US20200283500A1 - Variant icos ligand immunomodulatory proteins and related compositions and methods - Google Patents

Variant icos ligand immunomodulatory proteins and related compositions and methods Download PDF

Info

Publication number
US20200283500A1
US20200283500A1 US16/757,344 US201816757344A US2020283500A1 US 20200283500 A1 US20200283500 A1 US 20200283500A1 US 201816757344 A US201816757344 A US 201816757344A US 2020283500 A1 US2020283500 A1 US 2020283500A1
Authority
US
United States
Prior art keywords
polypeptide
variant
variant icosl
seq
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/757,344
Other languages
English (en)
Inventor
Lawrence Evans
Michael Kornacker
Ryan Swanson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Alpine Immune Sciences Inc
Original Assignee
Alpine Immune Sciences 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 Alpine Immune Sciences Inc filed Critical Alpine Immune Sciences Inc
Priority to US16/757,344 priority Critical patent/US20200283500A1/en
Assigned to ALPINE IMMUNE SCIENCES, INC. reassignment ALPINE IMMUNE SCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: EVANS, LAWRENCE, KORNACKER, MICHAEL, SWANSON, Ryan
Publication of US20200283500A1 publication Critical patent/US20200283500A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70532B7 molecules, e.g. CD80, CD86
    • 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
    • A61K35/48Reproductive organs
    • A61K35/54Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1774Immunoglobulin superfamily (e.g. CD2, CD4, CD8, ICAM molecules, B7 molecules, Fc-receptors, MHC-molecules)
    • 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
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4621Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
    • 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
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/46434Antigens related to induction of tolerance to non-self
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464403Receptors for growth factors
    • A61K39/464406Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ ErbB4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • 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
    • C12N5/0681Cells of the genital tract; Non-germinal cells from gonads
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • C07K2319/41Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation containing a Myc-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • C07K2319/43Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation containing a FLAG-tag

