US20190365861A1 - Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof - Google Patents

Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof Download PDF

Info

Publication number
US20190365861A1
US20190365861A1 US16/388,174 US201916388174A US2019365861A1 US 20190365861 A1 US20190365861 A1 US 20190365861A1 US 201916388174 A US201916388174 A US 201916388174A US 2019365861 A1 US2019365861 A1 US 2019365861A1
Authority
US
United States
Prior art keywords
amino acid
variant
substitutions
domain
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/388,174
Other languages
English (en)
Inventor
Matthew Bernett
John Desjarlais
Rumana Rashid
Rajat Varma
Christine Bonzon
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.)
Xencor Inc
Original Assignee
Xencor 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 Xencor Inc filed Critical Xencor Inc
Priority to US16/388,174 priority Critical patent/US20190365861A1/en
Publication of US20190365861A1 publication Critical patent/US20190365861A1/en
Assigned to XENCOR, INC. reassignment XENCOR, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DESJARLAIS, JOHN, VARMA, Rajat, BERNETT, MATTHEW J., BONZON, CHRISTINE, RASHID, Rumana
Priority to US17/209,047 priority patent/US20220040264A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • 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
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2086IL-13 to IL-16
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5443IL-15
    • 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
    • C07K14/7155Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2812Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2815Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD8
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/289Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD45
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/524CH2 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • 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
    • C07K2317/71Decreased effector function due to an Fc-modification
    • 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
    • C07K2317/72Increased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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

