e.g., Rosenberg et al., New Eng. J. of Med.319: 1676, 1988).
  • the optimal dosage and treatment regime for a particular patient can readily be determined by one skilled in the art of medicine by monitoring the patient for signs of disease and adjusting the treatment accordingly.
  • co-administration encompass administration of two or more active pharmaceutical ingredients (in a preferred embodiment of the present invention, for example, at least one potassium channel agonist in combination with a plurality of TILs) to a subject so that both active pharmaceutical ingredients and/or their metabolites are present in the subject at the same time.
  • Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which two or more active pharmaceutical ingredients are present. Simultaneous administration in separate compositions and administration in a composition in which both agents are present are preferred.
  • Cellularized or cellularization refers to the process of disaggregation whereby the solid tissue a multicellular material generally made up of multiple cell lineages/types is broken down into small numbers of cells including but not limited to one cell but could be multiple cells of various lineages or cell types in very small numbers i.e. clump of cells or cell aggregates.
  • Cellularized or cellularization refers to the process of disaggregation whereby the solid tissue a multicellular material generally made up of multiple cell lineages/types is broken down into small numbers of cells including but not limited to one cell but could be multiple cells of various lineages or cell types in very small numbers i.e. clump of cells or cell aggregates.
  • “Closed system” as used herein refers to a system that is closed to the outside environment. Any closed system appropriate for cell culture methods can be employed with the methods of the present invention. Closed systems include, for example, but are not limited to closed G-Rex containers or cell culture bags.
  • cryopreservation media or “cryopreservation medium” as used herein refers to any medium that can be used for cryopreservation of cells. Such media can include media comprising 2% to 10% DMSO. Exemplary media include CryoStor CS10, HypoThermosol, Bloodstor BS- 55 as well as combinations thereof.
  • cryopreserved TILs herein is meant that TILs, either primary, bulk, or expanded (REP TILs), are treated and stored in the range of about -190 °C. to -60 °C. General methods for cryopreservation are also described elsewhere herein, including in the Examples. For clarity, “cryopreserved TILs” are distinguishable from frozen tissue samples which may be used as a source of primary TILs.
  • “Depletion” as used herein refers to a process of a negative selection that separates the desired cells from the undesired cells which are labelled by one marker-binding fragment coupled to a solid phase.
  • Disaggregation or disaggregate refers to the transformation of solid tissue into a single cells or small cell number aggregates where a single cell as a spheroid has a diameter in the range of 5 ⁇ m, 6 ⁇ m, 7 ⁇ m, 8 ⁇ m, 9 ⁇ m, 10 ⁇ m, 20 ⁇ m, 30 ⁇ m, 40 ⁇ m, 50 ⁇ m, 60 ⁇ m, 70 ⁇ m, 80 ⁇ m, 90 ⁇ m, 100 ⁇ m, or more, wherein this is more usually between 7 to 20 ⁇ m.
  • an effective amount refers to that amount of a compound or combination of compounds as described herein that is sufficient to affect the intended application including, but not limited to, disease treatment.
  • a therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated (e.g., the weight, age and gender of the subject), the severity of the disease condition, or the manner of administration.
  • the term also applies to a dose that will induce a particular response in target cells (e.g., the reduction of platelet adhesion and/or cell migration).
  • “Engineered” as used herein refers to either addition of nucleic material or factors, which change the tissue derived cell function from their original function to have a new or improved function for its ultimate utility.
  • “Enzyme media” as used herein refers to media having enzymatic activity such as collagenase, trypsin, lipase, hyaluronidase, deoxyribonuclease, Liberase HI, pepsin, or mixtures thereof.
  • “Filtrate” as used herein refers to the material that passes through a filter, mesh or membrane.
  • “Flexible container” as used herein refers to a flexible packaging system in multiple formats with one or more different types of film. Each film type is selected to provide specific characteristics to preserve the physical, chemical, and functional characteristics of the sterile fluids, solid tissue derived cellular material and the container integrity depending upon the step of the process.
  • “Freezing solution” or “cryopreservation solution” also referred in the field to as the cryoprotectant is a solution that contains cryoprotective additives. These are generally permeable, non-toxic compounds which modify the physical stresses cells are exposed to during freezing in order to minimize freeze damage (i.e.
  • hematological malignancy refers to mammalian cancers and tumors of the hematopoietic and lymphoid tissues, including but not limited to tissues of the blood, bone marrow, lymph nodes, and lymphatic system.
  • Hematological malignancies are also referred to as “liquid tumors.” Hematological malignancies include, but are not limited to, acute lymphoblastic leukemia (ALL), chronic lymphocytic lymphoma (CLL), small lymphocytic lymphoma (SLL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute monocytic leukemia (AMoL), Hodgkin's lymphoma, and non-Hodgkin's lymphomas.
  • ALL acute lymphoblastic leukemia
  • CLL chronic lymphocytic lymphoma
  • SLL small lymphocytic lymphoma
  • AML acute myelogenous leukemia
  • CML chronic myelogenous leukemia
  • AoL acute monocytic leukemia
  • Hodgkin's lymphoma and non-Hodgkin's lymphomas.
  • B cell hematological malignancy refers to hematological
  • IL-2 refers to the T cell growth factor known as interleukin-2, and includes all forms of IL-2 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof. IL-2 is described, e.g., in Nelson, J. Immunol.2004, 172, 3983-88 and Malek, Annu. Rev. Immunol. 2008, 26, 453-79, the disclosures of which are incorporated by reference herein.
  • IL-2 encompasses human, recombinant forms of IL-2 such as aldesleukin (PROLEUKIN, available commercially from multiple suppliers in 22 million IU per single use vials), as well as the form of recombinant IL-2 commercially supplied by CellGenix, Inc., Portsmouth, N.H., USA (CELLGRO GMP) or ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-209-b) and other commercial equivalents from other vendors.
  • aldesleukin PROLEUKIN, available commercially from multiple suppliers in 22 million IU per single use vials
  • CELLGRO GMP CellGenix, Inc.
  • ProSpec-Tany TechnoGene Ltd. East Brunswick, N.J., USA
  • Aldesleukin (des-alanyl-1, serine-125 human IL-2) is a nonglycosylated human recombinant form of IL-2 with a molecular weight of approximately 15 kDa.
  • the term IL-2 also encompasses pegylated forms of IL-2, as described herein, including the pegylated IL2 prodrug NKTR-214, available from Nektar Therapeutics, South San Francisco, Calif., USA.
  • NKTR-214 and pegylated IL-2 suitable for use in the invention is described in U.S. Patent Application Publication No. US 2014/0328791 A1 and International Patent Application Publication No. WO 2012/065086 A1.
  • IL-4 also referred to herein as “IL4” refers to the cytokine known as interleukin 4, which is produced by Th2 T cells and by eosinophils, basophils, and mast cells. IL- 4 regulates the differentiation of naive helper T cells (Th0 cells) to Th2 T cells. Steinke and Borish, Respir. Res.2001, 2, 66-70.
  • Th2 T cells Upon activation by IL-4, Th2 T cells subsequently produce additional IL-4 in a positive feedback loop.
  • IL-4 also stimulates B cell proliferation and class II MHC expression, and induces class switching to IgE and IgG1 expression from B cells.
  • Recombinant human IL-4 suitable for use in the invention is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-211) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-15 recombinant protein, Cat. No. Gibco CTP0043).
  • IL-7 refers to a glycosylated tissue- derived cytokine known as interleukin 7, which may be obtained from stromal and epithelial cells, as well as from dendritic cells. Fry and Mackall, Blood 2002, 99, 3892-904. IL-7 can stimulate the development of T cells. IL-7 binds to the IL-7 receptor, a heterodimer consisting of IL-7 receptor alpha and common gamma chain receptor, which in a series of signals important for T cell development within the thymus and survival within the periphery.
  • IL-7 suitable for use in the invention is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-254) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-15 recombinant protein, Cat. No. Gibco PHC0071).
  • IL-12 also referred to herein as “IL12” refers to the T cell growth factor known as interleukin-12.
  • Interleukin (IL)-12 is a secreted heterodimeric cytokine comprised of 2 disulfide- linked glycosylated protein subunits, designated p35 and p40 for their approximate molecular weights.
  • IL-12 is produced primarily by antigen-presenting cells and drives cell- mediated immunity by binding to a two-chain receptor complex that is expressed on the surface of T cells or natural killer (NK) cells.
  • the IL-12 receptor beta-1 (IL-12Rpi) chain binds to the p40 subunit of IL-12, providing the primary interaction between IL-12 and its receptor. However, it is IL-12p35 ligation of the second receptor chain, IL-12RP2, that confers intracellular signaling.
  • IL-12 signaling concurrent with antigen presentation is thought to invoke T cell differentiation towards the T helper 1 (Thl) phenotype, characterized by interferon gamma (IFNy) production.
  • Thl cells are believed to promote immunity to some intracellular pathogens, generate complement-fixing antibody isotypes, and contribute to tumor immunosurveillance.
  • IFNy interferon gamma
  • IL-12 is thought to be a significant component to host defense immune mechanisms.
  • IL-12 is part of the IL-12 family of cytokines which also includes IL-23, IL-27, IL-35, IL-39.
  • IL-15 refers to the T cell growth factor known as interleukin-15, and includes all forms of IL-15 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof.
  • IL- 15 is described, e.g., in Fehniger and Caligiuri, Blood 2001, 97, 14-32, the disclosure of which is incorporated by reference herein.
  • IL-15 shares ⁇ and ⁇ signaling receptor subunits with IL-2.
  • Recombinant human IL-15 is a single, non-glycosylated polypeptide chain containing 114 amino acids (and an N-terminal methionine) with a molecular mass of 12.8 kDa.
  • Recombinant human IL-15 is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-230-b) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-15 recombinant protein, Cat. No.34-8159-82).
  • the term “IL-18” (also referred to herein as “IL18”) refers to the T cell growth factor known as interleukin-15.
  • Interleukin-18 is a proinflammatory cytokine that belongs to the IL-1 cytokine family, due to its structure, receptor family and signal transduction pathways. Related cytokines include IL-36, IL-37, IL-38.
  • IL-21 refers to the pleiotropic cytokine protein known as interleukin-21, and includes all forms of IL-21 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof. IL-21 is described, e.g., in Spolski and Leonard, Nat. Rev. Drug.
  • IL-21 is primarily produced by natural killer T cells and activated human CD4 + T cells.
  • Recombinant human IL-21 is a single, non-glycosylated polypeptide chain containing 132 amino acids with a molecular mass of 15.4 kDa.
  • Recombinant human IL-21 is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-408-b) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-21 recombinant protein, Cat. No.14-8219-80).
  • liquid tumor refers to an abnormal mass of cells that is fluid in nature.
  • Liquid tumor cancers include, but are not limited to, leukemias, myelomas, and lymphomas, as well as other hematological malignancies. TILs obtained from liquid tumors may also be referred to herein as marrow infiltrating lymphocytes (MILs).
  • MILs marrow infiltrating lymphocytes
  • “Ferromagnetic” materials are strongly susceptible to magnetic fields and are capable of retaining magnetic properties when the field is removed. “Paramagnetic” materials have only a weak magnetic susceptibility and when the field is removed quickly lose their weak magnetism. “Superparamagnetic” materials are highly magnetically susceptible, i.e. they become strongly magnetic when placed in a magnetic field, but, like paramagnetic materials, rapidly lose their magnetism. [00173] “Marker” as used herein refers to a cell antigen that is specifically expressed by a certain cell type. Preferentially, the marker is a cell surface marker, so that enrichment, isolation and/or detection of living cells can be performed.
  • Marker-binding fragment refers to any moiety that binds preferentially to the desired target molecule of the cell, i.e. the antigen.
  • the term moiety comprises, e.g., an antibody or antibody fragment.
  • antibody refers to polyclonal or monoclonal antibodies which can be generated by methods well known to the person skilled in the art.
  • the antibody may be of any species, e.g. murine, rat, sheep, human.
  • non-human antigen binding fragments are to be used, these can be humanized by any method known in the art.
  • the antibodies may also be modified antibodies (e.g. oligomers, reduced, oxidized and labelled antibodies).
  • antibody comprises both intact molecules and antibody fragments, such as Fab, Fab', F(ab')2, Fv and single- chain antibodies.
  • marker-binding fragment includes any moiety other than antibodies or antibody fragments that binds preferentially to the desired target molecule of the cell. Suitable moieties include, without limitation, oligonucleotides known as aptamers that bind to desired target molecules (Hermann and Pantel, 2000: Science 289: 820-825), carbohydrates, lectins or any other antigen binding protein (e.g. receptor-ligand interaction).
  • Media means various solutions known in the art of cell culturing, cell handling and stabilization used to reduce cell death, including but not limited to one or more of the following media Organ Preservation Solutions , selective lysis solutions, PBS, DMEM, HBSS, DPBS, RPMI, Iscove’s medium, X-VIVOTM, Lactated Ringer's solution, Ringer's acetate, saline, PLASMALYTETM solution, crystalloid solutions and IV fluids, colloid solutions and IV fluids, five percent dextrose in water (D5W), Hartmann's Solution.
  • the media can be standard cell media like the above mentioned-media or special media for e.g. primary human cell culture (e.g.
  • the media may have supplements or reagents well known in the art, e.g. albumins and transport proteins, amino acids and vitamins, antibiotics, attachments factors, growth factors and cytokines, hormones, metabolic inhibitors or solubilizing agents.
  • supplements or reagents well known in the art, e.g. albumins and transport proteins, amino acids and vitamins, antibiotics, attachments factors, growth factors and cytokines, hormones, metabolic inhibitors or solubilizing agents.
  • Various media are commercially available e. g. from ThermoFisher Scientific or Sigma-Aldrich.
  • microenvironment may refer to the solid or hematological tumor microenvironment as a whole or to an individual subset of cells within the microenvironment.
  • the tumor microenvironment refers to a complex mixture of “cells, soluble factors, signaling molecules, extracellular matrices, and mechanical cues that promote neoplastic transformation, support tumor growth and invasion, protect the tumor from host immunity, foster therapeutic resistance, and provide niches for dominant metastases to thrive,” as described in Swartz, et al., Cancer Res., 2012, 72, 2473.
  • tumors express antigens that should be recognized by T cells, tumor clearance by the immune system is rare because of immune suppression by the microenvironment.
  • Non-labelled or untouched refers to the cells which are not bound by one marker-binding fragment coupled to a solid phase.
  • the non-labelled, untouched cell fraction contains the desired target cells.
  • Non-target cells refers to cells which are specifically bound by one marker-binding fragment which is coupled to a solid phase that is used to remove an unwanted cell type.
  • OKT-3 refers to a monoclonal antibody or biosimilar or variant thereof, including human, humanized, chimeric, or murine antibodies, directed against the CD3 receptor in the T cell antigen receptor of mature T cells, and includes commercially-available forms such as OKT-3 (30 ng/mL, MACS GMP CD3 pure, Miltenyi Biotech, Inc., San Diego, Calif., USA) and muromonab or variants, conservative amino acid substitutions, glycoforms, or biosimilars thereof.
  • Particle refers to a solid phase such as colloidal particles, microspheres, nanoparticles, or beads.
  • peripheral blood mononuclear cells and “PBMCs” refers to a peripheral blood cell having a round nucleus, including lymphocytes (T cells, B cells, NK cells) and monocytes.
  • T cells, B cells, NK cells lymphocytes
  • monocytes irradiated allogeneic peripheral blood mononuclear cells.
  • PBMCs are a type of antigen-presenting cell.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient” are intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and inert ingredients.
  • pharmaceutically acceptable carriers or pharmaceutically acceptable excipients for active pharmaceutical ingredients is well known in the art. Except insofar as any conventional pharmaceutically acceptable carrier or pharmaceutically acceptable excipient is incompatible with the active pharmaceutical ingredient, its use in the therapeutic compositions of the invention is contemplated. Additional active pharmaceutical ingredients, such as other drugs, can also be incorporated into the described compositions and methods.
  • the term “population of cells” herein is meant a number of cells that share common traits. In general, populations generally range from l x l0 6 to l x l0 12 in number, with different TIL populations comprising different numbers.
  • “Positively separated” as used herein refers to the active separation of cells which are bound by one marker-binding fragment coupled to a solid phase and these cells are the required population of cells.
  • “Negatively separated” as used herein refers to the active separation of cells which are bound by one marker-binding fragment coupled to a solid phase and these cells are not the required population of cells.
  • “Purity” as used herein refers to the percentage of the target population or populations desired from the original solid tissue.
  • Rapid expansion means an increase in the number of antigen-specific TILs of at least about 3-fold (or 4-, 5-, 6-, 7-, 8-, or 9-fold) over a period of a week, more preferably at least about 10-fold (or 20-, 30-, 40-, 50-, 60-, 70-, 800-, or 90-fold) over a period of a week, more preferably at least about 100-fold (or 200-, 300-, 400-, 500-, 600-, 700-, 800-, or 900-fold) over a period of a week, or most preferably at least about 1000-fold or 2000-, 3000-, 4000-, 5000-, 6000-, 7000-, 8000-, or 9000-fold) over a period of a week.
  • “Regenerative medicine(s)”, “adoptive cell therapy(ies)” or “advanced therapy medicinal product(s)” are used interchangeably herein to refer to cellular material that is used for therapeutic purposes of one or more mammals either by: the action of a part of or all of the cellular material; the supportive actions of a part of or all of the cellular material with the aim to improve the wellbeing of the mammal after application.
  • the therapeutic cells can either be used directly or may require further processing, expansion and/or engineering to provide these actions.
  • “Sample” as used herein refers to a sample containing cells in any ratio. Preferentially, these cells are viable.
  • these cells can also be fixed or frozen cells which may be used for subsequent nucleic acids or protein extraction.
  • the samples may be from animals, especially mammals such as mouse, rats, or humans. Any compressible solid tissue that contains cells can be used.
  • the invention is illustrated mainly through the isolation of hematopoietic and cancer cells from solid tumor tissue. However, the invention relates to a method for isolation of a breadth of cells from any mammalian solid tissue.
  • Solid phase refers to the coupling of the marker-binding fragment, e.g. an antibody, bound to another substrate(s), e.g. particles, fluorophores, haptens like biotin, polymers, or larger surfaces such as culture dishes and microtiter plates.
  • the coupling results in direct immobilization of the antigen-binding fragment, e.g. if the antigen- binding fragment is coupled to a larger surface of a culture dish.
  • this coupling results in indirect immobilization, e.g. an antigen-binding fragment coupled directly or indirectly (via e.g. biotin) to a magnetic bead is immobilized if said bead is retained in a magnetic field.
  • the coupling of the antigen-binding fragment to other molecules results not in a direct or indirect immobilization but allows for enrichment, separation, isolation, and detection of cells according to the present invention, e.g.
  • Solid tissue refers to a piece or pieces of animal derived mammalian solid tissue which by its three dimensions i.e. length, breadth and thickness as a geometrical body is larger than the size of multiple individual cell based units and often contains connective materials such as collagen or a similar matrix that make up structure of the tissue whereby said solid tissue cannot flow through tubes or be collected by a syringe or similar small conduit or receptacle and is i.e.
  • Solid tumor refers to an abnormal mass of tissue that usually does not contain cysts or liquid areas. Solid tumors may be benign or malignant.
  • solid tumor cancer refers to malignant, neoplastic, or cancerous solid tumors. Solid tumor cancers include, but are not limited to, sarcomas, carcinomas, and lymphomas, such as cancers of the lung, breast, prostate, colon, rectum, and bladder.
  • the tissue structure of solid tumors includes interdependent tissue compartments including the parenchyma (cancer cells) and the supporting stromal cells in which the cancer cells are dispersed and which may provide a supporting microenvironment.
  • the cancer is selected from cervical cancer, head and neck cancer (including, for example, head and neck squamous cell carcinoma [HNSCC]) glioblastoma, ovarian cancer, sarcoma, pancreatic cancer, bladder cancer, breast cancer, triple negative breast cancer, and non- small cell lung carcinoma.
  • the tissue structure of solid tumors includes interdependent tissue compartments including the parenchyma (cancer cells) and the supporting stromal cells in which the cancer cells are dispersed and which may provide a supporting microenvironment.
  • cryopreserved TILs herein is meant a population of TILs that was previously cryopreserved and then treated to return to room temperature or higher, including but not limited to cell culture temperatures or temperatures wherein TILs may be administered to a patient.
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • Treatment covers any treatment of a disease in a mammal, particularly in a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development or progression; and (c) relieving the disease, i.e., causing regression of the disease and/or relieving one or more disease symptoms. “Treatment” is also meant to encompass delivery of an agent in order to provide for a pharmacologic effect, even in the absence of a disease or condition.
  • treatment encompasses delivery of a composition that can elicit an immune response or confer immunity in the absence of a disease condition, e.g., in the case of a vaccine.
  • TILs tumor infiltrating lymphocytes
  • lymphocytes cytotoxic T cells
  • Thi and Thi 7 CD4 + T cells natural killer cells
  • dendritic cells dendritic cells
  • Ml macrophages include both primary and secondary TILs.
  • Primary TILs are those that are obtained from patient tissue samples as outlined herein (sometimes referred to as “freshly harvested”), and “secondary TILs” are any TIL cell populations that have been expanded or proliferated as discussed herein, including, but not limited to bulk TILs and expanded TILs (“REP TILs” or “post-REP TILs”).
  • TIL cell populations can include genetically modified TILs.
  • TILs can generally be defined either biochemically, using cell surface markers, or functionally, by their ability to infiltrate tumors and effect treatment.
  • TILs can be generally categorized by expressing one or more of the following biomarkers: CD4, CD8, TCR ⁇ , CD27, CD28, CD56, CCR7, CD45Ra, CD62L, CD95, PD-1, and CD25. Additionally and alternatively, TILs can be functionally defined by their ability to infiltrate solid tumors upon reintroduction into a patient.
  • TILS may further be characterized by potency--for example, TILS may be considered potent or functional if in response to TCR engagement they produce, for example, interferon (IFN) release is greater than about 50 pg/mL, greater than about 100 pg/mL, greater than about 150 pg/mL, or greater than about 200 pg/mL, or more preferably individual cells can be Potency through intracellular staining for CD137, CD107a, INF- ⁇ TNF- ⁇ , and IL-2 following TCR induced stimulation by flow cytometry.
  • IFN interferon
  • Retentate refers to the material that does not pass through a filter, mesh or membrane.
  • “Ultimate utility” as used herein refers to manufacture of or direct use in regenerative medicines, adoptive cell therapies, ATMPs, diagnostic in vitro studies or scientific research.
  • the terms “protein,” “polypeptide,” and “peptide,” used interchangeably herein, include polymeric forms of amino acids of any length, including coded and non-coded amino acids and chemically or biochemically modified or derivatized amino acids. The terms also include polymers that have been modified, such as polypeptides having modified peptide backbones.
  • domain refers to any part of a protein or polypeptide having a particular function or structure.
  • Proteins are said to have an “N-terminus” (amino-terminus) and a “C-terminus” (carboxy-terminus or carboxyl-terminus).
  • N-terminus relates to the start of a protein or polypeptide, terminated by an amino acid with a free amine group (-NH2).
  • C- terminus relates to the end of an amino acid chain (protein or polypeptide), terminated by a free carboxyl group (-COOH).
  • nucleic acid and “polynucleotide,” used interchangeably herein, include polymeric forms of nucleotides of any length, including ribonucleotides, deoxyribonucleotides, or analogs or modified versions thereof. They include single-, double-, and multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, and polymers comprising purine bases, pyrimidine bases, or other natural, chemically modified, biochemically modified, non-natural, or derivatized nucleotide bases. Likewise, DNA and RNA can include natural, chemically modified, biochemically modified, non-natural, or derivatized nucleotide bases.
  • Nucleic acids are said to have “5’ ends” and “3’ ends” because mononucleotides are reacted to make oligonucleotides in a manner such that the 5’ phosphate of one mononucleotide pentose ring is attached to the 3’ oxygen of its neighbor in one direction via a phosphodiester linkage.
  • An end of an oligonucleotide is referred to as the “5’ end” if its 5’ phosphate is not linked to the 3’ oxygen of a mononucleotide pentose ring.
  • an end of an oligonucleotide is referred to as the “3’ end” if its 3’ oxygen is not linked to a 5’ phosphate of another mononucleotide pentose ring.
  • a nucleic acid sequence even if internal to a larger oligonucleotide, also may be said to have 5’ and 3’ ends.
  • discrete elements are referred to as being “upstream” or 5’ of the “downstream” or 3’ elements.
  • the term “viral vector” refers to a recombinant nucleic acid that includes at least one element of viral origin and includes elements sufficient for or permissive of packaging into a viral vector particle.
  • the vector and/or particle can be utilized for the purpose of transferring DNA, RNA, or other nucleic acids into cells in vitro, ex vivo, or in vivo. Numerous forms of viral vectors are known.
  • isolated with respect to cells, tissues, proteins, and nucleic acids includes cells, tissues, proteins, and nucleic acids that are relatively purified with respect to other bacterial, viral, cellular, or other components that may normally be present in situ, up to and including a substantially pure preparation of the cells, tissues, proteins, and nucleic acids.
  • isolated also includes cells, tissues, proteins, and nucleic acids that have no naturally occurring counterpart, have been chemically synthesized and are thus substantially uncontaminated by other cells, tissues, proteins, and nucleic acids, or has been separated or purified from most other components (e.g., cellular components or organism components) with which they are naturally accompanied (e.g., other cellular proteins, nucleic acids, or cellular or extracellular components).
  • Exogenous molecules or sequences include molecules or sequences that are not normally present in a cell in that form or are provided to a cell from an external source. Normal presence includes presence with respect to the particular developmental stage and environmental conditions of the cell.
  • exogenous molecule or sequence can include a mutated version of a corresponding endogenous sequence within the cell or can include a sequence corresponding to an endogenous sequence within the cell but in a different form (i.e., not within a chromosome).
  • endogenous molecules or sequences include molecules or sequences that are normally present in that form in a particular cell at a particular developmental stage under particular environmental conditions.
  • Compositions or methods “comprising” or “including” one or more recited elements may include other elements not specifically recited.
  • a composition that “comprises” or “includes” a protein may contain the protein alone or in combination with other ingredients.
  • TILs tumor infiltrating lymphocytes
  • methods for preparing a therapeutic population of tumor infiltrating lymphocytes comprise treating a first population of TILs with one or more compounds to improve T-cell fitness.
  • the compounds and methods described herein can be used, for example, in combination with the methods disclosed in WO 2021/123832, herein incorporated by reference in its entirety for all purposes.
  • TIL therapy clinical responses are limited by T-cell dysfunction and exhaustion.
  • T- cells and TILs gradually lose functionality and proliferation potential during in vitro or in vivo antigen stimulation and expansion, a process known as T cell differentiation or exhaustion.
  • TILs in tumor tissue might be already differentiated as a result of chronic tumor antigen stimulation.
  • TILs are massively expanded in vitro, which leads to further differentiation and exhaustion.
  • the methods described herein can be used to increase TIL persistence and functionality by improving T-cell fitness. Increased fitness can be defined as one or a combination of the following: increasing expansion/proliferation potential (longer persistence), inducing and maintaining a favorable phenotype (better functionality), and preserving tumor-reactivity.
  • rejuvenating factors can be introduced at any point during the TIL isolation and ex vivo expansion process.
  • such rejuvenating factors can be transgenes (e.g., lentivirus or lentiviral vector, or electroporation of mRNA), gene knockout compounds (e.g., electroporation of CRISPR/Cas ribonucleoprotein (RNP) complex), gene knockdown compounds (e.g., lentivirus or lentiviral vector, or electroporation of siRNA or ASO), or small molecules or cytokines.
  • transgenes e.g., lentivirus or lentiviral vector, or electroporation of mRNA
  • gene knockout compounds e.g., electroporation of CRISPR/Cas ribonucleoprotein (RNP) complex
  • gene knockdown compounds e.g., lentivirus or lentiviral vector, or electroporation of siRNA or ASO
  • small molecules or cytokines cytokines
  • T cells are derived from hematopoietic stem cells resident in bone marrow but subsequently migrate to and mature in the thymus. During the process of maturation, T cells undergo a series of selection events, thereby generating a diverse repertoire of T cells. These cells are then released into the peripheral circulation to carry out their specific functions as a part of the adaptive immune system.
  • T cells are not a homogeneous group of cells but consist of many lineages, of which the predominant types are defined by the expression of two further cell markers.
  • CD4 expressing T cells are generally termed helper (Th) and are thought to orchestrate many functions of the immune system by cell-cell contact and through the production of mediator molecules called cytokines.
  • CD8 T cells are considered to be cytotoxic (Tc) and are thought to be the cells which perform direct killing of target cells. These activities are all controlled through the T cell receptor/antigen/MHC interaction – consequently, upon successful recognition of a peptide/MHC on a target cell, CD4 and CD8 cells act in concert through cytokine production and cytotoxic activity to eliminate target cells, including virus infected and tumor cells.
  • T cells do not recognize intact proteins (antigens) but respond to short, protein fragments presented on the surface of target cells by specific proteins called the Major Histocompatibility Complex (MHC).
  • MHC Major Histocompatibility Complex
  • TCR antigen-specific T cell receptor
  • peptide short protein antigens presented by MHC molecules. Consequently, only when the correct peptide is presented on the surface of a target cell associated with the correct MHC molecule will the T cell activate its effector functions. Therefore, the frequency of tumor specific T cells are enriched in the tumor making it an ideal source for tumor specific T cells i.e.
  • TIL tumor-infiltrating lymphocytes
  • Tumor specific TIL are T cells isolated from a tumor of a patient with primary or metastatic cancer. In most cancer patients circulating tumor-specific T cells can hardly be detected in blood. However, certain cancers such as cutaneous melanoma appear to be immunogenic as it has the ability to induce significant numbers of T cells with anti-tumor activity during the natural course of the tumor growth, especially within the tumor areas (Muul et al., J Immunol.1987 Feb 1;138(3):989-95).
  • Tumor-reactive T cells “selected as T cell specific for the tumor” can be isolated from tumor material and expanded ex vivo into high numbers. Reports have shown that these cells contain anti-tumor reactivity, which can result in tumor destruction and clinical responses upon reinfusion into the patient (Dudley et al., Science.2002 Oct 25;298(5594):850-4. Epub 2002 Sep 19). In subsequent trials the importance of T cell characteristics was confirmed and the benefit of “young” rapidly growing cells “Young TILs” was confirmed whereby cells are “not selected for specificity” at all.
  • TIL or CD8 selected TIL of around 50% (Besser et al., Anticancer Res.2009 Jan;29(1):145-54; Dudley et al., Clin Cancer Res.2010 Dec 15;16(24):6122-31. doi: 10.1158/1078-0432.CCR-10-1297. Epub 2010 Jul 28). [00222] Studies by Andersen et al. (Cancer Res.2012 Apr 1;72(7):1642-50. doi: 10.1158/0008-5472.CAN-11-2614. Epub 2012 Feb 6) identified that melanoma specific T cells (for known cancer antigens) are enriched within the tumor compared with T cells in the peripheral blood.
  • the tumor of the ‘734 patent is also processed to fragments which have deteriorated internal cell populations. Furthermore, the TILs used for manufacturing will only be TILs expanded from tissue fragments and not any TILs retained in the interior. Therefore, the resulting cell population may not reflect the full diversity of the tumor microenvironment. [00224]
  • Harvesting TILs requires the aseptic disaggregation of solid tissue as a bulk tumor prior to the culture and expansion of the TIL population. The conditions during solid tissue disaggregation and time taken to harvest the cells have a substantial impact on the viability and recovery of the final cellularized material.
  • a solid tissue derived cell suspension that is obtained using conventional methods often includes a wide variety of different cell types, disaggregation media, tissue debris and/or fluids.
  • selection or enrichment techniques generally utilize one of: size, shape, density, adherence, strong protein-protein interactions (i.e. antibody-antigen interactions). For example, in some instances selection may be conducted by providing a growth supporting environment and by controlling the culture conditions or more complex cell marker interactions associated with semi-permanent or permanent coupling to magnetic or non-magnetic solid or semi-solid phase substrates.
  • any sorting technology can be used, for example, affinity chromatography or any other antibody-dependent separation technique known in the art. Any ligand-dependent separation technique known in the art may be used in conjunction with both positive and negative separation techniques that rely on the physical properties of the cells.
  • An especially potent sorting technology is magnetic cell sorting. Methods to separate cells magnetically are commercially available e.g. from Thermo Fisher, Miltenyi Biotech, Stemcell Technologies, Cellpro Seattle, Advanced Magnetics, Boston Scientific, or Quad Technologies. For example, monoclonal antibodies can be directly coupled to magnetic polystyrene particles like Dynal M 450 or similar magnetic particles and used, for example for cell separation.
  • Enriching, sorting and/or detecting cells from a sample includes using monoclonal antibodies in conjunction with colloidal superparamagnetic microparticles having an organic coating of, for example, polysaccharides (e.g. magnetic-activated cell sorting (MACS) technology (Miltenyi Biotec, Bergisch Gladbach, Germany)).
  • MCS magnetic-activated cell sorting
  • Particles e.g., nanobeads or MicroBeads
  • Particles can be either directly conjugated to monoclonal antibodies or used in combination with anti-immunoglobulin, avidin, or anti-hapten-specific MicroBeads, or coated with other mammalian molecules with selective binding properties.
  • Magnetic particle selection technologies such as those described above, allows cells to be positively or negatively separated by incubating them with magnetic nanoparticles coated with antibodies or other moieties directed against a particular surface marker. This causes the cells expressing this marker to attach to the magnetic nanoparticles. Afterwards the cell solution is placed within a solid or flexible container in a strong magnetic field.
  • the cells attach to the nanoparticles (expressing the marker) and stay on the column, while other cells (not expressing the marker) flow through.
  • the cells can be separated positively or negatively with respect to the particular marker(s).
  • the cells expressing the marker(s) of interest, which attached to the magnetic column are washed out to a separate vessel, after removing the column from the magnetic field.
  • the antibody or selective moiety used is directed against surface markers(s) which are known to be present on cells that are not of interest.
  • the cells expressing these antigens bind to the column and the fraction that goes through is collected, as it contains the cells of interest. As these cells are non-labelled by the selective antibodies or moiety(s) coupled to nanoparticles, they are “untouched”.
  • the known manual or semi-automated solid tissue processing steps are labor-intensive and require a knowledge of the art.
  • the processing requires strict regulated environmental conditions during handling of the cell cultures, for example tissue processing as a part of or prior to disaggregation, enzymatic digestion and transfer into storing devices, or incubation conditions for disaggregation/cellularization and viable tissue yields.
  • TILs tumor infiltrating lymphocytes
  • the methods can further administering a therapeutic amount of the therapeutic population of TILs to a subject with a cancer to treat the subject.
  • Some such methods comprise treating a first population of TILs with a compound or one or more compounds to improve T-cell fitness (i.e., an effective amount of the compound or the one or more compounds in order to improve T-cell fitness as described herein).
  • the methods can comprise treating the first population of TILs with the compound or the combination of compounds a single time or multiple times (e.g., at least 2 times, at least 3 times, at least 4 times, or at least 5 times). If a combination of compounds is used, the TILs can be treated with the compounds simultaneously or at different times.
  • populations of isolated TILs produced by such methods, or isolated populations of TILs that comprise the compound or the combination of compounds to improve T-cell fitness are also provided herein.
  • the compounds can be any suitable agent or inhibitory agent such as a nucleic acid such as DNA or messenger RNA, an antigen-binding protein such as an antibody, an inhibitory RNA such as an antisense oligonucleotide or an RNAi agent, a nuclease agent, or a small molecule inhibitor.
  • a nucleic acid such as DNA or messenger RNA
  • an antigen-binding protein such as an antibody
  • an inhibitory RNA such as an antisense oligonucleotide or an RNAi agent
  • nuclease agent i.g., RNAi agent, a nuclease agent, or a small molecule inhibitor.
  • Any one or more of the several successive molecular mechanisms involved in the expression of a given gene or polypeptide may be targeted as intended herein.
  • these may include targeting the gene sequence (e.g., targeting the polypeptide- encoding, non-coding and/or regulatory portions of the gene sequence), the transcription of the gene into RNA, the polyadenylation and where applicable splicing and/or other post- transcriptional modifications of the RNA into mRNA, the localization of the mRNA into cell cytoplasm, where applicable other post-transcriptional modifications of the mRNA, the translation of the mRNA into a polypeptide chain, where applicable post-translational modifications of the polypeptide, and/or folding of the polypeptide chain into the mature conformation of the polypeptide.
  • targeting the gene sequence e.g., targeting the polypeptide- encoding, non-coding and/or regulatory portions of the gene sequence
  • the transcription of the gene into RNA e.g., targeting the polypeptide- encoding, non-coding and/or regulatory portions of the gene sequence
  • the transcription of the gene into RNA e.g., targeting the polypeptide- encoding, non
  • the invention comprises mRNA transfection to modify T cells. Rabinowitz et al. (2009) Hum.
  • the present invention includes production of lentivirus modified TILs.
  • Lentiviruses are complex retroviruses that have the ability to infect and express their genes in both mitotic and post-mitotic cells.
  • the most commonly known lentivirus is the human immunodeficiency virus (HIV).
  • OptiMEM serum-free media and transfection can be done 4 hours later.
  • Cells can be transfected with 10. ⁇ g of lentiviral transfer plasmid (pCasES10) and the following packaging plasmids: 5 ⁇ g of pMD2.G (VSV-g pseudotype), and 7.5 ug of psPAX2 (gag/pol/rev/tat).
  • Transfection can be done in 4 mL OptiMEM with a cationic lipid delivery agent (50 uL Lipofectamine 2000 and 100 ul Plus reagent).
  • the media can be changed to antibiotic-free DMEM with 10% fetal bovine serum.
  • Lentivirus may be purified as follows. Viral supernatants can be harvested after 48 hours. Supernatants can be first cleared of debris and filtered through a 0.45 ⁇ m low protein binding (PVDF) filter. They can then be centrifuged in an ultracentrifuge for 2 hours at 24,000 rpm. Viral pellets can be resuspended in 50 ⁇ L of DMEM overnight at 4° C. They can then be aliquoted and immediately frozen at -80° C. [00238] TILs can be obtained from surgical specimens.
  • PVDF low protein binding
  • PBLs can be thawed from frozen stock stored at ⁇ 180°C and placed into culture in AIM-V and interleukin-2 (IL-2; Cetus, Emeryville, CA) at 300 IU/ml.
  • IL-2 interleukin-2
  • the cells can be either initially placed in medium with anti-CD3 antibody, OKT3 (Ortho Biotech, Bridgewater, NJ) at 50 ng/ml, or can be placed in OKT3 medium after transduction, at the initial changing of the culture medium.
  • OKT3 Ortho Biotech, Bridgewater, NJ
  • 1 ⁇ 10 6 cells can be adjusted to a final volume of 1 mL in a 24- well tissue culture-treated plate with the viral supernatant and Polybrene (final concentration, 8 ⁇ g/ml).
  • TILs can be transduced by centrifugation of the plates for 1.5 hr at 1000 ⁇ g, 32°C.
  • the plates can be placed in a 37°C, humidified 5% CO 2 incubator overnight, and the medium can be replaced the next day.
  • TILs can be subject to the rapid expansion protocol (REP) as previously described, using OKT3 (50 ng/ml), IL-2 (5000 IU/ml), and irradiated allogeneic peripheral blood mononuclear cells from three different donors (TIL:feeder ratio, 1:100).
  • REP rapid expansion protocol
  • cryopreserved TILs can be thawed and about 2, 3, 4, 5, 6 or 7 days after outgrowth, the TILs can be subject to lentivirus transduction.
  • transduction with lentivirus is performed at an MOI of about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 based on the total number of live cells.
  • the MOI is about 5.
  • IL-2 is added at a concentration of 1000, 2000, 3000, 4000, 5000 or 6000 IU/mL.
  • concentration of IL-2 is about 3000 IU/mL.
  • the lentivirus modified cells can be cryopreserved, subjected to outgrowth, and REP, then cryopreserved.
  • lentivirus modified TILs can be thawed and infused into a lymphodepleted patient, optionally prior to IL-2 infusion.
  • a non-limiting representative methodology is as follows: [00241] On day 0, digested tumor samples from each donor were thawed in complete TIL TCM, cells are washed once by centrifuging at 400 x g for 5 minutes, resuspended in fresh TIL TCM and counted using DRAQ7 dye and anti-CD2 antibody stains. Cells are then centrifuged at 400 x g for 5 minutes, resuspended at a concentration of 1 x 10 6 cells/mL, placed into an appropriate vessel with 3000 IU/mL IL-2 and rested for two days in a 5% CO 2 incubator set to 37°C.
  • cells are seeded in appropriate scale G-REX plates for REP using pooled irradiated allogeneic PBMCs from 10 healthy donors as feeders at a 200:1 ratio of feeders:TIL.
  • the media used for the REP is complete REP TIL TCM with anti-CD3 (OKT3) antibody added at a concentration of 30 ng/mL for activation.
  • IL-2 is added at a concentration of 3000 IU/mL and cells are placed in a 5% CO2 incubator set to 37°C.
  • the REP period was 12 days during which IL-2 (3000 IU/mL) was supplemented every 2-3 days.
  • antigen-binding protein includes any protein that binds to an antigen.
  • antigen-binding proteins include an antibody, an antigen-binding fragment of an antibody, a multispecific antibody (e.g., a bi-specific antibody), an scFV, a bis-scFV, a diabody, a triabody, a tetrabody, a V-NAR, a VHH, a VL, a F(ab), a F(ab)2, a DVD (dual variable domain antigen-binding protein), an SVD (single variable domain antigen-binding protein),or a bispecific T-cell engager (BiTE).
  • Nuclease agents can include, for example, a Clustered Regularly Interspersed Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas) (CRISPR/Cas) nuclease, a zinc finger nuclease (ZFN), or a Transcription Activator-Like Effector Nuclease (TALEN) that cleaves a target site in target gene.
  • CRISPR/Cas nuclease comprise a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene.
  • the CRISPR/Cas system can be, for example, a type II system (e.g., Cas9) or a type V system (e.g., Cas12a). See, e.g., Zetsche et al. (2015) Cell 163(3):759-771, WO 2013/176772, WO 2014/065596, WO 2016/106121, and WO 2019/067910, each of which is herein incorporated by reference in its entirety for all purposes.
  • the compounds used herein may be DNA targeting agents.
  • DNA targeting herein may be the specific introduction of a knock-out, edit, or knock-in at a particular DNA sequence, such as in a chromosome of a cell, using methods known in the art.
  • a knock-out as used herein represents a DNA sequence of a cell that has been rendered partially or completely inoperative by targeting using methods known in the art, such as with a Cas protein as described above. Such a DNA sequence prior to knock-out could have encoded an amino acid sequence, or could have had a regulatory function (e.g., promoter), for example.
  • a knock-out may be produced by an indel (insertion or deletion of nucleotide bases in a target DNA sequence through NHEJ), or by specific removal of sequence that reduces or completely destroys the function of sequence at or near the targeting site.
  • a knock-in represents the replacement or insertion of a DNA sequence at a specific DNA sequence in cell by targeting using methods known in the art, such as with a Cas protein.
  • RNAi agent is a composition that comprises a small double-stranded RNA or RNA-like (e.g., chemically modified RNA) oligonucleotide molecule capable of facilitating degradation or inhibition of translation of a target RNA, such as messenger RNA (mRNA), in a sequence-specific manner.
  • mRNA messenger RNA
  • the oligonucleotide in the RNAi agent is a polymer of linked nucleosides, each of which can be independently modified or unmodified.
  • RNAi agents operate through the RNA interference mechanism (i.e., inducing RNA interference through interaction with the RNA interference pathway machinery (RNA-induced silencing complex or RISC) of mammalian cells). While it is believed that RNAi agents, as that term is used herein, operate primarily through the RNA interference mechanism, the disclosed RNAi agents are not bound by or limited to any particular pathway or mechanism of action.
  • RNAi agents disclosed herein comprise a sense strand and an antisense strand, and include, but are not limited to, short interfering RNAs (siRNAs), double-stranded RNAs (dsRNA), micro RNAs (miRNAs), short hairpin RNAs (shRNA), and dicer substrates.
  • the antisense strand of the RNAi agents described herein is at least partially complementary to a sequence (i.e., a succession or order of nucleobases or nucleotides, described with a succession of letters using standard nomenclature) in the target RNA.
  • ASOs antisense oligonucleotides
  • RNAi RNA interference
  • RNAi agent associates with the RNA-induced silencing complex (RISC), one strand (the passenger strand) is lost, and the remaining strand (the guide strand) cooperates with RISC to bind complementary RNA.
  • Argonaute 2 (Ago2) the catalytic component of the RISC, then cleaves the target RNA.
  • the guide strand is always associated with either the complementary sense strand or a protein (RISC).
  • RISC complementary sense strand or a protein
  • an ASO must survive and function as a single strand.
  • ASOs bind to the target RNA and block ribosomes or other factors, such as splicing factors, from binding the RNA or recruit proteins such as nucleases.
  • a gapmer is an ASO oligonucleotide containing 2–5 chemically modified nucleotides (e.g. LNA or 2’-MOE) on each terminus flanking a central 8–10 base gap of DNA. After binding the target RNA, the DNA-RNA hybrid acts substrate for RNase H.
  • the treating or the compound(s) can have, for example, one or more (any combination thereof) or all of the following effects: (1) decreases expression or activity of Fas (FAS) or FasL (FASLG) (e.g., a FAS/FASLG inhibitory agent); (2) decreases expression or activity of a TGF ⁇ R (e.g., TGF ⁇ R1) or the TGF ⁇ signaling pathway (e.g., a TGF ⁇ /TGF ⁇ R1 inhibitory agent); (3) decreases expression or activity of interferon regulatory factor 7 (IRF7) (e.g., an IRF7 inhibitory agent); (4) decreases expression or activity of DNA-directed RNA polymerase III subunit RPC1 (POLR3A) (e.g., a POLR3A inhibitory agent); (5) decreases expression or activity of ETV7; (6) decreases expression or activity of ETV3; (7) decreases expression or activity of ASH2L; (8) decreases expression or activity of PML; (9) decreases expression or activity of Fa
  • the treating or the compound(s) can have 1 of the above effects. In some embodiments, the treating or the compound(s) can have 2 of the above effects. In some embodiments, the treating or the compound(s) can have 3 of the above effects. In some embodiments, the treating or the compound(s) can have 4 of the above effects. In some embodiments, the treating or the compound(s) can have 5 of the above effects. In some embodiments, the treating or the compound(s) can have 6 of the above effects. In some embodiments, the treating or the compound(s) can have 7 of the above effects. In some embodiments, the treating or the compound(s) can have 8 of the above effects. In some embodiments, the treating or the compound(s) can have 9 of the above effects.
  • the treating or the compound(s) can have 10 of the above effects. In some embodiments, the treating or the compound(s) can have 11 of the above effects. In some embodiments, the treating or the compound(s) can have 12 of the above effects. In some embodiments, the treating or the compound(s) can have 13 of the above effects. In some embodiments, the treating or the compound(s) can have 14 of the above effects. In some embodiments, the treating or the compound(s) can have 15 of the above effects. In some embodiments, the treating or the compound(s) can have 16 of the above effects. In some embodiments, the treating or the compound(s) can have 17 of the above effects. In some embodiments, the treating or the compound(s) can have 18 of the above effects.
  • the treating or the compound(s) can have 19 of the above effects. In some embodiments, the treating or the compound(s) can have 20 of the above effects. In some embodiments, the treating or the compound(s) can have 21 of the above effects. In some embodiments, the treating or the compound(s) can have 22 of the above effects. In some embodiments, the treating or the compound(s) can have 23 of the above effects. In some embodiments, the treating or the compound(s) can have 24 of the above effects.
  • the treating or the compound(s) can have, for example, one or more or all of the following effects: (i) decreases expression or activity of Fas (FAS) or FasL (FASLG) (e.g., a FAS/FASLG inhibitory agent); (ii) decreases expression or activity of a TGF ⁇ R (e.g., TGF ⁇ R1) or the TGF ⁇ signaling pathway (e.g., a TGF ⁇ /TGF ⁇ R1 inhibitory agent); (iii) decreases expression or activity of interferon regulatory factor 7 (IRF7) (e.g., an IRF7 inhibitory agent); and (iv) decreases expression or activity of DNA-directed RNA polymerase III subunit RPC1 (POLR3A) (e.g., a POLR3A inhibitory agent).
  • the treating or the compound(s) can have effect (i). In some embodiments, the treating or the compound(s) can have effect (ii). In some embodiments, the treating or the compound(s) can have effect (iii). In some embodiments, the treating or the compound(s) can have effect (iv). In some embodiments, the treating or the compound(s) can have effects (i) and (ii). In some embodiments, the treating or the compound(s) can have effects (i) and (iii). In some embodiments, the treating or the compound(s) can have effects (i) and (iv). In some embodiments, the treating or the compound(s) can have effects (ii) and (iii).
  • the treating or the compound(s) can have effects (ii) and (iv). In some embodiments, the treating or the compound(s) can have effects (iii) and (iv). In some embodiments, the treating or the compound(s) can have effects (i), (ii), and (iii). In some embodiments, the treating or the compound(s) can have effects (i), (ii), and (iv). In some embodiments, the treating or the compound(s) can have effects (i), (iii), and (iv). In some embodiments, the treating or the compound(s) can have effects (ii), (iii), and (iv).
  • the treating or the compound(s) can have effects (i), (ii), (iii), and (iv).
  • the one or more compounds i.e., exogenous compounds
  • a FAS/FASLG inhibitory agent can be anything that decreases FAS or FASLG expression or activity or that decreases FAS signaling pathway activity.
  • a TGF ⁇ /TGF ⁇ R1 inhibitory agent can be anything that decreases TGF ⁇ R1 or TGF ⁇ 1 expression or activity or that decreases TGF ⁇ signaling pathway activity.
  • An IRF7 inhibitory agent can be anything that decreases IRF7 expression or activity.
  • a POLR3A inhibitory agent can be anything that decreases POLR3A expression or activity. Examples of suitable inhibitory agents are provided in more detail below.
  • the compound(s) can comprise a FAS/FASLG inhibitory agent (i.e., FASLG signaling pathway inhibitory agent).
  • the compounds can comprise a TGF ⁇ /TGF ⁇ R1 inhibitory agent (i.e., a TGF ⁇ signaling pathway inhibitory agent).
  • the compound(s) can comprise an IRF7 inhibitory agent. In some embodiments, the compound(s) can comprise a POLR3A inhibitory agent. In some embodiments, the compounds can comprise a FAS/FASLG inhibitory agent and a TGF ⁇ /TGF ⁇ R1 inhibitory agent. In some embodiments, the compounds can comprise a FAS/FASLG inhibitory agent and an IRF7 inhibitory agent, the compounds can comprise a FAS/FASLG inhibitory agent and a POLR3A inhibitory agent. In some embodiments, the compounds can comprise a TGF ⁇ /TGF ⁇ R1 inhibitory agent and an IRF7 inhibitory agent.
  • the compounds can comprise a TGF ⁇ /TGF ⁇ R1 inhibitory agent and a POLR3A inhibitory agent. In some embodiments, the compounds can comprise an IRF7 inhibitory agent and a POLR3A inhibitory agent. In some embodiments, the compounds can comprise a FAS/FASLG inhibitory agent, a TGF ⁇ /TGF ⁇ R1 inhibitory agent, and an IRF7 inhibitory agent. In some embodiments, the compounds can comprise a FAS/FASLG inhibitory agent, a TGF ⁇ /TGF ⁇ R1 inhibitory agent, and a POLR3A inhibitory agent.
  • the compounds can comprise a FAS/FASLG inhibitory agent, an IRF7 inhibitory agent, and a POLR3A inhibitory agent.
  • the compounds can comprise a TGF ⁇ /TGF ⁇ R1 inhibitory agent, an IRF7 inhibitory agent, and a POLR3A inhibitory agent.
  • the compounds can comprise a FAS/FASLG inhibitory agent, a TGF ⁇ /TGF ⁇ R1 inhibitory agent, an IRF7 inhibitory agent, and a POLR3A inhibitory agent.
  • the treating or compound(s) decrease expression or activity of FAS or FASLG.
  • the treating or compound(s) can transiently decrease expression or activity of FAS or FASLG.
  • the treating or compound(s) can permanently decrease expression or activity of FAS or FASLG.
  • FAS tumor necrosis factor receptor superfamily member 6; UniProt ID P25445
  • FAS tumor necrosis factor receptor superfamily member 6
  • FADD recruits caspase-8 to the activated receptor.
  • the resulting death-inducing signaling complex (DISC) performs caspase-8 proteolytic activation which initiates the subsequent cascade of caspases (aspartate-specific cysteine proteases) mediating apoptosis.
  • FAS- mediated apoptosis may have a role in the induction of peripheral tolerance, in the antigen- stimulated suicide of mature T-cells, or both.
  • FASL tumor necrosis factor ligand superfamily member 6
  • FASLG National Cancer Institute
  • a FAS/FASLG inhibitory agent comprises a DNA encoding a dominant negative FAS mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative Fas mutant can be in a viral vector, such as a lentiviral vector.
  • the FAS/FASLG inhibitory agent comprises a messenger RNA encoding a dominant negative FAS mutant.
  • the dominant negative FAS mutant comprises a mutated FADD binding site, optionally wherein the dominant negative FAS mutant is Fas_D244V.
  • the dominant negative FAS mutant comprises a deleted DD domain, optionally wherein the dominant negative FAS mutant is Fas_del230-314.
  • the FAS/FASLG inhibitory agent comprises an anti-FAS antigen-binding protein, such as antibody.
  • the FAS/FASLG inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to a FAS or FASLG messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the FAS/FASLG inhibitory agent comprises a nuclease agent targeting a nuclease target site in a FAS or FASLG gene.
  • the nuclease agent can comprise a Cas protein (e.g. a Cas9 protein or a Cas12a protein) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding Fas or FasL.
  • the FAS/FASLG inhibitory agent comprises a small molecule Fas/FasL inhibitor, such as Kp-7- 6.
  • the treating or compound(s) decrease expression or activity of TGF ⁇ or TGF ⁇ R1.
  • the treating or compound(s) can transiently decrease expression or activity of TGF ⁇ or TGF ⁇ R1.
  • the treating or compound(s) can permanently decrease expression or activity of TGF ⁇ or TGF ⁇ R1.
  • TGF-beta receptor type-1 (UniProt ID P36897) is encoded by TGFBR1 (NCBI GeneID 7046).
  • TGFBR2 TGF-beta type II serine/threonine kinase receptor
  • TGFBR1 Activated TGFBR1 phosphorylates SMAD2 which dissociates from the receptor and interacts with SMAD4.
  • SMAD2-SMAD4 complex is subsequently translocated to the nucleus where it modulates the transcription of the TGF-beta-regulated genes. This constitutes the canonical SMAD-dependent TGF-beta signaling cascade. Also involved in non-canonical, SMAD-independent TGF-beta signaling pathways.
  • TGFBR1 induces TRAF6 autoubiquitination which in turn results in MAP3K7 ubiquitination and activation to trigger apoptosis. It also regulates epithelial to mesenchymal transition through a SMAD-independent signaling pathway through PARD6A phosphorylation and activation.
  • the TGF ⁇ signaling pathway is immunosuppressive and inhibits cell proliferation.
  • the TGF ⁇ ligand is pervasively expressed among all TIL subpopulations. The receptor is expressed primarily among Tem/Temra CD8s, activated CD8s, na ⁇ ve-like CD4s, and NK- like/Exhausted Delta-Gamma/CD8s.
  • a TGF ⁇ /TGF ⁇ R1 inhibitory agent comprises a DNA encoding a dominant negative TGF ⁇ R1 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative TGF ⁇ R1 mutant can be in a viral vector, such as a lentiviral vector.
  • a TGF ⁇ /TGF ⁇ R1 inhibitory agent comprises a messenger RNA encoding a dominant negative TGF ⁇ R1 mutant.
  • a TGF ⁇ /TGF ⁇ R1 inhibitory agent comprises an anti-TGF ⁇ R1 antigen-binding protein, such as an antibody.
  • the TGF ⁇ /TGF ⁇ R1 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to a TGF ⁇ R1 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the TGF ⁇ /TGF ⁇ R1 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding TGF ⁇ R1.
  • the nuclease agent can comprise a Cas protein and a guide RNA that forms a complex with the Cas protein (e.g., a Cas9 protein or a Cas12a protein) and targets a guide RNA target sequence in the gene encoding TGF ⁇ R1.
  • the TGF ⁇ /TGF ⁇ R1 inhibitory agent comprises a small molecule TGF ⁇ R1 inhibitor, such as SB431542.
  • the treating or compound(s) decrease expression or activity of IRF7.
  • the treating or compound(s) can transiently decrease expression or activity of IRF7.
  • the treating or compound(s) can permanently decrease expression or activity of IRF7.
  • Interferon regulatory factor 7 (Uniprot ID Q92985) is encoded by IRF7 (NCBI GeneID 3665).
  • IRF7 NCBI GeneID 3665.
  • Interferon regulatory factor 7 is a key transcriptional regulator of type I interferon (IFN)-dependent immune responses and plays a critical role in the innate immune response against DNA and RNA viruses.
  • an IRF7 inhibitory agent comprises a DNA encoding a dominant negative IRF7 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative IRF7 mutant is in a viral vector, such as a lentiviral vector.
  • the IRF7 inhibitory agent comprises a messenger RNA encoding a dominant negative IRF7 mutant.
  • the IRF7 inhibitory agent comprises an anti-IRF7 antigen-binding protein, such as an antibody.
  • the IRF7 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an IRF7 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the IRF7 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding IRF7, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF7.
  • a Cas protein e.g., Cas9 or Cas12a
  • the IRF7 inhibitory agent comprises a small molecule IRF7 inhibitor.
  • the treating or compound(s) decrease expression or activity of POLR3A.
  • the treating or compound(s) can transiently decrease expression or activity of POLR3A.
  • the treating or compound(s) can permanently decrease expression or activity of POLR3A.
  • POLR3A DNA-directed RNA polymerase III subunit RPC1; UniProt ID O14802
  • POLR3A NCBI GeneID 11128. It is a DNA-dependent RNA polymerase catalyzes the transcription of DNA into RNA using the four ribonucleoside triphosphates as substrates.
  • the POLR3A inhibitory agent comprises a DNA encoding a dominant negative POLR3A mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative POLR3A mutant can be in a viral vector, such as a lentiviral vector.
  • the POLR3A inhibitory agent comprises a messenger RNA encoding a dominant negative POLR3A mutant.
  • the POLR3A inhibitory agent comprises an anti-POLR3A antigen-binding protein, such as an antibody.
  • the POLR3A inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to a POLR3A messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the POLR3A inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding POLR3A, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding POLR3A.
  • the POLR3A inhibitory agent comprises a small molecule POLR3A inhibitor.
  • the treating or compound(s) decrease expression or activity of ETV7.
  • the treating or compound(s) can transiently decrease expression or activity of ETV7.
  • the treating or compound(s) can permanently decrease expression or activity of ETV7.
  • Transcription factor ETV7 (ETS translocation variant 7; ETS variant transcription factor; Uniprot ID Q9Y603) is encoded by ETV7 (NCBI GeneID 51513).
  • Transcription factor ETV7 is a transcriptional repressor that binds to the DNA sequence 5'-CCGGAAGT-3'.
  • an ETV7 inhibitory agent comprises a DNA encoding a dominant negative ETV7 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ETV7 mutant is in a viral vector, such as a lentiviral vector.
  • the ETV7 inhibitory agent comprises a messenger RNA encoding a dominant negative ETV7 mutant.
  • the ETV7 inhibitory agent comprises an anti- ETV7 antigen-binding protein, such as an antibody.
  • the ETV7 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ETV7 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the ETV7 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ETV7, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV7.
  • the ETV7 inhibitory agent comprises a small molecule ETV7 inhibitor.
  • the treating or compound(s) decrease expression or activity of ETV3.
  • the treating or compound(s) can transiently decrease expression or activity of ETV3.
  • the treating or compound(s) can permanently decrease expression or activity of ETV3.
  • ETS translocation variant 3 (Uniprot ID P41162) is encoded by ETV3 (NCBI GeneID 2117).
  • ETS translocation variant 3 is a transcriptional repressor that contributes to growth arrest during terminal macrophage differentiation by repressing target genes involved in Ras-dependent proliferation. It also represses MMP1 promoter activity.
  • an ETV3 inhibitory agent comprises a DNA encoding a dominant negative ETV3 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ETV3 mutant is in a viral vector, such as a lentiviral vector.
  • the ETV3 inhibitory agent comprises a messenger RNA encoding a dominant negative ETV3 mutant.
  • the ETV3 inhibitory agent comprises an anti-ETV3 antigen-binding protein, such as an antibody.
  • the ETV3 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ETV3 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the ETV3 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ETV3, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3.
  • the ETV3 inhibitory agent comprises a small molecule ETV3 inhibitor.
  • the treating or compound(s) decrease expression or activity of ASH2L.
  • the treating or compound(s) can transiently decrease expression or activity of ASH2L.
  • the treating or compound(s) can permanently decrease expression or activity of ASH2L.
  • Set1/Ash2 histone methyltransferase complex subunit ASH2 (ASH2 like, histone lysine methyltransferase complex subunit; Uniprot ID Q9UBL3) is encoded by ASH2L (NCBI GeneID 9070).
  • an ASH2L inhibitory agent comprises a DNA encoding a dominant negative ASH2L mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ASH2L mutant is in a viral vector, such as a lentiviral vector.
  • the ASH2L inhibitory agent comprises a messenger RNA encoding a dominant negative ASH2L mutant.
  • the ASH2L inhibitory agent comprises an anti-ASH2L antigen-binding protein, such as an antibody.
  • the ASH2L inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ASH2L messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the ASH2L inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ASH2L, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ASH2L.
  • the ASH2L inhibitory agent comprises a small molecule ASH2L inhibitor.
  • the treating or compound(s) decrease expression or activity of PML.
  • the treating or compound(s) can transiently decrease expression or activity of PML.
  • the treating or compound(s) can permanently decrease expression or activity of PML.
  • PML PML nuclear body scaffold; promyelocytic leukemia protein; Uniprot ID P29590
  • PML NCBI GeneID 5371
  • PML functions via its association with PML-nuclear bodies (PML-NBs) in a wide range of important cellular processes, including tumor suppression, transcriptional regulation, apoptosis, senescence, DNA damage response, and viral defense mechanisms. It acts as the scaffold of PML-NBs allowing other proteins to shuttle in and out, a process which is regulated by SUMO-mediated modifications and interactions.
  • an PML inhibitory agent comprises a DNA encoding a dominant negative PML mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative PML mutant is in a viral vector, such as a lentiviral vector.
  • the PML inhibitory agent comprises a messenger RNA encoding a dominant negative PML mutant.
  • the PML inhibitory agent comprises an anti-PML antigen-binding protein, such as an antibody.
  • the PML inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an PML messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the PML inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding PML, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding PML.
  • the PML inhibitory agent comprises a small molecule PML inhibitor.
  • the treating or compound(s) decrease expression or activity of STAT2.
  • the treating or compound(s) can transiently decrease expression or activity of STAT2.
  • the treating or compound(s) can permanently decrease expression or activity of STAT2.
  • STAT2 Signal transducer and activator of transcription 2 (Uniprot ID P52630) is encoded by STAT2 (NCBI GeneID 6773). STAT2 mediates signaling by type I interferons (IFN-alpha and IFN-beta). Following type I IFN binding to cell surface receptors, Jak kinases (TYK2 and JAK1) are activated, leading to tyrosine phosphorylation of STAT1 and STAT2. The phosphorylated STATs dimerize, associate with IRF9/ISGF3G to form a complex termed ISGF3 transcription factor, and then enter the nucleus.
  • IFN-alpha and IFN-beta type IFN binding to cell surface receptors
  • Jak kinases TYK2 and JAK1
  • the phosphorylated STATs dimerize, associate with IRF9/ISGF3G to form a complex termed ISGF3 transcription factor, and then enter the nucleus.
  • an STAT2 inhibitory agent comprises a DNA encoding a dominant negative STAT2 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative STAT2 mutant is in a viral vector, such as a lentiviral vector.
  • the STAT2 inhibitory agent comprises a messenger RNA encoding a dominant negative STAT2 mutant.
  • the STAT2 inhibitory agent comprises an anti-STAT2 antigen-binding protein, such as an antibody.
  • the STAT2 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an STAT2 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the STAT2 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding STAT2, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT2.
  • the STAT2 inhibitory agent comprises a small molecule STAT2 inhibitor.
  • the treating or compound(s) decrease expression or activity of SPI1.
  • the treating or compound(s) can transiently decrease expression or activity of SPI1.
  • the treating or compound(s) can permanently decrease expression or activity of SPI1.
  • Transcription factor PU.1 (Spi-1 proto-oncogene; Uniprot ID P17947) is encoded by SPI1 (NCBI GeneID 6688). Transcription factor PU.1 binds to the PU-box, a purine-rich DNA sequence (5'-GAGGAA-3') that can act as a lymphoid-specific enhancer.
  • an SPI1 inhibitory agent comprises a DNA encoding a dominant negative SPI1 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative SPI1 mutant is in a viral vector, such as a lentiviral vector.
  • the SPI1 inhibitory agent comprises a messenger RNA encoding a dominant negative SPI1 mutant.
  • the SPI1 inhibitory agent comprises an anti-SPI1 antigen-binding protein, such as an antibody.
  • the SPI1 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an SPI1 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the SPI1 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding SPI1, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SPI1.
  • the SPI1 inhibitory agent comprises a small molecule SPI1 inhibitor.
  • the treating or compound(s) decrease expression or activity of IRF9.
  • the treating or compound(s) can transiently decrease expression or activity of IRF9.
  • the treating or compound(s) can permanently decrease expression or activity of IRF9.
  • Interferon regulatory factor 9 (Uniprot ID Q00978) is encoded by IRF9 (NCBI GeneID 10379).
  • Interferon regulatory factor 9 is a transcription factor that plays an essential role in anti-viral immunity. It mediates signaling by type I IFNs (IFN-alpha and IFN-beta).
  • an IRF9 inhibitory agent comprises a DNA encoding a dominant negative IRF9 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative IRF9 mutant is in a viral vector, such as a lentiviral vector.
  • the IRF9 inhibitory agent comprises a messenger RNA encoding a dominant negative IRF9 mutant.
  • the IRF9 inhibitory agent comprises an anti-IRF9 antigen-binding protein, such as an antibody.