Definitions

  • the present disclosure relates to therapeutic compositions for modulating immune response in the treatment of cancer and immunological diseases.
  • the present disclosure relates to particular variants of ICOS Ligand (ICOSL) that exhibit improved binding, such as improved affinity or selectivity for one or both of the cognate binding partner proteins ICOS or CD28.
  • ICOSL ICOS Ligand
  • IS immunological synapse
  • APCs antigen-presenting cells
  • target cells and lymphocytes are of increasing medical interest.
  • cell surface proteins in the IS can involve the coordinated and often simultaneous interaction of multiple protein targets with a single protein to which they bind. IS interactions occur in close association with the junction of two cells, and a single protein in this structure can interact with both a protein on the same cell (cis) as well as a protein on the associated cell (trans), likely at the same time.
  • therapeutics are known that can modulate the IS, improved therapeutics are needed.
  • immunomodulatory proteins including soluble proteins or transmembrane immunomodulatory proteins capable of being expressed on cells, that meet such needs.
  • ICOSL ICOS Ligand
  • IgSF immunoglobulin superfamily
  • the variant ICOSL polypeptide exhibits altered binding to the ectodomain(s) of ICOS or CD28 compared to the binding of the ICOSL reference polypeptide for the same ectodomain(s). In some of any of the provided embodiments, the variant ICOSL polypeptide exhibits increased binding to the ectodomain(s) of ICOS or CD28 compared to the binding of the ICOSL reference polypeptide for the same ectodomain(s).
  • the C-terminal truncation is of at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 125 amino acid residues.
  • the ICOSL reference polypeptide is altered in or lacks a protease cleavage site set forth as amino acids 204-209 of SEQ ID NO:32.
  • the ICOSL reference polypeptide contains the sequence of amino acids set forth in SEQ ID NO: 545.
  • the ICOSL reference polypeptide consists of the sequence of amino acids set forth in SEQ ID NO: 545.
  • ICOSL ICOSL Ligand
  • IgSF immunoglobulin superfamily
  • the variant ICOSL polypeptide exhibits altered binding to one or more of its binding partner(s) compared to the binding of the ICOSL reference polypeptide for the one or more binding partner(s). In some of any of the provided embodiments, the variant ICOSL polypeptide exhibits increased binding to one or more of its binding partner(s) compared to the binding of the ICOSL reference polypeptide for the one or more binding partner(s).
  • the alteration includes a deletion of one or more contiguous amino acids corresponding to amino acids 204-209 with reference to SEQ ID NO: 32.
  • the ICOSL reference polypeptide comprises the sequence of amino acids set forth in any of SEQ ID NOS: 600-605. In some of any of the provided embodiments, the ICOSL reference polypeptide consists of the sequence of amino acids set forth in any of SEQ ID NOS: 600-605.
  • the alteration includes at least one amino acid substitution at one or both of position 207 and 208 corresponding to positions set forth in SEQ ID NO: 32.
  • the at least one amino acid substitution is N207A, N207G or L208G, or a conservative amino acid substitution thereof.
  • the reference ICOSL polypeptide contains the sequence of amino acids set forth in any of SEQ ID NOS: 623-628. In some of any of the provided embodiments, the reference ICOSL polypeptide consists of the sequence of amino acids set forth in any of SEQ ID NOS: 623-628.
  • the variant ICOSL polypeptide exhibits reduced proteolytic cleavage when expressed from a cell.
  • the cell is a mammalian cell.
  • the cell is a Chinese Hamster Ovary (CHO) cell line or a derivative thereof.
  • the amino acid modification is an amino acid substitution, insertion or deletion.
  • the one or more amino acid modifications are in a position corresponding to position(s) selected from 10, 11, 13, 16, 18, 20, 25, 27, 30, 33, 37, 38, 42, 43, 47, 52, 54, 57, 61, 62, 67, 71, 72, 74, 75, 77, 78, 80, 84, 89, 90, 92, 93, 94, 96, 97, 98, 99, 100, 102, 103, 107, 109, 110, 111, 113, 115, 116, 117, 119, 120, 121, 122, 126, 129, 130, 132, 133, 135, 138, 139, 140, 142, 143, 144, 146, 148, 151, 152, 153, 154, 155, 156, 158, 161, 164, 166, 168, 172, 173,
  • the one or more amino acid modifications are in a position corresponding to position(s) selected from 10, 11, 13, 16, 18, 20, 25, 26, 27, 30, 33, 37, 38, 42, 43, 47, 52, 54, 57, 61, 62, 67, 71, 72, 74, 75, 77, 78, 80, 84, 89, 90, 92, 93, 94, 96, 97, 98, 99, 100, 102, 103, 107, 109, 110, 111, 113, 115, 116, 117, 119, 120, 121, 122, 126, 129, 130, 132, 133, 135, 137, 138, 139, 140, 142, 143, 144, 146, 151, 152, 153, 154, 155, 156, 158, 161, 164, 166, 168, 172, 173, 175, 190, 192, 193, 194, 198, 201, 203,
  • the one or more amino acid modifications are selected from M10V, M10I, V11E, S13G, E16V, S18R, A20V, S25G, F27S, F27C, N30D, Y33del, Q37R, K42E, T43A, Y47H, N52A, N52C, N52D, N52G, N52H, N52K, N52L, N52M, N52Q, N52R, N52S, N52T, N52V, N52Y, S54A, S54P, N57A, N57D, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57Y, N57W, R61S, R61C, Y62F, L67P, A71T, G72R, L74Q, R75Q, D77G, F78L, L80P
  • the one or more amino acid modifications are selected from M10V, M10I, V11E, S13G, E16V, S18R, A20T, A20V, S25G, R26S, F27C, F27S, N30D, Y33del, Q37R, T38P, K42E, T43A, Y47H, N52A, N52C, N52D, N52G, N52H, N52K, N52L, N52M, N52P, N52Q, N52R, N52S, N52T, N52V, N52Y, S54A, S54F, S54P, N57A, N57D, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57W, N57Y, R61C, R61S, Y62F, L67P, A71T, G72R
  • the one or more amino acid modifications are in a position corresponding to position(s) 52, 57 or 100. In some of any of the provided embodiments, the one or more amino acid modifications are selected from N52A, N52C, N52D, N52G, N52H, N52K, N52L, N52M, N52Q, N52R, N52S, N52T, N52V, N52Y, N57A, N57D, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57Y, N57W, Q100A, Q100D, Q100G, Q100K, Q100L, Q100M, Q100N, Q100P, Q100R, Q100S, Q100T or Q100V.
  • the one or more amino acid modifications are selected from N52A, N52C, N52D, N52G, N52H, N52K, N52L, N52M, N52Q, N52R, N52S, N52T, N52V, N52Y, S54A, S54P, N57A, N57D, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57Y, N57W, Q100A, Q100D, Q100G, Q100K, Q100L, Q100M, Q100N, Q100P, Q100R, Q100S, Q100T or Q100V.
  • the one or more amino acid modifications are selected from among N52Y/N57Y/F138L/L203P, N52H/N57Y/Q100P, N52S/Y146C/Y152C, N52H/C198R, N52H/C140D/T225A, N52H/C198R/T225A, N52H/K92R, N52H/S99G, N57Y/Q100P, N52S/S130G/Y152C, N52S/Y152C, N52S/C198R, N52Y/N57Y/Y152C, N52Y/N57Y/H129P/C198R, N52H/L161P/C198R, N52S/T113E, N52D/S54P, N52K/L208P, N52S/Y152H, N52D/V151A, N52H/I143T, N52S/L80P, N52S/R75Q/L203P, N52S/D158G
  • the one or more amino acid modifications are selected from among N52Y/N57Y/F138L/L203P, N52H/N57Y/Q100P, N52S/Y146C/Y152C, N52H/C198R, N52H/C140D/T225A, N52H/C198R/T225A, N52H/K92R, N52H/S99G, N57Y/Q100P, N52S/S130G/Y152C, N52S/Y152C, N52S/C198R, N52Y/N57Y/Y152C, N52Y/N57Y/H129P/C198R, N52H/L161P/C198R, N52S/T113E, N52D/S54P, N52K/L208P, N52S/Y152H, N52D/V151A, N52H/I143T, N52S/L80P, F120S/Y152H/N201S, N
  • the one or more amino acid modifications are N52H/Q100R.
  • the variant ICOSL polypeptide has the sequence set forth in SEQ ID NO:567.
  • the one or more amino acid modifications are N52H/N57Y/Q100R.
  • the variant ICOSL polypeptide contains the sequence set forth in SEQ ID NO:565.
  • the one or more amino acid modifications are N52L/N57H/Q100R.
  • the variant ICOSL polypeptide contains the sequence set forth in SEQ ID NO: 761.
  • the one or more amino acid modifications is N52D.
  • the variant ICOSL polypeptide contains the sequence set forth in SEQ ID NO: 548.
  • the one or more amino acid modifications is N52H/N57Y/Q100P.
  • the variant ICOSL polypeptide contains the sequence set forth in SEQ ID NO: 570.
  • the one or more amino acid modifications are selected from among F120S/Y152H/N201S, E111del, Y33del, N168Q/N207Q, N84Q/N207Q, N155Q/N207Q, N119Q/N168Q, N119Q/N207Q, N119Q/N155Q, N84Q/N119Q, N84Q/N155Q/N168Q, N84Q/N168Q/N207Q, N84Q/N155H/N207Q, N155Q/N168Q/N207Q, N119Q N155Q/N168Q, N119Q/N168Q/N207Q, N84Q/N119Q/N207Q, N119Q/N155H/N207Q, N84Q/N119Q/N155Q, N84Q/N119Q/N155Q/N168Q, N84Q/N155Q/N168Q, N84Q/N119Q/N155Q/N168Q, N84Q/N155Q/N168Q, N
  • the one or more amino acid modifications are selected from C198R, D158G, E16V, E90A, F120S, F138L, F172S, H115R, H115X, I143T, 1143V, I224V, K156M, K42E, K92R, L102R, L203P, L208P, N194D, N30D, N52A, N52D, N52G, N52H, N52K, N52L, N52M, N52Q, N52R, N52S, N52T, N52Y, N57F, N57H, N57K, N57L, N57M, N57P, N57S, N57V, N57W, N57Y, Q100A, Q100D, Q100E, Q100K, Q100M, Q100P, Q100R, Q100S, Q100T, Q133H, R221I, R75Q, S54A, S54P, T113E, T225A, V110D
  • the one or more amino acid modifications are selected from among N52S, N52H, N52D, N52H/N57Y/Q100P, N52S/Y146C/Y152C, N52H/C198R, N52H/C198R/T225A, N52H/K92R, N57Y, N52S/C198R, N52S/T113E, S54A, N52D/S54P, N52K/L208P, N52H/I143T, N52S/R75Q/L203P, N52S/D158G, N52D/Q133H, N52H/N57Y/Q100R/V122A, N52H/N57Y/Q100R/F172S, N52H/N57Y/Q100R, N52S/N194D, N52H/N57Y/Q100R/L102R/V110D/H115R/C198R, N52S/E90A, N52S/F
  • the one or more amino acid modifications are selected from C198R, D158G, E16V, E90A, F120S, F138L, F172S, H115R, I143V, I224V, K156M, K42E, K92R, L102R, L203P, L208P, N194D, N30D, N52A, N52D, N52G, N52H, N52K, N52L, N52M, N52Q, N52R, N52S, N52T, N52Y, N57F, N57H, N57L, N57M, N57S, N57V, N57W, N57Y, Q100A, Q100E, Q100G, Q100K, Q100M, Q100P, Q100R, Q100S, Q133H, S212G, S54A, S54P, T113E, V110D, V122A, Y146C, Y152C, or T225A.
  • the one or more amino acid modifications are selected from among N52A/N57Y/Q100A, N52D/Q100S, N52G/Q100A, N52M/N57H/Q100S, N52M/N57W/Q100P, N52Q/N57S/Q100A, N52R/N57L/Q100A, N52S/N57H/Q100E, N52S/N57L/Q100S, N52S/N57M/Q100S, N52S/N57Y/Q100M, N52T/N57H/Q100S, N52R/N57F/Q100P, N52R/N57F/Q100T, N52R/N57W/Q100K, N52R/N57W, N52G/N57V, N52L/N57V, N52S/N57L/Q100G, N52T/N57K/Q100P, N52S, N52H, N52D, N52Y/N57Y/
  • the variant ICOSL polypeptide contains the sequence of amino acids set forth in any one of SEQ ID NOS: 546-599, 734-781, 783, 786, 788, 792, 796, 798, 800, 802, 804, 806, 808, 811, 813, 815, 817, 818, 820, 822, 824, 826, 827, 829, 831, 833, 834, 836, 838, 840-843, 845, 847, 848, 850-853, 855, 857, 907, 910, or a sequence of amino acids that exhibits at least 90%, 91%%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOS: 546-599, 734-781, 783, 786, 788, 792, 796, 798, 800, 802, 804, 806, 808, 811, 813, 815, 8
  • the variant ICOSL polypeptide consists of the sequence of amino acids set forth in any one of SEQ ID NOS: 546-599, 734-781, 783, 786, 788, 792, 796, 798, 800, 802, 804, 806, 808, 811, 813, 815, 817, 818, 820, 822, 824, 826, 827, 829, 831, 833, 834, 836, 838, 840-843, 845, 847, 848, 850-853, 855, 857, 907, 910, or a sequence of amino acids that exhibits at least 90%, 91%%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOS: 546-599, 734-781, 783, 786, 788, 792, 796, 798, 800, 802, 804, 806, 808, 811, 813, 815
  • the variant ICOSL polypeptide contains one or more amino acid modifications in an ICOSL reference polypeptide or a specific binding fragment thereof corresponding to amino acid modifications are selected from N52A, N52C, N52D, N52G, N52K, N52L, N52M, N52R, N52T, N52V, N57A, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57W, Q100A, Q100D, Q100G, Q100L, Q100M, Q100N, Q100R, Q100S, Q100T or Q100V with reference to SEQ ID NO:32.
  • the one or more amino acid modifications are selected from among N52A/N57F/Q100S, N52A, /N57H/Q100S, N52A/N57Y/Q100A, N52D/N57A/Q100A, N52D/Q100S, N52G/Q100A, N52H/Q100A, N52M/N57H/Q100S, N52M/N57W/Q100P, N52Q/N57F, N52Q/N57S/Q100A, N52R/N57L/Q100A, N52R/N57Y/Q100P, N52R/N57Y/Q100S, N52S/N57A/Q100A, N52S/N57H/Q100E, N52S/N57L/Q100S, N52S/N57M/Q100S, N52S/N57Y/Q100S, N52S/N57Y/Q100M, N52S/N57Y/Q100M, N52S/N
  • the ICOSL reference polypeptide is a mammalian ICOSL or a specific binding fragment thereof. In some examples, the ICOSL reference polypeptide is a human ICOSL or a specific binding fragment thereof.
  • the ICOSL reference polypeptide contains (i) the sequence of amino acids set forth in SEQ ID NO:32, (ii) a sequence of amino acids that has at least 95% sequence identity to SEQ ID NO:32; or (iii) a portion of (i) or (ii) comprising an IgV domain or IgC domain or specific binding fragments thereof or both.
  • the specific binding fragment of the IgV domain or IgC domain has a length of at least 50, 60, 70, 80, 90, 100, 110 or more amino acids; or the specific binding fragment of the IgV domain contains a length that is at least 80% of the length of the IgV domain set for as amino acids 19-129 of SEQ ID NO:5 and/or the specific binding fragment of the IgC domain comprises a length that is at least 80% of the length of the IgC domain set forth as amino acids 141-227 of SEQ ID NO:5.
  • the variant ICOSL polypeptide comprises the IgV domain or a specific fragment thereof and the IgC domain or a specific fragment thereof.
  • the variant ICOSL polypeptide contains the sequence of amino acids set forth in any one of SEQ ID NOS: 638-685, or a sequence of amino acids that exhibits at least 90%, 91%%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOS: 638-685.
  • the variant ICOSL polypeptide consists of the sequence of amino acids set forth in any one of SEQ ID NOS: 638-685, or a sequence of amino acids that exhibits at least 90%, 91%%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOS: 638-685.
  • the variant ICOSL polypeptide contains the IgV domain or a specific binding fragment thereof. In some of any of the provided embodiments, the variant ICOSL polypeptide contains the sequence of amino acids set forth in any one of SEQ ID NOS: 686-781, or a sequence of amino acids that exhibits at least 90%, 91%%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOS: 686-781.
  • the variant ICOSL polypeptide consists of the sequence of amino acids set forth in any one of SEQ ID NOS: 686-781, or a sequence of amino acids that exhibits at least 90%, 91%%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOS: 686-781.
  • the IgV domain or specific binding fragment thereof is the only ICOSL portion of the variant ICOSL polypeptide. In some examples, the IgC domain or specific binding fragment thereof is the only ICOSL portion of the variant ICOSL polypeptide.
  • the variant ICOSL polypeptide exhibits altered binding to the ectodomain of ICOS or CD28 compared to the binding of the ICOSL reference polypeptide for the same ectodomain. In some aspects, the variant ICOSL polypeptide exhibits increased binding to the ectodomain(s) of ICOS or CD28 compared to the binding of the ICOSL reference polypeptide for the same ectodomain(s). In some examples, the binding is increased more than 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold or 60-fold.
  • the ICOS is a human ICOS.
  • the CD28 is a human CD28.
  • the variant ICOSL polypeptide exhibits decreased binding to the ectodomain of CTLA-4 compared to the binding of the reference ICOSL polypeptide for the same ectodomain. In some examples, the binding is decreased more than 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold or 60-fold.
  • the CTLA-4 is a human CTLA-4.
  • the altered (increased or decreased) binding is altered (increased or decreased) binding affinity.
  • the variant ICOSL polypeptide comprises up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 amino acid modifications, optionally amino acid substitutions, insertions and/or deletions. In some cases, the variant ICOSL polypeptide exhibits at least or at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to the ICOSL reference polypeptide.
  • the variant ICOSL polypeptide is a soluble protein. In some of any of the provided embodiments, the variant ICOSL polypeptide lacks a transmembrane domain and intracellular signaling domain; and/or when expressed from a cell, the variant ICOSL polypeptide is not expressed on the surface of the cell.
  • the variant ICOSL polypeptide further contains transmembrane domain.
  • the transmembrane domain contains the sequence of amino acids set forth as residues 257-277 of SEQ ID NO:5 or a functional variant thereof that exhibits at least 85% sequence identity to residues 257-277 of SEQ ID NO:5.
  • the variant ICOSL polypeptide further contains a cytoplasmic signaling domain linked to the transmembrane domain.
  • the cytoplasmic signaling domain contains the sequence of amino acids set forth as residues 278-302 of SEQ ID NO:5 or a functional variant thereof that exhibits at least 85% sequence identity to residues 278-302 of SEQ ID NO:5.
  • the variant ICOSL polypeptide is deglycosylated or partially deglycosylated compared to the ICOSL reference sequence.
  • an immunomodulatory protein containing any of the provided variant ICOSL polypeptide and a half-life extending moiety.
  • the half-life extending moiety comprises a multimerization domain, albumin, an albumin-binding polypeptide, Pro/Ala/Ser (PAS), a C-terminal peptide (CTP) of the beta subunit of human chorionic gonadotropin, polyethylene glycol (PEG), long unstructured hydrophilic sequences of amino acids (XTEN), hydroxyethyl starch (HES), an albumin-binding small molecule, or a combination thereof.
  • the half-life extending moiety is or comprises Pro/Ala/Ser (PAS) and the variant ICOSL polypeptide is PASylated.
  • the half-life extending moiety contains the sequence set forth in SEQ ID NO:904.
  • the half-life extending moiety is or contains a multimerization domain.
  • the multimerization domain is selected from an Fc region of an immunoglobulin, a leucine zipper, an isoleucine zipper or a zinc finger.
  • the variant ICOSL polypeptide is linked, directly or indirectly via a linker, to the multimerization domain.
  • the immunomodulatory protein is a multimer containing a first variant ICOSL polypeptide linked to a first multimerization domain and a second variant ICOSL polypeptide linked to a second multimerization domain, wherein the first and second multimerization domains interact to form a multimer comprising the first and second variant ICOSL polypeptide.
  • the multimer is a dimer.
  • the first variant ICOSL polypeptide and the second variant ICOSL polypeptide are the same.
  • the dimer is a homodimer. In some instances, the dimer is a heterodimer.
  • the multimerization domain is or contains an Fc region of an immunoglobulin.
  • the Fc region is of an immunoglobulin G1 (IgG1) or an immunoglobulin G2 (IgG2) protein.
  • the immunoglobulin protein is human and/or the Fc region is human.
  • the Fc region contains the sequence of amino acids set forth in SEQ ID NO: 227 or a variant thereof that exhibits at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO:227.
  • the Fc region contains the sequence of amino acids set forth in SEQ ID NO: 226 or a variant thereof that exhibits at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO:226. In some of any of the provided embodiments, the Fc region exhibits one or more effector functions. In some of any of the provided embodiments, the Fc region exhibits one or more reduced effector function compared to a wildtype Fc region, optionally wherein the wildtype human Fc is of human IgG1.
  • the one or more effector function is selected from among antibody dependent cellular cytotoxicity (ADCC), complement dependent cytotoxicity, programmed cell death and cellular phagocytosis.
  • ADCC antibody dependent cellular cytotoxicity
  • the Fc region is a variant Fc region comprising one or more amino acid substitutions compared to the wildtype Fc region.
  • the one or more amino acid substitutions of the variant Fc region are selected from N297G, E233P/L234V/L235A/G236del/S267K or L234A/L235E/G237A, wherein the residue is numbered according to the EU index of Kabat.
  • the variant Fc region further contains the amino acid substitution C220S, wherein the residues are numbered according to the EU index of Kabat.
  • the Fc region contains the sequence of amino acid sequence set forth in any of SEQ ID NOS: 476-478 or a sequence of amino acids that exhibits at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any of SEQ ID NOS:476-478 and contains the amino acid substitutions.
  • the Fc region contains K447del, wherein the residue is numbered according to the EU index of Kabat.
  • the Fc region contains the sequence of amino acid sequence set forth in any of SEQ ID NOS: 632-634 or a sequence of amino acids that exhibits at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any of SEQ ID NOS: 632-634 and contains the amino acid substitutions.
  • the Fc region contains the sequence of amino acid sequence set forth in SEQ ID NOS: 474 or 637, or a sequence of amino acids that exhibits at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NOS: 474 or 637 and contains the amino acid substitutions.
  • the Fc region contains the sequence of amino acids set forth in SEQ ID NO: 478 or SEQ ID NO: 634. In some of any of the provided embodiments, the Fc region contains the sequence of amino acids set forth in SEQ ID NO: 477. In some of any of the provided embodiments, the Fc region contains the sequence of amino acids set forth in SEQ ID NO: 633. In some of any of the provided embodiments, the Fc region contains the sequence of amino acids set forth in SEQ ID NO: 474. In some of any of the provided embodiments, the Fc region contains the sequence of amino acids set forth in SEQ ID NO:637.
  • an immunomodulatory protein containing (a) a variant ICOSL polypeptide containing one or more amino acid modifications in an immunoglobulin superfamily (IgSF) domain of an ICOSL reference polypeptide, wherein the variant ICOSL polypeptide exhibits altered binding to the ectodomain(s) of ICOS or CD28 compared to the binding of the ICOSL reference polypeptide for the same ectodomain(s); and (b) a variant Fc region containing amino acid substitutions selected from N297G/K447del, E233P/L234V/L235A/G236del/S267K/K447del or L234A/L235E/G237A/K447del compared to wildtype human IgG1, wherein the residues are numbered according to the EU index of Kabat.
  • IgSF immunoglobulin superfamily
  • the immunomodulatory protein is a dimer.
  • the variant Fc region further contains the amino acid substitution C220S, wherein the residues are numbered according to the EU index of Kabat.
  • the Fc region contains the sequence of amino acid sequence set forth in any of SEQ ID NOS: 632-634 or a sequence of amino acids that exhibits at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any of SEQ ID NOS:632-634 and contains the amino acid substitutions.
  • the Fc region contains the sequence of amino acid sequence set forth in SEQ ID NOS: 474 or 637, or a sequence of amino acids that exhibits at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NOS: 474 or 637 and contains the amino acid substitutions.
  • the variant ICOSL polypeptide is linked, directly or indirectly via a linker, to the variant Fc region.
  • the linker contains 1 to 10 amino acids.
  • the linker is selected from AAA, G4S (SEQ ID NO: 636), (G 4 S) 2 (SEQ ID NO: 229) or GSGGGGS linker (SEQ ID NO: 635).
  • the linker is (G 4 S) 3 (SEQ ID NO: 228).
  • the linker is AAA. In some of any of the provided embodiments, the linker is G4S (SEQ ID NO:636). In some of any of the provided embodiments, the linker is (G 4 S) 2 (SEQ ID NO:229). In some of any of the provided embodiments, the linker is GSGGGGS linker (SEQ ID NO: 635).
  • the variant ICOSL polypeptide is or comprise an IgV domain.
  • the variant ICOSL polypeptide contains amino acid modifications N52H/Q100R.
  • the variant ICOSL polypeptide has the sequence set forth in SEQ ID NO: 567.
  • the variant ICOSL polypeptide comprises amino acid modifications N52H/N57Y/Q100R.
  • the variant ICOSL polypeptide has the sequence set forth in SEQ ID NO: 565.
  • the variant ICOSL polypeptide comprises amino acid modifications are N52L/N57H/Q100R. In some of any of the provided embodiments, the variant ICOSL polypeptide has the sequence set forth in SEQ ID NO: 761. In some of any of the provided embodiments, the variant ICOSL polypeptide comprises amino acid modifications N52H/N57Y/Q100P. In some of any of the provided embodiments, the variant ICOSL polypeptide has the sequence set forth in SEQ ID NO: 570. In some of any of the provided embodiments, the variant ICOSL polypeptide comprises the amino acid modification is N52D. In some of any of the provided embodiments, the polypeptide has the sequence set forth in SEQ ID NO: 548.
  • a variant ICOSL-Fc fusion protein that has the sequence of amino acids set forth in SEQ ID NO: 928.
  • a variant ICOSL-Fc fusion protein that has a sequence of amino acids that has at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to the sequence set forth in SEQ ID NO: 928.
  • the provided immunomodulatory proteins such as fusion proteins, e.g. variant ICOSL-Fc fusion proteins, binds CD28 and ICOS.
  • the variant ICOSL-Fc fusion protein exhibits increased binding affinity to CD28 and/or ICOS compared to a human wild-type ICOSL-Fc fusion protein, e.g. containing an ICOSL IgV portion set forth in SEQ ID NO:545 linked via a linker, e.g. set forth in SEQ ID NO:229, to an Fc region.
  • the Fc region is an inert or effectorless Fc containing the mutations L234A, L235E and L235E in a human IgG1 Fc, e.g. set forth in SEQ ID NO:637,
  • an immunomodulatory protein containing any of the variant ICOSL polypeptides linked to a second polypeptide comprising an immunoglobulin superfamily (IgSF) domain.
  • IgSF domain is affinity modified and exhibits altered binding to one or more of its cognate binding partner(s) compared to the unmodified or wild-type IgSF domain.
  • the IgSF domain exhibits increased binding to one or more of its cognate binding partner(s) compared to the unmodified or wild-type IgSF domain.
  • the variant ICOSL polypeptide is a first ICOSL variant polypeptide and the IgSF domain of the second polypeptide is an IgSF domain from a second variant ICOSL polypeptide provided herein, wherein the first and second ICOSL variant are the same or different.
  • the variant ICOSL polypeptide is capable of specifically binding to CD28 or ICOS and the IgSF domain of the second polypeptide is capable of binding to a binding partner other than one specifically bound by the ICOSL variant polypeptide.
  • the IgSF domain is from a member of the B7 family.
  • the IgSF domain is a tumor-localizing moiety that binds to a ligand expressed on a tumor or is an inflammatory-localizing moiety that binds to a ligand expressed on a cell or tissue of an inflammatory environment.
  • the ligand is B7H6.
  • the IgSF domain is from NKp30.
  • the IgSF domain of the second polypeptide is or comprises an IgV domain. In some of any of the provided embodiments, the IgSF domain of the second polypeptide is a variant NKp30 molecule containing L30V/A60V/S64P/S86G. In some of any of the provided embodiments, the IgSF domain of the second polypeptide has the sequence set forth in SEQ ID NO: 504.
  • the IgSF domain is or comprises an IgV domain.
  • the variant ICOSL polypeptide is or contains an IgV domain.
  • the variant ICOSL polypeptide is or comprise an IgV domain. In some of any of the provided embodiments, the variant ICOSL polypeptide contains amino acid modifications N52H/Q100R. In some of any of the provided embodiments, the variant ICOSL polypeptide has the sequence set forth in SEQ ID NO: 567. In some of any of the provided embodiments, the variant ICOSL polypeptide comprises amino acid modifications N52H/N57Y/Q100R. In some of any of the provided embodiments, the variant ICOSL polypeptide has the sequence set forth in SEQ ID NO: 565. In some of any of the provided embodiments, the variant ICOSL polypeptide comprises amino acid modifications are N52L/N57H/Q100R.
  • the variant ICOSL polypeptide has the sequence set forth in SEQ ID NO: 761. In some of any of the provided embodiments, the variant ICOSL polypeptide comprises amino acid modifications N52H/N57Y/Q100P. In some of any of the provided embodiments, the variant ICOSL polypeptide has the sequence set forth in SEQ ID NO: 570. In some of any of the provided embodiments, the variant ICOSL polypeptide comprises the amino acid modification is N52D. In some of any of the provided embodiments, the polypeptide has the sequence set forth in SEQ ID NO:548.
  • the immunomodulatory protein comprises a multimerization domain linked to one or both of the variant ICOSL polypeptide or the second polypeptide comprising the IgSF domain.
  • the multimerization domain is an Fc domain or a variant thereof with reduced effector function.
  • the immunomodulatory protein is dimeric. In some cases, the immunomodulatory protein is homodimeric. In some aspects, the immunomodulatory protein is heterodimeric.
  • a conjugate containing any of the provided variant ICOSL polypeptides or any of the provided immunomodulatory protein and a heterologous moiety containing any of the provided variant ICOSL polypeptides or any of the provided immunomodulatory protein and a heterologous moiety.
  • the variant ICOSL polypeptide is linked, directly or indirectly via a linker, to the heterologous moiety.
  • the targeting moiety is a protein, a peptide, nucleic acid, small molecule or nanoparticle.
  • the target moiety is a protein or a peptide.
  • the conjugate is a fusion protein.
  • a fusion protein containing any of the provided variant ICOSL polypeptides or any of the provided immunomodulatory protein and a heterologous moiety.
  • the moiety is a targeting moiety that specifically binds to a molecule on the surface of a cell.
  • the targeting moiety specifically binds to a molecule on the surface of an immune cell.
  • the immune cell is an antigen presenting cell or a lymphocyte.
  • the targeting moiety is a tumor-localizing moiety that binds to a molecule on the surface of a tumor.
  • the targeting moiety binds to a molecule HER1/EGFR, HER2/ERBB2, CD20, CD25 (IL-2R ⁇ receptor), CD33, CD52, CD133, CD206, CEA, CEACAM1, CEACAM3, CEACAM5, CEACAM6, cancer antigen 125 (CA125), alpha-fetoprotein (AFP), Lewis Y, TAG72, Caprin-1, mesothelin, PDGF receptor (PDGFR; such as PDGF-R ⁇ ), PD-1, PD-L1, CTLA-4, IL-2 receptor, vascular endothelial growth factor (VEGF), CD30, EpCAM, EphA2, Glypican-3, gpA33, mucins, CAIX, PSMA, folate-binding protein, gangliosides (such as GD2, GD3, GM1 and GM2), VEGF receptor (VEGFR), VEGFR2, VEGF-A, integrin ⁇ V ⁇
  • the targeting moiety is an antibody or antigen-binding fragment.
  • the antibody is selected from cetuximab, panitumumab, zalutumumab, nimotuzumab, trastuzumab, Ado-trastuzumab emtansine, Tositumomab (Bexxar®), Rituximab (Rituxan, Mabthera), Ibritumomab tiuxetan (Zevalin), Daclizumab (Zenapax), Gemtuzumab (Mylotarg), Alemtuzumab, CEA-scan Fab fragment, OC125 monoclonal antibody, ab75705, B72.3, Bevacizumab (Avastin®), Afatinib, Axitinib, Bosutinib, Cabozantinib, Ceritinib, Crizotini
  • the variant ICOSL polypeptide is linked, directly or indirectly via a linker, to the N-terminus of the heavy and/or light chain of the antibody or antigen-binding fragment. In some cases, the variant ICOSL polypeptide is linked, directly or indirectly via a linker, to the C-terminus of the heavy and/or light chain of the antibody or antigen binding fragment.
  • the conjugate is divalent, tetravalent, hexavalent or octavalent.
  • the heterologous moiety is or contains a label for detection or purification of the variant ICOSL polypeptide.
  • a monovalent fusion protein containing a variant ICOSL polypeptide containing one or more amino acid modifications in an immunoglobulin superfamily (IgSF) domain of an ICOSL reference polypeptide, wherein the variant ICOSL polypeptide exhibits altered binding to the ectodomain(s) of ICOS or CD28 compared to the binding of the ICOSL reference polypeptide for the same ectodomain(s); and a label for detection or purification of the variant ICOSL polypeptide.
  • the label for detection or purification is selected from a poly-histidine (His) tag, a FLAG-tag, a Myc-tag, or a fluorescent protein-tag.
  • the variant ICOSL polypeptide contains one or more amino acid modifications in a position corresponding to position(s) selected from 10, 11, 13, 16, 18, 20, 25, 27, 30, 33, 37, 38, 42, 43, 47, 52, 54, 57, 61, 62, 67, 71, 72, 74, 75, 77, 78, 80, 84, 89, 90, 92, 93, 94, 96, 97, 98, 99, 100, 102, 103, 107, 109, 110, 111, 113, 115, 116, 117, 119, 120, 121, 122, 126, 129, 130, 132, 133, 135, 138, 139, 140, 142, 143, 144, 146, 148, 151, 152, 153, 154, 155, 156, 158, 161, 164, 166, 168, 172, 173, 175, 190, 192, 193, 194, 198,
  • the variant ICOSL polypeptide contains one or more amino acid modifications are in a position corresponding to position(s) selected from 10, 11, 13, 16, 18, 20, 25, 26, 27, 30, 33, 37, 38, 42, 43, 47, 52, 54, 57, 61, 62, 67, 71, 72, 74, 75, 77, 78, 80, 84, 89, 90, 92, 93, 94, 96, 97, 98, 99, 100, 102, 103, 107, 109, 110, 111, 113, 115, 116, 117, 119, 120, 121, 122, 126, 129, 130, 132, 133, 135, 137, 138, 139, 140, 142, 143, 144, 146, 151, 152, 153, 154, 155, 156, 158, 161, 164, 166, 168, 172, 173, 175, 190, 192, 193, 194, 198
  • the one or more amino acid modifications are selected from M10V, M10I, V11E, S13G, E16V, S18R, A20V, S25G, F27S, F27C, N30D, Y33del, Q37R, K42E, T43A, Y47H, N52A, N52C, N52D, N52G, N52H, N52K, N52L, N52M, N52Q, N52R, N52S, N52T, N52V, N52Y, N52K, S54A, S54P, N57A, N57D, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57Y, N57W, R61S, R61C, Y62F, L67P, A71T, G72R, L74Q, R75Q, D77G, F78L, L80P, N
  • the one or more amino acid modifications are selected from M10V, M10I, V11E, S13G, E16V, S18R, A20T, A20V, S25G, R26S, F27C, F27S, N30D, Y33del, Q37R, T38P, K42E, T43A, Y47H, N52A, N52C, N52D, N52G, N52H, N52K, N52L, N52M, N52P, N52Q, N52R, N52S, N52T, N52V, N52Y, S54A, S54F, S54P, N57A, N57D, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57W, N57Y, R61C, R61S, Y62F, L67P, A71T, G72R, L74Q, R
  • the ICOSL reference polypeptide contains (i) the sequence of amino acids set forth in SEQ ID NO:32, (ii) a sequence of amino acids that has at least 95% sequence identity to SEQ ID NO:32; or (iii) a portion of (i) or (ii) comprising an IgV domain or IgC domain or specific binding fragments thereof or both.
  • the ICOSL reference polypeptide comprises the sequence of amino acids set forth in any of SEQ ID NOS: 196, 545, 600-605 and 623-628. In some aspects, the ICOSL reference polypeptide consists of the sequence of amino acids set forth in any of SEQ ID NOS: 32, 196, 545, 600-605 and 623-628.
  • nucleic acid molecule(s) encoding any of the provided variant ICOSL polypeptides, immunomodulatory proteins, or fusion proteins.
  • the nucleic acid molecules(s) is synthetic nucleic acid.
  • the nucleic acid molecule(s) is cDNA.
  • the vector containing any of the provided the nucleic acid molecule(s).
  • the vector is an expression vector.
  • the vector is a mammalian expression vector or a viral vector.
  • the cell containing any of the provided vectors.
  • the cell is a mammalian cell.
  • the cell is a Chinese Hamster Ovary (CHO) cell or a derivative thereof.
  • the method further includes isolating or purifying the protein from the cell.
  • the protein or immunomodulatory protein is purified from Chinese Hamster Ovary Cells or a derivative thereof.
  • a polynucleotide containing a nucleic acid encoding a variant ICOSL polypeptide containing a provided transmembrane domain and one or more nucleic acid encoding one or more chain of a recombinant antigen receptor.
  • the recombinant antigen receptor is a chimeric antigen receptor (CAR) or an engineered T cell receptor (TCR).
  • each of the nucleic acid encoding the variant ICOSL polypeptide and the one or more nucleic acid encoding one or more chain of the recombinant receptor is separated by a nucleic acid encoding a self-cleaving peptide or a peptide that causes ribosome skipping.
  • the polynucleotide contains the nucleic acid encoding the variant ICOSL polypeptide, a nucleic acid encoding a self-cleaving peptide or a peptide that causes ribosome skipping and a nucleic acid encoding a CAR.
  • the polynucleotide comprises the nucleic acid encoding the variant ICOSL polypeptide, a nucleic acid encoding a first self-cleaving peptide or a peptide that causes ribosome skipping, a nucleic acid encoding one of an engineered TCRalpha chain or an engineered TCRbeta chain, a nucleic acid encoding a second self-cleaving peptide or a peptide that causes ribosome skipping, and a nucleic acid encoding the other of the engineered TCRalpha chain or the engineered TCRbeta chain.
  • the encoded first and second self-cleaving peptide is the same.
  • the self-cleaving peptide or the peptide that causes ribosome skipping is a T2A, a P2A, a E2A or a F2A.
  • the vector is a viral vector.
  • the viral vector is a retroviral vector or a lentiviral vector.
  • an engineered cell containing any of the provided polynucleotides or vectors is also provided. Also provided is an engineered cell containing any of the provided variant ICOSL polypeptides, immunomodulatory proteins, or fusion proteins.
  • an engineered cell containing any of the provided nucleic acid molecules or the vectors.
  • the nucleic acid encoding the variant ICOSL polypeptide, immunomodulatory protein or fusion protein encodes a signal peptide.
  • the variant ICOSL polypeptide, immunomodulatory protein or fusion protein does not contain a transmembrane domain and/or is not expressed on the surface of the cell.
  • the variant ICOSL polypeptide, immunomodulatory protein or fusion protein is secreted from the engineered cell.
  • the engineered cell contains a variant ICOSL polypeptide containing a transmembrane domain.
  • the variant ICOSL polypeptide is expressed on the surface of the cell.
  • the cell is an immune cell.
  • the immune cell is an antigen presenting cell (APC) or a lymphocyte.
  • the engineered cell is primary cell.
  • the cell is a mammalian cell.
  • the cell is a human cell.
  • the lymphocyte is a T cell.
  • the engineered cell is an APC and the APC is an artificial APC.
  • the engineered cell further contains a chimeric antigen receptor (CAR) or an engineered T-cell receptor.
  • CAR chimeric antigen receptor
  • an infectious agent containing a nucleic acid molecule encoding a provided variant ICOSL polypeptide or a provided immunomodulatory protein, a provided the fusion protein.
  • the encoded variant ICOSL polypeptide, immunomodulatory protein or fusion protein does not contain a transmembrane domain and/or is not expressed on the surface of a cell in which it is expressed.
  • the encoded variant ICOSL polypeptide, immunomodulatory protein or fusion protein is secreted from the infectious agent when it is expressed.
  • the encoded variant ICOSL polypeptide comprises a transmembrane domain.
  • the encoded variant ICOSL polypeptide is expressed on the surface of a cell in which it is expressed.
  • the infectious agent is a bacteria or a virus.
  • the virus is an oncolytic virus.
  • the oncolytic virus is an adenoviruses, adeno-associated viruses, herpes viruses, Herpes Simplex Virus, Vesticular Stomatic virus, Reovirus, Newcastle Disease virus, parvovirus, measles virus, vesticular stomatitis virus (VSV), Coxsackie virus or a Vaccinia virus.
  • the virus specifically targets dendritic cells (DCs) and/or is dendritic cell-tropic.
  • DCs dendritic cells
  • the virus is a lentiviral vector that is pseudotyped with a modified Sindbis virus envelope product.
  • the infectious agent further contains a nucleic acid molecule encoding a further gene product that results in death of a target cell or that can augment or boost an immune response.
  • the further gene product is selected from an anticancer agent, anti-metastatic agent, an antiangiogenic agent, an immunomodulatory molecule, an immune checkpoint inhibitor, an antibody, a cytokine, a growth factor, an antigen, a cytotoxic gene product, a pro-apoptotic gene product, an anti-apoptotic gene product, a cell matrix degradative gene, genes for tissue regeneration or a reprogramming human somatic cells to pluripotency.
  • a pharmaceutical composition containing any of the variant ICOSL polypeptides, immunomodulatory proteins, conjugates or fusion proteins or any of the provided engineered cells or infectious agents.
  • the pharmaceutical composition contains a pharmaceutically acceptable excipient.
  • the pharmaceutical composition is sterile.
  • the vial is sealed.
  • kit containing any of the provided compositions and instructions for use is also provided. Also provided is a kit containing any of the provided articles of manufacture and instructions for use.
  • a method of modulating an immune response in a subject including administering the pharmaceutical composition to the subject. Also provided is a method of modulating an immune response in a subject including administering the engineered cells.
  • the engineered cells are autologous to the subject. In some of any of the provided embodiments, the engineered cells are allogenic to the subject.
  • the modulating the immune response treats a disease or condition in the subject. In some aspects, the immune response is increased.
  • an immunomodulatory protein or conjugate containing a variant ICOSL polypeptide linked to a tumor-localizing moiety is administered to the subject.
  • the tumor-localizing moiety is or comprises a binding molecule that recognizes a tumor antigen.
  • the binding molecule contains an antibody or an antigen-binding fragment thereof or comprises a wild-type IgSF domain or variant thereof.
  • the immunomodulatory protein or the conjugate or fusion protein is administered to the subject.
  • a variant ICOSL polypeptide that is a transmembrane immunomodulatory protein is administered to the subject.
  • the engineered cell containing a variant ICOSL polypeptide that is a transmembrane immunomodulatory protein is administered to the subject.
  • the disease or condition is a tumor or cancer.
  • the disease or condition is selected from melanoma, lung cancer, bladder cancer, a hematological malignancy, liver cancer, brain cancer, renal cancer, breast cancer, pancreatic cancer, colorectal cancer, spleen cancer, prostate cancer, testicular cancer, ovarian cancer, uterine cancer, gastric carcinoma, a musculoskeletal cancer, a head and neck cancer, a gastrointestinal cancer, a germ cell cancer, or an endocrine and neuroendocrine cancer.
  • the immune response is decreased.
  • a variant ICOSL polypeptide or immunomodulatory protein that is soluble is administered to the subject.
  • the soluble immunomodulatory protein is an immunomodulatory Fc fusion protein.
  • a provided variant ICOSL polypeptide, immunomodulatory protein, or fusion protein is administered to the subject.
  • an engineered cell containing a secretable variant ICOSL polypeptide is administered to the subject.
  • a provided engineered cell is administered to the subject.
  • an infectious agent encoding a variant ICOSL polypeptide that is a secretable immunomodulatory protein is administered to the subject, optionally under conditions in which the infectious agent infects a tumor cell or immune cell and the secretable immunomodulatory protein is secreted from the infected cell.
  • the disease or condition is an inflammatory or autoimmune disease or condition.
  • the disease or condition is an Antineutrophil cytoplasmic antibodies (ANCA)-associated vasculitis, a vasculitis, an autoimmune skin disease, transplantation, a Rheumatic disease, an inflammatory gastrointestinal disease, an inflammatory eye disease, an inflammatory neurological disease, an inflammatory pulmonary disease, an inflammatory endocrine disease, or an autoimmune hematological disease.
  • ANCA Antineutrophil cytoplasmic antibodies
  • the disease or condition is selected from inflammatory bowel disease, transplant, Crohn's disease, ulcerative colitis, multiple sclerosis, asthma, rheumatoid arthritis, or psoriasis.
  • FIG. 1 depicts impedance results reflecting cytotoxic killing activity of cells engineered with an anti-CD19 chimeric antigen receptor (CAR) alone or with an exemplary transmembrane immunomodulatory TIP (CD80-TIP or ICOSL-TIP) or the corresponding CD80 or ICOSL wild-type transmembrane protein following co-culture with target antigen-expressing cells. Impedance was assessed using the Acea Real-Time Cell Analyzer (RTCA), which measures the impedance variations in the culture media of a 96-well microelectronic plate (E-plate).
  • RTCA Acea Real-Time Cell Analyzer
  • FIG. 2A depicts that primary T cells are effectively transduced with viruses encoding both the CAR and TIP proteins.
  • Primary human T cells activated 48 hours with anti-CD3 plus anti-CD28 beads and were then tranduced with a Lenti-virus encoding an anti-CD19 CAR with a BFP reporter, plus a second Lenti-virus encoding and ICOSL TIP with a GFP reporter.
  • the FACs plot shows BFP expression on the y-axis and GFP expression on the x-axis and the percentage of T cells that fall into each quadrant are indicated.
  • Results show that the cultures include CAR only transduced cells (upper left quadrant), TIP only transduced cells (lower right quadrant), cells transduced with both viruses (upper right quadrant) and cells that were not transduced with either (lower left).
  • TIPs expressed on CAR-T cells provide costimulation to the CAR-T cells.
  • CAR-T cells with or without TIP co-transduction were labeled with Cell-Trace Far Red and incubated with the CD19+ NALM6 cell line to engage the CAR. Proliferation was assessed by the percentage of CAR-expressing cells that had diluted out the fluorescent dye.
  • FIG. 3A-3B demonstrate, via cytokine release, the costimulatory capacity of wild-type (WT) or variant ICOSL when coimmobilized with anti-CD3.
  • 10 nM anti-CD3 was wet coated to the wells of 96-well flat bottomed polystyrene tissue culture plates with 40 nM (arrows) or 10 nM WT or variant ICOSL. 100,000 purified CD4 + and CD8 + (pan) T-cells cells were added and supernatant was harvested 72 hours later for ELISA analysis for cytokine release.
  • FIG. 3A shows IFN-gamma and
  • FIG. 3B shows IL-17 protein levels secreted from pan T-cells.
  • Graphs are representative of typical IFN-gamma and IL-17 responses from pan T-cell costimulation.
  • FIG. 4A-4B demonstrate, via proliferation, the costimulatory capacity of wild-type (WT) or variant ICOSL when coimmobilized with anti-CD3.
  • CFSE-labeled pan T-cells were incubated in anti-CD3 and ICOSL coated plates as previously described for 72 hours. Cells were harvested, washed, stained with fluorescently conjugated anti-CD4 or anti-CD8 antibodies, and analyzed by flow cytometry. Gates and cytometer voltages were set using non-stimulated control CFSE-labeled T-cells. Proliferation was determined by CFSE dilution from control.
  • FIG. 4A shows percent of total proliferating (arrows), CD4 + (solid bar), and CD8 + cells (hatched bar) T-cells following 40 nM ICOSL costimulation.
  • FIG. 4B shows percent of total pan T-cell proliferation following 10 nM ICOSL costimulation. Graphs are representative of typical proliferative response from pan T-cell costimulation.
  • FIG. 5 depicts ICOSL vIgD candidate function in a human Mixed-Lymphocyte-Reation (MLR).
  • ICOSL variants and their mutations are listed on the x-axis, along with wild-type ICOSL, negative controls PDL2-Fc and human IgG, as well as the positive control benchmark molecule CTLA-Ig Belatacept.
  • the line across the graph represents the baseline amount of IFN-gamma detected in the supernatants of negative control cultures.
  • three different concentrations were tested with arrows indicating the highest concentration of protein in cultures at 40 nM.
  • the majority of ICOSL variant candidates show superior antagonistic activity at all three concentrations tested compared to belatacept as reflected by the lower concentration of IFN-gamma in those cultures.
  • FIG. 6A-6D depicts the inhibition of soluble ICOSL Fc-fusion proteins on B and T cell responses in a B-T co-culture assay.
  • FIG. 6A depicts soluble ICOSL Fc-fusion proteins inhibition of T cell-driven B cell proliferation.
  • Purified CD4+ T cells and B cells from a single donor were CFSE-labeled and co-incubated at a 1:1 ration in the presence or absence of the indicated mitogens with or without the indicated ICOSL Fc-fusion proteins.
  • Cells were stimulated with Staph enterotoxin B (SEB) at 100 ng/mL, Pokeweed mitogen (PWM) at 1 mg/mL, or both.
  • SEB Staph enterotoxin B
  • PWM Pokeweed mitogen
  • ICOSL Fc-fusion proteins were included at a final concentration of 40 nM and cultures were incubated for 7 days and subjected to FACS analysis. The number of divided B cells was determined from the number of cells in the cultures that had diluted their CFSE. All of the ICOSL Fc-fusion proteins tested except for wild-type reduced B cell proliferation.
  • FIG. 6B-6D show ICOSL Fc-fusion proteins inhibited cytokine T cell cytokine production in B-T co-cultures. Supernatants from the cultures described above were harvested on day 7 and analyzed for cytokine content using a LEGENDplex Human Th Cytokine Panel (Biolegend). T cell production of IL-5 ( FIG. 6B ), IL-13 ( FIG. 6C ) and IL-21 ( FIG. 6D ) is attenuated by inclusion of ICOSL Fc-fusion proteins.
  • FIG. 7A-7F depicts different endpoints in a mouse model of Graft Verse Host Disease (GVHD) where human PBMC cells were adoptively transferred into immunodeficient NSG murine hosts.
  • FIG. 7A shows survival curves of the treated animals. Aggressive disease course and subsequent mortality was observed in the saline control animals, with similar survival observed in the animals treated with wild-type ICOSL-Fc, as well as the N52H/I143T ICOSL variant. Variant N52H/N57Y/Q100P had improved survival rates comparable to the clinical benchmark belatacept.
  • FIG. 7A-7F shows survival curves of the treated animals. Aggressive disease course and subsequent mortality was observed in the saline control animals, with similar survival observed in the animals treated with wild-type ICOSL-Fc, as well as the N52H/I143T ICOSL variant. Variant N52H/N57Y/Q100P had improved survival rates comparable to the clinical benchmark belatacept.
  • FIG. 7B shows similar trends in body weight loss, with ICOSL variant N52H/N57Y/Q100P demonstrating similar weight maintenance as animals treated with belatacept, even though all other groups experienced rapid weight loss.
  • FIG. 7C shows clinical scores from standardized GVHD Disease Activity Index (DAI) observations, again showing lower scores in animals treated with the ICOSL variant N52H/N57Y/Q100P that are comparable to the clinical benchmark belatacept while the other groups of animals experienced higher DAI scores.
  • FIG. 7D depicts a flow cytometric measurement of CD4 and CD8 percentages in blood from experimental animals measured on day 14. The percentage of CD8 cells between experimental groups was largely the same, however, animals treated with ICOSL variant N52H/N57Y/Q100P and belatacept have lower percentages of CD4 cells compared to the other experimental groups.
  • FIG. 7E depicts survival curves from a similar experiment testing additional ICOSL variant molecules.
  • FIG. 7F depicts clinical scores from a similar experiment testing additional ICOSL variant molecules.
  • FIG. 8 shows localized costimulatory activity conveyed by the indicated variant stack molecule vIgD C-L, where C represents an ICOSL costimulatory domain and L represents a NKp30 localizing domain.
  • target K562 cells expressing the localizing surface protein, B7-H6, were cultured in the presence of anti-CD3 with human T cells and T cell activation was assessed by IFN-gamma levels in culture supernatants. Including anti-CD3 alone or no stack variant Fc molecules did not induce T cell activation. Similarly, cells cultured with only the wild-type localizing NKp30 domain alone or the wild-type costimulatory ICOSL domain alone as Fc fusion proteins did not result in T cell activation.
  • a stacked domain containing the wild-type version of both the costimulatory domain and localizing domain induced measurable IFN-gamma at the highest concentration tested, however, the variant localizing costimulatory stack induced greater than two fold higher IFN-gamma levels at the highest concentration, and IFN-gamma levels that were still observed as the concentrations were titrated down.
  • FIG. 9 summarizes changes in ear thickness in mice from a standard model of Delayed-Type Hypersensitivity (DTH).
  • DTH Delayed-Type Hypersensitivity
  • FIG. 10A-10C depicts various exemplary configurations of a variant IgSF domain (vIgD) conjugated to an antibody (V-Mab).
  • FIG. 10A shows various configurations in which a vIgD is linked, directly or indirectly, to the N- and/or C-terminus of the light chain of an antibody.
  • FIG. 10B shows various configurations in which a vIgD is linked, directly or indirectly, to the N- and/or C-terminus of the heavy chain of an antibody.
  • FIG. 10C depicts the resulting V-Mab configurations when a light chain of FIG. 10A and a heavy chain of FIG. 10B are co-expressed in a cell.
  • FIG. 11A-11B demonstrate V-Mab specificity for cognate binding partners. Binding assays were performed on Expi293 cells transiently transfected with DNA for mammalian surface expression of human HER2, CD28, CTLA-4, or ICOS. 200,000 transfected cells were incubated with 100,000 pM to 100 pM parental antibody (C1) or various V-Mabs (C2-9). Unbound antibody was removed, bound antibody detected with fluorescently conjugated anti-human IgG, and the cells were analyzed by flow cytometry for MFI and percentage positive based on Fc controls. FIG. 11A shows binding of the V-Mabs to HER2 transfectants at levels similar to the parental antibody.
  • FIG. 11B shows binding of the parental IgSF-domain (N52H/N57Y/Q100P) to its cognate partners is maintained or increased (C2, C3, C4, C5, C6, C8, C9) by V-Mabs.
  • FIG. 12 demonstrates V-Mab costimulatory and proliferative capacity when coimmobilized with anti-CD3.
  • 10 nM anti-CD3 was wet coated to the wells of 96-well flat bottomed polystyrene tissue culture plates with 30 nM to 3 nM parental antibody, V-Mabs, or Fc controls.
  • CFSE-labeled pan T-cells were added for 72 hours.
  • IFN-gamma secretion was measured by ELISA and total T-cell proliferation was measured by flow cytometric analysis of CFSE-dilution.
  • IFN-gamma secretion and proliferation of IgSF-domain (N52H/N57Y/Q100P) is greater than WT ICOSL.
  • V-Mabs demonstrate increased cytokine and proliferative costimulatory capacity over the parental IgSF.
  • FIG. 13A-13C depicts various formats of the provided variant IgSF domain molecules.
  • FIG. 13A depicts soluble molecules, including: (1) a variant IgSF domain (vIgD) fused to an Fc chain; (2) a stack molecule containing a first variant IgSF domain (first vIgD) and a second IgSF domain, such as a second variant IgSF domain (second vIgD); (3) a tumor targeting IgSF molecule containing a first variant IgSF domain (vIgD) and an IgSF domain that targets to a tumor antigen, such as an NKp30 IgSF domain; and (4) a variant IgSF domain (vIgD) linked to an antibody (V-Mab).
  • FIG. 13A depicts soluble molecules, including: (1) a variant IgSF domain (vIgD) fused to an Fc chain; (2) a stack molecule containing a first variant IgSF domain (first vIgD) and a second IgSF
  • FIG. 13B depicts a transmembrane immunomodulatory protein (TIP) containing a variant IgSF domain (vIgD), e.g., variant ICOSL, expressed on the surface of a cell.
  • TIP transmembrane immunomodulatory protein
  • vIgD variant IgSF domain
  • the cognate binding partner of the transmembrane bound vIgD is a costimulatory receptor, e.g. CD28
  • the TIP containing the vIgD e.g. ICOSL vIgD
  • FIG. 13C depicts a secreted immunomodulatory protein (SIP) in which a variant IgSF domain (vIgD), e.g., variant ICOSL, is secreted from a cell, such as a first T cell (e.g. CAR T cell).
  • vIgD variant IgSF domain
  • the cognate binding partner of the secreted vIgD is an activating receptor, e.g., CD28, which can be expressed on the first cell (e.g., T cell, such as a CAR T cell) and/or on a second cell (e.g. T cell; either endogenous or engineered, such as a CAR T cell).
  • the vIgD can be a V-domain (IgV) only, the combination of the V-domain (IgV) and C-domain (IgC), including the entire extracellular domain (ECD), or any combination of Ig domains of the IgSF superfamily member.
  • FIG. 14 depicts an exemplary schematic of the activity of a variant IgSF domain (vIgD) fused to an Fc (vIgD-Fc) in which the vIgD is a variant of an IgSF domain of ICOSL.
  • vIgD variant IgSF domain
  • Fc Fc
  • a soluble vIgD of ICOSL interacts with its cognate binding partners to block interactions of CD80 (B7-1)/CD86 (B7-2) or ICOSL with CD28 or ICOS, respectively, thereby blocking costimulation by the CD28 and/or ICOS costimulatory receptors.
  • FIG. 15 depicts an exemplary schematic of a stack molecule for localizing the variant IgSF domain (vIgD) to a tumor cell.
  • the stack molecule contains a first variant IgSF domain (first vIgD) and a second IgSF domain (e.g. a second vIgD) in which the second IgSF domain (e.g. a second vIgD) is a tumor-targeted IgSF domain that binds to a tumor antigen.
  • An exemplary tumor-targeted IgSF domain is an IgSF domain of NKp30, which binds to the tumor antigen B7-H6.
  • the vIgD is a variant of an IgSF domain of ICOSL.
  • binding of tumor-targeted IgSF domain to the surface of the tumor cell localizes the first vIgD on the tumor cell surface where it can interact with one or more of its cognate binding partners (e.g. CD28 or ICOS) expressed on the surface of an adjacent immune cell (e.g. T cell) to stimulate the costimulatory receptor.
  • cognate binding partners e.g. CD28 or ICOS
  • FIG. 16A depicts various exemplary configurations of a stack molecule containing a first variant IgSF domain (first vIgD), e.g. variant ICOSL, and a second IgSF domain, such as a second variant IgSF domain (second vIgD).
  • first vIgD first variant IgSF domain
  • second IgSF domain second variant IgSF domain
  • the first vIgD and second IgSF domain are independently linked, directly or indirectly, to the N- or C-terminus of an Fc region.
  • the Fc region is one that is capable of forming a homodimer with a matched Fc subunit by co-expression of the individual Fc regions in a cell.
  • the individual Fc regions contain mutations (e.g. “knob-into-hole” mutations in the CH3 domain), such that formation of the heterodimer is favored compared to homodimers when the individual Fc regions are co-expressed in a cell.
  • mutations e.g. “knob-into-hole” mutations in the CH3 domain
  • FIG. 16B depicts various exemplary configurations of a stack molecule containing a first variant IgSF domain (first vIgD), a second IgSF domain, such as a second variant IgSF domain (second vIgD), and a third IgSF domain, such as a third variant IgSF domain (third vIgD).
  • first vIgD, second IgSF, and third IgSF domains are independently linked, directly or indirectly, to the N- or C-terminus of an Fc region.
  • the Fc region is one that is capable of forming a homodimer with a matched Fc region by co-expression of the individual Fc regions in a cell.
  • FIG. 17 depicts an exemplary schematic of the activity of a variant IgSF domain (vIgD) conjugated to an antibody (V-Mab) in which the antibody (e.g. anti-HER2 antibody) binds to an antigen on the surface of the tumor cell.
  • the vIgD is a variant of an IgSF domain of ICOSL.
  • binding of the antibody to the surface of the tumor cell localizes the vIgD on the tumor cell surface where it can interact with one or more of its cognate binding partners expressed on the surface of an adjacent immune cell (e.g. T cell) to agonize receptor signaling.
  • an adjacent immune cell e.g. T cell
  • the variant IgSF domain is a variant of an IgSF domain of ICOSL. Binding of the ICOSL vIgD to CD28 or ICOS costimulatory receptors provides an agonist or costimulatory signal.
  • FIG. 18 depicts the Nanostring transcriptional signature of primary human T cells when incubated 10 nM anti-CD3 with 40 nM of an Fc-control protein, wild-type ICOSL-Fc, wild-type CD80-Fc, both of these proteins, or a variant ICOSL Fc-fusion proteins with mutations as indicated.
  • Total RNA from samples was prepared from harvested cells and the RNA was transferred to Nanostring and a Cancer Immune chip was used to quantitate transcripts of 750 gene in each sample. Altered transcripts include those whose level is above or below the diagonal line, including the noted transcripts.
  • FIG. 19 depicts transcript levels of exemplary transcripts upon incubation as described in FIG. 18 for the indicated times in the presence of the various immunomodulatory proteins.
  • FIG. 20A-20B demonstrates VmAb mediated T-cell proliferation when co-cultured with HER2 expressing targets.
  • CFSE-labeled pan T-cells were activated with K562-derived artificial target cells displaying cell surface anti-CD3 single chain Fv (OKT3) and HER2 in the presence of VmAbs or control proteins. Proliferation was measured by flow cytometric analysis of CFSE-dilution on CD4 + (left panel) or CD8 + (right panel) stained T-cells.
  • K562 cells were titrated and plated with T-cells for an effector:target (E:T) ratio of 40 to 1280:1.
  • VmAbs, parental IgSF domain, or WT ICOSL were added at 1000 pM.
  • K562 cells were added to T-cells for an E:T ratio of 160:1.
  • VmAbs or control proteins were titrated and added at 3000 to 37 pM.
  • FIG. 21 depicts the proliferation studies for T cells transduced with various IgSF domain-containing transmembrane immunomodulatory proteins (TIPs) and an exemplary recombinant E6-specific TCRs in primary human T cells.
  • TIPs IgSF domain-containing transmembrane immunomodulatory proteins
  • FIG. 22A-22G shows SEC analysis of proteolysis in variant ICOSL Fc-fusion molecules containing mutations N52H/N57Y/Q100R/F172S generated in various reference sequences, such as truncated ICOSL ECD Fc-fusion, an ICOSL IgV domain alone Fc-fusion, and/or ICOSL variant Fc fusion proteins with mutations at N207G/L208G with reference to the reference ICOSL extracellular domain (ECD) sequence set forth in SEQ ID NO:32. Molecules were expressed using ExpiCHO-S derived cells.
  • ECD extracellular domain
  • FIG. 23A-23B depicts the proliferation of CD4 and CD8 human T cells stimulated with K652 cells expressing variant ICOSL TIPs containing an ECD containing an affinity-modified IgSF with amino acid mutations corresponding to N52H/N57Y/Q100P (SEQ ID NO: 288), N52H/N57Y/Q100R (SEQ ID NO: 283), and E16V/N52H/N57Y/Q100R/V110D/H115R/Y152C/K156M/C198R (SEQ ID NO: 300).
  • FIG. 24A depicts binding of V-mAbs to HER2 and CD28.
  • FIG. 24B-24F depicts results of VmAb costimulation of T cells using a transfected cell system using Jurkat cells with an IL-2 promoter luciferase reporter, showing that V-mAbs provided a significant costimulatory signal in the presence of HER2+K562/OKT3 cells.
  • FIG. 25A-25D depicts binding of stack Fc-fusion molecules to cells expressing cognate binding partners B7H6 ( FIG. 25A ), ICOS ( FIG. 25B ), CD28 ( FIG. 25C ), and CTLA-4 ( FIG. 25D ).
  • FIG. 26A-26B depicts bioactivity studies for exemplary tested ICOSL/NKp30 stack proteins.
  • FIG. 27 depicts proliferation induced by ICOSL/NKp30 stack proteins as measured by flow cytometric analysis of CFSE-dilution on CD4+ or CD8+ stained T-cells.
  • FIG. 28 depicts anti-tumor effects of the combination of the tested ICOSL/NKp30 stack protein and mPD-1 mAb.
  • FIG. 29A-29E depicts anti-inflammatory activity of prophylactic dosing of the exemplary ICOSL IgV-Fc fusion molecule in the collagen-induced arthritis (CIA) model, including mean sum paw score ( FIG. 29A ), detected CII IgG ( FIG. 29B ), serum cytokine levels ( FIG. 29C ), CD44+ activated T cells or T FH cells ( FIG. 29D ), and fraction of B cells in the draining lymph node ( FIG. 29E ).
  • CIA collagen-induced arthritis
  • FIG. 30A-30D depicts anti-inflammatory activity of delayed dosing of the exemplary ICOSL IgV-Fc fusion molecule in the collagen-induced arthritis (CIA) model, including mean sum paw score ( FIG. 30A ) and serum cytokine levels ( FIG. 30C-30D ).
  • FIG. 31A-31D depicts anti-inflammatory activity of delayed dosing of the exemplary ICOSL IgV-Fc fusion molecule in the experimental autoimmune encephalomyelitis (EAE) model, including EAE score ( FIG. 31A ), flow cytometric analysis of inguinal lymph node T cells ( FIG. 31C ), and proinflammatory cytokines ( FIG. 31D ).
  • EAE experimental autoimmune encephalomyelitis
  • FIG. 32A-32B depicts survival and DAI score of Graft-versus-Host-Disease (GvHD) mice treated with various doses (20, 100, or 500 ⁇ g) of a variant ICOSL IgV-Fc molecule.
  • GvHD Graft-versus-Host-Disease
  • FIG. 33A-33F depicts results from flow cytometric analysis of Graft-versus-Host-Disease (GvHD) ratio of human cells/mouse cells in blood collected ( FIG. 33A ) or in total T cell count ( FIG. 33B ) at the end of the study, and assessment of ICOS+CD4+ or CD8+ cells ( FIG. 33C-33D ), or CD28+CD4+ or CD8+ cells ( FIG. 33E-33F ) from Graft-versus-Host-Disease (GvHD) mice treated with various doses (20, 100, or 500 ⁇ g) of a variant ICOSL IgV-Fc molecule.
  • GvHD Graft-versus-Host-Disease
  • FIG. 34A-34B depicts expression of activation or exhaustion markers of T cells from Graft-versus-Host-Disease (GvHD) mice treated with various doses (20, 100, or 500 ⁇ g) of a variant ICOSL IgV-Fc molecule.
  • GvHD Graft-versus-Host-Disease
  • FIG. 34C depicts the ratio of T effector cells (Teff) to T regulatory cells (Treg) from Graft-versus-Host-Disease (GvHD) mice treated with various doses (20, 100, or 500 ⁇ g) of a variant ICOSL IgV-Fc molecule.
  • FIG. 35A-35D depicts serum proinflammatory cytokines from Graft-versus-Host-Disease (GvHD) mice treated with various doses (20, 100, or 500 ⁇ g) of a variant ICOSL IgV-Fc molecule.
  • FIG. 35E depicts serum exposure of variant ICOSL IgV-Fc (N52H/N57Y/Q100R) in the GVHD model compared to normal mice.
  • FIG. 36A depicts DAI results and FIG. 36B depicts histology results from treatment with an exemplary variant ICOSL IgV-Fc on disease activity index (DAI) calculated from body weight and stool scores in a CD4+CD45RBhigh-induced colitis model.
  • DAI disease activity index
  • immunomodulatory proteins that are or comprise variants or mutants of ICOS ligand (ICOSL) or specific binding fragments thereof that exhibit activity to bind to at least one target ligand cognate binding partner (also called counter-structure protein).
  • the variant ICOSL polypeptides contain one or more amino acid modifications (e.g. amino acid substitutions, deletions or additions) compared to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the one or more amino acid modifications e.g. amino acid substitutions, deletions or additions
  • IgSF immunoglobulin superfamily
  • the variant ICOSL polypeptide exhibits altered, such as increased or decreased, binding activity or affinity for at least one cognate binding partner, such as at least one of ICOS, CD28, or CTLA-4.
  • the immunomodulatory proteins are soluble.
  • the immunomodulatory proteins are transmembrane immunomodulatory proteins capable of being expressed on the surface of cells.
  • also provided herein are one or more other immunomodulatory proteins that are conjugates or fusions containing a variant ICOSL polypeptide provided herein and one or more other moiety or polypeptide.
  • the variant ICOSL polypeptides and immunomodulatory proteins modulate an immunological immune response, such as an increased or decreased immune response.
  • the variant ICOSL polypeptides and immunomodulatory proteins provided herein can be used for the treatment of diseases or conditions that are associated with a dysregulated immune response.
  • the provided variant ICOSL polypeptides modulate T cell activation via interactions with costimulatory signaling molecules.
  • costimulatory signaling molecules In general, antigen specific T-cell activation requires two distinct signals. The first signal is provided by the interaction of the T-cell receptor (TCR) with major histocompatibility complex (MHC) associated antigens present on antigen presenting cells (APCs). The second signal is costimulatory to TCR engagement and necessary to avoid T-cell apoptosis or anergy.
  • TCR T-cell receptor
  • MHC major histocompatibility complex
  • the T cell-mediated immune response is initiated by antigen recognition by the T cell receptor (TCR) and is regulated by a balance of co-stimulatory and inhibitory signals (e.g., immune checkpoint receptors).
  • TCR T cell receptor
  • the immune system relies on immune checkpoint receptors to prevent autoimmunity (i.e., self-tolerance) and to protect tissues from excessive damage during an immune response, for example during an attack against a pathogenic infection.
  • these immunomodulatory proteins can be dysregulated in diseases and conditions, including tumors, as a mechanism for evading the immune system.
  • T-cell costimulatory receptors are known T-cell costimulatory receptors, which is the T-cell costimulatory receptor for the ligands B7-1 (CD80) and B7-2 (CD86) both of which are present on APCs.
  • CD28 is the T-cell costimulatory receptor for the ligands B7-1 (CD80) and B7-2 (CD86) both of which are present on APCs.
  • CTLA4 cytotoxic T-lymphocyte-associated protein 4
  • CTLA-4 acts to down-modulate the immune response.
  • ICOS inducible costimulator
  • ICOSL ICOS ligand
  • CD28 and CTLA-4 also are known to interact with ICOSL at a binding site that overlaps with the binding of ICOSL to the T-cell costimulatory receptor ICOS (Yao et al. (2011) Immunity, 34:729-740).
  • CD28 and ICOS are related CD28 family activating receptors and share some intracellular signaling motifs, costimulatory effects between CD28 and ICOS differ.
  • CD28 is expressed on both unactivated and activated T cells and its signaling is important for IL-2 production and subsequent T cell effector function.
  • ICOS is generally not expressed on the surface of T cells until after T cell activation, and signaling through ICOS on activated T cells supports specialized T cell subset differentiation.
  • costimulation by CD28 and ICOS yields overlapping and complementary effects.
  • T cells express the costimulatory molecules CD28 and ICOS, which interact with CD80/CD86 and ICOSL respectively, on antigen presenting cells (APC).
  • APC antigen presenting cells
  • professional APC i.e. dendritic cells, macrophages, and B cells