Definitions

  • Cytokines such as IL-2 and IL-15 function in aiding the proliferation and differentiation of B cells, T cells, and NK cells. Both cytokines exert their cell signaling function through binding to a trimeric complex consisting of two shared receptors, the common gamma chain ( ⁇ c; CD132) and IL-2 receptor beta-chain (IL-2R ⁇ ; CD122), as well as an alpha chain receptor unique to each cytokine: IL-2 receptor alpha (IL-2R ⁇ ; CD25) or IL-15 receptor alpha (IL-15R ⁇ ; CD215). Both cytokines are considered as potentially valuable therapeutics in oncology, and IL-2 has been approved for use in patients with metastatic renal-cell carcinoma and malignant melanoma.
  • Immune checkpoint proteins such as PD-1 are up-regulated following T cell activation to preclude autoimmunity by exhausting activated T cells upon binding to immune checkpoint ligands such as PD-L1.
  • immune checkpoint proteins are also up-regulated in tumor-infiltrating lymphocytes (TILs), and immune checkpoint ligands are overexpressed on tumor cells, contributing to immune escape by tumor cells.
  • TILs tumor-infiltrating lymphocytes
  • De-repression of TILs by blockade of immune checkpoint interactions by drugs such as Opdivo® (nivolumab) and Keytruda® (pembrolizumab) have proven highly effective in treatment of cancer.
  • checkpoint blockade therapies such as nivolumab and pembrolizumab, many patients still fail to achieve sufficient response to checkpoint blockade alone.
  • the present invention is directed to administering a novel IL-15/IL-15R ⁇ heterodimeric Fc fusion protein in combination with a checkpoint blockade antibody.
  • the checkpoint blockade antibody selected from the group consisting of an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM3 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, and an anti-CTLA-4 antibody.
  • provided herein is a method of treating cancer in a patient in need thereof comprising administering:
  • an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprising:
  • a therapeutically effective amount of a checkpoint blockade antibody selected from the group consisting of an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM3 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, and an anti-CTLA-4 antibody.
  • the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions D30N/E64Q/N65D. In some embodiments, the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions D30N/N65D. In some embodiments, the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions N4D/N65D.
  • the IL-15R ⁇ sushi domain comprises the amino acid sequence of SEQ ID NO:4.
  • the first and second variant Fc domains have S364K/E357Q:L368D/K370S substitutions. In some embodiments, the first variant Fc domain has S364K/E357Q substitutions and the second variant Fc domain L368D/K370S substitutions.
  • the first and second variant Fc domains each comprise M428L/N434S substitutions.
  • the first and second variant Fc domains each comprise E233P/L234V/L235A/G236del/S267K substitutions.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein and the checkpoint blockade antibody are administered concomitantly or sequentially.
  • the anti-PD-1 antibody is nivolumab, pembrolizumab, or pidilizumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 or XENP24045.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is nivolumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is pembrolizumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is pidilizumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is nivolumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is pembrolizumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is pidilizumab.
  • the cancer is metastatic cancer.
  • the cancer is selected from the group consisting of breast cancer, lung cancer, colon cancer, ovarian cancer, melanoma cancer, bladder cancer, renal cancer, kidney cancer, liver cancer, head and neck cancer, colorectal cancer, melanoma, pancreatic cancer, gastric carcinoma cancer, esophageal cancer, mesothelioma, prostate cancer, leukemia, lymphoma, and myeloma.
  • the method of treating cancer outlined herein results in a minimal level of vascular leakage in the patient.
  • the level of vascular leakage ranges from a 20% reduction or less in serum albumin in the patient following administration.
  • a method of treating cancer in a patient comprising administering a combination therapy comprising an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein and a checkpoint blockade antibody to the patient, wherein the IL-15/IL-15R ⁇ heterodimeric Fc fusion comprises:
  • the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions D30N/E64Q/N65D. In some embodiments, the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions D30N/N65D. In some embodiments, the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions N4D/N65D.
  • the IL-15R ⁇ sushi domain comprises the amino acid sequence of SEQ ID NO:4.
  • the first and second variant Fc domains have S364K/E357Q:L368D/K370S substitutions. In some embodiments, the first variant Fc domain has S364K/E357Q substitutions and the second variant Fc domain L368D/K370S substitutions.
  • the first and second variant Fc domains each comprise M428L/N434S substitutions.
  • the first and second variant Fc domains each comprise E233P/L234V/L235A/G236del/S267K substitutions.
  • the anti-PD-1 antibody is nivolumab, pembrolizumab, or pidilizumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) or XENP24045 (SEQ ID NOS 204 and 205).
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is nivolumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is pembrolizumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is pidilizumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is nivolumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is pembrolizumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is pidilizumab.
  • the cancer is metastatic cancer.
  • the cancer is selected from the group consisting of breast cancer, lung cancer, colon cancer, ovarian cancer, melanoma cancer, bladder cancer, renal cancer, kidney cancer, liver cancer, head and neck cancer, colorectal cancer, melanoma, pancreatic cancer, gastric carcinoma cancer, esophageal cancer, mesothelioma, prostate cancer, leukemia, lymphoma, and myeloma.
  • the method of treating cancer described herein results in a minimal level of vascular leakage in the patient.
  • the level of vascular leakage ranges from a 20% reduction or less in serum albumin in the patient following administration.
  • provided herein is a method of inducing T cell expansion in a patient comprising administering:
  • an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprising:
  • a therapeutically effective amount of a checkpoint blockade antibody selected from the group consisting of an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM3 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, and an anti-CTLA-4 antibody.
  • the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions D30N/E64Q/N65D. In some embodiments, the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions D30N/N65D. In some embodiments, the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions N4D/N65D.
  • the IL-15R ⁇ sushi domain comprises the amino acid sequence of SEQ ID NO:4.
  • the first and second variant Fc domains have S364K/E357Q:L368D/K370S substitutions. In some embodiments, the first variant Fc domain has S364K/E357Q substitutions and the second variant Fc domain L368D/K370S substitutions.
  • the first and second variant Fc domains each comprise M428L/N434S substitutions.
  • the first and second variant Fc domains each comprise E233P/L234V/L235A/G236del/S267K substitutions.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein and the checkpoint blockade antibody are administered concomitantly or sequentially.
  • the e anti-PD-1 antibody is nivolumab, pembrolizumab, or pidilizumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) or XENP24045 (SEQ ID NOS 204 and 205).
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is nivolumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is pembrolizumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is pidilizumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is nivolumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is pembrolizumab. In some embodiments, the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is pidilizumab.
  • the patient has cancer.
  • the cancer is metastatic cancer.
  • the cancer is selected from the group consisting of breast cancer, lung cancer, colon cancer, ovarian cancer, melanoma cancer, bladder cancer, renal cancer, kidney cancer, liver cancer, head and neck cancer, colorectal cancer, melanoma, pancreatic cancer, gastric carcinoma cancer, esophageal cancer, mesothelioma, prostate cancer, leukemia, lymphoma, and myeloma.
  • the T cell expansion is at least a 2-fold increase in T cells. In some embodiments, the T cell expansion ranges from a 2-fold to a 15-fold increase in T cells.
  • the method does not increase the likelihood of inducing hypoalbuminemia.
  • the T cells comprise tumor infiltrating lymphocytes.
  • the present invention provides a combination therapy comprising an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein and a checkpoint blockade antibody selected from an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM3 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, and an anti-CTLA-4 antibody.
  • a checkpoint blockade antibody selected from an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM3 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, and an anti-CTLA-4 antibody.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises:
  • the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions D30N/E64Q/N65D. In some embodiments, the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions D30N/N65D. In some embodiments, the variant IL-15 domain comprises the amino acid sequence of SEQ ID NO:2 and the amino acid substitutions N4D/N65D.
  • the IL-15R ⁇ sushi domain comprises the amino acid sequence of SEQ ID NO:4.
  • the first and second variant Fc domains have S364K/E357Q:L368D/K370S substitutions. In some embodiments, the first variant Fc domain has S364K/E357Q substitutions and the second variant Fc domain L368D/K370S substitutions.
  • the first and second variant Fc domains each comprise M428L/N434S substitutions.
  • the first and second variant Fc domains each comprise E233P/L234V/L235A/G236del/S267K substitutions.
  • the anti-PD-1 antibody is nivolumab, pembrolizumab, or pidilizumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) or XENP24045 (SEQ ID NOS 204 and 205).
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is nivolumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is pembrolizumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24306 (SEQ ID NOS 253 and 254) and the anti-PD-1 antibody is pidilizumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is nivolumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is pembrolizumab.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprises the amino acid sequence of XENP24045 (SEQ ID NOS 204 and 205) and the anti-PD-1 antibody is pidilizumab.
  • FIG. 1 depicts the structure of IL-15 in complex with its receptors IL-15R ⁇ (CD215), IL-15R ⁇ (CD122), and the common gamma chain (CD132).
  • FIG. 2A and FIG. 2B depict the sequences for IL-15 and its receptors.
  • FIG. 3A - FIG. 3E depict useful pairs of Fc heterodimerization variant sets (including skew and pI variants).
  • FIG. 3D and FIG. 3E there are variants for which there are no corresponding “monomer 2” variants; these are pI variants which can be used alone on either monomer.
  • FIG. 4 depict a list of isosteric variant antibody constant regions and their respective substitutions.
  • pI_( ) indicates lower pI variants, while pI_(+) indicates higher pI variants.
  • pI_(+) indicates higher pI variants.
  • FIG. 5 depict useful ablation variants that ablate Fc ⁇ R binding (sometimes referred to as “knock outs” or “KO” variants). Generally, ablation variants are found on both monomers, although in some cases they may be on only one monomer.
  • FIG. 6A - FIG. 6E show particularly useful embodiments of “non-cytokine” components of the invention.
  • FIG. 7 depicts a number of exemplary variable length linkers.
  • these linkers find use linking the C-terminus of IL-15 and/or IL-15R ⁇ (sushi) to the N-terminus of the Fc region.
  • these linkers find use fusing IL-15 to the IL-15R ⁇ (sushi).
  • FIG. 8A - FIG. 8D show the sequences of several useful IL-15/R ⁇ -Fc format backbones based on human IgG1, without the cytokine sequences (e.g., the II-15 and/or IL-15R ⁇ (sushi)).
  • Backbone 1 is based on human IgG1 (356E/358M allotype), and includes C220S on both chain, the S364K/E357Q:L368D/K370S skew variants, the Q295E/N384D/Q418E/N421D pI variants on the chain with L368D/K370S skew variants and the E233P/L234V/L235A/G236del/S267K ablation variants on both chains.
  • Backbone 2 is based on human IgG1 (356E/358M allotype), and includes C220S on both chain, the S364K:L368D/K370S skew variants, the Q295E/N384D/Q418E/N421D pI variants on the chain with L368D/K370S skew variants and the E233P/L234V/L235A/G236del/S267K ablation variants on both chains.
  • Backbone 3 is based on human IgG1 (356E/358M allotype), and includes C220S on both chain, the S364K:L368E/K370S skew variants, the Q295E/N384D/Q418E/N421D pI variants on the chain with L368E/K370S skew variants and the E233P/L234V/L235A/G236del/S267K ablation variants on both chains.
  • Backbone 4 is based on human IgG1 (356E/358M allotype), and includes C220S on both chain, the D401K:K360E/Q362E/T411E skew variants, the Q295E/N384D/Q418E/N421D pI variants on the chain with K360E/Q362E/T411E skew variants and the E233P/L234V/L235A/G236del/S267K ablation variants on both chains.
  • Backbone 5 is based on human IgG1 (356D/358L allotype), and includes C220S on both chain, the S364K/E357Q:L368D/K370S skew variants, the Q295E/N384D/Q418E/N421D pI variants on the chain with L368D/K370S skew variants and the E233P/L234V/L235A/G236del/S267K ablation variants on both chains.
  • Backbone 6 is based on human IgG1 (356E/358M allotype), and includes C220S on both chain, the S364K/E357Q:L368D/K370S skew variants, Q295E/N384D/Q418E/N421D pI variants on the chain with L368D/K370S skew variants and the E233P/L234V/L235A/G236del/S267K ablation variants on both chains, as well as an N297A variant on both chains.
  • Backbone 7 is identical to 6 except the mutation is N297S.
  • Backbone 8 is based on human IgG4, and includes the S364K/E357Q:L368D/K370S skew variants, the Q295E/N384D/Q418E/N421D pI variants on the chain with L368D/K370S skew variants, as well as a S228P (EU numbering, this is S241P in Kabat) variant on both chains that ablates Fab arm exchange as is known in the art.
  • Backbone 9 is based on human IgG2, and includes the S364K/E357Q:L368D/K370S skew variants, the Q295E/N384D/Q418E/N421D pI variants on the chain with L368D/K370S skew variants.
  • Backbone 10 is based on human IgG2, and includes the S364K/E357Q:L368D/K370S skew variants, the Q295E/N384D/Q418E/N421D pI variants on the chain with L368D/K370S skew variants as well as a S267K variant on both chains.
  • Backbone 11 is identical to backbone 1, except it includes M428L/N434S Xtend mutations.
  • Backbone 12 is based on human IgG1 (356E/358M allotype), and includes C220S on both identical chain, the E233P/L234V/L235A/G236del/S267K ablation variants on both identical chains.
  • Backbone 13 is based on human IgG1 (356E/358M allotype), and includes C220S on both chain, the S364K/E357Q:L368D/K370S skew variants, the P217R/P229R/N276K pI variants on the chain with S364K/E357Q skew variants and the E233P/L234V/L235A/G236del/S267K ablation variants on both chains.
  • any IL-15 and IL-15R ⁇ (sushi) pairs outlined herein including but not limited to IL-15/R ⁇ -heteroFc, ncIL-15/R ⁇ , scIL-15/R ⁇ , and dsIL-15/R ⁇ as schematically depicted in FIGS. 9A-9G and 39 .
  • any IL-15 and/or IL-15R ⁇ (sushi) variants can be incorporated into these FIG. 8A-8D backbones in any combination.
  • each of these backbones includes sequences that are 90, 95, 98 and 99% identical (as defined herein) to the recited sequences, and/or contain from 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 additional amino acid substitutions (as compared to the “parent” of the Figure, which, as will be appreciated by those in the art, already contain a number of amino acid modifications as compared to the parental human IgG1 (or IgG2 or IgG4, depending on the backbone). That is, the recited backbones may contain additional amino acid modifications (generally amino acid substitutions) in addition to the skew, pI and ablation variants contained within the backbones of this figure.
  • FIG. 9A - FIG. 9G depict several formats for the IL-15/R ⁇ -Fc fusion proteins of the present invention.
  • IL-15R ⁇ Heterodimeric Fc fusion or “IL-15/R ⁇ -heteroFc” ( FIG. 9A ) comprises IL-15 recombinantly fused to one side of a heterodimeric Fc and IL-15R ⁇ (sushi) recombinantly fused to the other side of a heterodimeric Fc.
  • the IL-15 and IL-15R ⁇ (sushi) may have a variable length Gly-Ser linker between the C-terminus and the N-terminus of the Fc region.
  • Single-chain IL-15/R ⁇ -Fc fusion or “scIL-15/R ⁇ -Fc” comprises IL-15R ⁇ (sushi) fused to IL-15 by a variable length linker (termed a “single-chain” IL-15/IL-15R ⁇ (sushi) complex or “scIL-15/R ⁇ ”) which is then fused to the N-terminus of a heterodimeric Fc-region, with the other side of the molecule being “Fc-only” or “empty Fc”.
  • Non-covalent IL-15/R ⁇ -Fc or “ncIL-15/R ⁇ -Fc” FIG.
  • FIG. 9C comprises IL-15R ⁇ (sushi) fused to a heterodimeric Fc region, while IL-15 is transfected separatedly so that a non-covalent IL-15/R ⁇ complex is formed, with the other side of the molecule being “Fc-only” or “empty Fc”.
  • Bivalent non-covalent IL-15/R ⁇ -Fc fusion or “bivalent ncIL-15/R ⁇ -Fc” ( FIG. 9D ) comprises IL-15R ⁇ (sushi) fused to the N-terminus of a homodimeric Fc region, while IL-15 is transfected separately so that a non-covalent IL-15/R ⁇ complex is formed.
  • Bivalent single-chain IL-15/R ⁇ -Fc fusion or “bivalent scIL-15/R ⁇ -Fc” comprises IL-15 fused to IL-15R ⁇ (sushi) by a variable length linker (termed a “single-chain” IL-15/IL-15R ⁇ (sushi) complex or “scIL-15/R ⁇ ”) which is then fused to the N-terminus of a homodimeric Fc-region.
  • Fc-non-covalent IL-15/R ⁇ fusion or “Fc-ncIL-15/R ⁇ ” FIG.
  • 9F comprises IL-15R ⁇ (sushi) fused to the C-terminus of a heterodimeric Fc region, while IL-15 is transfected separately so that a non-covalent IL-15/R ⁇ complex is formed, with the other side of the molecule being “Fc-only” or “empty Fc”.
  • Fc-single-chain IL-15/R ⁇ fusion or “Fc-scIL-15/R ⁇ ” FIG.
  • 9G comprises IL-15 fused to IL-15R ⁇ (sushi) by a variable length linker (termed a “single-chain” IL-15/IL-15R ⁇ (sushi) complex or “scIL-15/R ⁇ ”) which is then fused to the C-terminus of a heterodimeric Fc region, with the other side of the molecule being “Fc-only” or “empty Fc”.
  • a variable length linker termed a “single-chain” IL-15/IL-15R ⁇ (sushi) complex or “scIL-15/R ⁇ ”
  • FIG. 10 depicts sequences of XENP20818 and XENP21475, illustrative IL-15/R ⁇ -Fc fusion proteins of the “IL-15/R ⁇ -heteroFc” format, with additional sequences of XENP20819, XENP21471, XENP21472, XENP21473, XENP21474, XENP21476, XENP21477 being listed in WO2018071919 in FIGS. 104A-104D, respectively and as SEQ ID NOS: 418-423, 424-429, 430-435, 436-441, 442-447, 454-459, and 460-465, respectively.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 11 depicts sequences of XENP21478, an illustrative IL-15/R ⁇ -Fc fusion protein of the “scIL-15/R ⁇ -Fc” format, with additional sequences of XENP21993, XENP21994, XENP21995, XENP23174, XENP23175, XENP24477, and XENP24480 being listed in WO2018071919 in FIGS. 104G, 104H, 104AG, 104AU, and 104AV, respectively and as SEQ ID NOS: 514-518, 519-523, 524-528, 849-853, 1063-1067, and 1078-1082, respectively.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 12A - FIG. 12B depict sequences of XENP21479, XENP22366 and XENP24348, illustrative IL-15/R ⁇ -Fc fusion proteins of the “ncIL-15/R ⁇ -Fc” format.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 13 depicts sequences of XENP21978, an illustrative IL-15/R ⁇ -Fc fusion protein of the “bivalent ncIL-15/R ⁇ -Fc” format, with additional sequences of XENP21979 being listed in WO2018071919 in FIG. 104E and as SEQ ID NOS: 480-483.
  • IL-15 and IL-15R ⁇ (sushi) are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 14 depicts sequences of an illustrative IL-15/R ⁇ -Fc fusion protein of the “bivalent scIL-15/R ⁇ -Fc” format.
  • IL-15 and IL-15R ⁇ (sushi) are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 )
  • slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 15 depicts sequences of XENP22637, an illustrative IL-15/R ⁇ -Fc fusion protein of the “Fc-ncIL-15/R ⁇ ” format, with additional sequences of XENP22638 being listed in WO2018071919 in FIG. 104T and as SEQ ID NOS: 668-672.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 16 depicts sequences of an illustrative IL-15/R ⁇ -Fc fusion protein of the “Fc-scIL-15/R ⁇ ” format.
  • IL-15 and IL-15R ⁇ (sushi) are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 )
  • slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 17A - FIG. 17E provide data for an illustrative IL-15/R ⁇ -Fc fusion protein format for XENP20818.
  • FIG. 17A depicts the IL-15/R ⁇ -Fc fusion protein format for XENP20818.
  • FIG. 17B depicts the purity and homogeneity of XENP20818 as determined by SEC.
  • FIG. 17C depicts the purity and homogeneity of XENP20818 as determined by CEF.
  • FIG. 17D depicts the affinity of XENP20818 for IL-2R ⁇ as determined by Octet.
  • FIG. 17E depicts the stability of XENP20818 as determined by DSF.
  • FIG. 18A - FIG. 18E provide data for an illustrative IL-15/R ⁇ -Fc fusion protein format for XENP21478.
  • FIG. 18A depicts the IL-15/R ⁇ -Fc fusion protein format for XENP21478.
  • FIG. 18B depicts the purity and homogeneity of XENP21478 as determined by SEC.
  • FIG. 18C depicts the purity and homogeneity of XENP21478 as determined by CEF.
  • FIG. 18D depicts the affinity of XENP21478 for IL-2R ⁇ as determined by Octet.
  • FIG. 18E depicts the stability of XENP21478 as determined by DSF.
  • FIG. 19A - FIG. 19E provide data for an illustrative IL-15/R ⁇ -Fc fusion protein format for XENP21479.
  • FIG. 19A depicts the IL-15/R ⁇ -Fc fusion protein format for XENP21479.
  • FIG. 19B depicts the purity and homogeneity of XENP21479 as determined by SEC.
  • FIG. 19C depicts the purity and homogeneity of XENP21479 as determined by CEF.
  • FIG. 19D depicts the affinity of XENP21479 for IL-2R ⁇ as determined by Octet.
  • FIG. 19E depicts the stability of XENP21479 as determined by DSF.
  • FIG. 20A - FIG. 20C depict the induction of NK (CD56 + /CD16 + ) cells ( FIG. 20A ), CD4 + T cells ( FIG. 20B ), and CD8 + T cells ( FIG. 20C ) proliferation by illustrative IL-15/R ⁇ -Fc fusion proteins of the IL-15/R ⁇ -heteroFc format with different linker lengths based on Ki67 expression as measured by FACS.
  • FIG. 21A - FIG. 21C depict the induction of NK (CD56 + /CD16 + ) cells ( FIG. 21A ), CD4 + T cells ( FIG. 21B ), and CD8 + T cells ( FIG. 21C ) proliferation by illustrative IL-15/R ⁇ -Fc fusion proteins of the scIL-15/R ⁇ -Fc format (XENP21478) and the ncIL-15/R ⁇ -Fc format (XENP21479) based on Ki67 expression as measured by FACS.
  • FIG. 22 depicts enhancement of IL-2 secretion by illustrative IL-15/R ⁇ -Fc fusion proteins, an isotype control, and a bivalent anti-PD-1 antibody over PBS control in an SEB-stimulated PBMC assay.
  • FIG. 23 depicts the survival curve for PBMC-engrafted NSG mice following treatment with XENP20818 and recombinant IL-15.
  • FIG. 24 depicts the concentration of IFN ⁇ in serum of NSG mice on Day 7 after engraftment with human PBMCs and treatment with XENP20818 at the indicated concentrations.
  • FIGS. 25A-25C depict CD4 + T cell ( FIG. 25A ), CD8 + T cell ( FIG. 25B ), and CD45 + cell ( FIG. 25C ) counts in whole blood of human PBMC-engrafted NSG mice 7 days after treatment with XENP20818 at the indicated concentrations.
  • FIG. 26 depicts a structural model of the IL-15/R ⁇ heterodimer showing locations of engineered disulfide bond pairs.
  • FIG. 27 depicts sequences for illustrative IL-15R ⁇ (sushi) variants engineered with additional residues at the C-terminus to serve as a scaffold for engineering cysteine residues.
  • FIG. 28 depicts sequences for illustrative IL-15 variants engineered with cysteines in order to form covalent disulfide bonds with IL-15R ⁇ (sushi) variants engineered with cysteines.
  • FIG. 29 depicts sequences for illustrative IL-15R ⁇ (sushi) variants engineered with cysteines in order to form covalent disulfide bonds with IL-15 variants engineered with cysteines.
  • FIG. 30A - FIG. 30C depict IL-15/R ⁇ heterodimers with and without engineered disulfide bonds between IL-15 and IL-15R ⁇ (sushi).
  • Non-covalent IL-15/R ⁇ heterodimer or “ncIL-15/R ⁇ heterodimer” comprises IL-15R ⁇ (sushi) and IL-15 transfected separately and non-covalently linked.
  • Disulfide-bonded IL-15/R ⁇ heterodimer or “dsIL-15/R ⁇ heterodimer” comprises IL-15R ⁇ (sushi) and IL-15 transfected separately and covalently linked as a result of engineered cysteines.
  • Single-chain IL-15/R ⁇ heterodimer or “scIL-15/R ⁇ Heterodimer” comprises IL-15R ⁇ (sushi) fused to IL-15 by a variable length Gly-Ser linker.
  • FIG. 31 depicts sequences of XENP21996, an illustrative ncIL-15/R ⁇ heterodimer. It is important to note that these sequences were generated using polyhistidine (His ⁇ 6 or HHHHHH (SEQ ID NO: 10)) C-terminal tags at the C-terminus of IL-15R ⁇ (sushi).
  • FIG. 32 depicts sequences of XENP22004, XENP22005, XENP22006, XENP22008, and XENP22494, illustrative dsIL-15/R ⁇ heterodimers, with additional sequences of XENP22007, XENP22009, XENP22010, XENP22011, XENP22012, and XENP22493 depicted in WO2018071919 in FIGS. 104J, 104K, and 104I, respectively and as SEQ ID NOS: 543-544, 545-546, 547-548, 551-552, 553-554, AND 647-648, respectively. It is important to note that these sequences were generated using polyhistidine (His ⁇ 6 or HHHHHH (SEQ ID NO: 10)) C-terminal tags at the C-terminus of IL-15R ⁇ (sushi).
  • FIG. 33 depicts the sequence for XENP22049, an illustrative scIL-15/R ⁇ Heterodimer. It is important to note that these sequences were generated using polyhistidine (His ⁇ 6 or HHHHHH (SEQ ID NO: 10)) C-terminal tags at the C-terminus of IL-15. IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , and linker.
  • polyhistidine His ⁇ 6 or HHHHHHHH (SEQ ID NO: 10)
  • FIG. 34 depicts the purity and homogeneity of illustrative IL-15/R ⁇ heterodimers with and without engineered disulfide bonds as determined by CEF.
  • FIG. 35 depicts the purity and homogeneity of illustrative IL-15/R ⁇ heterodimers with and without engineered disulfide bonds as determined by CEF.
  • FIG. 36 depicts the stability and melting temperatures of illustrative IL-15/R ⁇ heterodimers with and without engineered disulfide bonds as indicated by melting curves from DSF.
  • FIG. 37 depicts the stability and melting temperatures of illustrative IL-15/R ⁇ heterodimers with and without engineered disulfide bonds as indicated by melting curves from DSF.
  • FIG. 38 depicts the expression yield, molecular weight, predicted change in affinity between IL-15 and IL-15R ⁇ (sushi) as calculated by MOE software, melting temperature, and affinity for IL-2R ⁇ for IL-15/R ⁇ heterodimers with and without engineered disulfide bonds. Mutations are indicated in parentheses after the relevant monomer.
  • FIG. 39A - FIG. 39D depict additional formats for the IL-15/R ⁇ -Fc fusion proteins of the present invention with engineered disulfide bonds.
  • Disulfide-bonded IL-15/R ⁇ heterodimeric Fc fusion or “dsIL-15/R ⁇ -heteroFc” ( FIG. 39A ) is the same as “IL-15/R ⁇ -heteroFc”, but wherein IL-15R ⁇ (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • Disulfide-bonded IL-15/R ⁇ Fc fusion or “dsIL-15/R ⁇ -Fc” FIG.
  • FIG. 39B is the same as “ncIL-15/R ⁇ -Fc”, but wherein IL-15R ⁇ (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • Bivalent disulfide-bonded IL-15/R ⁇ -Fc or “bivalent dsIL-15/R ⁇ -Fc” is the same as “bivalent ncIL-15/R ⁇ -Fc”, but wherein IL-15R ⁇ (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • Fc-disulfide-bonded IL-15/R ⁇ fusion or “Fc-dsIL-15/R ⁇ ” is the same as “Fc-ncIL-15/R ⁇ ”, but wherein IL-15R ⁇ (sushi) and IL-15 are further covalently linked as a result of engineered cysteines.
  • FIG. 40A - FIG. 40B depict sequences of XENP22013, XENP22014, XENP22015, and XENP22017, illustrative IL-15/R ⁇ -Fc fusion protein of the “dsIL-15/R ⁇ -heteroFc” format.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 41A - FIG. 41B depict sequences of XENP22357, XENP22358, XENP22359, XENP22684, and XENP22361, illustrative IL-15/R ⁇ -Fc fusion proteins of the “dsIL-15/R ⁇ -Fc” format. Additional sequences of XENPs 22360, 22362, 22363, 22364, 22365, 22366 are depicted in WO2018/071919 in FIGS.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 42 depicts sequences of XENP22634, XENP22635, and XENP22636, illustrative IL-15/R ⁇ -Fc fusion proteins of the “bivalent dsIL-15/R ⁇ -Fc” format. Additional sequences of XENP22687 are depicted in WO2018/071919 in FIG. 104V and as SEQ ID NOS:685-688, herein incorporated by reference in its entirety. IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 43 depicts sequences of XENP22639 and XENP22640, illustrative IL-15/R ⁇ -Fc fusion proteins of the “Fc-dsIL-15/R ⁇ ” format.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 44 depicts the purity and homogeneity of illustrative IL-15/R ⁇ -Fc fusion proteins with and without engineered disulfide bonds as determined by CEF.
  • FIG. 45A - FIG. 45C depict the induction of NK (CD56+/CD16+) cell ( FIG. 45A ), CD8 + T cell ( FIG. 45B ), and CD4 + T cell ( FIG. 45C ) proliferation by illustrative IL-15/R ⁇ -Fc fusion proteins with and without engineered disulfide bonds based on Ki67 expression as measured by FACS.
  • FIG. 46 depicts the structure of IL-15 complexed with IL-15R ⁇ , IL-2R ⁇ , and common gamma chain. Locations of substitutions designed to reduce potency are shown.
  • FIG. 47A - FIG. 47C depict sequences for illustrative IL-15 variants engineered for reduced potency. Included within each of these variant IL-15 sequences are sequences that are 90%, 95%, 98% and 99% identical (as defined herein) to the recited sequences, and/or contain from 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 additional amino acid substitutions. In a non-limiting example, the recited sequences may contain additional amino acid modifications such as those contributing to formation of covalent disulfide bonds as described in Example 2.
  • FIG. 48A - FIG. 48H depict sequences of XENP22816, XENP22819, XENP22820, XENP22821, XENP22822, XENP22829, XENP22834, XENP23554, XENP23557, XENP23561, XENP24018, XENP24019, XENP24045, XENP24051, and XENP24052, illustrative IL-15/R ⁇ -Fc fusion proteins of the “IL-15/R ⁇ -heteroFc” format engineered for lower potency.
  • 104Z 104AA, 104AC, 104AD, 104AE, 104AF, 104AJ, 104AK, 104AM, 104AN, and 104AO, and as SEQ ID NOS: 729-734, 741-746, 747-752, 777-782, 783-788, 789-794, 795-800, 801-806, 807-812, 819-824, 825-830, 831-836, 837-842, 887-892, 899-904, 905-910, 937-942, 955-960, 961-966, and 979-984, respectively.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 49A - FIG. 49D depict sequences of XENP24015, XENP24050, XENP24475, XENP24476, XENP24478, XENP24479, and XENP24481, illustrative IL-15/R ⁇ -Fc fusion proteins of the “scIL-15/R ⁇ -Fc” format engineered for lower potency. Additional sequences of XENPs 24013, 24014, 24016 are depicted in WO2018071919 in FIGS. 104AK and 104AL and as SEQ ID NOS: 914-921, 922-926, and 932-936.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 50A - FIG. 50B depict sequences of XENP24349, XENP24890, and XENP25138, illustrative IL-15/R ⁇ -Fc fusion proteins of the “ncIL-15/R ⁇ -Fc” format engineered for lower potency.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 51 depicts sequences of XENP22801 and XENP22802, illustrative ncIL-15/R ⁇ heterodimers engineered for lower potency. Additional sequences of XENPs 22791, 22792, 22793, 22794, 22795, 22796, 22803, 22804, 22805, 22806, 22807, 22808, 22809, 22810, 22811, 22812, 22813, 22814 are depicted in WO2018071919 in FIGS.
  • 104V, 104W, 104X, 104Y, and 104Z and as SEQ ID NOS: 689-690, 691-692, 693-694, 695-696, 697-698, 699-700, 705-706, 707-708, 709-710, 711-712, 713-714, 715-716, 717-718, 719-720, 721-722, 723-724, 725-726, and 727-728, respectively. It is important to note that these sequences were generated using polyhistidine (His ⁇ 6 or HHHHHH (SEQ ID NO: 10)) C-terminal tags at the C-terminus of IL-15R ⁇ (sushi).
  • FIG. 52 depicts sequences of XENP24342, an illustrative IL-15/R ⁇ -Fc fusion protein of the “bivalent ncIL-15/R ⁇ -Fc” format engineered for lower potency.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 53 depicts sequences of XENP23472 and XENP23473, illustrative IL-15/R ⁇ -Fc fusion proteins of the “dsIL-15/R ⁇ -Fc” format engineered for lower potency.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 54A - FIG. 54C depict the induction of NK cell ( FIG. 54A ), CD8 + (CD45RA ⁇ ) T cell ( FIG. 54B ), and CD4 + (CD45RA ⁇ ) T cell ( FIG. 54C ) proliferation by variant IL-15/R ⁇ -Fc fusion proteins based on Ki67 expression as measured by FACS.
  • FIG. 55 depicts EC50 for induction of NK and CD8 + T cells proliferation by variant IL-15/R ⁇ -Fc fusion proteins, and fold reduction in EC50 relative to XENP20818.
  • FIG. 56A - FIG. 56C depict the gating of lymphocytes and subpopulations for the experiments depicted in FIGS. 59A-59D .
  • FIG. 56A shows the gated lymphocyte population.
  • FIG. 56B shows the CD3-negative and CD3-positive subpopulations.
  • FIG. 56C shows the CD16-negative and CD16-positive subpopulations of the CD3-negative cells.
  • FIG. 57A - FIG. 57C depict the gating of CD3 + lymphocyte subpopulations for the experiments depicted in FIGS. 59A-59D .
  • FIG. 57A shows the CD4 + , CD8 + and ⁇ T cell subpopulations of the CD3 + T cells.
  • FIG. 57B shows the CD45RA( ⁇ ) and CD45RA(+) subpopulations of the CD4 + T cells.
  • FIG. 57C shows the CD45RA( ⁇ ) and CD45RA(+) subpopulations of the CD8 + T cells.
  • FIG. 58A - FIG. 58B depict CD69 and CD25 expression before ( FIG. 58A ) and after ( FIG. 58B ) incubation of human PBMCs with XENP22821.
  • FIG. 59A - FIG. 59D depict cell proliferation in human PBMCs incubated for four days with the indicated variant IL-15/R ⁇ -Fc fusion proteins.
  • FIGS. 59A-C show the percentage of proliferating NK cells (CD3-CD16+) ( FIG. 59A ), CD8+ T cells (CD3+CD8+CD45RA ⁇ ) ( FIG. 59B ) and CD4+ T cells (CD3+CD4+CD45RA ⁇ ) ( FIG. 59C ).
  • FIG. 59D shows the fold change in EC50 of various IL-15/IL-15R ⁇ Fc heterodimers relative to control (XENP20818).
  • FIG. 60A - FIG. 60D depict cell proliferation in human PBMCs incubated for three days with the indicated variant IL-15/R ⁇ -Fc fusion proteins.
  • FIGS. 60A-C show the percentage of proliferating CD8 + (CD45RA ⁇ ) T cells ( FIG. 60A ), CD4 + (CD45RA ⁇ ) T cells ( FIG. 60B ), ⁇ T cells ( FIG. 60C ), and NK cells ( FIG. 60D ).
  • FIG. 61A - FIG. 61C depict the percentage of Ki67 expression on CD8+ T cells ( FIG. 61A ), CD4+ T cells ( FIG. 61B ), and NK cells ( FIG. 61C ) following treatment with additional IL-15/R ⁇ variants.
  • FIG. 62A - FIG. 62E depict the percentage of Ki67 expression on ( FIG. 62A ) CD8+(CD45RA ⁇ ) T cells, ( FIG. 62B ) CD4+(CD45RA ⁇ ) T cells, ( FIG. 62C ) ⁇ T cells, ( FIG. 62D ) NK (CD16+CD8 ⁇ ) cells, and ( FIG. 62E ) NK (CD56+CD8 ⁇ ) cells following treatment with IL-15/R ⁇ variants.
  • FIG. 63A - FIG. 63E depict the percentage of Ki67 expression on CD8 + (CD45RA ⁇ ) T cells ( FIG. 63A ), CD4 + (CD45RA ⁇ ) T cells ( FIG. 63B ), ⁇ T cells ( FIG. 63C ), NK (CD16+CD8 ⁇ ) cells ( FIG. 63D ), and NK (CD56+CD8 ⁇ ) cells ( FIG. 63E ) following treatment with IL-15/R ⁇ variants.
  • FIG. 64A - FIG. 64D depict the percentage of Ki67 expression on ( FIG. 64A ) CD8+ T cells, ( FIG. 64B ) CD4+ T cells, ( FIG. 64C ) ⁇ T cells and ( FIG. 64D ) NK (CD16+) cells following treatment with additional IL-15/R ⁇ variants engineered for decreased potency with different linker lengths.
  • FIG. 65A - FIG. 65D depict the percentage of Ki67 expression on ( FIG. 65A ) CD8 + T cells, ( FIG. 65B ) CD4 + T cells, ( FIG. 65C ) ⁇ T cells and ( FIG. 65D ) NK (CD16+) cells following treatment with additional IL-15/R ⁇ variants.
  • FIG. 66A - FIG. 66D depict gating of lymphocytes and subpopulations thereof for the experiments depicted in FIGS. 67A-67C .
  • FIG. 66A shows gating of the lymphocyte population.
  • FIG. 66B shows CD4+ and CD8+ T cells.
  • FIG. 66C shows the CD45RA and CD27 expressing subpopulations of CD4+ T cells.
  • FIG. 66D shows the CD45RA and CD27 expressing subpopulations of CD8+ T cells.
  • FIG. 67A - FIG. 67C depict STAT5 phosphorylation on CD8 + T cells (CD45RA-CD27 ⁇ ) ( FIG. 67A ) and CD4 + T cells (CD45RA ⁇ CD27 ⁇ ) ( FIG. 67B ) following incubation of PBMCs for 4 days with the indicated variant IL-15/IL-15R ⁇ -Fc fusion proteins at the indicated concentrations.
  • FIG. 67C shows the fold change in EC50 of various IL-15/IL-15R ⁇ Fc heterodimers relative to control (XENP20818).
  • FIG. 68A - FIG. 68B depict STAT5 phosphorylation on CD8 + CD45RA ⁇ T cells ( FIG. 68A ) and CD4 + CD45RA ⁇ T cells ( FIG. 68B ) in mouse splenocytes following incubation with the indicated test articles.
  • FIG. 69 depicts IV-TV Dose PK of various IL-15/R ⁇ -Fc fusion proteins or controls in C57BL/6 mice at 0.1 mg/kg single dose.
  • FIG. 70 depicts the correlation of half-life vs NK cell potency.
  • FIG. 71 depicts the correlation of half-life vs mouse STAT5 signaling for IL-15/R ⁇ -Fc affinity variants.
  • FIG. 72 depicts the serum concentration of test articles 8 days after dosing of C57BL/6 albino mice.
  • FIG. 73 depicts the CD8 + T cell count in the spleen of C57BL/6 albino mice 8 days after dosing with the indicated test articles.
  • FIG. 74 shows that CD45 + cell levels are predictive of disease.
  • FIG. 75A - FIG. 75B depict the enhancement of engraftment by variant IL-15/R ⁇ -Fc fusion proteins as indicated by CD45 + cell counts on Day 4 ( FIG. 75A ) and Day 8 ( FIG. 75B ).
  • FIG. 76A - FIG. 76C depict IFN ⁇ levels on Day 4 ( FIG. 76A ), Day 7 ( FIG. 76B ), and Day 11 ( FIG. 76C ) after treatment of NSG mice engrafted with human PBMCs with the indicated variant IL-15/R ⁇ -Fc fusion proteins or control.
  • FIG. 77A - FIG. 77C depict CD45 + lymphocyte cell counts on Day 4 ( FIG. 77A ), Day 7 ( FIG. 77B ), and Day 11 ( FIG. 77C ) after treatment of NSG mice engrafted with human PBMCs with the indicated variant IL-15/R ⁇ -Fc fusion proteins or control.
  • FIG. 78A - FIG. 78C depict NK cell (CD16+CD56+CD45RA+) counts on Day 4 ( FIG. 78A ), Day 7 ( FIG. 78B ) and Day 11 ( FIG. 78C ) after treatment of NSG mice engrafted with human PBMCs with the indicated IL-15/R ⁇ -Fc fusion proteins or control.
  • FIG. 79A - FIG. 79B depict CD8 + T cell (CD8+CD45RA+) counts on Day 7 ( FIG. 79A ) and Day 11 ( FIG. 79B ) after treatment of NSG mice engrafted with human PBMCs with the indicated IL-15/R ⁇ -Fc fusion proteins or control.
  • FIG. 80A - FIG. 80B depict CD4+ T cell (CD4+CD45RA+) counts on Day 7 ( FIG. 80A ) and Day 11 ( FIG. 80B ) after treatment of NSG mice engrafted with human PBMCs with the indicated IL-15/R ⁇ -Fc fusion proteins or control.
  • FIG. 81 depicts IFN ⁇ level on Days 4, 7, and 11 in serum of huPBMC engrafted mice following treatment with additional variant IL-15/R ⁇ -Fc fusion proteins.
  • FIG. 82A - FIG. 82C depict CD8+ T cell count on Day 4 ( FIG. 82A ), Day 7 ( FIG. 82B ), and Day 11 ( FIG. 82C ) in whole blood of huPBMC engrafted mice following treatment with additional variant IL-15/R ⁇ -Fc fusion proteins.
  • FIG. 83A - FIG. 83C depict CD4+ T cell count on Day 4 ( FIG. 83A ), Day 7 ( FIG. 83B ), and Day 11 ( FIG. 83C ) in whole blood of huPBMC engrafted mice following treatment with additional variant IL-15/R ⁇ -Fc fusion proteins.
  • FIG. 84A - FIG. 84C depict CD45+ cell count on Day 4 ( FIG. 84A ), Day 7 ( FIG. 84B ), and Day 11 ( FIG. 84C ) in whole blood of huPBMC engrafted mice following treatment with additional variant IL-15/R ⁇ -Fc fusion proteins.
  • FIG. 85A - FIG. 85C depict the body weight as a percentage of initial body weight of huPBMC engrafted mice on Day 4 ( FIG. 85A ), Day 7 ( FIG. 85B ), and Day 11 ( FIG. 85C ) following treatment with additional IL-15/R ⁇ variants. Each point represents a single NSG mouse. Mice whose body weights dropped below 70% initial body weight were euthanized. Dead mice are represented as 70%.
