US20230241168A1 - April and baff inhibitory immunomodulatory proteins with and without a t cell inhibitory protein and methods of use thereof - Google Patents

April and baff inhibitory immunomodulatory proteins with and without a t cell inhibitory protein and methods of use thereof Download PDF

Info

Publication number
US20230241168A1
US20230241168A1 US17/923,208 US202117923208A US2023241168A1 US 20230241168 A1 US20230241168 A1 US 20230241168A1 US 202117923208 A US202117923208 A US 202117923208A US 2023241168 A1 US2023241168 A1 US 2023241168A1
Authority
US
United States
Prior art keywords
seq
immunomodulatory protein
amino acid
set forth
bcma
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/923,208
Other languages
English (en)
Inventor
Stacey DILLON
Mark Rixon
Lawrence Evans
Daniel William DEMONTE
Joseph L. Kuijper
Stanford L. PENG
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 US17/923,208 priority Critical patent/US20230241168A1/en
Publication of US20230241168A1 publication Critical patent/US20230241168A1/en
Assigned to ALPINE IMMUNE SCIENCES, INC. reassignment ALPINE IMMUNE SCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SWANSON, Ryan, DEMONTE, Daniel William, EVANS, LAWRENCE, KUIJPER, JOSEPH L., PENG, Stanford L., DILLON, STACEY, RIXON, MARK
Pending 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
    • C07K14/70521CD28, CD152
    • 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
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • 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
    • 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
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • 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
    • A61K47/51Medicinal 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 the non-active ingredient being a modifying agent
    • A61K47/62Medicinal 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 the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6425Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a receptor, e.g. CD4, a cell surface antigen, i.e. not a peptide ligand targeting the antigen, or a cell surface determinant, i.e. a part of the surface of a cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • 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

