WO2023178253A2 - Utilisation d'une molécule de liaison anti-pd-l1 multimère en combinaison avec une thérapie par anticorps monoclonal - Google Patents

Utilisation d'une molécule de liaison anti-pd-l1 multimère en combinaison avec une thérapie par anticorps monoclonal Download PDF

Info

Publication number
WO2023178253A2
WO2023178253A2 PCT/US2023/064548 US2023064548W WO2023178253A2 WO 2023178253 A2 WO2023178253 A2 WO 2023178253A2 US 2023064548 W US2023064548 W US 2023064548W WO 2023178253 A2 WO2023178253 A2 WO 2023178253A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
igm
amino acid
monoclonal antibody
acid sequence
Prior art date
Application number
PCT/US2023/064548
Other languages
English (en)
Other versions
WO2023178253A3 (fr
Inventor
Thierry Giffon
Angus SINCLAIR
Roel Funke
Original Assignee
Igm Biosciences, 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 Igm Biosciences, Inc. filed Critical Igm Biosciences, Inc.
Publication of WO2023178253A2 publication Critical patent/WO2023178253A2/fr
Publication of WO2023178253A3 publication Critical patent/WO2023178253A3/fr

Links

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
    • C07K16/2827Immunoglobulins [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 B7 molecules, e.g. CD80, CD86
    • 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
    • C07K16/2863Immunoglobulins [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 growth factors, growth regulators
    • 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
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • 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
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/35Valency
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • P-L1 Programmed cell death protein ligand 1
  • PD-L1 is a ligand and has been shown to downregulate T-cell activation and cytokine secretion upon binding to its receptor programmed cell death protein 1 (PD-1) (Freeman et al. (2000) J Exp Med 192: 1027-34; Latchman et al. (2001) Nat Immunol 2 261-8,' Carter et al. (2002) Eur J Immunol 32:634- 43; Ohigashi etal. (2005) Clin Cancer Res 11 :2947-53).
  • PD-1 programmed cell death protein 1
  • PD-L1 expression has been found in several murine and human cancers, including human lung, ovarian, and colon carcinoma, as well as various myelomas (Iwai et al. (2002) PNAS 99: 12293-7; Ohigashi et al. (2005) Clin Cancer Res 11 :2947-53).
  • PD-L1 has also been suggested to play a role in tumor immunity by increasing apoptosis of antigen-specific T-cell clones (Dong et al. (2002) Nat Med 8:793-800). As such, targeting of the interaction between PD-1 and PD-L1 is an area of particular interest for therapeutic intervention.
  • anti-PD-Ll and anti-PD-1 antibody therapies have been more effective than conventional therapies, the overall response rate (ORR) in two retrospective studies was 10.34%-29% of patients depending on the study and treatment group (PD-L1 negative patient populations had lower ORR). Further, relapse occurred in 87% of patients with stable disease (SD) or partial response (PR). Therefore, there is a need for methods for improving anti-PD-Ll and anti-PD-1 antibody therapies.
  • SD stable disease
  • PR partial response
  • a method for treating cancer in a subject in need of thereof comprising administering to the subject a combination therapy comprising: (a) an effective amount of a multimeric binding molecule comprising five bivalent binding units and a modified J-chain, where the modified J-chain comprises a J-chain or a functional fragment or variant thereof (“J”), an interleukin- 15 (IL-15) protein or receptor-binding fragment or variant thereof (“I”), and an interleukin- 15 receptor-a (IL-15Ra) fragment comprising the sushi domain or a variant thereof capable of associating with I (“R”), where J and at least one of I and R are associated as a fusion protein, where I and R can associate to function as an immunostimulatory agent, where each binding unit comprises two IgM heavy chains, each comprising a heavy chain variable region (VH-IgM) and an IgM constant region and two light chains, each comprising a light chain variable region (VL-IgM) and a light chain constant region,
  • VH-IgM
  • the monoclonal antibody comprises a heavy chain variable region (VH-MA) and a light chain variable region (VL-MA), where the VH-MA and VL- MA comprise six immunoglobulin complementarity determining regions (CDRs): monoclonal antibody heavy chain CDR 1 (HCDR1-MA), monoclonal antibody heavy chain CDR 2 (HCDR2-MA), monoclonal antibody heavy chain CDR 3 (HCDR3-MA), monoclonal antibody light chain CDR 1 (LCDR1-MA), monoclonal antibody light chain CDR 2 (LCDR2-MA), and monoclonal antibody light chain CDR 3 (LCDR3-MA), where the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3- MA comprise, respectively, the amino acid sequence of SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, SEQ ID NO: 209, SEQ ID NO: a heavy chain variable region (
  • the tumor antigen is CD38
  • the HCDR1-MA, HCDR2- MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, SEQ ID NO: 209, SEQ ID NO: 210, and SEQ ID NO: 211; or SEQ ID NO: 214, SEQ ID NO: 215, SEQ ID NO: 216, SEQ ID NO: 217, SEQ ID NO: 218, and SEQ ID NO: 219.
  • the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, SEQ ID NO: 209, SEQ ID NO: 210, and SEQ ID NO: 211.
  • the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 212 and SEQ ID NO: 213; or SEQ ID NO: 220 and SEQ ID NO: 221. In some embodiments, the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 212 and SEQ ID NO: 213.
  • the monoclonal antibody comprises daratumumab or isatuximab. In some embodiments, the monoclonal antibody comprises daratumumab. In some embodiments, the cancer is multiple myeloma.
  • the tumor antigen is HER2
  • the HCDR1-MA, HCDR2- MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 222, SEQ ID NO: 223, SEQ ID NO: 224, SEQ ID NO: 225, SEQ ID NO: 226, and SEQ ID NO: 227; or SEQ ID NO: 230, SEQ ID NO: 231, SEQ ID NO: 232, SEQ ID NO: 233, SEQ ID NO: 234, and SEQ ID NO: 235.
  • the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 222, SEQ ID NO: 223, SEQ ID NO: 224, SEQ ID NO: 225, SEQ ID NO: 226, and SEQ ID NO: 227.
  • the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 228 and SEQ ID NO: 229; or SEQ ID NO: 236 and SEQ ID NO: 237.
  • the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 228 and SEQ ID NO: 229.
  • the monoclonal antibody comprises trastuzumab or pertuzumab. In some embodiments, the monoclonal antibody comprises trastuzumab. In some embodiments, the cancer is breast cancer.
  • the tumor antigen is EGFR
  • the HCDR1-MA, HCDR2- MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 238, SEQ ID NO: 239, SEQ ID NO: 240, SEQ ID NO: 241, SEQ ID NO: 242, and SEQ ID NO: 243; SEQ ID NO: 246, SEQ ID NO: 247, SEQ ID NO: 248, SEQ ID NO: 249, SEQ ID NO: 250, and SEQ ID NO: 251; or SEQ ID NO: 254, SEQ ID NO: 255, SEQ ID NO: 256, SEQ ID NO: 257, SEQ ID NO: 258, and SEQ ID NO: 259.
  • the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 238, SEQ ID NO: 239, SEQ ID NO: 240, SEQ ID NO: 241, SEQ ID NO: 242, and SEQ ID NO: 243.
  • the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 244 and SEQ ID NO: 245; SEQ ID NO: 252 and SEQ ID NO: 253; or SEQ ID NO: 260 and SEQ ID NO: 261.
  • the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 244 and SEQ ID NO: 245.
  • the monoclonal antibody comprises cetuximab, necitumumab, or panitumumab. In some embodiments, the monoclonal antibody comprises cetuximab.
  • the cancer is skin cancer, head and neck cancer, or colorectal cancer.
  • J comprises the amino acid sequence SEQ ID NO: 7 or a functional fragment or variant thereof.
  • J is a variant J-chain or fragment thereof comprising one or more single amino acid substitutions, deletions, or insertions relative to SEQ ID NO: 7 that can affect serum half-life of the multimeric binding molecule; and where the multimeric binding molecule exhibits an increased serum half-life upon administration to an animal relative to a reference multimeric binding molecule that is identical except for the one or more single amino acid substitutions, deletions, or insertions, and is administered in the same way to the same animal species.
  • the J comprises an amino acid substitution at the amino acid position corresponding to amino acid Y102 of the mature wild-type human J-chain (SEQ ID NO: 7).
  • J is a variant human J-chain and comprises the amino acid sequence SEQ ID NO: 8 (“J*”).
  • I comprises the mature human IL- 15 amino acid sequence of SEQ ID NO: 16 or a receptor-binding variant or fragment thereof.
  • I comprises an amino acid substitution at a position corresponding to S73 of SEQ ID NO: 16.
  • the amino acid substitution comprises S73I in SEQ ID NO: 16.
  • I comprises the amino acid sequence SEQ ID NO: 30.
  • R comprises the amino acid sequence SEQ ID NO: 36 or a variant or fragment thereof that is capable of associating with human IL-15.
  • R consists essentially of or consists of the amino acid sequence SEQ ID NO: 36 or a variant thereof that is capable of associating with human IL-15.
  • J, I, and R are associated as a fusion protein.
  • the modified J-chain comprises the amino acid sequence SEQ ID NO: 51.
  • the VH-IgM and VL-IgM comprise six immunoglobulin complementarity determining regions (CDRs): IgM binding molecule heavy chain CDR 1 (HCDRl-IgM), IgM binding molecule heavy chain CDR 2 (HCDR2-IgM), IgM binding molecule heavy chain CDR 3 (HCDR3-IgM), IgM binding molecule light chain CDR 1 (LCDRl-IgM), IgM binding molecule light chain CDR 2 (LCDR2-IgM), and IgM binding molecule light chain CDR 3 (LCDR3-IgM), where the HCDRl-IgM, HCDR2-IgM, HCDR3-IgM, LCDRl-IgM, LCDR2-IgM, and LCDR3-IgM comprise, respectively, the amino acid sequence of SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO:
  • each IgM heavy chain constant region is a human IgM constant region comprising the amino acid sequence SEQ ID NO: 1, SEQ ID NO: 2, or a multimerizing variant or fragment thereof.
  • the multimeric binding molecule comprises a variant human IgM constant region, where the multimeric binding molecule has reduced CDC activity relative to a multimeric binding molecule comprising IgM heavy chain constant regions comprising the amino acid sequence SEQ ID NO: 1, SEQ ID NO: 2, or a multimerizing variant or fragment thereof.
  • each IgM heavy chain constant region comprises a variant of the amino acid sequence SEQ ID NO: 1 or SEQ ID NO: 2, where the variant comprises an amino acid substitution at position K315 of SEQ ID NO: 1 or SEQ ID NO: 2.
  • the amino acid substitution is K315D of SEQ ID NO: 1 or SEQ ID NO: 2.
  • the heavy chain comprises SEQ ID NO: 72
  • the light chain comprises SEQ ID NO: 73
  • the modified J-chain comprises SEQ ID NO: 51.
  • FIG. 1A show the killing curves of RPMI-8226 cells with a combination of PDL 1- IL-15 IGM1 and Daratumumab at various concentration.
  • FIG. IB shows a 3D surface plot of synergy scores for killing RPMI-8226 cells from a continuum of dose combinations of PDL 1 -IL- 15 IGM1 and Daratumumab in vitro, with valleys reflecting antagonism and hills representing synergy.
  • FIG. 2A show the killing curves of SKOV3 cells with a combination of PDL1-IL- 15 IGM1 and Trastuzumab at various concentration.
  • FIG. 2B shows a 3D surface plot of synergy scores for killing SKOV3 cells from a continuum of dose combinations of PDL 1- IL-15 IGM1 and Trastuzumab in vitro, with valleys reflecting antagonism and hills representing synergy.
  • FIG. 3A show the killing curves of A431 cells with a combination of PDL1-IL-15 IGM1 and Cetuximab at various concentration.
  • FIG. 3B shows a 3D surface plot of synergy scores for killing A431 cells from a continuum of dose combinations of PDL1- IL-15 IGM1 and Cetuximab in vitro, with valleys reflecting antagonism and hills representing synergy.
  • FIGS. 4A-4B show tumor volumes over time in a RPMI-8226 xenograft tumor model for vehicle, daratumumab (3 mg/kg), PD-L1-IL-15 IgM, and Daratumumab (3 mg/kg) + PD-L1-IL-15 IgM treated mice (FIG. 4A) and vehicle, daratumumab (10 mg/kg), PD-L1-IL-15 IgM, and Daratumumab (10 mg/kg) + PD-L1-IL-15 IgM (FIG. 4B) treated mice.
  • FIG. 4C shows the individual tumor sizes at Day 55 in all the treatment groups in a RPMI-8226 xenograft tumor model.
  • FIG. 4D shows the Kaplan-Meyer survival plot of all the treatment groups in a RPMI-8226 xenograft tumor model.
  • FIGS. 5A-5B show tumor volumes over time in a SKOV3 xenograft tumor model for vehicle, Trastuzumab (3 mg/kg), PD-L1-IL-15 IgM, and Trastuzumab (3 mg/kg) + PD- Ll-IL-15 IgM treated mice (FIG. 5A) and vehicle, Trastuzumab (10 mg/kg), PD-L1-IL- 15 IgM, and Trastuzumab (10 mg/kg) + PD-L1-IL-15 IgM (FIG. 5B) treated mice.
  • FIG. 5C shows the individual tumor sizes at Day 55 in all the treatment groups in a SKOV3 xenograft tumor model.
  • FIG. 5D shows the Kaplan-Meyer survival plot of all the treatment groups in a SKOV3 xenograft tumor model.
  • FIG. 6 shows tumor volumes over time in a A431 xenograft tumor model for vehicle, Cetuximab (3 or 10 mg/kg), PD-L1-IL-15 IgM, and Cetuximab (3 or 10 mg/kg) + PD-L1-IL-15 IgM treated mice.
  • a or “an” entity refers to one or more of that entity; for example, “a binding molecule,” is understood to represent one or more binding molecules.
  • a binding molecule is understood to represent one or more binding molecules.
  • the terms “a” (or “an”), “one or more,” and “at least one” can be used interchangeably herein.
  • polypeptide is intended to encompass a singular “polypeptide” as well as plural “polypeptides,” and refers to a molecule composed of monomers (amino acids) linearly linked by amide bonds (also known as peptide bonds).
  • polypeptide refers to any chain or chains of two or more amino acids and does not refer to a specific length of the product.
  • polypeptides dipeptides, tripeptides, oligopeptides, “protein,” “amino acid chain,” or any other term used to refer to a chain or chains of two or more amino acids are included within the definition of "polypeptide,” and the term “polypeptide” can be used instead of, or interchangeably with any of these terms.
  • polypeptide is also intended to refer to the products of post-expression modifications of the polypeptide, including without limitation glycosylation, acetylation, phosphorylation, amidation, and derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids.
  • a polypeptide can be derived from a biological source or produced by recombinant technology but is not necessarily translated from a designated nucleic acid sequence. It can be generated in any manner, including by chemical synthesis.
  • a polypeptide as disclosed herein can be of a size of about 3 or more, 5 or more, 10 or more, 20 or more, 25 or more, 50 or more, 75 or more, 100 or more, 200 or more, 500 or more, 1,000 or more, or 2,000 or more amino acids.
  • Polypeptides can have a defined three-dimensional structure, although they do not necessarily have such structure. Polypeptides with a defined three-dimensional structure are referred to as folded, and polypeptides which do not possess a defined three-dimensional structure, but rather can adopt a large number of different conformations and are referred to as unfolded.
  • glycoprotein refers to a protein coupled to at least one carbohydrate moiety that is attached to the protein via an oxygen-containing or a nitrogen-containing side chain of an amino acid, e.g., a serine or an asparagine.
  • an "isolated" polypeptide or a fragment, variant, or derivative thereof is intended a polypeptide that is not in its natural milieu. No particular level of purification is required.
  • an isolated polypeptide can be removed from its native or natural environment.
  • Recombinantly produced polypeptides and proteins expressed in host cells are considered isolated as disclosed herein, as are native or recombinant polypeptides which have been separated, fractionated, or partially or substantially purified by any suitable technique.
  • a non-naturally occurring polypeptide or any grammatical variants thereof, is a conditional definition that explicitly excludes, but only excludes, those forms of the polypeptide that are, or might be, determined or interpreted by a judge or an administrative or judicial body, to be “naturally-occurring.”
  • polypeptides disclosed herein are fragments, derivatives, analogs, or variants of the foregoing polypeptides, and any combination thereof.
  • fragment include any polypeptides which retain at least some of the properties of the corresponding native antibody or polypeptide, for example, specifically binding to an antigen. Fragments of polypeptides include, for example, proteolytic fragments, as well as deletion fragments, in addition to specific antibody fragments discussed elsewhere herein.
  • Variants of, e.g., a polypeptide include fragments as described above, and polypeptides with altered amino acid sequences due to amino acid substitutions, deletions, or insertions.
  • variants can be non-naturally occurring.
  • Non-naturally occurring variants can be produced using art- known mutagenesis techniques.
  • Variant polypeptides can comprise conservative or nonconservative amino acid substitutions, deletions, or additions.
  • Derivatives are polypeptides that have been altered to exhibit additional features not found on the original polypeptide. Examples include fusion proteins.
  • Variant polypeptides can also be referred to herein as "polypeptide analogs.”
  • a "derivative" of a polypeptide can also refer to a subject polypeptide having one or more amino acids chemically derivatized by reaction of a functional side group.
  • derivatives are those peptides that contain one or more derivatives of the twenty standard amino acids.
  • 4-hydroxyproline can be substituted for proline
  • 5-hydroxylysine can be substituted for lysine
  • 3- methylhistidine can be substituted for histidine