  • FIG. 86A - FIG. 86B depict percentage cyno CD8 + T cell ( FIG. 86A ) and cyno NK cell ( FIG. 86B ) expressing Ki67 following incubation with the indicated test articles.
  • FIG. 87A - FIG. 87E depict lymphocyte counts after dosing cynomolgus monkeys with XENP20818.
  • FIGS. 87A-E respectively show the fold change in absolute count of CD56+NK cells ( FIG. 87A ), CD16+NK cells ( FIG. 87B ), ⁇ T cells (CD45RA+CD3+CD4-CD8-) ( FIG. 87C ), CD8+ T cells ( FIG. 87D ), and CD4+ T cells ( FIG. 87E ).
  • FIG. 88A - FIG. 88E depict proliferation of CD56+NK cells ( FIG. 88A ), CD16+NK cells ( FIG. 88B ), CD8+ T cells (CD45RA+) ( FIG. 88C ), CD8+ T cells (CD45RA ⁇ ) ( FIG. 88D ), and CD4+ T cells (CD45RA ⁇ ) ( FIG. 88E ) after dosing cynomolgus monkeys with XENP20818.
  • FIG. 89A - FIG. 89E depict lymphocyte counts after dosing cynomolgus monkeys with XENP22819.
  • FIGS. 89A-89E show the fold change in absolute count of CD56+NK cells ( FIG. 89A ), CD16+NK cells ( FIG. 89B ), ⁇ T cells (CD45RA+CD3+CD4 ⁇ CD8 ⁇ ) ( FIG. 89C ), CD8+ T cells ( FIG. 89D ), and CD4+ T cells ( FIG. 89E ).
  • FIG. 90A - FIG. 90E depict proliferation of CD56+NK cells ( FIG. 90A ), CD16+NK cells ( FIG. 90B ), CD8+ T cells (CD45RA+) ( FIG. 90C ), CD8+ T cells (CD45RA ⁇ ) ( FIG. 90D ), and CD4+ T cells (CD45RA ⁇ ) ( FIG. 90E ) after dosing cynomolgus monkeys with XENP22819.
  • FIG. 91A - FIG. 91E depict lymphocyte counts after dosing cynomolgus monkeys with XENP22821.
  • FIGS. 91A-91E show the fold change in absolute count of CD56+NK cells ( FIG. 91A ), CD16+NK cells ( FIG. 91B ), ⁇ T cells (CD45RA+CD3+CD4 ⁇ CD8 ⁇ ) ( FIG. 91C ), CD8+ T cells ( FIG. 91D ), and CD4+ T cells ( FIG. 91E ).
  • FIG. 92A - FIG. 92E depict proliferation of CD56+NK cells ( FIG. 92A ), CD16+NK cells ( FIG. 92B ), CD8+ T cells (CD45RA+) ( FIG. 92C ), CD8+ T cells (CD45RA ⁇ ) ( FIG. 92D ), and CD4+ T cells (CD45RA ⁇ ) ( FIG. 92E ) after dosing cynomolgus monkeys with XENP22821.
  • FIG. 93A - FIG. 93E depict lymphocyte counts after dosing cynomolgus monkeys with XENP22822.
  • FIGS. 93A-E respectively show the fold change in absolute count of CD56+NK cells ( FIG. 93A ), CD16+NK cells ( FIG. 93B ), ⁇ T cells (CD45RA+CD3+CD4 ⁇ CD8 ⁇ ) ( FIG. 93C ), CD8+ T cells ( FIG. 93D ), and CD4+ T cells ( FIG. 93E ).
  • FIG. 94A - FIG. 94E depict proliferation of CD56+NK cells ( FIG. 94A ), CD16+NK cells ( FIG. 94B ), CD8 + T cells (CD45RA+) ( FIG. 94C ), CD8 + T cells (CD45RA ⁇ ) ( FIG. 94D ), and CD4 + T cells (CD45RA ⁇ ) ( FIG. 94E ) after dosing cynomolgus monkeys with XENP22822.
  • FIG. 95A - FIG. 95E depict lymphocyte counts after dosing cynomolgus monkeys with XENP22834.
  • FIGS. 95A-E respectively show the fold change in absolute count of CD56+NK cells ( FIG. 95A ), CD16+NK cells ( FIG. 95B ), ⁇ T cells (CD45RA+CD3+CD4 ⁇ CD8 ⁇ ) ( FIG. 95C ), CD8+ T cells ( FIG. 95D ), and CD4+ T cells ( FIG. 95E ).
  • FIG. 96A - FIG. 96E depict proliferation of CD56+NK cells ( FIG. 96A ), CD16+NK cells ( FIG. 96B ), CD8+ T cells (CD45RA+) ( FIG. 96C ), CD8+ T cells (CD45RA ⁇ ) ( FIG. 96D ), and CD4+ T cells (CD45RA ⁇ ) ( FIG. 96E ) after dosing cynomolgus monkeys with XENP22834.
  • FIG. 97A - FIG. 97E depict lymphocyte counts after dosing cynomolgus monkeys with XENP23343.
  • FIGS. 97A-E respectively show the fold change in absolute count of CD56 + NK cells ( FIG. 97A ), CD16 + NK cells ( FIG. 97B ), ⁇ T cells (CD45RA+CD3+CD4 ⁇ CD8 ⁇ ) ( FIG. 97C ), CD8 + T cells ( FIG. 97D ), and CD4 + T cells ( FIG. 97E ).
  • FIG. 98A - FIG. 98E depict proliferation of CD56+NK cells ( FIG. 98A ), CD16+NK cells ( FIG. 98B ), CD8+ T cells (CD45RA+) ( FIG. 98C ), CD8+ T cells (CD45RA ⁇ ) ( FIG. 98D ), and CD4+ T cells (CD45RA ⁇ ) ( FIG. 98E ) after dosing cynomolgus monkeys with XENP23343.
  • FIG. 99A - FIG. 99C depict sequences of XENP23343, XENP23504, XENP24113, XENP24301, XENP24306, and XENP24341, illustrative IL-15/R ⁇ -Fc fusion proteins of the “IL-15/R ⁇ -heteroFc” format with M428L/N434S substitutions.
  • IL-15 and IL-15R ⁇ (sushi) are underlined
  • linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 )
  • slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 100 depicts sequences of XENP25938 (also referred to as XENP24294), an illustrative IL-15/R ⁇ -Fc fusion protein of the “scIL-15/R ⁇ -Fc” format with M428L/N434S substitutions.
  • FIG. 101 depicts sequences of XENP24383, an illustrative IL-15/R ⁇ -Fc fusion protein of the “ncIL-15/R ⁇ -Fc” format with M428L/N434S substitutions.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 102 depicts sequences of XENP24346 and XENP24351, illustrative IL-15/R ⁇ -Fc fusion proteins of the “bivalent ncIL-15/R ⁇ -Fc” format with M428L/N434S substitutions.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in FIG. 7 ), and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and Fc regions.
  • FIG. 103A - FIG. 103C depict the percentage of Ki67 expression on human CD8 + T cells ( FIG. 103A ), human CD4 + T cells ( FIG. 103B ) and human NK cells ( FIG. 103C ) following treatment with IL-15/R ⁇ variants with M428L/N434S Fc mutations.
  • FIG. 104A - FIG. 104D depict the percentage of Ki67 expression on human CD8 + T cells ( FIG. 104A ), human CD4 + T cells ( FIG. 104B ), human NK cells ( FIG. 104C ), and human ⁇ T cells ( FIG. 104D ) following treatment with XmAb24306 (also referred to as XENP24306).
  • FIG. 105A - FIG. 105C depict the percentage of Ki67 expression on cyno CD8 + T cells ( FIG. 105A ), cyno CD4 + T cells ( FIG. 105B ) and cyno NK cells ( FIG. 105C ) following treatment with WT IL-15/R ⁇ -Fc and potency-reduced IL-15/R ⁇ variants with M428L/N434S Fc mutations.
  • FIG. 106 depicts the percentage of Ki67 expression on cyno CD8 ⁇ + CD45RA ⁇ T cells following treatment with XENP20818 or XmAb24306.
  • FIG. 107A - FIG. 107C depicts CD4+ T cell count on Day 4 ( FIG. 107A ) and Day 7 ( FIG. 107B ) in whole blood and Day 8 ( FIG. 107C ) in spleen of huPBMC engrafted mice following treatment with additional variant IL-15/R ⁇ -Fc fusion proteins.
  • FIG. 108A - FIG. 108C depict CD8 + T cell count on ( FIG. 108A ) Day 4 and ( FIG. 108B ) Day 7 in whole blood and ( FIG. 108C ) Day 8 in spleen of huPBMC engrafted mice following treatment with additional variant IL-15/R ⁇ -Fc fusion proteins.
  • FIG. 109A - FIG. 109C depict CD8+ T cell count on Day 4 ( FIG. 109A ) and Day 7 ( FIG. 109B ) in whole blood and Day 8 ( FIG. 109C ) in spleen of huPBMC engrafted mice following treatment with additional variant IL-15/R ⁇ -Fc fusion proteins.
  • FIG. 110A - FIG. 110F depicts the body weight as a percentage of initial body weight of huPBMC engrafted mice on Day ⁇ 2 ( FIG. 110A ), Day 1 ( FIG. 110B ), Day 5 ( FIG. 110C ), Day 8 ( FIG. 110D ) and Day 11 ( FIG. 110E ) following treatment with additional IL-15/R ⁇ variants. Each point represents a single NSG mouse.
  • FIG. 110F depicts a time-course of body weight in huPBMC engrafted mice following treatment with the IL-15/R ⁇ variants.
  • FIG. 111A - FIG. 111D depict the percentage of CD8+CD45RA ⁇ T cell ( FIG. 111A ), CD4+CD45RA ⁇ T cell ( FIG. 111B ), ⁇ T cell ( FIG. 111C ), and CD16+NK cell ( FIG. 111D ) expressing Ki67 following incubation with the indicated test articles.
  • FIG. 112A - FIG. 112D depict CD8+ T cell ( FIG. 112A ), CD4+ T cell ( FIG. 112B ), NK cell ( FIG. 112C ) and ⁇ T cell ( FIG. 112D ) counts in cynomolgus monkeys after treatment with IL-15/R ⁇ variants.
  • FIG. 113A-113H depicts the serum concentrations over time and half-lives of the indicated test articles in cynomolgus monkeys.
  • FIG. 114A - FIG. 114F depict the binding of AF647-labeled test articles (WT IL-15/R ⁇ -Fc and IL-15/R ⁇ -Fc affinity variants with Xtend and without domain linkers) to CD8+CD45RA ⁇ T cells ( FIG. 114A ), CD8+CD45RA+ T cells ( FIG. 114B ), CD4+CD45RA-T cells ( FIG. 114C ), CD4+CD45RA+ T cells ( FIG. 114D ), CD16+NK cells ( FIG. 114E ), and ⁇ T cells ( FIG. 114F ) in fresh and activated PBMC.
  • AF647-labeled test articles WT IL-15/R ⁇ -Fc and IL-15/R ⁇ -Fc affinity variants with Xtend and without domain linkers
  • FIG. 115A - FIG. 115F depict the binding of AF647-labeled test articles (WT IL-15/R ⁇ -Fc and IL-15/R ⁇ -Fc affinity variants with Xtend and with domain linkers) to ( FIG. 115A ) CD8 + CD45RA ⁇ T cells, ( FIG. 115B ) CD8 + CD45RA + T cells, ( FIG. 115C ) CD4 + CD45RA ⁇ T cells, ( FIG. 115D ) CD4 + CD45RA + T cells, ( FIG. 115E ) CD16 + NK cells, and ( FIG. 115F ) ⁇ T cells in fresh and activated PBMC.
  • AF647-labeled test articles WT IL-15/R ⁇ -Fc and IL-15/R ⁇ -Fc affinity variants with Xtend and with domain linkers
  • FIG. 116A - FIG. 116F depicts the binding of AF647-labeled test articles (WT IL-15/R ⁇ -Fc and IL-15/R ⁇ -Fc fusion proteins in additional formats with Xtend variants such as FcRn variants) to ( FIG. 116A ) CD8 + CD45RA ⁇ T cells, ( FIG. 116B ) CD8 + CD45RA + T cells, ( FIG. 116C ) CD4 + CD45RA ⁇ T cells, ( FIG. 116D ) CD4 + CD45RA + T cells, ( FIG. 116E ) CD16 + NK cells, and ( FIG. 116F ) ⁇ T cells in fresh and activated PBMC.
  • AF647-labeled test articles WT IL-15/R ⁇ -Fc and IL-15/R ⁇ -Fc fusion proteins in additional formats with Xtend variants such as FcRn variants
  • FIG. 117 depicts the binding of AF647-labeled test articles (including XENP24341 and XENP24113) to CD8 + CD45RA ⁇ T cells in activated PBMC.
  • FIG. 118A - FIG. 118B depicts CD25 expression on CD8 + T cells in ( FIG. 118A ) Group 1 (purified T cells incubated with parental MCF-7 tumor cells and indicated test articles) and ( FIG. 118B ) Group 2 (purified T cells incubated with pp65-expressing MCF-7 tumor cells and indicated test articles).
  • FIG. 119A - FIG. 119B depicts CD25 expression on CD4 + T cells in ( FIG. 119A ) Group 1 (purified T cells incubated with parental MCF-7 tumor cells and indicated test articles) and ( FIG. 119B ) Group 2 (purified T cells incubated with pp65-expressing MCF-7 tumor cells and indicated test articles).
  • FIG. 120A - FIG. 120B depict CD69 expression on CD8 + T cells in ( FIG. 120A ) Group 1 (purified T cells incubated with parental MCF-7 tumor cells and indicated test articles) and ( FIG. 120B ) Group 2 (purified T cells incubated with pp65-expressing MCF-7 tumor cells and indicated test articles).
  • FIG. 121A - FIG. 121B depict CD69 expression on CD4 + T cells in ( FIG. 121A ) Group 1 (purified T cells incubated with parental MCF-7 tumor cells and indicated test articles) and ( FIG. 121B ) Group 2 (purified T cells incubated with pp65-expressing MCF-7 tumor cells and indicated test articles).
  • FIG. 122A - FIG. 122B depict intracellular IFN ⁇ expression in CD8 + T cells in ( FIG. 122A ) Group 1 (purified T cells incubated with parental MCF-7 tumor cells and indicated test articles) and ( FIG. 122B ) Group 2 (purified T cells incubated with pp65-expressing MCF-7 tumor cells and indicated test articles).
  • FIG. 123A - FIG. 123B depict intracellular IFN ⁇ expression in CD4 + T cells in ( FIG. 123A ) Group 1 (purified T cells incubated with parental MCF-7 tumor cells and indicated test articles) and ( FIG. 123B ) Group 2 (purified T cells incubated with pp65-expressing MCF-7 tumor cells and indicated test articles).
  • FIG. 124A - FIG. 124C depict percentage of ( FIG. 124A ) Ki-67 + /IFN ⁇ ⁇ , ( FIG. 124B ) Ki-67 + /IFN ⁇ + , and ( FIG. 124C ) Ki-67 ⁇ /IFN ⁇ + fractions of CD8 + T cells in Group 1 (purified T cells incubated with parental MCF-7 tumor cells and indicated test articles).
  • FIG. 125A - FIG. 125C depicts percentage of ( FIG. 125A ) Ki-67 + /IFN ⁇ ⁇ , ( FIG. 125B ) Ki-67 + /IFN ⁇ + , and ( FIG. 125C ) Ki-67 ⁇ /IFN ⁇ + fractions of CD4 + T cells in Group 1 (purified T cells incubated with parental MCF-7 tumor cells and indicated test articles).
  • FIG. 126A - FIG. 126C depict percentage of ( FIG. 126A ) Ki-67 + /IFN ⁇ ⁇ , ( FIG. 126B ) Ki-67 + /IFN ⁇ + , and ( FIG. 126C ) Ki-67 ⁇ /IFN ⁇ + fractions of CD8 + T cells in Group 1 (purified T cells incubated with pp65-expressing MCF-7 tumor cells and indicated test articles).
  • FIG. 127A - FIG. 127C depict percentage of ( FIG. 127A ) Ki-67 + /IFN ⁇ ⁇ , ( FIG. 127B ) Ki-67 + /IFN ⁇ + , and ( FIG. 127C ) Ki-67 ⁇ /IFN ⁇ + fractions of CD4 + T cells in Group 1 (purified T cells incubated with pp65-expressing MCF-7 tumor cells and indicated test articles).
  • FIG. 128A - FIG. 128B depict CD107a expression on CD8 + T cells in ( FIG. 128A ) Group 1 (purified T cells incubated with parental MCF-7 tumor cells and indicated test articles) and ( FIG. 128B ) Group 2 (purified T cells incubated with pp65-expressing MCF-7 tumor cells and indicated test articles).
  • FIG. 129A - FIG. 129B depict CD107a expression on CD4 + T cells in ( FIG. 129A ) Group 1 (purified T cells incubated with parental MCF-7 tumor cells and indicated test articles) and ( FIG. 129B ) Group 2 (purified T cells incubated with pp65-expressing MCF-7 tumor cells and indicated test articles).
  • FIG. 130A - FIG. 130B depict remaining target cells [ FIG. 130A : parental MCF-7 tumor cells; FIG. 130B : pp65-expressing MCF-7 tumor cells] following incubation with purified T cells and indicated test articles.
  • FIG. 131A - FIG. 131B depict number of dead cells [ FIG. 131A : parental MCF-7 tumor cells; FIG. 131B : pp65-expressing MCF-7 tumor cells] following incubation with purified T cells and indicated test articles.
  • FIG. 132 depicts percentage of Ki-67 ⁇ /IFN ⁇ + fractions of CD8 + T cells following incubation of purified T cells with parental MCF-7 tumor cells or pp65-expressing MCF-7 tumor cells, with or without anti-HLA-A antibodies.
  • FIG. 133 depicts number of target cells (i.e. parental MCF-7 tumor cells or pp65-expressing MCF-7 tumor cells) following incubation of purified T cells with parental MCF-7 tumor cells or pp65-expressing MCF-7 tumor cells, with or without anti-HLA-A antibodies.
  • target cells i.e. parental MCF-7 tumor cells or pp65-expressing MCF-7 tumor cells
  • FIG. 134A - FIG. 134B depict CD25 expression on ( FIG. 134A ) CD8 + T cells and ( FIG. 134B ) CD4 + T cells following incubation of purified T cells with pp65-expressing MCF-7 tumor cells and indicated test articles.
  • FIG. 135A - FIG. 135B depict CD69 expression on ( FIG. 135A ) CD8 + T cells and ( FIG. 135B ) CD4 + T cells following incubation of purified T cells with pp65-expressing MCF-7 tumor cells and indicated test articles.
  • FIG. 136A - FIG. 136B depict Ki67 expression on ( FIG. 136A ) CD8 + T cells and ( FIG. 136B ) CD4 + T cells following incubation of purified T cells with pp65-expressing MCF-7 tumor cells and indicated test articles.
  • FIG. 137A - FIG. 137B depict percentage of Ki-67 + /IFN ⁇ + fractions of ( FIG. 137A ) CD8 + T cells and ( FIG. 137B ) CD4 + T cells following incubation of purified T cells with pp65-expressing MCF-7 tumor cells and indicated test articles.
  • FIG. 138A - FIG. 138B depict percentage of CD69 + /IFN ⁇ + fractions of ( FIG. 138A ) CD8 + T cells and ( FIG. 138B ) CD4 + T cells following incubation of purified T cells with pp65-expressing MCF-7 tumor cells and indicated test articles.
  • FIG. 139A - FIG. 139B depict percentage of CD69+/Ki67+ fractions of ( FIG. 139A ) CD8+ T cells and ( FIG. 139B ) CD4+ T cells following incubation of purified T cells with pp65-expressing MCF-7 tumor cells and indicated test articles.
  • FIG. 140 depicts remaining target cells (pp65-expressing MCF-7 tumor cells) following incubation with purified T cells and indicated test articles.
  • FIG. 141 depicts number of dead cells (pp65-expressing MCF-7 tumor cells) following incubation with purified T cells and indicated test articles.
  • FIG. 142A - FIG. 142B depict mean tumor volume ( FIG. 142A ) and change in tumor volume ( FIG. 142B ) in mice engrafted with pp65-expressing MCF-7 tumor cells and pp65-reactive human PBMCs following treatment with XENP24045, the non-Xtend analog of XmAb24306.
  • FIG. 143A - FIG. 143D depict CD45 + cell ( FIG. 143A ), CD4 + cell ( FIG. 143B ), CD8 + cell ( FIG. 143C ), and NK cell ( FIG. 143D ) counts in whole blood of mice engrafted with pp65-expressing MCF-7 tumor cells and pp65-reactive human PBMCs following treatment with XENP24045, the non-Xtend analog of XmAb24306.
  • FIG. 144 depicts the serum concentrations over time and half-lives of various test articles at various concentrations in cynomolgus monkeys.
  • FIG. 145 depicts the Cmax normalized serum concentrations over time of XENP22821 at 1 ⁇ and 3 ⁇ dose in cynomolgus monkeys.
  • FIG. 146A - FIG. 146E depict the mean fold-change in CD8 + T cell ( FIG. 146A ), CD4 + T cell ( FIG. 146B ), CD16 + NK cell ( FIG. 146C ), CD56 + NK cell ( FIG. 146D ), and ⁇ T cell ( FIG. 146E ) counts in cynomolgus monkeys following dosing with the indicated test articles.
  • FIG. 147A - FIG. 147B depict fold change in CD8 + T cells ( FIG. 147A ) and ⁇ T cells ( FIG. 147B ) in cyno whole blood over time following dosing with either 3 ⁇ dose or 0.6 ⁇ dose of XENP24306.
  • FIG. 148 depicts the percentage of CD8 ⁇ + CD45RA + T cells in cyno lymph nodes expressing Ki67 following dosing with 0.3 ⁇ dose XENP22821 and 0.6 ⁇ dose XENP24306.
  • FIG. 149 depicts the percentage of CD8 ⁇ + CD45RA + T cells in cyno whole blood expressing Ki67 (left axis) and the fold change in cell counts (right axis) following dosing with 0.3 ⁇ dose XENP22821 and 0.6 ⁇ dose XENP24306.
  • FIG. 150 depicts the percentage of various lymphocyte populations in cyno PBMC expressing Ki67 after dosing with 0.3 ⁇ dose XENP22821.
  • FIG. 151 depicts the percentage of various lymphocyte populations in cyno PBMC expressing Ki67 after dosing with 0.6 ⁇ dose XmAb24306.
  • FIG. 152 depicts an overlay of the percentage of CD8 ⁇ +CD45RA+ T cells in cyno whole blood expressing Ki67 and the serum concentration of XmAb24306 over time following dosing with 0.6 ⁇ dose XmAb24306.
  • FIG. 153 depicts fold-change in counts of CD8 + T cell, CD4 + T cell, CD56 + CD8 ⁇ + NK cell and ⁇ T cell in cyno whole blood over time following dosing with 0.6 ⁇ dose XmAb24306.
  • FIG. 154 depicts fold-change in counts of CD8 + T cell and Treg in cyno whole blood over time following dosing with 0.6 ⁇ dose XmAb24306.
  • FIG. 155A - FIG. 155C depict overlays of CD8 + T cell ( FIG. 155A ), CD4 + T cell ( FIG. 155B ), and CD16 + NK cell ( FIG. 155C ) counts over time in whole blood of cynomolgus monkeys following dosing with either XENP20818 or XmAb24306.
  • FIG. 156A - FIG. 156C depict the fold change in immune-related gene expression in ( FIG. 156A ) whole PBMC, ( FIG. 156B ) purified NK cells, and ( FIG. 156C ) purified CD8 + T cells following 24 hour treatment with either XENP20818 or XmAb24306 at their EC 50 concentrations as compared to untreated.
  • FIG. 157A - FIG. 157C depict the fold change in gene expression in ( FIG. 157A ) whole PBMC, ( FIG. 157B ) purified NK cells, and ( FIG. 157C ) purified CD8 + T cells following 48 hour treatment with either XENP20818 or XmAb24306 at their EC 50 concentrations as compared to untreated.
  • FIG. 158A - FIG. 159B depict the fold change in gene expression in whole PBMC following 48 hour treatment with ( FIG. 158A ) XmAb24306 or IL-15 and ( FIG. 158B ) XmAb24306 or IL-2 at their EC50 concentrations as compared to untreated.
  • FIG. 159A - FIG. 159B depict the proliferation of ( FIG. 159A ) CD8+ and ( FIG. 159B ) CD4+ responder T cells in the presence of XmAb24306 and various concentrations of rapamycin expanded Treg.
  • FIG. 160 depicts the sequences for XENP16432, a bivalent anti-PD-1 mAb with an ablation variant (E233P/L234V/L235A/G236del/S267K, “IgG1_PVA_/S267k”).
  • the CDRs are underlined. As noted herein and is true for every sequence herein containing CDRs, the exact identification of the CDR locations may be slightly different depending on the numbering used as is shown in Table 1, and thus included herein are not only the CDRs that are underlined but also CDRs included within the VH and VL domains using other numbering systems. Additionally, each CDR has its own SEQ ID NO: or sequence identifier in the sequence listing, and each VH and VL domain has its own SEQ ID NO: or sequence identifier in the sequence listing.
  • FIG. 161A - FIG. 161B depict CD8 + T cells in whole blood of mice on ( FIG. 161A ) Day 6 and ( FIG. 161B ) Day 10 after first dose of the indicated test articles.
  • FIG. 162A - FIG. 162B depict CD4 + T cells in whole blood of mice on ( FIG. 162A ) Day 6 and ( FIG. 162B ) Day 10 after first dose of the indicated test articles.
  • FIG. 163A - FIG. 163B depict CD45 + T cells in whole blood of mice on ( FIG. 163A ) Day 6 and ( FIG. 163B ) Day 10 after first dose of the indicated test articles.
  • FIG. 164A - FIG. 164B depict NK cells in whole blood of mice on ( FIG. 164A ) Day 6 and ( FIG. 164B ) Day 10 after first dose of the indicated test articles.
  • FIG. 165 depicts IFN ⁇ in serum of NSG mice on Day 7 after first dose of the indicated test articles.
  • FIG. 166A - FIG. 166D depict body weight of mice on Day 7 ( FIG. 166A ), Day 11 ( FIG. 166B ), Day 14 ( FIG. 166C ), and Day 18 ( FIG. 166D ) after first dose of the indicated test articles.
  • FIG. 167 depicts percentage of indicated lymphocyte populations expressing Ki67 following incubation with XENP20818.
  • FIG. 168 depicts STAT5 phosphorylation on CD8 + CD45RA + T cells following incubation with XENP20818, XENP22821, XENP24050, and XENP24306.
  • FIG. 169 depicts STAT5 phosphorylation on CD8 + CD45RA + T cells following incubation with XENP20818, XENP22819, XENP22821, and XENP22834.
  • FIG. 170 depicts percentage CD8 + CD45RA ⁇ T cells expressing Ki67 over time following incubation with XENP20818 or XENP24306 at their respective EC50.
  • FIG. 171 depicts BCL2 expression on CD8 + CD45RA ⁇ T cells over time following incubation with XENP20818 or XENP24306 at their respective EC50.
  • FIG. 172 depicts CD25 expression on CD8 + CD45RA ⁇ T cells over time following incubation with XENP20818 or XENP24306 at their respective EC50.
  • FIG. 173 depicts STAT5 phosphorylation on CD8 + T cells in fresh versus stimulated human PBMCs following incubation with XENP24045 or XENP20818.
  • FIG. 174 depicts percent proliferating CD8 + CD45RA ⁇ (as determined by CFSE dilution) following incubation of human PBMCs with indicated dose of XENP24306 and indicated dose of plate bound anti-CD3 (OKT3).
  • FIG. 175 depicts serum IFN ⁇ concentration in huPBMC-engrafted NSG mice on Day 10 after the first dosing with the indicated test articles at the indicated concentrations.
  • FIG. 176 depicts CD45 + cell count in blood of huPBMC-engrafted NSG mice on Day 17 after the first dosing with the indicated test articles at the indicated concentrations.
  • FIG. 177 depicts body weight (as a percentage of initial body weight) of huPBMC-engrafted NSG mice on Day 25 after the first dosing with the indicated test articles at the indicated concentrations.
  • FIG. 178 depicts serum IFN ⁇ concentration in huPBMC-engrafted NSG mice on Day 7 after the first dosing with the indicated test articles at the indicated concentrations.
  • FIG. 179A , FIG. 179B , FIG. 179C , and FIG. 179D depicts A) CD45 + cell, B) CD3 + T cell, C) CD4 + T cell, D) CD8 + T cell counts in blood of huPBMC-engrafted NSG mice on Day 10 after the first dosing with the indicated test articles at the indicated concentrations.
  • FIG. 180 depicts body weight (as a percentage of initial body weight) of huPBMC-engrafted NSG mice on Day 11 after the first dosing with the indicated test articles at the indicated concentrations.
  • FIG. 181 depicts a time course of body weight (as a percentage of initial body weight) of huPBMC-engrafted NSG mice following the first dosing with the indicated test articles at the indicated concentrations.
  • FIG. 182 depicts CD45 + cell counts on Day 21 in pp65-MCF7 and huPBMC-engrafted NSG mice dosed with XENP16432 and/or XENP24045.
  • FIG. 183 depicts tumor volume on Day 31 in pp65-MCF7 and huPBMC-engrafted NSG mice dosed with XENP16432 and/or XENP24045.
  • FIG. 184 depicts tumor volume over time (post-huPBMC engraftment) in pp65-MCF7 and huPBMC (1.5 ⁇ 10 6 )-engrafted NSG mice dosed with XENP16432 and/or XENP24045.
  • FIG. 185 depicts tumor volume over time (post-huPBMC engraftment) in pp65-MCF7 and huPBMC(5 ⁇ 10 6 )-engrafted NSG mice dosed with XENP16432 and/or XENP24045.
  • FIG. 186 depicts the sequences of XENP21993, an scIL-15/R ⁇ -Fc fusion comprising a wild-type IL-15.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in the Figures, and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and constant/Fc regions.
  • FIG. 187 depicts the sequences of XENP22853, an IL-15/R ⁇ -heteroFc fusion comprising a wild-type IL-15 and Xtend Fc (M428L/N434S) variant.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in the Figures, and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and constant/Fc regions.
  • FIG. 188 depicts the sequences of XENP24294, an scIL-15/R ⁇ -Fc fusion comprising an IL-15(N4D/N65D) variant and Xtend Fc (M428L/N434S) substitution.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in the Figures, and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and constant/Fc regions.
  • FIG. 189 depicts the serum concentration of the indicated test articles over time in cynomolgus monkeys following a first dose at the indicated relative concentrations.
  • FIG. 190 depicts relative serum concentrations of XENP22853 and corresponding WT non-Xtend XENP20818 over time.
  • FIG. 191 depicts sequences for illustrative IL-15 variants engineered for reduced potency and comprising a D30N substitution. Including within each of these variant IL-15 sequences are sequences that are 90%, 95%, 98% and 99% identical (as defined herein) to the recited sequences, and/or contain from 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 additional amino acid substitutions. In a nonlimiting example, the recited sequences may contain additional amino acid modifications such as those contributing to formation of covalent disulfide bonds as described in Example 2.
  • FIG. 192A - FIG. 192C depict illustrative scIL-15/R ⁇ -Fc fusions having IL-15 variants comprising D30N substitution.
  • IL-15 and IL-15R ⁇ (sushi) are underlined, linkers are double underlined (although as will be appreciated by those in the art, the linkers can be replaced by other linkers, some of which are depicted in the Figures, and slashes (/) indicate the border(s) between IL-15, IL-15R ⁇ , linkers, and constant/Fc regions.
  • FIG. 193A , FIG. 193B , FIG. 193C , FIG. 193D , FIG. 193E , FIG. 193F and FIG. 193G depict percentage of A) CD4+CD45RA ⁇ , B) CD4+CD45RA+, C) CD8+CD45RA ⁇ , D) CD8+CD45RA+, E) CD16+NK cells, F) CD56+NK cells, and G) ⁇ cells expression Ki67 following incubation with the indicated test articles.
  • FIG. 194A , FIG. 194B , FIG. 194C , FIG. 194D and FIG. 194E show that a mechanistic PK/RO/TMDD model depicts relative PD effects as discussed in Example 18.
  • the predicted RO-AUC values correlate very well with experimentally determined PD-AUC (pharmacodynamic AUC), indicating that RO-AUC is a target value to optimize, allowing a prediction of the optimal Kd value.
  • FIG. 195 shows a schematic illustrating the hypothesis that receptor-mediated internalization is a key factor that influences the PK/PD relationships of various IL-15s.
  • k prolif indicates proliferation rate.
  • kd eathCTL indicates rate of cell death.
  • K d indicates dissociation constant.
  • k el indicates rate of elimination.
  • k infusion indicates rate of infusion into central blood.
  • k syn indicates rate of synthesis of IL-15 receptors.
  • k deg indicates rate of degradation of IL-15 receptors.
  • FIG. 196 depicts overlay of PK data from multiple cynomolgus monkey studies which were input into simulations based on the model depicted in FIG. 195 .
  • FIG. 197 depicts that estimated KD values based on model depicted in FIG. 1 correlate linearly with experimentally measured EC50 values for binding.
  • FIG. 198 depicts simulated PK profiles, receptor occupancy (RO) curves, and pharmacodynamic profiles based on the model depicted in FIG. 195 .
  • the model demonstrates that reduced potency prolongs exposure, and that an optimal KD exists for maximal pharmacodynamic (i.e., T cell expansion).
  • FIG. 199A - FIG. 199F depict correlation between A) predicted RO-AUC values with experimentally determined PD-AUC (pharmacodynamic AUC), B) predicted RO-AUC values with experimentally determined PD-Max (peak CD8+ T cell counts), C) predicted RO-AUC values with experimentally determined ALB-min (nadir serum albumin concentration), D) predicted RO-Max (maximum receptor occupancy) values with experimentally determined PD-AUC, E) predicted RO-Max with experimentally determined PD-Max, and F) predicted RO-Max with experimentally determined ALB-Min.
  • the predicted RO-AUC values correlate very well with experimentally determined PD-AUC and PD-max, while the predicted RO-Max values correlate very well with experimentally determined ALB-Min.
  • FIG. 200 depicts a simulation scan (based on the model depicted in FIG. 195 ) over a variety of K D values was performed for either Xtend or non-Xtend versions of IL-15/R ⁇ -heteroFc fusions, revealing that the predicted optimal K D is approximately 200 nM, similar to the estimated K D value of XmAb24306.
  • FIG. 201A and FIG. 201B depict A) human CD8+ T cell counts and B) CD25 expression on human CD8+ T cells in pp65-MCF7 and huPBMC-engrafted NSG mice dosed with XENP16432 and/or XENP24045.
  • *** indicates p ⁇ 0.001 and ** indicates p ⁇ 0.01 as determined by an unpaired t-test. It was found that treatment with the combination of XENP24045 and anti-PD-1 mAb significantly enhanced CD8+ T cell expansion on later days, and induced earlier activation of CD8+ T cells.
  • FIG. 202A - FIG. 202C depict expansion of A) CD8+ T cells, B) CD16+NK cells, and C) lymphocytes in cynomolgus monkeys following dosing with 0.3 ⁇ dose XENP20818, 0.3 ⁇ dose XmAb24306, and 0.6 ⁇ dose XmAb24306.
  • the data shows enhanced pharmacodynamics conferred by reduced-potency XmAb24306.
  • FIG. 203 depicts the percent change in serum albumin (as an indicator of vascular leak) in cynomolgus monkeys following dosing with 0.3 ⁇ dose XENP20818, 0.3 ⁇ dose XmAb24306, and 0.6 ⁇ dose XmAb24306.
  • the data shows enhanced tolerability (as indicated by reduction in albumin drop) conferred by reduced-potency XmAb24306.
  • FIG. 204A - FIG. 204F depict A) CD45 + cell, B) CD3 + T cell, C) CD4 + T cell, D) CD8 + T cell, E) CD16 + CD56 + NK cell counts, and F) CD4 to CD8 ratio in blood of huPBMC-engrafted NSG mice on Day 7 after the first dosing with the indicated test articles at the indicated concentrations.
  • FIG. 205A - FIG. 205F depict A) CD45+ cell, B) CD3+ T cell, C) CD4+ T cell, D) CD8+ T cell, E) CD16+CD56+NK cell counts, and F) CD4 to CD8 ratio in blood of huPBMC-engrafted NSG mice on Day 14 after the first dosing with the indicated test articles at the indicated concentrations.
  • the data show that by Day 14, treatment with a combination of 0.3 mg/kg or 0.1 mg/kg XENP24306 and 3 mg/kg XENP16432 significantly enhanced CD3+ T cell expansion beyond treatment with 3 mg/kg XENP16432 alone (statistics were performed on log-transformed data using unpaired t-test).
  • FIG. 206A - FIG. 206F depict A) CD45+ cell, B) CD3+ T cell, C) CD4+ T cell, D) CD8+ T cell, E) CD16+CD56+NK cell counts, and F) CD4 to CD8 ratio in blood of huPBMC-engrafted NSG mice on Day 21 after the first dosing with the indicated test articles at the indicated concentrations.
  • FIG. 207A-207I depict the change in body weight (as an indicator of GVHD) of huPBMC-engrafted NSG mice on A) Day 3, B) Day 6, C) Day 10, D) Day 13, E) Day 17, F) Day 20, G) Day 25, H) Day 28, and I) Day 29 after the first dosing with the indicated test articles at the indicated concentrations.
  • the data show that by Day 10, treatment with a combination of 0.3 mg/kg, 0.1 mg/kg, or 0.01 mg/kg XENP24306 and 3 mg/kg XENP16432 significantly enhanced GVHD beyond treatment with 3 mg/kg XENP16432 alone (statistics were performed using unpaired t-test).
  • FIG. 208 depicts the change in body weight (as an indicator of GVHD) of huPBMC-engrafted NSG mice over time after dosing with the indicating test articles at the indicated concentrations.
  • FIG. 209A - FIG. 209C depict serum IFN ⁇ concentration in huPBMC-engrafted NSG mice on A) Day 7, B) Day 14, and C) Day 21 after the first dosing with the indicated test articles at the indicated concentrations.
  • FIG. 210A - FIG. 201F depict A) CD45+ cell, B) CD3+ T cell, C) CD4+ T cell, D) CD8+ T cell, E) CD16+CD56+NK cell counts, and F) CD4 to CD8 ratio in blood of pp65-MCF-7 and huPBMC-engrafted NSG mice on Day 14 after the first dosing with the indicated test articles at the indicated concentrations.
  • * denotes p ⁇ 0.05, unpaired t-test, indicated group in comparison to PBS-treated group; ⁇ denotes p ⁇ 0.04, unpaired t-test, indicated group in comparison to XENP16432-treated group. Data were log-transformed prior to statistical analysis.
  • FIG. 211A - FIG. 211F depict A) CD45+ cell, B) CD3+ T cell, C) CD4+ T cell, D) CD8+ T cell, E) CD16+CD56+NK cell counts, and F) CD4 to CD8 ratio in blood of pp65-MCF-7 and huPBMC-engrafted NSG mice on Day 21 after the first dosing with the indicated test articles at the indicated concentrations.
  • * denotes p ⁇ 0.05, unpaired t-test, indicated group in comparison to PBS-treated group; ⁇ denotes p ⁇ 0.04, unpaired t-test, indicated group in comparison to XENP16432-treated group. Data were log-transformed prior to statistical analysis.
  • FIG. 212A - FIG. 2121 depict the change in tumor volume (baseline-corrected) in pp65-MCF-7 and huPBMC-engrafted NSG mice on A) Day 4, B) Day 6, C) Day 8, D) Day 11, E) Day 13, F) Day 15, G) Day 19, H) Day 22, and I) Day 25 after the first dosing with the indicated test articles at the indicated concentrations.
  • the data show that by Day 11, treatment with a combination of 1.0 mg/kg, 0.3 mg/kg, or 0.1 mg/kg XENP24306 and 3 mg/kg XENP16432 significantly decreased tumor volume in comparison to treatment with XENP16432 alone (statistics were performed using unpaired t-test on baseline corrected tumor measurements).
  • FIG. 213A - FIG. 213B depict the A) change in tumor volume and B) change in body weight (as an indicator of GVHD) of pp65-MCF-7 and huPBMC-engrafted NSG mice over time after dosing with the indicating test articles at the indicated concentrations.
  • the data indicates that although all mice were dead in Groups C, F, and G, this corresponded to GVHD (as indicated by change in body weight).
  • FIG. 214A - FIG. 214C depict serum IFN ⁇ concentration in pp65-MCF-7 and huPBMC-engrafted NSG mice on A) Day 7, B) Day 14, and C) Day 21 after the first dosing with the indicated test articles at the indicated concentrations.
  • * denotes p ⁇ 0.05, unpaired t-test, indicated group in comparison to PBS-treated group; ⁇ denotes p ⁇ 0.04, unpaired t-test, indicated group in comparison to XENP16432-treated group. Data were log-transformed prior to statistical analysis.
  • ablation herein is meant a decrease or removal of binding and/or activity.
  • “ablating Fc ⁇ R binding” means the Fc region amino acid variant has less than 50% starting binding as compared to an Fc region not containing the specific variant, with less than 70-80-90-95-98% loss of binding being preferred, and in general, with the binding being below the level of detectable binding in a Biacore assay.
  • the Fc monomers of the invention retain binding to the FcRn.
  • ADCC antibody dependent cell-mediated cytotoxicity
  • ADCC antibody dependent cell-mediated cytotoxicity
  • the cell-mediated reaction wherein nonspecific cytotoxic cells that express Fc ⁇ Rs recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • ADCC is correlated with binding to Fc ⁇ RIIIa; increased binding to Fc ⁇ RIIIa leads to an increase in ADCC activity.
  • many embodiments of the invention ablate ADCC activity entirely.
  • ADCP antibody dependent cell-mediated phagocytosis as used herein is meant the cell-mediated reaction wherein nonspecific cytotoxic cells that express Fc ⁇ Rs recognize bound antibody on a target cell and subsequently cause phagocytosis of the target cell.
  • modification herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence or an alteration to a moiety chemically linked to a protein.
  • a modification may be an altered carbohydrate or PEG structure attached to a protein.
  • amino acid modification herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence.
  • the amino acid modification is always to an amino acid coded for by DNA, e.g., the 20 amino acids that have codons in DNA and RNA.
  • amino acid substitution or “substitution” herein is meant the replacement of an amino acid at a particular position in a parent polypeptide sequence with a different amino acid.
  • the substitution is to an amino acid that is not naturally occurring at the particular position, either not naturally occurring within the organism or in any organism.
  • the substitution E272Y or 272Y refers to a variant polypeptide, in this case an Fc variant, in which the glutamic acid at position 272 is replaced with tyrosine.
  • a protein which has been engineered to change the nucleic acid coding sequence, but not to change the starting amino acid is not an “amino acid substitution”; that is, despite the creation of a new gene encoding the same protein, if the protein has the same amino acid at the particular position that it started with, it is not an amino acid substitution.
  • substitutions can include naturally occurring amino acids and, in some cases, synthetic amino acids. Examples include U.S. Pat. No.
  • amino acid insertion or “insertion” as used herein is meant the addition of an amino acid residue or sequence at a particular position in a parent polypeptide sequence.
  • ⁇ 233E designates an insertion of glutamic acid after position 233 and before position 234.
  • ⁇ 233ADE or A233ADE designates an insertion of AlaAspGlu after position 233 and before position 234.
  • amino acid deletion or “deletion” as used herein is meant the removal of an amino acid residue or sequence at a particular position in a parent polypeptide sequence.
  • E233, E233#, E233( ) or E233del designates a deletion of glutamic acid at position 233.
  • EDA233 ⁇ or EDA233# designates a deletion of the sequence GluAspAla that begins at position 233.
  • variant protein a protein that differs from that of a parent protein by virtue of at least one amino acid modification.
  • Protein variant may refer to the protein itself, a composition comprising the protein, the amino sequence that encodes it, or the DNA or nucleic acid sequence that encodes it.
  • the protein variant has at least one amino acid modification compared to the parent protein, e.g., from about one to about seventy amino acid modifications, and preferably from about one to about five amino acid modifications compared to the parent.
  • the medication can be an addition, deletion, or substitution.
  • the parent polypeptide for example an Fc parent polypeptide
  • the protein variant sequence herein will preferably possess at least about 80% identity with a parent protein sequence, and most preferably at least about 90% identity, more preferably at least about 95-98-99% identity.
  • antibody variant or “variant antibody” as used herein is meant an antibody that differs from a parent antibody by virtue of at least one amino acid modification
  • IgG variant or “variant IgG” as used herein is meant an antibody that differs from a parent IgG (again, in many cases, from a human IgG sequence) by virtue of at least one amino acid modification
  • immunoglobulin variant or “variant immunoglobulin” as used herein is meant an immunoglobulin sequence that differs from that of a parent immunoglobulin sequence by virtue of at least one amino acid modification
  • Fc variant or “variant Fc” as used herein is meant a protein comprising an amino acid modification in an Fc domain.
  • the modification can be an addition, deletion, or substitution.
  • the Fc variants of the present invention are defined according to the amino acid modifications that compose them.
  • N434S or 434S is an Fc variant with the substitution for a serine residue at position 434 relative to the parent Fc polypeptide, wherein the numbering is according to the EU index.
  • M428L/N434S defines an Fc variant with the substitutions M428L and N434S relative to the parent Fc polypeptide.
  • the identity of the WT amino acid may be unspecified, in which case the aforementioned variant is referred to as 428L/434S.
  • substitutions are provided is arbitrary, that is to say that, for example, 428L/434S is the same Fc variant as 434S/428L, and so on.