  • CD80, CD86, and ICOSL and engage CD28+/ICOS+ T cells.
  • activated T cells can then differentiate into effector cells such as CD8+ cytotoxic T cells (CTL), IL-17A/F-secreting CD4+Th17 cells, or CD4+ follicular helper (T FH ) cells.
  • CTL cytotoxic T cells
  • T FH CD4+ follicular helper
  • T FH -expressing CD40L engage B cells in lymphoid follicles and release cytokines (e.g. IL-21) inducing differentiation of B cells to antibody (Ab)-secreting plasma cells.
  • Plasma cells can produce tissue-damaging antibodies, e.g., rheumatoid factor (RF) and anti-citrullinated peptide antibodies (ACPA) in humans, and anti-collagen (CII) antibodies in mice, which can form immune complexes and deposits in the joints and other tissues.
  • ICOSL can also be expressed on non-professional APCs, leading to T cell activation in non-lymphoid tissues and further damage to the tissues and joints.
  • CD4+Th1-, Th9- and Th17-cells are implicated as key contributors to multiple sclerosis (MS) by increasing inflammation within the CNS in both multiple sclerosis and experimental autoimmune encephalomyelitis and CD4+ ICOS+CXCR5+T follicular helper cells are increased in PBMC in relapsing-remitting and correlate with disease progression in secondary progressive MS.
  • ICOS gene expression in cerebrospinal fluid cells, in secondary progressive MS, and an increased percentage of total monocytes and monocytes expressing ICOSL is observed.
  • ICOSL also expressed on non-professional APCs, leading to T cell activation in non-lymphoid tissues and further tissue damage
  • ICOSL polypeptide are polypeptides that, when modified by one or more amino acid modifications of an IgSF domain of a reference ICOSL polypeptide, exhibit enhanced binding affinity for CD28 and/or ICOS.
  • the overall increase in ICOS binding in provided variants is less than the increase in CD28 binding because wild-type ICOSL already demonstrates substantially more binding affinity for ICOS than CD28.
  • various formats of the provided variant polypeptides As shown herein, alternative formats can facilitate manipulation of the immune response, and hence the therapeutic application. For example, delivery of enhanced ICOSL proteins in soluble formats is shown herein to antagonize T cell activation by inhibiting CD28 and/or ICOS signaling.
  • tethering of the variant ICOSL molecules to a surface facilitates T cell activation by providing a costimulatory signal.
  • Various tethering strategies are provided to localize delivery of a T cell costimulatory signal including, but not limited to, direct coating to plastic, use of another variant IgSF domain to localize to a plate-bound or cell surface expressed protein target, or fusion of the variant ICOSL to a tumor-specific monoclonal antibody.
  • the modulation of immune signaling achieved by the provided variant ICOSL polypeptides and immunomodulatory polypeptides offers advantages for treatment of inflammatory and autoimmune disorders and other diseases and conditions compared to other treatments.
  • therapies to intervene and alter the costimulatory effects of both receptors are constrained by the spatial orientation requirements as well as size limitations imposed by the confines of the immunological synapse.
  • existing therapeutic drugs, including antibody drugs may not be able to interact simultaneously with the multiple target proteins involved in modulating these interactions.
  • existing therapeutic drugs may only have the ability to antagonize but not agonize an immune response. Additionally, pharmacokinetic differences between drugs that independently target one or the other of these two receptors can create difficulties in properly maintaining a desired blood concentration of such drug combinations throughout the course of treatment.
  • the provided variant ICOSL polypeptides or immunomodulatory proteins modulate (e.g. increase or decrease) immunological activity induced by costimulatory receptors CD28 or ICOS.
  • the provided polypeptides overcome these constraints by providing variant ICOSL (inducible costimulator ligand) with altered (e.g. increased or decreased) binding affinities to both CD28 and ICOS, and, in some cases, CTLA-4, thereby agonizing or antagonizing the complementary effects of costimulation by receptors. Methods of making and using these variant ICOSL are also provided.
  • the provided molecules may also be more effective than other soluble therapeutic protein agents.
  • abatacept CLA-4-Fc
  • CTLA-4-Fc abatacept
  • IL-4-Fc a variant CTLA-4-Fc molecule, for transplant rejection.
  • CTLA-4-Fc proteins bind to CD80 and CD86 and prevent these costimulatory ligands from engaging and triggering only CD28.
  • Variant ICOSL polypeptides provided herein exhibit binding affinity and enhanced activity for both CD28 and ICOS.
  • an immunomodulatory polypeptide comprising a variant ICOSL polypeptide is provided in a format, e.g. as an Fc-fusion protein, to antagonize or block activity of its cognate binding partner, e.g. ICOS and/or CD28.
  • blocking or inhibiting costimulatory signaling via CD28 or ICOS may be useful to suppresses an immune response, which can be useful in the treatment of inflammatory or autoimmune disorders (e.g., multiple sclerosis or brain inflammation), or organ transplantation.
  • tethering variant ICOSL proteins to a surface can deliver a localized costimulatory signal, which, in some aspects, can be used to target tumor tissue to deliver localized costimulation to tumor infiltrating T cells.
  • Most primary tumors lack expression of costimulatory molecules such as CD80, CD86 or ICOSL, and thus T cell anti-tumor responses can be compromised by a lack of costimulation (Yu et al. (1998) Int. Immunol.
  • T cell responses can be enhanced in the absence of tumor-expressed costimulatory proteins.
  • affinity modified as used in the context of an immunoglobulin superfamily domain, means a mammalian immunoglobulin superfamily (IgSF) domain having an altered amino acid sequence (relative to the corresponding wild-type parental or unmodified IgSF domain) such that it has an increased or decreased binding affinity or avidity to at least one of its cognate binding partners (alternatively “counter-structures”) compared to the parental wild-type or unmodified (i.e., non-affinity modified) IgSF control domain. Included in this context is an affinity modified ICOSL IgSF domain.
  • IgSF mammalian immunoglobulin superfamily
  • the affinity-modified IgSF domain can contain 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 or more amino acid differences, such as amino acid substitutions, in a reference (e.g., unmodified) or wild-type IgSF domain.
  • An increase or decrease in binding affinity or avidity can be determined using well known binding assays such as flow cytometry. Larsen et al., American Journal of Transplantation, Vol 5: 443-453 (2005). See also, Linsley et al., Immunity, Vol 1: 793-801 (1994).
  • An increase in a protein's binding affinity or avidity to its cognate binding partner(s) is to a value at least 10% greater than that of the wild-type IgSF domain control and in some embodiments, at least 20%, 30%, 40%, 50%, 100%, 200%, 300%, 500%, 1000%, 5000%, or 10000% greater than that of the wild-type IgSF domain control value.
  • a decrease in a protein's binding affinity or avidity to at least one of its cognate binding partner is to a value no greater than 90% of the control but no less than 10% of the wild-type IgSF domain control value, and in some embodiments no greater than 80%, 70% 60%, 50%, 40%, 30%, or 20% but no less than 10% of the wild-type IgSF domain control value.
  • affinity-modified protein is altered in primary amino acid sequence by substitution, addition, or deletion of amino acid residues.
  • affinity modified IgSF domain is not be construed as imposing any condition for any particular starting composition or method by which the affinity-modified IgSF domain was created.
  • the affinity modified IgSF domains of the present invention are not limited to wild type IgSF domains that are then transformed to an affinity modified IgSF domain by any particular process of affinity modification.
  • An affinity modified IgSF domain polypeptide can, for example, be generated starting from wild type mammalian IgSF domain sequence information, then modeled in silico for binding to its cognate binding partner, and finally recombinantly or chemically synthesized to yield the affinity modified IgSF domain composition of matter.
  • an affinity modified IgSF domain can be created by site-directed mutagenesis of a wild-type IgSF domain.
  • affinity modified IgSF domain denotes a product and not necessarily a product produced by any given process.
  • a variety of techniques including recombinant methods, chemical synthesis, or combinations thereof, may be employed.
  • allogeneic as used herein means a cell or tissue that is removed from one organism and then infused or adoptively transferred into a genetically dissimilar organism of the same species.
  • the species is murine or human.
  • autologous means a cell or tissue that is removed from the same organism to which it is later infused or adoptively transferred.
  • An autologous cell or tissue can be altered by, for example, recombinant DNA methodologies, such that it is no longer genetically identical to the native cell or native tissue which is removed from the organism.
  • a native autologous T-cell can be genetically engineered by recombinant DNA techniques to become an autologous engineered cell expressing a transmembrane immunomodulatory protein and/or chimeric antigen receptor (CAR), which in some cases involves engineering a T-cell or TIL (tumor infiltrating lymphocyte).
  • CAR transmembrane immunomodulatory protein and/or chimeric antigen receptor
  • the engineered cells are then infused into a patient from which the native T-cell was isolated.
  • the organism is human or murine.
  • binding affinity means the specific binding affinity and specific binding avidity, respectively, of a protein for its counter-structure under specific binding conditions.
  • avidity refers to the accumulated strength of multiple affinities of individual non-covalent binding interactions, such as between ICOSL and its counter-structures ICOS and/or CD28. As such, avidity is distinct from affinity, which describes the strength of a single interaction.
  • an increase or attenuation in binding affinity of a variant ICOSL containing an affinity modified ICOSL IgSF domain to its counter-structure is determined relative to the binding affinity of the unmodified ICOSL, such as an unmodified ICOSL containing the native or wild-type IgSF domain, such as IgV domain.
  • Methods for determining binding affinity or avidity are known in art. See, for example, Larsen et al., American Journal of Transplantation, Vol 5: 443-453 (2005).
  • a variant ICOSL of the invention i.e.
  • a ICOSL protein containing an affinity modified IgSF domain specifically binds to CD28 and/or ICOS measured by flow cytometry with a binding affinity that yields a Mean Fluorescence Intensity (MFI) value at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% greater than a wild-type ICOSL control in a binding assay such as described in Example 6.
  • MFI Mean Fluorescence Intensity
  • biological half-life refers to the amount of time it takes for a substance, such as an immunomodulatory polypeptide comprising a variant ICOSL of the present invention, to lose half of its pharmacologic or physiologic activity or concentration.
  • Biological half-life can be affected by elimination, excretion, degradation (e.g., enzymatic) of the substance, or absorption and concentration in certain organs or tissues of the body.
  • biological half-life can be assessed by determining the time it takes for the blood plasma concentration of the substance to reach half its steady state level (“plasma half-life”).
  • Conjugates that can be used to derivatize and increase the biological half-life of polypeptides of the invention are known in the art and include, but are not limited to, polyethylene glycol (PEG), hydroxyethyl starch (HES), XTEN (extended recombinant peptides; see, WO2013130683), human serum albumin (HSA), bovine serum albumin (BSA), lipids (acylation), and poly-Pro-Ala-Ser (PAS), polyglutamic acid (glutamylation).
  • PEG polyethylene glycol
  • HES hydroxyethyl starch
  • XTEN extended recombinant peptides
  • HSA human serum albumin
  • BSA bovine serum albumin
  • lipids acylation
  • PAS poly-Pro-Ala-Ser
  • chimeric antigen receptor refers to an artificial (i.e., man-made) transmembrane protein expressed on a mammalian cell comprising at least an ectodomain, a transmembrane, and an endodomain.
  • the CAR protein includes a “spacer” which covalently links the ectodomain to the transmembrane domain.
  • a spacer is often a polypeptide linking the ectodomain to the transmembrane domain via peptide bonds.
  • the CAR is typically expressed on a mammalian lymphocyte.
  • the CAR is expressed on a mammalian cell such as a T-cell or a tumor infiltrating lymphocyte (TIL).
  • TIL tumor infiltrating lymphocyte
  • a CAR expressed on a T-cell is referred to herein as a “CAR T-cell” or “CAR-T.”
  • the CAR-T is a T helper cell, a cytotoxic T-cell, a natural killer T-cell, a memory T-cell, a regulatory T-cell, or a gamma delta T-cell.
  • a CAR-T with antigen binding specificity to the patient's tumor is typically engineered to express on a native T-cell obtained from the patient.
  • the engineered T-cell expressing the CAR is then infused back into the patient.
  • the CAR-T is thus often an autologous CAR-T although allogeneic CAR-T are included within the scope of the invention.
  • the ectodomain of a CAR comprises an antigen binding region, such as an antibody or antigen binding fragment thereof (e.g. scFv), that specifically binds under physiological conditions with a target antigen, such as a tumor specific antigen.
  • a biochemical chain of events i.e., signal transduction results in modulation of the immunological activity of the CAR-T.
  • CD3-z CD3-zeta chain
  • CAR-Ts can further comprise multiple signaling domains such as CD28, 41BB or OX40, to further modulate immunomodulatory response of the T-cell.
  • CD3-z comprises a conserved motif known as an immunoreceptor tyrosine-based activation motif (ITAM) which is involved in T-cell receptor signal transduction.
  • ITAM immunoreceptor tyrosine-based activation motif
  • cytokine when used in reference to cytokine production induced by the presence of two or more variant ICOSL of the invention in an in vitro assay, means the overall cytokine expression level irrespective of the cytokine production induced by individual variant ICOSL.
  • the cytokine being assayed is IFN-gamma in an in vitro primary T-cell assay such as described in Example 6 and Example 7.
  • cognate binding partner in reference to a polypeptide, such as in reference to an IgSF domain of a variant ICOSL, refers to at least one molecule (typically a native mammalian protein) to which the referenced polypeptide specifically binds under specific binding conditions.
  • a variant ICOSL containing an affinity modified IgSF domain specifically binds to the counter-structure of the corresponding native or wild-type ICOSL but with increased or attenuated affinity.
  • a species of ligand recognized and specifically binding to its cognate receptor under specific binding conditions is an example of a counter-structure or cognate binding partner of that receptor.
  • a “cognate cell surface binding partner” is a cognate binding partner expressed on a mammalian cell surface.
  • a “cell surface molecular species” is a cognate binding partner of ligands of the immunological synapse (IS), expressed on and by cells, such as mammalian cells, forming the immunological synapse.
  • conjugate refers the joining or linking together of two or more compounds resulting in the formation of another compound, by any joining or linking methods known in the art. It can also refer to a compound which is generated by the joining or linking together two or more compounds.
  • a variant ICOSL polypeptide linked directly or indirectly to one or more chemical moieties or polypeptide is an exemplary conjugate.
  • conjugates include fusion proteins, those produced by chemical conjugates and those produced by any other methods.
  • competitive binding means that a protein is capable of specifically binding to at least two cognate binding partners but that specific binding of one cognate binding partner inhibits, such as prevents or precludes, simultaneous binding of the second cognate binding partner. Thus, in some cases, it is not possible for a protein to bind the two cognate binding partners at the same time. Generally, competitive binders contain the same or overlapping binding site for specific binding but this is not a requirement. In some embodiments, competitive binding causes a measurable inhibition (partial or complete) of specific binding of a protein to one of its cognate binding partner due to specific binding of a second cognate binding partner. A variety of methods are known to quantify competitive binding such as ELISA (enzyme linked immunosorbent assay) assays.
  • ELISA enzyme linked immunosorbent assay
  • conservative amino acid substitution means an amino acid substitution in which an amino acid residue is substituted by another amino acid residue having a side chain R group with similar chemical properties (e.g., charge or hydrophobicity).
  • groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine, and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartic acid and glutamic acid; and 7) sulfur-containing side chains: cysteine and methionine.
  • Conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine.
  • corresponding to with reference to positions of a protein, such as recitation that nucleotides or amino acid positions “correspond to” nucleotides or amino acid positions in a disclosed sequence, such as set forth in the Sequence listing, refers to nucleotides or amino acid positions identified upon alignment with the disclosed sequence based on structural sequence alignment or using a standard alignment algorithm, such as the GAP algorithm.
  • corresponding residues can be determined by alignment of a reference sequence with the sequence set forth in SEQ ID NO: 32 (ECD domain) or set forth in SEQ ID NOs: 196 or 545 (IgV domain) by structural alignment methods as described herein. By aligning the sequences, one skilled in the art can identify corresponding residues, for example, using conserved and identical amino acid residues as guides.
  • decrease or “attenuate” “or suppress” as used herein means to decrease by a statistically significant amount.
  • a decrease can be at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of a control value, such as a non-zero control value.
  • a decreased activity can refer to one or more of cell cycle inhibition, reduced cell survival, reduced cell proliferation, reduced cytokine production, or reduced T-cell cytotoxicity, such as by a statistically significant amount.
  • reference to reduced immunological activity means to reduce interferon gamma (IFN-gamma) production compared to in the absence of treatment, such as by a statistically significant amount.
  • IFN-gamma interferon gamma
  • the immunological activity can be assessed in a mixed lymphocyte reaction (MLR) assay.
  • MLR mixed lymphocyte reaction
  • Methods of conducting MLR assays are known in the art. Wang et al., Cancer Immunol Res. 2014 September: 2(9):846-56. Other methods of assessing activities of lymphocytes are known in the art, including any assay as described herein.
  • an enhancement can be a decrease by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, or 100%, as compared to a control value, such as an untreated control value or a non-zero control value.
  • derivatives or “derivatized” refer to modification of a protein by covalently linking it, directly or indirectly, to a composition so as to alter such characteristics as biological half-life, bioavailability, immunogenicity, solubility, toxicity, potency, or efficacy while retaining or enhancing its therapeutic benefit.
  • derivatives of immunomodulatory polypeptides of the invention are within the scope of the invention and can be made by, for example, glycosylation, pegylation, lipidation, or Fc-fusion.
  • domain refers to a portion of a molecule, such as a protein or encoding nucleic acid, that is structurally and/or functionally distinct from other portions of the molecule and is identifiable.
  • domains include those portions of a polypeptide chain that can form an independently folded structure within a protein made up of one or more structural motifs and/or that is recognized by virtue of a functional activity, such as binding activity.
  • a protein can have one, or more than one, distinct domains.
  • a domain can be identified, defined or distinguished by homology of the primary sequence or structure to related family members, such as homology to motifs.
  • a domain can be distinguished by its function, such as an ability to interact with a biomolecule, such as a cognate binding partner.
  • a domain independently can exhibit a biological function or activity such that the domain independently or fused to another molecule can perform an activity, such as, for example binding.
  • a domain can be a linear sequence of amino acids or a non-linear sequence of amino acids.
  • Many polypeptides contain a plurality of domains. Such domains are known, and can be identified by those of skill in the art. For exemplification herein, definitions are provided, but it is understood that it is well within the skill in the art to recognize particular domains by name. If needed appropriate software can be employed to identify domains.
  • ectodomain refers to the region of a membrane protein, such as a transmembrane protein, that lies outside the vesicular membrane. Ectodomains often comprise binding domains that specifically bind to ligands or cell surface receptors, such as via a binding domain that specifically binds to the ligand or cell surface receptor.
  • the ectodomain of a cellular transmembrane protein is alternately referred to as an extracellular domain.
  • an effective amount refers to a quantity and/or concentration of a therapeutic composition of the invention, including a protein composition or cell composition, that when administered ex vivo (by contact with a cell from a patient) or in vivo (by administration into a patient) either alone (i.e., as a monotherapy) or in combination with additional therapeutic agents, yields a statistically significant decrease in disease progression as, for example, by ameliorating or eliminating symptoms and/or the cause of the disease.
  • An effective amount may be an amount that relieves, lessens, or alleviates at least one symptom or biological response or effect associated with a disease or disorder, prevents progression of the disease or disorder, or improves physical functioning of the patient.
  • the effective amount is an effective dose or number of cells administered to a patient by adoptive cell therapy.
  • the patient is a mammal such as a non-human primate or human patient.
  • endodomain refers to the region found in some membrane proteins, such as transmembrane proteins, that extends into the interior space defined by the cell surface membrane.
  • the endodomain In mammalian cells, the endodomain is the cytoplasmic region of the membrane protein. In cells, the endodomain interacts with intracellular constituents and can be play a role in signal transduction and thus, in some cases, can be an intracellular signaling domain.
  • the endodomain of a cellular transmembrane protein is alternately referred to as a cytoplasmic domain, which, in some cases, can be a cytoplasmic signaling domain.
  • an increased activity can be one or more of increase cell survival, cell proliferation, cytokine production, or T-cell cytotoxicity, such as by a statistically significant amount.
  • reference to increased immunological activity means to increase interferon gamma (IFN-gamma) production, such as by a statistically significant amount.
  • IFN-gamma interferon gamma
  • the immunological activity can be assessed in a mixed lymphocyte reaction (MLR) assay.
  • MLR mixed lymphocyte reaction
  • Methods of conducting MLR assays are known in the art. Wang et al., Cancer Immunol Res. 2014 September: 2(9):846-56. Other methods of assessing activities of lymphocytes are known in the art, including any assay as described herein.
  • an enhancement can be an increase of at least 10%, 20%, 30%, 40%, 50%, 75%, 100%, 200%, 300%, 400%, or 500% greater than a non-zero control value.
  • engineered cell refers to a mammalian cell that has been genetically modified by human intervention such as by recombinant DNA methods or viral transduction.
  • the cell is an immune cell, such as a lymphocyte (e.g. T cell, B cell, NK cell) or an antigen presenting cell (e.g. dendritic cell).
  • the cell can be a primary cell from a patient or can be a cell line.
  • an engineered cell of the invention comprises a variant ICOSL provided herein.
  • the variant ICOSL is a transmembrane immunomodulatory protein (hereinafter referred to as “TIP”) that is expressed on the engineered cell.
  • TIP transmembrane immunomodulatory protein
  • the TIP contains the extracellular domain or a portion thereof containing the IgV domain linked to a transmembrane domain (e.g., a ICOSL transmembrane domain) and, optionally, an intracellular signaling domain.
  • the TIP is formatted as a chimeric receptor containing a heterologous cytoplasmic signaling domain or endodomain.
  • an engineered cell is capable of expressing and secreting a immunomodulatory protein as described herein.
  • provided engineered cells also are cells further containing an engineered T-cell receptor (TCR) or chimeric antigen receptor (CAR).
  • engineered T-cell refers to a T-cell such as a T helper cell, cytotoxic T-cell (alternatively, cytotoxic T lymphocyte or CTL), natural killer T-cell, regulatory T-cell, memory T-cell, or gamma delta T-cell, that has been genetically modified by human intervention such as by recombinant DNA methods or viral transduction methods.
  • An engineered T-cell comprises a variant ICOSL transmembrane immunomodulatory protein (TIP) or secreted immunodulatory protein (SIP) of the present invention that is expressed on the T-cell and is engineered to modulate immunological activity of the engineered T-cell itself, or a mammalian cell to which the variant ICOSL expressed on the T-cell specifically binds.
  • An engineered T-cell can comprise a variant ICOSL secreted immunomodulatory protein (SIP) of the present invention that is expressed by and/or secreted by the T-cell and is engineered to modulate immunological activity of the engineered T-cell itself, or a mammalian cell to which the variant ICOSL when secreted by the T-cell, specifically binds.
  • engineered T-cell receptor refers to a T-cell receptor (TCR) engineered to specifically bind with a desired affinity to a major histocompatibility complex (MHC)/peptide target antigen that is selected, cloned, and/or subsequently introduced into a population of T-cells, often used for adoptive immunotherapy.
  • MHC major histocompatibility complex
  • CARs are engineered to bind target antigens in a MHC independent manner.
  • the term “expressed on” as used herein is used in reference to a protein expressed on the surface of a cell, such as a mammalian cell.
  • the protein is expressed as a membrane protein.
  • the expressed protein is a transmembrane protein.
  • the protein is conjugated to a small molecule moiety such as a drug or detectable label.
  • Proteins expressed on the surface of a cell can include cell-surface proteins such as cell surface receptors that are expressed on mammalian cells.
  • half-life extending moiety refers to a moiety of a polypeptide fusion or chemical conjugate that extends the half-life of a protein circulating in mammalian blood serum compared to the half-life of the protein that is not so conjugated to the moiety. In some embodiments, half-life is extended by greater than or greater than about 1.2-fold, 1.5-fold, 2.0-fold, 3.0-fold, 4.0-fold, 5.0-fold, or 6.0-fold. In some embodiments, half-life is extended by more than 6 hours, more than 12 hours, more than 24 hours, more than 48 hours, more than 72 hours, more than 96 hours or more than 1 week after in vivo administration compared to the protein without the half-life extending moiety.
  • the half-life refers to the amount of time it takes for the protein to lose half of its concentration, amount, or activity.
  • Half-life can be determined for example, by using an ELISA assay or an activity assay.
  • Exemplary half-life extending moieties include an Fc domain, a multimerization domain, polyethylene glycol (PEG), hydroxyethyl starch (HES), XTEN (extended recombinant peptides; see, WO2013130683), human serum albumin (HSA), bovine serum albumin (BSA), lipids (acylation), and poly-Pro-Ala-Ser (PAS), and polyglutamic acid (glutamylation).
  • immunological synapse or “immune synapse” as used herein means the interface between a mammalian cell that expresses MHC I (major histocompatibility complex) or MHC II, such as an antigen-presenting cell or tumor cell, and a mammalian lymphocyte such as an effector T cell or Natural Killer (NK) cell.
  • MHC I major histocompatibility complex
  • MHC II such as an antigen-presenting cell or tumor cell
  • a mammalian lymphocyte such as an effector T cell or Natural Killer (NK) cell.
  • NK Natural Killer
  • an Fc (fragment crystallizable) region or domain of an immunoglobulin molecule corresponds largely to the constant region of the immunoglobulin heavy chain, and is responsible for various functions, including the antibody's effector function(s).
  • the Fc domain contains part or all of a hinge domain of an immunoglobulin molecule plus a CH2 and a CH3 domain.
  • the Fc domain can form a dimer of two polypeptide chains joined by one or more disulfide bonds.
  • the Fc is a variant Fc that exhibits reduced (e.g. reduced greater than 30%, 40%, 50%, 60%, 70%, 80%, 90% or more) activity to facilitate an effector function.
  • reference to amino acid substitutions in an Fc region is by EU numbering system unless described with reference to a specific SEQ ID NO. EU numbering is known and is according to the most recently updated IMGT Scientific Chart (IMGT®, the international ImMunoGeneTics information System® http://www.imgt.org/IMGTScientificChart/Numbering/Hu_IGHGnber.html (created: 17 May 2001, last updated: 10 Jan. 2013) and the EU index as reported in Kabat, E. A. et al. Sequences of Proteins of Immunological interest. 5th ed. US Department of Health and Human Services, NIH publication No. 91-3242 (1991).
  • An immunoglobulin Fc fusion such as an immunomodulatory Fc fusion protein, is a molecule comprising one or more polypeptides (or one or more small molecules) operably linked to an Fc region of an immunoglobulin.
  • An Fc-fusion may comprise, for example, the Fc region of an antibody (which facilitates effector functions and pharmacokinetics) and a variant ICOSL.
  • An immunoglobulin Fc region may be linked indirectly or directly to one or more variant ICOSL or small molecules (fusion partners).
  • Various linkers are known in the art and can optionally be used to link an Fc to a fusion partner to generate an Fc-fusion.
  • Fc-fusions of identical species can be dimerized to form Fc-fusion homodimers, or using non-identical species to form Fc-fusion heterodimers.
  • the Fc is a mammalian Fc such as a murine or human Fc.
  • host cell refers to a cell that can be used to express a protein encoded by a recombinant expression vector.
  • a host cell can be a prokaryote, for example, E. coli , or it can be a eukaryote, for example, a single-celled eukaryote (e.g., a yeast or other fungus), a plant cell (e.g., a tobacco or tomato plant cell), an animal cell (e.g., a human cell, a monkey cell, a hamster cell, a rat cell, a mouse cell, or an insect cell) or a hybridoma.
  • host cells examples include Chinese hamster ovary (CHO) cells or their derivatives such as Veggie CHO and related cell lines which grow in serum-free media or CHO strain DX-B11, which is deficient in DHFR.
  • a host cell is a mammalian cell (e.g., a human cell, a monkey cell, a hamster cell, a rat cell, a mouse cell, or an insect cell).
  • immunoglobulin refers to a mammalian immunoglobulin protein including any of the five human classes of antibody: IgA (which includes subclasses IgA1 and IgA2), IgD, IgE, IgG (which includes subclasses IgG1, IgG2, IgG3, and IgG4), and IgM.
  • immunoglobulins that are less than full-length, whether wholly or partially synthetic (e.g., recombinant or chemical synthesis) or naturally produced, such as antigen binding fragment (Fab), variable fragment (Fv) containing V H and V L , the single chain variable fragment (scFv) containing V H and V L linked together in one chain, as well as other antibody V region fragments, such as Fab′, F(ab) 2 , F(ab′) 2 , dsFv diabody, Fc, and Fd polypeptide fragments.
  • Fab′ antigen binding fragment
  • Fv variable fragment
  • scFv single chain variable fragment
  • bispecific antibodies homobispecific and heterobispecific, are included within the meaning of the term.
  • immunoglobulin superfamily or “IgSF” as used herein means the group of cell surface and soluble proteins that are involved in the recognition, binding, or adhesion processes of cells. Molecules are categorized as members of this superfamily based on shared structural features with immunoglobulins (i.e., antibodies); they all possess a domain known as an immunoglobulin domain or fold. Members of the IgSF include cell surface antigen receptors, co-receptors and co-stimulatory molecules of the immune system, molecules involved in antigen presentation to lymphocytes, cell adhesion molecules, certain cytokine receptors and intracellular muscle proteins. They are commonly associated with roles in the immune system. Proteins in the immunological synapse are often members of the IgSF. IgSF can also be classified into “subfamilies” based on shared properties such as function. Such subfamilies typically consist of from 4 to 30 IgSF members.
  • IgSF domain or “immunoglobulin domain” or “Ig domain” as used herein refers to a structural domain of IgSF proteins. Ig domains are named after the immunoglobulin molecules. They contain about 70-110 amino acids and are categorized according to their size and function. Ig-domains possess a characteristic Ig-fold, which has a sandwich-like structure formed by two sheets of antiparallel beta strands. Interactions between hydrophobic amino acids on the inner side of the sandwich and highly conserved disulfide bonds formed between cysteine residues in the B and F strands, stabilize the Ig-fold.
  • Ig domains One end of the Ig domain has a section called the complementarity determining region that is important for the specificity of antibodies for their ligands.
  • the Ig like domains can be classified (into classes) as: IgV, IgC1, IgC2, or IgI. Most Ig domains are either variable (IgV) or constant (IgC). IgV domains with 9 beta strands are generally longer than IgC domains with 7 beta strands. Ig domains of some members of the IgSF resemble IgV domains in the amino acid sequence, yet are similar in size to IgC domains. These are called IgC2 domains, while standard IgC domains are called IgC1 domains. T-cell receptor (TCR) chains contain two Ig domains in the extracellular portion; one IgV domain at the N-terminus and one IgC1 domain adjacent to the cell membrane. ICOSL contains two Ig domains: IgV and IgC.
  • IgSF species as used herein means an ensemble of IgSF member proteins with identical or substantially identical primary amino acid sequence.
  • Each mammalian immunoglobulin superfamily (IgSF) member defines a unique identity of all IgSF species that belong to that IgSF member.
  • each IgSF family member is unique from other IgSF family members and, accordingly, each species of a particular IgSF family member is unique from the species of another IgSF family member. Nevertheless, variation between molecules that are of the same IgSF species may occur owing to differences in post-translational modification such as glycosylation, phosphorylation, ubiquitination, nitrosylation, methylation, acetylation, and lipidation.
  • a “cell surface IgSF species” is an IgSF species expressed on the surface of a cell, generally a mammalian cell.
  • immunological activity refers to one or more cell survival, cell proliferation, cytokine production (e.g. interferon-gamma), or T-cell cytotoxicity activities.
  • an immunological activity can mean the cell expression of cytokines, such as chemokines or interleukins.
  • Assays for determining enhancement or suppression of immunological activity include the MLR (mixed lymphocyte reaction) assays measuring interferon-gamma cytokine levels in culture supernatants (Wang et al., Cancer Immunol Res.
  • An immunomodulatory protein such as a variant ICOSL polypeptide containing an affinity modified IgSF domain, as provided herein can in some embodiments increase or, in alternative embodiments, decrease IFN-gamma (interferon-gamma) expression in a primary T-cell assay relative to a wild-type IgSF member or IgSF domain control.
  • IFN-gamma interferon-gamma
  • a Mixed Lymphocyte Reaction (MLR) assay can be used as described in Example 6.
  • MLR Mixed Lymphocyte Reaction
  • a soluble form of an affinity modified IgSF domain of the invention can be employed to determine its ability to antagonize and thereby decrease the IFN-gamma expression in a MLR as likewise described in Example 6.
  • a co-immobilization assay can be used.
  • a T-cell receptor signal in some embodiments by anti-CD3 antibody, is used in conjunction with a co-immobilized affinity modified IgSF domain, such as variant ICOSL, to determine the ability to increase IFN-gamma expression relative to a wild-type IgSF domain control.
  • a co-immobilized affinity modified IgSF domain such as variant ICOSL
  • Methods to assay the immunological activity of engineered cells including to evaluate the activity of a variant ICOSL transmembrane immunomodulatory protein, are known in the art and include, but are not limited to, the ability to expand T cells following antigen stimulation, sustain T cell expansion in the absence of re-stimulation, and anti-cancer activities in appropriate animal models.
  • Assays also include assays to assess cytotoxicity, including a standard 51 Cr-release assay (see e.g. Milone et al., (2009) Molecular Therapy 17: 1453-1464) or flow based cytotoxicity assays, or an impedance based cytotoxicity assay (Peper et al. (2014) Journal of Immunological Methods, 405:192-198).
  • assays to assess cytotoxicity including a standard 51 Cr-release assay (see e.g. Milone et al., (2009) Molecular Therapy 17: 1453-1464) or flow based cytotoxicity assays, or an impedance based cytotoxicity assay (Peper et al. (2014) Journal of Immunological Methods, 405:192-198).
  • an “immunomodulatory polypeptide” or “immunomodulatory protein” is a polypeptide or protein molecule that modulates immunological activity. By “modulation” or “modulating” an immune response is meant that immunological activity is either increased or decreased.
  • An immunomodulatory protein can be a single polypeptide chain or a multimer (dimers or higher order multimers) of at least two polypeptide chains covalently bonded to each other by, for example, interchain disulfide bonds. Thus, monomeric, dimeric, and higher order multimeric polypeptides are within the scope of the defined term. Multimeric polypeptides can be homomultimeric (of identical polypeptide chains) or heteromultimeric (of non-identical polypeptide chains).
  • An immunomodulatory protein of the invention comprises a variant ICOSL.
  • increase means to increase by a statistically significant amount. An increase can be at least 5%, 10%, 20%, 30%, 40%, 50%, 75%, 100%, or greater than a non-zero control value.
  • an “isoform” of ICOSL is one of a plurality of naturally occurring ICOSL polypeptides that differ in amino acid sequence. Isoforms can be the product of splice variants of an RNA transcript expressed by a single gene, or the expression product of highly similar but different genes yielding a functionally similar protein such as may occur from gene duplication. As used herein, the term “isoform” of ICOSL also refers to the product of different alleles of an ICOSL gene (e.g., ICOSLG).
  • lymphocyte means any of three subtypes of white blood cell in a mammalian immune system. They include natural killer cells (NK cells) (which function in cell-mediated, cytotoxic innate immunity), T cells (for cell-mediated, cytotoxic adaptive immunity), and B cells (for humoral, antibody-driven adaptive immunity). T cells include: T helper cells, cytotoxic T-cells, natural killer T-cells, memory T-cells, regulatory T-cells, or gamma delta T-cells. Innate lymphoid cells (ILC) are also included within the definition of lymphocyte.
  • NK cells natural killer cells
  • T cells for cell-mediated, cytotoxic adaptive immunity
  • B cells for humoral, antibody-driven adaptive immunity
  • T cells include: T helper cells, cytotoxic T-cells, natural killer T-cells, memory T-cells, regulatory T-cells, or gamma delta T-cells.
  • ILC Innate lymphoid cells
  • mammal specifically includes reference to at least one of a: human, chimpanzee, rhesus monkey, cynomolgus monkey, dog, cat, mouse, or rat.
  • membrane protein as used herein means a protein that, under physiological conditions, is attached directly or indirectly to a lipid bilayer.
  • a lipid bilayer that forms a membrane can be a biological membrane such as a eukaryotic (e.g., mammalian) cell membrane or an artificial (i.e., man-made) membrane such as that found on a liposome. Attachment of a membrane protein to the lipid bilayer can be by way of covalent attachment, or by way of non-covalent interactions such as hydrophobic or electrostatic interactions.
  • a membrane protein can be an integral membrane protein or a peripheral membrane protein. Membrane proteins that are peripheral membrane proteins are non-covalently attached to the lipid bilayer or non-covalently attached to an integral membrane protein.
  • a peripheral membrane protein forms a temporary attachment to the lipid bilayer such that under the range of conditions that are physiological in a mammal, peripheral membrane protein can associate and/or disassociate from the lipid bilayer.
  • integral membrane proteins form a substantially permanent attachment to the membrane's lipid bilayer such that under the range of conditions that are physiological in a mammal, integral membrane proteins do not disassociate from their attachment to the lipid bilayer.
  • a membrane protein can form an attachment to the membrane by way of one layer of the lipid bilayer (monotopic), or attached by way of both layers of the membrane (polytopic).
  • An integral membrane protein that interacts with only one lipid bilayer is an “integral monotopic protein”.
  • An integral membrane protein that interacts with both lipid bilayers is an “integral polytopic protein” alternatively referred to herein as a “transmembrane protein”.
  • modulating or “modulate” as used herein in the context of an immune response, such as a mammalian immune response, refer to any alteration, such as an increase or a decrease, of existing or potential immune responses that occurs as a result of administration of an immunomodulatory polypeptide comprising a variant ICOSL of the present invention or as a result of administration of engineered cells expresses an immunomodulatory protein, such as a variant ICOSL transmembrane immunomodulatory protein of the present invention.
  • an alteration such as an increase or decrease, of an immune response as compared to the immune response that occurs or is present in the absence of the administration of the immunomodulatory protein comprising the variant ICOSL or cells expressing such an immunomodulatory polypeptide.
  • Such modulation includes any induction, activation, suppression or alteration in degree or extent of immunological activity of an immune cell.
  • Immune cells include B cells, T cells, NK (natural killer) cells, NK T cells, professional antigen-presenting cells (APCs), and non-professional antigen-presenting cells, and inflammatory cells (neutrophils, macrophages, monocytes, eosinophils, and basophils).
  • Modulation includes any change imparted on an existing immune response, a developing immune response, a potential immune response, or the capacity to induce, regulate, influence, or respond to an immune response.
  • Modulation includes any alteration in the expression and/or function of genes, proteins and/or other molecules in immune cells as part of an immune response.
  • Modulation of an immune response or modulation of immunological activity includes, for example, the following: elimination, deletion, or sequestration of immune cells; induction or generation of immune cells that can modulate the functional capacity of other cells such as autoreactive lymphocytes, antigen presenting cells, or inflammatory cells; induction of an unresponsive state in immune cells (i.e., anergy); enhancing or suppressing the activity or function of immune cells, including but not limited to altering the pattern of proteins expressed by these cells. Examples include altered production and/or secretion of certain classes of molecules such as cytokines, chemokines, growth factors, transcription factors, kinases, costimulatory molecules, or other cell surface receptors or any combination of these modulatory events.
  • Modulation can be assessed, for example, by an alteration in IFN-gamma (interferon gamma) expression relative to the wild-type ICOSL control in a primary T cell assay (see, Zhao and Ji, Exp Cell Res. 2016 Jan. 1; 340(1) 132-138). Modulation can be assessed, for example, by an alteration of an immunological activity of engineered cells, such as an alteration in cytotoxic activity of engineered cells or an alteration in cytokine secretion of engineered cells relative to cells engineered with a wild-type ICOSL transmembrane protein.
  • molecular species as used herein means an ensemble of proteins with identical or substantially identical primary amino acid sequence.
  • Each mammalian immunoglobulin superfamily (IgSF) member defines a collection of identical or substantially identical molecular species.
  • IgSF immunoglobulin superfamily
  • human ICOSL is an IgSF member and each human ICOSL molecule is a molecule species of ICOS.
  • Variation between molecules that are of the same molecular species may occur owing to differences in post-translational modification such as glycosylation, phosphorylation, ubiquitination, nitrosylation, methylation, acetylation, and lipidation.
  • a “cell surface molecular species” is a molecular species expressed on the surface of a mammalian cell. Two or more different species of protein, each of which is present exclusively on one or exclusively the other (but not both) of the two mammalian cells forming the IS, are said to be in “cis” or “cis configuration” with each other.
  • Two different species of protein are said to be in “trans” or “trans configuration.”
  • Two different species of protein each of which is present on both of the two mammalian cells forming the IS are in both cis and trans configurations on these cells.
  • a “multimerization domain” refers to a sequence of amino acids that promotes stable interaction of a polypeptide molecule with one or more additional polypeptide molecules, each containing a complementary multimerization domain (e.g. a first multimerization domain and a second multimerization domain), which can be the same or a different multimerization domain.
  • the interactions between complementary multimerization domains e.g. interaction between a first multimerization domain and a second multimerization domain, form a stable protein-protein interaction to produce a multimer of the polypeptide molecule with the additional polypeptide molecule.
  • the multimerization domain is the same and interacts with itself to form a stable protein-protein interaction between two polypeptide chains.
  • a polypeptide is joined directly or indirectly to the multimerization domain.
  • multimerization domains include the immunoglobulin sequences or portions thereof, leucine zippers, hydrophobic regions, hydrophilic regions, and compatible protein-protein interaction domains.
  • the multimerization domain can be an immunoglobulin constant region or domain, such as, for example, the Fc domain or portions thereof from IgG, including IgG1, IgG2, IgG3 or IgG4 subtypes, IgA, IgE, IgD and IgM and modified forms thereof.
  • nucleic acid and “polynucleotide” are used interchangeably to refer to a polymer of nucleic acid residues (e.g., deoxyribonucleotides or ribonucleotides) in either single- or double-stranded form. Unless specifically limited, the terms encompass nucleic acids containing known analogues of natural nucleotides and that have similar binding properties to it and are metabolized in a manner similar to naturally-occurring nucleotides.
  • nucleic acid or polynucleotide encompasses cDNA or mRNA encoded by a gene.
  • non-competitive binding means the ability of a protein to specifically bind simultaneously to at least two cognate binding partners.
  • the protein is able to bind to at least two different cognate binding partners at the same time, although the binding interaction need not be for the same duration such that, in some cases, the protein is specifically bound to only one of the cognate binding partners.
  • the binding occurs under specific binding conditions.
  • the simultaneous binding is such that binding of one cognate binding partner does not substantially inhibit simultaneous binding to a second cognate binding partner.
  • non-competitive binding means that binding a second cognate binding partner to its binding site on the protein does not displace the binding of a first cognate binding partner to its binding site on the protein.
  • the first cognate binding partner specifically binds at an interaction site that does not overlap with the interaction site of the second cognate binding partner such that binding of the second cognate binding partner does not directly interfere with the binding of the first cognate binding partner.
  • any effect on binding of the cognate binding partner by the binding of the second cognate binding partner is through a mechanism other than direct interference with the binding of the first cognate binding partner.
  • Non-competitive inhibitor binds to a site other than the active site of the enzyme.
  • Non-competitive binding encompasses uncompetitive binding interactions in which a second cognate binding partner specifically binds at an interaction site that does not overlap with the binding of the first cognate binding partner but binds to the second interaction site only when the first interaction site is occupied by the first cognate binding partner.
  • composition refers to a composition suitable for pharmaceutical use in a mammalian subject, often a human.
  • a pharmaceutical composition typically comprises an effective amount of an active agent (e.g., an immunomodulatory polypeptide comprising a variant ICOSL or engineered cells expressing a variant ICOSL transmembrane immunomodulatory protein) and a carrier, excipient, or diluent.
  • an active agent e.g., an immunomodulatory polypeptide comprising a variant ICOSL or engineered cells expressing a variant ICOSL transmembrane immunomodulatory protein
  • carrier, excipient, or diluent is typically a pharmaceutically acceptable carrier, excipient or diluent, respectively.
  • polypeptide and protein are used interchangeably herein and refer to a molecular chain of two or more amino acids linked through peptide bonds. The terms do not refer to a specific length of the product. Thus, “peptides,” and “oligopeptides,” are included within the definition of polypeptide.
  • the terms include post-translational modifications of the polypeptide, for example, glycosylations, acetylations, phosphorylations and the like.
  • the terms also include molecules in which one or more amino acid analogs or non-canonical or unnatural amino acids are included as can be synthesized, or expressed recombinantly using known protein engineering techniques. In addition, proteins can be derivatized.
  • primary T-cell assay refers to an in vitro assay to measure interferon-gamma (“IFN-gamma”) expression.
  • IFN-gamma interferon-gamma
  • the assay used is an anti-CD3 coimmobilizaton assay.
  • primary T cells are stimulated by anti-CD3 immobilized with or without additional recombinant proteins.
  • Culture supernatants are harvested at timepoints, usually 24-72 hours.
  • the assay used is the MLR.
  • primary T cells are stimulated with allogeneic APC. Culture supernatants are harvested at timepoints, usually 24-72 hours. Human IFN-gamma levels are measured in culture supernatants by standard ELISA techniques. Commercial kits are available from vendors and the assay is performed according to manufacturer's recommendation.
  • nucleic acids such as encoding immunomodulatory proteins of the invention
  • purified as applied to nucleic acids, such as encoding immunomodulatory proteins of the invention, generally denotes a nucleic acid or polypeptide that is substantially free from other components as determined by analytical techniques well known in the art (e.g., a purified polypeptide or polynucleotide forms a discrete band in an electrophoretic gel, chromatographic eluate, and/or a media subjected to density gradient centrifugation).
  • nucleic acid or polypeptide that gives rise to essentially one band in an electrophoretic gel is “purified.”
  • a purified nucleic acid or protein of the invention is at least about 50% pure, usually at least about 75%, 80%, 85%, 90%, 95%, 96%, 99% or more pure (e.g., percent by weight or on a molar basis).
  • recombinant indicates that the material (e.g., a nucleic acid or a polypeptide) has been artificially (i.e., non-naturally) altered by human intervention. The alteration can be performed on the material within, or removed from, its natural environment or state.
  • a “recombinant nucleic acid” is one that is made by recombining nucleic acids, e.g., during cloning, affinity modification, DNA shuffling or other well-known molecular biological procedures.
  • a “recombinant DNA molecule,” is comprised of segments of DNA joined together by means of such molecular biological techniques.
  • recombinant protein or “recombinant polypeptide” as used herein refers to a protein molecule which is expressed using a recombinant DNA molecule.
  • a “recombinant host cell” is a cell that contains and/or expresses a recombinant nucleic acid or that is otherwise altered by genetic engineering, such as by introducing into the cell a nucleic acid molecule encoding a recombinant protein, such as a transmembrane immunomodulatory protein provided herein.
  • Transcriptional control signals in eukaryotes comprise “promoter” and “enhancer” elements. Promoters and enhancers consist of short arrays of DNA sequences that interact specifically with cellular proteins involved in transcription.
  • Promoter and enhancer elements have been isolated from a variety of eukaryotic sources including genes in yeast, insect and mammalian cells and viruses (analogous control elements, i.e., promoters, are also found in prokaryotes). The selection of a particular promoter and enhancer depends on what cell type is to be used to express the protein of interest.
  • the terms “in operable combination,” “in operable order” and “operably linked” as used herein refer to the linkage of nucleic acid sequences in such a manner or orientation that a nucleic acid molecule capable of directing the transcription of a given gene and/or the synthesis of a desired protein molecule is produced.
  • recombinant expression vector refers to a DNA molecule containing a desired coding sequence and appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequence in a particular host cell.
  • Nucleic acid sequences necessary for expression in prokaryotes include a promoter, optionally an operator sequence, a ribosome binding site and possibly other sequences.
  • Eukaryotic cells are known to utilize promoters, enhancers, and termination and polyadenylation signals.
  • a secretory signal peptide sequence can also, optionally, be encoded by the recombinant expression vector, operably linked to the coding sequence for the recombinant protein, such as a recombinant fusion protein, so that the expressed fusion protein can be secreted by the recombinant host cell, for easier isolation of the fusion protein from the cell, if desired.
  • the term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • the vectors are viral vectors, such as lentiviral vectors.
  • selectivity refers to the preference of a subject protein, or polypeptide, for specific binding of one substrate, such as one cognate binding partner, compared to specific binding for another substrate, such as a different cognate binding partner of the subject protein. Selectivity can be reflected as a ratio of the binding activity (e.g. binding affinity) of a subject protein and a first substrate, such as a first cognate binding partner, (e.g., K d1 ) and the binding activity (e.g. binding affinity) of the same subject protein with a second cognate binding partner (e.g., K d2 ).
  • a first cognate binding partner e.g., K d1
  • second cognate binding partner e.g., K d2
  • sequence identity refers to the sequence identity between genes or proteins at the nucleotide or amino acid level, respectively. “Sequence identity” is a measure of identity between proteins at the amino acid level and a measure of identity between nucleic acids at nucleotide level.
  • the protein sequence identity may be determined by comparing the amino acid sequence in a given position in each sequence when the sequences are aligned.
  • the nucleic acid sequence identity may be determined by comparing the nucleotide sequence in a given position in each sequence when the sequences are aligned. Methods for the alignment of sequences for comparison are well known in the art, such methods include GAP, BESTFIT, BLAST, FASTA and TFASTA.
  • the BLAST algorithm calculates percent sequence identity and performs a statistical analysis of the similarity between the two sequences.
  • the software for performing BLAST analysis is publicly available through the National Center for Biotechnology Information (NCBI) website.
  • soluble as used herein in reference to proteins, means that the protein is not a membrane protein.
  • a soluble protein contains only the extracellular domain of an IgSF family member receptor, or a portion thereof containing an IgSF domain or domains or specific-binding fragments thereof, but does not contain the transmembrane domain and/or is not capable of being expressed on the surface of a cell.
  • solubility of a protein can be improved by linkage or attachment, directly or indirectly via a linker, to an Fc domain, which, in some cases, also can improve the stability and/or half-life of the protein.
  • a soluble protein is an Fc fusion protein.
  • proteins as used herein with respect to polypeptides or nucleic acids means an ensemble of molecules with identical or substantially identical sequences. Variation between polypeptides that are of the same species may occur owing to differences in post-translational modification such as glycosylation, phosphorylation, ubiquitination, nitrosylation, methylation, acetylation, and lipidation. Slightly truncated sequences of polypeptides that differ (or encode a difference) from the full length species at the amino-terminus or carboxy-terminus by no more than 1, 2, or 3 amino acid residues are considered to be of a single species. Such microheterogeneities are a common feature of manufactured proteins.
  • specific binding fragment as used herein in reference to a full-length wild-type mammalian ICOSL polypeptide or an IgV or an IgC domain thereof, means a polypeptide having a subsequence of an IgV and/or IgC domain and that specifically binds in vitro and/or in vivo to a mammalian ICOS and/or mammalian CD28 such as a human or murine ICOS or CD28.
  • the specific binding fragment of ICOSL IgV or ICOSL IgC is at at least 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% the sequence length of the full-length wild-type sequence.
  • the specific binding fragment can be altered in sequence to form a variant ICOSL of the invention.
  • specifically binds means the ability of a protein, under specific binding conditions, to bind to a target protein such that its affinity or avidity is at least 5 times as great, but optionally at least 10, 20, 30, 40, 50, 100, 250 or 500 times as great, or even at least 1000 times as great as the average affinity or avidity of the same protein to a collection of random peptides or polypeptides of sufficient statistical size.
  • a specifically binding protein need not bind exclusively to a single target molecule but may specifically bind to a non-target molecule due to similarity in structural conformation between the target and non-target (e.g., paralogs or orthologs).
  • a polypeptide of the invention may specifically bind to more than one distinct species of target molecule due to cross-reactivity.
  • Solid-phase ELISA immunoassays or Biacore measurements can be used to determine specific binding between two proteins.
  • interactions between two binding proteins have dissociation constants (K d ) less than 1 ⁇ 10 ⁇ 5 M, and often as low as 1 ⁇ 10 ⁇ 12 M.
  • interactions between two binding proteins have dissociation constants of 1 ⁇ 10 ⁇ 6 M, 1 ⁇ 10 ⁇ 7 M, 1 ⁇ 10 ⁇ 8 M, 1 ⁇ 10 ⁇ 9 M, 1 ⁇ 10 ⁇ 10 M or 1 ⁇ 10 ⁇ 11 M.
  • surface expresses”, “surface expression” or “expressed on the surface” in reference to a mammalian cell expressing a polypeptide means that the polypeptide is expressed as a membrane protein.
  • the membrane protein is a transmembrane protein.
  • synthetic with reference to, for example, a synthetic nucleic acid molecule or a synthetic gene or a synthetic peptide refers to a nucleic acid molecule or polypeptide molecule that is produced by recombinant methods and/or by chemical synthesis methods.
  • targeting moiety refers to a composition that is covalently or non-covalently attached to, or physically encapsulates, a polypeptide comprising a variant ICOSL of the present invention.
  • the targeting moiety has specific binding affinity for a target molecule such as a target molecule expressed on a cell.
  • the target molecule is localized on a specific tissue or cell-type.
  • Targeting moieties include: antibodies, antigen binding fragment (Fab), variable fragment (Fv) containing V H and V L , the single chain variable fragment (scFv) containing V H and V L linked together in one chain, as well as other antibody V region fragments, such as Fab′, F(ab) 2 , F(ab′) 2 , dsFv diabody, nanobodies, soluble receptors, receptor ligands, affinity matured receptors or ligands, as well as small molecule ( ⁇ 500 dalton) compositions (e.g., specific binding receptor compositions).
  • Targeting moieties can also be attached covalently or non-covalently to the lipid membrane of liposomes that encapsulate a polypeptide of the present invention.
  • transmembrane protein as used herein means a membrane protein that substantially or completely spans a lipid bilayer such as those lipid bilayers found in a biological membrane such as a mammalian cell, or in an artificial construct such as a liposome.
  • the transmembrane protein comprises a transmembrane domain (“transmembrane domain”) by which it is integrated into the lipid bilayer and by which the integration is thermodynamically stable under physiological conditions.
  • Transmembrane domains are generally predictable from their amino acid sequence via any number of commercially available bioinformatics software applications on the basis of their elevated hydrophobicity relative to regions of the protein that interact with aqueous environments (e.g., cytosol, extracellular fluid).
  • a transmembrane domain is often a hydrophobic alpha helix that spans the membrane.
  • a transmembrane protein can pass through the both layers of the lipid bilayer once or multiple times.
  • a transmembrane protein includes the provided transmembrane immunomodulatory proteins described herein.
  • a transmembrane immunomodulatory protein of the invention further comprises an ectodomain and, in some embodiments, an endodomain.
  • treating means slowing, stopping or reversing the disease or disorders progression, as evidenced by decreasing, cessation or elimination of either clinical or diagnostic symptoms, by administration of a therapeutic composition (e.g. containing an immunomodulatory protein or engineered cells) of the invention either alone or in combination with another compound as described herein.
  • a therapeutic composition e.g. containing an immunomodulatory protein or engineered cells
  • Treatment also means a decrease in the severity of symptoms in an acute or chronic disease or disorder or a decrease in the relapse rate as for example in the case of a relapsing or remitting autoimmune disease course or a decrease in inflammation in the case of an inflammatory aspect of an autoimmune disease.
  • the terms “treatment” or, “inhibit,” “inhibiting” or “inhibition” of cancer refers to at least one of: a statistically significant decrease in the rate of tumor growth, a cessation of tumor growth, or a reduction in the size, mass, metabolic activity, or volume of the tumor, as measured by standard criteria such as, but not limited to, the Response Evaluation Criteria for Solid Tumors (RECIST), or a statistically significant increase in progression free survival (PFS) or overall survival (OS).
  • RECIST Response Evaluation Criteria for Solid Tumors
  • PFS progression free survival
  • OS overall survival
  • Preventing,” “prophylaxis,” or “prevention” of a disease or disorder as used in the context of this invention refers to the administration of an immunomodulatory polypeptide or engineered cells of the invention, either alone or in combination with another compound, to prevent the occurrence or onset of a disease or disorder or some or all of the symptoms of a disease or disorder or to lessen the likelihood of the onset of a disease or disorder.
  • tumor specific antigen refers to a counter-structure that is present primarily on tumor cells of a mammalian subject but generally not found on normal cells of the mammalian subject.
  • a tumor specific antigen need not be exclusive to tumor cells but the percentage of cells of a particular mammal that have the tumor specific antigen is sufficiently high or the levels of the tumor specific antigen on the surface of the tumor are sufficiently high such that it can be targeted by anti-tumor therapeutics, such as immunomodulatory polypeptides of the invention, and provide prevention or treatment of the mammal from the effects of the tumor.
  • At least 50% of the cells displaying a TSA are cancerous. In other embodiments, at least 60%, 70%, 80%, 85%, 90%, 95%, or 99% of the cells displaying a TSA are cancerous.
  • variant as used in reference to a variant ICOSL means an ICOSL, such as a mammalian (e.g., human or murine) ICOSL created by human intervention.
  • the variant ICOSL is a polypeptide having an altered amino acid sequence, relative to a reference (e.g. unmodified) or wild-type ICOSL.
  • the variant ICOSL is a polypeptide which differs from a reference ICOSL, such as a wild-type ICOSL isoform sequence, by one or more modifications, such as one or more amino acid substitutions, deletions, additions, or combinations thereof.
  • the variant ICOSL contains at least one affinity modified domain, whereby one or more of the amino acid differences occurs in an IgSF domain (e.g. IgV domain).
  • IgSF domain e.g. IgV domain
  • a variant ICOSL can contain 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 or more amino acid differences, such as amino acid substitutions.
  • a variant ICOSL polypeptide generally exhibits at least 50%, 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to a corresponding reference (e.g.
  • a variant ICOSL polypeptide exhibits at least 50%, 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to a corresponding reference (e.g. unmodified) or wild-type ICOSL, such as a reference ICOSL set forth in SEQ ID NO:32 or SEQ ID NOs: 196 or 545.
  • a corresponding reference e.g. unmodified
  • wild-type ICOSL such as a reference ICOSL set forth in SEQ ID NO:32 or SEQ ID NOs: 196 or 545.
  • Non-naturally occurring amino acids as well as naturally occurring amino acids are included within the scope of permissible substitutions or additions.
  • a variant ICOSL is not limited to any particular method of making and includes, for example, de novo chemical synthesis, de novo recombinant DNA techniques, or combinations thereof.
  • a variant ICOSL of the invention specifically binds to CD28, ICOS, and/or CTLA-4 of a mammalian species.
  • the altered amino acid sequence results in an altered (i.e., increased or decreased) binding affinity or avidity to ICOS and/or CD28 compared to the reference (e.g. unmodified) or wild-type ICOSL protein.
  • An increase or decrease in binding affinity or avidity can be determined using well known binding assays such as flow cytometry. Larsen et al., American Journal of Transplantation, Vol 5: 443-453 (2005). See also, Linsley et al., Immunity, Vol. 1(9): 793-801 (1994).
  • An increase in variant ICOSL binding affinity or avidity to ICOS and/or CD28 is to a value at least 5% greater than that of the reference (e.g. unmodified) or wild-type ICOSL and in some embodiments, at least 10%, 15%, 20%, 30%, 40%, 50%, 100% greater than that of the reference (e.g. unmodified) or wild-type ICOSL control value.
  • a decrease in ICOSL binding affinity or avidity to ICOS and/or CD28 is to a value no greater than 95% of the of the wild-type control values, and in some embodiments no greater than 80%, 70% 60%, 50%, 40%, 30%, 20%, 10%, 5%, or no detectable binding affinity or avidity of the wild-type ICOS and/or CD28 control values.
  • a variant ICOSL is altered in primary amino acid sequence by substitution, addition, or deletion of amino acid residues.
