WO2020065409A2 - Anti-cd40 binding molecules having engineered fc domains and therapeutic uses thereof - Google Patents

Anti-cd40 binding molecules having engineered fc domains and therapeutic uses thereof Download PDF

Info

Publication number
WO2020065409A2
WO2020065409A2 PCT/IB2019/001135 IB2019001135W WO2020065409A2 WO 2020065409 A2 WO2020065409 A2 WO 2020065409A2 IB 2019001135 W IB2019001135 W IB 2019001135W WO 2020065409 A2 WO2020065409 A2 WO 2020065409A2
Authority
WO
WIPO (PCT)
Prior art keywords
substitution
binding molecule
seq
antibody
binding
Prior art date
Application number
PCT/IB2019/001135
Other languages
English (en)
French (fr)
Other versions
WO2020065409A8 (en
WO2020065409A3 (en
Inventor
Jieyi Wang
Yi Wu
Original Assignee
Lyvgen Biopharma Co., Ltd.
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
Priority to CN202210955817.9A priority Critical patent/CN116178544A/zh
Application filed by Lyvgen Biopharma Co., Ltd. filed Critical Lyvgen Biopharma Co., Ltd.
Priority to CN201980064218.4A priority patent/CN112996818B/zh
Priority to JP2021517801A priority patent/JP2022503961A/ja
Priority to AU2019349874A priority patent/AU2019349874A1/en
Priority to KR1020217012243A priority patent/KR20210070317A/ko
Priority to EP19867441.8A priority patent/EP3856790A4/en
Priority to CN202210957160.XA priority patent/CN116063495A/zh
Priority to CA3114567A priority patent/CA3114567A1/en
Publication of WO2020065409A2 publication Critical patent/WO2020065409A2/en
Publication of WO2020065409A3 publication Critical patent/WO2020065409A3/en
Priority to US17/206,685 priority patent/US11186648B2/en
Publication of WO2020065409A8 publication Critical patent/WO2020065409A8/en
Priority to US17/536,974 priority patent/US20220081487A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/522CH1 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • CD40 Cluster of differentiation 40
  • APC antigen-presenting cell
  • CD40 is a member of the tumor necrosis factor (TNF)- receptor superfamily and is essential for various immune and inflammatory responses, including T cell-dependent immunoglobulin class switching, memory B cell development, and germinal center formation. Additionally, CD40 is found on the surface of tumor cells, such as B-lymphomas and about 70% of all solid tumors. Its activation has been shown to reverse tolerance to tumor-specific antigens, leading to antigen-specific antitumor immunity.
  • TNF tumor necrosis factor
  • Fc receptors are a family of immune cell surface proteins capable of binding to the Fc portion of antibodies. There are several different types of Fc receptors, including Fey receptors, Fca receptors, Fes receptors, and neonatal Fc receptors (FcRn), which have different binding activities to IgG, IgA, IgE, and IgG antibodies, respectively.
  • the Fey receptor subfamily includes FcyRI (CD64), FcyRIIA (CD32a), FcyRIIB (CD32b), FcyRIIB (CD32c), FcyRIIIA (CDl6a), and FcyRIIIB (CDl6b).
  • FcyRI has high binding affinity to IgGl and IgG3 antibodies, while the other FcyRs have low binding affinity to IgG antibodies.
  • FcyRIII receptors expressed on NK cells and macrophages, bind to antibodies that are attached to infected cells or invading pathogens and trigger antibody-mediated phagocytosis (ADCP) or antibody-dependent cell-mediated cytotoxicity (ADCC) of the immune cells, thereby leading to elimination of the infected cells or invading pathogens.
  • ADCP antibody-mediated phagocytosis
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • FcyRIIB receptors expressed on B cells and dendritic cells, can down regulate the activity of the immune cells when binding to IgG antibodies.
  • CD40-binding molecules e.g CD40 agonists or CD40 antagonists
  • CD40-binding molecules comprising an engineered (variant) Fc region (e.g. , an engineered Fc region of IgGl, IgG2, or IgG4 molecules) that exhibit desired hinge flexibility and preferred Fc receptor binding activity and/or selectivity, e.g., enhanced binding affinity and/or selected to FcyRIIB or substantially reduced binding affinity to one or more FcyR receptors.
  • Such CD40-binding molecules can be used for modulating immune responses (e.g. , selectively enhancing or selectively inhibiting an immune response) in subject in need of the treatment.
  • one aspect of the present disclosure provides a CD40-binding molecule (e.g., a CD40 agonist or a CD40 antagonist) comprising a CD40 binding moiety and an antibody heavy chain constant region comprising an engineered Fc region, which comprises at least one mutation at any of positions 220-331, for example, any of positions 228-329, as compared to the wild-type Fc region counterpart.
  • a CD40-binding molecule e.g., a CD40 agonist or a CD40 antagonist
  • an antibody heavy chain constant region comprising an engineered Fc region, which comprises at least one mutation at any of positions 220-331, for example, any of positions 228-329, as compared to the wild-type Fc region counterpart.
  • the numbering system is according to the EU index.
  • the CD40 binding moiety is the extracellular domain of a
  • CD40L which may comprise the amino acid sequence of SEQ ID NO: 140.
  • a CD40-binding molecule comprising such a CD40-binding moiety may be a CD40 agonist.
  • the CD40 binding moiety can be an anti-CD40 antibody fragment.
  • the anti-CD40 antibody fragment comprises a heavy chain that comprises a heavy chain variable region, which is linked to any of the engineered Fc regions described herein, and a light chain that comprises a light chain variable region, and a light chain constant region.
  • the variant Fc region may have an enhanced binding affinity and/or selectivity to FcyRIIB relative to the wild-type counterpart. In other instances, the variant Fc region may have substantially reduced binding affinity to one or more FcyR receptors. In some examples, the variant Fc region has low or no binding activity to all FcyR receptors.
  • the CD40-binding molecule may comprise a variant Fc region of an IgGl molecule (e.g.
  • a human IgGl molecule which may comprise a mutation comprising one or more of the following: (a) an amino acid substitution or deletion within positions 233-238 (e.g., 234-238); (b) a substitution at position 265; (c) a substitution at position 267; (d) a substitution at position 297; (e) a substitution at position 328, a substitution at position 329; or a combination thereof.
  • the Fc variant of an IgGl molecule may comprise one or more mutations at positions 220, 226, 229, 238, 273, 327, 330, and/or 331.
  • the mutation comprises substitutions at any of positions 233-236, a deletion at one or more of positions 236-238, or a combination thereof.
  • the substitutions at positions 233-235 may comprise E233P, L234V, L234F, L234A, L235A, and/or L235E.
  • the mutation comprises a deletion at one or more of positions 236-238.
  • the substitution at position 238 can be P238S
  • the substation at position 265 can be D265A, or a combination thereof.
  • the substitution is at position 267 and is S267E.
  • the substitution is at position 329 and is P329G.
  • the mutation may comprise a substitution at position 265, a substitution at position 297, or a combination thereof.
  • the substitution at position 265 may be D265A and the substitution at position 297 may be N297A.
  • the one or more substitutions at positions 220, 226, 229, 327, 330, and 331 can be C220S, C226S, C229S, A327G, A330S, and P331S.
  • Exemplary variant Fc regions derived from human IgGl may be one of Glml, Glm2, Glml7, Glm27, GlmAA, GlmAAG, G1N297A, Glm240, and Glm40.
  • the CD40-binding molecule may comprise an Fc region of an IgG2 molecule (e.g., a human IgG2 molecule), which may comprise a mutation comprising one or more of the following: (a) a deletion of one or more of positions 237 and 238; (b) a substitution at position 265; (b) a substitution at position 267; (c) a substitution at position 297; (d) a substitution at position 328; or a combination thereof.
  • the deletion is at position 237 or at both positions 237 and 238.
  • the Fc variant of an IgG2 molecule may comprise at least one mutation comprising a substitution at one or more of positions 233-235, 237, 238, 268, 273, 330, and 331.
  • the substitution at one or more of positions 233-235, 237, 238, 268, 273, 330, and 331 can be P233E, V234A, V234L, A235L, A235S, G237A, P238S, H268A, H268Q, V273E, A330S, and P331S.
  • the substitution is at position 267 and is S267E.
  • the substitution is at position 328 and is L328F.
  • the mutation may comprise a substitution at position 265, a substitution as position 297, or a combination thereof.
  • the substitution at position 265 may be D265A and the substitution at position 297 may be N297A.
  • the variant Fc region derived from IgG2 can be one of G2ml, G2ml7, G2ml8, G2ml9, G2m20, G2m27, G2m28, G2m29, G2m2040, G2m43, G2G4, G2mAA, and G2m40.
  • the CD40-binding molecule may comprise an Fc region of an IgG4 molecule (e.g., a human IgG4 molecule), which may comprise (a) an amino acid residue substitution at position 228; (b) a substitution or deletion at any one positions 235- 238; (c) a substitution at position 265; (d) a substitution at position 267; (e) a substitution at position 297; (e) a substitution at position 328; or a combination thereof.
  • the substitution at position 228 is S228P.
  • the deletion is at one or more of positions 236, 237, and 238 (e.g. , at both positions 236 and 237).
  • the Fc variant of an IgG4 molecule may comprise a substitution at one or more of positions 233-235, 237, and 273.
  • the substitution at one or more positions 233- 235, 237, and 273 may be E233P, F234V, F234A, L235S, L235E, L235A, G237A, and V273E.
  • the substitution at position 267 is S267E.
  • the substitution at position 328 is L328F.
  • the mutation may comprise a substitution at position 265, a substitution at position 297, or a combination thereof.
  • the substitution at position 265 may be D265A and the substitution at position 297 may be N297A.
  • the variant Fc region is one of G4ml, G4m2, G4m20, G4m28, G4m30, G4m4l, G4m42, G4m46, G4mPE, G4mAA, and G4m40.
  • any of the variant Fc regions described herein may exhibit an enhanced binding activity and/or an enhanced selectivity to FcyRIIB as compared with the wild-type Fc region.
  • the variant Fc regions described herein may have low or no binding activity to any of the FcyR receptors.
  • the variant Fc region may exhibit a decreased binding affinity to FcyRIIB.
  • the variant Fc region binds FcRn.
  • the anti-CD40 antibody fragment in any of the CD40-binding molecules described herein may be of a human antibody or a humanized antibody.
  • the anti-CD40 antibody is an agonist antibody.
  • the anti-CD40 antibody may comprise the same heavy chain complementary determining regions (HC CDRs) as those in SEQ ID NO: 128 or as those in any one of 19G6D6, 36G7B8, 13F1A7, 9F12D9, and 17C5C2, and/or the same light chain complementary determining regions (LC CDRs) as those in SEQ ID NO: 129 or those in any one of 19G6D6, 36G7B8, 13F1A7, 9F12D9, and 17C5C2.
  • the antibody fragment may comprise a heavy chain variable region of SEQ ID NO: 128 and/or a light chain variable region of SEQ ID NO: 129.
  • the antibody fragment may comprise the same heavy chain variable region and/or the same light chain variable region as one of 19G6D6, 36G7B8, 13F1A7, 9F12D9, and 17C5C2.
  • a pharmaceutical composition comprising any of the CD40-binding molecules described herein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition may be used to selectively modulate (e.g . , selectively activate or selectively inhibit) an immune response in a subject.
  • the present disclosure provides a method for selectively modulating (e.g. , selectively activating or selectively inhibiting) an immune response in a subject, the method comprising administering to a subject in need thereof an effective amount of a CD40-binding molecule as described herein.
  • the subject can be a human patient having or suspected of having a cancer and the CD40-binding molecule may be a CD40 agonist.
  • Exemplary cancers include lung cancer, stomach cancer, liver cancer, breast cancer, skin cancer, pancreatic cancer, brain cancer, prostate cancer, bladder cancer, colorectal cancer, sarcoma, bone cancer, lymphoma and a hematological cancer.
  • the subject can be a human patient having or suspected of having an immune-related disorder and the CD40-binding molecule may be a CD40 antagonist.
  • Exemplary immune disorders include autoimmune diseases, immune-deficiencies, or allergies.
  • the target disease for treatment is an autoimmune disease.
  • the disclosure provides an isolated anti-CD40 antibody, which binds to the same epitope of CD40 as a reference antibody selected from the group consisting of: 19G6D6, 36G7B8, 13F1A7, 9F12D9, and 17C5C2 or competes against the reference antibody from binding to the epitope.
  • the antibody comprises a heavy chain complementary determining region 1 (CDR1), a heavy chain complementary determining region 2 (CDR2), and a heavy chain complementary determining region 3 (CDR3), which collectively are at least 85% identical to the respective heavy chain CDRs of the reference antibody; and/or wherein the antibody comprises a light chain CDR1, a light chain CDR2, and a light chain CDR3, which collectively are at least 85% identical to the respective light chain CDRs of the reference antibody.
  • CDR1 heavy chain complementary determining region 1
  • CDR2 heavy chain complementary determining region 2
  • CDR3 heavy chain complementary determining region 3
  • CDR3 collectively comprise 10 or fewer amino acid mutations relative to the respective heavy chain CDRs of the reference antibody; and/or wherein the light chain CDR1, light chain CDR2, and light chain CDR3 collectively comprise 10 or fewer amino acid mutations relative to the respective light chain CDRs of the reference antibody
  • the antibody comprises a heavy chain variable region that is at least 85% identical to the heavy chain variable region of the reference antibody and/or a light chain variable region that is at least 85% identical to the light chain variable region of the reference antibody.
  • the heavy chain variable region comprises 10 or fewer amino acid residue mutations relative to the heavy chain variable region of the reference antibody; and/or and a light chain variable region that comprises 10 or fewer amino acid mutations relative to the light chain variable region of the reference antibody.
  • the anti-CD40 antibody disclosed herein may comprise the same heavy chain variable region CDRs as the reference antibody and/or the same light chain variable region CDRs as the reference antibody.
  • any of the anti-CD40 antibodies described herein may be a human antibody or a humanized antibody. Also provided herein is a pharmaceutical composition comprising any of the anti-CD40 antibodies described herein and a pharmaceutically acceptable carrier.
  • the instant disclosure features an isolated nucleic acid or set of nucleic acids which collectively encode any of the CD40 binding molecules or any of the anti-CD40 antibodies described herein.
  • the nucleic acid or set of nucleic acids may be located on one or two vectors, for example, expression vectors. Also provided herein are host cells comprising such vector(s).
  • compositions comprising one or more of the CD40-binding molecules described herein and/or one or more of the anti-CD40 antibodies disclosed herein for use in treating a target disorder as described herein, or uses of such CD40-binding molecules and/or anti-CD40 antibodies for manufacturing a medicament for use in treating the target disorder.
  • FIGs. 1A-1D are charts showing binding activity of various anti-CD40 antibody 383 IgG variants as indicated to different types of Fey receptors expressed on CHO-K1 cells at the various concentrations as indicated.
  • concentrations of each IgG variant, from left to right, are 0.1 pg/ml, 0.3 pg/ml, 1 pg/ml, 3 pg/ml, 10 pg/ml, and 30 pg/ml.
  • the two rightmost bars (“2nd only” and“blank”) served as controls.
  • FIG 1A binding affinity to FcyRI.
  • FIG. IB binding affinity to FcyRII.
  • FIG. 1C binding affinity to FcyRIIB.