  • amino acid position numbering is according to the EU index.
  • the EU index or EU index as in Kabat or EU numbering scheme refers to the numbering of the EU antibody (Edelman et al., 1969, Proc Natl Acad Sci USA 63:78-85, hereby entirely incorporated by reference).
  • the modification can be an addition, deletion, or substitution.
  • substitutions can include naturally occurring amino acids and, in some cases, synthetic amino acids.
  • protein at least two covalently attached amino acids, which includes proteins, polypeptides, oligopeptides and peptides.
  • the peptidyl group may comprise naturally occurring amino acids and peptide bonds, or synthetic peptidomimetic structures, i.e. “analogs”, such as peptoids (see Simon et al., PNAS USA 89(20):9367 (1992), entirely incorporated by reference).
  • the amino acids may either be naturally occurring or synthetic (e.g. not an amino acid that is coded for by DNA); as will be appreciated by those in the art.
  • homo-phenylalanine, citrulline, ornithine and noreleucine are considered synthetic amino acids for the purposes of the invention, and both D- and L-(R or S) configured amino acids may be utilized.
  • the variants of the present invention may comprise modifications that include the use of synthetic amino acids incorporated using, for example, the technologies developed by Schultz and colleagues, including but not limited to methods described by Cropp & Shultz, 2004, Trends Genet.
  • polypeptides may include synthetic derivatization of one or more side chains or termini, glycosylation, PEGylation, circular permutation, cyclization, linkers to other molecules, fusion to proteins or protein domains, and addition of peptide tags or labels.
  • a biologically functional molecule comprises two or more proteins, each protein may be referred to as a “monomer” or as a “subunit” or as a “domain”; and the biologically functional molecule may be referred to as a “complex”.
  • residue as used herein is meant a position in a protein and its associated amino acid identity.
  • Asparagine 297 also referred to as Asn297 or N297
  • Asn297 is a residue at position 297 in the human antibody IgG1.
  • IgG subclass modification or “isotype modification” as used herein is meant an amino acid modification that converts one amino acid of one IgG isotype to the corresponding amino acid in a different, aligned IgG isotype.
  • IgG1 comprises a tyrosine and IgG2 a phenylalanine at EU position 296, a F296Y substitution in IgG2 is considered an IgG subclass modification.
  • non-naturally occurring modification is meant an amino acid modification that is not isotypic.
  • the substitution 434S in IgG1, IgG2, IgG3, or IgG4 (or hybrids thereof) is considered a non-naturally occurring modification.
  • amino acid and “amino acid identity” as used herein is meant one of the 20 naturally occurring amino acids that are coded for by DNA and RNA.
  • effector function as used herein is meant a biochemical event that results from the interaction of an antibody Fc region with an Fc receptor or ligand. Effector functions include but are not limited to ADCC, ADCP, and CDC.
  • IgG Fc ligand or “Fc ligand” as used herein is meant a molecule, preferably a polypeptide, from any organism that binds to the Fc region of an IgG antibody to form an Fc/Fc ligand complex.
  • Fc ligands include but are not limited to Fc ⁇ RIs, Fc ⁇ RIIs, Fc ⁇ RIIIs, FcRn, C1q, C3, mannan binding lectin, mannose receptor, staphylococcal protein A, streptococcal protein G, and viral Fc ⁇ R.
  • Fc ligands also include Fc receptor homologs (FcRH), which are a family of Fc receptors that are homologous to the Fc ⁇ Rs (Davis et al., 2002, Immunological Reviews 190:123-136, entirely incorporated by reference).
  • Fc ligands may include undiscovered molecules that bind Fc.
  • Particular IgG Fc ligands are FcRn and Fc gamma receptors.
  • Fc gamma receptor any member of the family of proteins that bind the IgG antibody Fc region and is encoded by an Fc ⁇ R gene. In humans this family includes but is not limited to Fc ⁇ RI (CD64), including isoforms Fc ⁇ RIa, Fc ⁇ RIb, and Fc ⁇ RIc; Fc ⁇ RII (CD32), including isoforms Fc ⁇ RIIa (including allotypes H131 and R131), Fc ⁇ RIIb (including Fc ⁇ RIIb-1 and Fc ⁇ RIIb-2), and Fc ⁇ RIIc; and Fc ⁇ RIII (CD16), including isoforms Fc ⁇ RIIIa (including allotypes V158 and F158) and Fc ⁇ RIIIb (including allotypes Fc ⁇ RIIb-NA1 and Fc ⁇ RIIb-NA2) (Jefferis et al., 2002, Immunol Lett 82:57-65
  • An Fc ⁇ R may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys.
  • Mouse Fc ⁇ Rs include but are not limited to Fc ⁇ RI (CD64), Fc ⁇ RII (CD32), Fc ⁇ RIII (CD16), and Fc ⁇ RIII-2 (CD16-2), as well as any undiscovered mouse Fc ⁇ Rs or Fc ⁇ R isoforms or allotypes.
  • FcRn or “neonatal Fc receptor” as used herein is meant a protein that binds the IgG antibody Fc region and is encoded at least in part by an FcRn gene.
  • the FcRn may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys.
  • the functional FcRn protein comprises two polypeptides, often referred to as the heavy chain and light chain.
  • the light chain is beta-2-microglobulin ( ⁇ 2-microglobulin) and the heavy chain is encoded by the FcRn gene.
  • FcRn or an FcRn protein refers to the complex of FcRn heavy chain with ⁇ 2-microglobulin.
  • Fc variants can be used to increase binding to the FcRn, and in some cases, to increase serum half-life.
  • the Fc monomers of the invention retain binding to the FcRn (and, as noted below, can include amino acid variants to increase binding to the FcRn).
  • parent polypeptide as used herein is meant a starting polypeptide that is subsequently modified to generate a variant.
  • the parent polypeptide may be a naturally occurring polypeptide, or a variant or engineered version of a naturally occurring polypeptide.
  • Parent polypeptide may refer to the polypeptide itself, compositions that comprise the parent polypeptide, or the amino acid sequence that encodes it.
  • parent immunoglobulin as used herein is meant an unmodified immunoglobulin polypeptide that is modified to generate a variant
  • parent antibody as used herein is meant an unmodified antibody that is modified to generate a variant antibody. It should be noted that “parent antibody” includes known commercial, recombinantly produced antibodies as outlined below.
  • Fc or “Fc region” or “Fc domain” as used herein is meant the polypeptide comprising the constant region of an antibody, in some instances, excluding all or a portion of the first constant region immunoglobulin domain (e.g., CH1) or a portion thereof, and in some cases, further excluding all or a portion of the hinge.
  • an Fc can refer to the last two constant region immunoglobulin domains (e.g., CH2 and CH3) of IgA, IgD, and IgG, the last three constant region immunoglobulin domains of IgE and IgM, and optionally, all or a portion of the flexible hinge N-terminal to these domains.
  • Fc may include the J chain.
  • the Fc domain comprises immunoglobulin domains CH2 and CH3 (C ⁇ 2 and C ⁇ 3) and the lower hinge region between CH1 (C ⁇ 1) and CH2 (C ⁇ 2).
  • the human IgG heavy chain Fc region is usually defined to include residues E216, C226, or A231 to its carboxyl-terminus, wherein the numbering is according to the EU index as in Kabat.
  • amino acid modifications are made to the Fc region, for example to alter binding to one or more Fc ⁇ R or to the FcRn.
  • heavy constant region domains can be different among different numbering systems.
  • a useful comparison of heavy constant region numbering according to EU and Kabat is as below, see Edelman et al., 1969, Proc Natl Acad Sci USA 63:78-85 and Kabat et al., 1991, Sequences of Proteins of Immunological Interest, 5th Ed., United States Public Health Service, National Institutes of Health, Bethesda, entirely incorporated by reference.
  • fusion protein as used herein is meant covalent joining of at least two proteins. Fusion proteins may comprise artificial sequences, e.g. a domain linker, as described herein.
  • Fc fusion protein or “immunoadhesin” herein is meant a protein comprising an Fc region, generally linked (optionally through a domain linker, as described herein) to one or more different proteins, such as to IL-15 and/or IL-15R, as described herein.
  • two Fc fusion proteins can form a homodimeric Fc fusion protein or a heterodimeric Fc fusion protein with the latter being preferred.
  • one monomer of the heterodimeric Fc fusion protein comprises an Fc domain alone (e.g., an empty Fc domain) and the other monomer is an Fc fusion, comprising a variant Fc domain and a protein domain, such as a receptor, ligand or other binding partner.
  • position as used herein is meant a location in the sequence of a protein. Positions may be numbered sequentially, or according to an established format, for example the EU index for antibody numbering.
  • strandedness in the context of the monomers of the heterodimeric proteins of the invention herein is meant that, similar to the two strands of DNA that “match”, heterodimerization variants are incorporated into each monomer so as to preserve, create, and/or enhance the ability to “match” to form heterodimers.
  • steric variants that are “charge pairs” that can be utilized as well do not interfere with the pI variants, e.g. the charge variants that make a pI higher are put on the same “strand” or “monomer” to preserve both functionalities.
  • wild type or WT herein is meant an amino acid sequence or a nucleotide sequence that is found in nature, including allelic variations.
  • a WT protein has an amino acid sequence or a nucleotide sequence that has not been intentionally modified.
  • the heterodimeric proteins of the present invention are generally isolated or recombinant.
  • isolated when used to describe the various polypeptides disclosed herein, means a polypeptide that has been identified and separated and/or recovered from a cell or cell culture from which it was expressed. Ordinarily, an isolated polypeptide will be prepared by at least one purification step.
  • Recombinant means the proteins are generated using recombinant nucleic acid techniques in exogeneous host cells.
  • Percent (%) amino acid sequence identity with respect to a protein sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the specific (parental) sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. One particular program is the ALIGN-2 program outlined at paragraphs [0279] to [0280] of US Pub. No. 20160244525, hereby incorporated by reference.
  • invention sequence The degree of identity between an amino acid sequence of the present invention (“invention sequence”) and the parental amino acid sequence is calculated as the number of exact matches in an alignment of the two sequences, divided by the length of the “invention sequence,” or the length of the parental sequence, whichever is the shortest. The result is expressed in percent identity.
  • two or more amino acid sequences are at least 50%, 60%, 70%, 80%, or 90% identical. In some embodiments, two or more amino acid sequences are at least 95%, 97%, 98%, 99%, or even 100% identical.
  • antigen binding domain or “ABD” hereins is meant in part of an antigen binding molecule which confers its binding specificity to an antigen determinant.
  • antigen binding molecule refers in its broadest sense to any molecule that specifically binds to an antigenic determinant.
  • An antigen binding molecule may be a protein, carbohydrate, lipid, or other chemical compound.
  • antigen binding molecules are immunoglobulin and derivatives or fragments thereof, e.g., Fab and scFv. Additional examples of antigen binding molecules are receptors and ligands.
  • Specific binding or “specifically binds to” or is “specific for” a particular antigen or an epitope means binding that is measurably different from a non-specific interaction. Specific binding can be measured, for example, by determining binding of a molecule compared to binding of a control molecule, which generally is a molecule of similar structure that does not have binding activity. For example, specific binding can be determined by competition with a control molecule that is similar to the target.
  • the strength, or affinity, of specific binding can be expressed in terms of dissociation constant (K D ) of the interaction, wherein a smaller K D represents greater affinity and a larger K D represents lower affinity.
  • Binding properties can be determined by methods well known in the art such as bio-layer interferometry and surface plasmon resonance based methods. One such method entails measuring the rates of antigen-binding site/antigen or receptor/ligand complex association and dissociation, wherein rates depend on the concentration of the complex partners, the affinity of the interaction, and geometric parameters that equally influence the rate in both directions.
  • association rate (ka) and the dissociation rate (kd) can be determined, and the ratio of kd/ka is equal to the dissociation constant K D (See, e.g., Nature 361:186-187 (1993) and Davies et al. (1990) Annual Rev Biochem 59:439-473).
  • an antigen binding molecule that specifically binds an antigen will have a K D that is 20-, 50-, 100-, 500-, 1000-, 5,000-, 10,000- or more times greater for a control molecule relative to the antigen or epitope.
  • specific binding for a particular molecule or an epitope can be exhibited, for example, by an antigen binding molecule having a ka or association rate for an antigen or epitope of at least 20-, 50-, 100-, 500-, 1000-, 5,000-, 10,000- or more times greater for the epitope relative to a control.
  • epitope is herein meant a determinant that interacts with a specific antigen binding domain, for example variable region of an antibody molecule, known as a paratope.
  • Epitopes are groupings of molecules such as amino acids or sugar side chains and usually have specific structural characteristics, as well as specific charge characteristics.
  • a single molecule may have more than one epitope.
  • the epitope may comprise amino acid residues directly involved in the binding (also called immunodominant component of the epitope) and other amino acid residues, which are not directly involved in the binding, such as amino acid residues which are effectively blocked by the specifically antigen binding peptide; in other words, the amino acid residue is within the footprint of the specifically antigen binding peptide.
  • Epitopes may be either conformational or linear.
  • a conformational epitope is produced by spatially juxtaposed amino acids from different segments of the linear polypeptide chain.
  • a linear epitope is one produced by adjacent amino acid residues in a polypeptide chain. Conformational and nonconformational epitopes may be distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
  • Antigen binding molecules that recognize the same epitope can be verified in a simple immunoassay showing the ability of one antigen binding molecule to block the binding of another antigen binding molecule to a target antigen, for example “binning.”
  • the invention not only includes the enumerated antigen binding molecules and antigen binding domains herein, but those that compete for binding with the epitopes bound by the enumerated antigen binding molecules or antigen binding domains.
  • fused or “covalently linked” is herein meant that the components (e.g., IL-15 and an Fc domain) are linked by peptide bonds, either directly or via domain linkers, outlined herein.
  • single-chain refers to a molecule comprising amino acid monomers linearly linked by peptide bonds.
  • Antibodies that find use in the present invention can take on a number of formats as described herein, including traditional antibodies as well as antibody derivatives, fragments and mimetics, described herein and depicted in the figures.
  • Each tetramer is typically composed of two identical pairs of polypeptide chains, each pair having one “light” (typically having a molecular weight of about 25 kDa) and one “heavy” chain (typically having a molecular weight of about 50-70 kDa).
  • Human light chains are classified as kappa and lambda light chains.
  • the present invention is directed to bispecific antibodies that generally are based on the IgG class, which has several subclasses, including, but not limited to IgG1, IgG2, IgG3, and IgG4. In general, IgG1, IgG2 and IgG4 are used more frequently than IgG3.
  • IgG1 has different allotypes with polymorphisms at 356 (D or E) and 358 (L or M).
  • the sequences depicted herein use the 356E/358M allotype, however the other allotype is included herein. That is, any sequence inclusive of an IgG1 Fc domain included herein can have 356D/358L replacing the 356E/358M allotype.
  • cysteines at position 220 have at least one of the cysteines at position 220 replaced by a serine; generally this is the on the “scFv monomer” side for most of the sequences depicted herein, although it can also be on the “Fab monomer” side, or both, to reduce disulfide formation.
  • cysteines replaced (C220S).
  • isotype as used herein is meant any of the subclasses of immunoglobulins defined by the chemical and antigenic characteristics of their constant regions. It should be understood that therapeutic antibodies can also comprise hybrids of isotypes and/or subclasses. For example, as shown in US Publication 2009/0163699, incorporated by reference, the present invention the use of human IgG1/G2 hybrids.
  • the hypervariable region generally encompasses amino acid residues from about amino acid residues 24-34 (LCDR1; “L” denotes light chain), 50-56 (LCDR2) and 89-97 (LCDR3) in the light chain variable region and around about 31-35B (HCDR1; “H” denotes heavy chain), 50-65 (HCDR2), and 95-102 (HCDR3) in the heavy chain variable region; Kabat et al., SEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991) and/or those residues forming a hypervariable loop (e.g.
  • variable heavy and/or variable light sequence includes the disclosure of the associated (inherent) CDRs.
  • the disclosure of each variable heavy region is a disclosure of the vhCDRs (e.g. vhCDR1, vhCDR2 and vhCDR3) and the disclosure of each variable light region is a disclosure of the vlCDRs (e.g. vlCDR1, vlCDR2 and vlCDR3).
  • vlCDRs e.g. vlCDR1, vlCDR2 and vlCDR3
  • the Kabat numbering system is generally used when referring to a residue in the variable domain (approximately, residues 1-107 of the light chain variable region and residues 1-113 of the heavy chain variable region) and the EU numbering system for Fc regions (e.g, Kabat et al., supra (1991)).
  • Ig domain of the heavy chain is the hinge region.
  • hinge region or “hinge region” or “antibody hinge region” or “hinge domain” herein is meant the flexible polypeptide comprising the amino acids between the first and second constant domains of an antibody.
  • the IgG CH1 domain ends at EU position 215, and the IgG CH2 domain begins at residue EU position 231.
  • the antibody hinge is herein defined to include positions 216 (E216 in IgG1) to 230 (p230 in IgG1), wherein the numbering is according to the EU index as in Kabat.
  • a “hinge fragment” is used, which contains fewer amino acids at either or both of the N- and C-termini of the hinge domain.
  • pI variants can be made in the hinge region as well.
  • the light chain generally comprises two domains, the variable light domain (containing the light chain CDRs and together with the variable heavy domains forming the Fv region), and a constant light chain region (often referred to as CL or C ⁇ ).
  • Fc region Another region of interest for additional substitutions, outlined below, is the Fc region.
  • a “full CDR set” comprises the three variable light and three variable heavy CDRs, e.g. a vlCDR1, vlCDR2, vlCDR3, vhCDR1, vhCDR2 and vhCDR3. These can be part of a larger variable light or variable heavy domain, respectfully.
  • the variable heavy and variable light domains can be on separate polypeptide chains, when a heavy and light chain is used (for example when Fabs are used), or on a single polypeptide chain in the case of scFv sequences.
  • epitope binding site contributes to the formation of the antigen-binding, or more specifically, epitope binding site of antibodies.
  • Epitope refers to a determinant that interacts with a specific antigen binding site in the variable region of an antibody molecule known as a paratope. Epitopes are groupings of molecules such as amino acids or sugar side chains and usually have specific structural characteristics, as well as specific charge characteristics. A single antigen may have more than one epitope.
  • the epitope may comprise amino acid residues directly involved in the binding (also called immunodominant component of the epitope) and other amino acid residues, which are not directly involved in the binding, such as amino acid residues which are effectively blocked by the specifically antigen binding peptide; in other words, the amino acid residue is within the footprint of the specifically antigen binding peptide.
  • Epitopes may be either conformational or linear.
  • a conformational epitope is produced by spatially juxtaposed amino acids from different segments of the linear polypeptide chain.
  • a linear epitope is one produced by adjacent amino acid residues in a polypeptide chain. Conformational and nonconformational epitopes may be distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
  • Antibodies that recognize the same epitope can be verified in a simple immunoassay showing the ability of one antibody to block the binding of another antibody to a target antigen, for example “binning.”
  • the invention not only includes the enumerated antigen binding domains and antibodies herein, but those that compete for binding with the epitopes bound by the enumerated antigen binding domains.
  • the present invention provides different antibody domains.
  • the heterodimeric antibodies of the invention comprise different domains within the heavy and light chains, which can be overlapping as well. These domains include, but are not limited to, the Fc domain, the CH1 domain, the CH2 domain, the CH3 domain, the hinge domain, the heavy constant domain (CH1-hinge-Fc domain or CH1-hinge-CH2-CH3), the variable heavy domain, the variable light domain, the light constant domain, Fab domains and scFv domains.
  • the “Fc domain” includes the —CH2-CH3 domain, and optionally a hinge domain (—H—CH2-CH3).
  • a scFv when a scFv is attached to an Fc domain, it is the C-terminus of the scFv construct that is attached to all or part of the hinge of the Fc domain; for example, it is generally attached to the sequence EPKS (SEQ ID NO: 292) which is the beginning of the hinge.
  • the heavy chain comprises a variable heavy domain and a constant domain, which includes a CH1-optional hinge-Fc domain comprising a CH2-CH3.
  • the light chain comprises a variable light chain and the light constant domain.
  • a scFv comprises a variable heavy chain, an scFv linker, and a variable light domain.
  • the C-terminus of the variable heavy chain is attached to the N-terminus of the scFv linker, the C-terminus of which is attached to the N-terminus of a variable light chain (N-vh-linker-vl-C) although that can be switched (N-vl-linker-vh-C).
  • Some embodiments of the invention comprise at least one scFv domain, which, while not naturally occurring, generally includes a variable heavy domain and a variable light domain, linked together by a scFv linker.
  • a scFv linker As outlined herein, while the scFv domain is generally from N- to C-terminus oriented as vh-scFv linker-vl, this can be reversed for any of the scFv domains (or those constructed using vh and vl sequences from Fabs), to vl-scFv linker-vh, with optional linkers at one or both ends depending on the format.
  • linker peptide may predominantly include the following amino acid residues: Gly, Ser, Ala, or Thr.
  • the linker peptide should have a length that is adequate to link two molecules in such a way that they assume the correct conformation relative to one another so that they retain the desired activity.
  • the linker is from about 1 to 50 amino acids in length, preferably about 1 to 30 amino acids in length.
  • linkers of 1 to 20 amino acids in length may be used, with from about 5 to about 10 amino acids finding use in some embodiments.
  • Useful linkers include glycine-serine polymers, including for example (GS)n (SEQ ID NO: 11), (GSGGS)n (SEQ ID NO: 12), (GGGGS)n (SEQ ID NO: 13), and (GGGS)n (SEQ ID NO: 14), where n is an integer of at least one (and generally from 3 to 4), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers.
  • nonproteinaceous polymers including but not limited to polyethylene glycol (PEG), polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol, may find use as linkers.
  • PEG polyethylene glycol
  • polypropylene glycol polypropylene glycol
  • polyoxyalkylenes polyoxyalkylenes
  • copolymers of polyethylene glycol and polypropylene glycol may find use as linkers.
  • linker sequences may include any sequence of any length of CL/CH1 domain but not all residues of CL/CH1 domain; for example the first 5-12 amino acid residues of the CL/CH1 domains.
  • Linkers can be derived from immunoglobulin light chain, for example C ⁇ or C ⁇ .
  • Linkers can be derived from immunoglobulin heavy chains of any isotype, including for example C ⁇ 1, C ⁇ 2, C ⁇ 3, C ⁇ 4, C ⁇ 1, C ⁇ 2, C ⁇ , C ⁇ , and C ⁇ .
  • Linker sequences may also be derived from other proteins such as Ig-like proteins (e.g. TCR, FcR, KIR), hinge region-derived sequences, and other natural sequences from other proteins.
  • the linker is a “domain linker”, used to link any two domains as outlined herein together.
  • a domain linker that attaches the C-terminus of the CH1 domain of the Fab to the N-terminus of the scFv, with another optional domain linker attaching the C-terminus of the scFv to the CH2 domain (although in many embodiments the hinge is used as this domain linker).
  • a glycine-serine polymer as the domain linker, including for example (GS)n (SEQ ID NO: 15), (GSGGS)n (SEQ ID NO: 16), (GGGGS)n (SEQ ID NO: 17), and (GGGS)n (SEQ ID NO: 18), where n is an integer of at least one (and generally from 3 to 4 to 5) as well as any peptide sequence that allows for recombinant attachment of the two domains with sufficient length and flexibility to allow each domain to retain its biological function.
  • charged domain linkers as used in some embodiments of scFv linkers can be used.
  • the linker is a scFv linker, used to covalently attach the vh and vl domains as discussed herein.
  • the scFv linker is a charged scFv linker.
  • the present invention further provides charged scFv linkers, to facilitate the separation in pI between a first and a second monomer. That is, by incorporating a charged scFv linker, either positive or negative (or both, in the case of scaffolds that use scFvs on different monomers), this allows the monomer comprising the charged linker to alter the pI without making further changes in the Fc domains.
  • These charged linkers can be substituted into any scFv containing standard linkers. Again, as will be appreciated by those in the art, charged scFv linkers are used on the correct “strand” or monomer, according to the desired changes in pI.
  • the original pI of the Fv region for each of the desired antigen binding domains are calculated, and one is chosen to make an scFv, and depending on the pI, either positive or negative linkers are chosen.
  • Charged domain linkers can also be used to increase the pI separation of the monomers of the invention as well, and thus those included herein can be used in any embodiment herein where a linker is utilized.
  • heterodimeric antibodies meaning that the protein has at least two associated Fc sequences self-assembled into a heterodimeric Fc domain and at least two Fv regions, whether as Fabs or as scFvs.
  • the checkpoint blockade antibodies of the invention comprise a heavy chain variable region from a particular germline heavy chain immunoglobulin gene and/or a light chain variable region from a particular germline light chain immunoglobulin gene.
  • such antibodies may comprise or consist of a human antibody comprising heavy or light chain variable regions that are “the product of” or “derived from” a particular germline sequence.
  • a human antibody that is “the product of” or “derived from” a human germline immunoglobulin sequence can be identified as such by comparing the amino acid sequence of the human antibody to the amino acid sequences of human germline immunoglobulins and selecting the human germline immunoglobulin sequence that is closest in sequence (i.e., greatest % identity) to the sequence of the human antibody (using the methods outlined herein).
  • a human antibody that is “the product of” or “derived from” a particular human germline immunoglobulin sequence may contain amino acid differences as compared to the germline sequence, due to, for example, naturally-occurring somatic mutations or intentional introduction of site-directed mutation.
  • a humanized antibody typically is at least 90% identical in amino acids sequence to an amino acid sequence encoded by a human germline immunoglobulin gene and contains amino acid residues that identify the antibody as being derived from human sequences when compared to the germline immunoglobulin amino acid sequences of other species (e.g., murine germline sequences).
  • a humanized antibody may be at least 95%, 96%, 97%, 98%, or 99%, or even at least 96%, 97%, 98%, or 99% identical in amino acid sequence to the amino acid sequence encoded by the germline immunoglobulin gene.
  • a humanized antibody derived from a particular human germline sequence will display no more than 10-20 amino acid differences from the amino acid sequence encoded by the human germline immunoglobulin gene (prior to the introduction of any skew, pI and ablation variants herein; that is, the number of variants is generally low, prior to the introduction of the variants of the invention).
  • the humanized antibody may display no more than 5, or even no more than 4, 3, 2, or 1 amino acid difference from the amino acid sequence encoded by the germline immunoglobulin gene (again, prior to the introduction of any skew, p1 and ablation variants herein; that is, the number of variants is generally low, prior to the introduction of the variants of the invention).
  • the parent antibody has been affinity matured, as is known in the art.
  • Structure-based methods may be employed for humanization and affinity maturation, for example as described in U.S. Ser. No. 11/004,590.
  • Selection based methods may be employed to humanize and/or affinity mature antibody variable regions, including but not limited to methods described in Wu et al., 1999, J. Mol. Biol. 294:151-162; Baca et al., 1997, J. Biol. Chem. 272(16):10678-10684; Rosok et al., 1996, J. Biol. Chem. 271(37): 22611-22618; Rader et al., 1998, Proc. Natl. Acad. Sci.
  • the present invention relates to heterodimeric Fc fusion proteins that include IL-15 and IL-15 receptor alpha (IL-15R ⁇ ) protein domains in different orientations.
  • the Fc domains can be derived from IgG Fc domains, e.g., IgG1, IgG2, IgG3 or IgG4 Fc domains, with IgG1 Fc domains finding particular use in the invention.
  • each chain defines a constant region primarily responsible for effector function.
  • Kabat et al. collected numerous primary sequences of the variable regions of heavy chains and light chains. Based on the degree of conservation of the sequences, they classified individual primary sequences into the CDRs and the framework and made a list thereof (see SEQUENCES OF IMMUNOLOGICAL INTEREST, 5th edition, NIH publication, No. 91-3242, E. A. Kabat et al., entirely incorporated by reference).
  • the Kabat numbering system is generally used when referring to a residue in the variable domain (approximately, residues 1-107 of the light chain variable region and residues 1-113 of the heavy chain variable region) and the EU numbering system for Fc regions (e.g., Kabat et al., supra (1991)).
  • immunoglobulin domains in the heavy chain.
  • immunoglobulin (Ig) domain herein is meant a region of an immunoglobulin having a distinct tertiary structure.
  • the heavy chain domains including, the constant heavy (CH) domains and the hinge domains.
  • the IgG isotypes each have three CH regions. Accordingly, “CH” domains in the context of IgG are as follows: “CH1” refers to positions 118-215 according to the EU index as in Kabat. “Hinge” refers to positions 216-230 according to the EU index as in Kabat.
  • CH2 refers to positions 231-340 according to the EU index as in Kabat
  • CH3 refers to positions 341-447 according to the EU index as in Kabat.
  • Table 1 the exact numbering and placement of the heavy chain domains can be different among different numbering systems.
  • the pI variants can be in one or more of the CH regions, as well as the hinge region, discussed below.
  • Ig domain of the heavy chain is the hinge region.
  • hinge region or “hinge region” or “antibody hinge region” or “immunoglobulin hinge region” herein is meant the flexible polypeptide comprising the amino acids between the first and second heavy chain constant domains of an antibody.
  • the IgG CH1 domain ends at EU position 215, and the IgG CH2 domain begins at residue EU position 237.
  • the antibody hinge is herein defined to include positions 216 (E216 in IgG1) to 230 (P230 in IgG1), wherein the numbering is according to the EU index as in Kabat.
  • the hinge is included, generally referring to positions 216-230.
  • pI variants can be made in the hinge region as well.
  • the present invention provides different antibody domains.
  • the heterodimeric proteins of the invention comprise different domains, which can be overlapping as well. These domains include, but are not limited to, the Fc domain, the CH1 domain, the CH2 domain, the CH3 domain, the hinge domain, and the heavy constant domain (CH1-hinge-Fc domain or CH1-hinge-CH2-CH3).
  • the “Fc domain” includes the —CH2-CH3 domain, and optionally a hinge domain. In some embodiments, the Fc domain also includes a portion of the CH1 domain.
  • a protein fragment e.g., IL-15 or IL-15R ⁇ is attached to an Fc domain
  • it is the C-terminus of the IL-15 or IL-15R ⁇ construct that is attached to all or part of the hinge of the Fc domain; for example, it is generally attached to the sequence EPKSC (SEQ ID NO: 293) which is the beginning of the hinge.
  • a protein fragment, e.g., IL-15 or IL-15R ⁇ when a protein fragment, e.g., IL-15 or IL-15R ⁇ is attached to an Fc domain, it is the N-terminus of the protein fragment that is attached to the C-terminus of the CH3 domain.
  • the C-terminus of the IL-15 or IL-15R ⁇ protein fragment is attached to the N-terminus of a domain linker, the C-terminus of which is attached to the N-terminus of a constant Fc domain (N-IL-15 or IL-15R ⁇ protein fragment-linker-Fc domain-C) although that can be switched (N-Fc domain-linker-IL-15 or IL-15R ⁇ protein fragment-C).
  • C-terminus of a first protein fragment is attached to the N-terminus of a second protein fragment, optionally via a domain linker
  • the C-terminus of the second protein fragment is attached to the N-terminus of a constant Fc domain, optionally via a domain linker.
  • a constant Fc domain that is not attached to a first protein fragment or a second protein fragment is provided.
  • a heterodimer Fc fusion protein can contain two or more of the exemplary monomeric Fc domain proteins described herein.
  • the N-terminus of a first protein fragment is attached to the C-terminus of a second protein fragment, optionally via a domain linker
  • the N-terminus of the second protein fragment is attached to the C-terminus of a constant Fc domain, optionally via a domain linker.
  • the linker is a “domain linker”, used to link any two domains as outlined herein together, some of which are depicted in FIG. 87 . While any suitable linker can be used, many embodiments utilize a glycine-serine polymer, including for example (GS)n (SEQ ID NO: 19), (GSGGS)n (SEQ ID NO: 20), (GGGGS)n (SEQ ID NO: 21), and (GGGS)n (SEQ ID NO: 22), where n is an integer of at least one (and generally from 0 to 1 to 2 to 3 to 4 to 5) as well as any peptide sequence that allows for recombinant attachment of the two domains with sufficient length and flexibility to allow each domain to retain its biological function. In some cases, and with attention being paid to “strandedness”, as outlined below, the linker is a charged domain linker.
  • the present invention provides heterodimeric Fc fusion proteins that rely on the use of two different heavy chain variant Fc sequences, that will self-assemble to form a heterodimeric Fc domain fusion polypeptide.
  • heterodimeric Fc fusion proteins contain at least two constant domains which can be engineered to produce heterodimers, such as p1 engineering.
  • Other Fc domains that can be used include fragments that contain one or more of the CH1, CH2, CH3, and hinge domains of the invention that have been pI engineered.
  • the formats depicted in FIGS. 9A-9G, and 39A-39D are heterodimeric Fc fusion proteins, meaning that the protein has two associated Fc sequences self-assembled into a heterodimeric Fc domain and at least one protein fragment (e.g., 1, 2 or more protein fragments).
  • a first protein fragment is linked to a first Fc sequence and a second protein fragment is linked to a second Fc sequence.
  • a first protein fragment is linked to a first Fc sequence, and the first protein fragment is non-covalently attached to a second protein fragment that is not linked to an Fc sequence.
  • the heterodimeric Fc fusion protein contains a first protein fragment linked to a second protein fragment which is linked to a first Fc sequence, and a second Fc sequence that is not linked to either the first or second protein fragments.
  • the present invention is directed to novel constructs to provide heterodimeric Fc fusion proteins that allow binding to one or more binding partners, ligands or receptors.
  • the heterodimeric Fc fusion constructs are based on the self-assembling nature of the two Fc domains of the heavy chains of antibodies, e.g., two “monomers” that assemble into a “dimer”.
  • Heterodimeric Fc fusions are made by altering the amino acid sequence of each monomer as more fully discussed below.
  • the present invention is generally directed to the creation of heterodimeric Fc fusion proteins which can co-engage binding partner(s) or ligand(s) or receptor(s) in several ways, relying on amino acid variants in the constant regions that are different on each chain to promote heterodimeric formation and/or allow for ease of purification of heterodimers over the homodimers.
  • heterodimerization variants amino acid variants that lead to the production of heterodimers are referred to as “heterodimerization variants”.
  • heterodimerization variants can include steric variants (e.g. the “knobs and holes” or “skew” variants described below and the “charge pairs” variants described below) as well as “p1 variants”, which allows purification of homodimers away from heterodimers.
  • heterodimerization variants useful mechanisms for heterodimerization include “knobs and holes” (“KIH”; sometimes described herein as “skew” variants (see discussion in WO2014/145806), “electrostatic steering” or “charge pairs” as described in WO2014/145806, pI variants as described in WO2014/145806, and general additional Fc variants as outlined in WO2014/145806 and below.
  • embodiments of particular use in the present invention rely on sets of variants that include skew variants, that encourage heterodimerization formation over homodimerization formation, coupled with pI variants, which increase the pI difference between the two monomers and each dimeric species.
  • pI variants can be either contained within the constant and/or Fc domains of a monomer, or domain linkers can be used. That is, the invention provides pI variants that are on one or both of the monomers, and/or charged domain linkers as well.
  • additional amino acid engineering for alternative functionalities may also confer pI changes, such as Fc, FcRn and KO variants.
  • amino acid variants can be introduced into one or both of the monomer polypeptides; that is, the pI of one of the monomers (referred to herein for simplicity as “monomer A”) can be engineered away from monomer B, or both monomer A and B can be changed, with the pI of monomer A increasing and the pI of monomer B decreasing.
  • the pI changes of either or both monomers can be done by removing or adding a charged residue (e.g., a neutral amino acid is replaced by a positively or negatively charged amino acid residue, e.g., glutamine to glutamic acid), changing a charged residue from positive or negative to the opposite charge (e.g., aspartic acid to lysine) or changing a charged residue to a neutral residue (e.g., loss of a charge; lysine to serine.).