  • the term “variant” in the context of variant ICOSL is not be construed as imposing any condition for any particular starting composition or method by which the variant ICOSL is created.
  • a variant ICOSL can, for example, be generated starting from a reference ICOSL or wild type mammalian ICOSL sequence information, then modeled in silico for binding to ICOS and/or CD28, and finally recombinantly or chemically synthesized to yield a variant ICOSL of the present invention.
  • a variant ICOSL can be created by site-directed mutagenesis of a reference (e.g. unmodified) or wild-type ICOSL.
  • variant ICOSL denotes a composition and not necessarily a product produced by any given process.
  • a variety of techniques including recombinant methods, chemical synthesis, or combinations thereof, may be employed.
  • wild-type or “natural” or “native” as used herein is used in connection with biological materials such as nucleic acid molecules, proteins (e.g., ICOSL), IgSF members, host cells, and the like, refers to those which are found in nature and not modified by human intervention.
  • variant ICOSL polypeptides that exhibit altered (increased or decreased) binding activity or affinity for one or more of an ICOSL cognate binding partner.
  • the ICOSL cognate binding partner is one or more of CD28, ICOS, or CTLA-4.
  • the variant ICOSL polypeptide contains one or more amino acids modifications, such as one or more substitutions (alternatively, “mutations” or “replacements”), deletions or addition, in an immunoglobulin superfamily (IgSF) domain (IgD) relative to a wild-type or unmodified ICOSL polypeptide or a portion of a wild-type or unmodified ICOSL containing an immunoglobulin superfamily (IgSF) domain or a specific binding fragment thereof.
  • a provided variant ICOSL polypeptide is or comprises a variant IgD (hereinafter called “vIgD”) in which the one or more amino acid modifications (e.g. substitutions) is in an IgD.
  • the IgD comprises an IgV domain or an IgC (e.g. IgC2) domain or specific binding fragment of the IgV domain or the IgC (e.g. IgC2) domain, or combinations thereof.
  • the IgD can be an IgV only, the combination of the IgV and IgC, including the entire extracellular domain (ECD), or any combination of Ig domains of ICOSL.
  • Table 2 provides exemplary residues that correspond to IgV or IgC regions of ICOSL.
  • the variant ICOSL polypeptide contains an IgV domain or an IgC domain or specific binding fragments thereof in which the at least one of the amino acid modifications (e.g. substitutions) is in the IgV domain or IgC domain or a specific binding fragment thereof.
  • the IgV domain or IgC domain is an affinity-modified IgSF domain.
  • the variant is modified in one more IgSF domains relative to the sequence of a reference (e.g., unmodified) ICOSL sequence.
  • the reference (e.g., unmodified) ICOSL sequence is a wild-type ICOSL.
  • the reference (e.g., unmodified) or wild-type ICOSL has the sequence of a native ICOSL or an ortholog thereof.
  • the reference (e.g., unmodified) or wild-type ICOSL is or comprises the extracellular domain (ECD) of ICOSL or a portion thereof containing one or more IgSF domain (see Table 2).
  • the extracellular domain of a reference (e.g., unmodified) or wild-type ICOSL polypeptide comprises an IgV domain and an IgC domain or domains.
  • the variant ICOSL polypeptide need not comprise both the IgV domain and the IgC domain or domains.
  • the variant ICOSL polypeptide comprises or consists essentially of the IgV domain or a specific binding fragment thereof.
  • the variant ICOSL polypeptide comprises or consists essentially of the IgC domain or specific binding fragments thereof.
  • the variant ICOSL is soluble and lacks a transmembrane domain.
  • the variant ICOSL further comprises a transmembrane domain and, in some cases, also a cytoplasmic domain.
  • the reference (e.g., unmodified) or wild-type ICOSL sequence is a mammalian ICOSL sequence.
  • the reference (e.g., unmodified) or wild-type ICOSL sequence can be a mammalian ICOSL that includes, but is not limited to, human, mouse, cynomolgus monkey, or rat.
  • the reference (e.g., unmodified) or wildtype ICOSL sequence is human.
  • the reference (e.g., unmodified) or wild-type ICOSL sequence has (i) the sequence of amino acids set forth in SEQ ID NO:5 or a mature form thereof lacking the signal sequence, (ii) a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO:5 or the mature form thereof, or (iii) is a portion of (i) or (ii) containing an IgV domain or IgC domain or specific binding fragments thereof.
  • the reference ICOSL sequence is or comprises an extracellular domain of the ICOSL or a portion thereof.
  • the reference or wild-type ICOSL polypeptide comprises the amino acid sequence set forth in SEQ ID NO:32, or an ortholog thereof.
  • the reference (e.g., unmodified) or wild-type ICOSL polypeptide can comprise (i) the sequence of amino acids set forth in SEQ ID NO:32, (ii) a sequence of amino acids that has at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 32, or (iii) is a specific binding fragment of the sequence of (i) or (ii) comprising an IgV domain or an IgC domain.
  • the reference ICOSL polypeptide comprises a truncated extracellular domain comprising a C-terminal truncation with reference to the reference ICOSL extracellular domain sequence set forth in SEQ ID NO:32.
  • the C-terminal truncation is of at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 125 amino acid residues.
  • the C-terminal truncation is of at least 1, at least 5 at least 10, at least 15, at least 20, at least 25, at least 30, at least 35 amino acid residues.
  • the ICOSL polypeptide comprising a C-terminal truncation does not contain, beyond the C-terminus of the truncation point, contiguous amino acid residues of a wild-type ICOSL.
  • ICOSL reference sequences are those that are shorter than the full extracellular domain of a wild-type ICOSL, e.g. set forth in SEQ ID NO: 32.
  • the ICOSL polypeptide comprising a C-terminal truncation does not contain or is not fused to amino acid residues of an ICOSL domain beyond the extracellular domain.
  • the ICOSL reference polypeptide is altered, such as mutated or deleted, in one or more protease cleavage site.
  • wild-type ICOSL polypeptide contains a protease cleavage site that, in some cases, results in cleavage of the protein upon expression in cells, e.g. Chinese Hamster Ovary cells, thereby resulting in a heterogeneous product of multiple species, including species of different lengths or sizes.
  • cleavage of the ICOSL polypeptide may occur at the LQQN/LT protease cleavage site between residues 207 and 208 of SEQ ID NO: 32 (“/” indicates potential cleavage site).
  • the ICOSL reference polypeptide is altered in or lacks a protease cleavage site set forth as amino acids 204-209 of SEQ ID NO:32.
  • a truncated ICOSL polypeptide is more resistant to protease cleavage compared to a wild-type or non-truncated ICOSL polypeptide.
  • Exemplary truncated ICOSL polypeptide ECD truncations lacking all or a portion of the LQQN/LT protease cleavage site are provided in SEQ ID NOs: 600-606.
  • Truncation #2 (SEQ ID NO: 600) DTQEKEVRAMVGSDVELSCACPEGSRFDLNDVYVYWQTSESKTVVTYHIP QNSSLENVDSRYRNRALMSPAGMLRGDFSLRLFNVTPQDEQKFHCLVLSQ SLGFQEVLSVEVTLHVAANFSVPVVSAPHSPSQDELTFTCTSINGYPRPN VYWINKTDNSLLDQALQNDTVFLNMRGLYDVVSVLRIARTPSVNIGCCIE NVLLQQNL Truncation #3: (SEQ ID NO: 601) DTQEKEVRAMVGSDVELSCACPEGSRFDLNDVYVYWQTSESKTVVTYHIP QNSSLENVDSRYRNRALMSPAGMLRGDFSLRLFNVTPQDEQKFHCLVLSQ SLGFQEVLSVEVTLHVAANFSVPVVSAPHSPSQDELTFTCTSINGYPRPN VYWINKTDNSLLDQALQNDTVFLNMRGLYDV
  • the ICOSL reference polypeptide is altered in one or more amino acids corresponding to amino acids 204-209 with reference to SEQ ID NO:32.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g., substitution in a reference ICOSL or specific binding fragment thereof corresponding to position(s) 207 and/or 208 with reference to numbering of SEQ ID NO:32.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g., substitution, selected from N207A, N207G, L208G, or a conservative amino acid modification, e.g., substitution thereof.
  • the one or more amino acid modification e.g., substitution is N207A/L208G or N207G/L208G.
  • the full length reference ECDs or truncated reference ECDs of the variant ICOSL polypeptide are modified to contain one or more amino acid modifications, e.g., substitutions, selected from N207A, N207G, L208G, or a conservative amino acid modification.
  • Exemplary full length or truncated reference ECDs with one or more modifications are set forth in SEQ ID NOs: 607-628.
  • Exemplary reference sequences containing mutations at cleavage site N207 and/or L208 with reference to positions are set forth in SEQ ID NO: 32 are set forth in SEQ ID NOs: 624-628.
  • the provided modifications may reduce protease cleavage of the ICOSL polypeptide, such as cleavage that may occur at the LQQN/LT protease cleavage site.
  • combinations of the above truncation and modification strategies can be employed in a reference ICOSL ECD sequence.
  • the modifications e.g., substitutions, are made in a truncated reference ICOSL polypeptide such as exemplary reference ICOSL sequence set forth in SEQ ID NOs: 600-606.
  • Exemplary variant ICOSL polypeptide sequences with modifications at the potential protease cleavage site(s) N207 and/or L208 are set forth in SEQ ID NOs: 607-628.
  • the variant ICOSL polypeptide exhibits decreased protease cleavage compared to wild-type ICOSL polypeptide, such as containing the ECD sequence set forth in SEQ ID NO:32.
  • the reference (e.g., unmodified) or wildtype ICOSL polypeptide comprises an IgV domain or an IgC domain, or a specific binding fragment thereof.
  • an ICOSL reference polypeptide containing an IgV domain comprises the amino acid sequence set forth in SEQ ID NO: 196 (corresponding to amino acid residues 19-129 of SEQ ID NO:5), or an ortholog thereof.
  • the reference ICOSL polypeptide containing the IgV domain contains at least amino acids 1-112, 1-113, 1-114, 1-115, 1-116, 1-117, 1-118, 1-119, 1-120, 1-121, 1-122, with reference to numbering set forth in SEQ ID NO:32.
  • an ICOSL reference polypeptide containing an IgV domain comprises the amino acid sequence set forth in SEQ ID NO: 545 (corresponding to amino acid residues 19-140 of SEQ ID NO:5), or an ortholog thereof.
  • the IgV domain is the only IgSF domain of the ICOSL reference polypeptide.
  • the IgV domain of the reference (e.g., unmodified) or wild-type ICOSL polypeptide can contain (i) the sequence of amino acids set forth in SEQ ID NO: 196 or 545, (ii) a sequence of amino acids that has at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 196 or 545, or (iii) a specific binding fragment of the sequence of amino acids set forth in SEQ ID NO: 196 or 545 or a specific binding fragment of a sequence of (i) or (ii).
  • the reference (e.g., unmodified) IgV domain is capable of binding one or more ICOSL cognate binding proteins, such as one or more of CD28, ICOS, or CTLA-4.
  • the IgC domain of the reference (e.g., unmodified) or wild-type ICOSL polypeptide comprises the amino acid sequence set forth as residues 141-227 of SEQ ID NO: 5, or an ortholog thereof.
  • the IgC domain of the reference (e.g., unmodified) or wild-type ICOSL polypeptide can contain (i) the sequence of amino acids set forth residues 141-227 of SEQ ID NO: 5, (ii) a sequence of amino acids that has at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence identity to residues 141-227 of SEQ ID NO: 5, or (iii) (i) or (ii).
  • the reference IgV domain is capable of binding one or more ICOSL cognate binding proteins.
  • the reference (e.g., unmodified) or wild-type ICOSL polypeptide contains a specific binding fragment of ICOSL, such as a specific binding fragment of the IgV domain or the IgC domain.
  • the specific binding fragment can bind CD28, ICOS, and/or CTLA-4.
  • the specific binding fragment can have an amino acid length of at least 50 amino acids, such as at least 60, 70, 80, 90, 100, or 110 amino acids.
  • the specific binding fragment of the IgV domain contains an amino acid sequence that is at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% of the length of the IgV domain set forth as amino acids 19-129 of SEQ ID NO: 5.
  • the specific binding fragment of the IgC domain comprises an amino acid sequence that is at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% of the length of the IgC domain set forth as amino acids 141-227 of SEQ ID NO: 5.
  • the variant ICOSL polypeptide comprises the ECD domain, a truncated ECD domain, or a portion thereof comprising one or more affinity modified IgSF domains.
  • the variant ICOSL polypeptides can comprise an IgV domain or an IgC domain, in which one or more of the IgSF domains (IgV or IgC) or a specific binding fragment of the IgV domain or a specific binding fragment of the IgC domain contains the one or more amino acid modifications (e.g. substitutions).
  • the variant ICOSL polypeptides can comprise an IgV domain and an IgC domain, or a specific binding fragment of the IgV domain and a specific binding fragment of the IgC domain.
  • the variant ICOSL polypeptide comprises a full-length IgV domain. In some embodiments, the variant ICOSL polypeptide comprises a full-length IgC domain. In some embodiments, the variant ICOSL polypeptide comprises a specific binding fragment of the IgV domain. In some embodiments, the variant ICOSL polypeptide comprises a specific binding fragment of the IgC domain. In some embodiments, the variant ICOSL polypeptide comprises a full-length IgV domain and a full-length IgC domain. In some embodiments, the variant ICOSL polypeptide comprises a full-length IgV domain and a specific binding fragment of an IgC domain.
  • the variant ICOSL polypeptide comprises a specific binding fragment of an IgV domain and a full-length IgC domain. In some embodiments, the variant ICOSL polypeptide comprises a specific binding fragment of an IgV domain and a specific binding fragment of an IgC domain.
  • the one or more amino acid modifications (e.g., substitutions) of the variant ICOSL polypeptides can be located in any one or more of the ICOSL polypeptide domains.
  • one or more amino acid substitutions are located in the extracellular domain (ECD) of the variant ICOSL polypeptide, such as set forth in SEQ ID NO: 32.
  • one or more amino acid substitutions are located in the IgV domain or specific binding fragment of the IgV domain.
  • one or more amino acid modifications (e.g. substitutions) are located in the IgC domain or specific binding fragment of the IgC domain.
  • each of the various attributes of polypeptides are separately disclosed below (e.g., soluble, secretable and membrane bound polypeptides, affinity of ICOSL for CD28, ICOS, and CTLA-4, number of variations per polypeptide chain, number of linked polypeptide chains, the number and nature of amino acid alterations per variant ICOSL, etc.).
  • any particular polypeptide can comprise a combination of these independent attributes. It is understood that reference to amino acids, including to a specific sequence set forth as a SEQ ID NO used to describe domain organization of an IgSF domain are for illustrative purposes and are not meant to limit the scope of the embodiments provided.
  • polypeptides and the description of domains thereof are theoretically derived based on homology analysis and alignments with similar molecules. Thus, the exact locus can vary, and is not necessarily the same for each protein.
  • the specific IgSF domain such as specific IgV domain or IgC domain, can be several amino acids (such as one, two, three or four) longer or shorter.
  • variant ICOSL polypeptides containing at least one affinity-modified IgSF domain (e.g., IgV or IgC) or a specific binding fragment thereof in an IgSF domain contained in a reference (e.g., unmodified) or wild-type ICOSL polypeptide such that the variant ICOSL polypeptide exhibits altered (increased or decreased) binding activity or affinity for one or more ligands ICOS, CD28, or CTLA-4 compared to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • affinity-modified IgSF domain e.g., IgV or IgC
  • a reference e.g., unmodified
  • a variant ICOSL polypeptide has a binding affinity for CD28, ICOS, and/or CTLA-4 that differs from that of a reference (e.g., unmodified) or wild-type ICOSL polypeptide control sequence as determined by, for example, solid-phase ELISA immunoassays, flow cytometry or Biacore assays.
  • the variant ICOSL polypeptide has an increased binding affinity for CD28, ICOS, and/or CTLA-4.
  • the variant ICOSL polypeptide has a decreased binding affinity for CD28, ICOS, and/or CTLA-4, relative to a reference (unmodified) or wild-type ICOSL polypeptide.
  • the CD28, ICOS and/or the CTLA-4 can be a mammalian protein, such as a human protein or a murine protein.
  • Binding affinities for each of the cognate binding partners are independent; that is, in some embodiments, a variant ICOSL polypeptide has an increased binding affinity for one, two or three of CD28, ICOS, and/or CTLA-4, and a decreased binding affinity for one, two or three of CD28, ICOS, and CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the variant ICOSL polypeptide has an increased binding affinity for CD28, relative to a reference (e.g., unmodified) or wildtype ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has an increased binding affinity for ICOS, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has an increased binding affinity for CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the variant ICOSL polypeptide has a decreased binding affinity for CD28, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has a decreased binding affinity for ICOS, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has a decreased binding affinity for CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the variant ICOSL polypeptide has an increased binding affinity for CD28 and ICOS, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has an increased binding affinity for CD28 and a decreased binding affinity for ICOS, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has a decreased binding affinity for CD28 and ICOS, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has a decreased binding affinity for CD28 and an increased binding affinity for ICOS, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the variant ICOSL polypeptide has an increased binding affinity for CD28 and CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has an increased binding affinity for CD28 and a decreased binding affinity for CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has a decreased binding affinity for CD28 and CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has a decreased binding affinity for CD28 and an increased binding affinity for CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the variant ICOSL polypeptide has an increased binding affinity for ICOS and CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOS polypeptide. In some embodiments, the variant ICOSL polypeptide has an increased binding affinity for ICOS and a decreased binding affinity for CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has a decreased binding affinity for ICOS and CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has a decreased binding affinity for ICOS and an increased binding affinity for CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the variant ICOSL polypeptide has an increased binding affinity for CD28, ICOS, and CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has an increased binding affinity for CD28 and ICOS, and a decreased binding affinity for CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has an increased binding affinity for CD28 and CTLA-4, and a decreased binding affinity for ICOS, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the variant ICOSL polypeptide has a decreased binding affinity for CD28 and ICOS, and an increased binding affinity for CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has a decreased binding affinity for CD28 and an increased binding affinity for ICOS and CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has an increased binding affinity for CD28, and a decreased binding affinity for ICOS and CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the variant ICOSL polypeptide has a decreased binding affinity for CD28, CTLA-4, and ICOS, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide. In some embodiments, the variant ICOSL polypeptide has a decreased binding affinity for CD28, and an increased binding affinity for ICOS and CTLA-4, relative to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • a variant ICOSL polypeptide with increased or greater binding affinity to CD28, ICOS, and/or CTLA-4 will have an increase in binding affinity relative to the reference (e.g., unmodified) or wild-type ICOSL polypeptide control of at least about 5%, such as at least about 10%, 15%, 20%, 25%, 35%, or 50% for the CD28, ICOS, and/or CTLA-4.
  • the increase in binding affinity relative to the reference (e.g., unmodified) or wild-type ICOSL polypeptide is more than 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold 40-fold or 50-fold.
  • the reference (e.g., unmodified) or wild-type ICOSL polypeptide has the same sequence as the variant ICOSL polypeptide except that it does not contain the one or more amino acid modifications (e.g., substitutions).
  • a variant ICOSL polypeptide with reduced or decreased binding affinity to CD28, ICOS, and/or CTLA-4 will have decrease in binding affinity relative to the reference (e.g., unmodified) or wild-type ICOSL polypeptide control of at least 5%, such as at least about 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more for the CD28, ICOSL, and/or CTLA-4.
  • the decrease in binding affinity relative to the reference (e.g., unmodified) or wild-type ICOSL polypeptide is more than 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold 40-fold or 50-fold.
  • the reference (e.g., unmodified) or wild-type ICOSL polypeptide has the same sequence as the variant ICOSL polypeptide except that it does not contain the one or more amino acid modifications, e.g. substitutions.
  • the equilibrium dissociation constant (K d ) of any of the foregoing embodiments to CD28, ICOS, and/or CTLA-4 can be less than 1 ⁇ 10 ⁇ 5 M, 1 ⁇ 10 ⁇ 6 M, 1 ⁇ 10 ⁇ 7 M, 1 ⁇ 10 ⁇ 8 M, 1 ⁇ 10 ⁇ 9 M, 1 ⁇ 10 ⁇ 10 M or 1 ⁇ 10 ⁇ 11 M, or 1 ⁇ 10 ⁇ 12 M.
  • a variant ICOSL polypeptide has an increased or greater binding affinity to CD28.
  • a variant ICOSL polypeptide with increased or greater binding affinity to CD28 will have an increase in binding affinity relative to the reference (e.g., unmodified) or wild-type ICOSL polypeptide control of at least about 25%, such as at least about 30%, 40%, 50%, or 60% for CD28.
  • a variant ICOSL polypeptide with increased or greater binding affinity to CD28 has an equilibrium dissociation constant (K d ) of less than 200 pM, 300 pM, 400 pM, 500 pM, or 600 pM for CD28.
  • the variant polypeptide specifically binds to the ectodomain of one of ICOS, CD28 or CTLA4 with increased selectivity compared to the reference (e.g., unmodified) or wild-type ICOSL.
  • the increased selectivity is for CD28.
  • the increased selectivity comprises a greater ratio of binding of the variant ICOSL polypeptide for one cognate binding partner selected from among ICOS, CD28 and CTLA4 versus another of the cognate binding partner compared to the ratio of binding of the reference (e.g., unmodified) or wild-type ICOSL polypeptide for the one cognate binding partner versus the another of the cognate binding partner.
  • the ratio is greater by at least or at least about 1.5-fold, 2.0-fold, 3.0-fold, 4.0-fold, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 40-fold, 50-fold or more.
  • variant ICOSL polypeptides can be made, for example, by de novo peptide synthesis and thus does not necessarily require a modification, such as a “substitution”, in the sense of altering a codon to encode for the modification, e.g. substitution.
  • variant ICOSL polypeptides are designed or created is not limited to any particular method. In some embodiments, however, a reference (e.g., unmodified) or wild-type ICOSL encoding nucleic acid is mutagenized from reference (e.g., unmodified) or wild-type ICOSL genetic material and screened for desired specific binding affinity and/or induction of IFN-gamma expression or other functional activity.
  • a reference e.g., unmodified
  • wild-type ICOSL encoding nucleic acid is mutagenized from reference (e.g., unmodified) or wild-type ICOSL genetic material and screened for desired specific binding affinity and/or induction of IFN-gamma expression or other functional activity.
  • a variant ICOSL polypeptide is synthesized de novo utilizing protein or nucleic acid sequences available at any number of publicly available databases and then subsequently screened.
  • the National Center for Biotechnology Information provides such information and its website is publicly accessible via the internet as is the UniProtKB database as discussed previously.
  • amino acid modification are designated by amino acid position number corresponding to the numbering of positions of the reference ECD sequence set forth in SEQ ID NO:32. It is within the level of a skilled artisan to identify the corresponding position of a modification, e.g. amino acid substitution, in an ICOSL polypeptide, including portion thereof containing an IgSF domain (e.g. IgV) thereof, such as by alignment of a reference sequence (e.g. SEQ ID NO: 196, 545, 600-628) with SEQ ID NO:32. In the listing of modifications throughout this disclosure, the amino acid position is indicated in the middle, with the corresponding reference (e.g.
  • unmodified or wild-type amino acid listed before the number and the identified variant amino acid substitution listed after the number. If the modification is a deletion of the position a “del” is indicated and if the modification is an insertion at the position an “ins” is indicated. In some cases, an insertion is listed with the amino acid position indicated in the middle, with the corresponding reference amino acid listed before and after the number and the identified variant amino acid insertion listed after the unmodified (e.g. wild-type) amino acid.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution in a reference (e.g., unmodified) or wild-type ICOSL sequence.
  • the one or more amino acid modification, e.g. substitution can be in the ectodomain (extracellular domain) of the reference (e.g., unmodified) or wild-type ICOSL sequence.
  • the one or more amino acid modification, e.g. substitution is in the IgV domain or specific binding fragment thereof.
  • the one or more amino acid modification, e.g. substitution is in the IgC domain or specific binding fragment thereof.
  • some of the one or more amino acid modification, e.g. substitution is in the IgV domain or a specific binding fragment thereof, and some of the one or more amino acid modification, e.g. substitution are in the IgC domain or a specific binding fragment thereof.
  • the variant ICOSL polypeptide has up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid modification(s), e.g. substitution.
  • the modification, e.g. substitution can be in the IgV domain or the IgC domain.
  • the variant ICOSL polypeptide has up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid substitutions in the IgV domain or specific binding fragment thereof.
  • the variant ICOSL polypeptide has up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid substitutions in the IgC domain or specific binding fragment thereof.
  • the variant ICOSL polypeptide has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the reference (e.g., unmodified) or wild-type ICOSL polypeptide or specific binding fragment thereof, such as with the amino acid sequence of SEQ ID NO: 32, 196 or 545.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution in a reference ICOSL or specific binding fragment there of corresponding to position(s) 10, 11, 13, 16, 18, 20, 25, 27, 30, 33, 37, 42, 43, 47, 52, 54, 57, 61, 62, 67, 71, 72, 74, 77, 78, 75, 80, 84, 89, 90, 92, 93, 94, 96, 97, 98, 99, 100, 102, 103, 107, 109, 110, 111, 113, 115, 116, 117, 119, 120, 121, 122, 126, 129, 130, 132, 133, 135, 138, 139, 140, 142, 143, 144, 146, 151, 152, 153, 154, 155, 156, 158, 161, 166, 168, 172, 173, 175, 190, 192, 193, 194, 198,
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution in a reference ICOSL or specific binding fragment there of corresponding to position(s) 10, 11, 13, 16, 18, 20, 25, 26, 27, 30, 33, 37, 38, 42, 43, 47, 52, 54, 57, 61, 62, 67, 71, 72, 74, 75, 77, 78, 80, 84, 89, 90, 92, 93, 94, 96, 97, 98, 99, 100, 102, 103, 107, 109, 110, 111, 113, 115, 116, 117, 119, 120, 121, 122, 126, 129, 130, 132, 133, 135, 137, 138, 139, 140, 142, 143, 144, 146, 151, 152, 153, 154, 155, 156, 158, 161, 164, 166, 168, 172, 173, 175, 190,
  • such variant ICOSL polypeptides exhibit altered binding affinity to one or more of CD28, ICOS, and/or CTLA-4 compared to the reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the variant ICOSL polypeptide exhibits increased binding affinity to CD28, ICOS, and/or CTLA-4 compared to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the variant ICOSL polypeptide exhibits decreased binding affinity to CD28, ICOS, or CTLA-4 compared to a reference (e.g., unmodified) or wild-type ICOSL polypeptide.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from M10V, M10I, V11E, S13G, E16V, S18R, A20V, S25G, F27S, F27C, N30D, Y33del, Q37R, K42E, Y47H, T43A, N52A, N52C, N52D, N52G, N52H, N52L, N52K, N52M, N52P, N52Q, N52R, N52S, N52T, N52V, N52Y, S54A, S54P, N57A, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57W, N57Y, R61S, R61C, Y62F, L67P, A71T, G72R, L74Q, R75Q, D77
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from M10V, M10I, V11E, S13G, E16V, S18R, A20T, A20V, S25G, R26S, F27C, F27S, N30D, Y33del, Q37R, T38P, K42E, T43A, Y47H, N52A, N52C, N52D, N52G, N52H, N52K, N52L, N52M, N52P, N52Q, N52R, N52S, N52T, N52V, N52Y, S54A, S54F, S54P, N57A, N57D, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57W, N57Y, R61C, R61S, Y62F
  • a conservative amino acid modification is any amino acid that falls in the same class of amino acids as the substituted amino acids, other than the reference (e.g., unmodified) or wild-type amino acid.
  • the classes of amino acids are aliphatic (glycine, alanine, valine, leucine, and isoleucine), hydroxyl or sulfur-containing (serine, cysteine, threonine, and methionine), cyclic (proline), aromatic (phenylalanine, tyrosine, tryptophan), basic (histidine, lysine, and arginine), and acidic/amide (aspartate, glutamate, asparagine, and glutamine).
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from M10V, M10I, V11E, S13G, E16V, S18R, A20V, S25G, F27S, F27C, N30D, Y33del, Q37R, K42E, T43A, Y47H, N52A, N52C, N52D, N52G, N52H, N52L, N52K, N52M, N52P, N52Q, N52R, N52S, N52T, N52V, N52Y, S54A, S54P, N57A, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57W, N57Y, R61S, R61C, Y62F, L67P, A71T, G72R, L74Q, R75Q, D77
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from M10V, M10I, V11E, S13G, E16V, S18R, A20T, A20V, S25G, R26S, F27C, F27S, N30D, Y33del, Q37R, T38P, K42E, T43A, Y47H, N52A, N52C, N52D, N52G, N52H, N52K, N52L, N52M, N52P, N52Q, N52R, N52S, N52T, N52V, N52Y, S54A, S54F, S54P, N57A, N57D, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57W, N57Y, R61C, R61S, Y62F, L67
  • the one or more amino acid modification e.g. substitution is N52Y/N57Y/F138L/L203P, N52H/N57Y/Q100P, N52S/Y146C/Y152C, N52H/C198R, N52H/C140D/T225A, N52H/C198R/T225A, N52H/K92R, N52H/S99G, N57Y/Q100P, N52S/G103E, N52S/S130G/Y152C, N52S/Y152C, N52S/C198R, N52Y/N57Y/Y152C, N52Y/N57Y/H129P/C198R, N52H/L161P/C198R, N52S/T113E, N52D/S54P, N52K/L208P, N52S/Y152H, N52D/V151A, N52H/I143T, N52S/L80P, F120S/Y152
  • the one or more amino acid modifications are selected from among N52Y/N57Y/F138L/L203P, N52H/N57Y/Q100P, N52S/Y146C/Y152C, N52H/C198R, N52H/C140D/T225A, N52H/C198R/T225A, N52H/K92R, N52H/S99G, N57Y/Q100P, N52S/S130G/Y152C, N52S/Y152C, N52S/C198R, N52Y/N57Y/Y152C, N52Y/N57Y/H129P/C198R, N52H/L161P/C198R, N52S/T113E, N52D/S54P, N52K/L208P, N52S/Y152H, N52D/V151A, N52H/I143T, N52S/L80P, F120S/Y152H/N201S, N52S/R75
  • the variant ICOSL polypeptide comprises any of the mutations listed in Table 1.
  • Table 1 also provides exemplary sequences by reference to SEQ ID NO for the extracellular domain (ECD) or IgV domain of the reference (e.g., unmodified) or wild-type ICOSL or exemplary variant ICOSL polypeptides.
  • ECD extracellular domain
  • IgV IgV domain of the reference
  • wild-type ICOSL wild-type ICOSL
  • exemplary variant ICOSL polypeptides As indicated, the exact locus or residues corresponding to a given domain can vary, such as depending on the methods used to identify or classify the domain.
  • adjacent N- and/or C-terminal amino acids of a given domain e.g. IgV
  • adjacent N- and/or C-terminal amino acids of a given domain also can be included in a sequence of a variant IgSF polypeptide, such as to ensure proper folding of the domain when expressed.
  • the particular domain, such as the ECD domain, of a variant ICOSL polypeptide can be several amino acids longer or shorter, such as 1-10, e.g., 1, 2, 3, 4, 5, 6 or 7 amino acids longer or shorter, than the sequence of amino acids set forth in the respective SEQ ID NO.
  • the variant ICOSL polypeptide comprises any of the mutations listed in Table 1.
  • the mutations are made in a reference ICOSL containing the sequence of amino acids set forth in SEQ ID NO: 32, a reference ICOSL that contains the IgV domain of ICOSL set forth in SEQ ID NOs: 196 or 545, or a reference ICOSL that is truncated and/or modified containing the sequence of amino acids set forth in any of SEQ ID NOs: 600-628.
  • the variant ICOSL polypeptide comprises any of the extracellular domain (ECD) sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 109-142, 239, 280-325, 364-381, 387-424, 427-433, 435-470, 638-685).
  • ECD extracellular domain
  • the variant ICOSL polypeptide comprises a polypeptide sequence that exhibits at least 90% identity, at least 91% identity, at least 92% identity, at least 93% identity, at least 94% identity, at least 95% identity, such as at least 96% identity, 97% identity, 98% identity, or 99% identity to any of the extracellular domain (ECD) sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 109-142, 239, 280-325, 364-381, 387-424, 427-433, 435-470, 638-685) and contains the amino acid modification(s), e.g. substitution(s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • ECD extracellular domain
  • the variant ICOSL polypeptide comprises a specific binding fragment of any of the extracellular domain (ECD) sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 109-142, 239, 280-325, 364-381, 387-424, 427-433, 435-470, 638-685) and contains the amino acid modification(s), e.g. substitution (s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • ECD extracellular domain
  • the variant ICOSL polypeptide comprises any of the extracellular domain (ECD) sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 109-142, 239, 280-325, 364-381, 387-424, 427-433, 435-470, 638-685, 905, 908).
  • ECD extracellular domain
  • the variant ICOSL polypeptide comprises a polypeptide sequence that exhibits at least 90% identity, at least 91% identity, at least 92% identity, at least 93% identity, at least 94% identity, at least 95% identity, such as at least 96% identity, 97% identity, 98% identity, or 99% identity to any of the extracellular domain (ECD) sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 109-142, 239, 280-325, 364-381, 387-424, 427-433, 435-470, 638-685, 905, 908) and contains the amino acid modification(s), e.g.
  • ECD extracellular domain
  • the variant ICOSL polypeptide comprises a specific binding fragment of any of the extracellular domain (ECD) sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 109-142, 239, 280-325, 364-381, 387-424, 427-433, 435-470, 638-685, 905, 908) and contains the amino acid modification(s), e.g. substitution (s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • ECD extracellular domain
  • the variant ICOSL polypeptide comprises any of the IgV sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 197-199, 201-208, 210, 212, 240, 326-340, 382-386, 425-426, 434, 546-599, 686-857).
  • the variant ICOSL polypeptide comprises a polypeptide sequence that exhibits at least 90% identity, at least 91% identity, at least 92% identity, at least 93% identity, at least 94% identity, at least 95% identity, such as at least 96% identity, 97% identity, 98% identity, or 99% identity to any of the IgV sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 197-199, 201-208, 210, 212, 240, 326-340, 382-386, 425-426, 434, 546-599, 686-857) and contains the amino acid modification(s), e.g. substitution(s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • the variant ICOSL polypeptide comprises a specific binding fragment of any of the IgV sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 197-199, 201-208, 210, 212, 240, 326-340, 382-386, 425-426, 434, 546-599, 686-857) and contains the amino acid substitution(s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • the variant ICOSL polypeptide comprises any of the IgV sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 197-199, 201-208, 210, 212, 240, 326-340, 382-386, 425-426, 434, 546-599, 686-857, 906-907, 909-910).
  • the variant ICOSL polypeptide comprises a polypeptide sequence that exhibits at least 90% identity, at least 91% identity, at least 92% identity, at least 93% identity, at least 94% identity, at least 95% identity, such as at least 96% identity, 97% identity, 98% identity, or 99% identity to any of the IgV sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 197-199, 201-208, 210, 212, 240, 326-340, 382-386, 425-426, 434, 546-599, 686-857, 906-907, 909-910) and contains the amino acid modification(s), e.g.
  • the variant ICOSL polypeptide comprises a specific binding fragment of any of the IgV sequences listed in Table 1 (i.e., any one of SEQ ID NOS: 197-199, 201-208, 210, 212, 240, 326-340, 382-386, 425-426, 434, 546-599, 686-857, 906-907, 909-910) and contains the amino acid substitution(s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • Mutations designated with an “X” indicate the designated position contains a Q or the wild-type residue set forth in the corresponding position of SEQ ID NO: 32.
  • the variant ICOSL polypeptide exhibits increased affinity for the ectodomain of CD28 compared to the reference (e.g., unmodified) or wild-type ICOSL polypeptide, such as comprising the sequence set forth in SEQ ID NO: 32, 196, or 545.
  • the ICOSL polypeptide exhibits increased affinity for the ectodomain of ICOS compared to the reference (e.g., unmodified) or wild-type ICOSL, such as comprising the sequence set forth in SEQ ID NO: 32, 196, or 545.
  • the ICOSL polypeptide exhibits increased affinity for the ectodomain of CD28 and the ectodomain of ICOS compared to the reference (e.g., unmodified) or wild-type ICOSL, such as comprising the sequence set forth in SEQ ID NO: 32, 196, or 545.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution corresponding to position(s) 52, 54 or 57. In some embodiments, the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from N52H, N52D, N52Q, N52S, N52Y, N52K, S54A, S54P, or N57Y or a conservative amino acid modification, e.g. substitution thereof. In some embodiments, the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from N52H, N52D, N52S, N52K or N57Y or a conservative amino acid modification, e.g. substitution thereof.
  • the variant ICOSL polypeptide can contain one or more further amino acid modification, e.g. substitution in addition to an amino acid modification, e.g. substitution at a position corresponding to position 52, 54 or 57.
  • the one or more further amino acid modification e.g.
  • substitution is at a position corresponding to 10, 11, 13, 16, 18, 20, 25, 27, 30, 37, 42, 43, 47, 52, 54, 57, 61, 62, 67, 71, 72, 74, 75, 77, 78, 80, 84, 89, 90, 92, 93, 94, 96, 97, 98, 99, 100, 102, 103, 107, 109, 110, 113, 115, 116, 117, 119, 120, 121, 122, 126, 129, 130, 132, 133, 135, 138, 139, 140, 142, 143, 144, 146, 151, 152, 153, 154, 155, 156, 158, 161, 166, 168, 172, 173, 175, 190, 192, 193, 194, 198, 201, 203, 207, 208, 210, 212, 217, 218, 220, 221, 224, 225 or 227.
  • the one or more further amino acid modification e.g. substitution is at a position corresponding to 10, 11, 13, 16, 18, 20, 25, 26, 27, 30, 33, 37, 38, 42, 43, 47, 52, 54, 57, 61, 62, 67, 71, 72, 74, 75, 77, 78, 80, 84, 89, 90, 92, 93, 94, 96, 97, 98, 99, 100, 102, 103, 107, 109, 110, 111, 113, 115, 116, 117, 119, 120, 121, 122, 126, 129, 130, 132, 133, 135, 137, 138, 139, 140, 142, 143, 144, 146, 151, 152, 153, 154, 155, 156, 158, 161, 164, 166, 168, 172, 173, 175, 190, 192, 193, 194, 198, 201, 203, 207,
  • the variant ICOSL contains one or more further amino acid modification, e.g. substitution selected from M10V, M10I, V11E, S13G, E16V, S18R, A20V, S25G, F27S, F27C, N30D, Y33del, Q37R, K42E, T43A, Y47H, N52H, N52D, N52S, N52Y, N52K, N52Q, S54A, S54P, N57D, N57Y, R61S, R61C, Y62F, L67P, A71T, G72R, L74Q, R75Q, D77G, F78L, L80P, N84Q, D89G, E90A, K92R, F93L, H94E, H94D, L96F, L96I, V97A, L98F, S99G, Q100R, Q100K, Q100P, L102R, G103E, V107A, V107I,
  • the variant ICOSL contains one or more further amino acid modification, e.g. substitution selected from M10V, M10I, V11E, S13G, E16V, S18R, A20T, A20V, S25G, R26S, F27C, F27S, N30D, Y33del, Q37R, T38P, K42E, T43A, Y47H, N52A, N52C, N52D, N52G, N52H, N52K, N52L, N52M, N52P, N52Q, N52R, N52S, N52T, N52V, N52Y, S54A, S54F, S54P, N57A, N57D, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57W, N57Y, R61C, R61S, Y62F, L67P,
  • the variant ICOSL polypeptide further comprises one or more amino acid deletions corresponding to positions 140 of SEQ ID NO: 32.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from N52Y/N57Y/F138L/L203P, N52H/N57Y/Q100P, N52S/Y146C/Y152C, N52H/C198R, N52H/C140del/T225A, N52H/C198R/T225A, N52H/K92R, N57Y/Q100P, N52S/C198R, N52Y/N57Y/Y152C, N52Y/N57Y/H129P/C198R, N52H/L161P/C198R, N52S/T113E, N52S/S54P, N52K/L208P, N52S/Y152H, N52H/I143T, N52S/R75Q/L203P, N52S/D158G, N52D/Q133H, N52H/N57Y/Q100R/V110D/C198R/
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution in an reference ICOSL or specific binding fragment there of corresponding to position(s) 52, 57, or 100 with reference to numbering of SEQ ID NO:32. In some embodiments, the variant ICOSL polypeptide has one or more amino acid modification, e.g.
  • the one or more amino acid modification e.g.
  • substitution is N52Y/N57Y/F138L/L203P, N52H/N57Y/Q100P, N52S/Y146C/Y152C, N52H/C198R, N52H/C140D/T225A, N52H/C198R/T225A, N52H/K92R, N52H/S99G, N57Y/Q100P, N52S/S130G/Y152C, N52S/Y152C, N52S/C198R, N52Y/N57Y/Y152C, N52Y/N57Y/H129P/C198R, N52H/L161P/C198R, N52S/T113E, N52D/S54P, N52K/L208P, N52S/Y152H, N52D/V151A, N52H/I143T, N52S/L80P, N52S/R75Q/L203P, N52S/D158G, N52D/Q133H, N52S
  • the one or more amino acid modifications are selected from among N52Y/N57Y/F138L/L203P, N52H/N57Y/Q100P, N52S/Y146C/Y152C, N52H/C198R, N52H/C140D/T225A, N52H/C198R/T225A, N52H/K92R, N52H/S99G, N57Y/Q100P, N52S/S130G/Y152C, N52S/Y152C, N52S/C198R, N52Y/N57Y/Y152C, N52Y/N57Y/H129P/C198R, N52H/L161P/C198R, N52S/T113E, N52D/S54P, N52K/L208P, N52S/Y152H, N52D/V151A, N52H/I143T, N52S/L80P, F120S/Y152H/N201S, N52S/R75
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from N52A, N52C, N52D, N52G, N52K, N52L, N52M, N52R, N52T, N52V, N57A, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57W, Q100A, Q100D, Q100G, Q100L, Q100M, Q100N, Q100R, Q100S, Q100T or Q100V. with reference to SEQ ID NO:32.
  • the one or more amino acid modifications are selected from among N52A/N57F/Q100S, N52A/N57H/Q100S, N52A/N57Y/Q100A, N52D/N57A/Q100A, N52D/Q100S, N52G/Q100A, N52H/Q100A, N52M/N57H/Q100S, N52M/N57W/Q100P, N52Q/N57F, N52Q/N57S/Q100A, N52R/N57L/Q100A, N52R/N57Y/Q100P, N52R/N57Y/Q100S, N52S/N57A/Q100A, N52S/N57H/Q100E, N52S/N57L/Q100S, N52S/N57M/Q100S, N52S/N57Y/Q100S, N52S/N57Y/Q100M, N52S/N57Y/Q100V, N52T
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from N52H/N57Y/Q100R/C198R, N52H/N57Y/Q100R/V122A, N52H/N57Y/Q100R/F172S, N52Y/N57Y/F138L/L203P, V11E/N30D/N52H/N57Y/H94E/L96I/L98F/N194D/V210A/I218T, N52H/N57Y/Q100R/L102R/V110D/H115R/C198R, N52H/N57Y/Q100R, N52H/Q100R, N52H/N57Y/Q100R/V110D/C198R/S212G, N52H/N57Y/Q100R/L102R/V110D/H115R/C198R, E16V/N52H/N57Y/Q100R/V110D/
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from N52H/N57Y/Q100R/F172S, N52H/Q100R, or N52H/N57Y/Q100R/C198R. In some embodiments, the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from E16V/N52H/N57Y/Q100R/V1100D/H115R/Y152C/K156M/C198R, N52H/N57Y/Q100R, and N52H/N57Y/Q100P.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from N52H/N57Y/F138L/L203P, N52H/N57Y/Q100P, N52H/K92R, N52H/C140del/T225A, N52H/C198R/T225A, N52H/K92R, N57Y/Q100P, N52Y/N57Y/H129P/C198R, N52H/L161P/C198R, N52K/L208P or N52H/I143T.
  • substitution selected from N52H/N57Y/F138L/L203P amino acid modification, e.g. substitution selected from N52H/N57Y/F138L/L203P, N52H/N57Y/Q100P, N52H/K92R, N52H/C140del/T225A, N52H/C198R/T225A, N52H/K92R, N57Y/Q100P
  • the one or more amino acid modifications are selected from among F120S/Y152H/N201S, E111del, Y33del, N168Q/N207Q, N84Q/N207Q, N155Q/N207Q, N119Q/N168Q, N119Q/N207Q, N119Q/N155Q, N84Q/N119Q, N84Q/N155Q/N168Q, N84Q/N168Q/N207Q, N84Q/N155H/N207Q, N155Q/N168Q/N207Q, N119Q N155Q/N168Q, N119Q/N168Q/N207Q, N84Q/N119Q/N207Q, N119Q/N155H/N207Q, N84Q/N119Q/N155Q, N84Q/N119Q/N155Q/N168Q, N84Q/N119Q/N155Q/N168Q, N84Q/N155Q/N168Q, N84Q/N119Q/N155Q/N
  • the variant ICOSL polypeptide exhibits increased binding affinity for binding one of the ectodomains of CD28 or ICOS and exhibits decreased binding affinity for binding to the other of the ectodomains of CD28 or ICOS compared to the reference (e.g., unmodified) or wild-type ICOSL polypeptide, such as comprising the sequence set forth in SEQ ID NO: 32, 196, or 545.
  • the variant ICOSL polypeptide exhibits increased binding affinity for ICOS.
  • the one or more amino acid substitution is at a position corresponding to 16, 30, 42, 52, 54, 57, 75, 90, 92, 100, 102, 110, 113, 115, 120, 122, 133, 138, 143, 146, 152, 156, 158, 172, 194, 198, 203, 208, 221, 224, or 225.
  • the variant ICOSL contains one or more amino acid substitutions selected from C198R, D158G, E16V, E90A, F120S, F138L, F172S, H115R, H115X, I143T, I143V, I224V, K156M, K42E, K92R, L102R, L203P, L208P, N194D, N30D, N52A, N52D, N52G, N52H, N52K, N52L, N52M, N52Q, N52R, N52S, N52T, N52Y, N57F, N57H, N57K, N57L, N57M, N57P, N57S, N57V, N57W, N57Y, Q100A, Q100D, Q100E, Q100K, Q100M, Q100P, Q100P, Q100R, Q100S, Q100T, Q133H, R221I, R75Q, S54A, S54P, T113E, T
  • the variant ICOSL polypeptide has one or more amino acid substitutions selected from N52S, N52H, N52D, N52H/N57Y/Q100P, N52S/Y146C/Y152C, N52H/C198R, N52H/C198R/T225A, N52H/K92R, N57Y, N52S/C198R, N52S/T113E, S54A, N52D/S54P, N52K/L208P, N52H/I143T, N52S/R75Q/L203P, N52S/D158G, N52D/Q133H, N52H/N57Y/Q100R/V122A, N52H/N57Y/Q100R/F172S, N52H/N57Y/Q100R, N52S/N194D, N52H/N57Y/Q100R/L102R/V110D/H115R/C198R, N52S/E90A, N52S, N52S
  • the variant ICOSL polypeptide exhibits increased binding affinity for ICOS and exhibits decreased binding affinity for CD28.
  • the one or more further amino acid substitution is at a position corresponding to 52, 57, 80 100, 130, 152, 161 or 198.
  • the variant ICOSL contains one or more amino acid substitutions selected from N52S, N52H, N52Y, N52H, N57Y, L80P, Q100P Q100R, Q100K, V110D, S130G, Y152C, L161P, L161M, C198R, R221G, or a conservative amino acid substitution thereof.
  • the variant ICOSL polypeptide has one or more amino acid substitutions selected from N57Y/Q100P, N52S/S130G/Y152C, N52S/Y152C, N52Y/N57Y/Y152C, N52H/L161P/C198R, N52H/L161P/C198R, N52S/L80P, A20V/N52H/N57Y/Q100R/S109G, N52H/N57Y/R61S/Q100R/V110D/L173S, N52H/N57Y/Q100R/V107I/V110D/S132F/I154F/C198R/R221G, Q37R/N52H/N57Y/Q100R/V110N/S142F/C198R/D217V/R221G, N52H/N57Y/Q100R/V110D/C198R, F27S/N52H/N57Y/V110N, S18
  • the variant ICOSL polypeptide exhibits increased binding affinity for CD28.
  • the one or more amino acid substitution is at a position corresponding to 10, 11, 13, 16, 18, 20, 25, 27, 30, 36, 40, 41, 42, 52, 54, 57, 63, 70, 71, 72, 74, 77, 80, 81, 84, 88, 89, 90, 91, 92, 93, 94, 96, 98, 99, 100, 102, 107, 109, 110, 113, 114, 115, 117, 118, 119, 120, 121, 122, 126, 127, 129, 130, 132, 133, 135, 138, 139, 140, 143, 144, 146, 152, 153, 154, 155, 156, 158, 161, 166, 168, 169, 172, 173, 178, 190, 192, 193, 194, 198, 199, 201, 203, 207,
  • the variant ICOSL contains one or more amino acid substitutions selected from A117T, A20V, A71T, A91G, A91G, AE88D, C140del, C198R, D158G, D77G, D90K, E117G, E135K, E16V, E81A, E88D, E90A, F120I, F120S, F138L, F172S, F27C, F92Y, G72R, H115R, H115X, H129P, H94E, I118V, I127T, I143T, I143V, I154F, 1218N, I218T, I224V, K156M, K169E, K36G, K42E, K89R, K92R, K93R, L102R, L161P, L166Q, L173S, L203F, L203P, L208P, L209P, L40M, L70Q, L70R, L74Q, L80P, L96I,
  • the variant ICOSL polypeptide has one or more amino acid substitutions selected from N52S, N52H, N52D, N52Y/N57Y/F138L/L203P, N52H/N57Y/Q100P, N52S/Y146C/Y152C, N52H/C198R, N52H/C140del/T225A, N52H/C198R/T225A, N52H/K92R, N52H/S99G, N57Y, N57Y/Q100P, N52S/S130G/Y152C, N52S/Y152C, N52S/C198R, N52Y/N57Y/Y152C, N52Y/N57Y/H129P/C198R, N52H/L161P/C198R, N52S/T113E, S54A, N52D/S54P, N52K/L208P, N52S/Y152H, N52H/I143T, N52S/L80
  • the variant ICOSL polypeptide exhibits increased binding affinity for CD28 and exhibits decreased binding affinity for ICOS.
  • the one or more amino acid substitution is at a position corresponding to 52, 75 or 203.
  • the variant ICOSL contains one or more amino acid substitution selected from N52S, R75Q, L203F, or L203P.
  • the variant ICOSL polypeptide has amino acid substitutions N52S/R75Q/L203P.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution in an reference ICOSL or specific binding fragment there of corresponding to position(s) 16, 30, 42, 52, 57, 90, 100, 102, 110, 115, 120, 122, 138, 143, 152, 156, 172, 194, 198, 203, 221, or 224 with reference to numbering of SEQ ID NO: 32.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution in an reference ICOSL or specific binding fragment there of corresponding to position(s) 115, 172, or 198 with reference to numbering of SEQ ID NO:32.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from H115R, F172S or C198R.
  • the one or more amino acid modification, e.g. substitution is N52H/N57Y/Q100R/H115R/C198R, N52H/N57Y/Q100R/F172S/C198R, N52H/N57Y/Q100R/H115R/F172S/C198R, N52H/N57Y/Q100R/H115R/I143V/F172S/C198R, N52H/N57Y/Q100R/L102R/H115R/F172S/C198R, N52H/V122A/F172S/C198R, N52H/N57Y/Q100R/H115R/F172S/N194D, N52H/N57Y/H115R/F172S/C198R, N52H/N57Y/Q100R/H115R/F172S/
  • the variant ICOSL polypeptide comprises any of the extracellular domain (ECD) sequences set forth in SEQ ID NOS: 435-470.
  • the variant ICOSL polypeptide comprises a polypeptide sequence that exhibits at least 90% identity, at least 91% identity, at least 92% identity, at least 93% identity, at least 94% identity, at least 95% identity, such as at least 96% identity, 97% identity, 98% identity, or 99% identity to any of the extracellular domain (ECD) set forth in SEQ ID NOS: 435-470 and contains the amino acid modification(s), e.g. substitution(s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • the variant ICOSL polypeptide comprises a specific binding fragment of any of the extracellular domain (ECD) sequences set forth in SEQ ID NOS: 435-470 and contains the amino acid modification(s), e.g. substitution (s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • ECD extracellular domain
  • the variant ICOSL polypeptide exhibits increased binding affinity for CD28 and exhibits increased binding affinity for ICOS.
  • the one or more amino acid substitution is at a position corresponding to 16, 30, 42, 52, 54, 57, 90, 92, 100, 102, 110, 113, 115, 120, 122, 133, 138, 143, 146, 152, 156, 158, 172, 194, 198, 203, 208, 212, 224, or 225.
  • the variant ICOSL contains one or more amino acid substitutions selected from C198R, D158G, E16V, E90A, F120S, F138L, F172S, H115R, 1143V, I224V, K156M, K42E, K92R, L102R, L203P, L208P, N194D, N30D, N52A, N52D, N52G, N52H, N52K, N52L, N52M, N52Q, N52R, N52S, N52T, N52Y, N57F, N57H, N57L, N57M, N57S, N57V, N57W, N57Y, Q100A, Q100E, Q100G, Q100K, Q100M, Q100P, Q100R, Q100S, Q133H, S212G, S54A, S54P, T113E, T225A, V110D, V122A, Y146C, Y152C, or a conservative amino acid substitution thereof
  • the variant ICOSL polypeptide has one or more amino acid substitutions selected from N52A/N57Y/Q100A, N52D/Q100S, N52G/Q100A, N52M/N57H/Q100S, N52M/N57W/Q100P, N52Q/N57S/Q100A, N52R/N57L/Q100A, N52S/N57H/Q100E, N52S/N57L/Q100S, N52S/N57M/Q100S, N52S/N57Y/Q100M, N52T/N57H/Q100S, N52R/N57F/Q100P, N52R/N57F/Q100T, N52R/N57W/Q100K, N52R/N57W, N52G/N57V, N52L/N57V, N52S/N57L/Q100G, N52T/N57K/Q100P, N52S, N52H, N52D,
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution in a reference ICOSL or specific binding fragment there of corresponding to position(s) 52, 57, 100, 138, 198, or 203 with reference to numbering of SEQ ID NO:32.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from N52H, N52Y, N57Y, Q100R, Q100P, F138L, C198R, or L203P.
  • the one or more amino acid modification e.g.
  • substitution is Q100R, F138L/L203P, N52Y/F138L/L203P, N57Y/Q100R/C198R, N57Y/F138L/L203P, N52H, N57Y, N57Y/Q100P, Q100R/F138L, or L203P.
  • the variant ICOSL polypeptide comprises any of the extracellular domain (ECD) sequences set forth in SEQ ID NOS: 427-433.
  • the variant ICOSL polypeptide comprises a polypeptide sequence that exhibits at least 90% identity, at least 91% identity, at least 92% identity, at least 93% identity, at least 94% identity, at least 95% identity, such as at least 96% identity, 97% identity, 98% identity, or 99% identity to any of the extracellular domain (ECD) set forth in SEQ ID NOS: 427-433 and contains the amino acid modification(s), e.g. substitution(s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • the variant ICOSL polypeptide comprises a specific binding fragment of any of the extracellular domain (ECD) sequences set forth in SEQ ID NOS: 427-433 and contains the amino acid modification(s), e.g. substitution (s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • the variant ICOSL polypeptide comprises the IgV sequence set forth in SEQ ID NO: 434.
  • the variant ICOSL polypeptide comprises a polypeptide sequence that exhibits at least 90% identity, at least 91% identity, at least 92% identity, at least 93% identity, at least 94% identity, at least 95% identity, such as at least 96% identity, 97% identity, 98% identity, or 99% identity to the IgV sequence set forth in SEQ ID NO: 434 and contains the amino acid modification(s), e.g. substitution(s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • the variant ICOSL polypeptide comprises a specific binding fragment of the IgV sequence set forth in SEQ ID NO: 434 and contains the amino acid substitution(s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution in a reference ICOSL or specific binding fragment there of corresponding to position(s) 52, 84, 91, 119, 155, 168, 207 with reference to numbering of SEQ ID NO:32.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g. substitution selected from A91S, N52H, N52Q, N84Q, N119Q, N155H, N155Q, N168Q, N207Q.
  • the one or more amino acid modification e.g.
  • substitution is N84Q, N119Q, N168Q, N207Q, N52Q, N52Q/N207Q, N168Q/N207Q, N52Q/N168Q, N84Q/N207Q, N155Q/N207Q, N119Q/N168Q, N119Q/N207Q, N119Q/N155Q, N52Q/N84Q, N52Q/N119Q, N84Q/N119Q, N52Q/N84Q/N168Q, N52Q/N84Q/N207Q, N84Q/N155Q/N168Q, N84Q/N168Q/N207Q, N84Q/N155H/N207Q, N155Q/N168Q/N207Q, N119Q N155Q/N168Q, N119Q/N168Q/N207Q, N84Q/N119Q/N207Q, N119Q/N155H/N27Q, N4Q/N119Q1/N155Q, N52Q/N119Q/N155Q, N
  • the variant ICOSL polypeptide comprises any of the extracellular domain (ECD) sequences set forth in SEQ ID NOS: 387-424, 427-433, 435-470.
  • the variant ICOSL polypeptide comprises a polypeptide sequence that exhibits at least 90% identity, at least 91% identity, at least 92% identity, at least 93% identity, at least 94% identity, at least 95% identity, such as at least 96% identity, 97% identity, 98% identity, or 99% identity to any of the extracellular domain (ECD) set forth in SEQ ID NOS: 387-424, 427-433, 435-470 and contains the amino acid modification(s), e.g.
  • the variant ICOSL polypeptide comprises a specific binding fragment of any of the extracellular domain (ECD) sequences set forth in SEQ ID NOS: 387-424, 427-433, 435-470 and contains the amino acid modification(s), e.g. substitution (s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • the variant ICOSL polypeptide comprises any of the IgV sequences set forth in SEQ ID NOS: 425-426.
  • the variant ICOSL polypeptide comprises a polypeptide sequence that exhibits at least 90% identity, at least 91% identity, at least 92% identity, at least 93% identity, at least 94% identity, at least 95% identity, such as at least 96% identity, 97% identity, 98% identity, or 99% identity to any of the IgV sequences set forth in SEQ ID NO: 425-426 and contains the amino acid modification(s), e.g. substitution(s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • the variant ICOSL polypeptide comprises a specific binding fragment of any of the IgV sequences set forth in SEQ ID NO: 425-426 and contains the amino acid substitution(s) not present in the reference (e.g., unmodified) or wild-type ICOSL.
  • immunomodulatory polypeptide comprising a variant ICOSL can be formatted in a variety of ways, including as a soluble protein, a membrane bound protein, secreted protein, conjugate or fusion or for expression by an engineered cell or infectious agent as described elsewhere herein.
  • both immunomodulatory polypeptides comprising one or more vIgD of ICOSL or immunomodulatory polypeptides comprising multiple IgSF domains can be formatted in a variety of ways.
  • the particular format can be chosen for the desired therapeutic application.
  • an immunomodulatory polypeptide comprising a variant ICOSL polypeptide is provided in a format to antagonize or block activity of its cognate binding partner, e.g. CD28.
  • antagonism of CD28 may be useful to treat inflammation or autoimmunity.
  • an immunomodulatory polypeptide comprising a variant ICOSL polypeptide is provided in a format to agonize or stimulate activity of its cognate binding partner, e.g. CD28.
  • agonism of CD28 may be useful for treating oncology indications.
  • a skilled artisan can readily determine the activity of a particular format, such as for antagonizing or agonizing one or more specific cognate binding partner. Exemplary methods for assessing such activities are provided herein, including in the examples.
  • a soluble immunomodulatory polypeptide such as a variant ICOSL containing a vIgD
  • a liposome which itself can be conjugated to any one of or any combination of the provided conjugates (e.g., a targeting moiety).
  • the soluble or membrane bound immunomodulatory polypeptides of the invention are deglycosylated.
  • the variant ICOSL sequence is deglycosylated.
  • the IgV and/or IgC (e.g. IgC2) domain or domains of the variant ICOSL is deglycosylated.
  • Non-limiting examples of provided formats are described in FIGS. 13A-13C and further described below.
  • immunomodulatory polypeptides comprising a vIgD of ICOSL.
  • the immunomodulatory protein containing a variant ICOSL polypeptide is a soluble protein.
  • cell surface proteins typically have an intracellular, transmembrane, and extracellular domain (ECD) and that a soluble form of such proteins can be made using the extracellular domain or an immunologically active subsequence thereof.
  • ECD extracellular domain
  • the immunomodulatory protein containing a variant ICOSL polypeptide lacks a transmembrane domain or a portion of the transmembrane domain.
  • the immunomodulatory protein containing a variant ICOSL lacks the intracellular (cytoplasmic) domain or a portion of the intracellular domain. In some embodiments, the immunomodulatory protein containing the variant ICOSL polypeptide only contains the vIgD portion containing the ECD domain or a portion thereof containing an IgV domain and/or IgC (e.g. IgC2) domain or domains or specific binding fragments thereof containing the amino acid modification(s).
  • an immunomodulatory polypeptide comprising a variant ICOSL can include one or more variant ICOSL polypeptides.
  • one or more additional IgSF domain such as one or more additional vIgD, may be linked to a vIgD of ICOSL as provided herein.
  • both immunomodulatory polypeptides comprising one or more vIgD of ICOSL or immunomodulatory polypeptides comprising multiple IgSF domains can be formatted in a variety of ways, such as described in subsection C of Section III.
  • an immunomodulatory polypeptide comprising a variant ICOSL can include one or more variant ICOSL polypeptides of the invention.
  • a polypeptide of the invention will comprise exactly 1, 2, 3, 4, 5 variant ICOSL sequences.
  • at least two of the variant ICOSL sequences are identical variant ICOSL sequences.
  • the provided immunomodulatory polypeptide comprises two or more vIgD sequences of ICOSL.
  • Multiple variant ICOSL polypeptides within the polypeptide chain can be identical (i.e., the same species) to each other or be non-identical (i.e., different species) variant ICOSL sequences.
  • two, three, four, or more of the polypeptides of the invention can be covalently or non-covalently attached to each other.
  • monomeric, dimeric, and higher order (e.g., 3, 4, 5, or more) multimeric proteins are provided herein.
  • compositions comprising two or more polypeptides of the invention can be of an identical species or substantially identical species of polypeptide (e.g., a homodimer) or of non-identical species of polypeptides (e.g., a heterodimer).
  • a composition having a plurality of linked polypeptides of the invention can, as noted above, have one or more identical or non-identical variant ICOSL polypeptides of the invention in each polypeptide chain.
  • one or more additional IgSF domain may be linked to a vIgD of ICOSL as provided herein (hereinafter called a “stack” or “stacked” immunomodulatory protein).
  • the modular format of the provided immunomodulatory proteins provides flexibility for engineering or generating immunomodulatory proteins for modulating activity of multiple counterstrucutres (multiple cognate binding partners).
  • such “stack” molecules can be provided in a soluble format or, in some cases, may be provided as membrane bound or secreted proteins.
  • the immunomodulatory proteins can contain any of the variant ICOSL polypeptides provided herein linked, directly or indirectly, to one or more other immunoglobulin superfamily (IgSF) domain (“stacked” immunomodulatory protein construct and also called a “Type II” immunomodulatory protein). In some aspects, this can create unique multi-domain immunomodulatory proteins that bind two or more, such as three or more, cognate binding partners, thereby providing a multi-targeting modulation of the immune synapse.
  • IgSF immunoglobulin superfamily
  • an immunomodulatory protein comprises a combination (a “non-wild-type combination”) and/or arrangement (a “non-wild type arrangement” or “non-wild-type permutation”) of a variant ICOSL domain with one or more other affinity modified and/or non-affinity modified IgSF domain sequences of another IgSF family member (e.g. a mammalian IgSF family member) that are not found in wild-type IgSF family members.
  • the immunomodulatory protein contains 2, 3, 4, 5 or 6 immunoglobulin superfamily (IgSF) domains, where at least one of the IgSF domain is a variant ICOSL IgSF domain (vIgD of ICOSL) according to the provided description.
  • the sequences of the additional IgSF domains can be a modified IgSF domain that contains one or more amino acid modifications, e.g. substitutions, compared to a reference (e.g., unmodified) or wild-type IgSF domain.
  • the IgSF domain can be non-affinity modified (e.g., wild-type) or have been affinity modified.
  • the reference (e.g., unmodified) or wild-type IgSF domain can be from mouse, rat, cynomolgus monkey, or human origin, or combinations thereof.
  • the additional IgSF domains can be an IgSF domain of an IgSF family member set forth in Table 2.
  • the additional IgSF domain can be an affinity-modified IgSF domain containing one or more amino acid modifications, e.g. substitutions, compared to an IgSF domain contained in an IgSF family member set forth in Table 2.
  • the additional IgSF domain is an affinity or non-affinity modified IgSF domain contained in an IgSF family member of a family selected from Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, B7 family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (SIRP)
  • the additional IgSF domains are independently derived from an IgSF protein selected from the group consisting of CD80(B7-1), CD86(B7-2), CD274 (PD-L1, B7-H1), PDCD1LG2(PD-L2, CD273), ICOSLG (B7RP1, CD275, ICOSL, B7-H2), CD276(B7-H3), VTCN1(B7-H4), CD28, CTLA4, PDCD1(PD-1), ICOS, BTLA(CD272), CD4, CD8A(CD8-alpha), CD8B(CD8-beta), LAG3, HAVCR2(TIM-3), CEACAM1, TIGIT, PVR(CD155), PVRL2(CD112), CD226, CD2, CD160, CD200, CD200R1(CD200R), and NCR3 (NKp30).
  • IgSF protein selected from the group consisting of CD80(B7-1), CD86(B7-2), CD274 (PD-
  • the first column of Table 2 provides the name and, optionally, the name of some possible synonyms for that particular IgSF member.
  • the second column provides the protein identifier of the UniProtKB database, a publicly available database accessible via the internet at uniprot.org or, in some cases, the GenBank Number.
  • the Universal Protein Resource (UniProt) is a comprehensive resource for protein sequence and annotation data.
  • the UniProt databases include the UniProt Knowledgebase (UniProtKB). UniProt is a collaboration between the European Bioinformatics Institute (EMBL-EBI), the SIB Swiss Institute of Bioinformatics and the Protein Information Resource (PIR) and supported mainly by a grant from the U.S. National Institutes of Health (NIH).
  • GenBank is the NIH genetic sequence database, an annotated collection of all publicly available DNA sequences (Nucleic Acids Research, 2013 January; 41(D1):D36-42).
  • the third column provides the region where the indicated IgSF domain is located. The region is specified as a range where the domain is inclusive of the residues defining the range.
  • Column 3 also indicates the IgSF domain class for the specified IgSF region.
  • Column 4 provides the region where the indicated additional domains are located (signal peptide, S; extracellular domain, E; transmembrane domain, T; cytoplasmic domain, C). It is understood that description of domains can vary depending on the methods used to identify or classify the domain, and may be identified differently from different sources.
  • residues corresponding to a domain in Table 2 is for exemplification only and can be several amino acids (such as one, two, three or four) longer or shorter.
  • Column 5 indicates for some of the listed IgSF members, some of its cognate cell surface binding partners.
  • the provided immunomodulatory proteins in addition to containing a variant ICOSL polypeptide, also contains at least 1, 2, 3, 4, 5 or 6 additional immunoglobulin superfamily (IgSF) domains, such as an IgD domain of an IgSF family member set forth in Table 2.
  • IgSF immunoglobulin superfamily
  • the provided immunomodulatory protein contains at least one additional IgSF domain (e.g. second IgSF domain). In some embodiments, the provided immunomodulatory protein contains at least two additional IgSF domains (e.g. second and third IgSF domain). In some embodiments, the provided immunomodulatory protein contains at least three additional IgSF domains (e.g. second, third and fourth). In some embodiments, the provided immunomodulatory protein contains at least four additional IgSF domains (e.g. second, third, fourth and fifth). In some embodiments, the provided immunomodulatory protein contains at least five additional IgSF domains (e.g. second, third, fourth, fifth and sixth).
  • the provided immunomodulatory protein contains at least six additional IgSF domains (e.g. second, third, fourth, fifth, sixth and seventh).
  • each of the IgSF domains in the immunomodulatory protein are different.
  • at least one of the additional IgSF domain is the same as at least one other IgSF domain in the immunomodulatory protein.
  • each of the IgSF domains is from or derived from a different IgSF family member.
  • at least two of the IgSF domains is from or derived from the same IgSF family member.
  • the additional IgSF domain comprises an IgV domain or an IgC (e.g., IgC2) domain or domains, or a specific binding fragment of the IgV domain or a specific binding fragment of the IgC (e.g., IgC2) domain or domains.
  • the additional IgSF domain is or comprises a full-length IgV domain.
  • the additional IgSF domain is or comprises a full-length IgC (e.g., IgC2) domain or domains.
  • the additional IgSF domain is or comprises a specific binding fragment of the IgV domain.
  • the additional IgSF domain is or comprises a specific binding fragment of the IgC (e.g., IgC2) domain or domains.
  • the immunomodulatory protein contains at least two additional IgSF domains from a single (same) IgSF member.
  • the immunomodulatory protein contains an ECD or portion thereof of an IgSF member containing a full-length IgV domain and a full-length IgC (e.g., IgC2) domain or domains or specific binding fragments thereof.
  • the provided immunomodulatory proteins contain at least one additional IgSF domain (e.g. a second or, in some cases, also a third IgSF domain) in which at least one additional, e.g., second or third IgSF domain, is an IgSF domain set forth in a reference (e.g., unmodified) or wild-type IgSF domain or a specific binding fragment thereof contained in the sequence of amino acids set forth in any of SEQ ID NOS: 1-27 and 341.
  • the reference (e.g., unmodified) or wild-type IgSF domain is an IgV domain or an IgC domain, such as an IgC1 or IgC2 domain.