  • FIG. ID binding affinity to FcyRIIIA.
  • FIG. 2 is a chart showing stimulation of human CD40 activation in a reporter assay by a number of anti-CD40 antibody 383 IgG variants as indicated by IL8 secretion.
  • concentrations of each IgG variant correspond to: 0.003 pg/ml, 0.01 pg/ml, 0.03 pg/ml, 0.1 pg/ml, 0.3 pg/ml, 1 pg/ml, 3 pg/ml, and 10 pg/ml.
  • FIGs. 3A-3D are charts showing binding activity of a number of anti-CD40 antibody
  • FIG. 383 IgG variants as indicated to different types of Fey receptors expressed on CHO-K1 cells.
  • concentrations of each anti-CD40 antibody 383 IgG variant, from left to right, are 0.3 pg/ml, 1 pg/ml, 3 pg/ml, 10 pg/ml, and 30 pg/ml.
  • the two rightmost bars (“2nd only” and “blank”) served as controls.
  • FIG. 3A binding affinity to FcyRIIB.
  • FIG. 3B binding affinity to FcyRI.
  • FIG. 3C binding affinity to FcyRIIA.
  • FIG. 3D binding affinity to FcyRIIIA.
  • FIGs. 4A-4F are charts showing stimulation of human CD40 activation indicated by IL8 secretion in a reporter assay by a number of anti-CD40 antibody 383 IgG variants in solution and/or in co-culture with FcyRIIB expressing cells.
  • concentrations of each IgG variant, from left to right, correspond to: 0.003 pg/ml, 0.01 pg/ml, 0.03 pg/ml, 0.1 pg/ml, 0.3 pg/ml, and 1 pg/ml. Dash line indicates baseline reporter signal in each assay.
  • FIG 4A IgG variants IgGlm2, IgG2m20, IgGlm40, IgG4m40.
  • FIG. 4B IgG variants IgGlm240, IgG2m2040, IgG4m4l, IgG4m42, IgG2m43, IgG2m44, and IgG4mAA .
  • FIG. 4C IgG variants IgGl N297A, IgG2mAA, IgG2G4, IgG4mPE, and IgGlmAAG.
  • FIG. 4D IgG variants of IgGlm47, IgGlm48, IgGlm49, IgGlm50, and IgGlmAA.
  • FIG. 4E IgG variants of IgG4m46 and IgG4m30.
  • FIG. 4F IgG variants of IgGlm27, IgG2ml9, IgG2ml, IgG4m2 and IgG4m20.
  • FIGs. 5A-5D include a set of bar graphs showing the activity of exemplary anti-CD40 antibody 383 IgG variants as indicated in activation of human dendritic cells (DC) from a healthy donor by the antibodies either in solution (FIG. 5A and 5B) or in co-culture of CHO cells expressing human FcyRIIB (FIG. 5C and 5D).
  • DC activation is indicated by the bar graphs signal of IL-8 in the culture supernatant.
  • FIGs. 6A-6B are charts showing binding activity of anti-CD40 antibody 383 IgG variants as indicated to human CD40 expressed on CHO-K1 cells.
  • concentrations of each IgG variant, from left to right, are 0.01 pg/ml, 0.1 pg/ml, 1 pg/ml, and 10 pg/ml.
  • FIG. 6A a number of anti-CD40 antibody 383 IgG variants as indicated.
  • FIG. 6B a number of anti-
  • FIGs. 7A-7B are charts showing tumor growth curves of various anti-CD40 antibody 383 IgG variants as indicated in a mouse tumor model.
  • FIG. 7A a chart showing tumor volume changes at various time points after antibody treatment in homozygous B-hCD40 C57BL6 mice. The average ⁇ SEM of tumor sizes are shown.
  • FIG. 7B is a chart showing serum alanine transaminase (ALT, a liver enzyme released into serum upon liver damage) level after treatment of antibodies as shown in homozygous B-hCD40 C57BL6 mice. The average ⁇ SEM of tumor sizes are shown.
  • ALT serum alanine transaminase
  • CD40-binding molecules e.g, CD40 agonists or CD40 antagonists
  • CD40 binding moiety e.g., an extracellular domain of a CD40 ligand or an anti-CD40 antibody fragment
  • engineered (variant) Fc regions which may have altered binding affinity and/or specificity to one or more Fc receptors, for example, enhanced binding affinity to FcyRIIB, enhanced binding specificity to FcyRIIB, and/or substantially reduced binding affinity to one or more FcyR receptors (e.g., low or no binding activity to all FcyR receptors).
  • CD40-binding molecules are expected to exhibit unexpected, superior therapeutic activity.
  • CD40-binding molecules having a variant Fc region that has enhanced binding affinity to FcyRIIB would be expected to exhibit enhanced agonistic activity and CD40-binding molecules having a variant Fc region that has enhanced binding specificity to FcyRIIB would be expected to exhibit higher tumor selectivity relative to the wild-type counterpart.
  • CD40-binding molecules having low or no binding affinity to FcyR receptors would help activate CD40-positive immune cells in tumor microenvironment and/or block CD40-positive immune cells in immune disorders.
  • an Fc variant which may be of an IgG molecule (e.g., IgGl, IgG2, and IgG4 molecules such as human IgGl, human IgG2, and human IgG4 molecules), and uses thereof for modulating immune responses.
  • an Fc variant may have enhanced binding affinity to FcyRIIB (CD32B) relative to the wild- type counterpart and/or binding selectivity as relative to other Fc receptors such as FcyRIII (CD 16).
  • such an Fc variant may have substantially reduced binding affinity to one or more FcyR receptors (e.g., to all FcyR receptors) relative to the wild-type counterpart.
  • “Substantially reduced” means that the binding affinity of a Fc variant to a FcyR receptor is at least 60% lower (e.g. , 70% lower, 80% lower, 90% lower, 95% lower, 98% lower, or 99% lower) than the binding affinity of the wild-type counterpart to the same FcyR receptor.
  • the Fc variant may have low or no binding affinity to all FcyR receptors, /. e.. binding affinity cannot be detected by conventional assays or binding affinity is substantially low such that no significant bioactivity would be triggered.
  • CD40-binding molecules that comprise a CD40-binding moiety linked to an engineered Fc region having altered binding affinity and/or specificity to one or more Fc receptors as described herein.
  • Such CD40-binding molecules may be CD40 agonists, which are capable of triggering the signaling mediated by CD40/CD40L upon binding to cell surface CD40.
  • the CD40-binding molecules described herein are CD40 antagonists, which are capable of inhibiting the signaling mediated by
  • CD40/CD40L upon binding to cell surface CD40.
  • the CD40-binding molecules are useful in modulating (selectively modulating) immune responses when administered to a subject in need of the treatment.
  • a CD40-binding moiety as described herein may be any peptide or polypeptide that binds CD40, for example human CD40.
  • CD40 is an immune cell receptor well known in the art.
  • NCBI GenBank Accession Nos. P25942.1 and AAB08705.1 provide information for human and mouse CD40, respectively.
  • Provided below is an amino acid sequence of an exemplary human CD40 polypeptide.
  • CD40 polypeptides from other species are known in the art and can be obtained from publicly available gene databases, for example, GenBank, using either the human sequence or the mouse sequence as a query.
  • the CD40-binding moieties can be a polypeptide comprising an extracellular domain of a CD40 ligand (CD40L or CD 154).
  • CD40L is a membrane glycoprotein expressed on the surface of T cells. The molecule has been shown to stimulate B cell proliferation and the secretion of immunoglobulins in the presence of cytokines.
  • CD40L may induce cytokine production and tumoricidal activity in peripheral blood monocytes.
  • the sequence of CD40L and its extracellular domain are well known in the art. For example, NCBI GenBank Accession No. NP_000065.1 (extracellular domain, amino acids 47-261) provides information for human CD40L. Provided below is an amino acid sequence of an extracellular domain of an exemplary human CD40L.
  • Human CD40L (extracellular domain): HRRLDKIEDERNLHEDFVFMKTIQRCNTGERSLSLLNCEEIKSQFEGFVKDIMLNKEETKKENSFEMQKGDQNPQ IAAHVISEASSKTTSVLQWAEKGYYTMSNNLVTLENGKQLTVKRQGLYYIYAQVTFCSNREASSQAPFIASLCLK SPGRFERILLRAANTHSSAKPCGQQSIHLGGVFELQPGASVFVNVTDPSQVSHGTGFTSFGLLKL ( SEQ ID NO: 140)
  • CD40L polypeptides from other species are known in the art and can be obtained from publicly available gene databases, for example, GenBank, using the human sequence as a query.
  • the CD40-binding moiety described herein can comprise an anti-CD40 antibody.
  • anti-CD40 antibody refers to any antibody capable of binding to a CD40 polypeptide, which can be of a suitable source, for example, human or a non-human mammal (e.g., mouse, rat, rabbit, primate such as monkey, etc.).
  • the anti-CD40 antibodies described herein comprise a heavy chain that comprises a heavy chain variable domain, which is linked to any of the Fc variants described herein, and optionally a light chain that comprises a light chain variable region and a light chain constant region.
  • the heavy chain variable region (V H ) and optionally the light chain variable region (V L ) are usually involved in antigen (CD40 in this case) binding.
  • the V H and V L regions can be further subdivided into regions of hypervariability, also known as“complementarity determining regions” (“CDR”), interspersed with regions that are more conserved, which are known as“framework regions” (“FR”).
  • CDR complementarity determining regions
  • FR framework regions
  • Each V H and V L is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy- terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the extent of the framework region and CDRs can be precisely identified using methodology known in the art, for example, by the Rabat definition, the Chothia definition, the AbM definition, and/or the contact definition, all of which are well known in the art. See, e.g. , Rabat, E.
  • the anti-CD40 antibody as described herein can bind and inhibit the activity of CD40 by at least 50% (e.g., 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
  • the apparent inhibition constant (Ki app or K La ) which provides a measure of inhibitor potency, is related to the concentration of inhibitor required to reduce enzyme activity and is not dependent on enzyme concentrations.
  • the inhibitory activity of an anti-CD40 antibody described herein can be determined by routine methods known in the art.
  • the K, . a value of an antibody may be determined by measuring the inhibitory effect of different concentrations of the antibody on the extent of the reaction (e.g., enzyme activity); fitting the change in pseudo-first order rate constant (v) as a function of inhibitor concentration to the modified Morrison equation (Equation 1) yields an estimate of the apparent Ki value.
  • the Ki app can be obtained from the y-intercept extracted from a linear regression analysis of a plot of I app versus substrate concentration.
  • A is equivalent to v E, the initial velocity ( v,J of the enzymatic reaction in the absence of inhibitor (I) divided by the total enzyme concentration ( E ).
  • the anti-CD40 antibody described herein may have a Ki app value of 1000, 900, 800, 700, 600, 500, 400, 300, 200, 100, 50, 40, 30, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5 pM or less for the target antigen or antigen epitope.
  • any of the anti-CD40 antibodies may be further affinity matured to reduce the
  • the anti-CD40 antibody may suppress the signaling triggered by CD40/CD40L interaction by at least 50% (e.g. , 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
  • Such inhibitory activity can be determined by conventional methods or the assays described herein.
  • the antibodies described herein can be murine, rat, human, or any other origin (including chimeric or humanized antibodies). Such antibodies are non-naturally occurring, i.e., would not be produced in an animal without human act (e.g., immunizing such an animal with a desired antigen or fragment thereof or isolated from antibody libraries).
  • the anti-CD40 antibody is a humanized antibody, which may have one of more of the elements or characteristics described below or elsewhere herein.
  • Humanized antibodies refer to forms of non-human (e.g. , murine) antibodies that are specific chimeric immunoglobulins, immunoglobulin chains, or antigen-binding fragments thereof that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a CDR of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity.
  • the humanized antibody may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance.
  • the humanized antibody may comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin.
  • Antibodies may have Fc regions modified as described in WO 99/58572.
  • humanized antibodies have one or more CDRs (one, two, three, four, five, or six) which are altered with respect to the original antibody, which are also termed one or more CDRs “derived from” one or more CDRs from the original antibody. Humanized antibodies may also involve affinity maturation.
  • variable regions of V H and V L of a parent non-human antibody are subjected to three- dimensional molecular modeling analysis following methods known in the art.
  • framework amino acid residues predicted to be important for the formation of the correct CDR structures are identified using the same molecular modeling analysis.
  • human V H and V L chains having amino acid sequences that are homologous to those of the parent non-human antibody are identified from any antibody gene database using the parent V H and V L sequences as search queries. Human V H and V L acceptor genes are then selected.
  • the CDR regions within the selected human acceptor genes can be replaced with the CDR regions from the parent non-human antibody or functional variants thereof.
  • residues within the framework regions of the parent chain that are predicted to be important in interacting with the CDR regions can be used to substitute for the corresponding residues in the human acceptor genes.
  • the anti-CD40 antibodies described herein specifically bind to the corresponding target antigen or an epitope thereof, e.g. , CD40 antigen or epitope.
  • An antibody that“specifically binds” to an antigen or an epitope is a term well understood in the art. A molecule is said to exhibit“specific binding” if it reacts more frequently, more rapidly, with greater duration and/or with greater affinity with a particular target antigen than it does with alternative targets.
  • An antibody“specifically binds” to a target antigen or epitope if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances.
  • an antigen CD40
  • an antigenic epitope therein is an antibody that binds this target antigen with greater affinity, avidity, more readily, and/or with greater duration than it binds to other antigens or other epitopes in the same antigen. It is also understood with this definition that, for example, an antibody that specifically binds to a first target antigen may or may not specifically or preferentially bind to a second target antigen.
  • “specific binding” or“preferential binding” does not necessarily require (although it can include) exclusive binding.
  • an antibody that“specifically binds” to a target antigen or an epitope thereof may not bind to other antigens or other epitopes in the same antigen (i.e., only baseline binding activity can be detected in a conventional method).
  • the anti-CD40 antibodies described herein specifically bind to CD40.
  • the anti-CD40 antibody described herein specifically binds human CD40 or a fragment thereof as relative to the mouse counterpart, or vice versa (e.g., having a binding affinity at least 10-fold higher to one antigen than the other as determined in the same assay under the same assay conditions).
  • the anti-CD40 antibody described herein may cross-react to human and a non-human CD40 (e.g. , mouse), e.g. , the difference in binding affinity to the human and the non-human CD40 is less than 5-fold, e.g., less than 2-fold, or substantially similar.
  • a non-human CD40 e.g. , mouse
  • an anti-CD40 antibody as described herein has a suitable binding affinity for the target antigen (e.g., CD40) or antigenic epitopes thereof.
  • binding affinity refers to the apparent association constant or K A .
  • the K A is the reciprocal of the dissociation constant (K D ).
  • the anti-CD40 antibody described herein may have a binding affinity (K D ) of at least 10 5 , 10 -6 , 10 7 , 10 8 , 10 9 , 10 10 M, or lower for the target antigen or antigenic epitope.
  • An increased binding affinity corresponds to a decreased K D .