  • a charged residue e.g., a neutral amino acid is replaced by a positively or negatively charged amino acid residue, e.g., glutamine to glutamic acid
  • changing a charged residue from positive or negative to the opposite charge e.g., aspartic acid to lysine
  • changing a charged residue to a neutral residue e.g., loss of a charge; lysine to serine.
  • this embodiment of the present invention provides for creating a sufficient change in pI in at least one of the monomers such that heterodimers can be separated from homodimers.
  • this can be done by using a “wild type” heavy chain constant region and a variant region that has been engineered to either increase or decrease its pI (wt A:B+ or wt A:B ⁇ ), or by increasing one region and decreasing the other region (A+:B ⁇ or A ⁇ :B+).
  • a component of some embodiments of the present invention are amino acid variants in the constant regions that are directed to altering the isoelectric point (pI) of at least one, if not both, of the monomers of a dimeric protein by incorporating amino acid substitutions (“pI variants” or “pI substitutions”) into one or both of the monomers.
  • pI variants amino acid substitutions
  • pI substitutions amino acid substitutions
  • the number of pI variants to be included on each or both monomer(s) to get good separation will depend in part on the starting pI of the components. That is, to determine which monomer to engineer or in which “direction” (e.g., more positive or more negative), the sequences of the Fc domains, and in some cases, the protein domain(s) linked to the Fc domain are calculated and a decision is made from there. As is known in the art, different Fc domains and/or protein domains will have different starting pIs which are exploited in the present invention. In general, as outlined herein, the pIs are engineered to result in a total pI difference of each monomer of at least about 0.1 logs, with 0.2 to 0.5 being preferred as outlined herein.
  • heterodimers can be separated from homodimers on the basis of size. As shown in the Figures, for example, several of the formats allow separation of heterodimers and homodimers on the basis of size.
  • heterodimerization variants including skew and purification heterodimerization variants
  • the possibility of immunogenicity resulting from the pI variants is significantly reduced by importing pI variants from different IgG isotypes such that pI is changed without introducing significant immunogenicity.
  • an additional problem to be solved is the elucidation of low pI constant domains with high human sequence content, e.g. the minimization or avoidance of non-human residues at any particular position.
  • a side benefit that can occur with this pI engineering is also the extension of serum half-life and increased FcRn binding. That is, as described in U.S. Ser. No. 13/194,904 (incorporated by reference in its entirety), lowering the pI of antibody constant domains (including those found in antibodies and Fc fusions) can lead to longer serum retention in vivo. These pI variants for increased serum half-life also facilitate pI changes for purification.
  • the pI variants of the heterodimerization variants give an additional benefit for the analytics and quality control process of Fc fusion proteins, as the ability to either eliminate, minimize and distinguish when homodimers are present is significant. Similarly, the ability to reliably test the reproducibility of the heterodimeric Fc fusion protein production is important.
  • the present invention provides heterodimeric proteins, including heterodimeric Fc fusion proteins in a variety of formats, which utilize heterodimeric variants to allow for heterodimer formation and/or purification away from homodimers.
  • the heterodimeric fusion constructs are based on the self-assembling nature of the two Fc domains, e.g., two “monomers” that assemble into a “dimer”.
  • these sets do not necessarily behave as “knobs in holes” variants, with a one-to-one correspondence between a residue on one monomer and a residue on the other; that is, these pairs of sets form an interface between the two monomers that encourages heterodimer formation and discourages homodimer formation, allowing the percentage of heterodimers that spontaneously form under biological conditions to be over 90%, rather than the expected 50% (25% homodimer A/A:50% heterodimer A/B:25% homodimer B/B).
  • the formation of heterodimers can be facilitated by the addition of steric variants. That is, by changing amino acids in each heavy chain, different heavy chains are more likely to associate to form the heterodimeric structure than to form homodimers with the same Fc amino acid sequences.
  • Suitable steric variants are included in the FIG. 29 of U.S. Ser. No. 15/141,350, all of which is hereby incorporated by reference in its entirety, as well as in FIG. 84 .
  • knocks and holes referring to amino acid engineering that creates steric influences to favor heterodimeric formation and disfavor homodimeric formation, as described in U.S. Ser. No. 61/596,846, Ridgway et al., Protein Engineering 9(7):617 (1996); Atwell et al., J. Mol. Biol. 1997 270:26; U.S. Pat. No. 8,216,805, all of which are hereby incorporated by reference in their entirety.
  • the Figures identify a number of “monomer A-monomer B” pairs that rely on “knobs and holes”.
  • these “knobs and hole” mutations can be combined with disulfide bonds to skew formation to heterodimerization.
  • electrostatic steering An additional mechanism that finds use in the generation of heterodimers is sometimes referred to as “electrostatic steering” as described in Gunasekaran et al., J. Biol. Chem. 285(25):19637 (2010), hereby incorporated by reference in its entirety. This is sometimes referred to herein as “charge pairs”.
  • electrostatics are used to skew the formation towards heterodimerization. As those in the art will appreciate, these may also have an effect on pI, and thus on purification, and thus could in some cases also be considered pI variants. However, as these were generated to force heterodimerization and were not used as purification tools, they are classified as “steric variants”.
  • D221E/P228E/L368E paired with D221R/P228R/K409R e.g., these are “monomer corresponding sets”
  • C220E/P228E/368E paired with C220R/E224R/P228R/K409R e.g., these are “monomer corresponding sets”
  • Additional monomer A and monomer B variants can be combined with other variants, optionally and independently in any amount, such as pI variants outlined herein or other steric variants that are shown in FIG. 37 of US 2012/0149876, all of which are incorporated expressly by reference herein.
  • the steric variants outlined herein can be optionally and independently incorporated with any pI variant (or other variants such as Fc variants, FcRn variants, etc.) into one or both monomers, and can be independently and optionally included or excluded from the proteins of the invention.
  • FIGS. 3A-3E A list of suitable skew variants is found in FIGS. 3A-3E .
  • the pairs of sets including, but not limited to, S364K/E357Q:L368D/K370S; L368D/K370S:S364K; L368E/K370S:S364K; T411E/K360E/Q362E:D401K; L368D/K370S: S364K/E357L; K370S:S364K/E357Q; and T366S/L368A/Y407V:T366W (optionally including a bridging disulfide, T366S/L368A/Y407V/Y349C:T366W/S354C).
  • the pair “S364K/E357Q:L368D/K370S” means that one of the monomers has the double variant set S364K/E357Q and the other has the double variant set L368D/K370S; as above, the “strandedness” of these pairs depends on the starting pI.
  • pI variants those that increase the pI of the protein (basic changes) and those that decrease the pI of the protein (acidic changes).
  • basic changes those that increase the pI of the protein
  • acidic changes those that decrease the pI of the protein
  • all combinations of these variants can be used: one monomer may be wild type, or a variant that does not display a significantly different pI from wild-type, and the other can be either more basic or more acidic. Alternatively, each monomer may be changed, one to more basic and one to more acidic.
  • FIG. 30 Preferred combinations of pI variants are shown in FIG. 30 of U.S. Ser. No. 15/141,350, all of which are herein incorporated by reference in its entirety. As outlined herein and shown in the figures, these changes are shown relative to IgG1, but all isotypes can be altered this way, as well as isotype hybrids. In the case where the heavy chain constant domain is from IgG2-4, R133E and R133Q can also be used. pI variants are depicted in FIG. 4 .
  • a preferred combination of pI variants has one monomer comprising 208D/295E/384D/418E/421D variants (N208D/Q295E/N384D/Q418E/N421D when relative to human IgG1) if one of the Fc monomers includes a CH1 domain.
  • the second monomer comprising a positively charged domain linker, including (GKPGS) 4 (SEQ ID NO: 294).
  • the first monomer includes a CH1 domain, including position 208.
  • a preferred negative pI variant Fc set includes 295E/384D/418E/421D variants (Q295E/N384D/Q418E/N421D when relative to human IgG1).
  • mutations are made in the hinge domain of the Fc domain, including positions 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, and 230.
  • pI mutations and particularly substitutions can be made in one or more of positions 216-230, with 1, 2, 3, 4 or 5 mutations finding use in the present invention. Again, all possible combinations are contemplated, alone or with other pI variants in other domains.
  • substitutions that find use in lowering the pI of hinge domains include, but are not limited to, a deletion at position 221, a non-native valine or threonine at position 222, a deletion at position 223, a non-native glutamic acid at position 224, a deletion at position 225, a deletion at position 235 and a deletion or a non-native alanine at position 236.
  • pI substitutions are done in the hinge domain, and in others, these substitution(s) are added to other pI variants in other domains in any combination.
  • mutations can be made in the CH2 region, including positions 233, 234, 235, 236, 274, 296, 300, 309, 320, 322, 326, 327, 334 and 339. Again, all possible combinations of these 14 positions can be made; e.g., a pI antibody may have 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 CH2 pI substitutions.
  • substitutions that find use in lowering the pI of CH2 domains include, but are not limited to, a non-native glutamine or glutamic acid at position 274, a non-native phenylalanine at position 296, a non native phenylalanine at position 300, a non-native valine at position 309, a non-native glutamic acid at position 320, a non-native glutamic acid at position 322, a non-native glutamic acid at position 326, a non-native glycine at position 327, a non-native glutamic acid at position 334, a non native threonine at position 339, and all possible combinations within CH2 and with other domains.
  • the mutations can be independently and optionally selected from position 355, 359, 362, 384, 389, 392, 397, 418, 419, 444 and 447.
  • Specific substitutions that find use in lowering the pI of CH3 domains include, but are not limited to, a non native glutamine or glutamic acid at position 355, a non-native serine at position 384, a non-native asparagine or glutamic acid at position 392, a non-native methionine at position 397, a non native glutamic acid at position 419, a non native glutamic acid at position 359, a non native glutamic acid at position 362, a non native glutamic acid at position 389, a non native glutamic acid at position 418, a non native glutamic acid at position 444, and a deletion or non-native aspartic acid at position 447.
  • IgG1 is a common isotype for therapeutic antibodies for a variety of reasons, including high effector function.
  • the heavy constant region of IgG1 has a higher pI than that of IgG2 (8.10 versus 7.31).
  • IgG2 residues at particular positions into the IgG1 backbone By introducing IgG2 residues at particular positions into the IgG1 backbone, the pI of the resulting monomer is lowered (or increased) and additionally exhibits longer serum half-life.
  • IgG1 has a glycine (pI 5.97) at position 137
  • IgG2 has a glutamic acid (pI 3.22); importing the glutamic acid will affect the pI of the resulting protein.
  • pI 3.22 glutamic acid
  • a number of amino acid substitutions are generally required to significantly affect the pI of the variant Fc fusion protein.
  • even changes in IgG2 molecules allow for increased serum half-life.
  • non-isotypic amino acid changes are made, either to reduce the overall charge state of the resulting protein (e.g., by changing a higher pI amino acid to a lower pI amino acid), or to allow accommodations in structure for stability, etc. as is more further described below.
  • the pI of each monomer can depend on the pI of the variant heavy chain constant domain and the pI of the total monomer, including the variant heavy chain constant domain and the fusion partner.
  • the change in pI is calculated on the basis of the variant heavy chain constant domain, using the chart in the FIG. 19 of US Publ. App. No. 2014/0370013.
  • which monomer to engineer is generally decided by the inherent pI of each monomer.
  • the pI variant decreases the pI of the monomer, they can have the added benefit of improving serum retention in vivo.
  • Fc amino acid modification In addition to pI amino acid variants, there are a number of useful Fc amino acid modification that can be made for a variety of reasons, including, but not limited to, altering binding to one or more Fc ⁇ Rs, altered binding to FcRn, etc.
  • the proteins of the invention can include amino acid modifications, including the heterodimerization variants outlined herein, which includes the pI variants and steric variants.
  • Each set of variants can be independently and optionally included or excluded from any particular heterodimeric protein.
  • Fc substitutions that can be made to alter binding to one or more of the Fc ⁇ Rs.
  • Substitutions that result in increased binding as well as decreased binding can be useful.
  • ADCC antibody dependent cell-mediated cytotoxicity; the cell-mediated reaction wherein nonspecific cytotoxic cells that express Fc ⁇ Rs recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • Fc ⁇ RIIb an inhibitory receptor
  • Amino acid substitutions that find use in the present invention include those listed in U.S. Ser. No. 11/124,620 (particularly FIG. 41), Ser. Nos.
  • variants that find use include, but are not limited to, 236A, 239D, 239E, 332E, 332D, 239D/332E, 267D, 267E, 328F, 267E/328F, 236A/332E, 239D/332E/330Y, 239D, 332E/330L, 243A, 243L, 264A, 264V and 299T.
  • amino acid substitutions that increase affinity for Fc ⁇ RIIc can also be included in the Fc domain variants outlined herein.
  • the substitutions described in, for example, U.S. Ser. Nos. 11/124,620 and 14/578,305 are useful.
  • Fc substitutions that find use in increased binding to the FcRn and increased serum half-life, as specifically disclosed in U.S. Ser. No. 12/341,769, hereby incorporated by reference in its entirety, including, but not limited to, 434S, 434A, 428L, 308F, 259I, 428L/434S, 259I/308F, 436I/428L, 436I or V/434S, 436V/428L and 259I/308F/428L.
  • Fc ⁇ R ablation variants or “Fc knock out (FcKO or KO)” variants.
  • Fc ⁇ R ablation variants or “Fc knock out (FcKO or KO)” variants.
  • Fc ⁇ R ablation variants for some therapeutic applications, it is desirable to reduce or remove the normal binding of the Fc domain to one or more or all of the Fc ⁇ receptors (e.g., Fc ⁇ R1, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ RIIIa, etc.) to avoid additional mechanisms of action. That is, for example, in many embodiments, particularly in the use of immunomodulatory proteins, it is desirable to ablate Fc ⁇ RIIIa binding to eliminate or significantly reduce ADCC activity such that one of the Fc domains comprises one or more Fc ⁇ receptor ablation variants.
  • Fc ⁇ R ablation variants for some therapeutic applications, it is desirable to reduce or remove the normal binding of the Fc domain to one or more or all of the Fc ⁇ receptors (e.g., Fc
  • ablation variants are depicted in FIG. 31 of U.S. Ser. No. 15/141,350, all of which are herein incorporated by reference in its entirety, and each can be independently and optionally included or excluded, with preferred aspects utilizing ablation variants selected from the group consisting of G236R/L328R, E233P/L234V/L235A/G236del/S239K, E233P/L234V/L235A/G236del/S267K, E233P/L234V/L235A/G236del/S239K/A327G, E233P/L234V/L235A/G236del/S267K/A327G and E233P/L234V/L235A/G236del, according to the EU index.
  • ablation variants referenced herein ablate Fc ⁇ R binding but generally not FcRn binding.
  • Useful ablation variants that ablate Fc ⁇ R binding are depicted in FIG. 5 .
  • heterodimerization variants including skew and/or pI variants
  • skew and/or pI variants can be optionally and independently combined in any way, as long as they retain their “strandedness” or “monomer partition”.
  • all of these variants can be combined into any of the heterodimerization formats.
  • any of the heterodimerization variants, skew and pI may also be independently and optionally combined with Fc ablation variants, Fc variants, FcRn variants, as generally outlined herein.
  • a monomeric Fc domain can comprise a set of amino acid substitutions that includes C220S/S267K/L368D/K370S or C220S/S267K/S364K/E357Q.
  • heterodimeric Fc fusion proteins can comprise skew variants (e.g., a set of amino acid substitutions as shown in FIGS. 1A-1C of U.S. Ser. No. 15/141,350, all of which are herein incorporated by reference in its entirety), with particularly useful skew variants being selected from the group consisting of S364K/E357Q:L368D/K370S; L368D/K370S:S364K; L368E/K370S:S364K; T411E/K360E/Q362E:D401K; L368D/K370S:S364K/E357L, K370S:S364K/E357Q, T366S/L368A/Y407V:T366W; and T366S/L368A/Y407V:T366W (optionally including a bridging disulfide, T366S/L368A/Y407V
  • the Fc domain comprises one or more amino acid substitutions selected from the group consisting of: 236R, S239D, S239E, F243L, M252Y, V259I, S267D, S267E, S67K, S298A, V308F, L328F, L328R, 330L, I332D, I332E, M428L, N434A, N434S, 236R/L328R, S239D/I332E, 236R/L328F, V259I/V308F, S267E/L328F, M428L/N43S, Y436I/M428L, N436V/M428L, V436I/N434S, Y436V/N434S, S239D/I332E/330L, M252Y/S54T/T256E, V259I/V308F/M428L, E233P/
  • a particular combination of skew and pI variants that finds use in the present invention is T366S/L368A/Y407V:T366W (optionally including a bridging disulfide, T366S/L368A/Y407V/Y349C:T366W/S354C) with one monomer comprising Q295E/N384D/Q418E/N481D and the other a positively charged domain linker.
  • the “knobs in holes” variants do not change pI, and thus can be used on either monomer.
  • the Fc domain comprises amino acid substitutions comprising M428L/N434S.
  • each Fc domain of the untargeted IL-15/R ⁇ heterodimeric Fc fusion proteins comprises amino acid substitutions comprising M428L/N434S.
  • FIGS. 6A-6E Useful embodiments of the Fc domains of the heterodimeric Fc fusion proteins containing IL-15 and IL-15R ⁇ proteins described herein are provided in FIGS. 6A-6E .
  • the present invention provides heterodimeric Fc fusion proteins containing IL-15 and IL-15R ⁇ proteins.
  • the IL-15 complex can take several forms.
  • the IL-15 protein on its own is less stable than when complexed with the IL-15R ⁇ protein.
  • the IL-15R ⁇ protein contains a “sushi domain”, which is the shortest region of the receptor that retains IL-15 binding activity.
  • preferred embodiments herein include complexes that just use the sushi domain, the sequence of which is shown in the figures.
  • the IL-15 complexes generally comprises the IL-15 protein and the sushi domain of IL-15R ⁇ (unless otherwise noted that the full length sequence is used, “IL-15R ⁇ ”, “IL-15R ⁇ (sushi)” and “sushi” are used interchangeably throughout).
  • This complex can be used in three different formats. As shown in FIGS. 9A, 9C, 9D, and 9F , the IL-15 protein and the IL-15R ⁇ (sushi) are not covalently attached, but rather are self-assembled through regular ligand-ligand interactions. As is more fully described herein, it can be either the IL-15 domain or the sushi domain that is covalently linked to the Fc domain (generally using an optional domain linker).
  • each of the IL-15 and sushi domains can be engineered to contain a cysteine amino acid, that forms a disulfide bond to form the complex as is generally shown in FIGS. 39A-39D , again, with either the IL-15 domain or the sushi domain being covalently attached (using an optional domain linker) to the Fc domain.
  • the human IL-15 protein has the amino acid sequence set forth in NCBI Ref. Seq. No. NP_000576.1 or SEQ ID NO:1. In some cases, the coding sequence of human IL-15 is set forth in NCBI Ref. Seq. No. NM_000585.
  • An exemplary IL-15 protein of the Fc fusion heterodimeric protein outlined herein can have the amino acid sequence of SEQ ID NO:2 (mature IL-15) or amino acids 49-162 of SEQ ID NO:1. In some embodiments, the IL-15 protein has at least 90%, e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity to SEQ ID NO:2.
  • the IL-15 protein has the amino acid sequence of SEQ ID NO:2 and one or more amino acid substitutions selected from the group consisting of C42S, L45C, Q48C, V49C, L52C, E53C, E87C, and E89C.
  • the IL-15 protein of the Fc fusion protein can have 1, 2, 3, 4, 5, 6, 7, 8 or 9 amino acid substitutions.
  • the amino acid substitution(s) may be isosteric substitutions at the IL-15:IL-2P and IL-15:common gamma chain interface.
  • the human IL-15 protein such as the human mature IL-15 protein has one or more amino acid substitutions selected from the group consisting of N1D, N4D, D8N, D30N, D61N, E64Q, N65D, Q108E, and any combination thereof.
  • the human IL-15 protein of the Fc fusion protein has the amino acid sequence of SEQ ID NO:2 and amino acid substitutions N4D/N65D.
  • the human IL-15 protein of the Fc fusion protein has at least 97% or 98% sequence identity to SEQ ID NO:2 including N4D/N65D substitutions. In some embodiments, the human IL-15 protein of the Fc fusion protein has the amino acid sequence of SEQ ID NO:2 and amino acid substitutions D30N/N65D. In some cases, the human IL-15 protein of the Fc fusion protein has at least 97% or 98% sequence identity to SEQ ID NO:2 including D30N/N65D substitutions. In some embodiments, the human IL-15 protein of the Fc fusion protein has the amino acid sequence of SEQ ID NO:2 and amino acid substitutions D30N/E64Q/N65D. In some cases, the human IL-15 protein of the Fc fusion protein has at least 96% or 97% sequence identity to SEQ ID NO:2 including D30N/E64Q/N65D substitutions.
  • the human IL-15 protein such as a human mature IL-15 protein of the Fc fusion protein is identical to the amino acid sequence of SEQ ID NO:2. In some cases, the human IL-15 protein such as the human mature IL-15 protein has no amino acid substitutions.
  • the human mature IL-15 variant protein has one or more amino acid mutations (e.g., substitutions, insertions and/or deletions).
  • the mutation introduces a cysteine residue that can form a disulfide bond with human IL-15 receptor alpha (IL-15R ⁇ ) protein.
  • the human IL-15 receptor alpha (IL-15R ⁇ ) protein has the amino acid sequence set forth in NCBI Ref. Seq. No. NP_002180.1 or SEQ ID NO:3.
  • the coding sequence of human IL-15R ⁇ is set forth in NCBI Ref. Seq. No. NM_002189.3.
  • An exemplary IL-15R ⁇ protein of the Fc fusion heterodimeric protein outlined herein can comprise or consist of the sushi domain of SEQ ID NO:3 (e.g., amino acids 31-95 of SEQ ID NO:3), or in other words, the amino acid sequence of SEQ ID NO:4.
  • the IL-15R ⁇ protein has the amino acid sequence of SEQ ID NO:4 and an amino acid insertion selected from the group consisting of D96, P97, A98, D96/P97, D96/C97, D96/P97/A98, D96/P97/C98, and D96/C97/A98, wherein the amino acid position is relative to full-length human IL-15R ⁇ protein or SEQ ID NO:3.
  • amino acid(s) such as D (e.g., Asp), P (e.g., Pro), A (e.g., Ala), DP (e.g., Asp-Pro), DC (e.g., Asp-Cys), DPA (e.g., Asp-Pro-Ala), DPC (e.g., Asp-Pro-Cys), or DCA (e.g., Asp-Cys-Ala)
  • D e.g., Asp
  • P e.g., Pro
  • A e.g., Ala
  • DP e.g., Asp-Pro
  • DC e.g., Asp-Cys
  • DPA e.g., Asp-Pro-Ala
  • DPC e.g., Asp-Pro-Cys
  • DCA e.g., Asp-Cys-Ala
  • the IL-15R ⁇ protein has the amino acid sequence of SEQ ID NO:4 and one or more amino acid substitutions selected from the group consisting of K34C, A37C, G38C, 540C, and L42C, wherein the amino acid position is relative to SEQ ID NO:4.
  • the IL-15R ⁇ (sushi) protein of SEQ ID NO:4 can have 1, 2, 3, 4, 5, 6, 7, 8 or more amino acid mutations (e.g., substitutions, insertions and/or deletions).
  • SEQ ID NO: 1 is MRISKPHLRSISIQCYLCLLLNSHFLTEAGIHVFILGCFSAGLPKTEA NWVNVISDLKKIEDLIQSMHIDATLYTESDVHPSCKVTAMKCFLLELQ VISLESGDASIHDTVENLIILANNSLSSNGNVTESGCKECEELEEKNI KEFLQSFVHIVQMFINTS.
  • SEQ ID NO: 2 is NWVNVISDLKKIEDLIQSMHIDATLYTESDVHPSCKVTAMKCFLLELQ VISLESGDASIHDTVENLIILANNSLSSNGNVTESGCKECEELEEKNI KEFLQSFVHIVQMFINTS.
  • SEQ ID NO: 3 is MAPRRARGCRTLGLPALLLLLLLRPPATRGITCPPPMSVEHADIWVKS YSLYSRERYICNSGFKRKAGTSSLTECVLNKATNVAHWTTPSLKCIRD PALVHQRPAPPSTVTTAGVTPQPESLSPSGKEPAASSPSSNNTAATTA AIVPGSQLMPSKSPSTGTTEISSHESSHGTPSQTTAKNWELTASASHQ PPGVYPQGHSDTTVAISTSTVLLCGLSAVSLLACYLKSRQTPPLASVE MEAMEALPVTWGTSSRDEDLENCSHHL.
  • SEQ ID NO: 5 is ITCPPPMSVEHADIWVKSYSLYSRERYICNSGFKRKAGTSSLTECVLN KATNVAHWTTPSLKCIR.
  • the invention provides proteins comprising a human IL-15 variant with the amino acid variant D30N.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and a D30N substitution.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and at least a D30N substitution.
  • the invention provides proteins comprising a human IL-15 variant with the amino acid variant N1D.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and an N1D substitution.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and at least an N1D substitution.
  • the invention provides proteins comprising a human IL-15 variant with the amino acid variant N4D.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and an N4D substitution.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and at least an N4D substitution.
  • the invention provides proteins comprising a human IL-15 variant with the amino acid variant E64Q.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and an E64Q substitution.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and at least an E64Q substitution.
  • the invention provides proteins comprising a human IL-15 variant with the amino acid variant N65D.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and an N65D substitution.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and at least an N65D substitution.
  • the invention provides proteins comprising a human IL-15 variant with amino acid substitutions N1D/D30N.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and N1D/D30N substitutions.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and at least N1D/D30N substitutions.
  • the invention provides proteins comprising a human IL-15 variant with amino acid substitutions N4D/D30N.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and N4D/D30N substitutions.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and at least N4D/D30N substitutions.
  • the invention provides proteins comprising a human IL-15 variant with amino acid substitutions D30N/E64Q.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and D30N/E64Q substitutions.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and at least D30N/E64Q substitutions.
  • the invention provides proteins comprising a human IL-15 variant with amino acid substitutions D30N/N65D.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and D30N/N65D substitutions.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and at least D30N/N65D substitutions.
  • the invention provides proteins comprising a human IL-15 variant with amino acid substitutions D30N/E64Q/N65D.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and D30N/E64Q/N65D substitutions.
  • the protein comprises the amino acid sequence of SEQ ID NO:2 and at least D30N/E64Q/N65D substitutions.
  • the IL-15 protein and IL-15R ⁇ protein are attached together via a linker.
  • the proteins are not attached via a linker.
  • the IL-15 protein and IL-15R ⁇ protein are noncovalently attached.
  • the IL-15 protein is attached to an Fc domain via a linker. In certain embodiments, the IL-15 protein is attached to an Fc domain directly, such as without a linker. In particular embodiments, the IL-15 protein is attached to an Fc domain via a hinge region or a fragment thereof. In some embodiments, the IL-15R ⁇ protein is attached to an Fc domain via a linker. In other embodiments, the IL-15R ⁇ protein is attached to an Fc domain directly, such as without a linker. In particular embodiments, the IL-15R ⁇ protein is attached to an Fc domain via a hinge region or a fragment thereof. In some cases, a linker is not used to attach the IL-15 protein or IL-15R ⁇ protein to an Fc domain.
  • the linker is a “domain linker”, used to link any two domains as outlined herein together. While any suitable linker can be used, many embodiments utilize a glycine-serine polymer, including for example (GS)n (SEQ ID NO: 23), (GSGGS)n (SEQ ID NO: 24), (GGGGS)n (SEQ ID NO: 25), and (GGGS)n (SEQ ID NO: 26), where n is an integer of at least 0 (and generally from 0 to 1 to 2 to 3 to 4 to 5) as well as any peptide sequence that allows for recombinant attachment of the two domains with sufficient length and flexibility to allow each domain to retain its biological function.
  • GS glycine-serine polymer
  • useful linkers include (GGGGS) 0 (“GGGGS” disclosed as SEQ ID NO: 27) or (GGGGS) 1 (SEQ ID NO: 27) or (GGGGS) 2 (SEQ ID NO: 28).
  • Useful domain linkers are shown in FIG. 7 .
  • charged domain linkers can be used as discussed herein and shown in FIG. 7 and herein.
  • the heterodimeric fusion proteins of the invention have two functional components: an IL-15/IL-15R ⁇ (sushi) component and an Fc component, both of which can take different forms as outlined herein and both of which can be combined with the other component in any configuration.
  • the first and the second Fc domains can have a set of amino acid substitutions selected from the group consisting of a) S267K/L368D/K370S:S267K/LS364K/E357Q; b) S364K/E357Q:L368D/K370S; c) L368D/K370S:S364K; d) L368E/K370S:S364K; e) T411E/K360E/Q362E:D401K; f) L368D/K370S:S364K/E357L and g) K370S:S364K/E357Q, according to EU numbering.
  • the first Fc domain has L368D/K370S substitutions and the second Fc domain has S364K/E357Q substitutions. In some embodiments, the first Fc domain has S364K/E357Q substitutions and the second Fc domain has L368D/K370S substitutions.
  • the first and/or the second Fc domains have an additional set of amino acid substitutions comprising Q295E/N384D/Q418E/N421D, according to EU numbering.
  • the first and/or the second Fc domains have an additional set of amino acid substitutions consisting of G236R/L328R, E233P/L234V/L235A/G236del/S239K, E233P/L234V/L235A/G236del/S267K, E233P/L234V/L235A/G236del/S239K/A327G, E233P/L234V/L235A/G236del/S267K/A327G and E233P/L234V/L235A/G236del, according to EU numbering.
  • the first and/or second Fc domains have 428L/434S variants for half life extension.
  • the first and/or second Fc domains have M428L/N434S variants for half life extension.
  • the first Fc domain has M428L/N434S substitutions and the second Fc domain has M428L/N434S substitutions
  • the heterodimeric fusion protein comprises two monomers (two Fc monomers).
  • the first monomer comprises (from N- to C-terminus) IL-15-optional domain linker-CH2-CH3, where the domain linker sometimes comprises all or part of the hinge.
  • the second monomer comprises the IL-15/R ⁇ (sushi) domain-optional domain linker-CH2-CH3, where the domain linker sometimes comprises all or part of the hinge.
  • a preferred embodiment utilizes the skew variant pair S364K/E357Q:L368D/K370S.
  • the L368D/K370S variant is on the first monomer and the S354K/E357Q variant is on the second monomer.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 D30N/N65D variant, an IL-15R ⁇ (sushi) domain, and Fc monomers comprising the amino acid substitutions depicted in FIG. 6A .
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 D30N/N65D variant; a human IL-15R ⁇ (sushi) domain; an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 D30N/N65D variant; a human IL-15R ⁇ (sushi) domain, an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N43434
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q: L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 D30N/E64Q/N65D variant, an IL-15R ⁇ (sushi) domain, and Fc monomers comprising the amino acid substitutions depicted in FIG. 6A .
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 D30N/E64Q/N65D variant; a human IL-15R ⁇ (sushi) domain; an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 D30N/E64Q/N65D variant; a human IL-15R ⁇ (sushi) domain, an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L
  • a preferred embodiment utilizes the IL-15 N4D variant.
  • a preferred embodiment utilizes the IL-15 N4D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N4D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N4D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N4D variant, an IL-15R ⁇ (sushi) domain, and Fc monomers comprising the amino acid substitutions depicted in FIG. 6A .
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N4D variant; a human IL-15R ⁇ (sushi) domain; an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N4D variant; a human IL-15R ⁇ (sushi) domain, an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • a preferred embodiment utilizes the IL-15 N65D variant.
  • a preferred embodiment utilizes the IL-15 N65D variant, and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N65D variant, an IL-15R ⁇ (sushi) domain, and Fc monomers comprising the amino acid substitutions depicted in FIG. 6A .
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N65D variant; a human IL-15R ⁇ (sushi) domain; an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N65D variant; a human IL-15R ⁇ (sushi) domain, an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N4D/N65D variant, an IL-15R ⁇ (sushi) domain, and Fc monomers comprising the amino acid substitutions depicted in FIG. 6A .
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N4D/N65D variant; a human IL-15R ⁇ (sushi) domain; an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N4D/N65D variant; a human IL-15R ⁇ (sushi) domain, an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N43434
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N1D/N65D variant, an IL-15R ⁇ (sushi) domain, and Fc monomers comprising the amino acid substitutions depicted in FIG. 6A .
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N1D/N65D variant; a human IL-15R ⁇ (sushi) domain; an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N1D/N65D variant; a human IL-15R ⁇ (sushi) domain, an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N43434
  • a preferred embodiment utilizes the IL-15 Q108E variant.
  • a preferred embodiment utilizes the IL-15 Q108E variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 Q108E variant and the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on both monomers.
  • a preferred embodiment utilizes the IL-15 Q108E variant and the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on both monomers.
  • a preferred embodiment utilizes the IL-15 Q108E variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 Q108E variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 Q108E variant, an IL-15R ⁇ (sushi) domain, and Fc monomers comprising the amino acid substitutions depicted in FIG. 6A .
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 Q108E variant; a human IL-15R ⁇ (sushi) domain; an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 Q108E variant; a human IL-15R ⁇ (sushi) domain, an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • a preferred embodiment utilizes the IL-15 wildtype variant.
  • a preferred embodiment utilizes the IL-15 wildtype variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 wildtype variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 wildtype variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 wildtype variant, an IL-15R ⁇ (sushi) domain, and Fc monomers comprising the amino acid substitutions depicted in FIG. 6A .
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 wildtype variant; a human IL-15R ⁇ (sushi) domain; an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 wildtype variant; a human IL-15R ⁇ (sushi) domain, an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • FIG. 48A XENP22822 including chain 1 (17693) and chain 2 (15908)
  • FIG. 94A XENP23504 including chain 1 and chain 2)
  • FIG. 104AO XENP24045 including chain 1 and chain 2)
  • FIG. 104AQ XENP24306 including chain 1 and chain 2
  • FIG. 48A XENP22821 including chain 1 and chain 2)
  • FIG. 94A XENP23343 including chain 1 and chain 2)
  • FIG. 104AJ XENP23557 including chain 1 and chain 2)
  • FIG. 104AP XENP24113 including chain 1 and chain 2)
  • FIG. 104AP (XENP24051 including chain 1 and chain 2), FIG. 104AR (XENP24341 including chain 1 and chain 2), FIG. 104AP (XENP24052 including chain 1 and chain 2), and FIG. 104AP (XENP24301 including chain 1 and chain 2), all of which are herein incorporated by reference in its entirety.
  • FIG. 10 In the IL-15/R ⁇ -heteroFc format, preferred embodiments are shown in FIG. 10 (XENP22822 including chain 1 (15902) and chain 2 (15908)) and (XENP21475 including chain 1 (16479) and chain 2 (16481)).
  • the heterodimeric fusion protein comprises two monomers.
  • the first monomer comprises (from N- to C-terminus) IL-15/R ⁇ (sushi)-domain linker-IL-15-optional domain linker-CH2-CH3, where the domain linker sometimes comprises all or part of the hinge.
  • the second monomer comprises and “empty” Fc, comprising all or part of the hinge-CH2-CH3. This is referred to as “scIL-15/R ⁇ -Fc” with the “sc” standing for “single chain” (e.g. of the IL-15/R ⁇ sushi complex).
  • a preferred embodiment utilizes the IL-15 D30N/N65D variants.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variants and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S, and on each Fc monomer the 428L/434S variants.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S, and on each Fc monomer the M428L/N434S variants.
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 D30N/N65D variant and an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6B .
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 D30N/N65D variant and an IL-15R ⁇ (sushi) domain; an scIL-15/R ⁇ -Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 D30N/N65D variant and an IL-15R ⁇ (sushi) domain; an scIL-15/R ⁇ -Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an empty-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N43434; and an
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 D30N/E64Q/N65D variant and an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6B .
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 D30N/E64Q/N65D variant and an IL-15R ⁇ (sushi) domain; an scIL-15/R ⁇ -Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and Ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 D30N/E64Q/N65D variant and an IL-15R ⁇ (sushi) domain; an scIL-15/R ⁇ -Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an empty-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L
  • a preferred embodiment utilizes the IL-15 N65D variant.
  • a preferred embodiment utilizes the IL-15 N65D variant, and the skew variant pair S364K/E357Q:L368D/K370S
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N65D variant and an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6B .
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N65D variant and an IL-15R ⁇ (sushi) domain; an scIL-15/R ⁇ -Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N65D variant and an IL-15R ⁇ (sushi) domain; an scIL-15/R ⁇ -Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an empty-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N4D/N65D variant and an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6B .
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N4D/N65D variant and an IL-15R ⁇ (sushi) domain; an scIL-15/R ⁇ -Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N4D/N65D variant and an IL-15R ⁇ (sushi) domain; an scIL-15/R ⁇ -Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an empty-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N43434; and an