Definitions

  • the present disclosure provides immunomodulatory proteins that exhibit neutralizing activity of BAFF and APRIL (or BAFF/APRIL heterotrimers) alone, or also coupled with inhibition of T cell costimulation.
  • the immunomodulatory proteins include variant domains of B cell maturation antigen (BCMA) alone, and multi-domain immunomodulatory proteins that inhibit B cell responses and also can inhibit T cell costimulation.
  • the present disclosure also provides nucleic acids molecules encoding the immunomodulatory proteins.
  • the immunomodulatory proteins provide therapeutic utility for a variety of immunological diseases or conditions. Compositions and methods for making and using such proteins are provided.
  • Modulation of the immune response by intervening in the processes involving interactions between soluble ligands and their receptor is of increasing medical interest.
  • biologics used to enhance or suppress immune responses have generally been limited to antibodies (e.g., anti-PD-1 antibodies) or soluble receptors against a single cell surface molecule (e.g., Fc-CTLA-4).
  • Improved therapeutic agents that can modulate the immune response, and particularly B cell immune responses and, in some cases also T cell immune responses, are needed. Provided are embodiments that meet such needs.
  • an immunomodulatory protein that contains at least one T cell inhibitory molecule (TIM) that binds to a T cell stimulatory receptor, or a ligand of a T cell stimulatory receptor; and that antagonizes activity of a T cell stimulatory receptor; and at least one B cell inhibitory molecule (BIM) that binds to a ligand of a B cell stimulatory receptor and/or antagonizes activity of a B cell stimulatory receptor.
  • TIM T cell inhibitory molecule
  • BIM B cell inhibitory molecule
  • the immunomodulatory protein contains at least one T cell inhibitory molecule (TIM) that binds to a T cell stimulatory receptor, or a ligand of a T cell stimulatory receptor; or antagonizes activity of a T cell stimulatory receptor; and at least one B cell inhibitory molecule (BIM) that binds to a ligand of a B cell stimulatory receptor and/or antagonizes activity of a B cell stimulatory receptor.
  • TIM T cell inhibitory molecule
  • BIM B cell inhibitory molecule
  • the TIM binds to a ligand of a T cell stimulatory receptor.
  • the T cell stimulatory receptor is CD28; and the ligand of the T cell stimulatory receptor is CD80 or CD86.
  • the T cell stimulatory receptor is CD28; or the ligand of the T cell stimulatory receptor is CD80 or CD86.
  • the TIM is a CTLA-4 extracellular domain or a binding portion thereof that binds to CD80 or CD86.
  • the CTLA-4 extracellular domain or binding portion thereof consists of the sequence of amino acids set forth in SEQ ID NO:1 or SEQ ID NO:2, a variant CTLA-4 sequence of amino acids that has at least 85% sequence identity to SEQ ID NO:1 or SEQ ID NO:2; or a portion thereof containing an IgV domain.
  • the CTLA-4 extracellular domain consists of the sequence of amino acids set forth in SEQ ID NO: 1.
  • the CTLA-4 extracellular domain consists of a variant CTLA-4 sequence of amino acids that has at least 85% sequence identity to SEQ ID NO:1 or a portion thereof containing an IgV domain, wherein the variant sequence comprises one or more amino acid substitutions in SEQ ID NO:1 or the portion thereof containing the IgV domain.
  • the variant CTLA-4 sequence comprises the amino acid substitution C122S.
  • the CTLA-4 extracellular domain or the binding portion thereof is set forth in SEQ ID NO: 668.
  • the variant CTLA-4 binds to the ectodomain of CD80 and CD86, optionally wherein binding affinity to one or both of CD80 and CD86 is increased compared to the sequence set forth in SEQ ID NO:1 or the portion thereof containing the IgV domain.
  • the one or more amino acid substitutions in a variant CLTA-4 polypeptide comprise amino acid substitutions selected from L12F, R16H, G29W, T53S, M56T, N58S, L63P, L98Q, or Y105L, or combinations thereof.
  • the one or more amino acid substitutions comprise G29W, L98Q and Y105L.
  • the one or more amino acid substitutions are G29W/N58S/L63P/Q82R/L98Q/Y105L.
  • the one or more amino acid substitutions are L12F/R16H/G29W/M56T/L98Q/Y105L.
  • the one or more amino acid substitutions are G29W/L98Q/Y105L.
  • a variant CTLA-4 polypeptide is one in which the variant CTLA-4 polypeptide has at least about 85%, at least about 90% or at least about 95% sequence identity to SEQ ID NO: 1, and contains the one or more amino acid substitutions as described.
  • a variant CTLA-4 polypeptide is one in which the variant CTLA-4 polypeptide has at least about 85%, at least about 90% or at least about 95% sequence identity to SEQ ID NO:2, and contains the one or more amino acid substitutions as described.
  • the CTLA-4 extracellular domain or the binding portion thereof is set forth in any one of SEQ ID NO:92, SEQ ID NO: 112, SEQ ID NO: 165 or SEQ ID NO: 186 or a portion thereof comprising the IgV domain.
  • the variant CTLA-4 consists of the sequence set forth in SEQ ID NO:92 or a portion thereof containing the IgV domain. In some of any embodiments, the variant CTLA-4 consists of the sequence set forth in SEQ ID NO: 113 or a portion thereof containing the IgV domain. In some of any embodiments, the variant CTLA-4 consists of the sequence set forth in SEQ ID NO: 165 or a portion thereof containing the IgV domain. In some of any embodiments, the variant CTLA-4 consists of the sequence set forth in SEQ ID NO: 186 or a portion thereof containing the IgV domain.
  • the ligand of a B cell stimulatory receptor is APRIL or BAFF; and the B cell stimulatory receptor is TACI, BCMA, or BAFF-receptor. In some of any embodiments, the ligand of a B cell stimulatory receptor is APRIL or BAFF; or the B cell stimulatory receptor is TACI, BCMA, or BAFF-receptor.
  • the BIM is a TACI polypeptide that consists of the TACI extracellular domain or a binding portion thereof that binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • the BIM is a TACI extracellular domain or the binding portion thereof that has an extracellular domain sequence set forth as (i) the sequence of amino acids set forth in SEQ ID NO:709, (ii) a sequence of amino acids that has at least 95% sequence identity to SEQ ID NO:709; or (iii) a portion of (i) or (ii) comprising one or both of a CRD1 domain and CRD2 domain that binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • the BIM is a TACI extracellular domain or the binding portion thereof that comprises the CRD1 domain and the CRD2 domain.
  • a BIM that is a TACI polypeptide is one in which the TACI polypeptide is a truncated wild-type TACI extracellular domain that consists of the sequence set forth in SEQ ID NO: 516.
  • a BIM that is a TACI polypeptide is a truncated wild-type TACI extracellular domain or is a variant thereof, wherein the truncated wild-type TACI extracellular domain contains the cysteine rich domain 2 (CRD2) but lacks the entirety of the cysteine rich domain 1 (CRD1), and/or wherein the variant TACI polypeptide comprises one or more amino acid substitutions in the truncated wild-type TACI extracellular domain.
  • CCD2 cysteine rich domain 2
  • CCD1 cysteine rich domain 1
  • the a BIM that is a TACI polypeptide is a truncated wild-type TACI extracellular domain or is a variant thereof, wherein the truncated wild-type TACI extracellular domain consists of a contiguous sequence contained within amino acid residues 67-118 that includes amino acid residues 71-104, with reference to positions set forth in SEQ ID NO:709, wherein the variant TACI polypeptide comprises one or more amino acid substitutions in the truncated wild-type TACI extracellular domain.
  • a BIM that is a TACI polypeptide is one in which the truncated wild-type TACI extracellular domain is 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 59, 50 or 51 amino acids in length.
  • the truncated wild-type TACI extracellular domain consists of amino acid residues 68-110 set forth in SEQ ID NO: 709.
  • the TACI polypeptide consists of the sequence of amino acid set forth in SEQ ID NO:528 or is a variant thereof containing one or more amino acid substitutions in the sequence set forth in SEQ ID NO: 528.
  • a BIM that is a TACI polypeptide is one in which the TACI polypeptide is a truncated wild-type TACI extracellular domain that consists of the sequence set forth in SEQ ID NO: 528.
  • a BIM that is a TACI polypeptide is one in which the truncated TACI polypeptide or the variant thereof binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • a BIM that is a TACI polypeptide is one in which the TACI polypeptide is a variant TACI polypeptide, wherein the variant TACI polypeptide has increased binding affinity to one or both of APRIL and BAFF compared to the truncated TACI polypeptide.
  • the variant TACI polypeptide comprises one or more amino acid substitutions in the extracellular domain (ECD) of a reference TACI polypeptide or a specific binding fragment thereof at positions selected from among 74, 75, 76, 77, 78, 79, 82, 83, 84, 85, 86, 87, 88, 92, 95, 97, 98, 99, 101, 102 and 103, corresponding to numbering set forth in SEQ ID NO: 709.
  • ECD extracellular domain
  • the one or more amino acid substitutions are selected from E74V, Q75E, Q75R, G76S, K77E, F78Y, Y79F, L82H, L82P, L83S, R84G, R84L, R84Q, D85E, D85V, C86Y, I87L, I87M, S88N, I92V, Q95R, P97S, K98T, Q99E, A101D, Y102D, F103S, F103V, F103Y, or a conservative amino acid substitution thereof.
  • the one or more amino acid substitutions comprise at least one of E74V, K77E, Y79F, L82H, L82P, R84G, R84L, R84Q, D85V, or C86Y. In some of any embodiments, the one or more amino acid substitutions are D85E/K98T, I87L/K98T, L82P/I87L, G76S/P97S, K77E/R84L/F103Y, Y79F/Q99E, L83S/F103S, K77E/R84Q, K77E/A101D, K77E/F78Y/Y102D, Q75E/R84Q, Q75R/R84G/I92V, K77E/A101D/Y102D, R84Q/S88N/A101D, R84Q/F103V, K77E/Q95R/A101D or I87M/A101D.
  • the one or more amino acid substitutions are K77E/F78Y/Y102D. In some embodiments, the one or more amino acid substitutions are Q75E/R84Q. In some embodiments, a BIM that is a TACI polypeptide is the variant TACI polypeptide set forth in SEQ ID NO: 541. In some embodiments, a BIM that is a TACI polypeptide is the variant TACI polypeptide set forth in SEQ ID NO:542.
  • a BIM that is a TACI polypeptide is one in which the TACI polypeptide is a variant TACI polypeptide that comprises one or more amino acid substitutions in the extracellular domain (ECD) of a reference TACI polypeptide or a specific binding fragment thereof at positions selected from among 40, 59, 60, 61, 74, 75, 76, 77, 78, 79, 82, 83, 84, 85, 86, 87, 88, 92, 95, 97, 98, 99, 101, 102 and 103, corresponding to numbering of positions set forth in SEQ ID NO:709.
  • ECD extracellular domain
  • the one or more amino acid substitutions are selected from W40R, Q59R, R60G, T61P E74V, Q75E, Q75R, G76S, K77E, F78Y, Y79F, L82H, L82P, L83S, R84G, R84L, R84Q, D85E, D85V, C86Y, I87L, I87M, S88N, I92V, Q95R, P97S, K98T, Q99E, A101D, Y102D, F103S, F103V, F103Y, or a conservative amino acid substitution thereof.
  • the one or more amino acid substitutions comprise at least one of E74V, K77E, Y79F, L82H, L82P, R84G, R84L, R84Q, D85V or C86Y. In some of any embodiments, the one or more amino acid substitution comprise at least the amino acid substitution K77E. In some of any embodiments, the one or more amino acid substitution comprise at least the amino acid substitution R84G. In some of any embodiments, the one or more amino acid substitution comprise at least the amino acid substitution R84Q.
  • the reference TACI polypeptide is a truncated polypeptide consisting of the extracellular domain of TACI or a specific binding portion thereof that binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • the reference TACI polypeptide comprises (i) the sequence of amino acids set forth in SEQ ID NO:709, (ii) a sequence of amino acids that has at least 95% sequence identity to SEQ ID NO:709; or (iii) a portion of (i) or (ii) containing one or both of a CRD1 domain and CRD2 domain that binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • the reference TACI polypeptide lacks an N-terminal methionine.
  • the reference TACI polypeptide comprises the CRD1 domain and the CRD2 domain.
  • the reference TACI polypeptide comprises the sequence set forth in SEQ ID NO:516. In some of any embodiments, the reference TACI polypeptide consists of the sequence set forth in SEQ ID NO:516. In some of any embodiments, the reference TACI polypeptide consists essentially of the CRD2 domain. In some of any embodiments, the reference TACI polypeptide comprises the sequence set forth in SEQ ID NO:528. In some of any embodiments, the reference TACI polypeptide consists of the sequence set forth in SEQ ID NO:528.
  • the one or more amino acid substitutions are D85E/K98T, I87L/K98T, R60G/Q75E/L82P, R60G/C86Y, W40R/L82P/F103Y, W40R/Q59R/T61P/K98T, L82P/I87L, G76S/P97S, K77E/R84L/F103Y, Y79F/Q99E, L83S/F103S, K77E/R84Q, K77E/A101D, K77E/F78Y/Y102D, Q75E/R84Q, Q75R/R84G/I92V, K77E/A101D/Y102D, R84Q/S88N/A101D, R84Q/F103V, K77E/Q95R/A101D or I87M/A101D.
  • the one or more amino acid substitutions are K77E/F78Y/Y102D. In some of any embodiments, the one or more amino acid
  • a BIM that that is a TACI polypeptide is one in which the variant TACI polypeptide has at least about 85%, at least about 90% or at least about 95% sequence identity to SEQ ID NO:709, and contains the one or more amino acid substitutions as described.
  • a BIM that that is a TACI polypeptide is one in which the variant TACI polypeptide has at least about 85%, at least about 90% or at least about 95% sequence identity to SEQ ID NO:719, and contains the one or more amino acid substitutions as described.
  • a BIM that that is a TACI polypeptide is one in which the variant TACI polypeptide has at least about 85%, at least about 90% or at least about 95% sequence identity to SEQ ID NO:718, and contains the one or more amino acid substitutions as described.
  • a BIM that that is a TACI polypeptide is one in which the variant TACI polypeptide has at least about 85%, at least about 90% or at least about 95% sequence identity to SEQ ID NO:516, and contains the one or more amino acid substitutions as described.
  • a BIM that that is a TACI polypeptide is one in which the variant TACI polypeptide has at least about 85%, at least about 90% or at least about 95% sequence identity to SEQ ID NO:528, and contains the one or more amino acid substitutions as described.
  • a BIM that is a TACI polypeptide is one in which the variant TACI polypeptide has increased binding affinity to one or both of APRIL and BAFF compared to the reference TACI polypeptide. In some of any embodiments, the variant TACI polypeptide has increased binding affinity to APRIL. In some of any embodiments, the variant TACI polypeptide has increased binding affinity to BAFF. In some of any embodiments, the variant TACI polypeptide has increased binding affinity to APRIL and BAFF.
  • the increased binding affinity for BAFF or APRIL is independently 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.
  • a BIM that is a TACI polypeptide is one in which the variant TACI polypeptide comprises the sequence set forth in any one of SEQ ID NOS: 517-527, 536, 537, 682-701. In some of any embodiments, a BIM that is a TACI polypeptide is one in which the variant TACI polypeptide comprises the sequence set forth in any one of SEQ ID NOS: 529-535, 538-550, or 673-681.
  • a BIM that is a TACI polypeptide is one in which the variant TACI polypeptide consists or consists essentially of the sequence set forth in any one of SEQ ID NOS: 517-527, 536, 537, 682-701. In some of any embodiments, a BIM that is a TACI polypeptide is one in which the variant TACI polypeptide consists or consists essentially of the sequence set forth in any one of SEQ ID NOS: 529-535, 538-550, 673-681.
  • a BIM is a TACI polypeptide in which the variant TACI polypeptide is is set forth in any one of SEQ ID NO:535, SEQ ID NO: 541, SEQ ID NO:542, or SEQ ID NO:688.
  • a BIM is a TACI polypeptide is one in which the variant TACI polyeptide consists or consists essentially of the sequence set forth in SEQ ID NO:535.
  • a BIM that is a TACI polypeptide is one in which the variant TACI polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 541.
  • a BIM that is a TACI polypeptide is one in which the variant TACI polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO:542.
  • a BIM that is a TACI polypeptide is one in which the variant TACI polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO:688. In some of any embodiments, a BIM that is a TACI polypeptide is one in which the variant TACI polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO:535.
  • the BIM is a BCMA polypeptide that consists of the BCMA extracellular domain or a binding portion thereof that binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • the BCMA extracellular domain or the binding portion thereof is an extracellular domain sequence set forth as (i) the sequence of amino acids set forth in SEQ ID NO:356, (ii) a sequence of amino acids that has at least 95% sequence identity to SEQ ID NO:356; or (iii) a portion of (i) or (ii) comprising a CRD domain.
  • the BCMA polypeptide consists of the sequence set forth in SEQ ID NO:356.
  • the BCMA polypeptide is a variant BCMA polypeptide containing one or more amino acid substitutions in the extracellular domain (ECD) of a reference BCMA polypeptide or a specific binding fragment at positions selected from among 9, 10, 11, 14, 16, 19, 20, 22, 25, 27, 29, 30, 31, 32, 35, 36, 39, 43, 45, 46, 47 and 48, corresponding to numbering set forth in SEQ ID NO: 710.
  • an immunomodulatory protein containing a variant BCMA polypeptide wherein the variant BCMA polypeptide comprises one or more amino acid substitutions in the extracellular domain (ECD) of a reference BCMA polypeptide or a specific binding fragment thereof at positions selected from among 9, 10, 11, 14, 16, 19, 20, 22, 25, 27, 29, 30, 31, 32, 35, 36, 39, 43, 45, 46, 47 and 48, corresponding to numbering of positions set forth in SEQ ID NO:710.
  • the reference BCMA polypeptide is a polypeptide consisting of the extracellular domain of BCMA or a specific binding portion thereof that binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • the reference BCMA polypeptide comprises (i) the sequence of amino acids set forth in SEQ ID NO:710, (ii) a sequence of amino acids that has at least 95% 37a.equence identity to SEQ ID NO:710; or (iii) a portion of (i) or (ii) containing the CRD. In some of any embodiments, the reference BCMA lacks an N-terminal methionine.
  • the reference BCMA polypeptide comprises (i) the sequence of amino acids set forth in SEQ ID NO:356, (ii) a sequence of amino acids that has at least 95% sequence identity to SEQ ID NO:356; or (iii) a portion of (i) or (ii) containing the CRD. In some of any embodiments, the reference BCMA polypeptide comprises the sequence set forth in SEQ ID NO:356. In some of any embodiments, the reference BCMA polypeptide consists of the sequence set forth in SEQ ID NO:356.
  • the one or more amino acid substitutions of a BCMA polypeptide are selected from S9G, S9N, S9Y, Q10E, Q10P, N11D, N11S, F14Y, S16A, H19A, H19C, H19D, H19E, H19F, H19G, H19I, H19K, H19L, H19M, H19N, H19P, H19Q, H19R, H19S, H19T, H19V, H19W, H19Y, A20T, I22L, I22V, Q25E, Q25F, Q25G, Q25H, Q25I, Q25K, Q25L, Q25M, Q25S, Q25V, Q25Y, R27H, R27L, S29P, S30G, S30Y, N31D, N31G, N31H, N31K, N31L, N31M, N31P, N31S, N31V, N31Y, T
  • the one or more amino acid substitutions of a BCMA polypeptide comprise at least one substitution at position 19. In some embodiments, at least one substitution is selected from H19A, H19C, H19D, H19E, H19F, H19G, H19I, H19K, H19L, H19M, H19N, H19P, H19Q, H19R, H19S, H19T, H19V, H19W, H19Y. In some of any embodiments, the one or more amino acid substitution comprise at least the amino acid substitution H19L. In some of any embodiments, the one or more amino acid substitution comprise at least the amino acid substitution H19K. In some of any embodiments, the one or more amino acid substitution comprise at least the amino acid substitution H19R. In some of any embodiments, the one or more amino acid substitution comprise at least the amino acid substitution H19Y.
  • the one or more amino acid substitutions of a BCMA polypeptide comprise at least one substitution at position 25.
  • at least one substitution is selected from Q25E, Q25F, Q25G, Q25H, Q25I, Q25K, Q25L, Q25M, Q25S, Q25V, Q25Y.
  • the one or more amino acid substitutions of a BCMA polypeptide comprise at least one substitution at position 31.
  • at least one substitution is selected from N31D, N31G, N31H, N31K, N31L, N31M, N31P, N31S, N31V, N31Y.
  • the one or more amino acid substitutions of a BCMA polypeptide comprise at least one substitution at position 35. In some embodiments, at least one substitution is selected from L35A, L35M, L35P, L35S, L35V, L35Y.
  • the one or more amino acid substitutions of a BCMA polypeptide comprise at least one substitution at position 36.
  • at least one substitution is selected from T36A, T36G, T36N, T36M, T36S, T36V.
  • the one or more amino acid substitutions of a BCMA polypeptide are H19Y/S30G; H19Y/V45A; F14Y/H19Y; H19Y/V45D; H19Y/A43E; H19Y/T36A; H19Y/I22V; N11D/H19Y; H19Y/T36M; N11S/H19Y; H19Y/L35P/T46A; H19Y/N47D; S9D/H19Y; H19Y/S30G/V45D; H19Y/R39Q; H19Y/L35P; S9D/H19Y/R27H; Q10P/H19Y/Q25H; H19Y/R39L/N47D; N11D/H19Y/N47D; H19Y/T32S; N11S/H19Y/S29P; H19Y/R39Q/N47D; S16A/H19Y/R39Q; S9N
  • the one or more amino acid substitutions of a BCMA polypeptide comprise H19F, H19L, H19K, H19M, H19R, H10Y, N11D/H19Y/N47D, H19Y/R39Q/N47D; S16A/H19Y/R39Q, S9G/H19Y/T32S; H19Y/T36A/N47Y; or Q10E/H19Y/A20T/T36S.
  • the one or more amino acid substitutions of a BCMA polypeptide comprise S16A/H19Y/R39Q.
  • a variant BCMA polypeptide is one in which the variant BCMA polypeptide has at least about 85%, at least about 90% or at least about 95% sequence identity to SEQ ID NO:710, and contains the one or more amino acid substitutions as described.
  • a variant BCMA polypeptide is one in which the variant BCMA polypeptide has at least about 85%, at least about 90% or at least about 95% sequence identity to SEQ ID NO:356, and contains the one or more amino acid substitutions as described.
  • the variant BCMA polypeptide has up to 10 amino acid substitutions compared to the reference BCMA polypeptide. In some of any embodiments, the variant BCMA polypeptide has up to 5 amino acid substitutions compared to the reference BCMA polypeptide. In some of any embodiments, the variant BCMA polypeptide has at least 90% sequence identity to SEQ ID NO:356. In some of any embodiments, the variant BCMA polypeptide has at least 95% sequence identity to SEQ ID NO:356.
  • the variant BCMA polypeptide has increased binding affinity to one or both of APRIL and BAFF compared to the reference BCMA polypeptide. In some of any embodiments, the variant BCMA polypeptide has increased binding affinity to APRIL. In some of any embodiments, the variant BCMA polypeptide has increased binding affinity to BAFF. In some of any embodiments, the variant BCMA polypeptide has increased binding affinity to APRIL and BAFF. In some of any embodiments, the increased binding affinity for BAFF or APRIL is independently 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 variant BCMA polypeptide comprises the sequence set forth in any one of SEQ ID NOS: 357-435. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in any one of SEQ ID NOS: 357-435. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 357. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 377. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 380.
  • the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 381. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 390. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 391. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 396. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 402.
  • the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 405. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 406. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 407. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO:411. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO:405. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO:406.
  • the immunomodulatory protein contains a heterologous moiety that is linked to the at least one BCMA polypeptide.
  • the heterologous moiety is a half-life extending moiety, a multimerization domain, a targeting moiety that binds to a molecule on the surface of a cell, or a detectable label.
  • 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.
  • PEG polyethylene glycol
  • XTEN long unstructured hydrophilic sequences of amino acids
  • HES hydroxyethyl starch
  • albumin-binding small molecule or a combination thereof.
  • the immunomodulatory protein contains an Fc region of an immunoglobulin that is linked to the at least one BCMA polypeptide.
  • the at least one TIM comprises only one TIM. In some of any embodiments, the at least one TIM comprises 2, 3, 4, or 5 TIMs, optionally wherein each TIM is the same. In some of any embodiments, each TIM is linked directly or indirectly via a linker, optionally wherein the linker is a peptide linker. In some of any embodiments, the at least one BIM comprises only one BIM. In some of any embodiments, the at least one BIM comprises 2, 3, 4, or 5 BIMs, optionally wherein each BIM is the same. In some of any embodiments, each BIM is linked directly or indirectly via a linker, optionally wherein the linker is a peptide linker.
  • the linker is a peptide linker and the peptide linker is selected from GSGGS (SEQ ID NO: 592), GGGGS (G4S; SEQ ID NO: 593), GSGGGGS (SEQ ID NO: 590), GGGGSGGGGS (2 ⁇ GGGGS; SEQ ID NO: 594), GGGGSGGGGSGGGGS (3 ⁇ GGGGS; SEQ ID NO: 595), GGGGSGGGGSGGGGSGGGGS (4 ⁇ GGGGS, SEQ ID NO:600), GGGGSGGGGSGGGGSGGGGSGGGGS (5 ⁇ GGGGS, SEQ ID NO: 671), GGGGSSA (SEQ ID NO: 596) or combinations thereof.
  • the at least one TIM and the at least one BIM are linked directly or indirectly via a linker, optionally wherein the linker comprises a peptide linker and/or a multimerization moiety.
  • the linker comprises a peptide linker and the peptide linker is selected from GSGGS (SEQ ID NO: 592), GGGGS (G4S; SEQ ID NO: 593), GSGGGGS (SEQ ID NO: 590), GGGGSGGGGS (2 ⁇ GGGGS; SEQ ID NO: 594), GGGGSGGGGSGGGGS (3 ⁇ GGGGS; SEQ ID NO: 595), GGGGSGGGGSGGGGSGGGGS (4 ⁇ GGGGS, SEQ ID NO:600), GGGGSGGGGSGGGGSGGGGSGGGGS (5 ⁇ GGGGS, SEQ ID NO: 671), GGGGSSA (SEQ ID NO: 596) or combinations thereof.
  • the linker comprises a peptide linker and the peptide linker is selected from SEQ ID NO: 711 (1 ⁇ EAAAK), SEQ ID NO: 712 (2 ⁇ EAAAK), SEQ ID NO: 713 (3 ⁇ EAAAK), SEQ ID NO: 714 (4 ⁇ EAAAK), SEQ ID NO: 715 (5 ⁇ EAAAK), SEQ ID NO: 665 (6 ⁇ EAAAK).
  • the immunomodulatory protein is a monomer and/or comprises a single polypeptide chain.
  • the at least one TIM is amino-terminal to the at least one BIM in the polypeptide. In some of any embodiments, the at least one TIM is carboxy-terminal to the at least one BIM in the polypeptide.
  • the immunomodulatory protein further comprises a detectable label, optionally wherein the detectable label is a Flag tag, a His tag, or a myc tag.
  • the immunomodulatory protein comprises the sequence of amino acids set forth in any of SEQ ID NOS: 618-623, or a sequence that exhibits at least 85% 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity thereto and retains activity.
  • the immunomodulatory protein comprises the sequence of amino acids set forth in any of SEQ ID NOS: 703-708, or a sequence that exhibits at least 85% 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity thereto and retains activity.
  • the TIM and the BIM are linked by a multimerization domain.
  • the multimerization domain promotes dimerization, trimerization, tetramerization, or pentamerization.
  • the multimerization domain is an immunoglobulin Fc region.
  • the immunomodulatory protein is a dimer.
  • the immunoglobulin Fc region is a homodimeric Fc region. In some of any embodiments, the immunoglobulin Fc region is a heterodimeric Fc region.
  • the immunomodulatory protein is a homodimer, wherein each polypeptide of the dimer is the same.
  • each polypeptide comprises the at least one TIM and the at least one BIM and wherein the at least one TIM is amino-terminal to the at least one BIM in each polypeptide.
  • each polypeptide comprises the at least one TIM and the at least one BIM and wherein the at least one TIM is carboxy-terminal to the at least one BIM in each polypeptide.
  • the immunoglobulin Fc region is an IgG2 Fc domain.
  • the IgG2 Fc domain comprises the sequence of amino acids set forth in SEQ ID NO: 729 or 853 or a sequence of amino acids that exhibits at least 95% sequence identity to SEQ ID NO:729 or 853.
  • the IgG2 Fc domain is set forth in SEQ ID NO:729.
  • the IgG2 Fc domain is set forth in SEQ ID NO: 853.
  • the immunoglobulin Fc region is an IgG4 Fc domain.
  • the IgG4 Fc domain is a variant IgG4 Fc domain comprising the amino acid substitution S228P.
  • the IgG4 Fc domain comprises the sequence of amino acids set forth in SEQ ID NO: 731 or 854, or a sequence of amino acids that exhibits at least 95% sequence identity to SEQ ID NO: 731 or 854.
  • the IgG4 Fc domain is set forth in SEQ ID NO:731.
  • the IgG4 Fc domain is set forth in SEQ ID NO: 854.
  • the immunoglobulin Fc is an IgG1 Fc domain, or is a variant Fc that exhibits reduced binding affinity to an Fc receptor and/or reduced effector function, optionally as compared to a wild-type IgG1 Fc domain.
  • the immunoglobulin Fc is an IgG1 Fc domain and the Fc includes the amino acid sequence set forth in SEQ ID NO: 597.
  • the immunoglobulin Fc is an IgG4 Fc domain, either wild type or modified.
  • the immunoglobulin Fc is a variant IgG1 Fc domain containing one or more amino acid substitutions selected from L234A, L234V, L235A, L235E, G237A, S267K, R292C, N297G, and V302C, by EU numbering.
  • the immunoglobulin Fc region contains the amino acid substitutions L234A, L235E an G237A by EU numbering or the amino acid substitutions R292C, N297G and V302C by EU numbering.
  • the Fc is a variant Fc including the amino acid sequence set forth in SEQ ID NO:589.
  • the Fc is a variant Fc including the amino acid sequence set forth in SEQ ID NO: 855.
  • the immunomodulatory protein is a BCMA-Fc fusion proteion.
  • the BCMA-Fc fusion protein comprises the structure: BCMA polypeptide (BCMA)-Linker-Fc region.
  • the BCMA-Fc fusion protein is set forth in SEQ ID NO:629.
  • an immunomodulatory BCMA-Fc fusion protein that is a homodimer comprising two identical copies of the BCMA-Fc fusion protein set forth in SEQ ID NO: 629 linked by a covalent disulfide bond.
  • the immunomodulatory protein is a BCMA-Fc fusion protein that has the structure: (BCMA)-Linker-Fc region-Linker-(BCMA).
  • the BCMA-Fc fusion protein is set forth in SEQ ID NO: 809. In some embodiments, the BCMA-Fc fusion protein is set forth in SEQ ID NO: 812.
  • the BCMA-Fc fusion protein has the structure: (BCMA)-Linker-(BCMA)-Linker-Fc region. In some embodiments, the BCMA-Fc fusion protein is set forth in SEQ ID NO: 813.
  • the immunomodulatory protein that contains at least one TIM and at least one BIM comprises the sequence of amino acids set forth in any of SEQ ID NOS: 610-617, 624-627, 637, 638, 643, 644, 648, 653 and 654, or a sequence that exhibits at least 85% 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity thereto and retains activity.
  • the TIM is a wild-type CTLA-4 extracellular domain or a binding portion thereof
  • the BIM is a TACI extracellular domain or a binding portion thereof comprising amino acid substitutions K77E/F78Y/Y102D, Q75E/R84Q, or R84G, corresponding to positions set forth in SEQ ID NO: 709.
  • the TIM is set forth in SEQ ID NO:1 and the BIM is set forth in SEQ ID NO:535, 541, 542, or 688.
  • the immunomodulatory protein comprises the sequence set forth in SEQ ID NO:611, SEQ ID NO:788, SEQ ID NO: 789, SEQ ID NO: 790, or SEQ ID NO: 792.
  • an immunomodulatory protein that is a homodimer comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 611 linked by a covalent disulfide bond.
  • an immunomodulatory protein that is a homodimer comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 788 linked by a covalent disulfide bond.
  • an immunomodulatory protein that is a homodimer comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 789 linked by a covalent disulfide bond.
  • an immunomodulatory protein that is a homodimer comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 790 linked by a covalent disulfide bond.
  • an immunomodulatory protein that is a homodimer comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 792 linked by a covalent disulfide bond.
  • the TIM is a wild-type CTLA-4 extracellular domain or a binding portion thereof, and the BIM is a truncated TACI extracellular domain comprising the CRD2 domain.
  • the TIM is set forth in SEQ ID NO:1 and the BIM is set forth in SEQ ID NO:528.
  • the immunomodulatory protein comprises the sequence set forth in SEQ ID NO:759, SEQ ID NO: 853, SEQ ID NO: 854 or SEQ ID NO: 791.
  • an immunomodulatory protein that is a homodimer comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 759 linked by a covalent disulfide bond.
  • an immunomodulatory protein that is a homodimer comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 853 linked by a covalent disulfide bond.
  • an immunomodulatory protein that is a homodimer comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 854 linked by a covalent disulfide bond.
  • an immunomodulatory protein that is a homodimer comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 791 linked by a covalent disulfide bond.
  • the TIM is a CTLA-4 extracellular domain or a binding portion thereof, comprising amino acid substitutions G29W/L98Q/Y105L corresponding to positions set forth in SEQ ID NO:1
  • the BIM is a TACI extracellular domain or a binding portion thereof comprising amino acid substitutions K77E/F78Y/Y102D, Q75E/R84Q, or R84G, corresponding to positions set forth in SEQ ID NO:709.
  • the TIM is set forth in SEQ ID NO: 186 and the BIM is set forth in SEQ ID NO:535, 541, 542, or 688.
  • the immunomodulatory protein comprises he sequence set forth in SEQ ID NO:610.
  • an immunomodulatory protein comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 610 linked by a covalent disulfide bond.
  • the immunomodulatory protein that contains at least one TIM and at least one BIM comprises the sequence of amino acids set forth in any of SEQ ID NOS: 601-609, 631-636, 645-647, 649-652, 655-659, or a sequence that exhibits at least 85% 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity thereto and retains activity.
  • the TIM is a wild-type CTLA-4 extracellular domain or a binding portion thereof
  • the BIM is a BCMA extracellular domain or a binding portion thereof comprising the amino acid substitution H19L corresponding to positions set forth in SEQ ID NO:710.
  • the TIM is set forth in SEQ ID NO:1 and the BIM is set forth in SEQ ID NO:406.
  • the immunomodulatory protein comprises the sequence set forth in SEQ ID NO:602.
  • an immunomodulatory protein that is a homodimer comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 602 linked by a covalent disulfide bond.
  • the TIM is a CTLA-4 extracellular domain or a binding portion thereof comprising the amino acid substitutions G29W/L98Q/Y105L corresponding to positions set forth in SEQ ID NO: 1
  • the BIM is a BCMA extracellular domain or a binding portion thereof comprising the amino acid substitution H19L with reference to positions set forth in SEQ ID NO:710.
  • the TIM is set forth in SEQ ID NO: 186 and the BIM is set forth in SEQ ID NO:406.
  • the immunomodulatory protein comprises the sequence set forth in SEQ ID NO:601.
  • an immunomodulatory protein that is a homodimer comprising two identical copies of the Fc fusion protein set forth in SEQ ID NO: 601 linked by a covalent disulfide bond.
  • the immunomodulatory protein is a heterodimer, wherein each polypeptide of the dimer is linked to an immunoglobulin Fc domain individually containing one or more amino acid modifications in a wild-type Fc domain to effect heterodimer formation between the polypeptides.
  • the wild-type immunoglobulin Fc is an IgG1 Fc domain.
  • the one more amino acid modifications are selected from a knob-into-hole modification and a charge mutation to reduce or prevent self-association due to charge repulsion.
  • the Fc region further contains one or more amino acid substitutions to reduce binding affinity to an Fc receptor and/or reduce effector function, optionally as compared to a wild-type IgG1 Fc domain.
  • the one or more amino acid substitutions are selected from L234A, L234V, L235A, L235E, G237A, S267K, R292C, N297G, and V302C, by EU numbering.
  • the immunoglobulin Fc region contains the amino acid substitutions L234A, L235E an G237A by EU numbering or the amino acid substitutions R292C, N297G and V302C by EU numbering.
  • the heterodimer comprises a first polypeptide containing the sequence of amino acids set forth in SEQ ID NO: 662 or 663 and a second polypeptide containing the sequence of amino acids set forth in SEQ ID NO:660.
  • the immunomodulatory protein blocks binding of APRIL, BAFF, or an APRIL/BAFF heterotrimer to BCMA or TACI; and the immunomodulatory protein reduces the levels of circulating APRIL, BAFF, or an APRIL/BAFF in the blood following administration to a subject. In some of any embodiments, the immunomodulatory protein blocks binding of APRIL, BAFF, or an APRIL/BAFF heterotrimer to BCMA or TACI; or the immunomodulatory protein reduces the levels of circulating APRIL, BAFF, or an APRIL/BAFF in the blood following administration to a subject. In some of any embodiments, the immunomodulatory protein reduces or inhibits B cell maturation, differentiation and proliferation.
  • the immunomodulatory protein blocks binding of CD80 or CD86 to a costimulatory receptor, optionally wherein the costimulatory receptor is CD28; and the immunomodulatory protein reduces or inhibits T cell co-stimulation. In some of any embodiments, the immunomodulatory protein reduces or inhibits B cell maturation, differentiation or proliferation. In some of any embodiments, the immunomodulatory protein blocks binding of CD80 or CD86 to a costimulatory receptor, optionally wherein the costimulatory receptor is CD28; or the immunomodulatory protein reduces or inhibits T cell costimulation.
  • nucleic acid molecule(s) encoding the immunomodulatory protein of any of the embodiments described herein.
  • the nucleic acid molecule is a synthetic nucleic acid.
  • the nucleic acid molecule is a cDNA.
  • the vector is an expression vector.
  • the vector is a mammalian expression vector or a viral vector.
  • a cell containing the nucleic acid of any of any of the embodiments described herein or the vector of any of any of embodiments described herein.
  • the cell is a mammalian cell. In some of any embodiments, the cell is a human cell.
  • an immunomodulatory protein including introducing the nucleic acid molecule of any of the embodiments described herein or vector of any of the embodiments described herein into a host cell under conditions to express the protein in the cell.
  • the method includes isolating or purifying the immunomodulatory protein from the cell.
  • an immunomodulatory protein produced by the method of any of the embodiments described herein.
  • composition including the immunomodulatory protein of any of the embodiments described herein.
  • variant BCMA-Fc fusion protein including a variant BCMA polypeptide, an Fc region, and a linker between the BCMA polypeptide and Fc region, wherein the variant BCMA polypeptide comprises one or more amino acid substitutions in the extracellular domain (ECD) of an unmodified BCMA polypeptide or a specific binding fragment thereof corresponding to positions selected from among 9, 10, 11, 14, 16, 19, 20, 22, 25, 27, 29, 30, 31, 32, 35, 36, 39, 43, 45, 46, 47 and 48 with reference to positions set forth in SEQ ID NO:710.
  • ECD extracellular domain
  • the reference BCMA polypeptide is a polypeptide consisting of the extracellular domain of BCMA or a specific binding portion thereof that binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • the reference BCMA polypeptide comprises (i) the sequence of amino acids set forth in SEQ ID NO:710, (ii) a sequence of amino acids that has at least 95% 37a.equence identity to SEQ ID NO:710; or (iii) a portion of (i) or (ii) containing the CRD.
  • the reference BCMA lacks an N-terminal methionine.
  • the reference BCMA polypeptide comprises (i) the sequence of amino acids set forth in SEQ ID NO:356, (ii) a sequence of amino acids that has at least 95% sequence identity to SEQ ID NO:356; or (iii) a portion of (i) or (ii) containing the CRD. In some of any embodiments, the reference BCMA polypeptide comprises the sequence set forth in SEQ ID NO:356. In some of any embodiments, the reference BCMA polypeptide consists of the sequence set forth in SEQ ID NO:356.
  • the one or more amino acid substitutions are selected from S9G, S9N, S9Y, Q10E, Q10P, N11D, N11S, F14Y, S16A, H19A, H19C, H19D, H19E, H19F, H19G, H19I, H19K, H19L, H19M, H19N, H19P, H19Q, H19R, H19S, H19T, H19V, H19W, H19Y, A20T, I22L, I22V, Q25E, Q25F, Q25G, Q25H, Q25I, Q25K, Q25L, Q25M, Q25S, Q25V, Q25Y, R27H, R27L, S29P, S30G, S30Y, N31D, N31G, N31H, N31K, N31L, N31M, N31P, N31S, N31V, N31Y, T32I, T32S, L
  • the one or more amino acid substitutions comprise at least one substitution at position 19, optionally wherein the at least one substitution is selected from H19A, H19C, H19D, H19E, H19F, H19G, H19I, H19K, H19L, H19M, H19N, H19P, H19Q, H19R, H19S, H19T, H19V, H19W, H19Y.
  • the one or more amino acid substitution comprise at least the amino acid substitution H19L.
  • the one or more amino acid substitution comprise at least the amino acid substitution H19K.
  • the one or more amino acid substitution comprise at least the amino acid substitution H19R.
  • the one or more amino acid substitution comprise at least the amino acid substitution H19Y.
  • the one or more amino acid substitutions comprise at least one substitution at position 25, optionally wherein the at least one substitution is selected from Q25E, Q25F, Q25G, Q25H, Q25I, Q25K, Q25L, Q25M, Q25S, Q25V, Q25Y. In some of any embodiments, the one or more amino acid substitutions comprise at least one substitution at position 31, optionally wherein the at least one substitution is selected from N31D, N31G, N31H, N31K, N31L, N31M, N31P, N31S, N31V, N31Y.
  • the one or more amino acid substitutions comprise at least one substitution at position 35, optionally wherein the at least one substitution is selected from L35A, L35M, L35P, L35S, L35V, L35Y. In some of any embodiments, the one or more amino acid substitutions comprise at least one substitution at position 36, optionally wherein the at least one substitution is selected from T36A, T36G, T36N, T36M, T36S, T36V.
  • the one or more amino acid substitutions are H19Y/S30G; H19Y/V45A; F14Y/H19Y; H19Y/V45D; H19Y/A43E; H19Y/T36A; H19Y/I22V; N11D/H19Y; H19Y/T36M; N11S/H19Y; H19Y/L35P/T46A; H19Y/N47D; S9D/H19Y; H19Y/S30G/V45D; H19Y/R39Q; H19Y/L35P; S9D/H19Y/R27H; Q10P/H19Y/Q25H; H19Y/R39L/N47D; N11D/H19Y/N47D; H19Y/T32S; N11S/H19Y/S29P; H19Y/R39Q/N47D; S16A/H19Y/R39Q; S9N/H19Y/N31K
  • the one or more amino acid substitutions comprise S16A/H19Y/R39Q.
  • the variant BCMA polypeptide has increased binding affinity to one or both of APRIL and BAFF compared to the reference TACI polypeptide.
  • the variant BCMA polypeptide has increased binding affinity to APRIL. In some of any embodiments, the variant BCMA polypeptide has increased binding affinity to BAFF. In some of any embodiments, the variant BCMA polypeptide has increased binding affinity to APRIL and BAFF.
  • the increased binding affinity for BAFF or APRIL is independently 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 variant BCMA polypeptide comprises the sequence set forth in any one of SEQ ID NOS: 357-435. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in any one of SEQ ID NOS: 357-435. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO: 381.
  • the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO:411. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO:405. In some of any embodiments, the variant BCMA polypeptide consists or consists essentially of the sequence set forth in SEQ ID NO:406.
  • the linker comprises a peptide linker and the peptide linker is selected from GSGGS (SEQ ID NO: 592), GGGGS (G4S; SEQ ID NO: 593), GSGGGGS (SEQ ID NO: 590), GGGGSGGGGS (2 ⁇ GGGGS; SEQ ID NO: 594), GGGGSGGGGSGGGGS (3 ⁇ GGGGS; SEQ ID NO: 595), GGGGSGGGGSGGGGSGGGGS (4 ⁇ GGGGS, SEQ ID NO:600), GGGGSGGGGSGGGGSGGGGSGGGGS (5 ⁇ GGGGS, SEQ ID NO: 671), GGGGSSA (SEQ ID NO: 596) or combinations thereof.
  • the Fc fusion protein is a dimer. In some of any embodiments, the immunoglobulin Fc region is a homodimeric Fc region.
  • the immunoglobulin Fc is an IgG1 Fc domain, or is a variant Fc that exhibits reduced binding affinity to an Fc receptor and/or reduced effector function, optionally as compared to a wild-type IgG1 Fc domain.
  • the immunoglobulin Fc is an IgG1 Fc domain and the Fc includes the amino acid sequence set forth in SEQ ID NO: 597.
  • the immunoglobulin Fc is a variant IgG1 Fc domain containing one or more amino acid substitutions selected from L234A, L234V, L235A, L235E, G237A, S267K, R292C, N297G, and V302C, by EU numbering.
  • the immunoglobulin Fc region contains the amino acid substitutions L234A, L235E an G237A by EU numbering or the amino acid substitutions R292C, N297G and V302C by EU numbering.
  • the immunoglobulin Fc is set forth in SEQ ID NO:586.
  • the Fc is a variant Fc comprising the amino acid sequence set forth in SEQ ID NO:589.
  • the Fc fusion protein is a homodimer.
  • the Fc fusion protein neutralizes APRIL and BAFF.
  • the IC50 for neutralizing APRIL is less than 100 pM, less than 50 pM, less than 40 pM, less than 30 pM, less than 20 pM, less than 10 pM, less than 5 pM or less than 1 pM, or is any value between any of the foregoing; and/or the IC50 for neutralizing BAFF is less than 400 pM, less than 300 pM, less than 200 pM, less than 100 pM, less than 75 pM, less than 50 pM, less than 25 pm, or less than 10 pM, or is any value between any of the foregoing.
  • the Fc fusion protein blocks binding of APRIL, BAFF, or an APRIL/BAFF heterotrimer to BCMA or TACI; and the Fc fusion protein reduces the levels of circulating APRIL, BAFF, or an APRIL/BAFF in the blood following administration to a subject.
  • the immunoglobulin Fc is set forth in SEQ ID NO:586.
  • the Fc fusion protein blocks binding of APRIL, BAFF, or an APRIL/BAFF heterotrimer to BCMA or TACI; or the Fc fusion protein reduces the levels of circulating APRIL, BAFF, or an APRIL/BAFF in the blood following administration to a subject.
  • the immunomodulatory protein reduces or inhibits B cell maturation, differentiation and/or proliferation.
  • nucleic acid molecule(s) encoding the Fc fusion protein of any of the embodiments described herein.
  • the nucleic acid molecule is a synthetic nucleic acid.
  • the nucleic acid molecule is a cDNA.
  • the vector is an expression vector.
  • the vector is a mammalian expression vector to a viral vector.
  • a cell containing the nucleic acid of any of any of the embodiments described herein or the vector of any of any of any of the embodiments described herein.
  • the cell is a mammalian cell. In some of any embodiments, the cell is a human cell.
  • a method of producing an immunomodulatory protein including introducing the nucleic acid molecule of any of any of the embodiments provided herein or vector of any of any of the embodiments provided herein into a host cell under conditions to express the protein in the cell.
  • the method further includes isolating or purifying the Fc fusion protein from the cell.
  • a method of producing an Fc fusion protein including introducing the nucleic acid molecule of any of the embodiments provided herein or vector of any of the embodiments provided herein into a host cell under conditions to express the protein in the cell.
  • an Fc fusion protein produced by the method of any of the embodiments described herein.
  • a pharmaceutical composition including the Fc fusion protein of any of any of the embodiments described herein.
  • the pharmaceutical composition contains a pharmaceutically acceptable excipient.
  • the pharmaceutical composition is sterile.
  • an article of manufacture including the pharmaceutical composition of any of the embodiments described herein in a vial or container.
  • the vial or container is sealed.
  • kits including the pharmaceutical composition of any of any of the embodiments provided herein and instructions for use.
  • the kit includes the article of manufacture of any of the embodiments described herein and instructions for use.
  • a method of reducing an immune response in a subject including administering the immunomodulatory protein of any of the embodiments described herein to a subject in need thereof.
  • Provided heroine is a method of reducing an immune response in a subject, including administering the Fc fusion protein of any of the embodiments described herein to a subject in need thereof.
  • a method of reducing an immune response in a subject including administering the pharmaceutical composition of any of any of the embodiments described herein to a subject in need thereof.
  • a B cell immune response is reduced in the subject, whereby B cell maturation, differentiation and/or proliferation is reduced or inhibited.
  • circulating levels of APRIL, BAFF or an APRIL/BAFF heterotrimer are reduced in the subject.
  • a method of reducing circulating levels of APRIL, BAFF or an APRIL/BAFF heterotrimer in a subject including administering the pharmaceutical composition of any of any of the embodiments described herein to the subject.
  • a T cell immune response is reduced in the subject, whereby T cell co-stimulation is reduced or inhibited.
  • reducing the immune response treats a disease or condition in the subject.
  • a method of treating a disease, disorder or condition in a subject including administering the immunomodulatory protein of any of any of the embodiments described herein to a subject in need thereof.
  • Provided herein is a method of treating a disease, disorder or condition in a subject, including administering the Fc fusion protein of any of any of the embodiments described herein to a subject in need thereof.
  • a method of treating a disease, disorder or condition in a subject including administering the pharmaceutical composition of any of any of the embodiments described herein to a subject in need thereof.
  • any of the immunomodulatory proteins or pharmaceutical compositions containing same for use in treating a disease, disorder or condition in a subject. Also provided herein are uses of any of the immunomodulatory proteins or pharmaceutical compositions containing same for formulation of a medicament for treating a disease, disorder or condition in a subject.
  • the disease, disorder or condition is an autoimmune disease, an inflammatory condition, a B cell cancer, an antibody-mediated pathology, a renal disease, a graft rejection, graft versus host disease, or a viral infection.
  • the disease or condition that is treated may be any as described herein.
  • the disease or condition is an autoimmune disease selected from the group consisting of Systemic lupus erythematosus (SLE); Sjögren's syndrome, scleroderma, Multiple sclerosis, diabetes, polymyositis, primary biliary cirrhosis, IgA nephropathy, optic neuritis, amyloidosis, antiphospholipid antibody syndrome (APS), autoimmune polyglandular syndrome type II (APS II), autoimmune thyroid disease (AITD), Graves' disease, autoimmune adrenalitis and pemphigus vulgaris.
  • the disease or condition is a B cell cancer and the cancer is myeloma.
  • the type of myeloma includes multiple myeloma, plasmacytoma, multiple solitary plasmacytoma, and/or extramedullary myeloma. In some of any embodiments, the type of myeloma includes light chain myeloma, nonsecretory myeloma, and/or IgD or IgE myeloma.
  • FIG. 1 shows a schematic representation of a functional inhibition assay involving recombinant APRIL and BAFF by BCMA or TAC.
  • APRIL recombinant APRIL
  • BAFF recombinant APRIL
  • endogenous NF- ⁇ B transcription factors bind to the DNA response elements controlling transcription of a firefly luciferase gene.
  • Luciferase expression can be monitored, such as by detection with Bio-GloTM reagent and measurement using a Cytation 3 reader.
  • FIG. 2 shows exemplary human BCMA TD Fc fusion molecules for blockade of human APRIL (top panel) and BAFF (bottom panel) mediated signaling.
  • Exemplary BCMA TD Fc fusions were incubated with APRIL (2 nM) or BAFF (4 nM) for 20 mins (room temperature with shaking) and then added to wells containing 150,000 Jurkat/TACI/NF ⁇ B-luciferase cells for 5 hours.
  • FIGS. 3 A- 3 B show function of exemplary BCMA TD Fc fusion molecules alone or when stacked with CTLA-4 IgD for blockade of APRIL (top panel of each FIG) or BAFF (bottom panel of each FIG).
  • FIG. 3 A and FIG. 3 B show human BCMA TDs retain function when stacked with CTLA-4.
  • FIGS. 3 C- 3 D show function of exemplary TACI TD Fc fusion molecules when stacked with CTLA-4 IgD for blockade of APRIL (top panel of each FIG) or BAFF (bottom panel of each FIG).
  • FIG. 3 C and FIG. 3 D show human TACI TDs retain function when stacked with CTLA-4.
  • FIG. 4 shows human BCMA fusion molecules alone or BCMA or TACI TD Fc fusion molecules when stacked with CTLA-4 IgD for blockade of mouse APRIL (left panel) and BAFF (right panel) mediated signaling.
  • FIG. 5 A shows human BCMA TD Fc fusion molecules alone or when stacked with CTLA-4 IgD for blockade of human APRIL (top panel) and BAFF (bottom panel) mediated signaling relative to TACI 13-118-Fc, TACI 30-110-Fc, and belimumab.
  • FIG. 5 B shows human TACI TD Fc fusion molecules when stacked with CTLA-4 IgD for blockade of human APRIL (tope panel) and BAFF (bottom panel) mediated signaling relative to TACI 13-118-Fc, TACI 30-110-Fc, and belimumab.
  • FIG. 6 shows a schematic representation of a functional inhibition assay of CD80/CD86-CD28 mediated costimulation.