  • homoserine can be substituted for serine
  • ornithine can be substituted for lysine.
  • a "conservative amino acid substitution” is one in which one amino acid is replaced with another amino acid having a similar side chain.
  • Families of amino acids having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g.
  • threonine valine, isoleucine
  • aromatic side chains e.g, tyrosine, phenylalanine, tryptophan, histidine.
  • substitution of a phenylalanine for a tyrosine is a conservative substitution.
  • conservative substitutions in the sequences of the polypeptides and antibodies of the present disclosure do not abrogate the binding of the polypeptide or antibody containing the amino acid sequence, to the antigen to which the polypeptide or antibody binds.
  • Methods of identifying nucleotide and amino acid conservative substitutions which do not eliminate antigen-binding are well-known in the art (see, e.g, Brummell et al., Biochem.
  • polynucleotide is intended to encompass a singular nucleic acid as well as plural nucleic acids and refers to an isolated nucleic acid molecule or construct, e.g., messenger RNA (mRNA), cDNA, or plasmid DNA (pDNA).
  • mRNA messenger RNA
  • cDNA plasmid DNA
  • a polynucleotide can comprise a conventional phosphodiester bond or a non-conventional bond (e.g., an amide bond, such as found in peptide nucleic acids (PNA)).
  • PNA peptide nucleic acids
  • nucleic acid or “nucleic acid sequence” refer to any one or more nucleic acid segments, e.g., DNA or RNA fragments, present in a polynucleotide.
  • an "isolated" nucleic acid or polynucleotide is intended any form of the nucleic acid or polynucleotide that is separated from its native environment.
  • gel- purified polynucleotide, or a recombinant polynucleotide encoding a polypeptide contained in a vector would be considered to be “isolated.”
  • a polynucleotide segment e.g., a PCR product, which has been engineered to have restriction sites for cloning is considered to be “isolated.”
  • Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in heterologous host cells or purified (partially or substantially) polynucleotides in a non-native solution such as a buffer or saline.
  • Isolated RNA molecules include in vivo or in vitro RNA transcripts of polynucleotides, where the transcript is not one that would be found in nature. Isolated polynucleotides or nucleic acids further include such molecules produced synthetically.
  • polynucleotide or a nucleic acid can be or can include a regulatory element such as a promoter, ribosome binding site, or a transcription terminator.
  • a non-naturally occurring polynucleotide or any grammatical variants thereof, is a conditional definition that explicitly excludes, but only excludes, those forms of the nucleic acid or polynucleotide that are, or might be, determined or interpreted by a judge, or an administrative or judicial body, to be “naturally-occurring.”
  • a "coding region” is a portion of nucleic acid which consists of codons translated into amino acids. Although a “stop codon” (TAG, TGA, or TAA) is not translated into an amino acid, it can be considered to be part of a coding region, but any flanking sequences, for example promoters, ribosome binding sites, transcriptional terminators, introns, and the like, are not part of a coding region. Two or more coding regions can be present in a single polynucleotide construct, e.g., on a single vector, or in separate polynucleotide constructs, e.g., on separate (different) vectors.
  • any vector can contain a single coding region, or can comprise two or more coding regions, e.g., a single vector can separately encode an immunoglobulin heavy chain variable region and an immunoglobulin light chain variable region.
  • a vector, polynucleotide, or nucleic acid can include heterologous coding regions, either fused or unfused to another coding region.
  • Heterologous coding regions include without limitation, those encoding specialized elements or motifs, such as a secretory signal peptide or a heterologous functional domain.
  • the polynucleotide or nucleic acid is DNA.
  • a polynucleotide comprising a nucleic acid which encodes a polypeptide normally can include a promoter and/or other transcription or translation control elements operably associated with one or more coding regions.
  • An operable association is when a coding region for a gene product, e.g., a polypeptide, is associated with one or more regulatory sequences in such a way as to place expression of the gene product under the influence or control of the regulatory sequence(s).
  • Two DNA fragments are "operably associated" if induction of promoter function results in the transcription of mRNA encoding the desired gene product and if the nature of the linkage between the two DNA fragments does not interfere with the ability of the expression regulatory sequences to direct the expression of the gene product or interfere with the ability of the DNA template to be transcribed.
  • a promoter region would be operably associated with a nucleic acid encoding a polypeptide if the promoter was capable of effecting transcription of that nucleic acid.
  • the promoter can be a cell-specific promoter that directs substantial transcription of the DNA in predetermined cells.
  • Other transcription control elements besides a promoter, for example enhancers, operators, repressors, and transcription termination signals, can be operably associated with the polynucleotide to direct cell-specific transcription.
  • transcription control regions are known to those skilled in the art. These include, without limitation, transcription control regions which function in vertebrate cells, such as, but not limited to, promoter and enhancer segments from cytomegaloviruses (the immediate early promoter, in conjunction with intron-A), simian virus 40 (the early promoter), and retroviruses (such as Rous sarcoma virus).
  • transcription control regions include those derived from vertebrate genes such as actin, heat shock protein, bovine growth hormone and rabbit B-globin, as well as other sequences capable of controlling gene expression in eukaryotic cells.
  • transcription control regions include tissue-specific promoters and enhancers as well as lymphokine-inducible promoters (e.g., promoters inducible by interferons or interleukins).
  • tissue-specific promoters and enhancers as well as lymphokine-inducible promoters (e.g., promoters inducible by interferons or interleukins).
  • lymphokine-inducible promoters e.g., promoters inducible by interferons or interleukins.
  • translation control elements include, but are not limited to ribosome binding sites, translation initiation and termination codons, and elements derived from picomaviruses (particularly an internal ribosome entry site, or IRES, also referred to as a CITE sequence).
  • a polynucleotide can be RNA, for example, in the form of messenger RNA (mRNA), transfer RNA, or ribosomal RNA.
  • mRNA messenger RNA
  • transfer RNA transfer RNA
  • ribosomal RNA RNA
  • Polynucleotide and nucleic acid coding regions can be associated with additional coding regions which encode secretory or signal peptides, which direct the secretion of a polypeptide encoded by a polynucleotide as disclosed herein.
  • proteins secreted by mammalian cells have a signal peptide or secretory leader sequence which is cleaved from the mature protein once export of the growing protein chain across the rough endoplasmic reticulum has been initiated.
  • polypeptides secreted by vertebrate cells can have a signal peptide fused to the N-terminus of the polypeptide, which is cleaved from the complete or "full length" polypeptide to produce a secreted or "mature” form of the polypeptide.
  • the native signal peptide e.g., an immunoglobulin heavy chain or light chain signal peptide is used, or a functional derivative of that sequence that retains the ability to direct the secretion of the polypeptide that is operably associated with it.
  • a heterologous mammalian signal peptide, or a functional derivative thereof can be used.
  • the wild-type leader sequence can be substituted with the leader sequence of human tissue plasminogen activator (TP A) or mouse B-glucuronidase.
  • binding molecule refers in its broadest sense to a molecule that specifically binds to a binding target, e.g., an epitope or an antigenic determinant.
  • a binding molecule can comprise one of more “antigen-binding domains” described herein.
  • a non-limiting example of a binding molecule is an antibody or antibody-like molecule that retains antigen-specific binding, or an antibody-like molecule or fragment thereof as described in detail herein that retains antigen-specific binding.
  • a “binding molecule” comprises an antibody or antibody-like molecule as described in detail herein.
  • binding domain or “antigen-binding domain” (can be used interchangeably) refer to a region or fragment of a binding molecule e.g., an antibody or antibody-like molecule, that is necessary and sufficient to specifically bind to a binding target, e.g., an epitope.
  • an “Fv,” e.g., a heavy chain variable region and a light chain variable region of an antibody, either as two separate polypeptide subunits or as a single chain, is considered to be a “binding domain.”
  • Other antigen-binding domains include, without limitation, the heavy chain variable region (VHH) of an antibody derived from a camelid species, or six immunoglobulin complementarity determining regions (CDRs) expressed in a scaffold, e.g., a fibronectin scaffold.
  • a “binding molecule,” e.g., an antibody or antibody-like molecule as described herein can include one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, or even more “antigen-binding domains.”
  • a “binding unit-associated antigen-binding domain” refers to an antigen binding domain that is part of an antibody heavy chain and/or an antibody light chain.
  • J-chain-associated antigen-binding domain refers to an antigen binding domain that is associated with a modified J-chain as described herein, for example, a scFv fused to a wild-type human J-chain, or functional fragment or variant thereof.
  • antibody and "immunoglobulin” can be used interchangeably herein.
  • An antibody as provided in this disclosure must specifically bind to an antigen, z.e., it includes at least the variable domain of a heavy chain (for camelid species) or at least the variable domains of a heavy chain and a light chain.
  • Basic immunoglobulin structures in vertebrate systems are relatively well understood. See, e.g., Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988).
  • antibody encompasses anything ranging from a small antigenbinding fragment of an antibody to a full sized antibody, e.g., an IgG antibody that includes two IgG heavy chains or fragments thereof and two light chains, an IgA antibody that includes two, four, or eight IgA heavy chains or multimerizing fragments thereof and two, four, or eight light chains and optionally includes a J-chain or functional fragment or variant thereof and/or a secretory component, or an IgM antibody that includes ten or twelve IgM heavy chains or multimerizing fragments thereof and ten or twelve light chains and optionally includes a J-chain or functional fragment or variant thereof.
  • immunoglobulin comprises various broad classes of polypeptides that can be distinguished biochemically. Those skilled in the art will appreciate that heavy chains are classified as, e.g., gamma, mu, alpha, delta, or epsilon, (y, p, a, 6, a) with some subclasses among them (e.g., yl-y4 or al-a2). It is the nature of this chain that determines the "isotype" of the antibody as IgG, IgM, IgA, IgD, or IgE, respectively.
  • immunoglobulin subclasses e.g., IgGi, IgG2, IgGs, IgG4, IgAi, IgA2, etc. are well characterized and are known to confer functional specialization. Modified versions of each of these immunoglobulins are readily discernible to the skilled artisan in view of the instant disclosure and, accordingly, are within the scope of this disclosure.
  • Light chains are classified as either kappa or lambda (K, ). Each heavy chain class can be associated with either a kappa or lambda light chain.
  • the light and heavy chains are covalently bonded to each other via disulfide bonds, and the "tail" portions of the two heavy chains are bonded to each other by covalent disulfide linkages or non- covalent linkages when the immunoglobulins are expressed, e.g., by hybridomas, B cells or genetically engineered host cells.
  • the amino acid sequences run from an N-terminus at the forked ends of the Y configuration to the C-terminus at the bottom of each chain.
  • binding unit is used herein to refer to the portion of a binding molecule, e.g., an antibody or antibody-like molecule that corresponds to a standard “H2L2” immunoglobulin structure, e.g., two heavy chains or fragments thereof and two light chains or fragments thereof.
  • a binding unit can correspond to two heavy chains, e.g., in a cam elid antibody.
  • the terms “binding molecule” and “binding unit” are equivalent.
  • the binding molecule is multimeric, e.g., a dimeric or tetrameric IgA antibody or IgA-like antibody, a pentameric IgM antibody or IgM-like antibody, or a hexameric IgM antibody or IgM-like antibody
  • the binding molecule comprises two or more “binding units.” Two or four in the case of an IgA dimer or tetramer, or five or six in the case of an IgM pentamer or hexamer, respectively.
  • a binding unit need not include full-length antibody heavy and light chains, but will typically be bivalent, i.e., will include two “antigen -binding domains,” as defined above.
  • certain binding molecules provided in this disclosure are “dimeric” or “tetrameric,” and include two or four bivalent binding units that include IgA heavy chain constant regions or multimerizing fragments thereof.
  • Certain binding molecules provided in this disclosure are “pentameric” or “hexameric,” and include five or six bivalent binding units that include IgM heavy chain constant regions or multimerizing fragments thereof.
  • a binding molecule e.g, an antibody or antibody-like molecule comprising two or more, e.g, two, four, five, or six binding units, is referred to herein as “multimeric.”
  • J-chain refers to the J-chain associated with pentameric IgM or dimeric or tetrameric IgA antibodies of any animal species, any functional fragment thereof, derivative thereof, and/or variant thereof, including the mature human J- chain, the amino acid sequence of which is presented as SEQ ID NO: 7.
  • a functional fragment or a “functional variant” includes those fragments and variants that can associate with IgM heavy chain constant regions to form a pentameric IgM antibody (or alternatively can associate with IgA heavy chain constant regions to form a dimeric or tetrameric IgA antibody).
  • modified J-chain is used herein to refer to a derivative of a J-chain polypeptide comprising a heterologous moiety, e.g., a heterologous polypeptide, e.g., an extraneous binding domain introduced into the J-chain polypeptide.
  • the introduction can be achieved by any means, including direct or indirect fusion of the heterologous polypeptide or other moiety or by attachment through a peptide or chemical linker.
  • modified human J-chain encompasses, without limitation, a human J-chain comprising the amino acid sequence of SEQ ID NO: 7 or functional fragment thereof, or functional variant thereof, modified by the introduction of a heterologous moiety, e.g., a heterologous polypeptide, e.g., an extraneous binding domain.
  • a heterologous moiety e.g., a heterologous polypeptide, e.g., an extraneous binding domain.
  • the heterologous moiety does not interfere with efficient polymerization of IgM into a pentamer and binding of such polymers to a target.
  • Exemplary modified J-chains can be found, e.g., in U.S. Patent No. 9,951,134, 10,975,147, 10,400,038, and 10,618,978, and in U.S. Patent Application Publication No. US-2019-0185570, each of which is incorporated herein by reference in its entirety.
  • IgM-derived binding molecule As used herein, the terms “IgM-derived binding molecule,” “IgM-like antibody,” “IgM-like binding unit,” or “IgM-like heavy chain constant region” refer to a variant antibody-derived binding molecule, antibody, binding unit, or heavy chain constant region that still retains the structural portions of an IgM heavy chain necessary to confer the ability to bind to antigen and to form multimers, /. ⁇ ., hexamers, or in association with J-chain, form pentamers.
  • An IgM-like antibody or IgM-derived binding molecule typically includes at least the Cp4 and p tailpiece (ptp) domains of the IgM constant region and an antigen binding domain or subunit thereof but can include heavy chain constant region domains from other antibody isotypes, e.g, IgG, from the same species or from a different species.
  • An IgM-like antibody or IgM-derived binding molecule can likewise be an antibody fragment in which one or more constant regions are deleted, as long as the IgM- like antibody is capable of binding antigen and of forming hexamers and/or pentamers.
  • an IgM-like antibody or IgM-derived binding molecule can be, e.g, a hybrid IgM/IgG antibody or can be a “multimerizing fragment” of an IgM antibody.
  • the terms “valency,” “bivalent,” “monovalent,” “multivalent” and grammatical equivalents, refer to the number of antigen-binding domains in given binding molecule, e.g., an antibody or antibody-like molecule or in a given binding unit.
  • bivalent in reference to a given binding molecule, e.g., an IgM antibody, IgM-like antibody or multimerizing fragment thereof, denote the presence of two antigen-binding domains, four antigen-binding domains, and six antigenbinding domains, respectively.
  • a typical IgM antibody or IgM-like antibody or IgM- derived binding molecule where each binding unit is bivalent can have 10 or 12 valencies.