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N1D/N65D variant and an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6B .
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N1D/N65D variant and an IL-15R ⁇ (sushi) domain; an scIL-15/R ⁇ -Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the scIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N1D/N65D variant and an IL-15R ⁇ (sushi) domain; an scIL-15/R ⁇ -Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an empty-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N43434; and an
  • preferred embodiments include XENP21478 including chain 1 (16478) and chain 2 (8924) and is shown in FIG. 11 .
  • Other embodiments of this format include XENP21993, XENP21994, XENP21995, XENP23174, XENP23175, XENP24477, and XENP24480 and are shown in WO2018071919 in FIGS. 104G, 104H, 104AG, 104AU, and 104AV and amino acid sequences are found in SEQ ID NOS: 514-518, 519-523, 524-528, 849-853, 1063-1067, and 1078-1082, respectively.
  • the heterodimeric fusion protein comprises three monomers.
  • the first monomer comprises (from N- to C-terminus) IL-15/R ⁇ (sushi)-domain linker-CH2-CH3, where the domain linker sometimes comprises all or part of the hinge.
  • the second monomer comprises and “empty” Fc, comprising all or part of the hinge-CH2-CH3.
  • the third monomer is IL-15. This is referred to as “ncIL-15/R ⁇ -Fc” with the “nc” standing for “non-covalent”).
  • a preferred embodiment utilizes the skew variant pair S364K/E357Q:L368D/K370S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 D30N/E64Q/N65D variant, an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 D30N/E64Q/N65D variant, an IL-15R ⁇ (sushi) domain; an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 D30N/E64Q/N65D variant; an IL-15R ⁇ (sushi) domain; an empty-Fc comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N4D variant.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N4D variant, and the skew variant pair S364K/E357Q:L368D/K370S.
  • n the ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N4D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N4D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 N4D variant, an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 N4D variant, an IL-15R ⁇ (sushi) domain; an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N4D variant; an IL-15R ⁇ (sushi) domain; an empty-Fc comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N65D variant.
  • a preferred embodiment utilizes the IL-15 N65D variant, and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 N65D variant, an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 N65D variant, an IL-15R ⁇ (sushi) domain; an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N65D variant; an IL-15R ⁇ (sushi) domain; an empty-Fc comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N4D/N65D variant.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 N4D/N65D variant, an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 N4D/N65D variant, an IL-15R ⁇ (sushi) domain; an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N4D/N65D variant; an IL-15R ⁇ (sushi) domain; an empty-Fc comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer ablation an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N1D/N65D variant.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N1D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 N1D/N65D variant, an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 N1D/N65D variant, an IL-15R ⁇ (sushi) domain; an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 N1D/N65D variant; an IL-15R ⁇ (sushi) domain; an empty-Fc comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 variant Q108E.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on both monomers.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on both monomers.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 Q108E variant, an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 Q108E variant, an IL-15R ⁇ (sushi) domain; an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 Q108E variant; an IL-15R ⁇ (sushi) domain; an empty-Fc comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 wildtype variant.
  • a preferred embodiment utilizes the IL-15 wildtype variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 wildtype variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 wildtype variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 wildtype variant, an IL-15R ⁇ (sushi) domain; and Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 wildtype variant, an IL-15R ⁇ (sushi) domain; an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a single chain comprising a human mature IL-15 wildtype variant; an IL-15R ⁇ (sushi) domain; an empty-Fc comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • FIG. 104AS XENP24349including chain 1 and chain 2
  • FIG. 104AT XENP24383 including chain 1 and chain 2
  • FIG. 12A and FIG. 12B preferred embodiments are shown in FIG. 12A and FIG. 12B such as XENP21479 including chain 1 (16484) and chain 2 (8793) and chain 3 (16481); XENP22366 including chain 1 (16478) and chain 2 (8924) and chain 3 ( ); and XENP22366 including chain 1 (16484) and chain 2 (8793) and chain 3 (15908); and XENP24348.
  • the homodimeric fusion protein comprises four monomers.
  • the first and second monomers comprise (from N- to C-terminus) IL-15/R ⁇ (sushi)-domain linker-CH2-CH3, where the domain linker sometimes comprises all or part of the hinge.
  • the third and fourth monomers comprise IL-15. This is referred to as “bivalent ncIL-15/R ⁇ -Fc” with the “nc” standing for “non-covalent”).
  • a preferred embodiment utilizes the skew variant pair S364K/E357Q:L368D/K370S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 D30N/N65D variants.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 D30N/N65D variants and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 D30N/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 D30N/N65D variant, an IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 D30N/N65D variants; a first IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 D30N/N65D variants; a first IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N65D variant.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N65D variant, and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 N65D variant, an IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 N65D variants; a first IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 N65D variants; a first IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N4D/N65D variant.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N4D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N1D/N65D variant.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N1D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 N1D/N65D variant, an IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 N1D/N65D variants; a first IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 N1D/N65D variants; a first IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 Q108E variant.
  • ncIL-15/R ⁇ -Fc a preferred embodiment utilizes the IL-15 Q108E variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 Q108E variant and the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on both monomers.
  • a preferred embodiment utilizes the IL-15 Q108E variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 Q108E variant, an IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 Q108E variants; a first IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-Q108E variants; a first IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 wildtype variant.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 wildtype variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 wildtype variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 wildtype variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises a human mature IL-15 wildtype variant, an IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6C .
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 wildtype variants; a first IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent ncIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-wildtype variants; a first IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (sushi) domain and an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15R ⁇ (sushi)-Fc monomer comprising a human IL-15R ⁇ (ablation) domain and an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment is shown in WO2018071919 in FIG. 104E (XENP21979 including chain 1 and chain 2) and in the sequence listing as SEQ ID NOS: 480-483, all of which are herein incorporated by reference in its entirety.
  • ncIL-15/R ⁇ -Fc format preferred embodiments are shown in FIG. 13 such as XENP21978 including chain 1 (17023) and chain 2 (16484).
  • the homodimeric fusion protein comprises two monomers.
  • the first and second monomers comprise (from N- to C-terminus) IL-15/R ⁇ (sushi)-domain linker-IL-15-optional domain linker-CH2-CH3, where the domain linker sometimes comprises all or part of the hinge.
  • This is referred to as “bivalent scIL-15/R ⁇ -Fc” with the “sc” standing for “single chain” (e.g., of the IL-15/R ⁇ sushi complex).
  • a preferred embodiment utilizes the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on both monomers.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 D30N/N65D variants, two IL-15R ⁇ (sushi) domains; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6D .
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 D30N/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 D30N/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 D30N/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 D30N/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 D30N/E64Q/N65D variants, two IL-15R ⁇ (sushi) domains; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6D .
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 D30N/E64Q/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 D30N/E64Q/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 D30N/E64Q/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 D30N/E64Q/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428
  • a preferred embodiment utilizes the IL-15 N65D variant.
  • a preferred embodiment utilizes the IL-15 N65D variant, and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 N65D variants, two IL-15R ⁇ (sushi) domains; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6D .
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 N4D/N65D variants, two IL-15R ⁇ (sushi) domains; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6D .
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N4D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N4D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N4D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N4D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 N1D/N65D variants, two IL-15R ⁇ (sushi) domains; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6D .
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N1D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N1D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N1D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N1D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • a preferred embodiment utilizes the IL-15 Q108E variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 Q108E variant and the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on both monomers.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 Q108E variants, two IL-15R ⁇ (sushi) domains; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6D .
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 Q108E variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 Q108E variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 Q108E variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 Q108E variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • a preferred embodiment utilizes the IL-15 wildtype variant.
  • a preferred embodiment utilizes the IL-15 wildtype variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 wildtype variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 wildtype variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises two human mature IL-15 wildtype variants, two IL-15R ⁇ (sushi) domains; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6D .
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N1D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N1D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the bivalent scIL-15/R ⁇ -Fc fusion protein comprises a first IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N1D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and a second IL-15/R ⁇ (sushi)-Fc monomer comprising a human mature IL-15 N1D/N65D variant linked to a human IL-15R ⁇ (sushi) domain linked to an Fc monomer comprising an amino acid substitution C220S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • FIG. 14 In the bivalent scIL-15/R ⁇ -Fc format, a preferred embodiment is shown in FIG. 14 .
  • the heterodimeric fusion protein comprises three monomers.
  • the first monomer comprises (from N- to C-terminus) CH2-CH3-domain linker-IL-15/R ⁇ (sushi), wherein the Fc comprises all of part of the hinge.
  • the second monomer comprises and “empty” Fc, comprising all or part of the hinge-CH2-CH3.
  • the third monomer is IL-15. This is referred to as “ncIL-15/R ⁇ -Fc” with the “nc” standing for “non-covalent”).
  • ncIL-15/R ⁇ -Fc (or Fc-ncIL-15/R ⁇ ) format
  • a preferred embodiment utilizes the skew variant pair S364K/E357Q:L368D/K370S.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 D30N/N65D variant.
  • ncIL-15/R ⁇ -Fc format a preferred embodiment utilizes the IL-15 D30N/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • Fc-ncIL-15/R ⁇ format a preferred embodiment utilizes the IL-15 D30N/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on both monomers.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on both monomers.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises human mature IL-15 D30N/N65D variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 D30N/N65D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 D30N/N65D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M4
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises human mature IL-15 D30N/E64Q/N65D variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 D30N/E64Q/N65D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 D30N/E64Q/N65D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn
  • a preferred embodiment utilizes the IL-15 N4D variant.
  • a preferred embodiment utilizes the IL-15 N4D variant, and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N4D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N4D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises human mature IL-15 N4D variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 N4D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 N4D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N
  • a preferred embodiment utilizes the IL-15 N65D variant.
  • a preferred embodiment utilizes the IL-15 N65D variant, and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises human mature IL-15 N65D variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 N65D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 N65D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises human mature IL-15 N4D/N65D variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 N4D/N65D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 N4D/N65D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M4
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises human mature IL-15 N1D/N65D variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 N1D/N65D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 N1D/N65D variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M4
  • a preferred embodiment utilizes the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 Q108E variant.
  • a preferred embodiment utilizes the IL-15 Q108E variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 Q108E variant and the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on both monomers.
  • a preferred embodiment utilizes the IL-15 Q108E variant and the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on both monomers.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises human mature IL-15 Q108E variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 Q108E variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 Q108E variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N
  • a preferred embodiment utilizes the IL-15 wildtype variant.
  • a preferred embodiment utilizes the IL-15 wildtype variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 wildtype variant, the skew variant pair S364K/E357Q: L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 wildtype variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises human mature IL-15 wildtype variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 wildtype variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the ncIL-15/R ⁇ -Fc fusion protein comprises a human IL-15 wildtype variant, a Fc-IL-15/R ⁇ (sushi) monomer comprising a human IL-15R ⁇ (sushi) domain linked to to the C-terminus of an Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N
  • preferred embodiments include XENP22637 as chain 1 (17603) and chain 2 (8927) and chain 3 (16484) and is shown in FIG. 15 .
  • the heterodimeric fusion protein comprises two monomers.
  • the first monomer comprises (from N- to C-terminus) CH2-CH3-optional domain linker-IL-15/R ⁇ (sushi)-domain linker-IL-15, wherein the Fc comprises all of part of the hinge.
  • the second monomer comprises and “empty” Fc, comprising all or part of the hinge-CH2-CH3. This is referred to as “Fc-scIL-15/R ⁇ ” or “scIL-15/R ⁇ -Fc” with the “sc” standing for “single chain” (e.g. of the IL-15/R ⁇ sushi complex).
  • the Fc-scIL-15/R ⁇ fusion protein comprises human mature IL-15 variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/
  • a preferred embodiment utilizes the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on both monomers.
  • a preferred embodiment utilizes the IL-15 D30N/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on both monomers.
  • the Fc-scIL-15/R ⁇ fusion protein comprises human mature IL-15 D30N/N65D variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 D30N/N65D variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 D30N/N65D variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 D30N/E64Q/N65D variants, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the Fc-scIL-15/R ⁇ fusion protein comprises human mature IL-15 D30N/E64Q/N65D variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 D30N/E64Q/N65D variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 D30N/E64Q/N65D variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and
  • a preferred embodiment utilizes the IL-15 N65D variant.
  • a preferred embodiment utilizes the IL-15 N65D variant, and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the Fc-scIL-15/R ⁇ fusion protein comprises human mature IL-15 N65D variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 N65D variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 N65D variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M4
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N4D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the Fc-scIL-15/R ⁇ fusion protein comprises human mature IL-15 N4D/N65D variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 N4D/N65D variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 N4D/N65D variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on each Fc monomer.
  • a preferred embodiment utilizes the IL-15 N1D/N65D variant, the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on each Fc monomer.
  • the Fc-scIL-15/R ⁇ fusion protein comprises human mature IL-15 N1D/N65D variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 N1D/N65D variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 N1D/N65D variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn
  • a preferred embodiment utilizes the IL-15 variant Q108E.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on both monomers.
  • a preferred embodiment utilizes the IL-15 variant Q108E and the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on both monomers.
  • the Fc-scIL-15/R ⁇ fusion protein comprises human mature IL-15 Q108E variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 Q108E variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 Q108E variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M4
  • a preferred embodiment utilizes the IL-15 wildtype variant.
  • a preferred embodiment utilizes the IL-15 wildtype variant and the skew variant pair S364K/E357Q:L368D/K370S.
  • a preferred embodiment utilizes the IL-15 wildtype variant and the skew variant pair S364K/E357Q:L368D/K370S and the 428L/434S variants on both monomers.
  • a preferred embodiment utilizes the IL-15 wildtype variant and the skew variant pair S364K/E357Q:L368D/K370S and the M428L/N434S variants on both monomers.
  • the Fc-scIL-15/R ⁇ fusion protein comprises human mature IL-15 wildtype variant, IL-15R ⁇ (sushi) domain; and two Fc monomers comprising the amino acid substitutions depicted in FIG. 6E .
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, and ablation substitutions E233P/L234V/L235A/G236del/S267K, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 wildtype variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the Fc-scIL-15/R ⁇ fusion protein comprises a Fc-IL-15/R ⁇ (sushi) monomer comprising a Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants L368D/K370S, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N343S, such that the Fc domain is linked to a human IL-15R ⁇ (sushi) domain linked to a human mature IL-15 wildtype variant; and an empty-Fc monomer comprising an amino acid substitution C220S, heterodimer pI variants S364K/E357Q, isosteric pI substitutions P217R/P228R/N276K, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428
  • FIG. 16 a preferred embodiment is shown in FIG. 16 .
  • compositions comprising untargeted IL-15/IL-15R ⁇ -Fc fusion proteins in different formats and checkpoint blockade antibodies. Also provided are methods comprising administering untargeted IL-15/IL-15R ⁇ -Fc fusion proteins and checkpoint blockade antibodies to a subject, e.g., a human subject.
  • the checkpoint blockade antibody of the compositions described herein is selected from the group consisting of an anti-PD-1 antibody.
  • the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, or pidilizumab.
  • the untargeted IL-15/IL-15R ⁇ -Fc fusion protein of the compositions described herein is organized in a format described herein, such as an IL-15/IL-15R ⁇ -hetero Fc format, scIL-15/R ⁇ -Fc format, ncIL-15/R ⁇ -Fc format, bivalent ncIL-15/R ⁇ -Fc format, bivalent scIL-15/R ⁇ -Fc format, Fc-ncIL-15/R ⁇ format, or Fc-scIL-15/R ⁇ format.
  • the untargeted IL-15/IL-15R ⁇ -Fc fusion protein has an IL-15/IL-15R ⁇ -hetero Fc format.
  • the untargeted IL-15/IL-15R ⁇ -Fc fusion protein of the composition has an IL-15/IL-15R ⁇ -hetero Fc format and comprises a human mature IL-15 D30N/E64Q/N65D variant; a human IL-15R ⁇ (sushi) domain; an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K; and an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q, and ablation substitutions E233P/L234V/L235A/G236del/S267K.
  • the IL-15/R ⁇ -heteroFc fusion protein comprises a human mature IL-15 N65D variant; a human IL-15R ⁇ (sushi) domain, an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and an IL-15R ⁇ (sushi)-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions S364K/E357Q, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S.
  • the composition or method described herein comprises an anti-PD-1 antibody and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/E64Q/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI0 substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K
  • the composition comprises an anti-PD-1 antibody and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/E64Q/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodi
  • the composition or method described herein comprises nivolumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/E64Q/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K
  • the composition or method described herein comprises nivolumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/E64Q/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C2
  • the composition or method described herein comprises pembrolizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/E64Q/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/
  • the composition comprises pembrolizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/E64Q/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodi
  • the composition or method described herein comprises pidilizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/E64Q/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K
  • the composition or method described herein comprises pidilizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/E64Q/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C2
  • the composition or method described herein comprises an anti-PD-1 antibody and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 N4D/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI0 substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357
  • the composition comprises an anti-PD-1 antibody and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 N4D/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI
  • the composition or method described herein comprises nivolumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 N4D/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357
  • the composition or method described herein comprises nivolumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 N4D/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, hetero
  • the composition or method described herein comprises pembrolizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 N4D/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q
  • the composition or method described herein comprises pembrolizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 N4D/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterod
  • the composition or method described herein comprises pidilizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 N4D/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357
  • the composition or method described herein comprises pidilizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 N4D/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, hetero
  • the composition or method described herein comprises an anti-PD-1 antibody and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI0 substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357
  • the composition or method described herein comprises an anti-PD-1 antibody and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterod
  • the composition or method described herein comprises nivolumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357
  • the composition or method described herein comprises nivolumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, hetero
  • the composition or method described herein comprises pembrolizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357Q
  • the composition or method described herein comprises pembrolizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterod
  • the composition or method described herein comprises pidilizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, and ablation substitutions E233P/L234V/L235A/G236del/S267K; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, heterodimer pI substitutions S364K/E357
  • the composition or method described herein comprises pidilizumab and an untargeted IL-15/IL-15R ⁇ -Fc fusion protein that comprises: (a) first monomer that (from N- to C-terminal) comprises a human mature IL-15 D30N/N65D variant linked an IL-15-Fc monomer comprising an amino acid substitution C220S, heterodimer pI substitutions L368D/K370S, isosteric pI substitutions Q295E/N384D/Q418E/N421D, ablation substitutions E233P/L234V/L235A/G236del/S267K, and FcRn substitutions M428L/N434S; and (b) a second monomer that (from N- to C-terminal) comprises a human IL-15R ⁇ (sushi) domain linked to an IL-15R ⁇ (sushi)-Fc monomer comprising the amino acid substitutions C220S, hetero
  • the composition comprises an anti-PD-1 antibody selected from the group consisting of nivolumab, pembrolizumab, and pedilizumab, and XENP24306 (as will be understand from those in the art, including the sequence identifiers thereof).
  • the composition comprises nivolumab and XENP24306.
  • the composition comprises pembrolizumab and XENP24306.
  • the composition comprises pedilizumab and XENP24306.
  • the composition comprises an anti-PD-1 antibody selected from the group consisting of nivolumab, pembrolizumab, and pedilizumab, and XENP24045 (as will be understand from those in the art, including the sequence identifiers thereof).
  • the composition comprises nivolumab and XENP24045.
  • the composition comprises pembrolizumab and XENP24045.
  • the composition comprises pedilizumab and XENP24045.
  • the combination therapy or treatment comprises an anti-PD-1 antibody selected from the group consisting of nivolumab, pembrolizumab, and pedilizumab, and XENP24306 (as will be understand from those in the art, including the sequence identifiers thereof).
  • the combination therapy or treatment comprises nivolumab and XENP24306.
  • the combination therapy or treatment comprises pembrolizumab and XENP24306.
  • the combination therapy or treatment comprises pedilizumab and XENP24306.
  • the combination therapy or treatment comprises an anti-PD-1 antibody selected from the group consisting of nivolumab, pembrolizumab, and pedilizumab, and XENP24045 (as will be understand from those in the art, including the sequence identifiers thereof).
  • the combination therapy or treatment comprises nivolumab and XENP24045.
  • the combination therapy or treatment comprises pembrolizumab and XENP24045.
  • the combination therapy or treatment comprises pedilizumab and XENP24045.
  • the combination therapy is useful for treating cancer, inducing T cell expansion, and/or results in minimal vascular leakage in a subject.
  • the combination therapy is administered to a subject and induces T cell expansion in the subject.
  • the expanded T cell population e.g., the TIL population is greater than when the subject is administered either the anti-PD-1 antibody or the untargeted IL-15/IL-15R ⁇ -Fc fusion protein alone.
  • the combination therapy is administered to a subject and results in a minimal level of vascular leakage compared to administration either the anti-PD-1 antibody or the untargeted IL-15/IL-15R ⁇ -Fc fusion protein alone.
  • the method of treating cancer comprises an anti-PD-1 antibody selected from the group consisting of nivolumab, pembrolizumab, and pedilizumab, and XENP24306 (as will be understand from those in the art, including the sequence identifiers thereof).
  • the method of method of treating cancer cancer comprises nivolumab and XENP24306.
  • the composition comprises pembrolizumab and XENP24306.
  • the method of treating cancer comprises pedilizumab and XENP24306.
  • the method of treating cancer that comprises an anti-PD-1 antibody selected from the group consisting of nivolumab, pembrolizumab, and pedilizumab, and XENP24045 (as will be understand from those in the art, including the sequence identifiers thereof).
  • the method of treating cancer comprises nivolumab and XENP24045.
  • the method of treating cancer comprises pembrolizumab and XENP24045.
  • the method of treating cancer comprises pedilizumab and XENP24045.
  • the method for expanding T cells in a subject comprises an anti-PD-1 antibody selected from the group consisting of nivolumab, pembrolizumab, and pedilizumab, and XENP24306 (as will be understand from those in the art, including the sequence identifiers thereof).
  • the method for expanding T cells in a subject comprises nivolumab and XENP24306.
  • the method for expanding T cells in a subject comprises pembrolizumab and XENP24306.
  • the method for expanding T cells in a subject comprises pedilizumab and XENP24306.
  • the method for expanding T cells in a subject comprises an anti-PD-1 antibody selected from the group consisting of nivolumab, pembrolizumab, and pedilizumab, and XENP24045 (as will be understand from those in the art, including the sequence identifiers thereof).
  • the method for expanding T cells in a subject comprises nivolumab and XENP24045.
  • the method for expanding T cells in a subject comprises pembrolizumab and XENP24045.
  • the method for expanding T cells in a subject comprises pedilizumab and XENP24045.
  • a treatment of an anti-PD-1 antibody and an exemplary IL-15/R ⁇ -Fc fusion variant induces proliferation of T cells including activated T cells in a subject.
  • an anti-PD-1 antibody e.g., XENP16432
  • IL-15/R ⁇ -Fc fusion variants e.g., XENP24306
  • administration of an anti-PD-1 antibody and an exemplary IL-15/R ⁇ -Fc fusion variant induces IFN ⁇ production.
  • the level of IFN ⁇ production increases by administering an anti-PD-1 antibody and an IL-15/R ⁇ -Fc fusion protein.
  • the level of IFN ⁇ is higher upon administering a treatment comprising an anti-PD-1 antibody and an IL-15/R ⁇ -Fc fusion protein, compared to anti-PD-1 antibody or IL-15/R ⁇ -Fc fusion protein alone.
  • administering decreases tumor size.
  • tumor size is lower upon administering a treatment comprising an anti-PD-1 antibody and an IL-15/R ⁇ -Fc fusion protein, compared to anti-PD-1 antibody or IL-15/R ⁇ -Fc fusion protein alone.
  • the composition or method includes an anti-PD-1 antibody and any of the heterodimeric fusion proteins of the present invention.
  • the heterodimeric fusion proteins of the present invention can take on a wide variety of configurations, as are generally depicted in FIGS. 9A-9G and FIGS. 39A-39D .
  • the amino acid sequences of exemplary fusion proteins are provided in FIGS. 12A, 12B, 13, 14, 15, 16, 40A, 40B, 41A, 41B, 42, 43, 48A-48H, 49A-49D, 50A-50B, 51, 52, 53 , 99 A- 99 C, 100 , 101 , and 102 , as will be apparent from the sequence identifiers.
  • the composition or method includes an anti-PD-1 antibody and any exemplary “IL-15/R ⁇ hetero Fc” and “dsIL-15/R ⁇ hetero Fc” protein.
  • first fusion protein comprising an IL-15 protein domain covalently attached using a first domain linker to the N-terminus of a first Fc domain
  • second fusion protein comprising an IL-15R ⁇ protein domain covalently attached using a second domain linker to the N-terminus of a second Fc domain.
  • IL-15/R ⁇ hetero Fc and “dsIL-15/R ⁇ hetero Fc”) include, but are not limited to, XENP20818, XENP20819, XENP21471, XENP21472, XENP21473, XENP21474, XENP21475, XENP21476, XENP21477, XENP22013, XENP22815, XENP22816, XENP22817, XENP22818, XENP22819, XENP22820, XENP22821, XENP22822, XENP22823, XENP22824, XENP22825, XENP22826, XENP22827, XENP22828, XENP22829, XENP22830, XENP22831, XENP22832, XENP22833,
  • the composition or method comprises an anti-PD-1 antibody and an IL-15/R ⁇ heterodimeric Fc fusion protein having a “IL-15/R ⁇ hetero Fc” and “dsIL-15/R ⁇ hetero Fc” format of XENP22357, XENP22358, XENP22359, XENP22360, XENP22362, XENP22363, XENP22364, XENP22365, XENP22366, XENP22684, XENP22361, XENP22816, XENP22819, XENP22820, XENP22821, XENP22822, XENP22829, XENP22834, XENP23554, XENP23557, XENP23561, XENP24018, XENP24019, XENP24045, XENP24051, XENP24052, XENP23343
  • FIGS. 41A, 41B, 48A, 48B, 48C, 48D, 48E, 48F, 48G, 48H, 53, 90A, 99B, and 99C Exemplary amino acid sequences of such formats are set forth in FIGS. 41A, 41B, 48A, 48B, 48C, 48D, 48E, 48F, 48G, 48H, 53, 90A, 99B, and 99C , as will be apparent from the sequence identifiers.
  • the composition or method includes an anti-PD-1 antibody and an scIL-15/R ⁇ -Fc heterodimeric fusion protein comprising a fusion protein comprising a first protein domain covalently attached to the N-terminus of a second protein domain via a first domain linker that is covalently attached to the N-terminus of a first Fc domain via a second domain linker, and a second Fc domain (e.g., an empty Fc domain).
  • the first protein domain is an IL-15R ⁇ protein domain and the second protein domain is an IL-15 protein domain.
  • An exemplary embodiment of this format (“scIL-15/R ⁇ -Fc”) includes, but is not limited to, XENP21478 (including the corresponding sequence identifiers).
  • the composition or method comprises an anti-PD-1 antibody and an IL-15/R ⁇ heterodimeric Fc fusion protein having a “scIL-15/R ⁇ -Fc” format of XENP24013, XENP24014, XENP24016, XENP24015, XENP24050, XENP24475, XENP24476, XENP24478, XENP24479, XENP24481, and XENP25938 (including the corresponding sequence identifiers).
  • Exemplary amino acid sequences of such formats are set forth in FIGS. 49A, 49B, 49C, 49D, and 100 , as will be apparent from the sequence identifiers.
  • the composition or method includes an anti-PD-1 antibody and an ncIL-15/R ⁇ -Fc or dsIL-15/R ⁇ -Fc heterodimeric fusion protein comprising a first protein domain covalently attached to the N-terminus a first Fc domain via a domain linker, a second Fc domain (e.g., an empty Fc domain), and a second protein domain that is noncovalently attached to the first protein domain.
  • the first protein domain is an IL-15 protein domain and the second protein domain is an IL-15R ⁇ protein domain.
  • ncIL-15/R ⁇ -Fc includes, but is not limited to, XENP21479, XENP22357, XENP22354, XENP22355, XENP22356, XENP22357, XENP22358, XENP22359, XENP22360, XENP22361, XENP22362, XENP22363, XENP22364, XENP22365, XENP22366, XENP22637, XENP24348, XENP24349, and XENP24383 (including the corresponding sequence identifiers).
  • the IL-15/R ⁇ heterodimeric Fc fusion protein having a “ncIL-15/R ⁇ -Fc” format is XENP21479, XENP22366, XENP24348, XENP24383, XENP24349, XENP24890, and XENP25138 (including the corresponding sequence identifiers).
  • Exemplary amino acid sequences of such formats are set forth in FIGS. 12A, 12B, 50A, 50B, and 101 , as will be apparent from the sequence identifiers.
  • the composition or method includes an anti-PD-1 antibody and a bivalent ncIL-15/R ⁇ -Fc or bivalent dsIL-15/R ⁇ -Fc fusion protein comprising a first fusion protein comprising a first protein domain covalently attached to the N-terminus of said first Fc domain via a first domain linker, a second fusion protein comprising a second protein domain covalently attached to the N-terminus of said second Fc domain via a second domain linker, a third protein domain noncovalently attached to said first protein domain of said first fusion protein, and a fourth protein domain noncovalently attached to said second protein domain of said second fusion protein.
  • the first and second protein domains are IL-15R ⁇ protein domains
  • the third and fourth protein domains are IL-15 protein domains.
  • An exemplary embodiment of this format (“bivalent ncIL-15/R ⁇ -Fc” or “bivalent dsIL-15/R ⁇ -Fc”) includes, but is not limited to, XENP21978, XENP22634, XENP24342, and XENP24346 (including the corresponding sequence identifiers).
  • the IL-15/R ⁇ heterodimeric Fc fusion protein having a “bivalent ncIL-15/R ⁇ -Fc” format is XENP21978, XENP24342, XENP24346, and XENP24351 (including the corresponding sequence identifiers). Exemplary amino acid sequences of such formats are set forth in FIGS. 13, 52, and 102 , as will be apparent from the sequence identifiers.
  • bivalent scIL-15/R ⁇ -Fc Another useful format (“bivalent scIL-15/R ⁇ -Fc”) is outlined herein in FIG. 14 , as will be apparent from the sequence identifiers.
  • the composition or method includes an anti-PD-1 antibody and an Fc-ncIL-15/R ⁇ fusion protein or Fc-dsIL-15/R ⁇ fusion protein comprising a first fusion protein comprising a first Fc domain covalently attached to the N-terminus of a first protein domain using a domain linker, a second Fc domain (e.g., an empty Fc domain), and a second protein domain noncovalently attached to said first protein domain.
  • An exemplary embodiment of this format (“Fc-ncIL-15/R ⁇ ” or “Fc-dsIL-15/R ⁇ ”) includes, but is not limited to, XENP22637 and XENP22639, and those depicted in FIG. 15 , as will be apparent from the sequence identifiers.
  • the first protein and the second protein are attached via a linker ( FIG. 9G ).