  • the provided immunomodulatory proteins in addition to containing a variant ICOSL polypeptide, also contains at least one additional IgSF domain (e.g. a or, in some cases, also a third affinity-modified IgSF domain and so on) in which at least one additional IgSF domain is a vIgD that contains one or more amino acid modifications (e.g. substitution, deletion or mutation) compared to an IgSF domain in a reference (e.g., unmodified) or wild-type IgSF domain, such as an IgSF domain in an IgSF family member set forth in Table 2.
  • at least one additional IgSF domain e.g. a or, in some cases, also a third affinity-modified IgSF domain and so on
  • at least one additional IgSF domain is a vIgD that contains one or more amino acid modifications (e.g. substitution, deletion or mutation) compared to an IgSF domain in a reference (e.g., unmodified) or wild-type IgSF domain, such
  • the additional, e.g., second or third affinity-modified IgSF domain comprises at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to a reference (e.g., unmodified) or wild-type IgSF domain or a specific binding fragment thereof contained in the sequence of amino acids set forth in any of SEQ ID NOS: 1-27 and 341.
  • the reference (e.g., unmodified) or wild-type IgSF domain is an IgV domain or an IgC domain, such as an IgC1 or IgC2 domain.
  • the additional, e.g., second or third IgSF domain is an affinity-modified IgV domain and/or IgC domain.
  • the one or more additional IgSF domain is an affinity-modified IgSF domain that contains an IgV domain and/or an IgC (e.g., IgC2) domain or domains, or a specific binding fragment of the IgV domain and/or a specific binding fragment of the IgC (e.g., IgC2) domain or domains, in which the IgV and/or IgC domain contains the amino acid modification(s) (e.g., substitution(s)).
  • the one or more additional affinity-modified IgSF domain contains an IgV domain containing the amino acid modification(s) (e.g. substitution(s)).
  • the one or more additional affinity-modified IgSF domain include IgSF domains present in the ECD or a portion of the ECD of the corresponding reference IgSF family member, such as a full-length IgV domain and a full-length IgC (e.g., IgC2) domain or domains, or specific binding fragments thereof, in which one or both of the IgV and IgC contain the amino acid modification(s) (e.g. substitution(s)).
  • the particular domain or each of the particular domains e.g.
  • additional, e.g., second or third IgSF domain) of a variant IgSF domain polypeptide can be several amino acids longer or shorter, such as 1-10, e.g., 1, 2, 3, 4, 5, 6 or 7 amino acids longer or shorter, than the sequence of amino acids set forth in the respective SEQ ID NO.
  • the provided immunomodulatory protein contains at least one additional, (e.g., second or, in some cases, also a third IgSF domain and so on) or second IgSF domain that is a vIgD that contains one or more amino acid substitutions compared to an IgSF domain (e.g., IgV) of a reference (e.g., unmodified) or wild-type IgSF domain other than ICOSL.
  • additional e.g., second or, in some cases, also a third IgSF domain and so on
  • second IgSF domain that is a vIgD that contains one or more amino acid substitutions compared to an IgSF domain (e.g., IgV) of a reference (e.g., unmodified) or wild-type IgSF domain other than ICOSL.
  • the additional or second IgSF domain contains one or more amino acid substitutions compared to an IgSF domain in a reference (e.g., unmodified) or wild-type IgSF domain, such as an IgSF domain in an IgSF family member set forth in Table 2.
  • the additional or second affinity-modified IgSF domain comprises at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to a reference (e.g., unmodified) or wild-type IgSF domain or a specific binding fragment thereof contained in the sequence of amino acids set forth in any of SEQ ID NOS: 1-27.
  • the reference (e.g., unmodified) or wild-type IgSF domain is an IgV domain or an IgC domain, such as an IgC1 or IgC2 domain.
  • the additional or second IgSF domain is an affinity-modified IgV domain or IgC domain.
  • Tables 3-5 provide exemplary polypeptides containing one or more affinity-modified IgSF domains that can be used in stack constructs provided herein.
  • the one or more additional IgSF domain (e.g. second IgSF) domain is an IgSF domain (e.g. IgV) of another IgSF family member that binds or recognizes a tumor antigen.
  • the IgSF family member serves as a tumor-localizing moiety, thereby bringing the vIgD of ICOSL in close proximity to immune cells in the tumor microenvironment.
  • the additional IgSF domain (e.g. second IgSF) domain is an IgSF domain of NKp30, which binds or recognizes B7-H6 expressed on a tumor cell.
  • the at least one additional (e.g. second) IgSF domain e.g.
  • NKp30 is a vIgD that contains one or more amino acid modifications (e.g. substitutions, deletions or additions).
  • the one or more amino acid modifications increase binding affinity and/or selectivity to B7-H6 compared to reference IgSF domain, e.g. NKp30, such as by at least or at least about 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold 40-fold or 50-fold.
  • NKp30 polypeptides IgC-like IgV-like ECD domain domain SEQ ID SEQ ID SEQ ID Mutation(s) NO NO NO Wild-type 54 214 929 L30V/A60V/S64P/S86G 143 215 504 L30V 144 216 930 A60V 145 217 931 S64P 146 218 932 S86G 147 219 933
  • the number of such non-affinity modified or affinity modified IgSF domains present in a “stacked” immunomodulatory protein construct is at least 2, 3, 4, or 5 and in some embodiments exactly 2, 3, 4, or 5 IgSF domains (whereby determination of the number of affinity modified IgSF domains disregards any non-specific binding fractional sequences thereof and/or substantially immunologically inactive fractional sequences thereof).
  • the number of IgSF domains is at least 2 wherein the number of affinity modified and the number of non-affinity modified IgSF domains is each independently at least: 0, 1, 2, 3, 4, 5, or 6.
  • the number of affinity modified IgSF domains and the number of non-affinity modified IgSF domains, respectively, can be exactly or at least: 2:0 (affinity modified:wild-type), 0:2, 2:1, 1:2, 2:2, 2:3, 3:2, 2:4, 4:2, 1:1, 1:3, 3:1, 1:4, 4:1, 1:5, or 5:1.
  • At least two of the non-affinity modified and/or affinity modified IgSF domains are identical IgSF domains.
  • a stacked immunomodulatory protein provided herein comprises at least two affinity modified and/or non-affinity modified IgSF domains from a single IgSF member but in a non-wild-type arrangement (alternatively, “permutation”).
  • a non-wild type arrangement or permutation is an immunomodulatory protein comprising a non-wild-type order of affinity modified and/or non-affinity modified IgSF domain sequences relative to those found in the wild-type ICOSL whose IgSF domain sequences served as the source of the variant IgSF domains as provided herein.
  • the immunomodulatory protein can comprise an IgV proximal and an IgC distal to the transmembrane domain albeit in a non-affinity modified and/or affinity modified form.
  • the presence, in an immunomodulatory protein provided herein, of both non-wild-type combinations and non-wild-type arrangements of non-affinity modified and/or affinity modified IgSF domains is also within the scope of the provided subject matter.
  • the non-affinity modified and/or affinity modified IgSF domains are non-identical (i.e., different) IgSF domains.
  • Non-identical affinity modified IgSF domains specifically bind, under specific binding conditions, different cognate binding partners and are “non-identical” irrespective of whether or not the reference (e.g., unmodified) or wild-type IgSF domains from which they are engineered was the same.
  • a non-wild-type combination of at least two non-identical IgSF domains in an immunomodulatory protein can comprise at least one IgSF domain sequence whose origin is from and unique to one ICOSL, and at least one of a second IgSF domain sequence whose origin is from and unique to another IgSF family member that is not ICOSL, wherein the IgSF domains of the immunomodulatory protein are in non-affinity modified and/or affinity modified form.
  • the two non-identical IgSF domains originate from the same IgSF domain sequence but at least one is affinity modified such that they specifically bind to different cognate binding partners.
  • a plurality of non-affinity modified and/or affinity modified IgSF domains in a stacked immunomodulatory protein polypeptide chain need not be covalently linked directly to one another.
  • an intervening span of one or more amino acid residues indirectly covalently bonds the non-affinity modified and/or affinity modified IgSF domains to each other.
  • the linkage can be via the N-terminal to C-terminal residues.
  • the linkage can be made via side chains of amino acid residues that are not located at the N-terminus or C-terminus of the non-affinity modified and/or affinity modified IgSF domain.
  • linkages can be made via terminal or internal amino acid residues or combinations thereof.
  • the two or more IgSF domain including a vIgD of ICOSL and one or more additional IgSF domain (e.g. second or third variant IgSF domain) from another IgSF family member, are covalently or non-covalently linked.
  • the two or more IgSF domains are linked directly or indirectly, such as via a linker.
  • an intervening span of one or more amino acid residues indirectly covalently bonds IgSF domains to each other.
  • the linkage can be via the N-terminal to C-terminal residues.
  • the linkage can be made via side chains of amino acid residues that are not located at the N-terminus or C-terminus of the IgSF domain(s). Thus, linkages can be made via terminal or internal amino acid residues or combinations thereof.
  • the immunomodulatory protein contains at least two IgSF domains, each linked directly or indirectly via a linker. In some embodiments, the immunomodulatory protein contains at least three immunomodulatory proteins, each linked directly or indirectly via a linker.
  • FIGS. 16A and 16B Various configurations are shown in FIGS. 16A and 16B .
  • one or more “peptide linkers” link the vIgD of ICOSL and one or more additional IgSF domain (e.g. second or third variant IgSF domain).
  • a peptide linker can be a single amino acid residue or greater in length.
  • the peptide linker has at least one amino acid residue but is no more than 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues in length.
  • the linker is a flexible linker.
  • the linker is (in one-letter amino acid code): GGGGS (“4GS”; SEQ ID NO: 636) or multimers of the 4GS linker, such as repeats of 2, 3, 4, or 5 4GS linkers.
  • the peptide linker is (GGGGS) 2 or (GGGGS) 3 as set forth in SEQ ID NOs: 229 and 228, respectively.
  • the linker also can include a series of alanine residues alone or in addition to another peptide linker (such as a 4GS linker or multimer thereof). In some embodiments, the number of alanine residues in each series is: 2, 3, 4, 5, or 6 alanines.
  • the linker is a rigid linker.
  • the linker is an ⁇ -helical linker.
  • the linker is (in one-letter amino acid code): EAAAK or multimers of the EAAAK linker, such as repeats of 2, 3, 4, or 5 EAAAK linkers, such as set forth in SEQ ID NO: 629 (1 ⁇ EAAAK), SEQ ID NO: 630 (3 ⁇ EAAAK) or SEQ ID NO: 631 (5 ⁇ EAAAK).
  • the linker can further include amino acids introduced by cloning and/or from a restriction site, for example the linker can include the amino acids GS (in one-letter amino acid code) as introduced by use of the restriction site BAMHI.
  • the linker in one-letter amino acid code is GSGGGGS (SEQ ID NO: 635). In some examples, the linker is a 2 ⁇ GGGGS followed by three alanines (GGGGSGGGGSAAA; SEQ ID NO: 230).
  • the non-affinity modified and/or affinity modified IgSF domains are linked by “wild-type peptide linkers” inserted at the N-terminus and/or C-terminus of a second non-affinity modified and/or affinity modified IgSF domains.
  • wild-type peptide linkers inserted at the N-terminus and/or C-terminus of a second non-affinity modified and/or affinity modified IgSF domains.
  • a second leading peptide linker inserted at the N-terminus of the second IgSF domain and/or a second trailing sequence inserted at the C-terminus of the second non-affinity modified and/or affinity modified IgSF domain.
  • first and second non-affinity modified and/or affinity modified IgSF domains are derived from the same parental protein and are connected in the same orientation, wild-type peptide linkers between the first and second non-affinity modified and/or affinity modified IgSF domains are not duplicated.
  • the Type II immunomodulatory protein does not comprise either the first trailing wild-type peptide linker or the second leading wild-type peptide linker.
  • the Type II immunomodulatory protein comprises a first leading wild-type peptide linker inserted at the N-terminus of the first non-affinity modified and/or affinity modified IgSF domain, wherein the first leading wild-type peptide linker comprises at least 5 (such as at least about any of 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more) consecutive amino acids from the intervening sequence in the wild-type protein from which the first non-affinity modified and/or affinity modified IgSF domain is derived between the parental IgSF domain and the immediately preceding domain (such as a signal peptide or an IgSF domain).
  • the first leading wild-type peptide linker comprises the entire intervening sequence in the wild-type protein from which the first non-affinity modified and/or affinity modified IgSF domain is derived between the parental IgSF domain and the immediately preceding domain (such as a signal peptide or an IgSF domain).
  • the Type II immunomodulatory protein further comprises a first trailing wild-type peptide linker inserted at the C-terminus of the first non-affinity modified and/or affinity modified IgSF domain, wherein the first trailing wild-type peptide linker comprises at least 5 (such as at least about any of 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more) consecutive amino acids from the intervening sequence in the wild-type protein from which the first non-affinity modified and/or affinity modified IgSF domain is derived between the parental IgSF domain and the immediately following domain (such as an IgSF domain or a transmembrane domain).
  • first trailing wild-type peptide linker comprises at least 5 (such as at least about any of 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more) consecutive amino acids from the intervening sequence in the wild-type protein from which the first non-affinity modified and/or affinity modified IgSF domain is derived between the parental IgSF domain and the immediately following domain (such as an IgSF domain or a transmembrane domain
  • the first trailing wild-type peptide linker comprises the entire intervening sequence in the wild-type protein from which the first non-affinity modified and/or affinity modified IgSF domain is derived between the parental IgSF domain and the immediately following domain (such as an IgSF domain or a transmembrane domain).
  • the Type II immunomodulatory protein further comprises a second leading wild-type peptide linker inserted at the N-terminus of the second non-affinity modified and/or affinity modified IgSF domain, wherein the second leading wild-type peptide linker comprises at least 5 (such as at least about any of 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more) consecutive amino acids from the intervening sequence in the wild-type protein from which the second non-affinity modified and/or affinity modified IgSF domain is derived between the parental IgSF domain and the immediately preceding domain (such as a signal peptide or an IgSF domain).
  • the second leading wild-type peptide linker comprises at least 5 (such as at least about any of 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more) consecutive amino acids from the intervening sequence in the wild-type protein from which the second non-affinity modified and/or affinity modified IgSF domain is derived between the parental IgSF domain and the immediately preceding domain (such as a signal peptide or an IgSF domain).
  • the second leading wild-type peptide linker comprises the entire intervening sequence in the wild-type protein from which the second non-affinity modified and/or affinity modified IgSF domain is derived between the parental IgSF domain and the immediately preceding domain (such as a signal peptide or an IgSF domain).
  • the Type II immunomodulatory protein further comprises a second trailing wild-type peptide linker inserted at the C-terminus of the second non-affinity modified and/or affinity modified IgSF domain, wherein the second trailing wild-type peptide linker comprises at least 5 (such as at least about any of 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more) consecutive amino acids from the intervening sequence in the wild-type protein from which the second non-affinity modified and/or affinity modified IgSF domain is derived between the parental IgSF domain and the immediately following domain (such as an IgSF domain or a transmembrane domain).
  • the second trailing wild-type peptide linker comprises at least 5 (such as at least about any of 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more) consecutive amino acids from the intervening sequence in the wild-type protein from which the second non-affinity modified and/or affinity modified IgSF domain is derived between the parental IgSF domain and the immediately following domain (such as an IgSF domain or a transmembrane
  • the second trailing wild-type peptide linker comprises the entire intervening sequence in the wild-type protein from which the second non-affinity modified and/or affinity modified IgSF domain is derived between the parental IgSF domain and the immediately following domain (such as an IgSF domain or a transmembrane domain).
  • Exemplary of a leading sequence and trailing sequence for a Type II protein containing a CD80 IgSF domain is set forth in SEQ ID NO:231 and SEQ ID NO:232.
  • Exemplary of a leading sequence and trailing sequence for a Type II protein containing an ICOSL IgSF domain is set forth in SEQ ID NO: 233 and 234.
  • Exemplary of a leading sequence and a trailing sequence for a Type II protein containing a CD86 IgSF domain is set forth in any of SEQ ID NOS: 236-238.
  • Exemplary of a wild-type linker sequence for a Type II protein containing an NKp30 IgSF domain is set forth in SEQ ID NO: 235.
  • immunomodulatory proteins containing a variant ICOSL polypeptide that is monovalent.
  • the variant ICOSL polypeptide of the monovalent immunomodulatory protein is linked, directly or indirectly, to a further moiety.
  • the further moiety is a protein, peptide, small molecule or nucleic acid.
  • the monovalent immunomodulatory protein is a fusion protein.
  • the moiety is a half-life extending molecule.
  • exemplary of such half-life extending molecules include, but are not limited to, albumin, an albumin-binding polypeptide, Pro/Ala/Ser (PAS), a C-terminal peptide (CTP) of the beta subunit of human chorionic gonadotropin, polyethylene glycol (PEG), long unstructured hydrophilic sequences of amino acids (XTEN), hydroxyethyl starch (HES), an albumin-binding small molecule, or a combination thereof.
  • PAS Pro/Ala/Ser
  • CTP C-terminal peptide
  • PEG polyethylene glycol
  • XTEN long unstructured hydrophilic sequences of amino acids
  • HES hydroxyethyl starch
  • albumin-binding small molecule or a combination thereof.
  • the immunomodulatory polypeptide comprising a variant ICOSL can include conformationally disordered polypeptide sequences composed of the amino acids Pro, Ala, and Ser (See e.g., WO2008/155134; SEQ ID NO: 904).
  • the amino acid repeat is at least 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 or more amino acid residues, wherein each repeat comprises (an) Ala, Ser, and Pro residue(s).
  • an immunomodulatory protein is a PASylated protein wherein the variant ICOSL polypeptide is linked, directly or indirectly via a linker, to Pro/Ala/Ser (PAS).
  • PAS Pro/Ala/Ser
  • one or more additional linker structures may be used.
  • the moiety facilitates detection or purification of the variant ICOSL polypeptide.
  • the immunomodulatory polypeptide comprises a tag or fusion domain, e.g. affinity or purification tag, linked, directly or indirectly, to the N- and/or c-terminus of the ICOSL polypeptide.
  • a tag or fusion domain e.g. affinity or purification tag
  • suitable polypeptide tags and/or fusion domains include but are not limited to, a poly-histidine (His) tag, a FLAG-tag (SEQ ID NO: 865), a Myc-tag, and fluorescent protein-tags (e.g., EGFP, set forth in SEQ ID NOs: 858, 859, or 896).
  • the immunomodulatory polypeptide comprising a variant ICOSL comprises at least six histidine residues (set forth in SEQ ID NO: 864). In some cases, the immunomodulatory polypeptide comprising a variant ICOSL further comprises various combinations of moieties. For example, the immunomodulatory polypeptide comprising a variant ICOSL further comprises one or more polyhistidine-tag and FLAG tag.
  • the ICOSL polypeptide is linked to a modified immunoglobulin heavy chain constant region (Fc) that remains in monovalent form such as set forth in SEQ ID NO: 472.
  • Fc immunoglobulin heavy chain constant region
  • the immunomodulatory protein containing a variant ICOSL is multivalent, such as bivalent.
  • the immunomodulatory protein is linked, directly or indirectly via a linker, to a multimerization domain.
  • the mutlimerization domain increase half-life of the molecule.
  • Interaction of two or more variant ICOSL polypeptides can be facilitated by their linkage, either directly or indirectly, to any moiety or other polypeptide that are themselves able to interact to form a stable structure.
  • separate encoded variant ICOSL polypeptide chains can be joined by multimerization, whereby multimerization of the polypeptides is mediated by a multimerization domain.
  • the multimerization domain provides for the formation of a stable protein-protein interaction between a first variant ICOSL polypeptide and a second variant ICOSL polypeptide.
  • Homo- or heteromultimeric polypeptides can be generated from co-expression of separate variant ICOSL polypeptides.
  • the first and second variant ICOSL polypeptides can be the same or different.
  • a multimerization domain includes any capable of forming a stable protein-protein interaction.
  • the multimerization domains can interact via an immunoglobulin sequence (e.g. Fc domain; see e.g., International Patent Pub. Nos. WO 93/10151 and WO 2005/063816 US; U.S. Pub. No. 2006/0024298; U.S. Pat. No. 5,457,035); leucine zipper (e.g.
  • a multimerization domain can include an amino acid sequence comprising a protuberance complementary to an amino acid sequence comprising a hole, such as is described, for example, in U.S. Pat. No.
  • Such a multimerization region can be engineered such that steric interactions not only promote stable interaction, but further promote the formation of heterodimers over homodimers from a mixture of chimeric monomers.
  • protuberances are constructed by replacing small amino acid side chains from the interface of the first polypeptide with larger side chains (e.g., tyrosine or tryptophan).
  • Compensatory cavities of identical or similar size to the protuberances are optionally created on the interface of the second polypeptide by replacing large amino acid side chains with smaller ones (e.g., alanine or threonine).
  • exemplary multimerization domains are described below.
  • the variant ICOSL polypeptide can be joined anywhere, but typically via its N- or C-terminus, to the N- or C-terminus of a multimerization domain to form a chimeric polypeptide.
  • the linkage can be direct or indirect via a linker.
  • the chimeric polypeptide can be a fusion protein or can be formed by chemical linkage, such as through covalent or non-covalent interactions.
  • nucleic acid encoding all or part of a variant ICOSL polypeptide can be operably linked to nucleic acid encoding the multimerization domain sequence, directly or indirectly or optionally via a linker domain.
  • the construct encodes a chimeric protein where the C-terminus of the variant ICOSL polypeptide is joined to the N-terminus of the multimerization domain.
  • a construct can encode a chimeric protein where the N-terminus of the variant ICOSL polypeptide is joined to the N- or C-terminus of the multimerization domain.
  • a polypeptide multimer contains two chimeric proteins created by linking, directly or indirectly, two of the same or different variant ICOSL polypeptides directly or indirectly to a multimerization domain.
  • the multimerization domain is a polypeptide
  • a gene fusion encoding the variant ICOSL polypeptide and multimerization domain is inserted into an appropriate expression vector.
  • the resulting chimeric or fusion protein can be expressed in host cells transformed with the recombinant expression vector, and allowed to assemble into multimers, where the multimerization domains interact to form multivalent polypeptides.
  • Chemical linkage of multimerization domains to variant ICOSL polypeptides can be effected using heterobifunctional linkers.
  • the resulting chimeric polypeptides can be purified by any suitable method such as, for example, by affinity chromatography over Protein A or Protein G columns.
  • affinity chromatography over Protein A or Protein G columns.
  • two nucleic acid molecules encoding different polypeptides are transformed into cells, formation of homo- and heterodimers will occur.
  • Conditions for expression can be adjusted so that heterodimer formation is favored over homodimer formation.
  • the immunomodulatory protein comprises a variant ICOSL polypeptide attached to an Fc region of an immunoglobulin (yielding an “immunomodulatory Fc fusion,” such as an “ICOSL-Fc variant fusion,” also termed a ICOSL vIgD-Fc fusion).
  • the ICOSL-Fc variant fusion also comprises one or more additional IgSF domain(s), such as one or more additional vIgD linked to a vIgD of ICOSL.
  • the attachment of the variant ICOSL polypeptide or additional IgSF domain is at the N-terminus of the Fc.
  • the attachment of the variant ICOSL or additional IgSF domain polypeptide is at the C-terminus of the Fc. In some embodiments, two or more ICOSL or additional IgSF domain variant polypeptides (the same or different) are independently attached at the N-terminus and at the C-terminus.
  • the Fc is murine or human Fc.
  • the Fc is a mammalian or human IgG1, IgG2, IgG3, or IgG4 Fc regions.
  • the Fc is derived from IgG1, such as human IgG1.
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 226 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 226.
  • the Fc region contains one more modifications to alter (e.g. reduce) one or more of its normal functions.
  • the Fc region is responsible for effector functions, such as complement-dependent cytotoxicity (CDC) and antibody-dependent cell cytotoxicity (ADCC), in addition to the antigen-binding capacity, which is the main function of immunoglobulins.
  • the FcRn sequence present in the Fc region plays the role of regulating the IgG level in serum by increasing the in vivo half-life by conjugation to an in vivo FcRn receptor.
  • such functions can be reduced or altered in an Fc for use with the provided Fc fusion proteins.
  • one or more amino acid modifications may be introduced into the Fc region of an ICOSL-Fc variant fusion provided herein, thereby generating an Fc region variant.
  • the Fc region variant has decreased effector function.
  • changes or mutations to Fc sequences that can alter effector function.
  • WO 2000/42072, WO2006/019447, WO2012/125850, WO2015/107026, US2016/0017041 and Shields et al. J Biol. Chem. 9(2): 6591-6604 (2001) describe exemplary Fc variants with improved or diminished binding to FcRs. The contents of those publications are specifically incorporated herein by reference.
  • the provided variant ICOSL-Fc fusions comprise an Fc region that exhibits reduced effector functions (also called inert Fc or effectorless Fc), which makes it a desirable candidate for applications in which the half-life of the ICOSL-Fc variant fusion in vivo is important yet certain effector functions (such as CDC and ADCC) are unnecessary or deleterious.
  • Fc receptor (FcR) binding assays can be conducted to ensure that the ICOSL-Fc variant fusion lacks Fc ⁇ R binding (hence likely lacking ADCC activity), but retains FcRn binding ability.
  • NK cells express Fc ⁇ RIII only, whereas monocytes express Fc ⁇ RI, Fc ⁇ RII and Fc ⁇ RIII.
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9:457-492 (1991).
  • Non-limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in U.S. Pat. No. 5,500,362 (see, e.g. Hellstrom, I. et al. Proc. Nat'l Acad. Sci. USA 83:7059-7063 (1986)) and Hellstrom, I et al., Proc.
  • non-radioactive assay methods may be employed (see, for example, ACTITM non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, Calif.; and CytoTox 96TM non-radioactive cytotoxicity assay (Promega, Madison, Wis.).
  • Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells.
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in an animal model such as that disclosed in Clynes et al. Proc. Nat'l Acad. Sci. USA 95:652-656 (1998).
  • C1q binding assays may also be carried out to confirm that the ICOSL-Fc variant fusion is unable to bind C1q and hence lacks CDC activity. See, e.g., C1q and C3c binding ELISA in WO 2006/029879 and WO 2005/100402.
  • a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J. Immunol.
  • FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, S. B. et al., Int'l. Immunol. 18(12):1759-1769 (2006)).
  • ICOSL-Fc variant fusions with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 by EU numbering (U.S. Pat. No. 6,737,056).
  • Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327 by EU numbering, including the so-called “DANA” Fc mutant with substitution of residues 265 and 297 to alanine (U.S. Pat. No. 7,332,581).
  • the Fc region of ICOSL-Fc variant fusions has an Fc region in which any one or more of amino acids at positions 234, 235, 236, 237, 238, 239, 270, 297, 298, 325, and 329 (indicated by EU numbering) are substituted with different amino acids compared to the native Fc region.
  • Such alterations of Fc region are not limited to the above-described alterations, and include, for example, alterations such as deglycosylated chains (N297A and N297Q), IgG1-N297G, IgG1-L234A/L235A, IgG1-L234A/L235E/G237A, IgG1-A325A/A330S/P331S, IgG1-C226S/C229S, IgG1-C226S/C229S/E233P/L234V/L235A, IgG1-E233P/L234V/L235A/G236del/S267K, IgG1-L234F/L235E/P331S, IgG1-S267E/L328F, IgG2-V234A/G237A, IgG2-H268Q/V309L/A330S/A331S, IgG
  • a ICOSL-Fc variant fusion comprising a variant Fc region comprising one or more amino acid substitutions which increase half-life and/or improve binding to the neonatal Fc receptor (FcRn).
  • FcRn neonatal Fc receptor
  • Antibodies with increased half-lives and improved binding to FcRn are described in US2005/0014934A1 (Hinton et al.) or WO2015107026. Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn.
  • Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434 by EU numbering, e.g., substitution of Fc region residue 434 (U.S. Pat. No. 7,371,826).
  • the Fc region of a ICOSL-Fc variant fusion comprises one or more amino acid substitution E356D and M358L by EU numbering. In some embodiments, the Fc region of a ICOSL-Fc variant fusion comprises one or more amino acid substitutions C220S, C226S, and/or C229S by EU numbering. In some embodiments, the Fc region of a ICOSL variant fusion comprises one or more amino acid substitutions R292C and V302C. See also Duncan & Winter, Nature 322:738-40 (1988); U.S. Pat. Nos. 5,648,260; 5,624,821; and WO 94/29351 concerning other examples of Fc region variants.
  • the wild-type IgG1 Fc can be the Fc set forth in SEQ ID NO: 226 having an allotype containing residues Glu (E) and Met (M) at positions 356 and 358 by EU numbering (e.g., f allotype).
  • the wild-type IgG1 Fc contains amino acids of the human G1m1 allotype, such as residues containing Asp (D) and Leu (L) at positions 356 and 358, e.g. as set forth in SEQ ID NO: 927.
  • an Fc provided herein can contain amino acid substitutions E356D and M358L to reconstitute residues of allotype G1 ml (e.g., alpha allotype).
  • a wild-type Fc is modified by one or more amino acid substitutions to reduce effector activity or to render the Fc inert for Fc effector function.
  • Exemplary effectorless or inert mutations include those described herein.
  • effectorless mutations that can be included in an Fc of constructs provided herein are L234A, L235E and G237A by EU numbering.
  • a wild-type Fc is further modified by the removal of one or more cysteine residue, such as by replacement of the cysteine residues to a serine residue at position 220 (C220S) by EU numbering.
  • cysteine residues such as by replacement of the cysteine residues to a serine residue at position 220 (C220S) by EU numbering.
  • Exemplary inert Fc regions having reduced effector function are set forth in SEQ ID NO: 633 or 477 and SEQ ID NO: 474 or 637, which are based on allotypes set forth in SEQ ID NO: 226 or SEQ ID NO: 927, respectively.
  • an Fc region used in a construct provided herein can further lack a C-terminal lysine residue.
  • alterations are made in the Fc region that result in diminished C1q binding and/or Complement Dependent Cytotoxicity (CDC), e.g., as described in U.S. Pat. No. 6,194,551, WO 99/51642, and Idusogie et al., J. Immunol. 164: 4178-4184 (2000).
  • CDC Complement Dependent Cytotoxicity
  • a ICOSL-Fc variant fusion comprising a variant Fc region comprising one or more amino acid modifications, wherein the variant Fc region is derived from IgG1, such as human IgG.
  • the variant Fc region is derived from the amino acid sequence set forth in SEQ ID NO: 226.
  • the Fc exhibits reduced effector function.
  • the Fc contains at least one amino acid substitution that is N82G by numbering of SEQ ID NO: 226 (corresponding to N297G by EU numbering).
  • the Fc further contains at least one amino acid substitution that is R77C or V87C by numbering of SEQ ID NO: 226 (corresponding to R292C or V302C by EU numbering).
  • the variant Fc region further comprises a C5S amino acid modification by numbering of SEQ ID NO: 226 (corresponding to C220S by EU numbering).
  • the variant Fc region comprises the following amino acid modifications: V297G and one or more of the following amino acid modifications C220S, R292C or V302C by EU numbering (corresponding to N82G and one or more of the following amino acid modifications C5S, R77C or V87C with reference to SEQ ID NO:226), e.g.
  • the Fc region comprises the sequence set forth in SEQ ID NO:476.
  • the variant Fc region comprises one or more of the amino acid modifications C220S, L234A, L235E or G237A, e.g. the Fc region comprises the sequence set forth in SEQ ID NO:477.
  • the variant Fc region comprises one or more of the amino acid modifications C220S, E233P, L234V, L235A, G236del or S267K, e.g. the Fc region comprises the sequence set forth in SEQ ID NO:478.
  • the variant Fc comprises one or more of the amino acid modifications C220S, L234A, L235E, G237A, E356D or M358L, e.g. the Fc region comprises the sequence set forth in SEQ ID NO:474.
  • the Fc region lacks the C-terminal lysine corresponding to position 232 of the reference (e.g., unmodified) or wild-type Fc set forth in SEQ ID NO: 56 (corresponding to K447del by EU numbering).
  • the C-terminal lysine may be differentially removed during biosynthesis, removal of the C-terminal lysine residue results in a more homogenous product when the protein is expressed in cells.
  • such an Fc region can additionally include one or more additional modifications, e.g. amino acid substitutions, such as any as described. Exemplary of such an Fc region is set forth in SEQ ID NO: 632, 633, 634, or 637.
  • a ICOSL-Fc variant fusion comprising a variant Fc region in which the variant Fc comprises the sequence of amino acids set forth in any of SEQ ID NOS:474, 476, 477, 478, 507, 632, 633, 634, or 637 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any of SEQ ID NOS: 474, 476, 477, 478, 507, 632, 633, 634, or 637.
  • the Fc exhibits reduced effector function.
  • the Fc is derived from IgG2, such as human IgG2.
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 227 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 227.
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 505 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 505.
  • the IgG4 Fc is a stabilized Fc in which the CH3 domain of human IgG4 is substituted with the CH3 domain of human IgG1 and which exhibits inhibited aggregate formation, an antibody in which the CH3 and CH2 domains of human IgG4 are substituted with the CH3 and CH2 domains of human IgG1, respectively, or an antibody in which arginine at position 409 indicated in the EU index proposed by Kabat et al. of human IgG4 is substituted with lysine and which exhibits inhibited aggregate formation (see e.g. U.S. Pat. No. 8,911,726).
  • the Fc is an IgG4 containing the S228P mutation, which has been shown to prevent recombination between a therapeutic antibody and an endogenous IgG4 by Fab-arm exchange (see e.g. Labrijin et al. (2009) Nat. Biotechnol., 27(8) 767-71.)
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 506 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 506.
  • the variant ICOSL polypeptide is directly linked to the Fc sequence. In some embodiments, the variant ICOSL polypeptide is indirectly linked to the Fc sequence, such as via a linker. In some embodiments, one or more “peptide linkers” link the variant ICOSL polypeptide and the Fc domain. In some embodiments, a peptide linker can be a single amino acid residue or greater in length. In some embodiments, the peptide linker has at least one amino acid residue but is no more than 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues in length. In some embodiments, the linker is three alanines (AAA).
  • the linker is (in one-letter amino acid code): GGGGS (“4GS”; SEQ ID NO:636) or multimers of the 4GS linker, such as repeats of 2, 3, 4, 5 or 6 4GS linkers, such as set forth in SEQ ID NO: 229 (2 ⁇ GGGGS) or SEQ ID NO: 228 (3 ⁇ GGGGS).
  • the linker is a rigid linker.
  • the linker is an ⁇ -helical linker.
  • the linker is (in one-letter amino acid code): EAAAK or multimers of the EAAAK linker, such as repeats of 2, 3, 4, or 5 4GS linkers, such as set forth in SEQ ID NO: 629 (EAAAK) or SEQ ID NO: 630 (3 ⁇ EAAAK) or SEQ ID NO: 631 (5 ⁇ EAAAK).
  • linkers start with one or more EAAAK units and can be lengthened by addition of A, AA, AAA, AAAA, EAAAA and EAAAK sequences.
  • the linker can further include amino acids introduced by cloning and/or from a restriction site, for example the linker can include the amino acids GS (in one-letter amino acid code) as introduced by use of the restriction site BAMHI.
  • the linker in one-letter amino acid code
  • the linker is GSGGGGS (SEQ ID NO: 635).
  • the linker is a 2 ⁇ GGGGS followed by three alanines (GGGGSGGGGSAAA; SEQ ID NO: 230).
  • the variant ICOSL-Fc fusion protein is a dimer formed by two variant ICOSL Fc polypeptides linked to an Fc domain.
  • identical or substantially identical species (allowing for 3 or fewer N-terminus or C-terminus amino acid sequence differences) of ICOSL-Fc variant fusion polypeptides will be dimerized to create a homodimer.
  • the dimer is a homodimer in which the two variant ICOSL Fc polypeptides are the same.
  • different species of ICOSL-Fc variant fusion polypeptides can be dimerized to yield a heterodimer.
  • the dimer is a heterodimer in which the two variant ICOSL Fc polypeptides are different.
  • a variant ICOSL-Fc fusion protein containing a variant ICOSL polypeptide that includes one or more amino acid modifications in a reference ICOSL as described in Section II that is linked, directly or indirectly, to an Fc region.
  • the C-terminus of the variant ICOSL polypeptide is joined to the N-terminus of the Fc region.
  • the variant ICOSL of an ICOSL-Fc fusion contains one or more amino acid modifications in the sequence of amino acids the reference IgV domain set forth in SEQ ID NO:545.
  • such an immunomodulatory protein contains variant ICOSL polypeptide containing an IgV domain, such as an IgV domain set forth in any one of SEQ ID NOS: 197-199, 201-208, 210, 212, 240, 326-340, 382-386, 425-426, 434, 546-599, 686-857, 906-907, 909-910, or an IgV domain that has at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% to any of SEQ ID NOS: 197-199, 201-208, 210, 212, 240, 326-340, 382-386, 425-426, 434, 546-599, 686-857, 906-907, 909-910 and contains the one or more amino acid modifications of the respective SEQ ID NO.
  • an IgV domain such as an IgV domain set forth in any one of SEQ ID NOS
  • the variant ICOSL polypeptide has an IgSF domain (e.g. IgV domain) that exhibits increased binding affinity to CD28 or ICOS, such as any of the amino acid modifications described herein.
  • the variant ICOSL polypeptide has an IgSF domain (e.g. IgV domain) containing one or more amino acid modification, e.g. substitution in an reference ICOSL or specific binding fragment, corresponding to position(s) 52, 57, or 100 with reference to numbering of SEQ ID NO: 32.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g.
  • Exemplary of such variant molecules include any as described herein.
  • the variant ICOSL polypeptide contains the amino acid modifications N52H/N57Y/Q100R (e.g. is or includes an IgV domain set forth in SEQ ID NO: 565). In some embodiments, the variant ICOSL polypeptide contains the amino acid modifications N52D (e.g. is or includes an IgV domain set forth in SEQ ID NO: 548). In some embodiments, the variant ICOSL polypeptide contains the amino acid modifications N52H/Q100R (e.g. is or includes an IgV domain set forth in SEQ ID NO: 567). In some embodiments, the variant ICOSL polypeptide contains the amino acid modifications N52L/N57H/Q100R (e.g.
  • the variant ICOSL polypeptide contains the amino acid modifications N52H/N57Y/Q100P (e.g. is or includes an IgV domain set forth in SEQ ID NO: 570).
  • the Fc polypeptide is a variant of a human IgG1 Fc region that exhibits reduced effector functions, such as any as described.
  • the Fc region is a human IgG1 that contains the amino acid modifications N297G, E233P/L234V/L235A/G236del/S267K or L234A/L235E/G237A, wherein the residue is numbered according to the EU index of Kabat.
  • the variant IgG1 Fc region further contains the amino acid substitution C220S, wherein the residues are numbered according to the EU index of Kabat.
  • the Fc region contains K447del, wherein the residue is numbered according to the EU index of Kabat.
  • the Fc region contains the sequence of amino acid sequence set forth in any of SEQ ID NOS: 474, 476, 477, 478, 633 or 637 or a sequence of amino acids that exhibits at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any of SEQ ID NOS: 474, 476, 477, 478, 633 or 637 and contains the amino acid substitutions of the respective SEQ ID NO.
  • the linkage between the variant ICOSL IgSF e.g.
  • IgV polypeptide and the Fc can be via a peptide linker, such as any as described.
  • the linker is GGGGS (“4GS”; SEQ ID NO: 636), SEQ ID NO: 229 (2 ⁇ GGGGS) or SEQ ID NO: 228 (3 ⁇ GGGGS).
  • the C-terminus of the variant ICOSL polypeptide is joined to the N-terminus of the Fc region, such that the order of components is variant ICOSL-linker-Fc.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO:565, a linker set forth in SEQ ID NO:636 and an Fc polypeptide set forth in SEQ ID NO: 637.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO:565, a linker set forth in SEQ ID NO:636 and an Fc polypeptide set forth in SEQ ID NO: 474.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO:565, a linker set forth in SEQ ID NO:636 and an Fc polypeptide set forth in SEQ ID NO: 477.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO:565, a linker set forth in SEQ ID NO:636 and an Fc polypeptide set forth in SEQ ID NO: 633.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO:565, a linker set forth in SEQ ID NO: 229 and an Fc polypeptide set forth in SEQ ID NO: 637.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO:565, a linker set forth in SEQ ID NO:229 and an Fc polypeptide set forth in SEQ ID NO: 474.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO:565, a linker set forth in SEQ ID NO: 229 and an Fc polypeptide set forth in SEQ ID NO: 477.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO:565, a linker set forth in SEQ ID NO:229 and an Fc polypeptide set forth in SEQ ID NO: 633.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO:565, a linker set forth in SEQ ID NO:228 and an Fc polypeptide set forth in SEQ ID NO: 637.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO:565, a linker set forth in SEQ ID NO: 228 and an Fc polypeptide set forth in SEQ ID NO: 474.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO:565, a linker set forth in SEQ ID NO:228 and an Fc polypeptide set forth in SEQ ID NO: 477.
  • a variant ICOSL-Fc fusion protein e.g. variant ICOSL-linker-Fc, containing a variant ICOSL IgV domain set forth in SEQ ID NO: 565, a linker set forth in SEQ ID NO:228 and an Fc polypeptide set forth in SEQ ID NO: 633.
  • a variant ICOSL IgSF Fc fusion protein that has the sequence of amino acids set forth in SEQ ID NO: 928, or a sequence of amino acids that exhibits at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% to SEQ ID NO:928.
  • the variant ICOSL IgSF Fc fusion protein binds to CD28 and ICOS, such as with increased binding affinity compared to reference (wild-type) ICOSL-Fc fusion protein.
  • the variant ICOSL IgSF Fc fusion exhibits reduced Fc effector function compared to fusion with an Fc of a wild-type human IgG1.
  • a multi-domain stack immunomodulatory protein in which two or more IgSF domain, including a vIgD of ICOSL and one or more additional IgSF domain (e.g. second variant IgSF domain) from another IgSF family member, are linked or attached to an Fc to form an Fc fusion, which, upon expression in a cell can, in some aspects, produce a dimeric multi-domain stack immunomodulatory protein.
  • additional IgSF domain e.g. second variant IgSF domain
  • the variant ICOSL polypeptide and one or more additional IgSF domain are independently linked, directly or indirectly, to the N- or C-terminus of an Fc region. In some embodiments, the variant ICOSL polypeptide and at least one of the one or more additional IgSF domain are linked, directly or indirectly, and one of the variant ICOSL or and one of the one or more additional IgSF domain is also linked, directly or indirectly, to the N- or C-terminus of an Fc region.
  • the N- or C-terminus of the Fc region is linked to the variant ICOSL polypeptide or the one or more additional IgSF domain and the other of the N- or C-terminus of the Fc region is linked to the other of the ICOSL variant or another of the one or more additional IgSF domain.
  • linkage to the Fc is via a peptide linker, e.g. a peptide linker, such as described above.
  • linkage between the variant ICOSL and second IgSF domain is via a peptide linker, e.g. a peptide linker, such as described above.
  • linkage between the variant ICOSL and the one or more additional IgSF domain is via a peptide linker, e.g., a peptide linker, such as described above.
  • a peptide linker e.g., a peptide linker, such as described above.
  • the vIgD of ICOSL, the one or more additional IgSF domains, and the Fc domain can be linked together in any of numerous configurations as depicted in FIGS. 16A and 16B .
  • the ICOSL-Fc variant fusion can further contain a signal peptide, such as an exemplary signal peptide as contained in the sequence of amino acids set forth in SEQ ID NO: 59 or 225. Exemplary configurations are described in the Examples.
  • the stacked immunomodulatory protein is a dimer formed by two immunomodulatory Fc fusion polypeptides. Also provided are nucleic acid molecules encoding any of the stacked immunomodulatory proteins. In some embodiments, the dimeric multi-domain stack immunomodulatory protein can be produced in cells by expression, or in some cases co-expression, of stack immunomodulatory Fc region polypeptides, such as described further below.
  • the dimeric multi-domain stack immunomodulatory protein is divalent for each Fc subunit, monovalent for each subunit, or divalent for one subunit and tetravalent for the other.
  • the dimeric multi-domain stack immunomodulatory protein is a homodimeric multi-domain stack Fc protein. In some embodiments, the dimeric multi-domain stack immunomodulatory protein comprises a first stack immunomodulatory Fc fusion polypeptide and a second stack immunomodulatory Fc fusion polypeptide in which the first and second polypeptide are the same. In some embodiments, the Fc portion of the polypeptide can be any Fc as described above.
  • the multi-domain stack molecule contains a first Fc fusion polypeptide containing a variant ICOSL and a second fusion IgSF domain and a second Fc polypeptide containing the variant ICOSL and the second IgSF domain. In some embodiments, the multi-domain stack molecule contains a first Fc fusion polypeptide containing a variant ICOSL and a second IgSF domain, and a third IgSF domain and a second Fc fusion polypeptide containing the variant ICOSL, the second IgSF domain, and the third IgSF domain.
  • the Fc portion of the first and/or second fusion polypeptide can be any Fc as described above. In some embodiments, the Fc portion or region of the first and second fusion polypeptide is the same.
  • an immunomodulatory protein that is a ICOSL-NKp30 multi-domain stack containing any of the variant ICOSL polypeptides and one or more IgF domains of NKp30, e.g. wild-type or unmodified NKp30, such as an IgV domain set forth in SEQ ID NO: 929 or an ECD or a binding portion thereof set forth in SEQ ID NO: 215 or a binding portion thereof.
  • the one or more amino acid modifications include one or more of L30V, A60V, S64P, S86G, such as 1, 2, 3 or 4 of such amino acid modifications.
  • a variant NKp30 of the multi-domain stack polypeptides is or includes a variant IgV domain, such as a variant IgV domain set forth in any of SEQ ID NOS: 504, 930, 931, 932 or 933, or an IgV domain that has at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% to any of SEQ ID NOS:504, 930, 931, 932 or 933 and contains the one or more amino acid modifications of the respective SEQ ID NO.
  • a variant NKp30 of the multi-domain stack polypeptides is or includes a variant ECD domain, such as a variant ECD set forth in any of SEQ ID NOS: 215, 216, 217, 218 or 219, or an ECD domain that has at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% to any of SEQ ID NOS: 215, 216, 217, 218 or 219 and contains the one or more amino acid modifications of the respective SEQ ID NO.
  • a variant ECD domain such as a variant ECD set forth in any of SEQ ID NOS: 215, 216, 217, 218 or 219, or an ECD domain that has at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% to any of SEQ ID NOS: 215,
  • the variant ICOSL polypeptide can include any described in Section II containing a variant IgSF domain (e.g. IgV or ECD), such as including any of the amino acid modifications set forth in a Table 1.
  • a variant IgSF domain e.g. IgV or ECD
  • such an immunomodulatory protein contains a variant ICOSL polypeptide containing an ECD domain, such as an ECD domain set forth in any one of SEQ ID NOS: 109-142, 239, 280-325, 364-381, 387-424, 427-433, 435-470, 638-685, 905, 908, or an ECD domain that has at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% to any of SEQ ID NOS: 109-142, 239, 280-325, 364-381, 387-424, 427-433, 435-470, 638-685, 905, 908 and contains the one or more amino acid modifications of the respective SEQ ID NO.
  • an ECD domain such as an ECD domain set forth in any one of SEQ ID NOS: 109-142, 239, 280-325, 364-381, 387-424, 427-433
  • such an immunomodulatory protein contains a variant ICOSL polypeptide containing an IgV domain, such as an IgV domain set forth in any one of SEQ ID NOS: 197-199, 201-208, 210, 212, 240, 326-340, 382-386, 425-426, 434, 546-599, 686-857, 906-907, 909-910, or an IgV domain that has at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% to any of SEQ ID NOS: 197-199, 201-208, 210, 212, 240, 326-340, 382-386, 425-426, 434, 546-599, 686-857, 906-907, 909-910 and contains the one or more amino acid modifications of the respective SEQ ID NO.
  • an IgV domain such as an IgV domain set forth in any one of SEQ ID
  • the variant ICOSL polypeptide has an IgSF domain (e.g. IgV domain) exhibits increased binding affinity to CD28 or ICOS, such as any described herein.
  • the variant ICOSL polypeptide has an IgSF domain (e.g. IgV domain) containing one or more amino acid modification, e.g. substitution in an reference ICOSL or specific binding fragment, corresponding to position(s) 52, 57, or 100 with reference to numbering of SEQ ID NO:32.
  • the variant ICOSL polypeptide has one or more amino acid modification, e.g.
  • Exemplary of such variant molecules include any as described herein.
  • the variant ICOSL polypeptide contains the amino acid modifications N52D (e.g. is or includes an IgV domain set forth in SEQ ID NO:548), N52H/Q100R (e.g. is or includes an IgV domain set forth in SEQ ID NO:567), N52H/N57Y/Q100R (e.g. is or includes an IgV domain set forth in SEQ ID NO:565), or N52L/N57H/Q100R (e.g. is or includes an IgV domain set forth in SEQ ID NO:761).
  • N52D e.g. is or includes an IgV domain set forth in SEQ ID NO:548
  • N52H/Q100R e.g. is or includes an IgV domain set forth in SEQ ID NO:567
  • N52H/N57Y/Q100R e.g. is or includes an IgV domain set forth in SEQ ID NO:565
  • N52L/N57H/Q100R e.
  • the provided multi-domain stack immunomodulatory proteins such as an ICOSL-NKp30 multi-domain stack immunomodulatory protein
  • the Fc polypeptide is a variant of a human IgG1 Fc region that exhibits reduced effector functions, such as any as described.
  • the Fc region is a human IgG1 that contains the amino acid modifications N297G, E233P/L234V/L235A/G236del/S267K or L234A/L235E/G237A, wherein the residue is numbered according to the EU index of Kabat.
  • the variant IgG1 Fc region further contains the amino acid substitution C220S, wherein the residues are numbered according to the EU index of Kabat.
  • the Fc region contains K447del, wherein the residue is numbered according to the EU index of Kabat.
  • the Fc region contains the sequence of amino acid sequence set forth in any of SEQ ID NOS: 474, 476, 477, 478, 633 or 637 or a sequence of amino acids that exhibits at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any of SEQ ID NOS: 474, 476, 477, 478, 633 or 637 and contains the amino acid substitutions of the respective SEQ ID NO.
  • any of the provided ICOSL-NKp30 multi-domain stack immunomodulatory polypeptides can contain two copies of a polypeptide having the structure: variant ICOSL IgSF (e.g. IgV, such as set forth in SEQ ID NO: 548, 565, 567 or 761)—linker 1—variant NKp30 IgSF (e.g. IgV, such as set forth in SEQ ID NO: 504)—linker 2—Fc.
  • variant ICOSL IgSF e.g. IgV, such as set forth in SEQ ID NO: 548, 565, 567 or 761
  • linker 1 variant ICOSL IgSF
  • variant NKp30 IgSF e.g. IgV, such as set forth in SEQ ID NO: 504
  • linker 2 Fc.
  • any of the provided ICOSL-NKp30 multi-domain stack immunomodulatory polypeptides can contain two copies of a polypeptide having the structure: variant ICOSL IgSF (e.g. IgV, such as set forth in SEQ ID NO: 548, 565, 567 or 761)—linker 1—variant NKp30 IgSF (e.g. IgV, such as set forth in SEQ ID NO:504)—linker 1—variant NKp30 IgSF (e.g. IgV, such as set forth in SEQ ID NO:504)—linker 2—Fc.
  • linker 1 and linker 2 are peptide linkers, such as any as described.
  • linker 1 and linker 2 are the same. In some embodiments, linker 1 and linker 2 are different. In some embodiments, linker 1 is 3 ⁇ GGGGS (SEQ ID NO: 228). In some embodiments, linker 2 is GSGGGS (SEQ ID NO: 635).
  • Exemplary ICOSL-NKp30 multidomain stacks have the sequence of amino acids set forth in any of SEQ ID NOS: 912, 914, 916, 918, 920, 922, 924 or 926, or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% to any of SEQ ID NOS: 912, 914, 916, 918, 920, 922, 924 or 926.
  • any of the provided ICOSL-NKp30 multi-domain stack immunomodulatory proteins bind to ICOS and/or CD28 and bind to B7-H6.
  • the provided ICOSL-NKp30 multi-domain stack immunomodulatory proteins provide for a binding molecule capable of tumor localization adjacent to an immune cell that expresses ICOS and/or CD28 (e.g. a T cell).
  • such ICOSL-NKp30 multi-domain stack immunomodulatory proteins can be used to increase an immune response by engagement of ICOS and/or CD28 costimulatory receptors on T cells in a tumor microenvironment.
  • such ICOSL-NKp30 multi-domain stack immunomodulatory proteins, or pharmaceutical compositions thereof can be used to treat a tumor or cancer.
  • the multi-domain stack molecule is heterodimeric, comprising two different Fc fusion polypeptides, e.g. a first and a second Fc polypeptide, wherein at least one is an Fc fusion polypeptide containing at least one variant ICOSL polypeptide and/or at least one is an Fc polypeptide containing a second IgSF domain (e.g. second variant IgSF domain).
  • the first or second Fc fusion polypeptide further contains a third IgSF domain (e.g. third variant IgSF domain).
  • the multi-domain stack molecule contains a first Fc fusion polypeptide containing a variant ICOSL and a second Fc fusion polypeptide containing at a second IgSF domain, in which, in some cases, the first or second Fc fusion polypeptide additionally contains a third IgSF domain.
  • the multi-domain stack molecule contains a first Fc fusion polypeptide containing a variant ICOSL, a second IgSF domain, and in some cases, a third IgSF domain and a second Fc fusion polypeptide that is not linked to either a variant ICOSL polypeptide or an additional IgSF domain.
  • the Fc portion or region of the first and second fusion polypeptide is the same.
  • the Fc portion or region of the first and second fusion polypeptide is different.
  • the multi-domain stack molecule contains a first fusion Fc polypeptide containing 1, 2, 3, 4 or more variant ICOSL polypeptides and/or 1, 2, 3, 4 or more additional IgSF domains, wherein the total number of IgSF domains in the first stack Fc fusion polypeptide is greater than 2, 3, 4, 5, 6 or more.
  • the second stack Fc fusion polypeptide contains 1, 2, 3, 4 or more variant ICOSL polypeptides and/or 1, 2, 3, 4 or more second IgSF domains, wherein the total number of IgSF domains in the second stack Fc fusion polypeptide is greater than 2, 3, 4, 5, 6 or more.
  • the second Fc fusion polypeptide is not linked to either a variant ICOSL polypeptide or additional IgSF domain.
  • the heterodimeric stack molecule contains a first stack immunomodulatory Fc fusion polypeptide and a second stack immunomodulatory Fc fusion polypeptide in which the first and second polypeptide are different.
  • a heterodimeric stack molecule contains a first Fc polypeptide fusion containing an Fc region and a first variant ICOSL polypeptide and/or second IgSF domain (e.g. second variant IgSF domain) and a second Fc polypeptide fusion containing an Fc region and the other of the first variant ICOSL polypeptide or the second IgSF domain.
  • a heterodimeric stack molecule contains a first Fc polypeptide fusion containing an Fc region and a first variant ICOSL polypeptide and/or second IgSF domain (e.g. second variant IgSF domain) and a second Fc polypeptide fusion containing both the first variant ICOSL polypeptide and second IgSF domain (e.g. second variant IgSF domain) but in a different orientation or configuration from the first Fc region.
  • the first and/or second Fc fusion polypeptide also contains a third IgSF domain (e.g. third variant IgSF domain).
  • the Fc domain of one or both of the first and second stacked immunomodulatory Fc fusion polypeptide comprises a modification (e.g. substitution) such that the interface of the Fc molecule is modified to facilitate and/or promote heterodimerization.
  • modifications include introduction of a protuberance (knob) into a first Fc polypeptide and a cavity (hole) into a second Fc polypeptide such that the protuberance is positionable in the cavity to promote complexing of the first and second Fc-containing polypeptides.
  • Amino acids targeted for replacement and/or modification to create protuberances or cavities in a polypeptide are typically interface amino acids that interact or contact with one or more amino acids in the interface of a second polypeptide.
  • a sequence of amino acids is added preceding the Fc sequence for constructs in which the Fc sequence is the N-terminal portion of the sequence.
  • the sequence of amino acids HMSSVSAQ (SEQ ID NO:475) is added immediately preceding the Fc sequence for constructs in which the Fc sequence is the N-terminal portion of the sequence.
  • a heterodimeric stack molecule contains a first Fc polypeptide fusion containing an Fc region (knob) and a first variant ICOSL polypeptide and/or second IgSF domain (e.g.
  • a first polypeptide that is modified to contain protuberance (hole) amino acids include replacement of a native or original amino acid with an amino acid that has at least one side chain which projects from the interface of the first polypeptide and is therefore positionable in a compensatory cavity (hole) in an adjacent interface of a second polypeptide.
  • the replacement amino acid is one which has a larger side chain volume than the original amino acid residue.
  • the replacement residues for the formation of a protuberance are naturally occurring amino acid residues and include, for example, arginine (R), phenylalanine (F), tyrosine (Y), or tryptophan (W).
  • the original residue identified for replacement is an amino acid residue that has a small side chain such as, for example, alanine, asparagine, aspartic acid, glycine, serine, threonine, or valine.
  • a second polypeptide that is modified to contain a cavity is one that includes replacement of a native or original amino acid with an amino acid that has at least one side chain that is recessed from the interface of the second polypeptide and thus is able to accommodate a corresponding protuberance from the interface of a first polypeptide.
  • the replacement amino acid is one which has a smaller side chain volume than the original amino acid residue.
  • the replacement residues for the formation of a cavity are naturally occurring amino acids and include, for example, alanine (A), serine (S), threonine (T) and valine (V).
  • the original amino acid identified for replacement is an amino acid that has a large side chain such as, for example, tyrosine, arginine, phenylalanine, or typtophan.
  • the CH3 interface of human IgG1 involves sixteen residues on each domain located on four anti-parallel ⁇ -strands which buries 1090 ⁇ 2 from each surface (see e.g., Deisenhofer et al. (1981) Biochemistry, 20:2361-2370; Miller et al., (1990) J Mol. Biol., 216, 965-973; Ridgway et al., (1996) Prot. Engin., 9: 617-621; U.S. Pat. No. 5,731,168).
  • Modifications of a CH3 domain to create protuberances or cavities are described, for example, in U.S. Pat. No.
  • modifications of a CH3 domain to create protuberances or cavities are typically targeted to residues located on the two central anti-parallel ⁇ -strands. The aim is to minimize the risk that the protuberances which are created can be accommodated by protruding into the surrounding solvent rather than being accommodated by a compensatory cavity in the partner CH3 domain.
  • the heterodimeric molecule contains a T366W mutation in the CH3 domain of the “knobs chain” and T366S, L368A, Y407V mutations in the CH3 domain of the “hole chain”.
  • an additional interchain disulfide bridge between the CH3 domains can also be used (Merchant, A. M., et al., Nature Biotech. 16 (1998) 677-681) e.g. by introducing a Y349C mutation into the CH3 domain of the “knobs” or “hole” chain and a E356C mutation or a S354C mutation into the CH3 domain of the other chain.
  • the heterodimeric molecule contains S354C, T366W mutations in one of the two CH3 domains and Y349C, T366S, L368A, Y407V mutations in the other of the two CH3 domains. In some embodiments, the heterodimeric molecule comprises E356C, T366W mutations in one of the two CH3 domains and Y349C, T366S, L368A, Y407V mutations in the other of the two CH3 domains.
  • the heterodimeric molecule comprises Y349C, T366W mutations in one of the two CH3 domains and E356C, T366S, L368A, Y407V mutations in the other of the two CH3 domains. In some embodiments, the heterodimeric molecule comprises Y349C, T366W mutations in one of the two CH3 domains and S354C, T366S, L368A, Y407V mutations in the other of the two CH3 domains. Examples of other knobs-in-holes technologies are known in the art, e.g. as described by EP 1 870 459 A1.
  • the Fc regions of the heterodimeric molecule additionally can contain one or more other Fc mutation, such as any described above.
  • the heterodimer molecule contains an Fc region with a mutation that reduces effector function.
  • an Fc variant containing CH3 protuberance (knob) or cavity (hole) modifications can be joined to a stacked immunomodulatory polypeptide anywhere, but typically via its N- or C-terminus, to the N- or C-terminus of a first and/or second stacked immunomodulatory polypeptide, such as to form a fusion polypeptide.
  • the linkage can be direct or indirect via a linker.
  • a knob and hole molecule is generated by co-expression of a first stacked immunomodulatory polypeptide linked to an Fc variant containing CH3 protuberance modification(s) with a second stacked immunomodulatory polypeptide linked to an Fc variant containing CH3 cavity modification(s).
  • nucleic acid molecules encoding the variant ICOSL-Fc fusion protein are also provided.
  • a nucleic acid molecule encoding a variant ICOSL-Fc fusion protein is inserted into an appropriate expression vector.
  • the resulting variant ICOSL-Fc fusion protein can be expressed in host cells transformed with the expression where assembly between Fc domains occurs by interchain disulfide bonds formed between the Fc moieties to yield dimeric, such as divalent, variant ICOSL-Fc fusion proteins.
  • the resulting Fc fusion proteins can be easily purified by affinity chromatography over Protein A or Protein G columns.
  • additional steps for purification can be necessary.
  • the formation of heterodimers must be biochemically achieved since variant ICOSL molecules carrying the Fc-domain will be expressed as disulfide-linked homodimers as well.
  • homodimers can be reduced under conditions that favor the disruption of interchain disulfides, but do no effect intra-chain disulfides.
  • different variant-ICOSL Fc monomers are mixed in equimolar amounts and oxidized to form a mixture of homo- and heterodimers. The components of this mixture are separated by chromatographic techniques.
  • the formation of this type of heterodimer can be biased by genetically engineering and expressing Fc fusion molecules that contain a variant ICOSL polypeptide using knob-into-hole methods described below.
  • the variant polypeptides provided herein which are immunomodulatory proteins comprising variants of an Ig domain of the IgSF family (vIgD) can be conjugated with, such as fused directly or indirectly, to, a moiety, such as an effector moiety, such as another protein, directly or indirectly, to form a conjugate (“IgSF conjugate”).
  • a variant ICOSL immunomodulatory protein is provided as a conjugate in which is contained a vIgD of ICOSL linked, directly or indirectly, to a targeting agent or moiety, e.g. to an antibody or other binding molecules that specifically binds to a ligand, e.g.
  • the targeting agent e.g. antibody or other binding molecule
  • TILs tumor infiltrating lymphocytes
  • the attachment can be covalent or non-covalent, e.g., via a biotin-streptavidin non-covalent interaction.
  • the conjugate is a fusion protein of a variant ICOSL polypeptide linked, directly or via a linker, to another protein or polypeptide moiety.
  • the fusion protein is an ICOSL-Fc variant fusion, in which any two or more of the foregoing variant polypeptides can be attached to an Fc.
  • the IgSF conjugate such as fusion protein, comprises the ECD of a wildtype (full length or truncated) or a variant ICOSL polypeptide.
  • the IgSF conjugate, such as fusion protein comprises an IgV domain or an IgC (e.g., IgC2) domain or domains, or a specific binding fragment of the IgV domain or a specific binding fragment of the IgC (e.g., IgC2) domain or domains.
  • the IgSF conjugate, such as fusion protein comprises an IgV domain of ICOSL as set forth in SEQ ID NOs: 196 or 545.
  • the moiety can be a targeting moiety, a small molecule drug (non-polypeptide drug of less than 500 daltons molar mass), a toxin, a cytostatic agent, a cytotoxic agent, an immunosuppressive agent, a radioactive agent suitable for diagnostic purposes, a radioactive metal ion for therapeutic purposes, a prodrug-activating enzyme, an agent that increases biological half-life, or a diagnostic or detectable agent.
  • the effector moiety is a therapeutic agent, such as a cancer therapeutic agent, which is either cytotoxic, cytostatic or otherwise provides some therapeutic benefit.
  • the effector moiety is a targeting moiety or agent, such as an agent that targets a cell surface antigen, e.g., an antigen on the surface of a tumor cell.
  • the effector moiety is a label, which can generate a detectable signal, either directly or indirectly.
  • the effector moiety is a toxin.
  • the effector moiety is a protein, peptide, nucleic acid, small molecule or nanoparticle.
  • 1, 2, 3, 4, 5 or more effector moieties which can be the same or different, are conjugated, linked or fused to the variant polypeptide or protein to form an IgSF conjugate.
  • effector moieties can be attached to the variant polypeptide or immunomodulatory protein using various molecular biological or chemical conjugation and linkage methods known in the art and described below.
  • linkers such as peptide linkers, cleavable linkers, non-cleavable linkers or linkers that aid in the conjugation reaction, can be used to link or conjugate the effector moieties to the variant polypeptide or immunomodulatory protein.