  • a bivalent or multivalent binding molecule e.g., antibody or antibody-like molecule
  • epitope includes any molecular determinant capable of specific binding to an antigen-binding domain of an antibody or antibody-like molecule.
  • an epitope can include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, can have three-dimensional structural characteristics, and or specific charge characteristics.
  • An epitope is a region of a target that is bound by an antigen-binding domain of an antibody.
  • target is used in the broadest sense to include substances that can be bound by a binding molecule, e.g., an antibody or antibody -like molecule.
  • a target can be, e.g., a polypeptide, a nucleic acid, a carbohydrate, a lipid, or other molecule.
  • a “target” can, for example, be a cell, an organ, or an organism that comprises an epitope that can be bound by a binding molecule, e.g., an antibody or antibody-like molecule.
  • Both the light and heavy chains of an antibody or antibody-like molecule are divided into regions of structural and functional homology.
  • the terms "constant” and “variable” are used functionally but refer to particular structures of the molecule.
  • the variable regions of both the light (VL) and heavy (VH) chains determine antigen recognition and specificity.
  • the constant domains of the light chain (CL) and the heavy chain e.g., CHI, CH2, CH3, or CH4 confer biological properties such as the ability to multimerize, secretion, transplacental mobility, Fc receptor binding, complement binding, and the like.
  • the numbering of the constant region domains increases as they become more distal from the antigen-binding regions or amino-terminus of the antibody.
  • the N-terminal portion is a variable region and at the C-terminal portion is a constant region; the CH3 (or CH4 in the case of IgM) and CL domains actually comprise the carboxy -terminus of the heavy and light chain, respectively.
  • a “full length IgM antibody heavy chain” is a polypeptide that includes, in N- terminal to C terminal direction, an antibody heavy chain variable domain (VH), an antibody heavy chain constant domain 1 (CM1 or Cpl), an antibody heavy chain constant domain 2 (CM2 or Cp2), an antibody heavy chain constant domain 3 (CM3 or Cp3), and an antibody heavy chain constant domain 4 (CM4 or Cp4) that can include a p tailpiece.
  • VH antibody heavy chain variable domain
  • CM1 or Cpl an antibody heavy chain constant domain 1
  • CM2 or Cp2 an antibody heavy chain constant domain 2
  • CM3 or Cp3 an antibody heavy chain constant domain 3
  • CM4 or Cp4 an antibody heavy chain constant domain 4
  • variable region(s) form the antigen-binding domain of the antibody or antibody-like molecule, allowing it to selectively recognize and specifically bind epitopes on antigens. That is, the VL domain and VH domain, or an antigen-binding subset of the complementarity determining regions (CDRs), of a binding molecule, e.g., an antibody or antibody -like molecule combine to form the antigen-binding domain. More specifically, an antigen-binding domain can be defined by three CDRs on each of the VH and VL chains. Certain antibodies or antibody-like molecules form larger structures. For example, IgM heavy chains can form a pentameric or hexameric molecule that includes five or six H2L2 binding units and optionally a J-chain covalently connected via disulfide bonds.
  • the six “complementarity determining regions” or “CDRs” present in an antibody antigen-binding domain are short, non-contiguous sequences of amino acids that are specifically positioned to form the antigen-binding domain as the antibody assumes its three-dimensional configuration in an aqueous environment.
  • the remainder of the amino acids in the antigen-binding domain referred to as "framework" regions, show less inter- molecular variability.
  • the framework regions largely adopt a > -sheet conformation and the CDRs form loops that connect, and in some cases form part of, the > -sheet structure.
  • framework regions act to form a scaffold that provides for positioning the CDRs in correct orientation by inter-chain, non-covalent interactions.
  • the antigen-binding domain formed by the positioned CDRs defines a surface complementary to the epitope on the target antigen. This complementary surface promotes the non-covalent binding of the antibody to its cognate epitope.
  • the amino acids that make up the CDRs and the framework regions, respectively, can be readily identified for any given heavy or light chain variable region by one of ordinary skill in the art, since they have been defined in various different ways (see, e.g., "Sequences of Proteins of Immunological Interest," Kabat, E., etal., U.S. Department of Health and Human Services, (1983); and Chothia and Lesk, J. Mol. Biol., 796:901-917 (1987), which are incorporated herein by reference in their entireties).
  • CDR complementarity determining region
  • the Kabat and Chothia definitions include overlapping or subsets of amino acids when compared against each other. Nevertheless, application of either definition (or other definitions known to those of ordinary skill in the art) to refer to a CDR of an antibody or variant thereof is intended to be within the scope of the term as defined and used herein, unless otherwise indicated.
  • the appropriate amino acids which encompass the CDRs as defined by each of the above cited references are set forth below in Table 1 as a comparison. The exact amino acid numbers which encompass a particular CDR will vary depending on the sequence and size of the CDR. Those skilled in the art can routinely determine which amino acids comprise a particular CDR given the variable region amino acid sequence of the antibody.
  • Antibody variable domains can also be analyzed, e.g., using the IMGT information system (imgt_dot_cines_dot_fr/) (IMGT®/V-Quest) to identify variable region segments, including CDRs.
  • IMGT information system IMGT®/V-Quest
  • Kabat et al. also defined a numbering system for variable region and constant region sequences that is applicable to any antibody.
  • One of ordinary skill in the art can unambiguously assign this system of "Kabat numbering" to any variable region sequence, without reliance on any experimental data beyond the sequence itself.
  • Kabat numbering refers to the numbering system set forth by Kabat et al.. U.S. Dept, of Health and Human Services, "Sequence of Proteins of Immunological Interest" (1983). Unless use of the Kabat numbering system is explicitly noted, however, consecutive numbering is used for all amino acid sequences in this disclosure.
  • the Kabat numbering system for the human IgM constant domain can be found in Kabat, et al. “Tabulation and Analysis of Amino acid and nucleic acid Sequences of Precursors, V-Regions, C-Regions, J-Chain, T-Cell Receptors for Antigen, T-Cell Surface Antigens, ⁇ -2 Microglobulins, Major Histocompatibility Antigens, Thy-1, Complement, C-Reactive Protein, Thymopoietin, Integrins, Post-gamma Globulin, a-2 Macroglobulins, and Other Related Proteins,” U.S. Dept, of Health and Human Services (1991).
  • IgM constant regions can be numbered sequentially (i.e., amino acid #1 starting with the first amino acid of the constant region, or by using the Kabat numbering scheme.
  • SEQ ID NO: 1 allele IGHM*03
  • SEQ ID NO: 2 allele IGHM*04
  • Binding molecules e.g., antibodies, antibody-like molecules, antigen-binding fragments, variants, or derivatives thereof, and/or multimerizing fragments thereof include, but are not limited to, polyclonal, monoclonal, human, humanized, or chimeric antibodies, single chain antibodies, epitope-binding fragments, e.g., Fab, Fab' and F(ab')2, Fd, Fvs, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv), fragments comprising either a VL or VH domain, fragments produced by a Fab expression library. ScFv molecules are described, e.g., in US patent 5,892,019.
  • a binding molecule e.g., an antibody or antibody-like molecule binds to an epitope via its antigen-binding domain, and that the binding entails some complementarity between the antigen-binding domain and the epitope.
  • a binding molecule e.g. , an antibody or antibodylike molecule is said to "specifically bind” to an epitope when it binds to that epitope, via its antigen-binding domain more readily than it would bind to a random, unrelated epitope.
  • the term "specificity" is used herein to qualify the relative affinity by which a certain binding molecule binds to a certain epitope.
  • binding molecule "A” can be deemed to have a higher specificity for a given epitope than binding molecule "B,” or binding molecule “A” can be said to bind to epitope “C” with a higher specificity than it has for related epitope "D.”
  • a binding molecule e.g., an antibody or fragment, variant, or derivative thereof disclosed herein can be said to bind a target antigen with an off rate (k(off)) of less than or equal to 5 X 10' 2 sec' 1 , 10' 2 sec' 1 , 5 X 10' 3 sec' 1 , 10' 3 sec' 1 , 5 X 10' 4 sec' 1 , 10' 4 sec' 1 , 5 X 10' 5 sec' 1 , 10' 5 sec' 1 , 5 X 10' 6 sec' 1 , 10' 6 sec' 1 , 5 X 10' 7 sec' 1 or 10' 7 sec' 1 .
  • off rate k(off)
  • a binding molecule e.g., an antibody or antibody-like molecule disclosed herein can be said to bind a target antigen with an on rate (k(on)) of greater than or equal to 10 3 M' 1 sec' 1 , 5 X 10 3 M' 1 sec' 1 , 10 4 M' 1 sec' 1 , 5 X 10 4 M' 1 sec' 1 , 10 5 M' 1 sec' 1 , 5 X 10 5 M' 1 sec' l, 10 6 M' 1 sec' 1 , 5 X 10 6 M' 1 sec' 1 , or 10 7 M' 1 sec' 1 .
  • k(on) on rate
  • a binding molecule e.g., an antibody or antibody-like molecule is said to competitively inhibit binding of a reference antibody or antibody-like molecule to a given epitope if it preferentially binds to that epitope to the extent that it blocks, to some degree, binding of the reference antibody or antigen-binding fragment to the epitope.
  • Competitive inhibition can be determined by any method known in the art, for example, competition ELISA assays or OCTET assays.
  • a binding molecule can be said to competitively inhibit binding of the reference antibody or antibody-like molecule to a given epitope by at least 90%, at least 80%, at least 70%, at least 60%, or at least 50%.
  • the term "affinity” refers to a measure of the strength of the binding of an individual epitope with one or more antigen -binding domains, e.g., of an antibody or antibody-like molecule. See, e.g., Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988) at pages 27-28.
  • the term “avidity” refers to the overall stability of the complex between a population of antigenbinding domains and an antigen. See, e.g., Harlow at pages 29-34.
  • Avidity is related to both the affinity of individual antigen-binding domains in the population with specific epitopes, and the valencies of the immunoglobulins and the antigen.
  • the interaction between a bivalent monoclonal antibody and an antigen with a highly repeating epitope structure, such as a polymer would be one of high avidity.
  • the interaction between a multimeric antibody with four, eight, ten, or twelve valencies and a population of specific epitopes would be one of high avidity.
  • An interaction between a bivalent monoclonal antibody with a receptor present at a high density on a cell surface would also be of high avidity.
  • Binding molecules e.g., antibodies or fragments, variants, or derivatives thereof as disclosed herein can also be described or specified in terms of their cross-reactivity.
  • cross-reactivity refers to the ability of a binding molecule, e.g., an antibody or fragment, variant, or derivative thereof, specific for one antigen, to react with a second antigen; a measure of relatedness between two different antigenic substances.
  • a binding molecule is cross reactive if it binds to an epitope other than the one that induced its formation.
  • the cross-reactive epitope generally contains many of the same complementary structural features as the inducing epitope, and in some cases, can actually fit better than the original.
  • a binding molecule e.g., an antibody or fragment, variant, or derivative thereof can also be described or specified in terms of their binding affinity to an antigen.
  • a binding molecule can bind to an antigen with a dissociation constant or KD no greater than 5 x IO' 2 M, IO' 2 M, 5 x 10' 3 M, 10' 3 M, 5 x IO' 4 M, IO' 4 M, 5 x 10' 5 M, 10' 5 M, 5 x 10' 6 M, IO' 6 M, 5 x IO' 7 M, IO' 7 M, 5 x IO' 8 M, IO' 8 M, 5 x IO' 9 M, IO' 9 M, 5 x IO' 10 M, IO' 10 M, 5 x 10' 11 M, 10' 11 M, 5 x IO' 12 M, IO' 12 M, 5 x 10' 13 M, 10' 13 M, 5 x IO' 14 M, IO' 14 M
  • Antigen-binding fragments of a binding molecule or antibody as provided herein including single-chain antibodies or other antigen-binding domains that can exist alone or in combination with one or more of the following: hinge region, CHI, CH2, CH3, or CH4 domains, J-chain, or secretory component. Also included are antigen-binding fragments that can include any combination of variable region(s) sufficient to bind antigen with one or more of a hinge region, CHI, CH2, CH3, or CH4 domains, a J-chain, or a secretory component. Binding molecules, e.g., antibodies or antibody-like molecules can be from any animal origin including birds and mammals.
  • the antibodies can be, e.g., human, murine, donkey, rabbit, goat, guinea pig, camel, llama, horse, or chicken antibodies.
  • the variable region can be condricthoid in origin (e.g., from sharks).
  • "human” antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulins and can in some instances express endogenous immunoglobulins and some not, as described infra and, for example in, U.S. Pat. No. 5,939,598 by Kucherlapati et al.
  • an IgM or IgM-like antibody or IgM-derived binding molecule as provided herein can include an antigen-binding fragment of an antibody, e.g., a scFv, so long as the IgM or IgM-like antibody is able to form a multimer, e.g., a hexamer or a pentamer.
  • an antigen-binding fragment of an antibody e.g., a scFv
  • the term “heavy chain subunit” includes amino acid sequences derived from an immunoglobulin heavy chain.
  • a binding molecule e.g., an antibody or antibody-like molecule comprising a heavy chain subunit can include a VH domain and one or more of a CHI domain, a hinge (e.g., upper, middle, and/or lower hinge region) domain, a CH2 domain, a CH3 domain, a CH4 domain, a p tail-piece (ptp), or a variant or fragment thereof.
  • a binding molecule e.g., an antibody, antibody-like molecule, or fragment, variant, or derivative thereof can include without limitation, in addition to a VH domain, any combination of a CHI domain, a hinge, a CH2 domain; a CH3 domain; a CH4 domain; or a p tailpiece (ptp) of one or more antibody isotypes and/or species.
  • a VH domain any combination of a CHI domain, a hinge, a CH2 domain; a CH3 domain; a CH4 domain; or a p tailpiece (ptp) of one or more antibody isotypes and/or species.
  • a binding molecule e.g., an antibody, antibody-like molecule, or fragment, variant, or derivative thereof can include, in addition to a VH domain, one or more of a CHI domain, a CH2 domain, a CH3 domain, a CH4 domain, a p-tailpiece (ptp) domain and a J-chain.
  • a binding molecule e.g., antibody or antibody-like molecule provided in the disclosure can lack certain constant region portions, e.g., all or part of a CHI domain, a hinge, a CH2 domain, or a CH3 domain.
  • an IgM or IgM-like antibody as provided herein includes sufficient portions of an IgM heavy chain constant region to allow the IgM or IgM-like antibody to form a multimer, e.g., a hexamer or a pentamer, e.g., the IgM heavy chain constant region includes a “multimerizing fragment” of an IgM heavy chain constant region.
  • the term “light chain subunit” includes amino acid sequences derived from an immunoglobulin light chain.
  • the light chain subunit includes at least a VL, and can further include a CL (e.g., CK or CX domain.
  • Binding molecules e.g, antibodies, antibody-like molecules, antigen-binding fragments, variants, or derivatives thereof, or multimerizing fragments thereof can be described or specified in terms of the epitope(s) or portion(s) of an antigen that they recognize or specifically bind.
  • the portion of a target antigen that specifically interacts with the antigen-binding domain of an antibody is an "epitope," or an "antigenic determinant.”
  • a target antigen can comprise a single epitope or two or more epitopes, and can include any number of epitopes, depending on the size, conformation, and type of antigen.
  • hinge region includes the portion of a heavy chain molecule that joins the CHI domain to the CH2 domain in IgG, IgA, and IgD heavy chains. This hinge region comprises approximately 25 amino acids and is flexible, thus allowing the two N-terminal antigen-binding regions to move independently.
  • disulfide bond includes the covalent bond formed between two sulfur atoms.
  • the amino acid cysteine comprises a thiol group that can form a disulfide bond or bridge with a second thiol group.
  • chimeric antibody refers to an antibody in which the immunoreactive region or site is obtained or derived from a first species and the constant region (which can be intact, partial, or modified) is obtained from a second species.
  • the target binding region or site will be from a non-human source (e.g., mouse or primate) and the constant region is human.
  • multispecific antibody or “bispecific antibody” refer to an antibody or antibody-like molecule that has antigen-binding domains for two or more different epitopes within a single antibody molecule.
  • Other binding molecules in addition to the canonical antibody structure can be constructed with two binding specificities.
  • the term “engineered antibody” refers to an antibody in which the variable domain in either the heavy and light chain or both is altered by at least partial replacement of one or more amino acids in either the CDR or framework regions.