  • Jurkat/IL-2 cells stably express a luciferase reporter driven by the IL-2 promoter when stimulated with anti-CD3 and an anti-CD28 stimulus.
  • Receptor-mediated signaling results in IL-2 promoter-mediated luminescence, and bioluminescent signal can be detected and quantified, such as by using Bio-GloTM substrate and a luminometer.
  • FIG. 7 A and FIG. 7 B shows wild type or CTLA-4 vIgDs (IgSF domain) maintain function for blockade of CD80 (left panel) or CD86 (right panel) when included in multi-specific (stack) construct molecules with BCMA to TACI TDs.
  • FIGS. 8 A- 8 F show activity of CTLA-4 vIgD alone or when included in multi-specific (stack) molecules with BCMA or TACI TDs for inhibiting human follicular helper T (T FH ) and B-cells in an autologous T FH -B cell assay.
  • B-Tfh cell cultures were incubated for 7 days in the presence of titrated (100,000-32 pM) protein. Cultured cells were surface stained and analyzed by flow cytometry for: ( FIG. 8 A ) CD4+ T cell recovery, ( FIG. 8 B ) CD4+CD40L+ cell recovery, ( FIG. 8 C ) CD4+ICOS+ cell recovery, ( FIG.
  • FIG. 8 D CD19+ B cell recovery, and ( FIG. 8 E ) B cell activation/upregulation of CD86.
  • Supernatants were collected and levels of IgM secretion were determined by ELISA ( FIG. 8 F ).
  • Data represent an average ( ⁇ SEM) of three replicates for each condition.
  • FIG. 9 shows anti-KLH IgM antibody levels in serum at termination (Day 19) in a KLH immunization model. Statistical differences between each group were determined by 1-way ANOVA; only significant differences (p ⁇ 0.05) are listed.
  • FIG. 10 shows anti-KLH IgG1 antibody levels in serum at termination (Day 19) in a KLH immunization model. Statistical differences between each group were determined by 1-way ANOVA; only significant differences (p ⁇ 0.05) are listed.
  • FIG. 11 shows spleen weight at termination (Day 19) in the KLH model. Statistical differences between the Fc control and other test articles were determined by t-test; only significant (p ⁇ 0.05) differences are listed.
  • FIGS. 12 A- 12 H show flow cytometry analyses of splenocyte B cell and Tfh subsets at termination (Day 19) in the KLH model.
  • Spleens were processed and analyzed by flow cytometry for B220+ B cells ( FIG. 12 A , FIG. 12 E ); Marginal Zone (MZ) B cells ( FIG. 12 B , FIG. 12 F ); Germinal Center (GC) B cells ( FIG. 12 C , FIG. 12 G ); T Follicular Helper (Tfh) cells ( FIG. 12 D , FIG. 12 H ).
  • MZ Marginal Zone
  • GC Germinal Center
  • Tfh T Follicular Helper
  • FIGS. 13 A- 13 D show flow cytometry analyses of splenocyte T effector memory subsets at termination (Day 19) in a KLH model. Spleens were processed and analyzed by flow cytometry for CD4+( FIG. 13 A , FIG. 13 C ) and CD8 + ( FIG. 13 B , FIG. 13 D ) T effector memory (T em ) cells.
  • ‘Fc control’ Fc set forth in SEQ ID NO:589.
  • Statistically significant differences p ⁇ 0.05
  • Fc control or abatacept were calculated by 1-way ANOVA with uncorrected Fisher's LSD.
  • Kidneys were processed and analyzed by histology in replicate Periodic acid-Schiff (PAS)-stained sections, with individual component and total histology scores depicted in FIG. 14 F .
  • Frozen kidneys were also sectioned and stained for immunohistochemical analysis of mouse IgG and complement C3 glomerular deposition, as shown in FIG. 14 G and FIG. 14 H , respectively.
  • FIG. 14 I shows the histological score ⁇ SEM.
  • FIG. 15 depicts a schematic representations of an exemplary BCMA-Fc fusion protein.
  • FIG. 16 depicts schematic representations of exemplary Fc fusion formats of provided multi-domain (stack) immunomodulatory proteins.
  • FIG. 17 A and FIG. 17 B depict exemplary sequence alignments to identify corresponding residues in a sequence compared to a reference sequence.
  • the symbol “*” between two aligned amino acid indicates that the aligned amino acids are identical.
  • the symbol “ ⁇ ” indicates a gap in the alignment. Exemplary, non-limiting positions for amino acid substitution described herein are indicated with bold text. Based on the alignment of two similar sequences having identical residues in common, a skilled artisan can identify “corresponding” positions in a sequence by comparison to a reference sequence using conserved and identical amino acid residues as guides.
  • FIG. 17 A and FIG. 17 B depict exemplary sequence alignments to identify corresponding residues in a sequence compared to a reference sequence.
  • the symbol “*” between two aligned amino acid indicates that the aligned amino acids are identical.
  • 17 A provides an exemplary alignment of a reference TACI extracellular domain sequence set forth in SEQ ID NO:709 (containing the full extracellular domain with a CRD1 and CRD2 and an initiating methionine residue) with a TACI extracellular domain sequence set forth in SEQ ID NO:528 (containing only a single CRD, CRD2); aligning identical residues demonstrates, for example, that amino acid residue E7 in SEQ ID NO:528 corresponds to residue E74 in SEQ ID NO: 709, amino acid residue K10 in SEQ ID NO: 528 corresponds to residue K77 in SEQ ID NO:709, amino acid residue Y12 in SEQ ID NO: 528 corresponds to Y79 in SEQ ID NO:709, amino acid residue L15 in SEQ ID NO:528 corresponds to L82 in SEQ ID NO:709, amino acid residue R17 in SEQ ID NO: 528 corresponds to R84 in SEQ ID NO:709; and amino acid residue D16 in SEQ ID NO:528 correspond
  • FIG. 17 B provides an exemplary alignment of a reference BCMA extracellular domain sequence set forth in SEQ ID NO:710 (containing the full extracellular domain with a CRD and an initiating methionine residue) with a BCMA extracellular domain sequence set forth in SEQ ID NO:356 (without the initiating methionine); aligning identical residues demonstrates, for example, that amino acid residue H18 in SEQ ID NO:356 corresponds to residue H19 in SEQ ID NO: 710, and amino acid residue R38 in SEQ ID NO:356 corresponds to residue R39 in SEQ ID NO: 710. It is within the level of a skilled artisan to carry out similar alignments between two similar protein sequences to identify corresponding residues, including based on the exemplification and description herein.
  • FIGS. 18 A- 18 D show analysis of parameters assessed murine keyhole limpet hemocyanin (KLH) model. Serum-KLH IgM OD levels were assessed as primary response ( FIG. 18 A ) and secondary response ( FIG. 18 B ). Similarly, serum anti-KLH IgG1 OD levels were assessed as both primary response ( FIG. 18 C ) and secondary response ( FIG. 18 D ).
  • KLH murine keyhole limpet hemocyanin
  • FIGS. 19 A- 19 B shown analysis of harvested spleen assessed from the murine keyhole limpet hemocyanin (KLH) immunization model. Spleens were processed and analyzed by weight ( FIG. 19 A ) as well as total cell number ( FIG. 19 B ).
  • KLH murine keyhole limpet hemocyanin
  • FIG. 20 depicts analysis of spleens assessed for cellular subtype population makeup from the murine keyhole limpet hemocyanin (KLH) model and shows results of B cell subset numbers relative to the group mean.
  • KLH murine keyhole limpet hemocyanin
  • FIG. 21 depicts analysis of spleens assessed for cellular subtype phenotype makeup from the murine keyhole limpet hemocyanin (KLH) model and shows results for numbers of germinal center B cells and plasma cells ( FIG. 21 ).
  • KLH keyhole limpet hemocyanin
  • FIGS. 22 A- 22 D depict T cell numbers in the murine keyhole limpet hemocyanin (KLH) model.
  • KLH murine keyhole limpet hemocyanin
  • the splenic CD3+, CD8+, CD4+ and Follicular Helper T cells are depicted in FIG. 22 A , FIG. 22 B , FIG. 22 C , and FIG. 22 D , respectively.
  • FIG. 23 depicts Tcm and Tem cellular populations in the murine keyhole limpet hemocyanin (KLH) model.
  • FIGS. 24 A- 24 B and FIGS. 25 A- 25 B depict overall incidence and degree of sialadenitis ( FIGS. 24 A- 24 B ) and insulitis ( FIGS. 25 A- 25 B ) in diabetes-prone mice after treatment with 186-CTLA-4 Fc and CTLA4 186-GSG4S-Fc-(G4S)4-TACI 541 tested molecules.
  • FIG. 26 depicts mean blood glucose concentrations (mg/dL) as measured in the blood on Days 7, 8, 9 and 10 in diabetes-prone mice after treatment with 186-CTLA-4 Fc and CTLA4 186-GSG4S-Fc-(G4S)4-TACI 541 tested molecules.
  • FIG. 27 and FIGS. 28 A- 28 C depict results from an in vivo murine bm12 inducible SLE model tested with an exemplary CTLA4 186-GSG4S-Fc-(G4S)4-TACI 541 multi-domain molecule, and compared to a WT-TACI Fc only.
  • the BUN concentrations from serum collected at day 82 (study termination) are shown in FIG. 27 .
  • FIGS. 28 A- 28 C depicts levels of IgG2b ( FIG. 28 A ), IgG2c ( FIG. 28 B ), and IgG3 ( FIG. 28 C ) from serum collected from days 14, 42 and 82.
  • FIG. 29 depicts levels of anti-dsDNA antibody levels from serum collected in an in vivo murine bm12 inducible SLE model tested with an exemplary CTLA4 186-GSG4S-Fc-(G4S)4-TACI 541 multi-domain molecule.
  • immunomodulatory proteins that engage with one or more other immune receptor or ligand, e.g. on antigen-presenting cells or produced as soluble factors, to suppress or reduce B cell responses or activity and, in some cases, also T cell responses.
  • proteins that bind to BAFF or APRIL ligands to neutralize their activity and block or antagonize the activity of B cell stimulatory receptors such as TACI or BCMA.
  • the provided immunomodulatory proteins may be fusion proteins of a BCMA extracellular domain or binding portion thereof (hereinafter BCMA ECD) and a multimerization domain, such as an immunoglobulin Fc.
  • BCMA ECD BCMA extracellular domain or binding portion thereof
  • Fc immunoglobulin Fc
  • multi-domain immunomodulatory proteins also called “stack” immunomodulatory proteins
  • the immunomodulatory proteins provided herein can be used for the treatment of diseases, disorders or conditions that are associated with a dysregulated immune response, such as associated with inflammatory or autoimmune symptoms including an inflammatory or autoimmune disease.
  • the immune system relies on immune checkpoints 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.
  • autoimmunity i.e., self-tolerance
  • the immune system can become dysregulated and an abnormal immune response can be mounted against a normal body part or tissue, resulting in an autoimmune disease or condition or autoimmune symptoms.
  • an unwanted immune response can be mounted to a foreign tissue, such as a transplant, resulting in transplant rejection.
  • immunotherapy that alters immune cell activity can treat certain diseases, disorders and conditions in which the immune response is dysregulated.
  • inhibition or attenuation of an immune response such as a B cell response and/or T cell response
  • Therapeutic approaches that seek to modulate interactions between ligands and their receptors that mediate an immune response, including in the immune synapse are not entirely satisfactory.
  • therapies to intervene and alter the immunomodulatory effects of immune cell e.g. T cell or B cell, activation are constrained by the spatial orientation requirements as well as size limitations imposed by the confines of the immunological synapse.
  • existing therapeutic drugs may not be able to interact simultaneously with the multiple target proteins involved in modulating these interactions.
  • soluble receptors and antibodies generally bind competitively (e.g., to no more than one target species at a time) and therefore lack the ability to simultaneously bind multiple targets.
  • pharmacokinetic differences between drugs that independently target one of these receptors can create difficulties in properly maintaining a desired blood concentration of a drug combination targeting two different targets throughout the course of treatment.
  • BAFF and APRIL are TNF superfamily members that bind both TACI and BCMA on B cells; BAFF also binds a 3 rd receptor, BAFF-R. Together, BAFF and APRIL support B cell development, differentiation, and survival. Their co-neutralization dramatically reduces B cell function, including antibody production, whereas inhibition of either BAFF or APRIL alone mediates relatively modest effects. While CTLA-4-based therapeutics that block T cell costimulation provide safe and moderately effective T cell inhibition in many disease settings, and while B cell targeting therapies have demonstrated promising therapeutic potential, they are not entirely satisfactory.
  • the improved activity is mediated by increased or improved binding or interaction of the provided immunomodulatory proteins with BAFF and/or APRIL.
  • the provided immunomodulatory proteins include those that suppress BAFF and/or APRIL mediated activity either alone (e.g. BCMA-Fc), or coupled with inhibition of T cell costimulation.
  • multi-domain immunomodulatory protein of a B cell inhibitor molecule that is an extracellular domain portion that binds to BAFF and/or APRIL (e.g. a BCMA ECD), fused to a T cell inhibitory molecule (TIM) that is another domain that binds to a ligand of a T cell stimulatory receptor and/or to a T cell stimulatory receptor to antagonize or block T cell responses.
  • BIM B cell inhibitor molecule
  • TIM T cell inhibitory molecule
  • the provided immunomodulatory proteins provide for improved activity to modulate B cell responses alone, or together with modulation of T cell response.
  • the provided immunomodulatory proteins provide effective and durable disease suppression in the treatment of autoimmune or inflammatory diseases, including in severe B cell-related autoimmune diseases like SLE.
  • the provided embodiments are based on findings that directed evolution by affinity modification of TNFR domain (TD) of the ectodomain of certain molecules (e.g. BCMA) facilitated the development of molecules with improved affinity for APRIL and/or BAFF.
  • the affinity modification produces a variant BCMA that contains a variant TNFR domain (vTD). Fusion of such molecules with an immunoglobulin Fc results in immunomodulatory proteins that suppress B cell activity and response.
  • the provided embodiments also are based on findings that further including the TD domains, e.g.
  • wild-type (WT) TD or vTD as a multi-domain fusion with an immunoglobulin superfamily (IgSF) domain of a T cell inhibitiory molecule, such as a CTLA-4 extracellular domain, further potentiates immunosuppressive activity.
  • IgSF immunoglobulin superfamily
  • Such activity can be further improved by directed evolution by affinity modification of the IgSF domain of CTLA-4 to produce a variant IgSF domain (vIgD) to further enhance affinity for CD80 and/CD86 ligands, which are ligands of the CD28 co-stimulatory receptor and inhibitory CTLA-4 receptor.
  • affinity-modified as used in the context of a domain of a protein means a mammalian protein having an altered amino acid sequence in an extracellular domain or a specific binding portion thereof (relative to the corresponding wild-type parental or unmodified domain) such that it has an increased or decreased binding activity, such as binding affinity, to at least one of its binding partners (alternatively “counter-structures”) compared to the parental wild-type or unmodified (i.e., non-affinity modified domain) protein.
  • the affinity-modified 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 wild-type or unmodified domain.
  • An increase or decrease in binding activity can be determined using well known binding assays, including flow cytometry. Larsen et al., American Journal of Transplantation, Vol 5: 443-453 (2005). See also, Linsley et al., Immunity, 1: 7930801 (1994).
  • An increase in a protein's binding activity e.g.
  • affinity, to its binding partner(s) is to a value at least 10% greater than that of the wild-type 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 control value.
  • a decrease in a protein's binding activity, e.g. affinity, to at least one of its binding partner is to a value no greater than 90% of the control but no less than 10% of the wild-type 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 control value.
  • An affinity-modified protein is altered in primary amino acid sequence of the extracellular domain or a specific binding portion thereof by substitution, addition, or deletion of amino acid residues.
  • the term “affinity-modified” is not be construed as imposing any condition for any particular starting composition or method by which the affinity-modified protein was created.
  • an affinity-modified protein is not limited to wild-type protein domains that are then transformed to an affinity-modified domain by any particular process of affinity modification.
  • An affinity-modified domain polypeptide can, for example, be generated starting from wild-type mammalian domain sequence information, then modeled in silico for binding to its binding partner, and finally recombinantly or chemically synthesized to yield the affinity-modified domain composition of matter.
  • an affinity-modified domain can be created by site-directed mutagenesis of a wild-type domain.
  • affinity modified IgSF domain or an affinity modified TD 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.
  • affinity-modified IgSF domain refers to an affinity-modified domain of a member of the immunoglobulin superfamily (IgSF) protein having an altered amino acid sequence of an immunoglobulin domain (e.g. IgV) within the extracellular domain of the IgSF protein or a specific binding portion thereof (relative to the corresponding wild-type parental or unmodified domain) such that it has an increased or decreased binding activity, such as binding affinity, to at least one of its binding partners (alternatively “counter-structures”) compared to the parental wild-type or unmodified protein containing the non-affinity modified or unmodified IgSF domain.
  • IgSF immunoglobulin superfamily
  • affinity-modified TD domain refers to an affinity-modified domain of a member of the tumor necrosis receptor superfamily (TNFRSF) protein or a TNF ligand thereof having an altered amino acid sequence of a TNFR domain or of a TNF domain therein, respectively.
  • TNFRSF tumor necrosis receptor superfamily
  • an affinity-modified TD domain of a TNFRSF protein has an altered amino acid sequence of a TNFR domain composed of at least one cysteine rich domain (CRD) within the extracellular domain of the TNFRSF protein or a specific binding portion thereof (relative to the corresponding wild-type parental or unmodified domain) such that it has an increased or decreased binding activity, such as binding affinity, to at least one of its binding partners (alternatively “counter-structures”) compared to the parental wild-type or unmodified protein containing the non-affinity modified or unmodified TD domain.
  • CCD cysteine rich domain
  • B cell inhibitory molecule refers to a protein molecule that antagonizes or blocks the activity of a B cell stimulatory receptor.
  • the BIM can antagonize the activity of the B cell stimulatory receptor by binding directly to a cognate ligand of the B cell stimulatory receptor, thereby blocking or reducing the binding between the ligand and the B cell stimulatory receptor.
  • a BIM binds to APRIL and/or BAFF, which are ligands of the B cell stimulatory receptors B cell maturation antigen (BCMA), B cell activation factor receptor (BAFF-R), and transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI).
  • BCMA B cell maturation antigen
  • BAFF-R B cell activation factor receptor
  • TACI transmembrane activator and calcium modulator and cyclophilin ligand interactor
  • a BIM provided herein contains the extracellular domain or a portion thereof containing a TNF receptor super family domain (TD, e.g. CRD) of a B cell stimulatory receptor that binds to the cognate ligand APRIL and/or BAFF, and heterotrimers of APRIL and BAFF.
  • TD TNF receptor super family domain
  • a BIM includes the extracellular domain of TACI, or a portion of the extracellular domain of TACI containing a TD domain that binds to cognate ligands APRIL and/or BAFF, and heterotrimers of APRIL and BAFF.
  • a BIM includes the extracellular domain of BCMA, or a portion of the extracellular domain of BCMA containing a TD domain that binds to cognate ligands APRIL and/or BAFF, and heterotrimers of APRIL and BAFF.
  • a BIM also can include an affinity-modified variant of the extracellular domain or portion thereof of TACI or BCMA with one more amino acid modifications (e.g. amino acid substitutions) in the TD that increase binding affinity for the cognate ligand (e.g. APRIL and/or BAFF, and heterotrimers of APRIL and BAFF).
  • B cell stimulatory receptor refers to one or more of B cell maturation antigen (BCMA), B cell activation factor receptor (BAFF-R), and transmembrane activator and calcium modulatory and cyclophilin ligand interactor (TACI), which are related tumor necrosis factor (TNFR) superfamily receptors expressed on B cells. Engagement or ligation of these related receptors by their cognate ligands, BAFF and/or APRIL, or heterotrimers of APRIL and BAFF, regulate B cell homeostasis, including B cell survival, B cell maturation and differentiation and immunoglobulin class switching.
  • BCMA B cell maturation antigen
  • BAFF-R B cell activation factor receptor
  • TACI transmembrane activator and calcium modulatory and cyclophilin ligand interactor
  • a B cell stimulatory receptor generally contains an extracellular portion, a transmembrane domain and cytoplasmic region, in which the cytoplasmic region contains one or more TNF receptor associated factor (TRAF) binding sites.
  • TRAF TNF receptor associated factor
  • Recruitment of various TRAF molecules to the cytoplasmic domain can activate various transcription factors, such as NF- ⁇ B (e.g. NF- ⁇ B1 or NF- ⁇ B2), to mediate B cell signaling pathways regulating B cell homeostasis.
  • NF- ⁇ B e.g. NF- ⁇ B1 or NF- ⁇ B2
  • Binding refers to the participation of a molecule in any attractive interaction with another molecule, resulting in a stable association in which the two molecules are in close proximity to one another. Binding includes, but is not limited to, non-covalent bonds, covalent bonds (such as reversible and irreversible covalent bonds), and includes interactions between molecules such as, but not limited to, proteins, nucleic acids, carbohydrates, lipids, and small molecules, such as chemical compounds including drugs.
  • binding activity refer to characteristics of a molecule, e.g. a polypeptide, relating to whether or not, and how, it binds one or more binding partners.
  • a binding activity can include any measure of binding of one molecule for a binding partner. Binding activities include the ability to bind the binding partner(s), the affinity with which it binds to the binding partner (e.g. high affinity), the avidity with which it binds to the binding partner, the strength of the bond with the binding partner and/or specificity or selectivity for binding with the binding partner.
  • binding affinity means the specific binding affinity of a protein for its binding partner (i.e., its counter-structure) under specific binding conditions.
  • the binding affinity refers to the strength of the interaction between two or more molecules, such as binding partners, typically the strength of the noncovalent interactions between two binding partners.
  • An increase or attenuation in binding affinity of an affinity-modified domain, or an immunomodulatory protein containing an affinity-modified domain, to a binding partner is determined relative to the binding affinity of the unmodified domain (e.g., the native or wild-type IgSF domain or the native or wild-type TD domain).
  • binding affinity can be measured by flow cytometry, such as based on a Mean Fluorescence Intensity (MFI) in a flow binding assay.
  • MFI Mean Fluorescence Intensity
  • binding avidity means the specific binding avidity, of a protein for its binding partner (i.e., 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 a protein for its binding partner (i.e., its counter-structure). As such, avidity is distinct from affinity, which describes the strength of a single interaction.
  • biological half-life refers to the amount of time it takes for a substance, such as an immunomodulatory protein, to lose half of its pharmacologic or physiologic activity or concentration.
  • Biological half-life can be affected by elimination, excretion, degradation (e.g., enzymatic degradation/digestion) 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 a protein are known in the art and include, but are not limited to, multimerization domains (e.g.
  • Fc immunoglobulin 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), 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
  • cell surface counter-structure is a counter-structure (alternatively is a binding partner) expressed on a mammalian cell.
  • the cell surface binding partner is a transmembrane protein.
  • the cell surface binding partner is a receptor.
  • binding partner in reference to a protein, such as a receptor, soluble ligand, or to an extracellular domain or portion thereof or affinity-modified variant thereof, refers to at least one molecule (typically a native mammalian protein) to which the referenced protein specifically binds under specific binding conditions.
  • an affinity-modified domain, or an immunomodulatory protein containing an affinity-modified domain specifically binds to the binding partner of the corresponding domain of the native or wild-type protein but with increased or attenuated affinity.
  • a “cell surface binding partner” is a binding partner expressed on a mammalian cell. Typically, the cell surface binding partner is a transmembrane protein.
  • the cell surface binding partner is a receptor or a ligand of a receptor expressed on and by cells, such as mammalian cells, forming the immunological synapse, for example immune cells.
  • cis with reference to binding to cell surface molecules refers to binding to two or more different cell surface molecules, each of which is present on the surface of the same cell. In some embodiments, cis means that the two or more cell surface molecules are exclusively on one or exclusively the other (but not both) of the two mammalian cells forming the IS.
  • 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.
  • nucleotides or amino acid positions “correspond to” nucleotides or amino acid positions in a disclosed sequence 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.
  • aligning the sequences one skilled in the art can identify corresponding residues, for example, using conserved and identical amino acid residues as guides.
  • FIG. 17 A and FIG. 17 B exemplify identification of corresponding residues by aligning two sequences.
  • 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.
  • amino acids including to a specific sequence set forth as a SEQ ID NO used to describe domain organization (e.g. of an IgSF domain or a TD domain) are for illustrative purposes and are not meant to limit the scope of the embodiments provided. It is understood that polypeptides and the description of domains thereof are theoretically derived based on homology analysis and alignments with similar molecules. Also, in some cases, adjacent N- and/or C-terminal amino acids of a given domain (e.g. IgSF domain or TD) also can be included in a sequence, such as to ensure proper folding of the domain when expressed. Thus, the exact locus can vary, and is not necessarily the same for each protein.
  • a specific IgSF domain such as specific IgV domain or IgC domain
  • a specific TD domain such as specific CRD domain
  • ECD extracellular domain
  • a membrane protein such as a transmembrane protein
  • vesicular membrane e.g., the space outside of a cell
  • ECD ectodomain
  • a soluble immunomodulatory protein can contain ECD sequences of a membrane protein fused to another moiety, such as a multimerization domain, for example an Fc region.
  • Ectodomains often interact with specific ligands or specific cell surface receptors, such as via a binding domain that specifically binds to the ligand or cell surface receptor.
  • binding domains include immunoglobulin domains (IgD, also called an IgSF domain) or cysteine rich domains (CRDs).
  • Ectodomains of members of the immunoglobulin superfamily contain an IgD (e.g. IgV domain).
  • Ectodomains of members of the TNFR superfamily contain a TD domain (e.g. a CRD domain).
  • an ECD herein includes a full-length sequence of an ECD of a membrane protein as well as specific-binding fragments thereof containing an IgD or a CRD that bind to a ligand or cognate binding partner.
  • ⁇ ективное amount refers to a quantity and/or concentration of a therapeutic composition, such as containing an immunomodulatory protein or Fc fusion protein, 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 inhibition of disease progression as, for example, by ameliorating or eliminating symptoms and/or the cause of the disease.
  • a therapeutic composition such as containing an immunomodulatory protein or Fc fusion protein
  • An effective amount for treating a disease, condition or disorder may be an amount that relieves, lessens, or alleviates at least one symptom or biological response or effect associated with the disease, condition or disorder, prevents progression of the disease, condition or disorder, or improves physical functioning of the patient.
  • the effective amount is an effective dose or number of cells administered to a patient.
  • the patient is a human patient.
  • a fusion protein refers to a polypeptide encoded by a nucleic acid sequence containing a coding sequence for two or more proteins, in some cases 2, 3, 4, 5 or more protein, in which the coding sequences are in the same reading frame such that when the fusion construct is transcribed and translated in a host cell, the protein is produced containing the two or more proteins.
  • Each of the two or more proteins can be adjacent to another protein in the construct or separated by a linker polypeptide that contains, 1, 2, 3, or more, but typically fewer than 20, 15, 10, 9, 8, 7, or 6 amino acids.
  • the protein product encoded by a fusion construct is referred to as a fusion polypeptide.
  • a fusion protein in accord with the provided embodiments is an Fc fusion protein containing an affinity-modified domain (e.g. a variant of a BCMA or portion thereof containing a CRD) that is linked to an immunoglobulin Fc domain.
  • an affinity-modified domain e.g. a variant of a BCMA or portion thereof containing a CRD
  • 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 about 1.2-fold, about 1.5-fold, about 2.0-fold, about 3.0-fold, about 4.0-fold, about 5.0-fold, or about 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).
  • an Fc (fragment crystallizable) region or domain of an immunoglobulin molecule corresponds largely to the constant region of the immunoglobulin heavy chain, and which, in some cases, 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. In some cases for inclusion in a provided fusion protein, all or a portion of the Fc hinge sequence may be deleted.
  • 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.
  • 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 operably linked to an Fc region of an immunoglobulin.
  • An Fc-fusion may comprise, for example, an Fc region operably linked to a TIM or BIM of the provided immunomodulatory proteins.
  • An Fc-fusion may comprise, for example, an Fc region operably linked to a BCMA extracellular domain or portion thereof containing a CRD, including any of the provided affinity-modified variants thereof.
  • An immunoglobulin Fc region may be linked indirectly or directly to the one or more polypeptides.
  • 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.
  • Fc fusion of non-identical species e.g. knob into hole engineering
  • the Fc is a mammalian Fc such as a murine or human Fc.
  • host cell refers to any 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.
  • Examples of host cells 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.
  • immunosynapse or “immune synapse” (abbreviated “IS”) 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
  • NK Natural Killer
  • immunoglobulin as used herein is synonymous with the term “antibody” (abbreviated “Ab”) and refers to a mammalian immunoglobulin protein including any of the five human classes: IgA (which includes subclasses IgA1 and IgA2), IgD, IgE, IgG (which includes subclasses IgG1, IgG2, IgG3, and IgG4), and IgM.
  • the term is also inclusive of immunoglobulins that are less than full-length, whether wholly or partially synthetic (e.g., recombinant or chemical synthesis) or naturally produced, including any fragment thereof containing at least a portion of the variable heavy (VH) chain and/or variable light (VL) chain region of the immunoglobulin molecule that is sufficient to form an antigen binding site and, when assembled, to specifically bind antigen.
  • the antibody also can include all or a portion of the constant region.
  • Such fragments include antigen binding fragment (Fab), variable fragment (Fv) containing VH and VL, the single chain variable fragment (scFv) containing VH and VL 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
  • dsFv diabody Fc
  • Fd polypeptide fragments fragments include full-length antibody and antigen-binding fragments.
  • the term antibody also includes antibody compositions with polyepitopic specificity, multispecific antibodies (e.g., bispecific antibodies), diabodies, and single-chain molecules. Bispecific antibodies, homobispecific and heterobispecific, are included within the meaning of the term.
  • Antibodies include polyclonal antibodies or monoclonal antibodies. Antibody also includes synthetic antibodies or recombinantly produced antibodies. For the structure and properties of the different classes of antibodies, see e.g., Basic and Clinical Immunology, 8th Edition, Daniel P. Sties, Abba I. Terr and Tristram G. Parsolw (eds), Appleton & Lange, Norwalk, Conn., 1994, page 71 and Chapter 6.
  • full-length antibody is an antibody typically having two full-length heavy chains (e.g., VH-CH1-CH2-CH3 or VH-CH1-CH2-CH3-CH4) and two full-length light chains (VL-CL) and hinge regions, such as antibodies produced from mammalian species (e.g. human, mouse, rat, rabbit, non-human primate, etc.) by antibody secreting B cells and antibodies with the same domains that are produced synthetically.
  • whole antibodies include those with heavy and light chains including an Fc region.
  • the constant domains may be native sequence constant domains (e.g., human native sequence constant domains) or amino acid sequence variants thereof.
  • the intact antibody may have one or more effector functions.
  • an “antibody fragment” comprises a portion of an intact antibody, the antigen binding and/or the variable region of the intact antibody.
  • Antibody fragments include, but are not limited to, Fab fragments, Fab′ fragments, F(ab′) 2 fragments, Fv fragments, disulfide-linked Fvs (dsFv), Fd fragments, Fd′ fragments; diabodies; linear antibodies (see U.S. Pat. No. 5,641,870, Example 2; Zapata et al., Protein Eng.
  • single-chain antibody molecules including single-chain Fvs (scFv) or single-chain Fabs (scFab); antigen-binding fragments of any of the above and multispecific antibodies from antibody fragments.
  • scFv single-chain Fvs
  • scFab single-chain Fabs
  • “Fv” is composed of one heavy- and one light-chain variable region domain linked by non-covalent association. From the folding of these two domains emanate six complementarity determining regions (CDR) (3 in each from the heavy and light chain) that contribute the amino acid residues for antigen binding and confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although, in some cases, at a lower affinity than the entire binding site.
  • CDR complementarity determining regions
  • dsFv refers to an Fv with an engineered intermolecular disulfide bond, which stabilizes the V H -V L pair.
  • An “Fd fragment” is a fragment of an antibody containing a variable domain (V H ) and one constant region domain (C H 1) of an antibody heavy chain.
  • a “Fab fragment” is an antibody fragment that results from digestion of a full-length immunoglobulin with papain, or a fragment having the same structure that is produced synthetically, e.g., by recombinant methods.
  • a Fab fragment contains a light chain (containing a V L and C L ) and another chain containing a variable domain of a heavy chain (V H ) and one constant region domain of the heavy chain (C H 1).
  • a “F(ab′) 2 fragment” is an antibody fragment that results from digestion of an immunoglobulin with pepsin at pH 4.0-4.5, or a fragment having the same structure that is produced synthetically, e.g., by recombinant methods.
  • the F(ab′) 2 fragment essentially contains two Fab fragments where each heavy chain portion contains an additional few amino acids, including cysteine residues that form disulfide linkages joining the two fragments.
  • a “Fab′ fragment” is a fragment containing one half (one heavy chain and one light chain) of the F(ab′) 2 fragment.
  • Fd′ fragment is a fragment of an antibody containing one heavy chain portion of a F(ab′) 2 fragment.
  • Fv′ fragment is a fragment containing only the V H and V L domains of an antibody molecule.
  • an “scFv fragment” refers to an antibody fragment that contains a variable light chain (V L ) and variable heavy chain (V H ), covalently connected by a polypeptide linker in any order.
  • the linker is of a length such that the two variable domains are bridged without substantial interference.
  • Exemplary linkers are (Gly-Ser) n residues with some Glu or Lys residues dispersed throughout to increase solubility.
  • Diabodies are dimeric scFv; diabodies typically have shorter peptide linkers than scFvs, and preferentially dimerize.
  • 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. Many “non-antibody IgSF” members include cell surface proteins or receptors that are not antibodies. 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.
  • IgSF immunological synapse
  • 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 include from 4 to 30 IgSF members.
  • IgSF domain or “immunoglobulin domain” or “Ig domain” or “IgD” as used herein refers to a structural domain or domains 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.
  • one end of the Ig domain has a section called the complementarity determining region, which, in some aspects, is involved in 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.
  • a “non-antibody IgSF domain” refers to IgSF domain or domains present in proteins other than antibodies, which typically are present in the extracellular portion or domain of certain cell surface proteins.
  • the extracellular domain (ECD) of IgSF family members contains one or more Ig domains; hence, the term Ig domain is also used with reference to the ECD of such protein molecules.
  • Reference to a variant IgSF domain (vIgD) refers to a variant or modified sequence of an IgD.
  • immunological activity refers to one or more activities of immune cells, such as T cells or B cells, including, for example, activation, cell survival, cell proliferation, cytokine production (e.g. interferon-gamma), cytotoxicity activity, or ability to activate NF- ⁇ B pathway or other signaling cascade leading to activation of a transcription factor in the immune cell.
  • Assays to assess immunological activity of immunomodulatory proteins can be compared to control proteins with a known activity.
  • an “immunomodulatory protein” or “immunomodulatory polypeptide” is a protein that modulates immunological activity.
  • modulation or “modulating” an immune response is meant that immunological activity is either enhanced or suppressed.
  • Such modulation includes any induction, or alteration in degree or extent, or suppression of immunological activity of an immune cell, such as a B cell or a T cell.
  • soluble Fc fusion proteins herein may suppress immunological activity of either B cells, T cells or both B cells and T cells.
  • 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.
  • Multimeric proteins can be homomultimeric (of identical polypeptide chains) or heteromultimeric (of different polypeptide chains).
  • modification is in reference to modification of a sequence of amino acids of a polypeptide or a sequence of nucleotides in a nucleic acid molecule and includes a change in amino acids or nucleotides, respectively, of the sequence.
  • the amino acid modification or change may be a deletion, insertion, or replacement (substitution) of amino acids or nucleotides, respectively.
  • Methods of modifying a polypeptide are routine to those of skill in the art, such as by using recombinant DNA methodologies.
  • a “multimerization domain” refers to a sequence of amino acids that promotes the formation of a multimer of two or more polypeptides.
  • a multimerization domain includes sequences that promote 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.
  • Exemplary 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.
  • nucleic acid residues e.g., deoxyribonucleotides or ribonucleotides
  • 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.
  • a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions) and complementary nucleotide sequences as well as the sequence explicitly indicated.
  • degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues.
  • nucleic acid or polynucleotide encompasses cDNA or mRNA encoded by a gene.
  • operable combination refers to the linkage of nucleic acid sequences in such a manner or orientation that the segments are arranged so that they function in concert for their intended purposes.
  • the term refers to linkage of nucleic acids to produce a nucleic acid molecule capable of directing the transcription of a given gene and/or to produce a desired protein molecule that is functional.
  • segments of a DNA sequence e.g. a coding sequence and a regulatory sequence(s) are linked in such a way as to permit gene expression when the appropriate molecules (e.g. transcriptional activator proteins) are bound to the regulatory sequence.
  • 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 protein) and a carrier, excipient, or diluent.
  • the 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.
  • proteins can be derivatized as described herein by well-known organic chemistry techniques.
  • purified as applied to nucleic acids, such as encoding immunomodulatory proteins, or proteins (e.g. immunomodulatory proteins) 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 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 (e.g., an immunomodulatory protein) 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 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.
  • recombinant expression vector refers to a DNA molecule containing a desired coding sequence (e.g., encoding an immunomodulatory protein) and appropriate nucleic acid sequences necessary for the expression of an operably linked coding sequence in a particular 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 so that the expressed protein can be secreted by the recombinant host cell, such as for its expression as a secretable protein or for more facile isolation or purification of the immunomodulatory 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.
  • 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.
  • a percent sequence identity can be determined as the percentage of amino acid residues (or nucleotide residues) in a candidate sequence that are identical with the amino acid residues (or nucleotide residues) in a reference sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity.
  • Reference to sequence identity includes sequence identity across the full length of each of the sequences being compared. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • soluble as used herein in reference to proteins means that the protein is not a membrane protein or is not anchored in a cell membrane.
  • a protein can be constructed as a soluble protein by inclusion of only an extracellular domain or a portion thereof and without a transmembrane domain.
  • solubility of a protein can be improved by linkage or attachment, directly or indirectly via a linker, to an Fc domain or other half-life extending molecule, which, in some cases, also can improve the stability and/or half-life of the protein.
  • a soluble protein is an Fc fusion protein.
  • 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 10 times as great, but optionally 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 more than one target molecule. In some cases, a specifically binding protein may bind to a protein that has similarity in structural conformation with the target protein (e.g., paralogs or orthologs).
  • an immunomodulatory protein of the invention may specifically bind to more than one distinct species of target molecule due to cross-reactivity.
  • Solid-phase ELISA immunoassays ForteBio Octet or Biacore measurements can be used to determine specific binding between two proteins.
  • interactions between two binding proteins have dissociation constants (Kd) less than about 1 ⁇ 10 ⁇ 5 M, and often as low as about 1 ⁇ 10 ⁇ 12 M.
  • dissociation constants Kd
  • interactions between two binding proteins have dissociation constants of less than about 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 less.
  • specific binding fragment or “fragment” as used herein in reference to a protein means a polypeptide that is shorter than a full-length protein or a specific domain or region thereof and that specifically binds in vitro and/or in vivo to a binding partner of the full-length protein or of the specific domain or region.
  • a specific finding fragment is in reference to a fragment of a full length extracellular domain of a polypeptide or a binding domain of a polypeptide, but that still binds to a binding partner of the binding domain.
  • a specific binding fragment is in reference to a fragment of a full-length extracellular domain of an IgSF family member or a full-length IgSF domain thereof (e.g.
  • a specific binding fragment is in reference to a fragment of an extracellular domain of a full-length TNFR family member or a full-length TNFR domain (TD) thereof (e.g. CRD), but that still binds to a binding partner of the TNFR family member or of a CRD of an TNFR family member.
  • TD full-length TNFR domain
  • the specific binding fragment is at least about 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% the sequence length of the full-length sequence of the extracellular domain or of a domain or region of the extracellular domain.
  • 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.
  • a “subject” is a mammal, such as a human or other animal, and typically is human.
  • the subject can be male or female and can be any suitable age, including infant, juvenile, adolescent, adult, and geriatric subjects.
  • 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.
  • TNFRSF TNFRSF
  • CRD cysteine rich domains
  • Molecules are categorized as members of this superfamily based on the shared structural features that include the one or more cysteine rich domain (CRD) present in their N-terminal extracellular region, which often play a role in protein binding of their cognate binding partner or ligand.
  • a TNFRSF protein may have only one or several CRDs (e.g. CRD1, CRD2, etc.).
  • ECD or ectodomain of TNFRSF members contain between 1 and 6 pseudorepeats of CRDs.
  • BAFF-receptor and BCMA each contain one CRD while TACI contains two CRDs (CRD1 and CRD2).
  • TNFRSF members are usually trimeric or multimeric complexes that are stabilized by their intracysteine disulfide bonds. Binding of TNFRSF proteins to their ligands facilitates various biological activities in cells, such as the induction of apoptotic cell death or cell survival and proliferation.
  • TD refers to a structural domain or domains of TNFRSF proteins or of TNF family ligands.
  • a TD of a TNFRSF protein is a cysteine-rich domain (CRD) module of about 40 amino acids containing six (6) conserved cysteines.
  • CRD cysteine-rich domain
  • the six cysteines are involved in formation of intrachain disulphide bonds.
  • the extracellular domain (ECD) of TNFRSF members contains one or more CRD domains; hence, the term TD is also used with reference to the ECD of such protein molecules.
  • Reference to a variant TD refers to a variant or modified sequence of a TD.
  • T cell inhibitory molecule refers to a protein molecule that antagonizes or blocks the activity of a T cell stimulatory receptor.
  • the TIM can antagonize the activity of the T cell stimulatory receptor by binding directly to the T cell stimulatory receptor or a ligand of the T cell stimulatory receptor, thereby blocking or reducing the binding between the ligand and the T cell stimulatory receptor.
  • a TIM antagonizes or inhibits activity of a T cell costimulatory receptor, such as CD28.
  • a TIM provided herein contains the extracellular domain or a portion thereof containing an immunoglobulin superfamily (IgSF) domain, such as an IgV domain, of a cognate ligand of a T cell stimulatory receptor.
  • IgSF immunoglobulin superfamily
  • a TIM includes the extracellular domain of CTLA-4, or a portion of the extracellular domain of CTLA-4 containing an IgSF domain (e.g. IgV domain) that binds to a T cell stimulatory receptor (e.g. CD28).
  • a TIM also can include an affinity-modified variant of the extracellular domain or portion thereof of a cognate ligand of the T cell stimulatory receptor, e.g. CTLA-4, with one more amino acid modifications (e.g. amino acid substitutions) in the IgSF domain that increase binding affinity for the T cell stimulatory receptor (e.g. CD28).
  • a “T cell stimulatory receptor” refers to a cell surface molecule expressed on a T cell in which engagement or ligation of the molecule results in the direct or indirect activation of one or more tyrosine kinases in the cell and/or culminates in the induction or potentiation of one or more effector cell functions the T cell in which it is expressed.
  • a T cell stimulatory receptor generally contains an extracellular portion, a transmembrane domain and cytoplasmic region.
  • the cytoplasmic region contains an intracellular signaling domain that contains an immunoreceptor tyrosine-based activation motif (ITAM; defined by the sequence YXX(L/I)X6-8YXX(L/I)) or that otherwise is capable of interacting with or associating with one or more accessory proteins, such as one or more adaptor proteins, involved in or regulating tyrosine phosphorylation in a signal transduction pathway.
  • ITAM immunoreceptor tyrosine-based activation motif
  • a T cell stimulatory receptor interacts with or associates with an adaptor protein that contains an ITAM or an adaptor protein that contains one or more protein-binding domains, such as e.g., Src homology 2 (SH2) and SH3 domains, that bind specific amino acid sequences, e.g phosphotyrosine residues, within a protein in a signal transduction pathway.
  • an adaptor protein that contains an ITAM or an adaptor protein that contains one or more protein-binding domains, such as e.g., Src homology 2 (SH2) and SH3 domains, that bind specific amino acid sequences, e.g phosphotyrosine residues, within a protein in a signal transduction pathway.
  • adaptor proteins include, but are not limited to, Lck, Fyn, ZAP70, SLP76, PI3K, Grb2, PKC ⁇ and SHC1.
  • the T cell stimulatory receptor itself need not possess intrinsic enzymatic activity but may indirectly mediate enzymatic activities
  • T cell stimulatory receptor typically includes a T cell receptor (TCR), CD3, CD4, CD8, CD28, ICOS, or CD2.
  • T cell stimulatory receptor is a costimulatory receptor, such as CD28.
  • trans with reference to binding to cell surface molecules refers to binding to two different cell surface molecules, each of which is present on the surface of a different cell.
  • trans means that with respect to two different cell surface molecules, the first is exclusively present on one of the two mammalian cells forming the IS and the second is present exclusively on the second of the two mammalian cells forming the IS.
  • 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.
  • 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 an immunomodulatory protein or engineered cells of the present invention either alone or in combination with another compound as described herein.
  • Treating also means a decrease in the severity of symptoms in an acute or chronic disease, condition or disorder or a decrease in the relapse rate as for example in the case of a relapsing or remitting autoimmune disease course or inflammatory condition or a decrease in inflammation in the case of an inflammatory aspect of an autoimmune disease or inflammatory condition.
  • Preventing,” “prophylaxis,” or “prevention” of a disease or disorder as used in the context of this invention refers to the administration of an immunomodulatory protein, 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, condition or disorder or to lessen the likelihood of the onset of a disease, condition or disorder.
  • variant also “modified” or mutant,” which can be used interchangeably
  • a variant protein or polypeptide means a protein, such as a mammalian (e.g., human or murine) protein created by human intervention.
  • the variant is a polypeptide having an altered or modified amino acid sequence, such as by one or more amino acid substitutions, deletions, additions or combinations thereof, relative to an unmodified or wild-type protein or to a domain thereof.
  • a variant polypeptide 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 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 form of a wild-type or unmodified protein, such as a mature sequence thereof (lacking the signal sequence) or a portion thereof containing the extracellular domain or an binding domain thereof.
  • Non-naturally occurring amino acids as well as naturally occurring amino acids are included within the scope of permissible substitutions or additions.
  • a variant protein is not limited to any particular method of making and includes, for example, chemical synthesis, recombinant DNA techniques, or combinations thereof.
  • a variant protein of the invention specifically binds to at least one or more binding partners.
  • the altered amino acid sequence results in an altered (i.e., increased or decreased) binding activity, such as binding affinity or avidity, to the one or more binding partners.
  • a variant protein may thus be an “affinity-modified” protein as described herein.