  • the IgSF conjugate comprises the following components: (protein or polypeptide), (L) q and (effector moiety) m , wherein the protein or polypeptide is any of the described variant polypeptides or immunomodulatory proteins capable of binding one or more cognate counter structure ligands as described; L is a linker for linking the protein or polypeptide to the moiety; m is at least 1; q is 0 or more; and the resulting IgSF conjugate binds to the one or more counter structure ligands.
  • m is 1 to 4 and q is 0 to 8.
  • the linker is a peptide.
  • the effector moiety is a protein or polypeptide.
  • an IgSF conjugate comprising a variant polypeptide or immunomodulatory protein provided herein conjugated with a targeting agent that binds to a cell surface molecule, for example, for targeted delivery of the variant polypeptide or immunomodulatory protein to a specific cell.
  • the targeting agent is a molecule(s) that has the ability to localize and bind to a molecule present on a normal cell/tissue and/or tumor cell/tumor in a subject.
  • IgSF conjugates comprising a targeting agent can bind to a ligand (directly or indirectly), which is present on a cell, such as a tumor cell.
  • the targeting agents of the invention contemplated for use include antibodies, polypeptides, peptides, aptamers, other ligands, or any combination thereof, that can bind a component of a target cell or molecule.
  • the targeting agent binds a tumor cell(s) or can bind in the vicinity of a tumor cell(s) (e.g., tumor vasculature or tumor microenvironment) following administration to the subject.
  • the targeting agent may bind to a receptor or ligand on the surface of the cancer cell.
  • a targeting agent is selected which is specific for a noncancerous cells or tissue.
  • a targeting agent can be specific for a molecule present normally on a particular cell or tissue.
  • the same molecule can be present on normal and cancer cells.
  • Various cellular components and molecules are known.
  • an IgSF conjugate of the invention can operate by two separate mechanisms (targeting cancer and non-cancer cells).
  • an IgSF conjugate of the invention comprises a targeting agent which can bind/target a cellular component, such as a tumor antigen, a bacterial antigen, a viral antigen, a mycoplasm antigen, a fungal antigen, a prion antigen, an antigen from a parasite.
  • a cellular component, antigen or molecule can each be used to mean, a desired target for a targeting agent.
  • a targeting agent is specific for or binds to a component, which includes but is not limited to, epidermal growth factor receptor (EGFR, ErbB-1, HER1), ErbB-2 (HER2/neu), ErbB-3/HER3, ErbB-4/HER4, EGFR ligand family; insulin-like growth factor receptor (IGFR) family, IGF-binding proteins (IGFBPs), IGFR ligand family; platelet derived growth factor receptor (PDGFR) family, PDGFR ligand family; fibroblast growth factor receptor (FGFR) family, FGFR ligand family, vascular endothelial growth factor receptor (VEGFR) family, VEGF family; HGF receptor family; TRK receptor family; ephrin (EPH) receptor family; AXL receptor family; leukocyte tyrosine kinase (LTK) receptor family; TIE receptor family, angiopoietin 1,2; receptor tyrosine kinase-
  • TGF- ⁇ tumor glycosylation antigen
  • LHR LHR
  • DDR discoidin domain receptor
  • KLG KLG receptor family
  • RYK receptor MuSK receptor family
  • TGF- ⁇ tumor glycosylation antigen
  • TGF- ⁇ tumor glycosylation antigen
  • Cytokine receptors Class I (hematopoietin family) and Class II (interferon/IL-10 family) receptors, tumor necrosis factor (TNF) receptor superfamily (TNFRSF), death receptor family
  • CT cancer-testis
  • CT cancer-testis
  • EDA extradomain A
  • GPNMB low density lipid receptor/GDP-L fucose: 13-D-galactose 2- ⁇ -L-fucosyltransferase (LDLR/FUT) fusion protein, HLA-A2.
  • HLA-A*201-R170I HLA-AI 1 heat shock protein 70-2 mutated
  • HLA-AI heat shock protein 70-2 mutated
  • K1AA0205 MART2, melanoma ubiquitous mutated 1, 2, 3 (MUM-I, 2, 3), prostatic acid phosphatase (PAP), neo-PAP, Myosin class I, NFYC, OGT, OS-9, pml-RAR ⁇ fusion protein, PRDX5, PTPRK, K-ras (KRAS2), N-ras (NRAS), HRAS, RBAF600, SIRT2, SNRPD1, SYT-SSX1 or -SSX2 fusion protein, Triosephosphate Isomerase, BAGE, BAGK-1, BAGE-2,3,4,5, GAGE-1,2,3,4,5,6,7,8, GnT-V (aberrant N
  • a targeting agent is specific for or binds to a component, which includes, but is not limited to, HER1/EGFR, HER2/ERBB2, CD20, CD25 (IL-2R ⁇ receptor), CD33, CD52, CD133, CD206, CEA, CEACAM1, CEACAM3, CEACAM5, CEACAM6, cancer antigen 125 (CA125), alpha-fetoprotein (AFP), Lewis Y, TAG72, Caprin-1, mesothelin, PDGF receptor (PDGFR; such as PDGF-R ⁇ ), PD-1, PD-L1, CTLA-4, IL-2 receptor, vascular endothelial growth factor (VEGF), CD30, EpCAM, EphA2, Glypican-3, gpA33, mucins, CAIX, PSMA, folate-binding protein, gangliosides (such as GD2, GD3, GM1 and GM2), VEGF receptor (VEGFR), VEGFR2, VEGF-A
  • an IgSF conjugate through its targeting agent, will bind a cellular component of a tumor cell, tumor vasculature or tumor microenvironment, thereby promoting killing of targeted cells via modulation of the immune response, (e.g., by activation of co-stimulatory molecules or inhibition of negative regulatory molecules of immune cell activation), inhibition of survival signals (e.g., growth factor or cytokine or hormone receptor antagonists), activation of death signals, and/or immune-mediated cytotoxicity, such as through antibody dependent cellular cytotoxicity.
  • modulation of the immune response e.g., by activation of co-stimulatory molecules or inhibition of negative regulatory molecules of immune cell activation
  • survival signals e.g., growth factor or cytokine or hormone receptor antagonists
  • activation of death signals e.g., through antibody dependent cellular cytotoxicity.
  • Such IgSF conjugates can function through several mechanisms to prevent, reduce or eliminate tumor cells, such as to facilitate delivery of conjugated effector moieties to the tumor target, such as through receptor-mediated endocytosis of the IgSF conjugate; or such conjugates can recruit, bind, and/or activate immune cells (e.g. NK cells, monocytes/macrophages, dendritic cells, T cells, B cells). Moreover, in some instances one or more of the foregoing pathways may operate upon administration of one or more IgSF conjugates of the invention.
  • immune cells e.g. NK cells, monocytes/macrophages, dendritic cells, T cells, B cells.
  • an IgSF conjugate through its targeting agent, will be localized to, such as bind to, a cellular component of a tumor cell, tumor vasculature or tumor microenvironment, thereby modulating cells of the immune response in the vicinity of the tumor.
  • the targeting agent facilitates delivery of the conjugated IgSF (e.g. vIgD) to the tumor target, such as to interact with its cognate binding partner to alter signaling of immune cells (e.g. NK cells, monocytes/macrophages, dendritic cells, T cells, B cells) bearing the cognate binding partner.
  • localized delivery agonizes or stimulates the costimulatory receptor.
  • the targeting agent is an immunoglobulin.
  • immunoglobulin includes natural or artificial mono- or polyvalent antibodies including, but not limited to, polyclonal, monoclonal, multispecific, human, humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab′) fragments, fragments produced by a Fab expression library, single chain Fv (scFv); anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies of the invention), and epitope-binding fragments of any of the above.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, e.g., molecules that contain an antigen binding site that immunospecifically binds an antigen.
  • the immunoglobulin molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA, and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2) or subclass of immunoglobulin molecule.
  • an IgSF conjugate through its antibody targeting moiety, will bind a cellular component of a tumor cell, tumor vasculature or tumor microenvironment, thereby promoting apoptosis of targeted cells via modulation of the immune response, (e.g., by activation of co-stimulatory molecules or inhibition of negative regulatory molecules of immune cell activation), inhibition of survival signals (e.g., growth factor or cytokine or hormone receptor antagonists), activation of death signals, and/or immune-mediated cytotoxicity, such as through antibody dependent cellular cytotoxicity.
  • modulation of the immune response e.g., by activation of co-stimulatory molecules or inhibition of negative regulatory molecules of immune cell activation
  • survival signals e.g., growth factor or cytokine or hormone receptor antagonists
  • activation of death signals e.g., through antibody dependent cellular cytotoxicity.
  • Such IgSF conjugates can function through several mechanisms to prevent, reduce or eliminate tumor cells, such as to facilitate delivery of conjugated effector moieties to the tumor target, such as through receptor-mediated endocytosis of the IgSF conjugate; or such conjugates can recruit, bind, and/or activate immune cells (e.g. NK cells, monocytes/macrophages, dendritic cells, T cells, B cells).
  • immune cells e.g. NK cells, monocytes/macrophages, dendritic cells, T cells, B cells.
  • an IgSF conjugate through its antibody targeting moiety, will bind a cellular component of a tumor cell, tumor vasculature or tumor microenvironment, thereby modulating the immune response (e.g., by activation of co-stimulatory molecules or inhibition of negative regulatory molecules of immune cell activation).
  • such conjugates can recognize, bind, and/or modulate (e.g. inhibit or activate) immune cells (e.g. NK cells, monocytes/macrophages, dendritic cells, T cells, B cells).
  • Antibody targeting moieties of the invention include antibody fragments that include, but are not limited to, Fab, Fab′ and F(ab′)2, Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and fragments comprising either a VL or VH domain.
  • Antigen-binding antibody fragments, including single-chain antibodies may comprise the variable region(s) alone or in combination with the entirety or a portion of the following: hinge region, CH1, CH2, and CH3 domains. Also included in the invention are antigen-binding fragments also comprising any combination of variable region(s) with a hinge region, CH1, CH2, and CH3 domains.
  • the antibody targeting moieties of the invention may be from any animal origin including birds and mammals.
  • the antibody targeting moieties are human, murine (e.g., mouse and rat), donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken. Further, such antibodies may be humanized versions of animal antibodies.
  • the antibody targeting moieties of the invention may be monospecific, bispecific, trispecific, or of greater multispecificity.
  • an antibody/targeting moiety recruits, binds, and/or activates immune cells (e.g. NK cells, monocytes/macrophages, dendritic cells) via interactions between Fc (in antibodies) and Fc receptors (on immune cells) and via the conjugated variant polypeptides or immunomodulatory proteins provided herein.
  • an antibody/targeting moiety recognizes or binds a tumor agent via and localizes to the tumor cell the conjugated variant polypeptides or immunomodulatory proteins provided herein to facilitate modulation of immune cells in the vicinity of the tumor.
  • antibodies which can be incorporated into IgSF conjugates include but are not limited to antibodies such as Cetuximab (IMC-C225; Erbitux®), Trastuzumab (Herceptin®), Rituximab (Rituxan®; MabThera®), Bevacizumab (Avastin®), Alemtuzumab (Campath®; Campath-1H®; Mabcampath®), Pertuzumab (Perjeta®), Panitumumab (ABX-EGF; Vectibix®), Ranibizumab (Lucentis®), Ibritumomab, Ibritumomab tiuxetan, (Zevalin®), Tositumomab, Iodine I131 Tositumomab (BEXXAR®), Catumaxomab (Removab®), Dinutuximab (UnituxinTM), Gemtuzumab, Gemtuzumab,
  • the antibody or antigen-binding fragment of the provided conjugates is cetuximab, panitumumab, zalutumumab, nimotuzumab, trastuzumab, Ado-trastuzumab emtansine, Tositumomab (Bexxar®), Rituximab (Rituxan, Mabthera), Ibritumomab tiuxetan (Zevalin), Daclizumab (Zenapax), Gemtuzumab (Mylotarg), Alemtuzumab, CEA-scan Fab fragment, OC125 monoclonal antibody, ab75705, B72.3, Bevacizumab (Avastin®), Afatinib, Axitinib, Bosutinib, Cabozantinib, Cerit
  • PD-L1 antibodies or antigen binding fragments thereof can be incorporated into the IgSF conjugates.
  • PD-L1 antibodies which can be incorporated into IgSF conjugates include but are not limited to antibodies such as BMS-936559, 12A4, LY3300054, Atezolizumab (Tecentriq®), Avelumab (Bavencio®), Durvalumab (Imfinzi®). See, e.g., WO2007/005874, WO2017/034916, WO2010/077634, WO2013/079174, WO2011/066389, these references are incorporated by reference in their entirety.
  • the vIgD is linked, directly or indirectly, to the N- or C-terminus of the light and/or heavy chain of an anti-PD-L1 antibody.
  • the anti-PD-L antibody is BMS-936559, LY3300054, atezolizumab, avelumab or durvalumab.
  • Exemplary light chain and heavy chain of an anti-PD-L1 antibody atezolizumab are set forth in SEQ ID NO: 866 and 867, respectively.
  • Exemplary IgSF conjugates that include the anti-PD-L1 antibody Atezolizumab is set forth in SEQ ID NOs: 868-895.
  • the antibody targeting moiety is a full length antibody, or antigen-binding fragment thereof, containing an Fc domain.
  • the variant polypeptide or immunomodulatory protein is conjugated to the Fc portion of the antibody targeting moiety, such as by conjugation to the N-terminus of the Fc portion of the antibody.
  • the vIgD is linked, directly or indirectly, to the N- or C-terminus of the light and/or heavy chain of the antibody.
  • linkage can be via a peptide linker, such as any described above.
  • the linker can further include amino acids introduced by cloning and/or from a restriction site.
  • the linker may include additional amino acids on either end introduced by a restriction site.
  • the linker can include additional amino acids such as SA (in one-letter amino acid code) as introduced by use of the restriction site AFEI.
  • SA in one-letter amino acid code
  • FIG. 10A-10C depict exemplary configurations.
  • the antibody conjugate can be produced by co-expression of the heavy and light chain of the antibody in a cell.
  • the targeting agent is an aptamer molecule.
  • the aptamer is comprised of nucleic acids that function as a targeting agent.
  • an IgSF conjugate of the invention comprises an aptamer that is specific for a molecule on a tumor cell, tumor vasculature, and/or a tumor microenvironment.
  • the aptamer itself can comprise a biologically active sequence, in addition to the targeting module (sequence), wherein the biologically active sequence can induce an immune response to the target cell.
  • such an aptamer molecule is a dual use agent.
  • an IgSF conjugate of the invention comprises conjugation of an aptamer to an antibody, wherein the aptamer and the antibody are specific for binding to separate molecules on a tumor cell, tumor vasculature, tumor microenvironment, and/or immune cells.
  • aptamer includes DNA, RNA or peptides that are selected based on specific binding properties to a particular molecule.
  • an aptamer(s) can be selected for binding a particular gene or gene product in a tumor cell, tumor vasculature, tumor microenvironment, and/or an immune cell, as disclosed herein, where selection is made by methods known in the art and familiar to one of skill in the art.
  • the targeting agent is a peptide.
  • the variant polypeptides or immunomodulatory proteins provided herein can be conjugated to a peptide which can bind with a component of a cancer or tumor cells. Therefore, such IgSF conjugates of the invention comprise peptide targeting agents which binds to a cellular component of a tumor cell, tumor vasculature, and/or a component of a tumor microenvironment.
  • targeting agent peptides can be an antagonist or agonist of an integrin. Integrins, which comprise an alpha and a beta subunit, include numerous types well known to a skilled artisan.
  • the targeting agent is Vv ⁇ 3.
  • Integrin Vv ⁇ 3 is expressed on a variety of cells and has been shown to mediate several biologically relevant processes, including adhesion of osteoclasts to bone matrix, migration of vascular smooth muscle cells, and angiogenesis.
  • Suitable targeting molecules for integrins include RGD peptides or peptidomimetics as well as non-RGD peptides or peptidomimetics (see, e.g., U.S. Pat. Nos. 5,767,071 and 5,780,426) for other integrins such as V4. ⁇ i (VLA-4), V4-P7 (see, e.g., U.S. Pat. No.
  • an IgSF conjugate comprising a variant polypeptide or immunomodulatory protein provided herein conjugated with a therapeutic agent.
  • the therapeutic agent includes, for example, daunomycin, doxorubicin, methotrexate, and vindesine (Rowland et al., Cancer Immunol. Immunother. 21:183-187, 1986).
  • the therapeutic agent has an intracellular activity.
  • the IgSF conjugate is internalized and the therapeutic agent is a cytotoxin that blocks the protein synthesis of the cell, therein leading to cell death.
  • the therapeutic agent is a cytotoxin comprising a polypeptide having ribosome-inactivating activity including, for example, gelonin, bouganin, saporin, ricin, ricin A chain, bryodin, diphtheria toxin, restrictocin, Pseudomonas exotoxin A and variants thereof.
  • the therapeutic agent is a cytotoxin comprising a polypeptide having a ribosome-inactivating activity
  • the IgSF conjugate must be internalized upon binding to the target cell in order for the protein to be cytotoxic to the cells.
  • an IgSF conjugate comprising a variant polypeptide or immunomodulatory protein provided herein conjugated with a toxin.
  • the toxin includes, for example, bacterial toxins such as diphtheria toxin, plant toxins such as ricin, small molecule toxins such as geldanamycin (Mandler et al., J. Nat. Cancer Inst. 92(19):1573-1581 (2000); Mandler et al., Bioorganic & Med. Chem. Letters 10: 1025-1028 (2000); Mandler et al., Bioconjugate Chem.
  • toxins may exert their cytotoxic and cytostatic effects by mechanisms including tubulin binding, DNA binding, or topoisomerase inhibition.
  • an IgSF conjugate comprising a variant polypeptide or immunomodulatory protein provided herein conjugated with a label, which can generate a detectable signal, indirectly or directly.
  • IgSF conjugates can be used for research or diagnostic applications, such as for the in vivo detection of cancer.
  • the label is preferably capable of producing, either directly or indirectly, a detectable signal.
  • the label may be radio-opaque or a radioisotope, such as 3H, 14C, 32P, 35S, 1231, 1251, 131I; a fluorescent (fluorophore) or chemiluminescent (chromophore) compound, such as fluorescein isothiocyanate, rhodamine or luciferin; an enzyme, such as alkaline phosphatase, ⁇ -galactosidase or horseradish peroxidase; an imaging agent; or a metal ion.
  • a radioisotope such as 3H, 14C, 32P, 35S, 1231, 1251, 131I
  • a fluorescent (fluorophore) or chemiluminescent (chromophore) compound such as fluorescein isothiocyanate, rhodamine or luciferin
  • an enzyme such as alkaline phosphatase, ⁇ -galactosidase or horseradish peroxidas
  • the label is a radioactive atom for scintigraphic studies, for example 99Tc or 1231, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, MRI), such as zirconium-89, iodine-123, iodine-131, indium-111, fluorine-19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron.
  • NMR nuclear magnetic resonance
  • Zirconium-89 may be complexed to various metal chelating agents and conjugated to antibodies, e.g., for PET imaging (WO 2011/056983).
  • the IgSF conjugate is detectable indirectly.
  • a secondary antibody that is specific for the IgSF conjugate and contains a detectable label can be used to detect the IgSF conjugate.
  • the IgSF conjugates may be prepared using any methods known in the art. See, e.g., WO 2009/067800, WO 2011/133886, and U.S. Patent Application Publication No. 2014322129, incorporated by reference herein in their entirety.
  • variant polypeptides or immunomodulatory proteins of an IgSF conjugate may be “attached to” the effector moiety by any means by which the variant polypeptides or immunomodulatory proteins can be associated with, or linked to, the effector moiety.
  • the variant polypeptides or immunomodulatory proteins of an IgSF conjugate may be attached to the effector moiety by chemical or recombinant means. Chemical means for preparing fusions or conjugates are known in the art and can be used to prepare the IgSF conjugate.
  • the method used to conjugate the variant polypeptides or immunomodulatory proteins and effector moiety must be capable of joining the variant polypeptides or immunomodulatory proteins with the effector moiety without interfering with the ability of the variant polypeptides or immunomodulatory proteins to bind to their one or more counter structure ligands.
  • the variant polypeptides or immunomodulatory proteins of an IgSF conjugate may be linked indirectly to the effector moiety.
  • the variant polypeptides or immunomodulatory proteins of an IgSF conjugate may be directly linked to a liposome containing the effector moiety of one of several types.
  • the effector moiety(s) and/or the variant polypeptides or immunomodulatory proteins may also be bound to a solid surface.
  • the variant polypeptides or immunomodulatory proteins of an IgSF conjugate and the effector moiety are both proteins and can be conjugated using techniques well known in the art.
  • There are several hundred crosslinkers available that can conjugate two proteins. See for example “Chemistry of Protein Conjugation and Crosslinking,” 1991, Shans Wong, CRC Press, Ann Arbor).
  • the crosslinker is generally chosen based on the reactive functional groups available or inserted on the variant polypeptides or immunomodulatory proteins and/or effector moiety.
  • a photoactivatible crosslinker can be used.
  • Crosslinking agents known to the art include the homobifunctional agents: glutaraldehyde, dimethyladipimidate and Bis(diazobenzidine) and the heterobifunctional agents: m Maleimidobenzoyl-N-Hydroxysuccinimide and Sulfo-m Maleimidobenzoyl-N-Hydroxysuccinimide.
  • the variant polypeptides or immunomodulatory proteins of an IgSF conjugate may be engineered with specific residues for chemical attachment of the effector moiety.
  • Specific residues used for chemical attachment of molecule known to the art include lysine and cysteine.
  • the crosslinker is chosen based on the reactive functional groups inserted on the variant polypeptides or immunomodulatory proteins, and available on the effector moiety.
  • An IgSF conjugate may also be prepared using recombinant DNA techniques.
  • a DNA sequence encoding the variant polypeptides or immunomodulatory proteins is fused to a DNA sequence encoding the effector moiety, resulting in a chimeric DNA molecule.
  • the chimeric DNA sequence is transfected into a host cell that expresses the fusion protein.
  • the fusion protein can be recovered from the cell culture and purified using techniques known in the art.
  • the radio- or other labels may be incorporated in the conjugate in known ways.
  • the peptide may be biosynthesized or may be synthesized by chemical amino acid synthesis using suitable amino acid precursors involving, for example, fluorine-19 in place of hydrogen.
  • Labels such as 99Tc or 1231, 186Re, 188Re and 111In can be attached via a cysteine residue in the peptide.
  • Yttrium-90 can be attached via a lysine residue.
  • the IODOGEN method (Fraker et al., Biochem. Biophys. Res. Commun. 80:49-57 (1978)) can be used to incorporate iodine-123. “Monoclonal Antibodies in Immunoscintigraphy” (Chatal, CRC Press 1989) describes other methods in detail.
  • Conjugates of the variant polypeptides or immunomodulatory proteins and a cytotoxic agent may be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCl), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate),
  • a ricin immunotoxin can be prepared as described in Vitetta et al., Science 238:1098 (1987).
  • Carbon-14-labeled 1-p-isothiocyanatobenzyl-3-methyldiethylenetriaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See, e.g., WO94/11026.
  • the linker may be a “cleavable linker” facilitating release of the cytotoxic drug in the cell.
  • an acid-labile linker for example, an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari et al., Cancer Research 52:127-131 (1992); U.S. Pat. No. 5,208,020) may be used.
  • the IgSF conjugates of the invention expressly contemplate, but are not limited to, drug conjugates prepared with cross-linker reagents: BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl-(4-vinylsulfone)benzoate) which are commercially available (e.g., from Pierce Biotechnology, Inc., Rockford, Ill., U.S.A). See pages 467-498, 2003-2004 Applications Handbook and Catalog.
  • engineered cells which express the immunomodulatory variant ICOSL polypeptides (alternatively, “engineered cells”).
  • the variant ICOSL polypeptide is expressed on a cell, such as an immune cell (e.g. T cell or antigen presenting cell), in membrane-bound form, thereby providing a transmembrane immunomodulatory protein (hereinafter also called a “TIP”).
  • TIP transmembrane immunomodulatory protein
  • the variant ICOSL polypeptide is expressed in a cell, such as an immune cell (e.g.
  • a SIP can antagonize a cognate binding partner in the environment (e.g. tumor microenvironment) in which it is secreted.
  • an immunomodulatory polypeptide comprising a variant ICOSL can be a membrane bound protein.
  • the immunomodulatory polypeptide can be a transmembrane immunomodulatory polypeptide comprising a variant ICOSL in which is contained: an ectodomain containing at least one affinity modified IgSF domain (IgV or IgC), a transmembrane domain and, optionally, a cytoplasmic domain.
  • the transmembrane immunomodulatory protein can be expressed on the surface of an immune cell, such as a mammalian cell, including on the surface of a lymphocyte (e.g. T cell or NK cell) or antigen presenting cell.
  • the transmembrane immunomodulatory protein is expressed on the surface of a mammalian T-cell, including such T-cells as: a T helper cell, a cytotoxic T-cell (alternatively, cytotoxic T lymphocyte or CTL), a natural killer T-cell, a regulatory T-cell, a memory T-cell, or a gamma delta T-cell.
  • the mammalian cell is an antigen presenting cell (APC).
  • APC antigen presenting cell
  • the ectodomain (alternatively, “extracellular domain”) of comprises the one or more amino acid variations (e.g. amino acid substitutions) of the variant ICOSL of the invention.
  • a transmembrane protein will comprise an ectodomain that comprises one or more amino acid substitutions of a variant ICOSL of the invention.
  • the engineered cells express variant ICOSL polypeptides that are transmembrane immunomodulatory polypeptides (TIPs) that can be a membrane protein such as a transmembrane protein.
  • TIPs transmembrane immunomodulatory polypeptides
  • the ectodomain of a membrane protein comprises an extracellular domain or IgSF domain thereof of a variant ICOSL provided herein in which is contained one or more amino acid substitutions in at least one IgSF domain as described.
  • the transmembrane immunomodulatory proteins provided herein further contain a transmembrane domain linked to the ectodomain.
  • the transmembrane domain results in an encoded protein for cell surface expression on a cell.
  • the transmembrane domain is linked directly to the ectodomain. In some embodiments, the transmembrane domain is linked indirectly to the ectodomain via one or more linkers or spacers. In some embodiments, the transmembrane domain contains predominantly hydrophobic amino acid residues, such as leucine and valine.
  • a full length transmembrane anchor domain can be used to ensure that the TIPs will be expressed on the surface of the engineered cell, such as engineered T cell.
  • this could be from a particular native protein that is being affinity modified (e.g. ICOSL or other native IgSF protein), and simply fused to the sequence of the first membrane proximal domain in a similar fashion as the native IgSF protein (e.g. ICOSL).
  • the transmembrane immunomodulatory protein comprises a transmembrane domain of the corresponding reference (e.g., unmodified) or wild-type IgSF member, such as a transmembrane domain contained in the sequence of amino acids set forth in SEQ ID NO:5 (Table 2).
  • the membrane bound form comprises a transmembrane domain of the corresponding reference (e.g., unmodified) or wild-type polypeptide, such as corresponding to residues 257-277 of SEQ ID NO:5.
  • the transmembrane domain is a non-native transmembrane domain that is not the transmembrane domain of native ICOSL.
  • the transmembrane domain is derived from a transmembrane domain from another non-ICOSL family member polypeptide that is a membrane-bound or is a transmembrane protein.
  • a transmembrane anchor domain from another protein on T cells can be used.
  • the transmembrane domain is derived from CD8.
  • the transmembrane domain can further contain an extracellular portion of CD8 that serves as a spacer domain.
  • An exemplary CD8 derived transmembrane domain is set forth in SEQ ID NO: 246 or 483 or a portion thereof containing the CD8 transmembrane domain.
  • the transmembrane domain is a synthetic transmembrane domain.
  • the transmembrane immunomodulatory protein further contains an endodomain, such as a cytoplasmic signaling domain, linked to the transmembrane domain.
  • the cytoplasmic signaling domain induces cell signaling.
  • the endodomain of the transmembrane immunomodulatory protein comprises the cytoplasmic domain of the corresponding reference (e.g., unmodified) or wild-type polypeptide, such as a cytoplasmic domain contained in the sequence of amino acids set forth in SEQ ID NO:5 (see Table 2).
  • a provided transmembrane immunomodulatory protein that is or comprises a variant ICOSL comprises a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 257 and contains an ectodomain comprising at least one affinity-modified ICOSL IgSF domain as described and a transmembrane domain.
  • the transmembrane immunomodulatory protein contains any one or more amino acid substitutions in an IgSF domain (e.g. IgV domain) as described, including any set forth in Table 1.
  • the transmembrane immunomodulatory protein can further comprise a cytoplasmic domain as described. In some embodiments, the transmembrane immunomodulatory protein can further contain a signal peptide. In some embodiments, the signal peptide is the native signal peptide of wild-type IgSF member, such as contained in the sequence of amino acids set forth in SEQ ID NO: 5 (see e.g. Table 2).
  • transmembrane immunomodulatory proteins comprising the amino acid substitutions E16V/N52H/N57Y/Q100R/V110D/H115R/Y152C/K156M/C198R, N52H/N57Y/Q100R, or N52H/N57Y/Q100P.
  • the provided transmembrane immunomodulatory protein is or comprises a variant ICOSL comprising the sequence of amino acids set forth in SEQ ID NO:257, but in which is contained amino substitutions E16V/N52H/N57Y/Q100R/V110D/H115R/Y152C/K156M/C198R, N52H/N57Y/Q100R, or N52H/N57Y/Q100P at corresponding positions in SEQ ID NO:257, or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 257 and contains the amino acid substitutions E16V/N52H/N57Y/Q100R/V110D/H115R/Y152C/K156M/C198R, N52H/N57Y/Q100R, or N52H/N57Y/Q100
  • transmembrane immunomodulatory proteins comprising the sequence of amino acids set forth in SEQ ID NOS: 496 or 497 (each containing the amino acid substitution N52D), SEQ ID NOS: 498 or 499 (each containing the amino acid substitutions N52H/N57Y/Q100P), SEQ ID NOS: 500 or 501 (each containing the amino acid substitutions E16V/N52H/N57Y/Q100R/V110D/H115R/Y152C/K156M/C198R) or SEQ ID NOS: 502 or 503 (each containing the amino acid substitutions N52H/N57Y/Q100R), or a sequence of amino acids that comprises at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to any of SEQ ID NOS: 495-503 and that contains the indicated amino acid substitutions.
  • a nucleic acid molecule encoding such transmembrane immunomodulatory proteins comprises a nucleotide sequence that encodes a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 257 and contains an ectodomain comprising at least one affinity-modified IgSF domain as described, a transmembrane domain and, optionally, a cytoplasmic domain.
  • the nucleic acid molecule can further comprise a sequence of nucleotides encoding a signal peptide.
  • the signal peptide is the native signal peptide of the corresponding wild-type IgSF member (see e.g. Table 2).
  • a transmembrane immunomodulatory protein is a ICOSL TIP comprising i) the sequence of amino acids set forth in SEQ ID NO:383 or ii) a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 243 and that comprises the affinity-modified domain contained in SEQ ID NO: 243 or the amino acid substitutions therein.
  • a sequence of nucleotides set forth in SEQ ID NO: 244 i) a sequence that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 244 and that encodes a TIP that comprises the affinity-modified domain of SEQ ID NO: 243, or iii) a sequence of i) or ii) having degenerate codons.
  • CAR-related transmembrane immunomodulatory proteins in which the endodomain of a transmembrane immunomodulatory protein comprises a cytoplasmic signaling domain that comprises at least one ITAM (immunoreceptor tyrosine-based activation motif)-containing signaling domain.
  • ITAM is a conserved motif found in a number of protein signaling domains involved in signal transduction of immune cells, including in the CD3-zeta chain (“CD3-z”) involved in T-cell receptor signal transduction.
  • the endodomain comprises at CD3-zeta signaling domain.
  • the CD3-zeta signaling domain comprises the sequence of amino acids set forth in SEQ ID NO: 243 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% to SEQ ID NO:247 and retains the activity of T cell signaling.
  • the endodomain of a CAR-related transmembrane immunomodulatory protein can further comprise a costimulatory signaling domain to further modulate immunomodulatory responses of the T-cell.
  • the costimulatory signaling domain is CD28, ICOS, 41BB or OX40.
  • the costimulatory signaling domain is a derived from CD28 or 4-1BB and comprises the sequence of amino acids set forth in any of SEQ ID NOS: 484-487 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% to SEQ ID NO: 484-487 and retains the activity of T cell costimulatory signaling.
  • the provided CAR-related transmembrane immunomodulatory proteins have features of CARs to stimulate T cell signaling upon binding of an affinity modified IgSF domain to a cognate binding partner or counter structure.
  • upon specific binding by the affinity-modified IgSF domain to its counter structure can lead to changes in the immunological activity of the T-cell activity as reflected by changes in cytotoxicity, proliferation or cytokine production.
  • the transmembrane immunomodulatory protein does not contain an endodomain capable of mediating cytoplasmic signaling. In some embodiments, the transmembrane immunomodulatory protein lacks the signal transduction mechanism of the wild-type or unmodified polypeptide and therefore does not itself induce cell signaling. In some embodiments, the transmembrane immunomodulatory protein lacks an intracellular (cytoplasmic) domain or a portion of the intracellular domain of the corresponding reference (e.g., unmodified) or wild-type polypeptide, such as a cytoplasmic signaling domain contained in the sequence of amino acids set forth in SEQ ID NO:5 (see Table 2).
  • the transmembrane immunomodulatory protein does not contain an ITIM (immunoreceptor tyrosine-based inhibition motif), such as contained in certain inhibitory receptors, including inhibitory receptors of the IgSF family (e.g. PD-1 or TIGIT).
  • ITIM immunoglobulin-based inhibition motif
  • the transmembrane immunomodulatory protein only contains the ectodomain and the transmembrane domain, such as any as described.
  • the ICOSL variant immunomodulatory polypeptide containing any one or more of the amino acid mutations as described herein, is secretable, such as when expressed from a cell.
  • a variant ICOSL immunomodulatory protein does not comprise a transmembrane domain.
  • the variant ICOSL immunomodulatory protein is not conjugated to a half-life extending moiety (such as an Fc domain or a multimerization domain).
  • the variant ICOSL immunomodulatory protein comprises a signal peptide, e.g. an antibody signal peptide or other efficient signal sequence to get domains outside of cell.
  • the immunomodulatory protein comprises a signal peptide and is expressed by an engineered cell
  • the signal peptide causes the immunomodulatory protein to be secreted by the engineered cell.
  • the signal peptide, or a portion of the signal peptide is cleaved from the immunomodulatory protein with secretion.
  • the immunomodulatory protein can be encoded by a nucleic acid (which can be part of an expression vector).
  • the immunomodulatory protein is expressed and secreted by a cell (such as an immune cell, for example a primary immune cell).
  • variant ICOSL immunomodulatory proteins that further comprise a signal peptide.
  • a nucleic acid molecule encoding the variant ICOSL immunomodulatory protein operably connected to a secretion sequence encoding the signal peptide.
  • a signal peptide is a sequence on the N-terminus of an immunomodulatory protein that signals secretion of the immunomodulatory protein from a cell.
  • the signal peptide is about 5 to about 40 amino acids in length (such as about 5 to about 7, about 7 to about 10, about 10 to about 15, about 15 to about 20, about 20 to about 25, or about 25 to about 30, about 30 to about 35, or about 35 to about 40 amino acids in length).
  • the signal peptide is a native signal peptide from the corresponding wild-type ICOSL (see Table 6).
  • the signal peptide is a non-native signal peptide.
  • the non-native signal peptide is a mutant native signal peptide from the corresponding wild-type ICOSL, and can include one or more (such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more) substitutions insertions or deletions.
  • the non-native signal peptide is a signal peptide or mutant thereof of a family member from the same IgSF family as the wild-type IgSF family member.
  • the non-native signal peptide is a signal peptide or mutant thereof from an IgSF family member from a different IgSF family than the wild-type IgSF family member.
  • the signal peptide is a signal peptide or mutant thereof from a non-IgSF protein family, such as a signal peptide from an immunoglobulin (such as IgG heavy chain or IgG-kappa light chain), a cytokine (such as interleukin-2 (IL-2), or CD33), a serum albumin protein (e.g. HSA or albumin), a human azurocidin preprotein signal sequence, a luciferase, a trypsinogen (e.g. chymotrypsinogen or trypsinogen) or other signal peptide able to efficiently secrete a protein from a cell.
  • Exemplary signal peptides include any described in the Table 6.
  • the immunomodulatory protein comprises a signal peptide when expressed, and the signal peptide (or a portion thereof) is cleaved from the immunomodulatory protein upon secretion.
  • the engineered cells express a variant ICOSL polypeptide that is secreted from the cell.
  • a variant ICOSL polypeptide is encoded by a nucleic acid molecule encoding an immunomodulatory protein under the operable control of a signal sequence for secretion.
  • the encoded immunomodulatory protein is secreted when expressed from a cell.
  • the immunomodulatory protein encoded by the nucleic acid molecule does not comprise a transmembrane domain.
  • the immunomodulatory protein encoded by the nucleic acid molecule does not comprise a half-life extending moiety (such as an Fc domain or a multimerization domain).
  • the immunomodulatory protein encoded by the nucleic acid molecule comprises a signal peptide.
  • a nucleic acid of the invention further comprises nucleotide sequence that encodes a secretory or signal peptide operably linked to the nucleic acid encoding the immunomodulatory protein, thereby allowing for secretion of the immunomodulatory protein
  • the engineered cells express any of the provided immunomodulatory polypeptides.
  • the engineered cells express on their surface any of the provided transmembrane immunomodulatory polypeptides.
  • the engineered cells express and are capable of or are able to secrete the immunomodulatory protein from the cells under conditions suitable for secretion of the protein.
  • the immunomodulatory protein is expressed on or in a lymphocyte such as a tumor infiltrating lymphocyte (TIL), T-cell or NK cell, or on a myeloid cell.
  • TIL tumor infiltrating lymphocyte
  • the engineered cells are antigen presenting cells (APCs).
  • the engineered cells are engineered mammalian T-cells or engineered mammalian antigen presenting cells (APCs).
  • the engineered T-cells or APCs are human or murine cells.
  • engineered T-cells include, but are not limited to, T helper cell, cytotoxic T-cell (alternatively, cytotoxic T lymphocyte or CTL), natural killer T-cell, regulatory T-cell, memory T-cell, or gamma delta T-cell.
  • the engineered T cells are CD4+ or CD8+.
  • engineered T-cells also require a co-stimulatory signal which in some embodiments is provided by a variant ICOSL transmembrane immunomodulatory polypeptide expressed in membrane bound form as discussed previously.
  • the engineered APCs include, for example, MHC II expressing APCs such as macrophages, B cells, and dendritic cells, as well as artificial APCs (aAPCs) including both cellular and acellular (e.g., biodegradable polymeric microparticles) aAPCs.
  • APCs artificial APCs
  • aAPCs are synthetic versions of APCs that can act in a similar manner to APCs in that they present antigens to T-cells as well as activate them. Antigen presentation is performed by the MHC (Class I or Class II).
  • the antigen that is loaded onto the MHC is, in some embodiments, a tumor specific antigen or a tumor associated antigen.
  • the antigen loaded onto the MHC is recognized by a T-cell receptor (TCR) of a T cell, which, in some cases, can express ICOS, CD28, or other molecule recognized by the variant ICOSL polypeptides provided herein.
  • TCR T-cell receptor
  • Materials which can be used to engineer an aAPC include: poly (glycolic acid), poly(lactic-co-glycolic acid), iron-oxide, liposomes, lipid bilayers, sepharose, and polystyrene.
  • a cellular aAPC can be engineered to contain a TIP and TCR agonist which is used in adoptive cellular therapy.
  • a cellular aAPC can be engineered to contain a TIP and TCR agonist which is used in ex vivo expansion of human T cells, such as prior to administration, e.g., for reintroduction into the patient.
  • the aAPC may include expression of at least one anti-CD3 antibody clone, e.g. such as, for example, OKT3 and/or UCHT1.
  • the aAPCs may be inactivated (e.g. irradiated).
  • the TIP can include any variant IgSF domain that exhibits binding affinity for a cognate binding partner on a T cell.
  • an immunomodulatory protein provided herein such as a transmembrane immunomodulatory protein or a secretable immunomodulatory protein, is co-expressed or engineered into a cell that expresses an antigen-binding receptor, such as a recombinant receptor, such as a chimeric antigen receptor (CAR) or T cell receptor (TCR).
  • an antigen-binding receptor such as a recombinant receptor, such as a chimeric antigen receptor (CAR) or T cell receptor (TCR).
  • the engineered cell such as an engineered T cell, recognizes a desired antigen associated with cancer, inflammatory and autoimmune disorders, or a viral infection.
  • the antigen-binding receptor contains an antigen-binding moiety that specifically binds a tumor specific antigen or a tumor associated antigen.
  • the engineered T-cell is a CAR (chimeric antigen receptor) T-cell that contains an antigen-binding domain (e.g. scFv) that specifically binds to an antigen, such as a tumor specific antigen or tumor associated antigen.
  • an antigen-binding domain e.g. scFv
  • the antigen-binding domain is specific for a particular antigen, e.g., CD19.
  • a CAR is an anti-CD19 CAR, such as a CAR containing an anti-CD19 scFv set forth in SEQ ID NO:482 or SEQ ID NO:245.
  • the TIP protein is expressed in an engineered T-cell receptor cell or an engineered chimeric antigen receptor cell. In such embodiments, the engineered cell co-expresses the TIP and the CAR or TCR.
  • the SIP protein is expressed in an engineered T-cell receptor cell or an engineered chimeric antigen receptor cell. In such embodiments, the engineered cell co-expresses the SIP and the CAR or TCR.
  • Chimeric antigen receptors are recombinant receptors that include an antigen-binding domain (ectodomain), a transmembrane domain and an intracellular signaling region (endodomain) that is capable of inducing or mediating an activation signal to the T cell after the antigen is bound.
  • CAR-expressing cells are engineered to express an extracellular single chain variable fragment (scFv) with specificity for a particular tumor antigen linked to an intracellular signaling part comprising an activating domain and, in some cases, a costimulatory domain.
  • the costimulatory domain can be derived from, e.g., CD28, OX-40, 4-1BB/CD137 or inducible T cell costimulator (ICOS).
  • the activating domain can be derived from, e.g., CD3, such as CD3 zeta, epsilon, delta, gamma, or the like.
  • the CAR is designed to have two, three, four, or more costimulatory domains.
  • the CAR scFv can be designed to target an antigen expressed on a cell associated with a disease or condition, e.g. a tumor antigen, such as, for example, CD19, which is a transmembrane protein expressed by cells in the B cell lineage, including all normal B cells and B cell malignances, including but not limited to NHL, CLL, and non-T cell ALL.
  • a tumor antigen such as, for example, CD19
  • Example CAR+ T cell therapies and constructs are described in U.S. Patent Publication Nos. 2013/0287748, 2014/0227237, 2014/0099309, and 2014/0050708, and these references are incorporated by reference in their entirety.
  • the antigen-binding domain is an antibody or antigen-binding fragment thereof, such as a single chain fragment (scFv).
  • the antigen is expressed on a tumor or cancer cell.
  • exemplary of an antigen is CD19.
  • exemplary of a CAR is an anti-CD19 CAR, such as a CAR containing an anti-CD19 scFv set forth in SEQ ID NO: 245.
  • the CAR further contains a spacer, a transmembrane domain, and an intracellular signaling domain or region comprising an ITAM signaling domain, such as a CD3zeta signaling domain.
  • the CAR further includes a costimulatory signaling domain.
  • the CAR further contains a spacer or hinge, a transmembrane domain, and an intracellular signaling domain (endodomain) comprising an ITAM signaling domain, such as a CD3zeta signaling domain.
  • the CAR further includes a costimulatory signaling domain.
  • the costimulatory domain can be derived from, e.g., CD28, OX-40, 4-1BB/CD137 or inducible T cell costimulator (ICOS).
  • the CAR is designed to have two, three, four, or more costimulatory domains.
  • the CAR scFv can be designed to target an antigen expressed on a cell associated with a disease or condition, e.g.
  • a tumor antigen such as, for example, CD19, which is a transmembrane protein expressed by cells in the B cell lineage, including all normal B cells and B cell malignances, including but not limited to NHL, CLL, and non-T cell ALL.
  • Example CAR+ T cell therapies and constructs are described in U.S. Patent Publication Nos. 2013/0287748, 2014/0227237, 2014/0099309, and 2014/0050708, and these references are incorporated by reference in their entirety.
  • the spacer or hinge is present between the antigen-binding domain and the transmembrane domain, such as is between the antigen-binding domain and plasma membrane when expressed on a cell.
  • the spacer or hinge is derived from IgG subclass (such as IgG1 and IgG4, IgD or CD8 (see e.g., Qin et al. (2017) J. Hematol. Oncol., 10:68). In some embodiments, the spacer or hinge is derived from IgG1.
  • the spacer and transmembrane domain are the hinge and transmembrane domain derived from CD8, such as having an exemplary sequence set forth in SEQ ID NO: 246, 483, or 897 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO:246, 483, or 897.
  • the endodomain comprises at CD3-zeta signaling domain.
  • the CD3-zeta signaling domain comprises the sequence of amino acids set forth in SEQ ID NO: 243 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to SEQ ID NO: 247 and retains the activity of T cell signaling.
  • the endodomain of a CAR can further comprise a costimulatory signaling domain or region to further modulate immunomodulatory responses of the T-cell.
  • the costimulatory signaling domain is or comprises a costimulatory region, or is derived from a costimulatory region, of CD28, ICOS, 41BB or OX40.
  • the costimulatory signaling domain is a derived from CD28 or 4-1BB and comprises the sequence of amino acids set forth in any of SEQ ID NOS: 484-487 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to SEQ ID NO:484-487 and retains the activity of T cell costimulatory signaling.
  • a polynucleotide encoding an ICOSL polypeptide and encoding one or more proteins, such as a recombinant antigen receptor (e.g., chimeric antigen receptor (CAR) or engineered T cell receptor (TCR)), a marker, and one or more self-cleaving peptides.
  • a recombinant antigen receptor e.g., chimeric antigen receptor (CAR) or engineered T cell receptor (TCR)
  • CAR chimeric antigen receptor
  • TCR engineered T cell receptor
  • the construct encoding the CAR further encodes a second protein, such as a marker, e.g. detectable protein, separated from the CAR by a self-cleaving peptide sequence.
  • the nucleic acid encoding the variant ICOSL polypeptide is separated from the one or more sequence(s) that is a nucleic acid encoding a protein, wherein the protein encodes a recombinant antigen receptor (e.g., CAR or TCR), a marker, a cytokine, or a chemokine.
  • a recombinant antigen receptor e.g., CAR or TCR
  • Any of the nucleotide sequences can be in a vector, such as viral vector.
  • the viral vector is a lentiviral vector or retroviral vector.
  • the self-cleaving peptide sequence is an F2A, T2A, E2A or P2A self-cleaving peptide.
  • Exemplary sequences of a T2A self-cleaving peptide are set for the in any one of SEQ ID NOS: 250, 488, 860-862 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to any of SEQ ID NOS: 250, 488, 860-862.
  • the T2A is encoded by the sequence of nucleotides set forth in SEQ ID NO:249 or a sequence that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to any of SEQ ID NO: 249.
  • An exemplary sequence of a P2A self-cleaving peptide is set in SEQ ID NO: 863 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to SEQ ID NOS: 863.
  • nucleic acid construct that encodes more than one P2A self-cleaving peptide such as a P2A1 and P2A2
  • the nucleotide sequence P2A1 and P2A2 each encode the P2A set forth in SEQ ID NO:863
  • the nucleotide sequence may be different to avoid recombination between sequences.
  • the marker is a detectable protein, such as a fluorescent protein, e.g., a green fluorescent protein (GFP) or blue fluorescent protein (BFP).
  • a fluorescent protein e.g., a green fluorescent protein (GFP) or blue fluorescent protein (BFP).
  • GFP green fluorescent protein
  • BFP blue fluorescent protein
  • Exemplary sequences of a fluorescent protein marker are set forth in SEQ ID NO:489, 858, 859, 903 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to SEQ ID NO: 489, 858, 859, 903.
  • the CAR is an anti-CD19 CAR that has the sequence of amino acids set forth in any of SEQ ID NOS: 479, 490, 491, 492, 898, 899, 901, or 902 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to any one of SEQ ID NOS: 479, 490, 491, 492, 898, 899, 901, or 902.
  • the CAR is encoded by a sequence of nucleotides set forth in SEQ ID NO: 248 or 900 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to any one of SEQ ID NO: 248 or 900.
  • the engineered T-cell possesses a TCR, including a recombinant or engineered TCR.
  • the TCR can be a native TCR.
  • Those of skill in the art will recognize that generally native mammalian T-cell receptors comprise an alpha and a beta chain (or a gamma and a delta chain) involved in antigen specific recognition and binding.
  • the TCR is an engineered TCR that is modified.
  • the TCR of an engineered T-cell specifically binds to a tumor associated or tumor specific antigen presented by an APC.
  • the immunomodulatory polypeptides such as transmembrane immunomodulatory polypeptides or secretable immunomodulatory polypeptides
  • engineered cells such as engineered T cells or engineered APCs
  • a variety of methods to introduce a DNA construct into primary T cells are known in the art.
  • viral transduction or plasmid electroporation are employed.
  • the nucleic acid molecule encoding the immunomodulatory protein, or the expression vector comprises a signal peptide that localizes the expressed transmembrane immunomodulatory proteins to the cellular membrane or for secretion.
  • a nucleic acid encoding a transmembrane immunomodulatory proteins of the invention is sub-cloned into a viral vector, such as a retroviral vector, which allows expression in the host mammalian cell.
  • the expression vector can be introduced into a mammalian host cell and, under host cell culture conditions, the immunomodulatory protein is expressed on the surface or is secreted.
  • primary T-cells can be purified ex vivo (CD4 cells or CD8 cells or both) and stimulated with an activation protocol consisting of various TCR/CD28 agonists, such as anti-CD3/anti-CD28 coated beads.
  • an activation protocol consisting of various TCR/CD28 agonists, such as anti-CD3/anti-CD28 coated beads.
  • a recombinant expression vector containing an immunomodulatory polypeptide can be stably introduced into the primary T cells through art standard lentiviral or retroviral transduction protocols or plasmid electroporation strategies.
  • Cells can be monitored for immunomodulatory polypeptide expression by, for example, flow cytometry using anti-epitope tag or antibodies that cross-react with native parental molecule and polypeptides comprising variant ICOSL.
  • T-cells that express the immunomodulatory polypeptide can be enriched through sorting with anti-epitope tag antibodies or enriched for high or low expression depending on the application.
  • the engineered T-cell can be assayed for appropriate function by a variety of means.
  • the engineered CAR or TCR co-expression can be validated to show that this part of the engineered T cell was not significantly impacted by the expression of the immunomodulatory protein.
  • standard in vitro cytotoxicity, proliferation, or cytokine assays e.g., IFN-gamma expression
  • Exemplary standard endpoints are percent lysis of the tumor line, proliferation of the engineered T-cell, or IFN-gamma protein expression in culture supernatants.
  • an engineered construct which results in statistically significant increased lysis of tumor line, increased proliferation of the engineered T-cell, or increased IFN-gamma expression over the control construct can be selected for.
  • non-engineered, such as native primary or endogenous T-cells could also be incorporated into the same in vitro assay to measure the ability of the immunomodulatory polypeptide construct expressed on the engineered cells, such as engineered T-cells, to modulate activity, including, in some cases, to activate and generate effector function in bystander, native T-cells.
  • Increased expression of activation markers such as CD69, CD44, or CD62L could be monitored on endogenous T cells, and increased proliferation and/or cytokine production could indicate desired activity of the immunomodulatory protein expressed on the engineered T cells.
  • the similar assays can be used to compare the function of engineered T cells containing the CAR or TCR alone to those containing the CAR or TCR and a TIP construct.
  • these in vitro assays are performed by plating various ratios of the engineered T cell and a “tumor” cell line containing the cognate CAR or TCR antigen together in culture. Standard endpoints are percent lysis of the tumor line, proliferation of the engineered T cell, or IFN-gamma production in culture supernatants.
  • An engineered immunomodulatory protein which resulted in statistically significant increased lysis of tumor line, increased proliferation of the engineered T cell, or increased IFN-gamma production over the same TCR or CAR construct alone can be selected for.
  • Engineered human T cells can be analyzed in immunocompromised mice, like the NSG strain, which lacks mouse T, NK and B cells.
  • Engineered human T cells in which the CAR or TCR binds a target counter-structure on the xenograft and is co-expressed with the TIP affinity modified IgSF domain can be adoptively transferred in vivo at different cell numbers and ratios compared to the xenograft.
  • engraftment of CD19+ leukemia tumor lines containing a luciferase/GFP vector can be monitored through bioluminescence or ex vivo by flow cytometry.
  • the xenograft is introduced into the murine model, followed by the engineered T cells several days later.
  • Engineered T cells containing the immunomodulatory protein can be assayed for increased survival, tumor clearance, or expanded engineered T cells numbers relative to engineered T cells containing the CAR or TCR alone.
  • endogenous, native (i.e., non-engineered) human T cells could be co-adoptively transferred to look for successful epitope spreading in that population, resulting in better survival or tumor clearance.
  • infectious agents that contain nucleic acids encoding any of the variant polypeptides, such as ICOSL vIgD polypeptides, including secretable or transmembrane immunomodulatory proteins described herein.
  • infectious agents can deliver the nucleic acids encoding the variant immunomodulatory polypeptides described herein, such as ICOSL vIgD polypeptides, to a target cell in a subject, e.g., immune cell and/or antigen-presenting cell (APC) or tumor cell in a subject.
  • nucleic acids contained in such infectious agents, and/or nucleic acids for generation or modification of such infectious agents such as vectors and/or plasmids, and compositions containing such infectious agents.
  • a variant ICOSL polypeptide is expressed in an infectious agent (e.g. viral or bacterial agent) which, upon administration to a subject, is able to infect a cell in vivo, such as an immune cell (e.g. T cell or antigen presenting cell) or tumor, for delivery or expression of the variant polypeptide as a TIP or a SIP in the cell.
  • infectious agent e.g. viral or bacterial agent
  • an immune cell e.g. T cell or antigen presenting cell
  • tumor for delivery or expression of the variant polypeptide as a TIP or a SIP in the cell.
  • the infectious agent is a microorganism or a microbe. In some embodiments, the infectious agent is a virus or a bacterium. In some embodiments, the infectious agent is a virus. In some embodiments, the infectious agent is a bacterium. In some embodiments, such infectious agents can deliver nucleic acid sequences encoding any of the variant polypeptides, such as ICOSL vIgD polypeptides, including secretable or transmembrane immunomodulatory proteins, described herein. Thus, in some embodiments, the cell in a subject that is infected or contacted by the infectious agents can be rendered to express on the cell surface or secrete, the variant immunomodulatory polypeptides.
  • the infectious agent can also deliver one or more other therapeutics or nucleic acids encoding other therapeutics to the cell and/or to an environment within the subject.
  • other therapeutics that can be delivered by the infectious agents include cytokines or other immunomodulatory molecules.
  • the infectious agent e.g., virus or bacteria
  • the infectious agent can be administered to the subject.
  • the infectious agent can be introduced to cells from the subject ex vivo.
  • the variant polypeptides such as ICOSL vIgD polypeptides, including transmembrane immunomodulatory proteins, expressed by the cell infected by the infectious agent is a transmembrane protein and is surface expressed.
  • the variant polypeptides, such as ICOSL vIgD polypeptides, including secretable immunomodulatory proteins, expressed by the cell infected by the infectious agent is expressed and secreted from the cell.
  • the transmembrane immunomodulatory protein or secreted immunomodulatory protein can be any described herein.
  • the cells in the subject that are targeted by the infectious agent include a tumor cell, an immune cell, and/or an antigen-presenting cell (APC).
  • the infectious agent targets a cell in the tumor microenvironment (TME).
  • the infectious agent delivers the nucleic acids encoding the variant polypeptides, such as ICOSL vIgD polypeptides, including secretable or transmembrane immunomodulatory proteins, to an appropriate cell (for example, an APC, such as a cell that displays a peptide/MHC complex on its cell surface, such as a dendritic cell) or tissue (e.g., lymphoid tissue) that modulate an immune response and/or a specific cell-medicated immune response, e.g., CD4 and/or CD8 T cell response, which CD8 T cell response may include a cytotoxic T cell (CTL) response.
  • CTL cytotoxic T cell
  • the infectious agent targets an APC, such as a dendritic cell (DC).
  • the nucleic acid molecule delivered by the infectious agents described herein include appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequences encoding the variant immunomodulatory polypeptides, in a particular target cell, e.g., regulatory elements such as promoters.
  • the infectious agent that contains nucleic acid sequences encoding the immunomodulatory polypeptides can also contain nucleic acid sequences that encode one or more additional gene products, e.g., cytokines, prodrug converting enzymes, cytotoxins and/or detectable gene products.
  • the infectious agent is an oncolytic virus and the virus can include nucleic acid sequences encoding additional therapeutic gene products (see, e.g., Kim et al., (2009) Nat Rev Cancer 9:64-71; Garcia-Aragoncillo et al., (2010) Curr Opin Mol Ther 12:403-411; see U.S. Pat. Nos.
  • the additional gene product can be a therapeutic gene product that can result in death of the target cell (e.g., tumor cell) or gene products that can augment or boost or regulate an immune response (e.g., cytokine).
  • Exemplary gene products also include among an anticancer agent, an anti-metastatic agent, an antiangiogenic agent, an immunomodulatory molecule, an immune checkpoint inhibitor, an antibody, a cytokine, a growth factor, an antigen, a cytotoxic gene product, a pro-apoptotic gene product, an anti-apoptotic gene product, a cell matrix degradative gene, genes for tissue regeneration and reprogramming human somatic cells to pluripotency, and other genes described herein or known to one of skill in the art.
  • the additional gene product is Granulocyte-macrophage colony-stimulating factor (GM-CSF).
  • the infectious agent is a virus.
  • the infectious agent is an oncolytic virus, or a virus that targets particular cells, e.g., immune cells.
  • the infectious agent targets a tumor cell and/or cancer cell in the subject.
  • the infectious agent targets an immune cell or an antigen-presenting cell (APC).
  • APC antigen-presenting cell
  • the infectious agent is an oncolytic virus.
  • Oncolytic viruses are viruses that accumulate in tumor cells and replicate in tumor cells. By virtue of replication in the tumor cells, and optional delivery of nucleic acids encoding variant ICOSL polypeptides or immunomodulatory polypeptides described herein, tumor cells are lysed, and the tumor shrinks and can be eliminated.
  • Oncolytic viruses can also have a broad host and cell type range. For example, oncolytic viruses can accumulate in immunoprivileged cells or immunoprivileged tissues, including tumors and/or metastases, and also including wounded tissues and cells, thus allowing the delivery and expression of nucleic acids encoding the variant immunomodulatory polypeptides described herein in a broad range of cell types. Oncolytic viruses can also replicate in a tumor cell specific manner, resulting in tumor cell lysis and efficient tumor regression.
  • oncolytic viruses include adenoviruses, adeno-associated viruses, herpes viruses, Herpes Simplex Virus, vesticular stomatic virus, Reovirus, Newcastle Disease virus, parvovirus, measles virus, vesticular stomatitis virus (VSV), Coxsackie virus and Vaccinia virus.
  • oncolytic viruses can specifically colonize solid tumors, while not infecting other organs, and can be used as an infectious agent to deliver the nucleic acids encoding the variant immunomodulatory polypeptides described herein to such solid tumors.
  • Oncolytic viruses for use in delivering the nucleic acids encoding variant ICOSL polypeptides or immunomodulatory polypeptides described herein can be any of those known to one of skill in the art and include, for example, vesicular stomatitis virus, see, e.g., U.S. Pat. Nos. 7,731,974, 7,153,510, 6,653,103 and U.S. Pat. Pub. Nos. 2010/0178684, 2010/0172877, 2010/0113567, 2007/0098743, 20050260601, 20050220818 and EP Pat. Nos. 1385466, 1606411 and 1520175; herpes simplex virus, see, e.g., U.S. Pat. Nos.
  • Oncolytic viruses also include viruses that have been genetically altered to attenuate their virulence, to improve their safety profile, enhance their tumor specificity, and they have also been equipped with additional genes, for example cytotoxins, cytokines, prodrug converting enzymes to improve the overall efficacy of the viruses (see, e.g., Kirn et al., (2009) Nat Rev Cancer 9:64-71; Garcia-Aragoncillo et al., (2010) Curr Opin Mol Ther 12:403-411; see U.S. Pat. Nos. 7,588,767, 7,588,771, 7,662,398 and 7,754,221 and U.S. Pat. Publ. Nos.
  • the oncolytic viruses can be those that have been modified so that they selectively replicate in cancerous cells, and, thus, are oncolytic.
  • the oncolytic virus is an adenovirus that has been engineered to have modified tropism for tumor therapy and also as gene therapy vectors.
  • the infectious agent is a modified herpes simplex virus.
  • the infectious agent is a modified version of Talimogene laherparepvec (also known as T-Vec, Imlygic or OncoVex GM-CSF), that is modified to contain nucleic acids encoding any of the variant ICOSL polypeptides or immunomodulatory polypeptides described herein.
  • the infectious agent is a modified herpes simplex virus that is described, e.g., in WO 2007/052029, WO 1999/038955, US 2004/0063094, US 2014/0154216, or, variants thereof.
  • the infectious agent is a virus that targets a particular type of cells in a subject that is administered the virus, e.g., a virus that targets immune cells or antigen-presenting cells (APCs).
  • APCs antigen-presenting cells
  • DCs Dendritic cells
  • MHC major histocompatibility complex
  • the infectious agent is a virus that specifically can target DCs to deliver nucleic acids encoding the variant ICOSL polypeptide or immunomodulatory polypeptides for expression in DCs.
  • the virus is a lentivirus or a variant or derivative thereof, such as an integration-deficient lentiviral vector.
  • the virus is a lentivirus that is pseudotyped to efficiently bind to and productively infect cells expressing the cell surface marker dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), such as DCs.
  • DC-SIGN dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin
  • the virus is a lentivirus pseudotyped with a Sindbis virus E2 glycoprotein or modified form thereof, such as those described in WO 2013/149167.
  • the virus allows for delivery and expression of a sequence of interest (e.g., a nucleic acid encoding any of the variant ICOSL polypeptides or immunomodulatory polypeptides described herein) to a DC.
  • a sequence of interest e.g., a nucleic acid encoding any of the variant ICOSL polypeptides or immunomodulatory polypeptides described herein
  • the virus includes those described in WO 2008/011636, US 2011/0064763, Tareen et al. (2014) Mol. Ther., 22:575-587, or variants thereof.
  • Exemplary of a dendritic cell-tropic vector platform is ZVexTM
  • the infectious agent is a bacterium.
  • the bacteria can deliver nucleic acids encoding any of the variant immunomodulatory polypeptides described herein, e.g., variant ICOSL polypeptide or immunomodulatory polypeptide, to a target cell in the subject, such as a tumor cell, an immune cell, an antigen-presenting cell and/or a phagocytic cell.
  • the bacterium can be preferentially targeted to a specific environment within a subject, such as a tumor microenvironment (TME), for expression and/or secretion of the variant immunomodulatory polypeptides and/or to target specific cells in the environment for expression of the variant immunomodulatory polypeptides.
  • TEE tumor microenvironment
  • the bacterium delivers the nucleic acids to the cells via bacterial-mediated transfer of plasmid DNA to mammalian cells (also referred to as “bactofection”). For example, in some embodiments, delivery of genetic material is achieved through entry of the entire bacterium into target cells. In some embodiments, spontaneous or induced bacterial lysis can lead to the release of plasmid for subsequent eukaryotic cell expression. In some embodiments, the bacterium can deliver nucleic acids to non-phagocytic mammalian cells (e.g., tumor cells) and/or to phagocytic cells, e.g., certain immune cells and/or APCs.
  • non-phagocytic mammalian cells e.g., tumor cells
  • phagocytic cells e.g., certain immune cells and/or APCs.
  • the nucleic acids delivered by the bacterium can be transferred to the nucleus of the cell in the subject for expression.
  • the nucleic acids also include appropriate nucleic acid sequences necessary for the expression of the operably linked sequences encoding the variant immunomodulatory polypeptides in a particular host cell, e.g., regulatory elements such as promoters or enhancers.