  • the IRF9 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an IRF9 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the IRF9 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding IRF9, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF9.
  • the IRF9 inhibitory agent comprises a small molecule IRF9 inhibitor.
  • the treating or compound(s) decrease expression or activity of STAT1.
  • the treating or compound(s) can transiently decrease expression or activity of STAT1.
  • the treating or compound(s) can permanently decrease expression or activity of STAT1.
  • STAT1 Signal transducer and activator of transcription 1 (Uniprot ID P42224) is encoded by STAT1 (NCBI GeneID 6772). STAT1 mediates cellular responses to interferons (IFNs), cytokine KITLG/SCF and other cytokines and other growth factors. Following type I IFN (IFN-alpha and IFN-beta) binding to cell surface receptors, signaling via protein kinases leads to activation of Jak kinases (TYK2 and JAK1) and to tyrosine phosphorylation of STAT1 and STAT2.
  • ISGF3 The phosphorylated STATs dimerize and associate with ISGF3G/IRF-9 to form a complex termed ISGF3 transcription factor that enters the nucleus.
  • ISGF3 binds to the IFN stimulated response element (ISRE) to activate the transcription of IFN-stimulated genes (ISG), which drive the cell in an antiviral state.
  • IFN-gamma type II IFN (IFN-gamma)
  • STAT1 is tyrosine- and serine- phosphorylated.
  • an STAT1 inhibitory agent comprises a DNA encoding a dominant negative STAT1 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative STAT1 mutant is in a viral vector, such as a lentiviral vector.
  • the STAT1 inhibitory agent comprises a messenger RNA encoding a dominant negative STAT1 mutant.
  • the STAT1 inhibitory agent comprises an anti-STAT1 antigen-binding protein, such as an antibody.
  • the STAT1 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an STAT1 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the STAT1 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding STAT1, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT1.
  • the STAT1 inhibitory agent comprises a small molecule STAT1 inhibitor.
  • the treating or compound(s) decrease expression or activity of IRF4.
  • the treating or compound(s) can transiently decrease expression or activity of IRF4.
  • the treating or compound(s) can permanently decrease expression or activity of IRF4.
  • Interferon regulatory factor 4 (Uniprot ID Q15306) is encoded by IRF4 (NCBI GeneID 3662). Interferon regulatory factor 4 is a transcriptional activator. IRF4 binds to the interferon-stimulated response element (ISRE) of the MHC class I promoter. It also binds the immunoglobulin lambda light chain enhancer, together with PU.1. [00298] In some embodiments, an IRF4 inhibitory agent comprises a DNA encoding a dominant negative IRF4 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative IRF4 mutant is in a viral vector, such as a lentiviral vector.
  • a viral vector such as a lentiviral vector.
  • the IRF4 inhibitory agent comprises a messenger RNA encoding a dominant negative IRF4 mutant.
  • the IRF4 inhibitory agent comprises an anti-IRF4 antigen-binding protein, such as an antibody.
  • the IRF4 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an IRF4 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the IRF4 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding IRF4, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF4.
  • the IRF4 inhibitory agent comprises a small molecule IRF4 inhibitor.
  • the treating or compound(s) decrease expression or activity of JDP2.
  • the treating or compound(s) can transiently decrease expression or activity of JDP2.
  • the treating or compound(s) can permanently decrease expression or activity of JDP2.
  • Jun dimerization protein 2 (Uniprot ID Q8WYK2) is encoded by JDP2 (NCBI GeneID 122953).
  • Jun dimerization protein 2 is a component of the AP-1 transcription factor that represses transactivation mediated by the Jun family of proteins. It is involved in a variety of transcriptional responses associated with AP-1, such as UV-induced apoptosis, cell differentiation, tumorigenesis, and tumor suppression.
  • JDP2 can also function as a repressor by recruiting histone deacetylase 3/HDAC3 to the promoter region of JUN.
  • an JDP2 inhibitory agent comprises a DNA encoding a dominant negative JDP2 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative JDP2 mutant is in a viral vector, such as a lentiviral vector.
  • the JDP2 inhibitory agent comprises a messenger RNA encoding a dominant negative JDP2 mutant.
  • the JDP2 inhibitory agent comprises an anti-JDP2 antigen-binding protein, such as an antibody.
  • the JDP2 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an JDP2 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the JDP2 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding JDP2, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding JDP2.
  • the JDP2 inhibitory agent comprises a small molecule JDP2 inhibitor.
  • the treating or compound(s) decrease expression or activity of ZNF337.
  • the treating or compound(s) can transiently decrease expression or activity of ZNF337.
  • the treating or compound(s) can permanently decrease expression or activity of ZNF337.
  • Zinc finger protein 337 (Uniprot ID Q9Y3M9) is encoded by ZNF337 (NCBI GeneID 26152). Zinc finger protein 337 may be involved in transcriptional regulation.
  • an ZNF337 inhibitory agent comprises a DNA encoding a dominant negative ZNF337 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ZNF337 mutant is in a viral vector, such as a lentiviral vector.
  • the ZNF337 inhibitory agent comprises a messenger RNA encoding a dominant negative ZNF337 mutant.
  • the ZNF337 inhibitory agent comprises an anti-ZNF337 antigen-binding protein, such as an antibody.
  • the ZNF337 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ZNF337 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the ZNF337 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ZNF337, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF337.
  • the ZNF337 inhibitory agent comprises a small molecule ZNF337 inhibitor.
  • the treating or compound(s) decrease expression or activity of ETV2.
  • the treating or compound(s) can transiently decrease expression or activity of ETV2.
  • the treating or compound(s) can permanently decrease expression or activity of ETV2.
  • ETS translocation variant 2 (Uniprot ID O00321) is encoded by ETV2 (NCBI GeneID 2116). ETS translocation variant 2 binds to DNA sequences containing the consensus pentanucleotide 5'-CGGA[AT]-3'.
  • an ETV2 inhibitory agent comprises a DNA encoding a dominant negative ETV2 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ETV2 mutant is in a viral vector, such as a lentiviral vector.
  • the ETV2 inhibitory agent comprises a messenger RNA encoding a dominant negative ETV2 mutant.
  • the ETV2 inhibitory agent comprises an anti-ETV2 antigen-binding protein, such as an antibody.
  • the ETV2 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ETV2 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the ETV2 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ETV2, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV2.
  • the ETV2 inhibitory agent comprises a small molecule ETV2 inhibitor.
  • the treating or compound(s) decrease expression or activity of ETV3L.
  • the treating or compound(s) can transiently decrease expression or activity of ETV3L.
  • the treating or compound(s) can permanently decrease expression or activity of ETV3L.
  • ETS translocation variant 3-like protein (ETS variant transcription factor 3 like; Uniprot ID Q6ZN32) is encoded by ETV3L (NCBI GeneID 440695).
  • ETS translocation variant 3-like protein is a transcriptional regulator.
  • an ETV3L inhibitory agent comprises a DNA encoding a dominant negative ETV3L mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ETV3L mutant is in a viral vector, such as a lentiviral vector.
  • the ETV3L inhibitory agent comprises a messenger RNA encoding a dominant negative ETV3L mutant.
  • the ETV3L inhibitory agent comprises an anti-ETV3L antigen-binding protein, such as an antibody.
  • the ETV3L inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ETV3L messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the ETV3L inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ETV3L, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3L.
  • the ETV3L inhibitory agent comprises a small molecule ETV3L inhibitor.
  • the treating or compound(s) decrease expression or activity of SOX18.
  • the treating or compound(s) can transiently decrease expression or activity of SOX18.
  • the treating or compound(s) can permanently decrease expression or activity of SOX18.
  • SRY-box transcription factor 18 (Uniprot ID P35713) is encoded by SOX18 (NCBI GeneID 54345).
  • SRY-box transcription factor 18 is a transcriptional activator that binds to the consensus sequence 5'-AACAAAG-3' in the promoter of target genes and plays an essential role in embryonic cardiovascular development and lymphangiogenesis. It activates transcription of PROX1 and other genes coding for lymphatic endothelial markers.
  • an SOX18 inhibitory agent comprises a DNA encoding a dominant negative SOX18 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative SOX18 mutant is in a viral vector, such as a lentiviral vector.
  • the SOX18 inhibitory agent comprises a messenger RNA encoding a dominant negative SOX18 mutant.
  • the SOX18 inhibitory agent comprises an anti-SOX18 antigen-binding protein, such as an antibody.
  • the SOX18 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an SOX18 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the SOX18 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding SOX18, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SOX18.
  • a Cas protein e.g., Cas9 or Cas12a
  • the SOX18 inhibitory agent comprises a small molecule SOX18 inhibitor.
  • the treating or compound(s) decrease expression or activity of CEBPG.
  • the treating or compound(s) can transiently decrease expression or activity of CEBPG.
  • the treating or compound(s) can permanently decrease expression or activity of CEBPG.
  • CCAAT/enhancer-binding protein gamma (Uniprot ID P53567) is encoded by CEBPG (NCBI GeneID 1054).
  • CEBPG is a transcription factor that binds to the promoter and the enhancer regions of several target genes, including the enhancer element PRE-I (positive regulatory element-I) of the IL-4 gene, the promoter and the enhancer of the immunoglobulin heavy chain, and binds to GPE1, a cis-acting element in the G-CSF gene promoter.
  • an CEBPG inhibitory agent comprises a DNA encoding a dominant negative CEBPG mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative CEBPG mutant is in a viral vector, such as a lentiviral vector.
  • the CEBPG inhibitory agent comprises a messenger RNA encoding a dominant negative CEBPG mutant.
  • the CEBPG inhibitory agent comprises an anti-CEBPG antigen-binding protein, such as an antibody.
  • the CEBPG inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an CEBPG messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the CEBPG inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding CEBPG, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPG.
  • the CEBPG inhibitory agent comprises a small molecule CEBPG inhibitor.
  • the treating or compound(s) decrease expression or activity of CREB3L4.
  • the treating or compound(s) can transiently decrease expression or activity of CREB3L4.
  • the treating or compound(s) can permanently decrease expression or activity of CREB3L4.
  • Cyclic AMP-responsive element-binding protein 3-like protein 4 (Uniprot ID Q8TEY5) is encoded by CREB3L4 (NCBI GeneID 148327).
  • CREB3L4 is a transcriptional activator that may play a role in the unfolded protein response. It binds to the UPR element (UPRE) but not to CRE element.
  • UPRE UPR element
  • CREB3L4 preferentially binds DNA with to the consensus sequence 5'-T[GT]ACGT[GA][GT]-3' and has transcriptional activation activity from UPRE.
  • an CREB3L4 inhibitory agent comprises a DNA encoding a dominant negative CREB3L4 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative CREB3L4 mutant is in a viral vector, such as a lentiviral vector.
  • the CREB3L4 inhibitory agent comprises a messenger RNA encoding a dominant negative CREB3L4 mutant.
  • the CREB3L4 inhibitory agent comprises an anti-CREB3L4 antigen-binding protein, such as an antibody.
  • the CREB3L4 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an CREB3L4 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the CREB3L4 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding CREB3L4, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CREB3L4.
  • the CREB3L4 inhibitory agent comprises a small molecule CREB3L4 inhibitor.
  • the treating or compound(s) decrease expression or activity of CEBPB.
  • the treating or compound(s) can transiently decrease expression or activity of CEBPB.
  • the treating or compound(s) can permanently decrease expression or activity of CEBPB.
  • CCAAT/enhancer-binding protein beta (Uniprot ID P17676) is encoded by CEBPB (NCBI GeneID 1051).
  • CEBPB is an important transcription factor regulating the expression of genes involved in immune and inflammatory responses.
  • an CEBPB inhibitory agent comprises a DNA encoding a dominant negative CEBPB mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative CEBPB mutant is in a viral vector, such as a lentiviral vector.
  • the CEBPB inhibitory agent comprises a messenger RNA encoding a dominant negative CEBPB mutant.
  • the CEBPB inhibitory agent comprises an anti-CEBPB antigen-binding protein, such as an antibody.
  • the CEBPB inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an CEBPB messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the CEBPB inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding CEBPB, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPB.
  • a Cas protein e.g., Cas9 or Cas12a
  • the CEBPB inhibitory agent comprises a small molecule CEBPB inhibitor.
  • the treating or compound(s) decrease expression or activity of FOXD1.
  • the treating or compound(s) can transiently decrease expression or activity of FOXD1.
  • the treating or compound(s) can permanently decrease expression or activity of FOXD1.
  • Forkhead box protein D1 (Uniprot ID Q16676) is encoded by FOXD1 (NCBI GeneID 2297).
  • Forkhead box protein D1 is a transcription factor involved in regulation of gene expression in a variety of processes, including formation of positional identity in the developing retina, regionalization of the optic chiasm, morphogenesis of the kidney, and neuralization of ectodermal cells.
  • an FOXD1 inhibitory agent comprises a DNA encoding a dominant negative FOXD1 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative FOXD1 mutant is in a viral vector, such as a lentiviral vector.
  • the FOXD1 inhibitory agent comprises a messenger RNA encoding a dominant negative FOXD1 mutant.
  • the FOXD1 inhibitory agent comprises an anti-FOXD1 antigen-binding protein, such as an antibody.
  • the FOXD1 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an FOXD1 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the FOXD1 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding FOXD1, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FOXD1.
  • the FOXD1 inhibitory agent comprises a small molecule FOXD1 inhibitor.
  • the treating or compound(s) decrease expression or activity of EOMES.
  • the treating or compound(s) can transiently decrease expression or activity of EOMES.
  • the treating or compound(s) can permanently decrease expression or activity of EOMES.
  • Eomesodermin homolog (Uniprot ID O95936) is encoded by EOMES (NCBI GeneID 8320). Eomesodermin homolog functions as a transcriptional activator playing a crucial role during development, particularly in trophoblast differentiation and later in gastrulation, regulating both mesoderm delamination and endoderm specification.
  • an EOMES inhibitory agent comprises a DNA encoding a dominant negative EOMES mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative EOMES mutant is in a viral vector, such as a lentiviral vector.
  • the EOMES inhibitory agent comprises a messenger RNA encoding a dominant negative EOMES mutant.
  • the EOMES inhibitory agent comprises an anti-EOMES antigen-binding protein, such as an antibody.
  • the EOMES inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an EOMES messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the EOMES inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding EOMES, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding EOMES.
  • the EOMES inhibitory agent comprises a small molecule EOMES inhibitor.
  • the treating or compound(s) decrease expression or activity of ZNF683.
  • the treating or compound(s) can transiently decrease expression or activity of ZNF683.
  • the treating or compound(s) can permanently decrease expression or activity of ZNF683.
  • Zinc finger protein 683 also known as tissue-resident T-cell transcription regulator protein, (Uniprot ID Q8IZ20) is encoded by ZNF683 (NCBI GeneID 257101).
  • Tissue-resident T- cell transcription regulator protein is a transcription factor that mediates a transcriptional program in various innate and adaptive immune tissue-resident lymphocyte T-cell types such as tissue-resident memory T (Trm), natural killer (trNK), and natural killer T (NKT) cells and negatively regulates gene expression of proteins that promote the egress of tissue-resident T-cell populations from non-lymphoid organs. It plays a role in the development, retention, and long- term establishment of adaptive and innate tissue-resident lymphocyte T cell types in non- lymphoid organs, such as the skin and gut, but also in other non-barrier tissues like liver and kidney, and therefore may provide immediate immunological protection against reactivating infections or viral reinfection.
  • Tissue-resident T-cell transcription regulator protein plays a role in the differentiation of both thymic and peripheral NKT cells. It negatively regulates the accumulation of interferon-gamma (IFN-gamma) in NKT cells at steady state or after antigenic stimulation and positively regulates granzyme B production in NKT cells after innate stimulation. It associates with the transcriptional repressor PRDM1/BLIMP1 to chromatin at gene promoter regions.
  • an ZNF683 inhibitory agent comprises a DNA encoding a dominant negative ZNF683 mutant operably linked to a promoter active in the TILs.
  • the DNA encoding the dominant negative ZNF683 mutant is in a viral vector, such as a lentiviral vector.
  • the ZNF683 inhibitory agent comprises a messenger RNA encoding a dominant negative ZNF683 mutant.
  • the ZNF683 inhibitory agent comprises an anti-ZNF683 antigen-binding protein, such as an antibody.
  • the ZNF683 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ZNF683 messenger RNA, such as an antisense oligonucleotide or an RNAi agent.
  • the ZNF683 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ZNF683, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF683.
  • the ZNF683 inhibitory agent comprises a small molecule ZNF683 inhibitor.
  • the TILs can originate from a subject.
  • the TILs can be from a tumor biopsy, a lymph node, or ascites.
  • the tumor can be, for example, from a bladder cancer, a breast cancer, a cancer caused by human papilloma virus, a cervical cancer, a head and neck cancer, a lung cancer, a melanoma, an ovarian cancer, a non- small-cell lung cancer (NSCLC), a renal cancer, or a renal cell carcinoma.
  • the tumor biopsy is from a melanoma.
  • the methods can further comprise: (i) obtaining a refined tumor product by cryopreserving a resected tumor and disaggregating the cryopreserved tumor, disaggregating a resected tumor and cryopreserving the disaggregated tumor, cryopreserving a resected tumor and processing the tumor into multiple tumor fragments, or processing a resected tumor into multiple tumor fragments and cryopreserving the tumor fragments; and (ii) performing a first expansion by culturing the refined resected tumor product in a cell culture medium comprising IL-2 to produce the first population of TILs, optionally wherein the first population of TILs is treated with the one or more compounds during or subsequent to the first expansion.
  • Cryopreserving can comprise, for example: (1) cooling under conditions whereby heat release to, into, around or in an environment including cells, as media crystalizes, is minimized or avoided; (2) continuous cooling, from disaggregation temperature to about -80°C; (3) continuous cooling at a rate of about -2°C / min; (4) continuous cooling, from disaggregation temperature to about -80°C, at a rate of about -2°C / min; or (5) continuous cooling, from disaggregation temperature to about -80°C, or from disaggregation temperature to -80°C at a rate of about -2°C / min, wherein disaggregation temperature comprises a normal body temperature for an animal from which the tumor was resected, or room temperature or 20°C or 25°C , or normal human body temperature approximately 35°C or 36°C or 36.1°C to approximately 37°C or 37.1°C or 37.2°C or 37.3°C or below about 38.3°C.
  • Disaggregating can comprise, for example, physical disaggregation, enzymatic disaggregation, or physical and enzymatic disaggregation.
  • a single cell suspension is obtained from the refined resected tumor product and used in step (ii), or wherein the refined resected tumor product from step (i) comprises a single cell suspension.
  • the first expansion in step (ii) is performed for about two weeks.
  • the culturing in step (ii) includes adding IL-7, IL-12, IL-15, IL-18, IL-21, or a combination thereof.
  • such methods can further comprise: (iii) performing a second expansion by culturing the first population of TILs in a cell culture medium comprising IL-2, optionally wherein the first population of TILs is treated with the one or more compounds prior to, during, or subsequent to the second expansion.
  • the expanding in step (iii) comprises culturing the first population of TILs with IL-2, OKT-3, and antigen presenting cells (APCs).
  • the expanding in step (iii) is performed for about two weeks.
  • the culturing in step (iii) includes adding IL-7, IL-12, IL-15, IL-18, IL-21, or a combination thereof.
  • the method further comprises harvesting and/or cryopreserving the therapeutic population of TILs.
  • isolated therapeutic population of TILs obtained by or obtainable by any of the above methods, pharmaceutical formulations comprising a pharmaceutically acceptable excipient and any of the above isolated therapeutic populations of TILs, cryopreserved bags or intravenous infusion bags, containers, or vessels containing contents comprising any of the above isolated therapeutic populations of TILs, and methods of treating a cancer in a subject, comprising administering any of above isolated therapeutic population of TILs or the above pharmaceutical formulation to the subject.
  • the present invention relates to a method for isolating a therapeutic population of cryopreserved unmodified tumor infiltrating lymphocytes (UTIL) which may comprise: (a) resecting a tumor from a subject; (b) storing the resected tumor in a single use aseptic kit, wherein the aseptic kit comprises: a disaggregation module for receipt and processing of material comprising solid mammalian tissue; an optional enrichment module for filtration of disaggregated solid tissue material and segregation of non-disaggregated tissue and filtrate; and a stabilization module for optionally further processing and/or storing disaggregated product material, wherein each of the modules comprises one or more flexible containers connected by one or more conduits adapted to enable flow of the tissue material there between; and wherein each of the modules comprises one or more ports to permit aseptic input of media and/or reagents into the one or more flexible containers; (c) aseptically disaggregating the resected tumor
  • UTIL cryopreser
  • the disaggregation may comprise physical disaggregation, enzymatic disaggregation, or physical and enzymatic disaggregation.
  • the disaggregated tumor is cellularized or purified.
  • the present invention relates to tumor infiltrating lymphocytes (TILs) in particular unmodified TILs (UTILs), which may be isolated from tumors of a cancer patient or a metastatic cancer patient, involving autologous TILs generated from and returned to the same cancer patient.
  • TILs tumor infiltrating lymphocytes
  • UTILs unmodified TILs
  • the present invention also relates to methods for isolating a therapeutic population of cryopreserved TILs or UTILs and to TILs and UTILs obtained or obtainable via use of a device comprising a single use aseptic kit for processing of a resected tumor by the methods described herein.
  • TILs may initially be obtained from a patient tumor sample (“primary TILs”) and then expanded into a larger population for further manipulation as described herein, cryopreserved, restimulated as outlined herein and optionally evaluated for phenotype and metabolic parameters as an indication of TIL health.
  • a patient tumor sample may be obtained using methods known in the art, generally via surgical resection, needle biopsy or other means for obtaining a sample that contains a mixture of tumor and TIL cells.
  • the tumor sample may be from any solid tumor, including primary tumors, invasive tumors or metastatic tumors.
  • the tumor sample may also be a liquid tumor, such as a tumor obtained from a hematological malignancy.
  • the solid tumor may be of any cancer type, including, but not limited to, breast, ovary, cervical, pancreatic, prostate, colorectal, lung, brain, renal, stomach, and skin (including but not limited to squamous cell carcinoma, basal cell carcinoma, and melanoma).
  • TILs are obtained from malignant melanoma tumors, as these have been reported to have particularly high levels of TILs.
  • the production can generally involve a two-stage process. In stage 1, initial tumor material is dissected, placed in the aseptic kit having a disaggregation module, enzymatically digesting and/or fragmenting, and homogenizing the tumor in the disaggregation module to provide a single cell suspension.
  • the homogenized cells can be further purified within the aseptic kit in a separate enrichment module to remove components such as no longer required reagents; cell debris; non-disaggregated tissue, the cells can be directly cryopreserved to stabilize the starting material for TIL manufacture and storage in the stabilization module of the aseptic kit until Stage 2 is required.
  • Stage 2 generally involves growth of the TILs out of the resected tumor starting material (2 weeks), followed by a rapid expansion process of the TIL cells (rapid expansion protocol “REP” – 2 weeks).
  • the final product is washed and harvested prior to suspension in buffered saline, 8.5% HAS and 10% DMSO and cryopreserved to form a solid aseptic product that is thawed prior to infusion as a single dose with no further modification.
  • the core element is the TILs (i.e., tumor-derived T cells), which can target and eliminate tumor cells by a variety of methods utilized by T cells as a part of their normal function. These methods include direct methods (i.e. perforin-mediated cytotoxicity) and indirect methods (i.e. cytokine production).
  • the two other elements which contribute to the therapy are pre-conditioning chemotherapy and high dose intravenous IL-2. These two elements are thought to act by supporting engraftment of T cells in the patient after infusion: initially through conditioning chemotherapy which removes competing and regulating immune cells; followed by the IL-2 component which supports survival of T cells.
  • the structure of the cell therapy product is created by growing the TIL directly out of an enzyme digested tumor mass by means of growth supporting cell culture media and a T cell supporting growth factor Interleukin-2 (IL-2).
  • the product comprises an autologous T- cell based product where the T cells have been derived from a patient’s own cancer tissue and rapidly expanded to form a pure T cell population and T cells as defined by CD3 surface marker.
  • TILs in particular UTILs, may be produced in a two-stage process using a tumor biopsy as the starting material: Stage 1 (generally performed over 2-3 hours) initial collection and processing of tumor material using dissection, enzymatic digestion and homogenization via use of a kit and a semi-automatic device to produce a single cell suspension which can be directly cryopreserved using the stabilization module of the kit to stabilize the starting material for subsequent manufacture and Stage 2 which can occur days or years later.
  • Stage 1 generally performed over 2-3 hours
  • Stage 1 initial collection and processing of tumor material using dissection, enzymatic digestion and homogenization via use of a kit and a semi-automatic device to produce a single cell suspension which can be directly cryopreserved using the stabilization module of the kit to stabilize the starting material for subsequent manufacture
  • Stage 2 which can occur days or years later.
  • Stage 2 may be performed over 4 weeks, which may be a continuous process starting with thawing of the product of Stage 1 and growth of the TIL out of the tumor starting material (about 2 weeks) followed by a rapid expansion process of the TIL cells (about 2 weeks) to increase the number of cells and therefore dose.
  • the TILs in particular UTILs, are concentrated and washed prior to formulation as a liquid suspension of cells.
  • the aseptic drug product may be cryopreserved in a bag that will be thawed prior to intravenous infusion as a single dose with no further modification.
  • a cell suspension (containing both T cells and tumor cells) can be generated from the resected metastatic tumor using an enzyme mixture of DNase 1 and Collagenase (Type IV).
  • the combination of the repeated mechanical compression exposes additional surfaces for the enzymes to access and the enzymatic reaction speed up the process of turning a solid tissue into a cell suspension prior to optional cryopreservation.
  • a DMSO based cryoprotectant is added just prior to a controlled rate freezing cycle.
  • the enzymatic breakdown of the solid tissue may be by the selection and provision of one or more media enzyme solutions such as collagenase, trypsin, lipase, hyaluronidase, deoxyribonuclease, Liberase H1, pepsin, or any mixture thereof.
  • Enzymatic digestion of the resected metastatic tumor can occur in the disaggregation flexible containers of the semi-automatic device.
  • the media formulation for enzymatic digestion must be supplemented with enzymes that aid in protein breakdown causing the cell to cell boundaries to break down.
  • liquid formulations known in the art of cell culturing or cell handling can be used as the liquid formulation used for cell disaggregation and enzymatic digestion of solid tissues, including but not limited to one or more of the following media Organ Preservation Solutions, selective lysis solutions, PBS, DMEM, HBSS, DPBS, RPMI, Iscove’s medium, XVIVOTM, AIM-VTM, Lactated Ringer's solution, Ringer's acetate, saline, PLASMALYTETM solution, crystalloid solutions and IV fluids, colloid solutions and IV fluids, five percent dextrose in water (D5W), Hartmann's Solution, DMEM, HBSS, DPBS, RPMI, AIM-VTM, Iscove’s medium, XVIVOTM, each can be optionally supplemented with additional cell supporting factors e.g.
  • the media can be standard cell media like the above mentioned media or special media for e.g. primary human cell culture (e.g. for endothelia cells, hepatocytes or keratinocytes) or stem cells (e.g. dendritic cell maturation, hematopoietic expansion, keratinocytes, mesenchymal stem cells or T cells).
  • the media may have supplements or reagents well known in the art, e.g.
  • the liquid formulation required for enzymatic digestion must have sufficient calcium ions present in the of at least 0.1 mM up to 50 mM with an optimal range of 2 to 7 mM ideally 5 mM.
  • the solid tissue to be digested can be washed after disaggregation with a liquid formulation containing chelating agents EGTA and EDTA to remove adhesion factors and inhibitory proteins prior to washing and removal of EDTA and EGTA prior to enzymatic digestion.
  • the liquid formulation required for enzymatic digestion is more optimal with minimal chelating agents EGTA and EDTA which can severely inhibit enzyme activity by removing calcium ions required for enzyme stability and activity.
  • ⁇ - mercaptoethanol, cysteine and 8-hydroxyquinoline-5-sulfonate are other known inhibitory substances.
  • Processing of tumor material using dissection, enzymatic digestion and homogenization produces a single cell suspension of TILs, in particular UTILs, which can be directly cryopreserved to stabilize the starting material for subsequent processing via the first expansion of the cell suspension of TILs, in particular UTILs, in IL-2 to obtain a first population of TILs, in particular UTILs.
  • the methods can also comprise the step of cryopreserving the disaggregated tumor, e.g. the cell suspension.
  • Cryopreserving the disaggregated tumor is carried out on the same day as carrying out the step of aseptically disaggregating a tumor resected from a subject thereby producing a disaggregated tumor, wherein the resected tumor is sufficiently disaggregated if it can be cryopreserved without cell damage.
  • cryopreserving is carried out 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, or 22 hours following the step of disaggregating the tumor.
  • Cryopreservation of the disaggregated tumor as a single cell suspension obtained from the enzymatic disaggregation in the disaggregation module of the semi-automatic device, is carried out by cooling or maintaining the suspension at a temperature between 8 °C and at least -80 °C.
  • Disaggregation could be as quick as 5 mins but most usually 45 mins to 1 hour and the cryopreservation can be a quick as 60 mins or up to 150 mins.
  • the methods include storing the cryopreserved disaggregated tumor.
  • the device comprises at least one cell container for cryopreservation wherein the containers are a flexible container manufactured from resilient deformable material.
  • the final container is either transferred directly to a freezer -20 to -190 °C or more optimally located in the controlled rate freezing apparatus either associated with the device or supplied separately (manufactured by for example Planer Products or Asymptote Ltd) in which the temperature of the freezing chamber and the flexible storage container(s) employed to contain the enriched disaggregated solid tissue container is controlled either by: injecting a cold gas (normally nitrogen for example Planer products); or by removing heat away from the controlled cooling surface(s). Both methods result in the ability to accurately control with an error of less than 1 °C or more preferable 0.1 °C the freezing process at the required rate for the specific cell(s) to be frozen based on the freezing solution and the desired viability of the product.
  • a cold gas normally nitrogen for example Planer products
  • This cryopreservation process must consider the ice nucleation temperature which is ideally as close as possible to the melting temperature of the freezing solution.
  • ice nucleation temperature which is ideally as close as possible to the melting temperature of the freezing solution.
  • water is removed from the system as ice, and the concentration of the residual unfrozen solution increases.
  • concentration of the residual unfrozen solution increases.
  • aqueous solutions there exists a large temperature range in which ice co-exists with a concentrated aqueous solution.
  • the solution reaches the glass transition state at which point the freezing solution and cells move from a viscous solution to a solid-like state below this temperature the cells can undergo no further biological changes and hence are stabilized, for years potentially decades, until required.
  • temperatures at the start of cryopreservation include, without limitation, 40°C, 39°C, 38°C, 37°C, 36°C, 35°C, 34°C, 33°C, 32°C, 31°C, 30°C, 29°C, 28°C, 27°C, 26°C, 25°C, 24°C, 23°C, 22°C, 21°C, and 20°C, i.e., temperatures ranging from a mammalian body temperature to room temperature, and further include temperatures below room temperature, including but not limited to refrigeration temperatures such as, without limitation, 19°C, 18°C, 17°C, 16°C,
  • Target temperatures for cryogenic cooling include, without limitation, -60°C, -65°C, -70°C, -75°C, -80°C, -85°C, -90°C, and temperatures in between as well as colder temperatures down to the temperature of liquid nitrogen vapor storage (-195.79°C).
  • the methods and devices used according to the invention are designed or programmed to minimize the time from physiological temperature or digestion temperature to cryostorage temperature.
  • the methods and devices used according to the invention for cryopreservation are advantageously designed and programmed for cooling under conditions whereby heat release to, into, around or in an environment including cells, as media crystalizes, is minimized or avoided.
  • the methods and devices used according to the invention for cryopreservation are advantageously designed and programmed for cooling under conditions whereby heat release to, into, around or in an environment including cells, as media crystalizes, is minimized or avoided, for example by maintaining a pre-determined rate of temperature change of the cryopreservation media even as nucleation and crystallization of the media releases heat that resists temperature change.
  • regulating or programming a rate of temperature change includes regulating the rate of heat extraction from the cryopreservation sample to maintain a predetermined rate of temperature change.
  • the cooling rate of the cryopreservation sample is maintained by measuring the temperature of the cryopreservation sample and adjusting the rate of heat extraction through a phase change by a feedback process.
  • the cooling rate of the cryopreservation sample is maintained by anticipating a phase change and increasing the rate of heat extraction at the anticipated time of the phase change.
  • methods are designed and/or devices programmed for continuous cooling from disaggregation temperature down to a cryogenic target temperature.
  • Exemplary programmed cooling rates include, without limitation, -0.5°C/min, -1°C/min, - 1.5°C/min, -2°C/min, or -2.5°C/min.
  • the cooling rates are program targets and may vary over a cooling cycle.
  • the cooling rates may vary, for example by ⁇ 0.1°C/min, ⁇ 0.2°C/min, ⁇ 0.3°C/min, ⁇ 0.4°C/min, or ⁇ 0.5°C/min.
  • the cryopreservation temperature is -80°C ⁇ 10°C and the device is programmed to reduce temperature by 1°C/min or 1.5°C/min or 2°C/min or 1°C/min ⁇ 0.5°C/min or 1.5°C/min ⁇ 0.5°C/min or 2°C/min ⁇ 0.5°C/min.
  • Cryopreservation may be employed throughout TIL manufacture including but not limited to i) cryopreservation of a processed tumor sample for use at a later time by thawing and TIL expansion, ii) cryopreservation of a processed tumor sample for use at a later time by thawing and use of tumor cells, iii) cryopreservation of a processed tumor sample for later analysis, iv) cryopreservation of a pre-REP expansion culture for use at a later time by thawing and REP expansion, v) cryopreservation of a portion of a pre-REP expansion culture (such as but not limited to a predetermined portion or to excess cells above a predetermined amount from a pre-REP culture) for use at a later time by
  • Cryopreserved TIL intermediates, products, and samples may be washed upon thawing prior to use.
  • cryopreserved tumor digests are thawed, diluted in growth media, and washed one or more times.
  • washing comprises centrifugation and growth media change.
  • washing comprises filtration and growth media change.
  • wash media is mixed into then withdrawn from a closed TIL container, such as a bag or dish and replaced by fresh media.
  • the wash may be automated in a closed system or containers for TILs, wash media, and other components interconnected by tubes and valves.
  • TIL viability, and or TIL potency, upon thawing, dilution, and optional wash, cryopreserved TILs are held in culture prior to outgrowth (i.e. pre-REP expansion).
  • the hold time is chosen to maximize total viable cells or fold expansion measured by CD3.
  • the hold time may comprise or consist of from 2 to 4 hr. or from 4 to 6 hrs. or from 6 to 9 hrs. or from 9 to 12 hr. or from 12 to 18 hr. or from 18 to 24 hr.
  • the present methods provide for obtaining young TILs, which are capable of increased replication cycles upon administration to a subject/patient and as such may provide additional therapeutic benefits over older TILs (i.e., TILs which have further undergone more rounds of replication prior to administration to a subject/patient).
  • the diverse antigen receptors of T and B lymphocytes are produced by somatic recombination of a limited, but large number of gene segments. These gene segments: V (variable), D (diversity), J (joining), and C (constant), determine the binding specificity and downstream applications of immunoglobulins and T-cell receptors (TCRs).
  • the present invention provides a method for generating TILs which exhibit and increase the T-cell repertoire diversity.
  • the TILs obtained by the present method exhibit an increase in the T-cell repertoire diversity.
  • the TILs obtained by the present method exhibit an increase in the T-cell repertoire diversity as compared to freshly harvested TILs and/or TILs prepared using other methods than those provide.
  • the TILs obtained by the present method exhibit an increase in the T-cell repertoire diversity as compared to freshly harvested TILs and/or TILs.
  • the TILs obtained in the first expansion exhibit an increase in the T-cell repertoire diversity.
  • the increase in diversity is an increase in the immunoglobulin diversity and/or the T-cell receptor diversity.
  • the diversity is in the immunoglobulin is in the immunoglobulin heavy chain.
  • the diversity is in the immunoglobulin is in the immunoglobulin light chain.
  • the diversity is in the T-cell receptor.
  • the diversity is in one of the T-cell receptors selected from the group consisting of alpha, beta, gamma, and delta receptors.
  • TCR T- cell receptor
  • TCR T-cell receptor
  • beta T-cell receptor
  • TCRab TCRab (i.e., TCR ⁇ / ⁇ ).
  • the methods of the invention can also comprise the step of performing a first expansion by culturing the disaggregated tumor in a cell culture medium comprising IL-2 to produce a first population of TILs, in particular UTILs,.
  • the cells resulting from the steps described above are cultured in serum containing IL-2 under conditions that favor the growth of TILs over tumor and other cells.
  • the tumor digests are incubated in 2 mL wells in media comprising inactivated human AB serum with 6000 IU/mL of IL-2.
  • This primary cell population is cultured for a period of days, generally from 3 to 14 days, resulting in a bulk TIL population, generally about 1x10 8 bulk TIL cells.
  • this primary cell population is cultured for a period of 7 to 14 days, resulting in a bulk TIL population, generally about 1x10 8 bulk TIL cells. In some embodiments, this primary cell population is cultured for a period of 10 to 14 days, resulting in a bulk TIL population, generally about 1x10 8 bulk TIL cells. In some embodiments, this primary cell population is cultured for a period of about 11 days, resulting in a bulk TIL population, generally about 1x10 8 bulk TIL cells.
  • expansion of TILs may be performed using an initial bulk TIL expansion step as described below and herein, followed by a second expansion (including rapid expansion protocol (REP) steps and followed by restimulation REP steps) as described below and herein.
  • the cryopreserved disaggregated tumor tissue is thawed and resuspended 1:9 in T cell media (T cell culture media contract manufactured for Immetacyte supplemented with the following additives 10% FBS and 3000 IU/mL IL-2) prior to filtration through an inline 100-270 ⁇ m filter and centrifugation in a 50 mL centrifuge tube prior to resuspension in 20 mL.
  • a sample may be taken for flow cytometry analysis to quantify a number of HLA-A, B, C and CD58 + , and DRAQ7 ⁇ cells. In some embodiments this may be seeded using an alternative manual (such as but not limited to a hemocytometer) or alternative automated total viable cell counting device such as but not limited to NucleoCounterTM; Guava ® ; automated blood analysis and counter; pipette based cell counter such as but not limited to ScepterTM.
  • resuspended cryopreserved disaggregated tumor tissue is cultured in serum containing IL-2 under conditions that favor the growth of TILs over tumor and other cells.
  • the tumor digests are incubated in 2 mL wells in media comprising inactivated human AB serum (or, in some cases, as outlined herein, in the presence of an artificial antigen-presenting [aAPC] cell population) with 6000 IU/mL of IL-2.
  • This primary cell population is cultured for a period of days, generally from 10 to 14 days, resulting in a bulk TIL population, generally about 1x10 8 bulk TIL cells.
  • the growth media during the first expansion comprises IL-2 or a variant thereof.
  • the IL is recombinant human IL-2 (rhIL-2).
  • the IL-2 stock solution has a specific activity of 20-30x10 6 IU/mg for a 1 mg vial. In some embodiments the IL-2 stock solution has a specific activity of 20x10 6 IU/mg for a 1 mg vial. In some embodiments the IL-2 stock solution has a specific activity of 25x10 6 IU/mg for a 1 mg vial. In some embodiments the IL-2 stock solution has a specific activity of 30x10 6 IU/mg for a 1 mg vial. In some embodiments, the IL-2 stock solution has a final concentration of 4-8x10 6 IU/mg of IL-2.
  • the IL-2 stock solution has a final concentration of 5-7x10 6 IU/mg of IL-2. In some embodiments, the IL-2 stock solution has a final concentration of 6x10 6 IU/mg of IL-2.
  • the first expansion culture media comprises about 10,000 IU/mL of IL-2, about 9,000 IU/mL of IL-2, about 8,000 IU/mL of IL-2, about 7,000 IU/mL of IL-2, about 6000 IU/mL of IL-2 or about 5,000 IU/mL of IL-2. In some embodiments, the first expansion culture media comprises about 9,000 IU/mL of IL-2 to about 5,000 IU/mL of IL-2.
  • the first expansion culture media comprises about 8,000 IU/mL of IL-2 to about 6,000 IU/mL of IL-2. In some embodiments, the first expansion culture media comprises about 7,000 IU/mL of IL-2 to about 6,000 IU/mL of IL-2. In some embodiments, the first expansion culture media comprises about 6,000 IU/mL of IL-2. In an embodiment, the cell culture medium further comprises IL-2. In some embodiments, the cell culture medium comprises about 3000 IU/mL of IL-2. In an embodiment, the cell culture medium further comprises IL-2. In a preferred embodiment, the cell culture medium comprises about 3000 IU/mL of IL-2.
  • the cell culture medium comprises about 1000 IU/mL, about 1500 IU/mL, about 2000 IU/mL, about 2500 IU/mL, about 3000 IU/mL, about 3500 IU/mL, about 4000 IU/mL, about 4500 IU/mL, about 5000 IU/mL, about 5500 IU/mL, about 6000 IU/mL, about 6500 IU/mL, about 7000 IU/mL, about 7500 IU/mL, or about 8000 IU/mL of IL-2.
  • the cell culture medium comprises between 1000 and 2000 IU/mL, between 2000 and 3000 IU/mL, between 3000 and 4000 IU/mL, between 4000 and 5000 IU/mL, between 5000 and 6000 IU/mL, between 6000 and 7000 IU/mL, between 7000 and 8000 IU/mL, or about 8000 IU/mL of IL-2.
  • first expansion culture media comprises about 500 IU/mL of IL-12, about 400 IU/mL of IL-12, about 300 IU/mL of IL-12, about 200 IU/mL of IL-12, about 180 IU/mL of IL-12, about 160 IU/mL of IL-12, about 140 IU/mL of IL-12, about 120 IU/mL of IL-12, or about 100 IU/mL of IL-12.
  • the first expansion culture media comprises about 500 IU/mL of IL-12 to about 100 IU/mL of IL-12.
  • the first expansion culture media comprises about 400 IU/mL of IL-12 to about 100 IU/mL of IL-12. In some embodiments, the first expansion culture media comprises about 300 IU/mL of IL-12 to about 100 IU/mL of IL-12. In some embodiments, the first expansion culture media comprises about 200 IU/mL of IL-12. In some embodiments, the cell culture medium comprises about 180 IU/mL of IL-12. In an embodiment, the cell culture medium further comprises IL-12. In a preferred embodiment, the cell culture medium comprises about 180 IU/mL of IL-12.
  • first expansion culture media comprises about 500 IU/mL of IL-15, about 400 IU/mL of IL-15, about 300 IU/mL of IL-15, about 200 IU/mL of IL-15, about 180 IU/mL of IL-15, about 160 IU/mL of IL-15, about 140 IU/mL of IL-15, about 120 IU/mL of IL-15, or about 100 IU/mL of IL-15.
  • the first expansion culture media comprises about 500 IU/mL of IL-15 to about 100 IU/mL of IL-15.
  • the first expansion culture media comprises about 400 IU/mL of IL-15 to about 100 IU/mL of IL-15. In some embodiments, the first expansion culture media comprises about 300 IU/mL of IL-15 to about 100 IU/mL of IL-15. In some embodiments, the first expansion culture media comprises about 200 IU/mL of IL-15. In some embodiments, the cell culture medium comprises about 180 IU/mL of IL-15. In an embodiment, the cell culture medium further comprises IL-15. In a preferred embodiment, the cell culture medium comprises about 180 IU/mL of IL-15.
  • first expansion culture media comprises about 500 IU/mL of IL-18, about 400 IU/mL of IL-18, about 300 IU/mL of IL-18, about 200 IU/mL of IL-18, about 180 IU/mL of IL-18, about 160 IU/mL of IL-18, about 140 IU/mL of IL-18, about 120 IU/mL of IL-18, or about 100 IU/mL of IL-18.
  • the first expansion culture media comprises about 500 IU/mL of IL-18 to about 100 IU/mL of IL-18.
  • the first expansion culture media comprises about 400 IU/mL of IL-18 to about 100 IU/mL of IL-18. In some embodiments, the first expansion culture media comprises about 300 IU/mL of IL-18 to about 100 IU/mL of IL-18. In some embodiments, the first expansion culture media comprises about 200 IU/mL of IL-18. In some embodiments, the cell culture medium comprises about 180 IU/mL of IL-18. In an embodiment, the cell culture medium further comprises IL-18. In a preferred embodiment, the cell culture medium comprises about 180 IU/mL of IL-18.
  • first expansion culture media comprises about 20 IU/mL of IL-21, about 15 IU/mL of IL-21, about 12 IU/mL of IL-21, about 10 IU/mL of IL-21, about 5 IU/mL of IL-21, about 4 IU/mL of IL-21, about 3 IU/mL of IL-21, about 2 IU/mL of IL-21, about 1 IU/mL of IL-21, or about 0.5 IU/mL of IL-21.
  • the first expansion culture media comprises about 20 IU/mL of IL-21 to about 0.5 IU/mL of IL-21.
  • the first expansion culture media comprises about 15 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the first expansion culture media comprises about 12 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the first expansion culture media comprises about 10 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the first expansion culture media comprises about 5 IU/mL of IL-21 to about 1 IU/mL of IL-21. In some embodiments, the first expansion culture media comprises about 2 IU/mL of IL-21. In some embodiments, the cell culture medium comprises about 1 IU/mL of IL-21.
  • the cell culture medium comprises about 0.5 IU/mL of IL-21. In an embodiment, the cell culture medium further comprises IL-21. In a preferred embodiment, the cell culture medium comprises about 1 IU/mL of IL-21.
  • the culture media are combinations of interleukins, such as but not limited to, IL-2, IL-12, IL-15, IL-18 and IL-21.
  • cytokines are also contemplated, such as IL-23, IL-27, IL-35, IL-39, IL-18, IL-36, IL-37, IL-38, IFN-alpha, IFN-beta, IFN-gamma or a combination thereof along with IL-2, IL-12, IL-15, IL-18 and IL-21.
  • Antibodies such as Th2 blocking reagents, are also contemplated, such as but not limited to, IL-4 (aIL4), anti-IL-4 (aIL4R), anti-IL-5R (aIL5R), anti-IL-5 (aIL5), anti-IL13R (aIL13R), or anti-IL13 (aIL13).
  • the first TIL expansion can proceed for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days. In some embodiments, the first TIL expansion can proceed for 1 day to 14 days. In some embodiments, the first TIL expansion can proceed for 2 days to 14 days. In some embodiments, the first TIL expansion can proceed for 3 days to 14 days. In some embodiments, the first TIL expansion can proceed for 4 days to 14 days. In some embodiments, the first TIL expansion can proceed for 5 days to 14 days. In some embodiments, the first TIL expansion can proceed for 6 days to 14 days.
  • the first TIL expansion can proceed for 7 days to 14 days. In some embodiments, the first TIL expansion can proceed for 8 days to 14 days. In some embodiments, the first TIL expansion can proceed for 9 days to 14 days. In some embodiments, the first TIL expansion can proceed for 10 days to 14 days. In some embodiments, the first TIL expansion can proceed for 11 days to 14 days. In some embodiments, the first TIL expansion can proceed for 12 days to 14 days. In some embodiments, the first TIL expansion can proceed for 13 days to 14 days. In some embodiments, the first TIL expansion can proceed for 14 days. In some embodiments, the first TIL expansion can proceed for 1 day to 13 days. In some embodiments, the first TIL expansion can proceed for 2 days to 13 days.
  • the first TIL expansion can proceed for 3 days to 13 days. In some embodiments, the first TIL expansion can proceed for 4 days to 13 days. In some embodiments, the first TIL expansion can proceed for 5 days to 13 days. In some embodiments, the first TIL expansion can proceed for 6 days to 13 days. In some embodiments, the first TIL expansion can proceed for 7 days to 13 days. In some embodiments, the first TIL expansion can proceed for 8 days to 13 days. In some embodiments, the first TIL expansion can proceed for 9 days to 13 days. In some embodiments, the first TIL expansion can proceed for 10 days to 13 days. In some embodiments, the first TIL expansion can proceed for 11 days to 13 days. In some embodiments, the first TIL expansion can proceed for 12 days to 13 days.
  • the first TIL expansion can proceed for 1 day to 12 days. In some embodiments, the first TIL expansion can proceed for 2 days to 12 days. In some embodiments, the first TIL expansion can proceed for 3 days to 12 days. In some embodiments, the first TIL expansion can proceed for 4 days to 12 days. In some embodiments, the first TIL expansion can proceed for 5 days to 12 days. In some embodiments, the first TIL expansion can proceed for 6 days to 12 days. In some embodiments, the first TIL expansion can proceed for 7 days to 12 days. In some embodiments, the first TIL expansion can proceed for 8 days to 12 days. In some embodiments, the first TIL expansion can proceed for 9 days to 12 days. In some embodiments, the first TIL expansion can proceed for 10 days to 12 days.
  • the first TIL expansion can proceed for 11 days to 12 days. In some embodiments, the first TIL expansion can proceed for 1 day to 11 days. In some embodiments, the first TIL expansion can proceed for 2 days to 11 days. In some embodiments, the first TIL expansion can proceed for 3 days to 11 days. In some embodiments, the first TIL expansion can proceed for 4 days to 11 days. In some embodiments, the first TIL expansion can proceed for 5 days to 11 days. In some embodiments, the first TIL expansion can proceed for 6 days to 11 days. In some embodiments, the first TIL expansion can proceed for 7 days to 11 days. In some embodiments, the first TIL expansion can proceed for 8 days to 11 days. In some embodiments, the first TIL expansion can proceed for 9 days to 11 days.
  • the first TIL expansion can proceed for 10 days to 11 days. In some embodiments, the first TIL expansion can proceed for 11 days. In some embodiments, REP day 10 is 3 days following electroporation.
  • a combination of IL-2, IL-7, IL-15, and/or IL-21 are employed as a combination during the first expansion. In some embodiments, IL-2, IL-7, IL-15, and/or IL- 21 as well as any combinations thereof can be included during the first expansion. In some embodiments, a combination of IL-2, IL-15, and IL-21 are employed as a combination during the first expansion. [00371] In some embodiments, the first expansion is performed in a closed system bioreactor.
  • a closed system is employed for the TIL expansion, as described herein.
  • a single bioreactor is employed.
  • the single bioreactor employed is example a G-REX-10 or a G-REX-100 or advantageously the device of WO 2018/130845.
  • the closed system bioreactor is a single bioreactor.
  • the first TIL population (sometimes referred to as the bulk TIL population) or the second TIL population (which can in some embodiments include populations referred to as the REP TIL populations) can be subjected to genetic modifications for suitable treatments prior to expansion or after the first expansion and prior to the second expansion.
  • the PBMCs are cryopreserved. Cryopreservation enables prescreening and PBMC inventory maintenance and reduces the number of donors needed for TIL manufacture.
  • Disaggregated tumor tissue can be thawed.
  • the TILs obtained from the first expansion are stored until phenotyped for selection.
  • the TILs obtained from the first are not stored and proceed directly to the second expansion.
  • the methods comprise the step of performing a second expansion by culturing the first population of TILs, in particular UTILs, with additional IL-2, OKT-3, and antigen presenting cells (APCs), to produce a second population of TILs.
  • the TILs obtained from the first expansion are not cryopreserved after the first expansion and prior to the second expansion.
  • the transition from the first expansion to the second expansion occurs at about 3 days, 4, days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days after the cryopreserved 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs at about 3 days to 21 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs at about 4 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed.
  • the transition from the first expansion to the second expansion occurs at about 4 days to 10 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs at about 7 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs at about 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments the seeding of the REP culture occurs 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 days after the cryopreserved disaggregated tumor tissue is thawed.
  • the transition from the first expansion to the second expansion occurs at 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days after the cryopreserved disaggregated tumor tissue is thawed.
  • the transition from the first expansion to the second expansion occurs 1 day to 14 days after the cryopreserved disaggregated tumor tissue is thawed.
  • the first TIL expansion can proceed for 2 days to 14 days.
  • the transition from the first expansion to the second expansion occurs 3 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed.
  • the transition from the first expansion to the second expansion occurs 4 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 5 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 6 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 7 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed.
  • the transition from the first expansion to the second expansion occurs 8 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 9 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 10 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 11 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed.
  • the transition from the first expansion to the second expansion occurs 12 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 13 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 1 day to 11 days after the cryopreserved disaggregated tumor tissue is thawed.
  • the transition from the first expansion to the second expansion occurs 2 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 3 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 4 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 5 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed.
  • the transition from the first expansion to the second expansion occurs 6 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 7 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 8 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 9 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed.
  • the transition from the first expansion to the second expansion occurs 10 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 11 days after the cryopreserved disaggregated tumor tissue is thawed.
  • the TILs or a portion of the TILs from the first expansion are cryopreserved. In certain embodiments, the TILs are divided in two or more portions, one or more portion proceeding to the second expansion, and one or more portion cryopreserved to be used in a later second expansion. In certain embodiments, the number of cells at the end of the first expansion is determined and the culture divided accordingly.
  • the average potency of the TILs from the first expansion is determined and the culture is divided accordingly.
  • an predetermined minimum number or optimal number of TILs proceeds to the second expansion and the remaining TILs are cryopreserved, and later thawed and used in a further second expansion.
  • the cryopreserved TILs can alternatively be used in a first expansion followed by a second expansion.
  • the TILs are not stored after the first expansion and prior to the second expansion, and the TILs proceed directly to the second. In some embodiments, the transition occurs in closed system, as described herein.
  • the TILs from the first expansion, the second population of TILs proceeds directly into the second expansion with no transition period.
  • the transition from the first expansion to the second expansion is performed in a closed system bioreactor.
  • a closed system is employed for the TIL expansion, as described herein.
  • a single bioreactor is employed.
  • the single bioreactor employed is for example a G-REX-10 or a G-REX-100 or Xuri WAVE bioreactor.
  • the closed system bioreactor is a single bioreactor.
  • the TIL cell population is expanded in number after harvest and initial bulk processing.
  • the second expansion can include expansion processes generally referred to in the art as a rapid expansion process.
  • the second expansion is generally accomplished using a culture media comprising a number of components, including feeder cells, a cytokine source, and an anti-CD3 antibody, in a gas-permeable or gas exchanging container.
  • the second expansion or second TIL expansion of TIL can be performed using any TIL culture flasks or containers known by those of skill in the art.
  • the second TIL expansion can proceed for 0 days, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days.
  • the second TIL expansion can proceed for about 7 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 8 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 9 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 10 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 11 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 12 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 13 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 14 days. In some embodiments, the second TIL expansion can proceed for about 7 days to about 13 days.
  • the second TIL expansion can proceed for about 8 days to about 13 days. In some embodiments, the second TIL expansion can proceed for about 9 days to about 13 days. In some embodiments, the second TIL expansion can proceed for about 10 days to about 13 days. In some embodiments, the second TIL expansion can proceed for about 11 days to about 13 days. In some embodiments, the second TIL expansion can proceed for about 12 days to about 13 days. In some embodiments, the second TIL expansion can proceed for about 7 days to about 12 days. In some embodiments, the second TIL expansion can proceed for about 8 days to about 12 days. In some embodiments, the second TIL expansion can proceed for about 9 days to about 12 days. In some embodiments, the second TIL expansion can proceed for about 10 days to about 12 days.
  • the second TIL expansion can proceed for about 11 days to about 12 days. In some embodiments, the second TIL expansion can proceed for about 12 days. In some embodiments, the second TIL expansion can proceed for about 13 days. In some embodiments, the second TIL expansion can proceed for about 14 days. [00381] In an embodiment, the second expansion can be performed in a gas permeable container using the methods of the present disclosure. For example, TILs can be rapidly expanded using non-specific T-cell receptor stimulation in the presence of interleukin-2 (IL-2) or interleukin-7 (IL-7) or interleukin-15 (IL-15); or interleukin-12 (IL-12).
  • IL-2 interleukin-2
  • IL-7 interleukin-7
  • IL-15 interleukin-15
  • IL-12 interleukin-12
  • the non-specific T-cell receptor stimulus can include, for example, an anti-CD3 antibody, such as about 30 ng/ml of OKT3, a mouse monoclonal anti-CD3 antibody (commercially available from Ortho-McNeil, Raritan, N.J. or Miltenyi Biotech, Auburn, Calif.) or clone UHCT-1 (commercially available from BioLegend, San Diego, Calif., USA).
  • an anti-CD3 antibody such as about 30 ng/ml of OKT3
  • a mouse monoclonal anti-CD3 antibody commercially available from Ortho-McNeil, Raritan, N.J. or Miltenyi Biotech, Auburn, Calif.
  • clone UHCT-1 commercially available from BioLegend, San Diego, Calif., USA.
  • TILs can be expanded to induce further stimulation of the TILs in vitro by including one or more antigens during the second expansion, including antigenic portions thereof, such as epitope(s), of the cancer, which can be optionally expressed from a vector, such as a human leukocyte antigen A2 (HLA-A2) binding peptide, e.g., 0.3 ⁇ M MART-1:26-35 (27 L) or gpl 00:209-217 (210M), optionally in the presence of a T-cell growth factor, such as 300 IU/mL IL-2 or IL-15.
  • HLA-A2 human leukocyte antigen A2
  • TIL may include, e.g., NY-ESO-1, TRP-1, TRP-2, tyrosinase cancer antigen, MAGE-A3, SSX-2, and VEGFR2, or antigenic portions thereof.
  • TIL may also be rapidly expanded by re-stimulation with the same antigen(s) of the cancer pulsed onto HLA-A2-expressing antigen-presenting cells.
  • the TILs can be further re-stimulated with, e.g., example, irradiated, autologous lymphocytes or with irradiated HLA-A2+ allogeneic lymphocytes and IL-2.
  • the re-stimulation occurs as part of the second expansion.
  • the second expansion occurs in the presence of irradiated, autologous lymphocytes or with irradiated HLA-A2+ allogeneic lymphocytes and IL-2.
  • the cell culture medium further comprises IL-2. In some embodiments, the cell culture medium comprises about 3000 IU/mL of IL-2.
  • the cell culture medium comprises about 100 IU/mL, about 200 IU/mL, about 300 IU/mL, about 400 IU/mL, about 500 IU/mL, about 600 IU/mL, about 700 IU/mL, about 800 IU/mL, about 900 IU/mL, 1000 IU/mL, about 1500 IU/mL, about 2000 IU/mL, about 2500 IU/mL, about 3000 IU/mL, about 3500 IU/mL, about 4000 IU/mL, about 4500 IU/mL, about 5000 IU/mL, about 5500 IU/mL, about 6000 IU/mL, about 6500 IU/mL, about 7000 IU/mL, about 7500 IU/mL, or about 8000 IU/mL of IL-2.
  • the cell culture medium comprises between 1000 and 2000 IU/mL, between 2000 and 3000 IU/mL, between 3000 and 4000 IU/mL, between 4000 and 5000 IU/mL, between 5000 and 6000 IU/mL, between 6000 and 7000 IU/mL, between 7000 and 8000 IU/mL, or between 8000 IU/mL of IL-2.
  • the cell culture medium comprises OKT3 antibody. In some embodiments, the cell culture medium comprises about 30 ng/mL of OKT3 antibody.
  • the cell culture medium comprises about 0.1 ng/mL, about 0.5 ng/mL, about 1 ng/mL, about 2.5 ng/mL, about 5 ng/mL, about 7.5 ng/mL, about 10 ng/mL, about 15 ng/mL, about 20 ng/mL, about 25 ng/mL, about 30 ng/mL, about 35 ng/mL, about 40 ng/mL, about 50 ng/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, about 90 ng/mL, about 100 ng/mL, about 200 ng/mL, about 500 ng/mL, and about 1 ⁇ g/mL of OKT3 antibody.
  • the cell culture medium comprises between 0.1 ng/mL and 1 ng/mL, between 1 ng/mL and 5 ng/mL, between 5 ng/mL and 10 ng/mL, between 10 ng/mL and 20 ng/mL, between 20 ng/mL and 30 ng/mL, between 30 ng/mL and 40 ng/mL, between 40 ng/mL and 50 ng/mL, and between 50 ng/mL and 100 ng/mL of OKT3 antibody.
  • a combination of IL-2, IL-7, IL-15, and/or IL-21 are employed as a combination during the second expansion.
  • IL-2, IL-7, IL-15, and/or IL-21 as well as any combinations thereof can be included during the second expansion.
  • a combination of IL-2, IL-15, and IL-21 are employed as a combination during the second expansion.
  • IL-2, IL-15, and IL-21 as well as any combinations thereof can be included.
  • the second expansion can be conducted in a supplemented cell culture medium comprising IL-2, OKT-3, and antigen-presenting feeder cells.
  • the second expansion occurs in a supplemented cell culture medium.
  • the supplemented cell culture medium comprises IL-2, OKT-3, and antigen- presenting feeder cells.
  • the second cell culture medium comprises IL-2, OKT-3, and antigen-presenting cells (APCs; also referred to as antigen-presenting feeder cells).
  • APCs antigen-presenting feeder cells
  • the second expansion occurs in a cell culture medium comprising IL-2, OKT-3, and antigen-presenting feeder cells (i.e., antigen presenting cells).
  • the second expansion culture media comprises about 500 IU/mL of IL-15, about 400 IU/mL of IL-15, about 300 IU/mL of IL-15, about 200 IU/mL of IL- 15, about 180 IU/mL of IL-15, about 160 IU/mL of IL-15, about 140 IU/mL of IL-15, about 120 IU/mL of IL-15, or about 100 IU/mL of IL-15.
  • the second expansion culture media comprises about 500 IU/mL of IL-15 to about 100 IU/mL of IL-15.
  • the second expansion culture media comprises about 400 IU/mL of IL-15 to about 100 IU/mL of IL-15. In some embodiments, the second expansion culture media comprises about 300 IU/mL of IL-15 to about 100 IU/mL of IL-15. In some embodiments, the second expansion culture media comprises about 200 IU/mL of IL-15. In some embodiments, the cell culture medium comprises about 180 IU/mL of IL-15. In an embodiment, the cell culture medium further comprises IL-15. In a preferred embodiment, the cell culture medium comprises about 180 IU/mL of IL-15.
  • the second expansion culture media comprises about 20 IU/mL of IL-21, about 15 IU/mL of IL-21, about 12 IU/mL of IL-21, about 10 IU/mL of IL-21, about 5 IU/mL of IL-21, about 4 IU/mL of IL-21, about 3 IU/mL of IL-21, about 2 IU/mL of IL- 21, about 1 IU/mL of IL-21, or about 0.5 IU/mL of IL-21.
  • the second expansion culture media comprises about 20 IU/mL of IL-21 to about 0.5 IU/mL of IL-21.
  • the second expansion culture media comprises about 15 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the second expansion culture media comprises about 12 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the second expansion culture media comprises about 10 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the second expansion culture media comprises about 5 IU/mL of IL-21 to about 1 IU/mL of IL-21. In some embodiments, the second expansion culture media comprises about 2 IU/mL of IL-21. In some embodiments, the cell culture medium comprises about 1 IU/mL of IL-21.
  • the cell culture medium comprises about 0.5 IU/mL of IL-21. In an embodiment, the cell culture medium further comprises IL-21. In a preferred embodiment, the cell culture medium comprises about 1 IU/mL of IL-21.
  • the antigen-presenting feeder cells are PBMCs.
  • the ratio of TILs to PBMCs and/or antigen-presenting cells in the rapid expansion and/or the second expansion is about 1 to 25, about 1 to 50, about 1 to 100, about 1 to 125, about 1 to 150, about 1 to 175, about 1 to 200, about 1 to 225, about 1 to 250, about 1 to 275, about 1 to 300, about 1 to 325, about 1 to 350, about 1 to 375, about 1 to 400, or about 1 to 500.
  • the ratio of TILs to PBMCs in the rapid expansion and/or the second expansion is between 1 to 50 and 1 to 300.
  • the ratio of TILs to PBMCs in the rapid expansion and/or the second expansion is between 1 to 100 and 1 to 200.
  • the second expansion (which can include processes referred to as the REP process) is shortened to 0-14 days. In some embodiments, the second expansion is shortened to 7-11 days. [00390]
  • sets of containers which are interconnected and have specific separate functions maintain an aseptically closed system to process, optionally enrich but stabilize the disaggregated and cellularized tumor. Essentially the invention provides a rapid pre-sterilized environment to minimize the time required and risk of contamination or operator exposure during the processing of the resected tumor.
  • the aseptic kit allows for closed solid tissue processing, eliminating the risk of contamination of the final cellularized product compared to standard non-closed tissue processing, especially when the process is performed within a tissue retrieval/procurement site and requires storage prior to final cell processing for its ultimate utility. In addition, safety of the operator is increased due to reduction of direct contact with biological hazardous material, which may contain infectious organisms such as viruses.
  • the kit also enables either all of or a portion of the finally processed cellularized material to be stabilized for either transport or storage prior to being processed for its ultimate utility.
  • the invention will enable the resected tumor to be processed at the time of resection, or later if required, without impact upon the retrieval procedure or the viability of the cellularized tumor.
  • an optional enrichment via a form of physical purification to reduce impurities such as no longer required reagents; cell debris; non-disaggregated tumor tissue and fats can be employed.
  • the aseptic kit can have an optional enrichment module, prior to stabilization, for this purpose.
  • a single cell or small cell number aggregates can be enriched for stabilization after disaggregation by excluding particles and fluids of less than 5 ⁇ m or incompletely disaggregated material of or around 200 ⁇ m across or larger but this will vary upon the tissue and the efficiency of disaggregation and various embodiments in the form of tissue specific kits may be employed depending upon the tissue or ultimate utility of the disaggregated tumor.