  • wild-type or “natural” or “native,” which are used interchangeably, as used herein is used in connection with biological materials such as nucleic acid molecules, proteins, host cells, and the like, that are found in nature and not modified by human intervention.
  • BCMA immunomodulatory proteins that contain a portion of the extracellular domain (ECD) of the BCMA receptor, or a variant thereof, that bind to at least one BCMA cognate binding partner.
  • variant BCMA polypeptides that exhibit altered (e.g. increased) binding activity or affinity for one or more of a BCMA cognate binding partner.
  • the BCMA cognate binding partner is one or more of BAFF or APRIL or is a BAFF/APRIL heterotrimer.
  • the provided BCMA immunomodulatory proteins and polypeptides include soluble fusion proteins thereof in which the BCMA portion of the extracellular domain or variant thereof is linked to another moiety, such as an immunoglobulin Fc or other multimerization domain or half-life extending moiety.
  • the immunomodulatory protein is a BCMA-Fc fusion protein.
  • a BCMA-Fc fusion protein containing (1) a BCMA polypeptide composed of the extracellular domain of the BCMA receptor or a portion thereof, or a variant BCMA polypeptide thereof, that binds to at least one BCMA cognate binding partner, and (2) an Fc domain.
  • the BCMA polypeptide or variant BCMA polypeptide can be linked directly or indirectly (e.g. via a peptide linker) to the Fc domain.
  • BCMA is a tumor necrosis factor receptor family member characterized by having an extracellular domain (ECD) containing cysteine-rich pseudo-repeat domain (CRD).
  • ECD extracellular domain
  • CRD cysteine-rich pseudo-repeat domain
  • BCMA is a membrane bound receptor, which has an extracellular domain containing a single CRD, a transmembrane domain and a cytoplasmic domain that contains TRAF-binding sites for binding to TRAF signaling molecules.
  • BCMA binds to cognate ligands APRIL and BAFF, although binding to BAFF is with weaker affinity. It is reported that BCMA binds to BAFF with two to three orders of magnitude weaker binding than binding between BAFF and its other cognate receptors BAFF-R and TACI (Bossen and Schneider et al. 2006 Seminars in Immunology, 18:263-75).
  • the amino acid sequence of the full length BCMA is set forth in SEQ ID NO:667.
  • the protein is a type II membrane protein and lacks a signal peptide; following expression in eukaryotic cells the N-terminal methionine is removed.
  • a mature BCMA protein does not contain the N-terminal methionine as set forth in SEQ ID NO:667.
  • the extracellular domain of BCMA (amino acid residues 1-54 of SEQ ID NO:667; ECD set forth in SEQ ID NO:710) contains one cysteine rich domain (CRD, hereinafter also called a tumor necrosis family receptor domain or TD), which exhibits affinity for binding to APRIL and to a lesser extent BAFF.
  • the CRD contains amino acid residues 7-41 of the sequence set forth in SEQ ID NO:710.
  • the variant BCMA polypeptides provided herein contain one or more amino acid modifications, such as one or more substitutions (alternatively, “mutations” or “replacements”), deletions or additions in the extracellular domain of a reference BCMA polypeptide, such as a wild-type or unmodified BCMA polypeptide containing a CRD (hereinafter also called TD).
  • a provided variant BCMA polypeptide is or comprises a variant TD (“vTD”) in which the one or more amino acid modifications (e.g. substitutions) is in the CRD.
  • the reference (e.g. unmodified) BCMA sequence is a wild-type BCMA sequence or is a portion thereof that contains the CRD.
  • the reference (e.g., unmodified) BCMA is or comprises the extracellular domain (ECD) of BCMA or a portion thereof containing the CRD.
  • the variant BCMA polypeptide comprises or consists essentially of the CRD or a specific binding fragment thereof.
  • the variant BCMA is a soluble polypeptide and lacks a transmembrane domain.
  • the variant BCMA polypeptide further comprises a transmembrane domain and, in some cases, also a cytoplasmic domain.
  • the reference (e.g., unmodified) BCMA sequence is a mammalian BCMA sequence.
  • the reference (e.g., unmodified) BCMA sequence can be a mammalian BCMA that includes, but is not limited to, human, mouse, cynomolgus monkey, or rat.
  • the reference (e.g., unmodified) BCMA sequence is human.
  • the extracellular domain of an exemplary human BCMA sequence is set forth in SEQ ID NO:710.
  • the reference (e.g., unmodified) BCMA sequence has (i) the sequence of amino acids set forth in SEQ ID NO:710 or a sequence thereof that lacks the N-terminal methionine, (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:710 and that binds to APRIL or BAFF, or (iii) is a fragment or portion of (i) or (ii) containing a CRD, in which the portion binds to APRIL or BAFF.
  • the reference (e.g., unmodified) BCMA sequence lacks the N-terminal methionine as set forth in SEQ ID NO: 710.
  • BCMA Extracellular Domain SEQ ID NO: 710 MLQMAGQCSQNEYFDSLLHACIPCQLRCSSNTPPLTCQRYCNASVTNSV KGTNA
  • the reference (e.g., unmodified) BCMA sequence lacks the N-terminal methionine as set forth in SEQ ID NO: 710.
  • the reference (e.g., unmodified) BCMA sequence has (i) the sequence of amino acids set forth in SEQ ID NO:356, (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:356 and that binds to APRIL or BAFF, or (iii) is a fragment or portion of (i) or (ii) containing a CRD, in which the portion binds to APRIL or BAFF.
  • BCMA Extracellular Domain SEQ ID NO: 356 LQMAGQCSQNEYFDSLLHACIPCQLRCSSNTPPLTCQRYCNASVTNSVK GTNA
  • variant BCMA polypeptides are variant BCMA polypeptides.
  • immunomodulatory proteins such as BCMA-Fc fusion proteins, that contain a provided variant BCMA polypeptide.
  • the variant BCMA sequence has the sequence of the reference (e.g. unmodified) BCMA sequence, such as any described above, but additionally contains one more amino acid modifications, such as one or more amino acid substitutions.
  • variant BCMA polypeptides containing at least one affinity-modified TD domain (CRD) or a specific binding fragment thereof that contains one or more amino acid substitutions in a TD domain of a reference (e.g., unmodified or wild-type) BCMA polypeptide, such that the variant BCMA polypeptide exhibits altered (e.g. increased) binding activity or affinity for one or both of APRIL or BAFF compared to the reference (e.g., unmodified or wild-type) BCMA polypeptide.
  • CCD affinity-modified TD domain
  • a specific binding fragment thereof that contains one or more amino acid substitutions in a TD domain of a reference (e.g., unmodified or wild-type) BCMA polypeptide
  • a variant BCMA polypeptide has a binding affinity for APRIL and/or BAFF that differs from that of a reference (e.g., unmodified or wild-type) BCMA polypeptide control sequence as determined by, for example, solid-phase ELISA immunoassays, flow cytometry or Biacore assays. Binding affinities for each of the cognate binding partners are independent; that is, in some embodiments, a variant BCMA polypeptide has an increased binding affinity for one or both APRIL and BAFF, and a decreased or unchanged binding affinity for the other of APRIL or BAFF, relative to a reference (e.g., unmodified or wild-type) BCMA polypeptide.
  • a reference e.g., unmodified or wild-type
  • the variant BCMA polypeptide has an increased binding affinity for BAFF, relative to the reference (unmodified or wild-type) BCMA polypeptide. In some embodiments, the variant BCMA polypeptide has an increased binding affinity for APRIL relative to the reference (unmodified or wild-type) BCMA polypeptide. In some embodiments, the variant BCMA polypeptide has an increased binding affinity for APRIL and BAFF relative to the reference (unmodified or wild-type) BCMA polypeptide.
  • the cognate ligands BAFFF and/or APRIL can be a mammalian protein, such as a human protein or a murine protein.
  • a variant BCMA polypeptide with increased or greater binding affinity to APRIL and/or BAFF will have an increase in binding affinity relative to the reference (e.g., unmodified or wild-type) BCMA polypeptide control of at least about 5%, such as at least about 10%, 15%, 20%, 25%, 35%, or 50%.
  • the increase in binding affinity relative to the reference (e.g., unmodified or wild-type) BCMA 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) BCMA polypeptide has the same sequence as the variant BCMA 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 BAFF 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.
  • the K d of any of the foregoing embodiments to BAFF is less than at or about 1 ⁇ 10 ⁇ 9 M, 1 ⁇ 10 ⁇ 10 M or 1 ⁇ 10 ⁇ 11 M, or 1 ⁇ 10 ⁇ 12 M.
  • the K d of any of the foregoing embodiments to BAFF is between 1 ⁇ 10 ⁇ 9 M and at or about 1 ⁇ 10 ⁇ 12 M. In some embodiments, the K d of any of the foregoing embodiments to BAFF is at or about 1 ⁇ 10 ⁇ 9 M, at or about 2 ⁇ 10 ⁇ 9 M, at or about 4 ⁇ 10 ⁇ 9 M, at or about 6 ⁇ 10 ⁇ 9 M, at or about 8 ⁇ 10 ⁇ 9 M, at or about 1 ⁇ 10 ⁇ 10 M, at or about 2 ⁇ 10 ⁇ 10 M, at or about 4 ⁇ 10 ⁇ 10 M, at or about 6 ⁇ 10 ⁇ 10 M, at or about 8 ⁇ 10 ⁇ 10 M, at or about 1 ⁇ 10 ⁇ 11 M, at or about 2 ⁇ 10 ⁇ 11 M, at or about 4 ⁇ 10 ⁇ 11 M, at or about 6 ⁇ 10 ⁇ 11 M, at or about 8 ⁇ 10 ⁇ 11 M, or at or about 1 ⁇ 10 ⁇ 11 M, or any value between any of the foregoing.
  • a provided embodiment includes a variant BCMA polypeptide as described above and the K d to BAFF is decreased (higher binding affinity) by greater than or greater than about 1.5-fold, such as greater than or about 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold or more.
  • the equilibrium dissociation constant (K d ) of any of the foregoing embodiments to APRIL 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.
  • the K d of any of the foregoing embodiments to APRIL is less than at or about 1 ⁇ 10 ⁇ 9 M, 1 ⁇ 10 ⁇ 10 M or 1 ⁇ 10 ⁇ 11 M, or 1 ⁇ 10 ⁇ 12 M.
  • the K d of any of the foregoing embodiments to APRIL is between 1 ⁇ 10 ⁇ 9 M and at or about 1 ⁇ 10 ⁇ 12 M. In some embodiments, the K d of any of the foregoing embodiments to APRIL is at or about 1 ⁇ 10 ⁇ 9 M, at or about 2 ⁇ 10 ⁇ 9 M, at or about 4 ⁇ 10 ⁇ 9 M, at or about 6 ⁇ 10 ⁇ 9 M, at or about 8 ⁇ 10 ⁇ 9 M, at or about 1 ⁇ 10 ⁇ 10 M, at or about 2 ⁇ 10 ⁇ 10 M, at or about 4 ⁇ 10 ⁇ 10 M, at or about 6 ⁇ 10 ⁇ 10 M, at or about 8 ⁇ 10 ⁇ 10 M, at or about 1 ⁇ 10 ⁇ 11 M, at or about 2 ⁇ 10 ⁇ 12 M, at or about 4 ⁇ 10 ⁇ 11 M, at or about 6 ⁇ 10 ⁇ 11 M, at or about 8 ⁇ 10 ⁇ 11 M, or at or about 1 ⁇ 10 ⁇ 12 M, or any value between any of the foregoing.
  • a provided embodiment includes a variant BCMA polypeptide as described above and the K d to APRIL is decreased (higher binding affinity) by greater than or greater than about 1.5-fold, such as greater than or about 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold or more.
  • the reference (e.g., unmodified or wild-type) BCMA sequence does not necessarily have to be used as a starting composition to generate variant BCMA polypeptides described herein. Therefore, use of the term “modification”, such as “substitution” does not imply that the present embodiments are limited to a particular method of making variant BCMA polypeptides or immunomodulatory proteins containing the same.
  • Variant BCMA 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 BCMA polypeptides are designed or created is not limited to any particular method.
  • a reference (e.g., unmodified or wild-type) BCMA encoding nucleic acid is mutagenized from reference (e.g., unmodified or wild-type) BCMA genetic material and screened for desired specific binding affinity or other functional activity.
  • a variant BCMA 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 (s) in a variant BCMA polypeptide are designated by amino acid position number corresponding to the numbering of positions of the reference ECD sequence set forth in SEQ ID NO:710. It is within the level of a skilled artisan to identify the corresponding position of a modification, e.g. amino acid substitution, in an BCMA polypeptide, including portion thereof containing TD (CRD) thereof, such as by alignment of a reference sequence (e.g. SEQ ID NO:356) with SEQ ID NO:710. An alignment identifying corresponding residues is exemplified in FIG. 17 B . 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 BCMA polypeptide has one or more amino acid modification, e.g. substitution in a reference (e.g., unmodified or wild-type) BCMA sequence, such as any as described.
  • 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) BCMA sequence.
  • the one or more amino acid modification, e.g. substitution is in the CRD domain or specific binding fragment thereof.
  • the variant BCMA 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 CRD.
  • the variant BCMA 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 CRD or specific binding fragment thereof.
  • the variant BCMA polypeptide containing the one or more amino acid modifications e.g.
  • the variant BCMA polypeptide containing the one or more amino acid modifications e.g.
  • the variant BCMA polypeptide containing the one or more amino acid modifications as described has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 710.
  • the variant BCMA polypeptide containing the one or more amino acid modifications as described has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 356.
  • the variant BCMA polypeptide has one or more amino acid modification, e.g. substitution in a reference BCMA polypeptide or specific binding fragment there of corresponding to position(s) 9, 10, 11, 14, 16, 19, 20, 22, 25, 27, 29, 30, 31, 32, 35, 36, 39, 43, 45, 46, 47 or 48 with reference to numbering of SEQ ID NO:710.
  • the variant BCMA polypeptide has one or more amino acid modification, e.g.
  • 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 BCMA polypeptide includes at least one amino acid substitution at position 9. In some embodiment, the at least one amino acid substitution is S9G, S9N, S9Y.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 10. In some embodiment, the at least one amino acid substitution is Q10E, Q10P.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 11. In some embodiment, the at least one amino acid substitution is N11D, N11S.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 19.
  • the at least one amino acid substitution is H19A, H19C, H19D, H19E, H19F, H19G, H19I, H19K, H19L, H19M, H19N, H19P, H19Q, H19R, H19S, H19T, H19V, H19W, H19Y.
  • the at least one amino acid substitution is H19L.
  • the at least one amino acid substitution is H19K.
  • the at least one amino acid substitution is H19Q.
  • the at least one amino acid substitution is H19R.
  • the at least one amino acid substitution is H19Y.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 22.
  • the at least one amino acid substitution is I22L, I22V.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 25.
  • the at least one amino acid substitution is Q25E, Q25F, Q25G, Q25H, Q25I, Q25K, Q25L, Q25M, Q25S, Q25V, Q25Y.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 27.
  • the at least one amino acid substitution is R27H, R27L.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 30.
  • the at least one amino acid substitution is S30G, S30Y.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 31.
  • the at least one amino acid substitution is N31D, N31G, N31H, N31K, N31L, N31M, N31P, N31S, N31V, N31Y.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 32.
  • the at least one amino acid substitution is T32I, T32S.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 35.
  • the at least one amino acid substitution is L35A, L35M, L35P, L35S, L35V, L35Y.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 36.
  • the at least one amino acid substitution is T36A, T36G, T36N, T36M, T36S, T36V.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 39.
  • the at least one amino acid substitution is R39L, R39Q.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 43.
  • the at least one amino acid substitution is A43E, A43S.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 45.
  • the at least one amino acid substitution is V45A, V45D, V45I.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 46.
  • the at least one amino acid substitution is T46A, T46I.
  • the variant BCMA polypeptide includes at least one amino acid substitution at position 47. In some embodiment, the at least one amino acid substitution is N47D, N47Y.
  • the one or more amino acid substitutions comprise S16A/H19Y/R39Q.
  • the variant BCMA polypeptide comprises any of the mutations listed in Table 1.
  • Table 1 also provides exemplary sequences by reference to SEQ ID NO of the reference (e.g., unmodified) BCMA polypeptide, and exemplary variant BCMA polypeptides.
  • 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. CRD
  • CRD adjacent N- and/or C-terminal amino acids of a given domain
  • the particular domain, such as the ECD domain or a portion thereof containing the CRD1/CRD2 or CRD2 only, of a variant BCMA 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 BCMA polypeptide comprises any of the mutations listed in Table 1.
  • the mutations are made in a reference BCMA containing the sequence of amino acids set forth in SEQ ID NO: 710.
  • the mutations are made in a reference BCMA containing the sequence of amino acids set forth in SEQ ID NO: 356.
  • a variant BCMA polypeptide 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.
  • This principle also extends to the terms “addition” and “deletion” of an amino acid residue which likewise do not imply a particular method of making.
  • the means by which the vTDs are designed or created is not limited to any particular method.
  • a wild-type or unmodified TD encoding nucleic acid is mutagenized from wild-type or unmodified TD genetic material and screened for desired specific binding activity, e.g. binding affinity, and/or alteration of NF- ⁇ B modulation or other functional activity.
  • a vTD 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.
  • the variant BCMA polypeptide comprises an extracellular domain (ECD) sequences set forth in any one of SEQ ID NOS: 357-435.
  • ECD extracellular domain
  • the variant BCMA 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 one of SEQ ID NOS: 357-435, and retains the amino acid modification(s), e.g. substitution(s) therein not present in the reference (e.g., unmodified or wild-type) BCMA.
  • the variant BCMA polypeptide comprises a specific binding fragment of any one of SEQ ID NOS: 357-435, in which the specific binding fragment binds BAFF and/or APRIL and contains a contiguous sequence therein that contains the amino acid modification(s), e.g. substitution (s) therein not present in the reference (e.g., unmodified or wild-type) BCMA.
  • the variant BCMA polypeptide comprises the sequence set forth in SEQ ID NO:381. In some embodiments, the variant BCMA polypeptide consists essentially of the sequence set forth in SEQ ID NO:381. In some embodiments, the variant BCMA polypeptide consists of the sequence set forth in SEQ ID NO:381.
  • the variant BCMA polypeptide comprises the sequence set forth in SEQ ID NO:405. In some embodiments, the variant BCMA polypeptide consists essentially of the sequence set forth in SEQ ID NO:405. In some embodiments, the variant BCMA polypeptide consists of the sequence set forth in SEQ ID NO:405.
  • the variant BCMA polypeptide comprises the sequence set forth in SEQ ID NO:406. In some embodiments, the variant BCMA polypeptide consists essentially of the sequence set forth in SEQ ID NO:406. In some embodiments, the variant BCMA polypeptide consists of the sequence set forth in SEQ ID NO:406.
  • the variant BCMA polypeptide comprises the sequence set forth in SEQ ID NO:410. In some embodiments, the variant BCMA polypeptide consists essentially of the sequence set forth in SEQ ID NO:410. In some embodiments, the variant BCMA polypeptide consists of the sequence set forth in SEQ ID NO:410.
  • the variant BCMA polypeptide comprises the sequence set forth in SEQ ID NO:411. In some embodiments, the variant BCMA polypeptide consists essentially of the sequence set forth in SEQ ID NO:411. In some embodiments, the variant BCMA polypeptide consists of the sequence set forth in SEQ ID NO:411.
  • the variant BCMA polypeptide is encoded by a sequence of nucleotides set forth in any of SEQ ID NOS: 437-515. In some embodiments, the variant BCMA polypeptide is encoded by a sequence of nucleotides that exhibits at least 90% identity, at least 910% 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 one of SEQ ID NOS: 437-515, and retains the amino acid modification(s), e.g. substitution(s) therein not present in the reference (e.g., unmodified or wild-type) BCMA.
  • amino acid modification(s) e.g. substitution(s) therein not present in the reference (e.g., unmodified or wild-type) BCMA.
  • nucleic acid containing the sequence set forth in any of SEQ ID NOS: 437-515 or a 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 one of SEQ ID NOS: 437-515.
  • BCMA-Fc BCMA immunomodulatory protein
  • BIM ECD BCMA H19Y 357 437 358 BCMA H19Y
  • S30G 358 438 359 BCMA H19Y, V45A 359 439
  • BCMA F14Y H19Y 360
  • 440 BCMA H19Y, V45D 361 441 362 BCMA H19Y, A43E 362 442 363 BCMA H19Y, T36A 363 443 364 BCMA H19Y, I22V 364 444 365 BCMA N11D, H19Y 365 445 366 BCMA H19Y, T36M 366 446 367 BCMA N11S, H19Y 367 447 368 BCMA H19Y, L35P, T46A 368 448 369 BCMA H19Y,
  • BCMA ECD fusion sequences in which any of the above BCMA sequence is linked or fused to a multimerization domain, such as any described herein.
  • exemplary multimerization domains are described in Section IV.C.
  • the multimerization domain is an immunoglobulin (e.g. IgG1) Fc region, in which the fusion protein is a BCMA-Fc containing (1) a BCMA sequence containing any of the provided BCMA ECD sequences; and (2) an immunoglobulin Fc region.
  • BCMA-Fc fusion proteins containing (1) a BCMA sequence containing or consisting of any of the above described BCMA ECD polypeptide sequences, such as variant BCMA polypeptide; and (2) an immunoglobulin Fc region.
  • the BCMA-Fc fusion is a variant BCMA-Fc fusion containing or consisting of any of the above described variant BCMA polypeptides and an immunoglobulin Fc region.
  • a variant BCMA-Fc fusion sequence that contains (1) a BCMA ECD sequence that contains the sequence set forth in any one of SEQ ID NOS: 357-435, and (2) an immunoglobulin Fc region.
  • a variant BCMA-Fc fusion sequence that contains (1) a BCMA ECD sequence that consist or consists essentially of the sequence set forth in any one of SEQ ID NOS: 357-435, and (2) an immunoglobulin Fc region.
  • a variant BCMA-Fc fusion sequence that contains (1) a BCMA ECD sequence that contains the sequence set forth in any one of SEQ ID NOS: 357-435 and (2) an immunoglobulin Fc region.
  • a variant BCMA-Fc fusion sequence that contains (1) a BCMA ECD sequence that consists or consists essentially of the sequence set forth in any one of SEQ ID NOS: 357-435 and (2) an immunoglobulin Fc region.
  • the immunoglobulin Fc region can be a wild-type Fc of an immunoglobulin, such as an IgG1 Fc.
  • the Fc region can be a variant Fc that lacks effector function (also called “effectorless Fc”).
  • effectorless Fc also called “effectorless Fc”.
  • the Fc is murine or human Fc. In some embodiments, the Fc is a mammalian or human IgG1, lgG2, lgG3, or lgG4 Fc regions.
  • the Fc is derived from IgG1, such as human IgG1.
  • the Fc is an IgG1 Fc set forth in SEQ ID NO: 586 having an allotype containing residues Glu (E) and Met (M) at positions 356 and 358 by EU numbering.
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 586 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: 586.
  • the Fc is IgG1 Fc that 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:597.
  • an Fc provided herein can contain amino acid substitutions E356D and M358L to reconstitute residues of allotype G1 ml.
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 597 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: 597.
  • the Fc region has the amino acid sequence set forth in SEQ ID NO:597.
  • the variant Fc comprises the sequence set forth in SEQ ID NO: 755. In some embodiments, the variant Fc comprises the sequence set forth in SEQ ID NO:756. In some embodiments, an Fc region used in a construct provided herein can further lack a C-terminal lysine residue.
  • the Fc is derived from IgG2, such as human IgG2.
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 726 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: 726.
  • the Fc is derived from IgG4, such as human IgG4.
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 727 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: 727.
  • 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: 728 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: 728.
  • the Fc region is a variant Fc region in which 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, including in Section IV.C.
  • the Fc region of immunomodulatory proteins 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 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, IgG4-L235A/G237A/E3
  • the variant Fc region comprises the one or more amino acid modifications (e.g amino acid substitutions) is derived from a wild-type IgG1, such as a wild-type human IgG1.
  • the wild-type IgG1 Fc can be the Fc set forth in SEQ ID NO: 586 having an allotype containing residues Glu (E) and Met (M) at positions 356 and 358 by EU numbering.
  • the variant Fc region is derived from the amino acid sequence set forth in SEQ ID NO: 586.
  • the wild-type IgG1 Fc contains amino acids of the human Glml allotype, such as residues containing Asp (D) and Leu (L) at positions 356 and 358, e.g. as set forth in SEQ ID NO:597.
  • the variant Fc is derived from the amino acid sequence set forth in SEQ ID NO:597.
  • the Fc region lacks the C-terminal lysine corresponding to position 232 of the wild-type or unmodified Fc set forth in SEQ ID NO: 586 or 597 (corresponding to K447del by EU numbering).
  • the variant Fc region comprises a C5S amino acid modification of the wild-type or unmodified Fc region by numbering of SEQ ID NO: 586 (corresponding to C220S by EU numbering).
  • the Fc region is a variant Fc that contains at least one amino acid substitution that is N82G by numbering of SEQ ID NO: 586 (corresponding to N297G by EU numbering). In some embodiments, the Fc further contains at least one amino acid substitution that is R77C or V87C by numbering of SEQ ID NO: 586 (corresponding to R292C or V302C by EU numbering). In some embodiments, the variant Fc region further comprises a C5S amino acid modification by numbering of SEQ ID NO: 586 (corresponding to C220S by EU numbering).
  • the variant Fc region comprises the following amino acid modifications: N297G 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:586), e.g., the Fc region comprises the sequence set forth in SEQ ID NO:598.
  • the variant Fc contains the amino acid substitutions L234A/L235E/G237A, by EU numbering. In some embodiments, the variant Fc contains the amino acid substitutions A330S/P331S, by EU numbering. In some embodiments, the variant Fc contains the amino acid substitutions L234A/L235E/G237A/A330S/P331S (Gross et al. (2001) Immunity 15:289). In some embodiments, the variant Fc comprises the sequence set forth in SEQ ID NO: 757. In some embodiments, the variant Fc comprises the sequence set forth in SEQ ID NO:758. In some embodiments, an Fc region used in a construct provided herein can further lack a C-terminal lysine residue.
  • the Fc region is a variant Fc that includes mutations 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.
  • Exemplary inert Fc regions having reduced effector function are set forth in SEQ ID NO: 599 and SEQ ID NO:591, which are based on allotypes set forth in SEQ ID NO:586 or SEQ ID NO: 597, respectively.
  • an Fc region can further lack a C-terminal lysine residue.
  • 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:589, 591, 599 or 724.
  • the variant Fe comprises has the sequence set forth in SEQ ID NO: 589.
  • the variant Fc comprises has the sequence set forth in SEQ ID NO: 591.
  • the variant Fc comprises has the sequence set forth in SEQ ID NO: 599.
  • the variant Fc comprises has the sequence set forth in SEQ ID NO: 724.
  • the Fc region is a variant Fc that has the sequence set forth in SEQ ID NO:589.
  • the Fc region is a variant Fc region that comprises one or more of the amino acid modifications C220S, L235P, L234V, L235A, G236del or S267K, e.g. the Fc region comprises the sequence set forth in SEQ ID NO:722.
  • the Fc region lacks the C-terminal lysine corresponding to position 232 of the wild-type or unmodified Fc set forth in SEQ ID NO: 586 (corresponding to K447del by EU numbering).
  • the Fc region is a variant Fc region that comprises one or more of the amino acid modifications C220S, R292C, N297G, V302C.
  • the Fc region lacks the C-terminal lysine corresponding to position 232 of the wild-type or unmodified Fc set forth in SEQ ID NO: 586 (corresponding to K447del by EU numbering).
  • An exemplary variant Fc region is set forth in SEQ ID NO: 723.
  • the variant Fc region comprises one or more of the amino acid modifications C220S/E233P/L234V/L235A/G236del/S267K.
  • the Fc region lacks the C-terminal lysine corresponding to position 232 of the wild-type or unmodified Fc set forth in SEQ ID NO: 586 (corresponding to K447del by EU numbering).
  • An exemplary variant Fc region is set forth in SEQ ID NO: 725.
  • the Fc region is a variant Fc region containing any combination of the Fc mutations in Table 4. In some embodiments, the Fc region is a variant Fc region having the sequence set forth in any one of the SEQ ID NOs in Table 4.
  • a variant Fc region may be an effectorless Fc that exhibits reduced effector activity compared to a wild-type IgG1 set forth in SEQ ID NO:586 or SEQ ID NO:597.
  • the variant Fc comprises the sequence of amino acids set forth in any of SEQ ID NOS:591, 598, 599, 722, 589, 723, 724, or 725 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: 591, 598, 599722, 589, 723, 724, or 725.
  • the variant Fc has the sequence set forth in SEQ ID NO: 589.
  • the BCMA polypeptide such as the variant BCMA polypeptide
  • the BCMA polypeptide is directly linked to the Fc sequence.
  • the BCMA polypeptide such as the variant BCMA polypeptide
  • one or more “peptide linkers” link the BCMA polypeptide (e.g. variant BCMA polypeptide) and the Fc region.
  • 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. Exemplary linkers are set forth in subsection “Linker.”
  • the linker is (in one-letter amino acid code): GGGGS (“4GS”; SEQ ID NO: 593) or multimers of the 4GS linker, such as repeats of 2, 3, 4, or 5 4GS linkers.
  • the peptide linker is the peptide linker is (GGGGS) 2 (SEQ ID NO: 594), (GGGGS) 3 (SEQ ID NO: 595), (GGGGS) 4 (SEQ ID NO: 600) or (GGGGS) 5 (SEQ ID NO: 671).
  • 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).
  • the linker in one-letter amino acid code is GSGGGGS (SEQ ID NO: 590) or GGGGSSA (SEQ ID NO: 596). In some examples, the linker is a 2 ⁇ GGGGS followed by three alanines (GGGGSGGGGSAAA; SEQ ID NO:721).
  • a BCMA-Fc fusion protein that is a dimer formed by two identical BCMA Fc polypeptides (e.g. variant BCMA polypeptide) as described linked to an Fc domain.
  • identical species of any of the provided BCMA-Fc fusion polypeptides, e.g. variant BCMA-Fc fusion will be dimerized to create a homodimer.
  • the dimer is a homodimer in which the two BCMA Fc polypeptides, e.g. variant BCMA Fc polypeptides, are the same.
  • 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.
  • nucleic acid molecules encoding the BCMA-Fc fusion proteins e.g. variant BCMA-Fc fusion protein.
  • a nucleic acid molecule encoding a BCMA-Fc fusion protein, e.g. variant BCMA-Fc fusion protein is inserted into an appropriate expression vector.
  • the resulting BCMA-Fc fusion protein, e.g. variant BCMA-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, BCMA-Fc fusion proteins.
  • the resulting Fc fusion proteins can be easily purified by affinity chromatography over Protein A or Protein G columns.
  • the provided immunomodulatory protein when produced and expressed from a cells, is a homodimer containing two identical polypeptide chains.
  • FIG. 15 depicts the structure of an exemplary BCMA-Fc fusion protein provided herein.
  • multi-domain immunomodulatory proteins that contains (1) one or more B cell inhibitory molecule (BIM) that bind to a ligand of a B cell stimulatory receptor and (2) one or more T cell inhibitory molecule (BIM) that binds to a T cell stimulatory receptor or a ligand of a T cell stimulatory receptor.
  • BIM B cell inhibitory molecule
  • BIM T cell inhibitory molecule
  • multi-domain immunomodulatory proteins are those in which the BIM antagonizes, such as reduces or inhibits, the activity of a B cell stimulatory receptor, and in which the TIM antagonizes, such as reduces or inhibits, the activity of a T cell stimulatory receptor.
  • the provided immunomodulatory proteins combine B and T cell inhibitors into a single molecule.
  • the BIM and TIM are linked directly or indirectly.
  • the provided immunomodulatory protein can be a fusion protein in which the multidomain BIM and TIM components are further linked to another moiety, such as a multimerization domain or half-life extending molecule.
  • the multidomain immunomodulatiory protein is a BIM/TIM Fc fusion protein.
  • the provided molecules can be used to modulate B and T cell pathways to thereby treat autoimmune diseases, particularly automimmune diseases in which etiology is associated with B and T cell responses.
  • the B cell stimulatory receptor is a receptor expressed on B cells that stimulates B cell responses, such as B cell maturation and differentiation.
  • the B cell stimulatory receptor may be BAFF-R, BCMA and/or TAC.
  • the provided multidomain immunomodulatory proteins antagonize the activity of one or more of BAFF-R, BCMA or TACI.
  • the BIM binds to a ligand of BAFF-R, BCMA or TACI.
  • the ligand may be BAFF or APRIL, which are homotrimeric molecules common to members of the TNF superfamily. BAFF and APRIL are both mainly expressed by myeloid cells, and have been reported to act as costimulatory B cell factors.
  • BAFF and APRIL share two receptors TACI and BCMA; BAFF is also able to bind and stimulate BAFF-R.
  • the ligand may be a heterotrimer of BAFF and APRIL.
  • heterotrimeric complexes of APRIL and BAFF are found in serum, particularly in subjects with autoimmune disease such as those with systemic immune-based rheumatic diseases.
  • the T cell stimulatory receptors comprises an immunoreceptor tyrosine-based activation motif (ITAM) or interacts with an adaptor protein involved in signal transduction pathways in a T cell to transduce activation signals.
  • the T cell stimulatory receptor may be a costimulatory receptor expressed on T cells, such as CD28 or ICOS.
  • the provided multidomain immunomodulatory proteins antagonize the activity of the T cell stimulatory receptor, such as a T cell costimulatory receptor, e.g. CD28 or ICOS.
  • the TIM may bind to the costimulatory receptor or to a ligand of the costimulatory receptor.
  • the provided multi-domain immunomodulatory proteins can be generated so that the TIM binds the T cell stimulatory receptor directly.
  • the TIM may be a CD28 or ICOS binding molecule.
  • the provided multi-domain immunomodulatory proteins can be generated so that the TIM binds a ligand of the T cell stimulatory receptor, and thereby indirectly antagonizes or inhibits the T cell stimulatory receptor.
  • the TIM binds to CD80 or CD86, which are ligands of CD28.
  • the one or more TIM and/or BIM independently include an antibody or an antigen-binding antibody fragment.
  • the TIM and/or BIM can be a human antibody and/or an antibody that binds a human protein.
  • At least one of the TIM or BIM is not an antibody or antigen-binding fragment. In some embodiments, at least one of the TIM or BIM is or contains an extracellular domain of a cell surface molecule expressed on immune cells. For example, certain members of the non-antibody immunoglobulin superfamily (IgSF) are expressed on T cells or regulate activity of T cells. These include, for example, certain T cell costimulatory molecules or ligands thereof.
  • the TIM includes an (IgSF) domain (IgD) of an IgSF member (e.g. wild-type IgD), or a variant IgD (hereinafter called “vIgD”) in which is contained one or more amino acid modifications (e.g.
  • the BIM include a TNF receptor domain (TD) of a TNFR superfamily member (e.g. wild-type TD), or a variant TD (hereinafter called “vTD”) in which is contained one more amino acid modifications (e.g. substitutions) in an TD.
  • TD TNF receptor domain
  • vTD variant TD
  • the BIM can bind to a ligand of a B cell stimulatory receptor with at least a certain binding activity, such as binding affinity, as measured by any of a number of known methods.
  • the TIM can bind to a T cell stimulatory receptor or a ligand of a T cell stimulatory receptor with at least a certain binding activity, such as binding affinity, as measured by any of a number of known methods.
  • the affinity is represented by an equilibrium dissociation constant (K D ) or is represented by EC 50 .
  • a variety of assays are known for assessing binding activity, including binding affinity, and/or determining whether a binding molecule (e.g., a TIM or BIM) specifically binds to a particular binding partner.
  • a binding molecule e.g., a TIM or BIM
  • a BIAcore® instrument can be used to determine the binding kinetics and constants of a complex between two proteins using surface plasmon resonance (SPR) analysis (see, e.g., Scatchard et al., Ann. N. Y. Acad. Sci. 51:660, 1949; Wilson, Science 295:2103, 2002; Wolff et al., Cancer Res. 53:2560, 1993; and U.S. Pat. Nos. 5,283,173, 5,468,614, or the equivalent).
  • SPR surface plasmon resonance
  • Bio-Layer Interferometry using a ForteBio Octet system may be used, such as with streptavidin coated sensor and biotinylated recombinant protein domain.
  • suitable assays for measuring the binding of one protein to another include, for example, immunoassays such as enzyme linked immunosorbent assays (ELISA) and radioimmunoassays (RIA), or determination of binding by monitoring the change in the spectroscopic or optical properties of the proteins through fluorescence, UV absorption, circular dichroism, or nuclear magnetic resonance (NMR).
  • exemplary assays include, but are not limited to, Western blot, ELISA, analytical ultracentrifugation, spectroscopy, flow cytometry, sequencing and other methods for detection of expressed nucleic acids or binding of proteins.
  • the BIM and TIM independently exhibit a binding affinity for a binding partner with a K D (i.e., an equilibrium dissociation constant of a particular binding interaction with units of M; equal to the ratio of the off-rate [k off or k d ] to the on-rate [k on or k a ] for this association reaction, assuming bimolecular interaction) of equal to or less than 10 ⁇ 5 M.
  • a K D i.e., an equilibrium dissociation constant of a particular binding interaction with units of M; equal to the ratio of the off-rate [k off or k d ] to the on-rate [k on or k a ] for this association reaction, assuming bimolecular interaction
  • K D ranges from 10 ⁇ 6 M to 10 ⁇ 12 M, such as 10 ⁇ 7 M to 10 ⁇ 11 M, 10 ⁇ 8 M to 10 ⁇ 10 M, or 10 ⁇ 9 M to 10 ⁇ 10 M.
  • the on-rate (association rate constant; k on or k a ; units of 1/Ms) and the off-rate (dissociation rate constant; k off or k d ; units of 1/s) can be determined using any of the assay methods known in the art, for example, surface plasmon resonance (SPR).
  • SPR surface plasmon resonance
  • the BIM exhibits a binding affinity for a ligand of a B cell stimulatory receptor that is from or from about 0.001 nM to 1000 nM, such as from or from about 0.01 nM to about 500 nM, from or from about 0.01 nM to about 400 nM, from or from about 0.01 nM to about 100 nM, from or from about 0.01 nM to about 50 nM, from or from about 0.01 nM to about 10 nM, from or from about 0.01 nM to about 1 nM, from or from about 0.01 nM to about 0.1 nM, is from or from about 0.1 nM to about 500 nM, from or from about 0.1 nM to about 400 nM, from or from about 0.1 nM to about 100 nM, from or from about 0.1 nM to about 50 nM, from or from about 0.1 nM to about 10 nM, from or from about 0.1 nM to about 1
  • the binding affinity of the BIM for the inhibitory receptor is at or less than or about 400 nM, 300 nM, 200 nM, 100 nM, 50 nM, 40 nM, 30 nM, 25 nM, 20 nM, 19 nM, 18 nM, 17 nM, 16 nM, 15 nM, 14 nM, 13 nM, 12 nM, 11 nM, 10 nM, 9 nM, 8 nM, 7 nM, 6 nM, 5 nM, 4 nM, 3 nM, 2 nM, or 1 nM or less.
  • the TIM exhibits a binding affinity for a T cell stimulatory receptor or a ligand of a T cell stimulatory receptor that is from or from about 0.001 nM to about 1000 nM, such as from or from about 0.01 nM to about 500 nM, from or from about 0.01 nM to about 400 nM, from or from about 0.01 nM to about 100 nM, from or from about 0.01 nM to about 50 nM, from or from about 0.01 nM to about 10 nM, from or from about 0.01 nM to about 1 nM, from or from about 0.01 nM to about 0.1 nM, is from or from about 0.1 nM to about 500 nM, from or from about 0.1 nM to about 400 nM, from or from about 0.1 nM to about 100 nM, from or from about 0.1 nM to about 50 nM, from or from about 0.1 nM to about 10 nM, from or from about
  • the binding affinity of the TIM for the stimulatory receptor or a ligand of the T cell stimulatory receptor is at or less than or about 400 nM, 300 nM, 200 nM, 100 nM, 50 nM, 40 nM, 30 nM, 25 nM, 20 nM, 19 nM, 18 nM, 17 nM, 16 nM, 15 nM, 14 nM, 13 nM, 12 nM, 11 nM, 10 nM, 9 nM, 8 nM, 7 nM, 6 nM, 5 nM, 4 nM, 3 nM, 2 nM, or 1 nM or less.
  • the TIM, or the multidomain immunomodulatory protein containing the TIM is not an agonist of the T cell stimulatory (e.g. costimulatory) receptor.
  • the TIM, or the multidomain immunodulatory protein containing the TIM binds to a T cell costimulatory receptor, e.g. CD28 or ICOS, but exhibits a relatively low affinity for the T cell costimulatory receptor.
  • the TIM has a binding affinity for the T cell costimulatory receptor of greater than 1 ⁇ 10 ⁇ 9 M, such as between at or about 1 ⁇ 10 ⁇ 7 M and at or about 1 ⁇ 10 ⁇ 9 M.
  • the TIM has a binding affinity for the T cell costimulatory receptor of at or about 1 ⁇ 10 ⁇ 7 M, at or about 2.5 ⁇ 10 ⁇ 7 M, at or about 5 ⁇ 10 ⁇ 7 M, at or about 7.5 ⁇ 10 ⁇ 7 M, at or about 1 ⁇ 10 ⁇ 8 M, at or about 2.5 ⁇ 10 ⁇ 8 M, at or about 5 ⁇ 10 ⁇ 8 M, at or about 7.5 ⁇ 10 ⁇ 8 M, or at or about 1 ⁇ 10 ⁇ 9 M, or any value between any of the foregoing.
  • the TIM of the provided multidomain immunomodulatory proteins does not directly bind to a T cell costimulatory receptor. In some embodiments, the TIM of the provided multidomain immunomodulatory proteins binds to a ligand of the T cell costimulatory receptor.
  • the provided multi-domain immunomodulatory proteins can include the BIM and TIM in various configurations or formats, including formats with one or more further moieties.
  • the provided immunomodulatory proteins include polypeptides in which the one or more BIM is N-terminal to the TIM.
  • the one or more BIM is C-terminal to the TIM.
  • the one or more BIM and the one or more TIM can be linked directly or indirectly, via a linker.
  • the immunomodulatory proteins can be formatted as multimeric molecules via fusion with a multimerization domain, such as an Fc protein.
  • the multi-domain immunomodulatory proteins can be formatted as multimeric molecules, e.g., dimeric, trimer, tetrameric, or pentameric molecules. In some embodiments, the immunomodulatory proteins are formatted as a monomeric molecules containing single polypeptide fusions of the one or more BIM and the one or more TIM.
  • FIG. 16 depicts exemplary formats and configurations, all of which may be encompassed by a provided multi-domain immunomodulatory protein.
  • the provided immunomodulatory protein contains a BIM that binds to one or more ligands of a B cell stimulatory receptor.
  • the B cell stimulatory receptor is a member of the TNFRSF.
  • the one or more B cell stimulatory receptor is TACI and BCMA.
  • the ligand of the B cell stimulatory receptor is BAFF or APRIL.
  • the BIM binds to BAFF, APRIL and/or a BAFF/APRIL heterotrimer.
  • the BIM is able to binds to BAFF, APRIL and a BAFF/APRIL heterotrimer.
  • the BIM is an antibody or antigen-binding fragment that binds to the ligand of a B cell stimulatory receptor. In some embodiments, the BIM is an antibody or antigen-binding fragment that binds BAFF and/or APRIL, such as a human BAFF and/or human APRIL.
  • the BIM is or contains a binding partner of the ligand of the B cell stimulatory receptor.
  • the multi-domain immunomodulatory protein provided herein are soluble proteins and/or do not contain a portion that includes a transmembrane domain.
  • cell surface proteins including proteins of the TNFRSF such as B cell stimulatory receptors, e.g. BCMA and TACI, typically have an intracellular domain, a transmembrane domain, 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.
  • the BIM lacks a transmembrane domain or a portion of the transmembrane domain of the B cell stimulatory receptor, e.g. BCMA or TACI.
  • the BIM lacks the intracellular (cytoplasmic) domain or a portion of the intracellular domain of the B cell stimulatory receptor, e.g. BCMA or TACI.
  • the BIM only contains the ECD domain or a portion thereof containing a TD, such as a CRD, or specific binding fragments thereof.
  • the BIM is or contains an ECD of a B cell stimulatory receptor, or a specific binding portion or fragment thereof containing at least one TD (e.g. at least one CRD), that binds to a ligand of the B cell stimulatory receptor.
  • the BIM can contain an ECD of TACI or BCMA, or a specific binding portion or fragment of TACI or BCMA containing at least one TD (e.g. at least one CRD), that binds to APRIL, BAFF and/or an APRIL/BAFF heterotrimer.
  • the BIM consists or consists essentially of an ECD of a B cell stimulatory receptor, or a specific binding portion or fragment thereof containing at least one TD (e.g. at least one CRD), such as consists or consists essentially of the ECD of TACI or BCMA or a specific binding portion or fragment of the ECD of TACI or BCMA that contains at least one TD (e.g. at least one CRD).
  • the BIM is less than the full length sequence of the ECD of the B cell stimulatory receptor.
  • the BIM is or only contains one CRD or a specific binding fragment of the CRD.
  • the BIM consists or consists essentially of a CRD of a B cell stimulatory receptor, such as consists or consists essentially of only one CRD of TACI or BCMA.
  • the sequence of the BIM containing an ECD or binding portion or fragment thereof containing a TD is a mammalian sequence that includes, but is not limited to, human, mouse, cynomolgus monkey, or rat.
  • the BIM sequence is human and/or binds a human protein.
  • the BIM is or includes a vTD that is an affinity-modified domain that exhibits increased binding activity, such as increased binding affinity, for the ligand of the B cell stimulatory receptor compared to the binding activity of the unmodified or wild-type TD for the same molecule.
  • the BIM contains a vTD with one or more amino acid substitutions compared to a TD of a TNFRSF member, e.g. BCMA or TACI, in which, the one or more amino acid substitutions confer or result in increased binding affinity to a cognate ligand of the B cell stimulatory receptor.
  • the BIM is or contains a vTD that contains one or more amino acids modifications, such as one or more substitutions (alternatively, “mutations” or “replacements”), deletions or additions, relative to a wild-type or unmodified TD of a binding partner of a ligand of the B cell stimulatory receptor.
  • the vTD contains 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, such as amino acid substitutions, deletions or additions in an TD domain of an TNFRSF binding partner of a B cell stimulatory receptor.
  • the modifications e.g., substitutions
  • the vTD 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 modifications (e.g., substitutions) in the CRD or specific binding fragment thereof. In some embodiments, the vTD has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the wild-type or unmodified TD or specific binding fragment thereof.
  • Non-limiting examples of a BIM in the provided multidomain immunomodulatory proteins are described in the following subsections. Any of the described BIMs herein can be combined with a TIM as described in Section III.B.
  • the BIM is or contains a wild-type TACI ECD or a specific binding portion or fragment thereof containing at least one TD (e.g. at least one CRD) that binds to APRIL, BAFF and/or an APRIL/BAFF heterotrimer.
  • the BIM is or contains a variant TACI ECD or a specific binding portion or fragment thereof containing at least one TD (e.g. at least one CRD) that binds to APRIL, BAFF and/or an APRIL/BAFF heterotrimer.
  • the BIM is a TACI polypeptide or variant thereof with any of the sequences set forth herein.
  • TACI is a tumor necrosis factor receptor family member characterized by having an extracellular domain (ECD) containing cysteine-rich pseudo-repeat domains (CRDs).
  • ECD extracellular domain
  • TACI is a membrane bound receptor, which has an extracellular domain containing two cysteine-rich pseudorepeats (CRD1 and CRD2), a transmembrane domain and a cytoplasmic domain that interacts with CAML (calcium-modulator and cyclophilin ligand), an integral membrane protein located at intracellular vesicles which is a co-inducer of NF-AT activation when overexpressed in Jurkat cells.
  • TACI is associated with B cells and a subset of T cells.
  • the TACI receptor binds two members of the tumor necrosis factor (TNF) ligand family.
  • TNF tumor necrosis factor
  • BAFF B cell Activating Factor of the TNF Family
  • ZTNF4 ZTNF4, “neutrokine- ⁇ ,” “BLyS,” “TALL-1,” and “THANK”
  • the other ligand has been designated as APRIL, and also is variously designated as “ZTNF2” and “TNRF death ligand-1” (Hahne et al., J. Exp. Med. 188:1185 (1998); Kelly et al., Cancer Res. 60:1021 (2000)). Both ligands are also bound by the B-cell maturation receptor (BCMA) (Gross et al., Nature 404:995 (2000)). Binding of TACI receptor to its ligands BAFF or APRIL stimulates B cell responses, including T cell-independent B cell antibody responses, isotype switching, and B cell homeostasis.
  • BCMA B-cell maturation receptor
  • the amino acid sequence of full-length TACI is set forth in SEQ ID NO:666.
  • the protein is a type III membrane protein and lacks a signal peptide; following expression in eukaryotic cells the N-terminal methionine is removed.
  • a mature TACI protein does not contain the N-terminal methionine as set forth in SEQ ID NO:666.
  • the extracellular domain of TACI (amino acid residues 1-166 of SEQ ID NO:666; ECD set forth in SEQ ID NO:709) contains two cysteine rich domain (CRDs, hereinafter also called a tumor necrosis family receptor domain or TD), each of which exhibit affinity for binding to BAFF and APRIL.
  • the first cysteine rich domain contains amino acid residues 34-66 of the sequence set forth in SEQ ID NO:709.
  • the second cysteine rich domain (CRD2) corresponds to amino acids 71-104 of the sequence set forth in SEQ ID NO:709.
  • TACI also contains a stalk region of about 60 amino acids following the second cysteine repeat in the extracellular domain, corresponding to amino acid residues 105-165 of the sequence set forth in SEQ ID NO:709.
  • the BIM is a variant TACI polypeptide that contains one or more amino acid modifications, such as one or more substitutions (alternatively, “mutations” or “replacements”), deletions or additions in the extracellular domain of a reference TACI polypeptide, such as a wild-type or unmodified TACI polypeptide containing a CRD(s) (hereinafter also called TDs).
  • a provided BIM that is a variant TACI polypeptide is or comprises a variant TD (“vTD”) in which the one or more amino acid modifications (e.g. substitutions) is in a CRD.
  • the one or more amino acids modifications such as one or more substitutions (alternatively, “mutations” or “replacements”), deletions or addition, is in the CRD1 region. In some embodiments, the one or more amino acids modifications, such as one or more substitutions (alternatively, “mutations” or “replacements”), deletions or addition, is in the CRD2 region. In some embodiments, the one or more amino acids modifications, such as one or more substitutions (alternatively, “mutations” or “replacements”), deletions or addition, is in amino acids within both the CRD1 and CRD2 regions.
  • the reference (e.g. unmodified) TACI sequence is a wild-type TACI sequence or is a portion thereof that contains one or both CRDs.
  • the reference (e.g., unmodified) TACI is or comprises the extracellular domain (ECD) of TACI or a portion thereof containing one or both CRD domains.
  • the extracellular domain of a reference (e.g., unmodified) TACI polypeptide comprises a CRD1 and CRD2.
  • the variant TACI polypeptide need not comprise both the CRD1 and the CRD2.
  • the variant TACI polypeptide comprises or consists essentially of the CRD1 or a specific binding fragment thereof.
  • the variant TACI polypeptide comprises or consists essentially of the CRD2 or specific binding fragments thereof. In some embodiments, the variant TACI is a soluble polypeptide and lacks a transmembrane domain. In some embodiments, the variant TACI polypeptide further comprises a transmembrane domain and, in some cases, also a cytoplasmic domain.
  • the reference (e.g., unmodified) TACI sequence is a mammalian TACI sequence.
  • the reference (e.g., unmodified) TACI sequence can be a mammalian TACI that includes, but is not limited to, human, mouse, cynomolgus monkey, or rat.
  • the reference (e.g., unmodified) TACI sequence is human.
  • the extracellular domain of an exemplary human TACI sequence is set forth in SEQ ID NO:709.
  • the reference (e.g., unmodified) TACI sequence has (i) the sequence of amino acids set forth in SEQ ID NO:709 or a sequence thereof that lacks the N-terminal methionine, (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:709 and that binds to APRIL, BAFF or an APRIL/BAFF heterotrimer, or (iii) is a fragment or portion of (i) or (ii) containing a CRD1 and/or CRD2, in which the portion binds to APRIL, BAFF or an APRIL/BAFF heterotrimer.
  • the reference (e.g., unmodified) TACI sequence lacks the N-terminal methionine as set forth in SEQ ID NO: 709.