  • entire CDRs from an antibody of known specificity can be grafted into the framework regions of a heterologous antibody.
  • alternate CDRs can be derived from an antibody of the same class or even subclass as the antibody from which the framework regions are derived, CDRs can also be derived from an antibody of different class, e.g., from an antibody from a different species.
  • an engineered antibody in which one or more "donor" CDRs from a non-human antibody of known specificity are grafted into a human heavy or light chain framework region is referred to herein as a "humanized antibody.”
  • a humanized antibody In certain embodiments, not all the CDRs are replaced with the complete CDRs from the donor variable region and yet the antigen-binding capacity of the donor can still be transferred to the recipient variable domains. Exemplary methods of humanization are described in U.S. Pat. Nos. 5,585,089, 5,693,761, 5,693,762, and 6,180,370.
  • engineered includes manipulation of nucleic acid or polypeptide molecules by synthetic means (e.g., by recombinant techniques, in vitro peptide synthesis, by enzymatic or chemical coupling of peptides, nucleic acids, or glycans, or some combination of these techniques).
  • in-frame fusion refers to the joining of two or more polynucleotide open reading frames (ORFs) to form a continuous longer ORF, in a manner that maintains the translational reading frame of the original ORFs.
  • a recombinant fusion protein is a single protein containing two or more segments that correspond to polypeptides encoded by the original ORFs (which segments are not normally so joined in nature.) Although the reading frame is thus made continuous throughout the fused segments, the segments can be physically or spatially separated by, for example, in-frame linker sequence.
  • polynucleotides encoding the CDRs of an immunoglobulin variable region can be fused, in-frame, but be separated by a polynucleotide encoding at least one immunoglobulin framework region or additional CDR regions, as long as the "fused" CDRs are co-translated as part of a continuous polypeptide.
  • the term “associated” and grammatical equivalents refers to the interaction of two or more elements function together and that can be linked or fused, but can also be in proximity, e.g., interacting in trans without being connected in any particular way.
  • a "linear sequence” or a “sequence” is an order of amino acids in a polypeptide in an amino to carboxyl terminal direction in which amino acids that neighbor each other in the sequence are contiguous in the primary structure of the polypeptide.
  • a portion of a polypeptide that is “amino-terminal” or “N-terminal” to another portion of a polypeptide is that portion that comes earlier in the sequential polypeptide chain.
  • a portion of a polypeptide that is “carboxy -terminal” or “C- terminal” to another portion of a polypeptide is that portion that comes later in the sequential polypeptide chain.
  • the variable domain is “N-terminal” to the constant region
  • the constant region is “C-terminal” to the variable domain.
  • expression refers to a process by which a gene produces a biochemical, for example, a polypeptide.
  • the process includes any manifestation of the functional presence of the gene within the cell including, without limitation, gene knockdown as well as both transient expression and stable expression. It includes without limitation transcription of the gene into RNA, e.g., messenger RNA (mRNA), and the translation of such mRNA into polypeptide(s). If the final desired product is a biochemical, expression includes the creation of that biochemical and any precursors.
  • RNA messenger RNA
  • a gene product can be either a nucleic acid, e.g., a messenger RNA produced by transcription of a gene, or a polypeptide that is translated from a transcript.
  • Gene products described herein further include nucleic acids with post transcriptional modifications, e.g., polyadenylation, or polypeptides with post translational modifications, e.g., methylation, glycosylation, the addition of lipids, association with other protein subunits, proteolytic cleavage, and the like.
  • Terms such as “treating” or “treatment” or “to treat” or “alleviating” or “to alleviate” refer to therapeutic measures that cure, slow down, lessen symptoms of, and/or halt or slow the progression of an existing diagnosed disease, pathologic condition, or disorder.
  • Terms such as “prevent,” “prevention,” “avoid,” “deterrence” and the like refer to prophylactic or preventative measures that prevent the development of an undiagnosed targeted disease, pathologic condition, or disorder.
  • “a subject need of treatment” can include those already with the disease, pathologic condition.
  • the term “a subject in need of prevention” those subjects prone to have the disease, pathologic condition, or disorder and those in whom the disease, pathologic condition or disorder is to be prevented.
  • the terms “serum half-life” or “plasma half-life” refer to the time it takes (e.g., in minutes, hours, or days) following administration for the serum or plasma concentration of a protein or a drug, e.g., a binding molecule such as an antibody or antibody-like molecule as described herein, to be reduced by 50%.
  • Two half-lives can be described: the alpha half-life, a half-life, or ti/2a, which is the rate of decline in plasma concentrations due to the process of drug redistribution from the central compartment, e.g., the blood in the case of intravenous delivery, to a peripheral compartment (e.g. , a tissue or organ), and the beta half-life, P half-life, or ti/2p which is the rate of decline due to the processes of excretion or metabolism.
  • AUC area under the plasma drug concentration-time curve
  • MRT mean residence time
  • subject or “individual” or “animal” or “patient” or “mammal,” is meant any mammalian subject, for whom diagnosis, prognosis, or therapy is desired.
  • Mammalian subjects include humans, domestic animals, farm animals, and zoo, sports, or pet animals such as dogs, cats, guinea pigs, rabbits, rats, mice, horses, swine, cows, bears, and so on.
  • a method for treating cancer in a subject in need of thereof comprising administering to the subject a combination therapy comprising an effective amount of a multimeric binding molecule as described elsewhere herein, and an effective amount of a monoclonal antibody, where the monoclonal antibody specifically binds a tumor antigen, wherein the tumor antigen is cluster of differentiation 38 (CD38), human epidermal growth factor receptor 2 (HER2), or epidermal growth factor receptor (EGFR).
  • HER2 human epidermal growth factor receptor 2
  • EGFR epidermal growth factor receptor
  • the target antigen is CD38.
  • CD38 is expressed in various cancers, such as various hematologic malignancies including chronic lymphocytic leukemia (CLL), multiple myeloma (MM), Hodgkin’s lymphoma (HL), diffuse large B- cell lymphoma (DLBCL), and peripheral T-cell lymphoma (PTCL), and various solid tumors, including prostate cancer, non-small cell lung cancer (NSCLC), squamous cell carcinoma of the head and neck (SCCHN), ovarian cancer, and liver cancer and is therefore considered a potential target for directed therapeutics (Martin et al., Cells, 2019, 8: 1522).
  • Various anti-CD38 antibodies have been described in U.S.
  • MM Multiple Myeloma
  • the monoclonal antibody heavy chain variable region (VH- MA) comprises the CDRs of the heavy chain variable region (VH) of daratumumab and the monoclonal antibody light chain variable region (VL-MA) comprises the CDRs of the light chain variable region (VL) of daratumumab.
  • the VH-MA comprises the VH of daratumumab and the VL-MA comprises the VL of daratumumab.
  • the monoclonal antibody heavy chain variable region comprises the CDRs of the heavy chain variable region of isatuximab and the monoclonal antibody light chain variable region (VL-MA) comprises the CDRs of the light chain variable region of isatuximab.
  • the VH-MA comprises the VH of isatuximab and the VL-MA comprises the VL of isatuximab.
  • the VH-MA comprise immunoglobulin complementarity determining regions (CDRs): HCDR1-MA, HCDR2-MA, and HCDR3-MA and the VL- MA comprise immunoglobulin CDRs: LCDR1-MA, LCDR2-MA, and LCDR3-MA, where the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, SEQ ID NO: 209, SEQ ID NO: 210, and SEQ ID NO: 211; or SEQ ID NO: 214, SEQ ID NO: 215, SEQ ID NO: 216, SEQ ID NO: 217, SEQ ID NO: 218, and SEQ ID NO: 219.
  • CDRs immunoglobulin complementarity determining regions
  • the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, SEQ ID NO: 209, SEQ ID NO: 210, and SEQ ID NO: 211.
  • the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 212 and SEQ ID NO: 213; or SEQ ID NO: 220 and SEQ ID NO: 221.
  • the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 212 and SEQ ID NO: 213.
  • the monoclonal antibody comprises daratumumab or isatuximab.
  • the cancer to be treated is a cancer in which the malignant cells express or over-express CD38.
  • the cancer is a hematological cancer, such as chronic lymphocytic leukemia (CLL), multiple myeloma (MM), Hodgkin’s lymphoma (HL), diffuse large B-cell lymphoma (DLBCL), peripheral T-cell lymphoma (PTCL), and various solid tumors, including prostate cancer, non-small cell lung cancer (NSCLC), squamous cell carcinoma of the head and neck (SCCHN), ovarian cancer, and liver cancer.
  • the cancer is a hematological cancer.
  • the cancer is MM.
  • the target antigen is human epidermal growth factor receptor 2 (HER2).
  • HER2 is overexpressed in various cancers, such as cancers of the stomach, ovary, uterine serous endometrial carcinoma, colon, bladder, lung, uterine cervix, head and neck, and esophagus, but is most studied in breast cancer where it is overexpressed in 15- 30% of invasive breast cancers (Igbal et al., Mol Biol Int. 2014; 2014: 852748, doi: 10.1155/2014/852748).
  • Various anti-HER.2 antibodies have been described in U.S. Patent 7,435,797, U.S. Patent Application Publication No. 2003/0086924, and PCT Application Publication No. WO 2011/107957, which are incorporated herein by reference.
  • Two anti- HER.2 IgG antibodies, trastuzumab and pertuzumab have been approved for the treatment of HER2-positive breast cancer.
  • the monoclonal antibody heavy chain variable region (VH- MA) comprises the CDRs of the heavy chain variable region (VH) of trastuzumab and the monoclonal antibody light chain variable region (VL-MA) comprises the CDRs of the light chain variable region (VL) of trastuzumab.
  • the VH-MA comprises the VH of trastuzumab and the VL-MA comprises the VL of trastuzumab.
  • the monoclonal antibody heavy chain variable region comprises the CDRs of the heavy chain variable region of pertuzumab and the monoclonal antibody light chain variable region (VL-MA) comprises the CDRs of the light chain variable region of pertuzumab.
  • the VH-MA comprises the VH of pertuzumab and the VL-MA comprises the VL of pertuzumab.
  • the VH-MA comprise immunoglobulin complementarity determining regions (CDRs): HCDR1-MA, HCDR2-MA, and HCDR3-MA and the VL-MA comprise immunoglobulin CDRs: LCDR1-MA, LCDR2-MA, and LCDR3-MA, where the HCDR1-MA, HCDR2- MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 222, SEQ ID NO: 223, SEQ ID NO: 224, SEQ ID NO: 225, SEQ ID NO: 226, and SEQ ID NO: 227; or SEQ ID NO: 230, SEQ ID NO: 231, SEQ ID NO: 232, SEQ ID NO: 233, SEQ ID NO: 234, and SEQ ID NO: 235.
  • CDRs immunoglobulin complementarity determining regions
  • the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 222, SEQ ID NO: 223, SEQ ID NO: 224, SEQ ID NO: 225, SEQ ID NO: 226, and SEQ ID NO: 227.
  • the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 228 and SEQ ID NO: 229; or SEQ ID NO: 236 and SEQ ID NO: 237.
  • the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 228 and SEQ ID NO: 229.
  • the monoclonal antibody comprises trastuzumab or pertuzumab.
  • the cancer to be treated is a cancer in which the malignant cells express or over-express HER2.
  • the cancer is a stomach cancer, ovarian cancer, uterine cancer, colorectal cancer, bladder cancer, lung cancer, head and neck cancer, or esophageal cancer.
  • the cancer is breast cancer.
  • the target antigen is epidermal growth factor receptor (EGFR).
  • EGFR is expressed in various cancers, including skin cancer, colorectal cancer, head and neck cancer, lung cancer, and pancreatic cancer.
  • Various anti-EGFR antibodies have been described in U.S. Patents 7,598,350 and 9,109,026, and U.S. Patent Application Publication 2005-0142133, which are incorporated herein by reference.
  • Three anti-EGFR IgG antibodies, cetuximab, necitumumab, and panitumumab have been approved for the treatment of squamous non-small-cell lung carcinoma (NSCLC), squamous cell carcinoma of the head and neck (SCCHN), or colorectal cancer.
  • NSCLC non-small-cell lung carcinoma
  • SCCHN squamous cell carcinoma of the head and neck
  • colorectal cancer colorectal cancer.
  • the monoclonal antibody heavy chain variable region (VH- MA) comprises the CDRs of the heavy chain variable region (VH) of trastuzumab and the monoclonal antibody light chain variable region (VL-MA) comprises the CDRs of the light chain variable region (VL) of cetuximab.
  • the VH-MA comprises the VH of cetuximab and the VL-MA comprises the VL of cetuximab.
  • the monoclonal antibody heavy chain variable region (VH-MA) comprises the CDRs of the heavy chain variable region of necitumumab and the monoclonal antibody light chain variable region (VL-MA) comprises the CDRs of the light chain variable region of necitumumab.
  • the VH-MA comprises the VH of necitumumab and the VL-MA comprises the VL of necitumumab.
  • the monoclonal antibody heavy chain variable region (VH-MA) comprises the CDRs of the heavy chain variable region of panitumumab and the monoclonal antibody light chain variable region (VL-MA) comprises the CDRs of the light chain variable region of panitumumab.
  • the VH-MA comprises the VH of panitumumab and the VL-MA comprises the VL of panitumumab.
  • the VH-MA comprise immunoglobulin complementarity determining regions (CDRs): HCDR1-MA, HCDR2-MA, and HCDR3-MA and the VL- MA comprise immunoglobulin CDRs: LCDR1-MA, LCDR2-MA, and LCDR3-MA, where the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 238, SEQ ID NO: 239, SEQ ID NO: 240, SEQ ID NO: 241, SEQ ID NO: 242, and SEQ ID NO: 243; SEQ ID NO: 246, SEQ ID NO: 247, SEQ ID NO: 248, SEQ ID NO: 249, SEQ ID NO: 250, and SEQ ID NO: 251; or SEQ ID NO: 254, SEQ ID NO: 255, SEQ ID NO: 256, SEQ ID NO: 257
  • CDRs complementarity
  • the HCDR1- MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 238, SEQ ID NO: 239, SEQ ID NO: 240, SEQ ID NO: 241, SEQ ID NO: 242, and SEQ ID NO: 243.
  • the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 244 and SEQ ID NO: 245; SEQ ID NO: 252 and SEQ ID NO: 253; or SEQ ID NO: 260 and SEQ ID NO: 261.
  • the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 244 and SEQ ID NO: 245.
  • the monoclonal antibody comprises cetuximab, necitumumab, or panitumumab. In some embodiments, the monoclonal antibody comprises cetuximab.
  • the cancer to be treated is a cancer in which the malignant cells express or over-express EGFR.
  • the cancer is skin cancer, colorectal cancer, head and neck cancer, lung cancer, or pancreatic cancer.
  • the cancer is squamous non-small-cell lung carcinoma (NSCLC), squamous cell carcinoma of the head and neck (SCCHN), or colorectal cancer.
  • compositions for treatment of cancer vary depending upon many different factors, including means of administration, target site, physiological state of the subject, whether the subject is human or an animal, and other medications administered.
  • the subject is a human, but non-human mammals including transgenic mammals can also be treated.
  • the subject to be treated can be any mammal, in need of treatment, in certain embodiments, the subject is a human subject.
  • a preparation to be administered to a subject is the multimeric binding molecule comprising an ISA as described herein or a monoclonal antibody, administered in conventional dosage form, which can be combined with a pharmaceutical excipient, carrier or diluent as described elsewhere herein.
  • compositions comprising the multimeric binding molecule or monoclonal antibody can be administered by any suitable method, e.g., parenterally, intraventricularly, orally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the cancer is a hematologic cancer or a solid tumor cancer.
  • the hematologic cancer is a leukemia, lymphoma, myeloma, or myelodysplastic syndrome.
  • the leukemia is an acute myeloid leukemia, acute lymphoblastic leukemia, chronic myelogenous leukemia, or chronic lymphocytic leukemia.
  • the lymphoma is Hodgkin’s lymphoma or non-Hodgkin’s lymphoma.
  • the solid tumor cancer is lung cancer, such as non-small-cell lung cancer, urothelial cancer, renal cancer, liver cancer, colorectal cancer, ovarian cancer, gastric cancer, esophageal cancer, pancreatic cancer, thyroid cancer, breast cancer, or head and neck cancer, such as nasopharyngeal cancer.
  • the breast cancer is hormone receptor negative or triple negative breast cancer.
  • the cancer is skin cancer, such as melanoma.
  • the cancer is glioblastoma.
  • compositions comprising a multimeric binding molecule comprising an ISA as described herein may differ depending on the route of administration.
  • routes of administration include intratumoral, oral, parenteral, by inhalation or topical.
  • parenteral as used herein includes, e.g., intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, rectal, or vaginal administration. While these forms of administration are contemplated as suitable forms, another example of a form for administration would be a solution for injection, in particular for intratumoral, intravenous, or intraarterial injection or drip.