  • a single cell suspension is provided after step (c).
  • the first population of UTILs requires about 1-250 million UTILs, including 1-20 million UTILS, 20-40 million UTILS, 40-60 million UTILS, 60-80 million UTILS, 80-100 million UTILS, 100-125 million UTILS, 125-150 million UTILS, 150- 200 million UTILS, or 200-250 million UTILS.
  • step (e) may further comprise growth of the UTILs out of the resected tumor starting material followed by the rapid expansion of step (f).
  • step (e) may be performed for about two weeks and step (f) may be performed for about two weeks.
  • additional step (h) involves suspending the second population of UTILs.
  • the suspending may be in buffered saline, human serum albumin, and/or dimethylsulfoxide (DMSO).
  • DMSO dimethylsulfoxide
  • the present invention also may comprise a therapeutic population of cryopreserved UTILs obtained by any of the herein disclosed methods.
  • the therapeutic population may comprise about 5x10 9 to 5x10 10 of T cells.
  • the present invention also encompasses a cryopreserved bag of the herein disclosed therapeutic population.
  • the cryopreserved bag may be for use in intravenous infusion.
  • the present invention also encompasses a method for treating cancer which may comprise administering the herein disclosed therapeutic population or the herein disclosed cryopreserved bag.
  • the present invention also encompasses the herein disclosed therapeutic population, pharmaceutical composition or cryopreserved bag for use in the treatment of cancer.
  • the cancer may be bladder cancer, breast cancer, cancer caused by human papilloma virus, cervical cancer, head and neck cancer (including head and neck squamous cell carcinoma (HNSCC), lung cancer (including non-small-cell lung cancer (NSCLC)), melanoma, ovarian cancer, renal cancer, or renal cell carcinoma.
  • HNSCC head and neck cancer
  • NSCLC non-small-cell lung cancer
  • the one or more flexible containers of the aseptic kit comprise a resilient deformable material.
  • the one or more flexible containers of the disaggregation module of the aseptic kit comprises one or more sealable openings.
  • the one or more flexible containers of the disaggregation module and/or the stabilization module may also comprise a heat sealable weld.
  • the one or more flexible containers of the aseptic kit comprises internally rounded edges.
  • the one or more flexible containers of the disaggregation module of the aseptic kit comprises disaggregation surfaces adapted to mechanically crush and shear the solid tumor therein.
  • the one or more flexible containers of the enrichment module of the aseptic kit comprises a filter that retains a retentate of cellularized disaggregated solid tumor.
  • the one or more flexible containers of the stabilization module of the aseptic kit comprises media formulation for storage of viable cells in solution or in a cryopreserved state.
  • the aseptic kit further comprises a digital, electronic, or electromagnetic tag identifier.
  • the tag identifier can relate to a specific program that defines a type of disaggregation and/or enrichment and/or stabilization process, one or more types of media used in said processes, including an optional freezing solution suitable for controlled rate freezing.
  • the same flexible container can form part of one or more of the disaggregation module, the stabilization module, and the optional enrichment modules.
  • the disaggregation module of the aseptic kit comprises a first flexible container for receipt of the tissue to be processed.
  • the disaggregation module of the aseptic kit comprises a second flexible container comprising the media for disaggregation.
  • the optional enrichment module of the aseptic kit comprises the first flexible container and a third flexible container for receiving the enriched filtrate.
  • both the disaggregation module and the stabilization module of the aseptic kit comprise the second flexible container and the second flexible container comprises digestion media and stabilization media.
  • the stabilization module of the aseptic kit comprises a fourth flexible container comprising stabilization media.
  • the stabilization module of the aseptic kit also comprises the first flexible container and/or third flexible container for storing and/or undergoing cryopreservation.
  • the present invention also provides for a method for isolating a therapeutic population of cryopreserved TILs comprising: (a) resecting a tumor from a subject; (b) storing the resected tumor in a single use aseptic kit, wherein the aseptic kit comprises: a disaggregation module for receipt and processing of material comprising solid mammalian tissue; an optional enrichment module for filtration of disaggregated solid tissue material and segregation of non- disaggregated tissue and filtrate; and a stabilization module for optionally further processing and/or storing disaggregated product material, wherein each of the modules comprises one or more flexible containers connected by one or more conduits adapted to enable flow of the tissue material there between; and wherein each of the modules comprises one or more ports to permit aseptic input of media and/or reagents into the one or more flexible containers; (c) aseptically disaggregating the resected tumor in the disaggregation module thereby producing a disaggregated tumor, where
  • the TIL activator comprises an antigen presenting cell (APC), or an artificial antigen presenting cell (aAPC), or an antigen fragment or complex or an antibody.
  • the automated device further comprises a radio frequency identification tag reader for recognition of the aseptic kit so that it may be scanned and recognized during automated processing, such as within the automated device in embodiments of the present invention.
  • the tag provides information about the conditions and steps required to be auto processed, so simply by scanning the kit, any automated system used with the kit to process the tissue can be undertaken without further intervention or contamination. Once the tissue sample has been placed in the disaggregation module, it can for example be sealed, manually or automatically, before processing begins.
  • the programmable processor of the automated device can also recognize the aseptic kit via the tag and subsequently can execute the kit program defining the type of disaggregation, enrichment, and stabilization processes, and the respective media types required for said processes, which include an optional freezing solution suitable for controlled rate freezing.
  • the programmable processor of the automated device is adaptable to communicate with and control the disaggregation module, the enrichment module, and/or the stabilization module. Put another way, the kit is therefore readable by an automated device used to execute a specific fully automatic method for processing the tumor when inserted into such a device.
  • the programmable processor of the automated device can control the disaggregation module to enable a physical and/or biological breakdown of the solid tissue material.
  • This breakdown can be a physical or enzymatic breakdown of the solid tissue material.
  • Enzymatic breakdown of the solid tissue material can be by one or more media enzyme solutions selected from the group consisting of collagenase, trypsin, lipase, hyaluronidase, deoxyribonuclease, Liberase HI, pepsin, and mixtures thereof.
  • the programmable processor controls disaggregation surfaces within the disaggregation flexible containers that mechanically crush and shear the solid tissue. In some embodiments, the disaggregation surfaces are controlled by mechanical pistons.
  • the programmable processor controls the stabilization module to cryopreserve the enriched disaggregated solid tissue in the container.
  • This may include: sensors capable of recognizing whether a disaggregation process has been completed in the disaggregation module prior to transfer of the disaggregated solid tissue to the optional enrichment module; weight sensors to determine an amount of media required in the containers of one or more of the disaggregation module; the enrichment module; and/or the stabilization module and control the transfer of material between respective containers; sensors to control temperature within the containers of the one or more of the disaggregation module; the enrichment module; and/or the stabilization module; at least one bubble sensor to control transfer of media between the input and output ports of each container in the module; at least one pump, optionally a peristaltic pump, to control transfer of media between the input and output ports; pressure sensors to assess the pressure within the enrichment module; one or more valves to control a tangential flow filtration process within the enrichment module; and/or one or more clamps to control the transfer of media between the input and output ports of each module.
  • the programmable processor of the automated device is adapted to maintain an optimal storage temperature range in the stabilization module until the container is removed; or executes a controlled freezing step. This allows the UTILs to be stored for short periods (minutes to days) or stored for long periods (multiple days to years) prior to their ultimate utility depending on the type or stabilization process used with the stabilization module.
  • the automated device further comprises a user interface.
  • the interface can comprise a display screen to display instructions that guide a user to input parameters, confirm pre-programmed steps, warn of errors, or combinations thereof.
  • the automated device is adapted to be transportable and thus may comprise dimensions that permit easy maneuverability and/or aid movement such as wheels, tires, and/or handles.
  • the present invention also provides a semi-automatic aseptic tissue processing method for isolating a therapeutic population of cryopreserved UTILs comprising the steps of: (a) automatically determining aseptic disaggregation tissue processing steps and their associated conditions from a digital, electronic, or electromagnetic tag identifier associated with an aseptic processing kit, wherein the aseptic kit comprises: a disaggregation module for receipt and processing of material comprising solid mammalian tissue; an optional enrichment module for filtration of disaggregated solid tissue material and segregation of non-disaggregated tissue and filtrate; and a stabilization module for optionally further processing and/or storing disaggregated product material, wherein each of the modules comprises one or more flexible containers connected by one or more conduits adapted to enable flow of the tissue material there between; and wherein each of the
  • Flexible containers such as bags, may be used to process tissue materials. Processing may include treatments that may separate or breakdown tissue, for example, physical breakdown may be accomplished using agitation, e.g., gentle agitation, a biological and/or enzymatic breakdown may include enzymatic digestion, and/or extraction of components of the tissue materials in the bag.
  • a flexible container, such as a bag, for processing tissue may include one or more layers made of a sealable polymer having at least three edges of the flexible container which are sealed during manufacturing and an open edge on the flexible container through which tissue material is inserted during use.
  • One or more connectors may be used to couple the flexible container to at least one element through tubing.
  • a section of the flexible container proximate the open edge may be sealed or welded to form a seal.
  • the seal may have a width of at least a three mm and be positioned substantially parallel to the open edge and spaced away from the open edge of the flexible container. In some instances, the seal may have a width greater than about five mm.
  • a bag may be sealed after tissue is placed inside to have a seal of least 5 mm positioned proximate the open edge of the bag. The seal may be parallel to the open edge and spaced away from the open edge of the bag.
  • the flexible container may be further secured using a clamp having protrusions and positioned proximate the seal and spaced further from the open edge of the flexible container than the seal.
  • the seal and the flexible container are constructed such that the flexible container can withstand a 100 N force applied to the flexible container during use.
  • a clamp in conjunction with such a seal may be advantageous in some instances depending on the type of material used and/or a structure of the seal.
  • a combination of a seal and a clamp may be capable of withstanding a 100 N force applied to the flexible container.
  • the seal and the flexible container are constructed such that the flexible container can withstand a 75 N force applied to the flexible container during use.
  • a clamp in conjunction with such a seal may be advantageous in some instances depending on the type of material used and/or a structure of the seal.
  • a combination of a seal and a clamp may be capable of withstanding a 75 N force applied to the flexible container.
  • a flexible container may be used to hold tissue during processing such as disaggregation of the tissue material.
  • a flexible container such as a bag
  • a flexible container may be used for disaggregation of the tissue material, filtration of disaggregated tissue material, and/or segregation of non-disaggregated tissue and filtrate.
  • Flexible containers such as bags may be formed from a resilient deformable material.
  • Materials for use in flexible containers, such as bags may be selected for one or more properties including but not limited to sealability such as sealability due to heat welding, or use of radio frequency energy, gas permeability, flexibility for example low temperature flexibility (e.g., at - 150oC, or -195 oC), elasticity for example low temperature elasticity, chemical resistance, optical clarity, biocompatibility such as cytotoxicity, hemolytic activity, resistance to leaching, having low particulates, high transmissions rates for particular gases (e.g., Oxygen and/or Carbon dioxide), and/or complying with regulatory requirements.
  • Flexible containers, such as bags may include indicators.
  • Indicators may be used to identify samples, patients from whom the samples were derived, and/or to track progress of a particular sample through a treatment process. In some instances, indicators may be scanned by an automated or semi-automated system to track progress of a sample.
  • Marks may be used on a flexible container, such as a bag, to identify where the bag should be placed, treated, sealed, or any other action that may be taken with respect to a bag that includes tissue. Each bag may include multiple marks for sealing.
  • An open end of the bag may be sealed after tissue is inserted in the bag. Any seal may be formed using a sealing device (e.g., heater sealer) operating at a predetermined pressure, a predetermined temperature, and predetermined time frame.
  • a sealing device e.g., heater sealer
  • a flexible container such as a bag may be used as a disaggregation container for use as part of a disaggregation element that may also include a disaggregation device.
  • media and/or enzymes may be added to a bag within a disaggregation element of a device.
  • a bag may be used with a device that mechanically crushes tissue material placed in the flexible container.
  • tissue in a flexible container such as a bag may be sheared during disaggregation.
  • the flexible container may be configured to shear the tissue material.
  • kits may be used in a semi-automated or an automated process for the aseptic disaggregation, stabilization and/or optional enrichment of mammalian cells or cell aggregates.
  • a kit for extraction of a desired material from tissue may include a disaggregation element in which at least some tissue is treated to form a processed fluid, an enrichment element (e.g., a filter) capable of enriching at least some of the processed fluid to form the desired material, a stabilization element capable of storing a portion of the desired material, and an indicator tag positioned on at least one of the disaggregation element, the enrichment element, or the stabilization element capable of providing at least one of a source of tissue, a status of the tissue with respect to the process, or a identifier.
  • an enrichment element e.g., a filter
  • the desired material may be biological material or components of a particular size.
  • the desired material may be tumor infiltrating lymphocytes (TILs).
  • TILs tumor infiltrating lymphocytes
  • Different types of media may be used in the various processes conducted by the disaggregation element and the stabilization element.
  • a cryopreservation media may be provided to the kit and used in the stabilization element to control a rate freezing.
  • An automated device for semi-automated aseptic disaggregation and/or enrichment and/or stabilization of cells or cell aggregates from mammalian solid tissue may include a programmable processor and a kit that includes the flexible container described herein.
  • the automated device may further include an indicator tag reader.
  • an indicator tag reader may be positioned at any element (e.g., disaggregation, enriching, or stabilization of tissue material in the kit).
  • an automated device may further include radio frequency identification tag reader to recognize samples in flexible containers in the kit.
  • An automated device may include a programmable processor that is capable of recognizing indicators positioned on components of the kit such as a bag via an indicator tag such as a QR code.
  • a kit for use in an automated device may include a disaggregation flexible container or bag.
  • the programmable processor may control a disaggregation element and disaggregation flexible container to enable a physical and/or biological breakdown of the solid tissue.
  • a programmable processor may control elements of an automated device such that disaggregation surfaces positioned proximate a disaggregation flexible container may mechanically crush and shear the solid tissue in the disaggregation flexible container, optionally wherein the disaggregation surfaces are mechanical pistons.
  • Disaggregation elements of a system may be controlled by a processor such that tissue in the disaggregation flexible container to enable a physical and enzymatic breakdown of the solid tissue.
  • tissue in the disaggregation flexible container may be controlled by a processor such that tissue in the disaggregation flexible container to enable a physical and enzymatic breakdown of the solid tissue.
  • One or more media enzyme solutions selected from collagenase, trypsin, lipase, hyaluronidase, deoxyribonuclease, Liberase HI, pepsin, or mixtures thereof may be provided to the disaggregation flexible container to aid in enzymatic breakdown of tissue.
  • a system may include a kit that includes a disaggregation flexible container and a stabilization flexible container and a programmable processor.
  • the programmable processor may be adapted to control one or more of: the disaggregation element; the enrichment element; and the stabilization element.
  • a programmable processor may control a stabilization element to cryopreserve the enriched disaggregated solid tissue in the stabilization container. In some embodiments, a predetermined temperature may be programmed.
  • An automated device may include additional components in a multitude of combinations.
  • Components may include sensors capable of recognizing whether a disaggregation process has been completed in the disaggregation module prior to transfer of the disaggregated solid tissue to the optional enrichment element, weight sensors to determine an amount of media required in the containers of one or more of the disaggregation element, an enrichment element, and/or the stabilization element and control the transfer of material between respective containers, sensors to control temperature within the containers of the one or more of the disaggregation element; the enrichment element; and/or the stabilization element; at least one bubble sensor to control the transfer of media between the input and output ports of each container in the element; at least one pump, optionally a peristaltic pump, to control the transfer of media between the input and output ports; pressure sensors to assess the pressure within the enrichment element; one or more valves to control a tangential flow filtration process within the enrichment element; and/or one or more clamps to control the transfer of media between the input and output ports of each element.
  • An automated device may include a programmable processor is adapted to maintain an optimal storage temperature range in the stabilization module until the container is removed.
  • the programmable processor may execute a controlled freezing step.
  • an automated device may include a user interface.
  • An interface of an automated device may include a display screen to display instructions that guide a user to input parameters, confirm pre-programmed steps, warn of errors, or combinations thereof.
  • An automated device as described herein may be adapted to be transportable.
  • An automatic tissue processing method may include automatically determining conditions for processing steps and the associated conditions from a digital, electronic or electromagnetic tag indicator associated with a component of a kit.
  • a tissue sample may be placed into a flexible container of the kit having at least one open edge. After positioning tissue in the flexible container, the open edge may be sealed.
  • tissue may be processed by automatically executing one or more tissue processing steps by communicating information associated with the indicator and controlling conditions near the flexible container and/or positions of the flexible container. Further, addition of materials to the kit may be controlled based on information associated with indicators. At least some of the processed tissue may be filtered such that a filtered fluid is generated. At least some of the filtered fluid may be provided to a cryopreservative flexible container to stabilize the desired material present in the filtered fluid.
  • Processing as described herein may include agitation, extraction, and enzymatic digestion of at least a portion of the tissue sample in the flexible container. In some instances, this processing of tissue may result in the extraction of a desired material from a tissue sample. For example, tumor infiltrating lymphocytes (TILs) may be extracted from a tissue sample.
  • TILs tumor infiltrating lymphocytes
  • Flexible containers, such as bags, for use in the methods described herein may include heat-sealable material.
  • Tissue processing and extraction from the tissue materials using a cryopreservation kit may result isolation of the desired material. In particular, materials such as tumor infiltrating lymphocytes (TILs) may be the desired material.
  • a cryopreservation kit and/or components thereof described herein may be single use in an automated and/or a semi-automated process for the disaggregation, enrichment, and/or stabilization of cells or cell aggregates.
  • bags for use in a cryopreservation kit such as a collection bag may in some embodiments be used for multiple processes. For example, collection bags may be repeatedly sealed in different locations to create separate compartments for processing of a tissue sample such as a biopsy sample and/or solid tissue.
  • Flexible containers such as bags, for use in the invention described herein include a collection bag and a cryopreservation bag may include at least a portion made from a predetermined material such as a thermoplastic, polyolefin polymer, ethylene vinyl acetate (EVA), blends such as copolymers, for example, a vinyl acetate and polyolefin polymer blend (i.e., OriGen Biomedical EVO film), a material that includes EVA, and/or coextruded layers of sealable plastics.
  • a collection bag such as a tissue collection bag of the invention may include a bag for receiving tissue made from a predetermined material such as ethylene vinyl acetate (EVA) and/or a material including EVA.
  • bags, including collection bags may be made substantially from a vinyl acetate and polyolefin polymer blend.
  • a property of interest that may be used to select a material for cryopreservation kit component such as a collection bag and/or the associated tubing may relate to heat sealing.
  • Materials for use in the bag may be selected for a specific property and/or a selection of properties, for example, sealability such as heat sealability, gas permeability, flexibility for example low temperature flexibility, elasticity for example low temperature elasticity, chemical resistance, optical clarity, biocompatibility such as cytotoxicity, hemolytic activity, resistance to leaching, having low particulates.
  • materials may be selected for specific properties for use in a coextruded material to form at least one layer of a bag.
  • Layers may be constructed such that when constructed an interior layer of the bag is relatively biocompatible, that is the material on an inner surface of the bag is stable and does not leach into the contents of the bag.
  • a property of interest that may be used to select a material for kit component such as a collection bag, a cryopreservation bag, and/or the associated tubing may relate to sealing, for example heat sealing.
  • Bags, such as collection bags and/or cryopreservation bags, and any associated tubing may be generally clear, transparent, translucent, any color desired, or a combination thereof.
  • Tissue collection bags and/or tubing may be generally fabricated in ways analogous to the fabrication of closed and/or sealed blood and/or cryopreservation bags and the associated tubing.
  • Tubing in the invention may be constructed from any desired material including, but not limited to polyvinyl chloride (PVC).
  • PVC polyvinyl chloride
  • at least one end of a collection bag may be open for receiving tissue.
  • a tissue sample for example from a biopsy may be placed in the bag through the open end, for example, a top end.
  • the biopsy sample may be cancerous tissue from an animal (e.g., domestic animal such as dog or cat) or a human.
  • the bag may be sealed, and then may be processed. Processing may include agitation, e.g., gentle agitation, extraction, and/or enzymatic digestion of the tissue in the bag. Tissue processing and extraction of a desired material, such as tumor infiltrating lymphocytes (TILs), can be in a closed system. Advantageous or preferred embodiments may include indicators to identify the patient from whom the tissue was collected and/or marks to show where the collection bag may be clamped, sealed, acted upon by a device, and/or affixed in place in an instrument. [00470] In some embodiments, bag may be formed from a sealable material.
  • TILs tumor infiltrating lymphocytes
  • bag may be formed from materials including, but not limited to polymers such as synthetic polymers including aliphatic or semi-aromatic polyamides (e.g., Nylon), ethylene-vinyl acetate (EVA) and blends thereof, thermoplastic polyurethanes (TPU), polyethylenes (PE), a vinyl acetate and polyolefin polymer blends, and/or combinations of polymers. Portions of a bag may be sealed and/or welded with energy such as heat, radio frequency energy, high frequency (HF) energy, dielectric energy, and/or any other method known in the art.
  • a collection bag may be used as a processing and/or disaggregation bag.
  • Collection bags may have width in a range from about 4 cm to about 12 cm and a width in a range from about 10 cm to about 30 cm.
  • a collection bag for use in processing may have a width of about 7.8 cm and a length of about 20 cm.
  • a bag may be heat sealable, for example, using an EVA polymer or blends thereof, a vinyl acetate and polyolefin polymer blend, and/or one or more polyamides (Nylon).
  • Indicators may include, but are not limited to codes, letters, words, names, alphanumeric codes, numbers, images, bar codes, quick response (QR) codes, tags, trackers such as smart tracker tags or bluetooth trackers, and/or any indicator known in the art.
  • indicators may be printed on, etched on, and/or adhered to a surface of a component of a kit. Indicators may also be positioned on a bag using an adhesive, for example, a sticker or tracker may be placed on a bag and/or on multiple bags. Collection bags and/or cryopreservation kit may include multiple indicators such as numeric codes and/or QR codes. [00473] Indicators, for example QR codes, tags such as smart tags, and/or trackers may be used to identify a sample within a bag as well as to instruct a device's processor such that the device runs a specific program according to a type of disaggregation, enrichment, and/or stabilization processes that are conducted in cryopreservation kits.
  • cryopreservation kit and/or components thereof may include indicators that may be readable by an automated device. The device may then execute a specific fully automatic method for processing tissue when inserted to such a device. The invention is particularly useful in a sample processing, particularly automated processing.
  • the cryopreservation kit and/or components thereof described herein may be single use in an automated and/or a semi-automated process for the disaggregation, enrichment, and/or stabilization of cells or cell aggregates.
  • bags for use in a cryopreservation kit such as a collection bag may in some embodiments be used for multiple processes. For example, collection bags may be repeatedly sealed in different locations to create separate compartments for processing of a tissue sample such as a biopsy sample and/or solid tissue.
  • marks may be placed at various locations on bags, such as tissue collection bags to indicate where the bags may be sealed, clamped, and/or affixed to an object.
  • marks showing where a bag may be clamped, sealed, and/or affixed to an object, such as instrument may be positioned on the bag prior to use. For example, one or more marks may be positioned on a bag during manufacturing.
  • Positioners may be used to ensure that tissue material in bags can be treated properly during use, for example, positioning proximate an instrument. In some systems, the positioners may facilitate the use of the bags described herein in automated systems. In particular, positioners may be used to move bag through an automated system. [00476] Use of an indicator, such as a QR code may allow for tracking of process steps for a specific sample such that it is possible to follow the sample through a given process. [00477] Cells are transferred to a container for use in administration to a patient. In some embodiments, once a therapeutically sufficient number of TILs are obtained using the expansion methods described above, they are transferred to a container for use in administration to a patient.
  • TILs expanded using APCs of the present disclosure are administered to a patient as a pharmaceutical composition.
  • the pharmaceutical composition is a suspension of TILs in a sterile buffer.
  • TILs expanded using PBMCs of the present disclosure may be administered by any suitable route as known in the art.
  • the T-cells are administered as a single intra-arterial or intravenous infusion, which preferably lasts approximately 30 to 60 minutes.
  • Other suitable routes of administration include intraperitoneal, intrathecal, and intralymphatic.
  • TILs expanded using the methods of the present disclosure are administered to a patient as a pharmaceutical composition.
  • the pharmaceutical composition is a suspension of TILs in a sterile buffer.
  • TILs expanded using PBMCs of the present disclosure may be administered by any suitable route as known in the art.
  • the T-cells are administered as a single intra-arterial or intravenous infusion, which preferably lasts approximately 30 to 60 minutes.
  • Other suitable routes of administration include intraperitoneal, intrathecal, and intralymphatic administration.
  • Any suitable dose of TILs can be administered. In some embodiments, from about 2.3x10 10 to about 13.7x10 10 TILs are administered, with an average of around 7.8x10 10 TILs, particularly if the cancer is melanoma.
  • about 1.2x10 10 to about 4.3x10 10 of TILs are administered. In some embodiments, about 3x10 10 to about 12x10 10 TILs are administered. In some embodiments, about 4x10 10 to about 10x10 10 TILs are administered. In some embodiments, about 5x10 10 to about 8x10 10 TILs are administered. In some embodiments, about 6x10 10 to about 8x10 10 TILs are administered. In some embodiments, about 7x10 10 to about 8x10 10 TILs are administered. In some embodiments, the therapeutically effective dosage is about 2.3x10 10 to about 13.7x10 10 . In some embodiments, the therapeutically effective dosage is about 7.8x10 10 TILs, particularly of the cancer is melanoma.
  • the therapeutically effective dosage is about 1.2x10 10 to about 4.3x10 10 of TILs. In some embodiments, the therapeutically effective dosage is about 3x10 10 to about 12x10 10 TILs. In some embodiments, the therapeutically effective dosage is about 4x10 10 to about 10x10 10 TILs. In some embodiments, the therapeutically effective dosage is about 5x10 10 to about 8x10 10 TILs. In some embodiments, the therapeutically effective dosage is about 6x10 10 to about 8x10 10 TILs. In some embodiments, the therapeutically effective dosage is about 7x10 10 to about 8x10 10 TILs.
  • the number of the TILs provided in the pharmaceutical compositions of the invention is about 1x10 6 , 2x10 6 , 3x10 6 , 4x10 6 , 5x10 6 , 6x10 6 , 7x10 6 8x10 6 , 9x10 6 , 1x10 7 , 2x10 7 , 3x10 7 , 4x10 7 , 5x10 7 , 6x10 7 , 7x10 7 , 8x10 7 , 9x10 7 , 1x10 8 , 2x10 8 , 3x10 8 , 4x10 8 , 5x10 8 , 6x10 8 , 7x10 8 , 8x10 8 , 9x10 8 , 1x10 9 , 2x10 9 , 3x10 9 , 4x10 9 , 5x10 9 , 6x10 9 , 7x10 9 , 8x10 9 , 9x10 9 , 1x10 10 , 2x10 9 , 3x10 9 , 4x10 9
  • the number of the TILs provided in the pharmaceutical compositions of the invention is in the range of 1x10 6 to 5x10 6 , 5x10 6 to 1x10 7 , 1x10 7 to 5x10 7 , 5x10 7 to 1x10 8 , 1x10 8 to 5x10 8 , 5x10 8 to 1x10 9 , 1x10 9 to 5x10 9 , 5x10 9 to 1x10 10 , 1x10 10 to 5x10 10 , 5x10 10 to 1x10 11 , 5x10 11 to 1x10 12 , 1x10 12 to 5x10 12 , and 5x10 12 to 1x10 13 .
  • the concentration of the TILs provided in the pharmaceutical compositions of the invention is less than, for example, 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002% or 0.0001% w/w, w/v or v/v of the pharmaceutical composition.
  • the concentration of the TILs provided in the pharmaceutical compositions of the invention is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 11.75%, 11.50%, 11.25% 11%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25% 7%, 6.75%, 6.50%, 6.25% 6%, 5.75%, 5.50%, 5.25% 5%,
  • the concentration of the TILs provided in the pharmaceutical compositions of the invention is in the range from about 0.0001% to about 50%, about 0.001% to about 40%, about 0.01% to about 30%, about 0.02% to about 29%, about 0.03% to about 28%, about 0.04% to about 27%, about 0.05% to about 26%, about 0.06% to about 25%, about 0.07% to about 24%, about 0.08% to about 23%, about 0.09% to about 22%, about 0.1% to about 21%, about 0.2% to about 20%, about 0.3% to about 19%, about 0.4% to about 18%, about 0.5% to about 17%, about 0.6% to about 16%, about 0.7% to about 15%, about 0.8% to about 14%, about 0.9% to about 12% or about 1% to about 10% w/w, w/v or v/v of the pharmaceutical composition.
  • the concentration of the TILs provided in the pharmaceutical compositions of the invention is in the range from about 0.001% to about 10%, about 0.01% to about 5%, about 0.02% to about 4.5%, about 0.03% to about 4%, about 0.04% to about 3.5%, about 0.05% to about 3%, about 0.06% to about 2.5%, about 0.07% to about 2%, about 0.08% to about 1.5%, about 0.09% to about 1%, about 0.1% to about 0.9% w/w, w/v or v/v of the pharmaceutical composition.
  • the amount of the TILs provided in the pharmaceutical compositions of the invention is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g, 0.01
  • the amount of the TILs provided in the pharmaceutical compositions of the invention is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065
  • TILs provided in the pharmaceutical compositions of the invention are effective over a wide dosage range.
  • the exact dosage will depend upon the route of administration, the form in which the compound is administered, the gender and age of the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
  • the clinically-established dosages of the TILs may also be used if appropriate.
  • the amounts of the pharmaceutical compositions administered using the methods herein, such as the dosages of TILs will be dependent on the human or mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the active pharmaceutical ingredients and the discretion of the prescribing physician.
  • TILs may be administered in a single dose.
  • TILs may be administered in multiple doses. Dosing may be once, twice, three times, four times, five times, six times, or more than six times per year. Dosing may be once a month, once every two weeks, once a week, or once every other day. Administration of TILs may continue as long as necessary.
  • an effective dosage of TILs is about 1x10 6 , 2x10 6 , 3x10 6 , 4x10 6 , 5x10 6 , 6x10 6 , 7x10 6 , 8x10 6 , 9x10 6 , 1x10 7 , 2x10 7 , 3x10 7 , 4x10 7 , 5x10 7 , 6x10 7 , 7x10 7 , 8x10 7 , 9x10 7 , 1x10 8 , 2x10 8 , 3x10 8 , 4x10 8 , 5x10 8 , 6x10 8 , 7x10 8 , 8x10 8 , 9x10 8 , 1x10 9 , 2x10 9 , 3x10 9 , 4x10 9 , 5x10 9 , 6x10 9 , 7x10 9 , 8x10 9 , 9x10 9 , 1x10 10 , 2x10 9 , 3x10 9 , 4x10 9 , 5x10 9
  • an effective dosage of TILs is in the range of 1x10 6 to 5x10 6 , 5x10 6 to 1x10 7 , 1x10 7 to 5x10 7 , 5x10 7 to 1x10 8 , 1x10 8 to 5x10 8 , 5x10 8 to 1x10 9 , 1x10 9 to 5x10 9 , 5x10 9 to 1x10 10 , 1x10 10 to 5x10 10 , 5x10 10 to 1x10 11 , 5x10 11 to 1x10 12 , 1x10 12 to 5x10 12 , and 5x10 12 to 1x10 13 .
  • an effective dosage of TILs is in the range of about 0.01 mg/kg to about 4.3 mg/kg, about 0.15 mg/kg to about 3.6 mg/kg, about 0.3 mg/kg to about 3.2 mg/kg, about 0.35 mg/kg to about 2.85 mg/kg, about 0.15 mg/kg to about 2.85 mg/kg, about 0.3 mg to about 2.15 mg/kg, about 0.45 mg/kg to about 1.7 mg/kg, about 0.15 mg/kg to about 1.3 mg/kg, about 0.3 mg/kg to about 1.15 mg/kg, about 0.45 mg/kg to about 1 mg/kg, about 0.55 mg/kg to about 0.85 mg/kg, about 0.65 mg/kg to about 0.8 mg/kg, about 0.7 mg/kg to about 0.75 mg/kg, about 0.7 mg/kg to about 2.15 mg/kg, about 0.85 mg/kg to about 2 mg/kg, about 1 mg/kg to about 1.85 mg/kg, about 1.15 mg/kg to about 1.7 mg/kg,
  • an effective dosage of TILs is in the range of about 1 mg to about 500 mg, about 10 mg to about 300 mg, about 20 mg to about 250 mg, about 25 mg to about 200 mg, about 1 mg to about 50 mg, about 5 mg to about 45 mg, about 10 mg to about 40 mg, about 15 mg to about 35 mg, about 20 mg to about 30 mg, about 23 mg to about 28 mg, about 50 mg to about 150 mg, about 60 mg to about 140 mg, about 70 mg to about 130 mg, about 80 mg to about 120 mg, about 90 mg to about 110 mg, or about 95 mg to about 105 mg, about 98 mg to about 102 mg, about 150 mg to about 250 mg, about 160 mg to about 240 mg, about 170 mg to about 230 mg, about 180 mg to about 220 mg, about 190 mg to about 210 mg, about 195 mg to about 205 mg, or about 198 to about 207 mg.
  • An effective amount of the TILs may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, topically, by transplantation, or by inhalation.
  • agents having similar utilities including intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, topically, by transplantation, or by inhalation.
  • Additional exemplary and non-limiting procedures for collection of tumor material, cryopreservation, and TIL manufacture are provided below.
  • the starting material for TIL manufacturing is a disaggregated and cryopreserved cell suspension containing autologous TIL and tumor cells from an eligible patient.
  • An exemplary flow diagram is provided for collection and processing of the tumor starting material.
  • the tumor is surgically resected and then trimmed to remove visibly necrotic tissue, visibly healthy (non-cancerous) tissue, fat tissue, and excess blood.
  • the trimmed tumor weight should be greater than or equal to 2 grams ( ⁇ 2 grams).
  • Tumors weighing over 7 g may be divided into smaller portions and individually disaggregated.
  • Each tumor fragment is placed into an individual sterile bag containing media, collagenase and DNAse. Exemplary reagents are shown in the following table: [00498] The bag is then heat sealed and its contents are disaggregated to generate a homogeneous cell suspension containing tumor and TIL.
  • Disaggregation is performed by a device, such as the Tiss-U-Stor device described herein, which runs a program to deliver a defined number of repeated physical compression events, with a defined compression pressure over a defined duration to ensure enzyme access into the tumor tissue thereby accelerating enzymatic digestion. The number of cycles, pressure, temperature, and duration are recorded for each individual tumor. [00499]
  • the homogenized cell suspension is then aseptically filtered using a 200 ⁇ m filter (Baxter, RMC2159) and the filtrate passed aseptically into the cryopreservation bag.
  • BloodStor 55-5 Biolife Solutions, Bothell, WA is aseptically added to achieve 5% DMSO.
  • the cell suspension is then cryopreserved using the Tiss-U-Stor device with a defined cooling program, and the measured temperature profile is recorded for each individual cell suspension derived from each tumor portion.
  • the cryopreserved cell suspension is stored in vapor-phase of liquid nitrogen.
  • the cryopreserved cell suspension recommended storage condition is ⁇ -130°C.
  • the cell suspension is transported from the clinical site to the GMP cell therapy manufacturing site by a qualified courier service packaged in a container validated to ensure the cryopreserved cell suspension is maintained at ⁇ -130°C.
  • Tiss-u-Stor Resected tumors are evaluated for weight and condition.
  • a CS50N bag is opened, up to about 7g of tumor is added and the bag is then sealed.15 ml of EDM digest medium is added to the bag with 2 ⁇ l gentamicin/amphotericin per ml EDM by syringe via needleless port followed by removal of air from the from the bag into the syringe.
  • the tumor tissue and disaggregation media in the disaggregation bag is placed in the temperature controlled tissue disaggregator. The temperature is increased from ambient temperature to 35 ⁇ C at a rate of 1.5 ⁇ C/min and maintained at 35 ⁇ C for a total of about 45 minutes during which time the disaggregator is active at 240 cycles per minute.
  • the tumor material is filtered through an inline filter into a secondary freezing bag.1.5 ml of Blood stor (DMSO) is injected via a needleless port and air removed. [00505] 2 ml. of the suspension is withdrawn for testing. [00506] For optional cryopreservation, the cryobag is loaded into a freezing cassette and the freezing cassette placed in the Via freeze. The Via freeze is then cooled to -80 ⁇ C, preferably directly from 35 ⁇ C to -80 ⁇ C at a rate of -2 ⁇ C/min. [00507] The frozen cryobag is then transferred to liquid nitrogen storage.
  • DMSO Blood stor
  • T cell medium contains Albumin (human), human Holo Transferrin, and animal origin cholesterol.
  • the source plasma used to manufacture Albumin and Transferrin are sourced from the USA and the donors are tested for adventitious agents.
  • Cholesterol is sourced from sheep wool grease originating in Australia/New Zealand, which complies with USDA regulations prohibiting ruminant original material from countries with reported cases of transmission spongiform encephalopathy (TSE).
  • Fetal Bovine Serum (FBS) is sourced from Australia / New Zealand in compliance with the USDA regulations prohibiting ruminant original material from countries with reported cases of transmission spongiform encephalopathy (TSE).
  • the FBS is tested in compliance with 21 CFR part 113.47, specifically including: bluetongue virus, bovine adenovirus, bovine parvovirus, bovine respiratory syncytial virus, bovine viral diarrhea virus, rabies virus, reovirus, cytopathic agents, haemadsorbing agents.
  • the FBS is heat inactivated at 56°C for 30 minutes and triple 0.1 ⁇ m filtered to provide two orthogonal viral removal steps.
  • Human AB Serum is sourced from Valley Biomedical, an FDA registered establishment (1121958).
  • HBV Hepatitis B surface Antigen
  • HBV Hepatitis B Virus
  • NAT Nucleic acid Amplification Test
  • HIV-1 HIV-1 NAT
  • HCV anti-Hepatitis C Virus
  • HCV NAT HCV NAT
  • syphilis a test for syphilis by FDA approved methods.
  • the serum is heat inactivated at 56°C for 30 minutes and 0.1 ⁇ m filtered.
  • Irradiated Buffy Coat sourcing, preparation, shipment and storage The Scottish National Blood Transfusion Service (SNBTS) screens donors, collects the blood component, prepares and irradiates buffy coats.
  • SNBTS National Blood Transfusion Service
  • the SNBTS is licensed by the United Kingdom’s Human Tissue Authority (license number 11018) in accordance with the Blood, Safety and Quality Regulations (2005) to procure, process, test, store and distribute blood, blood components and tissues.
  • Healthy donor screening meets or exceeds the requirements described in the United States Code of Federal Regulations (CFR) Title 21 Part 1271.75 with the exception that donors live in the United Kingdom. While this presents a theoretical risk of sporadic Creutzfeldt-Jakob Disease (sCJD) or variant Creutzfeldt-Jakob Disease (vCJD), the United Kingdom has a robust national surveillance program.
  • the licensed blood establishment prepares clinical grade irradiated buffy coats which are suitable to treat patients with severe neutropenia.
  • blood is centrifuged to form three layers: the red blood cell layer, the buffy coat layer and the plasma layer.
  • Buffy coats from 10 donors are irradiated with 25 to 50 Gy irradiation to arrest cell growth.
  • the clinical grade irradiated buffy coats are prepared and shipped to the GMP manufacturing facility by overnight courier using a controlled temperature shipper including a temperature monitor. The shipment occurs one day before use in the manufacturing process.
  • the buffy coats are held at 15 – 30°C until use in manufacturing.
  • Irradiated Feeder Cell Preparation Irradiated Feeder Cell Preparation.
  • Buffy coats from up to ten unique donors are pooled, then centrifuged by Ficoll gradient density centrifugation to harvest peripheral blood mononuclear cells (PBMCs).
  • PBMCs peripheral blood mononuclear cells
  • Approximately 4 x 10 9 viable white blood cells are resuspended in TCM supplemented with approximately 8% human AB serum, 3000 IU/mL IL-2 and 30 ng OKT-3 in a closed static cell culture bag.
  • the PBMC are released per specification.
  • the PBMC are also tested for sterility and mycoplasma.
  • a sample of the formulated feeder cell, including media, IL-2 and OKT3 is removed. This sample is incubated and analyzed on days 13, 17 and 18 to confirm that the feeder cells do not expand.
  • Albumin human
  • HSA Human Serum Albumin
  • the cell suspension is seeded at approximately 0.25 x 10 6 to 0.75 x 10 6 viable cells/mL into TCM supplemented with 10% FBS, 0.25 ⁇ g/mL Amphotericin B with 10 ⁇ g/mL Gentamicin (Life Technologies, Grand Island, NY), and interleukin-2 (IL-2; aldesluekin) 3000 IU/mL (Clinigen, Love, Germany) and cultured in standard cell culture conditions (37°C, 5% CO 2 ). [00523] On day 5, half of the media is removed and replaced with TCM supplemented with 10% FBS, 0.50 ⁇ g/mL Amphotericin B, 20 ⁇ g/mL Gentamicin and 6000 IU/mL IL-2.
  • the TIL outgrowth culture is diluted with three times the volume to maintain approximately 0.1 x 10 6 to 2.0 x 10 6 viable cells/mL. If the cell concentration is ⁇ 1.5 x 10 6 viable cells/mL, half of the media is replaced. In either option, the media is TCM supplemented with 10% FBS, 0.50 ⁇ g/mL Amphotericin B, 20 ⁇ g/mL Gentamicin and 6000 IU/mL IL-2.
  • the TIL outgrowth culture is diluted with three times the volume to maintain approximately 0.1 x 10 6 to 2.0 x 10 6 viable cells/mL. If the cell concentration is ⁇ 1.5 x 10 6 viable cells/mL, half of the media is replaced. In either option, the media added is TCM supplemented with 10% FBS, 0.50 ⁇ g/mL Amphotericin B, 20 ⁇ g/mL Gentamicin and 6000 IU/mL IL-2.
  • TILs are activated using an anti-CD3 antibody (OKT3) to provide a CD3 specific stimulation when bound to the FC receptor of irradiated feeder cells from allogeneic peripheral blood mononuclear cells (PBMCs).
  • PBMCs peripheral blood mononuclear cells
  • the feeders provide a natural source of additional co- stimulation to support the added anti-CD3 (OKT-3).
  • 1 to 20 x 10 6 viable T cells from the TIL outgrowth Step 2 are added to 2.0 to 4.0 x 10 9 viable irradiated feeder cells (Section 8.1.4.4) using approximately 30 ⁇ 10 ng/mL OKT3, approximately 8% Human AB Serum and 3000 ⁇ 1000 IU/mL IL-2.
  • the TIL activation culture is incubated for 6 days at standard cell culture conditions.
  • the activated TILs continue expansion by aseptically adding the activated TIL cell suspension into a bioreactor containing T cell media supplemented with approximately 8% Human AB Serum and 3000 IU/mL IL-2.
  • the TIL expansion is provided a continuous feed of T cell media supplemented with 3000 IU/mL IL-2 until harvest.
  • TILs are harvested by washing the cells using SEFIATM. The cells are concentrated by centrifugation then washed 2-4 times using phosphate buffered saline (PBS) supplemented with 1% human serum albumin (HSA).
  • PBS phosphate buffered saline
  • HSA human serum albumin
  • the cells are then resuspended in PBS + 1% HSA to approximately 50-60 mL.
  • the washed and concentrated cells are aseptically transferred into a cryobag and a portion removed for lot release testing and retained samples.
  • drug product DP
  • the TILs are then cooled to 2-8°C and formulated, e.g.1:1 with cryoprotectant containing 16% HSA and 20% DMSO, to achieve a formulated product of ⁇ 5 x 10 9 viable cells suspended in approximately 10% DMSO and 8.5% HSA in PBS. A portion is removed for lot release testing and retained samples.
  • the cryobag is cooled to -80°C.
  • the following table shows examples of TIL manufacture process variations.
  • CryoStor based DMSO was then compared with Bloodstor 55-5, a DMSO based cryopreservative, and the higher concentration BloodStor product was selected since it was more concentrated thus allowing for a smaller cryobag.
  • Cryopreservation was then compared following a protocol that either held the material at 4°C for 10 minutes, then decreased the temperature at a rate of -1°C/min or decreased from 35°C to -80°C directly at a rate of -2°C/min. Post-thaw viability was similar between the two cryopreservation protocols used. [00536] During cooling, ice nucleation releases heat.
  • TIL potency analysis comprises evaluation of analytes characteristic of TIL activation, including but not limited to indicators of mechanism of action. Exemplary non-limiting mechanisms of action include tumor cell killing, cytokine secretion, proliferation, persistence, and properties indicative of the mechanisms.
  • Analysis can comprise enumeration of T cells and target cells, for example by flow cytometry, percent killing which can be observed by fluorescence or luminescence in plate-based or flow cytometry or other methods such as cartridge-based methods, characterization of individual cells to determine expression of markers including but not limited to expression of cytokines, cell surface markers, expression levels of genes that are induced in activated T-cells, including and not limited to reporter molecules engineered to be expressed under activating conditions, or other hallmarks of T cell activation.
  • Measures of TIL potency include TIL cellular composition and phenotype, such as but not limited to numbers and proportions of CD8 + cells, memory phenotype including without limitation effector memory and central memory, measures of cytotoxicity using various cell lines, cytotoxicity using patient specific tumor, expression of cytokines or a panels of cytokines, and cell proliferation and persistence.
  • a bioassay for quantification of TIL potency comprises multiparameter or polychromatic intracellular flow cytometry. Intracellular flow cytometry is particularly advantageous for assessment of T cell specific parameters on an individual cell basis and ensures accurate determination even in heterogeneous cell populations.
  • Multiparameter flow cytometry permits simultaneous detection or two or more components, which can include two or more cytokines, combined with high throughput.
  • Cartridge-based analytical technologies are also contemplated, such as but not limited to the cartridges manufactured by Chemometec chemometec.com/products/nucleocounter-nc-200-automated-cell-counter/ or Accellix accellix.com/technology/).
  • intracellular assays described herein are cell and cell type specific.
  • individual cytokine producing cells can be identified and enriched if desired.
  • the intracellular methods avoid cytotoxicity and effects of the methods on the assayed cells are reversible.
  • a TIL population is cocultured with cells engineered to activate T cells via CD3, the signaling component of the T-cell receptor (TCR).
  • a modified TIL population is cocultured with cells engineered to activate T cells as well as engage and activate a costimulatory receptor.
  • a convenient example of activating cells comprises K562 cells engineered to express a binding protein or antibody or antigen binding fragment thereof that binds to and activates the TCR.
  • the antibody comprises OKT3.
  • the antigen binding fragment comprises a single- chain variable fragment (scFv) from OKT3. Co-culture of ITIL-168 DP with stimulatory K562- OKT3 cells allows for T cell activation via TCR.
  • a negative control for example, without limitation, nontransduced clonal K562 cells, K562-NT.
  • the ratio of TILs to activating cells can be adjusted as needed.
  • the ration of TILs to activating cells is from 10:1 to 1:10.
  • Non-limiting examples include coculture of TILs with stimulatory K562-OKT3 cells in ratios such as 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, or 1:10.
  • the potency analysis method is used to determine potency of a TIL population cocultured with a “standard” cell type.
  • a “standard” cell type is a K562 cell engineered to express a ligand, such as but not limited to an antibody or antigen binding fragment thereof, such as an OKT3 antibody or antigen binding fragment thereof that binds to and activates a T-cell receptor on the TIL.
  • the potency analysis method is used to determine potency of a TIL population cocultured with tumor cells or cells engineered to express a tumor associated antigen.
  • the potency analysis method is used to determine potency of a TIL population cocultured with tumor cells from the same patient as the source of the TILs.
  • Potency can be reported as: [00546] [00547] AVG indicates the average potency determined by assay in triplicate. [00548] Potency may be calculated as the frequency of all viable CD2+ cells that are positive for one or more of CD137, CD107a, TNF- ⁇ and IFN- ⁇ , preferably CD107a and IFN- ⁇ . [00549] The potency analysis method can be applied at any stage of TIL manufacture. In certain embodiments, TIL manufacture comprises monitoring potency of the TIL manufacture from one culture step to the next. In certain embodiments, TIL manufacture comprising monitoring TIL potency throughout the TIL manufacture.
  • TIL manufacture may comprise measuring TIL potency to confirm or adjust the number of cells from a culture step used to seed a subsequent culture step.
  • TIL quality attributes include potency, viability, cell count and purity.
  • TIL manufacture comprises measuring the potency of TILs processed from a tumor.
  • TIL manufacture comprises measuring the potency of TILs from a pre-REP expansion culture.
  • TIL manufacture comprises measuring the potency of TILs during a pre- expansion REP.
  • TIL manufacture comprises measuring the potency of TILs at the end of a REP.
  • TIL manufacture comprises measuring the potency of TILs at the end of a second REP.
  • TIL manufacture comprises measuring TIL potency during REP, for example mid-REP. In certain embodiments, TIL manufacture comprises measuring TIL potency prior to cryopreservation and/or after thawing of a cryopreserved cells. In certain embodiments, TIL manufacture comprises measuring the potency of TIL drug product (TIL DP).
  • TIL DP TIL drug product
  • the potency testing at any stage of TIL manufacture may further comprise enrichment or isolation of more potent TILs, for example the top 40%, or the top 50%, or the top 60%, or the top 70%, or the top 80%, or the top 90% of the TILs. In certain embodiments, the enrichment or isolation comprises separation of TILs from inhibitory cells.
  • Non-limiting examples of analytes indicative of TIL activation and potency include IFN- ⁇ , CD107a, CD137 (4-1BB).
  • Other markers indicative or TIL activation or beneficial anti- tumor characteristics include, but are not limited to, IL-1beta, IL-2, IL-4, IL-6, IL-8, IL-10, IL- 12p70, granzyme A/B, perforin, caspase 3 and other chemokine markers.
  • CD107a aka lysosomal-associated membrane protein-1 or LAMP-1
  • LAMP-1 is a marker of degranulation of NK cells and CD8+ T-cells.
  • IFN- ⁇ is a pleiotropic cytokine with antiviral, antitumor, and immunomodulatory functions. IFN- ⁇ has been shown to increase the motility of antigen-specific CD8+ T-cells to the antigen-expressing (target) cells and enhance the killing of target cells. IFN- ⁇ concentration in the tumor microenvironments has been linked to better immune checkpoint blockade efficacy. comprises an indicator of T-cell activation. In an embodiment, there is an analysis of IFN- ⁇ and CD107a.
  • CD137 (4-1BB) is a member of the TNFR family and functions as a costimulatory molecule to promote proliferation and survival of activated T cells. Expression of CD137 on T cells is found in T cells that have recently been activated by TCR engagement.
  • TNF is a proinflammatory cytokine produced by activated T cells and indicative of robust antitumor activity.
  • Potency due to autocrine stimulation of TIL by cytokines or potency due to paracrine stimulation of anti-tumor effects mediated by other cells in the tumor microenvironment is detectable in Applicant’s method, although if there is high background in the T cell +K562 parenteral, Applicants have not yet observed it. Potency markers indicating persistence may be detected in a cell proliferation assay.
  • analytes that distinguish cell subsets are examined.
  • Non limiting examples are CD62L and CD45RO which in different combination can distinguish among effector cells (CD62L-, CD45RO-), effector memory cells (CD62L-, CD45RO+), central memory cells (CD62L+, CD45RO+) and stem cell memory cells (CD62L+, CD45RO-).
  • Cryopreserved cells are thawed typically provided a recovery period before potency testing of about 1-2 hr, 2-4 hr, 4-6 hr., 6-8 hr., 8-10 hr., 10-12 hr, or overnight (up to 24 hr), before testing.
  • thawed TIL are then mixed with a population of stimulatory cells (e.g. K562 or other non T cell line engineered with OKT3 scFv fragment) capable of engaging and stimulating the TILs via the TCR.
  • a population of stimulatory cells e.g. K562 or other non T cell line engineered with OKT3 scFv fragment
  • the number of cells post recovery going into the assay may be from about 1 x 10 5 , 2 x 10 5 , 3 x 10 5 , 4 x 10 5 , 5 x 10 5 , 6 x 10 5 , 7 x 10 5 , 8 x 10 5 , 9 x 10 5 , 1 x 10 6 , 2 x 10 6 , 3 x 10 6 , 4 x 10 6 , 5 x 10 6 , 6 x 10 6 , 7 x 10 6 , 8 x 10 6 , 9 x 10 7 , 1 x 10 7 , 2 x 10 7 , 3 x 10 7 , 4 x 10 7 , 5 x 10 7 , 6 x 10 7 , 7 x 10 7 , 8 x 10 7 , 9 x 10 7 , 1 x 10 8 , 2 x 10 8 , 3 x 10 8 , 4 x 10 8 , 5 x 10 5 , 6 x 10 7 , 7 x 10 7 , 8
  • the mixed cell composition may be incubated for about 8-10 hr., 10-12 hr, 12-14 hr., 14-16 hr., 16-18 hr., 18-20 hr., 20-22 hr., 22-24 hr., 24-26 hr., 26-28 hr., 28-30 hr., 30-32 hr., 32-34 hr. or 34-36 hr. with an inhibitor of protein transport inhibitors (e.g.
  • Brefeldin A and Monensin which may be at a concentration from about 10X, 20X, 30X, 40X, 50X, 60X, 70X, 80X, 90X, 100X, 200X, 300X, 400X, 500X, 600X, 700X, 800X, 900X, 1000X, 200X, 3000X, 4000X, 5000X, 6000X, 7000X, 8000X, 9000X or 10,000X) and optionally one or more reagents to monitor pertinent markers that identify degranulating cells post activation (e.g., anti-CD107a).
  • CD107a may be added to mark T cell degranulation prior to analyzing cell count, viability and/or cell purity, which may be determined by flow cytometry or a cartridge based method.
  • the incubation period may be about 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 13, 13.5, 14, 14.5, 15, 15.5, 16, 16.5, 17, 17.5, 18, 18.5, 19, 19.5, 20, 20.5, 21, 21.5, 22, 22.5, 23, 23.5, 24, 24.5, 25, 25.5, 26, 26.2, 27, 27.5, 28, 28.5, 29, 29.5, 30, 30.5, 31, 31.5, 32, 32.5, 33, 33.5, 34, 34.5, 35, 35.5, or 36 hours
  • the cell culture is treated to distinguish live and dead cells and the cells are permeabilized and fixed.
  • the concentration of the fixative and the time of fixing may be optimized and is within the purview of one of skill in the art.
  • the treatment may be for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 30, 31, 32, 33, 34 or 35 minutes.
  • Permeabilized cells are stained for intracellular and extracellular markers and the markers measured by flow cytometry or a cartridge based method.
  • the antibody cocktail used to stain the cells of potency markers e.g., CD2, TNFa, IFNg, CD137
  • PE PCP-eF710, APC, APC-Cy7, BV711, eFLOUR506, GFP etc.
  • concentration volume 0.5, 1.0, 1.2, 1.25, 1.3, 1.5, 1.75, 1.8, 1.9, 2.0, 2.5, 3, 3.5, 4 etc.
  • incubation time 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60 mins, etc.
  • a stain may be utilized to distinguish between live and dead cells.
  • LEF loss-of-function
  • Lentiviral transfer plasmids and LVV preps for selected candidates [00558]
  • protein sequences of the common or most abundant isoform in T cells were obtained from UniProt, reverse translated, and codon optimized before gene synthesis and molecular cloning into pSF.Lenti.MND.P2A.tCD34.KanR lentiviral vector (pIB1123).
  • pIB1146-1147 For loss of function genes (pIB1146-1147), one or several dominant negative mutants were first identified from published literatures.
  • the resultant LVV prep was titrated by transduction in serial dilutions on Jurkat cells followed by flow cytometric analysis of surface tCD34 expression. Master regulators [00560] Single-cell RNA-Seq was performed on TIL product samples from patients in MS Specials study. Unsupervised clustering analysis was performed on the resulting single-cell gene expression profiles which grouped cells into subpopulations with distinct transcriptional signatures. The fractional abundance of each cell subpopulation in each TIL product sample was calculated and correlated to clinical response which identified a list of cell subpopulations being correlated with response.
  • cell subpopulations were annotated based on prior knowledge which identified several proliferating cell subpopulations and a non-proliferating highly differentiated cell subpopulation.
  • Gene regulatory network analysis was performed on each single-cell gene expression profile to score the activity level of each transcription factor in each cell. The activity score of each transcription factor is averaged within each cell subpopulation and standardized across cell subpopulations. The top 20 most active transcription factors of each cell subpopulation were identified as the list of key transcription factors responsible for maintaining the transcriptional signature of that cell subpopulation.
  • a list of candidate genes to upregulate for viTIL is generated by combining the lists of key transcription factors of the cell subpopulations that were positively correlated with response and the proliferating cell subpopulations.
  • a list of candidate genes to downregulate for viTIL is generated by combining the lists of key transcription factors of the cell subpopulations that were negatively correlated with response and the non-proliferating highly differentiated cell subpopulations.
  • Repeated stimulation assay on healthy donor T cells to model T cell differentiation in vitro Peripheral blood mononuclear cells (PBMCs) and isolated T cells from two healthy donors were thawed and stimulated with various stimulation methods under different schemes.
  • PBMCs Peripheral blood mononuclear cells
  • PBMCs or isolated T cells were thawed and seeded at 1x10 6 cells with stimulation. Cells were then counted, washed, diluted to 1x10 6 cells/mL in fresh complete TCM, and repeatedly stimulated every 3 days for a total of 10 rounds of stimulation over 34 days.
  • PBMCs or isolated T cells were thawed and seeded at 1x10 6 cells with 1 round of stimulation. Cells were then counted, washed, and diluted to 1x10 6 cells/mL in fresh complete TCM every 3 days without further stimulation for a total of 34 days.
  • PBMCs or isolated T cells were thawed and seeded at 1x10 6 cells with 2 rounds of stimulation. Cells were then counted, washed, and diluted to 1x10 6 cells/mL in fresh complete TCM every 3 days without further stimulation for a total of 34 days.
  • a fraction of cells from all groups were collected for staining and flow cytometric analysis of surface markers CD3, CD4, CD8, CD69, CD25, 4-1BB, PD-1, TIM-3, LAG-3, CTLA-4, CD39, CD103, CD45RA, CCR7, CD45RO, CD62L, CD27, CD28, OX40, and CD127.
  • T cell serial stimulation assay Screening of candidates in healthy donor T cell serial stimulation assay [00566] Peripheral blood T cells isolated from two healthy donors were thawed and stimulated with Dynabeads Human T-Activator CD3/CD28 at 1:1 ratio in complete TCM for 3 days. On day 1 after stimulation, T cells were transduced with candidate LVV at MOI of 5. From day 3, T cells in “Repeat Stim” group were counted, washed, diluted to 1x10 6 cells/mL in fresh complete TCM, and repeatedly stimulated with Dynabeads at 1:1 ratio every 3 days for a total of 10 rounds of stimulation over 34 days.
  • T cells in “Round 1 Rest” group were counted, washed, and diluted to 1x10 6 cells/mL in fresh complete TCM every 3 days without further stimulation for a total of 34 days as control.
  • a fraction of T cells from both groups were collected for staining and flow cytometric analysis of surface tCD34 as a surrogate of transgene expression, along with surface markers CD3, CD4, CD8, CD69, CD25, 4-1BB, PD-1, TIM-3, LAG-3, CTLA-4, CD39, CD103, CD45RA, CCR7, CD45RO, CD62L, CD27, CD28, OX40, and CD127.
  • Total viable cell count, tCD34+%, and tCD34+ viable cell count were used to select positive hits.
  • Surface expression of 4-1BB, OX40, PD-1, TIM-3, LAG-3, and CTLA-4 in CD4+ and CD8+ T cell populations were compared between positive hits in “Repeat Stim” group.
  • Phenotypic subsets defined by CD45RA and CCR7 were also compared between positive hits in “Repeat Stim” group.
  • Screening of selected candidates during ex vivo TIL expansion [00568] Tumor digest from ovarian cancer and renal cell carcinoma were thawed and plated in 12-well plates at 1x10 6 cells/mL in complete TCM with 3000 IU/mL IL-2, incubated for 2 days.
  • GAF gain of function
  • LEF loss of function
  • protein sequences of the common or most abundant isoform in T cells were obtained from UniProt, reverse translated, and codon optimized before gene synthesis and molecular cloning into pSF.Lenti.MND.P2A.tCD34.KanR lentiviral vector (pIB1123).
  • pIB1146-1147 For loss of function genes (pIB1146-1147), one or several dominant negative mutants were identified from published literatures.
  • the wild-type and mutant protein sequences were then reverse translated, and codon optimized before gene synthesis and molecular cloning into pSF.Lenti.MND.P2A.tCD34.KanR lentiviral vector (pIB1123).
  • pIB1123 was used as an empty vector control.
  • TIL tumor-infiltrating lymphocyte
  • TCR T-cell receptor
  • TIL therapy infusion product composition TCR repertoire, and mediators of cell-cell interaction were characterized in a translational subanalysis of the compassionate use clinical series.
  • TCR repertoire clonality and diversity of TIL products were assessed using multiple metrics, including Gini coefficient, using the Immunarch package.
  • 9 TIL products were assessed using RNA-based bulk TCR sequencing and paired single-cell RNA and TCR sequencing techniques.
  • Putative antigen/tumor-reactive clones were inferred using GLIPH2 (grouping of lymphocyte interactions by paratope hotspots 2) algorithm 10 and publicly available TCR annotation databases including VDJdb public database.
  • Unsupervised clustering of cells and differential gene expression analysis was performed using the Seurat package.
  • Gene set over-representation analysis was performed using the clusterProfiler package.
  • 13 Gene regulatory network analysis was performed using the SCENIC package.
  • 14 Cell-cell interaction analysis was performed using the CellChat package.
  • TIL product samples Multiple subpopulations were previously undescribed in TIL product samples. Certain T-cell populations were found at frequencies that differed between products administered to patients who developed responses compared to those who did not achieve a response. See Figures 23A-23C. The frequency of C7 (MX1+OAS1+; Figure 23A), C9 (BBC3+CHAC1+; Figure 23B), or C7 and C9 (Figure 23C) T-cell subpopulations in TIL product samples differed between responders and non-responder. Low abundance of C7 TIL or C9 TIL subpopulations was associated with response ( Figures 23A and 23B). Low abundance of the combined C7 and C9 TIL subpopulations was also associated with response ( Figure 23C). References [00577] 1.
  • Raw FASTQ files were processed using the Cell Ranger software 7 with default parameters.
  • the resulting counts matrix were then analyzed using the Seurat 9 R package.
  • the counts matrix was first filtered based on the total number of genes and the fraction of mitochondria genes detected in each cell. Cells with a fraction of mitochondria gene detected being greater than 10% or with a total number of genes detected being less than 800 were excluded from further analysis.
  • the top 2000 highly variable genes in each TIL product sample were then identified and used as the anchor genes for sample integration to remove any technical batch effect.
  • Cell cycle scores of each cell were calculated and regressed out from the integrated counts matrix, followed by unsupervised clustering which partitions all cells into several cell subpopulations with distinct transcriptional signatures.
  • PBMC and isolated T cells from two donors were thawed and stimulated according to different schedules ( Figure 1) and various reagents. After the first round of stimulation (3 days), cells in “Repeat Stim” group were counted, washed, diluted to 1x10 6 cells/mL, and repeatedly stimulated every 3 days for a total of 10 rounds in 34 days. Cells in “Round 1 Rest” group were counted, washed, and diluted to 1x10 6 cells/mL every 3 days without further stimulation.
  • Stimulating reagents were OKT3+CD28 (GMP) antibodies; OKT3+CD28 (RUO) antibodies; OKT3+CD28 -> OKT3; OKT3 -> OKT3; Dynabeads Human T-Activator CD3/CD28 (DB); and TransAct (TA).
  • GMP a Good Manufacturing Practices-grade soluble CD28 antibody for ex vivo T cell activation
  • RUO a Research Use Only-grade CD28 antibody for ex vivo T cell activation.
  • T cells were then stimulated and LVV-transfected according to the schedule depicted in Figure 4. Isolated T cells from two healthy donors were thawed and stimulated using Dynabeads for 3 days.
  • T cells in “Repeat Stim” group were counted, washed, diluted to 1x10 6 cells/mL, and repeatedly stimulated every 3 days for a total of 10 rounds in 34 days.
  • T cells in “Round 1 Rest” group were counted, washed, and diluted to 1x10 6 cells/mL every 3 days without stimulation.
  • a fraction of T cells was stained and analyzed by flow cytometry for surface tCD34 expression along with markers of activation, exhaustion, and differentiation phenotypes.
  • Expansion curves were determined under repeat stimulation conditions for loss-of- function (LOF) clones pIB1146 - pBI1148 ( Figure 5).
  • pIB1123 is an empty vector control. Candidates identified as enabling enhanced proliferation were pIB1146 – dnFas (mutDD) and pIB1147 – dnFas (delDD).
  • Expansion curves were determined under stimulation with rest conditions for the same loss-of-function (LOF) clones pIB1146 - pBI1148 ( Figure 6).
  • pIB1123 is an empty vector control. Candidates identified as enabling enhanced proliferation included pIB1146 – dnFas (mutDD).
  • tCD34+ cells were enriched in T cells transduced with pIB1146 – dnFas (mutDD).
  • Figure 7A shows percent of tCD34+ cells measured by flow cytometry.
  • Figure 7B shows viable tCD34+ cell count from total viable cells.
  • T cell activation / exhaustion markers were measured in CD4+ and CD8+ populations of tCD34+ T cells ( Figures 8A-8F). T cell exhaustion markers were also measured in the CD4+ and CD8+ populations ( Figures 9A-9F).
  • FIGS. 13A-13D show fold expansion in outgrown (Figure 13A) and REP (Figure 13B) and CD3+ fold expansion in outgrowth (Figure 13C) and REP ( Figure 13D).
  • Enrichment of transduced populations in TILs from renal cell carcinoma during REP was determined. The proportion of LVV transfected tCD34+ cells during REP was compared on day 13 and day 25 and compared to an empty vector (EV) control which transfected only tCD34+ ( Figure 14A).
  • Figures 19A-19B show the proportions of CD4+ (Figure 19A) or CD8+ ( Figure 19B) TILs expressing CD127 at the end of outgrowth and REP.
  • Figures 20A-20B show the proportion of CD4+ ( Figure 20A) or CD8+ ( Figure 20B) TILs expressing CD27 at the end of outgrowth and REP.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Wood Science & Technology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • General Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Oncology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Provided herein are methods for preparing a therapeutic population of tumor infiltrating lymphocytes (TILs), populations of TILs produced by the methods, and methods of using the TILs to treat cancer in a subject. The methods comprise treating a first population of TILs with one or more compounds to improve T-cell fitness.