  • TACI Extracellular Domain SEQ ID NO: 709 MSGLGRSRRGGRSRVDQEERFPQGLWTGVAMRSCP EEQYWDPLLGTCMSCKTICNHQSQRTCAAFCRSLS CRKEQGKFYDHLLRDCISCASICGQHPKQCAYFCE NKLRSPVNLPPELRRQRSGEVENNSDNSGRYQGLE HRGSEASPALPGLKLSADQVALVYST
  • the reference (e.g. unmodified) TACI sequence is an extracellular domain sequence of TACI that is a portion of the ECD that contains an N-terminal deletion relative to the sequence of amino acids set forth in SEQ ID NO:709.
  • the N-terminal deletion is deletion of N-terminal amino acid residues 1-28 corresponding to residues set forth in SEQ ID NO:709.
  • the N-terminal deletion is deletion of N-terminal amino acid residues 1-29 corresponding to residues set forth in SEQ ID NO:709.
  • the N-terminal deletion is deletion of N-terminal amino acid residues 1-30 corresponding to residues set forth in SEQ ID NO:709.
  • the N-terminal deletion is deletion of N-terminal amino acid residues 1-31 corresponding to residues set forth in SEQ ID NO:709. In some embodiments, the N-terminal deletion is deletion of N-terminal amino acid residues 1-32 corresponding to residues set forth in SEQ ID NO:709. In some embodiments, the N-terminal deletion is deletion of N-terminal amino acid residues 1-33 corresponding to residues set forth in SEQ ID NO:709.
  • the reference (e.g. unmodified) TACI sequence is an ECD portion that contains deletion of one or more residues of the stalk portion of the TACI extracellular domain.
  • the reference (e.g. unmodified) TACI sequence is an ECD portion that lacks one or more contiguous C-terminal amino acid residues beginning at residue 105 and up to or including amino acid residue 166 corresponding to residues of the ECD sequence set forth in SEQ ID NO:709.
  • the reference (e.g. unmodified) TACI sequence contains an ECD portion having a contiguous sequence of amino acids that includes the CRD1 and/or CRD2 (e.g. CRD1 and CRD2 or CRD2 only) and only a segment or portion of the stalk sequence.
  • Suitable stalk segments include one or more amino acids of amino acid residues 105 to 154 of SEQ ID NO:709.
  • the stalk segment can consist of the following with reference to SEQ ID NO: 709: amino acid residue 105, amino acid residues 105 to 106, amino acid residues 105 to 107, amino acid residues 105 to 108, amino acid residues 105 to 109, amino acid residues 105 to 110, amino acid residues 105 to 111, amino acid residues 105 to 112, amino acid residues 105 to 113, amino acid residues 105 to 114, amino acid residues 105 to 115, amino acid residues 105 to 116, amino acid residues 105 to 117, amino acid residues 105 to 118, amino acid residues 105 to 119, amino acid residues 105 to 120, amino acid residues 105 to 121, amino acid residues 105 to 122, amino acid residues 105 to 123, amino acid residues 105 to 124, amino acid residues 105 to 125, amino acid residues 105 to 126, amino acid residues 105 to 127, amino acid residues 105 to 128, amino acid residue
  • the reference (e.g. unmodified) TACI sequence lacks or is mutated in one or more potential furin cleavage sites.
  • the reference (e.g. unmodified) TACI sequence is an ECD or portion that in which the arginine residue at position 119 is mutated, e.g. R119G.
  • the reference (e.g. unmodified) TACI sequence is an ECD or portion that in which the glutamine residue at position 121 is mutated, e.g. Q121P.
  • the reference (e.g. unmodified) TACI sequence is an ECD or portion that in which the arginine residue at position 122 is mutated, e.g. R122Q.
  • the reference TACI sequence is a TACI ECD sequence as set forth in international PCT publication No. WO2000/067034, WO2002/094852 or WO2008/154814.
  • the reference TACI sequence is a TACI ECD sequence that has or consists of the sequence set forth in SEQ ID NO:719.
  • TACI ECD (CRD1/CRD2): SEQ ID NO: 719 SRVDQEER FPQGLWTGVA MRSCPEEQYW DPLLGTCMSCKTICNHQSQR TCAAFCRSLS CRKEQGKFYD HLLRDCISCA SICGQHPKQ CAYFCENKLRS PVNLPPEL
  • the reference TACI sequence is a TACI ECD sequence that has or consists of the sequence set forth in SEQ ID NO:718.
  • TACI ECD (CRD1/CRD2): SEQ ID NO: 718 AMRSCPEEQYWDPLLGTCMSCKTICNHQSQRTCAA FCRSLSCRKEQGKFYDHLLRDCISCASICGQHPKQ CAYFCENKLRS
  • the reference TACI sequence is a TACI ECD sequence that has or consists of the sequence set forth in SEQ ID NO:516 (encoded by the sequence of nucleotides set forth in SEQ ID NO:551).
  • TACI ECD (CRD1/CRD2): SEQ ID NO: 516 VAMRSCPEEQYWDPLLGTCMSCKTICNHQSQRTCA AFCRSLSCRKEQGKFYDHLLRDCISCASICGQHPK QCAYFCENKLRS
  • the reference TACI sequence is an extracellular domain region of TACI that consists essentially of only the CRD2 sequence and that is deleted in or lacks the entirety of the sequence of the CRD1 and substantially all of the stalk region.
  • residues in the stalk region may contain a protease cleavage site, it was believed that at least the CRD1 and CRD2 was required for sufficient expression and/or binding activity of TACI for its cognate ligands.
  • international PCT publication No. WO2002/094852 demonstrated that a TACI molecule containing a CRD1 and CRD2, but in which the whole amino terminal region and a partial sequence of the stalk region was deleted, exhibited reduced protein degradation when expressed.
  • the BIM is a TACI polypeptide that is a portion of the TACI extracellular domain (ECD) region that contains the CRD2, with a deletion of the N-terminal region and CRD1 and deletion of one or more residues of the stalk portion of the TACI extracellular domain, e.g. relative to the sequence of amino acids set forth in SEQ ID NO:709.
  • the portion of the TACI extracellular domain that contains the CRD2 includes amino acid residues 71-104 corresponding to residues set forth in SEQ ID NO:709.
  • the BIM of the immunomodulatory protein is a TACI polypeptide that contains deletion of N-terminal amino acid residues 1-66 corresponding to residues set forth in SEQ ID NO:709. In provided embodiments, the BIM of the immunomodulatory protein is a TACI polypeptide that contains deletion of N-terminal amino acid residues 1-67 corresponding to residues set forth in SEQ ID NO:709. In provided embodiments, the BIM of the immunomodulatory protein is a TACI polypeptide that contains deletion of N-terminal amino acid residues 1-68 corresponding to residues set forth in SEQ ID NO:709.
  • the BIM of the immunomodulatory protein is a TACI polypeptide that contains deletion of N-terminal amino acid residues 1-69 corresponding to residues set forth in SEQ ID NO:709. In provided embodiments, the BIM of the immunomodulatory protein is a TACI polypeptide that contains deletion of N-terminal amino acid residues 1-70 corresponding to residues set forth in SEQ ID NO:709. In some of any such embodiments, the BIM of the immunomodulatory protein is a TACI polypeptide that lacks one or more contiguous C-terminal amino acid residues beginning at residue 105 and up to or including amino acid residue 166 corresponding to residues of the ECD sequence set forth in SEQ ID NO:709.
  • the BIM of an immunomodulatory protein provided herein is a TACI polypeptide with a sequence that contains an ECD portion having a contiguous sequence of amino acids of a TACI ECD that includes the CRD2 (e.g. residues 71-104 with reference to SEQ ID NO:709), but with a deletion of the N-terminal region and CRD1 and deletion of one or more residues of the stalk portion of the TACI extracellular domain, e.g. relative to the sequence of amino acids set forth in SEQ ID NO:709.
  • CRD2 e.g. residues 71-104 with reference to SEQ ID NO:709
  • the TACI ECD portion can consist of the following with reference to amino acid residues set forth in SEQ ID NO:709: amino acid residues 67 to 118, amino acid residues 67 to 117, amino acid residues 67 to 116, amino acid residues 67 to 115, amino acid residues 67 to 114, amino acid residues 67 to 113, amino acid residues 67 to 112, amino acid residues 67 to 111, amino acid residues 67 to 110, amino acid residues 67 to 109, amino acid residues 67 to 108, amino acid residues 67 to 107, amino acid residues 67 to 106, amino acid residues 67 to 105, or amino acid residues 67 to 104.
  • the TACI ECD portion can consist of the following with reference to residues set forth in SEQ ID NO: 709: amino acid residues 68 to 118, amino acid residues 68 to 117, amino acid residues 68 to 116, amino acid residues 68 to 115, amino acid residues 68 to 114, amino acid residues 68 to 113, amino acid residues 68 to 112, amino acid residues 68 to 111, amino acid residues 68 to 110, amino acid residues 68 to 109, amino acid residues 68 to 108, amino acid residues 68 to 107, amino acid residues 68 to 106, amino acid residues 68 to 105, or amino acid residues 68 to 104.
  • the TACI ECD portion can consist of the following with reference to residues set forth in SEQ ID NO: 709: amino acid residues 69 to 118, amino acid residues 69 to 117, amino acid residues 69 to 116, amino acid residues 69 to 115, amino acid residues 69 to 114, amino acid residues 69 to 113, amino acid residues 69 to 112, amino acid residues 69 to 111, amino acid residues 69 to 110, amino acid residues 69 to 109, amino acid residues 69 to 108, amino acid residues 69 to 107, amino acid residues 69 to 106, amino acid residues 69 to 105, or amino acid residues 69 to 104.
  • the TACI ECD portion can consist of the following with reference to residues set forth in SEQ ID NO: 709: amino acid residues 70 to 118, amino acid residues 70 to 117, amino acid residues 70 to 116, amino acid residues 70 to 115, amino acid residues 70 to 114, amino acid residues 70 to 113, amino acid residues 70 to 112, amino acid residues 70 to 111, amino acid residues 70 to 110, amino acid residues 70 to 109, amino acid residues 70 to 108, amino acid residues 70 to 107, amino acid residues 70 to 106, amino acid residues 70 to 105, or amino acid residues 70 to 104.
  • the TACI ECD portion can consist of the following with reference to residues set forth in SEQ ID NO: 709: amino acid residues 71 to 118, amino acid residues 71 to 117, amino acid residues 71 to 116, amino acid residues 71 to 115, amino acid residues 71 to 114, amino acid residues 71 to 113, amino acid residues 71 to 112, amino acid residues 71 to 111, amino acid residues 71 to 110, amino acid residues 71 to 109, amino acid residues 71 to 108, amino acid residues 71 to 107, amino acid residues 71 to 106, amino acid residues 71 to 105, or amino acid residues 71 to 104.
  • any of the above TACI ECD sequences also can be a TACI reference sequence in accord with a TIM that is a variant TACI in the immunomodulatory proteins provided herein, in which such immunomodulatory proteins contain a variant TACI polypeptide that is modified by one or more amino acid modification (e.g. substitution) as described herein compared to such TACI reference sequence.
  • TACI ECD sequence that has or consists of the sequence set forth in SEQ ID NO:528 (encoded by the sequence of nucleotides set forth in SEQ ID NO:563.
  • the reference TACI sequence has or consists of the sequence set forth in SEQ ID NO:528, in which a provided variant TACI polypeptide is modified by one or more amino acid modification (e.g. substitution) as described herein compared to such reference TACI sequence.
  • TACI ECD sequence (CRD2): SEQ ID NO: 528 SLSCRKEQGKFYDHLLRDCISCASICGQHPKQCAY FCENKLRS
  • variant TACI polypeptides Among BIMs in a provided immunomodulatory protein are variant TACI polypeptides.
  • the variant TACI sequence has the sequence of the reference (e.g. unmodified) TACI sequence, such as any described above, but additionally contains one more amino acid modifications, such as one or more amino acid substitutions.
  • a BIM provided herein may be a variant TACI polypeptide containing at least one affinity-modified TD domain (e.g., CRD1 and/or CRD2) or a specific binding fragment thereof that contains one or more amino acid substitutions in a TD domain of a reference (e.g., unmodified or wild-type) TACI polypeptide, such that the variant TACI polypeptide exhibits altered (e.g. increased) binding activity or affinity for one or both of APRIL or BAFF compared to the reference (e.g., unmodified or wild-type) TACI polypeptide.
  • a reference e.g., unmodified or wild-type
  • a BIM is a variant TACI polypeptide that has a binding affinity for APRIL and/or BAFF that differs from that of a reference (e.g., unmodified or wild-type) TACI polypeptide control sequence as determined by, for example, solid-phase ELISA immunoassays, flow cytometry or Biacore assays. Binding affinities for each of the cognate binding partners are independent; that is, in some embodiments, a variant TACI polypeptide has an increased binding affinity for one or both APRIL and BAFF, and a decreased or unchanged binding affinity for the other of APRIL or BAFF, relative to a reference (e.g., unmodified or wild-type) TACI polypeptide.
  • a reference e.g., unmodified or wild-type
  • the BIM is a variant TACI polypeptide that has an increased binding affinity for BAFF, relative to the reference (unmodified or wild-type) TACI polypeptide. In some embodiments, the BIM is a variant TACI polypeptide that has an increased binding affinity for APRIL relative to the reference (unmodified or wild-type) TACI polypeptide. In some embodiments, the BIM is a variant TACI polypeptide has an increased binding affinity for APRIL and BAFF relative to the reference (unmodified or wild-type) TACI polypeptide.
  • the cognate ligands BAFF and/or APRIL can be a mammalian protein, such as a human protein or a murine protein.
  • a BIM that is a variant TACI polypeptide with increased or greater binding affinity to APRIL and/or BAFF will have an increase in binding affinity relative to the reference (e.g., unmodified or wild-type) TACI polypeptide control of at least about 5%, such as at least about 10%, 15%, 20%, 25%, 35%, or 50%.
  • the increase in binding affinity relative to the reference (e.g., unmodified or wild-type) TACI 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) TACI polypeptide has the same sequence as the variant TACI 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 BAFF 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.
  • the K d of any of the foregoing embodiments to BAFF is less than at or about 1 ⁇ 10 ⁇ 9 M, 1 ⁇ 10 ⁇ 10 M or 1 ⁇ 10 ⁇ 11 M, or 1 ⁇ 10 ⁇ 12 M.
  • the K d of any of the foregoing embodiments to BAFF is between 1 ⁇ 10 ⁇ 9 M and at or about 1 ⁇ 10 ⁇ 12 M. In some embodiments, the K d of any of the foregoing embodiments to BAFF is at or about 1 ⁇ 10 ⁇ 9 M, at or about 2 ⁇ 10 ⁇ 9 M, at or about 4 ⁇ 10 ⁇ 9 M, at or about 6 ⁇ 10 ⁇ 9 M, at or about 8 ⁇ 10 ⁇ 9 M, at or about 1 ⁇ 10 ⁇ 10 M, at or about 2 ⁇ 10 ⁇ 10 M, at or about 4 ⁇ 10 ⁇ 10 M, at or about 6 ⁇ 10 ⁇ 10 M, at or about 8 ⁇ 10 ⁇ 10 M, at or about 1 ⁇ 10 ⁇ 11 M, at or about 2 ⁇ 10 ⁇ 11 M, at or about 4 ⁇ 10 ⁇ 10 M, at or about 6 ⁇ 10 ⁇ 11 M, at or about 8 ⁇ 10 ⁇ 11 M, or at or about 1 ⁇ 10 ⁇ 12 M, or any value between any of the foregoing.
  • a BIM in a provided embodiment is a variant TACI polypeptide as described above and the K d to BAFF is decreased (higher binding affinity) by greater than or greater than about 1.5-fold, such as greater than or about 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold or more.
  • the equilibrium dissociation constant (K d ) of any of the foregoing embodiments to APRIL 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.
  • the K d of any of the foregoing embodiments to APRIL is less than at or about 1 ⁇ 10 ⁇ 9 M, 1 ⁇ 10 ⁇ 10 M or 1 ⁇ 10 ⁇ 11 M, or 1 ⁇ 10 ⁇ 12 M.
  • the K d of any of the foregoing embodiments to APRIL is between 1 ⁇ 10 ⁇ 9 M and at or about 1 ⁇ 10 ⁇ 12 M. In some embodiments, the K d of any of the foregoing embodiments to APRIL is at or about 1 ⁇ 10 ⁇ 9 M, at or about 2 ⁇ 10 ⁇ 9 M, at or about 4 ⁇ 10 ⁇ 9 M, at or about 6 ⁇ 10 ⁇ 9 M, at or about 8 ⁇ 10 ⁇ 9 M, at or about 1 ⁇ 10 ⁇ 10 M, at or about 2 ⁇ 10 ⁇ 10 M, at or about 4 ⁇ 10 ⁇ 10 M, at or about 6 ⁇ 10 ⁇ 10 M, at or about 8 ⁇ 10 ⁇ 10 M, at or about 1 ⁇ 10 ⁇ 11 M, at or about 2 ⁇ 10 ⁇ 11 M, at or about 4 ⁇ 10 ⁇ 11 M, at or about 6 ⁇ 10 ⁇ 11 M, at or about 8 ⁇ 10 ⁇ 11 M, or at or about 1 ⁇ 10 ⁇ 12 M, or any value between any of the foregoing.
  • a BIM of a provided embodiment is a variant TACI polypeptide as described above and the K d to APRIL is decreased (higher binding affinity) by greater than or greater than about 1.5-fold, such as greater than or about 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold or more.
  • the reference (e.g., unmodified or wild-type) TACI sequence does not necessarily have to be used as a starting composition to generate a BIM that is a variant TACI polypeptide described herein. Therefore, use of the term “modification”, such as “substitution” does not imply that the present embodiments are limited to a particular method of making variant TACI polypeptides or immunomodulatory proteins containing the same.
  • Variant TACI 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 TACI polypeptides are designed or created is not limited to any particular method.
  • a reference (e.g., unmodified or wild-type) TACI encoding nucleic acid is mutagenized from reference (e.g., unmodified or wild-type) TACI genetic material and screened for desired specific binding affinity or other functional activity.
  • a variant TACI 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 (s) of a BIM that is a variant TACI polypeptide are designated by amino acid position number corresponding to the numbering of positions of the reference ECD sequence set forth in SEQ ID NO:709. It is within the level of a skilled artisan to identify the corresponding position of a modification, e.g. amino acid substitution, in an TACI polypeptide, including portion thereof containing TD (e.g. CRD1 and/or CRD2) thereof, such as by alignment of a reference sequence (e.g. SEQ ID NO:516 or 528) with SEQ ID NO:709. An alignment identifying corresponding residues is exemplified in FIG. 17 A .
  • 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.
  • a BIM is a variant TACI polypeptide that has one or more amino acid modification, e.g. substitution in a reference (e.g., unmodified or wild-type) TACI sequence, such as any as described.
  • 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) TACI sequence.
  • the one or more amino acid modification, e.g. substitution is in the CRD1 domain or specific binding fragment thereof.
  • the one or more amino acid modification, e.g. substitution is in the CRD2 domain or specific binding fragment thereof.
  • some of the one or more amino acid modification, e.g. substitution is in the CRD1 domain or a specific binding fragment thereof, and some of the one or more amino acid modification, e.g. substitution are in the CRD2 domain or a specific binding fragment thereof.
  • a BIM that is a variant TACI 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 CRD1 domain or the CRD2 domain.
  • a BIM that is a variant TACI 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 CRD1 domain or specific binding fragment thereof.
  • a BIM that is a variant TACI 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 CRD2 domain or specific binding fragment thereof.
  • the variant TACI polypeptide containing the one or more amino acid modifications e.g.
  • amino acid substitutions as described 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) TACI polypeptide or specific binding fragment thereof, such as with the amino acid sequence of SEQ ID NO: 516, 528 or 709.
  • the variant TACI polypeptide containing the one or more amino acid modifications e.g.
  • the variant TACI polypeptide containing the one or more amino acid modifications as described has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 709.
  • the variant TACI polypeptide containing the one or more amino acid modifications as described has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 516.
  • the variant TACI polypeptide containing the one or more amino acid modifications (e.g. amino acid substitutions) as described has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 528.
  • a BIM that is a variant TACI polypeptide has one or more amino acid modification, e.g. substitution in a reference TACI polypeptide or specific binding fragment there of corresponding to position(s) 40, 59, 60, 61, 74, 75, 76, 77, 78, 79, 82, 83, 84, 85, 86, 87, 88, 92, 95, 97, 98, 99, 101, 102 and 103 with reference to numbering of SEQ ID NO:709.
  • a BIM that is a variant TACI polypeptide has one or more amino acid modification, e.g.
  • the reference TACI polypeptide includes the CRD1 domain or CRD2 domain, for example the reference TACI polypeptide is set forth in SEQ ID NO: 516 or SEQ ID NO:709.
  • a BIM that is a variant TACI polypeptide has one or more amino acid modification, e.g. substitution in a reference TACI polypeptide or specific binding fragment there of corresponding to position(s) 74, 75, 76, 77, 78, 79, 82, 83, 84, 85, 86, 87, 88, 92, 95, 97, 98, 99, 101, 102 and 103 with reference to numbering of SEQ ID NO:709.
  • a BIM that is a variant TACI polypeptide has one or more amino acid modification, e.g.
  • the reference TACI polypeptide includes only the CRD2 domain but lacks the CRD1 domain, for example the reference TACI polypeptide is set forth in SEQ ID NO: 528. Accordingly, in some embodiments, a BIM that is a variant TACI polypeptide includes a portion of the ECD sequence of a TACI polypeptide that includes the CRD2 domain but lacks the CRD1 domain.
  • 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).
  • a BIM that is a variant TACI polypeptide includes at least one amino acid substitution at position 75 with reference to numbering of SEQ ID NO:709.
  • the amino acid substitution at position 75 confers increased binding to BAFF or APRIL compared to the reference (e.g. wildtype or unmodified) TACI polypeptide not containing the amino acid substitution.
  • the substituted amino acid is an acidic amino acid or amide, such as to a different acidic amino acid or amide compared to the reference (e.g. wildtype or unmodified) TACI polypeptide.
  • the substituted amino acid at position 75 is a glutamic acid (Glu, E).
  • the substituted amino acid at position 75 is an asparatic acid (Asp, D). In some embodiments, the substituted amino acid at position 75 is an asparagine (Asn, N). In some embodiments, the substituted amino acid at position 75 is a glutamine (Gln, Q).
  • a BIM that is a variant TACI polypeptide includes at least one amino acid substitution at position 77 with reference to numbering of SEQ ID NO:709.
  • the amino acid substitution at position 77 confers increased binding to BAFF or APRIL compared to the reference (e.g. wildtype or unmodified) TACI polypeptide not containing the amino acid substitution.
  • the substituted amino acid at position 77 is an acidic amino acid or amide.
  • the substituted amino acid at position 77 is a glutamic acid (Glu, E).
  • the substituted amino acid at position 77 is an asparatic acid (Asp, D).
  • the substituted amino acid at position 77 is an asparagine (Asn, N).
  • the substituted amino acid at position 77 is a glutamine (Gln, Q).
  • a BIM that is a variant TACI polypeptide includes at least one amino acid substitution at position 78 with reference to numbering of SEQ ID NO:709.
  • the amino acid substitution at position 78 confers increased binding to BAFF or APRIL compared to the reference (e.g. wildtype or unmodified) TACI polypeptide not containing the amino acid substitution.
  • the substituted amino acid at position 78 is an aromatic amino acid, such as to a different aromatic amino acid compared to the reference (e.g. wildtype or unmodified) TACI polypeptide.
  • the substituted amino acid at position 78 is a phenyalanine (Phe, F).
  • the substituted amino acid at position 78 is a tyrosine (Tyr, Y).
  • the substituted amino acid at position 78 is a tryptophan (Trp, W).
  • a BIM that is a variant TACI polypeptide includes at least one amino acid substitution at position 84 with reference to numbering of SEQ ID NO:709.
  • the amino acid substitution at position 84 confers increased binding to BAFF or APRIL compared to the reference (e.g. wildtype or unmodified) TACI polypeptide not containing the amino acid substitution.
  • the substituted amino acid at position 84 is an acidic amino acid or amide.
  • the substituted amino acid at position 84 is a glutamic acid (Glu, E).
  • the substituted amino acid at position 84 is an asparatic acid (Asp, D).
  • the substituted amino acid at position 84 is an asparagine (Asn, N).
  • the substituted amino acid at position 84 is a glutamine (Gln, Q).
  • a BIM that is a variant TACI polypeptide includes at least one amino acid substitution at position 102 with reference to numbering of SEQ ID NO:709.
  • the amino acid substitution at position 102 confers increased binding to BAFF or APRIL compared to the reference (e.g. wildtype or unmodified) TACI polypeptide not containing the amino acid substitution.
  • the substituted amino acid at position 102 is an acidic amino acid or amide.
  • the substituted amino acid at position 102 is a glutamic acid (Glu, E).
  • the substituted amino acid at position 102 is an asparatic acid (Asp, D).
  • the substituted amino acid at position 102 is an asparagine (Asn, N).
  • the substituted amino acid at position 102 is a glutamine (Gln, Q).
  • a BIM that is a variant TACI polypeptide includes at least one amino acid substitution E74V. In some embodiments, a BIM that is a variant TACI polypeptide includes at least one amino acid substitution Q75E. In some embodiments, a BIM that is a variant TACI polypeptide includes at least one amino acid substitution K77E. In some embodiments, a BIM that is a variant TACI polypeptide includes at least one amino acid substitution F78Y. In some embodiments, a BIM that is a variant TACI polypeptide includes at least one amino acid substitution Y79F. In some embodiments, a BIM that is a variant TACI polypeptide includes at least one amino acid substitution L82H.
  • a BIM that is a variant TACI polypeptide includes at least one amino acid substitution L82P. In some embodiments, a BIM that is a variant TACI polypeptide includes at least one amino acid substitution R84G. In some embodiments, a BIM that is a variant TACI polypeptide includes at least one amino acid substitution R84L. In some embodiments, a BIM that is a variant TACI polypeptide includes at least one amino acid substitution R84Q. In some embodiments, a BIM that is a variant TACI polypeptide includes at least one amino acid substitution D85V. In some embodiments, a BIM that is a variant TACI polypeptide includes at least one amino acid substitution C86Y.
  • a BIM that is a variant TACI polypeptide includes at least one amino acid substitution Y102D. In some embodiments, a BIM that is a variant TACI polypeptide contains two or more amino acid substitutions of any two or more of the foregoing. In some embodiments, a BIM that is a variant TACI polypeptide includes one or more amino acid substitution that is a conservative amino acid substitution of any of the foregoing. In provided embodiments, a BIM that is a variant TACI polypeptide includes the at least one amino acid substitution in any reference TACI polypeptide sequence as described. In some embodiments, the at least one amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 516.
  • the at least one amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 528. In some embodiments, the at least one amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 718. In some embodiments, the at least one amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 719.
  • a BIM that is a variant TACI polypeptide includes the amino acid substitution E74V. In some embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitution Q75E. In some embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitution K77E. In some embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitution F78Y. In some embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitution Y79F. In some embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitution L82H.
  • a BIM that is a variant TACI polypeptide includes the amino acid substitution L82P. In some embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitution R84G. In some embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitution R84L. In some embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitution R84Q. In some embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitution D85V. In some embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitution C86Y.
  • a BIM that is a variant TACI polypeptide includes the amino acid substitution Y102D. In some embodiments, a BIM that is a variant TACI polypeptide contains two or more amino acid substitutions of any two or more of the foregoing. In some embodiments, a BIM that is a variant TACI polypeptide includes one or more of amino acid substitution that is a conservative amino acid substitution of any of the foregoing. In provided embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitution in any reference TACI polypeptide sequence as described. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 516.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 528. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 718. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 719.In some embodiments, the amino acid substitutions are D85E/K98T. In some embodiments, the amino acid substitutions are I87L/K98T. In some embodiments, the amino acid substitutions are R60G/Q75E/L82P. In some embodiments, the amino acid substitutions are R60G/C86Y. In some embodiments, the amino acid substitutions are W40R/L82P/F103Y.
  • the amino acid substitutions are W40R/Q59R/T61P/K98T. In some embodiments, the amino acid substitutions are L82P/I87L. In some embodiments, the amino acid substitutions are G76S/P97S. In some embodiments, the amino acid substitutions are K77E/R84L/F103Y. In some embodiments, the amino acid substitutions are Y79F/Q99E. In some embodiments, the amino acid substitutions are L83S/F103S. In some embodiments, the amino acid substitutions are K77E/R84Q. In some embodiments, the amino acid substitutions are K77E/A101D. In some embodiments, the amino acid substitutions are K77E/F78Y/Y102D.
  • the amino acid substitutions are Q75E/R84Q. In some embodiments, the amino acid substitutions are Q75R/R84G/I92V. In some embodiments, the amino acid substitutions are K77E/A101D/Y102D. In some embodiments, the amino acid substitutions are R84Q/S88N/A101D. In some embodiments, the amino acid substitutions are R84Q/F103V. In some embodiments, the amino acid substitutions are K77E/Q95R/A101D. In some embodiments, the amino acid substitutions are I87M/A101D. In provided embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitutions in any reference TACI polypeptide sequence as described.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 516. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 528. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 718. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 719.
  • a BIM that is a variant TACI polypeptide includes the amino acid substitutions K77E and F78Y (K77E/F78Y). In provided embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitutions in any reference TACI polypeptide sequence as described. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 516. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 528. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 718. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 719.
  • a BIM that is a variant TACI polypeptide includes the amino acid substitutions K77E and Y102D (K77E/Y102D). In provided embodiments, a BIM that is a variant TACI polypeptide includes the amino acid substitutions in any reference TACI polypeptide sequence as described. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 516. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 528. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 718. In some embodiments, the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 719.
  • a BIM that is a variant TACI polypeptide contains the amino acid substitutions F78Y and Y102D (F78Y/Y012D).
  • a BIM that is a variant TACI polypeptide includes the amino acid substitutions in any reference TACI polypeptide sequence as described.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 516.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 528.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 718.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 719.
  • a BIM that is a variant TACI polypeptide contains the amino acid substations K77E, F78Y and Y102D (K77E/F78Y/Y102D).
  • a BIM that is a variant TACI polypeptide includes the amino acid substitutions in any reference TACI polypeptide sequence as described.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 516.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 528.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 718.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 719.
  • a BIM that is a variant TACI polypeptide contains the amino acid substitutions Q75E/R84Q.
  • a BIM that is a variant TACI polypeptide includes the amino acid substitutions in any reference TACI polypeptide sequence as described.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 516.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 528.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 718.
  • the amino acid substitution is in the reference TACI sequence set forth in SEQ ID NO: 719.
  • a BIM that is a variant TACI polypeptide comprises any of the mutations listed in Table 2.
  • Table 2 also provides exemplary sequences by reference to SEQ ID NO of the reference (e.g., unmodified) TACI polypeptide, and exemplary variant TACI polypeptides.
  • 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. CRD
  • CRD adjacent N- and/or C-terminal amino acids of a given domain
  • the particular domain, such as the ECD domain or a portion thereof containing the CRD1/CRD2 or CRD2 only, of a variant TACI 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.
  • a BIM that is a variant TACI polypeptide comprises any of the mutations (amino acid substitutions) listed in Table 2.
  • the mutations (amino acid substitutions) are made in a reference TACI containing the sequence of amino acids set forth in SEQ ID NO: 709.
  • the mutations (amino acid substitutions) are made a reference TACI that contains the CRD1 and CRD2 domain of TACI, for example as set forth in SEQ ID NO: 516.
  • the mutations are made in a reference TACI that is further truncated by deletion of N-terminal and C-terminal amino acid residues to retain the CRD2, for example as set forth in SEQ ID NO: 528.
  • a BIM that is a variant TACI polypeptide 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.
  • This principle also extends to the terms “addition” and “deletion” of an amino acid residue which likewise do not imply a particular method of making.
  • the means by which the vTDs are designed or created is not limited to any particular method.
  • a wild-type or unmodified TD encoding nucleic acid is mutagenized from wild-type or unmodified TD genetic material and screened for desired specific binding activity, e.g. binding affinity, and/or alteration of NF- ⁇ B modulation or other functional activity.
  • a vTD 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.
  • a BIM that is a variant TACI polypeptide comprises an extracellular domain (ECD) sequences containing a CRD1 and CRD2, such as a variant TACI polypeptide set forth in any one of SEQ ID NOS: 517-527, 536, 537, 682-701.
  • ECD extracellular domain
  • a BIM that is a variant TACI 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 one of SEQ ID NOS: 517-527, 536, 537, 682-701, and retains the amino acid modification(s), e.g. substitution(s) therein not present in the reference (e.g., unmodified or wild-type) TAC.
  • a BIM that is a variant TACI polypeptide comprises a specific binding fragment of any one of SEQ ID NOS: 517-527, 536, 537, 682-701, in which the specific binding fragment binds BAFF, APRIL or a BAFF/APRIL heterotrimer, and contains a contiguous sequence therein that contains the amino acid modification(s), e.g. substitution (s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • a BIM that is a variant TACI polypeptide consists or consists essentially of a variant TACI extracellular domain (ECD) sequences set forth in any one of SEQ ID NOS: 517-527, 536, 537, 682-701.
  • ECD extracellular domain
  • a BIM that is a variant TACI polypeptide consists or consists essentially of 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 one of SEQ ID NOS: 517-527, 536, 537, 682-701, and retains the amino acid modification(s), e.g. substitution(s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • a BIM that is a variant TACI polypeptide consists or consists essentially of a specific binding fragment of any one of SEQ ID NOS: 517-527, 536, 537, 682-701, in which the specific binding fragment binds BAFF, APRIL or an APRIL/BAFF heterotrimer and contains a contiguous sequence therein that contains the amino acid modification(s), e.g. substitution (s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • a BIM that is a variant TACI polypeptide comprises an extracellular domain (ECD) sequences containing a CRD2 but lacking the CRD1 of a reference TACI polypeptide, such as a variant TACI polypeptide set forth in any one of SEQ ID NOS: 529-535, 538-550, 673-681.
  • ECD extracellular domain
  • a BIM that is a variant TACI 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 one of SEQ ID NOS: 529-535, 538-550, 673-681, and retains the amino acid modification(s), e.g. substitution(s) therein not present in the reference (e.g., unmodified or wild-type) TAC.
  • a BIM that is a variant TACI polypeptide comprises a specific binding fragment of any one of SEQ ID NOS: 529-535, 538-550, 673-681, in which the specific binding fragment binds BAFF, APRIL or a BAFF/APRIL heterotrimer, and contains a contiguous sequence therein that contains the amino acid modification(s), e.g. substitution (s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • a BIM that is a variant TACI polypeptide consists or consists essentially of the sequence set forth in any one of SEQ ID NOS: 529-535, 538-550, 673-681.
  • a BIM that is a variant TACI polypeptide consists or consists essentially of 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 one of SEQ ID NOS: 529-535, 538-550, 673-681, and retains the amino acid modification(s), e.g.
  • a BIM that is a variant TACI polypeptide consists or consists essentially of a specific binding fragment of any one of SEQ ID NOS: 529-535, 538-550, 673-681, in which the specific binding fragment binds BAFF, APRIL or a BAFF/APRIL heterotrimer, and contains a contiguous sequence therein that contains the amino acid modification(s), e.g. substitution (s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • a BIM that is a variant TACI polypeptide is one in which the variant TACI polypeptide comprises the sequence set forth in SEQ ID NO:535. In some embodiments, the variant TACI polypeptide consists essentially of the sequence set forth in SEQ ID NO:535. In some embodiments, the variant TACI polypeptide consists of the sequence set forth in SEQ ID NO:535.
  • a BIM that is a variant TACI polypeptide is one in which the variant TACI polypeptide comprises the sequence set forth in SEQ ID NO:541. In some embodiments, the variant TACI polypeptide consists essentially of the sequence set forth in SEQ ID NO:541. In some embodiments, the variant TACI polypeptide consists of the sequence set forth in SEQ ID NO:541.
  • a BIM that is a variant TACI polypeptide is one in which the variant TACI polypeptide comprises the sequence set forth in SEQ ID NO:542. In some embodiments, the variant TACI polypeptide consists essentially of the sequence set forth in SEQ ID NO:542. In some embodiments, the variant TACI polypeptide consists of the sequence set forth in SEQ ID NO:542.
  • a BIM that is a variant TACI polypeptide is one in which the variant TACI polypeptide comprises the sequence set forth in SEQ ID NO:688.
  • the variant TACI polypeptide consists essentially of the sequence set forth in SEQ ID NO:688. In some embodiments, the variant TACI polypeptide consists of the sequence set forth in SEQ ID NO:688.
  • a BIM that is a variant TACI polypeptide is one in which the variant TACI polypeptide is encoded by a sequence of nucleotides set forth in any of SEQ ID NOS: 552-562, 571 or 572.
  • the variant TACI polypeptide is encoded by a sequence of nucleotides 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 one of SEQ ID NOS: 552-562, 571 or 572, and retains the amino acid modification(s), e.g. substitution(s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • a BIM that is a variant TACI polypeptide is one in which the variant TACI polypeptide is encoded by a sequence of nucleotides set forth in any of SEQ ID NOS: 564-570 or 573-585.
  • the variant TACI polypeptide is encoded by a sequence of nucleotides 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 one of SEQ ID NOS: 564-570 or 573-585, and retains the amino acid modification(s), e.g. substitution(s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • the BIM is or contains a wild-type or unmodified ECD of TACI or a specific binding portion or fragment thereof containing at least one TD (e.g. at least one CRD, such as the CRD 1 and/or CRD2) that binds to APRIL, BAFF and/or an APRIL/BAFF heterotrimer.
  • at least one TD e.g. at least one CRD, such as the CRD 1 and/or CRD2
  • the BIM contains the ECD sequence set forth in SEQ ID NO: 709, (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:709 and that binds to APRIL, BAFF or an APRIL/BAFF heterotirmer, or (iii) is a fragment or portion of (i) or (ii) containing a CRD 1 and/or CRD2, in which the portion binds to APRIL, BAFF or an APRI/BAFF heterotrimer.
  • the BIM is a TACI sequence that consists or consists essentially of the sequence set forth in SEQ ID NO: 709. In some embodiments, the BIM is a TACI sequence containing residues 2-166 of SEQ ID NO:709 that lacks the N-terminal methionine as set forth in SEQ ID NO: 709. In some embodiments, the BIM is a TACI sequence that consists or consists essentially of the sequence set forth as amino acids 2-166 of SEQ ID NO: 709.
  • the BIM is or contains a binding portion of the wild-type or unmodified ECD of TACI or a specific binding portion or fragment thereof that contains the CRD1 and CRD2 of TACI and that binds to APRIL, BAFF and/or an APRIL/BAFF heterotrimer.
  • the BIM contains the sequence set forth in SEQ ID NO: 719, (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:719 and that binds to APRIL, BAFF or an APRIL/BAFF heterotrimer, or (iii) is a fragment or portion of (i) or (ii) containing a portion of the CRD1 and/or CRD2, in which the portion binds to APRIL, BAFF or an APRI/BAFF heterotrimer.
  • the BIM comprises the sequence set forth in SEQ ID NO: 719.
  • the BIM is a TACI sequence that consists or consists essentially of the sequence set forth in SEQ ID NO: 719.
  • the BIM contains the sequence set forth in SEQ ID NO: 718, (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:718 and that binds to APRIL, BAFF or an APRIL/BAFF heterotrimer, or (iii) is a fragment or portion of (i) or (ii) containing a portion of the CRD1 and/or CRD2, in which the portion binds to APRIL, BAFF or an APRI/BAFF heterotrimer.
  • the BIM comprises the sequence set forth in SEQ ID NO: 718.
  • the BIM is a TACI sequence that consists or consists essentially of the sequence set forth in SEQ ID NO: 718.
  • the BIM contains the sequence set forth in SEQ ID NO: 516, (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:516 and that binds to APRIL, BAFF or an APRIL/BAFF heterotrimer, or (iii) is a fragment or portion of (i) or (ii) containing a portion of the CRD1 and/or CRD2, in which the portion binds to APRIL, BAFF or an APRI/BAFF heterotrimer.
  • the BIM comprises the sequence set forth in SEQ ID NO:
  • the BIM is or contains a binding portion of the wild-type or unmodified ECD of TACI or a specific binding portion or fragment thereof that contains only the CRD2 of TACI and that binds to APRIL, BAFF and/or an APRIL/BAFF heterotrimer.
  • the BIM contains the sequence set forth in SEQ ID NO: 528, (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:528 and that binds to APRIL, BAFF or an APRIL/BAFF heterotrimer, or (iii) is a fragment or portion of (i) or (ii) containing a portion of the CRD2, in which the portion binds to APRIL, BAFF or an APRI/BAFF heterotrimer.
  • the BIM comprises the sequence set forth in SEQ ID NO: 528.
  • the BIM is a TACI sequence that consists or consists essentially of the sequence set forth in SEQ ID NO: 528.
  • the BIM is a variant TACI containing an ECD or specific binding portion or fragment thereof having a vTD containing one or more amino acid substitutions (mutations or replacements) relative to a reference TACI sequence.
  • the reference TACI sequence can include any as described in Section II above.
  • the one more amino acid substitutions can include any of the amino acid substitutions described in Section II above.
  • the BIM can be a variant TACI containing an ECD or specific binding fragment thereof having a vTD containing any of the amino acid substitutions set forth in Table 2.
  • the mutations are made in a reference TACI containing the sequence of amino acids set forth in SEQ ID NO: 709.
  • the mutations are made in a reference TACI that contains the CRD1 and CRD2 domain of TACI, for example as set forth in SEQ ID NO: 516. In some examples, the mutations are made in a reference TACI that is further truncated by deletion of N-terminal and C-terminal amino acid residues to retain the CRD2, for example as set forth in SEQ ID NO: 528.
  • the BIM comprises an extracellular domain (ECD) sequence containing a CRD1 and CRD2, such as a variant TACI polypeptide set forth in any one of SEQ ID NOS: 517-527, 536, 537, 682-701.
  • the BIM is a variant TACI polypeptide that 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 one of SEQ ID NOS: 517-527, 536, 537, 682-701, and retains the amino acid modification(s), e.g.
  • the BIM is a variant TACI polypeptide that comprises a specific binding fragment of any one of SEQ ID NOS: 517-527, 536, 537, 682-701, in which the specific binding fragment binds BAFF, APRIL or a BAFF/APRIL heterotimer and contains a contiguous sequence therein that contains the amino acid modification(s), e.g. substitution (s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • the BIM is a variant TACI polypeptide that consists or consists essentially of a variant TACI extracellular domain (ECD) sequences set forth in any one of SEQ ID NOS: 517-527, 536, 537, 682-701.
  • ECD extracellular domain
  • the BIM is a variant TACI polypeptide that consists or consists essentially of 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 one of SEQ ID NOS: 517-527, 536, 537, 682-701, and retains the amino acid modification(s), e.g. substitution(s) therein not present in the reference (e.g., unmodified or wild-type) TAC.
  • the BIM is a variant TACI polypeptide that consists or consists essentially of a specific binding fragment of any one of SEQ ID NOS: 517-527, 536, 537, 682-701, in which the specific binding fragment binds BAFF, APRIL or an APRIL/BAFF heterotrimer and contains a contiguous sequence therein that contains the amino acid modification(s), e.g. substitution (s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • the BIM is a variant TACI polypeptide that comprises an extracellular domain (ECD) sequences containing a CRD2 but lacking the CRD1 of a reference TACI polypeptide, such as a variant TACI polypeptide set forth in any one of SEQ ID NOS: 529-535, 538-550, 673-681, 769-794.
  • ECD extracellular domain
  • the BIM is a variant TACI polypeptide that 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 one of SEQ ID NOS: 529-535, 538-550, 673-681, and retains the amino acid modification(s), e.g. substitution(s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • the BIM is a variant TACI polypeptide that comprises a specific binding fragment of any one of SEQ ID NOS: 529-535, 538-550, 673-681, 769-794, in which the specific binding fragment binds BAFF, APRIL or a BAFF/APRIL heterotimer and contains a contiguous sequence therein that contains the amino acid modification(s), e.g. substitution (s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • the BIM is a variant TACI polypeptide that consists or consists essentially of the sequence set forth in any one of SEQ ID NOS: 529-535, 538-550, 673-681, 769-794.
  • the BIM is a variant TACI polypeptide that consists or consists essentially of a polypeptide sequence that exhibits at least 90% identity, at least 910% 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 one of SEQ ID NOS: 529-535, 538-550, 673-681, 769-794, and retains the amino acid modification(s), e.g.
  • the BIM is a variant TACI polypeptide that consists or consists essentially of a specific binding fragment of any one of SEQ ID NOS: 529-535, 538-550, 673-681, 769-794, in which the specific binding fragment binds BAFF, APRIL or a BAFF/APRIL heterotrimer, and contains a contiguous sequence therein that contains the amino acid modification(s), e.g. substitution (s) therein not present in the reference (e.g., unmodified or wild-type) TACI.
  • the BIM is a variant TACI polypeptide that comprises the sequence set forth in SEQ ID NO:535. In some embodiments, the BIM is a variant TACI polypeptide that consists essentially of the sequence set forth in SEQ ID NO:535. In some embodiments, the BIM is a variant TACI polypeptide that consists of the sequence set forth in SEQ ID NO:535.
  • the BIM is a variant TACI polypeptide that comprises the sequence set forth in SEQ ID NO:541. In some embodiments, the BIM is a variant TACI polypeptide that consists essentially of the sequence set forth in SEQ ID NO:541. In some embodiments, the BIM is a variant TACI polypeptide that consists of the sequence set forth in SEQ ID NO:541.
  • the BIM is a variant TACI polypeptide that comprises the sequence set forth in SEQ ID NO:542. In some embodiments, the BIM is a variant TACI polypeptide that consists essentially of the sequence set forth in SEQ ID NO:542. In some embodiments, the BIM is a variant TACI polypeptide consists of the sequence set forth in SEQ ID NO:542.
  • the BIM is a variant TACI polypeptide that comprises the sequence set forth in SEQ ID NO:688. In some embodiments, the BIM is a variant TACI polypeptide that consists essentially of the sequence set forth in SEQ ID NO:688. In some embodiments, the BIM is a variant TACI polypeptide that consists of the sequence set forth in SEQ ID NO:688.
  • the BIM is or contains a wild-type BCMA ECD or a specific binding portion or fragment thereof containing a TD (e.g. a CRD) that binds to APRIL, BAFF and/or an APRIL/BAFF heterotrimer.
  • the BIM is or contains a variant BCMA ECD or a specific binding portion or fragment thereof containing a TD (e.g. a CRD) that binds to APRIL, BAFF and/or an APRIL/BAFF heterotrimer.
  • the BIM is a BCMA polypeptide or variant thereof with any of the sequences set forth in Section II above (e.g. Table 1).
  • the BIM is or contains a wild-type or unmodified ECD of BCMA or a specific binding portion or fragment thereof containing a TD (e.g. a CRD) that binds to APRIL, BAFF and/or an APRIL/BAFF heterotrimer.
  • a TD e.g. a CRD
  • the BIM contains the ECD sequence set forth in SEQ ID NO: 710, (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:710 and that binds to APRIL, BAFF or an APRIL/BAFF heterotrimer, or (iii) is a fragment or portion of (i) or (ii) containing a CRD, in which the portion binds to APRIL, BAFF or an APRI/BAFF heterotrimer.
  • the BIM is a BCMA sequence that consists or consists essentially of the sequence set forth in SEQ ID NO: 710. In some embodiments, the BIM is a BCMA sequence containing residues 2-54 of SEQ ID NO:710 that lacks the N-terminal methionine as set forth in SEQ ID NO: 710. In some embodiments, the BIM is a BCMA sequence that consists or consists essentially of the sequence set forth as amino acids 2-54 of SEQ ID NO: 710.
  • the BIM contains the sequence set forth in SEQ ID NO: 356, (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:356 and that binds to APRIL, BAFF or an APRIL/BAFF heterotrimer, or (iii) is a fragment or portion of (i) or (ii) containing a portion of a CRD, in which the portion binds to APRIL, BAFF or an APRI/BAFF heterotrimer.
  • the BIM comprises the sequence set forth in SEQ ID NO: 356.
  • the BIM is a BCMA sequence that consists or consists essentially of the sequence set forth in SEQ ID NO: 356.
  • the BIM is a variant BCMA containing an ECD or specific binding portion or fragment thereof having a vTD containing one or more amino acid substitutions (mutations or replacements) relative to a reference BCMA sequence.
  • the reference BCMA sequence can include any as described in Section II above.
  • the one more amino acid substitutions can include any of the amino acid substitutions described in Section II above.
  • the BIM can be a variant BCMA containing an ECD or specific binding fragment thereof having a vTD containing any of the amino acid substitutions set forth in Table 1.
  • the mutations are made in a reference BCMA containing the sequence of amino acids set forth in SEQ ID NO: 710.
  • the mutations are made in a reference BCMA that contains the CRD domain of BCMA.
  • the mutations are made in a reference BCMA set forth in SEQ ID NO: 356.
  • the BIM comprises a variant BCMA polypeptide set forth in any one of SEQ ID NOS: 357-435.
  • the BIM is a variant BCMA polypeptide that comprises a polypeptide sequence that exhibits at least 90% identity, at least 910% 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 one of SEQ ID NOS: 357-435, and retains the amino acid modification(s), e.g. substitution(s) therein not present in the reference (e.g., unmodified or wild-type) BCMA.
  • the BIM is a variant BCMA polypeptide that comprises a specific binding fragment of any one of SEQ ID NOS: 357-435, in which the specific binding fragment binds BAFF, APRIL or a BAFF/APRIL heterotimer and contains a contiguous sequence therein that contains the amino acid modification(s), e.g. substitution (s) therein not present in the reference (e.g., unmodified or wild-type) BCMA.
  • the BIM is a variant BCMA polypeptide that consists or consists essentially of a variant BCMA extracellular domain (ECD) sequences set forth in any one of SEQ ID NOS: 357-435.