  • the antibody has specificity for the first antigen (e.g. , a first protein in a first conformation or mimic thereof) relative to the second antigen (e.g., the same first protein in a second conformation or mimic thereof; or a second protein). Differences in binding affinity (e.g., for specificity or other comparisons) can be at least 1.5, 2, 3, 4, 5, 10, 15, 20, 37.5, 50, 70, 80, 91, 100, 500, 1000, 10,000 or 10 5 fold. In some embodiments, any of the anti-CD40 antibodies may be further affinity matured to increase the binding affinity of the antibody to the target antigen or antigenic epitope thereof.
  • Binding affinity can be determined by a variety of methods including equilibrium dialysis, equilibrium binding, gel filtration, ELISA, surface plasmon resonance, or spectroscopy (e.g., using a fluorescence assay).
  • Exemplary conditions for evaluating binding affinity are in HBS-P buffer (10 mM HEPES pH7.4, 150 mM NaCl, 0.005% (v/v) Surfactant P20). These techniques can be used to measure the concentration of bound binding protein as a function of target protein concentration.
  • the concentration of bound binding protein [Bound] is generally related to the concentration of free target protein ([Free]) by the following equation:
  • anti-CD40 antibody 383 V H and V L amino acid sequences; CDRs are indicated in boldface
  • anti-CD40 antibodies are provided below (CDRs are indicated in boldface).
  • the anti-CD40 antibodies described herein bind to the same epitope as any of the exemplary antibodies described herein or competes against the exemplary antibody from binding to the CD40 antigen.
  • An“epitope” refers to the site on a target antigen that is recognized and bound by an antibody.
  • the site can be entirely composed of amino acid components, entirely composed of chemical modifications of amino acids of the protein (e.g., glycosyl moieties), or composed of combinations thereof.
  • Overlapping epitopes include at least one common amino acid residue.
  • An epitope can be linear, which is typically 6-15 amino acids in length. Alternatively, the epitope can be conformational.
  • the epitope to which an antibody binds can be determined by routine technology, for example, the epitope mapping method (see, e.g. , descriptions below).
  • An antibody that binds the same epitope as an exemplary antibody described herein may bind to exactly the same epitope or a substantially overlapping epitope (e.g., containing less than 3 non-overlapping amino acid residue, less than 2 non-overlapping amino acid residues, or only 1 non-overlapping amino acid residue) as the exemplary antibody. Whether two antibodies compete against each other from binding to the cognate antigen can be determined by a competition assay, which is well known in the art.
  • the anti-CD40 antibodies disclosed herein comprise the same heavy chain and light chain CDRs as those in (a) SEQ ID NO: 128 and SEQ ID NO: 129, (b) SEQ ID NO: 130 and SEQ ID NO: 131 (19G6D6); (c) SEQ ID NO: 132 and SEQ ID NO:
  • the anti-CD40 antibody comprises a heavy chain variable domain of SEQ ID NO: 128, SEQ ID NO: 130, SEQ ID NO: 132, SEQ ID NO: 134, SEQ ID NO: 136, or SEQ ID NO: 138, and/or a light chain variable domain of SEQ ID NO: 129, SEQ ID NO: 131, SEQ ID NO: 133, SEQ ID NO: 135, SEQ ID NO: 137, or SEQ ID NO: 139.
  • a functional variant comprises substantially the same V H and V L CDRs as any one of the exemplary anti-CD40 antibodies.
  • it may comprise only up to 10 (e.g., 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1) amino acid residue variations in the total CDR regions of the antibody (collectively) and binds the same epitope of CD40 with substantially similar affinity (e.g., having a K D value in the same order).
  • amino acid residue variations are conservative amino acid residue substitutions.
  • a “conservative amino acid substitution” refers to an amino acid substitution that does not alter the relative charge or size characteristics of the protein in which the amino acid substitution is made.
  • Variants can be prepared according to methods for altering polypeptide sequence known to one of ordinary skill in the art such as are found in references which compile such methods, e.g. Molecular Cloning: A Laboratory Manual, J. Sambrook, et al, eds., Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1989, or Current Protocols in Molecular Biology, F.M. Ausubel, et al, eds., John Wiley & Sons, Inc., New York.
  • Conservative substitutions of amino acids include substitutions made amongst amino acids within the following groups: (a) M, I, L, V; (b) F, Y, W; (c) K, R, H; (d) A, G;
  • the anti-CD40 antibody described herein may comprise heavy chain CDRs, which in combination (collectively) have at least 80% (e.g., 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% and any incremental percent therein) sequence identity with the heavy chain CDRs in SEQ ID NO: 128, SEQ ID NO: 130, SEQ ID NO: 132, SEQ ID NO: 134, SEQ ID NO: 136, or SEQ ID NO: 138.
  • the anti-CD40 antibody may comprises light chain CDRs, which collectively have at least 80% (e.g., 85%, 90%,
  • the anti-CD40 antibody described herein may comprise a heavy chain variable domain that is at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%,
  • The“percent identity” of two amino acid sequences is determined using the algorithm of Karlin and Altschul Proc. Natl. Acad. Sci. USA 87:2264-68, 1990, modified as in Karlin and Altschul Proc. Natl. Acad. Sci. USA 90:5873-77, 1993. Such an algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. J.
  • the CD40-binding molecules described herein comprise a modified heavy chain constant region, in which the Fc domain is modified to modulate its binding affinity and/or specificity to Fc receptors such as FcyRs.
  • the Fc variants in the CD40-binding molecules described herein have enhanced selectivity to FcyRIIB relative to its wild-type counterpart.
  • An Fc fragment having selectivity to FcyRIIB, selectively binding to FcyRIIB, or specifically binding to FcyRIIB is a term well understood in the art.
  • a molecule is said to exhibit “selective binding” or“specific binding” if it reacts more frequently, more rapidly, with greater duration and/or with greater affinity with a particular target antigen (e.g., an FcyRIIB receptor) than it does with alternative targets (e.g., FcyRIII receptors).
  • an Fc fragment “specifically binds” to an Fc receptor if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other Fc receptors.
  • an Fc fragment that specifically (or preferentially) binds to FcyRIIB is an Fc fragment that binds this Fc receptor with greater affinity, avidity, more readily, and/or with greater duration than it binds to other Fc receptors. It is also understood with this definition that, for example, an Fc fragment that selectively or specifically binds to a first Fc receptor may or may not specifically or preferentially bind to a second Fc receptor.
  • an Fc fragment that“selectively binds,” or “specifically binds” to a target Fc receptor may not bind to other Fc receptors (i.e., binding not detectable by routine methods).
  • the variant Fc fragment does not bind to any FcyRs.
  • the Fc variants described herein may have enhanced selectivity to FcyRIIB relative to their wild-type counterparts (the wild-type parent Fc region in which mutations are introduced to produce the Fc variants).
  • the relative binding activity to FcyRIIB versus another Fc receptor (e.g., FcyRIII) of such an Fc variant is higher than the relative binding activity to FcyRIIB versus the other Fc receptor (e.g., FcyRIII) of the wild-type counterpart.
  • the Fc variant may have enhanced binding activity to FcyRIIB and/or decreased binding activity to another Fc receptor, for example, FcyRIII.
  • the Fc variants described herein may have decreased binding activity to both FcyRIIB and another Fc receptor (for example, FcyRIII); however, the level of decreased binding activity to the other Fc receptor (e.g., FcyRIII) is greater than the level of decreased binding activity to FcyRIIB.
  • FcyRIII another Fc receptor
  • an Fc variant as described herein has a suitable binding affinity for FcyRIIB, e.g. , enhanced as compared with the wild-type parent Fc from which the Fc variant is derived.
  • binding affinity refers to the apparent association constant or K A .
  • the K A is the reciprocal of the dissociation constant (K D ).
  • the Fc variant described herein may have a binding affinity (K D ) of at least 10 5 , 10 -6 , 10 7 , 10 8 , 10 9 , 10 10 M, or lower for FcyRIIB.
  • An increased binding affinity corresponds to a decreased K D .
  • Fc variants described herein have a higher binding affinity (a higher K A or smaller K D ) to FcyRIIB as compared to the binding affinity to FcyRIII (either FcyRIIIA or FcyRIIIB). Differences in binding affinity (e.g., for specificity or other comparisons) can be at least 1.5, 2, 3, 4, 5, 10, 15, 20, 37.5, 50, 70, 80, 91, 100, 500, 1000, 10,000 or 10 5 fold.
  • the Fc variants described herein may be designed by mutating one or more amino acid residues in the wild-type of human IgGl, IgG2, or IgG4 Fc fragments in light of the amino acid residues in the corresponding mouse IgG, for example, mouse IgGl.
  • a sequence comparison of human and mouse IgGs is provided below (SEQ ID NOs: 60-64, from top to bottom, each representing a combination of fragments 211-245, 260-278, and 320-332 of the corresponding Fc region):
  • the Fc variants described herein is a human IgGl, G2, or G4 Fc variants comprising one or more mutations (e.g. , amino acid substitutions, deletions, or additions) in the hinge domain of an Fc fragment.
  • Human IgGs contain a core motif of CPPC or CPSC in the hinge domain (positions 226-229 according to the EU index). Positions 216 to 225 are deemed as the upper portion of the hinge domain and positions 230-238 are deemed as the lower portion of the hinge domain.
  • the numbering system used herein, unless explicitly indicated, is according to the EU index.
  • the one or more mutations can be located in the upper portion of the hinge domain.
  • the one or more mutations can be located in the lower portion of the hinge domain.
  • the Fc region of any of the anti-CD40 molecules disclosed herein may comprise at least one mutation at any of positions 220-331, preferably at any of positions 228-329.
  • Such Fc variants may have altered binding affinity and/or specificity to one or more of Fc receptors (e.g., Fey I IB. Fc RI. FcyRIIA, or FcyRIIIA) as compared with their native counterparts.
  • the mutations to a human IgG Fc can be made according to the corresponding amino acid residues in the hinge domain of mouse IgGl.
  • mouse IgGl does not contain the GGP motif at positions 236-238. Accordingly, one or more of the residues in this GGP motif can be deleted from a human IgGl, IgG2, or IgG4 Fc fragment to produce the Fc variants described herein.
  • the human Fc variants may contain one or more amino acid substitutions in the upper portion, in the lower portion, or both of the hinge domain.
  • the Fc variant may comprise one or more amino acid substitutions at one or more of positions 233, 234, 235, and/or 236.
  • Such an amino acid substitution may be in combination with the deletion of one or more of the GGP motif (236-238) noted herein.
  • These mutations may be introduced into a human IgG2 or IgG4 Fc fragment to produce the Fc variants described herein.
  • the Fc variants described herein contains a deletion at one or more of the positions 236-238 (e.g., 236, 237, 238 or any combination thereof)
  • any of the mutations in the hinge domain described herein may be in combination with a mutation (e.g., amino acid substitutions) at one or more positions that are involved in interaction with an Fc receptor.
  • a mutation e.g., amino acid substitutions
  • positions include, but are not limited to, positions 265, 267, 273, 297, and 327-331, or a combination thereof.
  • Exemplary amino acid substitutions at those positions include D265A, S267E, V273E, N297A, L328F, P329G, A330S, and/or P331S.
  • the Fc variants disclosed herein can be derived from an IgGl molecule (e.g., human IgGl) and contain one or more mutations at positions at one or more of positions 220, 226, 229, 234-238, 265, 267, 297, and 327-331.
  • the Fc variant may comprise a substitution or deletion within positions 234-238, a substitution at position 265 (e.g., D265A), a substitution at position 267 (e.g., S267E), a substitution at position 297 (e.g., N297A), a substitution at position 328 (e.g.
  • the Fc variants derived from an IgGl molecule may comprise a deletion at one or more of positions 236-238.
  • the Fc variants disclosed herein can be derived from an IgG2 molecule (e.g., human IgG2) and contain one or more mutations at one or more of positions 233-235, 237-238, 265-268, 273, 297, 328, 330, and 331.
  • a Fc variant may comprise comprises a deletion within positions 237-238 (e.g. , a deletion at position 237 or a deletion of both positions 237 and 238), a substitution at position 265 (e.g.
  • the Fc variant derived from an IgG2 molecule may comprise a substitution at one or more of positions 233-235, 237, 238, 268, 273, 330, and 331 (e.g., P233E, V234A, V234L, A235L, A235S, G237A, P238S, H268A, H268Q, V273E, A330S, and P331S).
  • the Fc variants disclosed herein can be derived from an IgG4 molecule (e.g., human IgG4) and contain one or more mutations at one or more positions of 228, 233-238, 265, 267, 273, 297, and 328.
  • such a Fc variant may comprise a substitution at position 228 (e.g.
  • positions 235-238 e.g., a deletion at one of positions 236-238, for example, position 236 and position 237
  • a substitution at position 265 e.g., D265A
  • a substitution at position 267 e.g., S267E
  • a substitution at position 273 e.g., V273E
  • a substitution at position 297 e.g., N297A
  • a substitution at position 328 e.g., L328F
  • a substitution or deletion at any of positions 235-238 e.g., a deletion at one of positions 236-238, for example, position 236 and position 237
  • a substitution at position 265 e.g., D265A
  • a substitution at position 267 e.g., S267E
  • a substitution at position 273 e.g., V273E
  • a substitution at position 297 e.g., N297A
  • a substitution at position 328 e.
  • the Fc variant may comprise a substitution at one or more of positions 233-235 and 237 (e.g., E233P, F234V, F234A, L235S, L235E, L235A, and G237A).
  • positions 233-235 and 237 e.g., E233P, F234V, F234A, L235S, L235E, L235A, and G237A.
  • Fc variants derived from IgG2 or IgG4 molecules that contain one or more mutations at positions 265, 267, 273, 297, 328, and/or 329 are also within the scope of the present disclosure.
  • Such mutations may include amino acid substitutions at one or more of these positions, for example, D265A, S267E, V273E, N297A, L328F, and/or P329G.
  • any of the Fc variants disclosed herein may further comprise a mutation at position 309, for example, a substitution (e.g., V309L).
  • Such an Fc variant may be derived from an IgG2 molecule (e.g., human IgG2).
  • the mutation at position 309 may be in combination with mutations at one or more of positions 234, 268, 330, and 331. Examples of such Fc variants include G2m43.
  • an Fc variant described herein may comprise an amino acid sequence at least 85% identical (e.g 90%, 95%, 98%, 99%, or above) to that of its wild-type counterpart (e.g. , the Fc fragment of wild-type human IgGl, IgG2, or IgG4 described herein).
  • amino acid residue substitutions in an Fc variant described herein are conservative amino acid residue substitutions.
  • amino acid sequences of wild-type human IgGl, IgG2, and IgG4 Fc fragments and a number of exemplary hlgGl, hIgG2, and hIgG4 Fc variants (position 221 and onward based on EU numbering) are provided below: Amino acid sequence of wild-type human IgGl Fc fragment:
  • VDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVKFNWYVDGVEVH NAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 1)
  • VDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVQFNWYVDGVEVHNAKT KPREEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGLPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVS LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 2)
  • VDKKVEPKCCVECPPCPAPELLSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKP REEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSL SLSPGK (SEQ ID NO: 7)