Description

TUMOR-INFILTRATING LYMPHOCYTE (TIL) COMPOSITIONS AND USES THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of US Application 63/331,697, filed April 15, 2022, and US Application 63/341,770, filed May 13, 2022, each of which is herein incorporated by reference in its entirety for all purposes.
BACKGROUND
[0002] Tumor infiltrating lymphocytes (TILs) gradually lose functionality and proliferation potential during in vitro or in vivo antigen stimulation and expansion, a process known as T cell differentiation or exhaustion. Prior to excision, TILs in tumor tissue may be already differentiated and reaching exhaustion as a result of chronic tumor antigen stimulation. During manufacture, TILs are massively expanded in vitro, which leads to further differentiation and exhaustion. Thus viability and recovery of a desired product from tissue may be affected by the conditions during tissue collection, disaggregation, and harvesting of cells, and expansion of cells. There is a need to provide TIL preparations and methods of manufacture that provide improved functionality.
SUMMARY
[0003] The invention relates to rejuvenation of TILs. Tumor-reactive T cells acquired from tumor microenvironments may already be exhausted or losing their functionality on their way to exhaustion. Further, TILs prepared and selected in vitro for activity, such as high levels of IFN-y and efficient targeting of target cells may be less effective at causing regression of targeted tumors when adoptively transferred to a patient than TILs at earlier other stages of differentiation.. To overcome these obstacles, the invention provides methods and compositions for producing TIL populations exhibiting improved persistence and functionality. In one sense, the TILs can be considered “rejuvenated,” i.e., demonstrating high levels of in vitro and in vivo activity and less subject to exhaustion. In another sense, the TILs can be considered “prodigious,” i.e., exhibiting prolonged high levels of in vivo activity when administered to a subject. [0004] The invention provides TIL compositions having improved persistence and functionality. The invention provides methods and agents for generating such improved TIL compositions and methods of treatment that employ the improved TIL compositions. [0005] Without being bound by theory, T cells are typically divided into discreet subsets based on definitions that reflect their roles in immunity. Features of T cell immune responses include clonal expansion, contraction, exhaustion, and memory formation. Expanded T cell populations are not homogeneous but consist of short lived effector cells and a smaller population of memory precursors. The invention provides improved TIL populations by promoting expansion and persistence and reducing exhaustion of tumor-reactive T cells. [0006] In one aspect, provided are methods of preparing a therapeutic population of tumor infiltrating lymphocytes (TILs). Some such methods comprise treating a first population of TILs with one or more compounds to improve T-cell fitness. Some such methods comprise treating the first population of TILs with the one or more compounds multiple times. [0007] In some such methods, the one or more compounds comprise on or more (any combination thereof) or all of the following: (1) a FAS/FASLG inhibitory agent; (2) a TGFβ/TGFβR1 inhibitory agent; (3) an IRF7 inhibitory agent; (4) a POLR3A inhibitory agent; (5) an ETV7 inhibitory agent; (6) an ETV3 inhibitory agent; (7) an ASH2L inhibitory agent; (8) a PML inhibitory agent; (9) a STAT2 inhibitory agent; (10) a SPI1 inhibitory agent; (11) an IRF9 inhibitory agent; (12) a STAT1 inhibitory agent; (13) an IRF4 inhibitory agent; (14) a JDP2 inhibitory agent; (15) a ZNF337 inhibitory agent; (16) an ETV2 inhibitory agent; (17) an ETV3L inhibitory agent; (18) a SOX18 inhibitory agent; (19) a CEBPG inhibitory agent; (20) a CREB3L4 inhibitory agent; (21) a CEBPB inhibitory agent; (22) a FOXD1 inhibitory agent; (23) an EOMES inhibitory agent; and (24) a ZNF683 inhibitory agent. [0008] In some such methods, the one or more compounds comprise on or more or all of the following: (i) a FAS/FASLG inhibitory agent; (ii) a TGFβ/TGFβR1 inhibitory agent; (iii) an IRF7 inhibitory agent; and (iv) a POLR3A inhibitory agent. [0009] In some such methods, the treating decreases expression or activity of FAS or FASLG. In some such methods, the treating transiently decreases expression or activity of FAS or FASLG. In some such methods, the treating permanently decreases expression or activity of FAS or FASLG. [0010] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative FAS mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative FAS mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative FAS mutant. In some such methods, the dominant negative FAS mutant comprises a mutated FADD binding site, optionally wherein the dominant negative FAS mutant is FAS_D244V. In some such methods, the dominant negative FAS mutant comprises a deleted DD domain, optionally wherein the dominant negative FAS mutant is FAS_del230-314. In some such methods, the one or more compounds comprise an anti-FAS or anti-FASLG antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to a FAS or FASLG messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding FAS or FASLG. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FAS or FASLG. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule FAS/FASLG inhibitor. [0011] In some such methods, the treating decreases expression or activity of TGFβ1 or TGFβR1. In some such methods, the treating transiently decreases expression or activity of TGFβ1 or TGFβR1. In some such methods, the treating permanently decreases expression or activity of TGFβ1 or TGFβR1. [0012] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative TGFβR1 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative TGFβR1 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative TGFβR1 mutant. In some such methods, the one or more compounds comprise an anti-TGFβR1 or TGFβ1 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to a TGFβR1 or TGFβ1 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding TGFβR1 or TGFβ1. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding TGFβR1 or TGFβ1. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule TGFβR1 inhibitor, optionally wherein the small molecule 1 inhibitor is SB431542. [0013] In some such methods, the treating decreases expression or activity of IRF7. In some such methods, the treating transiently decreases expression or activity of IRF7. In some such methods, the treating permanently decreases expression or activity of IRF7. [0014] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative IRF7 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative IRF7 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative IRF7 mutant. In some such methods, the one or more compounds comprise an anti-IRF7 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF7 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF7. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF7. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule IRF7 inhibitor. [0015] In some such methods, the treating decreases expression or activity of POLR3A. In some such methods, the treating transiently decreases expression or activity of POLR3A. In some such methods, the treating permanently decreases expression or activity of POLR3A. [0016] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative POLR3A mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative POLR3A mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative POLR3A mutant. In some such methods, the one or more compounds comprise an anti-POLR3A antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to a POLR3A messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding POLR3A. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding POLR3A. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule POLR3A inhibitor. [0017] In some such methods, the treating decreases expression or activity of ETV7. In some such methods, the treating transiently decreases expression or activity of ETV7. In some such methods, the treating permanently decreases expression or activity of ETV7. [0018] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative ETV7 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative ETV7 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative ETV7 mutant. In some such methods, the one or more compounds comprise an anti-ETV7 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV7 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV7. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV7. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule ETV7 inhibitor. [0019] In some such methods, the treating decreases expression or activity of ETV3. In some such methods, the treating transiently decreases expression or activity of ETV3. In some such methods, the treating permanently decreases expression or activity of ETV3. [0020] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative ETV3 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative ETV3 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative ETV3 mutant. In some such methods, the one or more compounds comprise an anti-ETV3 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV3 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV3. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule ETV3 inhibitor. [0021] In some such methods, the treating decreases expression or activity of ASH2L. In some such methods, the treating transiently decreases expression or activity of ASH2L. In some such methods, the treating permanently decreases expression or activity of ASH2L. [0022] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative ASH2L mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative ASH2L mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative ASH2L mutant. In some such methods, the one or more compounds comprise an anti-ASH2L antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ASH2L messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ASH2L. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ASH2L. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule ASH2L inhibitor. [0023] In some such methods, the treating decreases expression or activity of PML. In some such methods, the treating transiently decreases expression or activity of PML. In some such methods, the treating permanently decreases expression or activity of PML. [0024] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative PML mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative PML mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative PML mutant. In some such methods, the one or more compounds comprise an anti-PML antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an PML messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding PML. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding PML. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule PML inhibitor. [0025] In some such methods, the treating decreases expression or activity of STAT2. In some such methods, the treating transiently decreases expression or activity of STAT2. In some such methods, the treating permanently decreases expression or activity of STAT2. [0026] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative STAT2 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative STAT2 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative STAT2 mutant. In some such methods, the one or more compounds comprise an anti-STAT2 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an STAT2 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding STAT2. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT2. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule STAT2 inhibitor. [0027] In some such methods, the treating decreases expression or activity of SPI1. In some such methods, the treating transiently decreases expression or activity of SPI1. In some such methods, the treating permanently decreases expression or activity of SPI1. [0028] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative SPI1 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative SPI1 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative SPI1 mutant. In some such methods, the one or more compounds comprise an anti-SPI1 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an SPI1 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding SPI1. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SPI1. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule SPI1 inhibitor. [0029] In some such methods, the treating decreases expression or activity of IRF9. In some such methods, the treating transiently decreases expression or activity of IRF9. In some such methods, the treating permanently decreases expression or activity of IRF9. [0030] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative IRF9 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative IRF9 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative IRF9 mutant. In some such methods, the one or more compounds comprise an anti-IRF9 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF9 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF9. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF9. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule IRF9 inhibitor. [0031] In some such methods, the treating decreases expression or activity of STAT1. In some such methods, the treating transiently decreases expression or activity of STAT1. In some such methods, the treating permanently decreases expression or activity of STAT1. [0032] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative STAT1 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative STAT1 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative STAT1 mutant. In some such methods, the one or more compounds comprise an anti-STAT1 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an STAT1 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding STAT1. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT1. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule STAT1 inhibitor. [0033] In some such methods, the treating decreases expression or activity of IRF4. In some such methods, the treating transiently decreases expression or activity of IRF4. In some such methods, the treating permanently decreases expression or activity of IRF4. [0034] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative IRF4 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative IRF4 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative IRF4 mutant. In some such methods, the one or more compounds comprise an anti-IRF4 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF4 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF4. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF4. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule IRF4 inhibitor. [0035] In some such methods, the treating decreases expression or activity of JDP2. In some such methods, the treating transiently decreases expression or activity of JDP2. In some such methods, the treating permanently decreases expression or activity of JDP2. [0036] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative JDP2 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative JDP2 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative JDP2 mutant. In some such methods, the one or more compounds comprise an anti-JDP2 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an JDP2 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding JDP2. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding JDP2. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule JDP2 inhibitor. [0037] In some such methods, the treating decreases expression or activity of ZNF337. In some such methods, the treating transiently decreases expression or activity of ZNF337. In some such methods, the treating permanently decreases expression or activity of ZNF337. [0038] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative ZNF337 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative ZNF337 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative ZNF337 mutant. In some such methods, the one or more compounds comprise an anti-ZNF337 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ZNF337 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ZNF337. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF337. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule ZNF337 inhibitor. [0039] In some such methods, the treating decreases expression or activity of ETV2. In some such methods, the treating transiently decreases expression or activity of ETV2. In some such methods, the treating permanently decreases expression or activity of ETV2. [0040] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative ETV2 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative ETV2 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative ETV2 mutant. In some such methods, the one or more compounds comprise an anti-ETV2 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV2 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV2. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV2. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule ETV2 inhibitor. [0041] In some such methods, the treating decreases expression or activity of ETV3L. In some such methods, the treating transiently decreases expression or activity of ETV3L. In some such methods, the treating permanently decreases expression or activity of ETV3L. [0042] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative ETV3L mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative ETV3L mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative ETV3L mutant. In some such methods, the one or more compounds comprise an anti-ETV3L antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV3L messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV3L. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3L. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule ETV3L inhibitor. [0043] In some such methods, the treating decreases expression or activity of SOX18. In some such methods, the treating transiently decreases expression or activity of SOX18. In some such methods, the treating permanently decreases expression or activity of SOX18. [0044] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative SOX18 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative SOX18 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative SOX18 mutant. In some such methods, the one or more compounds comprise an anti-SOX18 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an SOX18 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding SOX18. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SOX18. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule SOX18 inhibitor. [0045] In some such methods, the treating decreases expression or activity of CEBPG. In some such methods, the treating transiently decreases expression or activity of CEBPG. In some such methods, the treating permanently decreases expression or activity of CEBPG. [0046] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative CEBPG mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative CEBPG mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative CEBPG mutant. In some such methods, the one or more compounds comprise an anti-CEBPG antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an CEBPG messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CEBPG. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPG. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule CEBPG inhibitor. [0047] In some such methods, the treating decreases expression or activity of CREB3L4. In some such methods, the treating transiently decreases expression or activity of CREB3L4. In some such methods, the treating permanently decreases expression or activity of CREB3L4. [0048] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative CREB3L4 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative CREB3L4 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative CREB3L4 mutant. In some such methods, the one or more compounds comprise an anti-CREB3L4 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an CREB3L4 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CREB3L4. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CREB3L4. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule CREB3L4 inhibitor. [0049] In some such methods, the treating decreases expression or activity of CEBPB. In some such methods, the treating transiently decreases expression or activity of CEBPB. In some such methods, the treating permanently decreases expression or activity of CEBPB. [0050] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative CEBPB mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative CEBPB mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative CEBPB mutant. In some such methods, the one or more compounds comprise an anti-CEBPB antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an CEBPB messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CEBPB. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPB. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule CEBPB inhibitor. [0051] In some such methods, the treating decreases expression or activity of FOXD1. In some such methods, the treating transiently decreases expression or activity of FOXD1. In some such methods, the treating permanently decreases expression or activity of FOXD1. [0052] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative FOXD1 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative FOXD1 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative FOXD1 mutant. In some such methods, the one or more compounds comprise an anti-FOXD1 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an FOXD1 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding FOXD1. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FOXD1. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule FOXD1 inhibitor. [0053] In some such methods, the treating decreases expression or activity of EOMES. In some such methods, the treating transiently decreases expression or activity of EOMES. In some such methods, the treating permanently decreases expression or activity of EOMES. [0054] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative EOMES mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative EOMES mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative EOMES mutant. In some such methods, the one or more compounds comprise an anti-EOMES antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an EOMES messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding EOMES. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding EOMES. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule EOMES inhibitor. [0055] In some such methods, the treating decreases expression or activity of ZNF683. In some such methods, the treating transiently decreases expression or activity of ZNF683. In some such methods, the treating permanently decreases expression or activity of ZNF683. [0056] In some such methods, the one or more compounds comprise a DNA encoding a dominant negative ZNF683 mutant operably linked to a promoter active in the TILs. In some such methods, the DNA encoding the dominant negative ZNF683 mutant is in a viral vector. In some such methods, the viral vector is a lentiviral vector. In some such methods, the one or more compounds comprise a messenger RNA encoding a dominant negative ZNF683 mutant. In some such methods, the one or more compounds comprise an anti-ZNF683 antigen-binding protein. In some such methods, the antigen-binding protein comprises an antibody. In some such methods, the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an ZNF683 messenger RNA. In some such methods, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such methods, the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ZNF683. In some such methods, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF683. In some such methods, the Cas protein is a Cas9 protein or a Cas12a protein. In some such methods, the one or more compounds comprise a small molecule ZNF683 inhibitor. [0057] In some such methods, the TILs originate from a subject. In some such methods, the TILs are from a tumor biopsy, a lymph node, or ascites. In some such methods, the tumor is from a bladder cancer, a breast cancer, a cancer caused by human papilloma virus, a cervical cancer, a head and neck cancer, a lung cancer, a melanoma, an ovarian cancer, a non-small-cell lung cancer (NSCLC), a renal cancer, or a renal cell carcinoma. In some such methods, the tumor biopsy is from a melanoma. [0058] In some such methods, the method further comprises: (i) obtaining a refined tumor product by cryopreserving a resected tumor and disaggregating the cryopreserved tumor, disaggregating a resected tumor and cryopreserving the disaggregated tumor, cryopreserving a resected tumor and processing the tumor into multiple tumor fragments, or processing a resected tumor into multiple tumor fragments and cryopreserving the tumor fragments; and (ii) performing a first expansion by culturing the refined resected tumor product in a cell culture medium comprising IL-2 to produce the first population of TILs, optionally wherein the first population of TILs is treated with the one or more compounds during or subsequent to the first expansion. [0059] In some such methods, the cryopreserving comprises: (1) cooling under conditions whereby heat release to, into, around or in an environment including cells, as media crystalizes, is minimized or avoided; (2) continuous cooling, from disaggregation temperature to about - 80°C; (3) continuous cooling at a rate of about -2°C / min; (4) continuous cooling, from disaggregation temperature to about -80°C, at a rate of about -2°C / min; or (5) continuous cooling, from disaggregation temperature to about -80°C, or from disaggregation temperature to -80°C at a rate of about -2°C / min, wherein disaggregation temperature comprises a normal body temperature for an animal from which the tumor was resected, or room temperature or 20°C or 25°C , or normal human body temperature approximately 35°C or 36°C or 36.1°C to approximately 37°C or 37.1°C or 37.2°C or 37.3°C or below about 38.3°C. [0060] In some such methods, the disaggregating comprises physical disaggregation, enzymatic disaggregation, or physical and enzymatic disaggregation. In some such methods, a single cell suspension is obtained from the refined resected tumor product and used in step (ii), or wherein the refined resected tumor product from step (i) comprises a single cell suspension. [0061] In some such methods, the first expansion in step (ii) is performed for about two weeks. In some such methods, the culturing in step (ii) includes adding IL-7, IL-12, IL-15, IL- 18, IL-21, or a combination thereof. [0062] In some such methods, the method further comprises: (iii) performing a second expansion by culturing the first population of TILs in a cell culture medium comprising IL-2, optionally wherein the first population of TILs is treated with the one or more compounds prior to, during, or subsequent to the second expansion. In some such methods, the expanding in step (iii) comprises culturing the first population of TILs with IL-2, OKT-3, and antigen presenting cells (APCs). In some such methods, the expanding in step (iii) is performed for about two weeks. In some such methods, the culturing in step (iii) includes adding IL-7, IL-12, IL-15, IL- 18, IL-21, or a combination thereof. In some such methods, the method further comprises harvesting and/or cryopreserving the therapeutic population of TILs. [0063] In another aspect, provided are isolated therapeutic populations of TILs obtained by or obtainable by any of the above methods. [0064] In another aspect, provided are isolated therapeutic populations of TILs comprising one or more (any combination thereof) exogenous compounds to improve T-cell fitness. In some such populations, the one or more exogenous compounds comprise one or more or all of the following: (1) a FAS/FASLG inhibitory agent; (2) a TGFβ/TGFβR1 inhibitory agent; (3) an IRF7 inhibitory agent; (4) a POLR3A inhibitory agent; (5) an ETV7 inhibitory agent; (6) an ETV3 inhibitory agent; (7) an ASH2L inhibitory agent; (8) a PML inhibitory agent; (9) a STAT2 inhibitory agent; (10) a SPI1 inhibitory agent; (11) an IRF9 inhibitory agent; (12) a STAT1 inhibitory agent; (13) an IRF4 inhibitory agent; (14) a JDP2 inhibitory agent; (15) a ZNF337 inhibitory agent; (16) an ETV2 inhibitory agent; (17) an ETV3L inhibitory agent; (18) a SOX18 inhibitory agent; (19) a CEBPG inhibitory agent; (20) a CREB3L4 inhibitory agent; (21) a CEBPB inhibitory agent; (22) a FOXD1 inhibitory agent; (23) an EOMES inhibitory agent; and (24) a ZNF683 inhibitory agent. [0065] In another aspect, provided are isolated therapeutic populations of TILs comprising one or more exogenous compounds to improve T-cell fitness. In some such populations, the one or more exogenous compounds comprise one or more or all of the following: (i) a FAS/FASLG inhibitory agent; (ii) a TGFβ/TGFβR1 inhibitory agent; (iii) an IRF7 inhibitory agent; and (iv) a POLR3A inhibitory agent. [0066] In some such populations, the one or more exogenous compounds decrease expression or activity of FAS or FASLG. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of FAS or FASLG. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of FAS or FASLG. [0067] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative FAS mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative FAS mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative FAS mutant. In some such populations, the dominant negative FAS mutant comprises a mutated FADD binding site, optionally wherein the dominant negative FAS mutant is FAS_D244V. In some such populations, the dominant negative FAS mutant comprises a deleted DD domain, optionally wherein the dominant negative FAS mutant is FAS_del230-314. In some such populations, the one or more exogenous compounds comprise an anti-FAS or anti-FASLG antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to a FAS or FASLG messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding FAS or FASLG. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FAS or FASLG. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule FAS/FASLG inhibitor. [0068] In some such populations, the one or more exogenous compounds decrease expression or activity of TGFβ1 or TGFβR1. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of TGFβ1 or TGFβR1. In some such populations, the one or more exogenous compounds comprise permanently decrease expression or activity of TGFβ1 or TGFβR1. [0069] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative TGFβR1 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative TGFβR1 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative TGFβR1 mutant. In some such populations, the one or more exogenous compounds comprise an anti-TGFβR or anti-TGFβ1 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to a TGFβ1 or TGFβR1 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding TGFβ1 or TGFβR1. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding TGFβR. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule TGFβR1 inhibitor, optionally wherein the small molecule TGFβR1 inhibitor is SB431542. [0070] In some such populations, the one or more exogenous compounds decrease expression or activity of IRF7. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of IRF7. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of IRF7. [0071] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative IRF7 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative IRF7 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative IRF7 mutant. In some such populations, the one or more exogenous compounds comprise an anti-IRF7 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF7 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF7. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF7. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule IRF7 inhibitor. [0072] In some such populations, the one or more exogenous compounds decrease expression or activity of POLR3A. In some such populations, the one or more exogenous compounds comprise transiently decrease expression or activity of POLR3A. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of POLR3A. [0073] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative POLR3A mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative POLR3A mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative POLR3A mutant. In some such populations, the one or more exogenous compounds comprise an anti-POLR3A antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to a POLR3A messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding POLR3A. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding POLR3A. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule POLR3A inhibitor. [0074] In some such populations, the one or more exogenous compounds decrease expression or activity of ETV7. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ETV7. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ETV7. [0075] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative ETV7 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative ETV7 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ETV7 mutant. In some such populations, the one or more exogenous compounds comprise an anti-ETV7 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV7 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV7. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV7. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule ETV7 inhibitor. [0076] In some such populations, the one or more exogenous compounds decrease expression or activity of ETV3. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ETV3. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ETV3. [0077] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative ETV3 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative ETV3 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ETV3 mutant. In some such populations, the one or more exogenous compounds comprise an anti-ETV3 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV3 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV3. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule ETV3 inhibitor. [0078] In some such populations, the one or more exogenous compounds decrease expression or activity of ASH2L. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ASH2L. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ASH2L. [0079] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative ASH2L mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative ASH2L mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ASH2L mutant. In some such populations, the one or more exogenous compounds comprise an anti-ASH2L antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ASH2L messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ASH2L. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ASH2L. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule ASH2L inhibitor. [0080] In some such populations, the one or more exogenous compounds decrease expression or activity of PML. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of PML. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of PML. [0081] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative PML mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative PML mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative PML mutant. In some such populations, the one or more exogenous compounds comprise an anti-PML antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an PML messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding PML. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding PML. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule PML inhibitor. [0082] In some such populations, the one or more exogenous compounds decrease expression or activity of STAT2. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of STAT2. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of STAT2. [0083] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative STAT2 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative STAT2 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative STAT2 mutant. In some such populations, the one or more exogenous compounds comprise an anti-STAT2 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an STAT2 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding STAT2. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT2. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule STAT2 inhibitor. [0084] In some such populations, the one or more exogenous compounds decrease expression or activity of SPI1. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of SPI1. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of SPI1. [0085] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative SPI1 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative SPI1 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative SPI1 mutant. In some such populations, the one or more exogenous compounds comprise an anti-SPI1 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an SPI1 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding SPI1. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SPI1. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule SPI1 inhibitor. [0086] In some such populations, the one or more exogenous compounds decrease expression or activity of IRF9. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of IRF9. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of IRF9. [0087] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative IRF9 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative IRF9 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative IRF9 mutant. In some such populations, the one or more exogenous compounds comprise an anti-IRF9 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF9 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF9. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF9. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule IRF9 inhibitor. [0088] In some such populations, the one or more exogenous compounds decrease expression or activity of STAT1. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of STAT1. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of STAT1. [0089] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative STAT1 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative STAT1 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative STAT1 mutant. In some such populations, the one or more exogenous compounds comprise an anti-STAT1 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an STAT1 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding STAT1. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT1. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule STAT1 inhibitor. [0090] In some such populations, the one or more exogenous compounds decrease expression or activity of IRF4. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of IRF4. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of IRF4. [0091] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative IRF4 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative IRF4 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative IRF4 mutant. In some such populations, the one or more exogenous compounds comprise an anti-IRF4 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF4 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF4. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF4. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule IRF4 inhibitor. [0092] In some such populations, the one or more exogenous compounds decrease expression or activity of JDP2. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of JDP2. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of JDP2. [0093] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative JDP2 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative JDP2 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative JDP2 mutant. In some such populations, the one or more exogenous compounds comprise an anti-JDP2 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an JDP2 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding JDP2. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding JDP2. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule JDP2 inhibitor. [0094] In some such populations, the one or more exogenous compounds decrease expression or activity of ZNF337. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ZNF337. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ZNF337. [0095] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative ZNF337 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative ZNF337 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ZNF337 mutant. In some such populations, the one or more exogenous compounds comprise an anti-ZNF337 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ZNF337 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ZNF337. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF337. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule ZNF337 inhibitor. [0096] In some such populations, the one or more exogenous compounds decrease expression or activity of ETV2. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ETV2. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ETV2. [0097] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative ETV2 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative ETV2 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ETV2 mutant. In some such populations, the one or more exogenous compounds comprise an anti-ETV2 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV2 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV2. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV2. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule ETV2 inhibitor. [0098] In some such populations, the one or more exogenous compounds decrease expression or activity of ETV3L. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ETV3L. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ETV3L. [0099] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative ETV3L mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative ETV3L mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ETV3L mutant. In some such populations, the one or more exogenous compounds comprise an anti-ETV3L antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ETV3L messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ETV3L. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3L. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule ETV3L inhibitor. [00100] In some such populations, the one or more exogenous compounds decrease expression or activity of SOX18. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of SOX18. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of SOX18. [00101] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative SOX18 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative SOX18 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative SOX18 mutant. In some such populations, the one or more exogenous compounds comprise an anti-SOX18 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an SOX18 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding SOX18. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SOX18. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule SOX18 inhibitor. [00102] In some such populations, the one or more exogenous compounds decrease expression or activity of CEBPG. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of CEBPG. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of CEBPG. [00103] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative CEBPG mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative CEBPG mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative CEBPG mutant. In some such populations, the one or more exogenous compounds comprise an anti-CEBPG antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an CEBPG messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CEBPG. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPG. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule CEBPG inhibitor. [00104] In some such populations, the one or more exogenous compounds decrease expression or activity of CREB3L4. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of CREB3L4. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of CREB3L4. [00105] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative CREB3L4 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative CREB3L4 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative CREB3L4 mutant. In some such populations, the one or more exogenous compounds comprise an anti-CREB3L4 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an CREB3L4 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CREB3L4. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CREB3L4. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule CREB3L4 inhibitor. [00106] In some such populations, the one or more exogenous compounds decrease expression or activity of CEBPB. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of CEBPB. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of CEBPB. [00107] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative CEBPB mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative CEBPB mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative CEBPB mutant. In some such populations, the one or more exogenous compounds comprise an anti-CEBPB antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an CEBPB messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding CEBPB. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPB. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule CEBPB inhibitor. [00108] In some such populations, the one or more exogenous compounds decrease expression or activity of FOXD1. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of FOXD1. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of FOXD1. [00109] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative FOXD1 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative FOXD1 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative FOXD1 mutant. In some such populations, the one or more exogenous compounds comprise an anti-FOXD1 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an FOXD1 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding FOXD1. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FOXD1. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule FOXD1 inhibitor. [00110] In some such populations, the one or more exogenous compounds decrease expression or activity of EOMES. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of EOMES. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of EOMES. [00111] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative EOMES mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative EOMES mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative EOMES mutant. In some such populations, the one or more exogenous compounds comprise an anti-EOMES antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an EOMES messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding EOMES. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding EOMES. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule EOMES inhibitor. [00112] In some such populations, the one or more exogenous compounds decrease expression or activity of ZNF683. In some such populations, the one or more exogenous compounds transiently decrease expression or activity of ZNF683. In some such populations, the one or more exogenous compounds permanently decrease expression or activity of ZNF683. [00113] In some such populations, the one or more exogenous compounds comprise a DNA encoding a dominant negative ZNF683 mutant operably linked to a promoter active in the TILs. In some such populations, the DNA encoding the dominant negative ZNF683 mutant is in a viral vector. In some such populations, the viral vector is a lentiviral vector. In some such populations, the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative ZNF683 mutant. In some such populations, the one or more exogenous compounds comprise an anti-ZNF683 antigen-binding protein. In some such populations, the antigen-binding protein comprises an antibody. In some such populations, the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an ZNF683 messenger RNA. In some such populations, the inhibitory RNA is an antisense oligonucleotide or an RNAi agent. In some such populations, the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding ZNF683. In some such populations, the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF683. In some such populations, the Cas protein is a Cas9 protein or a Cas12a protein. In some such populations, the one or more exogenous compounds comprise a small molecule ZNF683 inhibitor. [00114] In some such populations, the TILs originate from a subject. In some such populations, the TILs are from a tumor biopsy, a lymph node, or ascites. In some such populations, the tumor is from a bladder cancer, a breast cancer, a cancer caused by human papilloma virus, a cervical cancer, a head and neck cancer, a lung cancer, a melanoma, an ovarian cancer, a non-small-cell lung cancer (NSCLC), a renal cancer, or a renal cell carcinoma. In some such populations, the tumor biopsy is from a melanoma. In some such populations, the population comprises about 5x109 to about 5x1010 TILs. [00115] In another aspect, provided are pharmaceutical formulations comprising a pharmaceutically acceptable excipient and any of the above isolated therapeutic population of TILs. [00116] In another aspect, provided are cryopreserved bags or an intravenous infusion bags, containers, or vessels containing contents comprising any of the above isolated therapeutic population of TILs. [00117] In another aspect, provided are methods of treating a cancer in a subject, comprising administering any of the above isolated therapeutic population of TILs or the above pharmaceutical formulation to the subject. [00118] In another aspect, provide are methods for treating a cancer in a subject, comprising: (a) preparing a therapeutic population of TILs according to any of the above methods; and (b) administering a therapeutic amount of the therapeutic population of TILs to the subject with the cancer. [00119] In some such methods, the TILs are autologous or allogeneic. In some such methods, the cancer is a bladder cancer, a breast cancer, a cancer caused by human papilloma virus, a cervical cancer, a head and neck cancer, a head and neck squamous cell carcinoma (HNSCC), a lung cancer, a melanoma, an ovarian cancer, a non-small-cell lung cancer (NSCLC), a renal cancer, or a renal cell carcinoma. In some such methods, the cancer is a melanoma. [00120] In some such methods, the subject is a human. In some such methods, the subject is a non-human mammal. In some such methods, the non-human mammal is a primate, a rodent, a rat, a mouse, a domesticated mammal, a domesticated cat, a domesticated dog, a domesticated horse, a guinea pig, a laboratory animal, or a companion animal. In some such methods, the subject is an adult or individual having secondary sexual characteristics. In some such methods, the subject is not an adult or not individual having secondary sexual characteristics, or is a child or is a not physically mature mammal. [00121] In some such methods, the administering is performed more than once. In some such methods, the administering is performed more than once over a course of time, wherein: (1) the course of time is a week and the administering is twice, thrice, four times or five times in the week; (2) the course of time is a month and the administering is twice, thrice of four times in a month; (3) the course of time is three, six, nine, or twelve months and the administering is performed once monthly or once weekly. In some such methods, the administering is intravenous administration. BRIEF DESCRIPTION OF THE DRAWINGS [00122] The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee. [00123] Figure 1. Experimental scheme of healthy donor T cell repeat stimulation assay. PBMC and isolated T cells from two donors were thawed and stimulated using various methods according to the depicted schedules. After the first three-day round of stimulation, cells in “Repeat Stim” group were counted, washed, diluted to 1x106 cells/mL, and repeatedly stimulated every 3 days for a total of 10 rounds in 34 days. Cells in “Round 1 Rest” group were counted, washed, and diluted to 1x106 cells/mL every 3 days without further stimulation. Cells in “Round 2 Stim” group were counted, washed, and diluted to 1x106 cells/mL every 3 days along with 1 more round of stimulation. On each day of dilution and final harvest, a fraction of the cells in each group was stained and analyzed by flow cytometry for surface markers of activation, exhaustion, and differentiation phenotypes. [00124] Figures 2A-2F. Cell expansion curves of PBMCs under different stimulation schemes. Total viable cell counts of PBMC from two different donors (Figures 2A, 2B, 2C for D2040, Figures 2D, 2E, 2F for D2740) were measured and plotted over time in “Repeat Stim” group (Figures 2A, 2D), “R1 Rest group” (Figures 2B, 2E), and “R2 Rest” group (Figures 2C, 2F). Comparisons between different stimulation methods are shown in each plot. Stimulation schemes: OKT3+CD28 (GMP) antibodies; OKT3+CD28 (RUO) antibodies; OKT3+CD28 -> OKT3; OKT3 -> OKT3; Dynabeads Human T-Activator CD3/CD28 (DB); and TransAct (TA). GMP = a Good Manufacturing Practices-grade soluble CD28 antibody for ex vivo T cell activation; RUO = a Research Use Only-grade CD28 antibody for ex vivo T cell activation. [00125] Figure 3A-3F. Cell expansion curves of T cells under different stimulation schemes. Total viable cell counts of T cells from two different donors (Figures 3A, 3B, 3C for D2040, Figures 3D, 3E, 3F for D2740) were measured and plotted over time in “Repeat Stim” group (Figures 3A, 3D), “R1 Rest group” (Figures 3B, 3E), and “R2 Rest” group (Figures 3C, 3F). Comparison between different stimulation methods are shown in each plot. [00126] Figure 4. Experimental scheme of healthy donor (HD) T cell repeat stimulation assay screening of selected candidates. Isolated T cells from two healthy donors were thawed and stimulated using Dynabeads for 3 days. On day 1, T cells were transduced with short-listed candidate LVVs at MOI=5. After the first round of stimulation, T cells in “Repeat Stim” group were counted, washed, diluted to 1x106 cells/mL, and repeatedly stimulated every 3 days for a total of 10 rounds in 34 days. T cells in “Round 1 Rest” group were counted, washed, and diluted to 1x106 cells/mL every 3 days without stimulation. On each day of dilution and final harvest, a fraction of T cells was stained and analyzed by flow cytometry for surface tCD34 expression along with markers of activation, exhaustion, and differentiation phenotypes. [00127] Figure 5. T cell expansion curves under repeat stimulation. Total viable cell count was measured and plotted over time in LOF candidate groups. pIB1123 is empty vector that served as a control. Candidates that enable enhanced proliferation, as a T cell expansion curve above pIB1123 control curve, were selected as candidate and highlighted below figure legend in each panel. [00128] Figure 6. T cell expansion curves under resting conditions. Total viable cell count was measured and plotted over time in LOF candidate groups. pIB1123 is empty vector that served as a control. Candidates that enable enhanced proliferation, as a T cell expansion curve above pIB1123 control curve, were selected as candidate and highlighted below figure legend in each panel. [00129] Figures 7A-7B. tCD34+ cells were enriched in T cells transduced positive hits under repeat stimulation. Percent of tCD34+ cells (Figure 7A) and viable tCD34+ cell count (Figure 7B) were measured by flow cytometry (Figure 7A), calculated from total viable cells (Figure 7B), and plotted over time. pIB1123 is empty vector that served as a control. Positive hits enabled continuous enrichment of transduced T cell populations (as measured by tCD34+), which highlights enhanced proliferation and growth advantage from positive hits. [00130] Figures 8A-8F. T cell activation markers in tCD34+ T cells from positive hit groups under repeat stimulation. Surface expression of 4-1BB (Figures 8A, 8D), PD-1 (Figures 8B, 8E), and OX40 (Figures 8C, 8F) were measured on tCD34+CD4+ (Figures 8A, 8B, 8C) and tCD34+CD8+ (Figures 8D, 8E, 8F) populations by flow cytometry and plotted over time. [00131] Figure 9A-9F. T cell exhaustion markers in tCD34+ T cells from positive hit groups under repeat stimulation. Surface expression of TIM-3 (Figures 9A, 9D), LAG-3 (Figures 9B, 9E), and CTLA-4 (Figures 9C, 9F) were measured on tCD34+CD4+ (Figures 9A, 9B, 9C) and tCD34+CD8+ (Figures 9D, 9E, 9F) populations by flow cytometry and plotted over time. [00132] Figures 10A-10B. T cell phenotypic subsets in tCD34+ T cells from positive hit groups under repeat stimulation. Surface expression of CCR7 and CD45RA were measured on tCD34+CD4+ (Figure 10A) and tCD34+CD8+ (Figure 10B) populations by flow cytometry. Relative abundance of Tn/Tscm (CCR7+CD45RA+), Tcm (CCR7+CD45RA-), Tem (CCR7- CD45RA-), and Temra (CCR7-CD45RA+) populations were plotted over time. [00133] Figure 11. Experimental scheme of screening selected short-listed candidates in TILs. Frozen tumor digest from two patients were thawed and expanded ex vivo using standard TIL manufacture protocol of 12-day outgrowth followed by 12-day REP. Cells were transduced at MOI=5 twice on day 3 and 4 with selected short-listed candidate LVV. On day 1, 13, and 25, total viable cells were counted, and a fraction of cells was stained and analyzed by flow cytometry for surface tCD34 expression along with markers of activation, exhaustion, and differentiation phenotypes. [00134] Figures 12A-12B. Cell expansion curves of TILs from renal cell carcinoma during Outgrowth (Figure 12A) and REP (Figure 12B). Total viable cell count from groups transduced with different candidates were plotted over time for comparison. [00135] Figures 13A-13D. Fold expansion of TILs from renal cell carcinoma during Outgrowth (Figure 13A, 13C) and REP (Figure 13B, 13D). Total viable cell count from groups transduced with different candidates were plotted over time for comparison. [00136] Figures 14A-14C. Enrichment of transduced populations in TILs from renal cell carcinoma during REP. (Figure 14A) Percent of tCD34+ population measured by flow cytometry on day 13 and 25, as compared among all groups. Any increase from day 13 to day 25 in groups 3-7 greater than that in group 2 was selected as positive hits. (Figure 14B) Relative tCD34+% enrichment of transduced populations from day 13 to day 25 was calculated as [(tCD34+% on day 25) – (tCD34+% on day 13)] / (tCD34+% on day13) * 100%. Any relative tCD34+% enrichment in groups 3-7 greater than that in group 2 was selected as positive hits. (Figure 14C) Fold change of tCD34+ viable count from day 13 to day 25 was calculated as [(tCD34+ viable count on day 25) – (tCD34+ viable count on day13)] / (tCD34+ viable count on day13) * 100%. Any relative tCD34+% enrichment in groups 3-7 greater than that in group 2 was selected as positive hits. [00137] Figures 15A-15B. Relative abundance of CD4+ and CD8+ cells in CD3+ populations by the end of Outgrowth (day 13, Figure 15A) and REP (day 25, Figure 15B). Surface expression of CD4 and CD8 were measured by flow cytometry and compared across different groups. [00138] Figures 16A-16B. PD-1+% in CD4+ (Figure 16A) and CD8+ (Figure 16B) populations by the end of Outgrowth (day 13) and REP (day 25). Surface expression of PD-1 were measured by flow cytometry and compared across different groups. [00139] Figures 17A-17B. TIM-3+% in CD4+ (Figure 17A) and CD8+ (Figure 17B) populations by the end of Outgrowth (day 13) and REP (day 25). Surface expression of TIM-3 were measured by flow cytometry and compared across different groups. [00140] Figures 18A-18B. LAG-3+% in CD4+ (Figure 18A) and CD8+ (Figure 18B) populations by the end of Outgrowth (day 13) and REP (day 25). Surface expression of LAG-3 were measured by flow cytometry and compared across different groups. [00141] Figures 19A-19B. CD127+% in CD4+ (Figure 19A) and CD8+ (Figure 19B) populations by the end of Outgrowth (day 13) and REP (day 25). Surface expression of CD127 were measured by flow cytometry and compared across different groups. [00142] Figures 20A-20B. CD27+% in CD4+ (Figure 20A) and CD8+ (Figure 20B) populations by the end of Outgrowth (day 13) and REP (day 25). Surface expression of CD27 were measured by flow cytometry and compared across different groups. [00143] Figures 21A-21D. T cell phenotypic subsets in CD4+ (Figures 21A, 21B) and CD8+ (Figures 21C, 21D) populations by the end of Outgrowth (day 13, Figures 21A, 21C) and REP (day 25, Figures 21B, 21D). Surface expression of CCR7 and CD45RA were measured by flow cytometry. Relative abundance of Tn/Tscm (CCR7+CD45RA+), Tcm (CCR7+CD45RA-), Tem (CCR7-CD45RA-), and Temra (CCR7-CD45RA+) populations were compared across different groups. [00144] Figures 22A-22B. Unsupervised clustering (Figure 22A) of the gene expression profile of individual cells identified cell subpopulations with distinct transcriptional profiles (Figure 22B) previously undescribed in TIL products. UMAP, uniform manifold approximation and projection. [00145] Figures 23A-23C. Frequency of C7 (MX1+OAS1+; Figure 23A), C9 (BBC3+CHAC1+; Figure 23B), or C7 and C9 (Figure 23C) T-cell subpopulations in TIL product samples. Low abundance of C7 TIL or C9 TIL subpopulations is associated with response (Figures 23A and 23B). Low abundance of the combined C7 and C9 TIL subpopulations is associated with response (Figure 23C). [00146] Figure 24. Gene regulatory network analysis of TIL products identified predicted master regulators of C7 and C9 T-cell subpopulations. FDR stands for false discovery rate which is p value adjusted using the Benjamini-Hochberg procedure to account for multiple hypothesis testing. DEFINITIONS [00147] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs. [00148] The term “anti-CD3 antibody” refers to an antibody or variant thereof, e.g., a monoclonal antibody and including human, humanized, chimeric, murine or mammalian antibodies which are directed against the CD3 receptor in the T cell antigen receptor of mature human T cells. Anti-CD3 antibodies include OKT-3, also known as muromonab. Anti-CD3 antibodies also include the UHCT1 clone, also known as T3 and CD3.epsilon. Other anti-CD3 antibodies include, for example, otelixizumab, teplizumab, and visilizumab. [00149] When “an anti-tumor effective amount,” “an tumor-inhibiting effective amount,” or “therapeutic amount” is indicated, the precise amount of the compositions of the present invention to be administered can be determined by a physician with consideration of individual differences in age, weight, tumor size, extent of infection or metastasis, and condition of the patient (subject). It can generally be stated that a pharmaceutical composition comprising the tumor infiltrating lymphocytes (e.g. secondary TILs or genetically modified cytotoxic lymphocytes) described herein may be administered at a dosage of 104 to 1011 cells/kg body weight (e.g., 105 to 106, 105 to 1010, 105 to 1011, 106 to 1010, 106 to 1011, 107 to 1011, 107 to 1010, 108 to 1011, 108 to 1010, 109 to 1011, or 109 to 1010 cells/kg body weight), including all integer values within those ranges. Tumor infiltrating lymphocytes (including in some cases, genetically modified cytotoxic lymphocytes) compositions may also be administered multiple times at these dosages. The tumor infiltrating lymphocytes (including in some cases, genetically) can be administered by using infusion techniques that are commonly known in immunotherapy (see, e.g., Rosenberg et al., New Eng. J. of Med.319: 1676, 1988). The optimal dosage and treatment regime for a particular patient can readily be determined by one skilled in the art of medicine by monitoring the patient for signs of disease and adjusting the treatment accordingly. [00150] The terms “co-administration,” “co-administering,” “administered in combination with,” “administering in combination with,” “simultaneous,” and “concurrent,” as used herein, encompass administration of two or more active pharmaceutical ingredients (in a preferred embodiment of the present invention, for example, at least one potassium channel agonist in combination with a plurality of TILs) to a subject so that both active pharmaceutical ingredients and/or their metabolites are present in the subject at the same time. Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which two or more active pharmaceutical ingredients are present. Simultaneous administration in separate compositions and administration in a composition in which both agents are present are preferred. [00151] “Cellularized or cellularization” as used herein refers to the process of disaggregation whereby the solid tissue a multicellular material generally made up of multiple cell lineages/types is broken down into small numbers of cells including but not limited to one cell but could be multiple cells of various lineages or cell types in very small numbers i.e. clump of cells or cell aggregates. [00152] “Closed system” as used herein refers to a system that is closed to the outside environment. Any closed system appropriate for cell culture methods can be employed with the methods of the present invention. Closed systems include, for example, but are not limited to closed G-Rex containers or cell culture bags. Once a tumor segment is added to the closed system, the system is not open to the outside environment until the TILs are ready to be administered to the patient. In an advantageous embodiment, the closed system is the system disclosed in PCT Publication No. WO 2018/130845. [00153] “Cryopreservation media” or “cryopreservation medium” as used herein refers to any medium that can be used for cryopreservation of cells. Such media can include media comprising 2% to 10% DMSO. Exemplary media include CryoStor CS10, HypoThermosol, Bloodstor BS- 55 as well as combinations thereof. [00154] The term “cryopreserved TILs” herein is meant that TILs, either primary, bulk, or expanded (REP TILs), are treated and stored in the range of about -190 °C. to -60 °C. General methods for cryopreservation are also described elsewhere herein, including in the Examples. For clarity, “cryopreserved TILs” are distinguishable from frozen tissue samples which may be used as a source of primary TILs. [00155] “Depletion” as used herein refers to a process of a negative selection that separates the desired cells from the undesired cells which are labelled by one marker-binding fragment coupled to a solid phase. [00156] “Disaggregation or disaggregate” as used herein refers to the transformation of solid tissue into a single cells or small cell number aggregates where a single cell as a spheroid has a diameter in the range of 5 μm, 6 μm, 7 μm, 8 μm, 9 μm, 10 μm, 20 μm, 30 μm, 40 μm, 50 μm, 60 μm, 70 μm, 80 μm, 90 μm, 100 μm, or more, wherein this is more usually between 7 to 20 μm. [00157] The term “effective amount” or “therapeutically effective amount” refers to that amount of a compound or combination of compounds as described herein that is sufficient to affect the intended application including, but not limited to, disease treatment. A therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated (e.g., the weight, age and gender of the subject), the severity of the disease condition, or the manner of administration. The term also applies to a dose that will induce a particular response in target cells (e.g., the reduction of platelet adhesion and/or cell migration). The specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether the compound is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which the compound is carried. [00158] “Engineered” as used herein refers to either addition of nucleic material or factors, which change the tissue derived cell function from their original function to have a new or improved function for its ultimate utility. [00159] “Enzyme media” as used herein refers to media having enzymatic activity such as collagenase, trypsin, lipase, hyaluronidase, deoxyribonuclease, Liberase HI, pepsin, or mixtures thereof. [00160] “Filtrate” as used herein refers to the material that passes through a filter, mesh or membrane. [00161] “Flexible container” as used herein refers to a flexible packaging system in multiple formats with one or more different types of film. Each film type is selected to provide specific characteristics to preserve the physical, chemical, and functional characteristics of the sterile fluids, solid tissue derived cellular material and the container integrity depending upon the step of the process. [00162] “Freezing solution” or “cryopreservation solution” also referred in the field to as the cryoprotectant is a solution that contains cryoprotective additives. These are generally permeable, non-toxic compounds which modify the physical stresses cells are exposed to during freezing in order to minimize freeze damage (i.e. due to ice formation) and are most commonly a % vol/vol of one or more of the following: dimethylsulphoxide (DMSO); ethylene glycol; glycerol; 2-methyl-2,4-pentanediol (MPD); propylene glycol; sucrose; and trehalose. [00163] The term “hematological malignancy” refers to mammalian cancers and tumors of the hematopoietic and lymphoid tissues, including but not limited to tissues of the blood, bone marrow, lymph nodes, and lymphatic system. Hematological malignancies are also referred to as “liquid tumors.” Hematological malignancies include, but are not limited to, acute lymphoblastic leukemia (ALL), chronic lymphocytic lymphoma (CLL), small lymphocytic lymphoma (SLL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute monocytic leukemia (AMoL), Hodgkin's lymphoma, and non-Hodgkin's lymphomas. The term “B cell hematological malignancy” refers to hematological malignancies that affect B cells. [00164] The term “IL-2” (also referred to herein as “IL2”) refers to the T cell growth factor known as interleukin-2, and includes all forms of IL-2 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof. IL-2 is described, e.g., in Nelson, J. Immunol.2004, 172, 3983-88 and Malek, Annu. Rev. Immunol. 2008, 26, 453-79, the disclosures of which are incorporated by reference herein. For example, the term IL-2 encompasses human, recombinant forms of IL-2 such as aldesleukin (PROLEUKIN, available commercially from multiple suppliers in 22 million IU per single use vials), as well as the form of recombinant IL-2 commercially supplied by CellGenix, Inc., Portsmouth, N.H., USA (CELLGRO GMP) or ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-209-b) and other commercial equivalents from other vendors. Aldesleukin (des-alanyl-1, serine-125 human IL-2) is a nonglycosylated human recombinant form of IL-2 with a molecular weight of approximately 15 kDa. The term IL-2 also encompasses pegylated forms of IL-2, as described herein, including the pegylated IL2 prodrug NKTR-214, available from Nektar Therapeutics, South San Francisco, Calif., USA. NKTR-214 and pegylated IL-2 suitable for use in the invention is described in U.S. Patent Application Publication No. US 2014/0328791 A1 and International Patent Application Publication No. WO 2012/065086 A1. Alternative forms of conjugated IL-2 suitable for use in the invention are described in U.S. Pat. Nos.4,766,106, 5,206,344, 5,089,261 and 4902,502. Formulations of IL-2 suitable for use in the invention are described in U.S. Pat. No.6,706,289. [00165] The term “IL-4” (also referred to herein as “IL4”) refers to the cytokine known as interleukin 4, which is produced by Th2 T cells and by eosinophils, basophils, and mast cells. IL- 4 regulates the differentiation of naive helper T cells (Th0 cells) to Th2 T cells. Steinke and Borish, Respir. Res.2001, 2, 66-70. Upon activation by IL-4, Th2 T cells subsequently produce additional IL-4 in a positive feedback loop. IL-4 also stimulates B cell proliferation and class II MHC expression, and induces class switching to IgE and IgG1 expression from B cells. Recombinant human IL-4 suitable for use in the invention is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-211) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-15 recombinant protein, Cat. No. Gibco CTP0043). [00166] The term “IL-7” (also referred to herein as “IL7”) refers to a glycosylated tissue- derived cytokine known as interleukin 7, which may be obtained from stromal and epithelial cells, as well as from dendritic cells. Fry and Mackall, Blood 2002, 99, 3892-904. IL-7 can stimulate the development of T cells. IL-7 binds to the IL-7 receptor, a heterodimer consisting of IL-7 receptor alpha and common gamma chain receptor, which in a series of signals important for T cell development within the thymus and survival within the periphery. Recombinant human IL-7 suitable for use in the invention is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-254) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-15 recombinant protein, Cat. No. Gibco PHC0071). [00167] The term “IL-12” (also referred to herein as “IL12”) refers to the T cell growth factor known as interleukin-12. Interleukin (IL)-12 is a secreted heterodimeric cytokine comprised of 2 disulfide- linked glycosylated protein subunits, designated p35 and p40 for their approximate molecular weights. IL-12 is produced primarily by antigen-presenting cells and drives cell- mediated immunity by binding to a two-chain receptor complex that is expressed on the surface of T cells or natural killer (NK) cells. The IL-12 receptor beta-1 (IL-12Rpi) chain binds to the p40 subunit of IL-12, providing the primary interaction between IL-12 and its receptor. However, it is IL-12p35 ligation of the second receptor chain, IL-12RP2, that confers intracellular signaling. IL-12 signaling concurrent with antigen presentation is thought to invoke T cell differentiation towards the T helper 1 (Thl) phenotype, characterized by interferon gamma (IFNy) production. Thl cells are believed to promote immunity to some intracellular pathogens, generate complement-fixing antibody isotypes, and contribute to tumor immunosurveillance. Thus, IL-12 is thought to be a significant component to host defense immune mechanisms. IL-12 is part of the IL-12 family of cytokines which also includes IL-23, IL-27, IL-35, IL-39. [00168] The term “IL-15” (also referred to herein as “IL15”) refers to the T cell growth factor known as interleukin-15, and includes all forms of IL-15 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof. IL- 15 is described, e.g., in Fehniger and Caligiuri, Blood 2001, 97, 14-32, the disclosure of which is incorporated by reference herein. IL-15 shares β and γ signaling receptor subunits with IL-2. Recombinant human IL-15 is a single, non-glycosylated polypeptide chain containing 114 amino acids (and an N-terminal methionine) with a molecular mass of 12.8 kDa. Recombinant human IL-15 is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-230-b) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-15 recombinant protein, Cat. No.34-8159-82). [00169] The term “IL-18” (also referred to herein as “IL18”) refers to the T cell growth factor known as interleukin-15. Interleukin-18 (IL-18) is a proinflammatory cytokine that belongs to the IL-1 cytokine family, due to its structure, receptor family and signal transduction pathways. Related cytokines include IL-36, IL-37, IL-38. [00170] The term “IL-21” (also referred to herein as “IL21”) refers to the pleiotropic cytokine protein known as interleukin-21, and includes all forms of IL-21 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof. IL-21 is described, e.g., in Spolski and Leonard, Nat. Rev. Drug. Disc.2014, 13, 379-95, the disclosure of which is incorporated by reference herein. IL-21 is primarily produced by natural killer T cells and activated human CD4+ T cells. Recombinant human IL-21 is a single, non-glycosylated polypeptide chain containing 132 amino acids with a molecular mass of 15.4 kDa. Recombinant human IL-21 is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-408-b) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-21 recombinant protein, Cat. No.14-8219-80). [00171] The term “liquid tumor” refers to an abnormal mass of cells that is fluid in nature. Liquid tumor cancers include, but are not limited to, leukemias, myelomas, and lymphomas, as well as other hematological malignancies. TILs obtained from liquid tumors may also be referred to herein as marrow infiltrating lymphocytes (MILs). [00172] “Magnetic” in “magnetic particle” as used herein refers to all subtypes of magnetic particles, which can be prepared with methods well known to the skilled person in the art, especially ferromagnetic particles, superparamagnetic particles and paramagnetic particles. “Ferromagnetic” materials are strongly susceptible to magnetic fields and are capable of retaining magnetic properties when the field is removed. “Paramagnetic” materials have only a weak magnetic susceptibility and when the field is removed quickly lose their weak magnetism. “Superparamagnetic” materials are highly magnetically susceptible, i.e. they become strongly magnetic when placed in a magnetic field, but, like paramagnetic materials, rapidly lose their magnetism. [00173] “Marker” as used herein refers to a cell antigen that is specifically expressed by a certain cell type. Preferentially, the marker is a cell surface marker, so that enrichment, isolation and/or detection of living cells can be performed. [00174] “Marker-binding fragment” as used herein refers to any moiety that binds preferentially to the desired target molecule of the cell, i.e. the antigen. The term moiety comprises, e.g., an antibody or antibody fragment. The term “antibody” as used herein refers to polyclonal or monoclonal antibodies which can be generated by methods well known to the person skilled in the art. The antibody may be of any species, e.g. murine, rat, sheep, human. For therapeutic purposes, if non-human antigen binding fragments are to be used, these can be humanized by any method known in the art. The antibodies may also be modified antibodies (e.g. oligomers, reduced, oxidized and labelled antibodies). The term “antibody” comprises both intact molecules and antibody fragments, such as Fab, Fab', F(ab')2, Fv and single- chain antibodies. Additionally, the term “marker-binding fragment” includes any moiety other than antibodies or antibody fragments that binds preferentially to the desired target molecule of the cell. Suitable moieties include, without limitation, oligonucleotides known as aptamers that bind to desired target molecules (Hermann and Pantel, 2000: Science 289: 820-825), carbohydrates, lectins or any other antigen binding protein (e.g. receptor-ligand interaction). [00175] “Media” means various solutions known in the art of cell culturing, cell handling and stabilization used to reduce cell death, including but not limited to one or more of the following media Organ Preservation Solutions , selective lysis solutions, PBS, DMEM, HBSS, DPBS, RPMI, Iscove’s medium, X-VIVO™, Lactated Ringer's solution, Ringer's acetate, saline, PLASMALYTE™ solution, crystalloid solutions and IV fluids, colloid solutions and IV fluids, five percent dextrose in water (D5W), Hartmann's Solution. The media can be standard cell media like the above mentioned-media or special media for e.g. primary human cell culture (e.g. for endothelia cells, hepatocytes, or keratinocytes) or stem cells (e.g. dendritic cell maturation, hematopoietic expansion, keratinocytes, mesenchymal stem cells or T cell expansion). The media may have supplements or reagents well known in the art, e.g. albumins and transport proteins, amino acids and vitamins, antibiotics, attachments factors, growth factors and cytokines, hormones, metabolic inhibitors or solubilizing agents. Various media are commercially available e. g. from ThermoFisher Scientific or Sigma-Aldrich. [00176] The term “microenvironment,” as used herein, may refer to the solid or hematological tumor microenvironment as a whole or to an individual subset of cells within the microenvironment. The tumor microenvironment, as used herein, refers to a complex mixture of “cells, soluble factors, signaling molecules, extracellular matrices, and mechanical cues that promote neoplastic transformation, support tumor growth and invasion, protect the tumor from host immunity, foster therapeutic resistance, and provide niches for dominant metastases to thrive,” as described in Swartz, et al., Cancer Res., 2012, 72, 2473. Although tumors express antigens that should be recognized by T cells, tumor clearance by the immune system is rare because of immune suppression by the microenvironment. [00177] The term “negatively separated” as used herein refers to the active separation of cells which are bound by one marker-binding fragment coupled to a solid phase and these cells are not the required population of cells. [00178] “Non-labelled” or “untouched” as used herein refers to the cells which are not bound by one marker-binding fragment coupled to a solid phase. The non-labelled, untouched cell fraction contains the desired target cells. [00179] “Non-target cells” as used herein refers to cells which are specifically bound by one marker-binding fragment which is coupled to a solid phase that is used to remove an unwanted cell type. [00180] “OKT-3” (also referred to herein as “OKT3”) refers to a monoclonal antibody or biosimilar or variant thereof, including human, humanized, chimeric, or murine antibodies, directed against the CD3 receptor in the T cell antigen receptor of mature T cells, and includes commercially-available forms such as OKT-3 (30 ng/mL, MACS GMP CD3 pure, Miltenyi Biotech, Inc., San Diego, Calif., USA) and muromonab or variants, conservative amino acid substitutions, glycoforms, or biosimilars thereof. [00181] “Particle” as used herein refers to a solid phase such as colloidal particles, microspheres, nanoparticles, or beads. Methods for generation of such particles are well known in the field of the art. The particles may be magnetic particles or have other selective properties. The particles may be in a solution or suspension or they may be in a lyophilized state prior to use in the present invention. The lyophilized particle is then reconstituted in convenient buffer before contacting the sample to be processed regarding the present invention. [00182] The terms “peripheral blood mononuclear cells” and “PBMCs” refers to a peripheral blood cell having a round nucleus, including lymphocytes (T cells, B cells, NK cells) and monocytes. Preferably, the peripheral blood mononuclear cells are irradiated allogeneic peripheral blood mononuclear cells. PBMCs are a type of antigen-presenting cell. [00183] The terms “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” are intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and inert ingredients. The use of such pharmaceutically acceptable carriers or pharmaceutically acceptable excipients for active pharmaceutical ingredients is well known in the art. Except insofar as any conventional pharmaceutically acceptable carrier or pharmaceutically acceptable excipient is incompatible with the active pharmaceutical ingredient, its use in the therapeutic compositions of the invention is contemplated. Additional active pharmaceutical ingredients, such as other drugs, can also be incorporated into the described compositions and methods. [00184] The term “population of cells” (including TILs) herein is meant a number of cells that share common traits. In general, populations generally range from l x l06 to l x l012 in number, with different TIL populations comprising different numbers. [00185] “Positively separated” as used herein refers to the active separation of cells which are bound by one marker-binding fragment coupled to a solid phase and these cells are the required population of cells. [00186] “Negatively separated” as used herein refers to the active separation of cells which are bound by one marker-binding fragment coupled to a solid phase and