  • a suitable pharmaceutical composition can comprise a buffer (e.g., acetate, phosphate, or citrate buffer), a surfactant (e.g., polysorbate), optionally a stabilizer agent (e.g., human albumin), etc.
  • a multimeric binding molecule comprising an ISA as provided herein can be administered in a pharmaceutically effective amount for the treatment of a subject in need thereof.
  • the disclosed multimeric binding molecule comprising an ISA can be formulated so as to facilitate administration and promote stability of the active agent.
  • Pharmaceutical compositions accordingly can comprise a pharmaceutically acceptable, non-toxic, sterile carrier such as physiological saline, non-toxic buffers, preservatives, and the like.
  • a pharmaceutically effective amount of a multimeric binding molecule comprising an ISA as provided herein means an amount sufficient to achieve effective binding to a target and to achieve a therapeutic benefit. Suitable formulations are described in Remington's Pharmaceutical Sciences, e.g., 21 st Edition (Lippincott Williams & Wilkins) (2005).
  • compositions provided herein can be orally administered in an acceptable dosage form including, e.g., capsules, tablets, aqueous suspensions, or solutions. Certain pharmaceutical compositions also can be administered by nasal aerosol or inhalation. Such compositions can be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other conventional solubilizing or dispersing agents.
  • the amount of a multimeric binding molecule comprising an ISA that can be combined with carrier materials to produce a single dosage form will vary depending, e.g., upon the subject treated and the particular mode of administration.
  • the composition can be administered as a single dose, multiple doses or over an established period of time in an infusion. Dosage regimens also can be adjusted to provide the optimum desired response (e.g., a therapeutic response).
  • a multimeric binding molecule comprising an ISA as provided herein can be administered to a subject in need of therapy in an amount sufficient to produce a therapeutic effect.
  • a multimeric binding molecule comprising an ISA as described herein can be administered to the subject in a conventional dosage form prepared by combining the multimeric binding molecule of the disclosure with a conventional pharmaceutically acceptable carrier or diluent according to known techniques.
  • the form and character of the pharmaceutically acceptable carrier or diluent can be dictated by the amount of active ingredient with which it is to be combined, the route of administration and other well-known variables.
  • IgM antibodies IgM-like antibodies, and IgM-derived binding molecules
  • IgM is the first immunoglobulin produced by B cells in response to stimulation by antigen and is naturally present at around 1.5 mg/ml in serum with a half-life of about 5 days.
  • IgM is a pentameric or hexameric molecule and thus includes five or six binding units.
  • An IgM binding unit typically includes two light and two heavy chains. While an IgG heavy chain constant region contains three heavy chain constant domains (CHI, CH2 and CH3), the heavy (p) constant region of IgM additionally contains a fourth constant domain (CH4) and includes a C-terminal p “tailpiece” (ptp).
  • the human IgM constant region typically comprises the amino acid sequence SEQ ID NO: 1 (IMGT allele IGHM*03, identical to, e.g., GenBank Accession No. pir
  • the human Cpl region ranges from about amino acid 5 to about amino acid 102 of SEQ ID NO: 1 or SEQ ID NO: 2; the human Cp2 region ranges from about amino acid 114 to about amino acid 205 of SEQ ID NO: 1 or SEQ ID NO: 2, the human Cp3 region ranges from about amino acid 224 to about amino acid 319 of SEQ ID NO: 1 or SEQ ID NO: 2, the Cp 4 region ranges from about amino acid 329 to about amino acid
  • Each IgM heavy chain constant region is associated with an antigen-binding domain, e.g., a scFv, or a subunit of an antigen-binding domain, e.g., a VH region.
  • an antigen-binding domain e.g., a scFv
  • a subunit of an antigen-binding domain e.g., a VH region.
  • IgM binding units can form a complex with an additional small polypeptide chain (the J-chain), or a functional fragment, variant, or derivative thereof, to form a pentameric IgM antibody or IgM-like antibody.
  • the precursor form of the human J-chain is presented as SEQ ID NO: 6.
  • the signal peptide (underlined) extends from amino acid 1 to about amino acid 22 of SEQ ID NO: 6, and the mature human J-chain extends from about amino acid 23 to amino acid 159 of SEQ ID NO: 6.
  • the mature human J-chain has the amino acid sequence SEQ ID NO: 7.
  • an IgM antibody or IgM-like antibody typically assembles into a hexamer, comprising six binding units and up to twelve binding unit-associated antigen-binding domains.
  • an IgM antibody or IgM-like antibody typically assembles into a pentamer, comprising five binding units and up to ten binding unit-associated antigenbinding domains, or more, if the J-chain is a modified J-chain comprising one or more heterologous polypeptides that can be, e.g., additional J-chain-associated antigen-binding domain(s).
  • the assembly of five or six IgM binding units into a pentameric or hexameric IgM antibody or IgM-like antibody is thought to involve interactions between the Cp4 and p tailpiece domains. See, e.g. , Braathen, R., etaL, J. Biol. Chem. 277:42755-42762 (2002).
  • the constant regions of a pentameric or hexameric IgM antibody or antibodylike molecule utilized in a method provided in this disclosure typically includes at least the Cp4 and/or p tailpiece domains.
  • a “multimerizing fragment” of an IgM heavy chain constant region thus includes at least the Cp4 domain and a ptp domain.
  • An IgM heavy chain constant region can additionally include a Cp3 domain or a fragment thereof, a Cp2 domain or a fragment thereof, and/or a Cpl domain or a fragment thereof.
  • a binding molecule e.g., an IgM antibody or IgM-like antibody as provided herein can include a complete IgM heavy (p) chain constant domain, e.g., SEQ ID NO: 1 or SEQ ID NO: 2, or a multimerizing variant, derivative, or analog thereof, e.g., as provided herein.
  • the disclosure provides a method that utilizes a pentameric IgM or IgM-like antibody comprising five bivalent binding units, where each binding unit includes two IgM heavy chain constant regions or multimerizing fragments or variants thereof, each associated with an antigen-binding domain or a subunit of an antigen-binding domain.
  • the two IgM heavy chain constant regions are human heavy chain constant regions.
  • the IgM or IgM-like antibody utilized in a method provided herein typically further includes a J-chain, or functional fragment or variant thereof.
  • the J-chain is a modified J-chain comprising a heterologous moiety, e.g., a J-chain-associated antigen binding domain.
  • the J-chain-associated antigen binding domain specifically binds to an immune effector cell, e.g., a CD8+ cytotoxic T cell or an NK cell.
  • the modified J-chain includes one or more heterologous moieties attached thereto, e.g., an immune stimulatory agent.
  • the J-chain can be mutated to affect, e.g., enhance, the serum half-life of the IgM or IgM-like antibody described herein, as discussed elsewhere in this disclosure. In certain embodiments the J- chain can be mutated to affect glycosylation, as discussed elsewhere in this disclosure.
  • An IgM heavy chain constant region can include one or more of a Cpl domain or fragment or variant thereof, a Cp2 domain or fragment or variant thereof, a Cp3 domain or fragment or variant thereof, a Cp4 domain or fragment or variant thereof, and/or a p tail piece (ptp) or fragment or variant thereof, provided that the constant region can serve a desired function in the IgM or IgM-like antibody, e.g., associate with second IgM constant region to form a binding unit with one, two, or more antigen-binding domain(s), and/or associate with other binding units and a J-chain to form a pentamer.
  • ptp p tail piece
  • the two IgM heavy chain constant regions or fragments or variants thereof within an individual binding unit each comprise a Cp4 domain or fragment or variant thereof, a p tailpiece (ptp) or fragment or variant thereof, or a combination of a Cp4 domain and a ptp or fragment or variant thereof.
  • the two IgM heavy chain constant regions or fragments or variants thereof within an individual binding unit each further comprise a Cp3 domain or fragment or variant thereof, a Cp2 domain or fragment or variant thereof, a Cpl domain or fragment or variant thereof, or any combination thereof.
  • the binding units of the IgM or IgM-like antibody comprise two light chains. In some embodiments, the binding units of the IgM or IgM-like antibody comprise two fragments of light chains. In some embodiments, the light chains are kappa light chains. In some embodiments, the light chains are lambda light chains. In some embodiments, the light chains are hybrid kappa and lambda light chains. In some embodiments, each binding unit comprises two immunoglobulin light chains each comprising a VL situated amino terminal to an immunoglobulin light chain constant region.
  • IgM antibodies IgM-like antibodies, and IgM-derived binding molecules with enhanced serum half-life
  • IgM-derived multimeric binding molecules e.g., antibodies or antibody-like utilized in the methods provided herein can be modified to have enhanced serum half-life.
  • IgM heavy chain constant region mutations that can enhance serum half-life of an IgM-derived binding molecule are disclosed in U.S. Patent No. 10,899,835, which is incorporated by reference herein in its entirety.
  • a variant IgM heavy chain constant region of an IgM-derived binding molecule as described herein can include an amino acid substitution at an amino acid position corresponding to amino acid S401, E402, E403, R344, and/or E345 of a wild-type human IgM constant region (e.g., SEQ ID NO: 1 or SEQ ID NO: 2).
  • an amino acid corresponding to amino acid S401, E402, E403, R344, and/or E345 of a wild-type human IgM constant region is meant the amino acid in the sequence of the IgM constant region of any species which is homologous to S401, E402, E403, R344, and/or E345 in the human IgM constant region.
  • amino acid corresponding to S401, E402, E403, R344, and/or E345 of SEQ ID NO: 1 or SEQ ID NO: 2 can be substituted with any amino acid, e.g., alanine.
  • IgM antibodies, IgM-like antibodies, and IgM-derived binding molecules with reduced CDC activity can be substituted with any amino acid, e.g., alanine.
  • IgM-derived multimeric binding molecules e.g., antibodies or antibody-like molecules utilized in the methods provided herein can be engineered to exhibit reduced complement-dependent cytotoxicity (CDC) activity to cells in the presence of complement, relative to a reference IgM antibody or IgM-like antibody with a corresponding reference human IgM constant region identical, except for the mutations conferring reduced CDC activity.
  • CDC complement-dependent cytotoxicity
  • These CDC mutations can be combined with any of the mutations to block N-linked glycosylation and/or to confer increased serum half-life as provided herein.
  • corresponding reference human IgM constant region is meant a human IgM constant region or portion thereof, e.g., a Cp3 domain, that is identical to the variant IgM constant region except for the modification or modifications in the constant region affecting CDC activity.
  • the variant human IgM constant region includes one or more amino acid substitutions, e.g., in the Cp3 domain, relative to a wild-type human IgM constant region as described, e.g., in U.S. Patent No. 11,401,337, which is incorporated herein by reference in its entirety.
  • Assays for measuring CDC are well known to those of ordinary skill in the art, and exemplary assays are described e.g., in U.S. Patent No. 11,401,337, which is incorporated by reference herein in its entirety.
  • a variant human IgM constant region conferring reduced CDC activity includes an amino acid substitution corresponding to the wild-type human IgM constant region at position L310, P311, P313, and/or K315 of SEQ ID NO: 1 (human IgM constant region allele IGHM*03) or SEQ ID NO: 2 (human IgM constant region allele IGHM*04).
  • a variant human IgM constant region conferring reduced CDC activity includes an amino acid substitution corresponding to the wild-type human IgM constant region at position P311 of SEQ ID NO: 1 or SEQ ID NO: 2.
  • the variant IgM constant region as provided herein contains an amino acid substitution corresponding to the wild-type human IgM constant region at position P313 of SEQ ID NO: 1 or SEQ ID NO: 2.
  • the variant IgM constant region as provided herein contains a combination of substitutions corresponding to the wild-type human IgM constant region at positions P311 of SEQ ID NO: 1 or SEQ ID NO: 2 and/or P313 of SEQ ID NO: 1 or SEQ ID NO: 2.
  • These proline residues can be independently substituted with any amino acid, e.g., with alanine, serine, or glycine.
  • a variant human IgM constant region conferring reduced CDC activity includes an amino acid substitution corresponding to the wild-type human IgM constant region at position K315 of SEQ ID NO: 1 or SEQ ID NO: 2.
  • the lysine residue can be independently substituted with any amino acid, e.g., with alanine, serine, glycine, or aspartic acid.
  • a variant human IgM constant region conferring reduced CDC activity includes an amino acid substitution corresponding to the wild-type human IgM constant region at position K315 of SEQ ID NO: 1 or SEQ ID NO: 2 with aspartic acid.
  • a variant human IgM constant region conferring reduced CDC activity includes an amino acid substitution corresponding to the wild-type human IgM constant region at position L310 of SEQ ID NO: 1 or SEQ ID NO: 2.
  • the lysine residue can be independently substituted with any amino acid, e.g., with alanine, serine, glycine, or aspartic acid.
  • a variant human IgM constant region conferring reduced CDC activity includes an amino acid substitution corresponding to the wild-type human IgM constant region at position L310 of SEQ ID NO: 1 or SEQ ID NO: 2 with aspartic acid.
  • IgM antibodies Glyco-modified IgM antibodies, IgM-like antibodies, and IgM-derived binding molecules
  • N-linked glycosylation motif comprises or consists of the amino acid sequence N- Xi-S/T, where N is asparagine, Xi is any amino acid except proline (P), and S/T is serine (S) or threonine (T).
  • S serine
  • T threonine
  • the glycan is attached to the nitrogen atom of the asparagine residue. See, e.g., Drickamer K, Taylor ME (2006), Introduction to Glycobiology (2nd ed.). Oxford University Press, USA.
  • N-linked glycosylation motifs occur in the human IgM heavy chain constant regions of SEQ ID NO: 1 or SEQ ID NO: 2 starting at positions 46 (“Nl”), 209 (“N2”), 272 (“N3”), 279 (“N4”), and 440 (“N5”). These five motifs are conserved in non- human primate IgM heavy chain constant regions, and four of the five are conserved in the mouse IgM heavy chain constant region. Accordingly, in some embodiments, IgM heavy chain constant regions of a multimeric binding molecule as provided herein comprise 5 N- linked glycosylation motifs: Nl, N2, N3, N4, and N5. In some embodiments, at least three of the N-linked glycosylation motifs e.g, Nl, N2, and N3) on each IgM heavy chain constant region are occupied by a complex glycan.
  • At least one, at least two, at least three, or at least four of the N- Xi-S/T motifs can include an amino acid insertion, deletion, or substitution that prevents glycosylation at that motif.
  • the IgM-derived multimeric binding molecule can include an amino acid insertion, deletion, or substitution at motif Nl, motif N2, motif N3, motif N5, or any combination of two or more, three or more, or all four of motifs Nl, N2, N3, or N5, where the amino acid insertion, deletion, or substitution prevents glycosylation at that motif.
  • the IgM constant region comprises one or more substitutions relative to a wild-type human IgM constant region at positions 46, 209, 272, or 440 of SEQ ID NO: 1 (human IgM constant region allele IGHM*03) or SEQ ID NO: 2 (human IgM constant region allele IGHM*04). See, e.g., US Patent Application Publication No. 2022-0306760, which is incorporated herein by reference in its entirety.
  • the J-chain of a pentameric an IgM or IgM-like antibody as described herein and utilized in the methods provided herein can be modified, e.g., by introduction of a heterologous moiety, or two or more heterologous moi eties, e.g., polypeptides, without interfering with the ability of the IgM or IgM-like antibody to assemble and bind to its binding target(s).
  • a heterologous moiety or two or more heterologous moi eties, e.g., polypeptides
  • IgM or IgM-like antibodies as provided herein can include a modified J-chain or functional fragment or variant thereof that further includes a heterologous moiety, e.g., a heterologous polypeptide, introduced into the J-chain or fragment or variant thereof.
  • heterologous moiety can be a peptide or polypeptide fused in frame or chemically conjugated to the J-chain or fragment or variant thereof.
  • the heterologous polypeptide can be fused to the J-chain or functional fragment or variant thereof.
  • the heterologous polypeptide is fused to the J-chain or functional fragment or variant thereof via a linker, e.g., a peptide linker consisting of least 5 amino acids, but typically no more than 25 amino acids.
  • the peptide linker consists of GGGGS (SEQ ID NO: 9), GGGGSGGGGS (SEQ ID NO: 10), GGGGSGGGGSGGGGS (SEQ ID NO: 11), GGGGS GGGGS GGGGS GGGGS (SEQ ID NO: 12), or GGGGS GGGGS GGGGS GGGGS GGGGS GGGGS (SEQ ID NO: 13), or GGSGGGGSGGGSGGGGSLQ (SEQ ID NO: 14).