  • VDKKVEPKYGPPCPPCPAPELLGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKT KPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTKNQVS LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 8)
  • Glm5 VDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEEKFNWYVDGVEVH NAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 9)
  • VDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCVWDVEHEDPEVKFNWYVDGVEVH NAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 10)
  • VDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVKFNWYVDGVEVH NAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKAFPAPIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 11)
  • VDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCVWDVEHEDPEVKFNWYVDGVEVH NAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKAFPAPIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 12)
  • VDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVKFNWYVDGVEVH NAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 21)
  • VDKKVEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVKFNWYVDGVEVH NAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALGAPIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 22)
  • VDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCVWAVSHEDPEVKFNWYVDGVEVH NAKTKPREEQYASTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 160)
  • VDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVKFNWYVDGVEVH NAKTKPREEQYaSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 180)
  • VDKKVEPKSsDKTHTsPPs PAPELLGGsSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVKFNWYVDGVEVH NAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 182)
  • VDKKVEPKSCDKTHTsPPs PAPpvaGGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVKFNWYVDGVEVH NAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 183)
  • VDKKVEPKSCDKTHTCPPCPAPEfeGGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVKFNWYVDGVEVH NAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAsIEKTI SKAKGQPREPQVYTLPPSREEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK (SEQ ID NO: 184)
  • VDKTVERKCCVECPPCPAPPVASVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVQFNWYVDGVEVHNAKTKP REEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGLPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSL SLSPGK (SEQ ID NO: 23)
  • VDKTVERKYGPPCPPCPAPPVAGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVQFNWYVDGVEVHNAKT KPREEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGLPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVS LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 26)
  • VDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEEQFNWYVDGVEVHNAKT KPREEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGLPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVS LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 27)
  • VDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMI SRTPEVTCVWDVEHEDPEVQFNWYVDGVEVHNAKT KPREEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGLPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVS LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 28)
  • VDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVQFNWYVDGVEVHNAKT KPREEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGFPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVS LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 29)
  • VDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMI SRTPEVTCVWDVEHEDPEVQFNWYVDGVEVHNAKT KPREEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGFPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVS LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 30)
  • VDKTVERKCCVECPPCPAPEVSSVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVQFNWYVDGVEVHNAKTKP REEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGFPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSL SLSPGK (SEQ ID NO: 31)
  • VDKTVERKCCVECPPCPAPPVASVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEEQFNWYVDGVEVHNAKTKP REEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGLPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSL SLSPGK (SEQ ID NO: 32)
  • VDKTVERKCCVECPPCPAPPVASVFLFPPKPKDTLMI SRTPEVTCVWDVEHEDPEVQFNWYVDGVEVHNAKTKP REEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGLPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSL SLSPGK (SEQ ID NO: 33) G2ml8:
  • VDKTVERKCCVECPPCPAPPVASVFLFPPKPKDTLMI SRTPEVTCVWDVSHEDPEVQFNWYVDGVEVHNAKTKP REEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGFPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSL SLSPGK (SEQ ID NO: 34)
  • VDKTVERKCCVECPPCPAPPVASVFLFPPKPKDTLMI SRTPEVTCVWDVEHEDPEVQFNWYVDGVEVHNAKTKP REEQFNSTFRWSVLTWHQDWLNGKEYKCKVSNKGFPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSL SLSPGK (SEQ ID NO: 35)
  • VDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMI SRTPEVTCVWAVSHEDPEVQFNWYVDGVEVHNAKT KPREEQFASTFRWSVLTWHQDWLNGKEYKCKVSNKGLPAPIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVS LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 161)
  • VDKTVERKCCVECPPCPAPPaaasSVFLFPPKPKDTLMI SRTPEVTCVWDVSaEDPEVQFNWYVDGVEVHNAKT KPREEQFNSTFRWSVLTVlHQDWLNGKEYKCKVSNKGLPssIEKTI SKTKGQPREPQVYTLPPSREEMTKNQVS LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 185)
  • VDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSqEDPEVQFNWYVDGVEVHNAKT KPREEQFNSTFRWSVLTVlHQDWLNGKEYKCKVSNKGLPs siEKTI SKTKGQPREPQVYTLPPSREEMTKNQVS LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 186)
  • VDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTLMI SRTPEVTCVWDVSQEDPEVQFNWYVDGVEVHNAKT KPREEQFNSTYRWSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTI SKAKGQPREPQVYTLPPSQEEMTKNQVS LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQK SLSLSLGK (SEQ ID NO: 188)
  • the Fc variants described herein may exhibit an enhanced binding activity to FcyRIIB as compared with the wild-type counterpart.
  • Examples include G2m2, G2m5, G2m7, G2m8, G2m9, G2ml 9, G2m44, Glm7, Glm9, Glm27, Glm45, Glm47, Glm49, Glm50, G4m7, G4m46 and G4mPE.
  • the Fc variants may have an enhanced selectivity to FcyRIIB as compared with their wild-type counterparts, for example, G2ml, G2m20, G2m43, G2m44, G2G4, G2mAA, Glm2, Glml5, Glml7, Glml8, Glml9,
  • Fc variants may retain the binding activity to FcRn.
  • Fc variants can be used for constructing therapeutic agents described herein capable of cross-linking immune receptors and FcyRIIB receptor.
  • certain Fc variants as described herein may have selectivity to FcyRIIB and/or apparent low or no binding activity to any FcyR.
  • Examples include Glm2, Glm25, Glm40, GlmAAG, Glm240, G2ml, G2m20, G2m40, G2m2040, G4m5, G4ml8, G4ml9, G4m20, G4m30, G4m40, G4m4l, and G4m42.
  • Such Fc variants may retain the binding activity to FcRn.
  • Therapeutic agents e.g. , antibodies
  • containing such Fc variants may be capable of cross-linking immune receptors and FcyRIIB receptor due to avidity effect.
  • binding activity of an Fc variant is “increased” or“decreased” as relative to its wild-type counterpart means that the binding activity of the Fc variant is higher or lower than that of the wild-type counterpart as determined by the same assay under the same experimental conditions and the variation is significant (e.g., biologically significant) as known to those skilled in the art.
  • binding activity of an Fc variant is“slightly increased” or“slightly decreased” as relative to its wild-type counterpart means that the binding activity of the Fc variant is higher or lower than that of the wild-type counterpart as determined by the same assay under the same experimental conditions and the variation is statistically significant but to a limited level (e.g., up to 10%).
  • Fc variant as described herein can be designed following the guidance provided herein and produced via routine recombinant technology. Its binding affinity and specificity to various Fc receptors can be determined via routine methods. See also Examples below.
  • the CD40-binding molecules (anti-CD40 molecules) disclosed herein may maintain the hinge region of the IgGl parent, contain one or more mutations (e.g, one or more deletions at positions 236-238 in light of mouse IgGl as discussed above) that lead to reduced binding affinity to Fc receptors such as FcyR2B but maintain residual binding activity to the Fc receptor.
  • Exemplary Fc variants having such properties include, but are not limited to, Glm2, G4m20, G4m30 and G4m46.
  • the CD40 binding moiety in such anti-CD molecules may be derived from an agonistic anti-CD40 antibody.
  • Such a CD40 binding moiety may be derived from the anti-CD40 antibody 383 disclosed herein.
  • the CD40 binding moiety comprises the same V H and V L CDRS as antibody 383.
  • the CD40 binding moiety may comprise the same VH and/or VL chains as antibody 383 (SEQ ID NOs: 128 and 129, respectively).
  • the CD40 binding moiety can be a functional variant of antibody 383 as disclosed herein.
  • Such CD40 binding molecules would have a number of advantageous features as demonstrated in the Examples below. Examples include enhanced anti-tumor efficacy with improved therapeutic window and reduced side effects (e.g., less liver toxicity) in vivo, which may be attributable to the baseline CD40 agonistic activity and the residual FcyRIIB binding activity. Too high CD40 agonist activity and/or FcyRIIB binding activity are expected to induce side effects such as cell toxicity.
  • the CD40-binding molecule disclosed herein may comprise a Fc variant domain derived from IgG2 (e.g., maintain the IgG2 hinge) and contain one or more mutations (e.g., deletions or substitutions) at one or more of positions 236-238 in light of the mouse counterpart to reduce binding affinity to Fc receptors, particularly binding affinity to FcyRIIB.
  • Fc variants have deletions at one or more of positions 236-238.
  • the Fc variants may have substitutions at positions 234, 237, 238, 268 or a combination thereof.
  • Such Fc variants may contain further mutations at one or more positions involved in binding to Fc receptors, for example, position 265 (e.g., D265A).
  • the Fc variants may maintain residual binding activity to a Fc receptor such as FcyRIIB (e.g. , G2m20 and G2m43).
  • the Fc variants may not bind to Fc receptors such as FcyRIIB (e.g., G2m40).
  • the CD40 binding moiety in such anti-CD molecules may be derived from an agonistic anti-CD40 antibody.
  • Such a CD40 binding moiety may be derived from the anti-CD40 antibody 383 disclosed herein.
  • the CD40 binding moiety comprises the same V H and V L CDRS as antibody 383.
  • the CD40 binding moiety may comprise the same VH and/or VL chains as antibody 383 (SEQ ID NOs: 128 and 129, respectively).
  • the CD40 binding moiety can be a functional variant of antibody 383 as disclosed herein.
  • Such CD40 binding molecules would have a number of advantageous features as demonstrated in the Examples below.
  • Examples include enhanced anti-tumor efficacy with improved therapeutic window and reduced side effects (e.g., less liver toxicity) in vivo, which may be attributable to the combination of baseline CD40 agonistic activity and the residual FcyRIIB binding activity (e.g., 383-IghuG2m20), or the combination of relatively high CD40 agonistic activity and no binding to FcyRIIB (e.g. , 383-IghuG2m40).
  • side effects e.g., less liver toxicity
  • Fc variants for Fey receptors can be examined in alternative assays.
  • CD40 reporter assays with or without co-culture of FcyRIIB expressing cells can be used to demonstrate effect of receptor cross-linking and activation. Because of avidity effect resulting from simultaneous binding of antibody to two targets (e.g., CD40 and FcyRIIB), this assay is more sensitive to detect FcyRIIB binding. Minimal or no apparent binding detectable by FACS may show positive results in the reporter assay.
  • CD40-binding molecules described herein may be prepared by conventional methodology, for example, recombinant technology. Some examples follow.
  • a coding sequence of the CD40L extracellular domain can be fused in frame with a coding sequence of a suitable Fc variant.
  • the coding sequence for the whole CD40-binding molecule can be cloned into a suitable expression vector, which can be introduced into a suitable host cell for protein expression.
  • Antibodies binding to CD40 can be prepared by any method known in the art. See, for example, Harlow and Lane, (1998) Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York.
  • antibodies specific to CD40 or an extracellular domain thereof can be made by the conventional hybridoma technology.
  • the full-length target receptor or a fragment thereof, optionally coupled to a carrier protein such as KLH, can be used to immunize a host animal for generating antibodies binding to that antigen.
  • the route and schedule of immunization of the host animal are generally in keeping with established and conventional techniques for antibody stimulation and production, as further described herein. General techniques for production of mouse, humanized, and human antibodies are known in the art and are described herein.
  • any mammalian subject including humans or antibody producing cells therefrom can be manipulated to serve as the basis for production of mammalian, including human hybridoma cell lines.
  • the host animal is inoculated intraperitoneally, intramuscularly, orally, subcutaneously, intraplantar, and/or intradermally with an amount of immunogen, including as described herein.
  • Hybridomas can be prepared from the lymphocytes and immortalized myeloma cells using the general somatic cell hybridization technique of Kohler, B. and Milstein, C. (1975) Nature 256:495-497 or as modified by Buck, D. W., et al, In Vitro, 18:377-381 (1982).
  • Available myeloma lines including but not limited to X63-Ag8.653 and those from the Salk Institute, Cell Distribution Center, San Diego, Calif., USA, may be used in the hybridization.
  • the technique involves fusing myeloma cells and lymphoid cells using a fusogen such as polyethylene glycol, or by electrical means well known to those skilled in the art.
  • the cells are separated from the fusion medium and grown in a selective growth medium, such as hypoxanthine-aminopterin-thymidine (HAT) medium, to eliminate unhybridized parent cells.
  • a selective growth medium such as hypoxanthine-aminopterin-thymidine (HAT) medium
  • HAT hypoxanthine-aminopterin-thymidine
  • Any of the media described herein, supplemented with or without serum, can be used for culturing hybridomas that secrete monoclonal antibodies.
  • EBV immortalized B cells may be used to produce the anti-immune cell receptor monoclonal antibodies described herein.
  • hybridomas are expanded and subcloned, if desired, and supernatants are assayed for anti -immunogen activity by conventional immunoassay procedures (e.g., radioimmunoassay, enzyme immunoassay, or fluorescence immunoassay).
  • immunoassay procedures e.g., radioimmunoassay, enzyme immunoassay, or fluorescence immunoassay.
  • Hybridomas that may be used as source of antibodies encompass all derivatives, progeny cells of the parent hybridomas that produce monoclonal antibodies capable of modulating the activity of the target immune cell receptor.
  • Hybridomas that produce such antibodies may be grown in vitro or in vivo using known procedures.
  • the monoclonal antibodies may be isolated from the culture media or body fluids, by conventional immunoglobulin purification procedures such as ammonium sulfate precipitation, gel electrophoresis, dialysis, chromatography, and ultrafiltration, if desired.
  • Undesired activity if present, can be removed, for example, by running the preparation over adsorbents made of the immunogen attached to a solid phase and eluting or releasing the desired antibodies off the immunogen.
  • a target antigen or a fragment containing the target amino acid sequence conjugated to a protein that is immunogenic in the species to be immunized e.g., keyhole limpet hemocyanin, serum album
  • an antibody (monoclonal or polyclonal) of interest (e.g., produced by a hybridoma) may be sequenced and the polynucleotide sequence may then be cloned into a vector for further construction of the anti-CD40 antibodies described herein.