  • the BIM is a variant BCMA polypeptide that consists or consists essentially of a polypeptide sequence that exhibits at least 90% identity, at least 910% 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 one of SEQ ID NOS: 357-435, and retains the amino acid modification(s), e.g.
  • the BIM is a variant BCMA polypeptide that consists or consists essentially of a specific binding fragment of any one of SEQ ID NOS: 357-435, in which the specific binding fragment binds BAFF, APRIL or an APRIL/BAFF heterotrimer and contains a contiguous sequence therein that contains the amino acid modification(s), e.g. substitution (s) therein not present in the reference (e.g., unmodified or wild-type) BCMA.
  • the BIM is a variant BCMA polypeptide that comprises the sequence set forth in SEQ ID NO:381. In some embodiments, the BIM is a variant BCMA polypeptide that consists essentially of the sequence set forth in SEQ ID NO:381. In some embodiments, the BIM is a variant BCMA polypeptide that consists of the sequence set forth in SEQ ID NO:381.
  • the BIM is a variant BCMA polypeptide that comprises the sequence set forth in SEQ ID NO:405. In some embodiments, the BIM is a variant BCMA polypeptide that consists essentially of the sequence set forth in SEQ ID NO:405. In some embodiments, the BIM is a variant BCMA polypeptide that consists of the sequence set forth in SEQ ID NO:405.
  • the BIM is a variant BCMA polypeptide that comprises the sequence set forth in SEQ ID NO:406. In some embodiments, the BIM is a variant BCMA polypeptide that consists essentially of the sequence set forth in SEQ ID NO:406. In some embodiments, the BIM is a variant BCMA polypeptide that consists of the sequence set forth in SEQ ID NO:406.
  • the BIM is a variant BCMA polypeptide that comprises the sequence set forth in SEQ ID NO:410. In some embodiments, the BIM is a variant BCMA polypeptide that consists essentially of the sequence set forth in SEQ ID NO:410. In some embodiments, the BIM is a variant BCMA polypeptide that consists of the sequence set forth in SEQ ID NO:410.
  • the BIM is a variant BCMA polypeptide that comprises the sequence set forth in SEQ ID NO:411. In some embodiments, the BIM is a variant BCMA polypeptide that consists essentially of the sequence set forth in SEQ ID NO:411. In some embodiments, the BIM is a variant BCMA polypeptide that consists of the sequence set forth in SEQ ID NO:411.
  • the provided immunomodulatory protein contains a TIM that binds to a T cell stimulatory receptor or to a ligand of a T cell stimulatory receptor.
  • the T cell stimulatory receptor comprises a cytoplasmic region containing an immunoreceptor tyrosine-based activation motif (ITAM) or a cytoplasmic region that interacts with one or more adaptor protein involved in a signal transduction pathway in a cell to induce, mediate or potentiate activation of a T cell.
  • the adaptor protein contains a binding domain specific to a phosphotyrosine residue in a cytoplasmic region of stimulatory receptor.
  • the T cell stimulatory receptor includes a component of a TCR complex or is a co-receptor or costimulatory molecule that augments or enhances TCR signaling.
  • the T cell stimulatory receptor is a TCR, CD3, CD4, CD8, CD28, ICOS or CD2, including any mammalian orthologs thereof.
  • the T cell stimulatory receptor target is a human TCR, human CD3, human CD4, human CD8, human CD28, human ICOS or human CD2.
  • the T cell stimulatory receptor is expressed on a human T cell.
  • the TIM binds directly to a T cell stimulatory receptor, such as directly to a component of a TCR complex or a co-receptor or costimulatory molecule that augments or enhances TCR signaling.
  • the TIM binds to a TCR, CD3, CD4, CD8, CD28, ICOS or CD2, including any mammalian orthologs thereof.
  • the TIM binds to a human TCR, human CD3, human CD4, human CD8, human CD28, human ICOS or human CD2.
  • the TIM binds to a ligand of a T cell stimulatory receptor. In some embodiments, the TIM binds to a ligand of a component of a TCR complex or a ligand of a co-receptor or costimulatory molecule that augments or enhances TCR signaling. In some embodiments, the TIM binds to a ligand of a TCR, CD3, CD4, CD8, CD28, ICOS or CD2 molecule, including such molecules expressed on a T cell, e.g. a human T cell. In some embodiments, the TIM binds to a ligand of CD28, such as a ligand of CD28 expressed on a T cell, e.g. a human T cell. In some embodiments, the ligand is a CD80 or a CD86, such as a human CD80 or human CD86. In some embodiments, the ligand is expressed on an APC.
  • the TIM is an antibody or antigen-binding fragment that binds to a T cell stimulatory receptor or binds to a ligand of a T cell stimulatory receptor.
  • the TIM is an antibody or antigen-binding fragment that binds to a TCR, CD3, CD4, CD8, CD28, ICOS or CD2, including any mammalian orthologs thereof.
  • the antibody or antigen-binding fragment binds to a human TCR, human CD3, human CD4, human CD8, human CD28, human ICOS or human CD2, including such molecules expressed on a human T cell.
  • the antibody or antigen-binding fragment binds to CD80 or CD86.
  • the antibody or antigen-binding fragment binds to a human CD80 or human CD86, including such molecules expressed on a human APC.
  • the TIM is or contains a binding partner of a T cell stimulatory receptor or a ligand of a T cell stimulatory receptor.
  • molecules that can be used as a TIM are extracellular domains of IgSF protein member, particularly IgSF members that are T cell stimulatory receptors or their ligands.
  • the TIM is or contains an IgD of an IgSF family member that binds to an T cell stimulatory receptor, such as binds to TCR, CD3, CD4, CD8, CD28, ICOS or CD2, or is a specific fragment or vIgD thereof that binds to the T cell stimulatory receptor.
  • the T cell stimulatory receptor is CD28 or ICOS and the TIM binds to CD28 or ICOS.
  • Exemplary IgSF family members that are binding partners of or that bind to CD28 or ICOS include, for example, CD80, CD86 and ICOSL, such as human CD80, CD86 or ICOSL.
  • the TIM is or contains an IgD of a wild-type CD80, CD86 or ICOSL or is or contains a vIgD thereof, wherein the TIM specifically binds to CD28.
  • the TIM is or contains an IgD of an IgSF family member that binds to a ligand of a T cell stimulatory receptor, such as binds to CD80 or CD86, or is a specific fragment or vIgD thereof that binds to the ligand of the T cell stimulatory receptor.
  • IgSF family members that are binding partners of or that bind to CD80 or CD86 include, for example, CTLA-4, such as human CTLA-4.
  • the TIM is or contains an IgD of a wild-type CTLA-4 or is or contains a vIgD thereof, wherein the TIM specifically binds to CD80 or CD86.
  • Exemplary sequences for inclusion as a TIM in the provided multi-domain immunomodulatory proteins include molecules described in International PCT published Appl. No. WO2019/074983.
  • the multi-domain immunomodulatory protein provided herein are soluble proteins and/or do not contain a portion that includes a transmembrane domain.
  • cell surface proteins including proteins of the IgSF, typically have an intracellular domain, a transmembrane domain, 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.
  • the TIM lacks a transmembrane domain or a portion of the transmembrane domain of an IgSF member.
  • the TIM lacks the intracellular (cytoplasmic) domain or a portion of the intracellular domain of an IgSF member.
  • the TIM only contains the ECD domain or a portion thereof containing an IgSF domain, such an IgV domain, or specific binding fragments thereof.
  • the TIM is or contains an ECD of an IgSF receptor, or a specific binding portion or fragment thereof containing at least one IgD (e.g. IgV), that binds to a ligand of the T cell stimulatory receptor.
  • the TIM can contain an ECD of CTLA-4, or a specific binding portion or fragment of CTLA-4 containing at least one IgD (e.g. IgV), that binds to CD80 or CD86.
  • the TIM consists or consists essentially of an ECD of a an IgSF receptor, or a specific binding portion or fragment thereof containing at least one IgD (e.g.
  • the TIM is less than the full length sequence of the ECD of the receptor of the ligand of the T cell stimulatory receptor.
  • the TIM is or only contains one vIgD (e.g. IgV) or a specific binding fragment of the only one vIgD (e.g. IgV).
  • the TIM consists or consists essentially of an IgV of a receptor of the ligand of the T cell stimulatory receptor, such as consists or consists essentially of only the IgV of CTLA-4.
  • the sequence of the TIM containing an ECD or binding portion or fragment thereof containing a IgD is a mammalian sequence that includes, but is not limited to, human, mouse, cynomolgus monkey, or rat.
  • the TIM sequence is human and/or binds a human protein.
  • the vIgD is an affinity-modified domain that exhibits increased binding activity, such as increased binding affinity, for the T cell stimulatory receptor or the ligand of the T cell stimulatory receptor compared to the binding activity of the unmodified or wild-type IgD for the same molecule.
  • the TIM contains a vIgD with one or more amino acid substitutions compared to an IgD of an IgSF member, e.g. CTLA-4 o, in which, the one or more amino acid substitutions confer or result in increased binding affinity to a cognate binding partner that is a T cell stimulatory receptor or a ligand of the T cell stimulatory receptor.
  • the TIM is or contains a vIgD that contains one or more amino acids modifications, such as one or more substitutions (alternatively, “mutations” or “replacements”), deletions or additions, in an IgD relative to a wild-type or unmodified IgD of a binding partner of the T cell stimulatory receptor or a ligand of the T cell stimulatory receptor.
  • the vIgD contains 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, such as amino acid substitutions, deletions or additions in an IgD domain of an IgSF binding partner of a T cell stimulatory receptor or a ligand of a T cell stimulatory receptor.
  • the modifications can be in the IgV domain or the IgC domain.
  • the vIgD 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 modifications (e.g., substitutions) in the IgV domain or specific binding fragment thereof.
  • the vIgD 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 modifications (e.g., substitutions) in the IgC domain or specific binding fragment thereof.
  • the vIgD has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the wild-type or unmodified IgD or specific binding fragment thereof.
  • Non-limiting examples of a TIM in the provided multidomain immunomodulatory proteins are described in the following subsections. Any of the described TIMs herein can be combined with a BIM as described in Section III.A.
  • Ligand-Binding TIM e.g. CTLA-4
  • immunomodulatory proteins containing a T cell molecule that binds to a ligand of a T cell stimulatory receptor.
  • the T cell stimulatory receptor is CD28, e.g. human CD28
  • the ligand of the T cell stimulatory receptor is CD80 or CD86, e.g. human CD80 or human CD86.
  • the TIM of the immunomodulatory protein binds to the extracellular portion or ectodomain of CD80 or CD86.
  • the TIM binds to CD80 or CD86 on the surface of a cell, such as on the surface of an APC.
  • the TIM is an antibody that binds CD80 or CD86 or is an antigen-binding antibody fragment thereof (e.g. Fab or scFv). In some embodiments, the antibody or antigen-binding antibody fragment binds human CD80 or human CD86. In some embodiments, the antibody is a single chain variable fragment (e.g. scFv) containing a V H and V L of an anti-CD80 antibody or antigen-binding fragment or an anti-CD86 antibody or antigen-binding fragment.
  • scFv single chain variable fragment
  • the TIM is or contains an IgD (e.g. IgV) or a specific binding fragment thereof, such as an unmodified or wild-type IgD or a vIgD or a specific binding fragment thereof, of an IgSF family member that binds CD80 or CD86, such as human CD80 or human CD86.
  • the TIM is or contains one or more IgD that is an IgD, or a vIgD thereof, of a CTLA-4 polypeptide, such as a wild-type CTLA-4, e.g. a human CTLA-4.
  • the TIM contains one or more IgD (e.g.
  • IgV that is an vIgD containing one or more amino acid modifications (e.g., substitutions, deletions or additions) compared to an IgD of a wild-type or unmodified CTLA-4, which, in some aspects, result in increased binding of the TIM to CD80 or CD86.
  • Exemplary IgDs or vIgDs of CTLA-4 binding partners for inclusion as an TIM in the provided immunomodulatory proteins are described.
  • the TIM is or contains a vIgD polypeptide that exhibit increased binding activity, such as binding affinity, for CD80 or CD86 compared to a corresponding wild-type or unmodified IgD.
  • Exemplary IgDs or vIgDs of CTLA-4 for use as a TIM in the provided multi-domain immunomodulatory proteins include molecules described in International PCT published Appl. No. WO2019/074983.
  • CTLA-4 has been exploited as a therapeutic drug for treating autoimmune disease by attenuating T cell activation through modulation of CD80 and/or CD86 interactions.
  • abatacept and belatacept are FDA approved therapeutics for use in rheumatoid arthritis and transplant setting, respectively.
  • Abatacept is wild-type CTLA-4 IgSF domain fused to an Fc portion of an antibody.
  • the sequence of the CTLA-4 portion of abatacept is set forth in SEQ ID NO: 1.
  • Belatacept is a modified variant of CTLA-4 IgSF domain, containing a substitution of tyrosine for the alanine at position 31 and a glutamic acid for the leucine at position 106 (A31Y/L106E), corresponding to positions 31 and 106 of the wild-type reference CTLA-4 ECD sequence set forth in SEQ ID NO: 1, to confer increased affinity toward CD80 and CD86 ligands (Kremer et al., N Engl J Med. 2003; 349(20):1907-1915; Larsen et al, Am J Transplant. 2005; 5(3):443-453).
  • the sequence of the CTLA-4 portion of belatacept is set forth as SEQ ID NO: 672.
  • the TIM is not the full length sequence of the CTLA-4.
  • the TIM is a soluble polypeptide, is not membrane-expressed and/or lacks the transmembrane and/or cytoplasmic domain of CTLA-4.
  • the TIM only contains an extracellular domain (ECD) or a specific binding fragment thereof containing a IgD or vIgD, such as only contains an IgV domain or an IgC domain or specific binding fragment thereof, or combinations thereof.
  • the TIM is or contains the ECD sequence set forth in SEQ ID NO:1 or is a specific binding fragment thereof. In some embodiments, the TIM is or contains the ECD sequence set forth in SEQ ID NO:2 or is a specific binding fragment thereof. In some embodiments, the TIM is or contains an IgD (e.g. IgV) sequence of CTLA-4, such as human CTLA-4. In some embodiments, the TIM is or contain an IgD (e.g. IgV) sequence set forth in SEQ ID NO: 191, or is a specific binding fragment thereof.
  • IgD e.g. IgV
  • the TIM contains the sequence set forth in SEQ ID NO: 1, (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:1 and that binds to CD80 or CD86, or (iii) is a fragment or portion of (i) or (ii) containing an IgD (e.g. IgV) in which the portion binds to CD80 or CD86.
  • the TIM comprises the sequence set forth in SEQ ID NO: 1.
  • the TIM is a CTLA-4 sequence that consists or consists essentially of the sequence set forth in SEQ ID NO: 1.
  • the TIM contains the sequence set forth in SEQ ID NO: 2, (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:2 and that binds to CD80 or CD86, or (iii) is a fragment or portion of (i) or (ii) containing an IgD (e.g. IgV) in which the portion binds to CD80 or CD86.
  • the TIM comprises the sequence set forth in SEQ ID NO: 2.
  • the TIM is a CTLA-4 sequence that consists or consists essentially of the sequence set forth in SEQ ID NO: 2.
  • the TIM contains the sequence set forth in SEQ ID NO: 191, (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: 191 and that binds to CD80 or CD86, or (iii) is a fragment or portion of (i) or (ii) containing an IgD (e.g. IgV) in which the portion binds to CD80 or CD86.
  • the TIM comprises the sequence set forth in SEQ ID NO: 191.
  • the TIM is a CTLA-4 sequence that consists or consists essentially of the sequence set forth in SEQ ID NO: 191.
  • the immunomodulatory protein contains a TIM that is or contains a vIgD containing one or more amino acid modifications, e.g. substitutions, in an IgD of a wild-type or unmodified CTLA-4.
  • modifications provided herein can be in an TIM containing an unmodified IgD set forth in SEQ ID NO: 1, 2 or 191 or in a sequence that has 85%, 85%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 1, 2 or 191.
  • an TIM containing a vIgD of CTLA-4 has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the sequence set forth in any of SEQ ID NOs: 1, 2 or 191.
  • the TIM is or contains a vIgD that is an affinity-modified IgSF domain that has an increased binding activity, such as binding affinity, for CD80 or CD86 relative to the binding activity of the wild-type or unmodified IgD for CD80 or CD86.
  • the increase in binding activity, e.g. binding affinity, for CD80 or CD86 is increased at least about 5%, such as at least about 10%, 15%, 20%, 25%, 35%, 50%, 75%, 100%, 200% or more.
  • the increase in binding activity e.g.
  • binding affinity 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 wild-type or unmodified IgD has the same sequence as the vIgD except that it does not contain the one or more amino acid modifications (e.g. substitutions).
  • the equilibrium dissociation constant (K d ) of the TIM to CD80 or CD86 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 or less.
  • the TIM binds to CD80 or CD86 with a K d of from or from about 100 pm to 5000 pm, 100 pm to 2000 pm, 100 pm to 1500 pm, 100 pm to 1000 pm, 100 pm to 800 pm, 100 pm to 500 pm, 100 pm to 400 pm, 400 pm to 4000 pm, 400 pm to 2000 pm, 400 pm to 1500 pm, 400 pm to 1000 pm, 400 pm to 800 pm, 400 pm to 500 pm, 500 pm to 5000 pm, 500 pm to 2000 pm, 500 pm to 1500 pm, 500 pm to 1000 pm, 500 pm to 800 pm, 800 pm to 5000 pm, 800 pm to 2000 pm, 800 pm to 1500 pm, 800 pm to 1000 pm, 1000 pm to 5000 pm, 1000 pm to 2000 pm, 1000 pm to 1500 pm, 1500 pm to 5000 pm, 1500 to 2000 pm to 2000 pm to 50000 pm.
  • the TIM binds to CD80 or CD86 with a K d of less than 200 pM, 300 pM, 400 pM, 500 pM. In some embodiments, the TIM binds to CD80 or CD86 with a K d of greater than or greater than about 500 pm but less than or less than about 2000 pm, such as from or from about 500 pm to 1500 pm, 500 pm to 1250 pm, 500 pm to 1000 pm, 500 pm to 750 pm, 750 pm to 1500 pm, 750 pm to 1250 pm, 750 pm to 1000 pm, 1000 pm to 2000 pm, 1000 pm to 1500 pm or 1500 pm to 2000 pm.
  • amino acid modification(s) present in a vIgD of a CTLA-4 ECD or an IgD (e.g. IgV) thereof are designated by amino acid position number corresponding to the numbering of positions of the unmodified ECD sequence set forth in SEQ ID NO:1 or 2. It is within the level of a skilled artisan to identify the corresponding position of a modification, e.g. amino acid substitution, in an ECD or a portion thereof containing an IgSF domain (e.g. IgV) thereof, such as by alignment of a reference sequence with SEQ ID NOs: 1 or 2. In the listing of modifications throughout this disclosure, the amino acid position is indicated in the middle, with the corresponding unmodified (e.g.
  • the TIM contains a vIgD that 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 modifications, e.g. substitutions.
  • the one or more amino acid modifications, e.g. substitutions can be in the ectodomain (extracellular domain) of the wild-type or unmodified CTLA-4.
  • the one or more amino acid modifications, e.g. substitutions are in the ECD domain of CTLA-4 or a specific binding fragment thereof.
  • the one or more amino acid modifications, e.g. substitutions are in the IgV domain of CTLA-4 or a specific binding fragment thereof.
  • the one or more amino acid modifications are in an IgC domain of CTLA-4 or a specific binding fragment thereof. In some embodiments, the one or more amino acid modifications, e.g. substitutions, are in the IgV domain of CTLA-4 or a specific binding fragment thereof and in an IgC domain or domains of CTLA-4 or a specific binding fragment thereof.
  • the TIM is or contains a vIgD that has one or more amino modification, e.g. substitutions, in an unmodified IgD of CTLA-4 or specific binding fragment thereof corresponding to position(s) 6, 10, 12, 14, 15, 16, 18, 19, 20, 22, 24, 26, 27, 28, 29, 30, 31, 33, 35, 37, 38, 41, 42, 43, 45, 46, 47, 48, 53, 54, 55, 56, 58, 59, 61, 63, 64, 65, 67, 69, 71, 72, 73, 75, 76, 82, 85, 86, 87, 89, 91, 93, 95, 96, 97, 98, 99, 105, 106, 108, 110, 113, 115, 116, 117,118, 119, 120, 121, 122, 124, 125 and/or 126 with reference to positions set forth in SEQ ID NO:1.
  • the TIM contains a vIgD that has one or more amino acid modification, e.g. substitutions, in an unmodified CTLA-4 or specific binding fragment thereof corresponding to position(s) 12, 18, 26, 29, 31, 53, 56, 58, 63, 72, 98, 99, 105, 106, and/or 117 with reference to positions set forth in SEQ ID NO:1 or 2.
  • a vIgD that has one or more amino acid modification, e.g. substitutions, in an unmodified CTLA-4 or specific binding fragment thereof corresponding to position(s) 12, 18, 26, 29, 31, 53, 56, 58, 63, 72, 98, 99, 105, 106, and/or 117 with reference to positions set forth in SEQ ID NO:1 or 2.
  • the TIM is or contains a vIgD of CTLA-4 that has one or more amino acid modifications selected from L12F, L12H, L12P, 118A, I18F, I18N, I18T, I18V, A26D, A26S, A26T, G29R, G29W, A31Y, T53S, M56K, M56L, M56R, M56T, M56V, N58D, N58S, L63H, L63P, S72G, S72T, L98Q, L98R, M99I, M99L, Y105F, Y105L, L106E, L106I, L106R, I117E, I117L, I117M, and/or I117T, or a conservative amino acid substitution thereof.
  • the TIM is or contains a vIgD of CTLA-4 that has one or more amino acid modifications selected from A6T, V10A, L12F, L12H, L12I, L12P, S14N, S15P, R16C, R16G, R16H, I18A, I18F, I18N, I18T, I18V, A19V, S20N, V22A, V22I, E24Q, A26D, A26S, A26T, S27P, P28L, G29R, G29W, K30R, A31Y, E33M, E33V, R35K, T37S, V38I, Q41L, A42S, A42T, A42V, D43N, Q45H, V46E, T47A, E48R, T53S, Y54F, M55R, M55T, M55V, M56K, M56L, M56R, M56T, M56V, N58D, N58
  • the TIM is or contains a vIgD that has one or more amino acid modification from L12F, L12H, L12I, L12P, 118A, 118F, 118N, 118T, 118V, A26D, A26S, A26T, G29R, G29W, E33M, E33V, T53S, M55R, M55T, M55V, M56K, M56L, M56R, M56T, M56V, N58D, N58S, L63H, L63P, S72G, S72T, M87A, M87K, M87T, M87V, L98Q, L98R, M99I, M99L, Y105F, Y105L, L106I, L106N, L106R, L106V, I117E, I117K, I117L, I117M, and/or I117T, or a conservative amino acid substitution thereof.
  • the TIM is or contains a vIgD that has one or more amino acid modifications selected from I12F, L12P, I18T, A26T, G29W, T53S, M55T, M56K, M56T, N58S, S72G, M99L, L63P, L98Q, Y105L, L106I, and/or I117L, or a conservative amino acid substitution thereof.
  • the TIM is or contains a vIgD has one or more amino acid modifications selected from L12P, I18T, A26T, G29W, A31Y, T53S, M55T, M56K, N58S, S72G, M99L, L63P, L98Q, Y105L, L106E, L106I, and/or I117L, or a conservative amino acid substitution thereof.
  • the TIM is or contains a vIgD that has one or more amino acid modifications selected from A26T, G29W, L63P, S72G, L98Q, M99L, Y105L and/or L106I, or a conservative amino acid substitution thereof.
  • the TIM is or contains a vIgD that has two or more amino acid modifications selected from among A6T, V10A, L12F, L12H, L12I, L12P, S14N, S15P, R16C, R16G, R16H, I18A, I18F, I18N, I18T, I18V, A19V, S20N, V22A, V22I, E24Q, A26D, A26S, A26T, S27P, P28L, G29R, G29W, K30R, A31Y, E33M, E33V, R35K, T37S, V38I, Q41L, A42S, A42T, A42V, D43N, Q45H, V46E, T47A, E48R, T53S, Y54F, M55R, M55T, M55V, M56K, M56L, M56R, M56T, M56V, N58D, N58S, E
  • the TIM is or contain a vIgD of CTLA-4 that has an amino acid substitution in an unmodified or wild-type CTLA-4 polypeptide or specific binding fragment thereof corresponding to A26T, G29W, T53S, L63P, S72G, L98Q, M99L, Y105L and/or L106I.
  • the TIM is or contains a vIgD of CTLA-4 that contains the amino acid substitutions A26T/G29W, A26T/T53S, A26T/L63P, A26T/S72G, A26T/L98Q, A26T/M99L, A26T/Y105L, A26T/L106I, A26T/G29W, G29W/T53S, G29W/L63P, G29W/S72G, G29W/L98Q, G29W/M99L, G29W/Y105L, G29W/L106I, A26T/T53S, G29W/T53S, T53S/L63P, T53S/S72G, T53S/L98Q, T53S/M99L, T53S/Y105L, or T53S/L106I, A26T/L63P, G29W/L63P, T53S/L63P, L63P/S72G, L
  • the amino acid modification(s), e.g. substitutions(s) are A31Y/L106E, A6T/A26T/M55T/M99L/Y105L, V10A/G29W/T53S/M56K/L63P/L98Q/Y105L/P121S, V10A/L63P/D64V/S72G/L98Q/M99L/Y105L, V10A/L63P/L98Q/Y105L, L12F/R16H/G29W/M56T/L98Q/Y105L, L12F/A26T/L63P/L98Q/Y105L/L106R, L12F/K30R/S72G/Q82R/L98Q/M99L/Y105L, L12H/I18V/A42T/M55T/N58D/L98R/Y105L/L106I/P121S, L12H/E33M/L98Q/Y105L,
  • the TIM is or contains a vIgD of CTLA-4 that additionally includes the amino acid modifications C122S with reference to positions set forth in SEQ ID NO:1 or 2.
  • the TIM is or contains a vIgD of CTLA-4 that includes the amino acid modifications C122S with reference to positions set forth in SEQ ID NO:1 or 2. In some embodiments, the TIM has the sequence of amino acids set forth in SEQ ID NO: 668.
  • the TIM is or contains an IgD (e.g. IgV) of wild-type CTLA-4 set forth in Table 3 or a vIgD thereof comprising any of the modifications (e.g. substitutions) listed in Table 3.
  • Table 3 also provides exemplary sequences by reference to SEQ ID NO for the extracellular domain (ECD) or IgV domain of CTLA-4.
  • ECD extracellular domain
  • IgV domain of CTLA-4 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 also can be included in a sequence of an TIM, such as to ensure proper folding of the domain when expressed.
  • the particular domain, such as the IgV domain, of a variant CTLA-4 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 TIM is or contains a variant CTLA-4 ECD set forth in any one of SEQ ID NOS: 3-190.
  • the TIM is or contains a 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 one of SEQ ID NOS: 3-190 and contains the amino acid modification(s), e.g. substitution(s) not present in the wild-type or unmodified CTLA-4, e.g. not present in SEQ ID NO:1 or 2.
  • the TIM is or contains a specific binding fragment of any of the ECD sequences set forth in any one of SEQ ID NOS: 3-190 and that contains the amino acid modification(s), e.g. substitution(s) not present in the wild-type or unmodified CTLA-4, e.g. not present in SEQ ID NO:1 or 2.
  • the TIM is or contains a variant IgV sequence set forth in any one of SEQ ID NOS: 192-355. In some embodiments, the TIM is or contains a 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 any one of SEQ ID NOS: 192-355 and contains the amino acid modification(s), e.g., substitution(s), not present in the wild-type or unmodified CTLA-4, e.g. not present in SEQ ID NO:191.
  • amino acid modification(s) e.g., substitution(s)
  • the TIM is a specific binding fragment of any of the IgV sequences set forth in any one of SEQ ID NOS: 192-355 and that contains the amino acid modification(s), e.g. substitution(s) not present in the wild-type or unmodified CTLA-4, e.g. set forth in SEQ ID NO:191.
  • the TIM is a variant CTLA-4 polypeptide that comprises the sequence set forth in SEQ ID NO:92. In some embodiments, the TIM is a variant CTLA-4 polypeptide that consists essentially of the sequence set forth in SEQ ID NO:92. In some embodiments, the TIM is a variant CTLA-4 polypeptide that consists of the sequence set forth in SEQ ID NO:92.
  • the TIM is a variant CTLA-4 polypeptide that comprises the sequence set forth in SEQ ID NO: 113. In some embodiments, the TIM is a variant CTLA-4 polypeptide that consists essentially of the sequence set forth in SEQ ID NO: 113. In some embodiments, the TIM is a variant CTLA-4 polypeptide that consists of the sequence set forth in SEQ ID NO: 113.
  • the TIM is a variant CTLA-4 polypeptide that comprises the sequence set forth in SEQ ID NO: 165. In some embodiments, the TIM is a variant CTLA-4 polypeptide that consists essentially of the sequence set forth in SEQ ID NO: 165. In some embodiments, the TIM is a variant CTLA-4 polypeptide that consists of the sequence set forth in SEQ ID NO: 165.
  • the TIM is a variant CTLA-4 polypeptide that comprises the sequence set forth in SEQ ID NO: 186. In some embodiments, the TIM is a variant CTLA-4 polypeptide that consists essentially of the sequence set forth in SEQ ID NO: 186. In some embodiments, the TIM is a variant CTLA-4 polypeptide that consists of the sequence set forth in SEQ ID NO: 186.
  • the multi-domain immunomodulatory proteins containing one or more BIM and one or more TIM provided herein can be formatted in a variety of ways, including as a single chain polypeptide fusion or as a multimeric (e.g. dimeric, trimeric, tetrameric, or pentameric) molecules.
  • the particular format is chosen such that the BIM of the immunomodulatory protein specifically binds to a ligand of a B cell stimulatory receptor and the TIM specifically binds to a T cell stimulatory receptor or a ligand of a T cell stimulatory receptor.
  • the particular format is chosen to effect attenuation of an activity of the T cell stimulatory receptor and the B cell stimulatory receptor, such as to reduce or decrease an immune response mediated by T cells and B cells, respectively.
  • the modular format of the provided immunomodulatory proteins provides flexibility for engineering or generating immunomodulatory proteins for modulating activity at an immune synapse involving interactions between a B cell stimulatory receptor and a T cell stimulatory receptor and their ligands.
  • the format of the multi-domain immunomodulatory protein is chosen to avoid crosslinking or engagement of the T cell stimulatory receptor.
  • the provided immunomodulatory proteins do not exhibit multivalent binding to the T cell stimulatory receptor.
  • a relatively smaller molecular weight, monomeric and/or single chain polypeptide fusion of the immunomodulatory protein is contemplated.
  • the provided multi-domain immunomodulatory proteins can include one or more BIM and one or more TIM.
  • an immunomodulatory protein can include one or more BIM described herein and any one or more TIM described herein.
  • the immunomodulatory protein comprises exactly 1, 2, 3, 4, 5 BIMs, which, in some aspects, are the same or are of identical sequence when a plurality are included.
  • each of a plurality of, e.g. 2, 3, 4, or 5, are linked directly or indirectly via a linker to another BIM.
  • the immunomodulatory proteins comprises exactly 1, 2, 3, 4, 5 TIMs, which, in some aspects, are the same or are of identical sequence when a plurality are included.
  • each of a plurality of TIMs, e.g. 2, 3, 4, or 5, are linked directly or indirectly via a linker to another TIM.
  • the multi-domain immunomodulatory protein contains a polypeptide that includes at least one BIM and at least one TIM.
  • at least one of the one or more BIM molecules are linked directly or indirectly via a linker to at least one of the one or more TIM.
  • the immunomodulatory protein includes a polypeptide containing a BIM linked directly or indirectly via a linker to a TIM, in either order.
  • at least one BIM is amino terminal to at least one TIM in the polypeptide.
  • at least one BIM is carboxy terminal to at least one TIM in the polypeptide.
  • two, three, four, or more of a polypeptides containing one or more BIM and/or one or more BIM can be covalently or non-covalently attached to each other.
  • at least one polypeptide chain contains one or more BIM and at least one polypeptide chain contains one or more TIM.
  • each of at least two polypeptide chain contains at least one BIM and at least one TIM.
  • exactly two polypeptides each containing one or more BIM and/or one or more TIM, can be covalently or non-covalently attached to each other to form a dimer.
  • the two polypeptides can be attached via a multimerization domain, in which, in some aspects, one or both of the BIM and TIM are linked directly or indirectly via a linker to the multimerization domain.
  • the multimerization domain can be the same or different.
  • the multimerization domain such as an Fc region, facilitates attachment of two polypeptide chains via interchain cysteine disulfide bond.
  • compositions comprising two or more polypeptides can be of an identical sequence or substantially identical sequence of polypeptide (e.g., a homodimer) or of a non-identical sequence of polypeptides (e.g., a heterodimer).
  • the multi-domain immunomodulatory protein can further include a tag or moiety.
  • Non-limiting examples of components for inclusion in provided formats are further described in the following subsections. Exemplary Fc-fusion formats of provided multi-domain immunomodulatory proteins are depicted in FIG. 16 .
  • linkers can be used to connect components of a polypeptide, such as any BIM and/or TIM provided herein.
  • a linker is a peptide or polypeptide sequence ⁇ e.g. a synthetic peptide or polypeptide sequence), or is a non-peptide linker able to connect two moieties.
  • a linker is used or chosen to maintain the structural flexibility and other conformational characteristics of the individual residues or at the secondary, tertiary, or quaternary structural levels of domains of the polypeptide fusion protein, such as in order to maintain functional properties of the immunomodulatory protein.
  • Linkers can also provide additional beneficial properties to the protein, such as increased protein expression in mammalian expression systems, improved biophysical properties such as stability and solubility, improved protein purification and detection and/or increased enzymatic activity.
  • two or more linkers can be linked in tandem.
  • the linkers can be peptide linker.
  • the linker includes chemical linking agents and heterobifunctional linking agents.
  • the linker is not cleavable.
  • a linker can contain one or more protease-cleavable sites, which can be located within the sequence of the linker or flanking the linker at either end of the linker sequence.
  • each of the linkers can be the same or different.
  • linkers or multiple linkers provide flexibility to the polypeptide molecule.
  • one or more “peptide linkers” link the BIM, TIM, or other moieties of the immunomodulatory protein.
  • 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. Linking moieties are described, for example, in Huston et al. (1988) PNAS 85:5879-5883, Whitlow et al. (1993) Protein Engineering 6:989-995, and Newton et al, (1996) Biochemistry 35:545-553.
  • Other suitable peptide linkers include any of those described in U.S. Pat. Nos. 4,751,180 or 4,935,233.
  • a peptide linker includes peptides (or polypeptides) ⁇ e.g., natural, or non-naturally occurring peptides) which includes an amino acid sequence that links or genetically fuses a first linear sequence of amino acids to a second linear sequence of amino acids to which it is not naturally linked or genetically fused in nature.
  • the peptide linker can include non-naturally occurring polypeptides which are modified forms of naturally occurring polypeptides (e.g., that includes a mutation such as an addition, substitution or deletion).
  • the peptide linker can include non-naturally occurring amino acids.
  • the peptide linker can include naturally occurring amino acids occurring in a linear sequence that does not occur in nature.
  • the peptide linker can include a naturally occurring polypeptide sequence.
  • Linking moieties can also include derivatives and analogs of the naturally occurring amino acids, as well as various non-naturally occurring amino acids (D- or L-), hydrophobic or non-hydrophobic, known in the art.
  • Exemplary peptide linkers are linkers with the formula Ser(Gly 4 Ser) n (or (Gly-Ser) n residues with some Glu or Lys residues dispersed throughout to increase solubility, where n can be an integer from 1 to 20, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20.
  • Other exemplary linkers include peptide linkers with the formula [(Gly) x -Ser y ] z where x is from 1 to 4, y is 0 or 1, and z is from 1 to 50.
  • the peptide linker includes the sequence G., where n can be an integer from 1 to 100.
  • the sequence of the peptide linker can be (GA). or (GGS) n .
  • the linker is (in one-letter amino acid code): GGGGS (“4GS”; SEQ ID NO: 593) or multimers of the 4GS linker, such as repeats of 2, 3, 4, or 5 4GS linkers.
  • the peptide linker is the peptide linker is (GGGGS) 2 (SEQ ID NO: 594), (GGGGS) 3 (SEQ ID NO: 595), (GGGGS) 4 (SEQ ID NO: 600) or (GGGGS) 5 (SEQ ID NO: 671).
  • 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).
  • 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 (SEQ ID NO:711) or multimers of the EAAAK linker, such as repeats of 2, 3, 4, 5 or 6 EAAAK linkers, such as set forth in SEQ ID NO: 712 (2 ⁇ EAAAK), SEQ ID NO: 713 (3 ⁇ EAAAK), SEQ ID NO: 714 (4 ⁇ EAAAK), SEQ ID NO: 715 (5 ⁇ EAAAK), or SEQ ID NO: 665 (6 ⁇ 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
  • the linker is GSGGGGS (SEQ ID NO: 590) or GGGGSSA (SEQ ID NO: 596).
  • the linker is a 2 ⁇ GGGGS followed by three alanines (GGGGSGGGGSAAA; SEQ ID NO:721).
  • a polynucleotide encoding a desired peptide linker can be inserted between, and in the same reading frame as a polynucleotide encoding any TIM, BIM or other moiety in the provided immunomodulatory protein and between another moiety, using any suitable conventional technique.
  • the immunomodulatory protein containing one or more BIM(s) and/or TIM(s) is multimeric, such as dimeric, trimeric, tetrameric, or pentameric.
  • the immunomodulatory protein comprises a first polypeptide and a second polypeptide.
  • the first and/or second polypeptide is or contains a BIM, TIM, or both.
  • the BIM and/or TIM is linked, directly or indirectly via a linker, to a multimerization domain.
  • the mutlimerization domain increase half-life of the molecule.
  • the linker can include any as described above.
  • the immunomodulatory protein provided herein is a dimer.
  • the immunomodulatory protein is a homodimer that contains a first and second polypeptide subunit that are the same, i.e. each has the same amino acid sequence containing the identical BIM(s) and TIM(s).
  • the homodimer can be formed by transforming a nucleic acid molecule encoding the variant polypeptide into a cell, which, upon secretion, results in covalent or non-covalent interaction between residues of polypeptide subunits to mediate formation of the dimer.
  • the immunomodulatory protein is a heterodimer that contains a first and second polypeptide subunit that are different.
  • one or both of the first or second polypeptide subunit contains a sequence of amino acids of a BIM and TIM.
  • both the first and second polypeptide subunit can contain a sequence of amino acids of a BIM and a sequence of amino acids of a TIM.
  • the heterodimer can be formed by transforming into a cell both a first nucleic acid molecule encoding a first polypeptide subunit and a second nucleic acid molecule encoding a second different polypeptide subunit.
  • the heterodimer is produced upon expression and secretion from a cell as a result of covalent or non-covalent interaction between residues of the two polypeptide subunits to mediate formation of the dimer.
  • a mixture of dimeric molecules is formed, including homodimers and heterodimers.
  • additional steps for purification can be necessary.
  • the first and second polypeptide can be engineered to include a tag with metal chelates or other epitope, where the tags are different.
  • the tagged domains can be used for rapid purification by metal-chelate chromatography, and/or by antibodies, to allow for detection by western blots, immunoprecipitation, or activity depletion/blocking in bioassays.
  • Interaction of two or more polypeptides of the immunomodulatory proteins 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 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 polypeptide and a second polypeptide.
  • the two or more individual polypeptides of the immunomodulatory proteins can be joined by multimerization, such as joined as dimeric, trimeric, tetrameric, or pentameric molecules.
  • the individual polypeptides are the same.
  • a trimeric molecule can be formed from three copies of the same individual polypeptide.
  • a tetrameric molecule is generated from four copies of the same individual polypeptides.
  • a pentameric molecule is generated from five copies of the same individual polypeptides.
  • the individual polypeptides of an immunomodulatory proteins containing a BIM and/or TIM are fused to a multimerization domain.
  • the multimerization domain may be one that facilities dimerization, trimerization, tetramerization, or pentamerization of the polypeptide chains.
  • the immunomodulatory protein forms a multimer, e.g., a dimer.
  • the dimer is a homodimer in which the two polypeptides of the immunomodoulatory protein are the same.
  • the dimer is a heterodimer in which the two polypeptides of the immunomodoulatory protein are 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 BIM and/or TIM 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 BIM and/or TIM 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 BIM and/or TIM is joined to the N-terminus of the multimerization domain.
  • a construct can encode a chimeric protein where the N-terminus of the BIM and/or TIM 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 BIM and/or TIM directly or indirectly to a multimerization domain.
  • the multimerization domain is a polypeptide
  • a gene fusion encoding the BIM and/or TIM 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 the BIM and/or TIM 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 BIM and/or TIM attached to an immunoglobulin Fc (yielding an “immunomodulatory Fc fusion.”)
  • the attachment of the BIM and/or TIM is at the N-terminus of the Fc.
  • the attachment of the BIM and/or TIM is at the C-terminus of the Fc.
  • two or more BIM and/or TIM are independently attached at the N-terminus and at the C-terminus.
  • homo- or heteromultimeric polypeptides can be generated from co-expression of separate BIM and/or TIM containing polypeptides.
  • the first and second polypeptides can be the same or different.
  • the first and/or second polypeptide each contains two or more BIM and/or TIM linked to the Fc sequence. In some embodiments, the first and/or second polypeptide each contains three TIMs and one BIM linked to the Fc sequence. Exemplary Fc fusion formats of provided multi-domain immunomodulatory proteins are depicted in FIG. 16 .
  • the Fc is murine or human Fc. In some embodiments, the Fc is a mammalian or human IgG1, lgG2, lgG3, or lgG4 Fc regions.
  • the Fc is derived from IgG1, such as human IgG1.
  • the Fc is an IgG1 Fc set forth in SEQ ID NO: 586 having an allotype containing residues Glu (E) and Met (M) at positions 356 and 358 by EU numbering.
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 586 or a sequence of amino acids that exhibits at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 586.
  • the Fc is IgG1 Fc that contains amino acids of the human G1 ml allotype, such as residues containing Asp (D) and Leu (L) at positions 356 and 358, e.g. as set forth in SEQ ID NO:597.
  • an Fc provided herein can contain amino acid substitutions E356D and M358L to reconstitute residues of allotype G1 ml.
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 597 or a sequence of amino acids that exhibits at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 597.
  • the Fc region has the amino acid sequence set forth in SEQ ID NO:597.
  • the variant Fc comprises the sequence set forth in SEQ ID NO: 755. In some embodiments, the variant Fc comprises the sequence set forth in SEQ ID NO:756. In some embodiments, an Fc region used in a construct provided herein can further lack a C-terminal lysine residue.
  • the Fc is derived from IgG2, such as human IgG2.
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 726 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: 726.
  • the Fc region is an IgG2 Fc region that has the sequence set forth in SEQ ID NO: 726.
  • the Fc region is an IgG2 Fc region that has the sequence set forth in SEQ ID NO: 822.
  • the Fe is derived from IgG4, such as human IgG4.
  • the Fc comprises the amino acid sequence set forth in SEQ ID NO: 727 or a sequence of amino acids that exhibits at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 727.
  • 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: 728 or a sequence of amino acids that exhibits at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 728.
  • the Fc region is an IgG4 Fc region set forth in SEQ ID NO:728.
  • the Fc region is an IgG4 Fc region set forth in SEQ ID NO:823.
  • 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, 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 00/42072, WO2006019447, WO2012125850, 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 immunomodulatory proteins comprise an Fc region that exhibits reduced effector functions, which makes it a desirable candidate for applications in which the half-life of the immunomodulatory protein in vivo is important yet certain effector functions (such as CDC and ADCC) are unnecessary or deleterious.
  • In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities.
  • Fc receptor (FcR) binding assays can be conducted to ensure that the immunomodulatory protein 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 2 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).
  • Clq binding assays may also be carried out to confirm that the immunomodulatory protein n is unable to bind Clq and hence lacks CDC activity. See, e.g., Clq 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)).
  • Immunomodulatory protein 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).
  • 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).
  • 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.
  • the Fc region of immunomodulatory proteins 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
  • an immunomodulatory protein 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 the immunomodulatory protein comprises one or more amino acid substitutions C220S, C226S and/or C229S by EU numbering. In some embodiments, the Fc region of the immunomodulatory protein 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.
  • alterations are made in the Fc region that result in diminished Clq 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
  • an immunomodulatory protein comprising a variant Fc region comprising one or more amino acid modifications, wherein the variant Fc region is derived from a wild-type IgG1, such as a wild-type human IgG1.
  • the wild-type IgG1 Fc can be the Fc set forth in SEQ ID NO: 586 having an allotype containing residues Glu (E) and Met (M) at positions 356 and 358 by EU numbering.
  • the variant Fc region is derived from the amino acid sequence set forth in SEQ ID NO: 586.
  • the wild-type IgG1 Fc contains amino acids of the human Glml allotype, such as residues containing Asp (D) and Leu (L) at positions 356 and 358, e.g. as set forth in SEQ ID NO:597.
  • the variant Fc is derived from the amino acid sequence set forth in SEQ ID NO:597.
  • the Fe region lacks the C-terminal lysine corresponding to position 232 of the wild-type or unmodified Fc set forth in SEQ ID NO: 586 or 597 (corresponding to K447del by EU numbering).
  • the Fc contains at least one amino acid substitution that is N82G by numbering of SEQ ID NO: 586 (corresponding to N297G by EU numbering). In some embodiments, the Fc further contains at least one amino acid substitution that is R77C or V87C by numbering of SEQ ID NO: 586 (corresponding to R292C or V302C by EU numbering). In some embodiments, the variant Fc region further comprises a C5S amino acid modification by numbering of SEQ ID NO: 586 (corresponding to C220S by EU numbering).
  • the variant Fc region comprises the following amino acid modifications: N297G 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:586), e.g., the Fc region comprises the sequence set forth in SEQ ID NO:598.
  • the variant Fc contains the amino acid substitutions L234A/L235E/G237A, by EU numbering. In some embodiments, the variant Fc contains the amino acid substitutions A330S/P331S, by EU numbering. In some embodiments, the variant Fc contains the amino acid substitutions L234A/L235E/G237A/A330S/P331S (Gross et al. (2001) Immunity 15:289). In some embodiments, the variant Fc comprises the sequence set forth in SEQ ID NO: 757. In some embodiments, the variant Fc comprises the sequence set forth in SEQ ID NO:758. In some embodiments, an Fc region used in a construct provided herein can further lack a C-terminal lysine residue.
  • the variant Fc using in immunomodulatory protein constructs provided herein can include effectorless mutations 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.
  • Exemplary inert Fc regions having reduced effector function are set forth in SEQ ID NO: 599 and SEQ ID NO:591, which are based on allotypes set forth in SEQ ID NO:586 or SEQ ID NO: 597, respectively.
  • an Fc region used in a construct provided herein can further lack a C-terminal lysine residue.
  • 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:589, 591, 599 or 724.
  • the variant Fc comprises has the sequence set forth in SEQ ID NO: 589.
  • the variant Fc comprises has the sequence set forth in SEQ ID NO: 591.
  • the variant Fc comprises has the sequence set forth in SEQ ID NO: 599.
  • the variant Fc comprises has the sequence set forth in SEQ ID NO: 724.
  • the Fe region is a variant Fc that has the sequence set forth in SEQ ID NO:589.
  • the Fc region is a variant Fc that has the sequence set forth in SEQ ID NO: 824.
  • the variant Fc region comprises one or more of the amino acid modifications C220S, L235P, L234V, L235A, G236del or S267K, e.g. the Fc region comprises the sequence set forth in SEQ ID NO:722.
  • the Fc region lacks the C-terminal lysine corresponding to position 232 of the wild-type or unmodified Fc set forth in SEQ ID NO: 586 (corresponding to K447del by EU numbering).
  • the variant Fc region comprises one or more of the amino acid modifications C220S, R292C, N297G, V302C.
  • the Fc region lacks the C-terminal lysine corresponding to position 232 of the wild-type or unmodified Fc set forth in SEQ ID NO: 586 (corresponding to K447del by EU numbering).
  • An exemplary variant Fc region for use in the immunomodulatory protein constructs is set forth in SEQ ID NO: 723.
  • the variant Fc region comprises one or more of the amino acid modifications C220S/E233P/L234V/L235A/G236del/S267K.
  • the Fc region lacks the C-terminal lysine corresponding to position 232 of the wild-type or unmodified Fc set forth in SEQ ID NO: 586 (corresponding to K447del by EU numbering).
  • An exemplary variant Fc region for use in the immunomodulatory protein constructs is set forth in SEQ ID NO: 725.
  • the immunomodulatory protein is a homodimer that contains a first immunomodulatory Fc fusion polypeptide and a second immunomodulatory Fc fusion polypeptide in which the first and second polypeptide are the same.
  • a first Fc polypeptide fusion contains an Fc region and one or more BIM and/or TIM and a second polypeptide fusion contains the same Fc region and the same one or more BIM and/or TIM.
  • the Fc region can be any as described above.
  • Fc Regions wild-type or variant (effectorless) 356E/358M 356D/358L allotype allotype Fc mutations (EU numbering) SEQ ID NO SEQ ID NO (wild-type) 586 597 (with C220S, K447del C220S, R292C, N297G, V302C 598 C220S, R292C, N297G, V302C, K447del 726 C220S, L234A, L235E, G237A 599 591 C220S, L234A, L235E, G237A, K447del 724 589 L234A, L235E, G237A, K447del, with deletion 824 of hinge C220S, L235P, L234V, L235A, G236del, 722 S267K C220S/E233P/L234V/L235A/G236del/S
  • an immunomodulatory protein comprising at least one BIM (e.g. as described in Section III.A), at least one TIM (e.g. any set forth in Section III. B), and variant Fc region that exhibits reduced effector activity compared to a wild-type IgG1 set forth in SEQ ID NO:586 or 597.
  • BIM e.g. as described in Section III.A
  • TIM e.g. any set forth in Section III. B
  • variant Fc region that exhibits reduced effector activity compared to a wild-type IgG1 set forth in SEQ ID NO:586 or 597.
  • the variant Fc comprises the sequence of amino acids set forth in any of SEQ ID NOS:591, 598, 599, 722, 589, 723, 724, 725, 757, 758 or 824 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: 591, 598, 599, 722, 589, 723, 724, 725, 757, 758 or 824.
  • an immunomodulatory protein comprising at least one BIM (e.g.
  • the provided immunomodulatory protein when produced and expressed from a cells, is a homodimer containing two identical polypeptide chains.
  • the immunomodulatory protein contains a first immunomodulatory Fc fusion polypeptide and a second immunomodulatory Fc fusion polypeptide in which the first and second polypeptide are different.
  • a first Fc polypeptide fusion contains an Fc region and one or more BIM and/or TIM and a second polypeptide fusion contains an Fc region and one or more BIM and/or TIM.
  • the Fc region can be a region that promotes or facilitates formation of heterodimers.
  • the Fc domain of one or both of the first and second immunomodulatory Fc fusion polypeptides comprise a modification (e.g. substitution) such that the interface of the Fc molecule is modified to facilitate and/or promote heterodimerization.
  • Methods to promote heterodimerization of Fc chains include mutagenesis of the Fc region, such as by including a set of “knob-into-hole” mutations or including mutations to effect electrostatic steering of the Fc to favor attractive interactions among different polypeptide chains.
  • the Fc region 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.
  • such Fc regions contain mutations C220S, L234A, L235E and/or G237A by EU numbering.
  • any of the above mutations in an Fc backbone can be made in an allotype containing residues Glu (E) and Met (M) at positions 356 and 358 by EU numbering.
  • any of the above mutations in an Fc backbone can be made in an allotype containing residue Asp (D) and Leu (L) at positions 356 and 358 by EU numbering.
  • 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 first polypeptide that is modified to contain protuberance (knob) 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 tryptophan.
  • 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.
  • the knob Fc may contain the sequence set forth in SEQ ID NO: 669, containing S354C and T366W, and a hole Fc set forth in SEQ ID NO: 670, containing mutations Y349C, T366S, L368A and Y407V).