  • heterologous moiety can be a chemical moiety conjugated to the J-chain.
  • Heterologous moi eties to be attached to a J-chain can include, without limitation, a binding moiety, e.g., an antibody or antigen -binding fragment thereof, e.g., a single chain Fv (scFv) molecule, a stabilizing peptide that can increase the half-life of the IgM or IgM-like antibody, or a chemical moiety such as a polymer or a cytotoxin.
  • heterologous moiety comprises a stabilizing peptide that can increase the half-life of the binding molecule, e.g., human serum albumin (HSA) or an HSA binding molecule.
  • HSA human serum albumin
  • a modified J-chain can further include a J-chain associated antigen-binding domain, e.g., a polypeptide capable of specifically binding to a target antigen.
  • a J-chain-associated antigen-binding domain can be an antibody or an antigen-binding fragment thereof, as described elsewhere herein.
  • the J-chain-associated antigen-binding domain can be a single chain Fv (scFv) antigen-binding domain or a single-chain antigen -binding domain derived, e.g., from a camelid or condricthoid antibody.
  • the J-chain-associated antigen-binding domain can be introduced into the J-chain at any location that allows the binding of the J-chain- associated antigen-binding domain to its binding target without interfering with J-chain function or the function of an associated multimeric binding molecule. Insertion locations include but are not limited to at or near the C-terminus, at or near the N-terminus or at an internal location that, based on the three-dimensional structure of the J-chain, is accessible. In certain embodiments, the J-chain-associated antigen-binding domain can be introduced into the mature human J-chain of SEQ ID NO: 7 between cysteine residues 92 and 101 of SEQ ID NO: 7.
  • the J-chain-associated antigen-binding domain can be introduced into the human J-chain of SEQ ID NO: 7 at or near a glycosylation site. In a further embodiment, the J-chain-associated antigen-binding domain can be introduced into the human J-chain of SEQ ID NO: 7 within about 10 amino acid residues from the C- terminus, or within about 10 amino acids from the N-terminus.
  • a modified J-chain can include a cytokine, e.g., interleukin-2 (IL-2) or interleukin- 15 (IL- 15), or a receptor-binding fragment or variant thereof, which in certain embodiments can be associated, either via binding or covalent attachment, to part of its receptor, e.g, the sushi domain of IL- 15 receptor-a.
  • IL-2 interleukin-2
  • IL- 15 interleukin- 15
  • a receptor-binding fragment or variant thereof which in certain embodiments can be associated, either via binding or covalent attachment, to part of its receptor, e.g, the sushi domain of IL- 15 receptor-a.
  • the J-chain of an IgM antibody, IgM-like antibody, or IgM- derived binding molecule as provided herein is a variant J-chain that comprises one or more amino acid substitutions that can alter, e.g, the serum half-life of an IgM antibody, IgM-like antibody, or IgM-derived binding molecule provided herein.
  • certain amino acid substitutions, deletions, or insertions can result in the IgM-derived binding molecule exhibiting an increased serum half-life upon administration to a subject animal relative to a reference IgM-derived binding molecule that is identical except for the one or more single amino acid substitutions, deletions, or insertions in the variant J-chain, and is administered using the same method to the same animal species.
  • the variant J-chain can include one, two, three, or four single amino acid substitutions, deletions, or insertions relative to the reference J-chain.
  • the multimeric binding molecule can comprise a variant J- chain sequence, such as a variant sequence described herein with reduced glycosylation or reduced binding to one or more polymeric Ig receptors (e.g., plgR, Fc alpha-mu receptor (FcapR), or Fc mu receptor (FcpR)).
  • a variant J-chain sequence such as a variant sequence described herein with reduced glycosylation or reduced binding to one or more polymeric Ig receptors (e.g., plgR, Fc alpha-mu receptor (FcapR), or Fc mu receptor (FcpR)).
  • the variant J-chain can comprise an amino acid substitution at the amino acid position corresponding to amino acid Y102 of the mature wild-type human J-chain (SEQ ID NO: 7).
  • an amino acid corresponding to amino acid Y102 of the mature wild-type human J-chain is meant the amino acid in the sequence of the J-chain of any species which is homologous to Y102 in the human J-chain. See U.S. Patent No. 10,899,835, which is incorporated herein by reference in its entirety. The position corresponding to Y102 in SEQ ID NO: 7 is conserved in the J-chain amino acid sequences of at least 43 other species. See FIG. 4 of U.S. Patent No. 9,951,134, which is incorporated by reference herein.
  • Certain mutations at the position corresponding to Y102 of SEQ ID NO: 7 can inhibit the binding of certain immunoglobulin receptors, e.g., the human or murine Fcap receptor, the murine Fcp receptor, and/or the human or murine polymeric Ig receptor (pig receptor) to an IgM pentamer comprising the mutant J-chain.
  • immunoglobulin receptors e.g., the human or murine Fcap receptor, the murine Fcp receptor, and/or the human or murine polymeric Ig receptor (pig receptor) to an IgM pentamer comprising the mutant J-chain.
  • IgM antibodies, IgM-like antibodies, and IgM- derived binding molecules comprising a mutation at the amino acid corresponding to Y102 of SEQ ID NO: 7 have an improved serum half-life when administered to an animal than a corresponding antibody, antibody-like molecule or binding molecule that is identical except for the substitution, and which is administered to the same species in the same manner.
  • the amino acid corresponding to Y102 of SEQ ID NO: 7 can be substituted with any amino acid.
  • the amino acid corresponding to Y102 of SEQ ID NO: 7 can be substituted with alanine (A), serine (S) or arginine (R).
  • the amino acid corresponding to Y102 of SEQ ID NO: 7 can be substituted with alanine.
  • the J-chain or functional fragment or variant thereof is a variant human J-chain and comprises the amino acid sequence SEQ ID NO: 8, a J chain referred to herein as “J*.” [0131] Wild-type J-chains typically include one N-linked glycosylation site.
  • a variant J-chain or functional fragment thereof of a multimeric binding molecule as provided herein includes a mutation within the asparagine (N)-linked glycosylation motif N-Xi-S/T, e.g., starting at the amino acid position corresponding to amino acid 49 (motif N6) of the mature human J-chain (SEQ ID NO: 7) or J* (SEQ ID NO: 8), where N is asparagine, Xi is any amino acid except proline, and S/T is serine or threonine, and where the mutation prevents glycosylation at that motif.
  • N asparagine
  • Xi is any amino acid except proline
  • S/T is serine or threonine
  • mutations preventing glycosylation at this site can result in the multimeric binding molecule as described herein, exhibiting an increased serum halflife upon administration to a subject animal relative to a reference multimeric binding molecule that is identical except for the mutation or mutations preventing glycosylation in the variant J-chain, and is administered in the same way to the same animal species.
  • the variant J-chain or functional fragment thereof of a binding molecule comprising a J-chain as described herein can include an amino acid substitution at the amino acid position corresponding to amino acid N49 or amino acid S51 of SEQ ID NO: 7 or SEQ ID NO: 8, provided that the amino acid corresponding to S51 is not substituted with threonine (T), or where the variant J-chain comprises amino acid substitutions at the amino acid positions corresponding to both amino acids N49 and S51 of SEQ ID NO: 7 or SEQ ID NO: 8.
  • T threonine
  • the position corresponding to N49 of SEQ ID NO: 7 or SEQ ID NO: 8 is substituted with any amino acid, e.g., alanine (A), glycine (G), threonine (T), serine (S) or aspartic acid (D).
  • alanine A
  • G glycine
  • T threonine
  • S serine
  • D aspartic acid
  • the position corresponding to N49 of SEQ ID NO: 7 or SEQ ID NO: 8 can be substituted with alanine (A).
  • the position corresponding to N49 of SEQ ID NO: 7 or SEQ ID NO: 8 can be substituted with aspartic acid (D).
  • the position corresponding to S51 of SEQ ID NO: 7 or SEQ ID NO: 8 is substituted with alanine (A) or glycine (G). In some embodiments, the position corresponding to S51 of SEQ ID NO: 7 or SEQ ID NO: 8 is substituted with alanine (A).
  • Multimeric binding molecules with a modified J-chain expressing an immune stimulatory agent Multimeric binding molecules with a modified J-chain expressing an immune stimulatory agent
  • This disclosure provides methods utilizing multimeric binding molecules with immune stimulatory properties.
  • the disclosure provides a method utilizing a multimeric binding molecule that includes five IgM or IgM-like bivalent binding units or multimerizing variants or fragments thereof and a modified J-chain.
  • Each binding unit includes two IgM heavy chain constant regions or multimerizing variants or fragments thereof, each associated with an antigen-binding domain for a total of ten antigen-binding domains, which can be the same or different, but in certain embodiments at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine or ten of the antigen-binding domains of the binding molecule specifically bind to PD-L1.
  • the antigen binding domains can be identical, or can be different, e.g., binding to different epitopes of the same target antigen.
  • the modified J-chain of the multimeric binding molecule of the provided method includes (a) a J-chain or functional fragment or variant thereof (“J”), and (b) an immunostimulatory agent (“ISA”), wherein J and the ISA are associated as a fusion protein.
  • J J-chain or functional fragment or variant thereof
  • ISA immunostimulatory agent
  • Exemplary ISAs are disclosed in PCT Publication No: WO 2021/030688, which is incorporated herein by reference in its entirety.
  • the term “ISA” can refer to the heterologous moiety fused to the J-chain that possesses immune stimulatory activity, or can refer to entire multimeric binding molecule, which possesses immune stimulatory activity.
  • the ISA comprises a cytokine, or a receptor-binding fragment or variant thereof.
  • the ISA can include interleukin- 15 (IL-15), interleukin-2 (IL-2), interferon (IFN)-a, interleukin 12 (IL- 12), interleukin-21 (IL-21), granulocyte macrophage colony-stimulating factor (GM-CSF), or any receptor-binding fragment or variant thereof.
  • the ISA can, in addition, include portions of a receptor subunit, or other immune stimulating moieties.
  • IL-15 complexed with the sushi domain of IL-15Ra, forms a highly potent ISA that can stimulate CD8+ T cells and NK cells.
  • the disclosure provides a modified J-chain comprising a J-chain or functional fragment or variant thereof (“J”), and (a) an interleukin- 15 (IL- 15) protein or receptor-binding fragment or variant thereof (“I”), and/or (b) an interleukin- 15 receptor-a (IL-15Ra) fragment comprising the sushi domain or a variant thereof capable of associating with I (“R”), wherein J and at least one of I and R are associated as a fusion protein, and wherein I and R can associate to function as the ISA.
  • J can be a wild-type J-chain of any species, e.g., a human J-chain comprising the amino acid sequence SEQ ID NO: 7 or a functional fragment or variant thereof.
  • J can be a variant J-chain or fragment thereof comprising one or more single amino acid substitutions, deletions, or insertions relative to a wild-type J-chain that can affect, e.g., the serum half-life of the multimeric binding molecule comprising the J-chain, as described in U.S. Patent No. 10,899,835.
  • J is a variant human J-chain and comprises the amino acid sequence SEQ ID NO: 8, also referred to herein as (“J*”).
  • the interleukin- 15 (IL- 15) protein or receptor-binding fragment or variant thereof (“I”) of the immune stimulatory agent is a wild-type human IL-15 protein comprising the amino acid sequence SEQ ID NO: 16.
  • a non-limiting example of a modified J-chain IS As comprising the wild-type human IL- 15 is SEQ ID NO: 37.
  • the potency of the ISA activity is modulated, e.g., altered or reduced.
  • “I” comprises a receptor binding variant of human IL- 15, in which receptor binding is reduced but not eliminated.
  • the receptor binding variant of human IL- 15 comprises at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, or at least nine single amino acid insertions, deletions, or substitutions, where the single amino acid insertions, deletions, or substitutions reduce the affinity of the IL- 15 variant for its receptor.
  • Variant versions of human IL-15 that achieve this goal are described in U.S. Patent No. 10,550,185.
  • the variant human IL-15 comprises at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, or at least nine single amino acid insertions, deletions, or substitutions, but no more than ten, single amino acid insertions, deletions, or substitutions.
  • I comprises a variant human IL- 15 comprising one, two, three, four, five, six, seven, eight or nine single amino acid substitutions.
  • the amino acid substitutions are at one or more of positions corresponding to Nl, N4, D8, D30, D61, E64, N65, N72, or Q108 of SEQ ID NO: 16.
  • the amino acid substitutions comprise one or more of substitutions N1D, N4D, D8N, D30N, D61N, E64Q, N65D, N72D, or Q108E, in SEQ ID NO: 16.
  • I comprises SEQ ID NO: 16 except for: a single amino acid substitution at a position selected from the group consisting of N1D, N4D, D8N, D30N, D61N, E64Q, N65D, N72D, and Q108E; two amino acid substitutions at positions selected from the group consisting of N4D/N65D and N1D/N65D; or three amino acid substitutions at positions D30N/E64Q/N65D.
  • I comprises the amino acid sequence SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, or SEQ ID NO: 29.
  • the J*RI ISA comprises the amino acid sequence SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50.
  • I comprises a variant human IL-15 comprising one, two, three, four, or five single amino acid substitutions.
  • the amino acid substitutions are at one or more of positions corresponding to N49, N71, S73, N79, or Ml 12 of SEQ ID NO: 16.
  • the amino acid substitutions comprise one or more of substitutions N49D, N71D, S73I, N79D, or M112D, in SEQ ID NO: 16.
  • I comprises SEQ ID NO: 16 except for: a single amino acid substitution at a position selected from the group consisting of N49D, N71D, S73I, N79D, or M112D; or four amino acid substitutions at positions N49D/N71D/ N79D/M112D.
  • I comprises the amino acid sequence SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, or SEQ ID NO: 35.
  • the J*RI ISA comprises the amino acid sequence SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, or SEQ ID NO: 56.
  • R comprises the sushi domain of the human IL-15 receptor-a.
  • R comprises the amino acid sequence SEQ ID NO: 36 or a variant or fragment thereof that is capable of associating with human IL-15.
  • R consists essentially of or consists of the amino acid sequence SEQ ID NO: 36 or a variant thereof that is capable of associating with human IL-15.
  • heterologous moieties are fused to the J-chain or variant or fragment thereof via a linker, a small, flexible chain of amino acids, typically comprising small amino acids, e.g., glycine (G) and/or serine (S).
  • the linker can comprise, consist of, or consist essentially of SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, or SEQ ID NO: 13.
  • linkers are typically employed between each element for a total of at least two linkers.
  • the at least two linkers can be the same or different.
  • at least one linker comprises, consists essentially of, or consists of the amino acid sequence
  • At least one linker comprises, consists essentially of, or consists of the amino acid sequence
  • the modified J-chain of the multimeric binding molecule comprises the amino acid sequence SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO:
  • SEQ ID NO: 42 SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, or SEQ ID NO: 56.
  • a multimeric binding molecule as utilized in a method provided herein includes at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, or ten heavy chain constant regions associated with a IgM heavy chain, each comprising a heavy chain variable region (VH-IgM) and at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, or ten light chains, each comprising a light chain variable region (VL-IgM), and where the associated VH-IgM and VL-IgM specifically bind to PD-L1 or a subunit thereof.
  • VH-IgM heavy chain variable region
  • VL-IgM light chain variable region
  • VH-IgM and the VL-IgM comprises the heavy chain variable region (VH) and the light chain variable region (VL) of the humanized anti-PD-Ll antibody h3C5, disclosed in U.S. Patent No. 10,954,302, which is incorporated herein by reference.
  • the VH-IgM and VL-IgM comprise six immunoglobulin complementarity determining regions (CDRs): IgM binding molecule heavy chain CDR 1 (HCDRl-IgM), IgM binding molecule heavy chain CDR 2 (HCDR2-IgM), IgM binding molecule heavy chain CDR 3 (HCDR3-IgM), IgM binding molecule light chain CDR 1 (LCDRl-IgM), IgM binding molecule light chain CDR 2 (LCDR2-IgM), and IgM binding molecule light chain CDR 3 (LCDR3-IgM), wherein the HCDRl-IgM, HCDR2-IgM, HCDR3-IgM, LCDRl-IgM, LCDR2-IgM, and LCDR3-IgM comprise, respectively, the amino acid sequence of SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61
  • the VH-IgM comprises the amino acid sequence SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 68, or SEQ ID NO: 69
  • the VL-IgM comprises the amino acid sequence SEQ ID NO: 70 or SEQ ID NO: 71.
  • the VH-IgM and VL-IgM comprise, respectively, the amino acid sequences of SEQ ID NO: 64 and SEQ ID NO: 71.