these cells are not the required population of cells. [00187] “Purity” as used herein refers to the percentage of the target population or populations desired from the original solid tissue. [00188] “Rapid expansion” means an increase in the number of antigen-specific TILs of at least about 3-fold (or 4-, 5-, 6-, 7-, 8-, or 9-fold) over a period of a week, more preferably at least about 10-fold (or 20-, 30-, 40-, 50-, 60-, 70-, 800-, or 90-fold) over a period of a week, more preferably at least about 100-fold (or 200-, 300-, 400-, 500-, 600-, 700-, 800-, or 900-fold) over a period of a week, or most preferably at least about 1000-fold or 2000-, 3000-, 4000-, 5000-, 6000-, 7000-, 8000-, or 9000-fold) over a period of a week. A number of rapid expansion protocols are outlined below. [00189] “Regenerative medicine(s)”, “adoptive cell therapy(ies)” or “advanced therapy medicinal product(s)” are used interchangeably herein to refer to cellular material that is used for therapeutic purposes of one or more mammals either by: the action of a part of or all of the cellular material; the supportive actions of a part of or all of the cellular material with the aim to improve the wellbeing of the mammal after application. The therapeutic cells can either be used directly or may require further processing, expansion and/or engineering to provide these actions. [00190] “Sample” as used herein refers to a sample containing cells in any ratio. Preferentially, these cells are viable. In some instances, these cells can also be fixed or frozen cells which may be used for subsequent nucleic acids or protein extraction. The samples may be from animals, especially mammals such as mouse, rats, or humans. Any compressible solid tissue that contains cells can be used. The invention is illustrated mainly through the isolation of hematopoietic and cancer cells from solid tumor tissue. However, the invention relates to a method for isolation of a breadth of cells from any mammalian solid tissue. [00191] “Solid phase” as used herein refers to the coupling of the marker-binding fragment, e.g. an antibody, bound to another substrate(s), e.g. particles, fluorophores, haptens like biotin, polymers, or larger surfaces such as culture dishes and microtiter plates. In some cases, the coupling results in direct immobilization of the antigen-binding fragment, e.g. if the antigen- binding fragment is coupled to a larger surface of a culture dish. In other cases, this coupling results in indirect immobilization, e.g. an antigen-binding fragment coupled directly or indirectly (via e.g. biotin) to a magnetic bead is immobilized if said bead is retained in a magnetic field. In further cases the coupling of the antigen-binding fragment to other molecules results not in a direct or indirect immobilization but allows for enrichment, separation, isolation, and detection of cells according to the present invention, e.g. if the marker-binding fragment is coupled to a chemical or physical moiety which then allows discrimination of labelled cells and non-labelled cells, e.g. via flow cytometry methods, like FACS sorting, or fluorescence microscopy. [00192] “Solid tissue” as used herein refers to a piece or pieces of animal derived mammalian solid tissue which by its three dimensions i.e. length, breadth and thickness as a geometrical body is larger than the size of multiple individual cell based units and often contains connective materials such as collagen or a similar matrix that make up structure of the tissue whereby said solid tissue cannot flow through tubes or be collected by a syringe or similar small conduit or receptacle and is i.e. with dimensions in the range of 500 μm, 1 mm, 2 mm, 3 mm, 4 mm, 5 mm, 1 cm, 2 cm, 3 cm, 4 cm, 5 cm, 10 cm, 20 cm, 30 cm, or more. [00193] “Solid tumor” refers to an abnormal mass of tissue that usually does not contain cysts or liquid areas. Solid tumors may be benign or malignant. The term "solid tumor cancer" refers to malignant, neoplastic, or cancerous solid tumors. Solid tumor cancers include, but are not limited to, sarcomas, carcinomas, and lymphomas, such as cancers of the lung, breast, prostate, colon, rectum, and bladder. The tissue structure of solid tumors includes interdependent tissue compartments including the parenchyma (cancer cells) and the supporting stromal cells in which the cancer cells are dispersed and which may provide a supporting microenvironment. In some embodiments, the cancer is selected from cervical cancer, head and neck cancer (including, for example, head and neck squamous cell carcinoma [HNSCC]) glioblastoma, ovarian cancer, sarcoma, pancreatic cancer, bladder cancer, breast cancer, triple negative breast cancer, and non- small cell lung carcinoma. The tissue structure of solid tumors includes interdependent tissue compartments including the parenchyma (cancer cells) and the supporting stromal cells in which the cancer cells are dispersed and which may provide a supporting microenvironment. [00194] By “thawed cryopreserved TILs” herein is meant a population of TILs that was previously cryopreserved and then treated to return to room temperature or higher, including but not limited to cell culture temperatures or temperatures wherein TILs may be administered to a patient. [00195] The terms “treatment”, “treating”, “treat”, and the like, refer to obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease. “Treatment”, as used herein, covers any treatment of a disease in a mammal, particularly in a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development or progression; and (c) relieving the disease, i.e., causing regression of the disease and/or relieving one or more disease symptoms. “Treatment” is also meant to encompass delivery of an agent in order to provide for a pharmacologic effect, even in the absence of a disease or condition. For example, “treatment” encompasses delivery of a composition that can elicit an immune response or confer immunity in the absence of a disease condition, e.g., in the case of a vaccine. [00196] By “tumor infiltrating lymphocytes” or “TILs” herein is meant a population of cells originally obtained as white blood cells that have left the bloodstream of a subject and migrated into a tumor. TILs include, but are not limited to, CD8+ cytotoxic T cells (lymphocytes), Thi and Thi 7 CD4+ T cells, natural killer cells, dendritic cells, and Ml macrophages. TILs include both primary and secondary TILs. “Primary TILs” are those that are obtained from patient tissue samples as outlined herein (sometimes referred to as “freshly harvested”), and “secondary TILs” are any TIL cell populations that have been expanded or proliferated as discussed herein, including, but not limited to bulk TILs and expanded TILs (“REP TILs” or “post-REP TILs”). TIL cell populations can include genetically modified TILs. TILs can generally be defined either biochemically, using cell surface markers, or functionally, by their ability to infiltrate tumors and effect treatment. TILs can be generally categorized by expressing one or more of the following biomarkers: CD4, CD8, TCR αβ, CD27, CD28, CD56, CCR7, CD45Ra, CD62L, CD95, PD-1, and CD25. Additionally and alternatively, TILs can be functionally defined by their ability to infiltrate solid tumors upon reintroduction into a patient. TILS may further be characterized by potency--for example, TILS may be considered potent or functional if in response to TCR engagement they produce, for example, interferon (IFN) release is greater than about 50 pg/mL, greater than about 100 pg/mL, greater than about 150 pg/mL, or greater than about 200 pg/mL, or more preferably individual cells can be Potency through intracellular staining for CD137, CD107a, INF-γ TNF-α, and IL-2 following TCR induced stimulation by flow cytometry. [00197] “Retentate” as used herein refers to the material that does not pass through a filter, mesh or membrane. [00198] “Ultimate utility” as used herein refers to manufacture of or direct use in regenerative medicines, adoptive cell therapies, ATMPs, diagnostic in vitro studies or scientific research. [00199] The terms “protein,” “polypeptide,” and “peptide,” used interchangeably herein, include polymeric forms of amino acids of any length, including coded and non-coded amino acids and chemically or biochemically modified or derivatized amino acids. The terms also include polymers that have been modified, such as polypeptides having modified peptide backbones. The term “domain” refers to any part of a protein or polypeptide having a particular function or structure. [00200] Proteins are said to have an “N-terminus” (amino-terminus) and a “C-terminus” (carboxy-terminus or carboxyl-terminus). The term “N-terminus” relates to the start of a protein or polypeptide, terminated by an amino acid with a free amine group (-NH2). The term “C- terminus” relates to the end of an amino acid chain (protein or polypeptide), terminated by a free carboxyl group (-COOH). [00201] The terms “nucleic acid” and “polynucleotide,” used interchangeably herein, include polymeric forms of nucleotides of any length, including ribonucleotides, deoxyribonucleotides, or analogs or modified versions thereof. They include single-, double-, and multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, and polymers comprising purine bases, pyrimidine bases, or other natural, chemically modified, biochemically modified, non-natural, or derivatized nucleotide bases. Likewise, DNA and RNA can include natural, chemically modified, biochemically modified, non-natural, or derivatized nucleotide bases. [00202] Nucleic acids are said to have “5’ ends” and “3’ ends” because mononucleotides are reacted to make oligonucleotides in a manner such that the 5’ phosphate of one mononucleotide pentose ring is attached to the 3’ oxygen of its neighbor in one direction via a phosphodiester linkage. An end of an oligonucleotide is referred to as the “5’ end” if its 5’ phosphate is not linked to the 3’ oxygen of a mononucleotide pentose ring. An end of an oligonucleotide is referred to as the “3’ end” if its 3’ oxygen is not linked to a 5’ phosphate of another mononucleotide pentose ring. A nucleic acid sequence, even if internal to a larger oligonucleotide, also may be said to have 5’ and 3’ ends. In either a linear or circular DNA molecule, discrete elements are referred to as being “upstream” or 5’ of the “downstream” or 3’ elements. [00203] The term “viral vector” refers to a recombinant nucleic acid that includes at least one element of viral origin and includes elements sufficient for or permissive of packaging into a viral vector particle. The vector and/or particle can be utilized for the purpose of transferring DNA, RNA, or other nucleic acids into cells in vitro, ex vivo, or in vivo. Numerous forms of viral vectors are known. [00204] The term “isolated” with respect to cells, tissues, proteins, and nucleic acids includes cells, tissues, proteins, and nucleic acids that are relatively purified with respect to other bacterial, viral, cellular, or other components that may normally be present in situ, up to and including a substantially pure preparation of the cells, tissues, proteins, and nucleic acids. The term “isolated” also includes cells, tissues, proteins, and nucleic acids that have no naturally occurring counterpart, have been chemically synthesized and are thus substantially uncontaminated by other cells, tissues, proteins, and nucleic acids, or has been separated or purified from most other components (e.g., cellular components or organism components) with which they are naturally accompanied (e.g., other cellular proteins, nucleic acids, or cellular or extracellular components). [00205] “Exogenous” molecules or sequences include molecules or sequences that are not normally present in a cell in that form or are provided to a cell from an external source. Normal presence includes presence with respect to the particular developmental stage and environmental conditions of the cell. An exogenous molecule or sequence, for example, can include a mutated version of a corresponding endogenous sequence within the cell or can include a sequence corresponding to an endogenous sequence within the cell but in a different form (i.e., not within a chromosome). In contrast, endogenous molecules or sequences include molecules or sequences that are normally present in that form in a particular cell at a particular developmental stage under particular environmental conditions. [00206] Compositions or methods “comprising” or “including” one or more recited elements may include other elements not specifically recited. For example, a composition that “comprises” or “includes” a protein may contain the protein alone or in combination with other ingredients. The transitional phrase “consisting essentially of” means that the scope of a claim is to be interpreted to encompass the specified elements recited in the claim and those that do not materially affect the basic and novel characteristic(s) of the claimed invention. Thus, the term “consisting essentially of” when used in a claim of this invention is not intended to be interpreted to be equivalent to “comprising.” [00207] “Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur and that the description includes instances in which the event or circumstance occurs and instances in which the event or circumstance does not. [00208] Designation of a range of values includes all integers within or defining the range, and all subranges defined by integers within the range. [00209] Unless otherwise apparent from the context, the term “about” encompasses values ± 5% of a stated value. [00210] The term “and/or” refers to and encompasses any and all possible combinations of one or more of the associated listed items, as well as the lack of combinations when interpreted in the alternative (“or”). [00211] The term “or” refers to any one member of a particular list. [00212] The singular forms of the articles “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. For example, the term “a protein” or “at least one protein” can include a plurality of proteins, including mixtures thereof. [00213] Statistically significant means p ≤0.05. DETAILED DESCRIPTION [00214] Provided herein are methods for preparing a therapeutic population of tumor infiltrating lymphocytes (TILs), populations of TILs produced by the methods, and methods of using the TILs to treat cancer in a subject. The methods comprise treating a first population of TILs with one or more compounds to improve T-cell fitness. The compounds and methods described herein can be used, for example, in combination with the methods disclosed in WO 2021/123832, herein incorporated by reference in its entirety for all purposes. [00215] TIL therapy clinical responses are limited by T-cell dysfunction and exhaustion. T- cells and TILs gradually lose functionality and proliferation potential during in vitro or in vivo antigen stimulation and expansion, a process known as T cell differentiation or exhaustion. Prior to excision, TILs in tumor tissue might be already differentiated as a result of chronic tumor antigen stimulation. During manufacturing, TILs are massively expanded in vitro, which leads to further differentiation and exhaustion. The methods described herein can be used to increase TIL persistence and functionality by improving T-cell fitness. Increased fitness can be defined as one or a combination of the following: increasing expansion/proliferation potential (longer persistence), inducing and maintaining a favorable phenotype (better functionality), and preserving tumor-reactivity. [00216] In the methods disclosed herein, rejuvenating factors (compounds) can be introduced at any point during the TIL isolation and ex vivo expansion process. As disclosed in more detail elsewhere herein, such rejuvenating factors can be transgenes (e.g., lentivirus or lentiviral vector, or electroporation of mRNA), gene knockout compounds (e.g., electroporation of CRISPR/Cas ribonucleoprotein (RNP) complex), gene knockdown compounds (e.g., lentivirus or lentiviral vector, or electroporation of siRNA or ASO), or small molecules or cytokines. Single rejuvenating factors or compound can be used, or a combination can be used, and the TILs can be treated early in the process or late in the process. [00217] T cells are derived from hematopoietic stem cells resident in bone marrow but subsequently migrate to and mature in the thymus. During the process of maturation, T cells undergo a series of selection events, thereby generating a diverse repertoire of T cells. These cells are then released into the peripheral circulation to carry out their specific functions as a part of the adaptive immune system [00218] T cells are not a homogeneous group of cells but consist of many lineages, of which the predominant types are defined by the expression of two further cell markers. CD4 expressing T cells are generally termed helper (Th) and are thought to orchestrate many functions of the immune system by cell-cell contact and through the production of mediator molecules called cytokines. CD8 T cells are considered to be cytotoxic (Tc) and are thought to be the cells which perform direct killing of target cells. These activities are all controlled through the T cell receptor/antigen/MHC interaction – consequently, upon successful recognition of a peptide/MHC on a target cell, CD4 and CD8 cells act in concert through cytokine production and cytotoxic activity to eliminate target cells, including virus infected and tumor cells. [00219] T cells do not recognize intact proteins (antigens) but respond to short, protein fragments presented on the surface of target cells by specific proteins called the Major Histocompatibility Complex (MHC). During the maturation process, T cells express on their cell surface an antigen-specific T cell receptor (TCR), which recognizes these short protein (peptide) antigens presented by MHC molecules. Consequently, only when the correct peptide is presented on the surface of a target cell associated with the correct MHC molecule will the T cell activate its effector functions. Therefore, the frequency of tumor specific T cells are enriched in the tumor making it an ideal source for tumor specific T cells i.e. tumor-infiltrating lymphocytes (TIL) (Andersen et al., Cancer Res.2012 Apr 1;72(7):1642-50. doi: 10.1158/0008-5472.CAN- 11-2614. Epub 2012 Feb 6). [00220] Of course, this is a highly simplified view and represents a short general overview of T cell function. The adaptive immune response does not act in isolation but requires extensive interaction with a range of immune and non-immune cells to facilitate the efficient trafficking of T cells to the required site of activity, to ensure that the correct immune response is initiated and that the immune response is controlled and turned off after it is needed. Therefore, even in patients where the manufactured TIL initiate an immune response to the tumor it may then be supported or dampened by the patient’s own immune system and the tumor environment. [00221] Tumor specific TIL are T cells isolated from a tumor of a patient with primary or metastatic cancer. In most cancer patients circulating tumor-specific T cells can hardly be detected in blood. However, certain cancers such as cutaneous melanoma appear to be immunogenic as it has the ability to induce significant numbers of T cells with anti-tumor activity during the natural course of the tumor growth, especially within the tumor areas (Muul et al., J Immunol.1987 Feb 1;138(3):989-95). Tumor-reactive T cells “selected as T cell specific for the tumor” can be isolated from tumor material and expanded ex vivo into high numbers. Reports have shown that these cells contain anti-tumor reactivity, which can result in tumor destruction and clinical responses upon reinfusion into the patient (Dudley et al., Science.2002 Oct 25;298(5594):850-4. Epub 2002 Sep 19). In subsequent trials the importance of T cell characteristics was confirmed and the benefit of “young” rapidly growing cells “Young TILs” was confirmed whereby cells are “not selected for specificity” at all. Remarkably this produces excellent response rates in TIL or CD8 selected TIL of around 50% (Besser et al., Anticancer Res.2009 Jan;29(1):145-54; Dudley et al., Clin Cancer Res.2010 Dec 15;16(24):6122-31. doi: 10.1158/1078-0432.CCR-10-1297. Epub 2010 Jul 28). [00222] Studies by Andersen et al. (Cancer Res.2012 Apr 1;72(7):1642-50. doi: 10.1158/0008-5472.CAN-11-2614. Epub 2012 Feb 6) identified that melanoma specific T cells (for known cancer antigens) are enriched within the tumor compared with T cells in the peripheral blood. This supports the dogma that the isolated TIL population are enriched tumor specific T cells resulting in an enhanced anti-tumor activity when compared with early trials in melanoma patients using T cells isolated from peripheral blood and expanded in similar levels of IL2 or intravenous IL-2 alone (LAK cells – Bordignon et al., Haematologica.1999 Dec;84(12):1110-49). [00223] US Patent No.10,398,734 relates to methods for expanding TILs and producing therapeutic populations of TILs. The tumor of the ‘734 patent is shipped as a bulk tumor, and the TILs inside the bulk tumor rapidly become oxygen deficient and deteriorate progressively over time. The tumor of the ‘734 patent is also processed to fragments which have deteriorated internal cell populations. Furthermore, the TILs used for manufacturing will only be TILs expanded from tissue fragments and not any TILs retained in the interior. Therefore, the resulting cell population may not reflect the full diversity of the tumor microenvironment. [00224] Harvesting TILs requires the aseptic disaggregation of solid tissue as a bulk tumor prior to the culture and expansion of the TIL population. The conditions during solid tissue disaggregation and time taken to harvest the cells have a substantial impact on the viability and recovery of the final cellularized material. A solid tissue derived cell suspension that is obtained using conventional methods often includes a wide variety of different cell types, disaggregation media, tissue debris and/or fluids. This may necessitate the use of selective targeting and/or isolation of cell types, for example, prior to manufacture of regenerative medicines, adoptive cell therapies, ATMPs, diagnostic in vitro studies and/or scientific research. [00225] Currently, selection or enrichment techniques generally utilize one of: size, shape, density, adherence, strong protein-protein interactions (i.e. antibody-antigen interactions). For example, in some instances selection may be conducted by providing a growth supporting environment and by controlling the culture conditions or more complex cell marker interactions associated with semi-permanent or permanent coupling to magnetic or non-magnetic solid or semi-solid phase substrates. [00226] For enrichment, isolation, or selection, any sorting technology can be used, for example, affinity chromatography or any other antibody-dependent separation technique known in the art. Any ligand-dependent separation technique known in the art may be used in conjunction with both positive and negative separation techniques that rely on the physical properties of the cells. An especially potent sorting technology is magnetic cell sorting. Methods to separate cells magnetically are commercially available e.g. from Thermo Fisher, Miltenyi Biotech, Stemcell Technologies, Cellpro Seattle, Advanced Magnetics, Boston Scientific, or Quad Technologies. For example, monoclonal antibodies can be directly coupled to magnetic polystyrene particles like Dynal M 450 or similar magnetic particles and used, for example for cell separation. The Dynabeads technology is not column based, instead these magnetic beads with attached cells enjoy liquid phase kinetics in a sample tube, and the cells are isolated by placing the tube on a magnetic rack. [00227] Enriching, sorting and/or detecting cells from a sample includes using monoclonal antibodies in conjunction with colloidal superparamagnetic microparticles having an organic coating of, for example, polysaccharides (e.g. magnetic-activated cell sorting (MACS) technology (Miltenyi Biotec, Bergisch Gladbach, Germany)). Particles (e.g., nanobeads or MicroBeads) can be either directly conjugated to monoclonal antibodies or used in combination with anti-immunoglobulin, avidin, or anti-hapten-specific MicroBeads, or coated with other mammalian molecules with selective binding properties. [00228] Magnetic particle selection technologies such as those described above, allows cells to be positively or negatively separated by incubating them with magnetic nanoparticles coated with antibodies or other moieties directed against a particular surface marker. This causes the cells expressing this marker to attach to the magnetic nanoparticles. Afterwards the cell solution is placed within a solid or flexible container in a strong magnetic field. In this step, the cells attach to the nanoparticles (expressing the marker) and stay on the column, while other cells (not expressing the marker) flow through. With this method, the cells can be separated positively or negatively with respect to the particular marker(s). [00229] In case of a positive selection the cells expressing the marker(s) of interest, which attached to the magnetic column, are washed out to a separate vessel, after removing the column from the magnetic field. [00230] In case of a negative selection the antibody or selective moiety used is directed against surface markers(s) which are known to be present on cells that are not of interest. After application of the cells/magnetic nanoparticles solution onto the column the cells expressing these antigens bind to the column and the fraction that goes through is collected, as it contains the cells of interest. As these cells are non-labelled by the selective antibodies or moiety(s) coupled to nanoparticles, they are “untouched”. The known manual or semi-automated solid tissue processing steps are labor-intensive and require a knowledge of the art. [00231] In addition, where the material is used for therapeutic purposes, the processing requires strict regulated environmental conditions during handling of the cell cultures, for example tissue processing as a part of or prior to disaggregation, enzymatic digestion and transfer into storing devices, or incubation conditions for disaggregation/cellularization and viable tissue yields. Typically, this process would require multiple pieces of laboratory and tissue processing equipment, and personnel with the skills and knowledge of the scientific art with critical stages contained within either hazard containment or tissue processing facility(s) aseptic environment(s) in order to perform the same activity safely and also minimize the risk of contamination(s). [00232] Provided herein are methods of preparing a therapeutic population of tumor infiltrating lymphocytes (TILs). As described in more detail elsewhere herein, the methods can further administering a therapeutic amount of the therapeutic population of TILs to a subject with a cancer to treat the subject. Some such methods comprise treating a first population of TILs with a compound or one or more compounds to improve T-cell fitness (i.e., an effective amount of the compound or the one or more compounds in order to improve T-cell fitness as described herein). The methods can comprise treating the first population of TILs with the compound or the combination of compounds a single time or multiple times (e.g., at least 2 times, at least 3 times, at least 4 times, or at least 5 times). If a combination of compounds is used, the TILs can be treated with the compounds simultaneously or at different times. Also provided herein are populations of isolated TILs produced by such methods, or isolated populations of TILs that comprise the compound or the combination of compounds to improve T-cell fitness. Also provided are in vitro cultures comprising a population of isolated TILs, the compound or combination of compounds (i.e., exogenous compound or combination of exogenous compounds), and a culture medium suitable for maintaining the TILs in culture. Such culture media are well known. [00233] The compounds can be any suitable agent or inhibitory agent such as a nucleic acid such as DNA or messenger RNA, an antigen-binding protein such as an antibody, an inhibitory RNA such as an antisense oligonucleotide or an RNAi agent, a nuclease agent, or a small molecule inhibitor. [00234] Any one or more of the several successive molecular mechanisms involved in the expression of a given gene or polypeptide may be targeted as intended herein. Without limitation, these may include targeting the gene sequence (e.g., targeting the polypeptide- encoding, non-coding and/or regulatory portions of the gene sequence), the transcription of the gene into RNA, the polyadenylation and where applicable splicing and/or other post- transcriptional modifications of the RNA into mRNA, the localization of the mRNA into cell cytoplasm, where applicable other post-transcriptional modifications of the mRNA, the translation of the mRNA into a polypeptide chain, where applicable post-translational modifications of the polypeptide, and/or folding of the polypeptide chain into the mature conformation of the polypeptide. For compartmentalized polypeptides, such as secreted polypeptides and transmembrane polypeptides, this may further include targeting trafficking of the polypeptides, i.e., the cellular mechanism by which polypeptides are transported to the appropriate sub-cellular compartment or organelle, membrane, e.g. the plasma membrane, or outside the cell. [00235] In certain embodiments, the invention comprises mRNA transfection to modify T cells. Rabinowitz et al. (2009) Hum. Gene Ther.20:51–61, describes transfection of in vitro transcribed (IVT) CAR mRNA into CD3+/CD4+ T cells and CD3+/CD8+ T cells by electroporation, resulting in high levels of surface expression of CARs. Rabinowitz exemplifies an mRNA transfection method useful for carrying out the instant invention. Lissandrello et al. (2020) Sci Rep. Oct 22;10(1):18045 describes a microfluidic continuous-flow electrotransfection device for delivery of mRNA into primary human T cells with up to 95% transfection efficiency with samples comprising up to 500 million T cells. Lissandrello exemplifies a bulk mRNA transfection method useful for carrying out the instant invention. [00236] The present invention includes production of lentivirus modified TILs. Lentiviruses are complex retroviruses that have the ability to infect and express their genes in both mitotic and post-mitotic cells. The most commonly known lentivirus is the human immunodeficiency virus (HIV). Lentiviruses may be prepared as follows. After cloning pSF-LV or equivalent lentivirus transfer plasmid backbone to contain the transgene and promoter of interest, HEK293FT at low passage (p=5) can be seeded in a T-75 flask to 50% confluence the day before transfection in DMEM with 10% fetal bovine serum and without antibiotics. After 20 hours, media can be changed to OptiMEM (serum-free) media and transfection can be done 4 hours later. Cells can be transfected with 10.μg of lentiviral transfer plasmid (pCasES10) and the following packaging plasmids: 5 µg of pMD2.G (VSV-g pseudotype), and 7.5 ug of psPAX2 (gag/pol/rev/tat). Transfection can be done in 4 mL OptiMEM with a cationic lipid delivery agent (50 uL Lipofectamine 2000 and 100 ul Plus reagent). After 6 hours, the media can be changed to antibiotic-free DMEM with 10% fetal bovine serum. These methods use serum during cell culture, but serum-free methods can be preferred. [00237] Lentivirus may be purified as follows. Viral supernatants can be harvested after 48 hours. Supernatants can be first cleared of debris and filtered through a 0.45 µm low protein binding (PVDF) filter. They can then be centrifuged in an ultracentrifuge for 2 hours at 24,000 rpm. Viral pellets can be resuspended in 50 µL of DMEM overnight at 4° C. They can then be aliquoted and immediately frozen at -80° C. [00238] TILs can be obtained from surgical specimens. PBLs can be thawed from frozen stock stored at −180°C and placed into culture in AIM-V and interleukin-2 (IL-2; Cetus, Emeryville, CA) at 300 IU/ml. For OKT3 stimulation, the cells can be either initially placed in medium with anti-CD3 antibody, OKT3 (Ortho Biotech, Bridgewater, NJ) at 50 ng/ml, or can be placed in OKT3 medium after transduction, at the initial changing of the culture medium. For transduction of the PBLs or TILs, 1 × 106 cells can be adjusted to a final volume of 1 mL in a 24- well tissue culture-treated plate with the viral supernatant and Polybrene (final concentration, 8 μg/ml). The cells can be transduced by centrifugation of the plates for 1.5 hr at 1000 × g, 32°C. The plates can be placed in a 37°C, humidified 5% CO2 incubator overnight, and the medium can be replaced the next day. TILs can be subject to the rapid expansion protocol (REP) as previously described, using OKT3 (50 ng/ml), IL-2 (5000 IU/ml), and irradiated allogeneic peripheral blood mononuclear cells from three different donors (TIL:feeder ratio, 1:100). [00239] In a particularly advantageous embodiment, cryopreserved TILs can be thawed and about 2, 3, 4, 5, 6 or 7 days after outgrowth, the TILs can be subject to lentivirus transduction. Advantageously, transduction with lentivirus is performed at an MOI of about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 based on the total number of live cells. Advantageously, the MOI is about 5. IL-2 is added at a concentration of 1000, 2000, 3000, 4000, 5000 or 6000 IU/mL. Advantageously the concentration of IL-2 is about 3000 IU/mL. After a rapid expansion protocol (REP), optionally using OKT3 (50 ng/ml), IL-2 (5000 IU/ml), and/or irradiated allogeneic peripheral blood mononuclear cells (TIL:feeder ratio, 1:200), the lentivirus modified cells can be cryopreserved, subjected to outgrowth, and REP, then cryopreserved. When required, lentivirus modified TILs can be thawed and infused into a lymphodepleted patient, optionally prior to IL-2 infusion. [00240] A non-limiting representative methodology is as follows: [00241] On day 0, digested tumor samples from each donor were thawed in complete TIL TCM, cells are washed once by centrifuging at 400 x g for 5 minutes, resuspended in fresh TIL TCM and counted using DRAQ7 dye and anti-CD2 antibody stains. Cells are then centrifuged at 400 x g for 5 minutes, resuspended at a concentration of 1 x 106 cells/mL, placed into an appropriate vessel with 3000 IU/mL IL-2 and rested for two days in a 5% CO2 incubator set to 37°C. [00242] On day 2, cells are collected, washed, centrifuged at 400 x g for 5 minutes, and resuspended in fresh complete TCM. The number of viable cells in each sample was determined, cells centrifuged at 400 x g for 5 minutes and resuspended at a concentration of 1 x 106 cells/mL. Transduction with lentivirus is performed at an MOI of 5 based on the total number of live cells. IL-2 is added at a concentration of 3000 IU/mL and cells are placed in a 5% CO2 incubator set to 37°C. [00243] On day 3, the cells were collected, washed once with complete TIL TCM, and resuspended in the same volume of fresh complete TIL TCM as on day 2 for the second day of transduction. Transduction is performed using lentivirus at an MOI of 5 and IL-2 at 3000 IU/mL was added to the cells prior to placing them in a 5% CO2 incubator set to 37°C for 8 days. IL-2 (3000 IU/mL) was added to the cells every 2-3 days until D12. [00244] On day 12 cells are collected, washed, resuspended in complete TIL TCM and counted. After determining the TIL numbers, cells are seeded in appropriate scale G-REX plates for REP using pooled irradiated allogeneic PBMCs from 10 healthy donors as feeders at a 200:1 ratio of feeders:TIL. The media used for the REP is complete REP TIL TCM with anti-CD3 (OKT3) antibody added at a concentration of 30 ng/mL for activation. IL-2 is added at a concentration of 3000 IU/mL and cells are placed in a 5% CO2 incubator set to 37°C. The REP period was 12 days during which IL-2 (3000 IU/mL) was supplemented every 2-3 days. [00245] On day 18 (i.e., day 6 of REP), 5 mL of medium from the 24 well G-REX plates or 25 mL of medium from the 6 well G-REX plates is removed without disturbing the cells and replaced with fresh complete REP TIL TCM and IL-2 (3000 IU/mL). On D24, TILs are harvested by centrifugation at 400 x g for 5 minutes and resuspended in fresh media for counting. TILs are resuspended in cryoprotectant and aliquoted to cryovials, cooled to -80°C overnight, and then transferred to -150 °C for short term storage. [00246] The term “antigen-binding protein” includes any protein that binds to an antigen. Examples of antigen-binding proteins include an antibody, an antigen-binding fragment of an antibody, a multispecific antibody (e.g., a bi-specific antibody), an scFV, a bis-scFV, a diabody, a triabody, a tetrabody, a V-NAR, a VHH, a VL, a F(ab), a F(ab)2, a DVD (dual variable domain antigen-binding protein), an SVD (single variable domain antigen-binding protein),or a bispecific T-cell engager (BiTE). [00247] Nuclease agents can include, for example, a Clustered Regularly Interspersed Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas) (CRISPR/Cas) nuclease, a zinc finger nuclease (ZFN), or a Transcription Activator-Like Effector Nuclease (TALEN) that cleaves a target site in target gene. CRISPR/Cas nuclease comprise a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene. The CRISPR/Cas system can be, for example, a type II system (e.g., Cas9) or a type V system (e.g., Cas12a). See, e.g., Zetsche et al. (2015) Cell 163(3):759-771, WO 2013/176772, WO 2014/065596, WO 2016/106121, and WO 2019/067910, each of which is herein incorporated by reference in its entirety for all purposes. [00248] The compounds used herein may be DNA targeting agents. DNA targeting herein may be the specific introduction of a knock-out, edit, or knock-in at a particular DNA sequence, such as in a chromosome of a cell, using methods known in the art. A knock-out as used herein represents a DNA sequence of a cell that has been rendered partially or completely inoperative by targeting using methods known in the art, such as with a Cas protein as described above. Such a DNA sequence prior to knock-out could have encoded an amino acid sequence, or could have had a regulatory function (e.g., promoter), for example. A knock-out may be produced by an indel (insertion or deletion of nucleotide bases in a target DNA sequence through NHEJ), or by specific removal of sequence that reduces or completely destroys the function of sequence at or near the targeting site. A knock-in represents the replacement or insertion of a DNA sequence at a specific DNA sequence in cell by targeting using methods known in the art, such as with a Cas protein. Examples of knock-ins include, but are not limited to, a specific insertion of a heterologous amino acid coding sequence in a coding region of a gene, or a specific insertion of a transcriptional regulatory element in a genetic locus. [00249] An “RNAi agent” is a composition that comprises a small double-stranded RNA or RNA-like (e.g., chemically modified RNA) oligonucleotide molecule capable of facilitating degradation or inhibition of translation of a target RNA, such as messenger RNA (mRNA), in a sequence-specific manner. The oligonucleotide in the RNAi agent is a polymer of linked nucleosides, each of which can be independently modified or unmodified. RNAi agents operate through the RNA interference mechanism (i.e., inducing RNA interference through interaction with the RNA interference pathway machinery (RNA-induced silencing complex or RISC) of mammalian cells). While it is believed that RNAi agents, as that term is used herein, operate primarily through the RNA interference mechanism, the disclosed RNAi agents are not bound by or limited to any particular pathway or mechanism of action. RNAi agents disclosed herein comprise a sense strand and an antisense strand, and include, but are not limited to, short interfering RNAs (siRNAs), double-stranded RNAs (dsRNA), micro RNAs (miRNAs), short hairpin RNAs (shRNA), and dicer substrates. The antisense strand of the RNAi agents described herein is at least partially complementary to a sequence (i.e., a succession or order of nucleobases or nucleotides, described with a succession of letters using standard nomenclature) in the target RNA. [00250] Single-stranded antisense oligonucleotides (ASOs) and RNA interference (RNAi) share a fundamental principle in that an oligonucleotide binds a target RNA through Watson- Crick base pairing. Without wishing to be bound by theory, during RNAi, a small RNA duplex (RNAi agent) associates with the RNA-induced silencing complex (RISC), one strand (the passenger strand) is lost, and the remaining strand (the guide strand) cooperates with RISC to bind complementary RNA. Argonaute 2 (Ago2), the catalytic component of the RISC, then cleaves the target RNA. The guide strand is always associated with either the complementary sense strand or a protein (RISC). In contrast, an ASO must survive and function as a single strand. ASOs bind to the target RNA and block ribosomes or other factors, such as splicing factors, from binding the RNA or recruit proteins such as nucleases. Different modifications and target regions are chosen for ASOs based on the desired mechanism of action. A gapmer is an ASO oligonucleotide containing 2–5 chemically modified nucleotides (e.g. LNA or 2’-MOE) on each terminus flanking a central 8–10 base gap of DNA. After binding the target RNA, the DNA-RNA hybrid acts substrate for RNase H. [00251] The treating or the compound(s) can have, for example, one or more (any combination thereof) or all of the following effects: (1) decreases expression or activity of Fas (FAS) or FasL (FASLG) (e.g., a FAS/FASLG inhibitory agent); (2) decreases expression or activity of a TGFβR (e.g., TGFβR1) or the TGFβ signaling pathway (e.g., a TGFβ/TGFβR1 inhibitory agent); (3) decreases expression or activity of interferon regulatory factor 7 (IRF7) (e.g., an IRF7 inhibitory agent); (4) decreases expression or activity of DNA-directed RNA polymerase III subunit RPC1 (POLR3A) (e.g., a POLR3A inhibitory agent); (5) decreases expression or activity of ETV7; (6) decreases expression or activity of ETV3; (7) decreases expression or activity of ASH2L; (8) decreases expression or activity of PML; (9) decreases expression or activity of STAT2; (10) decreases expression or activity of SPI1; (11) decreases expression or activity of IRF9; (12) decreases expression or activity of STAT1; (13) decreases expression or activity of IRF4; (14) decreases expression or activity of JDP2; (15) decreases expression or activity of ZNF337; (16) decreases expression or activity of ETV2; (17) decreases expression or activity of ETV3L; (18) decreases expression or activity of SOX18; (19) decreases expression or activity of CEBPG; (20) decreases expression or activity of CREB3L4; (21) decreases expression or activity of CEBPB; (22) decreases expression or activity of FOXD1; (23) decreases expression or activity of EOMES; and (24) decreases expression or activity of ZNF683. In some embodiments, the treating or the compound(s) can have 1 of the above effects. In some embodiments, the treating or the compound(s) can have 2 of the above effects. In some embodiments, the treating or the compound(s) can have 3 of the above effects. In some embodiments, the treating or the compound(s) can have 4 of the above effects. In some embodiments, the treating or the compound(s) can have 5 of the above effects. In some embodiments, the treating or the compound(s) can have 6 of the above effects. In some embodiments, the treating or the compound(s) can have 7 of the above effects. In some embodiments, the treating or the compound(s) can have 8 of the above effects. In some embodiments, the treating or the compound(s) can have 9 of the above effects. In some embodiments, the treating or the compound(s) can have 10 of the above effects. In some embodiments, the treating or the compound(s) can have 11 of the above effects. In some embodiments, the treating or the compound(s) can have 12 of the above effects. In some embodiments, the treating or the compound(s) can have 13 of the above effects. In some embodiments, the treating or the compound(s) can have 14 of the above effects. In some embodiments, the treating or the compound(s) can have 15 of the above effects. In some embodiments, the treating or the compound(s) can have 16 of the above effects. In some embodiments, the treating or the compound(s) can have 17 of the above effects. In some embodiments, the treating or the compound(s) can have 18 of the above effects. In some embodiments, the treating or the compound(s) can have 19 of the above effects. In some embodiments, the treating or the compound(s) can have 20 of the above effects. In some embodiments, the treating or the compound(s) can have 21 of the above effects. In some embodiments, the treating or the compound(s) can have 22 of the above effects. In some embodiments, the treating or the compound(s) can have 23 of the above effects. In some embodiments, the treating or the compound(s) can have 24 of the above effects. [00252] The treating or the compound(s) can have, for example, one or more or all of the following effects: (i) decreases expression or activity of Fas (FAS) or FasL (FASLG) (e.g., a FAS/FASLG inhibitory agent); (ii) decreases expression or activity of a TGFβR (e.g., TGFβR1) or the TGFβ signaling pathway (e.g., a TGFβ/TGFβR1 inhibitory agent); (iii) decreases expression or activity of interferon regulatory factor 7 (IRF7) (e.g., an IRF7 inhibitory agent); and (iv) decreases expression or activity of DNA-directed RNA polymerase III subunit RPC1 (POLR3A) (e.g., a POLR3A inhibitory agent). In some embodiments, the treating or the compound(s) can have effect (i). In some embodiments, the treating or the compound(s) can have effect (ii). In some embodiments, the treating or the compound(s) can have effect (iii). In some embodiments, the treating or the compound(s) can have effect (iv). In some embodiments, the treating or the compound(s) can have effects (i) and (ii). In some embodiments, the treating or the compound(s) can have effects (i) and (iii). In some embodiments, the treating or the compound(s) can have effects (i) and (iv). In some embodiments, the treating or the compound(s) can have effects (ii) and (iii). In some embodiments, the treating or the compound(s) can have effects (ii) and (iv). In some embodiments, the treating or the compound(s) can have effects (iii) and (iv). In some embodiments, the treating or the compound(s) can have effects (i), (ii), and (iii). In some embodiments, the treating or the compound(s) can have effects (i), (ii), and (iv). In some embodiments, the treating or the compound(s) can have effects (i), (iii), and (iv). In some embodiments, the treating or the compound(s) can have effects (ii), (iii), and (iv). In some embodiments, the treating or the compound(s) can have effects (i), (ii), (iii), and (iv). [00253] Likewise, the one or more compounds (i.e., exogenous compounds) can comprise one or more or all of the following: (i) a FAS/FASLG inhibitory agent; (ii) a TGFβ/TGFβR1 inhibitory agent; (iii) an IRF7 inhibitory agent; and (iv) a POLR3A inhibitory agent. A FAS/FASLG inhibitory agent can be anything that decreases FAS or FASLG expression or activity or that decreases FAS signaling pathway activity. A TGFβ/TGFβR1 inhibitory agent can be anything that decreases TGFβR1 or TGFβ1 expression or activity or that decreases TGFβ signaling pathway activity. An IRF7 inhibitory agent can be anything that decreases IRF7 expression or activity. A POLR3A inhibitory agent can be anything that decreases POLR3A expression or activity. Examples of suitable inhibitory agents are provided in more detail below. In some embodiments, the compound(s) can comprise a FAS/FASLG inhibitory agent (i.e., FASLG signaling pathway inhibitory agent). In some embodiments, the compounds can comprise a TGFβ/TGFβR1 inhibitory agent (i.e., a TGFβ signaling pathway inhibitory agent). In some embodiments, the compound(s) can comprise an IRF7 inhibitory agent. In some embodiments, the compound(s) can comprise a POLR3A inhibitory agent. In some embodiments, the compounds can comprise a FAS/FASLG inhibitory agent and a TGFβ/TGFβR1 inhibitory agent. In some embodiments, the compounds can comprise a FAS/FASLG inhibitory agent and an IRF7 inhibitory agent, the compounds can comprise a FAS/FASLG inhibitory agent and a POLR3A inhibitory agent. In some embodiments, the compounds can comprise a TGFβ/TGFβR1 inhibitory agent and an IRF7 inhibitory agent. In some embodiments, the compounds can comprise a TGFβ/TGFβR1 inhibitory agent and a POLR3A inhibitory agent. In some embodiments, the compounds can comprise an IRF7 inhibitory agent and a POLR3A inhibitory agent. In some embodiments, the compounds can comprise a FAS/FASLG inhibitory agent, a TGFβ/TGFβR1 inhibitory agent, and an IRF7 inhibitory agent. In some embodiments, the compounds can comprise a FAS/FASLG inhibitory agent, a TGFβ/TGFβR1 inhibitory agent, and a POLR3A inhibitory agent. In some embodiments, the compounds can comprise a FAS/FASLG inhibitory agent, an IRF7 inhibitory agent, and a POLR3A inhibitory agent. In some embodiments, the compounds can comprise a TGFβ/TGFβR1 inhibitory agent, an IRF7 inhibitory agent, and a POLR3A inhibitory agent. In some embodiments, the compounds can comprise a FAS/FASLG inhibitory agent, a TGFβ/TGFβR1 inhibitory agent, an IRF7 inhibitory agent, and a POLR3A inhibitory agent. [00254] In one example, the treating or compound(s) decrease expression or activity of FAS or FASLG. For example, the treating or compound(s) can transiently decrease expression or activity of FAS or FASLG. Alternatively, the treating or compound(s) can permanently decrease expression or activity of FAS or FASLG. [00255] FAS (tumor necrosis factor receptor superfamily member 6; UniProt ID P25445) is encoded by FAS (NCBI GeneID 355). It is the receptor for TNFSF6/FASLG. The adapter molecule FADD recruits caspase-8 to the activated receptor. The resulting death-inducing signaling complex (DISC) performs caspase-8 proteolytic activation which initiates the subsequent cascade of caspases (aspartate-specific cysteine proteases) mediating apoptosis. FAS- mediated apoptosis may have a role in the induction of peripheral tolerance, in the antigen- stimulated suicide of mature T-cells, or both. [00256] FASL (tumor necrosis factor ligand superfamily member 6) (UniProt ID P48023) is encoded by FASLG (NCBI GeneID 356). It is a cytokine that binds to TNFRSF6/FAS, a receptor that transduces the apoptotic signal into cells. It is involved in cytotoxic T-cell-mediated apoptosis, natural killer cell-mediated apoptosis and in T-cell development. [00257] The FASLG signaling pathway can induce apoptosis of T cells. The FASLG ligand is expressed among cytotoxic Delta-Gamma/CD8s and NK-like exhausted Delta-Gamma/CD8s. The FAS receptor is expressed among all TIL subpopulations. Inhibitors of the FASLG ligand, the FAS receptor, the ligand-receptor interaction as well as downstream effects of the ligand- receptor binding are all contemplated. [00258] In one embodiment, a FAS/FASLG inhibitory agent comprises a DNA encoding a dominant negative FAS mutant operably linked to a promoter active in the TILs. For example, the DNA encoding the dominant negative Fas mutant can be in a viral vector, such as a lentiviral vector. In another embodiment, the FAS/FASLG inhibitory agent comprises a messenger RNA encoding a dominant negative FAS mutant. In a specific example, the dominant negative FAS mutant comprises a mutated FADD binding site, optionally wherein the dominant negative FAS mutant is Fas_D244V. In another specific example, the dominant negative FAS mutant comprises a deleted DD domain, optionally wherein the dominant negative FAS mutant is Fas_del230-314. In other embodiments, the FAS/FASLG inhibitory agent comprises an anti-FAS antigen-binding protein, such as antibody. In other embodiments, the FAS/FASLG inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to a FAS or FASLG messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the FAS/FASLG inhibitory agent comprises a nuclease agent targeting a nuclease target site in a FAS or FASLG gene. For example, the nuclease agent can comprise a Cas protein (e.g. a Cas9 protein or a Cas12a protein) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding Fas or FasL. In other embodiments, the FAS/FASLG inhibitory agent comprises a small molecule Fas/FasL inhibitor, such as Kp-7- 6. [00259] In one example, the treating or compound(s) decrease expression or activity of TGFβ or TGFβR1. For example, the treating or compound(s) can transiently decrease expression or activity of TGFβ or TGFβR1. Alternatively, the treating or compound(s) can permanently decrease expression or activity of TGFβ or TGFβR1. [00260] TGF-beta receptor type-1 (UniProt ID P36897) is encoded by TGFBR1 (NCBI GeneID 7046). It is a transmembrane serine/threonine kinase forming with the TGF-beta type II serine/threonine kinase receptor, TGFBR2, the non-promiscuous receptor for the TGF-beta cytokines TGFB1, TGFB2 and TGFB3. It transduces the TGFB1, TGFB2 and TGFB3 signal from the cell surface to the cytoplasm and is thus regulating a plethora of physiological and pathological processes including cell cycle arrest in epithelial and hematopoietic cells, control of mesenchymal cell proliferation and differentiation, wound healing, extracellular matrix production, immunosuppression and carcinogenesis. The formation of the receptor complex composed of 2 TGFBR1 and 2 TGFBR2 molecules symmetrically bound to the cytokine dimer results in the phosphorylation and the activation of TGFBR1 by the constitutively active TGFBR2. Activated TGFBR1 phosphorylates SMAD2 which dissociates from the receptor and interacts with SMAD4. The SMAD2-SMAD4 complex is subsequently translocated to the nucleus where it modulates the transcription of the TGF-beta-regulated genes. This constitutes the canonical SMAD-dependent TGF-beta signaling cascade. Also involved in non-canonical, SMAD-independent TGF-beta signaling pathways. For instance, TGFBR1 induces TRAF6 autoubiquitination which in turn results in MAP3K7 ubiquitination and activation to trigger apoptosis. It also regulates epithelial to mesenchymal transition through a SMAD-independent signaling pathway through PARD6A phosphorylation and activation. [00261] The TGFβ signaling pathway is immunosuppressive and inhibits cell proliferation. The TGFβ ligand is pervasively expressed among all TIL subpopulations. The receptor is expressed primarily among Tem/Temra CD8s, activated CD8s, naïve-like CD4s, and NK- like/Exhausted Delta-Gamma/CD8s. Multiple drugs have been developed to block this signaling pathway (see below tables and Huang et al., Biomedicine & Pharmacotherapy Volume 134, February 2021, 111046). [00262] Antisense oligonucleotides, ligand traps and monoclonal neutralizing antibodies targeting TGFβ signaling pathway.
Figure imgf000072_0001
[00263] Vaccines targeting a TGFβ pathway.
Figure imgf000073_0001
[00264] Small molecule TGFβ inhibitors.
Figure imgf000074_0001
[00265] In some embodiments, a TGFβ/TGFβR1 inhibitory agent comprises a DNA encoding a dominant negative TGFβR1 mutant operably linked to a promoter active in the TILs. For example, the DNA encoding the dominant negative TGFβR1 mutant can be in a viral vector, such as a lentiviral vector. In some embodiments, a TGFβ/TGFβR1 inhibitory agent comprises a messenger RNA encoding a dominant negative TGFβR1 mutant. In other embodiments, a TGFβ/TGFβR1 inhibitory agent comprises an anti-TGFβR1 antigen-binding protein, such as an antibody. In some embodiments, the TGFβ/TGFβR1 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to a TGFβR1 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the TGFβ/TGFβR1 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding TGFβR1. For example, the nuclease agent can comprise a Cas protein and a guide RNA that forms a complex with the Cas protein (e.g., a Cas9 protein or a Cas12a protein) and targets a guide RNA target sequence in the gene encoding TGFβR1. In other embodiments, the TGFβ/TGFβR1 inhibitory agent comprises a small molecule TGFβR1 inhibitor, such as SB431542. [00266] In one example, the treating or compound(s) decrease expression or activity of IRF7. For example, the treating or compound(s) can transiently decrease expression or activity of IRF7. Alternatively, the treating or compound(s) can permanently decrease expression or activity of IRF7. [00267] Interferon regulatory factor 7 (Uniprot ID Q92985) is encoded by IRF7 (NCBI GeneID 3665). Interferon regulatory factor 7 is a key transcriptional regulator of type I interferon (IFN)-dependent immune responses and plays a critical role in the innate immune response against DNA and RNA viruses. It regulates the transcription of type I IFN genes (IFN-alpha and IFN-beta) and IFN-stimulated genes (ISG) by binding to an interferon-stimulated response element (ISRE) in their promoters. [00268] In some embodiments, an IRF7 inhibitory agent comprises a DNA encoding a dominant negative IRF7 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative IRF7 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the IRF7 inhibitory agent comprises a messenger RNA encoding a dominant negative IRF7 mutant. In other embodiments, the IRF7 inhibitory agent comprises an anti-IRF7 antigen-binding protein, such as an antibody. In other embodiments, the IRF7 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an IRF7 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the IRF7 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding IRF7, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF7. In other embodiments, the IRF7 inhibitory agent comprises a small molecule IRF7 inhibitor. [00269] In one example, the treating or compound(s) decrease expression or activity of POLR3A. For example, the treating or compound(s) can transiently decrease expression or activity of POLR3A. Alternatively, the treating or compound(s) can permanently decrease expression or activity of POLR3A. [00270] POLR3A (DNA-directed RNA polymerase III subunit RPC1; UniProt ID O14802) is encoded by POLR3A (NCBI GeneID 11128). It is a DNA-dependent RNA polymerase catalyzes the transcription of DNA into RNA using the four ribonucleoside triphosphates as substrates. [00271] In some embodiments, the POLR3A inhibitory agent comprises a DNA encoding a dominant negative POLR3A mutant operably linked to a promoter active in the TILs. For example, the DNA encoding the dominant negative POLR3A mutant can be in a viral vector, such as a lentiviral vector. In other embodiments, the POLR3A inhibitory agent comprises a messenger RNA encoding a dominant negative POLR3A mutant. In other embodiments, the POLR3A inhibitory agent comprises an anti-POLR3A antigen-binding protein, such as an antibody. In other embodiments, the POLR3A inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to a POLR3A messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the POLR3A inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding POLR3A, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding POLR3A. In other embodiments, the POLR3A inhibitory agent comprises a small molecule POLR3A inhibitor. [00272] In one example, the treating or compound(s) decrease expression or activity of ETV7. For example, the treating or compound(s) can transiently decrease expression or activity of ETV7. Alternatively, the treating or compound(s) can permanently decrease expression or activity of ETV7. [00273] Transcription factor ETV7 (ETS translocation variant 7; ETS variant transcription factor; Uniprot ID Q9Y603) is encoded by ETV7 (NCBI GeneID 51513). Transcription factor ETV7 is a transcriptional repressor that binds to the DNA sequence 5'-CCGGAAGT-3'. [00274] In some embodiments, an ETV7 inhibitory agent comprises a DNA encoding a dominant negative ETV7 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative ETV7 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the ETV7 inhibitory agent comprises a messenger RNA encoding a dominant negative ETV7 mutant. In other embodiments, the ETV7 inhibitory agent comprises an anti- ETV7 antigen-binding protein, such as an antibody. In other embodiments, the ETV7 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ETV7 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the ETV7 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ETV7, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV7. In other embodiments, the ETV7 inhibitory agent comprises a small molecule ETV7 inhibitor. [00275] In one example, the treating or compound(s) decrease expression or activity of ETV3. For example, the treating or compound(s) can transiently decrease expression or activity of ETV3. Alternatively, the treating or compound(s) can permanently decrease expression or activity of ETV3. [00276] ETS translocation variant 3 (Uniprot ID P41162) is encoded by ETV3 (NCBI GeneID 2117). ETS translocation variant 3 is a transcriptional repressor that contributes to growth arrest during terminal macrophage differentiation by repressing target genes involved in Ras-dependent proliferation. It also represses MMP1 promoter activity. [00277] In some embodiments, an ETV3 inhibitory agent comprises a DNA encoding a dominant negative ETV3 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative ETV3 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the ETV3 inhibitory agent comprises a messenger RNA encoding a dominant negative ETV3 mutant. In other embodiments, the ETV3 inhibitory agent comprises an anti-ETV3 antigen-binding protein, such as an antibody. In other embodiments, the ETV3 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ETV3 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the ETV3 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ETV3, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3. In other embodiments, the ETV3 inhibitory agent comprises a small molecule ETV3 inhibitor. [00278] In one example, the treating or compound(s) decrease expression or activity of ASH2L. For example, the treating or compound(s) can transiently decrease expression or activity of ASH2L. Alternatively, the treating or compound(s) can permanently decrease expression or activity of ASH2L. [00279] Set1/Ash2 histone methyltransferase complex subunit ASH2 (ASH2 like, histone lysine methyltransferase complex subunit; Uniprot ID Q9UBL3) is encoded by ASH2L (NCBI GeneID 9070). ASH2 is a component of the Set1/Ash2 histone methyltransferase (HMT) complex, a complex that specifically methylates 'Lys-4' of histone H3, but not if the neighboring 'Lys-9' residue is already methylated. [00280] In some embodiments, an ASH2L inhibitory agent comprises a DNA encoding a dominant negative ASH2L mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative ASH2L mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the ASH2L inhibitory agent comprises a messenger RNA encoding a dominant negative ASH2L mutant. In other embodiments, the ASH2L inhibitory agent comprises an anti-ASH2L antigen-binding protein, such as an antibody. In other embodiments, the ASH2L inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ASH2L messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the ASH2L inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ASH2L, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ASH2L. In other embodiments, the ASH2L inhibitory agent comprises a small molecule ASH2L inhibitor. [00281] In one example, the treating or compound(s) decrease expression or activity of PML. For example, the treating or compound(s) can transiently decrease expression or activity of PML. Alternatively, the treating or compound(s) can permanently decrease expression or activity of PML. [00282] Protein PML (PML nuclear body scaffold; promyelocytic leukemia protein; Uniprot ID P29590) is encoded by PML (NCBI GeneID 5371). PML functions via its association with PML-nuclear bodies (PML-NBs) in a wide range of important cellular processes, including tumor suppression, transcriptional regulation, apoptosis, senescence, DNA damage response, and viral defense mechanisms. It acts as the scaffold of PML-NBs allowing other proteins to shuttle in and out, a process which is regulated by SUMO-mediated modifications and interactions. [00283] In some embodiments, an PML inhibitory agent comprises a DNA encoding a dominant negative PML mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative PML mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the PML inhibitory agent comprises a messenger RNA encoding a dominant negative PML mutant. In other embodiments, the PML inhibitory agent comprises an anti-PML antigen-binding protein, such as an antibody. In other embodiments, the PML inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an PML messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the PML inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding PML, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding PML. In other embodiments, the PML inhibitory agent comprises a small molecule PML inhibitor. [00284] In one example, the treating or compound(s) decrease expression or activity of STAT2. For example, the treating or compound(s) can transiently decrease expression or activity of STAT2. Alternatively, the treating or compound(s) can permanently decrease expression or activity of STAT2. [00285] Signal transducer and activator of transcription 2 (Uniprot ID P52630) is encoded by STAT2 (NCBI GeneID 6773). STAT2 mediates signaling by type I interferons (IFN-alpha and IFN-beta). Following type I IFN binding to cell surface receptors, Jak kinases (TYK2 and JAK1) are activated, leading to tyrosine phosphorylation of STAT1 and STAT2. The phosphorylated STATs dimerize, associate with IRF9/ISGF3G to form a complex termed ISGF3 transcription factor, and then enter the nucleus. ISGF3 binds to the IFN stimulated response element (ISRE) to activate the transcription of interferon stimulated genes, which drive the cell in an antiviral state. [00286] In some embodiments, an STAT2 inhibitory agent comprises a DNA encoding a dominant negative STAT2 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative STAT2 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the STAT2 inhibitory agent comprises a messenger RNA encoding a dominant negative STAT2 mutant. In other embodiments, the STAT2 inhibitory agent comprises an anti-STAT2 antigen-binding protein, such as an antibody. In other embodiments, the STAT2 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an STAT2 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the STAT2 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding STAT2, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT2. In other embodiments, the STAT2 inhibitory agent comprises a small molecule STAT2 inhibitor. [00287] In one example, the treating or compound(s) decrease expression or activity of SPI1. For example, the treating or compound(s) can transiently decrease expression or activity of SPI1. Alternatively, the treating or compound(s) can permanently decrease expression or activity of SPI1. [00288] Transcription factor PU.1 (Spi-1 proto-oncogene; Uniprot ID P17947) is encoded by SPI1 (NCBI GeneID 6688). Transcription factor PU.1 binds to the PU-box, a purine-rich DNA sequence (5'-GAGGAA-3') that can act as a lymphoid-specific enhancer. This protein is a transcriptional activator that may be specifically involved in the differentiation or activation of macrophages or B-cells. [00289] In some embodiments, an SPI1 inhibitory agent comprises a DNA encoding a dominant negative SPI1 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative SPI1 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the SPI1 inhibitory agent comprises a messenger RNA encoding a dominant negative SPI1 mutant. In other embodiments, the SPI1 inhibitory agent comprises an anti-SPI1 antigen-binding protein, such as an antibody. In other embodiments, the SPI1 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an SPI1 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the SPI1 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding SPI1, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SPI1. In other embodiments, the SPI1 inhibitory agent comprises a small molecule SPI1 inhibitor. [00290] In one example, the treating or compound(s) decrease expression or activity of IRF9. For example, the treating or compound(s) can transiently decrease expression or activity of IRF9. Alternatively, the treating or compound(s) can permanently decrease expression or activity of IRF9. [00291] Interferon regulatory factor 9 (Uniprot ID Q00978) is encoded by IRF9 (NCBI GeneID 10379). Interferon regulatory factor 9 is a transcription factor that plays an essential role in anti-viral immunity. It mediates signaling by type I IFNs (IFN-alpha and IFN-beta). Following type I IFN binding to cell surface receptors, Jak kinases (TYK2 and JAK1) are activated, leading to tyrosine phosphorylation of STAT1 and STAT2. IRF9/ISGF3G associates with the phosphorylated STAT1:STAT2 dimer to form a complex termed ISGF3 transcription factor, that enters the nucleus. ISGF3 binds to the IFN stimulated response element (ISRE) to activate the transcription of interferon stimulated genes, which drive the cell in an antiviral state. [00292] In some embodiments, an IRF9 inhibitory agent comprises a DNA encoding a dominant negative IRF9 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative IRF9 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the IRF9 inhibitory agent comprises a messenger RNA encoding a dominant negative IRF9 mutant. In other embodiments, the IRF9 inhibitory agent comprises an anti-IRF9 antigen-binding protein, such as an antibody. In other embodiments, the IRF9 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an IRF9 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the IRF9 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding IRF9, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF9. In other embodiments, the IRF9 inhibitory agent comprises a small molecule IRF9 inhibitor. [00293] In one example, the treating or compound(s) decrease expression or activity of STAT1. For example, the treating or compound(s) can transiently decrease expression or activity of STAT1. Alternatively, the treating or compound(s) can permanently decrease expression or activity of STAT1. [00294] Signal transducer and activator of transcription 1 (Uniprot ID P42224) is encoded by STAT1 (NCBI GeneID 6772). STAT1 mediates cellular responses to interferons (IFNs), cytokine KITLG/SCF and other cytokines and other growth factors. Following type I IFN (IFN-alpha and IFN-beta) binding to cell surface receptors, signaling via protein kinases leads to activation of Jak kinases (TYK2 and JAK1) and to tyrosine phosphorylation of STAT1 and STAT2. The phosphorylated STATs dimerize and associate with ISGF3G/IRF-9 to form a complex termed ISGF3 transcription factor that enters the nucleus. ISGF3 binds to the IFN stimulated response element (ISRE) to activate the transcription of IFN-stimulated genes (ISG), which drive the cell in an antiviral state. In response to type II IFN (IFN-gamma), STAT1 is tyrosine- and serine- phosphorylated. It then forms a homodimer termed IFN-gamma-activated factor (GAF), migrates into the nucleus, and binds to the IFN gamma activated sequence (GAS) to drive the expression of the target genes, inducing a cellular antiviral state. It becomes activated in response to KITLG/SCF and KIT signaling. [00295] In some embodiments, an STAT1 inhibitory agent comprises a DNA encoding a dominant negative STAT1 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative STAT1 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the STAT1 inhibitory agent comprises a messenger RNA encoding a dominant negative STAT1 mutant. In other embodiments, the STAT1 inhibitory agent comprises an anti-STAT1 antigen-binding protein, such as an antibody. In other embodiments, the STAT1 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an STAT1 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the STAT1 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding STAT1, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding STAT1. In other embodiments, the STAT1 inhibitory agent comprises a small molecule STAT1 inhibitor. [00296] In one example, the treating or compound(s) decrease expression or activity of IRF4. For example, the treating or compound(s) can transiently decrease expression or activity of IRF4. Alternatively, the treating or compound(s) can permanently decrease expression or activity of IRF4. [00297] Interferon regulatory factor 4 (Uniprot ID Q15306) is encoded by IRF4 (NCBI GeneID 3662). Interferon regulatory factor 4 is a transcriptional activator. IRF4 binds to the interferon-stimulated response element (ISRE) of the MHC class I promoter. It also binds the immunoglobulin lambda light chain enhancer, together with PU.1. [00298] In some embodiments, an IRF4 inhibitory agent comprises a DNA encoding a dominant negative IRF4 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative IRF4 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the IRF4 inhibitory agent comprises a messenger RNA encoding a dominant negative IRF4 mutant. In other embodiments, the IRF4 inhibitory agent comprises an anti-IRF4 antigen-binding protein, such as an antibody. In other embodiments, the IRF4 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an IRF4 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the IRF4 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding IRF4, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF4. In other embodiments, the IRF4 inhibitory agent comprises a small molecule IRF4 inhibitor. [00299] In one example, the treating or compound(s) decrease expression or activity of JDP2. For example, the treating or compound(s) can transiently decrease expression or activity of JDP2. Alternatively, the treating or compound(s) can permanently decrease expression or activity of JDP2. [00300] Jun dimerization protein 2 (Uniprot ID Q8WYK2) is encoded by JDP2 (NCBI GeneID 122953). Jun dimerization protein 2 is a component of the AP-1 transcription factor that represses transactivation mediated by the Jun family of proteins. It is involved in a variety of transcriptional responses associated with AP-1, such as UV-induced apoptosis, cell differentiation, tumorigenesis, and tumor suppression. JDP2 can also function as a repressor by recruiting histone deacetylase 3/HDAC3 to the promoter region of JUN. [00301] In some embodiments, an JDP2 inhibitory agent comprises a DNA encoding a dominant negative JDP2 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative JDP2 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the JDP2 inhibitory agent comprises a messenger RNA encoding a dominant negative JDP2 mutant. In other embodiments, the JDP2 inhibitory agent comprises an anti-JDP2 antigen-binding protein, such as an antibody. In other embodiments, the JDP2 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an JDP2 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the JDP2 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding JDP2, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding JDP2. In other embodiments, the JDP2 inhibitory agent comprises a small molecule JDP2 inhibitor. [00302] In one example, the treating or compound(s) decrease expression or activity of ZNF337. For example, the treating or compound(s) can transiently decrease expression or activity of ZNF337. Alternatively, the treating or compound(s) can permanently decrease expression or activity of ZNF337. [00303] Zinc finger protein 337 (Uniprot ID Q9Y3M9) is encoded by ZNF337 (NCBI GeneID 26152). Zinc finger protein 337 may be involved in transcriptional regulation. [00304] In some embodiments, an ZNF337 inhibitory agent comprises a DNA encoding a dominant negative ZNF337 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative ZNF337 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the ZNF337 inhibitory agent comprises a messenger RNA encoding a dominant negative ZNF337 mutant. In other embodiments, the ZNF337 inhibitory agent comprises an anti-ZNF337 antigen-binding protein, such as an antibody. In other embodiments, the ZNF337 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ZNF337 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the ZNF337 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ZNF337, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF337. In other embodiments, the ZNF337 inhibitory agent comprises a small molecule ZNF337 inhibitor. [00305] In one example, the treating or compound(s) decrease expression or activity of ETV2. For example, the treating or compound(s) can transiently decrease expression or activity of ETV2. Alternatively, the treating or compound(s) can permanently decrease expression or activity of ETV2. [00306] ETS translocation variant 2 (Uniprot ID O00321) is encoded by ETV2 (NCBI GeneID 2116). ETS translocation variant 2 binds to DNA sequences containing the consensus pentanucleotide 5'-CGGA[AT]-3'. [00307] In some embodiments, an ETV2 inhibitory agent comprises a DNA encoding a dominant negative ETV2 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative ETV2 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the ETV2 inhibitory agent comprises a messenger RNA encoding a dominant negative ETV2 mutant. In other embodiments, the ETV2 inhibitory agent comprises an anti-ETV2 antigen-binding protein, such as an antibody. In other embodiments, the ETV2 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ETV2 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the ETV2 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ETV2, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV2. In other embodiments, the ETV2 inhibitory agent comprises a small molecule ETV2 inhibitor. [00308] In one example, the treating or compound(s) decrease expression or activity of ETV3L. For example, the treating or compound(s) can transiently decrease expression or activity of ETV3L. Alternatively, the treating or compound(s) can permanently decrease expression or activity of ETV3L. [00309] ETS translocation variant 3-like protein (ETS variant transcription factor 3 like; Uniprot ID Q6ZN32) is encoded by ETV3L (NCBI GeneID 440695). ETS translocation variant 3-like protein is a transcriptional regulator. [00310] In some embodiments, an ETV3L inhibitory agent comprises a DNA encoding a dominant negative ETV3L mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative ETV3L mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the ETV3L inhibitory agent comprises a messenger RNA encoding a dominant negative ETV3L mutant. In other embodiments, the ETV3L inhibitory agent comprises an anti-ETV3L antigen-binding protein, such as an antibody. In other embodiments, the ETV3L inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ETV3L messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the ETV3L inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ETV3L, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ETV3L. In other embodiments, the ETV3L inhibitory agent comprises a small molecule ETV3L inhibitor. [00311] In one example, the treating or compound(s) decrease expression or activity of SOX18. For example, the treating or compound(s) can transiently decrease expression or activity of SOX18. Alternatively, the treating or compound(s) can permanently decrease expression or activity of SOX18. [00312] SRY-box transcription factor 18 (Uniprot ID P35713) is encoded by SOX18 (NCBI GeneID 54345). SRY-box transcription factor 18 is a transcriptional activator that binds to the consensus sequence 5'-AACAAAG-3' in the promoter of target genes and plays an essential role in embryonic cardiovascular development and lymphangiogenesis. It activates transcription of PROX1 and other genes coding for lymphatic endothelial markers. SOX18 also plays an essential role in triggering the differentiation of lymph vessels, but it is not required for the maintenance of differentiated lymphatic endothelial cells. It plays an important role in postnatal angiogenesis, where it is functionally redundant with SOX17. Interaction with MEF2C enhances transcriptional activation of SOX18. [00313] In some embodiments, an SOX18 inhibitory agent comprises a DNA encoding a dominant negative SOX18 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative SOX18 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the SOX18 inhibitory agent comprises a messenger RNA encoding a dominant negative SOX18 mutant. In other embodiments, the SOX18 inhibitory agent comprises an anti-SOX18 antigen-binding protein, such as an antibody. In other embodiments, the SOX18 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an SOX18 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the SOX18 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding SOX18, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding SOX18. In other embodiments, the SOX18 inhibitory agent comprises a small molecule SOX18 inhibitor. [00314] In one example, the treating or compound(s) decrease expression or activity of CEBPG. For example, the treating or compound(s) can transiently decrease expression or activity of CEBPG. Alternatively, the treating or compound(s) can permanently decrease expression or activity of CEBPG. [00315] CCAAT/enhancer-binding protein gamma (Uniprot ID P53567) is encoded by CEBPG (NCBI GeneID 1054). CEBPG is a transcription factor that binds to the promoter and the enhancer regions of several target genes, including the enhancer element PRE-I (positive regulatory element-I) of the IL-4 gene, the promoter and the enhancer of the immunoglobulin heavy chain, and binds to GPE1, a cis-acting element in the G-CSF gene promoter. [00316] In some embodiments, an CEBPG inhibitory agent comprises a DNA encoding a dominant negative CEBPG mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative CEBPG mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the CEBPG inhibitory agent comprises a messenger RNA encoding a dominant negative CEBPG mutant. In other embodiments, the CEBPG inhibitory agent comprises an anti-CEBPG antigen-binding protein, such as an antibody. In other embodiments, the CEBPG inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an CEBPG messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the CEBPG inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding CEBPG, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPG. In other embodiments, the CEBPG inhibitory agent comprises a small molecule CEBPG inhibitor. [00317] In one example, the treating or compound(s) decrease expression or activity of CREB3L4. For example, the treating or compound(s) can transiently decrease expression or activity of CREB3L4. Alternatively, the treating or compound(s) can permanently decrease expression or activity of CREB3L4. [00318] Cyclic AMP-responsive element-binding protein 3-like protein 4 (Uniprot ID Q8TEY5) is encoded by CREB3L4 (NCBI GeneID 148327). CREB3L4 is a transcriptional activator that may play a role in the unfolded protein response. It binds to the UPR element (UPRE) but not to CRE element. CREB3L4 preferentially binds DNA with to the consensus sequence 5'-T[GT]ACGT[GA][GT]-3' and has transcriptional activation activity from UPRE. [00319] In some embodiments, an CREB3L4 inhibitory agent comprises a DNA encoding a dominant negative CREB3L4 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative CREB3L4 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the CREB3L4 inhibitory agent comprises a messenger RNA encoding a dominant negative CREB3L4 mutant. In other embodiments, the CREB3L4 inhibitory agent comprises an anti-CREB3L4 antigen-binding protein, such as an antibody. In other embodiments, the CREB3L4 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an CREB3L4 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the CREB3L4 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding CREB3L4, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CREB3L4. In other embodiments, the CREB3L4 inhibitory agent comprises a small molecule CREB3L4 inhibitor. [00320] In one example, the treating or compound(s) decrease expression or activity of CEBPB. For example, the treating or compound(s) can transiently decrease expression or activity of CEBPB. Alternatively, the treating or compound(s) can permanently decrease expression or activity of CEBPB. [00321] CCAAT/enhancer-binding