  • 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. In some embodiments, 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.
  • 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.
  • knobs-in-holes technologies are known in the art, e.g. as described by EP 1 870 459 A1.
  • an Fc variant containing CH3 protuberance (knob) or cavity(hole) modifications can be joined to a multi-domain immunomodulatory polypeptide anywhere, but typically via its N- or C-terminus, to the N- or C-terminus of the one or more BIM or TIM, 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).
  • knob and hole Fc polypeptides are set forth in SEQ ID NOs: 716, and 717, respectively.
  • the knob or hold Fc region lacks the C-terminal lysine corresponding to position 232 of the wild-type or unmodified Fc set forth in SEQ ID NO: 586 (corresponding to K447del by EU numbering).
  • Exemplary sequences for knob and hole Fc polypeptides are set forth in SEQ ID NOs: 669, and 670, respectively.
  • an immunomodulatory protein comprising a first polypeptide containing at least one BIM (e.g. as described in Section III.A) and/or at least one TIM (e.g. any set forth in Section III. B), and a first variant Fc set forth in SEQ ID NO:716; and a second polypeptide containing at least one BIM (e.g. as described in Section III.A) and/or at least one TIM (e.g. any set forth in Section III. B), and a second variant Fc set forth in SEQ ID NO:717.
  • an immunomodulatory protein comprising a first polypeptide containing at least one BIM (e.g.
  • the provided immunomodulatory protein when produced and expressed from a cells, is a heterodimer containing two different polypeptide chains.
  • one of the polypeptides can express a TIM and one of the polypeptides can express a BIM.
  • the Fc region of each polypeptide of a heterodimer includes a mutation to altered charge polarity across the Fc dimer interface such that coexpression of electrostatically matched Fc chains support favorable attractive interactions thereby promoting desired Fc heterodimer formation, whereas unfavorable repulsive charge interactions suppress unwanted Fc homodimer formation (Guneskaran et al. (2010) JBC, 285: 19637-19646).
  • at least one polypeptide containing an BIM and/or TIM is linked directly or indirectly to an Fc containing mutations to positively charged residues (e.g.
  • the other polypeptide of the heterodimer containing an BIM and/or TIM is linked directly or indirectly to an Fc containing mutations to negatively charged residues (e.g. K370D, K392D and K409D by EU numbering; designated D chain), such as set forth in SEQ ID NO:730.
  • an Fc containing mutations to negatively charged residues e.g. K370D, K392D and K409D by EU numbering; designated D chain
  • an immunomodulatory protein comprising a first polypeptide containing at least one BIM (e.g. as described in Section III.A) and/or at least one TIM (e.g. any set forth in Section III. B), and a first variant Fc set forth in SEQ ID NO:729; and a second polypeptide containing at least one BIM (e.g. as described in Section III.A) and/or at least one TIM (e.g. any set forth in Section III. B), and a second variant Fc set forth in SEQ ID NO:730.
  • the provided immunomodulatory protein when produced and expressed from a cells, is a heterodimer containing two different polypeptide chains.
  • one of the polypeptides can express a TIM and one of the polypeptides can express a BIM.
  • individual polypeptide of a multi-domain polypeptide or individual polypeptides of a single-domain polypeptide are linked to a multimerization domain that forms an immunomodulatory protein is a trimer, tetramer or pentamer.
  • the individual polypeptides of such a molecule are the same.
  • such a multimerization domain is a cartilage oligomeric matrix protein (COMP) assembly domain, a vasodilator-stimulated phosphoprotein (VASP) tetramerization domain or a ZymoZipper (ZZ) 12.6 domain.
  • cartilage oligomeric matrix protein (COMP) assembly domain a cartilage oligomeric matrix protein (COMP) assembly domain, a vasodilator-stimulated phosphoprotein (VASP) tetramerization domain or a ZymoZipper (ZZ) 12.6 domain.
  • VASP vasodilator-stimulated phosphoprotein
  • the multimerization domain is a portion of the cartilage oligomeric matrix protein (COMP) assembly domain (Voulgaraki et al., Immunology (2005) 115(3):337-346.
  • the COMP is or contains an amino acid sequence as set forth in SEQ ID NO: 734 (e.g. amino acids 29-72 of the full length COMP, Uniprot accession number P49747) or a sequence that has 85%, 85%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 734.
  • the multimerization domain is a vasodilator-stimulated phosphoprotein (VASP) tetramerization domain (Bachmann et al., J Biol Chem (1999) 274(33):23549-23557).
  • VASP vasodilator-stimulated phosphoprotein
  • the VASP is or contains an amino acid sequence as set forth in SEQ ID NO: 735 (e.g. amino acids 343-375 of the full length VASP; Uniprot accession number P50552) or a sequence that has 85%, 85%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 735.
  • the multimerization domain is a ZymoZipper (ZZ) 12.6 domain.
  • the ZZ domain is or contains an amino acid sequence as set forth in SEQ ID NO: 736 (See U.S. Pat. No. 7,655,439) or a sequence that has 85%, 85%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 736.
  • a first and second polypeptide of a heterodimeric Fc fusion protein can be linked to a moiety for detection and/or purification.
  • the first and second polypeptide are linked to different tags or moieties.
  • the tag or moiety of the first and second polypeptide is independently selected from a poly-histidine tag (HHHHHH; SEQ ID NO: 702), a flag-tag (DYKDDDDK; SEQ ID NO: 588), a Myc-tag, or fluorescent protein-tags (e.g., EGFP, set forth in SEQ ID NOs:731, 732 or 733).
  • the first polypeptide containing an BIM and the second polypeptide containing an TIM each further contain a moiety for detection and/or purification, such as a poly-histidine tag (HHHHHH; SEQ ID NO: 702) and/or a flag-tag (DYKDDDDK; SEQ ID NO: 588).
  • a moiety for detection and/or purification such as a poly-histidine tag (HHHHHH; SEQ ID NO: 702) and/or a flag-tag (DYKDDDDK; SEQ ID NO: 588).
  • the BIM and/or TIM is directly linked to the Fc sequence. In some embodiments, the BIM and/or TIM is indirectly linked to the Fc sequence, such as via a linker. In some embodiments, one or more “peptide linkers” link the BIM and/or TIM 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. Exemplary linkers are set forth in subsection “Linker.”
  • the linker is (in one-letter amino acid code): GGGGS (“4GS”; SEQ ID NO: 593) or multimers of the 4GS linker, such as repeats of 2, 3, 4, or 5 4GS linkers.
  • the peptide linker is the peptide linker is (GGGGS) 2 (SEQ ID NO: 594), (GGGGS) 3 (SEQ ID NO: 595), (GGGGS) 4 (SEQ ID NO: 600) or (GGGGS) 5 (SEQ ID NO: 671).
  • 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).
  • the linker in one-letter amino acid code is GSGGGGS (SEQ ID NO: 590) or GGGGSSA (SEQ ID NO: 596). In some examples, the linker is a 2 ⁇ GGGGS followed by three alanines (GGGGSGGGGSAAA; SEQ ID NO:721).
  • nucleic acid molecules encoding the immunomodulatory protein are also provided.
  • a nucleic acid molecule encoding the immunomodulatory protein is inserted into an appropriate expression vector.
  • the resulting immunomodulatory 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, immunomodulatory proteins.
  • the resulting immunomodulatory protein containing an BIM, TIM, and Fc can be easily purified by affinity chromatography over Protein A or Protein G columns.
  • additional steps for purification can be necessary.
  • immunomodulatory protein 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 immunomodulatory protein monomers are mixed in equimolar amounts and oxidized to form a mixture of homo- and heterodimers.
  • this mixture is separated by chromatographic techniques.
  • the formation of this type of heterodimer can be biased by genetically engineering and expressing immunomodulatory proteins containing Fc fusion molecules that contain one or more BIM and/or TIM using knob-into-hole methods described below.
  • the one or more polypeptides containing a BIM and/or TIM in the provided immunomodulatory proteins can further include a tag or moiety.
  • the further moiety is a protein, peptide, small molecule or nucleic acid.
  • the immunomodulatory protein is linked, directly or indirectly to more than one further moiety, such as 2, 3, 4, 5, or 6, further moieties.
  • 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 BIM and/or TIM 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 BIM and/or TIM are linked, directly or indirectly via a linker, to Pro/Ala/Ser (PAS).
  • one or more additional linker structures may be used.
  • the moiety facilitates detection or purification of the immunomodulatory protein.
  • the immunomodulatory protein such as at least one of or each polypeptide of a multimer (e.g. dimer, trimer, tetramer, or pentamer) thereof, comprises a tag or moiety, e.g. affinity or purification tag, linked.
  • a tag or moiety can be linked directly or indirectly via a linker to the N- and/or c-terminus of the polypeptide.
  • polypeptide tags and/or fusion domains include but are not limited to, a poly-histidine (His) tag (SEQ ID NO:702), a FLAG-tag (SEQ ID NO:588), a Myc-tag, and fluorescent protein-tags (e.g., EGFP, set forth in SEQ ID NOs:734, 735, or 736.
  • the tag is a His tag containing at least six histidine residues (set forth in SEQ ID NO:702).
  • the immunomodulatory protein comprising a BIM and TIM further comprises various combinations of moieties.
  • the immunomodulatory protein comprising BIM or TIM further comprises one or more polyhistidine-tag and FLAG tag.
  • the combination of moieties, such as two or more moieties can be included on the same polypeptide.
  • the combination of moieties, such as two or more moieties can be included on different polypeptide, such as in connection with embodiments relating to heterodimeric immunomodulatory polypeptides.
  • nucleic acids which encode any of the immunomodulatory proteins provided herein.
  • nucleic acids provided herein including all described below, are useful in recombinant production (e.g., expression) of immunomodulatory proteins provided herein.
  • nucleic acids provided herein, including all described below are useful in expression of immunomodulatory proteins provided herein, such as BCMA fusion proteins or multi-domain immunomodulatory proteins provided herein.
  • 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.
  • 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.
  • a heterologous (non-native) signal peptide can be added to the nucleic acid encoding the immunomodulatory protein. This may be desired, for example, in the case of expression of BCMA fusion proteins or provided multi-domain immunomodulatory proteins, which do not contain an amino terminal signal sequence.
  • the signal peptide is 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 express and, in some aspects, secret a protein from a cell.
  • exemplary signal peptides include any described in the Table 5.
  • 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.
  • recombinant expression vectors and recombinant host cells useful in producing the immunomodulatory proteins, such as BCMA fusion proteins or multi-domain immunomodulatory proteins provided herein.
  • the nucleic acids encoding the 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 encoding an immunomodulatory protein 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.
  • 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, biosafety 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 cell is a mammalian cell.
  • suitable mammalian host cells include African green monkey kidney cells (Vero; ATCC CRL 1587), human embryonic kidney cells (293-HEK; ATCC CRL 1573), baby hamster kidney cells (BHK-21, BHK-570; ATCC CRL 8544, ATCC CRL 10314), canine kidney cells (MDCK; ATCC CCL 34), Chinese hamster ovary cells (CHO-K1; ATCC CCL61; CHO DG44 (Chasin et al, Som. Cell. Molec. Genet.
  • GH1 rat pituitary cells
  • H-4-II-E rat hepatoma cells
  • COS-1 SV40-transformed monkey kidney cells
  • NIH-3T3 murine embryonic cells
  • 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.
  • 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 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).
  • 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 effecting 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.
  • Transcriptional regulatory sequences include a promoter region sufficient to direct the initiation of RNA synthesis.
  • Suitable eukaryotic promoters include the promoter of the mouse metallothionein I gene (Hamer et al, J. Molec. Appl Genet. 1:273 (1982)), the TK promoter of Herpes virus (McKnight, Cell 31:355 (1982)), the SV40 early promoter (Benoist et al, Nature 290:304 (1981)), the Rous sarcoma virus promoter (Gorman et al, Proc. Nat'l Acad. Sci.
  • cytomegalovirus promoter Fert al, Gene 45:101 (1980)
  • mouse mammary tumor virus promoter see, generally, Etcheverry, “Expression of Engineered Proteins in Mammalian Cell Culture,” in Protein Engineering: Principles and Practice, Cleland et al. (eds.), pages 163-181 (John Wiley & Sons, Inc. 1996).
  • One useful combination of a promoter and enhancer is provided by a myeloproliferative sarcoma virus promoter and a human cytomegalovirus enhancer.
  • a prokaryotic promoter such as the bacteriophage T3 RNA polymerase promoter
  • a prokaryotic promoter can be used to control production of an immunomodulatory protein in mammalian cells if the prokaryotic promoter is regulated by a eukaryotic promoter (Zhou et al, Mol Cell. Biol. 10:4529 (1990), and Kaufman et al, Nucl. Acids Res. 19:4485 (1991)).
  • An expression vector can be introduced into host cells using a variety of standard techniques including calcium phosphate transfection, liposome-mediated transfection, microprojectile-mediated delivery, electroporation, and the like.
  • the transfected cells can be selected and propagated to provide recombinant host cells that comprise the expression vector stably integrated in the host cell genome.
  • Techniques for introducing vectors into eukaryotic cells and techniques for selecting such stable transformants using a dominant selectable marker are described, for example, by Ausubel (1995) and by Murray (ed.), Gene Transfer and Expression Protocols (Humana Press 1991).
  • one suitable selectable marker is a gene that provides resistance to the antibiotic neomycin.
  • selection is carried out in the presence of a neomycin-type drug, such as G-418 or the like.
  • Selection systems can also be used to increase the expression level of the gene of interest, a process referred to as “amplification.” Amplification is carried out by culturing transfectants in the presence of a low level of the selective agent and then increasing the amount of selective agent to select for cells that produce high levels of the products of the introduced genes.
  • a suitable amplifiable selectable marker is dihydrofolate reductase, which confers resistance to methotrexate.
  • drugs resistance genes e.g., hygromycin resistance, multi-drug resistance, puromycin acetyltransferase
  • markers that introduce an altered phenotype such as green fluorescent protein, or cell surface proteins such as CD4, CD8, Class I MHC, placental alkaline phosphatase may be used to sort transfected cells from untransfected cells by such means as FACS sorting or magnetic bead separation technology.
  • 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.
  • compositions containing any of the provided immunomodulatory proteins 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.
  • 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.
  • buffers such as neutral buffered saline, phosphate buffered saline and the like
  • carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol
  • proteins such as glucose, mannose, sucrose or dextrans, mannitol
  • proteins such as glucose, mannose, sucrose or dextrans, mannitol
  • proteins such as glucose, mannose, sucrose or dextrans, mannitol
  • proteins such as glucose, mannose, sucrose or dextrans, mannitol
  • proteins such as glucose, mannose
  • 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 immunomodulatory proteins 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.
  • a pharmaceutically acceptable carrier may be a pharmaceutically acceptable material, composition, or vehicle.
  • 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.
  • 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.
  • a provided pharmaceutical formulation may, for example, be in a form suitable for intravenous infusion.
  • the dosage ofthe 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 pg of protein per kg subject body mass or more (such as about 2 pg of protein per kg subject body mass or more, about 5 pg of protein per kg subject body mass or more, about 10 pg 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).
  • the provided immunomodulatory proteins exhibit immunomodulatory activity.
  • the provided immunodulatory proteins such as BCMA fusion proteins or multi-domain immunomodulatory proteins, can modulate B cell activity, such as one or more of B cell proliferation, differentiation or survival.
  • the provided immunomodulatory proteins such as multi-domain immunomodulatory proteins, may additionally modulate T cell activation or response. In some embodiments, T cell activation or response is reduced, decreased or attenuated.
  • immunomodulatory proteins can be examined using a variety of approaches to assess the ability of the proteins to bind to cognate binding partners.
  • BCMA fusion proteins may be assessed for binding to APRIL or BAFF.
  • the proteins may be assessed for binding to the cognate binding partner, such as a ligand of a T cell stimulatory receptor (e.g. CD80 or CD86) or directly to a T cell stimulatory receptor (e.g. CD28), and/or to a ligand of a B cell stimulatory receptor (e.g. APRIL or BAFF).
  • binding molecule e.g., immunomodulatory protein
  • a binding partner e.g., APRIL or BAFF
  • binding affinity e.g., immumodulaotry protein
  • APRIL e.g., APRIL
  • BAFF a binding partner
  • Various binding assays are known and include, but are not limited to, for example, ELISA K D , KinExA, flow cytometry, and/or surface plasmon resonance devices), including those described herein.
  • Such methods include, but are not limited to, methods involving BIAcore®, Octet®, or flow cytometry.
  • a BIAcore® instrument can be used to determine the binding kinetics and constants of a complex between two proteins using surface plasmon resonance (SPR) analysis (see, e.g., Scatchard et al., Ann. N.Y. Acad. Sci. 51:660, 1949; Wilson, Science 295:2103, 2002; Wolff et al., Cancer Res. 53:2560, 1993; and U.S. Pat. Nos. 5,283,173, 5,468,614, or the equivalent).
  • SPR measures changes in the concentration of molecules at a sensor surface as molecules bind to or dissociate from the surface.
  • the change in the SPR signal is directly proportional to the change in mass concentration close to the surface, thereby allowing measurement of binding kinetics between two molecules.
  • the dissociation constant for the complex can be determined by monitoring changes in the refractive index with respect to time as buffer is passed over the chip.
  • suitable assays for measuring the binding of one protein to another include, for example, immunoassays such as enzyme linked immunosorbent assays (ELISA) and radioimmunoassays (RIA), or determination of binding by monitoring the change in the spectroscopic or optical properties of the proteins through fluorescence, UV absorption, circular dichroism, or nuclear magnetic resonance (NMR).
  • exemplary assays include, but are not limited to, Western blot, ELISA, analytical ultracentrifugation, spectroscopy, flow cytometry, sequencing and other methods for detection of expressed polynucleotides or binding of proteins.
  • immunomodulatory proteins also can be assessed in any of a variety of assess to assess modulation of T cell or B cell activity.
  • One such assay is a cell proliferation assay.
  • Cells are cultured in the presence or absence of a test compound (e.g. immunomodulatory protein), and cell proliferation is detected by, for example, measuring incorporation of tritiated thymidine or by colorimetric assay based on the metabolic breakdown of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) (Mosman, J. Immunol. Meth. 65: 55-63, 1983).
  • An alternative assay format uses cells that are further engineered to express a reporter gene.
  • the reporter gene is linked to a promoter element that is responsive to the receptor-linked pathway, and the assay detects activation of transcription of the reporter gene.
  • a promoter element that is responsive to the receptor-linked pathway
  • Numerous reporter genes that are easily assayed for in cell extracts are known in the art, for example, the E. coli lacZ, chloroamphenicol acetyl transferase (CAT) and serum response element (SRE) (see, e.g., Shaw et al., Cell 56:563-72, 1989).
  • An exemplary reporter gene is a luciferase gene (de Wet et al., Mol. Cell. Biol. 7:725, 1987).
  • Luciferase activity assay kits are commercially available from, for example, Promega Corp., Madison, Wis.
  • immunomodulatory proteins can be characterized by the ability to inhibit the stimulation of human B cells by soluble APRIL or BAFF, as described by Gross et al, international publication No. WO00/40716.
  • human B cells are isolated from peripheral blood mononuclear cells, such as using CD19 magnetic beads separation (e.g. Miltenyi Biotec Auburn, Calif.).
  • the purified B cells can be incubated under conditions of stimulation, e.g. in the presence of soluble APRIL, and further in the presence of titrated concentration of immunomodulatory protein.
  • the B cells can be labeled with a proliferation dye or can be labeled with 1 ⁇ Ci 3 H-thymidine to measure proliferation. The number of B cells can be determined over time.
  • Reporter cell lines that express a reporter gene under the operable control of a transcription factor can be made that express TACI or BCMA.
  • the reporter cell can include Jurkat and other B Lymphoma cell lines. Incubation of these cells with soluble BAFF or APRIL ligands signal through the reporter genes in these constructs. The effect of provided immunomodulatory proteins to modulate this signaling can be assessed.
  • animal models of autoimmune disease include, for example, MRL-1pr/1pr or NZB ⁇ NZW F1 congenic mouse strains which serve as a model of SLE (systemic lupus erythematosus).
  • SLE systemic lupus erythematosus
  • Such animal models are known in the art, see for example Autoimmune Disease Models A Guidebook, Cohen and Miller eds. Academic Press.
  • Offspring of a cross between New Zealand Black (NZB) and New Zealand White (NZW) mice develop a spontaneous form of SLE that closely resembles SLE in humans.
  • the offspring mice known as NZBW begin to develop IgM autoantibodies against T-cells at 1 month of age, and by 5-7 months of age, Ig anti-DNA autoantibodies are the dominant immunoglobulin. Polyclonal B-cell hyperactivity leads to overproduction of autoantibodies. The deposition of these autoantibodies, particularly ones directed against single stranded DNA is associated with the development of glomerulonephritis, which manifests clinically as proteinuria, azotemia, and death from renal failure. Kidney failure is the leading cause of death in mice affected with spontaneous SLE, and in the NZBW strain, this process is chronic and obliterative.
  • the disease is more rapid and severe in females than males, with mean survival of only 245 days as compared to 406 days for the males. While many of the female mice will be symptomatic (proteinuria) by 7-9 months of age, some can be much younger or older when they develop symptoms.
  • the fatal immune nephritis seen in the NZBW mice is very similar to the glomerulonephritis seen in human SLE, making this spontaneous murine model very attractive for testing of potential SLE therapeutics (Putterman and Naparstek, Murine Models of Spontaneous Systemic Lupus Erythematosus , Autoimmune Disease Models: A Guidebook, chapter 14, pp. 217-34, 1994; Mohan et al., J.
  • H2-Ab1 bm12 differs from H2-Ab1 b by 3 nucleotides, resulting in alteration of 3 amino acids in the p-chain of the MHC class II I-A molecule.
  • Alloactivation of donor bm12 CD4+ T cells by recipient antigen presenting cells leads to chronic GVHD with symptoms closely resembling SLE, including autoantibody production, changes in immune cell subsets, and mild kidney disease.
  • Glomerulonephritis with immune complex deposition develops late in the model, largely comprised of autoantigens bound to IgG1, IgG2b, IgG2c, and IgG3 antibodies. Endpoints of this model may include concentrations of anti-dsDNA antibodies, select IgG isotypes, blood urea nitrogen (BUN), and creatinine in serum, immune cell subset composition in the spleen and cervical LN, and kidney histology.
  • BUN blood urea nitrogen
  • mouse models for Sjögren's syndrome can be used.
  • the SjS disease as well as an accelerated onset of diabetes, can be induced in female diabetes-prone non-obese diabetic (NOD) mice using repeat dosing with anti-mouse (m) PD-L1 antibody, based on a modified version of a protocol published by Zhou et al., 2016 Sci. Rep. 6, 39105.
  • NOD non-obese diabetic
  • m anti-mouse
  • mice are injected intraperitoneally (IP) on Study Days 0, 2, 4, and 6 with 100 ⁇ g of anti-PD-L1 antibody and are treated on various days with provided immunomodulatory proteins. Na ⁇ ve mice are included as controls for the endpoint analyses.
  • mice are typically terminated on Study Day 10 and submandibular glands (SMG) and the pancreas from each mouse are collected for histopathology evaluation to assess for signs and severity of sialadenitis and insulitis. Blood glucose levels can be measured on various days.
  • mouse models for experimental allergic encephalomyelitis can be used.
  • the models resemble human multiple sclerosis, and produces demyelination as a result of T-cell activation to neuroproteins such as myelin basic protein (MBP), or proteolipid protein (PLP). Inoculation with antigen leads to induction of CD4+, class II MHC-restricted T-cells (Thl).
  • Changes in the protocol for EAE can produce acute, chronic-relapsing, or passive-transfer variants of the model (Weinberg et al., J. Immunol. 162:1818-26, 1999; Mijaba et al., Cell. Immunol. 186:94-102, 1999; and Glabinski, Meth. Enzym. 288:182-90, 1997).
  • Administration of provided immunomodulatory proteins to ameliorate symptoms and alterations to the course of disease can be assessed.
  • a collagen-induced arthritis (CIA) model can be used in which mice develop chronic inflammatory arthritis which closely resembles human rheumatoid arthritis (RA). Since CIA shares similar immunological and pathological features with RA, this makes it an ideal model for screening potential human anti-inflammatory compounds.
  • Another advantage in using the CIA model is that the mechanisms of pathogenesis are known. The T and B cell epitopes on type II collagen have been identified, and various immunological (delayed-type hypersensitivity and anti-collagen antibody) and inflammatory (cytokines, chemokines, and matrix-degrading enzymes) parameters relating to immune-mediating arthritis have been determined, and can be used to assess test compound efficacy in the models (Wooley, Curr. Opin.
  • models for bronchial infection can be created when mice are injected with ovalbumin and restimulated nasally with antigen which produces an asthmatic response in the bronchi similar to asthma.
  • Administration of provided immunomodulatory proteins to ameliorate symptoms and alterations to the course of disease can be assessed.
  • myasthenia gravis is another autoimmune disease for which murine models are available.
  • MG is a disorder of neuromuscular transmission involving the production of autoantibodies directed against the nicotinic acetylcholine receptor (AChR).
  • AChR nicotinic acetylcholine receptor
  • MG is acquired or inherited with clinical features including abnormal weakness and fatigue on exertion.
  • a mouse model of MG has been established. (Christadoss et al., Establishment of a Mouse Model of Myasthenia Gravis Which Mimics Human Myasthenia Gravis Pathogenesis for Immune Intervention , in Immunobiology of Proteins and Peptides VIII, Atassi and Bixler, eds., 1995, pp.
  • EMG Experimental autoimmune myasthenia gravis
  • AChR antibodies to AChR. These antibodies destroy the receptor leading to defective neuromuscular electrical impulses, resulting in muscle weakness.
  • mice are immunized with the nicotinic acetylcholine receptor. Clinical signs of MG become evident weeks after the second immunization.
  • EAMG is evaluated by several methods including measuring serum levels of AChR antibodies by radioimmunoassay (Christadoss and Dauphinee, J Immunol. 136:2437-40, 1986; and Lindstrom et al., Methods Enzymol.
  • T cell dependent and T cell independent immune response can be measured as described in Perez-Melgosa et al., J. Immunol. 163:1123-7, 1999.
  • Immune response in animals subjected to a regular antigen challenge for example, keyhole limpet hemacyanin (KLH), sheep red blood cells (SRBC), ovalbumin or collagen
  • KLH keyhole limpet hemacyanin
  • SRBC sheep red blood cells
  • ovalbumin or collagen ovalbumin
  • Pharmacokinetic studies can be used in association with radiolabeled immunomodulatory proteins to determine the distribution and half life of such polypeptides in vivo.
  • T cell activation assays can be employed in which IFN-gamma or other effector cytokine is measured.
  • the immunomodulatory proteins can be assessed for their ability to suppress or decrease effector cytokine secretion following T cell activation.
  • Assays for determining enhancement or suppression of immunological activity include 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 assays (Wang et al., Cancer Immunol Res.
  • T cell activation is associated with secretion of IFN-gamma cytokine
  • detecting IFN-gamma levels in culture supernatants from these in vitro human T cell assays can be assayed using commercial ELISA kits (Wu et al, Immunol Lett 2008 Apr. 15; 117(1): 57-62).
  • Assays also include assays to assess cytotoxicity, including a standard 51Cr-release assay (see e.g.
  • the assay used is anti-CD3 coimmobilization 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 a mixed lymphocyte reaction (MLR).
  • MLR mixed lymphocyte reaction
  • primary T cells are simulated with allogenic 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 can be performed according to manufacturer's recommendation.
  • a T cell reporter assay in assaying for the ability of an immunomodulatory protein to modulate, e.g. increase or decrease IFN-gamma expression, 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 a T cell costimulatory receptor. In some embodiments, the APCs are artificial APCs. Artificial APCs are well known to a skilled artisan. In some embodiments, artificial APCs are derived from one or more mammalian cell line, such as K562, CHO or 293 cells.
  • 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. 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 compositions containing the same, to a subject having a disease, condition, or disorder.
  • the disease or disorder is an autoimmune or inflammatory disease, condition or disorder.
  • the molecule or engineered cell is administered in an effective amount to effect treatment of the disease, condition or disorder.
  • Uses include uses of molecules containing an immunomodulatory protein, and in the preparation of a medicament in order to carry out such therapeutic methods.
  • the methods are carried out by administering a provided immunomodulatory protein, or compositions comprising the same, to the subject having or suspected of having the disease or condition.
  • the methods thereby treat the disease or condition or disorder in the subject.
  • Illustrative subjects include mammalian subjects, such as farm animals, domestic animals, and human patients.
  • the subject is a human subject.
  • the pharmaceutical 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, organ transplantation, viral infections, and/or bacterial infections in a mammal.
  • the pharmaceutical composition can modulate an immune response to treat the disease.
  • the pharmaceutical composition suppresses an immune response, which can be useful in the treatment of inflammatory or autoimmune disorders, or organ transplantation.
  • the provided methods are believed to have utility in a variety of applications, including, but not limited to, e.g., in prophylactic or therapeutic methods for treating a variety of immune system diseases or conditions in a mammal in which modulation or regulation of the immune system and immune system responses is beneficial.
  • suppressing an immune response can be beneficial in prophylactic and/or therapeutic methods for inhibiting rejection of a tissue, cell, or organ transplant from a donor by a recipient.
  • the mammalian subject is typically one with an immune system disease or condition, and administration is conducted to prevent further progression of the disease or condition.
  • the provided immunomodulatory proteins can be used for the treatment of autoimmune diseases, B cell cancers, immunomodulation, EBD and any antibody-mediated pathologies (e.g., ITCP, myasthenia gravis and the like), renal diseases, indirect T cell immune response, graft rejection, and graft versus host disease.
  • Administration of the immunomodulatory proteins can specifically regulate B cell responses during the immune response. Additionally, administration of provided immunomodulatory proteins can be used to modulate B cell development, development of other cells, antibody production, and cytokine production.
  • provided immunomodulatory proteins can also modulate T and B cell communication, such as by neutralizing the proliferative effects of BAFF or APRIL alone or, in the case of provided multi-domain immunomodulatory proteins also by neutralizing proliferative effects mediated by T cell stimulatory molecules such as by neutralizing the proliferative effects of CD80/CD86 on CD28.
  • 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 an immunomodulatory proteins that exhibits antagonist activity of a B cell stimulatory receptor and/or T cell stimulatory receptor, thereby decreasing or reducing an immune response.
  • the compositions can be used to treat an autoimmune disease.
  • administration of a therapeutic composition containing an immunomodulatory protein provided herein to a subject suffering from an immune system disease can result in suppression or inhibition of such immune system attack or biological responses associated therewith.
  • an immune system disease e.g., autoimmune disease
  • the resulting physical symptoms e.g., pain, joint inflammation, joint swelling or tenderness
  • the biological and physical damage resulting from or associated with the immune system attack can be decreased, retarded, or stopped.
  • the subject may be one with, susceptible to, or believed to present an immune system disease, disorder or condition, and administration is typically conducted to prevent progression of the disease, disorder or condition, inhibit or alleviate symptoms, signs, or biological responses associated therewith, prevent bodily damage potentially resulting therefrom, and/or maintain or improve the subject's physical functioning.
  • the disease or conditions that can be treated by the pharmaceutical composition described herein is any disease mediated by immune complex deposition (e.g. lupus nephritis, vasculitis); direct interference with a pathway (e.g. catastrophic antiphospholipid antibody syndrome, myasthenia gravis crisis; anti-Jo-1 disease); opsonization or direct damage to cells (e.g. Idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia); antibody-mediated rejection of an allograft (e.g. highly-sensitized renal transplant patients); or anti-drug antibodies to biologic replacement factors, vectors (e.g. anti-Factor 8).
  • immune complex deposition e.g. lupus nephritis, vasculitis
  • direct interference with a pathway e.g. catastrophic antiphospholipid antibody syndrome, myasthenia gravis crisis; anti-Jo-1 disease
  • opsonization or direct damage to cells e.g. Idiopathic thrombocytopen
  • the inflammatory and autoimmune disorders that can be treated by the pharmaceutical composition described herein is Systemic lupus erythematosus (SLE), including flare prevention without glucocorticoids; Sjögren's syndrome; Primary biliary cirrhosis (PBC); Systemic scleroderma; Polymyositis; Diabetes prevention; IgA nephropathy; IgA vasculitis; B cell cancers, for example myeloma; Multiple sclerosis or Optic neuritis.
  • SLE Systemic lupus erythematosus
  • PBC Primary biliary cirrhosis
  • Systemic scleroderma Polymyositis
  • Diabetes prevention IgA nephropathy
  • IgA vasculitis B cell cancers, for example myeloma
  • Multiple sclerosis or Optic neuritis for example myeloma.
  • the provided immunomodulatory proteins can be used to treat pre-B or B-cell leukemias, such as plasma cell leukemia, chronic or acute lymphocytic leukemia, myelomas such as multiple myeloma, plasma cell myeloma, endothelial myeloma and giant cell myeloma, and lymphomas such as non-Hodgkins lymphoma.
  • pre-B or B-cell leukemias such as plasma cell leukemia, chronic or acute lymphocytic leukemia, myelomas such as multiple myeloma, plasma cell myeloma, endothelial myeloma and giant cell myeloma, and lymphomas such as non-Hodgkins lymphoma.
  • the provided immunomodulatory proteins can be used as immunosuppressants to selectively block the action of B-lymphocytes for use in treating disease.
  • certain autoimmune diseases are characterized by production of autoantibodies, which contribute to tissue destruction and exacerbation of disease.
  • Autoantibodies can also lead to the occurrence of immune complex deposition complications and lead to many symptoms of systemic lupus erythematosus, including kidney failure, neuralgic symptoms and death. Modulating antibody production independent of cellular response would also be beneficial in many disease states.
  • B cells have also been shown to play a role in the secretion of arthritogenic immunoglobulins in rheumatoid arthritis.
  • Methods and uses of the provided immunomodulatory proteins to inhibit, block or neutralize action of B cell cells to thereby suppress antibody production would be beneficial in treatment of autoimmune diseases such as myasthenia gravis, rheumatoid arthritis, polyarticular-course juvenile rheumatoid arthritis, and psoriatic arthritis.
  • the provided immunomodulatory proteins can be used to block or neutralize the actions of B-cells in association with end stage renal diseases, which may or may not be associated with autoimmune diseases.
  • Such methods would also be useful for treating immunologic renal diseases.
  • Such methods would be useful for treating glomerulonephritis associated with diseases such as membranous nephropathy, IgA nephropathy or Berger's Disease, IgM nephropathy, IgA Vasculitis, Goodpasture's Disease, post-infectious glomerulonephritis, mesangioproliferative disease, chronic lymphoid leukemia, minimal-change nephrotic syndrome.
  • Such methods would also serve as therapeutic applications for treating secondary glomerulonephritis or vasculitis associated with such diseases as lupus, polyarteritis, Henoch-Schonlein, Scleroderma, HTV-related diseases, amyloidosis or hemolytic uremic syndrome.
  • the provided methods would also be useful as part of a therapeutic application for treating interstitial nephritis or pyelonephritis associated with chronic pyelonephritis, analgesic abuse, nephrocalcinosis, nephropathy caused by other agents, nephrolithiasis, or chronic or acute interstitial nephritis.
  • the methods provided herein also include use of the provided immunomodulatory proteins in the treatment of hypertensive or large vessel diseases, including renal artery stenosis or occlusion and cholesterol emboli or renal emboli.
  • the provided methods and uses also can be used for treatment of renal or urological neoplasms, multiple myelomas, lymphomas, light chain neuropathy or amyloidosis.
  • the provided immunomodulatory proteins also can be used for the treatment of asthma and other chronic airway diseases such as bronchitis and emphysema.
  • the provided immunomodulatory proteins can also be used to treat Sjögren's Syndrome.
  • methods and uses of the provided immunomodulatory proteins include immunosuppression, in particular for such therapeutic use as for graft-versus-host disease and graft rejection.
  • methods and uses of the provided immunomodulatory proteins include treatment of such autoimmune diseases as insulin dependent diabetes mellitus (IDDM) and Crohn's Disease. Methods provided herein would have additional therapeutic value for treating chronic inflammatory diseases, in particular to lessen joint pain, swelling, anemia and other associated symptoms as well as treating septic shock.
  • IDDM insulin dependent diabetes mellitus
  • Crohn's Disease Methods provided herein would have additional therapeutic value for treating chronic inflammatory diseases, in particular to lessen joint pain, swelling, anemia and other associated symptoms as well as treating septic shock.
  • the inflammatory and autoimmune disorders that can be treated by a pharmaceutical composition containing an immunomodulatory protein described herein include, but are not limited to, Achalasia; Addison's disease; Adult Still's disease; Agammaglobulinemia; Alopecia areata; Amyloidosis; Ankylosing spondylitis; Anti-GBM/Anti-TBM nephritis; Antiphospholipid syndrome; Autoimmune adrenalitis (Addison's disease); Autoimmune angioedema; Autoimmune dysautonomia; Autoimmune encephalomyelitis; Autoimmune hepatitis; Autoimmune inner ear disease (AIED); Autoimmune myocarditis; Autoimmune oophoritis; Autoimmune orchitis; Autoimmune pancreatitis; Autoimmune polyglandular syndrome type II (APS II); Autoimmune retinopathy; Autoimmune thyroid disease (AITD), i.
  • Hashimoto's disease Autoimmune urticarial; Axonal & neuronal neuropathy (AMAN); Baló disease; Behcet's disease; Benign mucosal pemphigoid; Bullous pemphigoid; Castleman disease (CD); Celiac disease; Chagas disease; Chronic inflammatory demyelinating polyneuropathy (CIDP); Chronic recurrent multifocal osteomyelitis (CRMO); Churg-Strauss Syndrome (CSS) or Eosinophilic Granulomatosis (EGPA); Cicatricial pemphigoid; Cogan's syndrome; Cold agglutinin disease; Congenital heart block; Coxsackie myocarditis; CREST syndrome; Crohn's disease; Dermatitis herpetiformis; Dermatomyositis; Devic's disease (neuromyelitis optica); Discoid lupus; Dressler's syndrome; Endometriosis; Eosinophilic esoph
  • the provided immunomodulatory proteins can be used to treat Scleroderma, Myasthenia gravis, GVHD (including acute GVHD or chronic GVHD), an immune response in connection with transplantation; Antiphospholipid Ab syndrome; Multiple sclerosis; Sjögren's syndrome; IgG4-related disease; Type I diabetes; Rheumatoid arthritis including glucocorticoid therapy (GC) RA or Acute lupus nephritis.
  • GVHD including acute GVHD or chronic GVHD
  • the provided immunomodulatory proteins can be used to treat Amyotrophic lateral sclerosis, Neuromyelitis optica, Transverse myelitis, CNS autoimmunity, Guillain-barre syndrome, Neurocystercercosis, Sarcoidosis (T/seroneg), Churg-Strauss Syndrome, Hashimoto's thyroiditis, Grave's disease, immune thrombocytopenia (ITP), Addison's Disease, Polymyositis, or Dermatomyositis.
  • TIM-BIM multi-domain immunomodulatory
  • the provided immunomodulatory proteins including BCMA-Fc fusion proteins and provided multi-domain immunomodulatory (e.g. TIM-BIM) fusion proteins, can be used to treat IgA nephropathy, chronic inflammatory demyelinating polyneuropathy (CIDP), antisynthetase disease such as Jo-1 syndrome, or ANCA vasculitis.
  • CIDP chronic inflammatory demyelinating polyneuropathy
  • ANCA vasculitis ANCA vasculitis
  • the provided immunomodulatory proteins can be used to treat a B cell cancer.
  • the B cell cancer is a cancer in which BAFF and APRIL are involved or implicated in providing an autocrine survival loop to the B cells.
  • the cancer is B cell chronic lymphocytic leukemia, non-Hodgkins' lymphoma or myeloma. In some embodiments, the cancer is myeloma.
  • a therapeutic amount of the pharmaceutical 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, extent of infection, and condition of the patient (subject).
  • 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.
  • compositions described herein may be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, intramedullary, intramuscularly, by intravenous (i.v.) injection, or intraperitoneally.
  • the therapeutic composition is administered to a patient by intradermal or subcutaneous injection.
  • the therapeutic composition is administered by i.v. injection.
  • 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 immunosuppressant agents).
  • the additional agent is a glucocorticoid (e.g., prednisone, dexamethasone, and hydrocortisone), cytostatic agent, such as a cytostatic agent that affect proliferation of T cells and/or B cells (e.g., purine analogs, alkylating agents, or antimetabolites), an antibody (e.g., anti-CD20, anti-CD25 or anti-CD3 monoclonal antibodies), cyclosporine, tacrolimus, sirolimus, everolimus, an interferon, an opiod, a TNF binding protein, mycophenolate, small biological agent, such as fingolimod or myriocin, cytokine, such as interferon beta-1a, an integrin agonist,
  • 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.
  • An immunomodulatory protein comprising:
  • TIM T cell inhibitory molecule
  • BIM B cell inhibitory molecule
  • the T cell stimulatory receptor is CD28; and/or
  • the ligand of the T cell stimulatory receptor is CD80 or CD86.
  • CTLA-4 extracellular domain or binding portion thereof consists of (i) the sequence of amino acids set forth in SEQ ID NO:1 or SEQ ID NO:2, (ii) a variant CTLA-4 sequence of amino acids that has at least 85% sequence identity to SEQ ID NO:1 or SEQ ID NO:2; or (iii) a portion thereof comprising an IgV domain.
  • CTLA-4 extracellular domain consists of a variant CTLA-4 sequence of amino acids that has at least 85% sequence identity to SEQ ID NO:1 or a portion thereof comprising an IgV domain, wherein the variant sequence comprises one or more amino acid substitutions in SEQ ID NO:1 or the portion thereof comprising the IgV domain.
  • the BIM is a TACI polypeptide that consists of the TACI extracellular domain or a binding portion thereof that binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • the TACI polypeptide is a truncated wild-type TACI extracellular domain or is a variant thereof, wherein the truncated wild-type TACI extracellular domain contains the cysteine rich domain 2 (CRD2) but lacks the entirety of the cysteine rich domain 1 (CRD1), wherein the variant TACI polypeptide comprises one or more amino acid substitutions in the truncated wild-type TACI extracellular domain.
  • TACI polypeptide is a truncated wild-type TACI extracellular domain or is a variant thereof, wherein the truncated wild-type TACI extracellular domain consists of a contiguous sequence contained within amino acid residues 67-118 that consists of amino acid residues 71-104, with reference to positions set forth in SEQ ID NO:709, wherein the variant TACI polypeptide comprises one or more amino acid substitutions in the truncated wild-type TACI extracellular domain.
  • the BIM is a BCMA polypeptide that consists of the BCMA extracellular domain or a binding portion thereof that binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • immunomodulatory protein of any of embodiments 14-16, wherein the truncated wild-type TACI extracellular domain is 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 59, 50 or 51 amino acids in length.
  • TACI polypeptide is a truncated wild-type TACI extracellular domain that consists of the sequence set forth in SEQ ID NO: 516.
  • TACI polypeptide is a truncated wild-type TACI extracellular domain that consists of the sequence set forth in SEQ ID NO:528.
  • TACI polypeptide is the variant TACI polypeptide, wherein the variant TACI polypeptide has increased binding affinity to one or both of APRIL and BAFF compared to the truncated TACI polypeptide.
  • the one or more amino acid substitutions are D85E/K98T, I87L/K98T, L82P/I87L, G76S/P97S, K77E/R84L/F103Y, Y79F/Q99E, L83S/F103
  • TACI polypeptide is a variant TACI polypeptide that comprises one or more amino acid substitutions in the extracellular domain (ECD) of a reference TACI polypeptide or a specific binding fragment thereof at positions selected from among 40, 59, 60, 61, 74, 75, 76, 77, 78, 79, 82, 83, 84, 85, 86, 87, 88, 92, 95, 97, 98, 99, 101, 102 and 103, corresponding to numbering of positions set forth in SEQ ID NO:709.
  • ECD extracellular domain
  • TACI polypeptide is a truncated polypeptide consisting of the extracellular domain of TACI or a specific binding portion thereof that binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • the reference TACI polypeptide comprises (i) the sequence of amino acids set forth in SEQ ID NO:709, (ii) a sequence of amino acids that has at least 95% sequence identity to SEQ ID NO:709; or (iii) a portion of (i) or (ii) comprising one or both of a CRD1 domain and CRD2 domain that binds to APRIL, BAFF, or a BAFF/APRIL heterotrimer.
  • immunomodulatory protein of any of embodiments 33-36 and 40, wherein the reference TACI polypeptide comprises the sequence set forth in SEQ ID NO:528.
  • immunomodulatory protein of any of embodiments 33-42 wherein the one or more amino acid substitutions are selected from W40R, Q59R, R60G, T61P E74V, Q75E, Q75R, G76S, K77E, F78Y, Y79F, L82H, L82P, L83S, R84G, R84L, R84Q, D85E, D85V, C86Y, I87L, I87M, S88N, I92V, Q95R, P97S, K98T, Q99E, A101D, Y102D, F103S, F103V, F103Y, or a conservative amino acid substitution thereof.
  • immunomodulatory protein of any of embodiments 33-44, wherein the one or more amino acid substitution comprise at least the amino acid substitution K77E.
  • variant TACI polypeptide comprises the sequence set forth in any one of SEQ ID NOS: 517-527, 536, 537, 682-701; or
  • the variant TACI polypeptide comprises the sequence set forth in any one of SEQ ID NOS: 529-535, 538-550, 673-681.
  • the variant TACI polypeptide consists or consists essentially of the sequence set forth in any one of SEQ ID NOS: 517-527, 536, 537, 682-701; or
  • the variant TACI polypeptide consists or consists essentially of the sequence set forth in any one of SEQ ID NOS: 529-535, 538-550, 673-681.
  • immunomodulatory protein of any of embodiments 15-16, 17-21 and 24-61 comprising a heterologous moiety that is linked to the at least one TACI polypeptide.
  • the immunomodulatory protein of embodiment 62, wherein the heterologous moiety is a half-life extending moiety, a multimerization domain, a targeting moiety that binds to a molecule on the surface of a cell, or a detectable label.
  • the immunomodulatory protein of embodiment 63 wherein 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.
  • BCMA polypeptide is a variant BCMA polypeptide comprising one or more amino acid substitutions in the extracellular domain (ECD) of a reference BCMA polypeptide or a specific binding fragment at positions selected from among 9, 10, 11, 14, 16, 19, 20, 22, 25, 27, 29, 30, 31, 32, 35, 36, 39, 43, 45, 46, 47 and 48, corresponding to numbering set forth in SEQ ID NO:710.
  • ECD extracellular domain
  • An immunomodulatory protein comprising a variant BCMA polypeptide, wherein the variant BCMA polypeptide comprises one or more amino acid substitutions in the extracellular domain (ECD) of a reference BCMA polypeptide or a specific binding fragment thereof at positions selected from among 9, 10, 11, 14, 16, 19, 20, 22, 25, 27, 29, 30, 31, 32, 35, 36, 39, 43, 45, 46, 47 and 48, corresponding to numbering of positions set forth in SEQ ID NO:710.
  • ECD extracellular domain
  • the immunomodulatory protein of any of embodiments 66-68, wherein the reference BCMA polypeptide comprises (i) the sequence of amino acids set forth in SEQ ID NO:710, (ii) a sequence of amino acids that has at least 95% 37a.equence identity to SEQ ID NO:710; or (iii) a portion of (i) or (ii) comprising the CRD.
  • the immunomodulatory protein of any of embodiments 66-70, wherein the reference BCMA polypeptide comprises (i) the sequence of amino acids set forth in SEQ ID NO:356, (ii) a sequence of amino acids that has at least 95% sequence identity to SEQ ID NO:356; or (iii) a portion of (i) or (ii) comprising the CRD.
  • immunomodulatory protein of any of embodiments 66-83, wherein the one or more amino acid substitutions are H19Y/S30G; H19Y/V45A; F14Y/H19Y; H19Y/V45D; H19Y/A43E; H19Y/T36A; H19Y/I22V; N11D/H19Y; H19Y/T36M; N11S/H19Y; H19Y/L35P/T46A; H19Y/N47D; S9D/H19Y; H19Y/S30G/V45D; H19Y/R39Q; H19Y/L35P; S9D/H19Y/R27H; Q10P/H19Y/Q25H; H19Y/R39L/N47D; N11D/H19Y/N47D; H19Y/T32S; N11S/H19Y/S29P; H19Y/R39Q/N47D; S16A/H19Y/R39
  • immunomodulatory protein of any of embodiments 66-75, 79, and 84, wherein the one or more amino acid substitutions comprise S16A/H19Y/R39Q.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Transplantation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
US17/923,208 2020-05-08 2021-05-07 April and baff inhibitory immunomodulatory proteins with and without a t cell inhibitory protein and methods of use thereof Pending US20230241168A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/923,208 US20230241168A1 (en) 2020-05-08 2021-05-07 April and baff inhibitory immunomodulatory proteins with and without a t cell inhibitory protein and methods of use thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US202063022373P 2020-05-08 2020-05-08
US202063034361P 2020-06-03 2020-06-03
US202063080643P 2020-09-18 2020-09-18
PCT/US2021/031432 WO2021226553A2 (fr) 2020-05-08 2021-05-07 Protéines immunomodulatrices inhibitrices de baff et april avec et sans protéine inhibitrice des lymphocytes t et leurs procédés d'utilisation
US17/923,208 US20230241168A1 (en) 2020-05-08 2021-05-07 April and baff inhibitory immunomodulatory proteins with and without a t cell inhibitory protein and methods of use thereof