  • the heavy chain comprises SEQ ID NO: 72.
  • the light chain comprises SEQ ID NO: 73.
  • the heavy chain comprises SEQ ID NO: 72
  • the light chain comprises SEQ ID NO: 73
  • the modified J-chain comprises SEQ ID NO: 51.
  • the PD-L1 antibody can comprise the CDRs within or the VH and VL sequences of SEQ ID NO: 74 and SEQ ID NO: 75, SEQ ID NO: 76 and SEQ ID NO: 77, SEQ ID NO: 78 and SEQ ID NO: 79, SEQ ID NO: 80 and SEQ ID NO: 81, SEQ ID NO: 82 and SEQ ID NO: 83, SEQ ID NO: 84 and SEQ ID NO: 85, SEQ ID NO: 86 and SEQ ID NO: 87, SEQ ID NO: 88 and SEQ ID NO: 89, SEQ ID NO: 90 and SEQ ID NO: 91, SEQ ID NO: 92 and SEQ ID NO: 93, SEQ ID NO: 94 and SEQ ID NO: 95, SEQ ID NO: 96 and SEQ ID
  • the at least four, at least five, at least six, at least seven, at least eight, at least nine or ten antigen-binding domains are identical.
  • This disclosure employs, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. See, for example, Green and Sambrook, ed. (2012) Molecular Cloning A Laboratory Manual (4 th ed.; Cold Spring Harbor Laboratory Press); Sambrook et al., ed. (1992) Molecular Cloning: A Laboratory Manual, (Cold Springs Harbor Laboratory, NY); D. N.
  • Embodiment 1 A method for treating cancer in a subject in need of thereof comprising administering to the subject a combination therapy comprising:
  • a multimeric binding molecule comprising five bivalent binding units and a modified J-chain
  • the modified J-chain comprises a J-chain or a functional fragment or variant thereof (“J”), an interleukin- 15 (IL- 15) protein or receptor-binding fragment or variant thereof (“I”), and an interleukin- 15 receptor-a (IL- 15Ra) fragment comprising the sushi domain or a variant thereof capable of associating with I (“R”), wherein J and at least one of I and R are associated as a fusion protein, wherein I and R can associate to function as an immunostimulatory agent, wherein each binding unit comprises two IgM heavy chains, each comprising a heavy chain variable region (VH-IgM) and an IgM constant region and two light chains, each comprising a light chain variable region (VL-IgM) and a light chain constant region, and wherein the associated VH-IgM and VL-IgM specifically bind to PD-L1
  • a monoclonal antibody specifically binds a tumor antigen, wherein the tumor antigen is cluster of differentiation 38 (CD38), human epidermal growth factor receptor 2 (HER2), or epidermal growth factor receptor (EGFR).
  • CD38 cluster of differentiation 38
  • HER2 human epidermal growth factor receptor 2
  • EGFR epidermal growth factor receptor
  • Embodiment 2 The method of Embodiment 1, wherein the monoclonal antibody comprises a heavy chain variable region (VH-MA) and a light chain variable region (VL- MA), wherein the VH-MA and VL-MA comprise six immunoglobulin complementarity determining regions (CDRs): monoclonal antibody heavy chain CDR 1 (HCDR1-MA), monoclonal antibody heavy chain CDR 2 (HCDR2-MA), monoclonal antibody heavy chain CDR 3 (HCDR3-MA), monoclonal antibody light chain CDR 1 (LCDR1-MA), monoclonal antibody light chain CDR 2 (LCDR2-MA), and monoclonal antibody light chain CDR 3 (LCDR3-MA), wherein the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208,
  • Embodiment s The method of Embodiment 2, wherein the tumor antigen is CD38, and wherein the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2- MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, SEQ ID NO: 209, SEQ ID NO: 210, and SEQ ID NO: 211; or SEQ ID NO: 214, SEQ ID NO: 215, SEQ ID NO: 216, SEQ ID NO: 217, SEQ ID NO: 218, and SEQ ID NO: 219.
  • Embodiment 4 The method of Embodiment 3, wherein the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, SEQ ID NO: 209, SEQ ID NO: 210, and SEQ ID NO: 211.
  • Embodiment 5 The method of Embodiment 3, wherein the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 212 and SEQ ID NO: 213; or SEQ ID NO: 220 and SEQ ID NO: 221.
  • Embodiment 6 The method of Embodiment 4, wherein the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 212 and SEQ ID NO: 213.
  • Embodiment 7 The method of Embodiment 5, wherein the monoclonal antibody comprises daratumumab or isatuximab.
  • Embodiment 8 The method of Embodiment 6, wherein the monoclonal antibody comprises daratumumab.
  • Embodiment 9 The method of any one of Embodiments 3 to 8, wherein the cancer is multiple myeloma.
  • Embodiment 10 The method of Embodiment 2, wherein the tumor antigen is HER2, and wherein the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2- MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 222, SEQ ID NO: 223, SEQ ID NO: 224, SEQ ID NO: 225, SEQ ID NO: 226, and SEQ ID NO: 227; or SEQ ID NO: 230, SEQ ID NO: 231, SEQ ID NO: 232, SEQ ID NO: 233, SEQ ID NO: 234, and SEQ ID NO: 235.
  • Embodiment 11 The method of Embodiment 10, wherein the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 222, SEQ ID NO: 223, SEQ ID NO: 224, SEQ ID NO: 225, SEQ ID NO: 226, and SEQ ID NO: 227.
  • Embodiment 12 The method of Embodiment 10, wherein the VH-MA and VL- MA comprise, respectively, the amino acid sequence of SEQ ID NO: 228 and SEQ ID NO: 229; or SEQ ID NO: 236 and SEQ ID NO: 237.
  • Embodiment 13 The method of Embodiment 11, wherein the VH-MA and VL- MA comprise, respectively, the amino acid sequence of SEQ ID NO: 228 and SEQ ID NO: 229.
  • Embodiment 14 The method of Embodiment 12, wherein the monoclonal antibody comprises trastuzumab or pertuzumab.
  • Embodiment 15 The method of Embodiment 13, wherein the monoclonal antibody comprises trastuzumab.
  • Embodiment 16 The method of any one of Embodiments 10 to 15, wherein the cancer is breast cancer.
  • Embodiment 17 The method of Embodiment 2, wherein the tumor antigen is EGFR, and wherein the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2- MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 238, SEQ ID NO: 239, SEQ ID NO: 240, SEQ ID NO: 241, SEQ ID NO: 242, and SEQ ID NO: 243; SEQ ID NO: 246, SEQ ID NO: 247, SEQ ID NO: 248, SEQ ID NO: 249, SEQ ID NO: 250, and SEQ ID NO: 251; or SEQ ID NO: 254, SEQ ID NO: 255, SEQ ID NO: 256, SEQ ID NO: 257, SEQ ID NO: 258, and SEQ ID NO: 259.
  • Embodiment 18 The method of Embodiment 17, wherein the HCDR1-MA, HCDR2-MA, HCDR3-MA, LCDR1-MA, LCDR2-MA, and LCDR3-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 238, SEQ ID NO: 239, SEQ ID NO: 240, SEQ ID NO: 241, SEQ ID NO: 242, and SEQ ID NO: 243.
  • Embodiment 19 The method of Embodiment 17, wherein the the VH-MA and VL-MA comprise, respectively, the amino acid sequence of SEQ ID NO: 244 and SEQ ID NO: 245; SEQ ID NO: 252 and SEQ ID NO: 253; or SEQ ID NO: 260 and SEQ ID NO: 261.
  • Embodiment 20 The method of Embodiment 18, wherein the VH-MA and VL- MA comprise, respectively, the amino acid sequence of SEQ ID NO: 244 and SEQ ID NO: 245.
  • Embodiment 21 The method of Embodiment 19, wherein the monoclonal antibody comprises cetuximab, necitumumab, or panitumumab.
  • Embodiment 22 The method of Embodiment 20, wherein the monoclonal antibody comprises cetuximab.
  • Embodiment 23 The method of any one of Embodiments 17 to 22, wherein the cancer is skin cancer, head and neck cancer, or colorectal cancer.
  • Embodiment 24 The method of any one of Embodiments 1 to 23, wherein J comprises the amino acid sequence SEQ ID NO: 7 or a functional fragment or variant thereof.
  • Embodiment 25 The method of any one of Embodiments 1 to 24, wherein J is a variant J-chain or fragment thereof comprising one or more single amino acid substitutions, deletions, or insertions relative to SEQ ID NO: 7 that can affect serum halflife of the multimeric binding molecule; and wherein the multimeric binding molecule exhibits an increased serum half-life upon administration to an animal relative to a reference multimeric binding molecule that is identical except for the one or more single amino acid substitutions, deletions, or insertions, and is administered in the same way to the same animal species.
  • Embodiment 26 The method of Embodiment 25, wherein the J comprises an amino acid substitution at the amino acid position corresponding to amino acid Y102 of the mature wild-type human J-chain (SEQ ID NO: 7).
  • Embodiment 27 The method of Embodiment 26, wherein J is a variant human J- chain and comprises the amino acid sequence SEQ ID NO: 8 (“J*”).
  • Embodiment 28 The method of any one of Embodiments 1 to 27, wherein I comprises the mature human IL-15 amino acid sequence of SEQ ID NO: 16 or a receptorbinding variant or fragment thereof.
  • Embodiment 29 The method of Embodiment 28, wherein I comprises an amino acid substitution at a position corresponding to S73 of SEQ ID NO: 16.
  • Embodiment 30 The method of Embodiment 29, wherein the amino acid substitution comprises S73I in SEQ ID NO: 16.
  • Embodiment 31 The method of Embodiment 30, wherein I comprises the amino acid sequence SEQ ID NO: 30.
  • Embodiment 32 The method of any one of Embodiments 1 to 31, wherein R comprises the amino acid sequence SEQ ID NO: 36 or a variant or fragment thereof that is capable of associating with human IL-15.
  • Embodiment 33 The method of any one of Embodiments 1 to 31, wherein R consists essentially of or consists of the amino acid sequence SEQ ID NO: 36 or a variant thereof that is capable of associating with human IL-15.
  • Embodiment 34 The method of any one of Embodiments 1 to 33, wherein J, I, and R are associated as a fusion protein.
  • Embodiment 35 The method of Embodiment 34, wherein the modified J-chain comprises the amino acid sequence SEQ ID NO: 51.
  • Embodiment 36 The method of any one of Embodiments 1 to 35, wherein the VH-IgM and VL-IgM comprise six immunoglobulin complementarity determining regions (CDRs): IgM binding molecule heavy chain CDR 1 (HCDRl-IgM), IgM binding molecule heavy chain CDR 2 (HCDR2-IgM), IgM binding molecule heavy chain CDR 3 (HCDR3-IgM), IgM binding molecule light chain CDR 1 (LCDRl-IgM), IgM binding molecule light chain CDR 2 (LCDR2-IgM), and IgM binding molecule light chain CDR 3 (LCDR3-IgM), wherein the HCDRl-IgM, HCDR2-IgM, HCDR3-IgM, LCDRl-IgM, LCDR2-IgM, and LCDR3-IgM comprise, respectively, the amino acid sequence of SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO:
  • Embodiment 37 The method of Embodiment 36, wherein the VH-IgM and VL- IgM comprise, respectively, the amino acid sequences of SEQ ID NO: 64 and SEQ ID NO: 71.
  • Embodiment 38 The method of any one of Embodiments 1 to 37, wherein each IgM heavy chain constant region is a human IgM constant region comprising the amino acid sequence SEQ ID NO: 1, SEQ ID NO: 2, or a multimerizing variant or fragment thereof.
  • Embodiment 39 The method of any one of Embodiments 1 to 38, the multimeric binding molecule comprises a variant human IgM constant region, wherein the multimeric binding molecule has reduced CDC activity relative to a multimeric binding molecule comprising IgM heavy chain constant regions comprising the amino acid sequence SEQ ID NO: 1, SEQ ID NO: 2, or a multimerizing variant or fragment thereof.
  • Embodiment 40 The method of Embodiment 39, wherein each IgM heavy chain constant region comprises a variant of the amino acid sequence SEQ ID NO: 1 or SEQ ID NO: 2, wherein the variant comprises an amino acid substitution at position K315 of SEQ ID NO: 1 or SEQ ID NO: 2.
  • Embodiment 41 The method of Embodiment 40, wherein the amino acid substitution is K315D of SEQ ID NO: 1 or SEQ ID NO: 2.
  • Embodiment 42 The method of any one of Embodiments 1 to 41, wherein the heavy chain comprises SEQ ID NO: 72, the light chain comprises SEQ ID NO: 73, and the modified J-chain comprises SEQ ID NO: 51.
  • Example 1 Construction IgM-based immunostimulatory agents (ISA) with a modified J-chain expressing IL- 15 and the IL-15Ra sushi domain
  • a modified J-chain expressing mature human IL-15 comprising an S73I mutation (SEQ ID NO: 30) and the sushi domain of human IL-15Ra (SEQ ID NO: 36) as a fusion protein fused to a variant of the mature human J-chain comprising a Y to A amino acid substitution at position 102 (“Y102A” or “J*,” amino acid sequence of the variant presented as SEQ ID NO: 8) was constructed (SEQ ID NO: 51).
  • a DNA construct encoding the modified J-chain was expressed along with DNA constructs encoding anti-PD-Ll IgM heavy chains and light chains comprising , respectively, the VH and VL amino acid sequences (SEQ ID NO: 64 and SEQ ID NO: 71, respectively) of humanized anti-human PD-L1 antibody h3C5, as disclosed in U.S. Patent No. 10,954,302, which is incorporated herein by reference in its entirety.
  • the full heavy and light chain comprised the amino acid sequences of SEQ ID NOs: 72 and 73, respectively.
  • the resulting binding molecule “PDL1-IL-15 IgMl” properly assembled as a pentamer.
  • Luc-labeled tumor cells (RPML8226, SKOV3, or A431) were added to wells in a 96-well plate and were used as targets in an antibody dependent cellular cytotoxicity (ADCC) and T cell dependent cytotoxicity (TDCC) in vitro killing assay.
  • ADCC antibody dependent cellular cytotoxicity
  • TDCC T cell dependent cytotoxicity
  • Human peripheral blood mononuclear cells (PBMCs) were thawed. The percent of NK cells, and thus percent effector cells for these assays, was determined using flow cytometry to detect CD56+. Using these calculations, an appropriate number of PBMCs were added to wells so that the effector cells to target ratio was 3 : 1.
  • a dilution series ofPDLl-IL-15 IgMl was added to the wells.
  • the plates were incubated for 4 days at 37 °C in a humidified 5% CO2 incubator.
  • a dilution series (0.1-1000 ng/mL) of the monoclonal antibodies daratumumab, trastuzumab, and cetuximab were added to the appropriate plates before a further 24 hours of incubation.
  • Cell viability was determined by addition of 50 pL/well One-Gio EX (Promega, catalog # E633 A). After mixing on a plate shaker (1 min, 700 rpm) luminescence was read on a Perkin Elmer 2104 multilabel plate reader.
  • the percentage of viable was calculated by dividing RLU with antibody by that of no antibody control and multiplying by 100.
  • the data were analyzed by non-linear regression with a 4-parameter logistic curve fit in Prism, version 7.03 (GraphPad). The resulting plots are shown in FIG. 1A, FIG. 2A, and FIG. 3A for daratumumab, trastuzumab, and cetuximab, respectively.
  • BI assumes the effect of each drug to act independently from one another.
  • the choice to use BI is based on the separate and distinct mechanisms of action of each monoclonal antibody when compared to PDL1-IL-15 IgMl.
  • Synergy scores from BI are generated on a continuum of dose combinations, with negative scores reflecting antagonism and positive scores representing synergy. These scores are visualized in 3D surface plots with valleys of antagonism and hills of synergy over the 2D dimension representing the continuum of dose combinations.
  • 3D surface plots for daratumumab, trastuzumab, and cetuximab are shown in FIGS. IB, 2B, and 3B, respectively.
  • PDL1-IL-15 IgM antitumor activity in combination with Daratumumab was evaluated in a RPMI-8226 human multiple myeloma xenograft model.
  • CB17 NOD/SCID mice were implanted subcutaneously with IxlO 7 RPMI-8226 cells in 1 : 1 PBS/Matrigel.
  • mice were randomized in groups of 10, and dosed intraperitoneally (ip) as shown in Table 2.
  • ip intraperitoneally
  • FIG. 4A The resulting tumor volumes over time for groups 1, 2, 4, and 5 are shown in FIG. 4A and for groups 1, 2, 3, and 6 are shown in FIG. 4B.
  • FIG. 4C A comparison of tumor volumes on day 55 is shown in FIG. 4C.
  • the probability of survival for each group over time is shown in FIG. 4D.
  • PDL1-IL-15 IgM antitumor activity in combination with Trastuzumab was evaluated in a SKOV3 human ovarian cancer xenograft model.
  • BALB/c nude mice were implanted subcutaneously with IxlO 7 SKOV-3 cells in 1 : 1 PBS/Matrigel.
  • mice were randomized in groups of 10, and dosed intraperitoneally (ip) as shown in Table 3.
  • ip intraperitoneally
  • FIG. 5A The resulting tumor volumes over time for groups 1, 2, 4, and 5 are shown in FIG. 5A and for groups 1, 2, 3, and 6 are shown in FIG. 5B.
  • FIG. 5C A comparison of tumor volumes on day 55 is shown in FIG. 5C.
  • the probability of survival for each group over time is shown in FIG. 5D.
  • PDL1-IL-15 IgM antitumor activity in combination with Cetuximab was evaluated in a A431 epidermoid carcinoma xenograft model.
  • CB 17/SCID mice were implanted subcutaneously with IxlO 7 A431 cells in 1 : 1 PBS/Matrigel.
  • mice were randomized in groups of 10, and dosed intraperitoneally (ip) as shown in Table 4.
  • ip intraperitoneally
  • PDL1-IL-15 IgM antitumor activity in combination with Cetuximab is evaluated in a A431 epidermoid carcinoma xenograft model.
  • Humanized MHC- /- NSG mice are implanted subcutaneously with IxlO 7 A431 cells in 1 : 1 PBS/Matrigel.
  • mice are randomized in groups of 10, and are implanted with IxlO 7 human peripheral blood mononuclear cells (PBMCs). After two days, mice are dosed intraperitoneally (ip) as shown in Table 5.
  • PBMCs peripheral blood mononuclear cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Mycology (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente divulgation concerne des méthodes de traitement du cancer à l'aide d'une polythérapie comprenant une molécule de liaison multivalente qui se lie à PD-L1 et comprend une chaîne J modifiée qui comprend un agent de stimulation immunitaire, et un anticorps monoclonal qui se lie spécifiquement à la classe de différenciation 38 (CD38), au récepteur 2 du facteur de croissance épidermique humain (HER2), ou au récepteur du facteur de croissance épidermique (EGFR).
PCT/US2023/064548 2022-03-18 2023-03-16 Utilisation d'une molécule de liaison anti-pd-l1 multimère en combinaison avec une thérapie par anticorps monoclonal WO2023178253A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263321607P 2022-03-18 2022-03-18
US63/321,607 2022-03-18