protein beta (Uniprot ID P17676) is encoded by CEBPB (NCBI GeneID 1051). CEBPB is an important transcription factor regulating the expression of genes involved in immune and inflammatory responses. It also plays a significant role in adipogenesis, as well as in the gluconeogenic pathway, liver regeneration, and hematopoiesis. The consensus recognition site is 5'-T[TG]NNGNAA[TG]-3'. Its functional capacity is governed by protein interactions and post-translational protein modifications. [00322] In some embodiments, an CEBPB inhibitory agent comprises a DNA encoding a dominant negative CEBPB mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative CEBPB mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the CEBPB inhibitory agent comprises a messenger RNA encoding a dominant negative CEBPB mutant. In other embodiments, the CEBPB inhibitory agent comprises an anti-CEBPB antigen-binding protein, such as an antibody. In other embodiments, the CEBPB inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an CEBPB messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the CEBPB inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding CEBPB, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding CEBPB. In other embodiments, the CEBPB inhibitory agent comprises a small molecule CEBPB inhibitor. [00323] In one example, the treating or compound(s) decrease expression or activity of FOXD1. For example, the treating or compound(s) can transiently decrease expression or activity of FOXD1. Alternatively, the treating or compound(s) can permanently decrease expression or activity of FOXD1. [00324] Forkhead box protein D1 (Uniprot ID Q16676) is encoded by FOXD1 (NCBI GeneID 2297). Forkhead box protein D1 is a transcription factor involved in regulation of gene expression in a variety of processes, including formation of positional identity in the developing retina, regionalization of the optic chiasm, morphogenesis of the kidney, and neuralization of ectodermal cells. [00325] In some embodiments, an FOXD1 inhibitory agent comprises a DNA encoding a dominant negative FOXD1 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative FOXD1 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the FOXD1 inhibitory agent comprises a messenger RNA encoding a dominant negative FOXD1 mutant. In other embodiments, the FOXD1 inhibitory agent comprises an anti-FOXD1 antigen-binding protein, such as an antibody. In other embodiments, the FOXD1 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an FOXD1 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the FOXD1 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding FOXD1, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FOXD1. In other embodiments, the FOXD1 inhibitory agent comprises a small molecule FOXD1 inhibitor. [00326] In one example, the treating or compound(s) decrease expression or activity of EOMES. For example, the treating or compound(s) can transiently decrease expression or activity of EOMES. Alternatively, the treating or compound(s) can permanently decrease expression or activity of EOMES. [00327] Eomesodermin homolog (Uniprot ID O95936) is encoded by EOMES (NCBI GeneID 8320). Eomesodermin homolog functions as a transcriptional activator playing a crucial role during development, particularly in trophoblast differentiation and later in gastrulation, regulating both mesoderm delamination and endoderm specification. It plays a role in brain development, being required for the specification and the proliferation of the intermediate progenitor cells and their progeny in the cerebral cortex. EOMES is also involved in the differentiation of CD8+ T-cells during immune response regulating the expression of lytic effector genes. [00328] In some embodiments, an EOMES inhibitory agent comprises a DNA encoding a dominant negative EOMES mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative EOMES mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the EOMES inhibitory agent comprises a messenger RNA encoding a dominant negative EOMES mutant. In other embodiments, the EOMES inhibitory agent comprises an anti-EOMES antigen-binding protein, such as an antibody. In other embodiments, the EOMES inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an EOMES messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the EOMES inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding EOMES, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding EOMES. In other embodiments, the EOMES inhibitory agent comprises a small molecule EOMES inhibitor. [00329] In one example, the treating or compound(s) decrease expression or activity of ZNF683. For example, the treating or compound(s) can transiently decrease expression or activity of ZNF683. Alternatively, the treating or compound(s) can permanently decrease expression or activity of ZNF683. [00330] Zinc finger protein 683, also known as tissue-resident T-cell transcription regulator protein, (Uniprot ID Q8IZ20) is encoded by ZNF683 (NCBI GeneID 257101). Tissue-resident T- cell transcription regulator protein is a transcription factor that mediates a transcriptional program in various innate and adaptive immune tissue-resident lymphocyte T-cell types such as tissue-resident memory T (Trm), natural killer (trNK), and natural killer T (NKT) cells and negatively regulates gene expression of proteins that promote the egress of tissue-resident T-cell populations from non-lymphoid organs. It plays a role in the development, retention, and long- term establishment of adaptive and innate tissue-resident lymphocyte T cell types in non- lymphoid organs, such as the skin and gut, but also in other non-barrier tissues like liver and kidney, and therefore may provide immediate immunological protection against reactivating infections or viral reinfection. Tissue-resident T-cell transcription regulator protein plays a role in the differentiation of both thymic and peripheral NKT cells. It negatively regulates the accumulation of interferon-gamma (IFN-gamma) in NKT cells at steady state or after antigenic stimulation and positively regulates granzyme B production in NKT cells after innate stimulation. It associates with the transcriptional repressor PRDM1/BLIMP1 to chromatin at gene promoter regions. [00331] In some embodiments, an ZNF683 inhibitory agent comprises a DNA encoding a dominant negative ZNF683 mutant operably linked to a promoter active in the TILs. In some embodiments, the DNA encoding the dominant negative ZNF683 mutant is in a viral vector, such as a lentiviral vector. In other embodiments, the ZNF683 inhibitory agent comprises a messenger RNA encoding a dominant negative ZNF683 mutant. In other embodiments, the ZNF683 inhibitory agent comprises an anti-ZNF683 antigen-binding protein, such as an antibody. In other embodiments, the ZNF683 inhibitory agent comprises an inhibitory RNA comprising a region of complementarity to an ZNF683 messenger RNA, such as an antisense oligonucleotide or an RNAi agent. In other embodiments, the ZNF683 inhibitory agent comprises a nuclease agent targeting a nuclease target site in a gene encoding ZNF683, such as a Cas protein (e.g., Cas9 or Cas12a) and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding ZNF683. In other embodiments, the ZNF683 inhibitory agent comprises a small molecule ZNF683 inhibitor. [00332] As described in more detail elsewhere herein, the TILs can originate from a subject. For example, the TILs can be from a tumor biopsy, a lymph node, or ascites. The tumor can be, for example, from a bladder cancer, a breast cancer, a cancer caused by human papilloma virus, a cervical cancer, a head and neck cancer, a lung cancer, a melanoma, an ovarian cancer, a non- small-cell lung cancer (NSCLC), a renal cancer, or a renal cell carcinoma. In a specific example, the tumor biopsy is from a melanoma. [00333] As described in more detail elsewhere herein, the methods can further comprise: (i) obtaining a refined tumor product by cryopreserving a resected tumor and disaggregating the cryopreserved tumor, disaggregating a resected tumor and cryopreserving the disaggregated tumor, cryopreserving a resected tumor and processing the tumor into multiple tumor fragments, or processing a resected tumor into multiple tumor fragments and cryopreserving the tumor fragments; and (ii) performing a first expansion by culturing the refined resected tumor product in a cell culture medium comprising IL-2 to produce the first population of TILs, optionally wherein the first population of TILs is treated with the one or more compounds during or subsequent to the first expansion. Cryopreserving can comprise, for example: (1) cooling under conditions whereby heat release to, into, around or in an environment including cells, as media crystalizes, is minimized or avoided; (2) continuous cooling, from disaggregation temperature to about -80°C; (3) continuous cooling at a rate of about -2°C / min; (4) continuous cooling, from disaggregation temperature to about -80°C, at a rate of about -2°C / min; or (5) continuous cooling, from disaggregation temperature to about -80°C, or from disaggregation temperature to -80°C at a rate of about -2°C / min, wherein disaggregation temperature comprises a normal body temperature for an animal from which the tumor was resected, or room temperature or 20°C or 25°C , or normal human body temperature approximately 35°C or 36°C or 36.1°C to approximately 37°C or 37.1°C or 37.2°C or 37.3°C or below about 38.3°C. Disaggregating can comprise, for example, physical disaggregation, enzymatic disaggregation, or physical and enzymatic disaggregation. In some methods, a single cell suspension is obtained from the refined resected tumor product and used in step (ii), or wherein the refined resected tumor product from step (i) comprises a single cell suspension. In some methods, the first expansion in step (ii) is performed for about two weeks. In some methods, the culturing in step (ii) includes adding IL-7, IL-12, IL-15, IL-18, IL-21, or a combination thereof. [00334] As described in more detail elsewhere herein, such methods can further comprise: (iii) performing a second expansion by culturing the first population of TILs in a cell culture medium comprising IL-2, optionally wherein the first population of TILs is treated with the one or more compounds prior to, during, or subsequent to the second expansion. In some methods, the expanding in step (iii) comprises culturing the first population of TILs with IL-2, OKT-3, and antigen presenting cells (APCs). In some methods, the expanding in step (iii) is performed for about two weeks. In some such methods, the culturing in step (iii) includes adding IL-7, IL-12, IL-15, IL-18, IL-21, or a combination thereof. In some methods, the method further comprises harvesting and/or cryopreserving the therapeutic population of TILs. [00335] As disclosed in more detail elsewhere herein, also provided are isolated therapeutic population of TILs obtained by or obtainable by any of the above methods, pharmaceutical formulations comprising a pharmaceutically acceptable excipient and any of the above isolated therapeutic populations of TILs, cryopreserved bags or intravenous infusion bags, containers, or vessels containing contents comprising any of the above isolated therapeutic populations of TILs, and methods of treating a cancer in a subject, comprising administering any of above isolated therapeutic population of TILs or the above pharmaceutical formulation to the subject. [00336] In some embodiments, the present invention relates to a method for isolating a therapeutic population of cryopreserved unmodified tumor infiltrating lymphocytes (UTIL) which may comprise: (a) resecting a tumor from a subject; (b) storing the resected tumor in a single use aseptic kit, wherein the aseptic kit comprises: a disaggregation module for receipt and processing of material comprising solid mammalian tissue; an optional enrichment module for filtration of disaggregated solid tissue material and segregation of non-disaggregated tissue and filtrate; and a stabilization module for optionally further processing and/or storing disaggregated product material, wherein each of the modules comprises one or more flexible containers connected by one or more conduits adapted to enable flow of the tissue material there between; and wherein each of the modules comprises one or more ports to permit aseptic input of media and/or reagents into the one or more flexible containers; (c) aseptically disaggregating the resected tumor in the disaggregation module thereby producing a disaggregated tumor, wherein the resected tumor is sufficiently disaggregated if it can be cryopreserved with a minimum of cell damage; (d) cryopreserving the disaggregated tumor in the stabilization module; (e) performing a first expansion by culturing the disaggregated tumor in a cell culture medium comprising IL-2 to produce a first population of UTILs; (f) optionally performing a second expansion by culturing the first population of UTILs with additional IL-2, OKT-3, and antigen presenting cells (APCs), to produce a second population of TILs; (g) harvesting and/or cryopreserving the second population of UTILs. [00337] The disaggregation may comprise physical disaggregation, enzymatic disaggregation, or physical and enzymatic disaggregation. In an advantageous embodiment, the disaggregated tumor is cellularized or purified. [00338] In some embodiments, the present invention relates to tumor infiltrating lymphocytes (TILs) in particular unmodified TILs (UTILs), which may be isolated from tumors of a cancer patient or a metastatic cancer patient, involving autologous TILs generated from and returned to the same cancer patient. The present invention also relates to methods for isolating a therapeutic population of cryopreserved TILs or UTILs and to TILs and UTILs obtained or obtainable via use of a device comprising a single use aseptic kit for processing of a resected tumor by the methods described herein. [00339] In general, TILs may initially be obtained from a patient tumor sample (“primary TILs”) and then expanded into a larger population for further manipulation as described herein, cryopreserved, restimulated as outlined herein and optionally evaluated for phenotype and metabolic parameters as an indication of TIL health. [00340] A patient tumor sample may be obtained using methods known in the art, generally via surgical resection, needle biopsy or other means for obtaining a sample that contains a mixture of tumor and TIL cells. In general, the tumor sample may be from any solid tumor, including primary tumors, invasive tumors or metastatic tumors. The tumor sample may also be a liquid tumor, such as a tumor obtained from a hematological malignancy. The solid tumor may be of any cancer type, including, but not limited to, breast, ovary, cervical, pancreatic, prostate, colorectal, lung, brain, renal, stomach, and skin (including but not limited to squamous cell carcinoma, basal cell carcinoma, and melanoma). In some embodiments, TILs are obtained from malignant melanoma tumors, as these have been reported to have particularly high levels of TILs. [00341] The production can generally involve a two-stage process. In stage 1, initial tumor material is dissected, placed in the aseptic kit having a disaggregation module, enzymatically digesting and/or fragmenting, and homogenizing the tumor in the disaggregation module to provide a single cell suspension. While the homogenized cells can be further purified within the aseptic kit in a separate enrichment module to remove components such as no longer required reagents; cell debris; non-disaggregated tissue, the cells can be directly cryopreserved to stabilize the starting material for TIL manufacture and storage in the stabilization module of the aseptic kit until Stage 2 is required. Stage 2 generally involves growth of the TILs out of the resected tumor starting material (2 weeks), followed by a rapid expansion process of the TIL cells (rapid expansion protocol “REP” – 2 weeks). The final product is washed and harvested prior to suspension in buffered saline, 8.5% HAS and 10% DMSO and cryopreserved to form a solid aseptic product that is thawed prior to infusion as a single dose with no further modification. [00342] There are at least three separate elements to the treatment that can potentially contribute to therapeutic activity. The core element is the TILs (i.e., tumor-derived T cells), which can target and eliminate tumor cells by a variety of methods utilized by T cells as a part of their normal function. These methods include direct methods (i.e. perforin-mediated cytotoxicity) and indirect methods (i.e. cytokine production). Which of these methods is the most important to in vivo anti-tumor effects is unclear although mouse models suggest that the production of interferon gamma is critical for effective therapy. The two other elements which contribute to the therapy are pre-conditioning chemotherapy and high dose intravenous IL-2. These two elements are thought to act by supporting engraftment of T cells in the patient after infusion: initially through conditioning chemotherapy which removes competing and regulating immune cells; followed by the IL-2 component which supports survival of T cells. [00343] The structure of the cell therapy product is created by growing the TIL directly out of an enzyme digested tumor mass by means of growth supporting cell culture media and a T cell supporting growth factor Interleukin-2 (IL-2). This enables tumor specific T cells to selectively survive and grow out of the tumor cell mixture, while T cells that do not recognize tumor antigens will not be stimulated and be selectively lost. The product comprises an autologous T- cell based product where the T cells have been derived from a patient’s own cancer tissue and rapidly expanded to form a pure T cell population and T cells as defined by CD3 surface marker. [00344] In brief, TILs, in particular UTILs, may be produced in a two-stage process using a tumor biopsy as the starting material: Stage 1 (generally performed over 2-3 hours) initial collection and processing of tumor material using dissection, enzymatic digestion and homogenization via use of a kit and a semi-automatic device to produce a single cell suspension which can be directly cryopreserved using the stabilization module of the kit to stabilize the starting material for subsequent manufacture and Stage 2 which can occur days or years later. Stage 2 may be performed over 4 weeks, which may be a continuous process starting with thawing of the product of Stage 1 and growth of the TIL out of the tumor starting material (about 2 weeks) followed by a rapid expansion process of the TIL cells (about 2 weeks) to increase the number of cells and therefore dose. The TILs, in particular UTILs, are concentrated and washed prior to formulation as a liquid suspension of cells. The aseptic drug product may be cryopreserved in a bag that will be thawed prior to intravenous infusion as a single dose with no further modification. [00345] For enzymatic digestion, a cell suspension (containing both T cells and tumor cells) can be generated from the resected metastatic tumor using an enzyme mixture of DNase 1 and Collagenase (Type IV). The combination of the repeated mechanical compression exposes additional surfaces for the enzymes to access and the enzymatic reaction speed up the process of turning a solid tissue into a cell suspension prior to optional cryopreservation. In one embodiment upon completion of the disaggregation step a DMSO based cryoprotectant is added just prior to a controlled rate freezing cycle. In some embodiments, the enzymatic breakdown of the solid tissue may be by the selection and provision of one or more media enzyme solutions such as collagenase, trypsin, lipase, hyaluronidase, deoxyribonuclease, Liberase H1, pepsin, or any mixture thereof. Enzymatic digestion of the resected metastatic tumor can occur in the disaggregation flexible containers of the semi-automatic device. [00346] By way of example, in another embodiment of the method of the invention, where the disaggregation process is being supplemented with enzymatic digestion the media formulation for enzymatic digestion must be supplemented with enzymes that aid in protein breakdown causing the cell to cell boundaries to break down. [00347] Various liquid formulations known in the art of cell culturing or cell handling can be used as the liquid formulation used for cell disaggregation and enzymatic digestion of solid tissues, including but not limited to one or more of the following media Organ Preservation Solutions, selective lysis solutions, PBS, DMEM, HBSS, DPBS, RPMI, Iscove’s medium, XVIVO™, AIM-V™, Lactated Ringer's solution, Ringer's acetate, saline, PLASMALYTE™ solution, crystalloid solutions and IV fluids, colloid solutions and IV fluids, five percent dextrose in water (D5W), Hartmann's Solution, DMEM, HBSS, DPBS, RPMI, AIM-V™, Iscove’s medium, XVIVO™, each can be optionally supplemented with additional cell supporting factors e.g. with fetal calf serum, human serum or serum substitutes or other nutrients or cytokines to aid in cell recovery and survival or specific cell depletion. The media can be standard cell media like the above mentioned media or special media for e.g. primary human cell culture (e.g. for endothelia cells, hepatocytes or keratinocytes) or stem cells (e.g. dendritic cell maturation, hematopoietic expansion, keratinocytes, mesenchymal stem cells or T cells). The media may have supplements or reagents well known in the art, e.g. albumins and transport proteins, amino acids and vitamins, metal-ion(s), antibiotics, attachments factors, de-attachment factors, surfactants, growth factors and cytokines, hormones or solubilizing agents. Various media are commercially available e.g. from ThermoFisher, Lonza, or Sigma-Aldrich or similar media manufacturers and suppliers. [00348] The liquid formulation required for enzymatic digestion must have sufficient calcium ions present in the of at least 0.1 mM up to 50 mM with an optimal range of 2 to 7 mM ideally 5 mM. [00349] The solid tissue to be digested can be washed after disaggregation with a liquid formulation containing chelating agents EGTA and EDTA to remove adhesion factors and inhibitory proteins prior to washing and removal of EDTA and EGTA prior to enzymatic digestion. [00350] The liquid formulation required for enzymatic digestion is more optimal with minimal chelating agents EGTA and EDTA which can severely inhibit enzyme activity by removing calcium ions required for enzyme stability and activity. In addition, β- mercaptoethanol, cysteine and 8-hydroxyquinoline-5-sulfonate are other known inhibitory substances. [00351] Processing of tumor material using dissection, enzymatic digestion and homogenization produces a single cell suspension of TILs, in particular UTILs, which can be directly cryopreserved to stabilize the starting material for subsequent processing via the first expansion of the cell suspension of TILs, in particular UTILs, in IL-2 to obtain a first population of TILs, in particular UTILs. [00352] The methods can also comprise the step of cryopreserving the disaggregated tumor, e.g. the cell suspension. Cryopreserving the disaggregated tumor is carried out on the same day as carrying out the step of aseptically disaggregating a tumor resected from a subject thereby producing a disaggregated tumor, wherein the resected tumor is sufficiently disaggregated if it can be cryopreserved without cell damage. For example, cryopreserving is carried out 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, or 22 hours following the step of disaggregating the tumor. Cryopreservation of the disaggregated tumor, as a single cell suspension obtained from the enzymatic disaggregation in the disaggregation module of the semi-automatic device, is carried out by cooling or maintaining the suspension at a temperature between 8 °C and at least -80 °C. Disaggregation could be as quick as 5 mins but most usually 45 mins to 1 hour and the cryopreservation can be a quick as 60 mins or up to 150 mins. In one embodiment, the methods include storing the cryopreserved disaggregated tumor. As described in preferred embodiments, the device comprises at least one cell container for cryopreservation wherein the containers are a flexible container manufactured from resilient deformable material. In this embodiment of the device, the final container is either transferred directly to a freezer -20 to -190 °C or more optimally located in the controlled rate freezing apparatus either associated with the device or supplied separately (manufactured by for example Planer Products or Asymptote Ltd) in which the temperature of the freezing chamber and the flexible storage container(s) employed to contain the enriched disaggregated solid tissue container is controlled either by: injecting a cold gas (normally nitrogen for example Planer products); or by removing heat away from the controlled cooling surface(s). Both methods result in the ability to accurately control with an error of less than 1 °C or more preferable 0.1 °C the freezing process at the required rate for the specific cell(s) to be frozen based on the freezing solution and the desired viability of the product. This cryopreservation process must consider the ice nucleation temperature which is ideally as close as possible to the melting temperature of the freezing solution. Followed by crystal growth in an aqueous solution, water is removed from the system as ice, and the concentration of the residual unfrozen solution increases. As the temperature is lowered, more ice forms, decreasing the residual non-frozen fraction which further increases in concentration. In aqueous solutions, there exists a large temperature range in which ice co-exists with a concentrated aqueous solution. Eventually through temperature reduction the solution reaches the glass transition state at which point the freezing solution and cells move from a viscous solution to a solid-like state below this temperature the cells can undergo no further biological changes and hence are stabilized, for years potentially decades, until required. [00353] Ice nucleation and crystal growth involves release of heat to the freezing solution and the cellular microenvironment and it is desirable to maintain cooling of cells and freezing solution even as the freezing fluid resists temperature changes while undergoing phase change. Depending on whether disaggregation includes enzymatic disaggregation, and what is the optimal temperature of enzymatic digestion for a given enzyme, enzyme concentration and tissue type, temperatures at the start of cryopreservation include, without limitation, 40°C, 39°C, 38°C, 37°C, 36°C, 35°C, 34°C, 33°C, 32°C, 31°C, 30°C, 29°C, 28°C, 27°C, 26°C, 25°C, 24°C, 23°C, 22°C, 21°C, and 20°C, i.e., temperatures ranging from a mammalian body temperature to room temperature, and further include temperatures below room temperature, including but not limited to refrigeration temperatures such as, without limitation, 19°C, 18°C, 17°C, 16°C, 15°C, 14°C, 13°C, 12°C, 11°C, 10°C, 9°C, 8°C, 7°C, 6°C, 5°C, 4°C, 3°C, and 2°C. Target temperatures for cryogenic cooling include, without limitation, -60°C, -65°C, -70°C, -75°C, -80°C, -85°C, -90°C, and temperatures in between as well as colder temperatures down to the temperature of liquid nitrogen vapor storage (-195.79°C). In certain embodiments, the methods and devices used according to the invention are designed or programmed to minimize the time from physiological temperature or digestion temperature to cryostorage temperature. In certain embodiments, the methods and devices used according to the invention for cryopreservation are advantageously designed and programmed for cooling under conditions whereby heat release to, into, around or in an environment including cells, as media crystalizes, is minimized or avoided. In certain embodiments, the methods and devices used according to the invention for cryopreservation are advantageously designed and programmed for cooling under conditions whereby heat release to, into, around or in an environment including cells, as media crystalizes, is minimized or avoided, for example by maintaining a pre-determined rate of temperature change of the cryopreservation media even as nucleation and crystallization of the media releases heat that resists temperature change. In certain embodiments, regulating or programming a rate of temperature change includes regulating the rate of heat extraction from the cryopreservation sample to maintain a predetermined rate of temperature change. In certain embodiments, the cooling rate of the cryopreservation sample is maintained by measuring the temperature of the cryopreservation sample and adjusting the rate of heat extraction through a phase change by a feedback process. In certain embodiments, the cooling rate of the cryopreservation sample is maintained by anticipating a phase change and increasing the rate of heat extraction at the anticipated time of the phase change. In certain embodiments, methods are designed and/or devices programmed for continuous cooling from disaggregation temperature down to a cryogenic target temperature. Exemplary programmed cooling rates include, without limitation, -0.5°C/min, -1°C/min, - 1.5°C/min, -2°C/min, or -2.5°C/min. The cooling rates are program targets and may vary over a cooling cycle. The cooling rates may vary, for example by ± 0.1°C/min, ± 0.2°C/min, ± 0.3°C/min, ± 0.4°C/min, or ± 0.5°C/min. In an embodiment of the invention, the cryopreservation temperature is -80°C ± 10°C and the device is programmed to reduce temperature by 1°C/min or 1.5°C/min or 2°C/min or 1°C/min ± 0.5°C/min or 1.5°C/min ± 0.5°C/min or 2°C/min ± 0.5°C/min. [00354] It will be evident that accurate controlled cooling of TILs is desired. Accordingly, to optimized measurement and control of heat transfer from the TILs, it is advantageous to employ optimize surface to volume ratios, employ cassettes to house cryopreservation containers and facilitate heat transfer, and optimally locate temperature sensors. [00355] Cryopreservation may be employed throughout TIL manufacture including but not limited to i) cryopreservation of a processed tumor sample for use at a later time by thawing and TIL expansion, ii) cryopreservation of a processed tumor sample for use at a later time by thawing and use of tumor cells, iii) cryopreservation of a processed tumor sample for later analysis, iv) cryopreservation of a pre-REP expansion culture for use at a later time by thawing and REP expansion, v) cryopreservation of a portion of a pre-REP expansion culture (such as but not limited to a predetermined portion or to excess cells above a predetermined amount from a pre-REP culture) for use at a later time by thawing and REP expansion, vi) cryopreservation of a post-REP culture for use at a later time in a subsequent pre-REP expansion or REP, or vii) cryopreservation of a post-REP culture for use at a later time by thawing and administering to a subject. [00356] Cryopreserved TIL intermediates, products, and samples may be washed upon thawing prior to use. In certain embodiments cryopreserved tumor digests are thawed, diluted in growth media, and washed one or more times. In certain embodiments, washing comprises centrifugation and growth media change. In certain embodiments, washing comprises filtration and growth media change. In certain embodiments, wash media is mixed into then withdrawn from a closed TIL container, such as a bag or dish and replaced by fresh media. The wash may be automated in a closed system or containers for TILs, wash media, and other components interconnected by tubes and valves. [00357] In certain embodiments, to increase proportions of TILs, TIL subsets. TIL viability, and or TIL potency, upon thawing, dilution, and optional wash, cryopreserved TILs are held in culture prior to outgrowth (i.e. pre-REP expansion). In certain embodiments, the hold time is chosen to maximize total viable cells or fold expansion measured by CD3. In certain embodiments, the hold time may comprise or consist of from 2 to 4 hr. or from 4 to 6 hrs. or from 6 to 9 hrs. or from 9 to 12 hr. or from 12 to 18 hr. or from 18 to 24 hr. [00358] In some embodiments, the present methods provide for obtaining young TILs, which are capable of increased replication cycles upon administration to a subject/patient and as such may provide additional therapeutic benefits over older TILs (i.e., TILs which have further undergone more rounds of replication prior to administration to a subject/patient). Features of young TILs have been described in the literature, for example Donia, at al., Scandinavian Journal of Immunology, 75:157-167 (2012); Dudley et al., Clin Cancer Res, 16:6122-6131 (2010); Huang et al., J Immunother, 28(3):258-267 (2005); Besser et al., Clin Cancer Res, 19(17):OF1- OF9 (2013); Besser et al., J Immunother 32:415-423 (2009); Robbins, et al., J Immunol 2004; 173:7125-7130; Shen et al., J Immunother, 30:123-129 (2007); Zhou, et al., J Immunother, 28:53-62 (2005); and Tran, et al., J Immunother, 31:742-751 (2008), all of which are incorporated herein by reference in their entireties. [00359] The diverse antigen receptors of T and B lymphocytes are produced by somatic recombination of a limited, but large number of gene segments. These gene segments: V (variable), D (diversity), J (joining), and C (constant), determine the binding specificity and downstream applications of immunoglobulins and T-cell receptors (TCRs). The present invention provides a method for generating TILs which exhibit and increase the T-cell repertoire diversity. In some embodiments, the TILs obtained by the present method exhibit an increase in the T-cell repertoire diversity. In some embodiments, the TILs obtained by the present method exhibit an increase in the T-cell repertoire diversity as compared to freshly harvested TILs and/or TILs prepared using other methods than those provide. In some embodiments, the TILs obtained by the present method exhibit an increase in the T-cell repertoire diversity as compared to freshly harvested TILs and/or TILs. In some embodiments, the TILs obtained in the first expansion exhibit an increase in the T-cell repertoire diversity. In some embodiments, the increase in diversity is an increase in the immunoglobulin diversity and/or the T-cell receptor diversity. In some embodiments, the diversity is in the immunoglobulin is in the immunoglobulin heavy chain. In some embodiments, the diversity is in the immunoglobulin is in the immunoglobulin light chain. In some embodiments, the diversity is in the T-cell receptor. In some embodiments, the diversity is in one of the T-cell receptors selected from the group consisting of alpha, beta, gamma, and delta receptors. In some embodiments, there is an increase in the expression of T- cell receptor (TCR) alpha and/or beta. In some embodiments, there is an increase in the expression of T-cell receptor (TCR) alpha. In some embodiments, there is an increase in the expression of T-cell receptor (TCR) beta. In some embodiments, there is an increase in the expression of TCRab (i.e., TCRα/β). [00360] The methods of the invention can also comprise the step of performing a first expansion by culturing the disaggregated tumor in a cell culture medium comprising IL-2 to produce a first population of TILs, in particular UTILs,. The cells resulting from the steps described above are cultured in serum containing IL-2 under conditions that favor the growth of TILs over tumor and other cells. In some embodiments, the tumor digests are incubated in 2 mL wells in media comprising inactivated human AB serum with 6000 IU/mL of IL-2. This primary cell population is cultured for a period of days, generally from 3 to 14 days, resulting in a bulk TIL population, generally about 1x108 bulk TIL cells. In some embodiments, this primary cell population is cultured for a period of 7 to 14 days, resulting in a bulk TIL population, generally about 1x108 bulk TIL cells. In some embodiments, this primary cell population is cultured for a period of 10 to 14 days, resulting in a bulk TIL population, generally about 1x108 bulk TIL cells. In some embodiments, this primary cell population is cultured for a period of about 11 days, resulting in a bulk TIL population, generally about 1x108 bulk TIL cells. [00361] In a preferred embodiment, expansion of TILs may be performed using an initial bulk TIL expansion step as described below and herein, followed by a second expansion (including rapid expansion protocol (REP) steps and followed by restimulation REP steps) as described below and herein. [00362] In an advantageous embodiment, the cryopreserved disaggregated tumor tissue is thawed and resuspended 1:9 in T cell media (T cell culture media contract manufactured for Immetacyte supplemented with the following additives 10% FBS and 3000 IU/mL IL-2) prior to filtration through an inline 100-270 μm filter and centrifugation in a 50 mL centrifuge tube prior to resuspension in 20 mL. A sample may be taken for flow cytometry analysis to quantify a number of HLA-A, B, C and CD58+, and DRAQ7¯ cells. In some embodiments this may be seeded using an alternative manual (such as but not limited to a hemocytometer) or alternative automated total viable cell counting device such as but not limited to NucleoCounter™; Guava®; automated blood analysis and counter; pipette based cell counter such as but not limited to Scepter™. [00363] In one embodiment, resuspended cryopreserved disaggregated tumor tissue is cultured in serum containing IL-2 under conditions that favor the growth of TILs over tumor and other cells. In some embodiments, the tumor digests are incubated in 2 mL wells in media comprising inactivated human AB serum (or, in some cases, as outlined herein, in the presence of an artificial antigen-presenting [aAPC] cell population) with 6000 IU/mL of IL-2. This primary cell population is cultured for a period of days, generally from 10 to 14 days, resulting in a bulk TIL population, generally about 1x108 bulk TIL cells. In some embodiments, the growth media during the first expansion comprises IL-2 or a variant thereof. In some embodiments, the IL is recombinant human IL-2 (rhIL-2). In some embodiments the IL-2 stock solution has a specific activity of 20-30x106 IU/mg for a 1 mg vial. In some embodiments the IL-2 stock solution has a specific activity of 20x106 IU/mg for a 1 mg vial. In some embodiments the IL-2 stock solution has a specific activity of 25x106 IU/mg for a 1 mg vial. In some embodiments the IL-2 stock solution has a specific activity of 30x106 IU/mg for a 1 mg vial. In some embodiments, the IL-2 stock solution has a final concentration of 4-8x106 IU/mg of IL-2. In some embodiments, the IL-2 stock solution has a final concentration of 5-7x106 IU/mg of IL-2. In some embodiments, the IL-2 stock solution has a final concentration of 6x106 IU/mg of IL-2. In some embodiments, the first expansion culture media comprises about 10,000 IU/mL of IL-2, about 9,000 IU/mL of IL-2, about 8,000 IU/mL of IL-2, about 7,000 IU/mL of IL-2, about 6000 IU/mL of IL-2 or about 5,000 IU/mL of IL-2. In some embodiments, the first expansion culture media comprises about 9,000 IU/mL of IL-2 to about 5,000 IU/mL of IL-2. In some embodiments, the first expansion culture media comprises about 8,000 IU/mL of IL-2 to about 6,000 IU/mL of IL-2. In some embodiments, the first expansion culture media comprises about 7,000 IU/mL of IL-2 to about 6,000 IU/mL of IL-2. In some embodiments, the first expansion culture media comprises about 6,000 IU/mL of IL-2. In an embodiment, the cell culture medium further comprises IL-2. In some embodiments, the cell culture medium comprises about 3000 IU/mL of IL-2. In an embodiment, the cell culture medium further comprises IL-2. In a preferred embodiment, the cell culture medium comprises about 3000 IU/mL of IL-2. In an embodiment, the cell culture medium comprises about 1000 IU/mL, about 1500 IU/mL, about 2000 IU/mL, about 2500 IU/mL, about 3000 IU/mL, about 3500 IU/mL, about 4000 IU/mL, about 4500 IU/mL, about 5000 IU/mL, about 5500 IU/mL, about 6000 IU/mL, about 6500 IU/mL, about 7000 IU/mL, about 7500 IU/mL, or about 8000 IU/mL of IL-2. In an embodiment, the cell culture medium comprises between 1000 and 2000 IU/mL, between 2000 and 3000 IU/mL, between 3000 and 4000 IU/mL, between 4000 and 5000 IU/mL, between 5000 and 6000 IU/mL, between 6000 and 7000 IU/mL, between 7000 and 8000 IU/mL, or about 8000 IU/mL of IL-2. [00364] In some embodiments, first expansion culture media comprises about 500 IU/mL of IL-12, about 400 IU/mL of IL-12, about 300 IU/mL of IL-12, about 200 IU/mL of IL-12, about 180 IU/mL of IL-12, about 160 IU/mL of IL-12, about 140 IU/mL of IL-12, about 120 IU/mL of IL-12, or about 100 IU/mL of IL-12. In some embodiments, the first expansion culture media comprises about 500 IU/mL of IL-12 to about 100 IU/mL of IL-12. In some embodiments, the first expansion culture media comprises about 400 IU/mL of IL-12 to about 100 IU/mL of IL-12. In some embodiments, the first expansion culture media comprises about 300 IU/mL of IL-12 to about 100 IU/mL of IL-12. In some embodiments, the first expansion culture media comprises about 200 IU/mL of IL-12. In some embodiments, the cell culture medium comprises about 180 IU/mL of IL-12. In an embodiment, the cell culture medium further comprises IL-12. In a preferred embodiment, the cell culture medium comprises about 180 IU/mL of IL-12. [00365] In some embodiments, first expansion culture media comprises about 500 IU/mL of IL-15, about 400 IU/mL of IL-15, about 300 IU/mL of IL-15, about 200 IU/mL of IL-15, about 180 IU/mL of IL-15, about 160 IU/mL of IL-15, about 140 IU/mL of IL-15, about 120 IU/mL of IL-15, or about 100 IU/mL of IL-15. In some embodiments, the first expansion culture media comprises about 500 IU/mL of IL-15 to about 100 IU/mL of IL-15. In some embodiments, the first expansion culture media comprises about 400 IU/mL of IL-15 to about 100 IU/mL of IL-15. In some embodiments, the first expansion culture media comprises about 300 IU/mL of IL-15 to about 100 IU/mL of IL-15. In some embodiments, the first expansion culture media comprises about 200 IU/mL of IL-15. In some embodiments, the cell culture medium comprises about 180 IU/mL of IL-15. In an embodiment, the cell culture medium further comprises IL-15. In a preferred embodiment, the cell culture medium comprises about 180 IU/mL of IL-15. [00366] In some embodiments, first expansion culture media comprises about 500 IU/mL of IL-18, about 400 IU/mL of IL-18, about 300 IU/mL of IL-18, about 200 IU/mL of IL-18, about 180 IU/mL of IL-18, about 160 IU/mL of IL-18, about 140 IU/mL of IL-18, about 120 IU/mL of IL-18, or about 100 IU/mL of IL-18. In some embodiments, the first expansion culture media comprises about 500 IU/mL of IL-18 to about 100 IU/mL of IL-18. In some embodiments, the first expansion culture media comprises about 400 IU/mL of IL-18 to about 100 IU/mL of IL-18. In some embodiments, the first expansion culture media comprises about 300 IU/mL of IL-18 to about 100 IU/mL of IL-18. In some embodiments, the first expansion culture media comprises about 200 IU/mL of IL-18. In some embodiments, the cell culture medium comprises about 180 IU/mL of IL-18. In an embodiment, the cell culture medium further comprises IL-18. In a preferred embodiment, the cell culture medium comprises about 180 IU/mL of IL-18. [00367] In some embodiments, first expansion culture media comprises about 20 IU/mL of IL-21, about 15 IU/mL of IL-21, about 12 IU/mL of IL-21, about 10 IU/mL of IL-21, about 5 IU/mL of IL-21, about 4 IU/mL of IL-21, about 3 IU/mL of IL-21, about 2 IU/mL of IL-21, about 1 IU/mL of IL-21, or about 0.5 IU/mL of IL-21. In some embodiments, the first expansion culture media comprises about 20 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the first expansion culture media comprises about 15 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the first expansion culture media comprises about 12 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the first expansion culture media comprises about 10 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the first expansion culture media comprises about 5 IU/mL of IL-21 to about 1 IU/mL of IL-21. In some embodiments, the first expansion culture media comprises about 2 IU/mL of IL-21. In some embodiments, the cell culture medium comprises about 1 IU/mL of IL-21. In some embodiments, the cell culture medium comprises about 0.5 IU/mL of IL-21. In an embodiment, the cell culture medium further comprises IL-21. In a preferred embodiment, the cell culture medium comprises about 1 IU/mL of IL-21. [00368] Also contemplated for the culture media are combinations of interleukins, such as but not limited to, IL-2, IL-12, IL-15, IL-18 and IL-21. Other cytokines are also contemplated, such as IL-23, IL-27, IL-35, IL-39, IL-18, IL-36, IL-37, IL-38, IFN-alpha, IFN-beta, IFN-gamma or a combination thereof along with IL-2, IL-12, IL-15, IL-18 and IL-21. Antibodies, such as Th2 blocking reagents, are also contemplated, such as but not limited to, IL-4 (aIL4), anti-IL-4 (aIL4R), anti-IL-5R (aIL5R), anti-IL-5 (aIL5), anti-IL13R (aIL13R), or anti-IL13 (aIL13). [00369] In some embodiments, the first TIL expansion can proceed for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days. In some embodiments, the first TIL expansion can proceed for 1 day to 14 days. In some embodiments, the first TIL expansion can proceed for 2 days to 14 days. In some embodiments, the first TIL expansion can proceed for 3 days to 14 days. In some embodiments, the first TIL expansion can proceed for 4 days to 14 days. In some embodiments, the first TIL expansion can proceed for 5 days to 14 days. In some embodiments, the first TIL expansion can proceed for 6 days to 14 days. In some embodiments, the first TIL expansion can proceed for 7 days to 14 days. In some embodiments, the first TIL expansion can proceed for 8 days to 14 days. In some embodiments, the first TIL expansion can proceed for 9 days to 14 days. In some embodiments, the first TIL expansion can proceed for 10 days to 14 days. In some embodiments, the first TIL expansion can proceed for 11 days to 14 days. In some embodiments, the first TIL expansion can proceed for 12 days to 14 days. In some embodiments, the first TIL expansion can proceed for 13 days to 14 days. In some embodiments, the first TIL expansion can proceed for 14 days. In some embodiments, the first TIL expansion can proceed for 1 day to 13 days. In some embodiments, the first TIL expansion can proceed for 2 days to 13 days. In some embodiments, the first TIL expansion can proceed for 3 days to 13 days. In some embodiments, the first TIL expansion can proceed for 4 days to 13 days. In some embodiments, the first TIL expansion can proceed for 5 days to 13 days. In some embodiments, the first TIL expansion can proceed for 6 days to 13 days. In some embodiments, the first TIL expansion can proceed for 7 days to 13 days. In some embodiments, the first TIL expansion can proceed for 8 days to 13 days. In some embodiments, the first TIL expansion can proceed for 9 days to 13 days. In some embodiments, the first TIL expansion can proceed for 10 days to 13 days. In some embodiments, the first TIL expansion can proceed for 11 days to 13 days. In some embodiments, the first TIL expansion can proceed for 12 days to 13 days. In some embodiments, the first TIL expansion can proceed for 1 day to 12 days. In some embodiments, the first TIL expansion can proceed for 2 days to 12 days. In some embodiments, the first TIL expansion can proceed for 3 days to 12 days. In some embodiments, the first TIL expansion can proceed for 4 days to 12 days. In some embodiments, the first TIL expansion can proceed for 5 days to 12 days. In some embodiments, the first TIL expansion can proceed for 6 days to 12 days. In some embodiments, the first TIL expansion can proceed for 7 days to 12 days. In some embodiments, the first TIL expansion can proceed for 8 days to 12 days. In some embodiments, the first TIL expansion can proceed for 9 days to 12 days. In some embodiments, the first TIL expansion can proceed for 10 days to 12 days. In some embodiments, the first TIL expansion can proceed for 11 days to 12 days. In some embodiments, the first TIL expansion can proceed for 1 day to 11 days. In some embodiments, the first TIL expansion can proceed for 2 days to 11 days. In some embodiments, the first TIL expansion can proceed for 3 days to 11 days. In some embodiments, the first TIL expansion can proceed for 4 days to 11 days. In some embodiments, the first TIL expansion can proceed for 5 days to 11 days. In some embodiments, the first TIL expansion can proceed for 6 days to 11 days. In some embodiments, the first TIL expansion can proceed for 7 days to 11 days. In some embodiments, the first TIL expansion can proceed for 8 days to 11 days. In some embodiments, the first TIL expansion can proceed for 9 days to 11 days. In some embodiments, the first TIL expansion can proceed for 10 days to 11 days. In some embodiments, the first TIL expansion can proceed for 11 days. In some embodiments, REP day 10 is 3 days following electroporation. [00370] In some embodiments, a combination of IL-2, IL-7, IL-15, and/or IL-21 are employed as a combination during the first expansion. In some embodiments, IL-2, IL-7, IL-15, and/or IL- 21 as well as any combinations thereof can be included during the first expansion. In some embodiments, a combination of IL-2, IL-15, and IL-21 are employed as a combination during the first expansion. [00371] In some embodiments, the first expansion is performed in a closed system bioreactor. In some embodiments, a closed system is employed for the TIL expansion, as described herein. In some embodiments, a single bioreactor is employed. In some embodiments, the single bioreactor employed is example a G-REX-10 or a G-REX-100 or advantageously the device of WO 2018/130845. In some embodiments, the closed system bioreactor is a single bioreactor. [00372] Advantageously, the TIL population obtained from the first expansion, referred to as the second TIL population, can be subjected to a second expansion (which can include expansions sometimes referred to as REP. Similarly, in the case where genetically modified TILs will be used in therapy, the first TIL population (sometimes referred to as the bulk TIL population) or the second TIL population (which can in some embodiments include populations referred to as the REP TIL populations) can be subjected to genetic modifications for suitable treatments prior to expansion or after the first expansion and prior to the second expansion. [00373] In certain embodiments, the PBMCs are cryopreserved. Cryopreservation enables prescreening and PBMC inventory maintenance and reduces the number of donors needed for TIL manufacture. [00374] Disaggregated tumor tissue can be thawed. In some embodiments, the TILs obtained from the first expansion are stored until phenotyped for selection. In some embodiments, the TILs obtained from the first are not stored and proceed directly to the second expansion. Thus, the methods comprise the step of performing a second expansion by culturing the first population of TILs, in particular UTILs, with additional IL-2, OKT-3, and antigen presenting cells (APCs), to produce a second population of TILs. In some embodiments, the TILs obtained from the first expansion are not cryopreserved after the first expansion and prior to the second expansion. In some embodiments, the transition from the first expansion to the second expansion occurs at about 3 days, 4, days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days after the cryopreserved 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs at about 3 days to 21 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs at about 4 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs at about 4 days to 10 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs at about 7 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs at about 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments the seeding of the REP culture occurs 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 days after the cryopreserved disaggregated tumor tissue is thawed. [00375] In some embodiments, the transition from the first expansion to the second expansion occurs at 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 1 day to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the first TIL expansion can proceed for 2 days to 14 days. In some embodiments, the transition from the first expansion to the second expansion occurs 3 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 4 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 5 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 6 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 7 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 8 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 9 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 10 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 11 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 12 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 13 days to 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 14 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 1 day to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 2 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 3 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 4 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 5 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 6 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 7 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 8 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 9 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 10 days to 11 days after the cryopreserved disaggregated tumor tissue is thawed. In some embodiments, the transition from the first expansion to the second expansion occurs 11 days after the cryopreserved disaggregated tumor tissue is thawed. [00376] In some embodiments, the TILs or a portion of the TILs from the first expansion are cryopreserved. In certain embodiments, the TILs are divided in two or more portions, one or more portion proceeding to the second expansion, and one or more portion cryopreserved to be used in a later second expansion. In certain embodiments, the number of cells at the end of the first expansion is determined and the culture divided accordingly. In certain embodiments, the average potency of the TILs from the first expansion is determined and the culture is divided accordingly. In certain embodiments, an predetermined minimum number or optimal number of TILs proceeds to the second expansion and the remaining TILs are cryopreserved, and later thawed and used in a further second expansion. In certain embodiments, depending on the number and/or activity of left-over TILs, the cryopreserved TILs, can alternatively be used in a first expansion followed by a second expansion. [00377] In some embodiments, the TILs are not stored after the first expansion and prior to the second expansion, and the TILs proceed directly to the second. In some embodiments, the transition occurs in closed system, as described herein. In some embodiments, the TILs from the first expansion, the second population of TILs, proceeds directly into the second expansion with no transition period. [00378] In some embodiments, the transition from the first expansion to the second expansion is performed in a closed system bioreactor. In some embodiments, a closed system is employed for the TIL expansion, as described herein. In some embodiments, a single bioreactor is employed. In some embodiments, the single bioreactor employed is for example a G-REX-10 or a G-REX-100 or Xuri WAVE bioreactor. In some embodiments, the closed system bioreactor is a single bioreactor. [00379] In some embodiments, the TIL cell population is expanded in number after harvest and initial bulk processing. This further expansion is referred to herein as the second expansion, which can include expansion processes generally referred to in the art as a rapid expansion process. The second expansion is generally accomplished using a culture media comprising a number of components, including feeder cells, a cytokine source, and an anti-CD3 antibody, in a gas-permeable or gas exchanging container. [00380] In some embodiments, the second expansion or second TIL expansion of TIL can be performed using any TIL culture flasks or containers known by those of skill in the art. In some embodiments, the second TIL expansion can proceed for 0 days, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days. In some embodiments, the second TIL expansion can proceed for about 7 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 8 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 9 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 10 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 11 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 12 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 13 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 14 days. In some embodiments, the second TIL expansion can proceed for about 7 days to about 13 days. In some embodiments, the second TIL expansion can proceed for about 8 days to about 13 days. In some embodiments, the second TIL expansion can proceed for about 9 days to about 13 days. In some embodiments, the second TIL expansion can proceed for about 10 days to about 13 days. In some embodiments, the second TIL expansion can proceed for about 11 days to about 13 days. In some embodiments, the second TIL expansion can proceed for about 12 days to about 13 days. In some embodiments, the second TIL expansion can proceed for about 7 days to about 12 days. In some embodiments, the second TIL expansion can proceed for about 8 days to about 12 days. In some embodiments, the second TIL expansion can proceed for about 9 days to about 12 days. In some embodiments, the second TIL expansion can proceed for about 10 days to about 12 days. In some embodiments, the second TIL expansion can proceed for about 11 days to about 12 days. In some embodiments, the second TIL expansion can proceed for about 12 days. In some embodiments, the second TIL expansion can proceed for about 13 days. In some embodiments, the second TIL expansion can proceed for about 14 days. [00381] In an embodiment, the second expansion can be performed in a gas permeable container using the methods of the present disclosure. For example, TILs can be rapidly expanded using non-specific T-cell receptor stimulation in the presence of interleukin-2 (IL-2) or interleukin-7 (IL-7) or interleukin-15 (IL-15); or interleukin-12 (IL-12). The non-specific T-cell receptor stimulus can include, for example, an anti-CD3 antibody, such as about 30 ng/ml of OKT3, a mouse monoclonal anti-CD3 antibody (commercially available from Ortho-McNeil, Raritan, N.J. or Miltenyi Biotech, Auburn, Calif.) or clone UHCT-1 (commercially available from BioLegend, San Diego, Calif., USA). TILs can be expanded to induce further stimulation of the TILs in vitro by including one or more antigens during the second expansion, including antigenic portions thereof, such as epitope(s), of the cancer, which can be optionally expressed from a vector, such as a human leukocyte antigen A2 (HLA-A2) binding peptide, e.g., 0.3 μM MART-1:26-35 (27 L) or gpl 00:209-217 (210M), optionally in the presence of a T-cell growth factor, such as 300 IU/mL IL-2 or IL-15. Other suitable antigens may include, e.g., NY-ESO-1, TRP-1, TRP-2, tyrosinase cancer antigen, MAGE-A3, SSX-2, and VEGFR2, or antigenic portions thereof. TIL may also be rapidly expanded by re-stimulation with the same antigen(s) of the cancer pulsed onto HLA-A2-expressing antigen-presenting cells. Alternatively, the TILs can be further re-stimulated with, e.g., example, irradiated, autologous lymphocytes or with irradiated HLA-A2+ allogeneic lymphocytes and IL-2. In some embodiments, the re-stimulation occurs as part of the second expansion. In some embodiments, the second expansion occurs in the presence of irradiated, autologous lymphocytes or with irradiated HLA-A2+ allogeneic lymphocytes and IL-2. [00382] In an embodiment, the cell culture medium further comprises IL-2. In some embodiments, the cell culture medium comprises about 3000 IU/mL of IL-2. In an embodiment, the cell culture medium comprises about 100 IU/mL, about 200 IU/mL, about 300 IU/mL, about 400 IU/mL, about 500 IU/mL, about 600 IU/mL, about 700 IU/mL, about 800 IU/mL, about 900 IU/mL, 1000 IU/mL, about 1500 IU/mL, about 2000 IU/mL, about 2500 IU/mL, about 3000 IU/mL, about 3500 IU/mL, about 4000 IU/mL, about 4500 IU/mL, about 5000 IU/mL, about 5500 IU/mL, about 6000 IU/mL, about 6500 IU/mL, about 7000 IU/mL, about 7500 IU/mL, or about 8000 IU/mL of IL-2. In an embodiment, the cell culture medium comprises between 1000 and 2000 IU/mL, between 2000 and 3000 IU/mL, between 3000 and 4000 IU/mL, between 4000 and 5000 IU/mL, between 5000 and 6000 IU/mL, between 6000 and 7000 IU/mL, between 7000 and 8000 IU/mL, or between 8000 IU/mL of IL-2. [00383] In an embodiment, the cell culture medium comprises OKT3 antibody. In some embodiments, the cell culture medium comprises about 30 ng/mL of OKT3 antibody. In an embodiment, the cell culture medium comprises about 0.1 ng/mL, about 0.5 ng/mL, about 1 ng/mL, about 2.5 ng/mL, about 5 ng/mL, about 7.5 ng/mL, about 10 ng/mL, about 15 ng/mL, about 20 ng/mL, about 25 ng/mL, about 30 ng/mL, about 35 ng/mL, about 40 ng/mL, about 50 ng/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, about 90 ng/mL, about 100 ng/mL, about 200 ng/mL, about 500 ng/mL, and about 1 μg/mL of OKT3 antibody. In an embodiment, the cell culture medium comprises between 0.1 ng/mL and 1 ng/mL, between 1 ng/mL and 5 ng/mL, between 5 ng/mL and 10 ng/mL, between 10 ng/mL and 20 ng/mL, between 20 ng/mL and 30 ng/mL, between 30 ng/mL and 40 ng/mL, between 40 ng/mL and 50 ng/mL, and between 50 ng/mL and 100 ng/mL of OKT3 antibody. [00384] In some embodiments, a combination of IL-2, IL-7, IL-15, and/or IL-21 are employed as a combination during the second expansion. In some embodiments, IL-2, IL-7, IL-15, and/or IL-21 as well as any combinations thereof can be included during the second expansion. In some embodiments, a combination of IL-2, IL-15, and IL-21 are employed as a combination during the second expansion. In some embodiments, IL-2, IL-15, and IL-21 as well as any combinations thereof can be included. [00385] In some embodiments, the second expansion can be conducted in a supplemented cell culture medium comprising IL-2, OKT-3, and antigen-presenting feeder cells. In some embodiments, the second expansion occurs in a supplemented cell culture medium. In some embodiments, the supplemented cell culture medium comprises IL-2, OKT-3, and antigen- presenting feeder cells. In some embodiments, the second cell culture medium comprises IL-2, OKT-3, and antigen-presenting cells (APCs; also referred to as antigen-presenting feeder cells). In some embodiments, the second expansion occurs in a cell culture medium comprising IL-2, OKT-3, and antigen-presenting feeder cells (i.e., antigen presenting cells). [00386] In some embodiments, the second expansion culture media comprises about 500 IU/mL of IL-15, about 400 IU/mL of IL-15, about 300 IU/mL of IL-15, about 200 IU/mL of IL- 15, about 180 IU/mL of IL-15, about 160 IU/mL of IL-15, about 140 IU/mL of IL-15, about 120 IU/mL of IL-15, or about 100 IU/mL of IL-15. In some embodiments, the second expansion culture media comprises about 500 IU/mL of IL-15 to about 100 IU/mL of IL-15. In some embodiments, the second expansion culture media comprises about 400 IU/mL of IL-15 to about 100 IU/mL of IL-15. In some embodiments, the second expansion culture media comprises about 300 IU/mL of IL-15 to about 100 IU/mL of IL-15. In some embodiments, the second expansion culture media comprises about 200 IU/mL of IL-15. In some embodiments, the cell culture medium comprises about 180 IU/mL of IL-15. In an embodiment, the cell culture medium further comprises IL-15. In a preferred embodiment, the cell culture medium comprises about 180 IU/mL of IL-15. [00387] In some embodiments, the second expansion culture media comprises about 20 IU/mL of IL-21, about 15 IU/mL of IL-21, about 12 IU/mL of IL-21, about 10 IU/mL of IL-21, about 5 IU/mL of IL-21, about 4 IU/mL of IL-21, about 3 IU/mL of IL-21, about 2 IU/mL of IL- 21, about 1 IU/mL of IL-21, or about 0.5 IU/mL of IL-21. In some embodiments, the second expansion culture media comprises about 20 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the second expansion culture media comprises about 15 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the second expansion culture media comprises about 12 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the second expansion culture media comprises about 10 IU/mL of IL-21 to about 0.5 IU/mL of IL-21. In some embodiments, the second expansion culture media comprises about 5 IU/mL of IL-21 to about 1 IU/mL of IL-21. In some embodiments, the second expansion culture media comprises about 2 IU/mL of IL-21. In some embodiments, the cell culture medium comprises about 1 IU/mL of IL-21. In some embodiments, the cell culture medium comprises about 0.5 IU/mL of IL-21. In an embodiment, the cell culture medium further comprises IL-21. In a preferred embodiment, the cell culture medium comprises about 1 IU/mL of IL-21. [00388] In some embodiments the antigen-presenting feeder cells (APCs) are PBMCs. In an embodiment, the ratio of TILs to PBMCs and/or antigen-presenting cells in the rapid expansion and/or the second expansion is about 1 to 25, about 1 to 50, about 1 to 100, about 1 to 125, about 1 to 150, about 1 to 175, about 1 to 200, about 1 to 225, about 1 to 250, about 1 to 275, about 1 to 300, about 1 to 325, about 1 to 350, about 1 to 375, about 1 to 400, or about 1 to 500. In an embodiment, the ratio of TILs to PBMCs in the rapid expansion and/or the second expansion is between 1 to 50 and 1 to 300. In an embodiment, the ratio of TILs to PBMCs in the rapid expansion and/or the second expansion is between 1 to 100 and 1 to 200. [00389] In some embodiments, the second expansion (which can include processes referred to as the REP process) is shortened to 0-14 days. In some embodiments, the second expansion is shortened to 7-11 days. [00390] In the present invention, sets of containers, which are interconnected and have specific separate functions maintain an aseptically closed system to process, optionally enrich but stabilize the disaggregated and cellularized tumor. Essentially the invention provides a rapid pre-sterilized environment to minimize the time required and risk of contamination or operator exposure during the processing of the resected tumor. [00391] The aseptic kit allows for closed solid tissue processing, eliminating the risk of contamination of the final cellularized product compared to standard non-closed tissue processing, especially when the process is performed within a tissue retrieval/procurement site and requires storage prior to final cell processing for its ultimate utility. In addition, safety of the operator is increased due to reduction of direct contact with biological hazardous material, which may contain infectious organisms such as viruses. The kit also enables either all of or a portion of the finally processed cellularized material to be stabilized for either transport or storage prior to being processed for its ultimate utility. [00392] The invention will enable the resected tumor to be processed at the time of resection, or later if required, without impact upon the retrieval procedure or the viability of the cellularized tumor. [00393] In some embodiments, an optional enrichment via a form of physical purification to reduce impurities such as no longer required reagents; cell debris; non-disaggregated tumor tissue and fats can be employed. The aseptic kit can have an optional enrichment module, prior to stabilization, for this purpose. A single cell or small cell number aggregates can be enriched for stabilization after disaggregation by excluding particles and fluids of less than 5 μm or incompletely disaggregated material of or around 200 μm across or larger but this will vary upon the tissue and the efficiency of disaggregation and various embodiments in the form of tissue specific kits may be employed depending upon the tissue or ultimate utility of the disaggregated tumor. [00394] In another embodiment, a single cell suspension is provided after step (c). [00395] In another embodiment, the first population of UTILs requires about 1-250 million UTILs, including 1-20 million UTILS, 20-40 million UTILS, 40-60 million UTILS, 60-80 million UTILS, 80-100 million UTILS, 100-125 million UTILS, 125-150 million UTILS, 150- 200 million UTILS, or 200-250 million UTILS. [00396] In another embodiment, step (e) may further comprise growth of the UTILs out of the resected tumor starting material followed by the rapid expansion of step (f). [00397] In another embodiment, step (e) may be performed for about two weeks and step (f) may be performed for about two weeks. [00398] In another embodiment, additional step (h) involves suspending the second population of UTILs. The suspending may be in buffered saline, human serum albumin, and/or dimethylsulfoxide (DMSO). [00399] The present invention also may comprise a therapeutic population of cryopreserved UTILs obtained by any of the herein disclosed methods. The therapeutic population may comprise about 5x109 to 5x1010 of T cells. [00400] The present invention also encompasses a cryopreserved bag of the herein disclosed therapeutic population. The cryopreserved bag may be for use in intravenous infusion. [00401] The present invention also encompasses a method for treating cancer which may comprise administering the herein disclosed therapeutic population or the herein disclosed cryopreserved bag. The present invention also encompasses the herein disclosed therapeutic population, pharmaceutical composition or cryopreserved bag for use in the treatment of cancer. The cancer may be bladder cancer, breast cancer, cancer caused by human papilloma virus, cervical cancer, head and neck cancer (including head and neck squamous cell carcinoma (HNSCC), lung cancer (including non-small-cell lung cancer (NSCLC)), melanoma, ovarian cancer, renal cancer, or renal cell carcinoma. [00402] In another embodiment, the one or more flexible containers of the aseptic kit comprise a resilient deformable material. [00403] In another embodiment, the one or more flexible containers of the disaggregation module of the aseptic kit comprises one or more sealable openings. The one or more flexible containers of the disaggregation module and/or the stabilization module may also comprise a heat sealable weld. [00404] In another embodiment, the one or more flexible containers of the aseptic kit comprises internally rounded edges. [00405] In another embodiment, the one or more flexible containers of the disaggregation module of the aseptic kit comprises disaggregation surfaces adapted to mechanically crush and shear the solid tumor therein. [00406] In another embodiment, the one or more flexible containers of the enrichment module of the aseptic kit comprises a filter that retains a retentate of cellularized disaggregated solid tumor. [00407] In another embodiment, the one or more flexible containers of the stabilization module of the aseptic kit comprises media formulation for storage of viable cells in solution or in a cryopreserved state. [00408] In another embodiment, the aseptic kit further comprises a digital, electronic, or electromagnetic tag identifier. The tag identifier can relate to a specific program that defines a type of disaggregation and/or enrichment and/or stabilization process, one or more types of media used in said processes, including an optional freezing solution suitable for controlled rate freezing. [00409] In another embodiment, the same flexible container can form part of one or more of the disaggregation module, the stabilization module, and the optional enrichment modules. [00410] In another embodiment, the disaggregation module of the aseptic kit comprises a first flexible container for receipt of the tissue to be processed. [00411] In another embodiment, the disaggregation module of the aseptic kit comprises a second flexible container comprising the media for disaggregation. [00412] In another embodiment, the optional enrichment module of the aseptic kit comprises the first flexible container and a third flexible container for receiving the enriched filtrate. [00413] In another embodiment, both the disaggregation module and the stabilization module of the aseptic kit comprise the second flexible container and the second flexible container comprises digestion media and stabilization media. [00414] In another embodiment, the stabilization module of the aseptic kit comprises a fourth flexible container comprising stabilization media. [00415] In another embodiment, the stabilization module of the aseptic kit also comprises the first flexible container and/or third flexible container for storing and/or undergoing cryopreservation. [00416] The present invention also provides for a method for isolating a therapeutic population of cryopreserved TILs comprising: (a) resecting a tumor from a subject; (b) storing the resected tumor in a single use aseptic kit, wherein the aseptic kit comprises: a disaggregation module for receipt and processing of material comprising solid mammalian tissue; an optional enrichment module for filtration of disaggregated solid tissue material and segregation of non- disaggregated tissue and filtrate; and a stabilization module for optionally further processing and/or storing disaggregated product material, wherein each of the modules comprises one or more flexible containers connected by one or more conduits adapted to enable flow of the tissue material there between; and wherein each of the modules comprises one or more ports to permit aseptic input of media and/or reagents into the one or more flexible containers; (c) aseptically disaggregating the resected tumor in the disaggregation module thereby producing a disaggregated tumor, wherein the resected tumor is sufficiently disaggregated if it can be cryopreserved without cell damage; (d) cryopreserving the disaggregated tumor in the stabilization module; (e) performing a first expansion by culturing the disaggregated tumor in a cell culture medium comprising IL-2 to produce a first population of TILs; (f) optionally performing a second expansion by culturing the first population of TILs with additional IL-2, OKT-3, and a TIL activator, to produce a second population of TILs; (g) harvesting and/or cryopreserving the second population of TILs. [00417] In certain non-limiting embodiments, the TIL activator comprises an antigen presenting cell (APC), or an artificial antigen presenting cell (aAPC), or an antigen fragment or complex or an antibody. [00418] In another embodiment, the automated device further comprises a radio frequency identification tag reader for recognition of the aseptic kit so that it may be scanned and recognized during automated processing, such as within the automated device in embodiments of the present invention. Crucially the tag provides information about the conditions and steps required to be auto processed, so simply by scanning the kit, any automated system used with the kit to process the tissue can be undertaken without further intervention or contamination. Once the tissue sample has been placed in the disaggregation module, it can for example be sealed, manually or automatically, before processing begins. [00419] The programmable processor of the automated device can also recognize the aseptic kit via the tag and subsequently can execute the kit program defining the type of disaggregation, enrichment, and stabilization processes, and the respective media types required for said processes, which include an optional freezing solution suitable for controlled rate freezing. The programmable processor of the automated device is adaptable to communicate with and control the disaggregation module, the enrichment module, and/or the stabilization module. Put another way, the kit is therefore readable by an automated device used to execute a specific fully automatic method for processing the tumor when inserted into such a device. [00420] The programmable processor of the automated device can control the disaggregation module to enable a physical and/or biological breakdown of the solid tissue material. This breakdown can be a physical or enzymatic breakdown of the solid tissue material. Enzymatic breakdown of the solid tissue material can be by one or more media enzyme solutions selected from the group consisting of collagenase, trypsin, lipase, hyaluronidase, deoxyribonuclease, Liberase HI, pepsin, and mixtures thereof. [00421] In another embodiment, the programmable processor controls disaggregation surfaces within the disaggregation flexible containers that mechanically crush and shear the solid tissue. In some embodiments, the disaggregation surfaces are controlled by mechanical pistons. [00422] In another embodiment, the programmable processor controls the stabilization module to cryopreserve the enriched disaggregated solid tissue in the container. This may be achieved using a programmable temperature setting, a condition which is determined by reading the tag of the kit inserted in the device. [00423] In another embodiment, to undertake different functions of the process, one or more of the additional components of the device and/or kit are provided and may be available in any combination. This may include: sensors capable of recognizing whether a disaggregation process has been completed in the disaggregation module prior to transfer of the disaggregated solid tissue to the optional enrichment module; weight sensors to determine an amount of media required in the containers of one or more of the disaggregation module; the enrichment module; and/or the stabilization module and control the transfer of material between respective containers; sensors to control temperature within the containers of the one or more of the disaggregation module; the enrichment module; and/or the stabilization module; at least one bubble sensor to control transfer of media between the input and output ports of each container in the module; at least one pump, optionally a peristaltic pump, to control transfer of media between the input and output ports; pressure sensors to assess the pressure within the enrichment module; one or more valves to control a tangential flow filtration process within the enrichment module; and/or one or more clamps to control the transfer of media between the input and output ports of each module. [00424] In another embodiment, the programmable processor of the automated device is adapted to maintain an optimal storage temperature range in the stabilization module until the container is removed; or executes a controlled freezing step. This allows the UTILs to be stored for short periods (minutes to days) or stored for long periods (multiple days to years) prior to their ultimate utility depending on the type or stabilization process used with the stabilization module. [00425] In another embodiment, the automated device further comprises a user interface. The interface can comprise a display screen to display instructions that guide a user to input parameters, confirm pre-programmed steps, warn of errors, or combinations thereof. [00426] In another embodiment, the automated device is adapted to be transportable and thus may comprise dimensions that permit easy maneuverability and/or aid movement such as wheels, tires, and/or handles. [00427] The present invention also provides a semi-automatic aseptic tissue processing method for isolating a therapeutic population of cryopreserved UTILs comprising the steps of: (a) automatically determining aseptic disaggregation tissue processing steps and their associated conditions from a digital, electronic, or electromagnetic tag identifier associated with an aseptic processing kit, wherein the aseptic kit comprises: a disaggregation module for receipt and processing of material comprising solid mammalian tissue; an optional enrichment module for filtration of disaggregated solid tissue material and segregation of non-disaggregated tissue and filtrate; and a stabilization module for optionally further processing and/or storing disaggregated product material, wherein each of the modules comprises one or more flexible containers connected by one or more conduits adapted to enable flow of the tissue material there between; and wherein each of the modules comprises one or more ports to permit aseptic input of media and/or reagents into the one or more flexible containers; (b) resecting a tumor from a subject; (c) placing the tumor into the flexible plastic container of the disaggregation module of the aseptic kit; (d) processing the tumor by automatically executing the one or more tissue processing steps by communicating with and controlling: the disaggregation module; wherein the resected tumor is aseptically disaggregated thereby producing a disaggregated tumor, wherein the resected tumor is sufficiently disaggregated if it can be cryopreserved without cell damage; the optional enrichment module wherein the disaggregated tumor is filtered to remove disaggregated solid tissue material and to segregate non-disaggregated tissue and filtrate; the stabilization module wherein the disaggregated tumor is cryopreserved; (e) performing a first expansion by culturing the disaggregated tumor in a cell culture medium comprising IL-2 to produce a first population of UTILs; (f) optionally performing a second expansion by culturing the first population of UTILs with additional IL-2, OKT-3, and antigen presenting cells (APCs), to produce a second population of TILs; (g) harvesting and/or cryopreserving the second population of UTILs. [00428] Flexible containers such as bags, may be used to process tissue materials. Processing may include treatments that may separate or breakdown tissue, for example, physical breakdown may be accomplished using agitation, e.g., gentle agitation, a biological and/or enzymatic breakdown may include enzymatic digestion, and/or extraction of components of the tissue materials in the bag. [00429] A flexible container, such as a bag, for processing tissue may include one or more layers made of a sealable polymer having at least three edges of the flexible container which are sealed during manufacturing and an open edge on the flexible container through which tissue material is inserted during use. One or more connectors may be used to couple the flexible container to at least one element through tubing. After tissue is placed in the flexible container, a section of the flexible container proximate the open edge may be sealed or welded to form a seal. The seal may have a width of at least a three mm and be positioned substantially parallel to the open edge and spaced away from the open edge of the flexible container. In some instances, the seal may have a width greater than about five mm. For example, a bag may be sealed after tissue is placed inside to have a seal of least 5 mm positioned proximate the open edge of the bag. The seal may be parallel to the open edge and spaced away from the open edge of the bag. [00430] The flexible container may be further secured using a clamp having protrusions and positioned proximate the seal and spaced further from the open edge of the flexible container than the seal. [00431] In some instances, the seal and the flexible container are constructed such that the flexible container can withstand a 100 N force applied to the flexible container during use. Using a clamp in conjunction with such a seal may be advantageous in some instances depending on the type of material used and/or a structure of the seal. Thus, during use of a flexible container, such as a bag, a combination of a seal and a clamp may be capable of withstanding a 100 N force applied to the flexible container. [00432] In some instances, the seal and the flexible container are constructed such that the flexible container can withstand a 75 N force applied to the flexible container during use. Using a clamp in conjunction with such a seal may be advantageous in some instances depending on the type of material used and/or a structure of the seal. Thus, during use of a flexible container, such as a bag, a combination of a seal and a clamp may be capable of withstanding a 75 N force applied to the flexible container. [00433] A flexible container may be used to hold tissue during processing such as disaggregation of the tissue material. [00434] In some embodiments, a flexible container, such as a bag, may be used for disaggregation of the tissue material, filtration of disaggregated tissue material, and/or segregation of non-disaggregated tissue and filtrate. [00435] Flexible containers such as bags may be formed from a resilient deformable material. Materials for use in flexible containers, such as bags may be selected for one or more properties including but not limited to sealability such as sealability due to heat welding, or use of radio frequency energy, gas permeability, flexibility for example low temperature flexibility (e.g., at - 150ºC, or -195 ºC), elasticity for example low temperature elasticity, chemical resistance, optical clarity, biocompatibility such as cytotoxicity, hemolytic activity, resistance to leaching, having low particulates, high transmissions rates for particular gases (e.g., Oxygen and/or Carbon dioxide), and/or complying with regulatory requirements. [00436] Flexible containers, such as bags, may include indicators. Indicators may be used to identify samples, patients from whom the samples were derived, and/or to track progress of a particular sample through a treatment process. In some instances, indicators may be scanned by an automated or semi-automated system to track progress of a sample. [00437] Marks may be used on a flexible container, such as a bag, to identify where the bag should be placed, treated, sealed, or any other action that may be taken with respect to a bag that includes tissue. Each bag may include multiple marks for sealing. [00438] An open end of the bag may be sealed after tissue is inserted in the bag. Any seal may be formed using a sealing device (e.g., heater sealer) operating at a predetermined pressure, a predetermined temperature, and predetermined time frame. [00439] In some instances, a flexible container, such as a bag may be used as a disaggregation container for use as part of a disaggregation element that may also include a disaggregation device. In some embodiments, media and/or enzymes may be added to a bag within a disaggregation element of a device. For example, a bag may be used with a device that mechanically crushes tissue material placed in the flexible container. [00440] In some embodiments, tissue in a flexible container such as a bag may be sheared during disaggregation. In particular, the flexible container may be configured to shear the tissue material. [00441] Flexible containers may be used in a semi-automated or an automated process for the aseptic disaggregation, stabilization and/or optional enrichment of mammalian cells or cell aggregates. [00442] A kit for extraction of a desired material from tissue may include a disaggregation element in which at least some tissue is treated to form a processed fluid, an enrichment element (e.g., a filter) capable of enriching at least some of the processed fluid to form the desired material, a stabilization element capable of storing a portion of the desired material, and an indicator tag positioned on at least one of the disaggregation element, the enrichment element, or the stabilization element capable of providing at least one of a source of tissue, a status of the tissue with respect to the process, or a identifier. [00443] The desired material may be biological material or components of a particular size. For example, the desired material may be tumor infiltrating lymphocytes (TILs). [00444] Different types of media may be used in the various processes conducted by the disaggregation element and the stabilization element. For example, a cryopreservation media may be provided to the kit and used in the stabilization element to control a rate freezing. [00445] Kit for use in a device where a disaggregation element may include a first flexible container and the stabilization element may include a second flexible container. [00446] An automated device for semi-automated aseptic disaggregation and/or enrichment and/or stabilization of cells or cell aggregates from mammalian solid tissue may include a programmable processor and a kit that includes the flexible container described herein. The automated device may further include an indicator tag reader. For example, an indicator tag reader may be positioned at any element (e.g., disaggregation, enriching, or stabilization of tissue material in the kit). [00447] In some instances, an automated device may further include radio frequency identification tag reader to recognize samples in flexible containers in the kit. [00448] An automated device may include a programmable processor that is capable of recognizing indicators positioned on components of the kit such as a bag via an indicator tag such as a QR code. After determining which sample is in the bag, the programmable processor subsequently executes a program defining the type of disaggregation, enrichment, and stabilization processes and provides the respective media types required for those processes. [00449] A kit for use in an automated device may include a disaggregation flexible container or bag. The programmable processor may control a disaggregation element and disaggregation flexible container to enable a physical and/or biological breakdown of the solid tissue. [00450] A programmable processor may control elements of an automated device such that disaggregation surfaces positioned proximate a disaggregation flexible container may mechanically crush and shear the solid tissue in the disaggregation flexible container, optionally wherein the disaggregation surfaces are mechanical pistons. [00451] Disaggregation elements of a system may be controlled by a processor such that tissue in the disaggregation flexible container to enable a physical and enzymatic breakdown of the solid tissue. One or more media enzyme solutions selected from collagenase, trypsin, lipase, hyaluronidase, deoxyribonuclease, Liberase HI, pepsin, or mixtures thereof may be provided to the disaggregation flexible container to aid in enzymatic breakdown of tissue. [00452] A system may include a kit that includes a disaggregation flexible container and a stabilization flexible container and a programmable processor. The programmable processor may be adapted to control one or more of: the disaggregation element; the enrichment element; and the stabilization element. [00453] A programmable processor may control a stabilization element to cryopreserve the enriched disaggregated solid tissue in the stabilization container. In some embodiments, a predetermined temperature may be programmed. [00454] An automated device may include additional components in a multitude of combinations. Components may include sensors capable of recognizing whether a disaggregation process has been completed in the disaggregation module prior to transfer of the disaggregated solid tissue to the optional enrichment element, weight sensors to determine an amount of media required in the containers of one or more of the disaggregation element, an enrichment element, and/or the stabilization element and control the transfer of material between respective containers, sensors to control temperature within the containers of the one or more of the disaggregation element; the enrichment element; and/or the stabilization element; at least one bubble sensor to control the transfer of media between the input and output ports of each container in the element; at least one pump, optionally a peristaltic pump, to control the transfer of media between the input and output ports; pressure sensors to assess the pressure within the enrichment element; one or more valves to control a tangential flow filtration process within the enrichment element; and/or one or more clamps to control the transfer of media between the input and output ports of each element.