  • the sequence encoding the antibody of interest may be maintained in vector in a host cell and the host cell can then be expanded and frozen for future use.
  • the polynucleotide sequence may be used for genetic manipulation to“humanize” the antibody or to improve the affinity (affinity maturation), or other characteristics of the antibody.
  • the constant region may be engineered to more resemble human constant regions to avoid immune response if the antibody is used in clinical trials and treatments in humans. It may be desirable to genetically manipulate the antibody sequence to obtain greater affinity to the target antigen and greater efficacy in inhibiting or activating the activity of the immune cell receptor. It will be apparent to one of skill in the art that one or more polynucleotide changes can be made to the antibody and still maintain its binding specificity to the target receptor.
  • Fully human antibodies can be obtained by using commercially available mice that have been engineered to express specific human immunoglobulin proteins.
  • Transgenic animals that are designed to produce a more desirable (e.g. , fully human antibodies) or more robust immune response may also be used for generation of humanized or human antibodies. Examples of such technology are Xenomouse R TM from Amgen, Inc. (Fremont, Calif.) and HuMAb-Mouse R TM and TC MouseTM from Medarex, Inc. (Princeton, N.J.).
  • antibodies may be made recombinantly by phage display or yeast technology. See, for example, U.S. Pat. Nos. 5,565,332; 5,580,717; 5,733,743; and 6,265,150; and Winter et al, (1994) Amur Rev. Immunol. 12:433-455.
  • antibody library technology such as the phage display technology
  • yeast display technology or mammalian cell display technology, can be used to isolated antibodies such as human antibodies specific to a target immune receptor.
  • variable regions of V H and V L of a parent non-human antibody are subjected to three- dimensional molecular modeling analysis following methods known in the art.
  • framework amino acid residues predicted to be important for the formation of the correct CDR structures are identified using the same molecular modeling analysis.
  • human VH and VL chains having amino acid sequences that are homologous to those of the parent non-human antibody are identified from any antibody gene database using the parent VH and VL sequences as search queries. Human VH and VL acceptor genes are then selected.
  • the CDR regions within the selected human acceptor genes can be replaced with the CDR regions from the parent non-human antibody or functional variants thereof.
  • residues within the framework regions of the parent chain that are predicted to be important in interacting with the CDR regions can be used to substitute for the corresponding residues in the human acceptor genes.
  • the coding sequence of its heavy chain can be fused in-frame with the coding sequence of a suitable Fc variant, which may selectively bind FcgRIIB or which does not bind any FcyRs, for example, any of the Fc variants described herein via routine recombinant technology.
  • the antibody is first investigated for its agonistic effect to activate the immune cell receptor to which it binds.
  • Such an agonistic antibody can be selected for making the CD40-binding molecules described herein to enhance the agonistic effects.
  • the antibody is first investigated for its antagonistic effect to inhibit the immune cell receptor to which it binds.
  • an antagonistic antibody can be selected for making the CD40-binding molecule described herein to down-regulate immune responses.
  • an Fc variant having low or no binding activity to any Fc receptor can be selected for making CD40 antagonists.
  • the resultant antibody molecules or CD40-binding molecules described herein can be produced via routine recombinant technology as exemplified below.
  • Nucleic acids encoding the heavy and light chain of an antibody or the polypeptide of a CD40-binding molecule as described herein can be cloned into one expression vector, each nucleotide sequence being in operable linkage to a suitable promoter.
  • each of the nucleotide sequences encoding the heavy chain and light chain is in operable linkage to a distinct prompter.
  • the nucleotide sequences encoding the heavy chain and the light chain can be in operable linkage with a single promoter, such that both heavy and light chains are expressed from the same promoter.
  • an internal ribosomal entry site IRS can be inserted between the heavy chain and light chain encoding sequences.
  • the nucleotide sequences encoding the two chains of the antibody are cloned into two vectors, which can be introduced into the same or different cells.
  • the two chains are expressed in different cells, each of them can be isolated from the host cells expressing such and the isolated heavy chains and light chains can be mixed and incubated under suitable conditions allowing for the formation of the antibody.
  • a nucleic acid sequence encoding one or all chains of an antibody can be cloned into a suitable expression vector in operable linkage with a suitable promoter using methods known in the art.
  • the nucleotide sequence and vector can be contacted, under suitable conditions, with a restriction enzyme to create complementary ends on each molecule that can pair with each other and be joined together with a ligase.
  • synthetic nucleic acid linkers can be ligated to the termini of a gene. These synthetic linkers contain nucleic acid sequences that correspond to a particular restriction site in the vector.
  • expression vectors/promoter would depend on the type of host cells for use in producing the antibodies.
  • promoters can be used for expression of the antibodies described herein, including, but not limited to, cytomegalovirus (CMV) intermediate early promoter, a viral LTR such as the Rous sarcoma virus LTR, HIV-LTR, HTLV-l LTR, the simian virus 40 (SV40) early promoter, E. coli lac UV5 promoter, and the herpes simplex tk virus promoter.
  • CMV cytomegalovirus
  • a viral LTR such as the Rous sarcoma virus LTR, HIV-LTR, HTLV-l LTR, the simian virus 40 (SV40) early promoter
  • E. coli lac UV5 promoter the herpes simplex tk virus promoter.
  • Regulatable promoters can also be used.
  • Such regulatable promoters include those using the lac repressor from E. coli as a transcription modulator to regulate transcription from lac operator-bearing mammalian cell promoters [Brown, M. et al, Cell, 49:603-612 (1987)], those using the tetracycline repressor (tetR) [Gossen, M., and Bujard, EL, Proc. Natl. Acad. Sci. USA 89:5547-5551 (1992); Yao, F. et al, Human Gene Therapy, 9: 1939-1950 (1998); Shockelt, P., et al, Proc. Natl. Acad. Sci.
  • Regulatable promoters that include a repressor with the operon can be used.
  • the lac repressor from E. coli can function as a transcriptional modulator to regulate transcription from lac operator-bearing mammalian cell promoters [M. Brown et al, Cell, 49:603-612 (1987)]; Gossen and Bujard (1992); [M. Gossen et al., Natl. Acad. Sci.
  • cytomegalovirus (hCMV) major immediate-early promoter to create a tetR-tet operator system to control gene expression in mammalian cells.
  • a tetracycline inducible switch is used.
  • the tetracycline repressor (tetR) alone, rather than the tetR- mammalian cell transcription factor fusion derivatives can function as potent trans-modulator to regulate gene expression in mammalian cells when the tetracycline operator is properly positioned downstream for the TATA element of the CMVIE promoter (Yao et al., Human Gene Therapy).
  • this tetracycline inducible switch does not require the use of a tetracycline repressor-mammalian cells transactivator or repressor fusion protein, which in some instances can be toxic to cells (Gossen et al., Natl. Acad. Sci. USA, 89:5547-5551 (1992); Shockett et al., Proc. Natl. Acad. Sci. USA, 92:6522-6526 (1995)), to achieve its regulatable effects.
  • the vector can contain, for example, some or all of the following: a selectable marker gene, such as the neomycin gene for selection of stable or transient transfectants in mammalian cells; enhancer/promoter sequences from the immediate early gene of human CMV for high levels of transcription; transcription termination and RNA processing signals from SV40 for mRNA stability; SV40 polyoma origins of replication and ColEl for proper episomal replication; internal ribosome binding sites (IRESes), versatile multiple cloning sites; and T7 and SP6 RNA promoters for in vitro transcription of sense and antisense RNA.
  • a selectable marker gene such as the neomycin gene for selection of stable or transient transfectants in mammalian cells
  • enhancer/promoter sequences from the immediate early gene of human CMV for high levels of transcription
  • transcription termination and RNA processing signals from SV40 for mRNA stability SV40 polyoma origins of replication and ColEl for proper episomal replication
  • One or more vectors comprising nucleic acids encoding any of the antibodies may be introduced into suitable host cells for producing the antibodies.
  • the host cells can be cultured under suitable conditions for expression of the antibody or any polypeptide chain thereof.
  • Such antibodies or polypeptide chains thereof can be recovered by the cultured cells (e.g., from the cells or the culture supernatant) via a conventional method, e.g., affinity purification.
  • polypeptide chains of the antibody can be incubated under suitable conditions for a suitable period of time allowing for production of the antibody.
  • methods for preparing an antibody described herein involve a recombinant expression vector that encodes both the heavy chain and the light chain of an antibody as described herein.
  • the recombinant expression vector can be introduced into a suitable host cell (e.g. , a dhfr- CHO cell) by a conventional method, e.g. , calcium phosphate- mediated transfection.
  • a suitable host cell e.g. , a dhfr- CHO cell
  • Positive transformant host cells can be selected and cultured under suitable conditions allowing for the expression of the two polypeptide chains that form the antibody, which can be recovered from the cells or from the culture medium.
  • the two chains recovered from the host cells can be incubated under suitable conditions allowing for the formation of the antibody.
  • two recombinant expression vectors are provided, one encoding the heavy chain of the anti-immune cell receptor antibody and the other encoding the light chain of the same antibody.
  • Both of the two recombinant expression vectors can be introduced into a suitable host cell (e.g., dhfr- CHO cell) by a conventional method, e.g., calcium phosphate- mediated transfection.
  • each of the expression vectors can be introduced into a suitable host cells. Positive transformants can be selected and cultured under suitable conditions allowing for the expression of the polypeptide chains of the antibody.
  • the antibody produced therein can be recovered from the host cells or from the culture medium.
  • the polypeptide chains can be recovered from the host cells or from the culture medium and then incubated under suitable conditions allowing for formation of the antibody.
  • the two expression vectors are introduced into different host cells, each of them can be recovered from the corresponding host cells or from the corresponding culture media. The two polypeptide chains can then be incubated under suitable conditions for formation of the antibody.
  • Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recovery of the antibodies from the culture medium.
  • some antibodies can be isolated by affinity chromatography with a Protein A or Protein G coupled matrix.
  • compositions comprising the CD40- binding molecules or any of the anti-CD40 antibodies described herein and uses of such for modulating immune responses triggered by CD40/CD40L signaling.
  • Such CD40-binding molecules as described herein or anti-CD40 antibodies as also described herein can be used for treating diseases such as cancer or immune-related disorders.
  • CD40-binding molecules or anti-CD40 antibodies as described herein can be mixed with a pharmaceutically acceptable carrier (excipient) to form a pharmaceutical composition for use in treating a target disease.
  • a pharmaceutically acceptable carrier excipient
  • “Acceptable” means that the carrier must be compatible with the active ingredient of the composition (and preferably, capable of stabilizing the active ingredient) and not deleterious to the subject to be treated.
  • compositions including buffers, which are well known in the art. See, e.g., Remington: The Science and Practice of Pharmacy 20th Ed. (2000) Lippincott Williams and Wilkins, Ed. K. E. Hoover.
  • compositions to be used in the present methods can comprise pharmaceutically acceptable carriers, excipients, or stabilizers in the form of lyophilized formulations or aqueous solutions.
  • pharmaceutically acceptable carriers excipients, or stabilizers in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations used, and may comprise buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
  • the pharmaceutical composition described herein comprises liposomes containing the antibodies (or the encoding nucleic acids) which can be prepared by methods known in the art, such as described in Epstein, et al, Proc. Natl. Acad. Sci. USA 82:3688 (1985); Hwang, et al, Proc. Natl. Acad. Sci. USA 77:4030 (1980); and U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556.
  • Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • PEG-PE PEG-derivatized phosphatidylethanolamine
  • the CD40-binding molecules, anti-CD40 antibodies, or the encoding nucleic acid(s), may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • macroemulsions for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl- methacrylate), or poly(v nylalcohol)), polylactides (U.S. Pat. No.
  • microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(-)-3 -hydroxy butyric acid.
  • compositions to be used for in vivo administration must be sterile. This is readily accomplished by, for example, filtration through sterile filtration membranes.
  • Therapeutic antibody compositions are generally placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic inj ection needle.
  • compositions described herein can be in unit dosage forms such as tablets, pills, capsules, powders, granules, solutions or suspensions, or suppositories, for oral, parenteral or rectal administration, or administration by inhalation or insufflation.
  • the principal active ingredient can be mixed with a pharmaceutical carrier, e.g., conventional tableting ingredients such as com starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g., water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a non-toxic pharmaceutically acceptable salt thereof.
  • a pharmaceutical carrier e.g., conventional tableting ingredients such as com starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g., water
  • preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention.
  • the tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer that serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
  • Suitable surface-active agents include, in particular, non-ionic agents, such as polyoxyethylenesorbitans (e.g., TweenTM 20, 40, 60, 80 or 85) and other sorbitans (e.g., SpanTM 20, 40, 60, 80 or 85).
  • Compositions with a surface-active agent will conveniently comprise between 0.05 and 5% surface-active agent, and can be between 0.1 and 2.5%. It will be appreciated that other ingredients may be added, for example mannitol or other pharmaceutically acceptable vehicles, if necessary.
  • Suitable emulsions may be prepared using commercially available fat emulsions, such as IntralipidTM, LiposynTM, InfonutrolTM, LipofundinTM and LipiphysanTM.
  • the active ingredient may be either dissolved in a pre-mixed emulsion composition or alternatively it may be dissolved in an oil (e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, com oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e.g., egg phospholipids, soybean phospholipids or soybean lecithin) and water.
  • an oil e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, com oil or almond oil
  • a phospholipid e.g., egg phospholipids, soybean phospholipids or soybean lecithin
  • Suitable emulsions will typically contain up to 20% oil, for example, between 5 and 20%.
  • the fat emulsion can comprise fat droplets between 0.1 and 1.0 pm, particularly 0.1 and 0.5 pm, and have a pH in the range of 5.5 to 8.0.
  • the emulsion compositions can be those prepared by mixing an antibody with IntralipidTM or the components thereof (soybean oil, egg phospholipids, glycerol and water).
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • compositions in preferably sterile pharmaceutically acceptable solvents may be nebulized by use of gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device may be attached to a face mask, tent or intermittent positive pressure breathing machine. Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner. IV. Therapeutic Applications
  • CD40-binding molecules or anti-CD40 antibodies disclosed herein may be used to modulating (e.g., enhancing or inhibiting) immune responses against invading pathogens and/or diseased cells such as cancer cells.
  • an effective amount of the pharmaceutical composition described herein can be administered to a subject (e.g., a human) in need of the treatment via a suitable route, such as intravenous administration, e.g., as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal,
  • a suitable route such as intravenous administration, e.g., as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal,
  • nebulizers for liquid formulations including jet nebulizers and ultrasonic nebulizers are useful for administration.
  • Liquid formulations can be directly nebulized and lyophilized powder can be nebulized after reconstitution.
  • the antibodies as described herein can be aerosolized using a fluorocarbon formulation and a metered dose inhaler, or inhaled as a lyophilized and milled powder.
  • the subject to be treated by the methods described herein can be a mammal, more preferably a human.
  • Mammals include, but are not limited to, farm animals, sport animals, pets, primates, horses, dogs, cats, mice and rats.
  • the subject is a human patient having or at risk for a cell-mediated disease or disorder, such as cancer including but not limited to lung cancer, stomach cancer, liver cancer, breast cancer, skin cancer, pancreatic cancer, brain cancer, prostate cancer, bladder cancer, or colorectal cancer.
  • a cell-mediated disease or disorder such as cancer including but not limited to lung cancer, stomach cancer, liver cancer, breast cancer, skin cancer, pancreatic cancer, brain cancer, prostate cancer, bladder cancer, or colorectal cancer.
  • cancers include, but are not limited to, breast cancer; biliary tract cancer; bladder cancer; brain cancer including glioblastomas and medulloblastomas; cervical cancer; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric cancer; hematological neoplasms including acute lymphocytic and myelogenous leukemia, e.g., B Cell CLL; T-cell acute lymphoblastic leukemia/lymphoma; hairy cell leukemia; chronic myelogenous leukemia, multiple myeloma; AIDS-associated leukemias and adult T-cell leukemia/lymphoma; intraepithelial neoplasms including Bowen's disease and Paget's disease; liver cancer; lung cancer; lymphomas including Hodgkin's disease and lymphocytic lymphomas; neuroblastomas; oral cancer including squamous cell carcinoma; ovarian cancer including those arising from
  • osteosarcoma skin cancer including melanoma, Merkel cell carcinoma, Kaposi’s sarcoma, basal cell carcinoma, and squamous cell cancer; testicular cancer including germinal tumors such as seminoma, non-seminoma (teratomas, choriocarcinomas), stromal tumors, and germ cell tumors; thyroid cancer including thyroid adenocarcinoma and medullar carcinoma; and renal cancer including adenocarcinoma and Wilms tumor.
  • a subject having a cancer can be identified by routine medical examination, e.g., laboratory tests, organ functional tests, CT scans, or ultrasounds.
  • the subject to be treated by the method described herein may be a human cancer patient who has undergone or is subjecting to an anti-cancer therapy, for example, chemotherapy, radiotherapy, immunotherapy, or surgery.
  • the subject is a human patient having or at risk for an immune- related disorder.
  • Immune-related disorders refer to a dysfunction of the immune system, including autoimmune diseases, immunodeficiencies, and/or allergies.
  • the immune-related disorder is an autoimmune disease.
  • immune-related disorders include, but are not limited to, rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), Myasthenia Gravis (MG), Graves’ Disease, Idiopathic
  • Thrombocytopenia Purpura ITP
  • Guillain-Barre Syndrome autoimmune myocarditis
  • Membrane Glomerulonephritis diabetes mellitus, Type I or Type II diabetes
  • multiple sclerosis Reynaud's syndrome, autoimmune thyroiditis, gastritis, Celiac Disease, Vitiligo, Hepatitis, primary biliary cirrhosis, inflammatory bowel disease, spondyloarthropathies, experimental autoimmune encephalomyelitis, immune neutropenia, juvenile onset diabetes, and immune responses associated with delayed hypersensitivity mediated by cytokines, T- lymphocytes typically found in tuberculosis, sarcoidosis, and polymyositis, polyarteritis, cutaneous vasculitis, pemphigus, pemphigold, Goodpasture's syndrome, Kawasaki's disease, systemic sclerosis, anti-phospholipid syndrome, Sjogren's syndrome, graft-versus-
  • a subject having an immune-related disorder can be identified by routine medical examination, e.g., with laboratory tests.
  • the subject to be treated by the method described herein may be a human subject with an immune-related disorder who has undergone or is subjecting to an immune- related disorder treatment, for example, immunosuppressive mediation, hormone replacement therapy, blood transfusions, anti-inflammatory medication, and/or pain medication.
  • an effective amount refers to the amount of each active agent required to confer therapeutic effect on the subject, either alone or in combination with one or more other active agents.
  • the therapeutic effect is modulating (e.g., activating) the target immune receptor, thereby triggering or enhancing immune responses mediated by the receptor.
  • Effective amounts vary, as recognized by those skilled in the art, depending on the particular condition being treated, the severity of the condition, the individual patient parameters including age, physical condition, size, gender and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose of the individual components or combinations thereof be used, that is, the highest safe dose according to sound medical judgment.
  • Empirical considerations such as the half-life, generally will contribute to the determination of the dosage.
  • molecules that are compatible with the human immune system such as humanized antibodies or fully human antibodies, may be used to prolong half-life of the antibody and to prevent the antibody being attacked by the host's immune system.
  • Frequency of administration may be determined and adjusted over the course of therapy, and is generally, but not necessarily, based on treatment and/or suppression and/or amelioration and/or delay of a target disease/disorder.
  • sustained continuous release formulations of an antibody may be appropriate.
  • formulations and devices for achieving sustained release are known in the art.
  • dosages for a CD40-binding molecule or an anti-CD40 antibody as described herein may be determined empirically in individuals who have been given one or more administration(s) of the antibody. Individuals are given incremental dosages of the agonist. To assess efficacy of the agonist, an indicator of the disease/disorder can be followed.
  • an initial candidate dosage can be about 2 mg/kg.
  • a typical daily dosage might range from about any of 0.1 pg/kg to 3 pg/kg to 30 pg/kg to 300 pg/kg to 3 mg/kg, to 30 mg/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment is sustained until a desired suppression of symptoms occurs or until sufficient therapeutic levels are achieved to alleviate a target disease or disorder, or a symptom thereof.
  • An exemplary dosing regimen comprises administering an initial dose of about 2 mg/kg, followed by a weekly maintenance dose of about 1 mg/kg of the antibody, or followed by a maintenance dose of about 1 mg/kg every other week.
  • other dosage regimens may be useful, depending on the pattern of pharmacokinetic decay that the practitioner wishes to achieve. For example, dosing from one-four times a week is contemplated. In some embodiments, dosing ranging from about 3 pg/mg to about 2 mg/kg (such as about 3 pg/mg, about 10 pg/mg, about 30 pg/mg, about 100 pg/mg, about 300 pg/mg, about 1 mg/kg, and about 2 mg/kg) may be used.
  • dosing frequency is once every week, every 2 weeks, every 4 weeks, every 5 weeks, every 6 weeks, every 7 weeks, every 8 weeks, every 9 weeks, or every 10 weeks; or once every month, every 2 months, or every 3 months, or longer.
  • the progress of this therapy is easily monitored by conventional techniques and assays.
  • the dosing regimen (including the therapeutic used) can vary over time.
  • doses ranging from about 0.3 to 5.00 mg/kg may be administered.
  • the dosage of the therapeutic agents such as antibodies described herein can be 10 mg/kg.
  • the particular dosage regimen i.e., dose, timing and repetition, will depend on the particular individual and that individual's medical history, as well as the properties of the individual agents (such as the half-life of the agent, and other considerations well known in the art).
  • the appropriate dosage of a CD40-binding molecule or anti-CD40 antibody as described herein will depend on the specific antibody, antibodies, and/or non-antibody peptide (or compositions thereof) employed, the type and severity of the disease/disorder, whether the CD40-binding molecule or anti-CD40 antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the agonist, and the discretion of the attending physician.
  • the clinician will administer a CD40-binding molecule or anti-CD40 antibody, until a dosage is reached that achieves the desired result.
  • the desired result is a decrease in thrombosis.
  • Administration of one or more CD40- binding molecule or anti-CD40 antibody can be continuous or intermittent, depending, for example, upon the recipient's physiological condition, whether the purpose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners.
  • the administration of a CD40-binding molecule or anti-CD40 antibody may be essentially continuous over a preselected period of time or may be in a series of spaced dose, e.g., either before, during, or after developing a target disease or disorder.
  • the term“treating” refers to the application or administration of a composition including one or more active agents to a subject, who has a target disease or disorder, a symptom of the disease/disorder, or a predisposition toward the disease/disorder, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect the disorder, the symptom of the disease, or the predisposition toward the disease or disorder.
  • Alleviating a target disease/disorder includes delaying the development or progression of the disease, or reducing disease severity. Alleviating the disease does not necessarily require curative results. As used therein, "delaying" the development of a target disease or disorder means to defer, hinder, slow, retard, stabilize, and/or postpone progression of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individuals being treated.
  • a method that“delays” or alleviates the development of a disease, or delays the onset of the disease is a method that reduces probability of developing one or more symptoms of the disease in a given time frame and/or reduces extent of the symptoms in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a number of subjects sufficient to give a statistically significant result.
  • “Development” or“progression” of a disease means initial manifestations and/or ensuing progression of the disease. Development of the disease can be detectable and assessed using standard clinical techniques as well known in the art. However, development also refers to progression that may be undetectable. For purpose of this disclosure, development or progression refers to the biological course of the symptoms. “Development” includes occurrence, recurrence, and onset. As used herein“onset” or“occurrence” of a target disease or disorder includes initial onset and/or recurrence.
  • the antibodies described herein are administered to a subject in need of the treatment at an amount sufficient to activate the activity of the target receptor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo.
  • compositions can be administered via other conventional routes, e.g., administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous,
  • intramuscular, intraarticular, intraarterial, intrasynovial, intrastemal, intrathecal, intralesional, and intracranial injection or infusion techniques can be administered to the subject via injectable depot routes of administration such as using 1-, 3-, or 6-month depot injectable or biodegradable materials and methods.
  • the pharmaceutical composition is administered intraocularly or intravitreally.
  • Injectable compositions may contain various carriers such as vegetable oils, dimethylactamide, dimethyformamide, ethyl lactate, ethyl carbonate, isopropyl myristate, ethanol, and polyols (glycerol, propylene glycol, liquid polyethylene glycol, and the like).
  • carriers such as vegetable oils, dimethylactamide, dimethyformamide, ethyl lactate, ethyl carbonate, isopropyl myristate, ethanol, and polyols (glycerol, propylene glycol, liquid polyethylene glycol, and the like).
  • water soluble antibodies can be administered by the drip method, whereby a pharmaceutical formulation containing the antibody and a physiologically acceptable excipient is infused.
  • Physiologically acceptable excipients may include, for example, 5% dextrose, 0.9% saline, Ringer’s solution or other suitable excipients.
  • Intramuscular preparations e.g., a sterile formulation of a suitable soluble salt form of the antibody
  • a pharmaceutical excipient such as Water-for- Injection, 0.9% saline, or 5% glucose solution.
  • the particular dosage regimen i.e., dose, timing and repetition, used in the method described herein will depend on the particular subject and that subject's medical history.
  • more than one antibody, or a combination of an antibody and another suitable therapeutic agent may be administered to a subject in need of the treatment.
  • the CD40-binding molecule or anti-CD40 antibody can also be used in conjunction with other agents that serve to enhance and/or complement the effectiveness of the agents. Treatment efficacy for a target disease/disorder can be assessed by methods well- known in the art.
  • the therapeutic agent described herein may be utilized in conjunction with other types of therapy for the target disease such as cancer. Additional anti-cancer therapy includes chemotherapy, surgery, radiation, gene therapy, and so forth. When a second therapeutic agent is used, such an agent can be administered simultaneously or sequentially (in any order) with the CD40-binding molecule or anti-CD40 antibody described herein.
  • suitable therapeutically effective dosages for each agent may be lowered due to the additive action or synergy.
  • the treatments of the disclosure can be combined with other immunomodulatory treatments such as, e.g. , therapeutic vaccines (including but not limited to GVAX, DC-based vaccines, etc.), or checkpoint inhibitors (including but not limited to agents that block CTLA4, PD1, LAG3, TIM3, etc.).
  • therapeutic vaccines including but not limited to GVAX, DC-based vaccines, etc.
  • checkpoint inhibitors including but not limited to agents that block CTLA4, PD1, LAG3, TIM3, etc.