Publications (1)

Publication Number Publication Date
US20230241168A1 true US20230241168A1 (en) 2023-08-03

Family

ID=76270049

Family Applications (3)

Application Number Title Priority Date Filing Date
US17/923,208 Pending US20230241168A1 (en) 2020-05-08 2021-05-07 April and baff inhibitory immunomodulatory proteins with and without a t cell inhibitory protein and methods of use thereof
US17/315,168 Pending US20210388054A1 (en) 2020-05-08 2021-05-07 April and baff inhibitory immunomodulatory proteins and methods of use thereof
US17/346,095 Active US11274140B2 (en) 2020-05-08 2021-06-11 APRIL and BAFF inhibitory immunomodulatory proteins and methods of use thereof

Family Applications After (2)

Application Number Title Priority Date Filing Date
US17/315,168 Pending US20210388054A1 (en) 2020-05-08 2021-05-07 April and baff inhibitory immunomodulatory proteins and methods of use thereof
US17/346,095 Active US11274140B2 (en) 2020-05-08 2021-06-11 APRIL and BAFF inhibitory immunomodulatory proteins and methods of use thereof

Country Status (15)

Country Link
US (3) US20230241168A1 (fr)
EP (2) EP4146684A2 (fr)
JP (2) JP2023525033A (fr)
KR (2) KR20230029621A (fr)
CN (4) CN116903727A (fr)
AU (2) AU2021267276A1 (fr)
BR (2) BR112022022433A2 (fr)
CA (2) CA3178885A1 (fr)
CL (1) CL2022003104A1 (fr)
CO (1) CO2022017780A2 (fr)
IL (2) IL297980A (fr)
MX (2) MX2022013999A (fr)
PE (1) PE20230494A1 (fr)
TW (1) TW202208414A (fr)
WO (2) WO2021226551A1 (fr)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2020536552A (ja) 2017-10-10 2020-12-17 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 Ctla−4変異型免疫調節タンパク質およびそれらの使用
EP3868403A4 (fr) * 2019-12-24 2022-04-06 RemeGen Co., Ltd. Protéine de fusion taci-fc et son utilisation
JP2024518163A (ja) * 2021-05-07 2024-04-25 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 TACI-Fc融合免疫調節タンパク質を用いた投薬および処置の方法
AU2022392003A1 (en) * 2021-11-17 2024-05-23 Aurinia Pharmaceuticals Inc. B cell activating factor (baff)-a proliferation inducing ligand (april) dual inhibitors
WO2023236967A1 (fr) * 2022-06-08 2023-12-14 荣昌生物制药(烟台)股份有限公司 Méthode de traitement de la myasthénie grave avec une protéine de fusion taci-fc
WO2024044721A1 (fr) * 2022-08-25 2024-02-29 University Of Washington Hydrogels injectables à base de protéines recombinantes pour une administration thérapeutique

Family Cites Families (137)

* 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
US4751180A (en) 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
US4935233A (en) 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
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
AU600575B2 (en) 1987-03-18 1990-08-16 Sb2, Inc. Altered antibodies
US5443964A (en) 1987-08-10 1995-08-22 Duke University Poxvirus insertion/expression vector
US5283173A (en) 1990-01-24 1994-02-01 The Research Foundation Of State University Of New York System to detect protein-protein interactions
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
US6407213B1 (en) 1991-06-14 2002-06-18 Genentech, Inc. Method for making humanized antibodies
US5262522A (en) 1991-11-22 1993-11-16 Immunex Corporation Receptor for oncostatin M and leukemia inhibitory factor
EP0714409A1 (fr) 1993-06-16 1996-06-05 Celltech Therapeutics Limited Anticorps
US5457035A (en) 1993-07-23 1995-10-10 Immunex Corporation Cytokine which is a ligand for OX40
EP0783573B1 (fr) 1994-09-23 2005-12-21 The University of British Columbia Procede d'accentuation de l'expression de molecules de classe i du complexe majeur d'histocompatibilite portant des peptides endogenes
ATE197765T1 (de) 1994-10-03 2000-12-15 Us Gov Health & Human Serv Zusammensetzung enthaltend ein antigen exprimierendes rekombinantes virus und ein immunstimulierendes molekül exprimierendes rekombinantes virus
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
BR9612752A (pt) 1996-10-25 2000-01-18 Human Genome Sciences Inc Neutrocina
US5969102A (en) 1997-03-03 1999-10-19 St. Jude Children's Research Hospital Lymphocyte surface receptor that binds CAML, nucleic acids encoding the same and methods of use thereof
IL132560A0 (en) 1997-05-02 2001-03-19 Genentech Inc A method for making multispecific antibodies having heteromultimeric and common components
ES2532910T3 (es) 1998-04-02 2015-04-01 Genentech, Inc. Variantes de anticuerpos y fragmentos de los mismos
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
GB2337755B (en) 1998-05-29 2003-10-29 Secr Defence Virus vaccine
EP2272859B1 (fr) 1998-08-07 2014-10-22 University of Washington Antigènes immunologiques du virus de l'herpès simplex et méthodes pour leur utilisation
US20060067933A1 (en) 1999-01-07 2006-03-30 Gross Jane A Soluble receptor BR43x2 and methods of using
KR100699524B1 (ko) 1999-01-07 2007-03-23 지모제넥틱스, 인코포레이티드 가용성 수용체 br43×2 및 이용 방법
US7833529B1 (en) * 1999-01-07 2010-11-16 Zymogenetics, Inc. Methods for inhibiting B lymphocyte proliferation with soluble ztnf4 receptor
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
ES2694002T3 (es) 1999-01-15 2018-12-17 Genentech, Inc. Polipéptido que comprende una región Fc de IgG1 humana variante
US20030022233A1 (en) 1999-04-30 2003-01-30 Raymond G. Goodwin Methods of use of the taci/taci-l interaction
ATE403715T1 (de) 1999-08-09 2008-08-15 Targeted Genetics Corp Erhöhung der expression einer einzelsträngigen, heterologen nukleotidsequenz von einem rekombinanten viralen vektor durch ausgestaltung der sequenz in einer art und weise, dass basenpaarungen innerhalb der sequenz entstehen
EP1255558B1 (fr) 2000-02-16 2006-06-14 Genentech, Inc. Anticorps anti-april et cellules hybridomes
EP1746106A3 (fr) 2000-04-27 2007-03-14 Biogen Idec MA Inc. TACI utilisée en tant qu'agent antitumoral
PT1297013E (pt) 2000-04-27 2006-12-29 Apoxis Sa Utilização de taci como um agente anti-tumor
US20040013674A1 (en) 2001-04-27 2004-01-22 Christine Ambrose Taci as an anti-tumor agent
AU2001261557B2 (en) 2000-05-12 2005-06-30 Amgen Inc. Methods and compositions of matter concerning april/g70, bcma, blys/agp-3, and taci
AU2001282856A1 (en) 2000-06-15 2001-12-24 Human Genome Sciences, Inc. Human tumor necrosis factor delta and epsilon
AU2002238052A1 (en) 2001-02-20 2002-09-04 Zymogenetics, Inc. Antibodies that bind both bcma and taci
JP2004537290A (ja) 2001-05-24 2004-12-16 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド 腫瘍壊死因子δ(APRIL)に対する抗体
BRPI0209933B8 (pt) 2001-05-24 2021-05-25 Zymogenetics Inc proteína de fusão, e, molécula de ácido nucleico
EA200601861A1 (ru) 2001-08-03 2007-02-27 Дженентек, Инк. ПОЛИПЕПТИДЫ TACIs И BR3 И ИХ ПРИМЕНЕНИЕ
ATE454447T1 (de) 2001-11-30 2010-01-15 Us Gov Health & Human Serv Peptidagonisten für prostata-spezifisches antigen und ihre verwendung
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
CN1692127A (zh) 2002-07-25 2005-11-02 健泰科生物技术公司 Taci抗体及其用途
ATE441428T1 (de) 2002-10-11 2009-09-15 Zymogenetics Inc Herstellung von homotrimeren fusionsproteinen
US7361740B2 (en) 2002-10-15 2008-04-22 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
EP1944320A1 (fr) 2002-12-16 2008-07-16 Genentech, Inc. Variantes de l'immunoglobuline et leurs utilisations
JP2006517191A (ja) 2002-12-30 2006-07-20 アムジエン・インコーポレーテツド 共刺激因子を用いた併用療法
US20050163775A1 (en) 2003-06-05 2005-07-28 Genentech, Inc. Combination therapy for B cell disorders
MXPA05013117A (es) 2003-06-05 2006-03-17 Genentech Inc Terapia de combinacion para trastornos de celulas-b.
EP1675609A1 (fr) 2003-10-20 2006-07-05 Biogen Idec MA Inc. Regimes therapeutiques pour antagonistes baff
ZA200604864B (en) 2003-12-19 2007-10-31 Genentech Inc Monovalent antibody fragments useful as therapeutics
SG172616A1 (en) 2004-04-13 2011-07-28 Hoffmann La Roche Anti-p-selectin antibodies
WO2006019447A1 (fr) 2004-07-15 2006-02-23 Xencor, Inc. Variantes genetiques de fc optimisees
TWI380996B (zh) 2004-09-17 2013-01-01 Hoffmann La Roche 抗ox40l抗體
EP1810979B1 (fr) 2004-09-22 2012-06-20 Kyowa Hakko Kirin Co., Ltd. ANTICORPS IgG4 HUMAINS STABILISES
EP1812472A1 (fr) 2004-11-04 2007-08-01 Genentech, Inc. Polypeptides se liant avec baff et/ou april
EP1819823A2 (fr) 2004-12-01 2007-08-22 Bayer Schering Pharma Aktiengesellschaft Generation de virus capables de replication a usage therapeutique
JP2008526205A (ja) * 2004-12-31 2008-07-24 ジェネンテック・インコーポレーテッド Br3に結合するポリペプチド及びその使用
AU2006232287B2 (en) 2005-03-31 2011-10-06 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
AU2006278229B2 (en) 2005-08-09 2011-10-27 Ares Trading S.A. Methods for treating B-cell malignancies using TACI-Ig fusion molecule
JP5118037B2 (ja) 2005-08-09 2013-01-16 ザイモジェネティクス, インコーポレイテッド Taci融合分子を用いた異常細胞増殖の処置及び予防のための方法
EP1924286A4 (fr) 2005-08-12 2010-01-13 Garvan Inst Med Res Procédé thérapeutique et/ou phropylactique pour le traitement d'une maladie auto-immune
US7655439B2 (en) 2005-09-09 2010-02-02 Zymogenetics, Inc. Trimerizing polypeptides
US9168286B2 (en) 2005-10-13 2015-10-27 Human Genome Sciences, Inc. Methods and compositions for use in treatment of patients with autoantibody positive disease
EP3037544A1 (fr) 2005-10-13 2016-06-29 Human Genome Sciences, Inc. Procedes et compositions destinees au traitement de patients atteints de lupus erythematosus systémique positifs pour des auto-anticorps
AU2007249223B2 (en) * 2006-05-15 2012-08-02 Ares Trading S.A. Methods for treating autoimmune diseases using a TACI-Ig fusion molecule
JP2010501623A (ja) 2006-08-28 2010-01-21 アレス トレーディング ソシエテ アノニム Fc含有タンパク質の精製法
EA017733B1 (ru) 2006-08-28 2013-02-28 Арес Трейдинг С.А. СПОСОБ ОЧИСТКИ Fc-СЛИТЫХ БЕЛКОВ
JP2010501622A (ja) 2006-08-28 2010-01-21 アレス トレーディング ソシエテ アノニム Fc−融合タンパク質の精製法
WO2008092117A2 (fr) 2007-01-25 2008-07-31 Xencor, Inc. Nouvelles insertions, délétions et substitutions d'immunoglobulines
CA2674213A1 (fr) 2007-01-26 2008-07-31 Merck Serono S.A. Purification de proteines hybrides fc-tact utilisant la technologie a base de corps huileux
RS52888B (en) 2007-03-27 2014-02-28 Zymogenetics Inc. COMBINATION OF BLYS INHIBITION AND MYPHOPHENOLATE MOFETILE FOR THE TREATMENT OF AUTOMMUNE DISEASE
MX2009013183A (es) * 2007-06-13 2010-01-20 Zymogenetics Inc Uso de la proteina de fusion taci-ig, tal como atacicept, para la elaboracion de un medicamento para tratar lupus eritematoso.
CN101323643B (zh) 2007-06-15 2010-12-01 烟台荣昌生物工程有限公司 优化的TACI-Fc融合蛋白
EA021222B8 (ru) 2007-06-21 2015-11-30 ИксЭль-ПРОТЕИН ГМБХ Биологически активные белки, имеющие повышенную стабильность in vivo и/или in vitro
EP2233149B1 (fr) 2007-10-16 2016-02-10 ZymoGenetics, Inc. Combinaison de l'activateur transmembranaire et modulateur calcique et interacteur du ligand de cyclophiline (TACI) et d'un agent anti-CD20 pour le traitement des maladies auto-immunes
JP2011500757A (ja) 2007-10-22 2011-01-06 メルク セローノ ソシエテ アノニム Fc含有タンパク質の精製方法
WO2009062926A1 (fr) 2007-11-12 2009-05-22 Ares Trading S.A. Protéines hybrides taci-immunoglobuline pour le traitement de rechute de la sclérose en plaques
EP2219675B1 (fr) 2007-11-12 2013-10-23 Ares Trading S.A. Formulations pour protéines hybrides taci-immunoglobuline
WO2009062916A1 (fr) 2007-11-12 2009-05-22 Ares Trading S.A. Protéines de fusion taci-immunoglobuline pour le traitement d'une névrite optique
EP2222861B1 (fr) 2007-12-11 2017-12-06 The University of North Carolina At Chapel Hill Vecteurs rétroviraux modifiés dépourvus de sequence riche en polypurine (ppt)
CA2720682A1 (fr) 2008-04-25 2009-10-29 Zymogenetics, Inc. Niveaux d'expression de la proteine bcma sur les lymphocytes b et utilisation dans de methodes diagnostiques
US8003335B2 (en) 2008-04-30 2011-08-23 Universite Paris-SUD11 Levels of APRIL in serum and use in diagnostic methods
EP2291657B1 (fr) 2008-05-01 2016-04-13 Zymogenetics, Inc. Concentrations sériques en hétérotrimères blys/april et utilisation dans des procédés de diagnostic
EP2540740B1 (fr) 2008-06-17 2014-09-10 Apogenix GmbH Récepteurs multimériques TNF
CA2763439A1 (fr) 2009-05-28 2010-12-02 Glaxo Group Limited Proteines se liant a un antigene
WO2011109280A1 (fr) * 2010-03-05 2011-09-09 Lerner Research Institute Procédés et compositions permettant de traiter les troubles d'origines immunes
CN101851278B (zh) 2010-05-26 2013-03-13 石药集团中奇制药技术(石家庄)有限公司 B细胞激活因子拮抗剂及其制备方法与用途
WO2011160086A2 (fr) 2010-06-18 2011-12-22 Human Genome Sciences, Inc. Utilisation d'antagonistes d'une protéine stimulant les lymphocytes b en vue du traitement de l'asthme et d'autres affections allergiques et inflammatoires du système respiratoire
WO2012032112A1 (fr) 2010-09-10 2012-03-15 Cellerix, S.A. Milieux de culture de cellules souches et procédés
CN102085368B (zh) 2011-01-19 2013-06-12 烟台荣昌生物工程有限公司 优化的TACI-Fc融合蛋白用于制备治疗系统性红斑狼疮药物的应用
CA2830254C (fr) 2011-03-16 2019-09-10 Amgen Inc. Variants de fc
EP3632463A1 (fr) 2011-04-08 2020-04-08 Immune Design Corp. Compositions immunogènes et leurs procédés d'utilisation pour induire des réponses immunitaires humorales et cellulaires
ES2876421T3 (es) 2011-04-13 2021-11-12 Bristol Myers Squibb Co Proteínas de fusión Fc que comprenden enlazadores o disposiciones nuevos
WO2013041029A1 (fr) * 2011-09-23 2013-03-28 Igenimed Pharmaceuticals Inc. Nouveaux variants de ctla4 solubles
TWI476001B (zh) 2011-12-26 2015-03-11 Ind Tech Res Inst 三倍體Fc融合蛋白及其用途
EP3406347A3 (fr) 2012-02-27 2019-02-13 Amunix Operating Inc. Compositions de conjugués xten et leurs procédés de fabrication
WO2014126871A1 (fr) 2013-02-12 2014-08-21 Bristol-Myers Squibb Company Procédés de repliement de protéine utilisant la filtration tangentielle
EP3617220B1 (fr) 2013-02-12 2021-03-24 Bristol-Myers Squibb Company Procédés de repliement de protéine à ph élevé
JP2016515120A (ja) 2013-03-15 2016-05-26 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. 抗アルファvベータ5抗体を用いた急性腎損傷の治療及び予防
BR112016016416A2 (pt) 2014-01-15 2017-10-03 Hoffmann La Roche VARIANTES DE REGIÕES-"Fc" COM 'FcRn' MODIFICADAS E PROPRIEDADES DE LIGAÇÃO DE PROTEÍNA "A" MANTIDAS
US10562954B2 (en) * 2014-05-12 2020-02-18 Shanghai Kanda Biotechnology Co., Ltd. Fusion protein inhibiting TACI-BAFF complex formation and preparation method therefor and use thereof
AU2015289773A1 (en) 2014-07-15 2017-02-02 Immune Design Corp. Prime-boost regimens with a TLR4 agonist adjuvant and a lentiviral vector
CN115058395A (zh) 2015-06-25 2022-09-16 美商生物细胞基因治疗有限公司 嵌合抗原受体(car)、组合物及其使用方法
MA42895A (fr) 2015-07-15 2018-05-23 Juno Therapeutics Inc Cellules modifiées pour thérapie cellulaire adoptive
TW202208440A (zh) * 2015-12-14 2022-03-01 美商宏觀基因股份有限公司 對於pd-1和ctla-4具有免疫反應性的雙特異性分子及其使用方法
CN109715657A (zh) 2016-04-15 2019-05-03 高山免疫科学股份有限公司 Cd80变体免疫调节蛋白及其用途
KR20230051602A (ko) 2016-04-15 2023-04-18 알파인 이뮨 사이언시즈, 인코포레이티드 Icos 리간드 변이체 면역조절 단백질 및 그의 용도
EP3452101A2 (fr) 2016-05-04 2019-03-13 CureVac AG Arn codant pour une protéine thérapeutique
JP7114490B2 (ja) 2016-06-24 2022-08-08 アイセル・ジーン・セラピューティクス・エルエルシー キメラ抗体受容体(CARs)の構成およびその使用方法
WO2018022945A1 (fr) 2016-07-28 2018-02-01 Alpine Immune Sciences, Inc. Protéines immunomodulatrices à variants de cd112 et utilisations associées
CA3032120A1 (fr) 2016-07-28 2018-02-01 Alpine Immune Sciences, Inc. Proteines immunomodulatrices a variants de cd155 et leurs utilisations
CN110352245A (zh) 2016-10-20 2019-10-18 高山免疫科学股份有限公司 可分泌变体免疫调节蛋白和工程化细胞疗法
CN110809581A (zh) 2017-03-16 2020-02-18 高山免疫科学股份有限公司 Pd-l2变体免疫调节蛋白及其用途
WO2018170026A2 (fr) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Protéines immunomodulatrices à variants de cd80 et leurs utilisations
JP2020509776A (ja) 2017-03-16 2020-04-02 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 Pd−l1バリアント免疫調節タンパク質及びその使用
CN110945030A (zh) 2017-06-20 2020-03-31 丹娜法伯癌症研究院 使用april-taci相互作用的调节剂调节调控性t细胞、调控性b细胞和免疫响应的方法
WO2019046818A1 (fr) 2017-09-01 2019-03-07 Dana-Farber Cancer Institute, Inc. Peptides immunogènes spécifiques des antigènes bcma et taci pour le traitement du cancer
JP2020536552A (ja) * 2017-10-10 2020-12-17 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 Ctla−4変異型免疫調節タンパク質およびそれらの使用
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
GB201720426D0 (en) 2017-12-07 2018-01-24 Hummingbird Bioscience Pte Ltd CD47 and BCMA antigen-binding molecules
AU2019205273B2 (en) 2018-01-03 2024-04-04 Alpine Immune Sciences, Inc. Multi-domain immunomodulatory proteins and methods of use thereof
SG11202007771VA (en) 2018-03-02 2020-09-29 Allogene Therapeutics Inc Inducible chimeric cytokine receptors
CN110522908A (zh) 2018-05-25 2019-12-03 荣昌生物制药(烟台)有限公司 TACI-Fc融合蛋白用于制备治疗视神经脊髓炎谱系疾病和多发性硬化之药物中的应用
US20210363219A1 (en) 2018-06-15 2021-11-25 Alpine Immune Sciences, Inc. Pd-1 variant immunomodulatory proteins and uses thereof
WO2020028572A2 (fr) 2018-08-01 2020-02-06 Unum Therapeutics Inc. Récepteurs de lymphocyte t couplés à un anticorps (actr) en combinaison avec des molécules costimulatrices trans et leurs utilisations thérapeutiques
WO2020047329A1 (fr) * 2018-08-29 2020-03-05 Shattuck Labs, Inc. Protéines chimériques comprenant des domaines extracellulaires et leurs utilisations
JP2022501361A (ja) 2018-09-19 2022-01-06 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 バリアントcd80融合タンパク質および関連構築物の方法および使用
CA3117978A1 (fr) 2018-11-08 2020-05-14 Juno Therapeutics, Inc. Procedes et combinaisons pour le traitement et la modulation de lymphocytes t
WO2020113141A2 (fr) 2018-11-30 2020-06-04 Alpine Immune Sciences, Inc. Protéines immunomodulatrices à variants cd86 et leurs utilisations
US20220218785A1 (en) 2019-04-17 2022-07-14 Alpine Immune Sciences, Inc. Methods and uses of variant icos ligand (icosl) fusion proteins
AU2020292421A1 (en) 2019-06-14 2022-01-27 Cugene, Inc. Novel interleukin-2 variants and bifunctional fusion molecules thereof
EP4056232A4 (fr) 2019-09-13 2023-09-13 Kyowa Kirin Co., Ltd. Variant de dcr3
CA3128113A1 (fr) 2019-12-10 2021-06-17 Remegen Co., Ltd. Formulation pharmaceutique de proteine de fusion taci-fc
EP3868403A4 (fr) 2019-12-24 2022-04-06 RemeGen Co., Ltd. Protéine de fusion taci-fc et son utilisation

Also Published As

Publication number Publication date
CA3178882A1 (fr) 2021-11-11
US11274140B2 (en) 2022-03-15
CN116903727A (zh) 2023-10-20
MX2022013999A (es) 2023-02-16
CN116194467A (zh) 2023-05-30
EP4146683A1 (fr) 2023-03-15
PE20230494A1 (es) 2023-03-23
KR20230029621A (ko) 2023-03-03
BR112022022524A2 (pt) 2022-12-13
AU2021267276A1 (en) 2022-12-15
KR20230029622A (ko) 2023-03-03
TW202208414A (zh) 2022-03-01
MX2022013998A (es) 2023-02-16
WO2021226553A3 (fr) 2022-04-28
JP2023525033A (ja) 2023-06-14
CO2022017780A2 (es) 2023-03-17
CA3178885A1 (fr) 2021-11-11
EP4146684A2 (fr) 2023-03-15
WO2021226553A2 (fr) 2021-11-11
WO2021226551A1 (fr) 2021-11-11
CL2022003104A1 (es) 2023-07-14
IL297981A (en) 2023-01-01
CN117736297A (zh) 2024-03-22
CN115812077A (zh) 2023-03-17
IL297980A (en) 2023-01-01
US20210363223A1 (en) 2021-11-25
BR112022022433A2 (pt) 2022-12-13
US20210388054A1 (en) 2021-12-16
JP2023525032A (ja) 2023-06-14
AU2021268033A1 (en) 2022-12-15

Similar Documents

Publication Publication Date Title
US20230241168A1 (en) April and baff inhibitory immunomodulatory proteins with and without a t cell inhibitory protein and methods of use thereof
JP7148539B2 (ja) 免疫抱合体
AU2019205273B2 (en) Multi-domain immunomodulatory proteins and methods of use thereof
JP2016529215A (ja) Ctla−4およびcd40の両方に特異的に結合可能である二重特異性分子
CN110831963A (zh) Pd-l1变体免疫调节蛋白及其用途
US20220218785A1 (en) Methods and uses of variant icos ligand (icosl) fusion proteins
JP2023138962A (ja) Ctla-4変異型免疫調節タンパク質およびそれらの使用
CN115916827A (zh) 免疫活化Fc结构域结合分子
JP2020536552A5 (fr)
US20230303648A1 (en) Bifunctional molecules comprising an il-7 variant
KR20240019124A (ko) Taci-fc 융합 면역조절 단백질의 투여 및 치료 방법
WO2024077018A2 (fr) Procédés et utilisations d'une protéine immunomodulatrice de fusion taci-fc
CN117915937A (zh) 使用TACI-Fc融合免疫调节蛋白的给药和治疗方法

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: AWAITING RESPONSE FOR INFORMALITY, FEE DEFICIENCY OR CRF ACTION

AS Assignment

Owner name: ALPINE IMMUNE SCIENCES, INC., WASHINGTON

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DILLON, STACEY;RIXON, MARK;EVANS, LAWRENCE;AND OTHERS;SIGNING DATES FROM 20240415 TO 20240417;REEL/FRAME:067167/0291