[00455] An automated device may include a programmable processor is adapted to maintain an optimal storage temperature range in the stabilization module until the container is removed. In an embodiment, the programmable processor may execute a controlled freezing step.
[00456] In some instances, an automated device may include a user interface. An interface of an automated device may include a display screen to display instructions that guide a user to input parameters, confirm pre-programmed steps, warn of errors, or combinations thereof. [00457] An automated device as described herein may be adapted to be transportable.
[00458] An automatic tissue processing method may include automatically determining conditions for processing steps and the associated conditions from a digital, electronic or electromagnetic tag indicator associated with a component of a kit. During use a tissue sample may be placed into a flexible container of the kit having at least one open edge. After positioning tissue in the flexible container, the open edge may be sealed. During use tissue may be processed by automatically executing one or more tissue processing steps by communicating information associated with the indicator and controlling conditions near the flexible container and/or positions of the flexible container. Further, addition of materials to the kit may be controlled based on information associated with indicators. At least some of the processed tissue may be filtered such that a filtered fluid is generated. At least some of the filtered fluid may be provided to a cryopreservative flexible container to stabilize the desired material present in the filtered fluid.
[00459] Processing as described herein may include agitation, extraction, and enzymatic digestion of at least a portion of the tissue sample in the flexible container. In some instances, this processing of tissue may result in the extraction of a desired material from a tissue sample. For example, tumor infiltrating lymphocytes (TILs) may be extracted from a tissue sample. [00460] Flexible containers, such as bags, for use in the methods described herein may include heat-sealable material. [00461] Tissue processing and extraction from the tissue materials using a cryopreservation kit may result isolation of the desired material. In particular, materials such as tumor infiltrating lymphocytes (TILs) may be the desired material. [00462] In some instances, a cryopreservation kit and/or components thereof described herein may be single use in an automated and/or a semi-automated process for the disaggregation, enrichment, and/or stabilization of cells or cell aggregates. In some embodiments, bags for use in a cryopreservation kit such as a collection bag may in some embodiments be used for multiple processes. For example, collection bags may be repeatedly sealed in different locations to create separate compartments for processing of a tissue sample such as a biopsy sample and/or solid tissue. [00463] Flexible containers, such as bags, for use in the invention described herein include a collection bag and a cryopreservation bag may include at least a portion made from a predetermined material such as a thermoplastic, polyolefin polymer, ethylene vinyl acetate (EVA), blends such as copolymers, for example, a vinyl acetate and polyolefin polymer blend (i.e., OriGen Biomedical EVO film), a material that includes EVA, and/or coextruded layers of sealable plastics. A collection bag, such as a tissue collection bag of the invention may include a bag for receiving tissue made from a predetermined material such as ethylene vinyl acetate (EVA) and/or a material including EVA. Materials for use in the bag may be selected for specific properties. In an embodiment, bags, including collection bags may be made substantially from a vinyl acetate and polyolefin polymer blend. For example, a property of interest that may be used to select a material for cryopreservation kit component such as a collection bag and/or the associated tubing may relate to heat sealing. [00464] Materials for use in the bag may be selected for a specific property and/or a selection of properties, for example, sealability such as heat sealability, gas permeability, flexibility for example low temperature flexibility, elasticity for example low temperature elasticity, chemical resistance, optical clarity, biocompatibility such as cytotoxicity, hemolytic activity, resistance to leaching, having low particulates. [00465] In some embodiments, materials may be selected for specific properties for use in a coextruded material to form at least one layer of a bag. Layers may be constructed such that when constructed an interior layer of the bag is relatively biocompatible, that is the material on an inner surface of the bag is stable and does not leach into the contents of the bag. [00466] For example, a property of interest that may be used to select a material for kit component such as a collection bag, a cryopreservation bag, and/or the associated tubing may relate to sealing, for example heat sealing. [00467] Bags, such as collection bags and/or cryopreservation bags, and any associated tubing may be generally clear, transparent, translucent, any color desired, or a combination thereof. Tissue collection bags and/or tubing may be generally fabricated in ways analogous to the fabrication of closed and/or sealed blood and/or cryopreservation bags and the associated tubing. Tubing in the invention may be constructed from any desired material including, but not limited to polyvinyl chloride (PVC). For example, PVC may be a desired material as PVC is advantageous for welding and/or sealing. [00468] In some embodiments, at least one end of a collection bag may be open for receiving tissue. In particular, in an embodiment, a tissue sample, for example from a biopsy may be placed in the bag through the open end, for example, a top end. In some cases, the biopsy sample may be cancerous tissue from an animal (e.g., domestic animal such as dog or cat) or a human. [00469] After tissue is positioned in the bag, the bag may be sealed, and then may be processed. Processing may include agitation, e.g., gentle agitation, extraction, and/or enzymatic digestion of the tissue in the bag. Tissue processing and extraction of a desired material, such as tumor infiltrating lymphocytes (TILs), can be in a closed system. Advantageous or preferred embodiments may include indicators to identify the patient from whom the tissue was collected and/or marks to show where the collection bag may be clamped, sealed, acted upon by a device, and/or affixed in place in an instrument. [00470] In some embodiments, bag may be formed from a sealable material. For example, bag may be formed from materials including, but not limited to polymers such as synthetic polymers including aliphatic or semi-aromatic polyamides (e.g., Nylon), ethylene-vinyl acetate (EVA) and blends thereof, thermoplastic polyurethanes (TPU), polyethylenes (PE), a vinyl acetate and polyolefin polymer blends, and/or combinations of polymers. Portions of a bag may be sealed and/or welded with energy such as heat, radio frequency energy, high frequency (HF) energy, dielectric energy, and/or any other method known in the art. [00471] A collection bag may be used as a processing and/or disaggregation bag. Collection bags may have width in a range from about 4 cm to about 12 cm and a width in a range from about 10 cm to about 30 cm. For example, a collection bag for use in processing may have a width of about 7.8 cm and a length of about 20 cm. In particular, a bag may be heat sealable, for example, using an EVA polymer or blends thereof, a vinyl acetate and polyolefin polymer blend, and/or one or more polyamides (Nylon). [00472] Indicators may include, but are not limited to codes, letters, words, names, alphanumeric codes, numbers, images, bar codes, quick response (QR) codes, tags, trackers such as smart tracker tags or bluetooth trackers, and/or any indicator known in the art. In some embodiments, indicators may be printed on, etched on, and/or adhered to a surface of a component of a kit. Indicators may also be positioned on a bag using an adhesive, for example, a sticker or tracker may be placed on a bag and/or on multiple bags. Collection bags and/or cryopreservation kit may include multiple indicators such as numeric codes and/or QR codes. [00473] Indicators, for example QR codes, tags such as smart tags, and/or trackers may be used to identify a sample within a bag as well as to instruct a device's processor such that the device runs a specific program according to a type of disaggregation, enrichment, and/or stabilization processes that are conducted in cryopreservation kits. Different types of media may be used in these processes, for example, enzyme media, tumor digest media and/or cryopreservation media which may allow for a controlled rate of freezing. In some embodiments, cryopreservation kit and/or components thereof may include indicators that may be readable by an automated device. The device may then execute a specific fully automatic method for processing tissue when inserted to such a device. The invention is particularly useful in a sample processing, particularly automated processing. In some instances, the cryopreservation kit and/or components thereof described herein may be single use in an automated and/or a semi-automated process for the disaggregation, enrichment, and/or stabilization of cells or cell aggregates. In some embodiments, bags for use in a cryopreservation kit such as a collection bag may in some embodiments be used for multiple processes. For example, collection bags may be repeatedly sealed in different locations to create separate compartments for processing of a tissue sample such as a biopsy sample and/or solid tissue. [00474] Further, marks may be placed at various locations on bags, such as tissue collection bags to indicate where the bags may be sealed, clamped, and/or affixed to an object. In some embodiments, marks showing where a bag may be clamped, sealed, and/or affixed to an object, such as instrument, may be positioned on the bag prior to use. For example, one or more marks may be positioned on a bag during manufacturing. [00475] Positioners may be used to ensure that tissue material in bags can be treated properly during use, for example, positioning proximate an instrument. In some systems, the positioners may facilitate the use of the bags described herein in automated systems. In particular, positioners may be used to move bag through an automated system. [00476] Use of an indicator, such as a QR code may allow for tracking of process steps for a specific sample such that it is possible to follow the sample through a given process. [00477] Cells are transferred to a container for use in administration to a patient. In some embodiments, once a therapeutically sufficient number of TILs are obtained using the expansion methods described above, they are transferred to a container for use in administration to a patient. [00478] In an embodiment, TILs expanded using APCs of the present disclosure are administered to a patient as a pharmaceutical composition. In an embodiment, the pharmaceutical composition is a suspension of TILs in a sterile buffer. TILs expanded using PBMCs of the present disclosure may be administered by any suitable route as known in the art. In some embodiments, the T-cells are administered as a single intra-arterial or intravenous infusion, which preferably lasts approximately 30 to 60 minutes. Other suitable routes of administration include intraperitoneal, intrathecal, and intralymphatic. [00479] In an embodiment, TILs expanded using the methods of the present disclosure are administered to a patient as a pharmaceutical composition. In an embodiment, the pharmaceutical composition is a suspension of TILs in a sterile buffer. TILs expanded using PBMCs of the present disclosure may be administered by any suitable route as known in the art. In some embodiments, the T-cells are administered as a single intra-arterial or intravenous infusion, which preferably lasts approximately 30 to 60 minutes. Other suitable routes of administration include intraperitoneal, intrathecal, and intralymphatic administration. [00480] Any suitable dose of TILs can be administered. In some embodiments, from about 2.3x1010 to about 13.7x1010 TILs are administered, with an average of around 7.8x1010 TILs, particularly if the cancer is melanoma. In an embodiment, about 1.2x1010 to about 4.3x1010 of TILs are administered. In some embodiments, about 3x1010 to about 12x1010 TILs are administered. In some embodiments, about 4x1010 to about 10x1010 TILs are administered. In some embodiments, about 5x1010 to about 8x1010 TILs are administered. In some embodiments, about 6x1010 to about 8x1010 TILs are administered. In some embodiments, about 7x1010 to about 8x1010 TILs are administered. In some embodiments, the therapeutically effective dosage is about 2.3x1010 to about 13.7x1010. In some embodiments, the therapeutically effective dosage is about 7.8x1010 TILs, particularly of the cancer is melanoma. In some embodiments, the therapeutically effective dosage is about 1.2x1010 to about 4.3x1010 of TILs. In some embodiments, the therapeutically effective dosage is about 3x1010 to about 12x1010 TILs. In some embodiments, the therapeutically effective dosage is about 4x1010 to about 10x1010 TILs. In some embodiments, the therapeutically effective dosage is about 5x1010 to about 8x1010 TILs. In some embodiments, the therapeutically effective dosage is about 6x1010 to about 8x1010 TILs. In some embodiments, the therapeutically effective dosage is about 7x1010 to about 8x1010 TILs. [00481] In some embodiments, the number of the TILs provided in the pharmaceutical compositions of the invention is about 1x106, 2x106, 3x106, 4x106, 5x106, 6x106, 7x1068x106, 9x106, 1x107, 2x107, 3x107, 4x107, 5x107, 6x107, 7x107, 8x107, 9x107, 1x108, 2x108, 3x108, 4x108, 5x108, 6x108, 7x108, 8x108, 9x108, 1x109, 2x109, 3x109, 4x109, 5x109, 6x109, 7x109, 8x109, 9x109, 1x1010, 2x1010, 3x1010, 4x1010, 5x1010, 6x1010, 7x1010, 8x1010, 9x1010, 1x1011, 2x1011, 3x1011, 4x1011, 5x1011, 6x1011, 7x1011, 8x1011, 9x1011, 1x1012, 2x1012, 3x1012, 4x1012, 5x1012, 6x1012, 7x1012, 8x1012, 9x1012, 1x1013, 2x1013, 3x1013, 4x1013, 5x1013, 6x1013, 7x1013, 8x1013, and 9x1013. In an embodiment, the number of the TILs provided in the pharmaceutical compositions of the invention is in the range of 1x106 to 5x106, 5x106 to 1x107, 1x107 to 5x107, 5x107to 1x108, 1x108 to 5x108, 5x108 to 1x109, 1x109 to 5x109, 5x109 to 1x1010, 1x1010 to 5x1010, 5x1010 to 1x1011, 5x1011 to 1x1012, 1x1012 to 5x1012, and 5x1012 to 1x1013. [00482] In some embodiments, the concentration of the TILs provided in the pharmaceutical compositions of the invention is less than, for example, 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002% or 0.0001% w/w, w/v or v/v of the pharmaceutical composition. [00483] In some embodiments, the concentration of the TILs provided in the pharmaceutical compositions of the invention is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 11.75%, 11.50%, 11.25% 11%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25% 7%, 6.75%, 6.50%, 6.25% 6%, 5.75%, 5.50%, 5.25% 5%, 4.75%, 4.50%, 4.25%, 4%, 3.75%, 3.50%, 3.25%, 3%, 2.75%, 2.50%, 2.25%, 2%, 1.75%, 1.50%, 125%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002% or 0.0001% w/w, w/v, or v/v of the pharmaceutical composition. [00484] In some embodiments, the concentration of the TILs provided in the pharmaceutical compositions of the invention is in the range from about 0.0001% to about 50%, about 0.001% to about 40%, about 0.01% to about 30%, about 0.02% to about 29%, about 0.03% to about 28%, about 0.04% to about 27%, about 0.05% to about 26%, about 0.06% to about 25%, about 0.07% to about 24%, about 0.08% to about 23%, about 0.09% to about 22%, about 0.1% to about 21%, about 0.2% to about 20%, about 0.3% to about 19%, about 0.4% to about 18%, about 0.5% to about 17%, about 0.6% to about 16%, about 0.7% to about 15%, about 0.8% to about 14%, about 0.9% to about 12% or about 1% to about 10% w/w, w/v or v/v of the pharmaceutical composition. [00485] In some embodiments, the concentration of the TILs provided in the pharmaceutical compositions of the invention is in the range from about 0.001% to about 10%, about 0.01% to about 5%, about 0.02% to about 4.5%, about 0.03% to about 4%, about 0.04% to about 3.5%, about 0.05% to about 3%, about 0.06% to about 2.5%, about 0.07% to about 2%, about 0.08% to about 1.5%, about 0.09% to about 1%, about 0.1% to about 0.9% w/w, w/v or v/v of the pharmaceutical composition. [00486] In some embodiments, the amount of the TILs provided in the pharmaceutical compositions of the invention is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g, 0.01 g, 0.009 g, 0.008 g, 0.007 g, 0.006 g, 0.005 g, 0.004 g, 0.003 g, 0.002 g, 0.001 g, 0.0009 g, 0.0008 g, 0.0007 g, 0.0006 g, 0.0005 g, 0.0004 g, 0.0003 g, 0.0002 g, or 0.0001 g. [00487] In some embodiments, the amount of the TILs provided in the pharmaceutical compositions of the invention is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065 g, 0.07 g, 0.075 g, 0.08 g, 0.085 g, 0.09 g, 0.095 g, 0.1 g, 0.15 g, 0.2 g, 0.25 g, 0.3 g, 0.35 g, 0.4 g, 0.45 g, 0.5 g, 0.55 g, 0.6 g, 0.65 g, 0.7 g, 0.75 g, 0.8 g, 0.85 g, 0.9 g, 0.95 g, 1 g, 1.5 g, 2 g, 2.5, 3 g, 3.5, 4 g, 4.5 g, 5 g, 5.5 g, 6 g, 6.5 g, 7 g, 7.5 g, 8 g, 8.5 g, 9 g, 9.5 g, or 10 g. [00488] The TILs provided in the pharmaceutical compositions of the invention are effective over a wide dosage range. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the gender and age of the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician. The clinically-established dosages of the TILs may also be used if appropriate. The amounts of the pharmaceutical compositions administered using the methods herein, such as the dosages of TILs, will be dependent on the human or mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the active pharmaceutical ingredients and the discretion of the prescribing physician. [00489] In some embodiments, TILs may be administered in a single dose. Such administration may be by injection, e.g., intravenous injection. In some embodiments, TILs may be administered in multiple doses. Dosing may be once, twice, three times, four times, five times, six times, or more than six times per year. Dosing may be once a month, once every two weeks, once a week, or once every other day. Administration of TILs may continue as long as necessary. [00490] In some embodiments, an effective dosage of TILs is about 1x106, 2x106, 3x106, 4x106, 5x106, 6x106, 7x106, 8x106, 9x106, 1x107, 2x107, 3x107, 4x107, 5x107, 6x107, 7x107, 8x107, 9x107, 1x108, 2x108, 3x108, 4x108, 5x108, 6x108, 7x108, 8x108, 9x108, 1x109, 2x109, 3x109, 4x109, 5x109, 6x109, 7x109, 8x109, 9x109, 1x1010, 2x1010, 3x1010, 4x1010, 5x1010, 6x1010, 7x10108x1010, 9x1010, 1x1011, 2x1011, 3x1011, 4x1011, 5x1011, 6x1011, 7x1011, 8x1011, 9x1011, 1x1012, 2x1012, 3x1012, 4x1012, 5x1012, 6x1012, 7x1012, 8x1012, 9x1012, 1x1013, 2x1013, 3x1013, 4x1013, 5x1013, 6x1013, 7x1013, 8x1013, and 9x1013. In some embodiments, an effective dosage of TILs is in the range of 1x106 to 5x106, 5x106 to 1x107, 1x107 to 5x107, 5x107 to 1x108, 1x108 to 5x108, 5x108to 1x109, 1x109 to 5x109, 5x109 to 1x1010, 1x1010 to 5x1010, 5x1010to 1x1011, 5x1011 to 1x1012, 1x1012 to 5x1012, and 5x1012 to 1x1013. [00491] In some embodiments, an effective dosage of TILs is in the range of about 0.01 mg/kg to about 4.3 mg/kg, about 0.15 mg/kg to about 3.6 mg/kg, about 0.3 mg/kg to about 3.2 mg/kg, about 0.35 mg/kg to about 2.85 mg/kg, about 0.15 mg/kg to about 2.85 mg/kg, about 0.3 mg to about 2.15 mg/kg, about 0.45 mg/kg to about 1.7 mg/kg, about 0.15 mg/kg to about 1.3 mg/kg, about 0.3 mg/kg to about 1.15 mg/kg, about 0.45 mg/kg to about 1 mg/kg, about 0.55 mg/kg to about 0.85 mg/kg, about 0.65 mg/kg to about 0.8 mg/kg, about 0.7 mg/kg to about 0.75 mg/kg, about 0.7 mg/kg to about 2.15 mg/kg, about 0.85 mg/kg to about 2 mg/kg, about 1 mg/kg to about 1.85 mg/kg, about 1.15 mg/kg to about 1.7 mg/kg, about 1.3 mg/kg mg to about 1.6 mg/kg, about 1.35 mg/kg to about 1.5 mg/kg, about 2.15 mg/kg to about 3.6 mg/kg, about 2.3 mg/kg to about 3.4 mg/kg, about 2.4 mg/kg to about 3.3 mg/kg, about 2.6 mg/kg to about 3.15 mg/kg, about 2.7 mg/kg to about 3 mg/kg, about 2.8 mg/kg to about 3 mg/kg, or about 2.85 mg/kg to about 2.95 mg/kg. [00492] In some embodiments, an effective dosage of TILs is in the range of about 1 mg to about 500 mg, about 10 mg to about 300 mg, about 20 mg to about 250 mg, about 25 mg to about 200 mg, about 1 mg to about 50 mg, about 5 mg to about 45 mg, about 10 mg to about 40 mg, about 15 mg to about 35 mg, about 20 mg to about 30 mg, about 23 mg to about 28 mg, about 50 mg to about 150 mg, about 60 mg to about 140 mg, about 70 mg to about 130 mg, about 80 mg to about 120 mg, about 90 mg to about 110 mg, or about 95 mg to about 105 mg, about 98 mg to about 102 mg, about 150 mg to about 250 mg, about 160 mg to about 240 mg, about 170 mg to about 230 mg, about 180 mg to about 220 mg, about 190 mg to about 210 mg, about 195 mg to about 205 mg, or about 198 to about 207 mg. [00493] An effective amount of the TILs may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, topically, by transplantation, or by inhalation. [00494] Additional exemplary and non-limiting procedures for collection of tumor material, cryopreservation, and TIL manufacture are provided below. [00495] The starting material for TIL manufacturing is a disaggregated and cryopreserved cell suspension containing autologous TIL and tumor cells from an eligible patient. An exemplary flow diagram is provided for collection and processing of the tumor starting material. [00496] The tumor is surgically resected and then trimmed to remove visibly necrotic tissue, visibly healthy (non-cancerous) tissue, fat tissue, and excess blood. The trimmed tumor weight should be greater than or equal to 2 grams (≥ 2 grams). Tumors weighing over 7 g may be divided into smaller portions and individually disaggregated. [00497] Each tumor fragment is placed into an individual sterile bag containing media, collagenase and DNAse. Exemplary reagents are shown in the following table:
Figure imgf000134_0001
[00498] The bag is then heat sealed and its contents are disaggregated to generate a homogeneous cell suspension containing tumor and TIL. Disaggregation is performed by a device, such as the Tiss-U-Stor device described herein, which runs a program to deliver a defined number of repeated physical compression events, with a defined compression pressure over a defined duration to ensure enzyme access into the tumor tissue thereby accelerating enzymatic digestion. The number of cycles, pressure, temperature, and duration are recorded for each individual tumor. [00499] The homogenized cell suspension is then aseptically filtered using a 200 μm filter (Baxter, RMC2159) and the filtrate passed aseptically into the cryopreservation bag. BloodStor 55-5 (Biolife Solutions, Bothell, WA) is aseptically added to achieve 5% DMSO. The cell suspension is then cryopreserved using the Tiss-U-Stor device with a defined cooling program, and the measured temperature profile is recorded for each individual cell suspension derived from each tumor portion. The cryopreserved cell suspension is stored in vapor-phase of liquid nitrogen. [00500] The cryopreserved cell suspension recommended storage condition is ≤ -130°C. [00501] The cell suspension is transported from the clinical site to the GMP cell therapy manufacturing site by a qualified courier service packaged in a container validated to ensure the cryopreserved cell suspension is maintained at ≤ -130°C. [00502] Tiss-u-Stor. Resected tumors are evaluated for weight and condition. For each tumor fragment, extraneous material is removed and the fragment weighed. A CS50N bag is opened, up to about 7g of tumor is added and the bag is then sealed.15 ml of EDM digest medium is added to the bag with 2μl gentamicin/amphotericin per ml EDM by syringe via needleless port followed by removal of air from the from the bag into the syringe. [00503] The tumor tissue and disaggregation media in the disaggregation bag is placed in the temperature controlled tissue disaggregator. The temperature is increased from ambient temperature to 35 ^C at a rate of 1.5 ^C/min and maintained at 35 ^C for a total of about 45 minutes during which time the disaggregator is active at 240 cycles per minute. [00504] Once disaggregated the tumor material is filtered through an inline filter into a secondary freezing bag.1.5 ml of Blood stor (DMSO) is injected via a needleless port and air removed. [00505] 2 ml. of the suspension is withdrawn for testing. [00506] For optional cryopreservation, the cryobag is loaded into a freezing cassette and the freezing cassette placed in the Via freeze. The Via freeze is then cooled to -80 ^C, preferably directly from 35 ^C to -80 ^C at a rate of -2 ^C/min. [00507] The frozen cryobag is then transferred to liquid nitrogen storage. [00508] Autologous tissue used for culturing in the United Kingdom (UK) should conform to HTA-GD-20, Guide to Quality and Safety Assurance for Human Tissue and Cells for Patient Treatment, established by the UK’s Human Tissue Authority with suitable consent, Chain of Identity, Chain of Custody and screening to confirm donors are negative for Hepatitis B virus, Hepatitis C virus, HIV-1 & 2, HTLV-1 & 2, and Syphilis. [00509] Manufacturing involves outgrowth and expansion from a cryopreserved cell suspension containing TILs and tumor cells derived from a resected tumor. If the tumor is greater than about 7 g, the resection process generates multiple cryopreserved cell suspensions, where each cell suspension derives from a 2 – 7 g tumor fragment. Typically, only one cell suspension is needed to be thawed for 1 TIL outgrowth while the remaining cryopreserved cell suspensions remain in GMP control and held at the recommended storage condition (vapor phase of liquid nitrogen). [00510] In certain embodiments the cell suspension has been filtered after disaggregation, prior to cryopreservation. Exemplary Manufacturing Raw Materials are provided in the following table:
Figure imgf000136_0001
[00511] T cell medium (TCM) contains Albumin (human), human Holo Transferrin, and animal origin cholesterol. The source plasma used to manufacture Albumin and Transferrin are sourced from the USA and the donors are tested for adventitious agents. [00512] Cholesterol is sourced from sheep wool grease originating in Australia/New Zealand, which complies with USDA regulations prohibiting ruminant original material from countries with reported cases of transmission spongiform encephalopathy (TSE). [00513] Fetal Bovine Serum (FBS) is sourced from Australia / New Zealand in compliance with the USDA regulations prohibiting ruminant original material from countries with reported cases of transmission spongiform encephalopathy (TSE). The FBS is tested in compliance with 21 CFR part 113.47, specifically including: bluetongue virus, bovine adenovirus, bovine parvovirus, bovine respiratory syncytial virus, bovine viral diarrhea virus, rabies virus, reovirus, cytopathic agents, haemadsorbing agents. The FBS is heat inactivated at 56°C for 30 minutes and triple 0.1 μm filtered to provide two orthogonal viral removal steps. [00514] Human AB Serum is sourced from Valley Biomedical, an FDA registered establishment (1121958). Each donor unit is tested for Hepatitis B surface Antigen (HBsAg), Hepatitis B Virus (HBV) Nucleic acid Amplification Test (NAT), anti-Human Immunodeficiency Virus (HIV) type 1 and 2, HIV-1 NAT, anti-Hepatitis C Virus (HCV), HCV NAT, and a test for syphilis by FDA approved methods. The serum is heat inactivated at 56°C for 30 minutes and 0.1 μm filtered. [00515] Irradiated Buffy Coat sourcing, preparation, shipment and storage: The Scottish National Blood Transfusion Service (SNBTS) screens donors, collects the blood component, prepares and irradiates buffy coats. The SNBTS is licensed by the United Kingdom’s Human Tissue Authority (license number 11018) in accordance with the Blood, Safety and Quality Regulations (2005) to procure, process, test, store and distribute blood, blood components and tissues. [00516] Healthy donor screening meets or exceeds the requirements described in the United States Code of Federal Regulations (CFR) Title 21 Part 1271.75 with the exception that donors live in the United Kingdom. While this presents a theoretical risk of sporadic Creutzfeldt-Jakob Disease (sCJD) or variant Creutzfeldt-Jakob Disease (vCJD), the United Kingdom has a robust national surveillance program. The most recent annual report, covering May 1990 to December 31st 2018 (National CJD Research & Surveillance Unit, 2018), confirms the incidence of sCJD in the UK is comparable to those observed elsewhere in the world, including countries that are free of bovine spongiform encephalopathy (BSE). There have been no reported cases of vCJD in 2017 through April 5th 2020, and only two cases identified nationally since January 1st 2012 (NCJDRSU Monthly Report, 2020). This rigorous surveillance network has eliminated transfusion transmitted vCJD infections with none reported since 2007 (National CJD Research & Surveillance Unit, 2018). Exemplary eligible donor testing meets 21 CFR Part 1271.85 requirements and adds Hepatitis E testing which is not required.
Figure imgf000138_0001
[00517] The licensed blood establishment prepares clinical grade irradiated buffy coats which are suitable to treat patients with severe neutropenia. To prepare the buffy coats, blood is centrifuged to form three layers: the red blood cell layer, the buffy coat layer and the plasma layer. Buffy coats from 10 donors are irradiated with 25 to 50 Gy irradiation to arrest cell growth. The clinical grade irradiated buffy coats are prepared and shipped to the GMP manufacturing facility by overnight courier using a controlled temperature shipper including a temperature monitor. The shipment occurs one day before use in the manufacturing process. [00518] Upon receipt, the buffy coats are held at 15 – 30°C until use in manufacturing. [00519] Irradiated Feeder Cell Preparation. Buffy coats from up to ten unique donors are pooled, then centrifuged by Ficoll gradient density centrifugation to harvest peripheral blood mononuclear cells (PBMCs). Approximately 4 x 109 viable white blood cells are resuspended in TCM supplemented with approximately 8% human AB serum, 3000 IU/mL IL-2 and 30 ng OKT-3 in a closed static cell culture bag. The PBMC are released per specification.
Figure imgf000138_0002
[00520] The PBMC are also tested for sterility and mycoplasma. Immediately prior to starting step 3, a sample of the formulated feeder cell, including media, IL-2 and OKT3, is removed. This sample is incubated and analyzed on days 13, 17 and 18 to confirm that the feeder cells do not expand. [00521] Albumin (human), also known as Human Serum Albumin (HSA), is sourced from US donors. All plasma donations are individually tested and non-reactive to HBsAg, anti-HIV 1, anti-HIV 2, and anti-HCV antibodies. Each plasma pool is tested and found negative for HBsAg, anti-HIV 1, anti-HIV 2, and HCV-RNA by NAT. The HSA product is manufactured according to GMP regulations fulfilling the production and testing criteria of US and European Pharmacopoeia. [00522] TIL Outgrowth. The cell suspension is seeded at approximately 0.25 x 106 to 0.75 x 106 viable cells/mL into TCM supplemented with 10% FBS, 0.25 μg/mL Amphotericin B with 10 μg/mL Gentamicin (Life Technologies, Grand Island, NY), and interleukin-2 (IL-2; aldesluekin) 3000 IU/mL (Clinigen, Nürnberg, Germany) and cultured in standard cell culture conditions (37°C, 5% CO2). [00523] On day 5, half of the media is removed and replaced with TCM supplemented with 10% FBS, 0.50 μg/mL Amphotericin B, 20 μg/mL Gentamicin and 6000 IU/mL IL-2. [00524] On day 7, if the cell concentration is > 1.5 x 106 viable cells/mL, the TIL outgrowth culture is diluted with three times the volume to maintain approximately 0.1 x 106 to 2.0 x 106 viable cells/mL. If the cell concentration is ≤ 1.5 x 106 viable cells/mL, half of the media is replaced. In either option, the media is TCM supplemented with 10% FBS, 0.50 μg/mL Amphotericin B, 20 μg/mL Gentamicin and 6000 IU/mL IL-2. [00525] On day 10, if the cell concentration is > 1.5 x 106 viable cells/mL, the TIL outgrowth culture is diluted with three times the volume to maintain approximately 0.1 x 106 to 2.0 x 106 viable cells/mL. If the cell concentration is ≤ 1.5 x 106 viable cells/mL, half of the media is replaced. In either option, the media added is TCM supplemented with 10% FBS, 0.50 μg/mL Amphotericin B, 20 μg/mL Gentamicin and 6000 IU/mL IL-2. [00526] TILs are activated using an anti-CD3 antibody (OKT3) to provide a CD3 specific stimulation when bound to the FC receptor of irradiated feeder cells from allogeneic peripheral blood mononuclear cells (PBMCs). The feeders provide a natural source of additional co- stimulation to support the added anti-CD3 (OKT-3). [00527] On day 12, 1 to 20 x 106 viable T cells from the TIL outgrowth Step 2 are added to 2.0 to 4.0 x 109 viable irradiated feeder cells (Section 8.1.4.4) using approximately 30 ± 10 ng/mL OKT3, approximately 8% Human AB Serum and 3000 ± 1000 IU/mL IL-2. The TIL activation culture is incubated for 6 days at standard cell culture conditions. [00528] On day 18, the activated TILs continue expansion by aseptically adding the activated TIL cell suspension into a bioreactor containing T cell media supplemented with approximately 8% Human AB Serum and 3000 IU/mL IL-2. [00529] On day 19, the TIL expansion is provided a continuous feed of T cell media supplemented with 3000 IU/mL IL-2 until harvest. [00530] TILs are harvested by washing the cells using SEFIATM. The cells are concentrated by centrifugation then washed 2-4 times using phosphate buffered saline (PBS) supplemented with 1% human serum albumin (HSA). The cells are then resuspended in PBS + 1% HSA to approximately 50-60 mL. [00531] The washed and concentrated cells are aseptically transferred into a cryobag and a portion removed for lot release testing and retained samples. To formulate drug product (DP) the TILs are then cooled to 2-8°C and formulated, e.g.1:1 with cryoprotectant containing 16% HSA and 20% DMSO, to achieve a formulated product of ≥ 5 x 109 viable cells suspended in approximately 10% DMSO and 8.5% HSA in PBS. A portion is removed for lot release testing and retained samples. The cryobag is cooled to -80°C. [00532] The following table shows examples of TIL manufacture process variations.
Figure imgf000140_0001
[00533] The following table shows Drug Product Data
Figure imgf000141_0001
[00534] Comparing cryopreserved and fresh cell suspensions, representative yields were consistent as demonstrated by similar drug substance yield, viability, and percent T cells. [00535] Optimization of Cryopreservation - As a surrogate to tumor material, isolated PBMCs were digested using the Tiss-U-Stor process and materials. Commercial cryopreservation agents (CPAs) were evaluated across a range of conditions to determine which reagent maximized post- thaw viability. The post-thaw viabilities of two CPAs, Cryostor10 and Stem Cell Banker DMSO free, were similar. CryoStor based DMSO was then compared with Bloodstor 55-5, a DMSO based cryopreservative, and the higher concentration BloodStor product was selected since it was more concentrated thus allowing for a smaller cryobag. Cryopreservation was then compared following a protocol that either held the material at 4℃ for 10 minutes, then decreased the temperature at a rate of -1℃/min or decreased from 35℃ to -80℃ directly at a rate of -2℃/min. Post-thaw viability was similar between the two cryopreservation protocols used. [00536] During cooling, ice nucleation releases heat. Undercooling, a phenomenon where the released heat appears to warm the solution, is associated with lower post-thaw recoveries. Temperature data was recorded from test articles during cryopreservation using both protocols. Undercooling was observed in both independent runs using the -1℃/min protocol, whereas the - 2℃/min cooling protocol recorded no undercooling event once, and in the second independent run, an undercooling event was observed to release less heat relative to the alternative protocol. [00537] The cryopreserved DP is transferred to vapor phase LN2 for storage and transport at ≤ -130°C. [00538] Sample sterility is tested and retained samples are frozen using a Coolcell® (Biocision, Larkspur, CA) at -80°C then transferred to vapor phase LN2 for storage purposes. [00539] In an aspect, the invention provides methods for evaluating TIL compositions. TIL potency analysis comprises evaluation of analytes characteristic of TIL activation, including but not limited to indicators of mechanism of action. Exemplary non-limiting mechanisms of action include tumor cell killing, cytokine secretion, proliferation, persistence, and properties indicative of the mechanisms. Analysis can comprise enumeration of T cells and target cells, for example by flow cytometry, percent killing which can be observed by fluorescence or luminescence in plate-based or flow cytometry or other methods such as cartridge-based methods, characterization of individual cells to determine expression of markers including but not limited to expression of cytokines, cell surface markers, expression levels of genes that are induced in activated T-cells, including and not limited to reporter molecules engineered to be expressed under activating conditions, or other hallmarks of T cell activation. [00540] Measures of TIL potency include TIL cellular composition and phenotype, such as but not limited to numbers and proportions of CD8+ cells, memory phenotype including without limitation effector memory and central memory, measures of cytotoxicity using various cell lines, cytotoxicity using patient specific tumor, expression of cytokines or a panels of cytokines, and cell proliferation and persistence. [00541] In certain embodiments, there is provided a bioassay for quantification of TIL potency. In certain embodiments, the bioassay comprises multiparameter or polychromatic intracellular flow cytometry. Intracellular flow cytometry is particularly advantageous for assessment of T cell specific parameters on an individual cell basis and ensures accurate determination even in heterogeneous cell populations. Multiparameter flow cytometry permits simultaneous detection or two or more components, which can include two or more cytokines, combined with high throughput. Cartridge-based analytical technologies are also contemplated, such as but not limited to the cartridges manufactured by Chemometec chemometec.com/products/nucleocounter-nc-200-automated-cell-counter/ or Accellix accellix.com/technology/). [00542] Unlike ELISAs and similar methods used on bulk supernatant, intracellular assays described herein are cell and cell type specific. Advantageously, individual cytokine producing cells can be identified and enriched if desired. In certain embodiments, the intracellular methods avoid cytotoxicity and effects of the methods on the assayed cells are reversible. [00543] In certain embodiments, a TIL population is cocultured with cells engineered to activate T cells via CD3, the signaling component of the T-cell receptor (TCR). In certain embodiments, a modified TIL population is cocultured with cells engineered to activate T cells as well as engage and activate a costimulatory receptor. A convenient example of activating cells comprises K562 cells engineered to express a binding protein or antibody or antigen binding fragment thereof that binds to and activates the TCR. In certain embodiments, the antibody comprises OKT3. In certain embodiments, the antigen binding fragment comprises a single- chain variable fragment (scFv) from OKT3. Co-culture of ITIL-168 DP with stimulatory K562- OKT3 cells allows for T cell activation via TCR. In certain embodiments, there is provided a negative control, for example, without limitation, nontransduced clonal K562 cells, K562-NT. The ratio of TILs to activating cells can be adjusted as needed. In certain embodiments, the ration of TILs to activating cells is from 10:1 to 1:10. Non-limiting examples include coculture of TILs with stimulatory K562-OKT3 cells in ratios such as 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, or 1:10. [00544] In certain embodiments, the potency analysis method is used to determine potency of a TIL population cocultured with a “standard” cell type. A non-limiting example is a K562 cell engineered to express a ligand, such as but not limited to an antibody or antigen binding fragment thereof, such as an OKT3 antibody or antigen binding fragment thereof that binds to and activates a T-cell receptor on the TIL. In certain embodiments, the potency analysis method is used to determine potency of a TIL population cocultured with tumor cells or cells engineered to express a tumor associated antigen. In certain embodiments, the potency analysis method is used to determine potency of a TIL population cocultured with tumor cells from the same patient as the source of the TILs. [00545] Potency can be reported as: [00546]
Figure imgf000144_0001
[00547] AVG indicates the average potency determined by assay in triplicate. [00548] Potency may be calculated as the frequency of all viable CD2+ cells that are positive for one or more of CD137, CD107a, TNF-α and IFN-γ, preferably CD107a and IFN-γ. [00549] The potency analysis method can be applied at any stage of TIL manufacture. In certain embodiments, TIL manufacture comprises monitoring potency of the TIL manufacture from one culture step to the next. In certain embodiments, TIL manufacture comprising monitoring TIL potency throughout the TIL manufacture. In some embodiments, TIL manufacture may comprise measuring TIL potency to confirm or adjust the number of cells from a culture step used to seed a subsequent culture step. TIL quality attributes include potency, viability, cell count and purity. In certain embodiments, TIL manufacture comprises measuring the potency of TILs processed from a tumor. In certain embodiments, TIL manufacture comprises measuring the potency of TILs from a pre-REP expansion culture. In certain embodiments, TIL manufacture comprises measuring the potency of TILs during a pre- expansion REP. In certain embodiments, TIL manufacture comprises measuring the potency of TILs at the end of a REP. In certain embodiments, TIL manufacture comprises measuring the potency of TILs at the end of a second REP. In certain embodiments, TIL manufacture comprises measuring TIL potency during REP, for example mid-REP. In certain embodiments, TIL manufacture comprises measuring TIL potency prior to cryopreservation and/or after thawing of a cryopreserved cells. In certain embodiments, TIL manufacture comprises measuring the potency of TIL drug product (TIL DP). The potency testing at any stage of TIL manufacture may further comprise enrichment or isolation of more potent TILs, for example the top 40%, or the top 50%, or the top 60%, or the top 70%, or the top 80%, or the top 90% of the TILs. In certain embodiments, the enrichment or isolation comprises separation of TILs from inhibitory cells. [00550] Non-limiting examples of analytes indicative of TIL activation and potency include IFN-γ, CD107a, CD137 (4-1BB). Other markers indicative or TIL activation or beneficial anti- tumor characteristics include, but are not limited to, IL-1beta, IL-2, IL-4, IL-6, IL-8, IL-10, IL- 12p70, granzyme A/B, perforin, caspase 3 and other chemokine markers. [00551] CD107a (aka lysosomal-associated membrane protein-1 or LAMP-1) is a marker of degranulation of NK cells and CD8+ T-cells. IFN-γ is a pleiotropic cytokine with antiviral, antitumor, and immunomodulatory functions. IFN-γ has been shown to increase the motility of antigen-specific CD8+ T-cells to the antigen-expressing (target) cells and enhance the killing of target cells. IFN-γ concentration in the tumor microenvironments has been linked to better immune checkpoint blockade efficacy. comprises an indicator of T-cell activation. In an embodiment, there is an analysis of IFN-γ and CD107a. CD137 (4-1BB) is a member of the TNFR family and functions as a costimulatory molecule to promote proliferation and survival of activated T cells. Expression of CD137 on T cells is found in T cells that have recently been activated by TCR engagement. TNF is a proinflammatory cytokine produced by activated T cells and indicative of robust antitumor activity. [00552] Potency due to autocrine stimulation of TIL by cytokines or potency due to paracrine stimulation of anti-tumor effects mediated by other cells in the tumor microenvironment is detectable in Applicant’s method, although if there is high background in the T cell +K562 parenteral, Applicants have not yet observed it. Potency markers indicating persistence may be detected in a cell proliferation assay. [00553] In certain embodiments, analytes that distinguish cell subsets are examined. Non limiting examples are CD62L and CD45RO which in different combination can distinguish among effector cells (CD62L-, CD45RO-), effector memory cells (CD62L-, CD45RO+), central memory cells (CD62L+, CD45RO+) and stem cell memory cells (CD62L+, CD45RO-). [00554] Other examples indicative of desirable subsets, activated subsets, cells preferred to be discarded include clearance subsets, such as B cells, monocytes, granulocytes, NK cells, Melanoma tumor cells and other subsets include, but are not limited to, CD3, CD4, CD8, CD95, CCR7 and CD45RO, to distinguish between naïve, T memory stem cells (SCM), effector, effector memory, and central memory subsets. [00555] An assay overview is provided for cryopreserved cells. As described above, the assay is suitable to determine TIL potency at any stage of manufacture, and includes TIL from any process, culture, or expansion step, and TIL fresh or cryopreserved. Cryopreserved cells are thawed typically provided a recovery period before potency testing of about 1-2 hr, 2-4 hr, 4-6 hr., 6-8 hr., 8-10 hr., 10-12 hr, or overnight (up to 24 hr), before testing. After the recovery period, or on day 2 (day 1 for fresh TIL), thawed TIL are then mixed with a population of stimulatory cells (e.g. K562 or other non T cell line engineered with OKT3 scFv fragment) capable of engaging and stimulating the TILs via the TCR. The number of cells post recovery going into the assay, for example transduced and untransduced cells, seeded in the assay may be from about 1 x 105, 2 x 105, 3 x 105, 4 x 105, 5 x 105, 6 x 105, 7 x 105, 8 x 105, 9 x 105, 1 x 106, 2 x 106, 3 x 106, 4 x 106, 5 x 106, 6 x 106, 7 x 106, 8 x 106, 9 x 107, 1 x 107, 2 x 107, 3 x 107, 4 x 107, 5 x 107, 6 x 107, 7 x 107, 8 x 107, 9 x 107, 1 x 108, 2 x 108, 3 x 108, 4 x 108, 5 x 108, 6 x 108, 7 x 108, 8 x 108, 9 x 108, 1 x 109 cells. The mixed cell composition may be incubated for about 8-10 hr., 10-12 hr, 12-14 hr., 14-16 hr., 16-18 hr., 18-20 hr., 20-22 hr., 22-24 hr., 24-26 hr., 26-28 hr., 28-30 hr., 30-32 hr., 32-34 hr. or 34-36 hr. with an inhibitor of protein transport inhibitors (e.g. Brefeldin A and Monensin which may be at a concentration from about 10X, 20X, 30X, 40X, 50X, 60X, 70X, 80X, 90X, 100X, 200X, 300X, 400X, 500X, 600X, 700X, 800X, 900X, 1000X, 200X, 3000X, 4000X, 5000X, 6000X, 7000X, 8000X, 9000X or 10,000X) and optionally one or more reagents to monitor pertinent markers that identify degranulating cells post activation (e.g., anti-CD107a). CD107a may be added to mark T cell degranulation prior to analyzing cell count, viability and/or cell purity, which may be determined by flow cytometry or a cartridge based method. The incubation period may be about 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 13, 13.5, 14, 14.5, 15, 15.5, 16, 16.5, 17, 17.5, 18, 18.5, 19, 19.5, 20, 20.5, 21, 21.5, 22, 22.5, 23, 23.5, 24, 24.5, 25, 25.5, 26, 26.2, 27, 27.5, 28, 28.5, 29, 29.5, 30, 30.5, 31, 31.5, 32, 32.5, 33, 33.5, 34, 34.5, 35, 35.5, or 36 hours After an effective incubation period, the cell culture is treated to distinguish live and dead cells and the cells are permeabilized and fixed. The concentration of the fixative and the time of fixing may be optimized and is within the purview of one of skill in the art. The treatment may be for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 30, 31, 32, 33, 34 or 35 minutes. Permeabilized cells are stained for intracellular and extracellular markers and the markers measured by flow cytometry or a cartridge based method. The antibody cocktail used to stain the cells of potency markers (e.g., CD2, TNFa, IFNg, CD137) can vary across different fluorophores (e.g. PE, PCP-eF710, APC, APC-Cy7, BV711, eFLOUR506, GFP etc.) concentration volume (0.5, 1.0, 1.2, 1.25, 1.3, 1.5, 1.75, 1.8, 1.9, 2.0, 2.5, 3, 3.5, 4 etc.) and incubation time (5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60 mins, etc.). A stain may be utilized to distinguish between live and dead cells. [00556] All patent filings, websites, other publications, accession numbers and the like cited above or below are incorporated by reference in their entirety for all purposes to the same extent as if each individual item were specifically and individually indicated to be so incorporated by reference. If different versions of a sequence are associated with an accession number at different times, the version associated with the accession number at the effective filing date of this application is meant. The effective filing date means the earlier of the actual filing date or filing date of a priority application referring to the accession number if applicable. Likewise, if different versions of a publication, website or the like are published at different times, the version most recently published at the effective filing date of the application is meant unless otherwise indicated. Any feature, step, element, embodiment, or aspect of the invention can be used in combination with any other unless specifically indicated otherwise. Although the present invention has been described in some detail by way of illustration and example for purposes of clarity and understanding, it will be apparent that certain changes and modifications may be practiced within the scope of the appended claims. EXAMPLES Example 1. Identification of agents and methods to increase TIL persistence and functionality Candidates [00557] Genes and small molecules with reported impact on T cell proliferation, persistence, functionality, or differentiation phenotype in healthy donor peripheral blood T cells, viral- specific T cells, tumor infiltrating lymphocytes, CAR- or TCR-modified T cells, and in vitro stem-cell-derived T cells were identified as candidate agents. Genes or small molecule targets enhancing proliferation, persistence, functionality, and delaying differentiation or exhaustion were categorized as gain-of-function (GOF) genes of which up-regulation via transgene or overexpression of constitutively activated mutants is needed. Genes or small molecule targets repressing proliferation, persistence, functionality, and promoting differentiation or exhaustion were categorized as loss-of-function (LOF) genes of which down-regulation via gene knock- down, gene knock-out, or overexpression of dominant negative mutants is needed. Cytokines, growth factors, therapeutic antibodies, and small molecule inhibitors of kinases, cell surface receptors, or epigenetic regulators that are reportedly regulating T cell proliferation, persistence, functionality, or differentiation were also included. Lentiviral transfer plasmids and LVV preps for selected candidates [00558] For GOF genes, protein sequences of the common or most abundant isoform in T cells were obtained from UniProt, reverse translated, and codon optimized before gene synthesis and molecular cloning into pSF.Lenti.MND.P2A.tCD34.KanR lentiviral vector (pIB1123). For loss of function genes (pIB1146-1147), one or several dominant negative mutants were first identified from published literatures. The wild-type and mutant protein sequences were then reverse translated, and codon optimized before gene synthesis and molecular cloning into pSF.Lenti.MND.P2A.tCD34.KanR lentiviral vector (pIB1123). Sanger sequencing was used to verify the sequence of resultant plasmids. [00559] To make LVV preps, 293T cells were co-transfected with the transfer plasmid and three helper plasmids using PEI. Culture supernatant was collected 72 hours after transfection, cleared through 0.45μm filtration, and concentrated using Lenti-X Concentrator. The resultant LVV prep was titrated by transduction in serial dilutions on Jurkat cells followed by flow cytometric analysis of surface tCD34 expression. Master regulators [00560] Single-cell RNA-Seq was performed on TIL product samples from patients in MS Specials study. Unsupervised clustering analysis was performed on the resulting single-cell gene expression profiles which grouped cells into subpopulations with distinct transcriptional signatures. The fractional abundance of each cell subpopulation in each TIL product sample was calculated and correlated to clinical response which identified a list of cell subpopulations being correlated with response. In addition, cell subpopulations were annotated based on prior knowledge which identified several proliferating cell subpopulations and a non-proliferating highly differentiated cell subpopulation. [00561] Gene regulatory network analysis was performed on each single-cell gene expression profile to score the activity level of each transcription factor in each cell. The activity score of each transcription factor is averaged within each cell subpopulation and standardized across cell subpopulations. The top 20 most active transcription factors of each cell subpopulation were identified as the list of key transcription factors responsible for maintaining the transcriptional signature of that cell subpopulation. [00562] A list of candidate genes to upregulate for viTIL is generated by combining the lists of key transcription factors of the cell subpopulations that were positively correlated with response and the proliferating cell subpopulations. A list of candidate genes to downregulate for viTIL is generated by combining the lists of key transcription factors of the cell subpopulations that were negatively correlated with response and the non-proliferating highly differentiated cell subpopulations. Repeated stimulation assay on healthy donor T cells to model T cell differentiation in vitro [00563] Peripheral blood mononuclear cells (PBMCs) and isolated T cells from two healthy donors were thawed and stimulated with various stimulation methods under different schemes. For stimulation with OKT3/CD28, 6-well cell culture plates were coated with 1.23 μg/mL GMP- or RUO-grade OKT3 in 1X HBSS at 4°C overnight and washed with 1X PBS before use. Cells were seeded on OKT3-coated plates in the presence of 1 μg/mL soluble anti-CD28 antibody in T cell medium with 200 IU/mL IL-2 (complete TCM). For stimulation with Dynabeads, cells were seeded with Dynabeads Human T-Activator CD3/CD28 at 1:1 ratio in complete TCM. For stimulation with TransAct, cells were seeded in the presence of 10 μL human T Cell TransAct per 1x106 cells in complete TCM. [00564] In “Repeat Stim” groups, PBMCs or isolated T cells were thawed and seeded at 1x106 cells with stimulation. Cells were then counted, washed, diluted to 1x106 cells/mL in fresh complete TCM, and repeatedly stimulated every 3 days for a total of 10 rounds of stimulation over 34 days. In “Round 1 Rest” groups, PBMCs or isolated T cells were thawed and seeded at 1x106 cells with 1 round of stimulation. Cells were then counted, washed, and diluted to 1x106 cells/mL in fresh complete TCM every 3 days without further stimulation for a total of 34 days. In “Round 2 Rest” groups, PBMCs or isolated T cells were thawed and seeded at 1x106 cells with 2 rounds of stimulation. Cells were then counted, washed, and diluted to 1x106 cells/mL in fresh complete TCM every 3 days without further stimulation for a total of 34 days. [00565] By the end of each round of stimulation, a fraction of cells from all groups were collected for staining and flow cytometric analysis of surface markers CD3, CD4, CD8, CD69, CD25, 4-1BB, PD-1, TIM-3, LAG-3, CTLA-4, CD39, CD103, CD45RA, CCR7, CD45RO, CD62L, CD27, CD28, OX40, and CD127. Total viable cell count was used to select the optimal repeated stimulation method and scheme. Screening of candidates in healthy donor T cell serial stimulation assay [00566] Peripheral blood T cells isolated from two healthy donors were thawed and stimulated with Dynabeads Human T-Activator CD3/CD28 at 1:1 ratio in complete TCM for 3 days. On day 1 after stimulation, T cells were transduced with candidate LVV at MOI of 5. From day 3, T cells in “Repeat Stim” group were counted, washed, diluted to 1x106 cells/mL in fresh complete TCM, and repeatedly stimulated with Dynabeads at 1:1 ratio every 3 days for a total of 10 rounds of stimulation over 34 days. T cells in “Round 1 Rest” group were counted, washed, and diluted to 1x106 cells/mL in fresh complete TCM every 3 days without further stimulation for a total of 34 days as control. [00567] By the end of each round of stimulation, a fraction of T cells from both groups were collected for staining and flow cytometric analysis of surface tCD34 as a surrogate of transgene expression, along with surface markers CD3, CD4, CD8, CD69, CD25, 4-1BB, PD-1, TIM-3, LAG-3, CTLA-4, CD39, CD103, CD45RA, CCR7, CD45RO, CD62L, CD27, CD28, OX40, and CD127. Total viable cell count, tCD34+%, and tCD34+ viable cell count were used to select positive hits. Surface expression of 4-1BB, OX40, PD-1, TIM-3, LAG-3, and CTLA-4 in CD4+ and CD8+ T cell populations were compared between positive hits in “Repeat Stim” group. Phenotypic subsets defined by CD45RA and CCR7 were also compared between positive hits in “Repeat Stim” group. Screening of selected candidates during ex vivo TIL expansion [00568] Tumor digest from ovarian cancer and renal cell carcinoma were thawed and plated in 12-well plates at 1x106 cells/mL in complete TCM with 3000 IU/mL IL-2, incubated for 2 days. On day 3 and 4, cells were transduced with selected short-listed candidate LVV at MOI=5. On day 3, 7, and 11, cells were counted, and fresh IL-2 was added to the culture at 3000 IU/mL. On day 13, cells were harvested, counted, and 0.1x106 cells were placed into REP with irradiated PBMC at 1:200 ratio in complete TCM with 3000 IU/mL IL-2 in 24-well G-Rex plates. On day 19, 21, and 23, cells were counted, diluted, and split into multiple wells on 24-well G-Rex plates while fresh IL-3 was added to the culture at 3000 IU/mL. On day 25, cells were harvested, counted, and cryopreserved in CryoStor CS10. [00569] On day 1, day 13, and day 25, a fraction of cells from all groups were collected for staining and flow cytometric analysis of surface tCD34 as a surrogate of transgene expression, along with surface markers of CD3, CD4, CD8, CD69, CD25, 4-1BB, PD-1, TIM-3, LAG-3, CTLA-4, CD39, CD103, CD45RA, CCR7, CD45RO, CD62L, CD27, CD28, OX40, and CD127. Total viable cell count, total viable cell fold expansion, and CD3+ cell fold expansion were analyzed. tCD34% and tCD34+ viable cell count were used to select positive hits. Relative abundance of CD4+ and CD8+ T cells, along with surface expression of PD-1, TIM-3, LAG-3, CTLA-4, CD127, and CD27 in CD4+ and CD8+ T cell populations, were compared between positive hits. Phenotypic subsets defined by CD45RA and CCR7 were also compared between positive hits. Example 2. Agent selection [00570] Curated redirection agents. Genes or small molecules that have shown evidence of promoting T cell proliferation, enhancing T cell persistence, boosting T cell functionality, or maintaining a less differentiated phenotype in unmodified or CAR-/TCR-modified T cells or TILs were selected from published literature as redirection agents. Based on their known impact on T cells, genes enhancing proliferation, persistence, functionality, or delaying differentiation are categorized into gain of function (GOF) group, while genes repressing proliferation, persistence, functionality or promoting differentiation are categorized into loss of function (LOF) groups. Small molecules with known functions on T cells were also included. The LOF group included Fas/FasL and TGFβR. [00571] Lentiviral transfer plasmids (Table 1) were designed for selected agents described above. For gain of function genes, protein sequences of the common or most abundant isoform in T cells were obtained from UniProt, reverse translated, and codon optimized before gene synthesis and molecular cloning into pSF.Lenti.MND.P2A.tCD34.KanR lentiviral vector (pIB1123). For loss of function genes (pIB1146-1147), one or several dominant negative mutants were identified from published literatures. The wild-type and mutant protein sequences were then reverse translated, and codon optimized before gene synthesis and molecular cloning into pSF.Lenti.MND.P2A.tCD34.KanR lentiviral vector (pIB1123). pIB1123 was used as an empty vector control.
Figure imgf000152_0001
[00572] Master regulators. A single-cell RNA-seq dataset was used to bioinformatically identify candidates. Predicted master regulators within the cell clusters correlated with favorable clinical response. These included IRF7 and POLR3A (LOF group). See Figure 24. [00573] Solid tumors are characterized by marked clonal heterogeneity with a variety of antigens, including neoantigens, which often differ between patients and cancer types.1 Melanoma has a high mutational burden and marked clonal heterogeneity both within individual patients and between patients.1,2 Despite treatment advances with targeted therapies and checkpoint inhibitors, most patients with advanced melanoma ultimately relapse and have limited treatment options,3 highlighting an unmet medical need. Autologous tumor-infiltrating lymphocyte (TIL) products are comprised of an unrestricted T-cell receptor (TCR) repertoire and can recognize a broad set of tumor-associated antigens, including neoantigens specific to each patient’s tumor.4 A retrospective analysis of a single-center, compassionate use clinical series of TILs for the treatment of advanced cutaneous melanoma in 21 patients demonstrated high clinical response rates, with a 67% overall response rate and 19% complete response rate, and a safety profile consistent with lymphodepletion and high-dose interleukin (IL)-2.5 There are 106 expected TCR-β clonotypes, (most of which are not well characterized by function or previously annotated) and of these, a small subset of clones from TILs will demonstrate anti-tumor activity.6,7 Furthermore, predictive models for anti-tumor reactivity have limited positive and negative predictive power.8 For these reasons, an unselected approach maximizes the potential for the limited number of clones with anti-tumor activity to reach the final TIL product. [00574] TIL therapy infusion product composition, TCR repertoire, and mediators of cell-cell interaction were characterized in a translational subanalysis of the compassionate use clinical series. Patients with histologically confirmed malignant melanoma and no standard-of-care treatment options underwent resection of ≥1 cm3 of tumor tissue for TIL production.5 Patients received lymphodepleting chemotherapy (cyclophosphamide 60 mg/kg/d ×2 days, fludarabine 25 mg/m2/d ×5 days) followed by TIL infusion and post-TIL short course of high-dose IL-2 (600,000-720,000 IU/kg) on a compassionate use basis.5 Efficacy was assessed locally through standard disease assessment imaging and retrospectively analyzed per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 when feasible.5 TCR repertoire clonality and diversity of TIL products were assessed using multiple metrics, including Gini coefficient, using the Immunarch package.9 TIL products were assessed using RNA-based bulk TCR sequencing and paired single-cell RNA and TCR sequencing techniques. Putative antigen/tumor-reactive clones were inferred using GLIPH2 (grouping of lymphocyte interactions by paratope hotspots 2) algorithm10 and publicly available TCR annotation databases including VDJdb public database.11 Unsupervised clustering of cells and differential gene expression analysis was performed using the Seurat package.12 Gene set over-representation analysis was performed using the clusterProfiler package.13 Gene regulatory network analysis was performed using the SCENIC package.14 Cell-cell interaction analysis was performed using the CellChat package.15 Descriptive statistical testing was performed using Wilcoxon test; P values were not adjusted for multiplicity. [00575] As of December 31, 2019, 21 patients with advanced cutaneous melanoma underwent treatment. Of 21 patients, 20 had assessable products for RNA-based bulk TCR sequencing, and 18 had assessable products for paired single-cell RNA and TCR sequencing. Technical replicates showed a high Morisita index, demonstrating the reproducibility of the TCR testing method.16 A low Morisita index was observed across patients, suggesting a minimal overlap in TCR repertoire and distinct TCR β-chain repertoires between TIL samples.16 [00576] Single-cell RNA sequencing analysis of TIL product samples identified several T-cell subpopulations with distinct gene expression profiles. See Figures 22A-22B. Multiple subpopulations were previously undescribed in TIL product samples. Certain T-cell populations were found at frequencies that differed between products administered to patients who developed responses compared to those who did not achieve a response. See Figures 23A-23C. The frequency of C7 (MX1+OAS1+; Figure 23A), C9 (BBC3+CHAC1+; Figure 23B), or C7 and C9 (Figure 23C) T-cell subpopulations in TIL product samples differed between responders and non-responder. Low abundance of C7 TIL or C9 TIL subpopulations was associated with response (Figures 23A and 23B). Low abundance of the combined C7 and C9 TIL subpopulations was also associated with response (Figure 23C). References [00577] 1. Alexandrov LB, et al. Nature.2013;500(7463):415-421. [00578] 2. Grzywa TM, et al. Transl Oncol.2017;10(6):956-975. [00579] 3. Gide TN, et al. Clin Cancer Res.2018;24(6):1260-1270. [00580] 4. Rohaan MW, et al. J Immunother Cancer.2018;6(1):102. [00581] 5. Hawkins RE, et al. Cancer Res.2021;81(13 suppl):LB150. [00582] 6. Freeman JD, et al. Genome Res.2009;19(10):1817-24. [00583] 7. Mora T, et al. Curr Opin Syst Biol.2019;18:104-110 [00584] 8. Lam H, et al. Cancer Discov.2021;11(3):696-713. [00585] 9. Nazarov V, et al. Immunomind/immunarch: 0.6.5: Basic single-cell support (0.6.5). Zenodo.2020. Available at: https://doi.org/10.5281/zenodo.3893991. Accessed March 22, 2022. [00586] 10. Huang H, et al. Nat Biotechnol.2020;38(10):1194-1202. [00587] 11. Shugay M, et al. Nucleic Acids Res.2018;46(D1):D419-D427. [00588] 12. Hao Y, et al. Cell.2021;184(13):3573-3587. [00589] 13. Yu G, et al. OMICS.2012;16(5):284-7. [00590] 14. Aibar S, et al. Nat Methods.2017;14(11):1083-1086. [00591] 15. Jin S, et al. Nat Commun.2021;12(1):1088. [00592] 16. Yang H, et al. Front Oncol.2021;11:537735 [00593] Cryopreserved TIL product samples were thawed and rested overnight in RPMI culture media supplemented with 10% FBS, 50 uM beta-Mercaptoethanol, 100 units/mL penicillin, 100 ug/mL streptomycin, and 1000 IU/mL IL-2, followed by a Percoll purification step to remove debris and dead cells. The purified cells were then loaded to the Chromium controller7 for single-cell sequencing library preparation. Vendor provided reagents and standard protocols were used to generate paired single-cell RNA and TCR sequencing libraries for each TIL product sample. Final libraries were sequenced using Illumina’s NovaSeq sequencer at Genewiz8. [00594] Raw FASTQ files were processed using the Cell Ranger software7 with default parameters. The resulting counts matrix were then analyzed using the Seurat9 R package. The counts matrix was first filtered based on the total number of genes and the fraction of mitochondria genes detected in each cell. Cells with a fraction of mitochondria gene detected being greater than 10% or with a total number of genes detected being less than 800 were excluded from further analysis. The top 2000 highly variable genes in each TIL product sample were then identified and used as the anchor genes for sample integration to remove any technical batch effect. Cell cycle scores of each cell were calculated and regressed out from the integrated counts matrix, followed by unsupervised clustering which partitions all cells into several cell subpopulations with distinct transcriptional signatures. [00595] Differential gene expression analyses were performed between each cell subpopulation and the rest of cells to identify a list of upregulated and downregulated genes associated with each cell subpopulation. Gene set overrepresentation analysis (gene ontology analysis) was performed using the clusterProfiler10 R package on the up- and down- regulated gene lists respectively to identify biological processes associated with each cell subpopulation. See Figure 24. Within each cell subpopulation, differential gene expression analysis and gene set overrepresentation analysis were performed between cells from responders and cells from non- responders. The fractional abundance of each cell subpopulation in each TIL product sample was calculated and correlated to clinical response. [00596] Gene regulatory network analysis (to identify predicted master regulators associated with each cell cluster) was performed using the SCENIC11 R package on each single-cell gene expression profile to score the activity level of each transcription factor in each cell. The activity score of each transcription factor is averaged within each cell subpopulation and standardized across cell subpopulations. The top 20 most active transcription factors of each cell subpopulation were identified as the list of key transcription factors responsible for maintaining the transcriptional signature of that cell subpopulation. See Figure 24. References [00597] 7. 10xgenomics.com/ note: The Chromium machine and reagents from 10X genomics was used to perform the library preparation step of paired single-cell RNA and TCR sequencing. The Cell Ranger software from 10X genomics was used to process raw FASTQ files. [00598] 8. genewiz.com/en note: Genewiz is the vendor that performed the sequencing of paired single-cell RNA and TCR sequencing library [00599] 9. satijalab.org/seurat/ note: Seurat is the R package that was used to perform the unsupervised clustering of single cell RNA sequencing data and the differential gene expression analysis between cell populations [00600] 10. bioconductor.org/packages/release/bioc/html/clusterProfiler.html note: clusterProfiler is the R package that was used to perform gene set over-representation analysis [00601] 11. Aibar et al, Nature Methods 14, 1083-1086 (2017) Example 3. GOF and LOF candidate in HD PBMCs and T cells [00602] PBMC and isolated T cells from two donors were thawed and stimulated according to different schedules (Figure 1) and various reagents. After the first round of stimulation (3 days), cells in “Repeat Stim” group were counted, washed, diluted to 1x106 cells/mL, and repeatedly stimulated every 3 days for a total of 10 rounds in 34 days. Cells in “Round 1 Rest” group were counted, washed, and diluted to 1x106 cells/mL every 3 days without further stimulation. Cells in “Round 2 Stim” group were counted, washed, and diluted to 1x106 cells/mL every 3 days and received one 1 more round of stimulation after the initial round. On each day of dilution and final harvest, a fraction of the cells in each group was stained and analyzed by flow cytometry for surface markers of activation, exhaustion, and differentiation phenotypes. [00603] Cell expansion curves for the three expansion schedules and stimulating reagent combinations are shown for PBMCs (Figures 2A-2F) and donor T cells (Figures 3A-3F). Stimulating reagents were OKT3+CD28 (GMP) antibodies; OKT3+CD28 (RUO) antibodies; OKT3+CD28 -> OKT3; OKT3 -> OKT3; Dynabeads Human T-Activator CD3/CD28 (DB); and TransAct (TA). GMP = a Good Manufacturing Practices-grade soluble CD28 antibody for ex vivo T cell activation; RUO = a Research Use Only-grade CD28 antibody for ex vivo T cell activation. [00604] T cells were then stimulated and LVV-transfected according to the schedule depicted in Figure 4. Isolated T cells from two healthy donors were thawed and stimulated using Dynabeads for 3 days. On day 1, T cells were transduced with candidate LVVs at MOI=5. After the first round of stimulation, T cells in “Repeat Stim” group were counted, washed, diluted to 1x106 cells/mL, and repeatedly stimulated every 3 days for a total of 10 rounds in 34 days. T cells in “Round 1 Rest” group were counted, washed, and diluted to 1x106 cells/mL every 3 days without stimulation. On each day of dilution and final harvest, a fraction of T cells was stained and analyzed by flow cytometry for surface tCD34 expression along with markers of activation, exhaustion, and differentiation phenotypes. [00605] Expansion curves were determined under repeat stimulation conditions for loss-of- function (LOF) clones pIB1146 - pBI1148 (Figure 5). pIB1123 is an empty vector control. Candidates identified as enabling enhanced proliferation were pIB1146 – dnFas (mutDD) and pIB1147 – dnFas (delDD). [00606] Expansion curves were determined under stimulation with rest conditions for the same loss-of-function (LOF) clones pIB1146 - pBI1148 (Figure 6). pIB1123 is an empty vector control. Candidates identified as enabling enhanced proliferation included pIB1146 – dnFas (mutDD). [00607] Expansion was specifically measured in the tCD34+ transfected TIL subpopulation under repeated stimulation. tCD34+ cells were enriched in T cells transduced with pIB1146 – dnFas (mutDD). Figure 7A shows percent of tCD34+ cells measured by flow cytometry. Figure 7B shows viable tCD34+ cell count from total viable cells. [00608] T cell activation / exhaustion markers were measured in CD4+ and CD8+ populations of tCD34+ T cells (Figures 8A-8F). T cell exhaustion markers were also measured in the CD4+ and CD8+ populations (Figures 9A-9F). Relative abundance of Tn/Tscm (CCR7+CD45RA+), Tcm (CCR7+CD45RA-), Tem (CCR7-CD45RA-), and Temra (CCR7-CD45RA+) populations were plotted over time (Figures 10A-10B). Example 4. GOF and LOF candidates in HD PBMCs and T cells [00609] Selected GOF and LOF candidates were expressed by LVV transfection in donor TILs. TILs were thawed and expanded ex vivo using a standard TIL manufacture protocol of 12- day outgrowth followed by 12-day REP (Figure 11). Cells were transfected at MOI=5 on day 3 and on day 4. On days 1, 13, and 25, total viable cells were counted, and a fraction of cells was stained and analyzed by flow cytometry for surface tCD34 expression along with markers of activation, exhaustion, and differentiation phenotypes. Figures 13A-13D show fold expansion in outgrown (Figure 13A) and REP (Figure 13B) and CD3+ fold expansion in outgrowth (Figure 13C) and REP (Figure 13D). [00610] Enrichment of transduced populations in TILs from renal cell carcinoma during REP was determined. The proportion of LVV transfected tCD34+ cells during REP was compared on day 13 and day 25 and compared to an empty vector (EV) control which transfected only tCD34+ (Figure 14A). Comparative enrichment (Figure 14B) and fold change (Figure 14C) of tCD34+ cells was determined for candidate transduced cells. [00611] The relative abundance of CD4+ and CD8+ subpopulations after outgrowth (Figure 15A) and after REP (Figure 15B) was compared among candidate transfected TILs. Overall, the proportion of CD8+ TILs expressing PD1 following REP was reduced (Figure 16B). The proportion of CD4+ TILs expressing TIM3 was reduced (Figure 17A). The proportion of CD4+ TILs and CD8+ TILs expressing LAG3 was reduced (Figures 18A-18B). Figures 19A-19B show the proportions of CD4+ (Figure 19A) or CD8+ (Figure 19B) TILs expressing CD127 at the end of outgrowth and REP. Figures 20A-20B show the proportion of CD4+ (Figure 20A) or CD8+ (Figure 20B) TILs expressing CD27 at the end of outgrowth and REP. [00612] Proportions of phenotypic subsets CCR7+CD45RA+ (naïve (Tn) / memory stem cell (Tscm)), CCR7+CD45RA- (central memory (Tcm)), CCR7-CD45RA- (effector memory (Tem)), and CCR7-CD45RA+ (effector (Temra) were compared. Figures 21A-21D show the proportions in CD4+ (Figures 21A, 21B) and CD8+ cells (Figures 21C, 21D) at the end of outgrowth (day 13, Figures 21A, 21C) and REP (day 25, Figures 21B, 21D).