  • the treatment of the present disclosure can be combined with a chemotherapeutic agent, for example, pyrimidine analogs (5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine), purine analogs, folate antagonists and related inhibitors (mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine (cladribine)); antiproliferative/antimitotic agents including natural products such as vinca alkaloids (vinblastine, vincristine, and vinorelbine), microtubule disruptors such as taxane (paclitaxel, docetaxel), vincristin, vinblastin, nocodazole, epothilones and navelbine, epidipodophyllotoxins (etoposide, teniposide), DNA damaging agents (actinomycin, amsacrine, anthracyclines, bleomycin, busulfan, camptothecin,
  • antibiotics such as dactinomycin (actinomycin D), daunorubicin, doxorubicin (adriamycin), idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin; enzymes (L- asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine); antiplatelet agents;
  • antiproliferative/antimitotic alkylating agents such as nitrogen mustards (mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil), ethylenimines and methylmelamines (hexamethylmelamine and thiotepa), alkyl sulfonates-busulfan, nitrosoureas (carmustine (BCNU) and analogs, streptozocin), trazenes-dacarbazinine (DTIC); antiproliferative/antimitotic antimetabolites such as folic acid analogs (methotrexate);
  • nitrogen mustards mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil
  • ethylenimines and methylmelamines hexamethylmelamine and thiotepa
  • alkyl sulfonates-busulfan nitrosoureas (carmustine (BCNU
  • platinum coordination complexes cisplatin, carboplatin
  • procarbazine hydroxyurea, mitotane, aminoglutethimide
  • hormones hormone analogs (estrogen, tamoxifen, goserelin, bicalutamide, nilutamide) and aromatase inhibitors (letrozole, anastrozole)
  • anticoagulants heparin, synthetic heparin salts and other inhibitors of thrombin
  • fibrinolytic agents such as tissue plasminogen activator, streptokinase and urokinase), aspirin, dipyridamole, ticlopidine, clopidogrel, abciximab
  • antimigratory agents antisecretory agents (breveldin);
  • immunosuppressives cyclosporine, tacrolimus (FK-506), sirolimus (rapamycin), azathioprine, mycophenolate mofetil
  • anti-angiogenic compounds e.g., TNP-470, genistein, bevacizumab
  • growth factor inhibitors e.g., fibroblast growth factor (FGF) inhibitors
  • angiotensin receptor blocker e.g., fibroblast growth factor (FGF) inhibitors
  • angiotensin receptor blocker e.g., fibroblast growth factor (FGF) inhibitors
  • angiotensin receptor blocker e.g., fibroblast growth factor (FGF) inhibitors
  • angiotensin receptor blocker e.g., fibroblast growth factor (FGF) inhibitors
  • angiotensin receptor blocker e.g., fibroblast growth factor (FGF) inhibitors
  • angiotensin receptor blocker e.g., fibroblast growth factor (
  • mitochondrial dysfunction inducers and caspase activators; and chromatin disruptors.
  • the kit can comprise instructions for use in accordance with any of the methods described herein.
  • the included instructions can comprise a description of administration of the therapeutic agent to treat, delay the onset, or alleviate a target disease as those described herein.
  • the kit may further comprise a description of selecting an individual suitable for treatment based on identifying whether that individual has the target disease.
  • the instructions comprise a description of administering a therapeutic agent such as an antibody to an individual at risk of the target disease.
  • the instructions relating to the use of the therapeutic agent generally include information as to dosage, dosing schedule, and route of administration for the intended treatment.
  • the containers may be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses.
  • Instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit), but machine- readable instructions (e.g., instructions carried on a magnetic or optical storage disk) are also acceptable.
  • the label or package insert indicates that the composition is used for treating, delaying the onset and/or alleviating a target disease or disorder such as cancer. Instructions may be provided for practicing any of the methods described herein
  • kits of this invention are in suitable packaging.
  • suitable packaging includes, but is not limited to, vials, bottles, j ars, flexible packaging (e.g. , sealed Mylar or plastic bags), and the like.
  • packages for use in combination with a specific device such as an inhaler, nasal administration device (e.g. , an atomizer) or an infusion device such as a minipump.
  • a kit may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the container may also have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • a sterile access port for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle.
  • At least one active agent in the composition is the therapeutic agent as those described herein.
  • Kits may optionally provide additional components such as buffers and interpretive information.
  • the kit comprises a container and a label or package insert(s) on or associated with the container.
  • the invention provides articles of manufacture comprising contents of the kits described above.
  • the cDNA sequences encoding the anti-CD40 antibody variable domain with various heavy chain CH1 and Fc region or human kappa light chain constant region were synthesized and cloned. CHO transient expression was carried out with plasmids containing the corresponding heavy and light chain sequences. These antibodies were purified by protein A affinity chromatography.
  • the amino acid sequences of the heavy chain (HC) and the light chain (LC) are provided below: 383-huIgG LC
  • VTKSFNRGEC SEQ ID NO: 1436
  • IVKSFNRNEC (SEQ ID NO: 155)
  • TMTRDTS I STAYMELNRLRSDDTAVYYCARDQPLGYCTNGVCSYFDYWGQGTLVTVSSASTKGPSVFP
  • GNVFSCSVMHEALHNHYTQKSLSLS PGK (SEQ ID NO: 197)
  • TMTRDTS I STAYMELNRLRSDDTAVYYCARDQPLGYCTNGVCSYFDYWGQGTLVTVSSASTKGPSVFP
  • TMTRDTS I STAYMELNRLRSDDTAVYYCARDQPLGYCTNGVCSYFDYWGQGTLVTVSSASTKGPSVFP
  • CHO cells were genetically engineered to express human FcyRs (FcyRI, FcyRIIA, FcyRIIB, and FcyRIII) using a lentivirus delivery system as known in the art.
  • anti-CD40 antibody 383 disclosed herein as an exemplary anti-CD40 moiety
  • a number of anti-CD40 IgG Fc mutants including 383-msIgGl, 383-msIgGl DANA, 383- msIgG2a, 383-msIgG2a DANA, 383-hIgG2, control huIgGl, 383-huIgGlm27, 383- huIgG2ml9, 383-huIgGlm2, 383-huIgG2ml, 383-huIgG2m20, 383-huIgG4m2, 383- huIgG4m20, 383-huIgG4m30, 383-huIgGlmAA, and 383-huIgG4SP (amino acid sequences provided above), were designed and constructed following the disclosures herein. These IgG mutants contain mutations in
  • overexpressing CHO cells were harvested using trypsin-EDTA and were suspended in cold staining buffer (3% BSA in PBS).
  • Test IgG mutants which were diluted in staining buffer, were added into the cells. The mixture was incubated at 4°C for 2 hours, and then washed twice with cold staining buffer and re-suspended in PE-labeled anti-human IgG followed by incubation at 4°C for 2 hours. The mixture was washed twice with staining buffer and re suspended in 2% PFA in PBS for FACS.
  • a CD40 reporter assay was developed, which involves reporter cells over-expressing human CD40 T cells.
  • This GS-H2- huCD40 reporter cells were re-suspended in assay buffer and cell density and viability were determined with trypan blue.
  • the cell suspension was diluted to 1 x 10 4 cells/mL with assay buffer (MEM containing 1% FBS).
  • the cells were added at lOOuL/well, such that the final cell number was 1000 cells/well in the assay plate (Nunc, Cat#l67425).
  • Serial working dilutions of the test samples were prepared in assay buffer at 2x final concentrations.
  • Samples were added at lOOuL/well test sample at 2x final concentrations to the assay plate.
  • the assay plate was incubated in 37°C, 5% C0 2 incubator for 18-20 hours. After the 18-20 hour incubation, 8 ul of the supernatant from each well of the assay plate was collected and added to HTRF detection assay plate (Nunc).
  • a Human Interleukin 8 (reporter of CD40 activation) detection assay was performed using a Human IL-8 Assay Kit (Cisbio,
  • CD40 reporter assay was also performed in coculture with FcyRIIB-expressing CHO cells and the results are shown in FIGs 4A-4D. Binding to CD40 and FcyR2B by the tested antibody molecules simultaneously in a microenvironment would affect individual binding due to the avidity effect, which refers to the accumulated strength of multiple affinities of individual non-covalent binding interactions.
  • the antibodies showed increased activity correlating to their binding activity to FcyRIIB. It was noted that antibodies with Fc variants with weak or no apparent binding to FcyRIIB exhibited enhanced agonist activity in co culture reporter assay, suggesting higher sensitivity of the reporter assay which may be attributable to the avidity effect. Therefore, the Fc structure of the antibody can affect the agonist activity of the antibodies with the same variable domains.
  • the exemplary anti-CD40 antibody Ig variants comprising various Fc mutants were tested in vitro for CD40 binding activities and agonistic activity as described in Examples above, as well as in vivo for antitumor efficacy and toxicity as in Example 6. Their activities in activating human dendritic cells were carried out as following.
  • Frozen human PBMC from healthy donors (Allcells, Cat#PB005F) were thawed and transferred to a 50ml tube with 25mL RPMI1640 media.
  • EasySepTM Human CD14 Positive Selection Kit II (Stemcell, Cat: 17858) were used to isolate B cells. Centrifuge the PBMC suspension for l5min at 250g, and discard the supernatant and resuspend the cells in l.5mL EasySepTM Buffer. Add Selection Cocktail to sample to al4 mL (17 x 100 mm) polystyrene round-bottom tube. lOOul/ml, l50ul for each sample. Transfer the cell from 50ml tube to the 14 mL (17 x 100 mm) polystyrene round-bottom tube, mix and incubate for l5min at RT.
  • the isolated CDl4 + cells from human PBMCs were adjust to lx 10 6 cell/ml and add GM-CSF and IL-4 were added into the cell culture media. Incubate the cells in a 37 °C, 5% C02 humidified incubator for 3 days. On day 3, the culture media were changed by transferring the culture supernatant to a centrifuge tube and then centrifuged at 250 x g for 5 minutes. lOml fresh media containing GN-CSF and IL-4 were used to suspend the cell pellet in the centrifuge tube and then added to the flask during the centrifugation so that the cell pellet thus formed would be resuspended in the lOml fresh media.
  • the resuspended cells were added back to the same well or flask and incubated for an additional 2 days. On day 5, the above steps were repeated for media change. The cells were further incubated for another additional 2 days. On day 7, immature dendritic cells can be observed and are ready to be used in the desired application.
  • IL-8 and IL12 P70 were detected with the HuIL-l2-P70 detection kit (Cisbio, Cat#: 62HIL12PEH) and HuIL-8 detection kit (Cisbio, Cat#: 62HIL08PEH) in the cell culture supernatant 24h later (day3).
  • the DC activation assay was also performed in co-culture with FcyR2B expressing CHO cells.
  • the tested CD40 antibody IgG mutants stimulated human CD40 activation at various degrees as evidenced by the secretion of IL8 from the DC culture after antibody incubation.
  • the magnitude of DC activation was influenced by Fc variants contained therein due to the hinge flexibility of each antibody isotype structure and interaction with Fc receptors expressed by the DC culture.
  • FACS were used to evaluate the binding properties of exemplary anti-CD40 antibody IgG variants.
  • CHO cells over-expressing human CD40 or cynomolgus monkey CD40 were harvested using trypsin-EDTA partial digestion followed by centrifugation at 1000 rpm for 5 minutes. The cells were resuspended in cold PBS-BSA (2%) at 5xl0 6 /ml and aliquoted out to lOOul/tube.
  • the chimeric anti-CD40 antibodies were diluted in PBS-BSA in three times (final concentrations were 0.01, 0.1, 1, and 10 ug/ml) and 50ul of each concentration was added to the CHO-CD40 cells. The cell solutions were mixed and incubated at 4°C in the dark for 2 hours.
  • Exemplary anti-CD40 antibodies with various Fc mutants were tested in mouse syngeneic tumor models in vivo to determine how Fc mutants could affect the efficacy and toxicity of these antibodies.
  • C57BL6 mice with human CD40 extracellular domains knock in were used to develop syngeneic mouse tumor models.
  • CD40 antibodies were administered by intraperitoneal injections and tumor sizes were measure during 4-6 weeks of antibody treatment. Tumor sizes were measured by caliber 2 times a week and calculated as tumor volume using formula of 0.5xlengthxwidth 2 .
  • Anti-tumor efficacy was evaluated between tumor sizes of the control group and antibody treatment group.
  • murine IgG2aDANA Fc null
  • Human G4m2, Glm2, G2m20 and G2m40 showed efficacy superior or comparable to mIgG2aDANA.
  • the potential of antibody induced liver toxicity was examined in the mouse model by measuring serum ALT level after antibody treatment.
  • Human G2 showed a very large ALT elevation with its value exceeded upper limit of the assay (>1000 U/L).
  • the efficacious Fc mutants including Glm2, G2m20 and G2m40 showed less or minimal ALT elevation.
  • G2m40 is FcyR null in both binding and DC activation assays.
  • residual FcgRIIB binding activity of G2m20 enhanced efficacy.
  • Fc variants G2ml and G2m43 are expected to have similar efficacy but improved toxicity window given their similar in vitro activity profiles to those of G2m20 and G2m40.
  • inventive embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto, inventive embodiments may be practiced otherwise than as specifically described and claimed.
  • inventive embodiments of the present disclosure are directed to each individual feature, system, article, material, kit, and/or method described herein.
  • a reference to“A and/or B”, when used in conjunction with open-ended language such as“comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
  • “or” should be understood to have the same meaning as“and/or” as defined above.
  • “or” or“and/or” shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as“only one of’ or“exactly one of,” or, when used in the claims,“consisting of,” will refer to the inclusion of exactly one element of a number or list of elements.
  • the phrase“at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements.
  • This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase“at least one” refers, whether related or unrelated to those elements specifically identified.
  • “at least one of A and B” can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
PCT/IB2019/001135 2018-09-28 2019-09-27 Anti-cd40 binding molecules having engineered fc domains and therapeutic uses thereof WO2020065409A2 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
EP19867441.8A EP3856790A4 (en) 2018-09-28 2019-09-27 ANTI-CD40 BINDING MOLECULES WITH MODIFIED CF DOMAINS AND THEIR THERAPEUTIC USES
CN201980064218.4A CN112996818B (zh) 2018-09-28 2019-09-27 具有经过工程化的Fc结构域的抗CD40结合分子及其治疗用途
JP2021517801A JP2022503961A (ja) 2018-09-28 2019-09-27 操作されたFcドメインを有する抗CD40結合分子およびその治療用途
AU2019349874A AU2019349874A1 (en) 2018-09-28 2019-09-27 Anti-CD40 binding molecules having engineered Fc domains and therapeutic uses thereof
KR1020217012243A KR20210070317A (ko) 2018-09-28 2019-09-27 조작된 Fc 도메인을 갖는 항-CD40 결합 분자 및 그의 치료적 용도
CN202210955817.9A CN116178544A (zh) 2018-09-28 2019-09-27 具有经过工程化的Fc结构域的抗CD40结合分子及其治疗用途
CN202210957160.XA CN116063495A (zh) 2018-09-28 2019-09-27 具有经过工程化的Fc结构域的抗CD40结合分子及其治疗用途
CA3114567A CA3114567A1 (en) 2018-09-28 2019-09-27 Anti-cd40 binding molecules having engineered fc domains and therapeutic uses thereof
US17/206,685 US11186648B2 (en) 2018-09-28 2021-03-19 Anti-CD40 antibody having engineered Fc domains and therapeutic uses thereof
US17/536,974 US20220081487A1 (en) 2018-09-28 2021-11-29 Anti-cd40 binding molecules having engineered fc domains and therapeutic uses thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CNPCT/CN2018/108285 2018-09-28
CN2018010828 2018-09-28
CNPCT/CN2018/010828 2018-09-28