  • the infectious agent that is a bacterium can deliver nucleic acids encoding the immunomodulatory proteins in the form of an RNA, such as a pre-made translation-competent RNA delivered to the cytoplasm of the target cell for translation by the target cell's machinery.
  • the bacterium can replicate and lyse the target cells, e.g., tumor cells.
  • the bacterium can contain and/or release nucleic acid sequences and/or gene products in the cytoplasm of the target cells, thereby killing the target cell, e.g., tumor cell.
  • the infectious agent is bacterium that can replicate specifically in a particular environment in the subject, e.g., tumor microenvironment (TME).
  • TEE tumor microenvironment
  • the bacterium can replicate specifically in anaerobic or hypoxic microenvironments.
  • conditions or factors present in particular environments e.g., aspartate, serine, citrate, ribose or galactose produced by cells in the TME, can act as chemoattractants to attract the bacterium to the environment.
  • the bacterium can express and/or secrete the immunomodulatory proteins described herein in the environment, e.g., TME.
  • the infectious agent is a bacterium that is a Listeria sp., a Bifidobacterium sp., an Escherichia sp., a Clostridium sp., a Salmonella sp., a Shigella sp., a Vibrio sp. or a Yersinia sp.
  • the bacterium is selected from among one or more of Listeria monocytogenes, Salmonella typhimurium, Salmonella choleraesuis, Escherichia coli, Vibrio cholera, Clostridium perfringens, Clostridium butyricum, Clostridium novyi, Clostridium acetobutylicum, Bifidobacterium infantis, Bifidobacterium longum and Bifidobacterium adolescentis .
  • the bacterium is an engineered bacterium.
  • the bacterium is an engineered bacterium such as those described in, e.g., Seow and Wood (2009) Molecular Therapy 17(5):767-777; Baban et al. (2010) Bioengineered Bugs 1:6, 385-394; Patyar et al. (2010) J Biomed Sci 17:21; Tangney et al. (2010) Bioengineered Bugs 1:4, 284-287; van Pijkeren et al. (2010) Hum Gene Ther. 21(4):405-416; WO 2012/149364; WO 2014/198002; U.S. Pat. Nos.
  • the bacterium can be modified to deliver nucleic acid sequences encoding any of the variant immunomodulatory polypeptides, conjugates and/or fusions provided herein, and/or to express such variant immunomodulatory polypeptides in the subject.
  • nucleic acids which encode any of the various provided embodiments of the variant ICOSL polypeptides or immunomodulatory polypeptides provided herein.
  • nucleic acids provided herein, including all described below are useful in recombinant production (e.g., expression) of variant ICOSL polypeptides or immunomodulatory polypeptides provided herein.
  • nucleic acids provided herein, including all described below are useful in expression of variant ICOSL polypeptides or immunomodulatory polypeptides provided herein in cells, such as in engineered cells, e.g. immune cells, or infectious agent cells.
  • the nucleic acids provided herein can be in the form of RNA or in the form of DNA, and include mRNA, cRNA, recombinant or synthetic RNA and DNA, and cDNA.
  • the nucleic acids provided herein are typically DNA molecules, and usually double-stranded DNA molecules. However, single-stranded DNA, single-stranded RNA, double-stranded RNA, and hybrid DNA/RNA nucleic acids or combinations thereof comprising any of the nucleotide sequences of the invention also are provided.
  • engineered cells such as engineered immune cells, containing any of the provided nucleic acid molecules or any of the variant ICOSL polypeptides or immunomodulatory polypeptides, such as any of the transmembrane immunomodulatory polypeptides or secretable immunomodulatory polypeptides.
  • infectious agents such as bacterial or viral cells, containing any of the provided nucleic acid molecules or any of the variant ICOSL polypeptides or immunomodulatory polypeptides, such as any of the transmembrane immunomodulatory polypeptides or secretable immunomodulatory polypeptides.
  • the nucleic acids encoding the variant polypeptides or immunomodulatory polypeptides provided herein can be introduced into cells using recombinant DNA and cloning techniques.
  • a recombinant DNA molecule encoding an immunomodulatory polypeptide is prepared. Methods of preparing such DNA molecules are well known in the art. For instance, sequences coding for the peptides could be excised from DNA using suitable restriction enzymes. Alternatively, the DNA molecule could be synthesized using chemical synthesis techniques, such as the phosphoramidite method. Also, a combination of these techniques could be used.
  • a recombinant or synthetic nucleic acid may be generated through polymerase chain reaction (PCR).
  • a DNA insert can be generated encoding one or more variant ICOSL polypeptides containing at least one affinity-modified IgSF domain and, in some embodiments, a signal peptide, a transmembrane domain and/or an endodomain in accord with the provided description.
  • This DNA insert can be cloned into an appropriate transduction/transfection vector as is known to those of skill in the art. Also provided are expression vectors containing the nucleic acid molecules.
  • the expression vectors are capable of expressing the immunomodulatory proteins in an appropriate cell under conditions suited to expression of the protein.
  • nucleic acid molecule or an expression vector comprises the DNA molecule that encodes the immunomodulatory protein operatively linked to appropriate expression control sequences. Methods of effecting this operative linking, either before or after the DNA molecule is inserted into the vector, are well known.
  • Expression control sequences include promoters, activators, enhancers, operators, ribosomal binding sites, start signals, stop signals, cap signals, polyadenylation signals, and other signals involved with the control of transcription or translation.
  • expression of the immunomodulatory protein is controlled by a promoter or enhancer to control or regulate expression.
  • the promoter is operably linked to the portion of the nucleic acid molecule encoding the variant polypeptide or immunomodulatory protein.
  • the promotor is a constitutively active promotor (such as a tissue-specific constitutively active promotor or other constitutive promotor).
  • the promotor is an inducible promotor, which may be responsive to an inducing agent (such as a T cell activation signal).
  • a constitutive promoter is operatively linked to the nucleic acid molecule encoding the variant polypeptide or immunomodulatory protein.
  • Exemplary constitutive promoters include the Simian vacuolating virus 40 (SV40) promoter, the cytomegalovirus (CMV) promoter, the ubiquitin C (UbC) promoter, and the EF-1 alpha (EF1a) promoter.
  • the constitutive promoter is tissue specific.
  • the promoter allows for constitutive expression of the immunomodulatory protein in specific tissues, such as immune cells, lymphocytes, or T cells. Exemplary tissue-specific promoters are described in U.S. Pat. No. 5,998,205, including, for example, a fetoprotein, DF3, tyrosinase, CEA, surfactant protein, and ErbB2 promoters.
  • an inducible promoter is operatively linked to the nucleic acid molecule encoding the variant polypeptide or immunomodulatory protein such that expression of the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.
  • the promoter can be a regulated promoter and transcription factor expression system, such as the published tetracycline-regulated systems or other regulatable systems (see, e.g. published International PCT Appl. No. WO 01/30843), to allow regulated expression of the encoded polypeptide.
  • An exemplary regulatable promoter system is the Tet-On (and Tet-Off) system available, for example, from Clontech (Palo Alto, Calif.).
  • This promoter system allows the regulated expression of the transgene controlled by tetracycline or tetracycline derivatives, such as doxycycline.
  • Other regulatable promoter systems are known (see e.g., published U.S. Application No. 2002-0168714, entitled “Regulation of Gene Expression Using Single-Chain, Monomeric, Ligand Dependent Polypeptide Switches,” which describes gene switches that contain ligand binding domains and transcriptional regulating domains, such as those from hormone receptors).
  • an engineered T cell comprises an expression vector encoding the immunomodulatory protein and a promotor operatively linked to control expression of the immunomodulatory protein.
  • the engineered T cell can be activated, for example by signaling through an engineered T cell receptor (TCR) or a chimeric antigen rector (CAR), and thereby triggering expression and secretion of the immunomodulatory protein through the responsive promotor.
  • TCR engineered T cell receptor
  • CAR chimeric antigen rector
  • an inducible promoter is operatively linked to the nucleic acid molecule encoding the immunomodulatory protein such that the immunomodulatory protein is expressed in response to a nuclear factor of activated T-cells (NFAT) or nuclear factor kappa-light-chain enhancer of activated B cells (NF- ⁇ B).
  • the inducible promoter comprises a binding site for NFAT or NF- ⁇ B.
  • the promoter is an NFAT or NF- ⁇ B promoter or a functional variant thereof.
  • the nucleic acids make it possible to control the expression of immunomodulatory protein while also reducing or eliminating the toxicity of the immunomodulatory protein.
  • engineered immune cells comprising the nucleic acids of the invention express and secrete the immunomodulatory protein only when the cell (e.g., a T-cell receptor (TCR) or a chimeric antigen receptor (CAR) expressed by the cell) is specifically stimulated by an antigen and/or the cell (e.g., the calcium signaling pathway of the cell) is non-specifically stimulated by, e.g., phorbol myristate acetate (PMA)/Ionomycin.
  • TCR T-cell receptor
  • CAR chimeric antigen receptor
  • PMA phorbol myristate acetate
  • the expression and, in some cases, secretion, of immunomodulatory protein can be controlled to occur only when and where it is needed (e.g., in the presence of an infectious disease-causing agent, cancer, or at a tumor site), which can decrease or avoid undesired immunomodulatory protein interactions.
  • the nucleic acid encoding an immunomodulatory protein described herein comprises a suitable nucleotide sequence that encodes a NFAT promoter, NF- ⁇ B promoter, or a functional variant thereof.
  • NFAT promoter as used herein means one or more NFAT responsive elements linked to a minimal promoter.
  • NF- ⁇ B promoter refers to one or more NF- ⁇ B responsive elements linked to a minimal promoter.
  • the minimal promoter of a gene is a minimal human IL-2 promoter or a CMV promoter.
  • the NFAT responsive elements may comprise, e.g., NFAT1, NFAT2, NFAT3, and/or NFAT4 responsive elements.
  • the NFAT promoter, NF- ⁇ B promoter, or a functional variant thereof may comprise any number of binding motifs, e.g., at least two, at least three, at least four, at least five, or at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, or up to twelve binding motifs.
  • a nucleic acid provided herein further comprises nucleotide sequence that encodes a secretory or signal peptide operably linked to the nucleic acid encoding an immunomodulatory polypeptide such that a resultant soluble immunomodulatory polypeptide is recovered from the culture medium, host cell, or host cell periplasm.
  • the appropriate expression control signals are chosen to allow for membrane expression of an immunomodulatory polypeptide.
  • the resulting expression vector having the DNA molecule thereon is used to transform, such as transduce, an appropriate cell.
  • the introduction can be performed using methods well known in the art. Exemplary methods include those for transfer of nucleic acids encoding the receptors, including via viral, e.g., retroviral or lentiviral, transduction, transposons, and electroporation.
  • the expression vector is a viral vector.
  • the nucleic acid is transferred into cells by lentiviral or retroviral transduction methods.
  • the selection of a cell is dependent upon a number of factors recognized by the art. These include, for example, compatibility with the chosen expression vector, toxicity of the peptides encoded by the DNA molecule, rate of transformation, ease of recovery of the peptides, expression characteristics, bio-safety and costs. A balance of these factors must be struck with the understanding that not all cells can be equally effective for the expression of a particular DNA sequence.
  • the host cells can be a variety of eukaryotic cells, such as in yeast cells, or with mammalian cells such as Chinese hamster ovary (CHO) or HEK293 cells.
  • the host cell is a suspension cell and the polypeptide is engineered or produced in cultured suspension, such as in cultured suspension CHO cells, e.g. CHO-S cells.
  • the cell line is a CHO cell line that is deficient in DHFR (DHFR ⁇ ), such as DG44 and DUXB11.
  • the cell is deficient in glutamine synthase (GS), e.g.
  • the CHO cells such as suspension CHO cells, may be CHO-S-2H2 cells, CHO-S-clone 14 cells, or ExpiCHO-S cells.
  • expressing the provided ICOSL polypeptides from CHO cells results in a more homogenous composition of produced proteins.
  • the provided ICOSL polypeptides results in a more homogenous product when the proteins are expressed from CHO cells compared to ICOSL polypeptides containing the full ECD reference sequence and/or containing the protease cleavage site (e.g., LQQN/LT).
  • at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% of the composition of produced proteins containing an ICOSL variant polypeptide produced herein have the same amino acid length or are the same size. Techniques to assess homogeneity of size include high performance liquid chromatography (HPLC), size exclusion chromatography, SDS page, or sequencing.
  • host cells can also be prokaryotic cells, such as with E. coli .
  • the transformed recombinant host is cultured under polypeptide expressing conditions, and then purified to obtain a soluble protein.
  • Recombinant host cells can be cultured under conventional fermentation conditions so that the desired polypeptides are expressed. Such fermentation conditions are well known in the art.
  • the polypeptides provided herein can be recovered and purified from recombinant cell cultures by any of a number of methods well known in the art, including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, and affinity chromatography. Protein refolding steps can be used, as desired, in completing configuration of the mature protein.
  • HPLC high performance liquid chromatography
  • the cell is an immune cell, such as any described above in connection with preparing engineered cells.
  • such engineered cells are primary cells.
  • the engineered cells are autologous to the subject.
  • the engineered cells are allogeneic to the subject.
  • the engineered cells are obtained from a subject, such as by leukapheresis, and transformed ex vivo for expression of the immunomodulatory polypeptide, e.g. transmembrane immunomodulatory polypeptide or secretable immunomodulatory polypeptide.
  • nucleic acids encoding any of the variant immunomodulatory polypeptides contained in infectious agents described herein.
  • the infectious agents deliver the nucleic acids to a cell in the subject, and/or permit expression of the encoded variant polypeptides in the cell.
  • nucleic acids that are used to generate, produce or modify such infectious agents.
  • vectors and/or plasmids that contain nucleic acids encoding the variant immunomodulatory polypeptides, for generation of the infectious agents, delivery to the cells in a subject and/or expression of the variant immunomodulatory polypeptides in the cells in the subject.
  • the provided nucleic acids are recombinant viral or bacterial vectors containing nucleic acid sequences encoding the variant immunomodulatory polypeptides.
  • the recombinant vectors can be used to produce an infectious agent that contains nucleic acid sequences encoding the variant immunomodulatory polypeptides and/or to be delivered to a target cell in the subject for expression by the target cell.
  • the recombinant vector is an expression vector.
  • the recombinant vector includes appropriate sequences necessary for generation and/or production of the infectious agent and expression in the target cell.
  • the recombinant vector is a plasmid or cosmid. Plasmid or cosmid containing nucleic acid sequences encoding the variant immunomodulatory polypeptides, as described herein, is readily constructed using standard techniques well known in the art. For generation of the infectious agent, the vector or genome can be constructed in a plasmid form that can then be transfected into a packaging or producer cell line or a host bacterium. The recombinant vectors can be generated using any of the recombinant techniques known in the art. In some embodiments, the vectors can include a prokaryotic origin of replication and/or a gene whose expression confers a detectable or selectable marker such as a drug resistance for propagation and/or selection in prokaryotic systems.
  • the recombinant vector is a viral vector.
  • exemplary recombinant viral vectors include a lentiviral vector genome, poxvirus vector genome, vaccinia virus vector genome, adenovirus vector genome, adenovirus-associated virus vector genome, herpes virus vector genome, and alpha virus vector genome.
  • Viral vectors can be live, attenuated, replication conditional or replication deficient, non-pathogenic (defective), replication competent viral vector, and/or is modified to express a heterologous gene product, e.g., the variant immunomodulatory polypeptides provided herein.
  • Vectors for generation of viruses also can be modified to alter attenuation of the virus, which includes any method of increasing or decreasing the transcriptional or translational load.
  • Exemplary viral vectors that can be used include modified vaccinia virus vectors (see, e.g., Guerra et al., J. Virol. 80:985-98 (2006); Tartaglia et al., AIDS Research and Human Retroviruses 8: 1445-47 (1992); Gheradi et al., J. Gen. Virol. 86:2925-36 (2005); Mayr et al., Infection 3:6-14 (1975); Hu et al., J. Virol. 75: 10300-308 (2001); U.S. Pat. Nos.
  • adenovirus vector or adenovirus-associated virus vectors see., e.g., Molin et al., J. Virol. 72:8358-61 (1998); Narumi et al., Am J. Respir. Cell Mol. Biol. 19:936-41 (1998); Mercier et al., Proc. Natl. Acad. Sci. USA 101:6188-93 (2004); U.S. Pat. Nos.
  • retroviral vectors including those based upon murine leukemia virus (MuLV), gibbon ape leukemia virus (GaLV), ecotropic retroviruses, simian immunodeficiency virus (SIV), human immunodeficiency virus (HIV), and combinations (see, e.g., Buchscher et al., J. Virol. 66:2731-39 (1992); Johann et al., J. Virol.
  • MiLV murine leukemia virus
  • GaLV gibbon ape leukemia virus
  • SIV simian immunodeficiency virus
  • HAV human immunodeficiency virus
  • HIV-1 Human Immunodeficiency Virus
  • HIV-2 feline immunodeficiency virus
  • FIV feline immunodeficiency virus
  • equine infectious anemia virus Simian Immunodeficiency Virus
  • SIV Simian Immunodeficiency Virus
  • maedi/visna virus see, e.g., Pfeifer et al., Annu. Rev. Genomics Hum. Genet. 2: 177-211 (2001); Zufferey et al., J. Virol. 72: 9873, 1998; Miyoshi et al., J. Virol.
  • the recombinant vector can include regulatory sequences, such as promoter or enhancer sequences, that can regulate the expression of the viral genome, such as in the case for RNA viruses, in the packaging cell line (see, e.g., U.S. Pat. Nos. 5,385,839 and 5,168,062).
  • regulatory sequences such as promoter or enhancer sequences, that can regulate the expression of the viral genome, such as in the case for RNA viruses, in the packaging cell line (see, e.g., U.S. Pat. Nos. 5,385,839 and 5,168,062).
  • the recombinant vector is an expression vector, e.g., an expression vector that permits expression of the encoded gene product when delivered into the target cell, e.g., a cell in the subject, e.g., a tumor cell, an immune cell and/or an APC.
  • the recombinant expression vectors contained in the infectious agent are capable of expressing the immunomodulatory proteins in the target cell in the subject, under conditions suited to expression of the protein.
  • nucleic acids or an expression vector comprises a nucleic acid sequence that encodes the immunomodulatory protein operatively linked to appropriate expression control sequences. Methods of affecting this operative linking, either before or after the nucleic acid sequence encoding the immunomodulatory protein is inserted into the vector, are well known.
  • Expression control sequences include promoters, activators, enhancers, operators, ribosomal binding sites, start signals, stop signals, cap signals, polyadenylation signals, and other signals involved with the control of transcription or translation.
  • the promoter can be operably linked to the portion of the nucleic acid sequence encoding the immunomodulatory protein.
  • the promotor is a constitutively active promotor in the target cell (such as a tissue-specific constitutively active promotor or other constitutive promotor).
  • the recombinant expression vector may also include, lymphoid tissue-specific transcriptional regulatory elements (TRE) such as a B lymphocyte, T lymphocyte, or dendritic cell specific TRE. Lymphoid tissue specific TRE are known in the art (see, e.g., Thompson et al., Mol. Cell. Biol. 12:1043-53 (1992); Todd et al., J. Exp. Med. 177:1663-74 (1993); Penix et al., J. Exp. Med. 178:1483-96 (1993)).
  • TRE lymphoid tissue-specific transcriptional regulatory elements
  • the promotor is an inducible promotor, which may be responsive to an inducing agent (such as a T cell activation signal).
  • an inducing agent such as a T cell activation signal.
  • nucleic acids delivered to the target cell in the subject e.g., tumor cell, immune cell and/or APC, can be operably linked to any of the regulatory elements described above.
  • the vector is a bacterial vector, e.g., a bacterial plasmid or cosmid.
  • the bacterial vector is delivered to the target cell, e.g., tumor cells, immune cells and/or APCs, via bacterial-mediated transfer of plasmid DNA to mammalian cells (also referred to as “bactofection”).
  • the delivered bacterial vector also contains appropriate expression control sequences for expression in the target cells, such as a promoter sequence and/or enhancer sequences, or any regulatory or control sequences described above.
  • the bacterial vector contains appropriate expression control sequences for expression and/or secretion of the encoded variant polypeptides in the infectious agent, e.g., the bacterium.
  • polypeptides provided herein can also be made by synthetic methods.
  • Solid phase synthesis is the preferred technique of making individual peptides since it is the most cost-effective method of making small peptides.
  • well known solid phase synthesis techniques include the use of protecting groups, linkers, and solid phase supports, as well as specific protection and deprotection reaction conditions, linker cleavage conditions, use of scavengers, and other aspects of solid phase peptide synthesis. Peptides can then be assembled into the polypeptides as provided herein.
  • the variant ICOSL polypeptides provided herein exhibit immunomodulatory activity to modulate T cell activation.
  • ICOSL polypeptides modulate IFN-gamma expression in a primary T cell assay relative to a reference (e.g., unmodified) or wild-type ICOSL control.
  • modulation of IFN-gamma expression can increase or decrease IFN-gamma expression relative to the control.
  • Assays to determine specific binding and IFN-gamma expression are well-known in the art and include the MLR (mixed lymphocyte reaction) assays measuring interferon-gamma cytokine levels in culture supernatants (Wang et al., Cancer Immunol Res. 2014 September: 2(9):846-56), SEB (staphylococcal enterotoxin B) T cell stimulation assay (Wang et al., Cancer Immunol Res. 2014 September: 2(9):846-56), and anti-CD3 T cell stimulation assays (Li and Kurlander, J Transl Med. 2010: 8: 104).
  • MLR mixed lymphocyte reaction
  • SEB staphylococcal enterotoxin B
  • a variant ICOSL polypeptide can in some embodiments increase or, in alternative embodiments, decrease IFN-gamma (interferon-gamma) expression in a primary T-cell assay relative to a wild-type ICOSL control.
  • the polypeptide can increase IFN-gamma expression and, in alternative embodiments, decrease IFN-gamma expression in a primary T-cell assay relative to a wild-type ICOSL control.
  • the polypeptide can increase IFN-gamma expression and, in alternative embodiments, decrease IFN-gamma expression in a primary T-cell assay relative to a wild-type ICOSL control.
  • the format of the primary T-cell assay used to determine an increase in IFN-gamma expression can differ from that employed to assay for a decrease in IFN-gamma expression.
  • a Mixed Lymphocyte Reaction (MLR) assay can be used as described in Example 6.
  • MLR Mixed Lymphocyte Reaction
  • a soluble form of a variant ICOSL can be employed to determine the ability of the variant ICOSL to antagonize and thereby decrease the IFN-gamma expression in a MLR as likewise described in Example 6.
  • a co-immobilization assay in assaying for the ability of a variant ICOSL to increase IFN-gamma expression in a primary T-cell assay, can be used as described in Example 6.
  • a TCR signal provided in some embodiments by anti-CD3 antibody, is used in conjunction with a co-immobilized variant ICOSL to determine the ability to increase IFN-gamma expression relative to an ICOSL control.
  • a soluble form of a variant ICOSL that is multimerized to a degree to provide multivalent binding can be employed to determine the ability of the variant ICOSL to agonize and thereby increase the IFN-gamma expression in a MLR as likewise described in Example 6.
  • a T cell reporter assay in assaying for the ability of a variant ICOSL to modulate an increase or decrease IFN-gamma expression a T cell reporter assay can be used.
  • the T cell is a Jurkat T cell line or is derived from Jurkat T cell lines.
  • the reporter cell line e.g. Jurkat reporter cell
  • the reporter T cells also contain a reporter construct containing an inducible promoter responsive to T cell activation operably linked to a reporter.
  • the reporter is a fluorescent or luminescent reporter.
  • the reporter is luciferase.
  • the promoter is responsive to CD3 signaling. In some embodiments, the promoter is an NFAT promoter. In some embodiments, the promoter is responsive to costimulatory signaling, e.g. CD28 costimulatory signaling. In some embodiments, the promoter is an IL-2 promoter.
  • a reporter cell line is stimulated, such as by co-incubation with antigen presenting cells (APCs) expressing the wild-type ligand of the inhibitory receptor, e.g. ICOSL.
  • APCs antigen presenting cells
  • the APCs are artificial APCs.
  • Artificial APCs are well known to a skilled artisan.
  • artificial APCs are derived from one or more mammalian cell line, such as K562, CHO or 293 cells.
  • the Jurkat reporter cells are co-incubated with artificial APCs overexpressing the inhibitory ligand in the presence of the variant IgSF domain molecule or immunomodulatory protein, e.g., variant ICOSL polypeptide or immunomodulatory protein.
  • reporter expression is monitored, such as by determining the luminescence or fluorescence of the cells.
  • normal interactions between its inhibitory receptor and ligand result in a repression of or decrease in the reporter signal, such as compared to control, e.g. reporter expression by co-incubation of control T cells and APCs in which the inhibitory receptor and ligand interaction is not present, e.g. APCs that do not overexpress ICOSL.
  • a variant ICOSL polypeptide or immunomodulatory protein provided herein antagonizes the interaction, e.g. when provided in soluble form as a variant ICOSL-Fc or when expressed from the APC as a secretable immunomodulatory protein, thereby resulting in an increase in the reporter signal compared to the absence of the variant ICOSL polypeptide or immunomodulatory protein.
  • certain formats of a variant ICOSL polypeptide or immunomodulatory protein as provided herein may provide an agonist activity, thereby decreasing reporter expression compared to the absence of the variant ICOSL polypeptide or immunomodulatory protein.
  • the control typically involves use of the reference ICOSL, such as a wild-type of native ICOSL isoform from the same mammalian species from which the variant ICOSL was derived or developed. Irrespective of whether the binding affinity to either one or both of ICOS and CD28 is increased or decreased, a variant ICOSL in some embodiments will increase IFN-gamma expression and, in alternative embodiments, decrease IFN-gamma expression in a primary T-cell assay relative to a wild-type ICOSL control.
  • the reference ICOSL such as a wild-type of native ICOSL isoform from the same mammalian species from which the variant ICOSL was derived or developed.
  • a variant ICOSL increases IFN-gamma expression (i.e., protein expression) relative to a reference (e.g., unmodified) or wild-type ICOSL control by at least: 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or higher.
  • a variant ICOSL decreases IFN-gamma expression (i.e. protein expression) relative to a wild-type or unmodified ICOSL control by at least: 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or higher.
  • the wild-type ICOSL control is murine ICOSL, such as would typically be used for a variant ICOSL altered in sequence from that of a wild-type murine ICOSL sequence.
  • the wild-type ICOSL control is human ICOSL, such as would typically be used for a variant ICOSL altered in sequence from that of a wild-type human ICOSL sequence such as an ICOSL sequence comprising the sequence of amino acids of SEQ ID NO:32 or SEQ ID NO:196 or 545.
  • compositions containing any of the variant ICOSL polypeptides, immunodulatory proteins, conjugates, engineered cells or infectious agents described herein can further comprise a pharmaceutically acceptable excipient.
  • the pharmaceutical composition can contain one or more excipients for modifying, maintaining or preserving, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption, or penetration of the composition.
  • a pharmaceutical composition containing cells may differ from a pharmaceutical composition containing a protein.
  • the pharmaceutical composition is a solid, such as a powder, capsule, or tablet.
  • the components of the pharmaceutical composition can be lyophilized.
  • the solid pharmaceutical composition is reconstituted or dissolved in a liquid prior to administration.
  • the pharmaceutical composition is a liquid, for example variant ICOSL polypeptides dissolved in an aqueous solution (such as physiological saline or Ringer's solution).
  • the pH of the pharmaceutical composition is between about 4.0 and about 8.5 (such as between about 4.0 and about 5.0, between about 4.5 and about 5.5, between about 5.0 and about 6.0, between about 5.5 and about 6.5, between about 6.0 and about 7.0, between about 6.5 and about 7.5, between about 7.0 and about 8.0, or between about 7.5 and about 8.5).
  • the pharmaceutical composition comprises a pharmaceutically-acceptable excipient, for example a filler, binder, coating, preservative, lubricant, flavoring agent, sweetening agent, coloring agent, a solvent, a buffering agent, a chelating agent, or stabilizer.
  • a pharmaceutically-acceptable excipient for example a filler, binder, coating, preservative, lubricant, flavoring agent, sweetening agent, coloring agent, a solvent, a buffering agent, a chelating agent, or stabilizer.
  • pharmaceutically-acceptable fillers include cellulose, dibasic calcium phosphate, calcium carbonate, microcrystalline cellulose, sucrose, lactose, glucose, mannitol, sorbitol, maltol, pregelatinized starch, corn starch, or potato starch.
  • Examples of pharmaceutically-acceptable binders include polyvinylpyrrolidone, starch, lactose, xylitol, sorbitol, maltitol, gelatin, sucrose, polyethylene glycol, methyl cellulose, or cellulose.
  • Examples of pharmaceutically-acceptable coatings include hydroxypropyl methylcellulose (HPMC), shellac, corn protein zein, or gelatin.
  • Examples of pharmaceutically-acceptable disintegrants include polyvinylpyrrolidone, carboxymethyl cellulose, or sodium starch glycolate.
  • Examples of pharmaceutically-acceptable lubricants include polyethylene glycol, magnesium stearate, or stearic acid.
  • Examples of pharmaceutically-acceptable preservatives include methyl parabens, ethyl parabens, propyl paraben, benzoic acid, or sorbic acid.
  • Examples of pharmaceutically-acceptable sweetening agents include sucrose, saccharine, aspartame, or sorbitol.
  • Examples of pharmaceutically-acceptable buffering agents include carbonates, citrates, gluconates, acetates, phosphates, or tartrates.
  • the pharmaceutical composition further comprises an agent for the controlled or sustained release of the product, such as injectable microspheres, bio-erodible particles, polymeric compounds (polylactic acid, polyglycolic acid), beads, or liposomes.
  • an agent for the controlled or sustained release of the product such as injectable microspheres, bio-erodible particles, polymeric compounds (polylactic acid, polyglycolic acid), beads, or liposomes.
  • the pharmaceutical composition is sterile. Sterilization may be accomplished by filtration through sterile filtration membranes or radiation. Where the composition is lyophilized, sterilization using this method may be conducted either prior to or following lyophilization and reconstitution.
  • the composition for parenteral administration may be stored in lyophilized form or in solution.
  • parenteral compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • compositions containing the transmembrane immunomodulatory proteins including engineered cells expressing such transmembrane immunomodulatory proteins.
  • the pharmaceutical compositions and formulations include one or more optional pharmaceutically acceptable carrier or excipient.
  • Such compositions may comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives.
  • Compositions of the present invention are preferably formulated for intravenous administration.
  • the pharmaceutical composition contains infectious agents containing nucleic acid sequences encoding the immunomodulatory variant polypeptides. In some embodiments, the pharmaceutical composition contains a dose of infectious agents suitable for administration to a subject that is suitable for treatment. In some embodiments, the pharmaceutical composition contains an infectious agent that is a virus, at a single or multiple dosage amount, of between about between or between about 1 ⁇ 10 5 and about 1 ⁇ 10 12 plaque-forming units (pfu), 1 ⁇ 10 6 and 1 ⁇ 10 10 pfu, or 1 ⁇ 10 7 and 1 ⁇ 10 10 pfu, each inclusive, such as at least or at least about or at about 1 ⁇ 10 6 , 1 ⁇ 10 7 , 1 ⁇ 10 8 , 1 ⁇ 10 9 , 2 ⁇ 10 9 , 3 ⁇ 10 9 , 4 ⁇ 10 9 , 5 ⁇ 10 9 pfu or about 1 ⁇ 10 10 pfu.
  • infectious agent that is a virus, at a single or multiple dosage amount, of between about between or between about 1 ⁇ 10 5 and about 1 ⁇ 10 12 plaque-forming units (p
  • the pharmaceutical composition can contain a virus concentration of from or from about 10 5 to about 10 10 pfu/mL, for example, 5 ⁇ 10 6 to 5 ⁇ 10 9 or 1 ⁇ 10 7 to 1 ⁇ 10 9 pfu/mL, such as at least or at least about or at about 10 6 pfu/mL, 10 7 pfu/mL, 10 8 pfu/mL or 10 9 pfu/mL.
  • the pharmaceutical composition contains an infectious agent that is a bacterium, at a single or multiple dosage amount, of between about between or between about 1 ⁇ 10 3 and about 1 ⁇ 10 9 colony-forming units (cfu), 1 ⁇ 10 4 and 1 ⁇ 10 9 cfu, or 1 ⁇ 10 5 and 1 ⁇ 10 7 cfu, each inclusive, such as at least or at least about or at about 1 ⁇ 10 4 , 1 ⁇ 10 5 , 1 ⁇ 10 6 , 1 ⁇ 10 7 , 1 ⁇ 10 8 or 1 ⁇ 10 9 cfu.
  • infectious agent that is a bacterium, at a single or multiple dosage amount, of between about between or between about 1 ⁇ 10 3 and about 1 ⁇ 10 9 colony-forming units (cfu), 1 ⁇ 10 4 and 1 ⁇ 10 9 cfu, or 1 ⁇ 10 5 and 1 ⁇ 10 7 cfu, each inclusive, such as at least or at least about or at about 1 ⁇ 10 4 , 1 ⁇ 10 5 , 1 ⁇ 10 6 , 1 ⁇ 10 7 , 1 ⁇ 10 8 or 1 ⁇
  • the pharmaceutical composition can contain a bacterial concentration of from or from about 10 3 to about 10 8 cfu/mL, for example, 5 ⁇ 10 5 to 5 ⁇ 10 7 or 1 ⁇ 10 6 to 1 ⁇ 10 7 cfu/mL, such as at least or at least about or at about 10 5 cfu/mL, 10 6 cfu/mL, 10 7 cfu/mL or 10 8 cfu/mL
  • a pharmaceutically acceptable carrier may be a pharmaceutically acceptable material, composition, or vehicle that is involved in carrying or transporting cells of interest from one tissue, organ, or portion of the body to another tissue, organ, or portion of the body.
  • the carrier may be a liquid or solid filler, diluent, excipient, solvent, or encapsulating material, or some combination thereof.
  • Each component of the carrier must be “pharmaceutically acceptable” in that it must be compatible with the other ingredients of the formulation. It also must be suitable for contact with any tissue, organ, or portion of the body that it may encounter, meaning that it must not carry a risk of toxicity, irritation, allergic response, immunogenicity, or any other complication that excessively outweighs its therapeutic benefits.
  • the pharmaceutical composition is administered to a subject.
  • dosages and routes of administration of the pharmaceutical composition are determined according to the size and condition of the subject, according to standard pharmaceutical practice.
  • the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models such as mice, rats, rabbits, dogs, pigs, or monkeys. An animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. The exact dosage will be determined in light of factors related to the subject requiring treatment. Dosage and administration are adjusted to provide sufficient levels of the active compound or to maintain the desired effect. Factors that may be taken into account include the severity of the disease state, the general health of the subject, the age, weight, and gender of the subject, time and frequency of administration, drug combination(s), reaction sensitivities, and response to therapy.
  • compositions may be administered every 3 to 4 days, every week, or biweekly depending on the half-life and clearance rate of the particular formulation. The frequency of dosing will depend upon the pharmacokinetic parameters of the molecule in the formulation used. Typically, a composition is administered until a dosage is reached that achieves the desired effect. The composition may therefore be administered as a single dose, or as multiple doses (at the same or different concentrations/dosages) over time, or as a continuous infusion. Further refinement of the appropriate dosage is routinely made. Appropriate dosages may be ascertained through use of appropriate dose-response data.
  • a number of biomarkers or physiological markers for therapeutic effect can be monitored including T cell activation or proliferation, cytokine synthesis or production (e.g., production of TNF- ⁇ , IFN- ⁇ , IL-2), induction of various activation markers (e.g., CD25, IL-2 receptor), inflammation, joint swelling or tenderness, serum level of C-reactive protein, anti-collagen antibody production, and/or T cell-dependent antibody response(s).
  • the pharmaceutical composition is administered to a subject through any route, including orally, transdermally, by inhalation, intravenously, intra-arterially, intramuscularly, direct application to a wound site, application to a surgical site, intraperitoneally, by suppository, subcutaneously, intradermally, transcutaneously, by nebulization, intrapleurally, intraventricularly, intra-articularly, intraocularly, or intraspinally.
  • the dosage of the pharmaceutical composition is a single dose or a repeated dose.
  • the doses are given to a subject once per day, twice per day, three times per day, or four or more times per day.
  • about 1 or more (such as about 2 or more, about 3 or more, about 4 or more, about 5 or more, about 6 or more, or about 7 or more) doses are given in a week.
  • multiple doses are given over the course of days, weeks, months, or years.
  • a course of treatment is about 1 or more doses (such as about 2 or more does, about 3 or more doses, about 4 or more doses, about 5 or more doses, about 7 or more doses, about 10 or more doses, about 15 or more doses, about 25 or more doses, about 40 or more doses, about 50 or more doses, or about 100 or more doses).
  • an administered dose of the pharmaceutical composition is about 1 ⁇ g of protein per kg subject body mass or more (such as about 2 ⁇ g of protein per kg subject body mass or more, about 5 ⁇ g of protein per kg subject body mass or more, about 10 ⁇ g of protein per kg subject body mass or more, about 25 ⁇ g of protein per kg subject body mass or more, about 50 ⁇ g of protein per kg subject body mass or more, about 100 ⁇ g of protein per kg subject body mass or more, about 250 ⁇ g of protein per kg subject body mass or more, about 500 ⁇ g of protein per kg subject body mass or more, about 1 mg of protein per kg subject body mass or more, about 2 mg of protein per kg subject body mass or more, or about 5 mg of protein per kg subject body mass or more).
  • a therapeutic amount of a cell composition is administered.
  • 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).
  • a pharmaceutical composition comprising engineered cells, e.g. T cells, as described herein may be administered at a dosage of 10 4 to 10 9 cells/kg body weight, such as 10 5 to 10 6 cells/kg body weight, including all integer values within those ranges.
  • Engineered cell compositions, such as T cell compositions may also be administered multiple times at these dosages.
  • the cells 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.
  • a variety of means are known for determining if administration of a therapeutic composition of the invention sufficiently modulates immunological activity by eliminating, sequestering, or inactivating immune cells mediating or capable of mediating an undesired immune response; inducing, generating, or turning on immune cells that mediate or are capable of mediating a protective immune response; changing the physical or functional properties of immune cells; or a combination of these effects.
  • measurements of the modulation of immunological activity include, but are not limited to, examination of the presence or absence of immune cell populations (using flow cytometry, immunohistochemistry, histology, electron microscopy, polymerase chain reaction (PCR)); measurement of the functional capacity of immune cells including ability or resistance to proliferate or divide in response to a signal (such as using T-cell proliferation assays and pepscan analysis based on 3H-thymidine incorporation following stimulation with anti-CD3 antibody, anti-T-cell receptor antibody, anti-CD28 antibody, calcium ionophores, PMA (phorbol 12-myristate 13-acetate) antigen presenting cells loaded with a peptide or protein antigen; B cell proliferation assays); measurement of the ability to kill or lyse other cells (such as cytotoxic T cell assays); measurements of the cytokines, chemokines, cell surface molecules, antibodies and other products of the cells (e.g., by flow cytometry, enzyme-linked immunosorbent assays, Western blot
  • Suitable packaging for compositions such as ophthalmic compositions
  • suitable packaging for compositions are known in the art, and include, for example, vials (such as sealed vials), vessels, ampules, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. These articles of manufacture may further be sterilized and/or sealed.
  • kits comprising the pharmaceutical compositions (or articles of manufacture) described herein, which may further comprise instruction(s) on methods of using the composition, such as uses described herein.
  • the kits described herein may also include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for performing any methods described herein.
  • compositions described herein can be used in a variety of therapeutic applications, such as the treatment of a disease.
  • the pharmaceutical composition is used to treat inflammatory or autoimmune disorders, cancer, organ transplantation, viral infections, and/or bacterial infections in a mammal.
  • the pharmaceutical composition can modulate (e.g. increase or decrease) an immune response to treat the disease.
  • Such methods and uses include therapeutic methods and uses, for example, involving administration of the molecules or engineered cells, or compositions containing the same, to a subject having a disease, condition, or disorder.
  • the disease or disorder is an autoimmune or inflammatory disease or disorder.
  • the disease or disorder is a tumor or cancer.
  • the molecule or engineered cell is administered in an effective amount to effect treatment of the disease or disorder.
  • Uses include uses of molecules containing a variant ICOSL polypeptide, immunomodulatory protein, conjugate, engineered cell and infectious agents in such methods and treatments, and in the preparation of a medicament in order to carry out such therapeutic methods.
  • the methods are carried out by administering a variant ICOSL polypeptide, immunomodulatory protein, conjugate, engineered cell, and infectious agent, or compositions comprising the same, to the subject having or suspected of having the disease or condition. In some embodiments, the methods thereby treat the disease or condition or disorder in the subject.
  • the provided methods are applicable to therapeutic administration of variant ICOSL polypeptides, the immunomodulatory proteins, the conjugates, the engineered cells and infectious agents described herein. It is within the level of a skilled artisan, in view of the provided disclosure, to choose a format for the indication depending on the type of modulation of the immune response, e.g. increase or decrease that is desired.
  • a pharmaceutical composition provided herein that stimulates the immune response is administered, which can be useful, for example, in the treatment of cancer, viral infections, or bacterial infections.
  • the pharmaceutical composition contains a variant ICOSL polypeptide in a format that exhibits agonist activity of its cognate binding partner CD28 or ICOS and/or that stimulates or initiates costimulatory signaling via CD28 or ICOS.
  • Exemplary formats of an ICOSL polypeptide for use in connection with such therapeutic applications include, for example, an immunomodulatory protein or “stack” of a variant ICOSL polypeptide and an IgSF domain or variant thereof that binds to a tumor antigen (e.g.
  • Nkp30 or affinity-modified variant (also called a “tumor-localizing IgSF domain), a conjugate containing a variant ICOSL polypeptide linked to a tumor-targeting moiety (also called a tumor-localizing moiety), an engineered cell expressing a transmembrane immunomodulatory protein or an infectious agent comprising a nucleic acid molecule encoding a transmembrane immunomodulatory protein, such as for expression of the transmembrane immunomodulatory protein in an infected cell (e.g. tumor cell or APC, e.g. dendritic cell).
  • an infected cell e.g. tumor cell or APC, e.g. dendritic cell.
  • compositions comprising engineered cells and the methods described herein can be used in adoptive cell transfer applications.
  • cell compositions comprising engineered cells can be used in associated methods to, for example, modulate immunological activity in an immunotherapy approach to the treatment of, for example, a mammalian cancer or, in other embodiments the treatment of autoimmune disorders.
  • the methods employed generally comprise a method of contacting a TIP of the present invention with a mammalian cell under conditions that are permissive to specific binding of the affinity modified IgSF domain and modulation of the immunological activity of the mammalian cell.
  • immune cells such as tumor infiltrating lymphocytes (TILs), T-cells (including CD8+ or CD4+ T-cells), or APCs
  • TILs tumor infiltrating lymphocytes
  • T-cells including CD8+ or CD4+ T-cells
  • APCs APCs
  • the engineered cells can then contact a mammalian cell, such as an APC, a second lymphocyte or tumor cell in which modulation of immunological activity is desired under conditions that are permissive of specific binding of the affinity modified IgSF domain to a counter-structure on the mammalian cell such that immunological activity can be modulated in the mammalian cell.
  • Cells can be contacted in vivo or ex vivo.
  • the engineered cells are autologous cells. In other embodiments, the cells are allogeneic. In some embodiments, the cells are autologous engineered cells reinfused into the mammal from which it was isolated. In some embodiments, the cells are allogenic engineered cells infused into the mammal. In some embodiments, the cells are harvested from a patient's blood or tumor, engineered to express a polypeptide (such as the variant ICOSL polypeptide, immunomodulatory protein, or conjugate as described herein), expanded in an in vitro culture system (for example, by stimulating the cells), and reinfused into the patient to mediate tumor destruction.
  • a polypeptide such as the variant ICOSL polypeptide, immunomodulatory protein, or conjugate as described herein
  • the methods are conducted by adoptive cell transfer wherein cells expressing the TIP (e.g., a T-cell) are infused back into the patient.
  • the therapeutic compositions and methods of the invention are used in the treatment of a mammalian patient of cancers such as lymphoma, lymphoid leukemia, myeloid leukemia, cervical cancer, neuroblastoma, or multiple myeloma.
  • the pharmaceutical composition can be used to inhibit growth of mammalian cancer cells (such as human cancer cells).
  • a method of treating cancer can include administering an effective amount of any of the pharmaceutical compositions described herein to a subject with cancer.
  • the effective amount of the pharmaceutical composition can be administered to inhibit, halt, or reverse progression of cancers.
  • the cancers that can be treated by the pharmaceutical compositions and the treatment methods described herein include, but are not limited to, melanoma, bladder cancer, hematological malignancies (leukemia, lymphoma, myeloma), liver cancer, brain cancer, renal cancer, breast cancer, pancreatic cancer (adenocarcinoma), colorectal cancer, lung cancer (small cell lung cancer and non-small-cell lung cancer), spleen cancer, cancer of the thymus or blood cells (i.e., leukemia), prostate cancer, testicular cancer, ovarian cancer, uterine cancer, gastric carcinoma, a musculoskeletal cancer, a head and neck cancer, a gastrointestinal cancer, a germ cell cancer, or an endocrine and neuroendocrine cancer.
  • melanoma bladder cancer
  • hematological malignancies leukemia, lymphoma, myeloma
  • liver cancer brain cancer
  • renal cancer breast cancer
  • pancreatic cancer adenocarcinom
  • the cancer is Ewing's sarcoma. In some embodiments, the cancer is selected from melanoma, lung cancer, bladder cancer, and a hematological malignancy. In some embodiments, the cancer is a lymphoma, lymphoid leukemia, myeloid leukemia, cervical cancer, neuroblastoma, or multiple myeloma.
  • Human cancer cells can be treated in vivo, or ex vivo.
  • tissue or fluids containing cancer cells are treated outside the body and then the tissue or fluids are reintroduced back into the patient.
  • the cancer is treated in a human patient in vivo by administration of the therapeutic composition into the patient.
  • the pharmaceutical composition is administered as a monotherapy (i.e., as a single agent) or as a combination therapy (i.e., in combination with one or more additional anticancer agents, such as a chemotherapeutic drug, a cancer vaccine, or an immune checkpoint inhibitor.
  • the pharmaceutical composition can also be administered with radiation therapy.
  • the pharmaceutical composition is administered as a monotherapy (i.e., as a single agent) or as a combination therapy (i.e., in combination with one or more additional anticancer agents, such as a chemotherapeutic drug, a cancer vaccine, or an immune checkpoint inhibitor.
  • the pharmaceutical composition can also be administered with radiation therapy.
  • the immune checkpoint inhibitor blocks PD-1 interactions with PD-L1 and/or PD-L2.
  • the immune checkpoint inhibitor is an antibody or antigen binding fragment thereof that specifically binds PD-1, PD-L1 or PD-L2.
  • the immune checkpoint inhibitor is an anti-PD-1 antibody, such as nivolumab or pembrolizumab or an antigen binding fragment thereof. In some cases, the immune checkpoint inhibitor blocks or is an antagonist of CTLA-4, such as is an anti-CTLA-4 antibody or antigen binding fragment thereof. In some aspects of the present disclosure, the immune checkpoint inhibitor is tremelimumab or ipilimumab.
  • the pharmaceutical composition suppresses an immune response, which can be useful in the treatment of inflammatory or autoimmune disorders, or organ transplantation.
  • the pharmaceutical composition contains a variant ICOSL polypeptide in a format that exhibits antagonist activity of its cognate binding partner CD28 or ICOS and/or that blocks or inhibits costimulatory signaling via CD28 or ICOS.
  • exemplary formats of an ICOSL polypeptide for use in connection with such therapeutic applications include, for example, a variant ICOSL polypeptide that is soluble (e.g.
  • variant ICOSL-Fc fusion protein an immunomodulatory protein or “stack” of a variant ICOSL polypeptide and another IgSF domain, including soluble forms thereof that are Fc fusions, an engineered cell expressing a secretable immunomodulatory protein, or an infectious agent comprising a nucleic acid molecule encoding a secretable immunomodulatory protein, such as for expression and secretion of the secretable immunomodulatory protein in an infected cell (e.g. tumor cell or APC, e.g. dendritic cell).
  • an infected cell e.g. tumor cell or APC, e.g. dendritic cell.
  • the inflammatory or autoimmune disorder is antineutrophil cytoplasmic antibodies (ANCA)-associated vasculitis, a vasculitis, an autoimmune skin disease, transplantation, a Rheumatic disease, an inflammatory gastrointestinal disease, an inflammatory eye disease, an inflammatory neurological disease, an inflammatory pulmonary disease, an inflammatory endocrine disease, or an autoimmune hematological disease.
  • ANCA antineutrophil cytoplasmic antibodies
  • the inflammatory and autoimmune disorders that can be treated by the pharmaceutical composition described herein is Addison's Disease, allergies, alopecia areata, Alzheimer's, antineutrophil cytoplasmic antibodies (ANCA)-associated vasculitis, ankylosing spondylitis, antiphospholipid syndrome (Hughes Syndrome), asthma, atherosclerosis, rheumatoid arthritis, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune inner ear disease, autoimmune lymphoproliferative syndrome, autoimmune myocarditis, autoimmune oophoritis, autoimmune orchitis, azoospermia, Behcet's Disease, Berger's Disease, bullous pemphigoid, cardiomyopathy, cardiovascular disease, celiac Sprue/coeliac disease, chronic fatigue immune dysfunction syndrome (CFIDS), chronic idiopathic polyneuritis, chronic inflammatory demyelinating, polyradicalneuropathy (CIDP), chronic relapsing
  • the inflammatory or autoimmune disorder is a chronic autoimmune disease.
  • the inflammatory or autoimmune disorder is Sjogren's Syndrome (pSS) or Systemic Lupus Erythematosus (SLE).
  • the inflammatory or autoimmune disorder is an inflammatory bowel disease (IBD).
  • the inflammatory or autoimmune disorder is Crohn's Disease.
  • the inflammatory or autoimmune disorder is an IBD-related disease or disorder, e.g. interstitial lung disease (ILD).
  • the inflammatory or autoimmune disorder is psoriatic arthritis or rheumatoid arthritis.
  • the pharmaceutical composition is administered to modulate an autoimmune condition.
  • suppressing an immune response can be beneficial in methods for inhibiting rejection of a tissue, cell, or organ transplant from a donor by a recipient.
  • the pharmaceutical compositions described herein are used to limit or prevent graft-related or transplant related diseases or disorders, such as graft versus host disease (GvHD).
  • the pharmaceutical compositions are used to suppress autoimmune rejection of transplanted or grafted bone marrow, organs, skin, muscle, neurons, islets, or parenchymal cells.
  • a pharmaceutical composition provided herein such as a variant ICOSL IgSF (e.g. IgV) Fc fusion protein provided herein, is used to treat psoriatic arthritis (PsA).
  • PsA affects one or more joints, such as fingers, toes, arms or legs, including elbows, wrists, hands and feet, or sacroliliac joint.
  • the PsA is mild and/or affects four or less joints.
  • the PsA is moderate and/or affects four or more joints.
  • a subject with PsA may exhibit pain, stiffness or inflammation in the spine or neck, or in the one more joints.
  • a pharmaceutical composition provided herein such as a variant ICOSL IgSF (e.g. IgV) Fc fusion protein provided herein, is used to treat rheumatoid arthritis (RA).
  • RA rheumatoid arthritis
  • RA affects joints, lining of joints, and/or non-joint structures in the body (e.g., skin, eyes, lungs, heart, kidneys, salivary glands, nerve tissue, bone marrow or blood vessels).
  • RA or RA symptoms are chronic.
  • a pharmaceutical composition provided herein such as a variant ICOSL IgSF (e.g. IgV) Fc fusion protein provided herein, is used to treat GVHD.
  • the GVHD is acute GVHD (aGVHD).
  • aGVHD occurs after allogeneic hematopoietic stem cell transplant (HSCT) and/or a reaction of donor immune cells against host tissues.
  • HSCT allogeneic hematopoietic stem cell transplant
  • the aGVHD manifests in the skin, liver or gastrointestinal tract.
  • a pharmaceutical composition provided herein such as a variant ICOSL IgSF (e.g. IgV) Fc fusion protein provided herein, is used to treat an autoimmune condition associated with an organ transplant.
  • treating the autoimmune condition associated with an organ transplant may prolong the survival of the host and transplanted organ.
  • treating the autoimmune condition associated with an organ transplant includes prophylaxis of or inhibiting or preventing transplant rejections by a subject that is the recipient of the organ transplant.
  • a pharmaceutical composition provided herein such as a variant ICOSL IgSF (e.g. IgV) Fc fusion protein provided herein, is used to treat an inflammatory bowel disease (IBD).
  • a pharmaceutical composition provided herein such as a variant ICOSL IgSF (e.g. IgV) Fc fusion protein provided herein, is used to treat Crohn's disease.
  • the Crohn's disease can include a subtype from Crohn's colitis, Crohn's enteritis, Crohn's iletis or Crohn's enterocolitis.
  • a pharmaceutical composition provided herein such as a variant ICOSL IgSF (e.g. IgV) Fc fusion protein provided herein, is used to treat systemic lupus erythematosus (SLE).
  • a pharmaceutical composition provided herein such as a variant ICOSL IgSF (e.g. IgV) Fc fusion protein provided herein, is used to treat Sjogren's Syndrome.
  • a variant ICOS Ligand (ICOSL) polypeptide comprising one or more amino acid modifications in an immunoglobulin superfamily (IgSF) domain of an ICOSL reference polypeptide, wherein the ICOSL reference polypeptide is a truncated extracellular domain comprising a contiguous sequence of amino acids comprising amino acids 1-112 and a C-terminal truncation of at least 25 amino acids with reference to the ICOSL extracellular domain sequence set forth in SEQ ID NO: 32.
  • IgSF immunoglobulin superfamily
  • variant ICOSL polypeptide of embodiment 1 wherein the variant ICOSL polypeptide exhibits altered binding to the ectodomain(s) of ICOS or CD28 compared to the binding of the ICOSL reference polypeptide for the same ectodomain(s).
  • variant ICOSL polypeptide of embodiment 1 or embodiment 2 wherein the variant ICOSL polypeptide exhibits increased binding to the ectodomain(s) of ICOS or CD28 compared to the binding of the ICOSL reference polypeptide for the same ectodomain(s).
  • a variant ICOSL Ligand (ICOSL) polypeptide comprising one or more amino acid modifications in an ICOSL reference polypeptide, wherein the ICOSL reference polypeptide consists of the sequence of amino acids set forth in SEQ ID NO: 545.
  • a variant ICOS Ligand (ICOSL) polypeptide comprising one or more amino acid modifications in an immunoglobulin superfamily (IgSF) domain of an ICOSL reference polypeptide, wherein the ICOSL reference polypeptide is altered in one or more amino acids corresponding to amino acids 204-209 with reference to SEQ ID NO: 32.
  • IgSF immunoglobulin superfamily
  • variant ICOSL polypeptide of embodiment 8 or embodiment 9 wherein the variant ICOSL polypeptide exhibits altered binding to one or more of its binding partner(s) compared to the binding of the ICOSL reference polypeptide for the one or more binding partner(s).
  • variant ICOSL polypeptide of any of embodiments 5-7 and 9-18 wherein the variant ICOSL polypeptide exhibits reduced proteolytic cleavage when expressed from a cell, optionally compared to a full-length extracellular domain of the variant ICOSL polypeptide when expressed from the same cell.
  • variant ICOSL polypeptide of embodiment 19 or embodiment 20, wherein the cell is a Chinese Hamster Ovary (CHO) cell line or a derivative thereof.
  • variant ICOSL polypeptide of any of embodiments 1-25 wherein the one or more amino acid modifications are selected from N52A, N52C, N52D, N52G, N52H, N52K, N52L, N52M, N52Q, N52R, N52S, N52T, N52V, N52Y, N52K, S54A, S54P, N57A, N57D, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57Y, N57W, Q100A, Q100D, Q100G, Q100K, Q100L, Q100M, Q100N, Q100P, Q100R, Q100S, Q100T or Q100V.
  • the one or more amino acid modifications are selected from N52A, N52C, N52D, N52G, N52H, N52K, N52L, N52M, N52Q, N52R,
  • variant ICOSL polypeptide of any of embodiments 1-29 wherein the one or more amino acid modifications are selected from among N52S, N52H, N52D, N52H/N57Y/Q100P, N52S/Y146C/Y152C, N52H/C198R, N52H/C198R/T225A, N52H/K92R, N57Y, N52S/C198R, N52S/T113E, S54A, N52D/S54P, N52K/L208P, N52H/I143T, N52S/R75Q/L203P, N52S/D158G, N52D/Q133H, N52H/N57Y/Q100R/V122A, N52H/N57Y/Q100R/F172S, N52H/N57Y/Q100R, N52S/N194D, N52H/N57Y/Q100R/L102R/V110D/H115R/C198R, N52S
  • variant ICOSL polypeptide of any of embodiments 1-30 wherein the variant ICOSL polypeptide exhibits increased binding to the ectodomain of ICOS or CD28 compared to the binding of the reference ICOSL polypeptide to the same ectodomain.
  • a variant ICOS Ligand (ICOSL) polypeptide comprising an IgV domain or specific binding fragment thereof, an IgC domain or a specific binding fragment thereof, or both, wherein the variant ICOSL polypeptide comprises one or more amino acid modifications in an ICOSL reference polypeptide or a specific binding fragment thereof corresponding to amino acid modifications are selected from N52A, N52C, N52D, N52G, N52K, N52L, N52M, N52R, N52T, N52V, N57A, N57E, N57F, N57H, N57K, N57L, N57M, N57P, N57Q, N57S, N57T, N57V, N57W, Q100A, Q100D, Q100G, Q100L, Q100M, Q100N, Q100R, Q100S, Q100T or Q100V. with reference to SEQ ID NO:32.
  • variant ICOSL polypeptide of embodiment 37 wherein the one or more amino acid modifications are selected from among N52A/N57F/Q100S, N52A, /N57H/Q100S, N52A/N57Y/Q100A, N52D/N57A/Q100A, N52D/Q100S, N52G/Q100A, N52H/Q100A, N52M/N57H/Q100S, N52M/N57W/Q100P, N52Q/N57F, N52Q/N57S/Q100A, N52R/N57L/Q100A, N52R/N57Y/Q100P, N52R/N57Y/Q100S, N52S/N57A/Q100A, N52S/N57H/Q100E, N52S/N57L/Q100S, N52S/N57M/Q100S, N52S/N57Y/Q100S, N52S/N57Y/Q100M, N52S/N
  • variant ICOSL polypeptide of any one of embodiments 37-40 wherein the ICOSL reference polypeptide comprises (i) the sequence of amino acids set forth in SEQ ID NO:32, (ii) a sequence of amino acids that has at least 95% sequence identity to SEQ ID NO:32; or (iii) a portion of (i) or (ii) comprising an IgV domain or IgC domain or specific binding fragments thereof or both.
  • the specific binding fragment of the IgV domain or IgC domain has a length of at least 50, 60, 70, 80, 90, 100, 110 or more amino acids; or
  • the specific binding fragment of the IgV domain comprises a length that is at least 80% of the length of the IgV domain set for as amino acids 19-129 of SEQ ID NO:5 and/or the specific binding fragment of the IgC domain comprises a length that is at least 80% of the length of the IgC domain set forth as amino acids 141-227 of SEQ ID NO:5.
  • variant ICOSL polypeptide of any of embodiments 37-42 wherein the variant ICOSL polypeptide comprises the IgV domain or a specific fragment thereof and the IgC domain or a specific fragment thereof.
  • variant ICOSL polypeptide of any of embodiments 37-43 wherein the variant ICOSL polypeptide comprises the sequence of amino acids set forth in any one of SEQ ID NOS: 638-685, 905, 908, or a sequence of amino acids that exhibits at least 90%, 91%%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOS: 638-685, 905, 908.
  • variant ICOSL polypeptide of any of embodiments 37-43 wherein the variant ICOSL polypeptide consists of the sequence of amino acids set forth in any one of SEQ ID NOS: 638-685, 905, 908, or a sequence of amino acids that exhibits at least 90%, 91%%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOS: 638-685, 905, 908.
  • variant ICOSL polypeptide of any of embodiments 37-43 and 46 wherein the variant ICOSL polypeptide comprises the sequence of amino acids set forth in any one of SEQ ID NOS: 686-781, 907, 910, or a sequence of amino acids that exhibits at least 90%, 91%%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOS: 686-781, 907, 910.
  • variant ICOSL polypeptide of any of embodiments 37-43 and 46 wherein the variant ICOSL polypeptide consists of the sequence of amino acids set forth in any one of SEQ ID NOS: 686-781, 907, 910, or a sequence of amino acids that exhibits at least 90%, 91%%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOS: 686-781, 907, 910.
  • variant ICOSL polypeptide of any of embodiments 37-51 wherein the variant ICOSL polypeptide exhibits increased binding to the ectodomain(s) of ICOS or CD28 compared to the binding of the ICOSL reference polypeptide for the same ectodomain(s).
  • variant ICOSL polypeptide of any of embodiments 1-55 wherein the variant ICOSL polypeptide exhibits decreased binding to the ectodomain of CTLA-4 compared to the binding of the reference ICOSL polypeptide for the same ectodomain.
  • variant ICOSL polypeptide of embodiment 56 wherein the binding is decreased more than 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold or 60-fold.
  • variant ICOSL polypeptide of any of embodiments 1-59 wherein the variant ICOSL polypeptide comprises up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 amino acid modifications, optionally amino acid substitutions, insertions and/or deletions.
  • variant ICOSL polypeptide of any of embodiments 1-60 wherein the variant ICOSL polypeptide exhibits at least or at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to the ICOSL reference polypeptide.
  • variant ICOSL polypeptide of any of embodiments 1-61 that is a soluble protein.
  • the variant ICOSL polypeptide lacks a transmembrane domain and intracellular signaling domain
  • the variant ICOSL polypeptide when expressed from a cell, the variant ICOSL polypeptide is not expressed on the surface of the cell.
  • variant ICOSL polypeptide of embodiment 64 wherein the transmembrane domain comprises the sequence of amino acids set forth as residues 257-277 of SEQ ID NO:5 or a functional variant thereof that exhibits at least 85% sequence identity to residues 257-277 of SEQ ID NO:5.
  • variant ICOSL polypeptide of embodiment 64 or embodiment 65 further comprising a cytoplasmic signaling domain linked to the transmembrane domain.
  • variant ICOSL polypeptide of embodiment 66, wherein the cytoplasmic signaling domain comprises the sequence of amino acids set forth as residues 278-302 of SEQ ID NO:5 or a functional variant thereof that exhibits at least 85% sequence identity to residues 278-302 of SEQ ID NO:5.
  • variant ICOSL polypeptide of any of embodiments 1-67 that is deglycosylated or partially deglycosylated compared to the ICOSL reference sequence.
  • An immunomodulatory protein comprising the variant ICOSL polypeptide of any of embodiments 1-68 and a half-life extending moiety.
  • the half-life extending moiety comprises a multimerization domain, albumin, an albumin-binding polypeptide, Pro/Ala/Ser (PAS), a C-terminal peptide (CTP) of the beta subunit of human chorionic gonadotropin, polyethylene glycol (PEG), long unstructured hydrophilic sequences of amino acids (XTEN), hydroxyethyl starch (HES), an albumin-binding small molecule, or a combination thereof.
  • PAS Pro/Ala/Ser
  • CTP C-terminal peptide
  • PEG polyethylene glycol
  • XTEN long unstructured hydrophilic sequences of amino acids
  • HES hydroxyethyl starch
  • albumin-binding small molecule or a combination thereof.
  • PAS Pro/Ala/Ser
  • immunomodulatory protein of embodiment 71, wherein the half-life extending moiety comprises the sequence set forth in SEQ ID NO: 904.
  • the immunomodulatory protein of embodiment 73, wherein the multimerization domain is selected from an Fc region of an immunoglobulin, a leucine zipper, an isoleucine zipper or a zinc finger.
  • immunomodulatory protein of any of embodiments 73-75 wherein the immunomodulatory protein is a multimer comprising a first variant ICOSL polypeptide linked to a first multimerization domain and a second variant ICOSL polypeptide linked to a second multimerization domain, wherein the first and second multimerization domains interact to form a multimer comprising the first and second variant ICOSL polypeptide.
US16/757,344 2017-10-18 2018-10-17 Variant icos ligand immunomodulatory proteins and related compositions and methods Abandoned US20200283500A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/757,344 US20200283500A1 (en) 2017-10-18 2018-10-17 Variant icos ligand immunomodulatory proteins and related compositions and methods