  • the IL-15/R ⁇ heterodimeric Fc fusion protein having a “Fc-ncIL-15/R ⁇ ” format is XENP22637 and XENP22638. Exemplary amino acid sequences of such formats are set forth in FIG. 15 , as will be apparent from the sequence identifiers.
  • the IL-15/R ⁇ heterodimeric Fc fusion protein having a “Fc-scIL-15/R ⁇ ” format is set forth in FIG. 16 , as will be apparent from the sequence identifiers.
  • the IL-15/R ⁇ heterodimeric Fc fusion protein having a “Fc-dsIL-15/R ⁇ ” is XENP22639 and XENP22640 Exemplary amino acid sequences of such formats are set forth in FIG. 42 , as will be apparent from the sequence identifiers.
  • the first domain linker and the second domain linker can be the same or different.
  • the first Fc domain and the second Fc domain of the heterodimeric protein can have different amino acid sequences.
  • the Fc domains of the present invention comprise IgG Fc domains, e.g., IgG1, IgG2, IgG3 or IgG4 Fc domains, with IgG1, IgG2, and IgG4 Fc domains finding particular use in some embodiments.
  • the first and second Fc domains comprising a set of amino acid substitutions selected from the group consisting of: L368D/K370S and S364K; L368D/K370S and S364K/E357L; L368D/K370S and S364K/E357Q; T411E/K360E/Q362E and D401K; L368E/K370S and S364K; K370S and S364K/E357Q; K370S and S364K/E357Q; S267K/L368D/K370S and S267K/S364K/E357Q, according to EU numbering.
  • the first and/or the second Fc domains of any of the heterodimeric Fc fusion formats outlined herein can have an additional set of amino acid substitutions comprising Q295E/N384D/Q418E/N421D, according to EU numbering.
  • the first and/or the second Fc domains have an additional set of amino acid substitutions consisting of G236R/L328R, E233P/L234V/L235A/G236del/S239K, E233P/L234V/L235A/G236del/S267K, E233P/L234V/L235A/G236del/S239K/A327G, E233P/L234V/L235A/G236del/S267K/A327G and E233P/L234V/L235A/G236del, according to EU numbering.
  • compositions can further comprise ablation variants, pI variants, charged variants, isotypic variants, etc.
  • compositions comprising anti-PD-1 antibodies and IL-15/R ⁇ -Fc heterodimeric fusion proteins with engineered disulfide bonds at the IL-15/R ⁇ interface (see, e.g., Example 2).
  • IL-15/R ⁇ -Fc fusion proteins can induce or promote proliferation of immune cells including NK cells, CD8 + T cells, and CD4 + T cells.
  • compositions comprising anti-PD-1 antibodies and IL-15/R ⁇ -Fc fusion proteins engineered for decreased potency (also referred to as “IL-15/R ⁇ -Fc affinity variants”; see, e.g., Example 3).
  • IL-15/R ⁇ -Fc affinity variants also referred to as “IL-15/R ⁇ -Fc affinity variants”; see, e.g., Example 3.
  • the IL-15/R ⁇ -Fc fusion protein variants show improved pharmacokinetics, decreased potency, and prolonged (e.g., increased) half-life.
  • compositions comprising anti-PD-1 antibodies and IL-15/R ⁇ -Fc fusion variants engineered for decreased potency are also engineered with an Xtend (FcRn) Fc substitution such that each Fc monomer comprises amino acid substitutions M428L/N434S.
  • FcRn Xtend
  • Exemplary embodiments of such IL-15/R ⁇ -Xtend Fc variants are depicted in FIGS. 99A-99C, 100, 101, and 102 (see also Table 6).
  • Administration of IL-15/R ⁇ -Xtend Fc variants induces proliferation of T cells including activated T cells in a subject.
  • IL-15/R ⁇ -Fc affinity variants with domain linker can exhibit extended expansion of lymphocytes in vivo as compared to corresponding IL-15/R ⁇ -Fc affinity variants without linker (hinge only; e.g., XENP24341).
  • IL-15/R ⁇ -Fc affinity variants can provide prolonged T cell pharmacology.
  • the IL-15/R ⁇ -Fc fusion proteins of the present invention can preferentially bind to T cells and NK cells, and in some cases, selectively target activated T cells in a cancer environment in a subject.
  • IL-15/R ⁇ -Fc fusion variants e.g., IL-15/R ⁇ -Fc affinity variants
  • domain linkers e.g., XENP24341
  • IL-15/R ⁇ -Fc fusion proteins described herein can enhance anti-tumor effects of activated T cells (see Examples 5B and 5C).
  • IL-15/R ⁇ -Fc fusion proteins described herein can induce the proliferation of CD8 + T cells (e.g., activated CD8 + T cells) and CD4 + T cells (e.g., activated CD4 + T cells).
  • CD8 + T cells e.g., activated CD8 + T cells
  • CD4 + T cells e.g., activated CD4 + T cells
  • the IL-15/R ⁇ -Fc fusion proteins can induce proliferation of CD8 + T cells over CD4 + T cells.
  • IL-15/R ⁇ -Fc fusion proteins described herein can induce the proliferation of CD8 + T cells (e.g., CD69 + /IFN ⁇ + fractions and/or CD69 +/ Ki-67 + fractions of CD8 + T cells) and CD4 + T cells (e.g., CD69 + /IFN ⁇ + fractions and/or CD69 +/ Ki-67 + fractions of CD4 + T cells).
  • CD8 + T cells e.g., CD69 + /IFN ⁇ + fractions and/or CD69 +/ Ki-67 + fractions of CD8 + T cells
  • CD4 + T cells e.g., CD69 + /IFN ⁇ + fractions and/or CD69 +/ Ki-67 + fractions of CD4 + T cells.
  • the IL-15/R ⁇ -Fc fusion proteins of the invention can promote killing of tumor cells and can selectively expand activated lymphocytes including tumor-infiltrating lymphocytes.
  • the IL-15/R ⁇ -Fc fusion proteins can prefer
  • IL-15/R ⁇ -Fc fusion proteins described herein can enhance anti-tumor effects in subjects that are administered such proteins. As illustrated in the Examples described below (see Example 5D), treatment with IL-15/R ⁇ -Fc fusion proteins significantly reduced tumor growth in subjects having tumors compared to treatment without such IL-15/R ⁇ -Fc fusion proteins.
  • IL-15/R ⁇ -Fc fusion proteins with and without Xtend-Fc substitutions described herein can enhance both pharmacodynamics and pharmacokinetics in subjects that are administered such proteins.
  • IL-15/R ⁇ -Fc fusion proteins with Xtend (FcRn) substitutions e.g., M428L/N434S variants on each Fc monomer
  • XmAb24306 and XENP23343 exhibited a longer serum half-life in subjects compared to corresponding IL-15/R ⁇ -Fc fusion proteins without Xtend substitutions.
  • Xtend substitutions significantly improved exposure as indicated by increased half-life.
  • reduced potency IL-15/R ⁇ -Fc variants such as XENP22821 can expand lymphocyte counts for a greater duration than wild-type IL-15/R ⁇ -Fc fusion proteins described herein such as XENP20818.
  • XENP23343 the Xtend-analog of XENP22821, further enhanced the duration of lymphocyte expansion beyond XENP22821.
  • IL-15/R ⁇ -Fc fusion proteins such as, but not limited to XmAb24306 (also referred to as XENP24306) can overcome Treg suppression of anti-CD3 induced effector T cell proliferation.
  • IL-15/R ⁇ -Fc fusion proteins such as, but not limited to XENP24045 can promote leukocyte expansion and exacerbate xenogeneic GVHD over a range of dose levels.
  • combination therapy of XENP24045 and an anti-PD-1 antibody such as XENP13432 showed synergy (e.g., a synergic effect), particularly at a low dose.
  • the IL-15/R ⁇ -Fc fusion protein of the present invention is XENP23557 (see, FIG. 48E ).
  • XENP23557 comprises chain 1 (human_IL15_N4D/N65D_(GGGGS)1_Fc(216)_IgG1_pI( ⁇ )_Isosteric_A_C220 S/PVA_/S267K/L368D/K370S (18786)) and chain 2 (human_IL15R ⁇ (Sushi)_(GGGGS)1_Fc(216)_IgG1_C220 S/PVA_/S267K/S364K/E357Q (15908)).
  • the IL-15/R ⁇ -Fc fusion protein of the present invention is XENP24045 (see, FIG. 48E ).
  • XENP24045 comprises chain 1 (human_IL15_D30N/E64Q/N65D_(GGGGS)1_Fc(216)_IgG1_pI( ⁇ )_Isosteric_A_C220S/PVA_/S267K/L368D/K370S) and chain 2 (human_IL15R ⁇ (Sushi)_(GGGGS)1_Fc(216)_IgG1_C220S/PVA_/S267K/S364K/E357Q).
  • the IL-15/R ⁇ -Fc fusion protein of the present invention is XENP24113 (see, FIG. 99B ).
  • XENP24113 comprises chain 1 (human_IL15_N4D/N65D_(GGGGS)1_Fc(216)_IgG1_pI( ⁇ )_Isosteric_A_C220S/PVA_/S267K/L368D/K370S/M428L/N434S) and chain 2 (human_IL15R ⁇ (Sushi)_(GGGGS)1_Fc(216)_IgG1_C220S/PVA_/S267K/S364K/E357Q/M428 L/N434S).
  • the IL-15/R ⁇ -Fc fusion protein of the present invention is XENP24306 (see, FIG. 99C ).
  • XENP24306 comprises chain 1 (human_IL15_D30N/E64Q/N65D_(GGGGS)1_Fc(216)_IgG1_pI( ⁇ )_Isosteric_A_C220S/PVA_/S267K/L368D/K370S/M428L/N434S) and chain 2 (human_IL15R ⁇ (Sushi)_(GGGGS)1_Fc(216)_IgG1_C220S/PVA_/5267K/5364K/E357Q/M428 L/N434S).
  • the present invention provides a heterodimeric protein comprising a) a first fusion protein comprising a first protein domain and a first Fc domain, wherein the first protein domain is covalently attached to the N-terminus of the first Fc domain using a first domain linker; b) a second fusion protein comprising a second protein domain and a second Fc domain, wherein the second protein domain is covalently attached to the N-terminus of the Fc domain using a second domain linker; wherein the first and the second Fc domains have a set of amino acid substitutions selected from the group consisting of S267K/L368D/K370S:S267K/LS364K/E357Q; S364K/E357Q:L368D/K370S; L368D/K370S:S364K; L368E/K370S:S364K; T411E/K360E/Q362E:D401K; L368D/K
  • the first protein domain is covalently attached to the N-terminus of the first Fc domain directly and without using the first domain linker and/or the second protein domain is covalently attached to the N-terminus of the second Fc domain directly and without using the second domain linker.
  • the invention also provides a checkpoint blockade antibody such as an anti-PD-1 antibody.
  • the heterodimeric protein is selected from the group consisting of XENP20818, XENP20819, XENP21471, XENP21472, XENP21473, XENP21474, XENP21475, XENP21476, XENP21477, XENP22013, XENP22815, XENP22816, XENP22817, XENP22818, XENP22819, XENP22820, XENP22821, XENP22822, XENP22823, XENP22824, XENP22825, XENP22826, XENP22827, XENP22828, XENP22829, XENP22830, XENP22831, XENP22832, XENP22833, XENP22834, XENP23343, XENP23554,
  • the invention provides a heterodimeric protein comprising: a) a fusion protein comprising a first protein domain, a second protein domain, and a first Fc domain, wherein the first protein domain is covalently attached to the N-terminus of the second protein domain using a first domain linker, and wherein the second protein domain is covalently attached to the N-terminus of the first Fc domain using a second domain linker; b) a second Fc domain; wherein the first and the second Fc domains have a set of amino acid substitutions selected from the group consisting of S267K/L368D/K370S:S267K/LS364K/E357Q; S364K/E357Q:L368D/K370S; L368D/K370S:S364K; L368E/K370S:S364K; T411E/K360E/Q362E:D401K; L368D/K370S:S364K
  • the heterodimeric protein can be XENP21478.
  • the invention provides a heterodimeric protein comprising: a) a fusion protein comprising a first protein domain and a first Fc domain, wherein the first protein domain is covalently attached to the N-terminus of the first Fc domain using a domain linker; b) a second Fc domain; and c) a second protein domain noncovalently attached to the first protein domain; wherein the first and the second Fc domains have a set of amino acid substitutions selected from the group consisting of S267K/L368D/K370S:S267K/LS364K/E357Q; S364K/E357Q:L368D/K370S; L368D/K370S:S364K; L368E/K370S:S364K; T411E/K360E/Q362E:D401K; L368D/K370S:S364K/E357L and K370S:S364K/E357Q, according
  • the heterodimer protein can be selected from the group consisting of XENP21479, XENP22357, XENP22354, XENP22355, XENP22356, XENP22357, XENP22358, XENP22359, XENP22360, XENP22361, XENP22362, XENP22363, XENP22364, XENP22365, XENP22366, and XENP22637 (including the corresponding sequence identifiers).
  • the invention provides a heterodimeric protein comprising: a) a first fusion protein comprising a first protein domain and a first Fc domain, wherein the first protein domain is covalently attached to the N-terminus of said first Fc domain using a domain linker; b) a second fusion protein comprising a second heavy chain comprising a second protein domain and a first second heavy chain comprising a second Fc domain, wherein the second protein domain is covalently attached to the C-terminus of the second Fc domain using a domain linker; c) a third protein domain noncovalently attached to the first protein domain of the first fusion protein; and d) a fourth protein domain noncovalently attached to the second protein domain of the second fusion protein, wherein the first and the second Fc domains have a set of amino acid substitutions selected from the group consisting of S267K/L368D/K370S:S267K/LS364K/E357Q; S364K/E3
  • the invention provides a heterodimeric protein comprising: a) a first fusion protein comprising a first Fc domain and a first protein domain, wherein the first Fc domain is covalently attached to the N-terminus of the first protein domain using a domain linker; b) a second Fc domain, and c) a second protein domain noncovalently attached to the first protein domain of the first fusion protein; wherein the first and the second Fc domains have a set of amino acid substitutions selected from the group consisting of S267K/L368D/K370S:S267K/LS364K/E357Q; S364K/E357Q:L368D/K370S; L368D/K370S:S364K; L368E/K370S:S364K; T411E/K360E/Q362E:D401K; L368D/K370S:S364K/E357L and K370S:S364K;
  • the first and/or the second Fc domains can have an additional set of amino acid substitutions comprising Q295E/N384D/Q418E/N421D, according to EU numbering.
  • the first and/or the second Fc domains have an additional set of amino acid substitutions consisting of G236R/L328R, E233P/L234V/L235A/G236del/S239K, E233P/L234V/L235A/G236del/S267K, E233P/L234V/L235A/G236del/S239K/A327G, E233P/L234V/L235A/G236del/S267K/A327G and E233P/L234V/L235A/G236del, according to EU numbering.
  • the IL-15 protein has a polypeptide sequence selected from the group consisting of full-length human IL-15 and a truncated human IL-15
  • the IL-15R ⁇ protein has a polypeptide sequence selected from the group consisting of full-length human IL-15R ⁇ and the sushi domain of human IL-15R ⁇ .
  • the IL-15 protein and the IL1-5R ⁇ protein have a set of amino acid substitutions or additions selected from the group consisting of E87C:D96/P97/C98; E87C:D96/C97/A98; V49C:S40C; L52C:S40C; E89C:K34C; Q48C:G38C; E53C:L42C; C42S:A37C; and L45C:A37C, respectively.
  • the present invention provides a checkpoint blockade antibody and a heterodimeric protein selected from the group consisting of XENP20818, XENP20819, XENP21471, XENP21472, XENP21473, XENP21474, XENP21475, XENP21476, XENP21477, XENP21478, XENP21479, XENP21978, XENP22013, XENP22015, XENP22017, XENP22354, XENP22355, XENP22356, XENP22357, XENP22358, XENP22359, XENP22360, XENP22361, XENP22362, XENP22363, XENP22364, XENP22365, XENP22366, XENP22637, and XENP22639 (including the corresponding sequence identifier
  • the present invention provides a heterodimeric protein selected from the group consisting of XENP20818, XENP20819, XENP21471, XENP21472, XENP21473, XENP21474, XENP21475, XENP21476, XENP21477, XENP22013, XENP22815, XENP22816, XENP22817, XENP22818, XENP22819, XENP22820, XENP22821, XENP22822, XENP22823, XENP22824, XENP22825, XENP22826, XENP22827, XENP22828, XENP22829, XENP22830, XENP22831, XENP22832, XENP22833, XENP22834, XENP23343, XEN
  • a method of treating cancer in a patient in need thereof and minimizing the level of vascular leakage in said patient comprises administering a therapeutically effective amount of a checkpoint blockade antibody, e.g., an anti-PD-1 antibody and a therapeutically effective amount of an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein described herein or a pharmaceutical composition described herein to the patient.
  • a checkpoint blockade antibody e.g., an anti-PD-1 antibody and a therapeutically effective amount of an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein described herein or a pharmaceutical composition described herein
  • the method also includes administering a therapeutically effective amount of a checkpoint blockade antibody.
  • the checkpoint blockade antibody is selected from an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM3 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, and an anti-CTLA-4 antibody.
  • the anti-PD-1 antibody is nivolumab, pembrolizumab, or pidilizumab.
  • the anti-PD-L1 antibody is atezolizumab, avelumab, or durbalumab.
  • the level of vascular leakage ranges from a 20% reduction or less, e.g., a 20% reduction, a 19% reduction, a 18% reduction, a 17% reduction, a 16% reduction, a 20% reduction, a 20% reduction, a 20% reduction, a 20% reduction, a 15% reduction, a 14% reduction, a 13% reduction, a 12% reduction, a 11% reduction, a 10% reduction, or less reduction in serum albumin in the patient following administration of an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein and an anti-PD-1 antibody. In some instances, such a reduction in serum albumin occurs at day 1 to day 15 after administration.
  • the reduction occurs (or is detectable) at day 1, day 2, day 3, day 4, day 5, day 6, day 7, day 8, day 9, day 10, day 11, day 12, day 13, day 14, day 15 or later after administration of an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein and an anti-PD-1 antibody.
  • the patient exhibits a 2-fold to 15-fold increase, e.g., a 2-fold, a 3-fold, a 4-fold, a 5-fold, a 6-fold, a 7-fold, an 8-fold, a 9-fold, a 10-fold, a 11-fold, a 12-fold, a 13-fold, a 14-fold, or a 15-fold increase in lymphocytes following administration of an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein and an anti-PD-1 antibody.
  • such an increase in lymphocytes occurs at day 1 to day 15 after administration.
  • the increase in lymphocytes occurs (or is detectable) at day 1, day 2, day 3, day 4, day 5, day 6, day 7, day 8, day 9, day 10, day 11, day 12, day 13, day 14, day 15 or later after administration.
  • the patient exhibits a 2-fold to 13-fold increase, e.g., a 2-fold, a 3-fold, a 4-fold, a 5-fold, a 6-fold, a 7-fold, an 8-fold, a 9-fold, a 10-fold, a 11-fold, a 12-fold, or a 13-fold increase in peripheral CD8+ T cells following administration of an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein and an anti-PD-1 antibody.
  • a 2-fold to 13-fold increase e.g., a 2-fold, a 3-fold, a 4-fold, a 5-fold, a 6-fold, a 7-fold, an 8-fold, a 9-fold, a 10-fold, a 11-fold, a 12-fold, or a 13-fold increase in peripheral CD8+ T cells following administration of an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein and an anti-PD-1 antibody.
  • such an increase in peripheral CD8+ T cells occurs at day
  • a method of inducing T cell expansion in a patient in need thereof without increasing the likelihood of inducing hypoalbuminemia comprising administering a therapeutically effective amount of an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein described herein or a pharmaceutical composition described herein to said patient.
  • the method also includes administering a therapeutically effective amount of a checkpoint blockade antibody.
  • the checkpoint blockade antibody is selected from an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM3 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, and an anti-CTLA-4 antibody.
  • the anti-PD-1 antibody is nivolumab, pembrolizumab, or pidilizumab.
  • the T cell expansion is at least a 2-fold, e.g., a 2-fold, a 3-fold, a 4-fold, a 5-fold, a 6-fold, a 7-fold, a 8-fold, a 9-fold, a 10-fold, a 11-fold, a 12-fold, a 13-fold, a 14-fold, a 15-fold, a 16-fold, a 17-fold, a 18-fold, a 19-fold, a 20-fold, a 21-fold, or more fold increase in T cells.
  • a 2-fold e.g., a 2-fold, a 3-fold, a 4-fold, a 5-fold, a 6-fold, a 7-fold, a 8-fold, a 9-fold, a 10-fold, a 11-fold, a 12-fold, a 13-fold, a 14-fold, a 15-fold, a 16-fold, a 17-fold, a 18-fold, a 19-fold, a
  • the T cell expansion ranges from a 2-fold to a 15-fold increase, e.g., a 2-fold, a 3-fold, a 4-fold, a 5-fold, a 6-fold, a 7-fold, an 8-fold, a 9-fold, a 10-fold, a 11-fold, a 12-fold, a 13-fold, a 14-fold, or a 15-fold increase in T cells.
  • a 2-fold to a 15-fold increase e.g., a 2-fold, a 3-fold, a 4-fold, a 5-fold, a 6-fold, a 7-fold, an 8-fold, a 9-fold, a 10-fold, a 11-fold, a 12-fold, a 13-fold, a 14-fold, or a 15-fold increase in T cells.
  • such an expansion in T cells occurs at day 1 to day 15 after administration of an IL-15/IL-15R ⁇ heterodimeric Fc fusion and a checkpoint blockade antibody, e.
  • the expansion in T cells occurs (or is detectable) at day 1, day 2, day 3, day 4, day 5, day 6, day 7, day 8, day 9, day 10, day 11, day 12, day 13, day 14, day 15 or later after administration of an IL-15/IL-15R ⁇ heterodimeric Fc fusion and a checkpoint blockade antibody, e.g., an anti-PD-1 antibody.
  • a checkpoint blockade antibody e.g., an anti-PD-1 antibody.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein described herein in combination with an anti-PD-1 antibody enhances lymphocytes expansion in comparison to a control IL-15 or IL-15R ⁇ containing protein, and also results in a reduction in albumin drop (data depicted in FIG. 203 ).
  • the reduction in albumin drop indicates a superior therapeutic index for the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein and anti-PD-1 antibody.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein in combination with the anti-PD-1 antibody promotes at least a 3-fold (200%) increase in peripheral CD8 + T cells.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein in combination with the anti-PD-1 antibody mediated or facilitated the extent of albumin decrease as not to exceed a 15% reduction.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein in combination with the anti-PD-1 antibody caused or induced a decrease in albumin levels that is lower than a control IL-15 or IL-15R ⁇ containing protein.
  • a patient described herein is administered IL-15/IL-15R ⁇ heterodimeric Fc fusion protein and an anti-PD-1 antibody which promotes an at least 11-fold (1000%) increase of peripheral CD8 + T cells while maintaining albumin levels above a 20% decrease.
  • the present invention provides in a method of inducing T cell expansion in a patient by administering a checkpoint blockade antibody and an IL-15 protein in complex with an IL-15R ⁇ protein, the improvement comprising administering to the patient an anti-PD-1 antibody and an IL-15 variant protein in complex with an IL-15R ⁇ protein, wherein the IL-15 variant protein has reduced affinity such that the patient has a reduced likelihood of hypoalbuminemia.
  • the variant IL-15 has one or more amino acid substitutions selected from the group consisting of N1D, N4D, D8N, D30N, D61N, E64Q, N65D, and Q108E.
  • the variant IL-15 has an N1D substitution or at least an N1D substitution.
  • the variant IL-15 has an N4D substitution or at least an N4D substitution.
  • the variant IL-15 has an N1D substitution or at least an D8N substitution.
  • the variant IL-15 has an D30N substitution or at least an D30N substitution.
  • the variant IL-15 has an D61N substitution or at least an D61N substitution.
  • the variant IL-15 has an E64Q substitution or at least an E64Q substitution. In some embodiments, the variant IL-15 has an N65D substitution or at least an N65D substitution. In some embodiments, the variant IL-15 has an Q108E substitution or at least an Q108E substitution.
  • the IL-15 variant protein has an amino acid substitution(s) selected from the group consisting of N4D, N65D, N4D/N65D, D30N/N65D, and D30N/E64Q/N65D.
  • the variant IL-15 has an N4D substitution.
  • the variant IL-15 has an N65D substitution.
  • the variant IL-15 has N4D/N65D substitutions.
  • the variant IL-15 has D30N/N65D substitutions.
  • the variant IL-15 has D30N/E64Q/N65D substitutions.
  • the IL-15 variant protein in complex with an IL-15R ⁇ protein is any one of the IL-15/IL-15R ⁇ heterodimeric Fc fusion proteins described herein.
  • the present invention provides an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein comprising (a) a first fusion protein comprising a first protein domain and a first Fc domain, wherein the first protein domain is covalently attached to the N-terminus of the first Fc domain using a first domain linker; (b) a second fusion protein comprising a second protein domain and a second Fc domain, wherein the second protein domain is covalently attached to the N-terminus of the Fc domain using a second domain linker; wherein the first and the second Fc domains have a set of amino acid substitutions selected from the group consisting of S267K/L368D/K370S:S267K/LS364K/E357Q; S364K/E357Q:L368D/K370S; L368D/K370S:S364K; L368E/K370S:S364K; T411T/E360E/Q362E
  • the first protein domain is covalently attached to the N-terminus of the first Fc domain directly and without using the first domain linker and/or the second protein domain is covalently attached to the N-terminus of the second Fc domain directly and without using the second domain linker.
  • the first and/or the second Fc domains have an additional set of amino acid substitutions comprising Q295E/N384D/Q418E/N421D, according to EU numbering. In some embodiments, wherein the first and/or the second Fc domains have an additional set of amino acid substitutions consisting of G236R/L328R, E233P/L234V/L235A/G236del/S239K, E233P/L234V/L235A/G236del/S267K, E233P/L234V/L235A/G236del/S239K/A327G, E233P/L234V/L235A/G236del/S267K/A327G and E233P/L234V/L235A/G236del, according to EU numbering. In some embodiments, the first and/or the second Fc domains have an additional amino acid substitution M428L/N434S,
  • the IL-15 protein has a polypeptide sequence selected from the group consisting of full-length human IL-15 protein and a truncated human IL-15 protein
  • said IL-15R ⁇ protein has a polypeptide sequence selected from the group consisting of full-length human IL-15R ⁇ protein and the sushi domain of human IL-15R ⁇ protein.
  • the IL-15 protein has one or more amino acid substitutions selected from the group consisting of N1D, N4D, D8N, D30N, D61N, E64Q, N65D, and Q108E.
  • IL-15 protein has an amino acid substitution selected from the group consisting of N4D, N65D, N4D/N65D, and D30N/E64Q/N65D.
  • the IL-15 protein and the IL-15R ⁇ protein have a set of amino acid substitutions or additions selected from the group consisting of E87C:D96/P97/C98; E87C: D96/C97/A98; V49C:S40C; L52C:S40C; E89C:K34C; Q48C:G38C; E53C:L42C; C42S:A37C; and L45C:A37C, respectively.
  • the first protein domain is covalently attached to the N-terminus of the first Fc domain directly and without using the first domain linker and/or the second protein domain is covalently attached to the N-terminus of the second Fc domain directly and without using the second domain linker.
  • the IL-15/IL-15R ⁇ heterodimeric Fc fusion protein is selected from the group consisting of XENP20818, XENP22013, XENP22014, XENP22015, XENP22017, XENP23343, XENP23504, XENP23557, XENP24045, XENP24113, XENP24301, XENP24306, and XENP24341.
  • Nucleic acids, expression vectors and host cells are all provided as well, in addition to methods of making these proteins and treating patients with them.
  • the present invention provides a pharmaceutical composition comprising any one of the IL-15/IL-15R ⁇ Fc fusion heterodimeric proteins described herein and a pharmaceutically acceptable carrier.
  • the present invention provides a pharmaceutical composition comprising an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein selected from the group consisting of XENP20818, XENP22013, XENP22014, XENP22015, XENP22017, XENP23343, XENP23504, XENP23557, XENP24045, XENP24113, XENP24301, XENP24306, and XENP24341; and a pharmaceutically acceptable carrier.
  • the present invention provides a method of treating cancer in a patient in need thereof comprising administering a therapeutically effective amount of any of the IL-15/IL-15R ⁇ heterodimeric Fc fusion proteins described herein or a pharmaceutical composition thereof to the patient.
  • the method also comprises administering a therapeutically effective amount of a checkpoint blockade antibody.
  • the checkpoint blockade antibody is selected from an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM3 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, and an anti-CTLA-4 antibody.
  • the anti-PD-1 antibody is nivolumab, pembrolizumab, or pidilizumab.
  • the anti-PD-L1 antibody is atezolizumab, avelumab, or durbalumab.
  • the present invention provides a method of treating cancer in a patient in need thereof and minimizing the level of vascular leakage in the patient.
  • the method comprises administering a therapeutically effective amount of an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein described herein or a pharmaceutical composition described herein to the patient.
  • the method also comprises administering a therapeutically effective amount of a checkpoint blockade antibody.
  • the checkpoint blockade antibody is selected from an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM3 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, and an anti-CTLA-4 antibody.
  • the anti-PD-1 antibody is nivolumab, pembrolizumab, or pidilizumab.
  • the anti-PD-L1 antibody is atezolizumab, avelumab, or durbalumab.
  • the level of vascular leakage ranges from a 20% reduction or less in serum albumin in the patient following administration.
  • the patient exhibits a 2-fold to 15-fold increase in lymphocytes following administration.
  • the patient exhibits a 2-fold to 13-fold increase in peripheral CD8+ T cells following administration.
  • the present invention provides a method of inducing T cell expansion in a patient in need thereof without increasing the likelihood of inducing hypoalbuminemia comprising administering a therapeutically effective amount of an IL-15/IL-15R ⁇ heterodimeric Fc fusion protein described herein or a pharmaceutical composition described herein to the patient.
  • the method also comprises administering a therapeutically effective amount of a checkpoint blockade antibody.
  • the checkpoint blockade antibody is selected from an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-TIM3 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, and an anti-CTLA-4 antibody.
  • the anti-PD-1 antibody is nivolumab, pembrolizumab, or pidilizumab.
  • the anti-PD-L1 antibody is atezolizumab, avelumab, or durbalumab.
  • the T cell expansion is at least a 2-fold increase in T cells. In some embodiments, the T cell expansion ranges from a 2-fold to a 15-fold increase in T cells.
  • the present invention provides in a method of inducing T cell expansion in a patient by administering an IL-15 protein in complex with an IL-15R ⁇ protein, the improvement comprising administering to the patient an IL-15 variant protein in complex with an IL-15R ⁇ protein, wherein the IL-15 variant protein has reduced affinity such that the patient has a reduced likelihood of hypoalbuminemia.
  • the variant IL-15 has one or more amino acid substitutions selected from the group consisting of N1D, N4D, D8N, D30N, D61N, E64Q, N65D, and Q108E.
  • the IL-15 variant protein has an amino acid substitution(s) selected from the group consisting of N4D, N65D, N4D/N65D, and D30N/E64Q/N65D.
  • the IL-15 variant protein in complex with an IL-15R ⁇ protein is any one of the IL-15/IL-15R ⁇ heterodimeric Fc fusion proteins described herein.
  • the invention further provides nucleic acid compositions encoding the heterodimeric Fc fusion protein of the invention (or, in the case of a monomer Fc domain protein, nucleic acids encoding those as well).
  • nucleic acid compositions will depend on the format of the heterodimeric protein.
  • the format requires three amino acid sequences
  • three nucleic acid sequences can be incorporated into one or more expression vectors for expression.
  • only two nucleic acids are needed; again, they can be put into one or two expression vectors.
  • the nucleic acids encoding the components of the invention can be incorporated into expression vectors as is known in the art, and depending on the host cells used to produce the heterodimeric Fc fusion proteins of the invention. Generally the nucleic acids are operably linked to any number of regulatory elements (promoters, origin of replication, selectable markers, ribosomal binding sites, inducers, etc.).
  • the expression vectors can be extra-chromosomal or integrating vectors.
  • nucleic acids and/or expression vectors of the invention are then transformed into any number of different types of host cells as is well known in the art, including mammalian, bacterial, yeast, insect and/or fungal cells, with mammalian cells (e.g., CHO cells), finding use in many embodiments.
  • mammalian cells e.g., CHO cells
  • nucleic acids encoding each monomer are each contained within a single expression vector, generally under different or the same promoter controls. In embodiments of particular use in the present invention, each of these two or three nucleic acids are contained on a different expression vector.
  • the heterodimeric Fc fusion protein of the invention are made by culturing host cells comprising the expression vector(s) as is well known in the art. Once produced, traditional fusion protein or antibody purification steps are done, including an ion exchange chromotography step. As discussed herein, having the pIs of the two monomers differ by at least 0.5 can allow separation by ion exchange chromatography or isoelectric focusing, or other methods sensitive to isoelectric point.
  • substitutions that alter the isoelectric point (p1) of each monomer so that each monomer has a different p1 and the heterodimer also has a distinct p1, thus facilitating isoelectric purification of the heterodimer (e.g., anionic exchange chromatography, cationic exchange chromatography). These substitutions also aid in the determination and monitoring of any contaminating homodimers post-purification (e.g., IEF gels, cIEF, and analytical IEX columns).
  • the heterodimeric Fc fusion proteins of the invention are administered to patients with cancer, and efficacy is assessed, in a number of ways as described herein.
  • efficacy is assessed, in a number of ways as described herein.
  • standard assays of efficacy can be run, such as cancer load, size of tumor, evaluation of presence or extent of metastasis, etc.
  • immuno-oncology treatments can be assessed on the basis of immune status evaluations as well. This can be done in a number of ways, including both in vitro and in vivo assays. For example, evaluation of changes in immune status (e.g., presence of ICOS+ CD4+ T cells following ipi treatment) along with “old fashioned” measurements such as tumor burden, size, invasiveness, LN involvement, metastasis, etc. can be done.
  • any or all of the following can be evaluated: the inhibitory effects of PVRIG on CD4 + T cell activation or proliferation, CD8 + T (CTL) cell activation or proliferation, CD8 + T cell-mediated cytotoxic activity and/or CTL mediated cell depletion, NK cell activity and NK mediated cell depletion, the potentiating effects of PVRIG on Treg cell differentiation and proliferation and Treg- or myeloid derived suppressor cell (MDSC)-mediated immunosuppression or immune tolerance, and/or the effects of PVRIG on proinflammatory cytokine production by immune cells, e.g., IL-2, IFN- ⁇ or TNF- ⁇ production by T or other immune cells.
  • CTL CD8 + T
  • CTL CTL cytotoxic activity and/or CTL mediated cell depletion
  • NK cell activity and NK mediated cell depletion the potentiating effects of PVRIG on Treg cell differentiation and proliferation and Treg- or myeloid derived suppressor cell (MDSC)-mediated immunos
  • assessment of treatment is done by evaluating immune cell proliferation, using for example, CFSE dilution method, Ki67 intracellular staining of immune effector cells, and 3 H-thymidine incorporation method.
  • assessment of treatment is done by evaluating the increase in gene expression or increased protein levels of activation-associated markers, including one or more of: CD25, CD69, CD137, ICOS, PD1, GITR, OX40, and cell degranulation measured by surface expression of CD107A.
  • gene expression assays are done as is known in the art.
  • assessment of treatment is done by assessing cytotoxic activity measured by target cell viability detection via estimating numerous cell parameters such as enzyme activity (including protease activity), cell membrane permeability, cell adherence, ATP production, co-enzyme production, and nucleotide uptake activity.
  • enzyme activity including protease activity
  • cell membrane permeability cell permeability
  • cell adherence cell adherence
  • ATP production co-enzyme production
  • nucleotide uptake activity include, but are not limited to, Trypan Blue or PI staining, 51 Cr or 35 S release method, LDH activity, MTT and/or WST assays, Calcein-AM assay, Luminescent based assay, and others.
  • assessment of treatment is done by assessing T cell activity measured by cytokine production, measure either intracellularly in culture supernatant using cytokines including, but not limited to, IFN ⁇ , TNF ⁇ , GM-CSF, IL2, IL6, IL4, IL5, IL10, IL13 using well known techniques.
  • cytokines including, but not limited to, IFN ⁇ , TNF ⁇ , GM-CSF, IL2, IL6, IL4, IL5, IL10, IL13 using well known techniques.
  • assessment of treatment can be done using assays that evaluate one or more of the following: (i) increases in immune response, (ii) increases in activation of ⁇ and/or ⁇ T cells, (iii) increases in cytotoxic T cell activity, (iv) increases in NK and/or NKT cell activity, (v) alleviation of ⁇ and/or ⁇ T-cell suppression, (vi) increases in pro-inflammatory cytokine secretion, (vii) increases in IL-2 secretion; (viii) increases in interferon- ⁇ production, (ix) increases in Th1 response, (x) decreases in Th2 response, (xi) decreases or eliminates cell number and/or activity of at least one of regulatory T cells (Tregs).
  • T cell activation is assessed using a Mixed Lymphocyte Reaction (MLR) assay as is known in the art.
  • MLR Mixed Lymphocyte Reaction
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in immune response as measured for an example by phosphorylation or de-phosphorylation of different factors, or by measuring other post translational modifications.
  • IL-15 mediates IFN ⁇ expression and secretion through phosphorylation of STAT5.
  • the signaling pathway assay measures increases or decreases in immune response as indicated by phosphorylation of STAT5.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in activation of ⁇ and/or ⁇ T cells as measured for an example by cytokine secretion or by proliferation or by changes in expression of activation markers like for an example CD137, CD107a, PD1, etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in cytotoxic T cell activity as measured for an example by direct killing of target cells like for an example cancer cells or by cytokine secretion or by proliferation or by changes in expression of activation markers like for an example CD137, CD107a, PD1, etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in NK and/or NKT cell activity as measured for an example by direct killing of target cells like for an example cancer cells or by cytokine secretion or by changes in expression of activation markers like for an example CD107a, etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in ⁇ and/or ⁇ T-cell suppression, as measured for an example by cytokine secretion or by proliferation or by changes in expression of activation markers like for an example CD137, CD107a, PD1, etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in pro-inflammatory cytokine secretion as measured for example by ELISA or by Luminex or by Multiplex bead based methods or by intracellular staining and FACS analysis or by Alispot etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.
  • the signaling pathway assay measures increases or decreases in IL-2 secretion as measured for example by ELISA or by Luminex or by Multiplex bead based methods or by intracellular staining and FACS analysis or by Alispot etc.
  • An increase in activity indicates immunostimulatory activity. Appropriate increases in activity are outlined below.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Endocrinology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US16/388,174 2018-04-18 2019-04-18 Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof Abandoned US20190365861A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/388,174 US20190365861A1 (en) 2018-04-18 2019-04-18 Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof
US17/209,047 US20220040264A1 (en) 2018-04-18 2021-03-22 Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201862659563P 2018-04-18 2018-04-18
US201862684143P 2018-06-12 2018-06-12
US201862724396P 2018-08-29 2018-08-29
US201862756800P 2018-11-07 2018-11-07
US16/388,174 US20190365861A1 (en) 2018-04-18 2019-04-18 Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/209,047 Continuation US20220040264A1 (en) 2018-04-18 2021-03-22 Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof

Publications (1)

Publication Number Publication Date
US20190365861A1 true US20190365861A1 (en) 2019-12-05

Family

ID=68239907

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/388,174 Abandoned US20190365861A1 (en) 2018-04-18 2019-04-18 Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof
US17/209,047 Pending US20220040264A1 (en) 2018-04-18 2021-03-22 Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/209,047 Pending US20220040264A1 (en) 2018-04-18 2021-03-22 Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof

Country Status (12)

Country Link
US (2) US20190365861A1 (he)
EP (1) EP3781596A1 (he)
KR (1) KR20210003170A (he)
CN (1) CN112105645A (he)
AU (1) AU2019255744A1 (he)
CA (1) CA3097625A1 (he)
IL (2) IL310398A (he)
MA (1) MA52289A (he)
MX (1) MX2020010912A (he)
SG (1) SG11202010159RA (he)
TW (1) TW202011984A (he)
WO (1) WO2019204592A1 (he)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11136384B2 (en) * 2016-11-30 2021-10-05 Mereo Biopharma 5, Inc. Methods for treatment of cancer comprising TIGIT-binding agents
WO2023010031A1 (en) * 2021-07-28 2023-02-02 Genentech, Inc. Il15/il15r alpha heterodimeric fc-fusion proteins for the treatment of blood cancers
CN115806627A (zh) * 2022-08-03 2023-03-17 深圳市先康达生命科学有限公司 一种自分泌IL-15与anti-LAG3结合的融合蛋白及其应用
US11932675B2 (en) 2019-10-11 2024-03-19 Genentech, Inc. PD-1 targeted IL-15/IL-15Rα Fc fusion proteins with improved properties

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20191034A1 (es) 2016-10-14 2019-08-05 Xencor Inc Proteinas de fusion heterodimericas biespecificas que contienen proteinas de fusion fc il-15/il-15r y fragmentos de anticuerpo pd-1
AU2019256539A1 (en) 2018-04-18 2020-11-26 Xencor, Inc. PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
EP3781598A1 (en) 2018-04-18 2021-02-24 Xencor, Inc. Tim-3 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and tim-3 antigen binding domains
CN113423734A (zh) 2018-10-12 2021-09-21 Xencor股份有限公司 靶向PD-1的IL-15/IL-15RαFC融合蛋白及其在联合疗法中的应用
EP3897853A1 (en) 2018-12-20 2021-10-27 Xencor, Inc. Targeted heterodimeric fc fusion proteins containing il-15/il-15ra and nkg2d antigen binding domains
WO2021092719A1 (zh) * 2019-11-11 2021-05-20 王盛典 一种靶向抗原特异性t细胞诱导其向记忆干性细胞分化的融合蛋白
JP2023502712A (ja) * 2019-11-21 2023-01-25 インサーム(インスティテュ ナシオナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシェ メディカル) 抗pd-1/il-15免疫サイトカインによる新しいpd-1標的免疫療法
US20230048046A1 (en) * 2019-12-13 2023-02-16 Cugene Inc Novel interleukin-15 (il-15) fusion proteins and uses thereof
CA3164353A1 (en) * 2019-12-13 2021-06-17 Cugene Inc. Cytokine-based bioactivatable drugs and methods of uses thereof
WO2021142476A1 (en) * 2020-01-12 2021-07-15 Dragonfly Therapeutics, Inc. Single-chain polypeptides
JP2023511439A (ja) * 2020-01-28 2023-03-17 ジェネンテック, インコーポレイテッド がんの治療のためのil15/il15rアルファヘテロ二量体fc融合タンパク質
WO2021253360A1 (en) * 2020-06-18 2021-12-23 Proviva Therapeutics (Hong Kong) Limited Activatable procytokines
CN113321736B (zh) * 2020-12-30 2024-01-09 苏州复融生物技术有限公司 一种长效化白介素15融合蛋白及其制备方法和应用
CA3206844A1 (en) * 2021-02-05 2022-08-11 John Li Il-15 fusion proteins and methods of making and using same
BR112023027312A2 (pt) * 2021-06-23 2024-03-12 Cytune Pharma Imunocitocina
WO2023015198A1 (en) * 2021-08-04 2023-02-09 Genentech, Inc. Il15/il15r alpha heterodimeric fc-fusion proteins for the expansion of nk cells in the treatment of solid tumours
WO2023160721A1 (en) * 2022-02-28 2023-08-31 Wuxi Biologics (Shanghai) Co., Ltd. Heterodimeric polypeptide complexes comprising il-15 variants and uses thereof
WO2023239228A1 (ko) * 2022-06-10 2023-12-14 메디맵바이오 주식회사 항-tigit 항체, 인터류킨-15, 및 인터류킨-15 수용체 알파 스시 도메인을 포함하는 융합 단백질 및 이의 용도
WO2024011179A1 (en) * 2022-07-07 2024-01-11 Genentech, Inc. Combinations of il15/il15r alpha heterodimeric fc-fusion proteins and fcrh5xcd3 bispecific antibodies for the treatment of blood cancers
WO2024064200A2 (en) * 2022-09-20 2024-03-28 The Texas A&M University System Darpin-containing compositions and methods thereof
CN116554356B (zh) * 2023-05-16 2024-01-23 武汉大学 一种hyperIL-15与sCD4及Fc的融合蛋白及其应用

Family Cites Families (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5595756A (en) * 1993-12-22 1997-01-21 Inex Pharmaceuticals Corporation Liposomal compositions for enhanced retention of bioactive agents
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5811097A (en) 1995-07-25 1998-09-22 The Regents Of The University Of California Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
EP0915987A2 (en) 1997-04-21 1999-05-19 Donlar Corporation POLY-($g(a)-L-ASPARTIC ACID), POLY-($g(a)-L-GLUTAMIC ACID) AND COPOLYMERS OF L-ASP AND L-GLU, METHOD FOR THEIR PRODUCTION AND THEIR USE
ES2308857T3 (es) 1998-12-03 2008-12-01 The Regents Of The University Of California Estimulacion de celulas t contra autoantigenos mediante el uso de agentes bloqueadores de ctla-4.
EA006972B1 (ru) 1998-12-23 2006-06-30 Пфайзер Инк. Моноклональное антитело человека к ctla-4 и способы его применения
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
US7605238B2 (en) 1999-08-24 2009-10-20 Medarex, Inc. Human CTLA-4 antibodies and their uses
US7449443B2 (en) 2000-03-23 2008-11-11 California Institute Of Technology Method for stabilization of proteins using non-natural amino acids
US6586207B2 (en) 2000-05-26 2003-07-01 California Institute Of Technology Overexpression of aminoacyl-tRNA synthetases for efficient production of engineered proteins containing amino acid analogues
EP1490677A4 (en) 2002-02-27 2006-01-18 California Inst Of Techn COMPUTER METHOD FOR DESIGNING ENZYMES FOR THE INCORPORATION OF AMINO ACID ANALOGUES INTO PROTEINS
ES2367430T3 (es) 2002-12-23 2011-11-03 Wyeth Llc Anticuerpos contra pd-1 y sus usos.
ES2737837T3 (es) 2003-10-09 2020-01-16 Ambrx Inc Derivados poliméricos
JP2007519422A (ja) 2004-02-02 2007-07-19 アンブレツクス・インコーポレイテツド 修飾されたヒト四螺旋バンドルポリペプチド及びそれらの使用
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
CN101213297B (zh) 2005-05-09 2013-02-13 小野药品工业株式会社 程序性死亡-1(pd-1)的人单克隆抗体及单独使用或与其它免疫治疗剂联合使用抗pd-1抗体来治疗癌症的方法
KR101411165B1 (ko) 2005-07-01 2014-06-25 메다렉스, 엘.엘.시. 예정 사멸 리간드 1 (피디-엘1)에 대한 인간 모노클로날항체
US9244059B2 (en) 2007-04-30 2016-01-26 Immutep Parc Club Orsay Cytotoxic anti-LAG-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
EP1987839A1 (en) 2007-04-30 2008-11-05 I.N.S.E.R.M. Institut National de la Sante et de la Recherche Medicale Cytotoxic anti-LAG-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
KR101586617B1 (ko) 2007-06-18 2016-01-20 머크 샤프 앤 도메 비.브이. 사람 프로그램된 사멸 수용체 pd-1에 대한 항체
HUE034465T2 (en) 2008-02-11 2018-02-28 Cure Tech Ltd Monoclonal antibodies for tumor treatment
EP2262837A4 (en) 2008-03-12 2011-04-06 Merck Sharp & Dohme PD-1 BINDING PROTEINS
AR072999A1 (es) 2008-08-11 2010-10-06 Medarex Inc Anticuerpos humanos que se unen al gen 3 de activacion linfocitaria (lag-3) y los usos de estos
CN102203125A (zh) 2008-08-25 2011-09-28 安普利穆尼股份有限公司 Pd-1拮抗剂及其使用方法
NZ591130A (en) 2008-08-25 2012-09-28 Amplimmune Inc Compositions comprising a PD-1 antagonists and cyclophosphamide and methods of use thereof
PE20120341A1 (es) 2008-12-09 2012-04-24 Genentech Inc Anticuerpos anti-pd-l1 y su uso para mejorar la funcion de celulas t
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
MX343747B (es) 2009-11-24 2016-11-22 Medimmune Ltd Agentes de union diana contra b7-h1.
TWI629483B (zh) 2010-06-11 2018-07-11 協和醱酵麒麟有限公司 anti-TIM-3 antibody
RS59589B1 (sr) 2010-11-05 2019-12-31 Zymeworks Inc Dizajniranje stabilnog heterodimernog antitela sa mutacijama u fc domenu
RS61033B1 (sr) 2011-11-28 2020-12-31 Merck Patent Gmbh Antitela na pd-l1 i njihova upotreba
AR091649A1 (es) 2012-07-02 2015-02-18 Bristol Myers Squibb Co Optimizacion de anticuerpos que se fijan al gen de activacion de linfocitos 3 (lag-3) y sus usos
WO2014110601A1 (en) 2013-01-14 2014-07-17 Xencor, Inc. Novel heterodimeric proteins
PL3587448T3 (pl) 2013-03-15 2021-11-29 Xencor, Inc. Białka heterodimeryczne
JP6329343B2 (ja) 2013-06-13 2018-05-23 任天堂株式会社 画像処理システム、画像処理装置、画像処理プログラム、および画像処理方法
EP4269441A3 (en) * 2013-08-08 2024-01-24 Cytune Pharma Il-15 and il-15ralpha sushi domain based on modulokines
CA2935599A1 (en) * 2014-01-08 2015-07-16 Shanghai Hengrui Pharmaceutical Co., Ltd. Il-15 heterodimeric protein and uses thereof
JOP20200094A1 (ar) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc جزيئات جسم مضاد لـ pd-1 واستخداماتها
JOP20200096A1 (ar) 2014-01-31 2017-06-16 Children’S Medical Center Corp جزيئات جسم مضاد لـ tim-3 واستخداماتها
US20150259420A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
ME03666B (me) 2014-03-28 2020-10-20 Xencor Inc Bispecifična antitela koja se vezuju za cd38 i cd3
EP3673915A1 (en) * 2014-06-30 2020-07-01 Altor BioScience Corporation Il-15-based molecules and methods of use thereof
BR112017000703A2 (pt) 2014-07-16 2017-11-14 Genentech Inc métodos para tratar ou retardar a progressão do câncer, para reduzir ou inibir a reincidência do câncer, para tratar ou retardar a progressão da imunidade tumoral e para aumentar, intensificar ou estimular uma resposta ou função imune e kit
BR112017007379A2 (pt) 2014-10-14 2017-12-19 Dana Farber Cancer Inst Inc moléculas de anticorpo para pd-l1 e usos das mesmas
MX2017003478A (es) 2014-11-05 2018-02-01 Genentech Inc Anticuerpos anti-fgfr2/3 y metodos para su uso.
US10259887B2 (en) * 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
AU2015369683C1 (en) 2014-12-23 2024-06-20 Bristol-Myers Squibb Company Antibodies to TIGIT
TWI715587B (zh) 2015-05-28 2021-01-11 美商安可美德藥物股份有限公司 Tigit結合劑和彼之用途
CA2994858C (en) 2015-09-25 2024-01-23 Genentech, Inc. Anti-tigit antibodies and methods of use
PE20191034A1 (es) 2016-10-14 2019-08-05 Xencor Inc Proteinas de fusion heterodimericas biespecificas que contienen proteinas de fusion fc il-15/il-15r y fragmentos de anticuerpo pd-1

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11136384B2 (en) * 2016-11-30 2021-10-05 Mereo Biopharma 5, Inc. Methods for treatment of cancer comprising TIGIT-binding agents
US11230596B2 (en) * 2016-11-30 2022-01-25 Mereo Biopharma 5, Inc. Methods for treatment of cancer comprising TIGIT-binding agents
US11932675B2 (en) 2019-10-11 2024-03-19 Genentech, Inc. PD-1 targeted IL-15/IL-15Rα Fc fusion proteins with improved properties
WO2023010031A1 (en) * 2021-07-28 2023-02-02 Genentech, Inc. Il15/il15r alpha heterodimeric fc-fusion proteins for the treatment of blood cancers
CN115806627A (zh) * 2022-08-03 2023-03-17 深圳市先康达生命科学有限公司 一种自分泌IL-15与anti-LAG3结合的融合蛋白及其应用

Also Published As

Publication number Publication date
WO2019204592A8 (en) 2020-11-05
IL278090A (he) 2020-11-30
MA52289A (fr) 2021-02-24
CA3097625A1 (en) 2019-10-24
IL278090B2 (he) 2024-07-01
WO2019204592A1 (en) 2019-10-24
TW202011984A (zh) 2020-04-01
IL278090B1 (he) 2024-03-01
CN112105645A (zh) 2020-12-18
AU2019255744A1 (en) 2020-11-26
MX2020010912A (es) 2021-01-29
JP2021522175A (ja) 2021-08-30
US20220040264A1 (en) 2022-02-10
IL310398A (he) 2024-03-01
SG11202010159RA (en) 2020-11-27
EP3781596A1 (en) 2021-02-24
AU2019255744A8 (en) 2021-04-22
KR20210003170A (ko) 2021-01-11
WO2019204592A9 (en) 2019-12-12

Similar Documents

Publication Publication Date Title
US20220040264A1 (en) Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof
US11524991B2 (en) PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
US20220073588A1 (en) TARGETED HETERODIMERIC Fc FUSION PROTEINS CONTAINING IL-15 IL-15alpha AND ANTIGEN BINDING DOMAINS
US11584794B2 (en) Bispecific heterodimeric fusion proteins containing IL-15-IL-15Ralpha Fc-fusion proteins and immune checkpoint antibody fragments
US11618776B2 (en) Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15RA and NKG2D antigen binding domains
US11851466B2 (en) Targeted IL-12 heterodimeric Fc-fusion proteins
WO2017210485A1 (en) Bispecific antibodies that bind cd20 and cd3 for use in the treatment of lymphoma
US11505595B2 (en) TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains
US20230279071A1 (en) LAG-3 TARGETED HETERODIMERIC FUSION PROTEINS CONTAINING IL-15/IL-15RA Fc-FUSION PROTEINS AND LAG-3 ANTIGEN BINDING DOMAINS
TW202128757A (zh) 具有改善之特性的 PD-1 標靶 IL-15/IL-15Rα FC 融合蛋白
JP7516254B2 (ja) Il-15/il-15raヘテロ二量体fc融合タンパク質およびその使用
TW202400660A (zh) Cd3/bcma/cd38 三特異性抗體

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: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

AS Assignment

Owner name: XENCOR, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BERNETT, MATTHEW J.;RASHID, RUMANA;DESJARLAIS, JOHN;AND OTHERS;SIGNING DATES FROM 20200903 TO 20200916;REEL/FRAME:055556/0013

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE