WO2024146955A1 - Molécules de liaison à l'antigène - Google Patents

Molécules de liaison à l'antigène Download PDF

Info

Publication number
WO2024146955A1
WO2024146955A1 PCT/EP2024/050235 EP2024050235W WO2024146955A1 WO 2024146955 A1 WO2024146955 A1 WO 2024146955A1 EP 2024050235 W EP2024050235 W EP 2024050235W WO 2024146955 A1 WO2024146955 A1 WO 2024146955A1
Authority
WO
WIPO (PCT)
Prior art keywords
antigen
binding molecule
binding
cell
receptor
Prior art date
Application number
PCT/EP2024/050235
Other languages
English (en)
Inventor
John Edward Connolly
Anna-Marie FAIRHURST
Xiaohua Zhou
Original Assignee
Twain Therapeutics Pte. Ltd.
CLEGG, Richard Ian
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 Twain Therapeutics Pte. Ltd., CLEGG, Richard Ian filed Critical Twain Therapeutics Pte. Ltd.
Publication of WO2024146955A1 publication Critical patent/WO2024146955A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/22Immunoglobulins specific features characterized by taxonomic origin from camelids, e.g. camel, llama or dromedary
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin

Definitions

  • the present disclosure relates to the fields of molecular biology and methods of medical treatment and prophylaxis.
  • the present disclosure relates to antigen-binding molecules that bind to polypeptides of ⁇ c-containing cytokine receptors.
  • Background Interleukins play a central role in maintaining T cell homeostasis and mediating proper immune responses.
  • interleukins and associated cytokines serve as the means of communication for innate and adaptive immune cells as well as non-immune cells and tissues.
  • interleukins have a critical role in cancer development, progression and control (Briukhovetska D. et al. Nat Rev Cancer 21, 481–499 (2021)). Additionally, interleukins have a critical role in autoimmune diseases (Moudgil and Choubey. J Interferon Cytokine Res. (2011) 31(10): 695–703). The use of interleukins in therapy has shown much promise but has been associated with drawbacks and disappointing results.
  • IL-2 was the first interleukin to be approved for cancer treatment, although its use entails major safety concerns. The high dose of IL-2 that is required for effective treatment of certain diseases is highly toxic.
  • VLS vascular leak syndrome
  • IL-15 monogamma chain, ⁇ c, or CD132
  • CD132 common cytokine receptor polypeptide that is common to the cytokine receptor complexes of at least six different interleukin receptors (i.e.
  • ⁇ c can form functional receptors for cytokine proteins and can transmit signals from one cell to another and direct programs of cellular differentiation.
  • Heterodimerization of ⁇ c and other polypeptide(s) is necessary and sufficient for effective signal transduction through the interaction of their cytoplasmic domains and subsequent kinase activation of multiple signaling pathways.
  • heterodimerization of IL-7R ⁇ and ⁇ c is necessary for effective IL-7 signal transduction.
  • Cytokines that signal through cytokine receptors comprising ⁇ c i.e.
  • IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21 have been shown to be implicated in numerous autoimmune diseases such as multiple sclerosis (MS), Sjögren's syndrome (SS), and ulcerative colitis.
  • MS multiple sclerosis
  • SS Sjögren's syndrome
  • IL-7R ⁇ Both IL-7 and the IL-7 receptor alpha (IL-7R ⁇ ) have been shown to be overexpressed in the cerebrospinal fluid and labial salivary glands of patients with MS and SS. This has led to the initiation of numerous clinical trials relating to immunotherapies which target IL-7R ⁇ . For example, clinical trials were conducted to investigate the effects of IL-7R ⁇ antagonists, such as NCT02045732, NCT03980080, and NCT01808482.
  • Antigen-binding molecules that bind to ⁇ c and IL-2R ⁇ are disclosed e.g. in WO 2017/021540 A1. Summary In one aspect, the present disclosure provides an antigen-binding molecule, optionally isolated, comprising: (i) a ⁇ c-binding moiety, and (ii) a moiety that binds to IL-7R ⁇ . In some embodiments, the antigen-binding molecule is an agonist of a ⁇ c-containing cytokine receptor.
  • the antigen-binding molecule is an antagonist of a ⁇ c-containing cytokine receptor. In some embodiments, the antigen-binding molecule further comprises: (iii) an antigen-binding moiety that binds to a target antigen other than a ⁇ c-containing cytokine receptor polypeptide. In some embodiments, the target antigen other than a ⁇ c-containing cytokine receptor polypeptide is a disease-associated antigen or an antigen expressed by an immune cell.
  • CAR chimeric antigen receptor
  • the present disclosure also provides a nucleic acid, or a plurality of nucleic acids, optionally isolated, encoding an antigen-binding molecule according to the present disclosure or a CAR according to the present disclosure.
  • the present disclosure also provides an expression vector, or a plurality of expression vectors, comprising a nucleic acid or a plurality of nucleic acids according to the present disclosure.
  • the present disclosure also provides a cell comprising an antigen-binding molecule according to the present disclosure, a CAR according to the present disclosure, a nucleic acid or a plurality of nucleic acids according to the present disclosure, or an expression vector or a plurality of expression vectors according to the present disclosure.
  • the present disclosure also provides an antigen-binding molecule according to the present disclosure, a CAR according to the present disclosure, a nucleic acid or a plurality of nucleic acids according to the present disclosure, an expression vector or a plurality of expression vectors according to the present disclosure, a cell according to the present disclosure, or a composition according to the present disclosure, for use in a method of treatment or prophylaxis.
  • the present disclosure also provides a use of an antigen-binding molecule according to the present disclosure, a CAR according to the present disclosure, a nucleic acid or a plurality of nucleic acids according to the present disclosure, an expression vector or a plurality of expression vectors according to the present disclosure, a cell according to the present disclosure, or a composition according to the present disclosure, in the manufacture of a medicament for use in a method of treatment or prophylaxis.
  • the present disclosure also provides a method of treatment or prophylaxis, comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an antigen-binding molecule according to the present disclosure, a CAR according to the present disclosure, a nucleic acid or a plurality of nucleic acids according to the present disclosure, an expression vector or a plurality of expression vectors according to the present disclosure, a cell according to the present disclosure, or a composition according to the present disclosure.
  • Mewburn ref.008537086 4 is a method of treating or preventing a disease/condition characterised by T cell dysfunction, a cancer, an infectious disease, or an autoimmune disease.
  • the cancer is selected from the group consisting of: colon cancer, colon carcinoma, colorectal cancer, nasopharyngeal carcinoma, cervical carcinoma, oropharyngeal carcinoma, gastric carcinoma, hepatocellular carcinoma, head and neck cancer, head and neck squamous cell carcinoma (HNSCC), oral cancer, laryngeal cancer, prostate cancer, lung cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, urothelial carcinoma, melanoma, advanced melanoma, renal cell carcinoma, ovarian cancer or mesothelioma.
  • HNSCC head and neck squamous cell carcinoma
  • the antigen-binding molecule is an inhibitor of signalling mediated by a ⁇ c- containing cytokine receptor.
  • the ⁇ c-containing cytokine receptor comprises IL- 2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ , or IL-7R ⁇ .
  • the ⁇ c-containing cytokine receptor comprises ⁇ c and further comprises IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ , or IL-7R ⁇ .
  • the ⁇ c-binding moiety comprises a heavy chain variable (VH) region and a light chain variable (VL) region that binds to ⁇ c.
  • the antigen-binding moiety comprises an Fv and/or a Fab that binds to ⁇ c.
  • the ⁇ c-binding moiety comprises a heavy chain variable (VH) region and a light chain variable (VL) region of an antibody that binds to ⁇ c.
  • the antigen-binding moiety comprises an Fv and/or a Fab of an antibody that binds to ⁇ c.
  • the present disclosure also provides a method for inhibiting signalling mediated by a ⁇ c-containing cytokine receptor, comprising contacting a cell expressing a polypeptide of a ⁇ c-containing cytokine receptor in vitro, in vivo or ex vivo with an antigen-binding molecule, optionally isolated, comprising: (i) a ⁇ c-binding moiety, and (ii) an IL-7R ⁇ -binding moiety.
  • the frequency of ⁇ c forming a complex with IL-2R ⁇ , IL-2R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL- 15R ⁇ , and/or IL-21R ⁇ is inhibited.
  • signalling mediated by IL-2R ⁇ , IL-2R ⁇ , IL-4R ⁇ , IL-7R ⁇ , IL-9R ⁇ , IL-15R ⁇ , and/or IL-21R ⁇ is inhibited.
  • IL-2, IL-4, IL-7, IL-9, IL-15, and/or IL-21 mediated signalling is inhibited.
  • the present disclosure includes the combination of the aspects and preferred features described except where such a combination is clearly impermissible or expressly avoided.
  • the IL-7R ⁇ -binding moiety as disclosed herein inhibits signalling mediated by IL-7R ⁇ , and the ⁇ c-binding moiety reduces the availability of ⁇ c by binding and sequestering ⁇ c.
  • Sequestering ⁇ c reduces the availability of ⁇ c to form a heteromeric complex with a polypeptide of the ⁇ c receptor family (e.g. IL-2R ⁇ , IL-2R ⁇ , IL-4R ⁇ , IL-7R ⁇ , IL-9R ⁇ , IL-15R ⁇ , or IL-21R ⁇ ). Therefore, the disclosed antigen binding molecules may affect the signalling of multiple ⁇ c-associated cytokine receptors, not just IL-7R ⁇ . As a result, non-specific agonism is reduced.
  • All receptors of the ⁇ c receptor family comprise ⁇ c as a constituent polypeptide.
  • Janus kinas 3 (JAK3) associates with ⁇ c, and upon activation of a ⁇ c-containing cytokine receptor, JAK3 becomes phosphorylated and activated. Phosphorylated JAK3 then phosphorylates and activates downstream signalling proteins such as STAT5, and also triggers signalling through the MAPK/ERK and PI3K/Akt signal transduction pathways. Signalling through ⁇ c family receptors promotes immune cell activation, proliferation and survival.
  • ⁇ c may be characterised as having at least 70% sequence identity, preferably one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to the amino acid sequence of the reference protein.
  • a ‘fragment’ generally refers to a fraction of the reference protein.
  • a ‘variant’ generally refers to a protein having an amino acid sequence comprising one or more amino acid substitutions, insertions, deletions or other modifications relative to the amino acid sequence of the reference protein, but retaining a considerable degree of sequence identity (e.g. at least 60%) to the amino acid sequence of the reference protein.
  • Isoforms, fragments, variants or homologues of a given reference protein may optionally be characterised as having at least 70%, preferably one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to the amino acid sequence of an immature or mature (i.e. after processing to remove signal peptide) form of a specified isoform of the relevant protein from a given species, e.g. human.
  • Isoforms, fragments, variants or homologues of ⁇ c may optionally be characterised as having at least 70%, preferably one of 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to the amino acid sequence of an immature or mature ⁇ c isoform from a given species, e.g. human.
  • Isoforms, fragments, variants or homologues may optionally be functional isoforms, fragments, variants or homologues, e.g. having a functional property/activity of the reference ⁇ c (e.g.
  • an isoform, fragment, variant or homologue of ⁇ c may display one or more of: association with one or more of IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ , or IL-7R ⁇ , or binding to one or more of IL-2, IL-15, IL-4, IL-9, IL-21 or IL-7.
  • the type I IL-4 receptor (containing IL-4R ⁇ and ⁇ c) also activates STAT5 signalling. Additionally, IL-4 (via type I IL-4 receptor) activates IRS2 efficiently, therefore IL-4 subsequently activates various pathways including Sos/Ras, PI3K/Akt, PKB/mTOR, or PKC.
  • Interleukin-7 IL-7
  • IL-7 Interleukin-7
  • IL-7 a molecule known for its growth-promoting effects on progenitors of B cells, plays a vital role in health maintenance and disease prevention, and congenital deficiency of IL-7 signaling leads to profound immunodeficiency. Elevated IL-7 levels have been associated with poor prognosis of a number of cancers (Zarogoulidis et al., J Cancer.
  • ⁇ c-containing cytokine receptor- mediated signalling may be mediated by a polypeptide complex comprising IL-15, ⁇ c, IL-2R ⁇ and IL-15R ⁇ (i.e. a IL-15: ⁇ c:IL-2R ⁇ :IL-15R ⁇ complex).
  • IL-15: ⁇ c:IL-2R ⁇ :IL-15R ⁇ - mediated signalling i.e. signalling mediated by binding of IL-15 to the IL-15 receptor.
  • ⁇ c-containing cytokine receptor-mediated signalling may be mediated by a polypeptide complex comprising ⁇ c and IL-4R ⁇ (i.e. a ⁇ c:IL-4R ⁇ complex).
  • an ‘antigen-binding molecule’ refers to a molecule that binds to (a) given target antigen(s).
  • the antigen-binding molecules of the present disclosure comprise one or more antigen- binding moieties, through which the antigen-binding molecule binds to its target antigen(s).
  • Antigen-binding moieties may comprise, or may be derived from, antibodies (i.e. immunoglobulins (Igs)) and antigen-binding fragments thereof.
  • a replacement amino acid may be a non-naturally occurring amino acid residue – i.e. an amino acid residue other than those recited in the preceding sentence.
  • non-naturally occurring amino acid residues include norleucine, ornithine, norvaline, homoserine, aib, and other amino acid residue analogues such as those described in Ellman et al., Meth. Enzym. (1991) 202:301-336.
  • a substitution may be biochemically conservative.
  • an antigen-binding molecule may be a bispecific antibody conjugate (e.g. an IgG2, F(ab’)2 or CovX-Body), a bispecific IgG or IgG-like molecule (e.g. an IgG, scFv 4 -Ig, IgG-scFv, scFv-IgG, DVD-Ig, IgG-sVD, sVD- IgG, 2 in 1-IgG, mAb 2 , or Tandemab common LC), an asymmetric bispecific IgG or IgG-like molecule (e.g.
  • a bispecific antibody conjugate e.g. an IgG2, F(ab’)2 or CovX-Body
  • a bispecific IgG or IgG-like molecule e.g. an IgG, scFv 4 -Ig, IgG-scFv, scFv-IgG, DVD
  • the multispecific antigen-binding molecule is minimalistic bispecific antibody, such as a tandem scFV-scFv, a tandem VHH-VHH, or a tandem VHH-scFv antibody.
  • a tandem multispecific antigen-binding molecule (e.g., a tandem scFv) comprises two (or more) binding moieties (e.g., scFv and/or VHH moieties) and a linker.
  • the multispecific antigen- binding molecule is provided in a tandem format, where binding moieties are joined by a linker.
  • a P2C4 scFv can be combined with a P1A3 scFv in at least the following orientations: (i) P2C4VL–P2C4VH–linker–P1A3VL–P1A3VH, (ii) P1A3VL–P1A3VH– linker–P2C4VL– P2C4VH, (iii) P2C4VH–P2C4VL–linker–P1A3VH–P1A3VL, and (iv) P1A3VH–P1A3VL– linker–P2C4VH– P2C4VL.
  • the multispecific antigen-binding molecule comprises a linker between binding moieties, for example, a linker between a ⁇ c-binding moiety and a moiety that binds to a polypeptide of a Mewburn ref.008537086 29 ⁇ c-containing cytokine receptor other than ⁇ c (e.g., an IL-2R ⁇ -binding moiety).
  • linkers are described by Brinkmann and Kontermann (MAbs. (2017) 9(2):182-212), which is hereby incorporated by reference in its entirety.
  • the linker is a linker described in Brinkmann and Kontermann (MAbs. (2017) 9(2):182-212).
  • the linker is an amino acid linker. In some embodiments, the linker is a flexible linker. In some embodiments, the linker is a rigid linker. In some embodiments, the linker is a short flexible linker. In some embodiments, the linker is a long rigid linker. In some embodiments, the flexible linker is rich in small or polar amino acids such as Gly and/or Ser to provide flexibility and solubility. In some embodiments, the linker is a glycine-rich linker. In some embodiments, the linker is a serine-rich linker. In some embodiments, the linker is an amino acid linker in which at least 50% of the total amino acids are glycine amino acids, e.g.
  • the linker comprises or consists of a GGGGSGGGS (SEQ ID NO:452) amino acid sequence. In some embodiments, the linker comprises or consists of a (G3S)4 (SEQ ID NO:453) amino acid sequence. In some embodiments, the linker comprises or consists of a GGGSG (SEQ ID NO:454) amino acid sequence. In some embodiments, the linker sequence comprises at least one glycine residue and/or at least one serine residue. In some embodiments, the linker sequence comprises or consists of glycine and serine residues.
  • the linker sequence comprises one or more (e.g., 1, 2, 3, 4, 5 or 6) copies (e.g., in tandem) of the sequence motif G4S.
  • the linker sequence comprises or consists of (G4S)4 or (G4S)6.
  • the linker sequence has a length of 1-2, 1-3, 1-4, 1-5, 1-10, 1-15, 1-20, 1-25, or 1-30 amino acids.
  • the linker is a rigid linker.
  • the rigid linker forms an alpha helical structure between binding moieties. Rigid linkers are discussed by Arai et al. (Protein Engineering, Design and Selection, 14(8), 2001, 529–532), which is hereby incorporated by reference in its entirety.
  • the linker is a linker described in Arai et al. (Protein Engineering, Design and Selection, 14(8), 2001, 529–532).
  • the linker comprises or consists of an A(EAAAK)5A (SEQ ID NO:451) amino acid sequence.
  • the linker comprises an EAAAK (SEQ ID NO:455) amino acid sequence.
  • the linker comprises or consists of an A(EAAAK) 2 A (SEQ ID NO:456) amino acid Mewburn ref.008537086 30 sequence. In some embodiments, the linker comprises or consists of an A(EAAAK)3A (SEQ ID NO:457) amino acid sequence. In some embodiments, the linker comprises or consists of an A(EAAAK)4A (SEQ ID NO:458) amino acid sequence. In some embodiments, the linker comprises or consists of an A(EAAAK)5A (SEQ ID NO:451) amino acid sequence. In some embodiments, the linker has a length of at least 3 amino acids. In some embodiments, the linker has a maximum length of 50 amino acids.
  • the linker has a minimum length of one of 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, or 49 amino acids. In some embodiments, the linker has a maximum length of one of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 amino acids.
  • the linker has a length between 3 and 20 amino acids, 4 and 20 amino acids, 5 and 20 amino acids, 6 and 20 amino acids, 7 and 20 amino acids, 8 and 20 amino acids, 9 and 20 amino acids, 10 and 20 amino acids, 11 and 20 amino acids, 12 and 20 amino acids, 13 and 20 amino acids, 14 and 20 amino acids, 15 and 20 amino acids, 16 and 20 amino acids, 17 and 20 amino acids, 18 and 20 amino acids, or 19 and 20 amino acids.
  • Methods for producing multispecific antigen-binding molecules include chemical crosslinking of antigen- binding molecules or antibody fragments, e.g. with reducible disulphide or non-reducible thioether bonds, for example as described in Segal and Bast, (2001) Current Protocols in Immunology. Chapter 2:2.13.1– 2.13.16, which is hereby incorporated by reference in its entirety.
  • the light and heavy chain variable domains for the antigen-binding fragment Mewburn ref.008537086 32 capable of binding ⁇ c, and the light and heavy chain variable domains for the antigen-binding fragment capable of binding to another target protein), and including sequences encoding a suitable linker or dimerization domain between the antigen-binding fragments can be prepared by molecular cloning techniques.
  • Recombinant bispecific antibody can thereafter be produced by expression (e.g. in vitro) of the construct in a suitable host cell (e.g. a mammalian host cell), and expressed recombinant bispecific antibody can then optionally be purified.
  • the ⁇ c-binding moiety comprises a polypeptide comprising a VH region comprising HC-CDR1, HC-CDR2 and HC-CDR3 as indicated for one of binding moieties A1-1 to A1-27 in column A of Table A1, optionally wherein 1 or 2 or 3 amino acids in HC-CDR1, and/or 1 or 2 or 3 amino acids in HC-CDR2, and/or 1 or 2 or 3 amino acids in HC-CDR3 are substituted with another amino acid.
  • the ⁇ c-binding moiety comprises a polypeptide or polypeptides comprising: (i) a VH region comprising HC-FR1, HC-FR2, HC-FR3 and HC-FR4 as indicated in column A of Table B1, and (ii) a VL region comprising LC-FR1, LC-FR2, LC-FR3 and LC-FR4 as indicated in column B of Table B1, wherein the sequences of columns A and B are selected from the same row of Table B1 (i.e., wherein the sequences of columns A and B are of the same binding moiety selected from B1-1 to B1-27).
  • the ⁇ c-binding moiety of the present disclosure comprises a polypeptide or polypeptides comprising a VH region, and a VL region of a clone selected from: P1A3, P1A3_B3, P1A3_E8, P1A3_E9, P2B9, P1A3_B4, P1A3_FW2, P1A10, P1B6, P1C10, P1D7, P1E8, P2B2, P2B7, P2D11, P2F10, P2H4, P2D3, P1G4, P1B12, P1C7, P1A3_A, P1A3_Q, P1A3_AQ, P1A3_ANQ, P1A10_AQ, and P1A10_ANQ, as shown in Table C1 herein.
  • the IL-7R ⁇ -binding moiety of the present disclosure comprises a polypeptide or polypeptides comprising the heavy chain CDRs, and the light chain CDRs, of an antigen binding Mewburn ref.008537086 34 molecule, antibody, and/or clone described herein.
  • the IL-7R ⁇ -binding moiety of the present disclosure comprises a polypeptide or polypeptides comprising the heavy chain CDRs, and the light chain CDRs, of an antigen binding molecule, antibody, and/or clone described in Belarif et al, Nature Communications 9, 4483.2018, Herold et al., JCI Insight.2019 Dec 19; 4(24): e126054, Ellis et al., Br J Clin Pharmacol.2019 Feb;85(2):304-315, Hixon et al, Leukemia.2020 Jan; 34(1): 35–49, US10392441B2, WO2011104687A1, US20120282254A1, WO2015/189302A1, WO2016/059512A1, or US 9,150,653, which are hereby incorporated by reference in their entirety.
  • the IL-7R ⁇ -binding moiety of the present disclosure comprises a polypeptide or polypeptides comprising a VH region, and a VL region, of an antigen binding molecule, antibody, and/or clone described herein.
  • the IL-7R ⁇ -binding moiety of the present disclosure comprises a polypeptide or polypeptides comprising a VH region, and a VL region, of an antigen binding molecule, antibody, and/or clone described in Belarif et al, Nature Communications 9, 4483.2018, Herold et al., JCI Insight.2019 Dec 19; 4(24): e126054, Ellis et al., Br J Clin Pharmacol.2019 Feb;85(2):304- 315, Hixon et al, Leukemia.2020 Jan; 34(1): 35–49, US10392441B2, WO2011104687A1, US20120282254A1, WO2015/189302A1, WO2016/059512A1, or US 9,150,653, which are hereby incorporated by reference in their entirety.
  • the IL-7R ⁇ -binding moiety comprises a polypeptide comprising a VH region comprising heavy chain CDR1 of SEQ ID NOs 359, 376, 386, or 398, heavy chain CDR2 of SEQ ID NOs 360, 377, 387, or 399, and heavy chain CDR3 of SEQ ID NOs 361, 362, 378, 388, 389, 390, 391, 392, 393, 394, or 400, optionally wherein 1 or 2 or 3 amino acids in heavy chain CDR1, and/or 1 or 2 or 3 amino acids in heavy chain CDR2, and/or 1 or 2 or 3 amino acids in heavy chain CDR3 are substituted with another amino acid.
  • the IL-7R ⁇ -binding moiety of the present disclosure comprises a polypeptide comprising a VL region comprising light chain CDR1, light chain CDR2, and light chain CDR3, and a VH region comprising heavy chain CDR1, heavy chain CDR2, and heavy chain CDR3 of a clone selected from: N13B2-h1, N13B2-h2, N13B2-h3, Pf1, 1A11, 1A11 VH3 N98D, 1A11 VH3 N98E, 1A11 VH3 F100bE, 1A11 VH3 F100bH, 1A11 VH3 F100bI, 1A11 VH3 F100bV, 6A3, 1A11.L1, 1A11.L2, 1A11.L3, 1A11.L4, 1A11.L5, 1A11.L6, 1A11.L7, 1A11.L8, 1A11.L9, 6A3.L0, 6A3.L1, 6A3.L2, 6A3.L3, or 6A3.L27.
  • the IL-7R ⁇ -binding moiety of the present disclosure comprises a polypeptide comprising a VH region comprising, Mewburn ref.008537086 35 HC-CDR1: FTLSDYYMA (SEQ ID NO:359); HC-CDR2: TISASGLRTYYPDSVK (SEQ ID NO:360); HC-CDR3: PMSAHYGFNYFDY (SEQ ID NO:361) or PLSAHYGFNYFDY (SEQ ID NO:362); and a VL region comprising: LC-CDR1: RTSEDIYNGLA (SEQ ID NO:363) or CRTSEDIYQGLA (SEQ ID NO:364); LC-CDR2: SANSLHI (SEQ ID NO:365) or SANTLHI (SEQ ID NO:366); LC-CDR3: QQYYDYPLA (SEQ ID NO:367); In some embodiments, the IL-7R ⁇ -binding moiety of the VH region compris
  • the IL-7R ⁇ -binding moiety comprises a polypeptide comprising a VH region having at least 70%, e.g. one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to the amino acid sequence of SEQ ID NO: 368, 369, 370, 382, 404, 405, 406, 407, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, 431, 432, 433, and/or 434.
  • the IL-7R ⁇ -binding moiety comprises a polypeptide comprising a VL region having at least 70%, e.g. one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to the amino acid sequence of SEQ ID NO: 371, 372, 373, 383, 435, 436, 437, 438, 439, 440, 441, 442, 443, 444, 445, 446, 447, and/or 448.
  • the IL-7R ⁇ -binding moiety comprises a polypeptide comprising a VH region having at least 70%, e.g. one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to the amino acid sequence of SEQ ID NO: 368, 369, 370, 382, 404, 405, 406, 407, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, 431, 432, 433, and/or 434, and a polypeptide comprising a VL region having at least 70%, e.g.
  • the IL-7R ⁇ -binding moiety comprises a polypeptide comprising a VH region having at least 70%, e.g.
  • the IL-7R ⁇ -binding moiety comprises a polypeptide comprising a VH region having at least 70%, e.g.
  • the IL-7R ⁇ -binding moiety comprises a VH region having at least 70%, e.g.
  • the IL-7R ⁇ -binding moiety comprises a VH region having at least 70%, e.g.
  • the IL-7R ⁇ -binding moiety comprises a VH region having at least 70%, e.g. one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to SEQ ID NO:370, and a VL region having at least 70%, e.g. one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to SEQ ID NO:373.
  • the IL-7R ⁇ -binding moiety comprises a VH region having at least 70%, e.g. one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to SEQ ID NO:382, and a VL region having at least 70%, e.g. one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to SEQ ID NO:383.
  • the IL-7R ⁇ -binding moiety comprises a VH region having at least 70%, e.g. one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to SEQ ID NO:404, and a VL region having at least 70%, e.g. one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to SEQ ID NO:435.
  • the IL-7R ⁇ -binding moiety binds an epitope comprising an amino acid sequence having at least 70%, e.g. one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to SEQ ID NO:375.
  • the antigen-binding molecule according to the present disclosure comprises: (i) a ⁇ c-binding moiety according to an embodiment described herein, and (ii) a moiety that binds to IL-7R ⁇ . In some embodiments, the antigen-binding molecule according to the present disclosure comprises: (i) a ⁇ c-binding moiety according to an embodiment as described herein, and (ii) an IL-7R ⁇ -binding moiety according to an embodiment described herein.
  • the immunoglobulin heavy chain constant sequence is, or is derived from, the heavy chain constant sequence of a human IgG1 allotype (e.g. G1m1, G1m2, G1m3 or G1m17).
  • the antigen-binding molecule of the present disclosure comprises one or more regions of an immunoglobulin light chain constant sequence.
  • the immunoglobulin light chain constant sequence is human immunoglobulin kappa constant (IGKC; C ⁇ ).
  • the immunoglobulin light chain constant sequence is a human immunoglobulin lambda constant (IGLC; C ⁇ ), e.g. IGLC1, IGLC2, IGLC3, IGLC6 or IGLC7.
  • the antigen-binding molecules of the present disclosure comprise an Fc region. In some embodiments, the antigen-binding molecules of the present disclosure do not comprise an Fc region.
  • an ‘Fc region’ refers to a polypeptide complex formed by interaction between two polypeptides, each polypeptide comprising the CH2-CH3 region of an immunoglobulin (Ig) heavy chain constant sequence.
  • a ‘CH2 region’ refers to an amino acid sequence corresponding to the CH2 region of an immunoglobulin (Ig).
  • the CH2 region is the region of an Ig formed by positions 231 to 340 of the immunoglobulin constant region, according to the EU numbering system described in Edelman et al., Proc Natl Acad Sci USA (1969) 63(1):78-85.
  • a ‘CH3 region’ refers to an amino acid sequence corresponding to the CH3 region of an immunoglobulin (Ig).
  • the CH3 region is the region of an Ig formed by positions 341 to 447 of the immunoglobulin constant region, according to the EU numbering system described in Edelman et al., Proc Natl Acad Sci USA (1969) 63(1):78-85.
  • a ‘CH2-CH3 region’ refers to an amino acid sequence corresponding to the CH2 and CH3 regions of an immunoglobulin (Ig).
  • the CH2- CH3 region is the region of an Ig formed by positions 231 to 447 of the immunoglobulin constant region, according to the EU numbering system described in Edelman et al., Proc Natl Acad Sci USA (1969) 63(1):78-85.
  • a CH2 region, CH3 region and/or a CH2-CH3 region according to the present disclosure corresponds to the CH2 region/CH3 region/CH2-CH3 region of an IgG (e.g. IgG1, IgG2, IgG3, IgG4), IgA (e.g.
  • the CH2 region, CH3 region and/or a CH2-CH3 region corresponds to the CH2 region/CH3 region/CH2-CH3 region of a human IgG (e.g. hIgG1, hIgG2, hIgG3, hIgG4), hIgA (e.g. hIgA1, hIgA2), hIgD, hIgE or hIgM.
  • a human IgG e.g. hIgG1, hIgG2, hIgG3, hIgG4
  • hIgA e.g. hIgA1, hIgA2
  • hIgD hIgE or hIgM.
  • Fc-mediated functions include Fc receptor binding, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), complement-dependent Mewburn ref.008537086 40 cytotoxicity (CDC), formation of the membrane attack complex (MAC), cell degranulation, cytokine and/or chemokine production, and antigen processing and presentation.
  • ADCC antibody-dependent cellular cytotoxicity
  • ADCP antibody-dependent cell-mediated phagocytosis
  • CDC complement-dependent Mewburn ref.008537086 40 cytotoxicity
  • MAC membrane attack complex
  • the antigen-binding molecule of the present disclosure comprises an Fc region comprising modification to increase or reduce an Fc-mediated function as compared to an antigen-binding molecule comprising the corresponding unmodified Fc region.
  • the modification may be present in one or both of the polypeptide chains which together form the Fc region.
  • the antigen-binding molecule of the present disclosure comprises an Fc region comprising modification.
  • the antigen-binding molecule of the present disclosure comprises an Fc region comprising modification in one or more of the CH2 and/or CH3 regions.
  • the Fc region comprises modification to increase an Fc-mediated function.
  • the Fc region comprises modification to increase ADCC.
  • the Fc region comprises modification to increase ADCP.
  • the Fc region comprises modification to increase CDC.
  • an antigen-binding molecule comprising an Fc region comprising modification to increase an Fc-mediated function induces an increased level of the relevant effector function as compared to an antigen-binding molecule comprising the corresponding unmodified Fc region.
  • the Fc region comprises modification to increase binding to an Fc receptor.
  • the Fc region comprises modification to increase binding to an Fc ⁇ receptor.
  • the Fc region comprises modification to increase binding to one or more of Fc ⁇ RI, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ RIIc, Fc ⁇ RIIIa and Fc ⁇ RIIIb.
  • the Fc region comprises modification to increase binding to Fc ⁇ RIIIa. In some embodiments, the Fc region comprises modification to increase binding to Fc ⁇ RIIa. In some embodiments, the Fc region comprises modification to increase binding to Fc ⁇ RIIb. In some embodiments, the Fc region comprises modification to increase binding to FcRn. In Mewburn ref.008537086 41 some embodiments, the Fc region comprises modification to increase binding to a complement protein. In some embodiments, the Fc region comprises modification to increase binding to C1q. In some embodiments, the Fc region comprises modification to promote hexamerisation of the antigen-binding molecule. In some embodiments, the Fc region comprises modification to increase antigen-binding molecule half-life.
  • the Fc region comprises modification corresponding to the combination of substitutions described in Natsume et al., Cancer Res. (2008) 68(10):3863-72. In some embodiments, the Fc region comprises modification corresponding to the combination of substitutions E345R/E430G/S440Y as described in Diebolder et al. Science (2014) 343(6176):1260-3. In some embodiments, the Fc region comprises modification corresponding to the combination of substitutions M252Y/S254T/T256E as described in Dall’Acqua et al. J Immunol. (2002) 169:5171–5180.
  • the Fc region comprises modification corresponding to the combination of substitutions M428L/N434S as described in Zalevsky et al. Nat Biotechnol. (2010) 28:157–159.
  • the Fc region comprises modification corresponding to the combination of substitutions S267E/L328F as described in Chu et al., Mol Immunol. (2008) 45:3926–3933. In some embodiments, the Fc region comprises modification corresponding to the combination of substitutions N325S/L328F as described in Shang et al. Biol Chem. (2014) 289:15309–15318. In some embodiments, the Fc region comprises modification to reduce/prevent an Fc-mediated function. In some embodiments, the Fc region comprises modification to reduce/prevent ADCC. In some embodiments, the Fc region comprises modification to reduce/prevent ADCP. In some embodiments, the Fc region comprises modification to reduce/prevent CDC.
  • an antigen-binding molecule comprising an Fc region comprising modification to reduce/prevent an Fc-mediated function induces a reduced level of the relevant effector function as compared to an antigen-binding molecule comprising the corresponding unmodified Fc region.
  • the Fc region comprises modification to reduce/prevent binding to an Fc receptor.
  • the Fc region comprises modification to reduce/prevent binding to an Fc ⁇ receptor.
  • the Fc region comprises modification to reduce/prevent binding to one or more of Fc ⁇ RI, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ RIIc, Fc ⁇ RIIIa and Fc ⁇ RIIIb.
  • the Fc region comprises modification to reduce/prevent binding to Fc ⁇ RIIIa. In some embodiments, the Fc region comprises modification to reduce/prevent binding to Fc ⁇ RIIa. In some embodiments, the Fc region comprises modification to reduce/prevent binding to Fc ⁇ RIIb. In some embodiments, the Fc region comprises modification to reduce/prevent binding to a complement protein. In some embodiments, the Fc region comprises modification to reduce/prevent binding to C1q. In some embodiments, the Fc region comprises modification to reduce/prevent glycosylation of the amino acid residue corresponding to N297. In some embodiments, the Fc region is not able to induce one or more Fc-mediated functions (i.e.
  • antigen-binding molecules comprising such Fc regions also lack the ability to induce the relevant function(s). Such antigen-binding molecules may be described as being devoid of the relevant function(s).
  • the Fc region is not able to induce ADCC.
  • the Fc region is not able to induce ADCP.
  • the Fc region is not able to induce CDC.
  • the Fc region is not able to induce ADCC and/or is not able to induce ADCP and/or is not able to induce CDC.
  • the Fc region is not able to bind to an Fc receptor.
  • the Fc region is not able to bind to an Fc ⁇ receptor. In some embodiments, the Fc region is not able to bind to one or more of Fc ⁇ RI, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ RIIc, Fc ⁇ RIIIa and Fc ⁇ RIIIb. In some embodiments, the Fc region is not able to bind to Fc ⁇ RIIIa. In some embodiments, the Fc region is not able to bind to Fc ⁇ RIIa. In some embodiments, the Fc region is not able to bind to Fc ⁇ RIIb. In some embodiments, the Fc region is not able to bind to FcRn.
  • the Fc region is not able to bind to a complement Mewburn ref.008537086 43 protein. In some embodiments, the Fc region is not able to bind to C1q. In some embodiments, the Fc region is not glycosylated at the amino acid residue corresponding to N297. In some embodiments, the Fc region comprises modification corresponding to N297A or N297Q or N297G as described in Leabman et al., MAbs. (2013) 5:896–903. In some embodiments, the Fc region comprises modification corresponding to L235E as described in Alegre et al., J Immunol. (1992) 148:3461–3468.
  • the Fc region comprises modification corresponding to the combination of substitutions L234A/L235A or F234A/L235A as described in Xu et al., Cell Immunol. (2000) 200:16–26.
  • the Fc region comprises modification corresponding to P329A or P329G as described in Schlothauer et al., Protein Engineering, Design and Selection (2016), 29(10):457–466.
  • the Fc region comprises modification corresponding to the combination of substitutions L234A/L235A/P329G as described in Lo et al. J. Biol. Chem (2017) 292(9):3900-3908.
  • the Fc region comprises modification corresponding to the combination of substitutions described in Rother et al., Nat Biotechnol. (2007) 25:1256–1264. In some embodiments, the Fc region comprises modification corresponding to the combination of substitutions S228P/L235E as described in Newman et al., Clin. Immunol. (2001) 98:164–174. In some embodiments, the Fc region comprises modification corresponding to the combination of substitutions H268Q/V309L/A330S/P331S as described in An et al., MAbs. (2009) 1:572–579.
  • F234A/L235A in human IgG4 are known to disrupt binding of Fc to Fc ⁇ receptors and inhibit ADCC, ADCP, and also to reduce C1q binding and thus CDC (Schlothauer et al., Protein Engineering, Design and Selection (2016) 29(10):457–466, hereby incorporated by reference in entirety).
  • the substitutions ‘P329G’ and ‘P329A’ reduce C1q binding (and thereby CDC).
  • Substitution of ‘N297’ with ‘A’, ‘G’ or ‘Q’ is known to eliminate glycosylation, and thereby reduce Fc binding to C1q and Fc ⁇ receptors, and thus CDC and ADCC. Lo et al. J. Biol.
  • the Fc region comprises modification corresponding to the substitution S228P as described in Silva et al., J Biol Chem.
  • the Fc region comprises modification corresponding to the combination of substitutions L234A/L235A. In some embodiments, the Fc region comprises modification corresponding to the substitution P329G. In some embodiments, the Fc region comprises modification corresponding to the substitution N297Q. In some embodiments, the Fc region comprises modification corresponding to the combination of substitutions L234A/L235A/P329G. In some embodiments, the Fc region comprises modification corresponding to the combination of substitutions L234A/L235A/P329G/N297Q.
  • the Fc region comprises modification corresponding to the combination of substitutions L234A/L235E/G237A/A330S/P331S. In some embodiments, the Fc region comprises modification corresponding to the substitution S228P, e.g. in IgG4. In some embodiments, the Fc region comprises a CH2-CH3 region comprising an amino acid difference at one or more of the following positions, relative to the amino acid sequence of a CH2-CH3 region of a reference Fc region: 234 or 235 (according to the EU numbering system). In some embodiments, the Fc region comprises a CH2-CH3 region comprising one or more of the following specified amino acid residues: A234 or A235 (according to the EU numbering system).
  • the Fc region comprises a CH2-CH3 region comprising A234 and A235. In some embodiments, the Fc region comprises a CH2-CH3 region comprising one or more of the following amino acid substitutions, relative to the amino acid sequence of a CH2-CH3 region of the reference Fc region: L234A or L235A (according to the EU numbering system). In some embodiments, the Fc region comprises a CH2-CH3 region comprising the following amino acid substitutions, relative to the amino acid sequence of a CH2-CH3 region of the reference Fc region: L234A and L235A (according to the EU numbering system). In some embodiments – particularly embodiments in which the antigen-binding molecule is a multispecific (e.g.
  • the antigen-binding molecule comprises an Fc region comprising modification in one or more of the CH2 and CH3 regions promoting association of the Fc region.
  • Recombinant co-expression of constituent polypeptides of an antigen-binding molecule and subsequent association leads to several possible combinations.
  • modification(s) promoting association of the desired combination of heavy chain polypeptides.
  • Modifications may promote e.g. hydrophobic and/or electrostatic interaction between Mewburn ref.008537086 45 CH2 and/or CH3 regions of different polypeptide chains. Suitable modifications are described e.g.
  • the antigen-binding molecule of the present disclosure comprises an Fc region comprising paired substitutions in the CH3 regions of the Fc region according to one of the following formats, as shown in Table 1 of Ha et al., Front Immnol. (2016) 7:394: KiH, KiHs-s, HA-TF, ZW1, 7.8.60, DD-KK, EW-RVT, EW-RVTs-s, SEED or A107.
  • the multispecific e.g.
  • one of the CH3 regions of the Fc region of the antigen-binding molecule of the present disclosure comprises the substitution (numbering of positions/substitutions in the Fc region herein is according to the EU numbering system as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed.
  • one of the CH3 regions comprises the substitution S354C
  • the other CH3 region of the Fc region comprises the substitution Y349C.
  • Introduction of these cysteine residues results in formation of a disulfide bridge between the two CH3 regions of the Fc region, further stabilizing the heterodimer (Carter, J Immunol Methods (2001) 248:7-15).
  • one of the CH3 regions comprises the substitutions K392D and K409D
  • the other CH3 region of the Fc region comprises the substitutions E356K and D399K.
  • ‘DDKK’ knob-into-hole technology is described e.g. in WO 2014/131694 A1, and promotes assembly of the heavy chains providing the complementary amino acid residues.
  • the antigen-binding molecule of the present disclosure comprises an Fc region modified as described in Labrijn et al., Proc Natl Acad Sci USA. (2013) 110(13):5145-50, referred to as ‘Duobody’ format.
  • one of the CH3 regions comprises the substitution K409R
  • the other CH3 region of the Fc region comprises the substitution K405L.
  • the antigen-binding molecule of the present disclosure comprises an Fc region modified as described in Strop et al., J Mol Biol. (2012) 420(3):204-19, so-called ‘EEE-RRR’ format.
  • one of the CH3 regions comprises the substitutions D221E, P228E and L368E, and the other CH3 region of the Fc region comprises the substitutions D221R, P228R and K409R.
  • the antigen-binding molecule comprises an Fc region comprising the ‘EW-RVT’ modification described in Choi et al., Mol Cancer Ther. (2013) 12(12):2748–59.
  • one of the CH3 regions comprises the substitutions K360E and K409W, and the other CH3 region of the Fc region comprises the substitutions Q347R, D399V and F405T.
  • one of the CH3 regions comprises the substitutions T350V, L351Y, F405A and Y407V
  • the other CH3 region of the Fc region comprises the substitutions T350V, T366L, K392L and T394W (see e.g. Von Kreudenstein et al., MAbs (2013) 5(5):646–54).
  • one of the CH3 regions comprises the substitutions K360D, D399M and Y407A
  • the other CH3 region of the Fc region comprises the substitutions E345R, Q347R, T366V and K409V (see e.g. Leaver-Fay et al., Structure (2016) 24(4):641–51).
  • one of the CH3 regions comprises the substitutions K370E and K409W
  • the other CH3 region of the Fc region comprises the substitutions E357N, D399V and F405T (see e.g. Choi et al., PLoS One (2015) 10(12):e0145349).
  • the antigen-binding molecule of the present disclosure comprises an Fc region comprising modification to increase stability (e.g. thermostability and/or freeze-thaw stability).
  • the antigen-binding molecule comprises modification to one or more of the CH2 and CH3 regions to increase stability (e.g. thermostability and/or freeze-thaw stability).
  • the antigen-binding molecule of the present disclosure comprises CH3 regions (e.g. within an Fc region, e.g. within CH2-CH3 regions forming an Fc region) comprising paired CH3 region 'KiH' or 'KiHS-S' modifications.
  • Such paired CH3 regions may comprise a CH3 region comprising a knob modification, and a CH3 region comprising a hole modification.
  • a CH3 region comprising a knob modification comprises a tryptophan or tyrosine residue at position 366 (i.e.366W or 366Y).
  • the knob modification is or comprises T366W or T366Y.
  • a CH3 region comprising a knob modification comprises 366W. In some embodiments, the knob modification is or comprises T366W. In some embodiments, a CH3 region comprising a hole modification comprises 407V, 407A, 407S or 407T; 366S, 366V or 366A; and 368A, 368V, 368S or 368T. In some embodiments, the hole modification is or comprises Y407V, Y407A, Y407S or Y407T; T366S, T366V or T366A; and L368A, L368V, L368S or L368T.
  • a CH3 region comprising a hole modification comprises 407V, 366S and 368A.
  • the hole modification is or comprises Y407V, T366S, and L368A.
  • the antigen-binding molecule of the present disclosure comprise CH3 region(s) (e.g. within an Fc region, e.g. within CH2-CH3 region(s) of an Fc region) comprising modification for the formation of an interchain disulfide bond (i.e. between polypeptides comprising CH2-CH3 regions forming the Fc region).
  • Such modification may comprise the introduction of one or more cysteine residues into one or both of the CH3 regions of the constituent polypeptides of a polypeptide complex of the present disclosure. More particularly, such modification may have the result that the CH3:CH3 interface formed between the CH3 regions of polypeptides of polypeptide complexes of the present disclosure comprises a disulfide bond, formed between cysteine residues (one from each polypeptide).
  • one of the CH3 regions comprises 349C
  • the other CH3 region comprises 354C.
  • one of the CH3 regions comprises Y349C
  • the other CH3 region comprises S354C.
  • a CH3 region comprising a knob modification comprises 366W and S354C.
  • a CH3 region comprising a hole modification comprises Y407V, T366S, L368A and Y349C.
  • Further antigen-binding moiety In some embodiments, the antigen-binding molecule comprises a further antigen-binding moiety, sometimes described as a third antigen-binding moiety. In some embodiments, the further antigen-binding moiety binds to a target antigen other than a ⁇ c- containing cytokine receptor polypeptide (e.g. a target antigen which is not ⁇ c, IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ or IL-7R ⁇ ).
  • a target antigen which is not ⁇ c, IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ or IL-7R ⁇ .
  • the antigen-binding molecule of the present disclosure comprises (i) a ⁇ c-binding moiety, (ii) a moiety that binds to IL-7R ⁇ ), and (iii) a moiety that binds to a target antigen (e.g. an antigen that is not a ⁇ c-containing cytokine receptor polypeptide).
  • a target antigen e.g. an antigen that is not a ⁇ c-containing cytokine receptor polypeptide.
  • Mewburn ref.008537086 48 It will be appreciated that an effect of moiety (iii) is to localise the antigen-binding molecule to cells expressing its target. This can be useful to direct the effect of moieties (i) and (ii) of the antigen-binding molecule to cells expressing the target for moiety (iii).
  • moiety (ii) is an IL-7R ⁇ -binding moiety and wherein moiety (iii) is a CD8-binding moiety
  • the effect of moiety (iii) is to target the ⁇ c:IL-7R ⁇ receptor agonist/antagonist activity conferred by moieties (i) and (ii) to CD8+ T cells.
  • Moiety (iii) can also be employed to target the antigen-binding molecule to an anatomical site/tissue/organ of interest. This can be useful to direct the effect of moieties (i) and (ii) of the antigen-binding molecule to such regions.
  • moiety (ii) is an IL-7R ⁇ -binding moiety and wherein moiety (iii) is a cancer cell antigen-binding moiety
  • the effect of moiety (iii) is to target the ⁇ c:IL- 7R ⁇ receptor agonist/antagonist activity conferred by moieties (i) and (ii) to ⁇ c:IL-7R ⁇ receptor-expressing cells in the proximity of the cells expressing the cancer cell antigen.
  • moiety (iii) is employed to target/localise the antigen-binding molecule to, and/or increase the local concentration of the antigen-binding molecule in the proximity of, a cell comprising/expressing the target antigen for moiety (iii).
  • the target for moiety (iii) may be any target antigen.
  • the target antigen may be a peptide/polypeptide, glycoprotein, lipoprotein, glycan, glycolipid, lipid, or fragment thereof.
  • the antigen is preferably expressed at the cell surface of a cell expressing the antigen.
  • the target antigen is a disease-associated antigen or an antigen expressed by an immune cell.
  • a ‘disease-associated antigen’ refers to an antigen whose presence is indicative of a given disease/disease state, or an antigen for which an elevated level of the antigen is positively-correlated with a given disease/disease state.
  • the disease-associated antigen may be an antigen whose expression is associated with the development, progression or severity of symptoms of a given disease.
  • the disease- associated antigen may be associated with the cause or pathology of the disease, or may be expressed abnormally as a consequence of the disease.
  • a disease-associated antigen may be an antigen of an infectious agent or pathogen, a cancer-associated antigen or an autoimmune disease-associated antigen.
  • disease-associated antigens examples include CD49a, CD161, KLRG1, CRTH2, CD69, CCR6, CXCR5, NKG2D, CXCR4, CXCL12, CXCR3, CCR9, CD1c, NKp46, CD103, CD14, CD16, CD4, CD8, HLA-DR/DP/DQ or CD19.
  • the disease-associated antigen is an antigen of a pathogen.
  • the pathogen may be prokaryotic (bacteria), eukaryotic (e.g. protozoan, helminth, fungus), virus or prion.
  • the pathogen is an intracellular pathogen.
  • the pathogen is a virus, e.g. a virus as described hereinabove. In some embodiments the pathogen is a bacterium.
  • the target antigen is a cancer-associated antigen.
  • a cancer-associated antigen is an antigen whose expression or overexpression is associated with cancer.
  • the cancer-associated antigen is a receptor molecule, e.g. a cell surface receptor.
  • the cancer-associated antigen is a cell signalling molecule, e.g. a cytokine, chemokine, interferon, interleukin or lymphokine.
  • the cancer-associated antigen is a growth factor or a hormone.
  • the cancer-associated antigen is a viral antigen.
  • a cancer cell antigen may be abnormally expressed by a cancer cell (e.g. the cancer cell antigen may be expressed with abnormal localisation), or may be expressed with an abnormal structure by a cancer cell.
  • a cancer cell antigen may be capable of eliciting an immune response.
  • the antigen is expressed at the cell surface of the cancer cell (i.e. the cancer cell antigen is a cancer cell surface antigen).
  • the part of the antigen which is bound by the antigen-binding molecule described herein is displayed on the external surface of the cancer cell (i.e. is extracellular).
  • the cancer cell antigen may be a cancer-associated antigen.
  • the cancer cell antigen is an antigen whose expression is associated with the development, progression or severity of symptoms of a cancer.
  • the cancer- associated antigen may be associated with the cause or pathology of the cancer, or may be expressed abnormally as a consequence of the cancer.
  • the cancer cell antigen is an antigen whose expression is upregulated (e.g. at the RNA and/or protein level) by cells of a cancer, e.g. as compared to the level of expression by comparable non-cancerous cells (e.g. non-cancerous cells derived from the same tissue/cell type).
  • the cancer-associated antigen may be preferentially expressed by cancerous cells, and not expressed by comparable non-cancerous cells (e.g.
  • the cancer- associated antigen may be the product of a mutated oncogene or mutated tumor suppressor gene.
  • the cancer-associated antigen may be the product of an overexpressed cellular protein, a cancer antigen produced by an oncogenic virus, an oncofetal antigen, or a cell surface glycolipid or glycoprotein. Cancer-associated antigens are reviewed by Zarour HM, DeLeo A, Finn OJ, et al. Categories of Tumor Antigens. In: Kufe DW, Pollock RE, Weichselbaum RR, et al., editors. Holland-Frei Cancer Medicine.6th edition.
  • Cancer-associated antigens include oncofetal antigens: CEA, Immature laminin receptor, TAG-72; oncoviral antigens such as HPV E6 and E7; overexpressed proteins: BING-4, calcium-activated chloride channel 2, cyclin-B1, 9D7, Ep-CAM, EphA3, HER2/neu, telomerase, mesothelin, SAP-1, survivin; cancer-testis antigens: BAGE, CAGE, GAGE, MAGE, SAGE, XAGE, CT9, CT10, NY-ESO-1, PRAME, SSX-2; lineage restricted antigens: MART1, Gp100, tyrosinase, TRP-1/2, MC1R, prostate specific antigen; mutated antigens: ⁇ -catenin, BRCA1/2, CDK4, CML66, Fibronectin, MART-2, p53, Ras, TGF- ⁇ RII; post-translation
  • cancer cell antigens include heat-shock protein 70 (HSP70), heat-shock protein 90 (HSP90), glucose-regulated protein 78 (GRP78), vimentin, nucleolin, feto-acinar pancreatic protein (FAPP), alkaline Mewburn ref.008537086 50 phosphatase placental-like 2 (ALPPL-2), siglec-5, stress-induced phosphoprotein 1 (STIP1), protein tyrosine kinase 7 (PTK7), and cyclophilin B.
  • the cancer cell antigen is a cancer cell antigen described in Zhao and Cao, Front Immunol. (2019) 10:2250, which is hereby incorporated by reference in its entirety.
  • the target antigen is an immune cell surface molecule.
  • An immune cell surface molecule is any molecule which is expressed in or at the cell membrane of an immune cell.
  • the part of the immune cell surface molecule which is bound by the antigen-binding moiety is on the external surface of the immune cell (i.e. is extracellular).
  • the immune cell surface molecule may be expressed at the cell surface of any immune cell.
  • the immune cell may be a cell of hematopoietic origin, e.g. a neutrophil, eosinophil, basophil, dendritic cell, lymphocyte, or monocyte.
  • the lymphocyte may be e.g.
  • the immune cell may express a CD3 polypeptide (e.g. CD3 ⁇ CD3 ⁇ CD3 ⁇ or CD3 ⁇ ), a TCR polypeptide (TCR ⁇ or TCR ⁇ ), CD27, CD28, CD4 or CD8.
  • the immune cell is a T cell, e.g. a CD3+ T cell.
  • the T cell is a CD3+, CD4+ T cell.
  • the T cell is a CD3+, CD8+ T cell.
  • the T cell is a T helper cell (TH cell).
  • the T cell is a cytotoxic T cell (e.g. a cytotoxic T lymphocyte (CTL)).
  • CTL cytotoxic T lymphocyte
  • the immune cell is a T cell or an NK cell.
  • the immune cell surface molecule may be a CD3-TCR complex polypeptide, e.g. TCR ⁇ , TCR ⁇ , TCR ⁇ , TCR ⁇ , TRAC, TRBC1, TRBC2, TRGC1, TRGC2, TRDC, CD3 ⁇ , CD3 ⁇ , CD3 ⁇ or CD3 ⁇ .
  • the immune cell surface molecule is CD3, CD8, CD4 or CD28.
  • the immune cell surface molecule is a checkpoint molecule (e.g. PD-1, CTLA-4, LAG-3, TIM-3, VISTA, TIGIT or BTLA), or a ligand thereof.
  • the immune cell surface molecule is a costimulatory molecule (e.g. CD28, OX40, 4-1BB, ICOS or CD27), or a ligand thereof.
  • the target antigen is selected from PD-1, 4-1BB and CD8.
  • Chimeric antigen receptors The present disclosure also provides Chimeric Antigen Receptors (CARs). CARs are recombinant receptors that provide both antigen-binding and T cell activating functions.
  • CARs comprise an antigen-binding region linked to a cell membrane anchor region and a signalling region.
  • An optional hinge region may provide separation between the antigen- binding region and cell membrane anchor region, and may act as a flexible linker.
  • the antigen-binding domain of a CAR according to the present disclosure comprises or consists of an antigen-binding molecule as described herein. Accordingly, a CAR according to the present disclosure comprises an antigen-binding molecule as described herein.
  • an antigen-binding molecule according to the present disclosure forms, or is comprised in, the antigen-binding domain of the CAR. Accordingly, in some embodiments, the antigen- binding molecule of the present disclosure is comprised in a CAR. It will also be appreciated that an antigen-binding molecule according to the present disclosure may be a CAR. A CAR having an antigen-binding domain comprising or consisting of an antigen-binding molecule of the present disclosure is an antigen-binding molecule.
  • the antigen-binding domain of the CAR of the present disclosure may be provided with any suitable format, e.g. scFv, scFab, etc.
  • the cell membrane anchor region is provided between the antigen-binding region and the signalling region of the CAR and provides for anchoring the CAR to the cell membrane of a cell expressing a CAR, with the antigen-binding region in the extracellular space, and signalling region inside the cell.
  • the CAR comprises a cell membrane anchor region comprising or consisting of an amino acid sequence which comprises, consists of, or is derived from, the transmembrane region amino acid sequence for one of CD3- ⁇ , CD4, CD8 or CD28.
  • a region which is ‘derived from’ a reference amino acid sequence comprises an amino acid sequence having at least 60%, e.g.
  • the signalling region of a CAR allows for activation of the T cell.
  • the CAR signalling regions may comprise the amino acid sequence of the intracellular domain of CD3- ⁇ , which provides immunoreceptor tyrosine-based activation motifs (ITAMs) for phosphorylation and activation of the CAR-expressing T cell.
  • ITAMs immunoreceptor tyrosine-based activation motifs
  • Signalling regions comprising sequences of other ITAM-containing proteins such as Fc ⁇ RI have also been employed in CARs (Haynes et al., J Immunol. (2001) 166(1):182-187).
  • the CAR of the present disclosure comprises one or more co-stimulatory sequences comprising or consisting of an amino acid sequence which comprises, consists of, or is derived from, the amino acid sequence of the intracellular domain of one or more of CD28, OX40, 4-1BB, ICOS and CD27.
  • An optional hinge region may provide separation between the antigen-binding domain and the transmembrane domain, and may act as a flexible linker. Hinge regions may be derived from IgG1.
  • an antigen-binding molecule described herein may possess one or more of the following properties: binds to ⁇ c; binds to a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c; binds to IL-7R ⁇ ; binds to ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c; binds to ⁇ c and IL-7R ⁇ ; binds to ⁇ c-expressing cells; binds to cells expressing a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c; binds to IL-7R ⁇ -expressing cells; binds to cells expressing ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c; binds to cells expressing ⁇ c and IL-7R ⁇ ; binds to cells expressing a receptor comprising ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor
  • IL-2R ⁇ selected from IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ and IL-7R ⁇ ); inhibits signalling mediated by IL-7R ⁇ ; inhibits signalling mediated by IL-4R ⁇ ; inhibits signalling mediated by IL-21R ⁇ ; Mewburn ref.008537086 53 inhibits signalling mediated by IL-7R ⁇ and signalling mediated by one or more of IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , and/or IL-21R ⁇ ; inhibits IL-2, IL-4, IL-7, IL-9, IL-15, and/or IL-21 mediated signalling; inhibits IL-7 mediated signalling; inhibits IL-4 mediated signalling; inhibits IL-21 mediated signalling; inhibits IL-7 mediated signalling and inhibits IL-2, IL-4, IL-9, IL-15, and/or IL
  • IL-2R ⁇ increases multimerization of ⁇ c and IL-7R ⁇ ; decreases multimerization of ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c (e.g.
  • IL-2R ⁇ selected from IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ and IL-7R ⁇
  • decreases multimerization of ⁇ c and IL-7R ⁇ increases signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); increases signalling mediated by a ⁇ c:IL-7R ⁇ receptor; decreases signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); decreases signalling mediated by a ⁇ c:IL-7R ⁇ receptor; increases proliferation, survival and/or effector activity of cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ Mewburn ref.008537086 54 receptor ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL- 21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • increases proliferation, survival and/or effector activity of cells expressing IL-7R ⁇ decreases proliferation, survival and/or effector activity of cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL- 21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); decreases proliferation, survival and/or effector activity of cells expressing IL-7R ⁇ ; reduces expression of one or more markers of immune cell exhaustion by cells expressing a ⁇ c- containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); increases expression of one or more markers of immune cell exhaustion by cells expressing a ⁇ c- containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); increases expression of one or more markers of immune cell exhaustion by cells expressing IL- 7R ⁇ ; decreases multimerization of ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c (e.g.
  • IL-2R ⁇ decreases signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds
  • ⁇ c and/or a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c (e.g. selected from IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ , and IL-7R ⁇ )); increases cell killing/depletion of, and/or reduces the number/proportion of, cells comprising/expressing ⁇ c, and/or IL-7R ⁇ ; increased stability and/or half-life compared to one or more ⁇ c family cytokines (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • decreased upregulation of signalling mediated by a ⁇ c:IL-7R ⁇ receptor compared to IL-7 decreased upregulation of proliferation, survival and/or effector activity of cells expressing a ⁇ c- containing cytokine receptor to which the antigen-binding molecule binds
  • an antigen-binding molecule/moiety described herein binds to ⁇ c and/or IL-7R ⁇ with sub- nanomolar affinity, i.e. KD ⁇ 1 x 10 -9 M.
  • an antigen-binding molecule/moiety described herein binds to ⁇ c and/or IL-7R ⁇ with an affinity in the picomolar range, i.e.
  • the IL-7R ⁇ -binding moiety binds an epitope comprising an amino acid sequence having at least 70%, e.g. one of ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 91%, ⁇ 92%, ⁇ 93%, ⁇ 94%, ⁇ 95%, ⁇ 96%, ⁇ 97%, ⁇ 98%, ⁇ 99% or 100% amino acid sequence identity to SEQ ID NO:374.
  • the IL-7R ⁇ -binding moiety binds an epitope comprising an amino acid sequence having at least 70%, e.g.
  • the mutimerization of ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c is heteromultimerization, as the constituent polypeptides of the multimer are non-identical.
  • Mewburn ref.008537086 60 the multimers formed by mutimerization of ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c in accordance with the present disclosure are heteromultimers, rather than homomultimers.
  • the antigen-binding molecules of the present disclosure may promote mutimerization of ⁇ c and IL-7R ⁇ through binding to the respective polypeptides, through its constituent antigen-binding moieties.
  • antigen binding molecules may also be referred to as ‘antagonists’ of, or having ‘antagonistic’ or ‘inhibitory’ activity with respect to, the relevant ⁇ c-containing cytokine receptor.
  • antigen binding molecule binds and sequesters ⁇ c, this reduces the availability of ⁇ c to form a heteromeric complex with a polypeptide of the ⁇ c receptor family (e.g., IL-2R ⁇ , IL-2R ⁇ , IL-4R ⁇ , IL-7R ⁇ , IL-9R ⁇ , IL-15R ⁇ , or IL-21R ⁇ ). Therefore, the disclosed antigen binding molecules may affect the signalling of all ⁇ c-associated cytokine receptors, not just IL-7R ⁇ . As a result, non-specific agonism is prevented.
  • the antigen-binding molecule inhibits signalling mediated by ⁇ c:IL-7R ⁇ and ⁇ c:IL-2R ⁇ :IL-2R ⁇ . In some embodiments, the antigen-binding molecule inhibits signalling mediated by ⁇ c:IL-7R ⁇ and ⁇ c:IL-2R ⁇ :IL-15R ⁇ . In some embodiments, the antigen-binding molecule inhibits signalling mediated by ⁇ c:IL-7R ⁇ and ⁇ c:IL-4R ⁇ . In some embodiments, the antigen-binding molecule inhibits signalling mediated by ⁇ c:IL-7R ⁇ and ⁇ c:IL-9R ⁇ .
  • the antigen-binding molecule inhibits IL-7 signalling and IL-21 signalling. Mewburn ref.008537086 65 In some embodiments, the antigen-binding molecule inhibits IL-7 mediated signalling. In some embodiments, the antigen-binding molecule inhibits IL-2 mediated signalling. In some embodiments, the antigen-binding molecule inhibits IL-4 mediated signalling. In some embodiments, the antigen-binding molecule inhibits IL-9 mediated signalling. In some embodiments, the antigen-binding molecule inhibits IL-15 mediated signalling. In some embodiments, the antigen-binding molecule inhibits IL-21 mediated signalling.
  • Cytokine signalling can be analysed using cells expressing the relevant cytokine receptor, for example the relevant cytokine receptor for assaying IL-7 signalling would be the ⁇ c:IL-7R ⁇ receptor.
  • Suitable assays include e.g. assays for detecting the phosphorylation/activity/expression of factors which are phosphorylated/activated/expressed as a consequence of signalling through the ⁇ c-containing cytokine receptor.
  • cytokine signalling can be investigated by evaluating phosphorylation of one or more signal transduction molecules of a signal transduction pathway triggered by signalling through the relevant ⁇ c- containing cytokine receptor (e.g. the JAK/STAT, MAPK/ERK or PI3K/Akt pathways).
  • an ‘increased’ level of signalling/proliferation/survival/effector activity refers to a level of signalling/proliferation/survival/effector activity which is greater than 1 times, e.g. one of ⁇ 1.01 times, ⁇ 1.02 times, ⁇ 1.03 times, ⁇ 1.04 times, ⁇ 1.05 times, ⁇ 1.1 times, ⁇ 1.2 times, ⁇ 1.3 times, ⁇ 1.4 times, ⁇ 1.5 times, ⁇ 1.6 times, ⁇ 1.7 times, ⁇ 1.8 times, ⁇ 1.9 times, ⁇ 2 times, ⁇ 3 times, ⁇ 4 times, ⁇ 5 times, ⁇ 6 times, ⁇ 7 times, ⁇ 8 times, ⁇ 9 times or ⁇ 10 times the level of signalling/proliferation/survival/effector activity observed in the absence of the antigen-binding molecule, or in the presence of an appropriate control antigen-binding molecule (e.g.
  • Markers of immune cell exhaustion include e.g. immune checkpoint molecules (e.g. PD-1, CTLA-4, LAG-3, TIM-3, VISTA, TIGIT and BTLA), CD160 and CD244.
  • the cell surface expression of one or more markers of immune cell exhaustion may be evaluated, e.g. by flow cytometry.
  • a ‘reduction’ or ‘increase’ in the level of expression of one or more markers of immune cell exhaustion is relative to the level displayed (i.e. by cells of the same type) in the absence of the antigen- binding molecule, or in the presence of an appropriate control antigen-binding molecule (e.g.
  • an appropriate control antigen-binding molecule e.g. an antigen-binding molecule known not to influence expression of one or more markers of immune cell exhaustion.
  • the antigen-binding molecule of the present disclosure is able to decrease multimerization of ⁇ c and IL-7R ⁇ , decrease signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g. ⁇ c:IL-7R ⁇ receptor), and/or decrease proliferation, survival and/or effector activity of cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g. ⁇ c:IL-7R ⁇ receptor) by a mechanism not requiring or involving Fc-mediated function (i.e. independently of Fc-mediated function).
  • the antigen-binding molecule is able to achieve one or more of the effects recited in the preceding sentence in an Fc region- independent manner.
  • Mewburn ref.008537086 69 The ability of an antigen-binding molecule to decrease multimerization of ⁇ c and IL-7R ⁇ , decrease signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g. ⁇ c:IL-7R ⁇ receptor), and/or decrease proliferation, survival and/or effector activity of cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-7R ⁇ receptor by a mechanism not requiring/involving Fc-mediated function can be evaluated e.g. by analysing the ability of the antigen-binding molecule provided in a format lacking a functional Fc region to achieve one or more of the specified effects.
  • the relevant functional property(/ies) can be investigated using an antigen-binding molecule comprising a ‘silent’ Fc region (e.g. comprising L234A, L235A and P329G substitutions), or using an antigen-binding molecule provided in a format lacking an Fc region (e.g. scFv, Fab, etc.).
  • the antigen-binding molecule decreases multimerization of ⁇ c and IL-7R ⁇ , decreases signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g. ⁇ c:IL-7R ⁇ receptor, and/or decreases proliferation, survival and/or effector activity of cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g. ⁇ c:IL-7R ⁇ receptor) by a mechanism not involving ADCC, ADCP and/or CDC.
  • the antigen-binding molecule decreases multimerization of ⁇ c and IL-7R ⁇ , decreases signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g. ⁇ c:IL-7R ⁇ receptor), and/or decreases proliferation, survival and/or effector activity of cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g. ⁇ c:IL-7R ⁇ receptor) by a mechanism not requiring binding of the antigen-binding molecule to an Fc receptor (e.g. not requiring binding of the antigen-binding molecule to an Fc ⁇ receptor; e.g.
  • the antigen-binding molecule of the present disclosure does not induce ADCC, ADCP or CDC of cells comprising/expressing one or more of the target antigens of its constituent antigen- binding moieties (i.e. ⁇ c, and/or IL-7R ⁇ ). Antigen-binding molecules which do not induce (i.e.
  • ADCC/ADCP/CDC elicit substantially no ADCC/ADCP/CDC activity against the relevant cell type, e.g. as determined by analysis in an appropriate assay for the relevant activity.
  • substantially no ADCC/ADCP/CDC activity refers to a level of ADCC/ADCP/CDC that is not significantly greater than ADCC/ADCP/CDC determined for an appropriate negative control molecule in a given assay (e.g. an antigen-binding molecule lacking an Fc region, or an antigen-binding molecule comprising a ‘silent’ Fc region (e.g.
  • ‘Substantially no activity’ may be a level of the relevant activity which is ⁇ 5 Mewburn ref.008537086 70 times, e.g. ⁇ 4 times, ⁇ 3 times, ⁇ 2.5 times, ⁇ 2 times or ⁇ 1.5 times the level of activity determined for an appropriate negative control molecule in a given assay.
  • the ability of, and extent to which, a given antigen-binding molecule is able to induce ADCC of a given target cell type can be analysed e.g.
  • an antigen-binding molecule according to the present disclosure may increase (i.e. upregulate, enhance, potentiate) cell killing of cells comprising/expressing one or more of the target antigens of its constituent antigen-binding moieties (i.e. ⁇ c, and/or IL-7R ⁇ ).
  • an ‘increased’ level of cell killing refers to a level of cell killing which is greater than 1 times, e.g.
  • an appropriate control antigen-binding molecule e.g. an antigen-binding molecule known not to influence killing of such cells.
  • an antigen-binding molecule according to the present disclosure is capable of reducing the number/proportion of cells comprising/expressing one or more of the target antigens of its constituent antigen-binding moieties (i.e. ⁇ c, and/or IL-7R ⁇ ).
  • the antigen-binding molecule is capable of depleting/enhancing depletion of such cells.
  • an ‘reduced’ number/proportion of cells refers to a number/proportion of cells which is less than 1 times, e.g. one of e.g.
  • Antigen-binding molecules according to the present disclosure may comprise one or more moieties for potentiating a reduction in the number/proportion of cells comprising/expressing one or more of the target antigens of its constituent antigen-binding moieties (i.e. ⁇ c, and/or a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c (e.g. selected from IL-7R ⁇ ).
  • an antigen-binding molecule according to the present disclosure may e.g. comprise an Fc region and/or a drug moiety.
  • an antigen-binding molecule according to the present disclosure comprises an Fc region capable of potentiating/directing one or more of ADCC, ADCP, CDC against, and/or potentiating formation of a MAC on or cell degranulation of, a cell comprising/expressing one or more of the target antigens of its constituent antigen-binding moieties (i.e. ⁇ c, and/or IL-7R ⁇ ).
  • an antigen-binding molecule according to the present disclosure comprises a drug moiety. The antigen-binding molecule may be conjugated to the drug moiety. Antibody-drug conjugates are reviewed e.g.
  • the drug moiety is or comprises a cytotoxic agent, such that the antigen-binding molecule displays cytotoxicity to a cell comprising/expressing one or more of the target antigens of its constituent antigen-binding moieties (i.e. ⁇ c, and/or IL-7R ⁇ ).
  • the drug moiety is or comprises a chemotherapeutic agent.
  • an antigen-binding molecule according to the present disclosure comprises an immune cell-engaging moiety.
  • the antigen-binding molecule comprises a CD3 polypeptide-binding moiety (e.g.
  • the antigen-binding molecule of the present disclosure is more effective at increasing signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g. ⁇ c:IL-7R ⁇ receptor) as compared to a cytokine that binds to the ⁇ c-containing cytokine receptor.
  • the antigen-binding molecule of the present disclosure is less effective at increasing signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g. ⁇ c:IL-7R ⁇ receptor) as compared to a cytokine that binds to the ⁇ c-containing cytokine receptor.
  • the antigen-binding molecule of the present disclosure is more effective at increasing upregulation of proliferation, survival and/or effector activity of cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g. ⁇ c:IL-7R ⁇ receptor) compared to a cytokine that binds to the ⁇ c-containing cytokine receptor.
  • the antigen-binding molecule of the present disclosure is less effective at increasing upregulation of proliferation, survival Mewburn ref.008537086 73 and/or effector activity of cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • the antigen-binding molecule of the present disclosure is more effective at reducing the expression of one or more markers of immune cell exhaustion by cells expressing a ⁇ c- containing cytokine receptor to which the antigen-binding molecule binds (e.g. ⁇ c:IL-7R ⁇ receptor) compared to a cytokine that binds to the ⁇ c-containing cytokine receptor.
  • the antigen-binding molecule increases signalling mediated by ⁇ c:IL-7R ⁇ receptor and/or increases cell proliferation, survival and/or effector activity of cells expressing a ⁇ c:IL-7R ⁇ receptor with an EC50 which is greater than 1 times, e.g.
  • the antigen-binding molecule reduces Mewburn ref.008537086 74 expression of one or more markers of immune cell exhaustion by cells expressing a ⁇ c:IL-7R ⁇ receptor with an IC50 which is greater than 1 times, e.g.
  • administration of the antigen-binding molecule is associated with a reduction in the number of cancer cells (e.g. a reduction in cancer burden) in vivo, e.g. as compared to an appropriate control condition. In some embodiments, administration of the antigen-binding molecule is associated with an increase in the killing of cancer cells in vivo, e.g. as compared to an appropriate control condition. In some embodiments, administration of the antigen-binding molecule is associated with a reduction in pathogen load in vivo, e.g. as compared to an appropriate control condition.
  • administration of the antigen-binding molecule is associated with a reduction in the number of cells of a pathogen and/or the number of cells infected with a pathogen in vivo, e.g. as compared to an appropriate control condition.
  • administration of an antigen-binding molecule according to the present disclosure may be associated with one or more of the following, as compared to an appropriate control condition: inhibition of the development/progression of the cancer, a delay to/prevention of onset of the cancer, a reduction in/delay to/prevention of tumor growth, a reduction in/delay to/prevention of tissue invasion, a reduction in/delay to/prevention of metastasis, a reduction in the severity of one or more symptoms of the cancer, a reduction in the number of cancer cells, a reduction in the cancer burden, a reduction in tumour size/volume, and/or an increase in survival of subjects having the cancer (e.g. progression free survival or overall survival), e.g. as determined in an appropriate model.
  • inhibition of the development/progression of the cancer e.g. progression free survival or overall survival
  • the linker comprises or consists of a GGGGS (SEQ ID NO:240) amino acid sequence. In some embodiments, the linker comprises or consists of a GGGGSGGGGSGGGGS (SEQ ID NO:141) amino acid sequence. In some embodiments, the linker comprises a GGGGSGGGGSGGGGSGGGGS (SEQ ID NO:239) amino acid sequence. In some embodiments, the linker comprises or consists of a NSGAAA (SEQ ID NO:461) amino acid sequence. In some embodiments, the linker between a VH sequence and a VL sequence comprises or consists of a GGGGS (SEQ ID NO:240) amino acid sequence.
  • the linker between a VH sequence and a VL sequence comprises or consists of a GGGGSGGGGSGGGGS (SEQ ID NO:141) amino acid sequence.
  • scFV comprises a GGGGSGGGGSGGGGS (SEQ ID NO:141) amino acid sequence.
  • the linker between a VH sequence and a VL sequence comprises or consists of a GGGGSGGGGSGGGGSGGGGS (SEQ ID NO:239) amino acid sequence.
  • scFV comprises a GGGGSGGGGSGGGGSGGGGS (SEQ ID NO:239) amino acid sequence.
  • the linker sequence between antigen-binding moieties comprises or consists of a flexible linker. In some embodiments, the linker sequence between antigen-binding moieties comprises or Mewburn ref.008537086 76 consists of a GGGGS (SEQ ID NO:240) amino acid sequence. In some embodiments, the linker sequence between scFv molecules comprises or consists of a GGGGS (SEQ ID NO:240) amino acid sequence. In some embodiments, the linker sequence between VHH molecules comprises or consists of a GGGGS (SEQ ID NO:240) amino acid sequence.
  • the linker sequence between VHH molecules comprises or consists of an A(EAAAK)5A (SEQ ID NO:451) amino acid sequence.
  • the linker sequence between an scFv molecule and a VHH molecule comprises or consists of an A(EAAAK) 5 A (SEQ ID NO:451) amino acid sequence.
  • the antigen-binding molecules and polypeptides of the present disclosure may comprise amino acid sequence(s) to facilitate expression, folding, trafficking, processing, purification or detection of the antigen-binding molecule/polypeptide.
  • antigen-binding molecules and polypeptides of the present disclosure may additionally comprise a sequence of amino acids forming a detectable moiety, e.g. as described hereinbelow.
  • the signal peptide may be present at the N-terminus of the antigen-binding molecule/polypeptide and may be present in the newly synthesised antigen-binding molecule/polypeptide.
  • the signal peptide provides for efficient trafficking of the antigen-binding molecule/polypeptide. Signal peptides are often removed by cleavage, and thus are not comprised in the mature antigen-binding molecule/polypeptide.
  • the antigen-binding molecule or polypeptide of the present disclosure comprises a detectable moiety.
  • GFP green fluorescent protein
  • Eu europium
  • Tb terbium
  • Sm samarium
  • tetramethyl rhodamine Texas Red
  • 4-methyl umbelliferone 7-amino-4-methyl coumarin
  • Cy3 Cy5
  • Radiolabels include radioisotopes such as Hydrogen 3 , Sulfur 35 , Carbon 14 , Phosphorus 32 , Iodine 123 , Iodine 125 , Iodine 126 , Iodine 131 , Iodine 133 , Bromine 77 , Technetium 99m , Indium 111 , Indium 113m , Gallium 67 , Gallium 68 , Ruthenium 95 , Ruthenium 97 , Ruthenium 103 , Ruthenium 105 , Mercury 207 , Mercury 203 , Rhenium 99m , Rhenium 101 , Rhenium 105 , Scandium 47 , Tellurium 121m , Tellurium 122m , Tellurium 125m , Thulium 165 , Thuliuml 167 , Thulium 168 , Copper 67 , Fluorine 18 , Yttrium 90 , Palladium 100 , Bismuth 217 and Anti
  • Luminescent labels include as radioluminescent, chemiluminescent (e.g. acridinium ester, luminol, isoluminol) and bioluminescent labels.
  • Immuno-detectable labels include haptens, peptides/polypeptides, antibodies, receptors and ligands such as biotin, avidin, streptavidin or digoxigenin.
  • Nucleic acid labels include aptamers.
  • the antigen-binding molecule/polypeptide comprises an epitope tag, e.g.
  • the vector may facilitate delivery of the nucleic acid(s) encoding a polypeptide according to the present disclosure to a cell.
  • the vector may be an expression vector comprising elements required for expressing a polypeptide according to the present disclosure.
  • the vector may comprise elements facilitating integration of the nucleic acid(s) into the genomic DNA of the cell into which the vector is introduced.
  • Nucleic acids and vectors according to the present disclosure may be provided in purified or isolated form, i.e. from other nucleic acid, or naturally-occurring biological material. Mewburn ref.008537086 79
  • a vector may be a vector for expression of the nucleic acid in the cell (i.e. an expression vector).
  • production of the antigen-binding molecule may comprise transcription Mewburn ref.008537086 81 and translation of more than one polypeptide, and subsequent association of the polypeptide chains to form the antigen-binding molecule.
  • any cell suitable for the expression of polypeptides may be used.
  • the cell may be a prokaryote or eukaryote.
  • the cell is a prokaryotic cell, such as a cell of archaea or bacteria.
  • the bacteria may be Gram-negative bacteria such as bacteria of the family Enterobacteriaceae, for example Escherichia coli.
  • polypeptide(s) of interest may be desirable to isolate the polypeptide(s) of interest from the supernatant or culture medium, which may contain other protein and non-protein components.
  • a common approach to separating protein components from a supernatant or culture medium is by precipitation. Proteins of different solubilities are precipitated at different concentrations of precipitating agent such as ammonium sulfate. For example, at low concentrations of precipitating agent, water soluble proteins are extracted. Thus, by adding different increasing concentrations of precipitating agent, proteins of different solubilities may be distinguished. Dialysis may be subsequently used to remove ammonium sulfate from the separated proteins. Other methods for distinguishing different proteins are known in the art, for example ion exchange chromatography and size chromatography.
  • the methods may prevent development of the disease/condition a later stage (e.g. a chronic stage or metastasis).
  • Treatment may, for example, be reduction in the development or progression of a disease/condition, alleviation of the symptoms of a disease/condition or reduction in the pathology of a disease/condition.
  • Treatment or alleviation of a disease/condition may be effective to prevent progression of the disease/condition, e.g. to prevent worsening of the condition or to slow the rate of development.
  • treatment or alleviation may lead to an improvement in the disease/condition, e.g.
  • the antigen-binding molecules of the present disclosure find use in the treatment/prevention of diseases/conditions that would derive therapeutic or prophylactic benefit from an increase in the level of signalling mediated by the ⁇ c- containing cytokine receptor to which the antigen-binding molecule binds.
  • the antigen-binding molecule comprises (i) a ⁇ c-binding moiety and (ii) an IL-7R ⁇ - binding moiety
  • the antigen-binding molecule finds use in the treatment/prevention of diseases/conditions that would derive therapeutic or prophylactic benefit from an increase in signalling mediated by the ⁇ c:IL- 7R ⁇ receptor.
  • the antigen-binding molecules of the present disclosure find use in the treatment/prevention of diseases/conditions that would derive therapeutic or prophylactic benefit from a decrease in the level of signalling mediated by the ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • IL-2 IL-2R ⁇ receptor
  • IL-15R ⁇ IL-4R ⁇ receptor
  • ⁇ c:IL-9R ⁇ receptor ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • the articles of the present disclosure find use in the treatment/prevention of diseases/conditions that would derive therapeutic or prophylactic benefit from an decrease in the proliferation and/or population expansion of, and decrease in the survival of and/or an decrease in the number/proportion and/or activity of cells expressing a ⁇ c-containing cytokine receptor (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • a decreased number/proportion/level of activity of cells expressing a ⁇ c-containing cytokine receptor e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • a decreased number/proportion/level of activity of lymphocytes e.g., lymphocytes, a decreased number/proportion/level of Mewburn ref.008537086 88 activity of effector immune cells, a decreased number/proportion/level of activity of T cells (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • a decreased number/proportion/level of activity of lymphocytes a decreased number/proportion/level of activity of effector immune cells
  • T cells e.g. effector T cells
  • ISCs an increased number/proportion/level of activity of ISCs
  • B cells an increased number/proportion/level of activity of B cells
  • NK cells a decreased number/proportion/level of activity of NK cells.
  • the disease/condition to be treated/prevented in accordance with the present disclosure may be a disease/condition in which one or more of the following is positively-associated with the onset, development or progression of the disease/condition, and/or severity of one or more symptoms of the disease/condition, or is a risk factor for the onset, development or progression of the disease/condition: an increased level of signalling mediated by a ⁇ c family cytokine (e.g. selected from IL-2, IL-15, IL-4, IL-9, IL-21 and IL-7), an increased level of signalling mediated by ⁇ c-containing cytokine receptor (e.g.
  • a ⁇ c family cytokine e.g. selected from IL-2, IL-15, IL-4, IL-9, IL-21 and IL-7
  • ⁇ c-containing cytokine receptor e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • an increased number/proportion/level of activity of cells expressing a ⁇ c-containing cytokine receptor e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL- 2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • an increased number/proportion/level of activity of lymphocytes an increased number/proportion/level of activity of effector immune cells, an increased number/proportion/level of activity of T cells (e.g. effector T cells), and/or an increased number/proportion/level of activity of NK cells.
  • the disease/condition to be treated/prevented in accordance with the present disclosure is a disease/condition characterised by one or more of the following: an increased level of signalling mediated by a ⁇ c family cytokine (e.g. selected from IL-2, IL-15, IL-4, IL-9, IL-21 and IL- 7), an increased level of signalling mediated by ⁇ c-containing cytokine receptor (e.g.
  • ⁇ c:IL- 2R ⁇ receptor selected from ⁇ c:IL- 2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • an increased number/proportion/level of activity of cells expressing a ⁇ c-containing cytokine receptor e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL- 15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • an increased number/proportion/level of activity of lymphocytes an increased number/proportion/level of activity of Mewburn ref.008537086 89 effector immune cells
  • T cells e.g. effector T cells
  • NK cells an increased number/proportion/level of activity of NK cells.
  • the ‘decrease’/’increase’ may be relative to the level observed in the healthy, non-diseased state, e.g. as determined in a healthy control subject, and/or in equivalent non- diseased tissue.
  • methods are provided which are for, or which comprise (e.g. in the context of treatment/prevention of a disease/condition described herein), one or more of the following: increasing multimerization of ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c (e.g.
  • IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ and IL-7R ⁇ decreasing multimerization of ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c (e.g. selected from IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ and IL-7R ⁇ ); increasing signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); decreasing signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); increasing proliferation, survival and/or effector activity of cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL- 21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); decreasing proliferation, survival and/or effector activity of cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL- 21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); reducing expression of one or more markers of immune cell exhaustion by cells expressing a ⁇ c- containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); increasing expression of one or more markers of immune cell exhaustion by cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); increasing cell killing/depletion of, and/or reducing the number/proportion of, cells comprising/expressing one or more of the target antigens of its constituent antigen-binding moieties (i.e. ⁇ c, and/or a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c (e.g.
  • agents according to the present disclosure for use in such methods, and the use of agents according to the present disclosure in the manufacture of pharmaceutical compositions or medicaments for use in such methods. It will be appreciated that the methods may comprise administering an antigen-binding molecule, nucleic acid, expression vector, cell or composition described herein to a subject. Similarly, one or more of the following may be observed in a subject following therapeutic or prophylactic intervention in accordance with the present disclosure (e.g.
  • an increased level of multimerization of ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c e.g. selected from IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ and IL-7R ⁇
  • a decreased level of multimerization of ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c e.g.
  • IL-2R ⁇ selected from IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ and IL-7R ⁇
  • an increased level of signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL- 2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • an decreased level of signalling mediated by a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL- 2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor
  • an increased level of proliferation, survival and/or effector activity of cells expressing a ⁇ c- containing cytokine receptor to which the antigen-binding molecule binds e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); an increased level of expression of one or more markers of immune cell exhaustion by cells expressing a ⁇ c-containing cytokine receptor to which the antigen-binding molecule binds (e.g.
  • Mewburn ref.008537086 91 increased cell killing/depletion of, and/or a reduced number/proportion of, cells comprising/expressing one or more of the target antigens of its constituent antigen-binding moieties (i.e. ⁇ c, and/or a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c (e.g.
  • ⁇ c:IL-2R ⁇ receptor selected from ⁇ c:IL-2R ⁇ receptor, ⁇ c:IL-2R ⁇ :IL-2R ⁇ , ⁇ c:IL-2R ⁇ :IL-15R ⁇ , ⁇ c:IL-4R ⁇ receptor, ⁇ c:IL-9R ⁇ receptor, ⁇ c:IL-21R ⁇ receptor and ⁇ c:IL-7R ⁇ receptor); inhibiting proliferation, survival and/or effector activity of cells expressing IL-2R ⁇ , IL-2R ⁇ , IL- 15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ and/or IL-7R ⁇ ; and/or increasing expression of one or more markers of immune cell exhaustion by cells expressing a ⁇ c-containing cytokine receptor (e.g.
  • the disease/condition to be treated/prevented in accordance with the present disclosure may be lymphocytopenia, or a disease/condition characterised by lymphocytopenia.
  • Lymphocytopenia may be defined as a total lymphocyte count of ⁇ 1000/mcL (1 x 10 9 /L) in adults or ⁇ 3000/mcL ( ⁇ 3 x 10 9 /L) in children ⁇ 2 years.
  • the disease/condition to be treated/prevented in accordance with the present disclosure is a disease/condition characterised by T cell dysfunction, a cancer, infection, or an autoimmune disease/disorder. In some embodiments, the disease/condition to be treated/prevented in accordance with the present disclosure is an autoimmune disease.
  • the autoimmune disease is Crohn’s disease, Sjögren’s syndrome, lupus, systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), multiple sclerosis (MS), alopecia areata, psoriasis, psoriatic arthritis, myasthenia gravis, sarcoidosis, type 1 diabetes, ulcerative colitis, Addison’s disease, Graves’ disease, Hashimoto’s thyroiditis, Autoimmune vasculitis, Pernicious anemia, and/or Celiac disease.
  • IL-7 is a critical survival factor for lymphocytes and recent studies suggest that targeting the IL-7/IL-7R ⁇ pathway holds promise for the treatment of autoimmune diseases.
  • IL-9-producing cells contribute to a group of autoimmune disorders including systemic lupus erythematosus (SLE), multiple sclerosis (MS), inflammatory bowel diseases (IBD), rheumatoid arthritis (RA) and psoriasis; IL-9 inhibitors may potentially be used for the clinical treatment of allergic diseases, autoimmune diseases or cancers (Deng et al. Hum Immunol. (2017) 78(2):120-128). Mewburn ref.008537086 94 IL-15 plays important roles in innate and adaptative immune responses, and it has been implicated in the pathology of various autoimmune diseases such as rheumatoid arthritis, autoimmune diabetes, inflammatory bowel disease, coeliac disease and psoriasis.
  • SLE systemic lupus erythematosus
  • MS multiple sclerosis
  • IBD inflammatory bowel diseases
  • RA rheumatoid arthritis
  • psoriasis psoriasis
  • Pathological inflammation may refer to inflammation which is implicated in (i.e. which positively contributes to) the pathology of a disease.
  • Inflammation to be treated/prevented in accordance with the present disclosure can be of any tissue/organ of the body.
  • the inflammation is of the lung (e.g. bronchioles, alveoli), airways (e.g.
  • inflammation may be of an organ or tissue of the gastrointestinal system, e.g. of the liver, bowel, small intestine, large intestine, colon, or pancreas. In some embodiments, inflammation may be of the eye. In some embodiments, inflammation may be of the skin. In some embodiments, inflammation may be of an organ or tissue of the nervous system, e.g. the brain. In some embodiments, inflammation may be of the bone marrow. In some embodiments, inflammation may be of the joints. In some embodiments, inflammation may be of an organ or tissue of the urinary system, e.g. the kidneys.
  • the cancer to be treated may be colon cancer, colon carcinoma, colorectal cancer, nasopharyngeal carcinoma, cervical carcinoma, oropharyngeal carcinoma, gastric carcinoma, hepatocellular carcinoma, head and neck cancer, head and neck squamous cell carcinoma (HNSCC), oral cancer, laryngeal cancer, prostate cancer, lung cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, urothelial carcinoma, melanoma, advanced melanoma, renal cell carcinoma, ovarian cancer or mesothelioma.
  • a cancer according to the present disclosure is selected from: gastric cancer (e.g.
  • gastric carcinoma gastric adenocarcinoma, gastrointestinal adenocarcinoma
  • head and neck cancer e.g. head and neck squamous cell carcinoma
  • breast cancer ovarian cancer (e.g. ovarian carcinoma)
  • lung cancer e.g. NSCLC, lung adenocarcinoma, squamous lung cell carcinoma
  • melanoma prostate cancer
  • oral cavity cancer e.g. oropharyngeal cancer
  • renal cancer e.g. renal cell carcinoma
  • colorectal cancer e.g. colorectal carcinoma
  • oesophageal cancer pancreatic cancer, a solid cancer and a liquid cancer (i.e. a hematological cancer).
  • the bacterial infection may be sepsis or tuberculosis.
  • viral infections that may be treated include infection by influenza virus, measles virus, hepatitis B virus (HBV), hepatitis C virus (HCV), human immunodeficiency virus (HIV), lymphocytic choriomeningitis virus (LCMV), Herpes simplex virus or human papilloma virus (HPV).
  • the method may additionally comprise administering to a subject a therapeutically or prophylactically effective amount of an antigen-binding molecule according to the present disclosure.
  • the disease/condition to be treated/prevented in accordance with the present disclosure may be a disease characterised by an increased number/proportion and/or activity of cells expressing a ⁇ c-containing cytokine receptor (e.g.
  • the disease/condition to be treated/prevented in accordance with the present disclosure may be lymphocytosis, or a disease/condition characterised by lymphocytosis. Lymphocytosis may be defined as a total lymphocyte count of > 3000/mcL ( 1 x 10 9 /L) in adults or > 9000/mcL ( ⁇ 3 x 10 9 /L) in children ⁇ 2 years.
  • chronic lymphocytic leukemia acute lymphocytic leukemia, acute lymphoblastic leukemia
  • lymphoma infectious disease
  • EBV infection infectious mononucleosis hepatitis (e.g. hepatitis A, hepatitis B, hepatitis C), CMV infection, HIV/AIDS, syphilis, pertussis, toxoplasmosis, Chagas disease, tuberculosis, brucellosis, hypothyroidism, autoimmune disease and rheumatoid arthritis.
  • the drug may be formulated as a pharmaceutical composition or medicament.
  • the formulation may comprise one or more drugs (e.g. one or more active agents) together with one or more pharmaceutically acceptable diluents, excipients or carriers.
  • Chemotherapy may involve administration of more than one drug.
  • a drug may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated.
  • the chemotherapy may be administered by one or more routes of administration, e.g. parenteral, intravenous injection, oral, subcutaneous, intradermal or intratumoral.
  • the chemotherapy may be administered according to a treatment regime.
  • the antigen-binding molecules and antigen-binding moieties of the present disclosure may inhibit the formation of a complex containing ⁇ c (e.g. a heteromeric complex of ⁇ c and another polypeptide).
  • a complex containing ⁇ c e.g. a heteromeric complex of ⁇ c and another polypeptide.
  • Signalling mediated by a ⁇ c-containing cytokine receptor only occurs if ⁇ c has formed a complex with another component of the ⁇ c-containing cytokine receptor (e.g. IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ and IL-7R ⁇ ).
  • IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ and IL-7R ⁇ are heteromeric complexes.
  • the method may further comprise comparing the determined amount against a standard or reference value as part of the diagnostic or prognostic evaluation.
  • Other diagnostic/prognostic tests may be used in conjunction with those described herein to enhance the accuracy of the diagnosis or prognosis or to confirm a result obtained by using the tests described herein.
  • a sample may be taken from any tissue or bodily fluid.
  • the sample may comprise or may be derived from: a quantity of blood; a quantity of serum derived from the individual’s blood which may comprise the fluid portion of the blood obtained after removal of the fibrin clot and blood cells; a tissue sample or biopsy; pleural fluid; cerebrospinal fluid (CSF); or cells isolated from said individual.
  • CSF cerebrospinal fluid
  • an amino acid sequence, or a region of a polypeptide which ‘corresponds’ to a specified reference amino acid sequence or region of a polypeptide has at least 60%, e.g. one of at least 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of the amino acid sequence/polypeptide/region.
  • An amino acid sequence/region/position of a polypeptide/amino acid sequence which ‘corresponds’ to a specified reference amino acid sequence/region/position of a polypeptide/amino acid sequence can be identified by sequence alignment of the subject sequence to the reference sequence, e.g.
  • An antigen-binding molecule optionally isolated, comprising: (i) a ⁇ c-binding moiety, and (ii) a moiety that binds to a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c.
  • Para 2a The antigen-binding molecule according to para 1a, wherein the polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c is selected from: IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ , and IL-7R ⁇ .
  • Para 3a The antigen-binding molecule according to para 1a, wherein the polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c is selected from: IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ , and IL-7R ⁇ .
  • the antigen-binding molecule according to para 1a wherein the polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c is not IL-2R ⁇ .
  • Para 4a The antigen-binding molecule according to any one of paras 1a to 3a, wherein the antigen-binding molecule is an agonist of an ⁇ c-containing cytokine receptor, or an antagonist of an ⁇ c-containing cytokine receptor.
  • Para 5a The antigen-binding molecule according to para 1a, wherein the polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c is not IL-2R ⁇ .
  • Para 9a An expression vector, or a plurality of expression vectors, comprising a nucleic acid or a plurality of nucleic acids according to para 8a.
  • Para 10a A cell comprising an antigen-binding molecule according to any one of paras 1a to 6a, a CAR according to para 7a, a nucleic acid or a plurality of nucleic acids according to para 8a, or an expression vector or a plurality of expression vectors according to para 9a. Mewburn ref.008537086 151 Para 11a.
  • a method comprising culturing a cell according to para 9a under conditions suitable for expression of an antigen-binding molecule or CAR by the cell.
  • Para 12a A composition comprising an antigen-binding molecule according to any one of paras 1a to 6a, a CAR according to para 7a, a nucleic acid or a plurality of nucleic acids according to para 8a, an expression vector or a plurality of expression vectors according to para 9a, or a cell according to para 10a, and a pharmaceutically acceptable carrier, diluent, excipient or adjuvant.
  • Para 13a A composition comprising an antigen-binding molecule according to any one of paras 1a to 6a, a CAR according to para 7a, a nucleic acid or a plurality of nucleic acids according to para 8a, an expression vector or a plurality of expression vectors according to para 9a, or a cell according to para 10a, and a pharmaceutically acceptable carrier, diluent, ex
  • An antigen-binding molecule according to any one of paras 1a to 6a, a CAR according to para 7a, a nucleic acid or a plurality of nucleic acids according to para 8a, an expression vector or a plurality of expression vectors according to para 9a, a cell according to para 10a, or a composition according to para 12a, for use in a method of treatment or prophylaxis.
  • Para 14a An antigen-binding molecule according to any one of paras 1a to 6a, a CAR according to para 7a, a nucleic acid or a plurality of nucleic acids according to para 8a, an expression vector or a plurality of expression vectors according to para 9a, a cell according to para 10a, or a composition according to para 12a, for use in a method of treatment or prophylaxis.
  • an antigen-binding molecule according to any one of paras 1a to 6a, a CAR according to para 7a, a nucleic acid or a plurality of nucleic acids according to para 8a, an expression vector or a plurality of expression vectors according to para 9a, a cell according to para 10a, or a composition according to para 12a, in the manufacture of a medicament for use in a method of treatment or prophylaxis.
  • Para 15a Use of an antigen-binding molecule according to any one of paras 1a to 6a, a CAR according to para 7a, a nucleic acid or a plurality of nucleic acids according to para 8a, an expression vector or a plurality of expression vectors according to para 9a, a cell according to para 10a, or a composition according to para 12a, in the manufacture of a medicament for use in a method of treatment or prophylaxis.
  • Para 15a Use of an antigen-binding molecule according to any one of paras 1
  • Para 17a The antigen-binding molecule, nucleic acid or plurality thereof, expression vector or plurality thereof, cell, or composition for use according to para 13a, the use according to para 14a or the method according to para 15a, wherein the method of treatment or prophylaxis is a method of treating or preventing a disease/condition characterised by T cell dysfunction, a cancer, an infectious disease, or an autoimmune disease. Para 17a.
  • the cancer is selected from the group consisting of: colon cancer, colon carcinoma, colorectal cancer, nasopharyngeal carcinoma, cervical carcinoma, oropharyngeal carcinoma, gastric carcinoma, hepatocellular carcinoma, head and neck cancer, head and
  • Para 22a A method for decreasing the proliferation, survival and/or effector activity of a cell expressing a ⁇ c-containing cytokine receptor, comprising contacting a cell expressing a ⁇ c-containing cytokine receptor in vitro, in vivo or ex vivo with an antigen-binding molecule according to any one of paras 1a to 6a.
  • Para 23a The method according to any one of paras 19a to 22a, wherein the cell is an effector immune cell.
  • Para 24a The method according to any one of paras 19a to 23a, wherein the cell is a T cell or a NK cell.
  • Para 25a A method for decreasing the proliferation, survival and/or effector activity of a cell expressing a ⁇ c-containing cytokine receptor, comprising contacting a cell expressing a ⁇ c-containing cytokine receptor in vitro, in vivo or ex vivo with an antigen-binding molecule according to any one of paras 1a to 6a.
  • a method of promoting heteromultimerization of ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor complex comprising contacting ⁇ c and a polypeptide of a ⁇ c-containing cytokine receptor complex in vitro, in vivo or ex vivo with an antigen-binding molecule according to any one of paras 1a to 6a, or a CAR according to para 7a.
  • Mewburn ref.008537086 153 Numbered paragraphs – Part B The following numbered paragraphs (paras.) contain statements of broad combinations of the inventive technical features herein disclosed: Para 1b.
  • An antigen-binding molecule optionally isolated, comprising: (i) a ⁇ c-binding moiety, and (ii) an IL-7R ⁇ -binding moiety, wherein the antigen-binding molecule is an inhibitor of signalling mediated by a ⁇ c- containing cytokine receptor.
  • Para 2b The antigen-binding molecule according to para 1b, wherein the ⁇ c-containing cytokine receptor comprises IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ , or IL-7R ⁇ .
  • Para 3b The antigen-binding molecule according to para 1b, wherein the ⁇ c-containing cytokine receptor comprises IL-2R ⁇ , IL-2R ⁇ , IL-15R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-21R ⁇ , or IL-7R ⁇ .
  • antigen-binding molecule according to para 1b or para 2b, wherein the ⁇ c-binding moiety comprises a heavy chain variable (VH) region and a light chain variable (VL) region of an antibody that binds to ⁇ c, optionally wherein antigen-binding moiety comprises an Fv and/or a Fab of an antibody that binds to ⁇ c.
  • VH heavy chain variable
  • VL light chain variable
  • antigen-binding moiety comprises an Fv and/or a Fab of an antibody that binds to ⁇ c.
  • the antigen-binding molecule according to para 5b wherein the target antigen other than a ⁇ c and IL-7R ⁇ is a disease-associated antigen, or an antigen expressed by an immune cell.
  • Para 7b A chimeric antigen receptor (CAR), comprising an antigen-binding molecule according to any one of paras 1b to 6b.
  • Para 8b A nucleic acid, or a plurality of nucleic acids, optionally isolated, encoding an antigen- binding molecule according to any one of paras 1b to 6b, or a CAR according to para 7b.
  • Para 9b An expression vector, or a plurality of expression vectors, comprising a nucleic acid or a plurality of nucleic acids according to para 8b.
  • Para 10b A cell comprising an antigen-binding molecule according to any one of paras 1b to 6b, a CAR according to para 7b, a nucleic acid or a plurality of nucleic acids according to para 8b, or an expression vector or a plurality of expression vectors according to para 9b.
  • Para 11b A method comprising culturing a cell according to para 10b under conditions suitable for expression of an antigen-binding molecule or CAR by the cell.
  • Para 12b A method comprising culturing a cell according to para 10b under conditions suitable for expression of an antigen-binding molecule or CAR by the cell.
  • An antigen-binding molecule according to any one of paras 1b to 6b, a CAR according to para 7b, a nucleic acid or a plurality of nucleic acids according to para 8b, an expression vector or a plurality of expression vectors according to para 9b, a cell according to para 10b, or a composition according to para 12b, for use in a method of treatment or prophylaxis.
  • Para 14b An antigen-binding molecule according to any one of paras 1b to 6b, a CAR according to para 7b, a nucleic acid or a plurality of nucleic acids according to para 8b, an expression vector or a plurality of expression vectors according to para 9b, a cell according to para 10b, or a composition according to para 12b, for use in a method of treatment or prophylaxis.
  • an antigen-binding molecule according to any one of paras 1b to 6b, a CAR according to para 7b, a nucleic acid or a plurality of nucleic acids according to para 8b, an expression vector or a plurality of expression vectors according to para 9b, a cell according to para 10b, or a composition according to para 12b, in the manufacture of a medicament for use in a method of treatment or prophylaxis.
  • a method of treatment or prophylaxis comprising administering to a subject in need thereof a therapeutically or prophylactically effective amount of an antigen-binding molecule according to any one of paras 1b to 6b, a CAR according to para 7b, a nucleic acid or a plurality of nucleic acids according to para 8b, an expression vector or a plurality of expression vectors according to para 9b, a cell according to para 10b, or a composition according to para 12b.
  • Para 16b is a therapeutically or prophylactically effective amount of an antigen-binding molecule according to any one of paras 1b to 6b, a CAR according to para 7b, a nucleic acid or a plurality of nucleic acids according to para 8b, an expression vector or a plurality of expression vectors according to para 9b, a cell according to para 10b, or a composition according to para 12b.
  • the autoimmune disease is selected from Crohn’s disease, Sjögren’s syndrome, lupus, systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, alopecia areata,
  • Para 19b The antigen-binding molecule, nucleic acid or plurality thereof, expression vector or plurality thereof, cell, or composition for use, the use, or the method according to any one of paras 13b to 18b, wherein ⁇ c is sequestered by the antigen binding molecule.
  • Para 20b The antigen-binding molecule, nucleic acid or plurality thereof, expression vector or plurality thereof, cell, or composition for use, the use, or the method according to any one of paras 13b to para 19b, wherein the frequency of ⁇ c forming a complex with IL-2R ⁇ , IL-2R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-15R ⁇ , and/or IL-21R ⁇ is inhibited.
  • Mewburn ref.008537086 156 Para 21b.
  • Para 22b An in vitro complex, optionally isolated, comprising an antigen-binding molecule according to any one of paras 1b to 6b, a CAR according to para 7b, bound to ⁇ c and IL-7R ⁇ .
  • a method comprising contacting ⁇ c in vitro, in vivo or ex vivo with an antigen-binding molecule, optionally isolated, comprising: (i) a ⁇ c-binding moiety, and (ii) an IL-7R ⁇ -binding moiety.
  • Para 24b A method of sequestering ⁇ c, comprising contacting ⁇ c in vitro, in vivo or ex vivo with an antigen-binding molecule, optionally isolated, comprising: (i) a ⁇ c-binding moiety, and (ii) an IL-7R ⁇ - binding moiety.
  • a method of inhibiting the formation of a heteromeric complex of ⁇ c and IL-2R ⁇ , IL-2R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-15R ⁇ , and/or IL-21R ⁇ comprising contacting ⁇ c in vitro, in vivo or ex vivo with an antigen-binding molecule, optionally isolated, comprising: (i) a ⁇ c-binding moiety, and (ii) an IL-7R ⁇ - binding moiety.
  • para 26b or para 27b wherein the polypeptide of the ⁇ c receptor family is ⁇ c, IL-2R ⁇ , IL-2R ⁇ , IL-4R ⁇ , IL-7R ⁇ , IL-9R ⁇ , IL-15R ⁇ , or IL-21R ⁇ .
  • Para 29b The method according to any one of paras 23b to 28b, wherein the ⁇ c and/or the polypeptide of a ⁇ c-containing cytokine receptor is expressed by an effector immune cell.
  • Para 30b The method according to para 29b, wherein the effector immune cell is an innate lymphoid cell (ILC), T cell, B cell, or NK cell. Mewburn ref.008537086 157 Para 31b.
  • ILC innate lymphoid cell
  • any one of paras 23b to 30b wherein the antigen-binding molecule is the antigen-binding molecule according to any one of paras 1b to 6b, or a cell comprising a CAR according to para 7b.
  • Para 32b The method according to any one of paras 23b to 31b, wherein the frequency of ⁇ c forming a complex with IL-2R ⁇ , IL-2R ⁇ , IL-4R ⁇ , IL-9R ⁇ , IL-15R ⁇ , and/or IL-21R ⁇ is inhibited.
  • Para 33b The method according to any one of paras 23b to 30b, wherein the antigen-binding molecule is the antigen-binding molecule according to any one of paras 1b to 6b, or a cell comprising a CAR according to para 7b.
  • Example 1 Production and characterisation of bispecific ⁇ c- and CD122- binding antibodies in WO 2017/021540 A1, WO 2019/092181 A1, WO 2020/094834 A1 and WO 2020/094836 A1
  • the production and characterisation of bispecific, ⁇ c- and CD122- binding antibody agonists of the intermediate-affinity ⁇ c/IL-2R ⁇ receptor is described e.g. in WO 2017/021540 A1, WO 2019/092181 A1, WO 2020/094834 A1 and WO 2020/094836 A1, which are hereby incorporated by reference in their entirety.
  • Example 3 and Figure 21A demonstrate binding of bispecific antibodies comprising a ⁇ c-binding arm and/or a CD122-binding arm to cells expressing ⁇ c or CD122, as determined by flow cytometric analysis.
  • Example 4 and Figure 20 show that bispecific molecules comprising various different combinations of ⁇ c- binding and CD122-binding arms bind to recombinant ⁇ c or CD122, and Table 1 of Example 1 summarises the affinity of binding to the respective proteins by the bispecific antibodies, as determined by Surface Plasmon Resonance analysis.
  • Example 6.1 and Figure 23 show that a bispecific anti- ⁇ c, anti- CD122 antibody triggered signalling in IL-2 receptor-expressing cells (as determined by analysis of STAT5 phosphorylation) with a different activation profile as compared to IL-2, particularly activating signalling in NK and CD8+ T cells to a greater extent as proportion of activation of signalling in Tregs.
  • Example 6.2 and Figure 24 show that a bispecific anti- ⁇ c, anti-CD122 antibody promoted proliferation of NK92 cells in vitro, and that bispecific molecules comprising one of the arms and an irrelevant second arm did not stimulate such cell proliferation.
  • Example 6.2 and Figures 25B and 25C demonstrate that bispecific molecules comprising a ⁇ c-binding arm and a CD122-binding arm provided in scFv-Fc(KiH)-scFv or tandem scFv-scFv format, and provided with linkers of various different lengths (i.e. between the scFv moiety and the constant regions in the KiH format molecules, and between the two scFv moieties in the tandem scFv-scFv format molecules) induce in vitro proliferation of NK92 cells with similar efficiency.
  • linkers of various different lengths i.e. between the scFv moiety and the constant regions in the KiH format molecules, and between the two scFv moieties in the tandem scFv-scFv format molecules
  • Example 1 of WO 2019/092181 A1 discloses the production of bispecific anti- ⁇ c, anti-CD122 antibodies, in various different formats (particularly scFv-FcKiH-scFv format, CrossMab format and Duobody format).
  • Example 2.1 and Figures 1A and 1B report the ability of bispecific anti- ⁇ c, anti-CD122 antibodies comprising different ⁇ c-binding and a CD122-binding clones and provided in different antibody formats to bind to recombinant ⁇ c or CD122, as determined by ELISA.
  • Figures 8A to 8G show that the bispecific anti- ⁇ c, anti-CD122 antibodies dose-dependent proliferation of non-specifically activated T cells, with more pronounced expansion of CD8+ T cells than CD4+ T cells.
  • the bispecific anti- ⁇ c, anti-CD122 antibodies did not induce significant proliferation of Tregs, and they yielded expanded populations with higher ratios of CD8+ T cells to Tregs as compared to those expanded with IL-2 or IL-15.
  • Figure 8H shows that pre-expanded human Tregs were not expanded by treatment with the bispecific anti- ⁇ c, anti-CD122 antibodies, whereas they expanded in a dose-dependent fashion in response to treatment with IL-2 or IL-15.
  • Example 3.3 and Figures 9A to 9I similarly disclose preferential expansion of CD8+ T cells over Tregs, and CD4+ T cell proliferation following treatment of non-specifically activated primary human PBMCs treated with the bispecific anti- ⁇ c, anti-CD122 antibodies.
  • Example 3.4 and Figures 10A to 10G demonstrate that bispecific anti- ⁇ c, anti-CD122 antibodies stimulated in vitro proliferation of antigen-specific (particularly EBV-specific) CD4+ and CD8+ T cells in a dose-dependent manner, and to also induce proliferation of NK cells within the virus-specific T cell population.
  • Example 3.5 and Figures 11A to 11K show that a bispecific anti- ⁇ c, anti-CD122 antibody stimulated dose- dependent proliferation of non-specifically activated (i.e.
  • mice administered bispecific anti- ⁇ c, anti-CD122 antibodies again had a lower organ tumor burden compared to mice administered IL-2 or isotype control antibody, and higher numbers of CD8+ T cells of mice administered bispecific anti- ⁇ c, anti-CD122 antibodies were shown to be expressing effector molecules (IFN ⁇ , CD107a, perforin) relative to CD8+ T cells from mice administered IL-2 or isotype control antibody.
  • effector molecules IFN ⁇ , CD107a, perforin
  • mice were injected subcutaneously (SC) into flanks of NSG mice to establish a highly aggressive and Mewburn ref.008537086 164 metastatic disease that closely mimics EBV-driven lymphoid malignancies in humans, with dissemination to the spleen, liver and tertiary lymph nodes (typically absent in NSG mice).
  • Mice were started on the specified treatment regimen once the growth at the injection side became palpable (typically 6-8mm).
  • T cells were produced by in vitro stimulation of PBMCs from the same EBV-seropositive donor as above, with irradiated EBV-BLCLs. Tregs were expanded according to the protocol described in Section 3.4 to ensure sufficient numbers for injection.
  • EBV-BCL tumor- engrafted mice were treated with EBV specific T cells and Treg cells in the presence of IL-2 or agonist bispecific ⁇ c- and IL-2R ⁇ - binding antibodies (Adk-1 or Adk-2).
  • IL-2 preferentially stimulates Tregs and hence prevents elimination of tumor.
  • agonist bispecific ⁇ c- and CD122- binding antibodies do not stimulate Tregs and yield clearance of tumors in vivo.
  • 2.3 NHP T-cell proliferation in vivo Cynomolgus macaques were injected by i.v. with a single dose of agonist bispecific ⁇ c- and IL-2R ⁇ - binding antibodies at 5 mg/kg through the femoral artery.
  • Bispecific anti- ⁇ c, anti- IL-7R ⁇ antibodies Bispecific antibodies with a ⁇ c-binding moiety and an IL-7R ⁇ -binding moiety are produced. The binding characteristics of bispecific antibodies comprising a ⁇ c-binding arm and a IL-7R ⁇ -binding arm to cells expressing ⁇ c or IL-7R ⁇ , are determined.
  • Bispecific antibodies comprising a ⁇ c-binding arm and a IL-7R ⁇ -binding arm are shown to sequester ⁇ c, to inhibit signalling mediated by a ⁇ c-containing cytokine receptor, to inhibit heteromeric ⁇ c complex formation, and shown to inhibit the proliferation, survival and/or effector activity of a cell expressing a polypeptide of a ⁇ c-containing cytokine receptor.
  • Example 4 Cell line and assay development To assay functional properties of antibodies comprising a ⁇ c-binding moiety, and a moiety that binds to a polypeptide of a ⁇ c-containing cytokine receptor other than ⁇ c, suitable cell lines are required.
  • HEK 293 Cytokine Reporter Cells are designed to provide a simple, rapid, and reliable method to monitor the activation of signaling pathways induced by key cytokines. Cytokine reporter cells enable the detection of these biologically active cytokines and can also be used to screen for compounds exhibiting agonist and antagonist activities.
  • HEK-Blue IL-2R ⁇ Cells, HEK-Blue IL-7 Cells, HEK-Blue IL-9 Cells, and HEK-Blue IL-21 Cells are commercially available (InvivoGen, UK). These cytokine reporter cells are derived from the HEK293 cell line.
  • a HEK-Blue IL-15R ⁇ /IL-2R ⁇ line was generated for the first time to use in the examples of this specification. This HEK-Blue IL- 15R ⁇ /IL-2R ⁇ line was engineered to overexpress IL-15R ⁇ , IL-2R ⁇ , ⁇ c, JAK3 and STAT5. A HEK-Blue IL-4/IL-7 line was also generated.
  • This HEK-Blue IL-4/IL-7 line was engineered to overexpress IL-4R ⁇ , IL- 7R ⁇ , ⁇ c, JAK3 and STAT5.
  • a HEK-Blue IL-21R ⁇ /IL-7R ⁇ line was generated for the first time to use in the examples of this specification.
  • This HEK-Blue IL-21R ⁇ /IL-7R ⁇ line was engineered to overexpress IL- 21R ⁇ , IL-7R ⁇ , ⁇ c, JAK3 and STAT5.
  • the cell surface expression of cytokine receptor polypeptides on commercial and in-house produced cell lines was assayed to validate the cell lines before they were used in subsequent cell signalling assays.
  • HEK-Blue IL-2R ⁇ , HEK-Blue IL-7, HEK-Blue IL-9, HEK-Blue IL-21, HEK-Blue IL-15R ⁇ /IL-2R ⁇ , HEK-Blue IL-21R ⁇ /IL-7R ⁇ , and HEK-Blue IL-4/IL-7 cell lines enabled the completion of experimental assays based on the well-known QUANTI-Blue assay to quantify the level of STAT5 phosphorylation after treatment with the specified bispecific antibodies.
  • a general schematic overview of the cell signalling assays performed in Example 4 is provided in Figure 8. Further details are provided within the text of the Example 5.
  • Bispecific ⁇ c- and IL-7R ⁇ - binding antibodies Generation of bispecific ⁇ c- and IL-R ⁇ - binding antibodies
  • Bispecific antibodies comprising different combinations of ⁇ c-binding arm (TWNg, REGg, or SYNg), and IL-7R ⁇ -binding arm (OSE7R, PFZ7R, GSK7R, or NIH7R1) were generated, using different linkers.
  • ⁇ c-binding arms TWNg comprises the VH and VL sequences of anti- ⁇ c clone P1A3-AQ described herein.
  • REGg comprises the VH and VL sequences of anti- ⁇ c antibody REGN7257.
  • bispecific ⁇ c- and IL-7R ⁇ - binding antibodies demonstrated agonist activity, with treatment resulting in upregulation of STAT5 phosphorylation ( Figures 9A-D). These data demonstrate that bispecific ⁇ c- and IL-7R ⁇ - binding antibodies are capable of upregulating signalling mediated by a ⁇ c:IL-7R ⁇ receptor.
  • Antagonist assay HEK-Blue IL-4/IL-7 Cells were cultured, harvested, washed, and then cultured with Doxycycline (100ng/ml) for 24 hours to induce IL-4Ra expression.

Landscapes

  • Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Transplantation (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention concerne des molécules de liaison à l'antigène comprenant une fraction de liaison à γc, et une fraction qui se lie à IL-7Rα. L'invention concerne également des compositions comprenant de telles molécules de liaison à l'antigène, ainsi que des utilisations et des méthodes y ayant recours.
PCT/EP2024/050235 2023-01-06 2024-01-05 Molécules de liaison à l'antigène WO2024146955A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202363437470P 2023-01-06 2023-01-06
US63/437,470 2023-01-06
US202363462365P 2023-04-27 2023-04-27
US63/462,365 2023-04-27

Publications (1)

Publication Number Publication Date
WO2024146955A1 true WO2024146955A1 (fr) 2024-07-11

Family

ID=89620314

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2024/050235 WO2024146955A1 (fr) 2023-01-06 2024-01-05 Molécules de liaison à l'antigène

Country Status (1)

Country Link
WO (1) WO2024146955A1 (fr)

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7695936B2 (en) 1995-03-01 2010-04-13 Genentech, Inc. Knobs and holes heteromeric polypeptides
WO2011104687A1 (fr) 2010-02-24 2011-09-01 Rinat Neuroscience Corporation Anticorps récepteurs anti-il-7 antagonistes et procédés
US20120282254A1 (en) 2010-01-28 2012-11-08 Ian Kirby Cd127 binding proteins
WO2014131694A1 (fr) 2013-02-26 2014-09-04 Roche Glycart Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t
US20150044231A1 (en) 2011-06-06 2015-02-12 Novo Nordisk A/S Therapeutic Antibodies
WO2015189302A1 (fr) 2014-06-10 2015-12-17 Effimune Anticorps anti-cd127
WO2016059512A1 (fr) 2014-10-18 2016-04-21 Pfizer Inc. Compositions d'anticorps anti-il-7r
WO2017021540A1 (fr) 2015-08-06 2017-02-09 Agency For Science, Technology And Research Anticorps anti-il-2-r beta/chaîne gamma commune
WO2019092181A1 (fr) 2017-11-10 2019-05-16 Agency For Science, Technology And Research Anticorps anti-il-2-r bêta/chaîne gamma commune
US10392441B2 (en) 2015-10-07 2019-08-27 United States Of America, As Represented By The Secretary, Department Of Health And Human Services IL-7R-alpha specific antibodies for treating acute lymphoblastic leukemia
WO2020094834A1 (fr) 2018-11-09 2020-05-14 Euchloe Bio Pte. Ltd. Anticorps anti-il-2r bêta/chaîne gamma commune
WO2020094836A1 (fr) 2018-11-09 2020-05-14 Euchloe Bio Pte. Ltd. Anticorps anti-il-2-r bêta/chaîne gamma commune

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7695936B2 (en) 1995-03-01 2010-04-13 Genentech, Inc. Knobs and holes heteromeric polypeptides
US20120282254A1 (en) 2010-01-28 2012-11-08 Ian Kirby Cd127 binding proteins
US9150653B2 (en) 2010-01-28 2015-10-06 Glaxo Group Limited CD127 binding proteins
WO2011104687A1 (fr) 2010-02-24 2011-09-01 Rinat Neuroscience Corporation Anticorps récepteurs anti-il-7 antagonistes et procédés
US20150044231A1 (en) 2011-06-06 2015-02-12 Novo Nordisk A/S Therapeutic Antibodies
WO2014131694A1 (fr) 2013-02-26 2014-09-04 Roche Glycart Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t
WO2015189302A1 (fr) 2014-06-10 2015-12-17 Effimune Anticorps anti-cd127
WO2016059512A1 (fr) 2014-10-18 2016-04-21 Pfizer Inc. Compositions d'anticorps anti-il-7r
WO2017021540A1 (fr) 2015-08-06 2017-02-09 Agency For Science, Technology And Research Anticorps anti-il-2-r beta/chaîne gamma commune
EP3428193A1 (fr) * 2015-08-06 2019-01-16 Agency For Science, Technology And Research Anticorps anti-il-2-r beta/chaîne gamma commune
US10392441B2 (en) 2015-10-07 2019-08-27 United States Of America, As Represented By The Secretary, Department Of Health And Human Services IL-7R-alpha specific antibodies for treating acute lymphoblastic leukemia
WO2019092181A1 (fr) 2017-11-10 2019-05-16 Agency For Science, Technology And Research Anticorps anti-il-2-r bêta/chaîne gamma commune
WO2020094834A1 (fr) 2018-11-09 2020-05-14 Euchloe Bio Pte. Ltd. Anticorps anti-il-2r bêta/chaîne gamma commune
WO2020094836A1 (fr) 2018-11-09 2020-05-14 Euchloe Bio Pte. Ltd. Anticorps anti-il-2-r bêta/chaîne gamma commune

Non-Patent Citations (146)

* Cited by examiner, † Cited by third party
Title
"Uniprot", Database accession no. P16871-1
"UniProt", Database accession no. P31785-1
"UniProtKB", Database accession no. P01863-1
ADUNYAH ET AL., BIOCHEM BIOPHYS RES COMMUN, vol. 232, 1997, pages 754 - 8
AKKAPEDDI ET AL., LEUKEMIA, vol. 33, no. 9, 2019, pages 2155 - 2168
ALEGRE ET AL., J IMMUNOL, vol. 148, 1992, pages 3461 - 3468
ALLARD-CHAMARD ET AL., CYTOKINE, vol. 136, 2020, pages 155258
AN ET AL., MABS, vol. 1, 2009, pages 572 - 579
ARAI ET AL., PROTEIN ENGINEERING, DESIGN AND SELECTION, vol. 14, no. 8, 2001, pages 529 - 532
BANKAITIS ET AL., CLIN EXP METASTASIS, vol. 32, no. 8, 2015, pages 847 - 856
BELARIF ET AL., NAT. COMMUN., vol. 9, 2018, pages 4483
BELARIF ET AL., NATURE COMMUNICATIONS, vol. 9, 2018, pages 4483
BELARIF LYSSIA ET AL: "IL-7 receptor blockade blunts antigen-specific memory T cell responses and chronic inflammation in primates", NATURE COMMUNICATIONS, vol. 9, no. 1, 26 October 2018 (2018-10-26), UK, pages 1 - 13, XP093114550, ISSN: 2041-1723, Retrieved from the Internet <URL:https://www.nature.com/articles/s41467-018-06804-y> DOI: 10.1038/s41467-018-06804-y *
BOERSMA ET AL., J BIOL CHEM., vol. 286, 2011, pages 41273 - 85
BRINKMANNKONTERMANN, MABS, vol. 9, no. 2, 2017, pages 182 - 212
BRIUKHOVETSKA D ET AL., NAT REV CANCER, vol. 21, 2021, pages 481 - 499
BUCK ET AL., BIOTECHNIQUES, vol. 44, no. 7, 2008, pages 927 - 9
CARTER, J IMMUNOL METH., vol. 248, 2001, pages 7 - 15
CARTER, J IMMUNOL METHODS, vol. 248, 2001, pages 7 - 15
CERIGNOLI ET AL., PLOS ONE, vol. 13, no. 3, 2018, pages e0193498
CHAKRABORTY ET AL., INT J MOL SCI, vol. 20, no. 9, 2019, pages 2113
CHANDRUDU ET AL., MOLECULES, vol. 18, 2013, pages 4373 - 4388
CHEN ET AL., ADV DRUG DELIV REV., vol. 65, no. 10, 2013, pages 1357 - 1369
CHEN ET AL., ONCOTARGET, vol. 9, no. 6, 2018, pages 7204 - 7218
CHIA WK ET AL., MOLECULAR THERAPY, vol. 22, no. 1, 2014, pages 132 - 139
CHOI ET AL., MOL CANCER THER, vol. 12, no. 12, 2013, pages 2748 - 59
CHOTHIA ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CHU ET AL., MOL IMMUNOL, vol. 45, 2008, pages 3926 - 3933
CIRUELA, CURR OPIN BIOTECHNOL, vol. 19, no. 4, 2008, pages 338 - 43
COBBOLD ET AL., J EXP MED., vol. 202, 2005, pages 379 - 386
CONCEPCION ET AL., COMB CHEM HIGH THROUGHPUT SCREEN, vol. 12, no. 8, 2009, pages 791 - 800
CROCE, J IMMUNOL RES, vol. 2015, 2015, pages 696578
DAI ET AL., J NAT CANCER INST, vol. 108, no. 7, 2016, pages djv439
DALL'ACQUA ET AL., J IMMUNOL., vol. 169, 2002, pages 5171 - 5180
DAVIS ET AL., CANCER J, vol. 21, no. 6, 2015, pages 486 - 491
DAVIS ET AL., PROTEIN ENG DES SEL, vol. 23, no. 4, 2010, pages 195 - 202
DENG ET AL., CELL MOL IMMUNOL, vol. 12, no. 1, 2015, pages 31 - 39
DENG ET AL., HUM IMMUNOL, vol. 78, no. 2, 2017, pages 120 - 128
DIEBOLDER ET AL., SCIENCE, vol. 343, no. 6176, 2014, pages 1260 - 3
DIXON ET AL., ACS CHEM BIOL, vol. 11, no. 2, 2016, pages 400 - 408
DOOMS ET AL., J AUTOIMMUN, vol. 45, 2013, pages 40 - 8
DOTTI ET AL., IMMUNOL REV, vol. 257, no. 1, 2014, pages 107 - 126
EDELMAN ET AL., PROC NATL ACAD SCI USA, vol. 63, no. 1, 1969, pages 78 - 85
EDELMAN ET AL., PROC NATL ACAD SCI USA., vol. 63, no. 1, 1969, pages 78 - 85
EIKASSARGRESS, J IMMUNOTOXICOL, vol. 7, no. 1, 2010, pages 1 - 7
ELLIS ET AL., BR J CLIN PHARMACOL, vol. 85, no. 2, 2019, pages 304 - 315
ELLIS ET AL., BR J CLIN PHARMACOL, vol. 85, no. 2, February 2019 (2019-02-01), pages 304 - 315
ELLMAN ET AL., METH. ENZYM., vol. 202, 1991, pages 301 - 336
EMANUEL ET AL., MABS, vol. 3, 2011, pages 38 - 48
FLOCH-RAMONDOU ET AL., HEMASPHERE, vol. 6, 2022, pages 694 - 695
FRANKSIPPL, BIOINFORMATICS, vol. 24, 2008, pages 2172 - 2176
FRENCH, METHODS MOL MED, vol. 40, 2000, pages 333 - 339
FRENZEL ET AL., FRONT IMMUNOL, vol. 4, 2013, pages 217
FULCHERWONG, IMMUNOL CELL BIOL, vol. 77, no. 6, 1999, pages 559 - 564
GAO ET AL.: "Sec. Autoimmune and Autoinflammatory Disorders", FRONT IMMUNOL, 2022
GERSHONI ET AL., BIODRUGS, vol. 21, no. 3, 2007, pages 145 - 156
GHORESCHI ET AL., NAT MED, vol. 9, 2003, pages 40 - 6
GIRI ET AL., EMBO J, vol. 14, 1995, pages 3654 - 63
GREENSAMBROOK: "Molecular Cloning: A Laboratory Manual", 2012, COLD SPRING HARBOR PRESS
GU ET AL., MOL CELL BIOL, vol. 20, 2000, pages 7109 - 20
HE ET AL., PROC NATL ACAD SCI USA., vol. 92, no. 12, 1995, pages 5689 - 5693
HEROLD ET AL., JCI INSIGHT, vol. 4, no. 24, 19 December 2019 (2019-12-19), pages e126054
HEROLD ET AL., JCI INSIGHT, vol. 4, no. 24, 2019, pages e126054
HIXON ET AL., LEUKEMIA, vol. 34, no. 1, January 2020 (2020-01-01), pages 35 - 49
HORNIGFARBER-SCHWARZ, METHODS MOL BIOL, vol. 907, 2012, pages 411 - 442
IDUSOGIE ET AL., J IMMUNOL., vol. 166, no. 1, 2001, pages 2571 - 187
ISHII ET AL., INT IMMUNOL., vol. 6, no. 8, 1994, pages 1273 - 1277
JEDEMA ET AL., BLOOD, vol. 103, 2004, pages 2677 - 82
JEFFERIS ET AL., IMMUNOL REV, vol. 163, 1998, pages 59 - 76
JERABEK-WILLEMSEN ET AL., ASSAY DRUG DEVTECHNOL, vol. 9, no. 4, 2011, pages 342 - 353
JIANG ET AL., CYTOKINE GROWTH FACTOR REV, vol. 16, 2005, pages 513 - 533
JUNTTILA, FRONT IMMUNOL, vol. 9, 2018, pages 888
KALOSJUNE, IMMUNITY, vol. 39, no. 1, 2013, pages 49 - 60
KAMEN ET AL., J IMMUNOL, vol. 198, no. 157, 2017, pages 17
KATOHSTANDLEY, MOLECULAR BIOLOGY AND EVOLUTION, vol. 30, no. 4, 2013, pages 772 - 780
KEEGAN ET AL., FAC REV, vol. 10, 2021, pages 71
KHORYATI ET AL., SCI IMMUNOL, vol. 5, no. 50, 2020, pages eaba5264
KONTERMANN, MABS, vol. 4, no. 2, 2012, pages 182 - 197
KUNERTREINHART, APPL MICROBIOL BIOTECHNOL, vol. 100, 2016, pages 3451 - 3461
LABRIJN ET AL., PROC NATL ACAD SCI USA, vol. 110, no. 13, 2013, pages 5145 - 50
LAD ET AL., J BIOMOL SCREEN, vol. 20, no. 4, 2015, pages 498 - 507
LASSMANNSONNHAMMER, BMC BIOINFORMATICS, vol. 6, no. 298, 2005
LAZAR ET AL., PROC NATL ACAD SCI USA., vol. 103, 2006, pages 4005 - 4010
LE SAOUT ET AL., JCI INSIGHT, vol. 2, no. 22, 2017, pages e96228
LEAVER-FAY ET AL., STRUCTURE, vol. 24, no. 4, 2016, pages 641 - 51
LEE, PATHOLOGY & ONCOLOGY RESEARCH, vol. 26, 2020, pages 2017 - 2022
LEFRANC ET AL., DEV. COMP. IMMUNOL., vol. 27, 2003, pages 55 - 77
LEFRANC ET AL., NUCLEIC ACIDS RES, vol. 43, 2015, pages D413 - 22
LEONARD ET AL., IMMUNITY, vol. 50, no. 4, 2019, pages 832 - 850
LINLEONARD, COLD SPRING HARB PERSPECT BIOL, vol. 10, no. 9, 2018, pages a028449
LO ET AL., J. BIOL. CHEM, vol. 292, no. 9, 2017, pages 3900 - 3908
MA ET AL., FRONT IMMUNOL, vol. 12, 2021, pages 626616
MARY CAROLINE ET AL: "Abstract 2287: Bispecific anti-PD1 checkpoint inhibitors antibodies (BiCKI), an optimized platform designed to tackle anti-PD-(L)1 primary and secondary resistance mechanisms", CANCER RESEARCH, vol. 80, no. 16 Suppl, 27 April 2020 (2020-04-27), XP055839165, Retrieved from the Internet <URL:https://cancerres.aacrjournals.org/content/80/16_Supplement/2287> DOI: 10.1158/1538-7445.AM2020-2287 *
MAUS ET AL., ANNU REV IMMUNOL., vol. 32, 2014, pages 189 - 225
MISHRA ET AL., CLIN CANCER RES, vol. 20, no. 8, 2014, pages 2044 - 2050
MOORE ET AL., MABS, vol. 2, no. 2, 2010, pages 181 - 9
MOORE ET AL., MABS, vol. 3, no. 6, 2011, pages 546 - 57
MORGANBOYERINAS, BIOMEDICINES, vol. 4, no. 3, 2016, pages 14
MOUDGILCHOUBEY, J INTERFERON CYTOKINE RES, vol. 31, no. 10, 2011, pages 695 - 703
NAT METHODS, vol. 5, no. 2, 2008, pages 135 - 146
NATSUME ET AL., CANCER RES., vol. 68, no. 10, 2008, pages 3863 - 72
NEWMAN ET AL., CLIN. IMMUNOL., vol. 98, 2001, pages 164 - 174
NOTREDAME ET AL., J MOL BIOL., vol. 302, 2000, pages 205 - 217
PARSLOW ET AL., BIOMEDICINES, vol. 4, no. 3, September 2016 (2016-09-01), pages 14
PETERSEN ET AL., NATURE METHODS, vol. 8, 2011, pages 785 - 786
PULLIAM ET AL., IMMUNOL LETT, vol. 169, 2016, pages 61 - 72
RETTER ET AL., NUCL. ACIDS RES., vol. 33, 2005, pages D671 - D674
REVERDATTO ET AL., CURR TOP MED CHEM., vol. 15, no. 12, 2015, pages 1082 - 1101
RICH ET AL., ANAL BIOCHEM., vol. 373, no. 1, 2008, pages 112 - 20
RICHARDS ET AL., MOL CANCER THER, vol. 7, 2008, pages 2517 - 2527
ROTHER ET AL., NAT BIOTECHNOL., vol. 25, 2007, pages 1256 - 1264
SALIMITCHISON, PNAS USA, vol. 105, no. 7, 2008, pages 2415 - 2420
SCHLOTHAUER ET AL., PROTEIN ENGINEERING, DESIGN AND SELECTION, vol. 29, no. 10, 2016, pages 457 - 466
SHANG ET AL., BIOL CHEM, vol. 289, 2014, pages 15309 - 15318
SHIELDS ET AL., J BIOL CHEM., vol. 276, 2001, pages 6591 - 6604
SHIROKI ET AL., J IMMUNOL, vol. 178, 2007, pages 1349 - 1356
SHIROKI ET AL., J IMMUNOL., vol. 178, 2007, pages 1349 - 1356
SILVA ET AL., J BIOL CHEM., vol. 290, no. 9, 2015, pages 5462 - 5469
SKOROMBOLASFRELINGER, EXPERT REV CLIN IMMUNOL, vol. 10, no. 2, 2014, pages 207 - 217
SODING, J, BIOINFORMATICS, vol. 21, 2005, pages 951 - 960
SOUDERS ET AL., MABS, vol. 7, no. 5, 2015, pages 912 - 921
STAVENHAGEN ET AL., CANCER RES, vol. 67, 2007, pages 8882 - 8890
STROP ET AL., J MOL BIOL., vol. 420, no. 3, 2012, pages 204 - 19
SUDO ET AL., PROC NATL ACAD SCI USA., vol. 90, no. 19, 1993, pages 9125 - 9129
THIAGARAJAN ET AL., MABS, vol. 8, no. 6, 2016, pages 1088 - 1097
THIRUKKUMARAN ET AL., FRONT CHEM, vol. 7, 2020, pages 938
VAFA ET AL., METHODS, vol. 65, 2014, pages 114 - 126
VIERARAJEWSKY, EURJ IMMUNOL, vol. 18, no. 2, 1988, pages 313 - 6
VON KREUDENSTEIN ET AL., MABS, vol. 5, no. 5, 2013, pages 646 - 903
WAICKMAN ADAM T. ET AL: "The Cytokine Receptor IL-7R[alpha] Impairs IL-2 Receptor Signaling and Constrains the In Vitro Differentiation of Foxp3+ Treg Cells", ISCIENCE, vol. 23, no. 8, 1 August 2020 (2020-08-01), US, pages 101421, XP093141681, ISSN: 2589-0042, DOI: 10.1016/j.isci.2020.101421 *
WAICKMAN ET AL., CELL MOL LIFE SCI, vol. 73, no. 2, 2016, pages 253 - 269
WAICKMAN ET AL., ISCIENCE, vol. 23, no. 8, 2020, pages 101421
WALDMAN, FRONT IMMUNOL, vol. 11, 19 May 2020 (2020-05-19), pages 868
WANG ET AL., PROTEIN CELL, vol. 9, no. 1, 2018, pages 63 - 73
WHERRY, NATURE IMMUNOLOGY, vol. 12, no. 6, 2011, pages 492 - 499
XIA ET AL., FRONT IMMUNOL, vol. 10, 2019, pages 1719
XU ET AL., CELL IMMUNOL, vol. 200, 2000, pages 16 - 26
YAMASHITA ET AL., SCIENTIFIC REPORTS, vol. 6, 2016, pages 19772
YEN ET AL., CELL, vol. 185, no. 8, 2022, pages 1414 - 1430
ZALEVSKY ET AL., NAT BIOTECHNOL., vol. 28, 2010, pages 157 - 159
ZARITSKAYA ET AL., EXPERT REV VACCINES, vol. 9, no. 6, 2011, pages 601 - 616
ZAROGOULIDIS ET AL., J CANCER, vol. 5, no. 9, 2014, pages 765 - 773
ZAROUR HMDELEO AFINN OJ ET AL.: "Holland-Frei Cancer Medicine", 2003, BC DECKER, article "Categories of Tumor Antigens"
ZEMELLA ET AL., CHEMBIOCHEM, vol. 16, no. 17, 2015, pages 2420 - 2431
ZHANG ET AL., NAT BIOMED ENG, vol. 5, no. 11, 2021, pages 1288 - 1305
ZHOUROSSI, NAT REV DRUG DISCOV, vol. 16, no. 3, 2017, pages 181 - 202

Similar Documents

Publication Publication Date Title
US12030939B2 (en) Bispecific antibody binding BCMA and CD3
US20240158538A1 (en) Vista antigen-binding molecules
US11873346B2 (en) VISTA antigen-binding molecules
JP6703520B2 (ja) Cd3イプシロンおよびbcmaに対する二特異性抗体
US11780933B2 (en) HER3 antigen-binding molecules
JP2024028543A (ja) 二官能性タンパク質およびその作製
JP2023099051A (ja) Il2rベータ/共通ガンマ鎖抗体
WO2021104371A1 (fr) Anticorps anti-cd3 et anti-bcma, et protéines de liaison bispécifiques fabriquées à partir de ceux-ci
TW202118788A (zh) 包含血管舒緩素相關肽酶2抗原結合域之蛋白質及其用途
TWI644923B (zh) 抗cd52之抗體
US9944712B2 (en) TLR3 binding agents
US20230374147A1 (en) Bcma/taci antigen-binding molecules
CN118510815A (zh) 免疫缀合物及其用途
WO2024146955A1 (fr) Molécules de liaison à l&#39;antigène
WO2024146957A1 (fr) Molécules de liaison à l&#39;antigène
WO2024109657A1 (fr) Anticorps anti-ccr8 et son utilisation
JP7278623B2 (ja) 抗cd27抗体およびその使用
US20210388083A1 (en) Engineered fc
WO2024121233A1 (fr) Molécules de liaison à l&#39;antigène gp130
WO2024035342A1 (fr) Molécules de liaison à l&#39;antigène b7-h3

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: 24700547

Country of ref document: EP

Kind code of ref document: A1