Publications (2)

Publication Number Publication Date
WO2023178253A2 true WO2023178253A2 (fr) 2023-09-21
WO2023178253A3 WO2023178253A3 (fr) 2023-10-19

Family

ID=88024527

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/064548 WO2023178253A2 (fr) 2022-03-18 2023-03-16 Utilisation d'une molécule de liaison anti-pd-l1 multimère en combinaison avec une thérapie par anticorps monoclonal

Country Status (1)

Country Link
WO (1) WO2023178253A2 (fr)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102037541B1 (ko) * 2011-10-28 2019-10-29 테바 파마슈티컬즈 오스트레일리아 피티와이 엘티디 폴리펩티드 구축물 및 이의 용도
US10954302B2 (en) * 2016-05-09 2021-03-23 Igm Biosciences, Inc. Anti-PD-L1 antibodies
US20190048055A1 (en) * 2017-03-31 2019-02-14 Altor Bioscience Corporation Alt-803 in combination with anti-cd38 antibody for cancer therapies

Also Published As

Publication number Publication date
WO2023178253A3 (fr) 2023-10-19

Similar Documents

Publication Publication Date Title
US11578131B2 (en) Polynucleotides encoding death domain-containing receptor-5 (DR5) binding molecules
EP3265575B1 (fr) Molécules fixant pdk20 et leurs utilisations
US20210388098A1 (en) Multimeric cd137/4-1bb binding molecules and uses thereof
US20230203119A1 (en) Immunostimulatory multimeric binding molecules
KR20200123782A (ko) 암치료를 위해 화학치료제와 병용된 다중결합 항-dr5 결합분자의 용도
JP2019528683A (ja) 多量体gitr結合分子及びその使用
US20220289856A1 (en) Multimeric bispecific anti-cd123 binding molecules and uses thereof
US20220372142A1 (en) Multimeric antibodies with enhanced selectivity for cells with high target density
KR20230005228A (ko) Pd-1 효능제 다량체 결합 분자
JP2024508709A (ja) 抗cd123結合分子及びその使用法
WO2023178253A2 (fr) Utilisation d'une molécule de liaison anti-pd-l1 multimère en combinaison avec une thérapie par anticorps monoclonal
WO2023064900A1 (fr) Méthodes d'utilisation de molécules de liaison anti-pd-l1 multimères
AU2023216348A1 (en) Anti-cd38 binding molecules and uses thereof
WO2023150677A2 (fr) Molécules de liaison anti-cd38 et leurs utilisations

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23771675

Country of ref document: EP

Kind code of ref document: A2