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/206,685 Continuation US11186648B2 (en) 2018-09-28 2021-03-19 Anti-CD40 antibody having engineered Fc domains and therapeutic uses thereof

Publications (3)

Publication Number Publication Date
WO2020065409A2 true WO2020065409A2 (en) 2020-04-02
WO2020065409A3 WO2020065409A3 (en) 2020-05-07
WO2020065409A8 WO2020065409A8 (en) 2021-06-17

Family

ID=69950020

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2019/001135 WO2020065409A2 (en) 2018-09-28 2019-09-27 Anti-cd40 binding molecules having engineered fc domains and therapeutic uses thereof

Country Status (4)

Country Link
JP (1) JP2022503961A (ja)
AU (1) AU2019349874A1 (ja)
CA (1) CA3114567A1 (ja)
WO (1) WO2020065409A2 (ja)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022133543A1 (en) * 2020-12-23 2022-06-30 The Macfarlane Burnet Institute For Medical Research And Public Health Ltd Modified immunoglobulin with affinity for fcgammariib and method of use thereof
US11976123B2 (en) 2018-04-20 2024-05-07 Lyvgen Biopharma Holdings Limited Anti-CD40 antibodies and uses thereof

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5580717A (en) 1990-05-01 1996-12-03 Affymax Technologies N.V. Recombinant library screening methods
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
WO1999058572A1 (en) 1998-05-08 1999-11-18 Cambridge University Technical Services Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
US6265150B1 (en) 1995-06-07 2001-07-24 Becton Dickinson & Company Phage antibodies

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008091954A2 (en) * 2007-01-23 2008-07-31 Xencor, Inc. Optimized cd40 antibodies and methods of using the same
RU2745801C2 (ru) * 2015-05-04 2021-04-01 Аподжиникс Аг Одноцепочечные белки-агонисты рецептора cd40
EA201890162A1 (ru) * 2015-06-29 2018-07-31 Бристол-Маерс Сквибб Компани Антитела к cd40 с повышенной агонистической активностью

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5580717A (en) 1990-05-01 1996-12-03 Affymax Technologies N.V. Recombinant library screening methods
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US6265150B1 (en) 1995-06-07 2001-07-24 Becton Dickinson & Company Phage antibodies
WO1999058572A1 (en) 1998-05-08 1999-11-18 Cambridge University Technical Services Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis

Non-Patent Citations (37)

* Cited by examiner, † Cited by third party
Title
"Antibodies: a practical approach", 1988, IRL PRESS
"Cell and Tissue Culture: Laboratory Procedures", 1993, J. WILEY AND SONS
"GenBank", Database accession no. NP_000065.1
"Gene Transfer Vectors for Mammalian Cells", 1987
"Handbook of Experimental Immunology", 1994, ACADEMIC PRESS
"Harrison's Principles of Internal Medicine", 2001, MCGRAW HILL
"Oligonucleotide Synthesis", 1984
"Physician's Desk Reference", 2005
"Remington's The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS AND WILKINS
"The Antibodies", 1995, HARWOOD ACADEMIC PUBLISHERS
"The Merck Manual of Diagnosis and Therapy", 1992, MERCK RESEARCH LABORATORIES
AL-LAZIKANI ET AL., J. MOLEC. BIOL., vol. 273, 1997, pages 927 - 948
ALMAGRO, J. MOL. RECOGNIT., vol. 17, 2004, pages 132 - 143
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 10
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, no. 17, 1997, pages 3389 - 3402
BROWN, M. ET AL., CELL, vol. 49, 1987, pages 603 - 612
BUCK, D. W. ET AL., VITRO, vol. 18, 1982, pages 377 - 381
C. A. JANEWAYP. TRAVERS: "Immunobiology", 1997
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 877
CHOTHIA, C. ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
E. HARLOWD. LANE: "Using antibodies: a laboratory manual", 1999, COLD SPRING HARBOR LABORATORY PRESS
EPSTEIN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 82, 1985, pages 3688
GOSSEN, M.BUJARD, H., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 5547 - 5551
HWANG ET AL., PROC. NATL. ACAD. SCI. USA, vol. 77, 1980, pages 4030
J. P. MATHERP. E. ROBERTS: "Introduction to Cell and Tissue Culture", 1998, ACADEMIC PRESS
KABAT, E.A.: "Sequences of Proteins of Immunological Interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES
KARLINALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 2264 - 68
KARLINALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 5873 - 77
KOHLER, B.MILSTEIN, C., NATURE, vol. 256, 1975, pages 495 - 497
M. GOSSEN ET AL., NATL. ACAD. SCI. USA, vol. 89, 1992, pages 5547 - 5551
MCCAFFERTY ET AL., NATURE, vol. 348, 1990, pages 552 - 553
QUEEN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 10029 - 10033
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
SHOCKELT, P. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 92, 1995, pages 6522 - 6526
WINTER ET AL., ANNU. REV. IMMUNOL., vol. 12, 1994, pages 433 - 455
YAO ET AL., HUMAN GENE THERAPY
YAO, F. ET AL., HUMAN GENE THERAPY, vol. 9, 1998, pages 1939 - 1950

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11976123B2 (en) 2018-04-20 2024-05-07 Lyvgen Biopharma Holdings Limited Anti-CD40 antibodies and uses thereof
WO2022133543A1 (en) * 2020-12-23 2022-06-30 The Macfarlane Burnet Institute For Medical Research And Public Health Ltd Modified immunoglobulin with affinity for fcgammariib and method of use thereof

Also Published As

Publication number Publication date
JP2022503961A (ja) 2022-01-12
WO2020065409A8 (en) 2021-06-17
WO2020065409A3 (en) 2020-05-07
AU2019349874A1 (en) 2021-04-29
CA3114567A1 (en) 2020-04-02

Similar Documents

Publication Publication Date Title
US20220372155A1 (en) Anti-cd40 binding molecules and bi-specific antibodies comprising such
WO2020069382A1 (en) Anti-cd137 binding molecules having engineered fc domains and therapeutic uses thereof
US11505615B2 (en) Anti-CD137 antibodies and uses thereof
US20220081487A1 (en) Anti-cd40 binding molecules having engineered fc domains and therapeutic uses thereof
US20200392227A1 (en) Therapeutic agents and methods for enhancing immune responses in tumor microenvironment
AU2019349874A1 (en) Anti-CD40 binding molecules having engineered Fc domains and therapeutic uses thereof
US11976123B2 (en) Anti-CD40 antibodies and uses thereof
US20230406950A1 (en) Bi-specific antibodies comprising anti-cd137 binding molecules

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: 19867441

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2021517801

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3114567

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20217012243

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019349874

Country of ref document: AU

Date of ref document: 20190927

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019867441

Country of ref document: EP

Effective date: 20210428