Claims

We claim: 1. A method for preparing a therapeutic population of tumor infiltrating lymphocytes (TILs), comprising treating a first population of TILs with one or more compounds to improve T-cell fitness, optionally wherein the method comprises treating the first population of TILs with the one or more compounds multiple times.
2. The method of claim 1, wherein the one or more compounds comprise on or more or all of the following: (i) a FAS/FASLG inhibitory agent; (ii) a TGFβ/TGFβR1 inhibitory agent; (iii) an IRF7 inhibitory agent; and (iv) a POLR3A inhibitory agent.
3. The method of claim 1 or 2, wherein the treating decreases expression or activity of FAS or FASLG.
4. The method of any one of claims 1-3, wherein the treating transiently decreases expression or activity of FAS or FASLG.
5. The method of any one of claims 1-3, wherein the treating permanently decreases expression or activity of FAS or FASLG.
6. The method of any one of claims 1-5, wherein the one or more compounds comprise a DNA encoding a dominant negative FAS mutant operably linked to a promoter active in the TILs.
7. The method of claim 6, wherein the DNA encoding the dominant negative FAS mutant is in a viral vector.
8. The method of claim 7, wherein the viral vector is a lentiviral vector.
9. The method of any one of claims 1-4, wherein the one or more compounds comprise a messenger RNA encoding a dominant negative FAS mutant.
10. The method of any one of claims 6-9, wherein the dominant negative FAS mutant comprises a mutated FADD binding site, optionally wherein the dominant negative FAS mutant is FAS_D244V.
11. The method of any one of claim 6-9, wherein the dominant negative FAS mutant comprises a deleted DD domain, optionally wherein the dominant negative FAS mutant is FAS_del230-314.
12. The method of any one of claims 1-4, wherein the one or more compounds comprise an anti-FAS or anti-FASLG antigen-binding protein.
13. The method of claim 12, wherein the antigen-binding protein comprises an antibody.
14. The method of any one of claims 1-4, wherein the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to a FAS or FASLG messenger RNA.
15. The method of claim 14, wherein the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
16. The method of any one of claims 1-5, wherein the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding FAS or FASLG.
17. The method of claim 16, wherein the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FAS or FASLG.
18. The method of claim 17, wherein the Cas protein is a Cas9 protein or a Cas12a protein.
19. The method of any one of claims 1-4, wherein the one or more compounds comprise comprises a small molecule FAS/FASLG inhibitor.
20. The method of any one of claims 1-19, wherein the treating decreases expression or activity of TGFβ1 or TGFβR1.
21. The method of any one of claims 1-20, wherein the treating transiently decreases expression or activity of TGFβ1 or TGFβR1.
22. The method of any one of claims 1-20, wherein the treating permanently decreases expression or activity of TGFβ1 or TGFβR1.
23. The method of any one of claims 1-22, wherein the one or more compounds comprise comprises a DNA encoding a dominant negative TGFβR1 mutant operably linked to a promoter active in the TILs.
24. The method of claim 23, wherein the DNA encoding the dominant negative TGFβR1 mutant is in a viral vector.
25. The method of claim 24, wherein the viral vector is a lentiviral vector.
26. The method of any one of claims 1-21, wherein the one or more compounds comprise a messenger RNA encoding a dominant negative TGFβR1 mutant.
27. The method of any one of claims 1-21, wherein the one or more compounds comprise an anti-TGFβR1 or an anti-TGFβ1 antigen-binding protein.
28. The method of claim 27, wherein the antigen-binding protein comprises an antibody.
29. The method of any one of claims 1-21, wherein the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to a TGFβR1 or TGFβ1 messenger RNA.
30. The method of claim 29, wherein the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
31. The method of any one of claims 1-22, wherein the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding TGFβR1 or TGFβ1.
32. The method of claim 31, wherein the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding TGFβR1 or TGFβ1.
33. The method of claim 32, wherein the Cas protein is a Cas9 protein or a Cas12a protein.
34. The method of any one of claims 1-21, wherein the one or more compounds comprise a small molecule TGFβR1 inhibitor, optionally wherein the small molecule TGFβR1 inhibitor is SB431542.
35. The method of any one of claims 1-34, wherein the treating decreases expression or activity of IRF7.
36. The method of any one of claims 1-35, wherein the treating transiently decreases expression or activity of IRF7.
37. The method of any one of claims 1-35, wherein the treating permanently decreases expression or activity of IRF7.
38. The method of any one of claims 1-37, wherein the one or more compounds comprise a DNA encoding a dominant negative IRF7 mutant operably linked to a promoter active in the TILs.
39. The method of claim 38, wherein the DNA encoding the dominant negative IRF7 mutant is in a viral vector.
40. The method of claim 39, wherein the viral vector is a lentiviral vector.
41. The method of any one of claims 1-36, wherein the one or more compounds comprise a messenger RNA encoding a dominant negative IRF7 mutant.
42. The method of any one of claims 1-36, wherein the one or more compounds comprise an anti-IRF7 antigen-binding protein.
43. The method of claim 42, wherein the antigen-binding protein comprises an antibody.
44. The method of any one of claims 1-36, wherein the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF7 messenger RNA.
45. The method of claim 44, wherein the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
46. The method of any one of claims 1-37, wherein the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF7.
47. The method of claim 46, wherein the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF7.
48. The method of claim 47, wherein the Cas protein is a Cas9 protein or a
Cas 12a protein.
49. The method of any one of claims 1-36, wherein the one or more compounds comprise a small molecule IRF7 inhibitor.
50. The method of any one of claims 1-49, wherein the treating decreases expression or activity of POLR3A.
51. The method of any one of claims 1-50, wherein the treating transiently decreases expression or activity of POLR3A.
52. The method of any one of claims 1-50, wherein the treating permanently decreases expression or activity of POLR3A.
53. The method of any one of claims 1-52, wherein the one or more compounds comprise a DNA encoding a dominant negative POLR3A mutant operably linked to a promoter active in the TILs.
54. The method of claim 53, wherein the DNA encoding the dominant negative P0LR3 A mutant is in a viral vector.
55. The method of claim 54, wherein the viral vector is a lentiviral vector.
56. The method of any one of claims 1-51, wherein the one or more compounds comprise a messenger RNA encoding a dominant negative POLR3 A mutant.
57. The method of any one of claims 1-51, wherein the one or more compounds comprise an anti-POLR3A antigen-binding protein.
58. The method of claim 57, wherein the antigen-binding protein comprises an antibody.
59. The method of any one of claims 1-51, wherein the one or more compounds comprise an inhibitory RNA comprising a region of complementarity to a POLR3 A messenger RNA.
60. The method of claim 59, wherein the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
61. The method of any one of claims 1-52, wherein the one or more compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding P0LR3A.
62. The method of claim 61, wherein the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding P0LR3 A.
63. The method of claim 62, wherein the Cas protein is a Cas9 protein or a
Cas 12a protein.
64. The method of any one of claims 1-51, wherein the one or more compounds comprise a small molecule POLR3A inhibitor.
65. The method of any one of claims 1-64, wherein the TILs originate from a subject.
66. The method of any one of claims 1-65, wherein the TILs are from a tumor biopsy, a lymph node, or ascites.
67. The method of claim 66, wherein the tumor is from a bladder cancer, a breast cancer, a cancer caused by human papilloma virus, a cervical cancer, a head and neck cancer, a lung cancer, a melanoma, an ovarian cancer, a non-small-cell lung cancer (NSCLC), a renal cancer, or a renal cell carcinoma.
68. The method of claim 66, wherein the tumor biopsy is from a melanoma.
69. The method of any one of claims 1-68, wherein the method further comprises: (i) obtaining a refined tumor product by cryopreserving a resected tumor and disaggregating the cryopreserved tumor, disaggregating a resected tumor and cryopreserving the disaggregated tumor, cryopreserving a resected tumor and processing the tumor into multiple tumor fragments, or processing a resected tumor into multiple tumor fragments and cryopreserving the tumor fragments; and (ii) performing a first expansion by culturing the refined resected tumor product in a cell culture medium comprising IL-2 to produce the first population of TILs, optionally wherein the first population of TILs is treated with the one or more compounds during or subsequent to the first expansion.
70. The method of claim 69, wherein the cryopreserving comprises: (1) cooling under conditions whereby heat release to, into, around or in an environment including cells, as media crystalizes, is minimized or avoided; (2) continuous cooling, from disaggregation temperature to about -80°C; (3) continuous cooling at a rate of about -2°C / min; (4) continuous cooling, from disaggregation temperature to about -80°C, at a rate of about -2°C / min; or (5) continuous cooling, from disaggregation temperature to about -80°C, or from disaggregation temperature to -80°C at a rate of about -2°C / min, wherein disaggregation temperature comprises a normal body temperature for an animal from which the tumor was resected, or room temperature or 20°C or 25°C , or normal human body temperature approximately 35°C or 36°C or 36.1°C to approximately 37°C or 37.1°C or 37.2°C or 37.3°C or below about 38.3°C.
71. The method of claim 69 or 70, wherein the disaggregating comprises physical disaggregation, enzymatic disaggregation, or physical and enzymatic disaggregation.
72. The method of any one of claims 69-71, wherein a single cell suspension is obtained from the refined resected tumor product and used in step (ii), or wherein the refined resected tumor product from step (i) comprises a single cell suspension.
73. The method of any one of claims 69-72, wherein the first expansion in step (ii) is performed for about two weeks.
74. The method of any one of claims 69-73, wherein the culturing in step (ii) includes adding IL-7, IL-12, IL-15, IL-18, IL-21, or a combination thereof.
75. The method of any one of claims 69-74, further comprising: (iii) performing a second expansion by culturing the first population of TILs in a cell culture medium comprising IL-2, optionally wherein the first population of TILs is treated with the one or more compounds prior to, during, or subsequent to the second expansion.
76. The method of any one of claims 69-75, wherein the expanding in step (iii) comprises culturing the first population of TILs with IL-2, OKT-3, and antigen presenting cells (APCs).
77. The method of claim 75 or 76, wherein the expanding in step (iii) is performed for about two weeks.
78. The method of any one of claims 75-77, wherein the culturing in step (iii) includes adding IL-7, IL-12, IL-15, IL-18, IL-21, or a combination thereof.
79. The method of any one of claims 1-78, further comprising harvesting and/or cryopreserving the therapeutic population of TILs.
80. An isolated therapeutic population of TILs obtained by the method of any one of claims 1-79.
81. An isolated therapeutic population of TILs comprising one or more exogenous compounds to improve T-cell fitness.
82. The isolated therapeutic population of claim 81, wherein the one or more exogenous compounds comprise one or more or all of the following: (i) a FAS/FASLG inhibitory agent; (ii) a TGFβ/TGFβR1 inhibitory agent; (iii) an IRF7 inhibitory agent; and (iv) a POLR3A inhibitory agent.
83. The isolated therapeutic population of claim 81 or 82, wherein the one or more exogenous compounds decrease expression or activity of FAS or FASLR.
84. The isolated therapeutic population of any one of claims 81-83, wherein the one or more exogenous compounds transiently decrease expression or activity of FAS or FASLR.
85. The isolated therapeutic population of any one of claims 81-83, wherein the one or more exogenous compounds permanently decrease expression or activity of FAS or FASLR.
86. The isolated therapeutic population of any one of claims 81-85, wherein the one or more exogenous compounds comprise a DNA encoding a dominant negative FAS mutant operably linked to a promoter active in the TILs.
87. The isolated therapeutic population of claim 86, wherein the DNA encoding the dominant negative Fas mutant is in a viral vector.
88. The isolated therapeutic population of claim 87, wherein the viral vector is a lentiviral vector.
89. The isolated therapeutic population of any one of claims 81-84, wherein the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative FAS mutant.
90. The isolated therapeutic population of any one of claims 86-89, wherein the dominant negative FAS mutant comprises a mutated FADD binding site, optionally wherein the dominant negative FAS mutant is Fas_D244V.
91. The isolated therapeutic population of any one of claim 86-89, wherein the dominant negative FAS mutant comprises a deleted DD domain, optionally wherein the dominant negative FAS mutant is Fas_del230-314.
92. The isolated therapeutic population of any one of claims 81-84, wherein the one or more exogenous compounds comprise an anti-FAS or anti-FASLG antigen-binding protein.
93. The isolated therapeutic population of claim 92, wherein the antigen- binding protein comprises an antibody.
94. The isolated therapeutic population of any one of claims 81-84, wherein the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to a FAS or FASLG messenger RNA.
95. The isolated therapeutic population of claim 94, wherein the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
96. The isolated therapeutic population of any one of claims 81-85, wherein the one or more exogenous compounds comprise comprises a nuclease agent targeting a nuclease target site in a gene encoding FAS or FASLG.
97. The isolated therapeutic population of claim 96, wherein the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding FAS or FASLG.
98. The isolated therapeutic population of claim 97, wherein the Cas protein is a Cas9 protein or a Cas12a protein.
99. The isolated therapeutic population of any one of claims 81-84, wherein the one or more exogenous compounds comprise a small molecule FAS/FASLG inhibitor.
100. The isolated therapeutic population of any one of claims 81-99, wherein the one or more exogenous compounds decrease expression or activity of TGFβ1 or TGFβR1.
101. The isolated therapeutic population of any one of claims 81-100, wherein the one or more exogenous compounds transiently decrease expression or activity of TGFβ1 or TGFβR1.
102. The isolated therapeutic population of any one of claims 81-100, wherein the one or more exogenous compounds permanently decrease expression or activity of TGFβ1 or TGFβR1.
103. The isolated therapeutic population of any one of claims 81-102, wherein the one or more exogenous compounds comprise a DNA encoding a dominant negative TGFβR1 mutant operably linked to a promoter active in the TILs.
104. The isolated therapeutic population of claim 103, wherein the DNA encoding the dominant negative TGFβR1 mutant is in a viral vector.
105. The isolated therapeutic population of claim 104, wherein the viral vector is a lentiviral vector.
106. The isolated therapeutic population of any one of claims 81-101, wherein the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative TGFβR1 mutant.
107. The isolated therapeutic population of any one of claims 81-101, wherein the one or more exogenous compounds comprise an anti-TGFβR1 or anti-TGFβ1 antigen- binding protein.
108. The isolated therapeutic population of claim 107, wherein the antigen- binding protein comprises an antibody.
109. The isolated therapeutic population of any one of claims 81-101, wherein the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to a TGFβR1 or TGFβ1 messenger RNA.
110. The isolated therapeutic population of claim 109, wherein the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
111. The isolated therapeutic population of any one of claims 81-102, wherein the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding TGFβR1 or TGFβ1.
112. The isolated therapeutic population of claim 111, wherein the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding TGFβR1 or TGFβ1.
113. The isolated therapeutic population of claim 112, wherein the Cas protein is a Cas9 protein or a Cas12a protein.
114. The isolated therapeutic population of any one of claims 81-101, wherein the one or more exogenous compounds comprise a small molecule TGFβR1 inhibitor, optionally wherein the small molecule TGFβR1 inhibitor is SB431542.
115. The isolated therapeutic population of any one of claims 81-114, wherein the one or more exogenous compounds decrease expression or activity of IRF7.
116. The isolated therapeutic population of any one of claims 81-115, wherein the one or more exogenous compounds transiently decrease expression or activity of IRF7.
117. The isolated therapeutic population of any one of claims 81-115, wherein the one or more exogenous compounds permanently decrease expression or activity of IRF7.
118. The isolated therapeutic population of any one of claims 81-117, wherein the one or more exogenous compounds comprise a DNA encoding a dominant negative IRF7 mutant operably linked to a promoter active in the TILs.
119. The isolated therapeutic population of claim 118, wherein the DNA encoding the dominant negative IRF7 mutant is in a viral vector.
120. The isolated therapeutic population of claim 119, wherein the viral vector is a lentiviral vector.
121. The isolated therapeutic population of any one of claims 81-116, wherein the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative IRF7 mutant.
122. The isolated therapeutic population of any one of claims 81-116, wherein the one or more exogenous compounds comprise an anti-IRF7 antigen-binding protein.
123. The isolated therapeutic population of claim 122, wherein the antigen- binding protein comprises an antibody.
124. The isolated therapeutic population of any one of claims 81-116, wherein the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to an IRF7 messenger RNA.
125. The isolated therapeutic population of claim 124, wherein the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
126. The isolated therapeutic population of any one of claims 81-117, wherein the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding IRF7.
127. The isolated therapeutic population of claim 126, wherein the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding IRF7.
128. The isolated therapeutic population of claim 127, wherein the Cas protein is a Cas9 protein or a Cas12a protein.
129. The isolated therapeutic population of any one of claims 81-116, wherein the one or more exogenous compounds comprise a small molecule IRF7 inhibitor.
130. The isolated therapeutic population of any one of claims 81-129, wherein the one or more exogenous compounds decrease expression or activity of POLR3A.
131. The isolated therapeutic population of any one of claims 81-130, wherein the one or more exogenous compounds transiently decrease expression or activity of POLR3A.
132. The isolated therapeutic population of any one of claims 81-130, wherein the one or more exogenous compounds permanently decrease expression or activity of POLR3A.
133. The isolated therapeutic population of any one of claims 81-132, wherein the one or more exogenous compounds comprise a DNA encoding a dominant negative POLR3A mutant operably linked to a promoter active in the TILs.
134. The isolated therapeutic population of claim 133, wherein the DNA encoding the dominant negative POLR3A mutant is in a viral vector.
135. The isolated therapeutic population of claim 134, wherein the viral vector is a lentiviral vector.
136. The isolated therapeutic population of any one of claims 81-131, wherein the one or more exogenous compounds comprise a messenger RNA encoding a dominant negative POLR3A mutant.
137. The isolated therapeutic population of any one of claims 81-131, wherein the one or more exogenous compounds comprise an anti-POLR3A antigen-binding protein.
138. The isolated therapeutic population of claim 137, wherein the antigenbinding protein comprises an antibody.
139. The isolated therapeutic population of any one of claims 81-131, wherein the one or more exogenous compounds comprise an inhibitory RNA comprising a region of complementarity to a P0LR3 A messenger RNA.
140. The isolated therapeutic population of claim 139, wherein the inhibitory RNA is an antisense oligonucleotide or an RNAi agent.
141. The isolated therapeutic population of any one of claims 81-132, wherein the one or more exogenous compounds comprise a nuclease agent targeting a nuclease target site in a gene encoding P0LR3 A.
142. The isolated therapeutic population of claim 141, wherein the nuclease agent comprises a Cas protein and a guide RNA that forms a complex with the Cas protein and targets a guide RNA target sequence in the gene encoding P0LR3 A.
143. The isolated therapeutic population of claim 142, wherein the Cas protein is a Cas9 protein or a Cas12a protein.
144. The isolated therapeutic population of any one of claims 81-131, wherein the one or more exogenous compounds comprise a small molecule P0LR3 A inhibitor.
145. The isolated therapeutic population of any one of claims 81-144, wherein the TILs originate from a subject.
146. The isolated therapeutic population of any one of claims 81-145, wherein the TILs are from a tumor biopsy, a lymph node, or ascites.
147. The isolated therapeutic population of claim 146, wherein the tumor is from a bladder cancer, a breast cancer, a cancer caused by human papilloma virus, a cervical cancer, a head and neck cancer, a lung cancer, a melanoma, an ovarian cancer, a non-small-cell lung cancer (NSCLC), a renal cancer, or a renal cell carcinoma.
148. The isolated therapeutic population of claim 146, wherein the tumor biopsy is from a melanoma.
149. The isolated therapeutic population of TILs of any one of claims 80-148, wherein the population comprises about 5x109 to about 5x1010 TILs.
150. A pharmaceutical formulation comprising a pharmaceutically acceptable excipient and the isolated therapeutic population of TILs of any one of claims 80-149.
151. A cryopreserved bag or an intravenous infusion bag, container, or vessel containing contents comprising the isolated therapeutic population of TILs of any one of claims 80-149.
152. A method of treating a cancer in a subject, comprising administering the isolated therapeutic population of TILs of any one of claims 80-149 or the pharmaceutical formulation of claim 150 to the subject.
153. A method for treating a cancer in a subject, comprising: (a) preparing a therapeutic population of TILs according to the method of any one of claims 1-79; and (b) administering a therapeutic amount of the therapeutic population of TILs to the subject with the cancer.
154. The method of claim 152 or 153, wherein the TILs are autologous or allogeneic.
155. The method of any one of claims 152-154, wherein the cancer is a bladder cancer, a breast cancer, a cancer caused by human papilloma virus, a cervical cancer, a head and neck cancer, a head and neck squamous cell carcinoma (HNSCC), a lung cancer, a melanoma, an ovarian cancer, a non-small-cell lung cancer (NSCLC), a renal cancer, or a renal cell carcinoma.
156. The method of any one of claims 152-154, wherein the cancer is a melanoma.
157. The method of any one of claims 152-156, wherein the subject is a human.
158. The method of any one of claims 152-156, wherein the subject is a non- human mammal.
159. The method of claim 158, wherein the non-human mammal is a primate, a rodent, a rat, a mouse, a domesticated mammal, a domesticated cat, a domesticated dog, a domesticated horse, a guinea pig, a laboratory animal, or a companion animal.
160. The method of any one of claims 152-159, wherein the subject is an adult or individual having secondary sexual characteristics.
161. The method of any one of claims 152-159, wherein the subject is not an adult or not individual having secondary sexual characteristics, or is a child or is a not physically mature mammal.
162. The method of any one of claims 152-161, wherein the administering is performed more than once.
163. The method of claim 162, wherein the administering is performed more than once over a course of time, wherein: (1) the course of time is a week and the administering is twice, thrice, four times or five times in the week; (2) the course of time is a month and the administering is twice, thrice of four times in a month; (3) the course of time is three, six, nine, or twelve months and the administering is performed once monthly or once weekly.
164. The method of any one of claims 152-163, wherein the administering is intravenous administration.
PCT/US2023/018458 2022-04-15 2023-04-13 Tumor-infiltrating lymphocyte (til) compositions and uses thereof WO2023200929A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263331697P 2022-04-15 2022-04-15
US63/331,697 2022-04-15
US202263341770P 2022-05-13 2022-05-13
US63/341,770 2022-05-13

Publications (1)

Publication Number Publication Date
WO2023200929A1 true WO2023200929A1 (en) 2023-10-19

Family

ID=88330226

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/018458 WO2023200929A1 (en) 2022-04-15 2023-04-13 Tumor-infiltrating lymphocyte (til) compositions and uses thereof

Country Status (1)

Country Link
WO (1) WO2023200929A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014074785A1 (en) * 2012-11-08 2014-05-15 Ludwig Institute For Cancer Research Ltd. Methods of predicting outcome and treating breast cancer
WO2021081378A1 (en) * 2019-10-25 2021-04-29 Iovance Biotherapeutics, Inc. Gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
US20220033775A1 (en) * 2018-11-05 2022-02-03 Iovance Biotherapeutics, Inc. Expansion of tils utilizing akt pathways inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014074785A1 (en) * 2012-11-08 2014-05-15 Ludwig Institute For Cancer Research Ltd. Methods of predicting outcome and treating breast cancer
US20220033775A1 (en) * 2018-11-05 2022-02-03 Iovance Biotherapeutics, Inc. Expansion of tils utilizing akt pathways inhibitors
WO2021081378A1 (en) * 2019-10-25 2021-04-29 Iovance Biotherapeutics, Inc. Gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy

Similar Documents

Publication Publication Date Title
Lamers-Kok et al. Natural killer cells in clinical development as non-engineered, engineered, and combination therapies
CN111601883B (en) Amplification of TIL from fine needle aspirates and small biopsies
KR102575976B1 (en) Proliferation method of natural killer cells
KR20220119439A (en) Apparatus and method for isolating tumor-infiltrating lymphocytes and uses thereof
JP6073417B2 (en) Spontaneous killing cell proliferation method and composition for spontaneous killing cell proliferation
EP3000876B1 (en) Method for preparing nk cells
EP1666589B1 (en) Process for producing cytotoxic lymphocytes
Davis et al. Interleukin-7 permits Th1/Tc1 maturation and promotes ex vivo expansion of cord blood T cells: a critical step toward adoptive immunotherapy after cord blood transplantation
CN113454209A (en) Preparation and therapeutic use of universal double negative T cells
CN113766919A (en) Manufacture of anti-BCMA CAR T cells
WO2022130015A2 (en) Processing of tumor infiltrating lymphocytes
Shimasaki et al. Engineering of natural killer cells for clinical application
US20070196335A1 (en) Immune Modulation By Regulating Expression Of The "Minor" Gene In Immune Dendritic Cells
WO2023200929A1 (en) Tumor-infiltrating lymphocyte (til) compositions and uses thereof
US20200297768A1 (en) Cd28 t cell cultures, compositions, and methods of using thereof
EP3943932A1 (en) Method for providing immune cells
WO2023200928A1 (en) Methods for identifying tcr repertoire and compositions and uses thereof
WO2011021503A1 (en) Pharmaceutical composition containing transiently surviving ctl
EP3941487B1 (en) Cd28 t cell cultures, compositions, and methods of using thereof
JP7584437B2 (en) Production of anti-BCMA CAR T cells
TW202317756A (en) Methods of isolating of tumor infiltrating lymphocytes and use thereof
AU2022409848A1 (en) Processing of tumor infiltrating lymphocytes
WO2022271847A1 (en) Processing of tumor infiltrating lymphocytes
CN117858901A (en) Method for preparing cells expressing chimeric antigen receptor
CN117480246A (en) Methods and compositions for T cell co-culture potency determination and use with cell therapy products

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: 23788934

Country of ref document: EP

Kind code of ref document: A1