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762574161P 2017-10-18 2017-10-18
PCT/US2018/056381 WO2019079520A2 (en) 2017-10-18 2018-10-17 ICOS VARIANT LIGAND IMMUNOMODULATORY IMMUNOMODULATORY PROTEINS, COMPOSITIONS AND METHODS THEREOF
US16/757,344 US20200283500A1 (en) 2017-10-18 2018-10-17 Variant icos ligand immunomodulatory proteins and related compositions and methods

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/056381 A-371-Of-International WO2019079520A2 (en) 2017-10-18 2018-10-17 ICOS VARIANT LIGAND IMMUNOMODULATORY IMMUNOMODULATORY PROTEINS, COMPOSITIONS AND METHODS THEREOF

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/560,209 Continuation US11613566B2 (en) 2017-10-18 2021-12-22 Variant ICOS ligand immunomodulatory proteins and related compositions and methods

Publications (1)

Publication Number Publication Date
US20200283500A1 true US20200283500A1 (en) 2020-09-10

Family

ID=64316982

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/757,344 Abandoned US20200283500A1 (en) 2017-10-18 2018-10-17 Variant icos ligand immunomodulatory proteins and related compositions and methods
US17/560,209 Active US11613566B2 (en) 2017-10-18 2021-12-22 Variant ICOS ligand immunomodulatory proteins and related compositions and methods
US18/172,258 Pending US20230235012A1 (en) 2017-10-18 2023-02-21 Variant icos ligand immunomodulatory proteins and related compositions and methods

Family Applications After (2)

Application Number Title Priority Date Filing Date
US17/560,209 Active US11613566B2 (en) 2017-10-18 2021-12-22 Variant ICOS ligand immunomodulatory proteins and related compositions and methods
US18/172,258 Pending US20230235012A1 (en) 2017-10-18 2023-02-21 Variant icos ligand immunomodulatory proteins and related compositions and methods

Country Status (17)

Country Link
US (3) US20200283500A1 (zh)
EP (1) EP3697810A2 (zh)
JP (2) JP7282760B2 (zh)
KR (1) KR20200085777A (zh)
CN (1) CN111712515A (zh)
AR (1) AR113455A1 (zh)
AU (2) AU2018351000B2 (zh)
BR (1) BR112020007542A2 (zh)
CA (1) CA3078517A1 (zh)
EA (1) EA202090974A1 (zh)
IL (1) IL273726A (zh)
MA (1) MA50404A (zh)
MX (1) MX2020004540A (zh)
PH (1) PH12020550449A1 (zh)
SG (1) SG11202003078VA (zh)
TW (1) TW201925223A (zh)
WO (1) WO2019079520A2 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11319359B2 (en) 2015-04-17 2022-05-03 Alpine Immune Sciences, Inc. Immunomodulatory proteins with tunable affinities
US11753458B2 (en) 2017-10-10 2023-09-12 Alpine Immune Sciences, Inc. CTLA-4 variant immunomodulatory proteins and uses thereof

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017181148A2 (en) 2016-04-15 2017-10-19 Alpine Immune Sciences, Inc. Icos ligand variant immunomodulatory proteins and uses thereof
TW201925223A (zh) 2017-10-18 2019-07-01 美商艾爾潘免疫科學有限公司 變異型icos 配位體免疫調節蛋白及相關組合物及方法
AU2020257238A1 (en) * 2019-04-17 2021-12-02 Alpine Immune Sciences, Inc. Methods and uses of variant ICOS Ligand (ICOSL) fusion proteins
US20220267444A1 (en) * 2019-08-02 2022-08-25 CTTQ-Akeso (ShangHai) Biomed. Tech. Co., Ltd. Anti-pd-1 antibody and medical use thereof
EP4118105A2 (en) * 2020-03-09 2023-01-18 Pfizer Inc. Cd80-fc fusion protein and uses thereof
WO2021226551A1 (en) 2020-05-08 2021-11-11 Alpine Immune Sciences, Inc. April and baff inhibitory immunomodulatory proteins and methods of use thereof
CA3191448A1 (en) * 2020-08-12 2022-02-17 Migal Galilee Research Institute Ltd. Alloreactive immune cell-distancing device and uses thereof for protecting donor-derived cells from allorejection
KR20230155960A (ko) * 2022-05-03 2023-11-13 고려대학교 산학협력단 Icos와의 결합력이 향상된 icos-l 변이체

Family Cites Families (191)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7045313B1 (en) 1982-11-30 2006-05-16 The United States Of America As Represented By The Department Of Health And Human Services Recombinant vaccinia virus containing a chimeric gene having foreign DNA flanked by vaccinia regulatory DNA
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
DE3883899T3 (de) 1987-03-18 1999-04-22 Sb2 Inc Geänderte antikörper.
US5443964A (en) 1987-08-10 1995-08-22 Duke University Poxvirus insertion/expression vector
US6218525B1 (en) 1988-02-25 2001-04-17 The General Hospital Corporation Nucleic acid encoding CD28
US5716826A (en) 1988-03-21 1998-02-10 Chiron Viagene, Inc. Recombinant retroviruses
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US6410010B1 (en) 1992-10-13 2002-06-25 Board Of Regents, The University Of Texas System Recombinant P53 adenovirus compositions
JP3399943B2 (ja) 1991-05-06 2003-04-28 アメリカ合衆国 癌胎児性抗原を発現する組換えウイルスとその使用方法
LU91067I2 (fr) 1991-06-14 2004-04-02 Genentech Inc Trastuzumab et ses variantes et dérivés immuno chimiques y compris les immotoxines
US6887471B1 (en) 1991-06-27 2005-05-03 Bristol-Myers Squibb Company Method to inhibit T cell interactions with soluble B7
US5262522A (en) 1991-11-22 1993-11-16 Immunex Corporation Receptor for oncostatin M and leukemia inhibitory factor
EP1005870B1 (en) 1992-11-13 2009-01-21 Biogen Idec Inc. Therapeutic application of chimeric antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5773253A (en) 1993-01-22 1998-06-30 Bristol-Myers Squibb Company MYPPPY variants of CTL A4 and uses thereof
EP0714409A1 (en) 1993-06-16 1996-06-05 Celltech Therapeutics Limited Antibodies
US5457035A (en) 1993-07-23 1995-10-10 Immunex Corporation Cytokine which is a ligand for OX40
US20020159979A1 (en) 1994-06-06 2002-10-31 Children's Hospital, Inc. Adeno-associated virus materials and methods
DE69534702T2 (de) 1994-09-23 2006-08-24 The University Of British Columbia, Vancouver Verfahren zur erhöhung der expression von endogenen peptiden tragenden mhc klasse i molekulen
ES2154738T3 (es) 1994-10-03 2001-04-16 Us Gov Health & Human Serv Composicion que comprende un virus recombinante que expresa un antigeno y un virus recombinante que expresa una molecula inmunoestimuladora.
US5998205A (en) 1994-11-28 1999-12-07 Genetic Therapy, Inc. Vectors for tissue-specific replication
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US7153510B1 (en) 1995-05-04 2006-12-26 Yale University Recombinant vesiculoviruses and their uses
US5767071A (en) 1995-06-07 1998-06-16 Ixsys Incorporated Sevenmer cyclic peptide inhibitors of diseases involving αv β3
US5780426A (en) 1995-06-07 1998-07-14 Ixsys, Incorporated Fivemer cyclic peptide inhibitors of diseases involving αv β3
FR2735789B1 (fr) 1995-06-23 1997-07-25 Centre Nat Rech Scient Adenovirus recombinants, leur utilisation pour preparer des aav, lignee cellulaire complementaire et compositions pharmaceutiques les contenant
CA2230758A1 (en) 1995-09-08 1997-03-13 Genzyme Corporation Improved aav vectors for gene therapy
US7001765B2 (en) 1996-03-06 2006-02-21 Medigene Ag Adeno-associated virus vector for boosting immunogenicity of cells
US6294170B1 (en) 1997-08-08 2001-09-25 Amgen Inc. Composition and method for treating inflammatory diseases
EP0979281B1 (en) 1997-05-02 2005-07-20 Genentech, Inc. A method for making multispecific antibodies having heteromultimeric and common components
US5891432A (en) 1997-07-29 1999-04-06 The Immune Response Corporation Membrane-bound cytokine compositions comprising GM=CSF and methods of modulating an immune response using same
EP1012236A1 (en) 1997-08-15 2000-06-28 Rubicon Laboratory Inc. Retrovirus and viral vectors
CA2304168A1 (en) 1997-09-19 1999-04-01 The Trustees Of The University Of Pennsylvania Methods and cell line useful for production of recombinant adeno-associated viruses
GB9801930D0 (en) 1998-01-29 1998-03-25 Univ London Mutant herpes simplex viruses and uses thereof
JP2002510481A (ja) 1998-04-02 2002-04-09 ジェネンテック・インコーポレーテッド 抗体変異体及びその断片
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US20040063094A1 (en) 1998-05-20 2004-04-01 Biovex Limited Mutant herpes simplex viruses and uses thereof
US6436392B1 (en) 1998-05-20 2002-08-20 University Of Iowa Research Foundation Adeno-associated virus vectors
GB2337755B (en) 1998-05-29 2003-10-29 Secr Defence Virus vaccine
JP2002522451A (ja) 1998-08-07 2002-07-23 ユニバーシティ オブ ワシントン 免疫学的単純ヘルペスウイルス抗原とその使用法
DE69941905D1 (de) 1998-11-10 2010-02-25 Univ North Carolina Virusvektoren und verfahren für ihre herstellung und verabreichung.
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
PL209786B1 (pl) 1999-01-15 2011-10-31 Genentech Inc Przeciwciało zawierające wariant regionu Fc ludzkiej IgG1, przeciwciało wiążące czynnik wzrostu śródbłonka naczyń oraz immunoadhezyna
US8624010B1 (en) 1999-02-03 2014-01-07 Steven K. Yoshinaga Nucleic acids encoding B7RP1
GB9904582D0 (en) 1999-02-26 1999-04-21 Nycomed Imaging As Process
US6428968B1 (en) 1999-03-15 2002-08-06 The Trustees Of The University Of Pennsylvania Combined therapy with a chemotherapeutic agent and an oncolytic virus for killing tumor cells in a subject
DE19933288A1 (de) 1999-07-15 2001-01-18 Medigene Ag Strukturprotein von Adeno-assoziiertem Virus mit veränderter Antigenität, seine Herstellung und Verwendung
US6365619B1 (en) 1999-07-22 2002-04-02 Novartis Ag Treatment of arteriosclerosis
JP4827353B2 (ja) 1999-08-09 2011-11-30 ターゲティッド ジェネティクス コーポレイション 鎖内塩基対を形成するような配列の設計による、組換えウイルスベクターからの一本鎖の異種ヌクレオチド配列の発現の増大
US6521749B1 (en) 1999-09-21 2003-02-18 Genetics Institute, Inc. GL50 nucleic acids and uses therefor
US7241447B1 (en) 1999-10-07 2007-07-10 University Of Iowa Research Foundation Adeno-associated virus vectors and uses thereof
US7329728B1 (en) 1999-10-25 2008-02-12 The Scripps Research Institute Ligand activated transcriptional regulator proteins
US6723316B2 (en) 1999-12-22 2004-04-20 Onyx Pharmaceuticals, Inc. Herpes simplex virus-1 Glycoprotein C mutants for treating unwanted hyperproliferative cell growth
CN1250732C (zh) 2000-01-21 2006-04-12 拜奥维克斯有限公司 病毒株
US7306902B2 (en) 2002-06-28 2007-12-11 Oncolyties Biotech Inc. Oncolytic viruses as phenotyping agents for neoplasms
US7094874B2 (en) 2000-05-26 2006-08-22 Bristol-Myers Squibb Co. Soluble CTLA4 mutant molecules
JP2004513878A (ja) 2000-06-23 2004-05-13 マキシジェン, インコーポレイテッド 新規同時刺激分子
US7183376B2 (en) 2000-06-23 2007-02-27 Maxygen, Inc. Variant B7 co-stimulatory molecules
EP1372696B1 (en) 2000-07-03 2008-07-23 Bristol-Myers Squibb Company Methods for treating rheumatic diseases using a soluble ctla4 molecule
US20040022787A1 (en) 2000-07-03 2004-02-05 Robert Cohen Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
US7011972B2 (en) 2000-07-18 2006-03-14 The Scripps Research Institute Fusion polypeptide comprising two ligand binding domains
US6635750B1 (en) 2000-07-20 2003-10-21 Millennium Pharmaceuticals, Inc. B7-H2 nucleic acids, members of the B7 family
US6653103B2 (en) 2001-03-30 2003-11-25 Wisconsin Alumni Research Foundation Inhibition of nucleocytoplasmic transport by vesicular stomatitis virus M protein-like polypeptides
DE60239394D1 (de) 2001-05-11 2011-04-21 Wellstat Biologics Corp Onkolytische virustherapie
EP1497426A2 (en) 2001-06-22 2005-01-19 Maxygen, Inc. Co-stimulatory molecules
EP1411880B1 (en) 2001-07-11 2018-04-25 University of Miami Recombinant vsv for the treatment of tumor cells
CN1304559C (zh) 2001-10-09 2007-03-14 杭州康科生物技术有限公司 表达热休克蛋白的溶瘤微生物及其应用
US7247615B2 (en) 2001-11-30 2007-07-24 United States Of America, Represented By The Secretary, Department Of Health And Human Services Peptide agonists of prostate-specific antigen and uses therefor
US7235631B2 (en) 2002-02-07 2007-06-26 Mayo Foundation For Medical Education And Research ICOS mutants
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
CA2479763A1 (en) 2002-03-27 2003-10-09 Baylor College Of Medicine Potent oncolytic herpes simplex virus for cancer therapy
US20030225260A1 (en) 2002-04-30 2003-12-04 Snyder Richard O. Production of recombinant AAV virions
MXPA04012881A (es) 2002-06-21 2005-07-05 Wellstat Biologics Corp Administracion de virus terapeuticos.
EP1391213A1 (en) 2002-08-21 2004-02-25 Boehringer Ingelheim International GmbH Compositions and methods for treating cancer using maytansinoid CD44 antibody immunoconjugates and chemotherapeutic agents
CN1820078A (zh) 2002-09-09 2006-08-16 田纳西大学研究基金会 弹状病毒的重组突变体及其应用方法
US7361740B2 (en) 2002-10-15 2008-04-22 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
CN103833854B (zh) 2002-12-16 2017-12-12 健泰科生物技术公司 免疫球蛋白变体及其用途
EP1606411B1 (en) 2003-03-27 2008-12-10 Ottawa Health Research Institute Mutant vesicular stomatitis viruses and use thereof
US7731974B2 (en) 2003-03-27 2010-06-08 Ottawa Hospital Research Institute Mutant vesicular stomatitis viruses and uses thereof
ATE545699T1 (de) 2003-06-18 2012-03-15 Genelux Corp Modifizierte, rekombinante vacciniaviren und deren verwendungen
PT3211085T (pt) 2003-09-30 2021-06-17 Univ Pennsylvania Clados de vírus adeno-associados (aav), sequências, vetores que as contêm, e suas utilizações
US20070003520A1 (en) 2003-11-17 2007-01-04 Brown Susanne M Mutant viruses
US7897146B2 (en) 2003-11-17 2011-03-01 Crusade Laboratories Limited Treatment using herpes simplex virus
GB0326798D0 (en) 2003-11-17 2003-12-24 Crusade Lab Ltd Methods for generating mutant virus
NZ547438A (en) 2003-12-19 2010-01-29 Genentech Inc Monovalent antibody fragments useful as therapeutics
CA2885854C (en) 2004-04-13 2017-02-21 F. Hoffmann-La Roche Ag Anti-p-selectin antibodies
WO2005116224A2 (en) 2004-05-18 2005-12-08 Children's Memorial Hospital Tetracycline-regulated adeno-associated viral (aav) vectors for gene delivery to the nervous system
US7427396B2 (en) 2004-06-03 2008-09-23 Genzyme Corporation AAV vectors for gene delivery to the lung
US7731952B2 (en) 2004-06-24 2010-06-08 New York University Avirulent oncolytic herpes simplex virus strains engineered to counter the innate host response
EP3342782B1 (en) 2004-07-15 2022-08-17 Xencor, Inc. Optimized fc variants
TWI309240B (en) 2004-09-17 2009-05-01 Hoffmann La Roche Anti-ox40l antibodies
US8911726B2 (en) 2004-09-22 2014-12-16 Kyowa Hakko Kirin Co., Ltd Stabilized human Igg4 antibodies
US20100111856A1 (en) 2004-09-23 2010-05-06 Herman Gill Zirconium-radiolabeled, cysteine engineered antibody conjugates
US20090208924A1 (en) 2004-12-01 2009-08-20 Bayer Schering Pharma Aktiengesellschaft Generation of Replication Competent Viruses for Therapeutic Use
EP3050963B1 (en) 2005-03-31 2019-09-18 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
AU2006252406B2 (en) 2005-06-01 2012-05-17 California Institute Of Technology Method of targeted gene delivery using viral vectors
AU2006262021C1 (en) 2005-06-23 2012-05-10 The University Of Houston Use of mutant Herpes Simplex Virus-2 for cancer therapy
JP5252635B2 (ja) 2005-07-01 2013-07-31 メダレックス インコーポレーティッド プログラム死リガンド1(pd−l1)に対するヒトモノクローナル抗体
US7943374B2 (en) 2005-08-21 2011-05-17 Markus Hildinger Super-size adeno-associated viral vector harboring a recombinant genome larger than 5.7 kb
GB0522476D0 (en) 2005-11-03 2005-12-14 Biovex Ltd Oncolytic herpes virus vectors
ES2439641T3 (es) 2005-12-20 2014-01-24 Bristol-Myers Squibb Company Composiciones y procedimientos de producción de una composición
BRPI0714495B8 (pt) 2006-07-21 2021-05-25 California Inst Of Techn lentivírus deficiente para replicação recombinante pseudotipado
EP2097517B1 (en) 2006-10-16 2014-06-04 Genelux Corporation Recombinant Lister strain vaccinia virus encoding an anti-VEGF single chain antibody
WO2008140621A2 (en) 2006-12-21 2008-11-20 Mount Sinai School Of Medicine Of New York University Transgenic oncolytic viruses and uses thereof
WO2008092117A2 (en) 2007-01-25 2008-07-31 Xencor, Inc. Immunoglobulins with modifications in the fcr binding region
US8163292B2 (en) 2007-02-16 2012-04-24 Virttu Biologics Limited Herpes simplex viruses and methods of viral replication
HUE038506T2 (hu) 2007-03-30 2018-10-29 Memorial Sloan Kettering Cancer Center Kostimuláló ligand konstitutív expressziója adoptív módon átvitt T-limfocitákon
CA2690627C (en) 2007-06-15 2014-12-02 Genelux Corporation Microorganisms for imaging and/or treatment of tumors
DK2173890T3 (da) 2007-06-21 2011-06-27 Univ Muenchen Tech Biologisk aktive proteiner med forhøjet stabilitet in vivo og/eller in vitro
AU2008293885A1 (en) 2007-07-13 2009-03-05 The John Hopkins University B7-DC variants
EP2202297B1 (en) 2007-07-18 2014-05-14 Genelux Corporation Use of a chemotherapeutic agent in the preparation of a medicament for treating or ameliorating an adverse side effect associated with oncolytic viral therapy
TWI464262B (zh) 2007-09-26 2014-12-11 中外製藥股份有限公司 抗體固定區的變異
EP2212696B1 (en) 2007-10-25 2013-12-04 Genelux Corporation Systems and methods for viral therapy
PT2222697E (pt) 2007-11-01 2013-02-15 Perseid Therapeutics Llc Polipeptídeos imunossupressores e ácidos nucleicos
CN101925612A (zh) 2007-11-27 2010-12-22 维文蒂阿生物技术公司 针对癌相关的nfkbib变体的表位的抗体及其用途
TR201802323T4 (tr) 2007-12-11 2018-03-21 Univ North Carolina Chapel Hill Polipürin yolu modifiye edilmiş retroviral vektörler.
US8313896B2 (en) 2008-04-04 2012-11-20 The General Hospital Corporation Oncolytic herpes simplex virus immunotherapy in the treatment of brain cancer
EP2300023A2 (en) 2008-05-16 2011-03-30 Genelux Corporation Microorganisms for preventing and treating neoplasms accompanying cellular therapy
WO2009148488A2 (en) 2008-05-29 2009-12-10 The General Hospital Corporation Use of oncolytic herpes viruses for killing cancer stem cells
EP3936122A1 (en) 2008-11-24 2022-01-12 Massachusetts Institute Of Technology Methods and compositions for localized agent delivery
LT4209510T (lt) 2008-12-09 2024-03-12 F. Hoffmann-La Roche Ag Anti-pd-l1 antikūnai ir jų panaudojimas t ląstelių funkcijos pagerinimui
US8481297B2 (en) 2009-01-08 2013-07-09 Yale University Compositions and methods of use of an oncolytic vesicular stomatitis virus
EP2283810A1 (en) 2009-07-16 2011-02-16 University College Cork-National University of Ireland, Cork Orally administered bacteria as vehicles for systemic delivery of agents
US20150359909A1 (en) 2009-07-16 2015-12-17 University College Cork-National University Of Ireland, Cork Orally administered bacteria as vehicles for systemic delivery of agents
PL2456786T5 (pl) 2009-07-24 2017-10-31 Immune Design Corp Wektory lentiwirusowe pseudotypowane glikoproteiną otoczki wirusa sindbis
CA2769822C (en) * 2009-08-13 2019-02-19 The Johns Hopkins University Methods of modulating immune function
JP5960597B2 (ja) 2009-09-30 2016-08-02 メモリアル スローン−ケタリング キャンサー センター 癌治療のための併用免疫療法
PT2504364T (pt) 2009-11-24 2017-11-14 Medimmune Ltd Agentes de ligação direcionados contra b7-h1
WO2011097477A1 (en) 2010-02-04 2011-08-11 The Trustees Of The University Of Pennsylvania Icos critically regulates the expansion and function of inflammatory human th17 cells
CN113372451A (zh) 2010-02-19 2021-09-10 Xencor公司 新颖ctla4-ig免疫粘附素
WO2011113019A2 (en) 2010-03-12 2011-09-15 Abbott Biotherapeutics Corp. Ctla4 proteins and their uses
TWI586806B (zh) 2010-04-23 2017-06-11 建南德克公司 異多聚體蛋白質之製造
JP5947311B2 (ja) 2010-12-09 2016-07-06 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア 癌を治療するためのキメラ抗原受容体改変t細胞の使用
US9402865B2 (en) 2011-01-18 2016-08-02 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
CA2830254C (en) 2011-03-16 2019-09-10 Amgen Inc. Fc variants
CA2832307A1 (en) 2011-04-08 2012-10-18 Immune Design Corp. Immunogenic compositions and methods of using the compositions for inducing humoral and cellular immune responses
WO2012149364A1 (en) 2011-04-28 2012-11-01 Diamond Don J Tumor associated vaccines and compositions for disrupting tumor-derived immunosuppression for use in combination cancer immunotherapy
TWI496886B (zh) 2011-06-24 2015-08-21 Taipei Veteran General Hospital 提升感染性與惡性疾病之治療之免疫反應
CN103796681B (zh) 2011-06-30 2018-07-20 建新公司 T细胞活化的抑制剂
US9428574B2 (en) 2011-06-30 2016-08-30 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
US10421960B2 (en) 2011-09-16 2019-09-24 The Trustees Of The University Of Pennsylvania RNA engineered T cells for the treatment of cancer
US8956619B2 (en) 2011-10-25 2015-02-17 University Of Maryland, Baltimore County Soluble CD80 as a therapeutic to reverse immune supression in cancer patients
AU2012344260B2 (en) 2011-11-28 2017-09-07 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
US10172953B2 (en) 2012-02-27 2019-01-08 Amunix Operating Inc. XTEN conjugate compositions and methods of making same
EA038702B1 (ru) 2012-03-30 2021-10-07 Иммьюн Дизайн Корп. Лентивирусные векторные частицы, имеющие улучшенную эффективность трансдукции клеток, экспрессирующих dc-sign
WO2013156054A1 (en) * 2012-04-16 2013-10-24 Universität Stuttgart The igm and ige heavy chain domain 2 as covalently linked homodimerization modules for the generation of fusion proteins with dual specificity
BR112014026718B1 (pt) 2012-05-11 2021-05-18 Medimmune Limited. polipeptídeo isolado de ctla-4, célula hospedeira, composição e uso de um polipeptídeo ctla-4
JP2015522026A (ja) 2012-06-27 2015-08-03 オルバン バイオテック エルエルシー 糖尿病を処置するためのctla4融合タンパク質
US10117896B2 (en) 2012-10-05 2018-11-06 The Trustees Of The University Of Pennsylvania Use of a trans-signaling approach in chimeric antigen receptors
CN104968364A (zh) 2012-12-03 2015-10-07 百时美施贵宝公司 强化免疫调变性Fc融合蛋白的抗癌活性
CA2892831A1 (en) 2012-12-04 2014-06-12 Oncomed Pharmaceuticals, Inc. Immunotherapy with binding agents
WO2014138188A1 (en) 2013-03-07 2014-09-12 The General Hospital Corporation Human ctla4 mutants and use thereof
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
CA2903546A1 (en) 2013-03-15 2014-09-25 Biogen Ma Inc. Treatment and prevention of acute kidney injury using anti-alpha v beta 5 antibodies
WO2014198002A1 (en) 2013-06-14 2014-12-18 Ottawa Hospital Research Institute A bacterium producing an interferon binding protein and uses thereof
GB201311475D0 (en) 2013-06-27 2013-08-14 Alligator Bioscience Ab Polypeptides
WO2014207748A1 (en) 2013-06-27 2014-12-31 Alexander Biro Soluble ctla-4 molecules and derivatives thereof for treatment of minimal change disease
CA2918216C (en) 2013-07-15 2023-03-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-human papillomavirus 16 e6 t cell receptors
WO2015103438A2 (en) 2014-01-02 2015-07-09 Genelux Corporation Oncolytic virus adjunct therapy with agents that increase virus infectivity
JP6786392B2 (ja) 2014-01-15 2020-11-18 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft FcRn結合特性が改変され、プロテインA結合特性が保持されているFc領域変異体
US20160340422A1 (en) 2014-01-28 2016-11-24 Beijing Hanmi Pharmaceutical Co., Ltd. Bifunctional fusion protein, preparation method therefor, and use thereof
ES2792849T3 (es) 2014-02-10 2020-11-12 Univ Emory Expresión de polipéptido químico con receptores de linfocitos variables en células inmunes y usos para tratar el cáncer
KR20230022270A (ko) 2014-03-28 2023-02-14 젠코어 인코포레이티드 Cd38 및 cd3에 결합하는 이중특이적 항체
BR112016023450A2 (pt) 2014-04-25 2017-10-17 Bristol Myers Squibb Co uso de compostos ctla4 para alcançar remissão livre de fármacos em indivíduos com artrite reumatoide precoce
EP3169352A1 (en) 2014-07-15 2017-05-24 Immune Design Corp. Prime-boost regimens with a tlr4 agonist adjuvant and a lentiviral vector
HUE045108T2 (hu) 2014-07-16 2019-12-30 Transgene Sa Onkolitikus vírus immunellenõrzõpont-modulátorok expresszálására
EP3177640B1 (en) 2014-08-08 2020-05-06 The Board of Trustees of the Leland Stanford Junior University High affinity pd-1 agents and methods of use
KR20170048396A (ko) 2014-09-03 2017-05-08 버베리안 노딕 에이/에스 재조합 변형된 백시니아 바이러스 앙카라 (mva) 필로바이러스 백신
US20160145344A1 (en) 2014-10-20 2016-05-26 University Of Southern California Murine and human innate lymphoid cells and lung inflammation
KR20180004094A (ko) 2014-11-06 2018-01-10 칠드런스 내셔널 메디컬 센터 암 및 자가면역 질환을 위한 면역 요법
EP3020816A1 (en) 2014-11-11 2016-05-18 University College Cork Bacterial mediated gene therapy
WO2016118577A1 (en) 2015-01-22 2016-07-28 Medimmune, Llc Thymosin-beta-four fusion proteins
US10953045B2 (en) 2015-04-02 2021-03-23 Cancure Limited Agents and compositions for eliciting an immune response
WO2016164428A1 (en) 2015-04-06 2016-10-13 The Board Of Trustees Of The Leland Stanford Junior University Receptor-based antagonists of the programmed cell death 1 (pd-1) pathway
PT3283508T (pt) * 2015-04-17 2021-06-21 Alpine Immune Sciences Inc Proteínas imuno modulatórias com afinidades afináveis
TWI715587B (zh) 2015-05-28 2021-01-11 美商安可美德藥物股份有限公司 Tigit結合劑和彼之用途
GEP20227419B (en) 2015-07-30 2022-10-10 Macrogenics Inc Pd-1-binding molecules and methods of use thereof
AU2016303497A1 (en) 2015-07-31 2018-03-01 Tarveda Therapeutics, Inc. Compositions and methods for immuno-oncology therapies
AR105654A1 (es) 2015-08-24 2017-10-25 Lilly Co Eli Anticuerpos pd-l1 (ligando 1 de muerte celular programada)
CN108513576A (zh) 2015-09-14 2018-09-07 高山免疫科学股份有限公司 可调变体免疫球蛋白超家族结构域和工程改造的细胞治疗
RU2750675C1 (ru) 2015-10-02 2021-07-01 Симфоген А/С Антитела против pd-1 и композиции
JP2019521643A (ja) 2016-04-15 2019-08-08 アルパイン イミューン サイエンシズ インコーポレイテッド Cd80バリアント免疫調節タンパク質およびその使用
WO2017181148A2 (en) 2016-04-15 2017-10-19 Alpine Immune Sciences, Inc. Icos ligand variant immunomodulatory proteins and uses thereof
EP3458095A4 (en) 2016-05-18 2019-11-27 Albert Einstein College of Medicine PD-L1 POLYPEPTIDE VARIANTS, T-LYMPHOCYTE MODULATOR MULTIMERIC POLYPEPTIDES AND METHODS OF USING SAME
CN110088127A (zh) 2016-07-28 2019-08-02 高山免疫科学股份有限公司 Cd155变体免疫调节蛋白及其用途
US11834490B2 (en) 2016-07-28 2023-12-05 Alpine Immune Sciences, Inc. CD112 variant immunomodulatory proteins and uses thereof
SG11201903407XA (en) 2016-10-20 2019-05-30 Alpine Immune Sciences Inc Secretable variant immunomodulatory proteins and engineered cell therapy
TW201925223A (zh) 2017-10-18 2019-07-01 美商艾爾潘免疫科學有限公司 變異型icos 配位體免疫調節蛋白及相關組合物及方法
US20230101432A1 (en) 2018-01-03 2023-03-30 Alpine Immune Sciences, Inc. Multi-domain immunomodulatory proteins and methods of use thereof
WO2019241758A1 (en) 2018-06-15 2019-12-19 Alpine Immune Sciences, Inc. Pd-1 variant immunomodulatory proteins and uses thereof
AU2020257238A1 (en) 2019-04-17 2021-12-02 Alpine Immune Sciences, Inc. Methods and uses of variant ICOS Ligand (ICOSL) fusion proteins

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11319359B2 (en) 2015-04-17 2022-05-03 Alpine Immune Sciences, Inc. Immunomodulatory proteins with tunable affinities
US11753458B2 (en) 2017-10-10 2023-09-12 Alpine Immune Sciences, Inc. CTLA-4 variant immunomodulatory proteins and uses thereof

Also Published As

Publication number Publication date
AU2024201301A1 (en) 2024-03-21
US11613566B2 (en) 2023-03-28
JP2021500024A (ja) 2021-01-07
BR112020007542A2 (pt) 2020-12-01
CN111712515A (zh) 2020-09-25
CA3078517A1 (en) 2019-04-25
WO2019079520A3 (en) 2019-05-23
WO2019079520A2 (en) 2019-04-25
EA202090974A1 (ru) 2020-08-05
EP3697810A2 (en) 2020-08-26
MA50404A (fr) 2020-08-26
TW201925223A (zh) 2019-07-01
US20230235012A1 (en) 2023-07-27
AU2018351000A1 (en) 2020-04-30
JP2023055821A (ja) 2023-04-18
KR20200085777A (ko) 2020-07-15
IL273726A (en) 2020-05-31
SG11202003078VA (en) 2020-05-28
JP7282760B2 (ja) 2023-05-29
PH12020550449A1 (en) 2021-05-17
MX2020004540A (es) 2020-08-03
AR113455A1 (es) 2020-05-06
US20220112265A1 (en) 2022-04-14
AU2018351000B2 (en) 2023-11-30

Similar Documents

Publication Publication Date Title
US11359022B2 (en) CD80 variant immunomodulatory proteins and uses thereof
US20210188995A1 (en) Icos ligand variant immunomodulatory proteins and uses thereof
US20230381229A1 (en) Cd155 variant immunomodulatory proteins and uses thereof
US11613566B2 (en) Variant ICOS ligand immunomodulatory proteins and related compositions and methods
AU2018236224B2 (en) PD-L1 variant immunomodulatory proteins and uses thereof
US11834490B2 (en) CD112 variant immunomodulatory proteins and uses thereof
US11471488B2 (en) CD155 variant immunomodulatory proteins and uses thereof
US11732022B2 (en) PD-L2 variant immunomodulatory proteins and uses thereof
US20220372106A1 (en) Cd86 variant immunomodulatory proteins and uses thereof
EA044356B1 (ru) Вариантные иммуномодулирующие белки лиганда icos и их применение

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALPINE IMMUNE SCIENCES, INC., WASHINGTON

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:EVANS, LAWRENCE;KORNACKER, MICHAEL;SWANSON, RYAN;SIGNING DATES FROM 20190116 TO 20190325;REEL/FRAME:053031/0044

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

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION