WO2020207470A1 - 抗cd40抗体及其用途 - Google Patents
抗cd40抗体及其用途 Download PDFInfo
- Publication number
- WO2020207470A1 WO2020207470A1 PCT/CN2020/084186 CN2020084186W WO2020207470A1 WO 2020207470 A1 WO2020207470 A1 WO 2020207470A1 CN 2020084186 W CN2020084186 W CN 2020084186W WO 2020207470 A1 WO2020207470 A1 WO 2020207470A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- seq
- amino acid
- antibody
- acid sequence
- cells
- Prior art date
Links
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2878—Immunoglobulins [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
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39533—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
- A61K39/3955—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/21—Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/24—Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/33—Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
- C07K2317/565—Complementarity determining region [CDR]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/72—Increased effector function due to an Fc-modification
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
- C07K2317/732—Antibody-dependent cellular cytotoxicity [ADCC]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/74—Inducing cell proliferation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/75—Agonist effect on antigen
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
- C07K2317/92—Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
Definitions
- the present invention relates to novel antibodies and antibody fragments that specifically bind to CD40, and compositions containing the antibodies or antibody fragments, as well as medicines, combination products or kits.
- the present invention relates to nucleic acids encoding the antibodies or antibody fragments thereof, host cells containing them, and related uses.
- the present invention relates to the therapeutic and diagnostic uses of these antibodies and antibody fragments.
- T cells The complete activation of T cells requires two signals: antigen-presenting cells (APC) uptake and process tumor antigens to form MHC-antigen complexes, which are presented to T cells and combined with TCR on the surface of T cells.
- APC antigen-presenting cells
- the first signal; the second or costimulatory signal is transmitted through the interaction between CD28 and B7-1(CD80)/B7-2(CD86), as well as the costimulatory factors CD40, OX40, GITR, etc. and their ligands (Smith-Garvin , JE, GAKoretzky, and MSJordan, T cell activation. Annu Rev Immunol, 2009.27: p.591-619).
- T cells may undergo non-responsiveness (anergy) or programmed cell death (apoptosis) after antigen stimulation.
- CD40 is a member of the TNF receptor (TNFR) superfamily. It is mainly expressed on B cells, dendritic cells (DC cells), monocytes and macrophages and other antigen-presenting cells (Grewal, ISand RAFlavell, CD40 and CD154 in cell-mediated immunity. Annu Rev Immunol, 1998.16: p.111-35). CD40 forms a trimer on the cell surface, and the corresponding ligand CD40L (ie CD154) is mainly expressed on the surface of activated T cells. The interaction of CD40 and CD40L is a costimulatory signal for T cell activation. Depending on the specific cell type, CD40 participates in the role leading to specific gene expression patterns.
- TNFR TNF receptor
- CD40L and CD40 on T cells can activate a variety of pathways, including NF- ⁇ B (nuclear factor ⁇ B), MAPK (mitogen-activated protein kinase) and STAT3 (signal transducer and activator of transcription 3), etc.
- NF- ⁇ B nuclear factor ⁇ B
- MAPK mitogen-activated protein kinase
- STAT3 signal transducer and activator of transcription 3
- CD40 is not only expressed by normal immune cells, but also by many malignant cells. Specifically, CD40 is overexpressed in the following diseases: B-line NHL, chronic lymphocytic leukemia (CLL), hairy cell leukemia (HCL), Hodgkin’s disease, multiple myeloma and bladder cancer, kidney cancer, ovarian cancer , Cervical cancer, breast cancer, lung cancer, nasopharyngeal cancer and malignant melanoma (Hassan, SB, et al., Anti-CD40-mediated cancer immunotherapy: an update of recent and longoing clinical trials.ImmunopharmacolImmunotoxicol, 2014.36(2) : P.96-104).
- CLL chronic lymphocytic leukemia
- HCL hairy cell leukemia
- Hodgkin’s disease multiple myeloma and bladder cancer
- kidney cancer ovarian cancer
- Cervical cancer breast cancer
- lung cancer nasopharyngeal cancer
- CD40 agonist antibodies can fight tumor cells through a variety of mechanisms: First, CD40 agonist antibodies mediate stronger anti-tumor effects by activating the immune system. Specifically, CD40 agonist antibodies can activate DC cells to increase their antigen presentation ability, which is manifested in the increased expression of costimulatory molecules, such as the B7 family (CD80, CD86), and the promotion of cytokine secretion, such as interleukin 12. This will lead to a significant T cell response (Fong, L. and EG Engleman, Dendritic cells in cancer immunotherapy.
- costimulatory molecules such as the B7 family (CD80, CD86)
- cytokine secretion such as interleukin 12. This will lead to a significant T cell response (Fong, L. and EG Engleman, Dendritic cells in cancer immunotherapy.
- CD40 agonist antibodies can promote the proliferation and immunity of resting B cells Globulin class changes, antibody secretion, and have an impact on the development of germinal centers and the survival of memory B cells, all of which are essential for humoral immune response (Beatty, GL, Y. Li, and KBLong, Cancer immunotherapy: activating innate and adaptive immunity through CD40 agonists. Expert Rev Anticancer Ther, 2017.17(2): p.175-186).
- CD40 agonist antibody binds to CD40 expressed on the surface of tumor cells, it mediates antibody-dependent cytotoxicity (ADCC), and killer cells directly eliminate tumor cells with high CD40 expression (Vonderheide, RHand MJ Glennie, Agonistic CD40). antibodies and cancer therapy. Clin Cancer Res, 2013.19(5): p.1035-43).
- CD40 agonist antibody binds to CD40 expressed on the surface of tumor cells, it directly inhibits tumor growth and promotes its apoptosis.
- the CD40/CD40L signaling pathway can block the cell cycle of tumor cells and stop cells at G2-M
- the interaction of CD40/CD40L can also promote the increase of Fas expression on the surface of tumor cells, and inhibit the growth of high-expressing CD40 tumor cells and promote their apoptosis through the Fas/FasL signaling pathway (Eliopoulos, AG, et al., CD40 induces apoptosis in carcinoma) Cells through activation of cytotoxic ligands of the tumor necrosis factor superfamily. Mol Cell Biol, 2000.20(15): p.5503-15).
- CD40 agonist antibodies can be divided into two types according to their different agonistic methods.
- the first type of CD40 agonist activity does not depend on cross-linking of Fc receptors (such as CP-870893 and CDX-1140), although the former shows encouraging anti-cancer activity Efficacy, but there is a dose-limiting toxicity, which can lead to systemic immune disorders, venous thromboembolism and cytokine release syndrome (Cytokine Release Syndrome, CRS); the other type requires Fc receptor cross-linking to have CD40 Agonistic activity.
- Fc receptors such as CP-870893 and CDX-1140
- CRS Cytokine Release Syndrome
- CD40 antibodies in the field, such as Pfizer’s CP870893, there is still a need for some new CD40 antibodies with properties comparable to or better than existing antibodies, especially CD40 that relies on Fc receptor cross-linking.
- Antibodies especially antibodies with better anti-cancer properties and higher safety.
- the present invention therefore provides a new antibody that binds to CD40 (especially human CD40 or rhesus CD40), and antigen-binding fragments thereof.
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises a heavy chain variable region (VH), wherein the VH comprises
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises a light chain variable region (VL), wherein the VL comprises:
- LCDR3 contains at least one and no more than 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably conservative substitutions).
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises a heavy chain variable region VH and a light chain variable region VL, wherein
- the VH includes
- HCDR3 contains at least one and no more than 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably conservative substitutions);
- the VL includes:
- LCDR3 contains at least one and no more than 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably conservative substitutions).
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises a heavy chain variable region VH and/or a light chain variable region VL, wherein
- the VH includes
- HCDR3 contains at least one and no more than 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably conservative substitutions);
- the VL includes:
- LCDR3 contains at least one and no more than 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably conservative substitutions).
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises a heavy chain variable region VH and a light chain variable region VL, wherein
- the VH includes
- HCDR3 contains at least one and no more than 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably conservative substitutions);
- the VL includes:
- LCDR3 contains at least one and no more than 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably conservative substitutions).
- the VH comprises an amino acid sequence selected from SEQ ID NO: 13, 58, 60, 62, or 14, or consists of the amino acid sequence.
- VL comprises an amino acid sequence selected from SEQ ID NO: 15, 64, 66 or 16, or consists of said amino acid sequence.
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises
- the three complementary regions determine the LCDR region.
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises:
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises a heavy chain variable region (VH) and/or a light chain variable region (VL), wherein
- the VH includes complementarity determining regions (CDRs) HCDR1, HCDR2, and HCDR3, wherein HCDR1 includes the amino acid sequence of SEQ ID NO: 1 or 2, or consists of the amino acid sequence, or HCDR1 includes the amino acid sequence of SEQ ID NO: 1.
- CDRs complementarity determining regions
- amino acid sequence of 2 has one, two or three changes (preferably amino acid substitutions, preferably conservative substitutions) amino acid sequence
- HCDR2 includes the amino acid sequence of SEQ ID NO: 3 or 4, or consists of the amino acid sequence
- HCDR2 includes an amino acid sequence with one, two or three changes (preferably amino acid substitutions, preferably conservative substitutions) compared to the amino acid sequence selected from SEQ ID NO: 3 or 4
- HCDR3 includes SEQ ID NOs: 5, 51, The amino acid sequence of 52, 55 or 6 or consists of the amino acid sequence, or HCDR3 contains one, two or three changes (preferably amino acid sequence) compared with the amino acid sequence of SEQ ID NO: 5, 51, 52, 55 or 6. Substitution, preferably conservative substitution) amino acid sequence;
- VL includes complementarity determining regions (CDR) LCDR1, LCDR2 and LCDR3, where LCDR1 includes or consists of the amino acid sequence of SEQ ID NO: 7 or 8, or LCDR1 includes the same as SEQ ID NO: 7.
- CDR complementarity determining regions
- the amino acid sequence of 8 has one, two or three changes (preferably amino acid substitutions, preferably conservative substitutions) amino acid sequence;
- LCDR2 comprises or consists of the amino acid sequence of SEQ ID NO: 9 or 10, or LCDR2 includes an amino acid sequence with one, two or three changes (preferably amino acid substitutions, preferably conservative substitutions) compared with the amino acid sequence of SEQ ID NO: 9 or 10;
- LCDR3 includes an amino acid sequence selected from SEQ ID NOs: 11, 53, 54
- the amino acid sequence of, 56 or 12 or consists of the amino acid sequence, or LCDR3 contains one, two or three changes compared with the amino acid sequence of SEQ ID NO: 11, 53, 54, 56 or 12 (preferably amino acid substitution , Preferably conservative substitution) amino acid sequence.
- the present invention provides an anti-CD40 antibody or antigen-binding fragment thereof, which comprises a heavy chain variable region (VH) and a light chain variable region (VL), wherein
- the VH includes
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO: 1, SEQ ID NO: 3 and SEQ ID NO: 5; or
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO: 1, SEQ ID NO: 3 and SEQ ID NO: 51; or
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO: 1, SEQ ID NO: 3 and SEQ ID NO: 52; or
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO: 2, SEQ ID NO: 4 and SEQ ID NO: 6; or
- the VL includes
- LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequence: SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 11; or
- LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequence: SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 53; or
- LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequences: SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 54; or
- LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequences: SEQ ID NO: 8, SEQ ID NO: 10 and SEQ ID NO: 12; or
- the present invention provides an anti-CD40 antibody or antigen-binding fragment thereof, which comprises a heavy chain variable region (VH) and a light chain variable region (VL), wherein
- the VH includes
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO: 1, SEQ ID NO: 3 and SEQ ID NO: 5; or
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO: 1, SEQ ID NO: 3 and SEQ ID NO: 51; or
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO: 1, SEQ ID NO: 3 and SEQ ID NO: 52; or
- the VL includes
- LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequence: SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 11; or
- LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequence: SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 53; or
- LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequences: SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 54; or
- the present invention provides an anti-CD40 antibody or antigen-binding fragment thereof, which comprises a heavy chain variable region (VH) and a light chain variable region (VL), wherein
- the VH includes
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO: 2, SEQ ID NO: 4 and SEQ ID NO: 6; or
- the VL includes
- LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequences: SEQ ID NO: 8, SEQ ID NO: 10 and SEQ ID NO: 12; or
- the present invention provides an anti-CD40 antibody or antigen-binding fragment thereof, which comprises
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO:1, SEQ ID NO: 3 and SEQ ID NO: 5, and LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequences: SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 11; or
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO:1, SEQ ID NO: 3 and SEQ ID NO: 51, and LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequences: SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 53; or
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO:1, SEQ ID NO: 3 and SEQ ID NO: 52, and/or LCDR1, LCDR2 and LCDR2 and LCDR3: SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 54; or
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO: 2, SEQ ID NO: 4 and SEQ ID NO: 6, and LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequences: SEQ ID NO: 8, SEQ ID NO: 10 and SEQ ID NO: 12;
- the present invention provides an anti-CD40 antibody or antigen-binding fragment thereof, which comprises a heavy chain variable region (VH) and a light chain variable region (VL), wherein
- the VH includes
- HCDR1, HCDR2 and HCDR3 comprising or consisting of the following sequences: SEQ ID NO: 1, SEQ ID NO: 3 and SEQ ID NO: 55; or
- the VL includes
- LCDR1, LCDR2 and LCDR3 comprising or consisting of the following sequences: SEQ ID NO: 7, SEQ ID NO: 9 and SEQ ID NO: 56; or
- amino acid sequence shown in SEQ ID NO: 55 is as follows:
- ARERVGA-X1-PTYYY-X2-X3-DV wherein X1, X2 or X3 can be any amino acid, preferably, wherein X1 can be T, N, W, K, A or Y, more preferably T or N; wherein X2 Preferably it is W or Y; wherein X3 can be W, Y, M, F, T, more preferably M, W or Y.
- amino acid sequence shown in SEQ ID NO: 56 is as follows:
- X1, X2, X3 and X4 can be any amino acids, preferably, where X1 can be Q, N or P, more preferably Q or N; X2 can Is G, Q, F, S, Y or M; more preferably G or Q; X3 can be E, N, T, S or K; more preferably E or N; X4 can be T, Q, V , L, P or E; more preferably T, Q or V.
- the present invention provides an anti-CD40 antibody or antigen-binding fragment thereof, which comprises a heavy chain variable region (VH) and a light chain variable region (VL), wherein the VH comprises a complementarity determining region (CDR) HCDR1, HCDR2 and HCDR3 and said VL comprises (CDR) LCDR1, LCDR2 and LCDR3, wherein the combination of HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3 contained in said antibody or antigen-binding fragment thereof is as follows (Table A) Shown:
- Table A Exemplary combinations of HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3 in the antibody of the present invention or its antigen-binding fragment
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises a heavy chain variable region VH and/or a light chain variable region VL, wherein,
- (iii) Comprising one or more (preferably not more than 10, more preferably not more than 5, 4, 3, 2, and an amino acid sequence selected from SEQ ID NO: 13, 58, 60, 62 or 14). 1) the amino acid sequence of the amino acid change (preferably amino acid substitution, more preferably amino acid conservative substitution), preferably, the amino acid change does not occur in the CDR region, preferably, the amino acid change occurs in the FR region;
- amino acid sequence comprising SEQ ID NO: 15, 64, 66 or 16 has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical amino acid sequence or consisting of it;
- (iii) Comprising one or more (preferably no more than 10, more preferably no more than 5, 4, 3, 2, 1) compared with an amino acid sequence selected from SEQ ID NO: 15, 64, 66 or 16.
- the amino acid change does not occur in the CDR region, and preferably, the amino acid change occurs in the FR region.
- the heavy chain variable region VH of the anti-CD40 antibody or antigen-binding fragment thereof of the present invention is provided.
- amino acid sequence comprising SEQ ID NO: 13, 58, 60, or 62 has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical amino acid sequence or consisting of it; or
- (iii) Comprising one or more (preferably not more than 10, more preferably not more than 5, 4, 3, 2, 1) compared with an amino acid sequence selected from SEQ ID NO: 13, 58, 60 or 62
- the amino acid change does not occur in the CDR region, and preferably, the amino acid change occurs in the FR region.
- the light chain variable region VL of the anti-CD40 antibody or antigen-binding fragment thereof of the present invention is provided.
- amino acid sequence comprising SEQ ID NO: 15, 64 or 66 has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%
- the heavy chain variable region VH of the anti-CD40 antibody or antigen-binding fragment thereof of the present invention is provided.
- amino acid changes preferably Amino acid substitutions, more preferably conservative substitutions of amino acids
- the amino acid changes do not occur in the CDR region, preferably, the amino acid changes occur in the FR region.
- the light chain variable region VL of the anti-CD40 antibody or antigen-binding fragment thereof of the present invention is provided.
- amino acid changes (preferably not more than 5, 4, 3, 2, 1) compared with the amino acid sequence selected from SEQ ID NO: 16 Amino acid substitutions, more preferably conservative substitutions of amino acids), preferably, the amino acid changes do not occur in the CDR region, preferably, the amino acid changes occur in the FR region.
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises a heavy chain variable region VH and/or a light chain variable region VL, wherein,
- the heavy chain variable region VH includes or consists of the amino acid sequence of SEQ ID NO: 13, and the light chain variable region VL includes or consists of the amino acid sequence of SEQ ID NO: 15;
- the heavy chain variable region VH comprises or consists of the amino acid sequence of SEQ ID NO: 58
- the light chain variable region VL comprises or consists of the amino acid sequence of SEQ ID NO: 64;
- the heavy chain variable region VH comprises or consists of the amino acid sequence of SEQ ID NO: 60
- the light chain variable region VL comprises or consists of the amino acid sequence of SEQ ID NO: 66;
- the heavy chain variable region VH includes or consists of the amino acid sequence of SEQ ID NO: 62, and the light chain variable region VL includes or consists of the amino acid sequence of SEQ ID NO: 66;
- the heavy chain variable region VH includes or consists of the amino acid sequence of SEQ ID NO: 14, and the light chain variable region VL includes or consists of the amino acid sequence of SEQ ID NO: 16.
- the amino acid sequence of the heavy chain variable region of the antibody of the present invention has at least 90%, 91%, 92% of the amino acid sequence of the heavy chain variable region in (i)-(v) described above. %, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity, or its amino acid sequence is compared with the above-mentioned (i)-(v) heavy chain variable region Amino acid sequence, comprising one or more (preferably not more than 10, more preferably not more than 5, 4, 3, 2, 1) amino acid changes (preferably amino acid substitutions, more preferably amino acid conservative substitutions) amino acid sequence, preferably Preferably, the amino acid change does not occur in the CDR region, and preferably, the amino acid change occurs in the FR region.
- the amino acid sequence of the light chain variable region of the antibody of the present invention and the amino acid sequence of the light chain variable region in (i)-(v) described above have at least 90%, 91%, 92%. %, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity, or its amino acid sequence is compared with the light chain variable region in (i)-(v) described above Amino acid sequence, comprising one or more (preferably not more than 10, more preferably not more than 5, 4, 3, 2, 1) amino acid changes (preferably amino acid substitutions, more preferably amino acid conservative substitutions) amino acid sequence, preferably Preferably, the amino acid change does not occur in the CDR region, and preferably, the amino acid change occurs in the FR region.
- the present invention provides an anti-CD40 antibody or antigen-binding fragment thereof, which comprises a heavy chain variable region (VH) and a light chain variable region (VL), wherein the antibody or antigen-binding fragment thereof comprises
- VH heavy chain variable region
- VL light chain variable region
- Table B Exemplary combinations of heavy chain variable region VH and light chain variable region VL in the antibody or antigen-binding fragment of the present invention
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises a heavy chain and/or light chain, wherein
- amino acid sequence comprising SEQ ID NO: 17, 18, 19, 67, 69, 70, or 20 has at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical amino acid sequence or consisting of;
- amino acid sequence of amino acid changes (preferably amino acid substitutions, more preferably amino acid conservative substitutions), preferably, the amino acid changes do not occur in the CDR region of the heavy chain, more preferably, The amino acid change does not occur in the variable region of the heavy chain, and most preferably, the amino acid change occurs in the constant region of the heavy chain;
- amino acid sequence comprising SEQ ID NO: 21, 68, 71, or 22 has at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical amino acid sequence or consisting of it;
- (iii) Comprising one or more (preferably not more than 20 or 10, more preferably not more than 5, 4, 3, 2) compared with an amino acid sequence selected from SEQ ID NO: 21, 68, 71 or 22 , 1) amino acid change (preferably amino acid substitution, more preferably amino acid conservative substitution) amino acid sequence, preferably, the amino acid change does not occur in the CDR region of the light chain, more preferably, the amino acid change does not occur in In the light chain variable region, most preferably, the heavy chain amino acid changes occur in the light chain constant region.
- amino acid change preferably amino acid substitution, more preferably amino acid conservative substitution
- the heavy chain contained in the anti-CD40 antibody or antigen-binding fragment thereof of the present invention is the heavy chain contained in the anti-CD40 antibody or antigen-binding fragment thereof of the present invention.
- amino acid sequence comprising and being selected from SEQ ID NO: 17, 18, 19, 67, 69 or 70 has at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96% , 97%, 98%, or 99% identical amino acid sequence or consisting of it;
- (iii) Comprising one or more (preferably not more than 20 or 10, more preferably not more than 5, 4) compared with an amino acid sequence selected from SEQ ID NO: 17, 18, 19, 67, 69 or 70 , 3, 2, 1) amino acid sequence of amino acid changes (preferably amino acid substitutions, more preferably amino acid conservative substitutions), preferably, the amino acid changes do not occur in the CDR region of the heavy chain, more preferably, the amino acid The change does not occur in the variable region of the heavy chain, and most preferably, the amino acid change of the heavy chain occurs in the constant region of the heavy chain.
- amino acid sequence of amino acid changes preferably amino acid substitutions, more preferably amino acid conservative substitutions
- the light chain contained in the anti-CD40 antibody or antigen-binding fragment thereof of the present invention is the light chain contained in the anti-CD40 antibody or antigen-binding fragment thereof of the present invention.
- amino acid sequence comprising and selected from SEQ ID NO: 21, 68 or 71 has at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% Or 99% identical amino acid sequence or consisting of it;
- (iii) Comprising one or more (preferably not more than 20 or 10, more preferably not more than 5, 4, 3, 2, 1) compared with an amino acid sequence selected from SEQ ID NO: 21, 68 or 71 (Preferably amino acid substitution, more preferably amino acid conservative substitution) amino acid sequence, preferably, the amino acid change does not occur in the CDR region of the light chain, more preferably, the amino acid change does not occur in the light chain In the variable region, most preferably, the heavy chain amino acid changes occur in the light chain constant region.
- the heavy chain contained in the anti-CD40 antibody or antigen-binding fragment thereof of the present invention is the heavy chain contained in the anti-CD40 antibody or antigen-binding fragment thereof of the present invention.
- (iii) Comprising one or more (preferably not more than 20 or 10, more preferably not more than 5, 4, 3, 2, 1) amino acids compared to the amino acid sequence selected from SEQ ID NO: 20 Change the amino acid sequence (preferably amino acid substitution, more preferably amino acid conservative substitution), preferably, the amino acid change does not occur in the CDR region of the heavy chain, more preferably, the amino acid change does not occur in the heavy chain variable region Most preferably, the heavy chain amino acid changes occur in the constant region of the heavy chain.
- the light chain contained in the anti-CD40 antibody or antigen-binding fragment thereof of the present invention is the light chain contained in the anti-CD40 antibody or antigen-binding fragment thereof of the present invention.
- (iii) Comprising one or more (preferably not more than 20 or 10, more preferably not more than 5, 4, 3, 2, 1) amino acids compared to the amino acid sequence selected from SEQ ID NO: 22 Change the amino acid sequence (preferably amino acid substitution, more preferably amino acid conservative substitution), preferably, the amino acid change does not occur in the CDR region of the light chain, more preferably, the amino acid change does not occur in the light chain variable region Most preferably, the amino acid change of the heavy chain occurs in the constant region of the light chain.
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention comprises a heavy chain and/or light chain, wherein,
- the heavy chain comprises or consists of the amino acid sequence of SEQ ID NO: 17, 18, 19, 67, 69 or 70
- the light chain comprises or consists of the amino acid sequence of SEQ ID NO: 21, 68 or 71
- the heavy chain comprises or consists of the amino acid sequence of SEQ ID NO: 17, 18 or 19, and the light chain comprises or consists of the amino acid sequence of SEQ ID NO: 21;
- the heavy chain includes or consists of the amino acid sequence of SEQ ID NO: 67, and the light chain includes or consists of the amino acid sequence of SEQ ID NO: 68;
- the heavy chain comprises or consists of the amino acid sequence of SEQ ID NO: 69 or 70, and the light chain comprises or consists of the amino acid sequence of SEQ ID NO: 71;
- the heavy chain includes or consists of the amino acid sequence of SEQ ID NO: 20, and the light chain includes or consists of the amino acid sequence of SEQ ID NO: 22.
- the amino acid sequence of the heavy chain of the antibody of the present invention is at least 85%, 90%, 91%, 92%, 93%, 94% of the amino acid sequence of the heavy chain in (i)-(v) above. , 95%, 96%, 97%, 98%, or 99% identity, or its amino acid sequence is compared with the heavy chain amino acid sequence in (i)-(v) above, and contains 1 or more (preferably not more than 20 or 10, more preferably no more than 5, 4, 3, 2, 1) amino acid changes (preferably amino acid substitutions, more preferably amino acid conservative substitutions) amino acid sequence, preferably, the amino acid changes do not occur in the heavy In the CDR region of the chain, more preferably, the amino acid change does not occur in the variable region of the heavy chain, and most preferably, the amino acid change occurs in the constant region of the heavy chain.
- the amino acid sequence of the light chain of the antibody of the present invention is at least 85%, 90%, 91%, 92%, 93%, 94% of the amino acid sequence of the heavy chain in (i)-(v) above. , 95%, 96%, 97%, 98%, or 99% identity, or its amino acid sequence is compared with the light chain amino acid sequence in (i)-(v) above, and contains one or more (preferably not more than 20 or 10, more preferably no more than 5, 4, 3, 2, 1) amino acid changes (preferably amino acid substitutions, more preferably amino acid conservative substitutions) amino acid sequence, preferably, the amino acid changes do not occur in light In the CDR region of the chain, more preferably, the amino acid change does not occur in the variable region of the light chain, and most preferably, the amino acid change occurs in the constant region of the light chain.
- the present invention provides an anti-CD40 antibody or antigen-binding fragment thereof, which comprises a heavy chain and a light chain, wherein the combination of the heavy chain and light chain contained in the antibody or the antigen-binding fragment thereof is as follows (Table C) shows:
- Table C Exemplary combinations of heavy and light chains in the antibodies of the invention or antigen-binding fragments thereof
- the heavy chain and/or light chain of the anti-CD40 antibody or fragment thereof of the present invention further comprises a signal peptide sequence, for example, the signal peptide sequence comprises or consists of the amino acid sequence shown in SEQ ID NO: 43.
- the amino acid changes described herein include amino acid substitutions, insertions or deletions.
- the amino acid changes described herein are amino acid substitutions, preferably conservative substitutions.
- the changes occur in the CDR regions (especially CDR3 regions) of the heavy and/or light chains of the antibody. In some embodiments, there may be 1, 2, or 3 changes in the CDR regions, such as the CDR3 region.
- the amino acid changes described in the present invention occur in regions outside the CDR (for example, in FR).
- the change occurs in the FR region of the antibody, such as the FR region of the antibody heavy chain and/or light chain variable region, such as FR1, FR2, FR2, or F4.
- the change occurs in the FR2 region.
- amino acid changes described in the present invention occur in regions outside the variable region of the heavy chain and/or outside the variable region of the light chain, for example, in the constant region of the heavy chain and/or light chain.
- substitutions are conservative substitutions.
- Conservative substitution refers to the replacement of an amino acid by another amino acid in the same category, for example, an acidic amino acid is replaced by another acidic amino acid, a basic amino acid is replaced by another basic amino acid, or a neutral amino acid is replaced by another neutral amino acid. Replacement. Exemplary substitutions are shown in Table D below:
- the substitution occurs in the CDR region of the antibody.
- the obtained variant has a modification (e.g., improvement) in certain biological properties (e.g., increased affinity) relative to the parent antibody and/or will have certain biological properties substantially retained of the parent antibody.
- An exemplary substitution variant is an affinity matured antibody.
- the substitution occurs in the CDR3 region of the heavy chain and/or light chain of the antibody. In some embodiments, there may be 1, 2, or 3 substitutions in the CDR3 region.
- the substitution occurs in the FR region of the antibody, such as the FR region of the antibody heavy chain and/or light chain variable region, such as FR1, FR2, FR2, or F4. In some embodiments, the substitution occurs in the FR2 region. In some embodiments, there may be 1, 2, or 3 substitutions in the FR region.
- the invention provides antibodies comprising the variable region sequence disclosed herein or the variable region sequence having the CDR disclosed herein, and the constant domain of a modified Fc region, the modified Fc Compared to its affinity for other Fc receptors (ie, activated receptors), the region has an enhanced affinity for Fc ⁇ RIIb.
- modified Fc region Compared to its affinity for other Fc receptors (ie, activated receptors), the region has an enhanced affinity for Fc ⁇ RIIb.
- Such agonistic anti-CD40 antibodies with enhanced Fc ⁇ RIIb specificity are expected to exhibit excellent efficacy in cancer treatment and chronic infections (Li and Ravetch (2011) Science 333: 1030; White et al. (2011) J. Immunol. 187 : 1754).
- Fc ⁇ RIIb-specific agonistic anti-CD40 antibodies can exhibit enhanced auxiliary effects by increasing the proliferation of cytotoxic CD8 + T cells and the maturation of activated dendritic cells, leading to enhanced anti-tumor responses .
- the signal enhancement of the FcR-mediated agonist CD40 antibody due to the cross-linking of the present invention can be a major contributor to therapeutic efficacy.
- Cross-linking of FcR-conjugated CD40 agonist antibodies through the Fc portion of the antibody can increase signal strength and thereby enhance cell activation.
- Exemplary mutations in the Fc sequence include, for example, E233D, G237D, H268D, P271G, A330R, S267E, and/or L328F.
- the antibody of the present invention comprises a human IgG1 constant domain with mutations that enhance the specificity of FcyRIIb, the mutations including E233D, G237D, H268D, P271G and A330R, or S267E and L328F. See, for example, SEQ ID NO: 18 or 19 for the sequence of an exemplary antibody heavy chain comprising a mutated human IgG1 constant domain that enhances Fc ⁇ RIIb specificity.
- the antibodies provided herein can be further modified to contain other non-protein moieties known and readily available in the art.
- the part suitable for antibody derivatization includes, but is not limited to, water-soluble polymers.
- water-soluble polymers include, but are not limited to, polyethylene glycol (PEG), ethylene glycol/propylene glycol copolymer, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinylpyrrolidone, poly -1,3-dioxane, poly-1,3,6-trioxane, ethylene/maleic anhydride copolymer, polyamino acid (homopolymer or random copolymer), and dextran or poly(n-ethylene Pyrrolidone) polyethylene glycol, propylene glycol homopolymer, polypropylene oxide/ethylene oxide copolymer, polyoxyethylated polyol (such as glycerin), polyvinyl alcohol, and mixtures thereof.
- PEG polyethylene glycol
- the antibodies provided herein are modified to increase or decrease the degree to which the antibody is glycosylated.
- the addition or deletion of glycosylation sites of the antibody can be conveniently achieved by changing the amino acid sequence to create or remove one or more glycosylation sites.
- the carbohydrate attached to it can be changed.
- modifications to remove unwanted glycosylation sites may be useful, such as removing fucose moieties to improve antibody-dependent cellular cytotoxicity (ADCC) functions (see Shield et al. (2002) JBC277:26733 ).
- ADCC antibody-dependent cellular cytotoxicity
- galactosidation can be modified to modify complement dependent cytotoxicity (CDC).
- cysteine engineered antibody such as a "thioMAb"
- Cysteine engineered antibodies can be generated as described, for example, in U.S. Patent No. 7,521,541.
- the anti-CD40 antibody or antigen-binding fragment thereof of the present invention shows the same or similar binding affinity and/or specificity as the antibody of the present invention for CD40; and/or inhibits (eg, competitively inhibits) the antibody of the present invention It binds to CD40 and/or binds to the same or overlapping epitope with the antibody of the present invention; and/or competes with the antibody of the present invention for binding to CD40; and/or has one or more biological characteristics of the antibody of the present invention.
- the anti-CD40 antibody of the present invention is an antibody in the form of IgG1 or an antibody in the form of IgG2 or an antibody in the form of IgG3 or an antibody in the form of IgG4.
- the anti-CD40 antibody is a monoclonal antibody.
- the anti-CD40 antibody is humanized.
- Different methods for humanizing antibodies are known to the skilled person, as reviewed by Almagro & Fransson, the contents of which are fully incorporated herein by reference (Almagro JC and Fransson J (2008) Frontiers in Bioscience 13: 1619-1633).
- the anti-CD40 antibody is a human antibody.
- Human antibodies are generally described in van Dijk and van de Winkel, Curr. Opin. Pharmacol 5: 368-74 (2001) and Lonberg, Curr. Opin. Immunol 20: 450-459 (2008).
- the anti-CD40 antibody is a chimeric antibody.
- the anti-CD40 antibody of the present invention also encompasses its antibody fragments, preferably antibody fragments selected from the group consisting of Fab, Fab', Fab'-SH, Fv, single chain antibodies (such as scFv) or (Fab' ) 2. Single domain antibody, diabody (dAb) or linear antibody.
- the anti-CD40 antibody molecule is in the form of a bispecific or multispecific antibody molecule.
- the bispecific antibody molecule has a first binding specificity for CD40 and a second binding specificity for PD-1 or PD-L1 or PD-L2 or OX40 or 4-1BB or GITR or the like.
- the bispecific antibody molecule binds to CD40 and TNF or IL-17.
- Multispecific antibody molecules can have any combination of binding specificities for the aforementioned molecules.
- the antibodies of the invention have one or more of the following characteristics:
- CD40-expressing cells in a cross-linked or constitutive form, preferably in a cross-linked form, such as activating the NFkappa-B signaling pathway; in some embodiments, flow cytometry is used for detection; in some embodiments
- the reporter cells used are cells expressing NF- ⁇ B-GFP and hCD40;
- its equilibrium dissociation constant is less than or equal to about 5 ⁇ 10 -7 M, 4.5 ⁇ 10 -7 M, 4.4 ⁇ 10 -7 M, 4.3 ⁇ 10 -7 M, 4.2 ⁇ 10 -7 M, 4.1 ⁇ 10 -7 M, 4 ⁇ 10 -7 M, 3.9 ⁇ 10 -7 M, 3.8 ⁇ 10 -7 M, 3.7 ⁇ 10 -7 M, 3.6 ⁇ 10 -7 M, 3.5 ⁇ 10 -7 M, or 3.4 ⁇ 10 -7 M; in some embodiments, the measurement is a surface plasmon resonance technique.
- binding to CD40 on cells expressing CD40 is less than or equal to about 50nM, 40nM, 30nM, 20nM, 15nM, 14nM, 13nM, 12nM, 10nM, 9nM, 8nM.
- flow cytometry is used in combination; in some embodiments, the CD40-expressing cells are 293 cells, such as 293FT cells.
- the EC50 is less than or equal to about 4nM, 3.5nM, 3nM, 2.9nM, 2.8nM, 2.7nM, or 2.6nM; in some embodiments, it is measured using flow cytometry.
- the tumor cell is a Raji cell or Ramos cell.
- Having agonist activity such as significantly activating (e.g., human) B cells or T cells or dendritic cells.
- the CD40 antibody has one or more of the aforementioned properties when cross-linked.
- the present invention also encompasses antibodies conjugated to other substances ("immunoconjugates").
- Cytotoxic agents include any agents that are harmful to cells. Examples of cytotoxic agents (such as chemotherapeutics) or other substances suitable for forming immunoconjugates are known in the art, see, for example, WO2017/004006 or WO2017/059243.
- the tag is for example a tag sequence, such as a peptide.
- the tag amino acid sequence is a hexahistidine peptide, such as the tag provided in the pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), etc. Many of them are commercially available of. As described in Gentz et al., 1989, Proc. Natl. Acad. Sci. USA 86:821-824, for example, hexahistidine provides convenient purification of the fusion protein.
- peptide tags used for purification include, but are not limited to, hemagglutinin ("HA") tags, which correspond to epitopes derived from influenza hemagglutinin protein (Wilson et al., 1984, Cell 37:767) and "flag" label.
- HA hemagglutinin
- the indicated label may be a diagnostic agent or a detectable agent.
- the obtained antibody conjugate can be used as part of a clinical test method (such as determining the efficacy of a specific therapy) for monitoring or predicting the onset, formation, progression, and/or severity of a disease or disorder.
- Detectable or diagnostic agents include, but are not limited to, various enzymes, such as horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase or acetylcholinesterase; prosthetic groups, such as streptavidin/biotin And avidin/biotin; fluorescent substances, such as but not limited to umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinamide fluorescein, dansyl chloride or phycoerythrin; luminescence Substances, such as but not limited to luminol; bioluminescent substances, such as but not limited to luciferase, luciferin, and aequorin; radioactive substances, such as but not limited to iodine ( 131 I, 125 I, 123 I and 121 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 115 In, 113 In
- the therapeutic agent includes chemotherapeutics, cytokines, cytotoxic agents, other antibodies, small molecule drugs, or immunomodulators.
- the antibody molecule of the present invention can be conjugated to a therapeutic moiety (therapeutic agent) such as radioactive metal ions, such as ⁇ -emitters such as 213 Bi or can be used to make radioactive metal ions (including but not limited to 131 In, 131 LU, 131 Y, 131 Ho, 131 Sm) macrocyclic chelating agents conjugated to polypeptides.
- a therapeutic moiety such as radioactive metal ions, such as ⁇ -emitters such as 213 Bi or can be used to make radioactive metal ions (including but not limited to 131 In, 131 LU, 131 Y, 131 Ho, 131 Sm) macrocyclic chelating agents conjugated to polypeptides.
- the macrocyclic chelating agent is 1,4,7,10-tetraazacyclododecane-N,N',N",N"'-tetraacetic acid (DOTA), which can be passed through a linker The molecule attaches to the antibody.
- the invention provides a nucleic acid encoding any antibody or fragment or any chain thereof described herein.
- a vector comprising the nucleic acid is provided.
- the vector is an expression vector, such as a pFuse vector.
- a host cell comprising the nucleic acid or the vector is provided.
- the host cell is eukaryotic.
- the host cell is selected from yeast cells, mammalian cells (such as CHO cells or 293 cells) or other cells suitable for preparing antibodies or antigen-binding fragments thereof.
- the host cell is prokaryotic.
- the nucleic acid of the present invention includes:
- the nucleic acid selected from SEQ ID NO: 39-42, or the nucleic acid selected from SEQ ID NO: 39-42 has at least 85%, 90%, 91%, 92%, 93%, 94%, 95% %, 96%, 97%, 98%, or 99% identical nucleic acids.
- the present invention also covers nucleic acids that hybridize with the following nucleic acids under stringent conditions or have one or more substitutions (such as conservative substitutions), deletions or insertions with the following nucleic acids: comprising a code selected from SEQ ID NO: 1- A nucleic acid comprising a nucleic acid sequence of the amino acid sequence shown in any one of 9; or a nucleic acid comprising a coding and an amino acid sequence selected from any one of SEQ ID NO: 13-22 with at least 85%, 90%, 91%, 92% , 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the nucleic acid sequence of the amino acid sequence; or the nucleic acid comprising the nucleic acid sequence selected from the group consisting of SEQ ID NO: 39-42 Nucleic acid, or comprising a nucleic acid sequence selected from SEQ ID NO: 39-42 with at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 9
- one or more vectors comprising the nucleic acid are provided.
- the vector is an expression vector, such as a eukaryotic expression vector.
- Vectors include, but are not limited to, viruses, plasmids, cosmids, lambda phage, or yeast artificial chromosomes (YAC).
- YAC yeast artificial chromosomes
- pFuse vector Once the expression vector or DNA sequence for expression has been prepared, the expression vector can be transfected or introduced into a suitable host cell. Various techniques can be used to achieve this goal, such as protoplast fusion, calcium phosphate precipitation, electroporation, retroviral transduction, viral transfection, gene gun, lipid-based transfection or other conventional techniques.
- the cells are grown in culture and screened for appropriate activity.
- the methods and conditions for culturing the transfected cells produced and for recovering the antibody molecules produced are known to those skilled in the art and can be based on the methods known in this specification and the prior art, depending on the specific expression vector and Changes or optimization of mammalian host cells.
- the marker may, for example, provide prototrophy, biocidal resistance (e.g., antibiotics), or heavy metal (e.g. copper) resistance, etc., to the auxotrophic host.
- the selectable marker gene can be directly linked to the DNA sequence to be expressed or introduced into the same cell by co-transformation. Additional elements may also be required for optimal mRNA synthesis. These elements can include splicing signals, as well as transcription promoters, enhancers, and termination signals.
- a host cell comprising one or more polynucleotides of the invention.
- a host cell comprising the expression vector of the invention is provided.
- the host cell is selected from yeast cells, mammalian cells, or other cells suitable for preparing antibodies or antigen-binding fragments thereof.
- the present invention provides a method for preparing the antibody molecule of the present invention or a fragment thereof (preferred antigen-binding fragment), wherein the method includes a method suitable for expressing the antibody molecule of the present invention or a fragment thereof (preferred antigen-binding fragment).
- the host cell is cultured under the condition of nucleic acid, and the antibody or fragment thereof (preferably antigen-binding fragment) is optionally isolated.
- the method further comprises recovering the antibody molecule of the invention or a fragment thereof (preferably an antigen-binding fragment) from the host cell.
- a method for preparing an antibody molecule of the present invention comprises, under conditions suitable for expression of the antibody, culturing the antibody encoding the antibody (for example, any one polypeptide chain and/or multiple polypeptide chains)
- the nucleic acid or the host cell containing the expression vector of the nucleic acid, as provided above, and the antibody is optionally recovered from the host cell (or host cell culture medium).
- the nucleic acid encoding the antibody (such as the antibody described above, such as any one polypeptide chain and/or multiple polypeptide chains) is isolated, and inserted into one or more vectors for use in the host Further cloning and/or expression in the cell.
- Such nucleic acids are easily isolated and sequenced using conventional procedures (for example, by using oligonucleotide probes capable of specifically binding to genes encoding antibody heavy and light chains).
- the antibody molecules prepared as described herein can be purified by known existing techniques such as high performance liquid chromatography, ion exchange chromatography, gel electrophoresis, affinity chromatography, size exclusion chromatography and the like.
- the actual conditions used to purify a particular protein also depend on factors such as net charge, hydrophobicity, and hydrophilicity, and these will be obvious to those skilled in the art.
- the purity of the antibody molecule of the present invention can be determined by any one of a variety of well-known analytical methods, including size exclusion chromatography, gel electrophoresis, high performance liquid chromatography, and the like.
- the present invention also provides methods for identifying, screening or characterizing the physical/chemical properties and/or biological activities of the antibody molecules of the present invention.
- the antibody of the present invention is tested for its antigen binding activity, for example, by a known method such as ELISA, Western blot, and the like.
- the binding to CD40 can be determined using methods known in the art, and exemplary methods are disclosed herein.
- surface plasmon resonance assays e.g., affinity measurement
- ELISA assays are used.
- the invention also provides an assay method for identifying anti-CD40 antibodies with biological activity.
- Biological activities may include, for example, binding to CD40 (such as binding to human and/or rhesus CD40), improving CD40-mediated signal transduction (such as improving NFkappa-B signaling pathway), and reducing expression of CD40 by directly inducing tumor cell apoptosis.
- Cells such as Raji or Ramos cells
- activating dendritic cells or B cells or T cells such as by increasing T cell cytokine production
- promoting T cells such as CD8 + T cells, such as activated CD8 + T cells
- B cells proliferate or inhibit tumor growth.
- the antibodies of the invention are tested for such biological activity.
- T cells eg CD8+ T cells
- dendritic cells or B cells can be measured using methods known in the art. For example, by the level (eg, expression) of the cell activation marker CD8 (T cell) or CD86 (dendritic cell or B cell). Methods known in the art can also be used to measure CD40 signaling (for example, NF- ⁇ B signaling pathway) to measure T cell activation.
- a transgenic cell expressing human CD40 and a reporter gene including the NF-kappa B promoter fused to a reporter gene (e.g., ⁇ -luciferase, GFP) is generated.
- the addition of anti-CD40 antibodies to cells leads to an increase in NF-kappa B transcription, which is detected using assays for reporter genes (such as luciferase reporter assays).
- the ADCC effect of antibodies can be determined using methods known in the art. For example, by detecting its induction of tumor cell apoptosis (for example, by apoptosis marker molecules, such as CD95), the inhibition of tumor cell growth, or the inhibition of tumor growth in vivo.
- apoptosis marker molecules such as CD95
- T cells eg, CD8+ T cells, such as activated CD8+ T cells
- B cells eg, CD8+ T cells, such as activated CD8+ T cells
- the proliferation of B cells can be measured by a luminescence method, such as the CellTiter-Glo luminescence method.
- the proliferation of CD8+ T cells can be detected by the OVA-specific OT-I cell method.
- Cells for use in any of the above in vitro assays include cell lines that naturally express CD40 or are engineered to express CD40. Such cells include T cells that naturally express CD40L (for example, CD8+ T cells, such as activated CD8+ T cells), B cells or dendritic cells that naturally express CD40. Such cells also include cell lines that express CD40 and are transfected with CD40-encoding DNA that does not normally express CD40.
- T cells that naturally express CD40L for example, CD8+ T cells, such as activated CD8+ T cells
- B cells or dendritic cells that naturally express CD40.
- Such cells also include cell lines that express CD40 and are transfected with CD40-encoding DNA that does not normally express CD40.
- immunoconjugates of the present invention can be used to replace or supplement the anti-CD40 antibody to perform any of the aforementioned assays.
- any of the aforementioned assays can be performed using anti-CD40 antibodies and other active agents.
- the invention provides pharmaceutical compositions and combination products comprising the antibodies of the invention.
- the present invention provides a composition comprising any antibody molecule described herein or a fragment thereof (preferably an antigen-binding fragment thereof) or an immunoconjugate thereof, preferably the composition is a pharmaceutical composition.
- the composition further comprises pharmaceutical excipients.
- the present invention also includes compositions (including pharmaceutical compositions or pharmaceutical preparations) comprising the antibodies of the present invention or immunoconjugates thereof and/or compositions (including pharmaceutical compositions or pharmaceutical preparations) comprising polynucleotides encoding the antibodies of the present invention .
- the composition comprises one or more antibodies of the invention or fragments thereof or one or more polynucleotides encoding one or more antibodies or fragments thereof of the invention.
- compositions may also contain suitable pharmaceutical excipients, such as pharmaceutical carriers and pharmaceutical excipients known in the art, including buffers.
- pharmaceutical carrier includes any and all solvents, dispersion media, isotonic and absorption delaying agents, etc. that are physiologically compatible.
- Pharmaceutical carriers suitable for the present invention can be sterile liquids such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil, and the like. When the pharmaceutical composition is administered intravenously, water is the preferred carrier. It is also possible to use saline solutions and aqueous dextrose and glycerol solutions as liquid carriers, especially for injectable solutions.
- Suitable excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glyceryl monostearate, talc, sodium chloride, dry skim milk, glycerin , Propylene, glycol, water, ethanol, etc.
- excipients see also "Handbook of Pharmaceutical Excipients", fifth edition, R.C. Rowe, P.J. Seskey and S.C. Owen, Pharmaceutical Press, London, Chicago.
- composition may also contain small amounts of wetting or emulsifying agents, or pH buffering agents.
- composition of the invention can be in a variety of forms. These forms include, for example, liquid, semi-solid and solid dosage forms such as liquid solutions (for example, injectable solutions and infusible solutions), dispersions or suspensions, liposomes, and suppositories.
- liquid solutions for example, injectable solutions and infusible solutions
- dispersions or suspensions for example, liposomes, and suppositories.
- the preferred form depends on the intended mode of administration and therapeutic use. Commonly preferred compositions are in the form of injectable solutions or infusible solutions.
- the preferred mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal (i.p.), intramuscular) injection.
- the antibody molecule is administered by intravenous infusion or injection.
- the antibody molecule is administered by intramuscular, intraperitoneal or subcutaneous injection.
- the agonistic antibody that specifically binds to human CD40 of the present invention can be lyophilized for storage, and can be restored in a suitable carrier before use. This technique has been proven to be effective for conventional protein preparations, and well-known freeze-drying and reconstitution techniques can be used.
- the pharmaceutical composition or formulation of the present invention may also contain other therapeutic agents that are required for the specific indication being treated, preferably those that have complementary activities that do not adversely affect each other.
- therapeutic agents preferably those that have complementary activities that do not adversely affect each other.
- chemotherapeutics cytotoxic agents, vaccines, other antibodies, anti-infective agents, small molecule drugs or immunomodulators, etc.
- the present invention also provides a combination product, which comprises the antibody of the present invention or an antigen-binding fragment thereof, or an immunoconjugate thereof, and one or more other therapeutic agents (such as chemotherapeutics, other antibodies, Cytotoxic agents, vaccines, anti-infective agents, small molecule drugs or immunomodulators, etc.).
- a combination product which comprises the antibody of the present invention or an antigen-binding fragment thereof, or an immunoconjugate thereof, and one or more other therapeutic agents (such as chemotherapeutics, other antibodies, Cytotoxic agents, vaccines, anti-infective agents, small molecule drugs or immunomodulators, etc.).
- two or more components of the combination product may be administered to the subject in combination sequentially, separately, or simultaneously.
- the present invention also provides a kit comprising the antibody, pharmaceutical composition, immunoconjugate or combination product of the present invention, and optionally a package insert to guide administration.
- the present invention also provides a pharmaceutical product comprising the antibody, pharmaceutical composition, immunoconjugate, and combination product of the present invention.
- the pharmaceutical product further includes a package insert to guide administration.
- the other therapeutic agent includes, for example, one or more of the following: anti-CTLA-4 antibody, anti-PD-1 antibody, anti-PD-L1 antibody, anti-TIGIT antibody, anti-OX40 (also known as CD134, TNFRSF4, ACT35 and/or TXGP1L) antibody, anti-LAG-3 antibody, anti-CD73 antibody, anti-CD137 antibody, anti-CD27 antibody, anti-CSF-1R antibody, TLR agonist, or small molecule antagonist of IDO or TGF ⁇ . See also WO2017/059243 or WO2017/004006 for examples of therapeutic agents that can be combined with the antibody of the present invention.
- the present invention also provides a method for enhancing an immune response (e.g., antigen-specific T cell response) in a subject, which comprises administering to the subject an effective amount of the anti-CD40 antibody or antigen thereof of the present invention By binding the fragments, the immune response in the individual is enhanced.
- an immune response e.g., antigen-specific T cell response
- the individual has a tumor.
- the subject has a viral infection, such as a chronic viral infection.
- the present invention provides a method for activating T cells (such as CD8+ T cells) and/or dendritic cells and/or B cells in a subject using the antibodies of the present invention, which comprises giving the subject An effective amount of the anti-CD40 antibody or antigen-binding fragment thereof of the present invention is administered.
- the present invention also provides a method for promoting the proliferation of T cells (such as activated CD8+ T cells) or B cells using the method of the present invention, which comprises administering to a subject an effective amount of the anti-CD40 antibody or antigen-binding fragment thereof of the present invention .
- T cell responses such as antigen-specific T cell responses
- the T cells may be T eff cells, such as CD4+T eff cells, CD8+T eff cells, T helper (T h ) cells, and T toxic (T c ) cells.
- the present invention provides the use of the antibody molecules of the present invention to treat or prevent diseases that require modulation (e.g., enhancement) of an immune response in a subject, such as tumors or infections, such as chronic infections. Therefore, the present invention also provides a method for inhibiting tumor growth in a subject, which comprises administering the anti-CD40 antibody or antigen-binding fragment thereof of the present invention to the subject so that tumor growth is inhibited.
- diseases that require modulation e.g., enhancement
- an immune response in a subject such as tumors or infections, such as chronic infections. Therefore, the present invention also provides a method for inhibiting tumor growth in a subject, which comprises administering the anti-CD40 antibody or antigen-binding fragment thereof of the present invention to the subject so that tumor growth is inhibited.
- the disease is a CD40-related disease, for example, the disease is a disease in which CD40 expression or activity is reduced (e.g., compared to healthy controls), or the disease is a disease in which CD40 gene and/or protein levels are reduced (E.g. compared to healthy controls); or the disease benefits from activating CD40 activity, such as activating the CD40 signaling pathway, and/or activating T cells or B cells or dendritic cells.
- the present invention relates to a method of activating antigen activity or activating an antigen-mediated signaling pathway in an individual, the method comprising administering to the subject an effective amount of an antibody or antigen-binding fragment thereof disclosed herein.
- activating CD40 activity or activating CD40-mediated signaling pathway refers to activating the CD40 signaling pathway.
- the antibody or antigen-binding fragment thereof of the present invention can cause antibody-dependent cell-mediated cytotoxicity (ADCC), thereby killing tumor cells. Therefore, the present invention also relates to a method of treating tumors, which comprises administering an effective amount of the antibody or antigen-binding fragment thereof of the present invention to an individual.
- ADCC antibody-dependent cell-mediated cytotoxicity
- the present invention relates to a method of preventing or treating a tumor (e.g., cancer) in a subject, the method comprising administering to the subject an effective amount of the antibody molecule or pharmaceutical composition or immunoconjugate disclosed herein.
- a tumor e.g., cancer
- the tumor is cancer.
- the tumor may be a solid tumor or a liquid tumor, such as a malignant hematological tumor.
- the tumor is an immunogenic tumor.
- the tumor is non-immunogenic.
- the tumor is PD-L1 positive.
- the tumor is PD-L1 negative.
- tumors treated and/or prevented with antibody molecules include, but are not limited to, solid tumors, hematological cancers (e.g., leukemia, lymphoma, myeloma, e.g., multiple myeloma), and metastatic lesions.
- the cancer is a solid tumor.
- solid tumors include malignant tumors, for example, sarcomas and cancers of multiple organ systems, such as those that invade the lung, breast, ovary, lymphoid, gastrointestinal (for example, colon), anus, genitals, and genitourinary tract (for example, Kidney, bladder epithelium, bladder cells, prostate), pharynx, CNS (for example, brain, neural or glial cells), head and neck, skin (for example, melanoma), nasopharyngeal (for example, differentiated or undifferentiated Metastatic or locally recurrent nasopharyngeal carcinoma) and those of the pancreas, as well as adenocarcinomas, including malignant tumors such as colon cancer, rectal cancer, renal cell carcinoma, liver cancer, non-small cell lung cancer, small bowel cancer and esophageal cancer.
- the cancer can be early, middle or late or metastatic cancer.
- the cancer is selected from colorectal cancer (e.g., CRC), melanoma, e.g., advanced melanoma (e.g., stage II-IV melanoma) or HLA-A2-positive-melanoma; pancreas Cancer, for example, advanced pancreatic cancer; breast cancer, for example, metastatic breast cancer or triple negative breast cancer; head and neck cancer (e.g., HNSCC); esophageal cancer; renal cell carcinoma (RCC), for example, renal clear cell carcinoma (ccRCC) Or metastatic renal cell carcinoma (MRCC); lung cancer (e.g., NSCLC); cervical cancer; bladder cancer; or hematological malignancies, e.g., leukemia (e.g., lymphocytic leukemia) or lymphoma (e.g., Hodgkin’s lymphoma) (HL), non-Hodgkin's lymphoma (NHL), diffuse large B-cell lymphoma
- CRC
- examples of cancer further include, but are not limited to, B-cell proliferative disorders, which further include, but are not limited to, lymphoma (eg, B-cell non-Hodgkin's lymphoma (NHL)) and lymphocytic leukemia.
- lymphoma eg, B-cell non-Hodgkin's lymphoma (NHL)
- NHL lymphocytic leukemia
- the tumor is a tumor that requires activation of T cells or B cells or dendritic cells, such as cancer, such as a tumor or cancer with T cell dysfunction.
- the tumor is a tumor in which the expression or activity of OX40 is reduced.
- the tumor is a tumor that benefits from activation of the OX40 signaling pathway, such as cancer.
- the cancer described herein is lymphoma, colon cancer, colorectal cancer, rectal cancer, lung cancer (e.g., non-small cell lung cancer), liver cancer, gastric cancer, and metastatic cancers thereof.
- the antibodies or antigen-binding fragments thereof of the present invention are not suitable for the treatment of blood cancers with CD40 expression, which may be exacerbated by treatment with CD40 agonists.
- Certain cancers may be known to express CD40 and therefore experience such exacerbations and therefore can be excluded in category.
- specific tumor samples are tested for CD40 expression and are excluded from therapies with CD40 antibodies of the invention based on the test results.
- compositions disclosed herein can be used to treat metastatic lesions associated with the aforementioned cancers.
- the antibodies or antigen-binding fragments thereof that specifically bind to human CD40 of the present invention can also be administered prophylactically to reduce the risk of cancer, delay the onset of events in cancer progression, and/or reduce the risk of recurrence after cancer remission. This may be especially useful for patients whose tumors are difficult to locate and are known to have tumors due to other biological factors.
- the present invention relates to a method of preventing or treating an infectious disease in a subject, the method comprising administering to the subject an effective amount of the antibody molecule or pharmaceutical composition or immunoconjugate disclosed herein Or combination products or kits.
- the infectious disease is a chronic infection.
- the infection is a viral infection.
- the infection is acute or chronic.
- the chronic infection is a persistent infection, a latent infection, or a slow infection.
- the chronic infection is caused by a pathogen selected from bacteria, viruses, fungi, and protozoa.
- viruses include HIV, hepatitis (hepatitis A, hepatitis B, and hepatitis C), herpes viruses (e.g., VZV, HSV-1, HAV-6, HSV-II and CMV, Epstein-Barr virus), adenovirus, influenza Virus, flavivirus, Echo virus, rhinovirus, Coxsackie virus, coronavirus, respiratory syncytial virus, mumps virus, rotavirus, measles virus, rubella virus, parvovirus, vaccinia virus, HTLV virus, dengue fever virus , Papilloma virus, molluscum virus, polio virus, rabies virus, JC virus and arbo encephalitis virus; some examples of bacteria include Chlamydia, Rickettsia, Mycobacterium, Staphylococcus, Streptococcus, Pneumococcus , Meningococcus and Neisseria
- herpes viruses e
- the infectious diseases treated and/or prevented with antibody molecules include pathogens for which no effective vaccine currently exists or pathogens for which conventional vaccines are not fully effective. These include but are not limited to HIV, (A, B and C) hepatitis, influenza, herpes, Giardia, malaria, Leishmania, Staphylococcus aureus, Pseudomonas aeruginosa (Pseudomonas aeruginosa).
- the antibody or pharmaceutical composition or immunoconjugate or combination product or kit of the present invention can also be administered in combination with one or more other therapies, such as treatment modalities and/or other therapeutic agents, for use herein Said prevention and/or treatment.
- therapies such as treatment modalities and/or other therapeutic agents, for use herein Said prevention and/or treatment.
- the treatment modality includes surgery (e.g., tumor resection); radiation therapy (e.g., external particle beam therapy, which involves three-dimensional conformal radiotherapy in which the irradiation area is designed), localized irradiation (e.g., directed to a preselected target Or organ irradiation) or focused irradiation) and so on.
- radiation therapy e.g., external particle beam therapy, which involves three-dimensional conformal radiotherapy in which the irradiation area is designed
- localized irradiation e.g., directed to a preselected target Or organ irradiation
- focused irradiation can be selected from stereotactic radiosurgery, segmented stereotactic radiosurgery, and intensity-modulated radiotherapy.
- the focused irradiation may have a radiation source selected from particle beams (protons), cobalt-60 (photons), and linear accelerators (X-rays).
- Radiation therapy can be administered by one of several methods or a combination of methods, including but not limited to external particle beam therapy, internal radiation therapy, implant irradiation, stereotactic radiosurgery, whole body radiation therapy, radiotherapy, and permanent or transient Interstitial brachytherapy.
- the therapeutic agent is selected from chemotherapeutics, cytotoxic agents, vaccines, other antibodies, anti-infective agents, or immunomodulators (e.g., activators of costimulatory molecules or inhibitors of immune checkpoint molecules).
- Exemplary other antibodies include, but are not limited to, immune checkpoint inhibitors (e.g., anti-CTLA-4, anti-TIM-3, anti-CEACAM); antibodies that stimulate immune cells (e.g., agonistic GITR antibody or CD137 antibody); anti-cancer Antibodies (e.g., rituximab Trastuzumab Tositumomab Ibritomab Alemtuzumab Iparizumab Bevacizumab Erlotinib Cetuximab and many more.
- the other antibodies may be anti-PD-L1 antibodies, anti-LAG-3 antibodies, anti-PD-1 antibodies, or anti-CLA-4 antibodies.
- Exemplary vaccines include but are not limited to cancer vaccines.
- the vaccine can be a DNA-based vaccine, an RNA-based vaccine or a virus-transduction-based vaccine.
- Cancer vaccines can be preventive or therapeutic, such as cancer cells, purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), cells, and cells transfected with genes encoding immunostimulatory cytokines (He et al. (2004) J. Immunol. 173: 4919-28).
- Non-limiting examples of tumor vaccines that can be used include peptides of melanoma antigens, such as peptides of gp100, MAGE antigen, Trp-2, MART1 and/or tyrosinase, or transfected to express the cytokine GM-CSF Tumor cells.
- the cancer vaccine is a peptide cancer vaccine, which in some embodiments is a personalized peptide vaccine.
- the peptide cancer vaccine is a multivalent long peptide, a multipeptide, a peptide mixture, a hybrid peptide, or a peptide-pulsed dendritic cell vaccine (see, for example, Yamada et al., Cancer Sci, 104: 14-21 , 2013).
- the administration of the antibody or fragment thereof of the invention is combined with the administration of a tumor antigen.
- the antigen can be, for example, a tumor antigen, a viral antigen, a bacterial antigen, or an antigen from a pathogen.
- the tumor antigen comprises a protein.
- the tumor antigen comprises nucleic acid.
- the tumor antigen is a tumor cell.
- anti-infective agents include, but are not limited to, antiviral agents, antifungal agents, antiprotozoal agents, antibacterial agents, such as the nucleoside analogue zidovudine (AST), ganciclovir, foscarnet or cidovir etc.
- antiviral agents include, but are not limited to, antiviral agents, antifungal agents, antiprotozoal agents, antibacterial agents, such as the nucleoside analogue zidovudine (AST), ganciclovir, foscarnet or cidovir etc.
- Immunomodulators include immune checkpoint molecular inhibitors and costimulatory molecular activators.
- the inhibitor of immune checkpoint molecules is CTLA-4, TIM-3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CEACAM (e.g., CEACAM-1 and/or CEACAM-5) and/ Or TGFR inhibitor.
- the inhibition of molecules can be done at the DNA, RNA or protein level.
- inhibitory nucleic acids eg, dsRNA, siRNA, or shRNA
- the inhibitor of the immune checkpoint molecule is a polypeptide that binds to the immune checkpoint molecule, for example, a soluble ligand or antibody or antibody fragment.
- the immunomodulator is an activator or agonist of a costimulatory molecule.
- the agonist of the costimulatory molecule is selected from agonists of the following molecules (for example, agonistic antibodies or antigen-binding fragments thereof, or soluble fusions): OX40, CD2, CD27, CDS, ICAM-1, LFA -1 (CD11a/CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD30, CD40, BAFFR, HVEM, CD7, LIGHT, NKG2C, SLAMF7, NKp80, CD160, B7-H3 or CD83 ligand.
- the antibodies or fragments thereof of the present invention can be administered in combination with treatments comprising adoptive transfer of T cells expressing chimeric antigen receptors (CAR) (eg, cytotoxic T cells or CTL).
- CAR chimeric antigen receptors
- the antibodies or fragments thereof of the present invention can be administered in combination with anti-tumor agents or oncolytic viruses.
- CD40 agonism can be combined with other forms of immunotherapy such as cytokine therapy (eg interferon, GM-CSF, G-CSF, IL-2) or bispecific antibody therapy, which provides enhanced tumor Antigen presentation.
- cytokine therapy eg interferon, GM-CSF, G-CSF, IL-2
- bispecific antibody therapy which provides enhanced tumor Antigen presentation. See, for example, Holliger (1993) Proc. Natl. Acad. Sci. USA 90: 6444-6448; Poljak (1994) Structure 2: 1121-1123.
- the antibodies or fragments thereof of the present invention can be combined with conventional methods for enhancing host immune function, including but not limited to: (i) APC enhancement, such as (a) injection of encoding heterologous MHC into tumors DNA of allogeneic antigen, or (b) transfected biopsy tumor cells with genes that increase the possibility of immune antigen recognition (such as immunostimulatory cytokines, GM-CSF, costimulatory molecules B7.1, B7.2), ( iii) Adoptive cellular immunotherapy, or treatment with activated tumor-specific T cells.
- APC enhancement such as (a) injection of encoding heterologous MHC into tumors DNA of allogeneic antigen, or (b) transfected biopsy tumor cells with genes that increase the possibility of immune antigen recognition (such as immunostimulatory cytokines, GM-CSF, costimulatory molecules B7.1, B7.2),
- Adoptive cellular immunotherapy or treatment with activated tumor-specific T cells.
- Adoptive cellular immunotherapy includes the isolation of tumor-infiltrating host T lymphocytes, such as stimulation by IL-2 or tumor or both, to expand the population in vitro; in addition, dysfunctional isolated T cells can also be used in vitro by applying the antibody of the present invention To activate, the T cells thus activated can then be re-administered to the host.
- T lymphocytes such as stimulation by IL-2 or tumor or both
- Such combination therapies encompass combined administration (in which two or more therapeutic agents are contained in the same formulation or separate formulations), and separate administration, in which case, other therapies, such as treatment modality and
- other therapies such as treatment modality and
- the administration of the antibody of the invention occurs before, at the same time, and/or after the therapeutic agent.
- Antibody molecules and/or other therapies, such as therapeutic agents or treatment modalities can be administered during active disease or during remission or less active disease.
- the antibody molecule can be administered before other treatments, concurrently with other treatments, after treatment, or during disease remission.
- immunoconjugates or compositions or combination products or kits of the present invention can be used to replace or supplement the antibodies of the present invention for any treatment.
- the administration mode of the antibodies of the present invention can be any suitable route, such as parenteral administration, such as intradermal, intramuscular, and intraperitoneal , Intravenous or subcutaneous, mucosal (oral, intranasal, intravaginal, rectal) or other methods as understood by those skilled in the art.
- parenteral administration such as intradermal, intramuscular, and intraperitoneal
- Intravenous or subcutaneous such as intravenous or subcutaneous, mucosal (oral, intranasal, intravaginal, rectal) or other methods as understood by those skilled in the art.
- the agonistic antibody that specifically binds to CD40 can be administered intratumorally to the drainage site of the lymph node for local delivery to the tumor using known methods.
- the agonistic antibody that specifically binds to human CD40 of the present invention can be administered to a patient by any suitable route, for example, parenterally by intravenous (i.v.) infusion or bolus injection, intramuscular or subcutaneous or intraperitoneal injection.
- i.v. intravenous
- bolus injection intramuscular or subcutaneous or intraperitoneal injection.
- medication schedules are covered herein, including, but not limited to, single administration or multiple administrations at multiple time points, bolus administration and pulse infusion.
- the appropriate dose of the antibody of the present invention (when used alone or in combination with one or more other therapeutic agents) will depend on the type of disease to be treated, the type of antibody, the severity and progress of the disease , Whether the antibody is administered for prophylactic or therapeutic purposes, previous treatments, the patient's clinical history and response to the antibody, and the judgment of the attending physician.
- the antibody is suitably administered to the patient in one treatment or over a series of treatments. Generally, the clinician administers the composition until the dose to achieve the desired effect is achieved.
- the antibody of the present invention can therefore be administered in a single dose, or in two or more doses (which may contain the same or different amounts of the desired molecule) within a certain period of time, or by continuous infusion via an implanted device or catheter.
- the appropriate dose can be determined by using appropriate dose response data.
- the antibody can be administered to the patient for an extended period of time. In certain embodiments, the antibody is administered every week, every two weeks, every month, every two months, every three months, every four months, every five months, or every six months.
- the anti-CD40 antibodies or antigen-binding fragments thereof provided herein can be used to detect the presence of CD40 in biological samples.
- detection includes quantitative or qualitative detection. Exemplary detection methods may involve immunohistochemistry, immunocytochemistry, flow cytometry (for example, FACS), antibody molecule complexed magnetic beads, ELISA assays Method, PCR-technology (for example, RT-PCR).
- the biological sample is blood, serum, or other liquid samples of biological origin.
- the biological sample comprises cells or tissues.
- the biological sample is derived from the relevant lesions of the diseases described herein (e.g., tumors or infections)
- CD40 is human CD40 or cyno CD40.
- the method includes contacting a biological sample with an anti-CD40 antibody or antigen-binding fragment thereof as described herein under conditions that allow the anti-CD40 antibody to bind to CD40, and detecting whether the anti-CD40 antibody is between the anti-CD40 antibody and CD40 Whether to form a complex. The formation of the complex indicates the presence of CD40.
- the method can be an in vitro or in vivo method.
- anti-CD40 antibodies are used to select subjects suitable for treatment with anti-CD40 antibodies, for example where CD40 is the biomarker used to select the subject.
- the antibodies of the invention can be used to diagnose the diseases described herein, for example to evaluate (eg, monitor) the treatment or progression, diagnosis, and/or staging of the diseases described herein in a subject.
- labeled anti-CD40 antibodies are provided.
- Labels include, but are not limited to, labels or parts that are directly detected (such as fluorescent labels, chromophore labels, electron dense labels, chemiluminescence labels, and radioactive labels), and parts that are indirectly detected, such as enzymes or ligands, for example, Through enzymatic reactions or molecular interactions.
- Exemplary labels include, but are not limited to, the radioisotopes 32 P, 14 C, 125 I, 3 H and 131 I, fluorophores such as rare earth chelates or fluorescein and its derivatives, rhodamine and its derivatives, dansyl ( dansyl), umbelliferone, luceriferase, for example, firefly luciferase and bacterial luciferase (US Patent No.
- the sample is obtained before treatment with the anti-CD40 antibody. In some embodiments, the sample is obtained prior to treatment with the disease drugs described herein. In some embodiments, the sample is formalin fixed, paraffin coated (FFPE). In some embodiments, the sample is a biopsy (e.g., a core biopsy), a surgical specimen (e.g., a specimen from a surgical resection), or a fine needle aspirate.
- FFPE formalin fixed, paraffin coated
- the sample is a biopsy (e.g., a core biopsy), a surgical specimen (e.g., a specimen from a surgical resection), or a fine needle aspirate.
- CD40 is detected before treatment, for example, before the initial treatment or before some treatment after the treatment interval.
- a detection kit comprising the antibody of the present invention or an antigen-binding fragment thereof is provided for the diagnosis of diseases described herein, such as tumors or infections.
- a method of treating diseases described herein, such as tumors or infections comprising: testing a subject (e.g., a sample) (e.g., a sample of the subject) for the presence of CD40, thereby Determine the CD40 value, compare the CD40 value to the control value, and if the CD40 value is less than the control value, then administer to the subject a therapeutically effective amount of an anti-CD40 antibody optionally in combination with one or more other therapies (for example, as described herein The aforementioned anti-CD40 antibodies), thereby treating the diseases described herein, such as tumors or infections.
- the present invention therefore also relates to the use of the antibody or antigen-binding fragment thereof of the present invention in the above-mentioned method, and the use of the antibody or antigen-binding fragment thereof of the present invention in the preparation of a medicine or composition or combination product or kit used in the above-mentioned method Use, and/or use of the antibody or antigen-binding fragment thereof of the present invention in preparing a kit for diagnosing the diseases described herein.
- the methods and uses applicable to the antibody or antigen-binding fragment thereof of the present invention are also applicable to immunoconjugates, compositions, combination products or kits comprising the antibody or antigen-binding fragment thereof of the present invention.
- Figure 1 shows the monoclonal activation of Jurkat/NF- ⁇ B-GFP+hCD40 reporter cells by HEL and CD40L produced by 293F cells measured by flow cytometry.
- the reporter cells have GFP expression after activation.
- the GFP positive rate is detected by FITC channel .
- Figure 2 shows the positive rate of CD40-binding antibodies in the antibody library obtained from the second round ( Figure 2A) and the third round ( Figure 2B) of phage display enrichment by phage ELISA.
- the binding signal of antibody to CD40 is greater than that of BSA binding signal. More than three times is defined as a positive antibody.
- Figure 3 shows that the supernatant containing negative control N27 and positive CD40 agonist antibody produced by the Jurkat/NF- ⁇ B-GFP+hCD40 reporter cell monoclonal was determined by flow cytometry to Jurkat/NF- ⁇ B-GFP+hCD40 reporter cell Activation.
- the fluorescence intensity of GFP detected by the FITC channel represents the activation degree of the monoclonal.
- Figure 4 shows the determination of the properties of the NK003 polymer of the antibody of the present invention produced by 293F cells by size exclusion chromatography.
- Figure 5A shows that the antibody NK003 of the present invention produced by 293F cells specifically binds to CD40, but does not bind or lowly binds to other TNFR family members OX40, 4-1BB, and GITR.
- Figure 5B shows the specific binding of the antibody NK004 of the present invention produced by 293F cells to CD40 measured by ELISA.
- Figure 6 shows that the antibody NK003 of the present invention produced by 293F cells competitively binds to CD40 with CD40L as determined by ELISA.
- Figure 7A shows that the antibody NK003 of the present invention produced by 293F cells was determined by flow cytometry to activate Jurkat/NF- ⁇ B-GFP+hCD40 reporter cells in a cross-linked form
- Figure 7B shows the flow cytometry measurement of 293F cells produced by The invented antibody NK004 does not rely on cross-linking and activates Jurkat/NF- ⁇ B-GFP+hCD40 reporter cells in a constitutive manner.
- the FITC channel detects GFP
- MFI is defined as the product of the Geometrical Mean of GFP-positive cells and the percentage of GFP-positive cells.
- Figure 8 shows the binding of the antibody NK003 of the present invention produced by 293F cells to 293FT cells overexpressing human CD40 ( Figure 8A) and 293FT cells overexpressing rhesus CD40 ( Figure 8B) measured by flow cytometry.
- Figure 9 shows that the antibody NK003 of the present invention produced by 293F cells induced apoptosis of Raji cells (Figure 9A) and Ramos cells ( Figure 9B) by flow cytometry, and the apoptosis index is the expression of CD95.
- MFI is defined as the product of the geometric mean number of CD95 positive cells and the percentage of CD95 positive cells.
- Figure 10 shows the determination of the antibody NK003 of the present invention produced by 293F cells by flow cytometry on dendritic cells in PBMC when no crosslinker (Figure 10A) and crosslinker anti-Fc ( Figure 10B) are added. activation.
- the index of dendritic cell activation is the expression of CD86.
- MFI is defined as the product of the geometric mean number of CD86 positive cells and the percentage of CD86 positive cells.
- Figure 11 shows the proliferation effect of the antibody NK003 of the present invention produced by 293F cells on B cells in PBMC measured by the CellTiter-Glo method.
- the cell proliferation index is fluorescence intensity (RLU). The larger the RLU value, the more cells.
- Fig. 12 shows the activation of B cells in PBMC by flow cytometry measurement of the antibody NK003 of the present invention produced by 293F cells without the addition of a cross-linking agent (Fig. 12A) and the addition of a cross-linking agent anti-Fc (Fig. 12B).
- the B cell activation index is the expression of CD86.
- MFI is defined as the product of the geometric mean number of CD86 positive cells and the percentage of CD86 positive cells.
- Figure 13 shows the antibody NK003 and negative control HEL of the present invention produced in 293F cells while being inoculated with Raji cells in SCID mice, individual mouse tumor growth curves (Figure 13A) and mouse survival rate statistics (Figure 13B) .
- Figure 14 shows the immune system activation of the antibody NK003 produced in 293F cells and negative control HEL in MC38 tumor-bearing CD40 humanized mice, the number of OT1 cells, the ratio of OT1 cells to CD8 cells (OT1/CD8 + Ratio) and the increase in the ratio of CD8 cells to CD4 cells (CD8/CD4 ratio) can indicate that the immune system is activated.
- Figure 15 shows the tumor growth curve of individual mice (Figure 15A) and the weight change of the antibody NK003, negative control HEL and positive control CP870893 produced in 293F cells administered to MC38 tumor-bearing CD40 humanized mice Curve ( Figure 15B).
- Figure 16 shows that the antibodies of the present invention NK003, Fc ⁇ RIIB enhanced mutant NK003-V12, Fc ⁇ RIIA/Fc ⁇ RIIB enhanced mutant NK003-S267E/L328F and negative control HEL produced by 293F cells determined by flow cytometry were measured in 293FT-Fc ⁇ RIIA cells, respectively ( Figure 16A) and 293FT-Fc ⁇ RIIB cells ( Figure 16B) cross-linked to activate Jurkat/NF- ⁇ B-GFP+hCD40 reporter cells.
- FITC channel detects GFP
- MFI is defined as the product of the geometric mean number of GFP-positive cells and the percentage of GFP-positive cells.
- Figure 17 shows the plasmid map of NK003 inserted into the pcomb3 vector in Fab format.
- Figure 18 shows the positive rate of CD40-binding antibodies in the antibody library obtained from the third round of phage display measured by phage ELISA.
- the binding signal of antibodies to CD40 is more than three times the signal to BSA is defined as positive antibodies.
- Figure 19 shows the third-generation sequencing results of NK003VH (Figure 19A) and VL ( Figure 19B) complementarity determining region CDR3 affinity maturation.
- Figure 20 shows that the antibodies NK003-AM-9 and NK003-AM-18 of the present invention produced by 293F cells were determined by flow cytometry to activate Jurkat/NF- ⁇ B-GFP+hCD40 reporter cells in a cross-linked form, and the FITC channel detects GFP, MFI is defined as the product of the Geometrical Mean of GFP-positive cells and the percentage of GFP-positive cells.
- Figure 21 shows the third-generation sequencing results of NK003VH (Figure 21A) and VL (Figure 21B) framework region affinity maturation.
- Figure 22 shows that the antibody NK003-AM-18-EP1 of the present invention produced by 293F cells is determined by flow cytometry to activate Jurkat/NF- ⁇ B-GFP+hCD40 reporter cells in a cross-linked form.
- the FITC channel detects GFP, and MFI is defined as GFP The product of the Geometrical Mean of positive cells and the percentage of GFP positive cells.
- the term “comprising” or “including” means including the stated elements, integers or steps, but does not exclude any other elements, integers or steps.
- the term “comprising” or “including” when used, unless otherwise specified, it also covers the combination of the stated elements, integers or steps.
- an antibody variable region that "comprises” a specific sequence it is also intended to encompass the antibody variable region composed of the specific sequence.
- CD40 is known in the art, such as human CD40 or rhesus CD40.
- CD40 is also known as tumor necrosis factor receptor superfamily member 5 (TNFRSF5), CD4OL receptor, or CD154 receptor.
- TNFRSF5 tumor necrosis factor receptor superfamily member 5
- CD4OL receptor CD4OL receptor
- CD154 receptor CD154 receptor.
- the human full-length CD40 protein is a type I membrane protein with 277 amino acids, see, for example, NCBI, NM_001250.5.
- NCBI NM_001265862.1.
- anti-CD40 antibody refers to antibodies that are capable of having sufficient affinity Binding (human or rhesus monkey) CD40 subunit or fragments thereof so that the antibody can be used as a diagnostic and/or therapeutic agent in targeting (human or rhesus monkey) CD40.
- the degree of binding of an anti-CD40 antibody to a non-(human or rhesus monkey) CD40 protein is about 10%, about 20%, or about 30% of the binding of the antibody to (human or rhesus monkey) CD40 protein. , About 40%, about 50%, about 60%, about 70%, about 80%, or about 90% or more, as measured, for example, by radioimmunoassay (RIA) or biooptical interferometry or MSD assay.
- RIA radioimmunoassay
- Antibodies that specifically bind to human CD40 may have cross-reactivity with other related antigens, for example, with the same antigen from other species (homologous) (such as rhesus monkeys). Although monospecific antibodies specifically bind to one antigen or one epitope, bispecific antibodies specifically bind to two different antigens or two different epitopes.
- a “complementarity determining region” or “CDR region” or “CDR” is an antibody variable domain that is hypervariable in sequence and forms a structurally defined loop ("hypervariable loop") and/or contains antigen contact residues ( "Antigen contact point”) area.
- CDR is mainly responsible for binding to antigen epitopes.
- the CDRs of the heavy and light chains are usually called CDR1, CDR2, and CDR3, and are numbered sequentially from the N-terminus.
- the CDRs located in the variable domain of the antibody heavy chain are called HCDR1, HCDR2, and HCDR3, and the CDRs located in the variable domain of the antibody light chain are called LCDR1, LCDR2, and LCDR3.
- each CDR can be determined using any one or a combination of many well-known antibody CDR assignment systems, which include For example: Chothia based on the three-dimensional structure of antibodies and the topology of CDR loops (Chothia et al.
- the residues of each CDR are as follows.
- the CDR can also be determined based on having the same Kabat numbering position as a reference CDR sequence (for example, any of the exemplary CDRs of the present invention).
- variable region residues in an antibody refers to residue positions in the variable region of an antibody (including heavy chain variable region residues and light chain variable region residues).
- the CDR of the antibody of the present invention determines the boundary by IMGT rules, for example, the IMGT database is used to determine the boundary.
- the CDR boundaries of the variable regions of the same antibody obtained based on different assignment systems may be different. That is, the CDR sequences of the variable regions of the same antibody defined under different assignment systems are different. Therefore, when it comes to defining antibodies with specific CDR sequences defined in the present invention, the scope of the antibodies also covers antibodies whose variable region sequences include the specific CDR sequences, but due to the application of different schemes (for example Different assignment system rules or combinations) cause the claimed CDR boundary to be different from the specific CDR boundary defined in the present invention.
- Fc region or “Fc domain” or “Fc” refers to the C-terminal region of the heavy chain of an antibody, which mediates the binding of immunoglobulin to host tissues or factors, including binding to various cells located in the immune system (e.g., effector Fc receptors on cells) or bind to the first component of the classical complement system (C1q). Therefore, the Fc region contains the constant region of an antibody excluding the first constant region immunoglobulin domain (e.g., CH1 or CL).
- first constant region immunoglobulin domain e.g., CH1 or CL
- the Fc region contains the CH2 and CH3 constant regions in each of the two heavy chains of the antibody; the IgM and IgE Fc regions contain three heavy chain constant domains (CH structure) in each polypeptide chain. Domain 2-4).
- the Fc region contains immunoglobulin domains C ⁇ 2 and C ⁇ 3 and the hinge between C ⁇ 1 and C ⁇ 2.
- the Fc region of a human IgG heavy chain is usually defined as the amino acid residue at position C226 or P230 (or the amino acid between these two amino acids) to the carboxyl group of the heavy chain
- the fragments at the end where numbering is based on the EU index in Kabat. Kabat et al. (1991) Sequences of Proteins of Immunological Interest, National Institutes of Health, Bethesda, MD; see also Figures 3c-3f of US Patent Application Publication No. 2008/0248028.
- the CH2 domain of the Fc region of human IgG extends from about amino acid 231 to about amino acid 340, and the CH3 domain is located at the C-terminal side of the CH2 domain in the Fc region, that is, it extends from about amino acid 341 to about amino acid 447 (including C-terminal lysine).
- the Fc region can be a native sequence Fc, including any allotype variant, or a variant Fc (such as a non-naturally occurring Fc).
- Fc can also refer to this region in isolation or in the context of Fc-containing protein polypeptides such as "Fc region-containing binding proteins" also known as "Fc fusion proteins” (eg, antibodies or immunoadhesins).
- epitopes within protein antigens can be formed from contiguous amino acids (usually linear epitopes) or discontinuous amino acids (usually conformational epitopes) juxtaposed by the tertiary folding of the protein. Epitopes formed by consecutive amino acids usually (but not always) remain exposed to denaturing solvents, while epitopes formed by tertiary folding are usually lost when treated with denaturing solvents. Epitopes usually include at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acids in a unique spatial conformation.
- An antibody that binds to the same or overlapping epitope refers to an antibody that blocks 50%, 60%, 70%, 80%, 90%, or 95% of the reference antibody in a competition assay. Antigen binding, on the contrary, the reference antibody blocks 50%, 60%, 70%, 80%, 90% or 95% of the binding of the antibody to its antigen in a competition assay.
- An antibody that competes with a reference antibody for binding to its antigen refers to an antibody that blocks 50%, 60%, 70%, 80%, 90%, or 95% or more of the binding of the reference antibody to its antigen in a competition assay. Conversely, the reference antibody blocks 50%, 60%, 70%, 80%, 90%, or 95% of the binding of the antibody to its antigen in a competition assay.
- Many types of competitive binding assays can be used to determine whether one antibody competes with another, such as: solid-phase direct or indirect radioimmunoassay (RIA), solid-phase direct or indirect enzyme immunoassay (EIA), sandwich competition Determination (see, for example, Stahli et al., 1983, Methods in Enzymology 9:242-253).
- An antibody that inhibits (for example, competitively inhibits) the binding of a reference antibody to its antigen refers to an antibody that inhibits 50%, 60%, 70%, 80%, 90%, or 95% or more of the binding of the reference antibody to its antigen .
- the reference antibody inhibits 50%, 60%, 70%, 80%, 90% or 95% of the binding of the antibody to its antigen.
- the binding of an antibody to its antigen can be measured by affinity (e.g. equilibrium dissociation constant). Methods for determining affinity are known in the art.
- An antibody that shows the same or similar binding affinity and/or specificity as a reference antibody refers to an antibody that can have at least 50%, 60%, 70%, 80%, 90%, or 95% or more of the binding of the reference antibody Affinity and/or specificity. This can be determined by any method known in the art for determining binding affinity and/or specificity.
- Antibody in the form of IgG refers to the form of IgG to which the constant region of the heavy chain of the antibody belongs.
- the heavy chain constant regions of all antibodies of the same type are the same, and the heavy chain constant regions of antibodies of different types are different.
- an antibody in the form of IgG1 means that the Ig domain of its heavy chain constant region is the Ig domain of IgG1.
- Human antibody refers to an antibody having variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences. In addition, if the antibody contains a constant region, the constant region is also derived from human germline immunoglobulin sequences.
- a “humanized” antibody refers to an antibody in which some, most or all of the amino acids outside the CDR domain of a non-human antibody (such as a mouse antibody) have been replaced by corresponding amino acids derived from human immunoglobulin. In one embodiment of the humanized form of the antibody, some, most, or all of the amino acids outside the CDR domain have been replaced by amino acids from human immunoglobulins, while some, most of the amino acids in one or more CDR regions Or all amino acids have not changed. Small additions, deletions, insertions, substitutions, or modifications of amino acids are allowed, as long as they do not eliminate the ability of the antibody to bind to a specific antigen. "Humanized” antibodies retain antigen specificity similar to the original antibody.
- chimeric antibody refers to an antibody in which the variable region is derived from one species and the constant region is derived from another species, such as an antibody in which the variable region is derived from a mouse antibody and the constant region is derived from a human antibody .
- antibody fragment refers to a molecule that is different from an intact antibody, which contains a part of an intact antibody and binds to the antigen to which the intact antibody binds.
- antigen-binding fragment refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (eg, human CD40).
- antibody fragments include, but are not limited to, Fv, Fab, Fab', Fab'-SH, F(ab') 2 ; diabody; linear antibody; single chain antibody (such as scFv); single domain antibody; bivalent or bispecific Antibodies or fragments thereof; camelid antibodies; and bispecific antibodies or multispecific antibodies formed from antibody fragments.
- multispecific refers to an antibody that specifically binds to at least two different antigens or two different epitopes within an antigen, such as three, four, or five different antigens or epitopes.
- bispecific refers to antibodies that specifically bind to two different antigens or two different epitopes within the same antigen. Bispecific antibodies can be cross-reactive to other related antigens, or can bind epitopes shared between two or more different antigens.
- cross-linking refers to the higher-order multimerization of CD40 on cells induced by the binding of an antibody that specifically binds human CD40 to cis or trans Fc ⁇ RIIb, resulting in the induction of CD40 agonistic activity.
- the anti-human F(ab')2 as described herein can be used as a cross-linking agent to evaluate cross-linking in vitro.
- cross-reactivity refers to the ability of the antibodies described herein to bind CD40 from different species.
- the antibodies described herein that bind human CD40 also bind CD40 from another species (e.g., cynomolgus CD40).
- cross-reactivity can be measured by detecting specific reactivity or binding with purified antigens in binding assays (e.g. SPR, ELISA) or otherwise functionally interacting with cells that physiologically express CD40.
- binding assays e.g. SPR, ELISA
- Methods for determining cross-reactivity include standard binding assays as described herein, for example by Surface plasmon resonance (SPR) analysis use 2000SPR instrument (Biacore AB, Uppsala, Sweden) or flow cytometry technology.
- SPR Surface plasmon resonance
- an “immunoconjugate” is an antibody conjugated to one or more other substances, including but not limited to cytotoxic agents or labels.
- label refers to a compound or composition that is directly or indirectly conjugated or fused to a reagent (such as a polynucleotide probe or antibody) and facilitates the detection of the reagent to which it is conjugated or fused.
- the label itself may be detectable (e.g., a radioisotope label or a fluorescent label) or, in the case of enzymatic labeling, may catalyze a chemical change of a detectable substrate compound or composition.
- the term is intended to cover the direct labeling of a probe or antibody by coupling (ie, physically linking) a detectable substance to the probe or antibody and the indirect labeling of the probe or antibody by reaction with another reagent that is directly labeled.
- Examples of indirect labels include detection of primary antibodies using fluorescently labeled secondary antibodies and end labeling of DNA probes with biotin so that they can be detected with fluorescently labeled streptavidin.
- Vector refers to a polynucleotide that can replicate within a biological system or can move between such systems.
- Vector polynucleotides usually contain elements such as origin of replication, polyadenylation signals or selection markers, whose function is to promote the replication or maintenance of these polynucleotides in biological systems. Examples of such biological systems may include cells, viruses, animals, plants, and biological systems reconstructed with biological components capable of replicating vectors.
- the polynucleotide containing the vector may be a DNA or RNA molecule or a hybrid of these molecules.
- Expression vector refers to a vector that can be used in a biological system or a reconstructed biological system to direct the translation of the polypeptide encoded by the polynucleotide sequence present in the expression vector.
- an “isolated” antibody is an antibody that has been separated from a component of its natural environment.
- the antibody is purified to more than 95% or 99% purity, such as by, for example, electrophoresis (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatography (e.g., ion exchange or reverse phase HPLC) confirmed.
- electrophoresis e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
- chromatography e.g., ion exchange or reverse phase HPLC
- nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment.
- An isolated nucleic acid includes a nucleic acid molecule contained in a cell that normally contains the nucleic acid molecule, but the nucleic acid molecule exists outside the chromosome or at a chromosomal location different from its natural chromosomal location.
- the sequences are aligned for optimal comparison purposes (for example, the first and second amino acid sequences or nucleic acid sequences can be used for optimal alignment. Gaps can be introduced in one or both or non-homologous sequences can be discarded for comparison purposes).
- the length of the compared reference sequence is at least 30%, preferably at least 40%, more preferably at least 50%, 60% and even more preferably at least 70%, 80% , 90%, 100% of the reference sequence length.
- the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide at the corresponding position in the second sequence, then the molecules are identical at this position.
- Mathematical algorithms can be used to achieve sequence comparison and percent identity calculation between two sequences.
- the Needlema and Wunsch ((1970) J.Mol.Biol.48:444-453) algorithm (at http://www.gcg.com) that has been integrated into the GAP program of the GCG software package is used. (Available), use Blossum 62 matrix or PAM250 matrix and gap weight 16, 14, 12, 10, 8, 6 or 4 and length weight 1, 2, 3, 4, 5 or 6, to determine the difference between two amino acid sequences Percent identity.
- the GAP program in the GCG software package (available at http://www.gcg.com) is used, the NWSgapdna.CMP matrix and gap weights 40, 50, 60, 70 or 80 are used. Length weights 1, 2, 3, 4, 5, or 6, determine the percent identity between two nucleotide sequences.
- a particularly preferred parameter set (and a parameter set that should be used unless otherwise specified) is a Blossum 62 scoring matrix with a gap penalty of 12, a gap extension penalty of 4, and a frameshift gap penalty of 5.
- nucleic acid sequences and protein sequences described herein can be further used as "query sequences" to perform searches against public databases, for example, to identify other family member sequences or related sequences.
- hybridizes under low stringency, medium stringency, high stringency, or very high stringency conditions describes hybridization and washing conditions. Instructions for performing hybridization reactions can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6, which is incorporated by reference. Aqueous and non-aqueous methods are described in the references and either method can be used.
- the specific hybridization conditions mentioned in this article are as follows: 1) Low stringency hybridization conditions are in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by at least 50°C (for low stringency conditions, you can Increase the washing temperature to 55°C) Wash twice in 0.2X SSC, 0.1% SDS; 2) Medium stringency hybridization conditions are about 45°C in 6X SSC, and then at 60°C in 0.2X SSC, 0.1% SDS Wash one or more times in medium; 3) High-stringency hybridization conditions are in 6X SSC at about 45°C, and then wash one or more times in 0.2X SSC, 0.1% SDS at 65°C; and preferably 4) Very high The stringent hybridization conditions are washing one or more times in 0.5M sodium phosphate, 7% SDS at 65°C, and then in 0.2X SSC, 0.1% SDS at 65°C.
- the very high stringency condition (4) is the preferred condition and one that should be used unless otherwise specified.
- host cell refers to cells into which exogenous nucleic acid is introduced, including the progeny of such cells.
- Host cells include “transformants” and “transformed cells”, which include primary transformed cells and progeny derived therefrom, regardless of the number of passages.
- the offspring may not be exactly the same as the parent cell in nucleic acid content, but may contain mutations. Included herein is the mutant progeny with the same function or biological activity screened or selected in the initially transformed cell.
- Suitable host cells suitable for use in the present invention include prokaryotic microorganisms such as E. coli.
- the host cell can also be eukaryotic microorganisms such as filamentous fungi or yeast, or various eukaryotic cells, such as insect cells. Vertebrate cells can also be used as hosts. For example, a mammalian cell line that has been adapted to grow in suspension can be used.
- Examples of useful mammalian host cell lines include SV40 transformed monkey kidney CV1 line (COS-7); human embryonic kidney line (HEK 293 or 293F cells), 293 cells, baby hamster kidney cells (BHK), monkey kidney cells ( CV1), African green monkey kidney cells (VERO-76), human cervical cancer cells (HELA), canine kidney cells (MDCK), Buffalo rat liver cells (BRL 3A), human lung cells (W138), human liver Cells (Hep G2), Chinese Hamster Ovary cells (CHO cells), CHOK1SV cells, CHOK1SV GS-KO cells, CHOS cells, NSO cells, myeloma cell lines such as Y0, NS0, P3X63 and Sp2/0.
- the host cell is a CHO cell, such as CHOS cell CHOK1SV cell or CHOK1SV GS-KO, or the host cell is 293 cell, such as HEK293 cell.
- agonist refers to an antibody that specifically binds to human CD40, and induces the proliferation or activation of B cells and/or dendritic cells (DC) or T cells when binding to CD40.
- the proliferation or activation of B cells and DC and T cells can be measured by measuring increased B cell proliferation, or measuring any surface markers CD23, CD80, CD83, CD86 on B cells, or CD80, CD83, CD86 and CD80 on DC. Up-regulation of HLA-DR. When compared with a control sample that does not contain antibodies, the agonist can induce B cell and/or DC and/or T cell activation in a statistically significant manner.
- “Inhibition of tumor cell/tumor growth” refers to the growth of tumor cells or tumors in vitro or in vivo when contacted with a therapeutic agent or a combination of therapeutic agents, compared with the growth of the same tumor cell or tumor in the absence of the therapeutic agent. The measured drop.
- the inhibition of tumor cells or tumor growth in vitro or in vivo can be at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100%.
- therapeutic agent encompasses any substance that is effective in preventing or treating diseases, such as tumors (such as cancer) and infections (such as chronic infections), including chemotherapeutics, cytotoxic agents, vaccines, other antibodies, and anti-infections. Active agents, small molecule drugs or immunomodulators.
- “Chemotherapeutic agents” include chemical compounds useful in the treatment of immune system diseases, including but not limited to alkylating agents; antimetabolites; anti-microtubule inhibitors, natural products; antibiotics; enzymes; miscellaneous agents; hormones and antagonists; Anti-estrogens; anti-androgens; non-steroidal anti-androgens, topoisomerase inhibitors, receptor tyrosine kinase inhibitors, angiogenesis inhibitors, etc.
- chemotherapeutics of the invention such as Anastrozole Bicalutamide Bleomycin Sulfate Busulfan Busulfan injection Capecitabine N4-Pentoxycarbonyl-5-deoxy-5-fluorocytidine, carboplatin Carmustine Chlorambucil Cisplatin Claribine Cyclophosphamide Cytarabine, Cytosine Arabinoside Cytarabine liposome injection dacarbazine Dactinomycin (actinomycin D, Cosmegan), daunorubicin hydrochloride Daunorubicin Citrate Liposome Injection Dexamethasone, Docetaxel Doxorubicin hydrochloride Etoposide Fludarabine phosphate 5-fluorouracil Flutamide tezacitibine, gemcitabine (difedeoxycytidine), hydroxyurea Idabycin Ifosfamide Irinotecan L-Asparaginase Leucovorin, Melphalan
- This definition also includes anti-hormonal drugs such as anti-estrogens used to modulate or inhibit the effects of hormones on tumors, including, for example, tamoxifen, raloxifene, aromatase inhibitor 4(5)-imidazole, 4-hydroxyl Tamoxifen, trovoxifene, keoxifene, LY117018, onlastone and toremifene and antiandrogens such as flutamide, nilutamide, bicalutamide, leuprolide acetate and ge Serrelin; and a pharmaceutically acceptable salt, acid or derivative of any of the above.
- anti-hormonal drugs such as anti-estrogens used to modulate or inhibit the effects of hormones on tumors, including, for example, tamoxifen, raloxifene, aromatase inhibitor 4(5)-imidazole, 4-hydroxyl Tamoxifen, trovoxifene, keoxifene, LY117018, onlastone and
- cytotoxic agent refers to a substance that inhibits or prevents cell function and/or causes cell death or destruction.
- cytotoxic agents include, but are not limited to: radioisotopes, such as iodine (131 I, 125 I, 123 I and 121 I), carbon (14 C), sulfur (35 S), tritium (3 H), indium ( 115 In, 113 In, 112 In and 111 In), technetium ( 99 Tc), thallium ( 201 Ti), gallium ( 68 Ga, 67 Ga), palladium ( 103 Pd), molybdenum ( 99 Mo), xenon ( 133 Xe) ), fluorine ( 18 F), 153 Sm, 177 Lu, 159 Gd, 149 Pm, 140 La, 175 Yb, 166 Ho, 90 Y, 47Sc, 186 Re, 188 Re, 142 Pr, 105 Rh, 97 Ru, 68 Ge, 57 Co, 65 Zn, 85 S
- exemplary cytotoxic agents of the present invention include anti-microtubule drugs, topoisomerase inhibitors, antimetabolites, mitotic inhibitors, alkylating agents, anthracyclines, vinblastine alkaloids , Intercalators, agents capable of interfering with signal transduction pathways, pro-apoptotic agents, proteasome inhibitors, and irradiation (e.g., local or whole body irradiation (e.g., gamma radiation).
- irradiation e.g., local or whole body irradiation (e.g., gamma radiation).
- small molecule drugs refers to low molecular weight organic compounds capable of regulating biological processes.
- Small molecules are defined as molecules with a molecular weight of less than 10 kD, usually less than 2 kD and preferably less than 1 kD.
- Small molecules include, but are not limited to, inorganic molecules, organic molecules, organic molecules containing inorganic components, molecules containing radioactive atoms, synthetic molecules, peptide mimetics, and antibody mimetics. As a therapeutic agent, small molecules can penetrate cells better than large molecules, are less susceptible to degradation, and are less likely to trigger an immune response.
- anti-infective active agent includes any molecule that specifically inhibits or eliminates the growth of microorganisms, such as viruses, bacteria, fungi, or protozoa, such as parasites, at the applied concentration and dosing interval, but is not lethal to the host.
- anti-infective agent includes antibiotics, antibacterial agents, antiviral agents, antifungal agents, and antiprotozoal agents.
- the anti-infective active agent is non-toxic to the host at the applied concentration and dosing interval.
- Antibacterial anti-infective agents or antibacterial agents can be broadly classified as bactericidal (ie, direct killing) or bacteriostatic (ie, preventing division). Antibacterial anti-infective active agents can be further classified as narrow-spectrum antibacterial agents (i.e., affect only small bacterial subtypes, for example, Gram-negative, etc.) or broad-spectrum antibacterial agents (i.e., affect a wide range of species). Examples include amikacin, gentamicin, geldanamycin, herbimycin, mupirocin, nitrofurantoin, pyrazinamide, quinupristin/dalfopristin, rifampicin/isophos Amide or tinidazole, etc.
- antiviral agent includes any substance that inhibits or eliminates the growth, pathogenicity, and/or survival of a virus. This includes, for example, acyclovir, cidofovir, zidovudine, didanosine (ddI, VIDEX), zacitabine (ddC, HIVID), stavudine (d4T, ZERIT), lamie Fudine (3TC, EPIVIR)), abacavir (ZIAGEN), emtricitabine (EMTRIVA) and so on.
- antifungal agent includes any substance that inhibits or eliminates the growth, pathogenicity, and/or survival of fungi. This includes, for example, natamycin, schizotonin, filipin, nystatin, amphotericin B, candesine, patchouli, neem seed oil, coconut Oil (Coconut Oil) etc.
- antiprotozoal agent includes any substance that inhibits or eliminates the growth, morbidity, and/or survival of protozoan organisms, such as parasites.
- antiprotozoal agents include antimalarial agents such as quinine, quinidine and the like.
- immunomodulator refers to natural or synthetic active agents or drugs that inhibit or modulate the immune response.
- the immune response can be a humoral response or a cellular response.
- Immunomodulators include immunosuppressive agents.
- immune checkpoint molecule refers to a type of inhibitory signal molecule present in the immune system, which avoids tissue damage by regulating the persistence and intensity of the immune response in peripheral tissues, and participates in maintaining tolerance to self-antigens (Pardoll DM. , The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer, 2012, 12(4): 252-264).
- Immune checkpoint molecules include but are not limited to programmed death 1 (PD-1), PD-L1, PD-L2, cytotoxic T lymphocyte antigen 4 (CTLA-4), LAG-3 and TIM-3.
- costimulatory molecule refers to a corresponding binding partner on T cells that specifically binds to a costimulatory ligand to mediate a costimulatory response (such as but not limited to proliferation) of the T cell.
- Co-stimulatory molecules are cell surface molecules that contribute to an effective immune response in addition to antigen receptors or their ligands.
- Co-stimulatory molecules include but are not limited to MHC class I molecules, TNF receptor proteins, immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocyte activation molecules (SLAM proteins), activated NK cell receptors, OX40 , CD40, GITR, 4-1BB (ie CD137), CD27 and CD28.
- the "costimulatory molecule” is OX40, GITR, 4-1BB (ie CD137), CD27, and/or CD28.
- cytokine is a general term for proteins that are released by a cell population and act as intercellular mediators on another cell.
- cytokines are lymphokines, monocytes, interleukins (IL), such as IL-1, IL-1 ⁇ , IL-2, IL-3, IL-4, IL-5, IL-6, IL- 7, IL-8, IL-9, IL-11, IL-12, IL-15; tumor necrosis factor, such as TNF- ⁇ or TNF- ⁇ ; and other polypeptide factors, including LIF and kit ligand (KL) and Gamma interferon.
- IL interleukins
- cytokine includes proteins from natural sources or from recombinant cell culture and biologically active equivalents of natural sequence cytokines, including small molecular entities produced by artificial synthesis, and their pharmaceutically acceptable Derivatives and salts.
- inhibitor or "antagonist” includes substances that reduce certain parameters (e.g., activity) of a given molecule (e.g., immune checkpoint molecule).
- this term includes substances that cause the given molecule to be inhibited by at least 5%, 10%, 20%, 30%, 40% or more of the activity (eg, PD-L1 activity). Therefore, the inhibitory effect does not have to be 100%.
- activator includes substances that increase certain parameters (e.g., activity) of a given molecule (e.g., co-stimulatory molecule). For example, this term includes substances that increase the activity of a given molecule by at least 5%, 10%, 20%, 30%, 40%, or more (eg, OX40 activity). Therefore, the activation does not have to be 100%.
- pharmaceutical excipients refers to diluents, adjuvants (for example Freund's adjuvant (complete and incomplete)), excipients, carriers or stabilizers, etc. administered together with the active substance.
- composition refers to a composition that is present in a form that allows the biological activity of the active ingredients contained therein to be effective, and does not contain additional substances that have unacceptable toxicity to the subject to which the composition is administered. Ingredients.
- combination product refers to a fixed or non-fixed combination in the form of a dosage unit or a kit of parts for combined administration, in which two or more therapeutic agents can be independently at the same time or within a time interval Separate administration, especially when these time intervals allow the combination partner to exhibit cooperation, for example, a synergistic effect.
- fixed combination means that the antibody of the present invention and the combination partner (such as other therapeutic agents, such as immunomodulators, such as immunosuppressive agents or anti-inflammatory agents) are simultaneously administered to a patient in the form of a single entity or dosage.
- non-fixed combination means that the antibody of the present invention and the combination partner (such as other therapeutic agents, such as immunomodulators, such as immunosuppressive agents or anti-inflammatory agents) are administered to a patient simultaneously, concurrently or sequentially as separate entities, without specific Time constraints, where such administration provides therapeutically effective levels of both compounds in the patient.
- combination partner such as other therapeutic agents, such as immunomodulators, such as immunosuppressive agents or anti-inflammatory agents
- cocktail therapy such as the administration of three or more therapeutic agents.
- the drug combination is a non-fixed combination.
- Immunogenicity refers to the ability of a particular substance to elicit an immune response. Tumors are immunogenic, and enhancing tumor immunogenicity helps clear tumor cells through an immune response.
- Immuno response refers to a biological response in vertebrates against foreign substances that protects the organism from these substances and diseases caused by them.
- the immune response is determined by cells of the immune system (for example, T lymphocytes, B lymphocytes, natural killer (NK) cells, macrophages, eosinophils, mast cells, dendritic cells, or neutrophils) and by these Mediated by the action of soluble macromolecules (including antibodies, cytokines, and complement) produced by cells or liver, which lead to the selective targeting, binding, damage, destruction and/or elimination of invading pathogens, cells or infected pathogens from vertebrate organisms Tissues, cancerous or other abnormal cells, or (in the case of autoimmune or pathological inflammation) normal human cells or tissues.
- T lymphocytes for example, T lymphocytes, B lymphocytes, natural killer (NK) cells, macrophages, eosinophils, mast cells, dendritic cells, or neutrophils
- soluble macromolecules including antibodies
- T effector T cells for example, effector T cells or Th cells, such as CD4+ or CD8+ T cells, or suppression or depletion of Treg cells.
- T effector ("T eff ") cells refer to T cells with cytolytic activity (such as CD4+ and CD8+ T cells) and T helper (Th) cells (which secrete cytokines and activate and guide other immune cells), but Does not include regulatory T cells (Treg cells).
- the term "effective amount” refers to the amount or dose of the antibody or fragment or conjugate or composition of the present invention that, after administration to the patient in single or multiple doses, produces the desired effect in the patient in need of treatment or prevention.
- the effective amount can be easily determined by the attending physician as a person skilled in the art by considering various factors such as the species of mammal; its size, age and general health; the specific disease involved; the degree or severity of the disease; The response of the individual patient; the specific antibody administered; the mode of administration; the bioavailability characteristics of the administered formulation; the selected dosing regimen; and the use of any concomitant therapy.
- Therapeutically effective amount refers to the amount that is effective to achieve the desired therapeutic result at the required dose and for the required period of time.
- the therapeutically effective amount of the antibody or antibody fragment or its conjugate or composition may vary according to various factors such as disease state, the age, sex and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual.
- a therapeutically effective amount is also an amount in which any toxic or harmful effects of the antibody or antibody fragment or its conjugate or composition are less than the therapeutically beneficial effects.
- a "therapeutically effective amount” preferably inhibits a measurable parameter (such as swelling rate, etc.) by at least about 20%, more preferably at least about 40%, even more preferably at least about 50%, 60%, or 70%. % And still more preferably at least about 80% or 90%.
- a measurable parameter such as swelling rate, etc.
- the ability of a compound to inhibit a measurable parameter can be evaluated in an animal model system predicting efficacy in human autoimmune diseases or inflammation.
- prophylactically effective amount refers to an amount that effectively achieves the desired preventive result at the required dose and for the required period of time. Generally, since the prophylactic dose is used in the subject before or at an earlier stage of the disease, the prophylactically effective amount will be less than the therapeutically effective amount.
- the "individual” or “subject” described herein can be used interchangeably, including mammals. Mammals include, but are not limited to, domestic animals (for example, cattle, sheep, cats, dogs, and horses), primates (for example, human and non-human primates such as monkeys), rabbits, and rodents (for example, , Mice and rats).
- the individual or subject may be a mammal, e.g., a primate, preferably a higher primate, e.g., a human (e.g., suffering from a disease described herein or having a disease described herein Risk of patients).
- the subject has or is at risk of suffering from a disease described herein (eg, a tumor or infection as described herein).
- the subject has received or has received other treatments, such as chemotherapy treatment and/or radiation therapy.
- the subject is immunocompromised due to infection or is at risk of being immunocompromised due to infection.
- combination therapy refers to the administration of two or more therapeutic agents or treatment modalities (e.g. radiation therapy or surgery) to treat IL-23 related diseases as described in this disclosure.
- administration includes co-administration of these therapeutic agents in a substantially simultaneous manner, for example, in a single capsule having a fixed ratio of active ingredients.
- administration includes co-administration of the respective active ingredients in multiple or in separate containers (e.g., tablets, capsules, powders and liquids).
- the powder and/or liquid can be reconstituted or diluted to the desired dose before administration.
- such administration also includes the use of each type of therapeutic agent in a sequential manner at approximately the same time or at different times. In either case, the treatment regimen will provide the beneficial effects of the drug combination in the treatment of the conditions or conditions described herein.
- treatment refers to slowing, interrupting, blocking, alleviating, stopping, reducing, or reversing the progression or severity of an existing symptom, disorder, condition, or disease.
- prevention includes the inhibition of the occurrence or development of a disease or condition or the symptoms of a specific disease or condition.
- subjects with a family history of immune system disease are candidates for preventive regimens.
- prevention refers to the occurrence of symptoms or symptoms of immune system diseases (autoimmune diseases or inflammation), especially in the presence of immune system diseases (autoimmune diseases or inflammation).
- Pre-drug administration in subjects at risk of immune disease or inflammation.
- Subject/patient sample refers to a collection of cells or fluids obtained from a patient or subject.
- the source of the tissue or cell sample can be solid tissue, such as fresh, frozen and/or preserved organ or tissue samples or biopsy samples or puncture samples; blood or any blood component; body fluids such as cerebrospinal fluid, amniotic fluid (amniotic fluid) ), peritoneal fluid (ascites), or interstitial fluid; cells from the subject's pregnancy or development at any time.
- Tissue samples may contain compounds that are not naturally mixed with tissues in nature, such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, and so on.
- NF- ⁇ B transcription factor binding elements in the promoter of the NF- ⁇ B-GFP reporter gene lentiviral system (QIAGEN, CCS-013G).
- the activation of this promoter can drive the expression of GFP.
- Jurkat cells ATCC, TIB-152
- Jurkat cells infected with lentivirus were screened with puromycin (InvivoGen, ant-pr-1).
- TNF ⁇ protein (Sino Biological, 10602-HNAE) to stimulate the cells selected with puromycin, and 24 hours later, flow cytometry (BD, FACSAria III) sorts the GFP fluorescent cell subsets to obtain Jurkat/NF- ⁇ B-GFP cells.
- the positive control CD40L sequence used herein comes from patent WO 2016/177771 (SEQ ID NO: 15), and the negative control HEL sequence is shown in Table 10.
- the DNA sequences of CD40L and HEL are fully synthesized in Jinweizhi Company.
- the DNA sequence was inserted into the eukaryotic expression vector pFUSE (InvivoGen, pfuse-hg1fc1) and transfected with PEI into 293F cells (Thermo Fisher Scientific, R79007) using standard procedures; Freestyle medium (Lifetechnologies, 12338026) ) Culture the cells on a shaker at 37°C. After 7 days of culture, the supernatant was collected and purified on the AKTA system (GE) using a Superdex TM 200 Increase prepacked column (GE, 28-9909-44).
- CD40 signal transduction can activate the NF- ⁇ B pathway. If CD40 is successfully expressed on the Jurkat/NF- ⁇ B-GFP cell membrane surface, CD40L stimulation will induce GFP expression.
- the constructed Jurkat/NF- ⁇ B-GFP cells sorted as described above were infected with the human CD40 lentiviral supernatant obtained above, and after 16 hours, 100nM of the above-obtained CD40L protein (as a positive control) and HEL (as a negative control) were added ) Stimulate, 24h later, flow cytometer single cell sorting the cells with the strongest GFP fluorescence into 96-well plates.
- the cells in the above 96-well plate were cultured in RPMI1640 medium (Lifetechnologies, C11875500CP) containing 10% fetal bovine serum (Biological Industries, 04-001-1A) for 2 weeks, and after the single cells grew up, 100 nM of the above was added separately Trimerize CD40L protein (as a positive control) and HEL (as a negative control), select a CD40L-positive and HEL-negative single clone as Jurkat/NF- ⁇ B-GFP+hCD40 (referred to as NF- ⁇ B-GFP+hCD40) cells For follow-up experiments. The results are shown in Figure 1. The positive rate of Jurkat/NF- ⁇ B-GFP+hCD40 monoclonal GFP after CD40L stimulation was 94.7%, and HEL stimulation was not activated.
- the peripheral blood of 30 healthy adults was purchased from Tianjin Blood Center and Shanghai Miaotong Biological Company, and PBMC was obtained by centrifugation with ficoll separation solution (Haoyang, LDS1075). Centrifugation conditions: 20°C, 2000rpm, up 5 down 0 mode for 30 minutes.
- the human natural antibody library was constructed using the PBMC obtained as described above, and the library was constructed by a conventional method (phage display, Tim Clackson and Henry B. Lowman). The obtained antibody heavy chain and light chain variable regions are randomly combined and displayed on the N-terminus of phage capsid protein pIII in the form of single-chain antibody scFv to obtain a phage display antibody library with a storage capacity of 10 10 .
- Phage screening is a conventional technology (Phage Display: A Laboratory Manual, Carlos F Barbas III).
- the biotinylated CD40 protein (acrobiosystems, TN5-H82F9) was incubated with the phage display antibody library obtained as described above for 2 hours at room temperature. After incubation, add 150ul streptavidin magnetic beads Dynabeads M280 (Lifetechnologies, 11205D) directly, and incubate on a mixer at room temperature for 30 minutes.
- PBS-tween PBS: Lifetechnologies, 70011044; Tween: Sigma-Aldrich, 9005-64-5
- PBS Lifetechnologies, 70011044; Tween: Sigma-Aldrich, 9005-64-5
- PBS Lifetechnologies, 70011044; Tween: Sigma-Aldrich, 9005-64-5
- PH2.2 glycine-hydrochloric acid
- Bacteriophage conjugated The eluted phage was used to infect E. coli XL1-blue (Agilent, 200236), and the helper phage VCSM13 (Agilent, 200251) was added for amplification and used in the next round of screening. There are three rounds of screening. After three rounds, the antibody phages that bind to CD40 were enriched, and the screening results are shown in Table 1.
- An ELISA plate (Corning, 3690) was coated with 1 ⁇ g/ml of human CD40 (Acrobiosystems, CD0-H5228) and incubated overnight at 4°C. Add the overnight induced phage supernatant to the ELISA plate after overnight incubation, incubate at room temperature for 1 hour, and wash 8 times with 0.05% PBST.
- BSA Solarbio, A8020-100
- HRP-conjugated anti-M13 GE, 27-9421-01, M13 is phage capsid protein
- a positive clone was defined as a CD40 binding signal that was more than 3 times that of the BSA control. The results are shown in Figure 2. The positive rates of the second and third rounds of screening were 6.9% and 27.6%, respectively.
- N27 ScFv into secretory lentiviral vector pCDH as a negative control.
- the sequence of N27 ScFv is shown in Table 11.
- the lentiviral core plasmid and helper plasmids pPACKH1-GAG, pPACKH1-REV, pVSVG were co-transfected into 293FT cells at a ratio of 1:1:1:1, and the medium was changed 6 hours after transfection.
- the cells were cultured at 37°C with 10% fetal bovine serum ( In the DMEM medium of Biological Industries, 04-001-1A), continue to culture for 48 hours to collect the supernatant containing the CD40-binding antibody lentiviral library; infect HEK293 cells with 50,000 pg of the CD40-binding antibody lentiviral library, and after infection for 16 hours, Centrifuge to remove the medium containing the lentivirus, add fresh medium, and sort the infected HEK293 cells into a 96-well plate using flow sorting technology so that each well contains only one infected HEK293 cell. Culture the cells for 3 weeks. After the cells reach a certain number, take the supernatant.
- 10% fetal bovine serum In the DMEM medium of Biological Industries, 04-001-1A
- the supernatant was added with 3 ⁇ 10 5 Jurkat/NF- ⁇ B-GFP+hCD40 reporter cells obtained as described above, and 2.5 ⁇ g/ml goat anti-human Fc (SouthernBiotech, SBA-2048-01) secondary antibody was added for cross-linking
- the antibody secreted by the cells enhances the activation strength of the agonist antibody; the activity of the cell supernatant is tested after 24 hours of culture, and the detection result of the positive antibody is shown in Figure 3.
- the antibody gene is extracted from the HEK293 cells with positive cell supernatant activity and constructed into pFUSE
- the vector was sequenced to obtain the positive antibody VH and VL sequences, and the two positive antibodies obtained were numbered NK003 and NK004.
- Table 4 to Table 8 for their sequences (including CDR, VH/VL, heavy chain, and light chain), please refer to Table 4 to Table 8 in the sequence list the sequence of.
- the selected agonist antibodies NK003, NK004 heavy chain and light chain DNA (Jin Weizhi) were synthesized and cloned into the vector pFUSE.
- PEI was used to convert the plasmids containing heavy and light chains. They were transiently co-transfected into 293F suspension cells at 1:1 to express full-length antibody.
- the AKTA system was purified using Superdex TM 200Increase prepacked column.
- NK003 The binding selectivity of NK003 was evaluated by direct ELISA with TNFR family members OX40, 4-1BB and GITR. Coated on the ELISA plate containing 1 ⁇ g/ml human OX40 (Acrobiosystems, OX0-H5224), 4-1BB (Acrobiosystems, 41B-H5227), GITR (Acrobiosystems, GIR-H5228) and CD40 (Acrobiosystems, CD0-H5228) Incubate in PBS solution overnight at 4°C.
- human OX40 Acrobiosystems, OX0-H5224
- 4-1BB Acrobiosystems, 41B-H5227
- GITR Acrobiosystems, GIR-H5228
- CD40 Acrobiosystems, CD0-H5228
- NK003 diluted to different concentrations with PBS as obtained in Example 4: 0.625 ⁇ g/ml, 1.25 ⁇ g/ml, 2.5 ⁇ g/ml and 5 ⁇ g/ml, after incubating for 30min at room temperature, wash with 0.05% PBS-Tween 8 times. Added goat anti-human HRP-conjugated Fc antibody (SouthernBiotech, 2048-05) to detect bound NK003. As shown in Figure 5A, NK003 selectively binds to human CD40, with low or no binding to other tested TNFR family proteins.
- NK004 In order to evaluate the binding of NK004 to CD40, a 1 ⁇ g/ml human CD40 (Acrobiosystems, CD0-H5228) PB solution was coated on an ELISA plate and incubated overnight at 4°C. Add the PBS obtained in Example 4 and dilute to different concentrations NK004: 0.002nM, 0.016nM, 0.13nM, 1nM, 8nM, 64nM and 500nM. HEL is a negative control antibody. After incubating at room temperature for 30 minutes, wash with 0.05% PBS-Tween 8 times . Add goat anti-human HRP-conjugated Fc antibody to detect bound NK004. As shown in Figure 5B, NK004 can bind to human CD40.
- ELISA was used to evaluate the effect of NK003 on the binding of CD40L and CD40.
- NK003 and NK004 activate NF- ⁇ B-GFP+hCD40 reporter cell line in cross-linked form
- flow cytometry was used to detect the activation of NF- ⁇ B-GFP+hCD40 reporter cell lines by NK003 and NK004 antibodies, and to determine the EC50 of the two antibodies, the specific steps are as follows:
- Example 4 Take 3 ⁇ 10 5 cells/tube to obtain NF- ⁇ B-GFP+hCD40 reporter cells as in Example 1, and add NK003 or NK004 diluted to different concentrations as obtained in Example 4: 0.001 ⁇ g/ml, 0.005 ⁇ g /ml, 0.01 ⁇ g/ml, 0.05 ⁇ g/ml, 0.1 ⁇ g/ml, 0.5 ⁇ g/ml, 1 ⁇ g/ml, 5 ⁇ g/ml and 10 ⁇ g/ml, 10 ⁇ g/ml, using HEL antibody as a negative control; for cross-linking, Each group was added with a 2.5 ⁇ g/ml secondary antibody, namely goat anti-human Fc.
- a 2.5 ⁇ g/ml secondary antibody namely goat anti-human Fc.
- NK004 on CD40 does not depend on the cross-linking of the secondary antibody, and activates the NF- ⁇ B-GFP+hCD40 reporter cell line in a constitutive mode.
- the EC50 when the secondary antibody is added is 4Nm ( Figure 7B).
- Biacore T200 (GE Healthcare) is used to detect the affinity of antibodies NK003 and NK004. Flow NK003 and NK004 through the Protein A chip at 10 ⁇ L/min and capture them on the chip.
- Use Running buffer HBS-EP+, GE
- the concentration is set to 2 ⁇ M, 1 ⁇ M, 500nM, 250nM, 125nM, 62.5nM, 31.25nM.
- the above-mentioned CD40 of different concentrations was flowed through the antibody-captured chip at a flow rate of 30 ⁇ L/min, combined for 120s, and then the chip was flowed through the Running buffer at a flow rate of 30 ⁇ L/min for 240s, and the antigen gradually dissociated from the antibody-captured chip.
- Data processing was performed using BIAevaluation software S200, which is the supporting software of Biacore T200 instrument, to calculate the binding constant (Ka), dissociation constant (Kd) and equilibrium dissociation constant (KD). The results are shown in Table 2.
- the binding of 293FT cells expressing rhesus and human CD40 to NK003 was detected by flow cytometry, and the cross-reactivity of NK003 with rhesus and human CD40 was determined.
- the CDS regions of rhesus monkey CD40 (NCBI, NM_001265862.1) and human CD40 (NCBI, NM_001250.5) were cloned into pCDH vector, and were transiently transfected into 293FT cells with PEI using standard procedures. After 6 hours of staining, the medium was changed and cultured in DMEM medium containing 10% fetal bovine serum (Biological Industries, 04-001-1A) at 37°C. After 48 hours of expression, cells expressing rhesus monkey CD40 and cells expressing human CD40 were obtained. .
- NK003 diluted to different concentrations with PBS prepared as described in Example 4: 0.001 ⁇ g/ml, 0.005 ⁇ g /ml, 0.01 ⁇ g/ml, 0.05 ⁇ g/ml, 0.1 ⁇ g/ml, 0.5 ⁇ g/ml, 1 ⁇ g/ml, 5 ⁇ g/ml and 10 ⁇ g/ml, HEL 10 ⁇ g/ml as a negative control, the cells and different concentrations of NK003 4 Incubate at °C for 30 min, wash with PBS three times, add Alexa Fluro 488 goat anti-human Fc fluorescent secondary antibody (Lifetechnologies, A11013) at 1:100, incubate at 4°C for 30 min in the dark, and analyze by flow cytometer.
- GraphPad Prism 6.0 was used to fit the curve and calculate the EC50. The results are shown in Figure 8. It can be seen that NK003 cross-reacts with rhesus CD40 (Figure 8B) and human CD40 ( Figure 8A). The EC50 combined with rhesus CD40 is 8nM , And human CD40 binding EC50 is 10nM.
- Lymphoma cells Raji and Ramos have CD40 expression on the membrane surface.
- Raji cells ATCC, CRL-7936
- Ramos cells ATCC, CRL-1596
- Raji and Ramos cells were seeded on a 24-well plate at a density of 3 ⁇ 10 5 /mL, cultured in RPMI 1640 medium containing 10% fetal bovine serum (Lifetechnologies, 10091148), and the cells were seeded at the same time as in Example 4 above
- the prepared PBS is diluted to different concentrations of NK003: 0.03 ⁇ g/ml, 0.06 ⁇ g/ml, 0.09 ⁇ g/ml, 0.3 ⁇ g/ml, 0.6 ⁇ g/ml, 0.9 ⁇ g/ml, 3 ⁇ g/ml, 6 ⁇ g/ml and 9 ⁇ g/ml, HEL9 ⁇ g/ml were used as negative controls, and goat anti-human Fc was added as a crosslinking agent at a concentration of 2.5 ⁇ g/ml to each well.
- NK003 promoted the apoptosis of Raji with an EC50 of 2.6 nM ( Figure 9A); NK003 promoted the apoptosis of Ramos cells with an EC50 of 3 nM ( Figure 9B).
- PBMC Human peripheral blood mononuclear cells
- NK003 to activate dendritic cells
- the isolation of DC cells is a conventional method, specifically as follows: PBMC (Hemacare, PB009C-3) is cultured in RPMI 1640 medium containing 10% fetal bovine serum (Lifetechnologies, 10091148) at 37°C, and adherent mononuclei are harvested 6 hours later Cells; remove the supernatant, add serum-free RPMI1640 medium containing 100ng/mL GM-CSF (R&D Systems, 204-IL) and 10ng/mL IL4 (R&D Systems, 215-GM), and culture at 37°C; three days later Replace half of the medium, supplement GM-CSF and IL4 to 100ng/mL and 10ng/mL; on the sixth day of culture, change the suspended cells to a new culture flask, and use the same 100ng/mL GM-CSF and 10ng /mL
- the DC cells obtained by the above induction were seeded in a 96-well plate at 1 ⁇ 10 5 cells/well, and different concentrations of NK003 were added: 0.01 ⁇ g/ml, 0.05 ⁇ g/ml, 0.1 ⁇ g/ml, 0.5 ⁇ g/ml, 1 ⁇ g/ml, 10 ⁇ g/ml and 100 ⁇ g/ml, 100 ⁇ g/ml HEL were used as negative controls; for cross-linking, each group was added 2.5 ⁇ g/ml goat anti-human Fc at the same time.
- NK003 can significantly activate human DC cells, with an EC50 of 2nM ( Figure 10A), and an EC50 of 8nM after adding crosslinker (goat anti-human Fc, anti-Fc) ( Figure 10B) And the cross-linked form of NK003 activates DC with a significantly stronger strength than uncross-linked (MFI is significantly higher).
- PBMC Human peripheral blood mononuclear cells
- CD19 immunomagnetic bead kit (Miltenyi Biotec, 130-050-301) was used to sort CD19 + B cells from human PBMC (Hemacare, PB009C-3) according to the kit instructions.
- the B cells obtained by the above sorting were cultured in RPMI1640 medium containing 10% fetal bovine serum (Lifetechnologies, 10091148), and 10ng/mL IL4 was added. Inoculate B cells in a 96-well plate at a rate of 1 ⁇ 10 5 cells/well, and add different concentrations of NK003: 0.01 ⁇ g/ml, 0.05 ⁇ g/ml, 0.1 ⁇ g/ml, 0.5 ⁇ g/ml, 1 ⁇ g/ml, 10 ⁇ g /ml and 100 ⁇ g/ml, 100 ⁇ g/ml HEL as a negative control.
- NK003 can promote the proliferation of human B cells with an EC50 of 4 nM.
- PBMC peripheral blood mononuclear cells
- CD19 + B cells were sorted from human PBMC using CD19 immunomagnetic bead kit.
- the B cells obtained by the above sorting were cultured in RPMI 1640 medium containing 10% fetal bovine serum (Lifetechnologies, 10091148). Inoculate B cells in a 96-well plate at a rate of 1 ⁇ 10 5 cells/well, and add different concentrations of NK003: 0.01 ⁇ g/ml, 0.05 ⁇ g/ml, 0.1 ⁇ g/ml, 0.5 ⁇ g/ml, 1 ⁇ g/ml, 10 ⁇ g /ml and 100 ⁇ g/ml, 100 ⁇ g/ml HEL as a negative control.
- EC50 is 70Nm without crosslinking agent (goat anti-human Fc, anti-Fc) ( Figure 12A), and EC50 is 4nM after crosslinking agent ( Figure 12A). 12B), and the cross-linked form of NK003 activates B cells with significantly stronger strength than uncross-linked (significantly higher CD86 expression).
- Antibody-dependent cell-mediated cytotoxicity refers to that the Fab fragment of an antibody binds to the epitope of tumor cells, and its Fc fragment binds to the FcR on the surface of killer cells (NK cells, macrophages, etc.) to mediate killing The cell directly kills the target cell.
- NK cells killer cells
- SCID mice to inoculate tumor cells expressing CD40.
- SCID mice Eight five-week-old male SCID mice were purchased from Weitong Lihua and raised in an SPF animal room. After one week of feeding, they were randomly divided into two groups. The back was inoculated with Raji cells subcutaneously in the number of 2.5 ⁇ 10 6 cells per mouse. Four cells in each group were inoculated. The day of inoculation was recorded as Day 0. The first group was administered NK003 antibody, and the second group was administered HEL control antibody.
- NK003 activates the immune system in CD40 humanized mice
- the CD40 humanized mouse model was constructed and provided by Li Fubin of Shanghai Jiaotong University, and was kept in an SPF animal room.
- OT-1 mice were provided by Li Fubin of Shanghai Jiaotong University and were kept in an SPF animal room. The OT-1 mice were killed by cervical dislocation and their spleens were removed, and the spleen was crushed to collect spleen cells.
- Use the immunomagnetic bead kit (R&D Systems, MAGM203) to isolate CD8 + T cells according to the kit instructions, that is, OT-1CD8 + T cells.
- mice Eight-week-old male CD40 humanized mice were selected to be injected with OT-1 CD8 + T cells in the tail vein at the number of 2 ⁇ 10 6 /mouse, and 0.1 mg/kg DEC-OVA (Sigma-Aldrich, SAB4700735) and 5mg/kg NK003 (the same dose of HEL is the control antibody). Seven days after cell inoculation, the mice were sacrificed by cervical spine method to separate their spleens. Grind the spleen to obtain a single cell suspension.
- CD45.1 + CD8 + TCR-V ⁇ 2 + subgroup is OT-1CD8 + T cells (OT1 cells).
- the CD45.1 + CD8 + subpopulation is CD8 cells, and the CD45.1 + CD4 + subpopulation is CD4 cells.
- the results are shown in Figure 14. The proportion of OT-1CD8 + T cells in the NK003 group increased significantly, the number of cells increased significantly, and the ratio of CD8 to CD4 increased correspondingly. NK003 can activate CD40 humanized mouse immune system.
- NK003 inhibits tumor growth in CD40 humanized mice
- CD40 humanized mice constructed and provided by Li Fubin of Shanghai Jiaotong University as described in 7.2 to carry the tumor cell MC38 (Institute of Basic Medicine, Chinese Academy of Medical Sciences, Basic Medical Cell Center, 3111C0001CCC000523) inoculation.
- the positive control anti-CD40 antibody CP870893 sequence used herein comes from patent US 7338660 (see SEQ ID NO: 46 and SEQ ID NO: 48 for light chain and heavy chain sequences). Transfect the light chain and heavy chain of CP870893 synthesized by Jin Weizhi into pFUSE vector, and transiently co-transfect 293F suspension cells at 1:1 to express the full-length antibody. After 1 week of expression, use Superdex TM 200 Increase pre-installed on the AKTA system Column purification (the specific steps are the same as those described in Example 4).
- mice Eight-week-old male CD40 humanized mice were subcutaneously inoculated with MC38 cells at the back of 2 ⁇ 10 6 /mouse.
- Tumor inhibition rate (average volume of control group-average volume of experimental group)/average volume of control group ⁇ 100%.
- NK003-V12, NK003-S267E/L328F Fc region mutants increase binding to Fc ⁇ RIIB and CD40 agonist activity
- glutamic acid (E) at residue 233 in the Fc region of NK003 was mutated to aspartic acid (D)
- residue at 237 Glycine (G) was mutated to aspartic acid (D)
- histidine (H) at residue 268 was mutated to aspartic acid (D)
- proline (P) at residue 271 was mutated to glycine (G)
- the 330th residue alanine (A) was mutated to arginine (R), resulting in the NK003-V12 mutant.
- NK003Fc region IgG1, EU numbering
- E glutamic acid
- L 328th residue Leucine
- F phenylalanine
- the light and heavy chains of the NK003-V12 and NK003-S267E/L328F mutants synthesized by Jin Weizhi were transfected into the pFUSE vector according to 1: 1 Transient co-transfection of 293F suspension cells to express the full-length antibody, and after one week of expression, it was purified on the AKTA system using a Superdex TM 200Increase prepacked column (the specific steps are the same as those described in Example 4).
- the Fc ⁇ RIIA (NCBI, NM_001136219.1) and Fc ⁇ RIIB (NCBI, NM_004001.4) CDS regions synthesized by Jin Weizhi were cloned into pCDH vector, and transiently transfected into 293FT cells with PEI using standard procedures, and the medium was changed after 6 hours of transfection. It was cultured in DMEM medium containing 10% fetal bovine serum (Biological Industries, 04-001-1A) at 37°C. After 48 hours of expression, cells expressing Fc ⁇ RIIA and 293FT cells expressing Fc ⁇ RIIB were obtained.
- NK003, NK003-V12 and NK003-S267E/L328F were used as negative controls; after co-cultivation at 37°C for 24 hours, washed with PBS three times and analyzed by flow cytometry to detect the MFI of GFP. The results are shown in Figure 16. Compared with wild-type NK003, NK003-V12, NK003-S267E/L328F have significantly enhanced agonist activity.
- a mutation library was constructed for the complementarity determining regions and framework regions of NK003 VH and VL, and inserted into the pcomb3 phage vector (Biovector Inc. 108925) in the form of Fab ( Figure 17).
- the phage affinity maturation method was used to complete the optimization of NK003.
- the light chain VL and CL fragments were amplified using the primer pair VL1/VL2, CL1/CL2, respectively. Reaction conditions: 95°C for 2min, 1cycle; 95°C for 30s, 65°C for 30s, 72°C for 10s, 35cycle; 72°C for 5min, 1cycle. Use Tiangen Recovery Kit to recover PCR target fragments.
- the primers are as follows:
- VL1 5’-GCGGCCGAGCTCGATGTTGTGATGACTCAG-3’
- VL2 5'-GGTCCCCTGGCCAAA AGT GTA CGG AGT TCA TAG ACC TTG CAT
- NK003 light chain mutation library The above two parts of PCR products were subjected to overlap PCR amplification to obtain the NK003 light chain mutation library. Reaction conditions: 95°C 2min, 1cycle; 95°C 30s, 65°C 30s, 72°C 20s, 8cycle; adding VL1/CL2 primer, 95°C 30s, 65°C 30s, 72°C 20s, 27cycle; 72°C 5min, 1cycle. SacI (NEB, R3156L) and PacI (NEB, R0547L) were used to digest the NK003 light chain mutation library and pcomb3 vector with double restriction enzymes, and the Tiangen recovery kit was used to recover the PCR target fragments.
- the mutation library gene and the carrier were ligated at a molar ratio of 3:1 with T4 ligase (NEB, M0202L) at 25°C for 3 hours.
- the ligation product was transferred to the XL1-Blue electroporation competence by electroporation method, and the library capacity was 5 ⁇ 10 5 light chain mutation library, named pcomb3-NK003-LCDR3.
- VH1/VH2 and CH1-1/CH1-2 heavy chain fragments were amplified using the primer pairs VH1/VH2, CH1-1/CH1-2, respectively. Reaction conditions: 95°C for 2min, 1cycle; 95°C for 30s, 65°C for 30s, 72°C for 10s, 35cycle; 72°C for 5min, 1cycle.
- the primers are as follows:
- VH1 5’-TGCAGCTGCTCGAGCAGGTACAGCTGGTGCAGTC-3’
- VH2 5'-CTTTGCCCCAGACGTC CAT GTA GTA GTA GTA GTA GTA GGT TCA AGT AGC TCC CAC TCT TTC TCTCGCACAG-3' (two random mutations at the horizontal line to MNN, high-throughput synthesis by Jinweizhi)
- CH1-1 5’-GACGTCTGGGGCAAAGGGACCACGGTC-3’
- the above two parts of PCR products were amplified by overlap PCR to obtain the NK003 heavy chain mutation library.
- the reaction conditions 95°C 2min, 1cycle; 95°C 30s, 65°C 30s, 72°C 20s, 8cycle; add VH1/CH1-2 primer, 95 °C30s, 65°C30s, 72°C20s, 27cycle; 72°C5min, 1cycle.
- SpeI NEB, R3133L
- XhoI NEB, R0146L
- the mutant library gene and the carrier were ligated at a molar ratio of 3:1 with T4 ligase at 25°C for 3h.
- the ligation product was transferred to XL1-Blue electrocompetence by electroporation, and the library capacity was 2.4 ⁇ 10 7 light and heavy chain double mutation library, named pcomb3-NK003-AM.
- the phage eluted in the third round was infected with XL1-blue, and then coated with a plate. After mixing with a scraper, the NK003-AM plasmid was extracted using the Tiangen plasmid small extraction kit. After double digestion with SacI and SpeI, the target fragment of about 1500 bp was recovered and sent to Jinweizhi for third-generation sequencing.
- the sequencing results are shown in Figure 19.
- the heavy chain and light chain DNA of antibodies NK003-AM-9 and NK003-AM-18 (Jin Weizhi) selected were synthesized and cloned into the vector pFUSE.
- the specific method of purification and expression is shown in Example 4.
- flow cytometry was used to detect the activation of the NF- ⁇ B-GFP+hCD40 reporter cell line by NK003-AM-9 and NK003-AM-18 antibodies.
- the specific steps are as follows:
- NK003-AM-9, NK003-AM-18 and NK003 0.01 ⁇ g/ml, 0.05 ⁇ g/ml, 0.1 ⁇ g/ml, 0.5 ⁇ g/ml and 1 ⁇ g/ml; for cross-linking, each group was added with a 2.5 ⁇ g/ml secondary antibody at the same time, namely goat anti-human Fc (SouthernBiotech, SBA-2048-01).
- NK003-AM-9 and NK003-AM-18 activate the NF- ⁇ B-GFP+hCD40 reporter cell line in a cross-linked form.
- the EC50 is 0.25nM and 0.3nM, respectively. The activities of the two are similar, and both are more similar than the wild type. NK003 improved by nearly 10 times.
- NK003 light chain DNA (SEQ ID NO: 74) as template, using random mutation PCR kit (Agilent Technologies, 200550) and primer pairs VL1/VL2, VL3/VL4 to carry out Nested PCR amplification of light chain VL region, reaction conditions :95°C2min, 1cycle; 95°C30s, 65°C30s, 72°C30s, 28cycle; 72°C10min, 1cycle.
- primer pair CL1/CL2 was used to amplify light chain CL region, reaction conditions: 95°C 2min, 1cycle; 95°C 30s, 65°C 30s, 72°C 30s, 30cycle ; 72°C5min, 1cycle.
- the primers are as follows:
- VL1 5’-CTATCGCGATTGCAGTGGCACTGGCTG-3’
- VL2 5’-CCAGATTTCAACTGCTCATCAGATGGC-3’
- VL3 5’-CTACCGTGGCCCAGGCGGCCGAGCTC-3’
- VL4 5’-GAAGACAGATGGTGCAGCCACAGTTCG-3’
- the VL and CL PCR products were subjected to overlap PCR amplification to obtain the NK003 light chain mutation library. Reaction conditions: 95°C for 2min, 1cycle; 95°C for 30s, 65°C for 30s, 72°C for 40s, 8cycle; adding VL3/CL2 primer, 95°C for 30s, 65°C for 30s, 72°C for 40s, 27cycle; 72°C for 10min, 1cycle.
- the NK003 light chain mutation library and pcomb3 vector were double digested with SacI and PacI, and the target fragments of PCR were recovered using the Tiangen recovery kit.
- the mutant library gene and the carrier were ligated at a molar ratio of 3:1 with T4 ligase at 25°C for 3h.
- the ligation product was transferred to XL1-Blue electrocompetent state by electroporation, and the library capacity was 1 ⁇ 10 5 light chain mutation library, named pcomb3-NK003-VL.
- NK003 heavy chain DNA (SEQ ID NO: 75) as a template, using primer pairs VH1/VH2, VH3/VH4 to perform Nested PCR amplification of the heavy chain VH region, reaction conditions 3: 95°C 2min, 1 cycle; 95°C 30s, 65°C30s, 72°C30s, 28cycle; 72°C10min, 1cycle.
- the primer pair CH1-1/CH1-2 was used to amplify the heavy chain CH1 region, reaction conditions: 95°C 2min, 1 cycle; 95°C 30s, 65°C 30s, 72 °C30s, 32cycle; 72°C10min, 1cycle.
- Use Tiangen Recovery Kit to recover PCR target fragments.
- the primers are as follows:
- VH1 5’-GCCGCTGGATTGTTATTACTCGCTGC-3’
- VH2 5’-CAGAGGTGCTCTTGGAGGAGGGTGCC-3’
- VH3 5’-GCCATGGCCGAGGTGCAGCTGCTCGAG-3’
- VH4 5’-GAAGACCGATGGGCCCTTGGTGGAGGC-3’
- CH1-1 5’-GCCTCCACCAAGGGCCCATCGGTCTTC-3’
- the above-mentioned VH and CH1 PCR products were subjected to overlap PCR amplification to obtain the NK003 heavy chain mutation library.
- Reaction conditions 95°C2min, 1cycle; 95°C30s, 65°C30s, 72°C40s, 8cycle; adding VH3/CH1-2 primer, 95°C30s, 65°C30s, 72°C40s, 27cycle; 72°C10min, 1cycle .
- the NK003 heavy chain mutation library and pcomb3-NK003-VL vector were double-enzyme digested with SpeI and XhoI, and the target fragments of PCR were recovered using Tiangen Recovery Kit.
- the mutant library gene and the carrier were ligated at a molar ratio of 3:1 with T4 ligase at 25°C for 3h.
- the ligated product was transferred to XL1-Blue electrocompetent state by electroporation method, and the library capacity was 2.4 ⁇ 10 7 light and heavy chain double mutation library, named pcomb3-NK003-EP.
- the phage eluted in the third round was infected with XL1-blue, and then coated with a plate. After mixing with a scraper, the NK003-EP plasmid was extracted using the Tiangen plasmid small extraction kit. After double digestion with SacI and SpeI, the target fragment of about 1500 bp was recovered and sent to Jinweizhi for third-generation sequencing. The sequencing results are shown in Figure 21. According to the results of the third-generation sequencing, the antibody NK003-AM-18-EP1 heavy chain DNA (Jin Weizhi) was synthesized and cloned into the vector pFUSE.
- NK003-AM-18-EP1 The plasmids containing the light chain of NK003-AM-18 and the heavy chain of NK003-AM-18-EP1 were transiently co-transfected into 293F suspension cells with PEI using standard procedures to express the full-length NK003-AM- 18-EP1 antibody. See Example 4 for specific methods of purification and expression. See sequence list 6 for NK003-AM-18-EP1 VH sequence.
- flow cytometry was used to detect the activation of the NF- ⁇ B-GFP+hCD40 reporter cell line by the NK003-AM-18-EP1 antibody.
- the specific steps are as follows:
- NK003-AM-18, NK003-AM-18-EP1 and NK003 0.0075nM, 0.025 nM, 0.075nM, 0.25nM, 0.75nM, 2.5nM and 7.5nM; for cross-linking, each group was added with a 2.5 ⁇ g/ml secondary antibody at the same time, namely goat anti-human Fc.
- NK003-AM-18-EP1 activates the NF- ⁇ B-GFP+hCD40 reporter cell line in a cross-linked form with an EC50 of 0.35 nM, which is similar to the activity of NK003-AM-18.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Immunology (AREA)
- Organic Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Engineering & Computer Science (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Molecular Biology (AREA)
- Genetics & Genomics (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Endocrinology (AREA)
- General Chemical & Material Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Peptides Or Proteins (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Preparation Of Compounds By Using Micro-Organisms (AREA)
Abstract
Description
原始残基 | 示例性置换 | 优选的保守氨基酸置换 |
Ala(A) | Val、Leu、Ile | Val |
Arg(R) | Lys、Gln、Asn | Lys |
Asn(N) | Gln、His、Asp、Lys、Arg | Gln |
Asp(D) | Glu、Asn | Glu |
Cys(C) | Ser、Ala | Ser |
Gln(Q) | Asn、Glu | Asn |
Glu(E) | Asp、Gln | Asp |
Gly(G) | Ala | Ala |
His(H) | Asn、Gln、Lys、Arg | Arg |
Ile(I) | Leu、Val、Met、Ala、Phe、正亮氨酸 | Leu |
Leu(L) | 正亮氨酸、Ile、Val、Met、Ala、Phe | Ile |
Lys(K) | Arg、Gln、Asn | Arg |
Met(M) | Leu、Phe、Ile | Leu |
Phe(F) | Trp、Leu、Val、Ile、Ala、Tyr | Tyr |
Pro(P) | Ala | Ala |
Ser(S) | Thr | Thr |
Thr(T) | Val、Ser | Ser |
Trp(W) | Tyr、Phe | Tyr |
Tyr(Y) | Trp、Phe、Thr、Ser | Phe |
Val(V) | Ile、Leu、Met、Phe、Ala、正亮氨酸 | Leu |
传感器 | 抗体 | 抗原 | ka(1/Ms) | kd(1/s) | K D(M) |
Protein A | NK003 | CD40 | 3.67E+04 | 1.21E-02 | 3.31E-07 |
Protein A | NK004 | CD40 | 8.87E+04 | 3.90E-02 | 4.40E-07 |
Claims (22)
- 结合CD40的抗体或其抗原结合片段,其包含(i)如SEQ ID NO:13、58、60或62所示的重链可变区的3个互补决定区HCDR1、HCDR2和HCDR3,以及如SEQ ID NO:15、64或66所示的轻链可变区的3个互补决定区LCDR1、LCDR2和LCDR3;(ii)如SEQ ID NO:13所示的重链可变区的3个互补决定区HCDR1、HCDR2和HCDR3,以及如SEQ ID NO:15所示的轻链可变区的3个互补决定区LCDR1、LCDR2和LCDR3;(iii)如SEQ ID NO:58所示的重链可变区的3个互补决定区HCDR1、HCDR2和HCDR3,以及如SEQ ID NO:64所示的轻链可变区的3个互补决定区LCDR1、LCDR2和LCDR3;(iv)如SEQ ID NO:60或62所示的重链可变区的3个互补决定区HCDR1、HCDR2和HCDR3,以及如SEQ ID NO:66所示的轻链可变区的3个互补决定区LCDR1、LCDR2和LCDR3;(v)如SEQ ID NO:14所示的重链可变区的3个互补决定区HCDR1、HCDR2和HCDR3,以及如SEQ ID NO:16所示的轻链可变区的3个互补决定区LCDR1、LCDR2和LCDR3;(vi)(i)-(v)中任一项的CDR组合,其中相比HCDR3和/或LCDR3,在所述序列上包含至少一个且不超过3、2或1个氨基酸改变(优选地氨基酸置换,优选地保守置换)。
- 结合CD40的抗体或其抗原结合片段,其包含(i)包含或由下述序列组成的HCDR1、HCDR2和HCDR3:SEQ ID NO:1、SEQ ID NO:3和SEQ ID NO:5;或者(ii)包含或由下述序列组成的HCDR1、HCDR2和HCDR3:SEQ ID NO:1、SEQ ID NO:3和SEQ ID NO:51;或者(iii)包含或由下述序列组成的HCDR1、HCDR2和HCDR3:SEQ ID NO:1、SEQ ID NO:3和SEQ ID NO:52;或者(iv)(i)-(iii)中任一项的HCDR组合,其中在所述HCDR3包含至少一个且不超过3、2或1个氨基酸改变(优选氨基酸置换,优选保守置换);和(i)包含或由下述序列组成的LCDR1、LCDR2和LCDR3:SEQ ID NO:7、SEQ ID NO:9和SEQ ID NO:11;或者(ii)包含或由下述序列组成的LCDR1、LCDR2和LCDR3:SEQ ID NO:7、SEQ ID NO:9和SEQ ID NO:53;或者(iii)包含或由下述序列组成的LCDR1、LCDR2和LCDR3:SEQ ID NO:7、SEQ ID NO:9和SEQ ID NO:54;或者(iv)(i)-(iii)中任一项的LCDR组合,其中在所述LCDR3包含至少一个且不超过3、2或1 个氨基酸改变(优选氨基酸置换,优选保守置换)。
- 结合CD40的抗体或其抗原结合片段,其包含(i)包含或由下述序列组成的HCDR1、HCDR2和HCDR3:SEQ ID NO:1、SEQ ID NO:3和SEQ ID NO:5,以及包含或由下述序列组成的LCDR1、LCDR2和LCDR3:SEQ ID NO:7、SEQ ID NO:9和SEQ ID NO:11;或者(ii)包含或由下述序列组成的HCDR1、HCDR2和HCDR3:SEQ ID NO:1、SEQ ID NO:3和SEQ ID NO:51,以及包含或由下述序列组成的LCDR1、LCDR2和LCDR3:SEQ ID NO:7、SEQ ID NO:9和SEQ ID NO:53;或者(iii)包含或由下述序列组成的HCDR1、HCDR2和HCDR3:SEQ ID NO:1、SEQ ID NO:3和SEQ ID NO:52,以及包含或由下述序列组成的LCDR1、LCDR2和LCDR3:SEQ ID NO:7、SEQ ID NO:9和SEQ ID NO:54;或者(iv)包含或由下述序列组成的HCDR1、HCDR2和HCDR3:SEQ ID NO:2、SEQ ID NO:4和SEQ ID NO:6,以及包含或由下述序列组成的LCDR1、LCDR2和LCDR3:SEQ ID NO:8、SEQ ID NO:10和SEQ ID NO:12;(v)包含或由下述序列组成的HCDR1、HCDR2和HCDR3:SEQ ID NO:1、SEQ ID NO:3和SEQ ID NO:55,以及包含或由下述序列组成的LCDR1、LCDR2和LCDR3:SEQ ID NO:7、SEQ ID NO:9和SEQ ID NO:56;(vi)(i)-(iv)中任一项的HCDR和LCDR组合,其中在所述HCDR3和/或LCDR3中包含至少一个且不超过3、2或1个氨基酸改变(优选氨基酸置换,优选保守置换)。
- 权利要求1至3中任一项的抗体或其抗原结合片段,其包含重链可变区VH和/或轻链可变区VL,其中,(a)重链可变区VH(i)包含与选自SEQ ID NO:13、58、60、62或14的氨基酸序列具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的氨基酸序列或由其组成;或者(ii)包含选自SEQ ID NO:13、58、60、62或14的氨基酸序列或由其组成;或者(iii)包含与选自SEQ ID NO:13、58、60、62或14的氨基酸序列相比具有1个或多个(优选不超过10个,更优选不超过5、4、3、2、1个)的氨基酸改变(优选氨基酸置换,更优选氨基酸保守置换)的氨基酸序列,优选地,所述氨基酸改变不发生在CDR区中,优选地,所述氨基酸改变发生在FR区中;和/或(b)轻链可变区VL(i)包含与选自SEQ ID NO:15、64、66或16的氨基酸序列具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的氨基酸序列或由其组成;(ii)包含选自SEQ ID NO:15、64、66或16的氨基酸序列或由其组成;或者(iii)包含与选自SEQ ID NO:15、64、66或16的氨基酸序列相比具有1个或多个(优选不超过10个,更优选不超过5、4、3、2、1个)的氨基酸改变(优选氨基酸置换,更优选氨基酸保守置换)的氨基酸序列,优选地,所述氨基酸改变不发生在CDR区中,优选地,所述 氨基酸改变发生在FR区中。
- 权利要求4的抗体或其抗原结合片段,其中(i)重链可变区VH包含SEQ ID NO:13、58、60或62的氨基酸序列或由其组成,和轻链可变区VL包含选自SEQ ID NO:15、64或66的氨基酸序列或由其组成;(ii)重链可变区VH包含SEQ ID NO:13的氨基酸序列或由其组成,和轻链可变区VL包含SEQ ID NO:15的氨基酸序列或由其组成;(iii)重链可变区VH包含SEQ ID NO:58的氨基酸序列或由其组成,和轻链可变区VL包含SEQ ID NO:64的氨基酸序列或由其组成;(iv)重链可变区VH包含SEQ ID NO:60的氨基酸序列或由其组成,和轻链可变区VL包含SEQ ID NO:66的氨基酸序列或由其组成;(v)重链可变区VH包含SEQ ID NO:62的氨基酸序列或由其组成,和轻链可变区VL包含SEQ ID NO:66的氨基酸序列或由其组成;(vi)重链可变区VH包含SEQ ID NO:14的氨基酸序列或由其组成,和轻链可变区VL包含SEQ ID NO:16的氨基酸序列或由其组成。
- 权利要求1至5中任一项的抗体或其抗原结合片段,其包含重链和/或轻链,其中(a)重链(i)包含与选自SEQ ID NO:17、18、19、67、69、70或20的氨基酸序列具有至少85%、90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的氨基酸序列或由其组成;(ii)包含选自SEQ ID NO:17、18、19、67、69、70或20的氨基酸序列或由其组成;或者(iii)包含与选自SEQ ID NO:17、18、19、67、69、70或20的氨基酸序列相比具有1个或多个(优选不超过20个或10个,更优选不超过5、4、3、2、1个)的氨基酸改变(优选氨基酸置换,更优选氨基酸保守置换)的氨基酸序列,优选地,所述氨基酸改变不发生在重链的CDR区中,更优选地,所述氨基酸改变不发生在重链可变区中,最优选地,所述氨基酸改变发生在重链恒定区中;和/或(b)轻链(i)包含与选自SEQ ID NO:21、68、71或22的氨基酸序列具有至少85%、90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的氨基酸序列或由其组成;(ii)包含选自SEQ ID NO:21、68、71或22的氨基酸序列或由其组成;或者(iii)包含与选自SEQ ID NO:21、68、71或22的氨基酸序列相比具有1个或多个(优选不超过20个或10个,更优选不超过5、4、3、2、1个)的氨基酸改变(优选氨基酸置换,更优选氨基酸保守置换)的氨基酸序列,优选地,所述氨基酸改变不发生在轻链的CDR区中,更优选地,所述氨基酸改变不发生在轻链可变区中,最优选的,所述重链氨基酸改变发生在轻链恒定区中。
- 权利要求6的抗体或其抗原结合片段,其中(i)重链包含SEQ ID NO:17、18、19、67、69或70的氨基酸序列或由其组成,和轻链包含SEQ ID NO:21、68或71的氨基酸序列或由其组成,(ii)重链包含SEQ ID NO:17、18或19的氨基酸序列或由其组成,和轻链包含SEQ ID NO:21的氨基酸序列或由其组成;(iii)重链包含SEQ ID NO:67的氨基酸序列或由其组成,和轻链包含SEQ ID NO:68的氨基酸序列或由其组成;(iv)重链包含SEQ ID NO:69或70的氨基酸序列或由其组成,和轻链包含SEQ ID NO:71的氨基酸序列或由其组成;(v)重链包含SEQ ID NO:20的氨基酸序列或由其组成,和轻链包含SEQ ID NO:22的氨基酸序列或由其组成。
- 权利要求1至7中任一项的结合CD40的抗体或其抗原结合片段,其中所述抗体是单克隆抗体。
- 权利要求1至8中任一项的结合CD40的抗体或其抗原结合片段,其中所述抗体是人源化的抗体或人抗体或嵌合抗体。
- 权利要求1至9中任一项的抗体或其抗原结合片段,其中所述抗原结合片段是选自以下的抗体片段:Fab、Fab’、Fab’-SH、Fv、单链抗体(例如scFv)或(Fab’) 2、单结构域抗体、diabody(dAb)或线性抗体。
- 分离的核酸,其编码权利要求1至10中任一项的结合CD40的抗体的任一或多条链或其抗原结合片段。
- 载体,其包含权利要求11的核酸,优选地所述载体是表达载体。
- 宿主细胞,其包含权利要求11的核酸或权利要求12的载体,优选地,所述宿主细胞是原核的或真核的,更优选的选自酵母细胞、哺乳动物细胞(例如所述宿主细胞是CHO细胞,例如CHOS细胞CHOK1SV细胞或CHOK1SV GS-KO,或所述宿主细胞是293细胞,例如HEK293细胞)或适用于制备抗体或其抗原结合片段的其它细胞。
- 制备结合CD40的抗体或其抗原结合片段的方法,所述方法包括在适于表达抗体的情况下,培养包含编码权利要求1-10中任一项的抗体或其抗原结合片段的核酸的宿主细胞,任选地所述方法还包括从所述宿主细胞回收所述抗体或其抗原结合片段。
- 免疫缀合物,其包含权利要求1至10中任一项的结合CD40的抗体或其抗原结合片段和其它物质,例如治疗剂或标记。
- 药物组合物,其包含权利要求1至10中任一项的结合CD40的抗体或其抗原结合片段或权利要求13的免疫缀合物,以及任选地一种或多种其它治疗剂,以及任选地药用辅料。
- 组合产品,其包含权利要求1至10中任一项的结合CD40的抗体或其抗原结合片段 或权利要求15的免疫缀合物,以及一种或多种其它治疗剂。
- 权利要求15的免疫缀合物、权利要求16的药物组合物或权利要求17的组合产品,其中所述治疗剂选自化疗剂、其他抗体、细胞毒性剂、疫苗、抗感染活性剂、小分子药物或免疫调节剂。
- 在受试者中增强免疫应答(例如抗原特异性T细胞应答)的方法,其包括向受试者施用有效量的权利要求1-10中任一项的抗体或其抗原结合片段、或权利要求16或18的免疫缀合物、或权利要求16或18的药物组合物,或权利要求17或18的组合产品。
- 预防或治疗受试者的需要在受试者中调节(例如增强)免疫应答的疾病或CD40相关疾病的方法,所述方法包括向所述受试者施用有效量的权利要求1-10中任一项的抗体或其抗原结合片段、或权利要求15或18的免疫缀合物、或权利要求16或18的药物组合物,或权利要求17或18的组合产品,优选地,所述疾病为肿瘤,例如淋巴瘤、结肠癌、结直肠癌、直肠癌、肺癌(例如非小细胞肺癌)、肝癌、胃癌及其转移性癌症;或感染,例如慢性感染,或病毒感染。
- 权利要求20所述的方法,其还包括向所述受试者联合施用一种或多种疗法,例如治疗方式和/或其它治疗剂,优选地,治疗方式包括手术治疗和/或放射疗法,和/或其它治疗剂选自化疗剂、其他抗体、细胞毒性剂、疫苗、抗感染活性剂、小分子药物或免疫调节剂。
- 检测试剂盒,其包含权利要求1-10中任一项的抗体或其抗原结合片段或权利要求15的免疫缀合物。
Priority Applications (7)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US17/594,255 US20220324988A1 (en) | 2019-04-10 | 2020-04-10 | Anti-CD40 antibodies and uses thereof |
CN202080027681.4A CN113728008B (zh) | 2019-04-10 | 2020-04-10 | 抗cd40抗体及其用途 |
EP20786934.8A EP3954705A4 (en) | 2019-04-10 | 2020-04-10 | ANTI-CD40 ANTIBODIES AND ITS USE |
CA3136491A CA3136491A1 (en) | 2019-04-10 | 2020-04-10 | Anti-cd40 antibodies and uses thereof |
AU2020256828A AU2020256828A1 (en) | 2019-04-10 | 2020-04-10 | Anti-CD40 antibody and use thereof |
SG11202111188VA SG11202111188VA (en) | 2019-04-10 | 2020-04-10 | Anti-cd40 antibody and use thereof |
JP2021560225A JP7535060B2 (ja) | 2019-04-10 | 2020-04-10 | 抗cd40抗体およびその使用 |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201910284441.1 | 2019-04-10 | ||
CN201910284441 | 2019-04-10 | ||
CN202010262466 | 2020-04-03 | ||
CN202010262466.4 | 2020-04-03 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2020207470A1 true WO2020207470A1 (zh) | 2020-10-15 |
Family
ID=72752143
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2020/084186 WO2020207470A1 (zh) | 2019-04-10 | 2020-04-10 | 抗cd40抗体及其用途 |
Country Status (9)
Country | Link |
---|---|
US (1) | US20220324988A1 (zh) |
EP (1) | EP3954705A4 (zh) |
JP (1) | JP7535060B2 (zh) |
CN (1) | CN113728008B (zh) |
AU (1) | AU2020256828A1 (zh) |
CA (1) | CA3136491A1 (zh) |
SG (1) | SG11202111188VA (zh) |
TW (1) | TW202103733A (zh) |
WO (1) | WO2020207470A1 (zh) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2022262099A1 (zh) * | 2021-06-17 | 2022-12-22 | 南京蓝盾生物科技有限公司 | 抗cd70内化的抗体、抗体偶联物及其应用 |
Families Citing this family (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR102373590B1 (ko) * | 2020-09-22 | 2022-03-14 | (주)아이엠지티 | 음이온성 고분자를 이용한 신규한 나노 입자, 이의 제조 방법 및 조성물 |
CN117229396A (zh) * | 2022-06-06 | 2023-12-15 | 普米斯生物技术(珠海)有限公司 | 抗cd40抗体及其用途 |
US20240279350A1 (en) * | 2023-02-16 | 2024-08-22 | Sanofi | CD40-Binding Proteins |
Citations (13)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4737456A (en) | 1985-05-09 | 1988-04-12 | Syntex (U.S.A.) Inc. | Reducing interference in ligand-receptor binding assays |
US6326482B1 (en) | 1999-04-23 | 2001-12-04 | Genentech, Inc. | SH2 domain-containing peptides |
US7338660B2 (en) | 2001-11-09 | 2008-03-04 | Abgenix, Inc. | Methods of treating cancer and enhancing immune responses with antibodies that bind CD40 |
US20080248028A1 (en) | 2004-03-24 | 2008-10-09 | Xencor, Inc. | Immunoglobulin Variants Outside the Fc Region |
US7521541B2 (en) | 2004-09-23 | 2009-04-21 | Genetech Inc. | Cysteine engineered antibodies and conjugates |
CN103635488A (zh) * | 2011-04-29 | 2014-03-12 | 埃派斯进有限公司 | 抗-cd40抗体及其使用方法 |
WO2014184545A2 (en) | 2013-05-14 | 2014-11-20 | Isis Innovation Limited | Antibodies |
WO2016177771A1 (en) | 2015-05-04 | 2016-11-10 | Apogenix Ag | Single-chain cd40-receptor agonist proteins |
WO2017004006A1 (en) | 2015-06-29 | 2017-01-05 | Bristol-Myers Squibb Company | Antibodies to cd40 |
WO2017059196A2 (en) * | 2015-09-30 | 2017-04-06 | Janssen Biotech, Inc. | Antagonistic antibodies specifically binding human cd40 and methods of use |
WO2017205742A1 (en) * | 2016-05-27 | 2017-11-30 | Abbvie Biotherapeutics Inc. | Anti-cd40 antibodies and their uses |
CN109069621A (zh) * | 2015-09-04 | 2018-12-21 | 普里玛托普医疗股份有限公司 | 人源化抗-cd40抗体及其用途 |
CN109265552A (zh) * | 2012-10-30 | 2019-01-25 | 埃派斯进有限公司 | 抗-cd40抗体及其使用方法 |
Family Cites Families (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP3283527B1 (en) * | 2015-04-13 | 2021-01-13 | Five Prime Therapeutics, Inc. | Combination therapy for cancer |
EA201890162A1 (ru) * | 2015-06-29 | 2018-07-31 | Бристол-Маерс Сквибб Компани | Антитела к cd40 с повышенной агонистической активностью |
US10865244B2 (en) * | 2016-04-18 | 2020-12-15 | Celldex Therapeutics, Inc. | Nucleic acids encoding agonistic antibodies that bind CD40 |
CN107474136B (zh) * | 2016-06-08 | 2023-03-07 | 上海交通大学医学院 | 增强激动型抗体活性的抗体重链恒定区序列 |
EA202090791A1 (ru) * | 2017-09-19 | 2020-06-17 | Мэб Дискавери Гмбх | Агонистические антитела против cd40 |
-
2020
- 2020-04-10 US US17/594,255 patent/US20220324988A1/en active Pending
- 2020-04-10 TW TW109112187A patent/TW202103733A/zh unknown
- 2020-04-10 WO PCT/CN2020/084186 patent/WO2020207470A1/zh unknown
- 2020-04-10 JP JP2021560225A patent/JP7535060B2/ja active Active
- 2020-04-10 SG SG11202111188VA patent/SG11202111188VA/en unknown
- 2020-04-10 CN CN202080027681.4A patent/CN113728008B/zh active Active
- 2020-04-10 CA CA3136491A patent/CA3136491A1/en active Pending
- 2020-04-10 EP EP20786934.8A patent/EP3954705A4/en active Pending
- 2020-04-10 AU AU2020256828A patent/AU2020256828A1/en active Pending
Patent Citations (14)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4737456A (en) | 1985-05-09 | 1988-04-12 | Syntex (U.S.A.) Inc. | Reducing interference in ligand-receptor binding assays |
US6326482B1 (en) | 1999-04-23 | 2001-12-04 | Genentech, Inc. | SH2 domain-containing peptides |
US7338660B2 (en) | 2001-11-09 | 2008-03-04 | Abgenix, Inc. | Methods of treating cancer and enhancing immune responses with antibodies that bind CD40 |
US20080248028A1 (en) | 2004-03-24 | 2008-10-09 | Xencor, Inc. | Immunoglobulin Variants Outside the Fc Region |
US7521541B2 (en) | 2004-09-23 | 2009-04-21 | Genetech Inc. | Cysteine engineered antibodies and conjugates |
CN103635488A (zh) * | 2011-04-29 | 2014-03-12 | 埃派斯进有限公司 | 抗-cd40抗体及其使用方法 |
CN109265552A (zh) * | 2012-10-30 | 2019-01-25 | 埃派斯进有限公司 | 抗-cd40抗体及其使用方法 |
WO2014184545A2 (en) | 2013-05-14 | 2014-11-20 | Isis Innovation Limited | Antibodies |
WO2016177771A1 (en) | 2015-05-04 | 2016-11-10 | Apogenix Ag | Single-chain cd40-receptor agonist proteins |
WO2017004006A1 (en) | 2015-06-29 | 2017-01-05 | Bristol-Myers Squibb Company | Antibodies to cd40 |
CN109069621A (zh) * | 2015-09-04 | 2018-12-21 | 普里玛托普医疗股份有限公司 | 人源化抗-cd40抗体及其用途 |
WO2017059196A2 (en) * | 2015-09-30 | 2017-04-06 | Janssen Biotech, Inc. | Antagonistic antibodies specifically binding human cd40 and methods of use |
WO2017059243A2 (en) | 2015-09-30 | 2017-04-06 | Janssen Biotech, Inc. | Agonistic antibodies specifically binding human cd40 and methods of use |
WO2017205742A1 (en) * | 2016-05-27 | 2017-11-30 | Abbvie Biotherapeutics Inc. | Anti-cd40 antibodies and their uses |
Non-Patent Citations (43)
Title |
---|
AL-LAZIKANI ET AL.: "Standard conformations for the canonical structures of immunoglobulins", JOURNAL OF MOLECULAR BIOLOGY, vol. 273, 1997, pages 927 - 948, XP004461383, DOI: 10.1006/jmbi.1997.1354 |
ALMAGRO JCFRANSSON J, FRONTIERS IN BIOSCIENCE, vol. 13, 2008, pages 1619 - 1633 |
ANNU REV IMMUNOL, vol. 16, 1998, pages 111 - 35 |
APOSTOLOPOULOS ET AL., CURR. MED. CHEM., vol. 9, 2002, pages 411 - 420 |
ARNON ET AL.: "Monoclonal Antibodies And Cancer Therapy", 1985, ALAN R. LISS, INC., article "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", pages: 243 - 256 |
BEATTY, G.L.Y. LIK.B. LONG: "Cancer immunization: activating in and adaptive immunity through CD40 agonists", EXT REV ANTICANCER THER, vol. 17, no. 2, 2017, pages 175, XP055753521, DOI: 10.1080/14737140.2017.1270208 |
CASSET ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 307, 2003, pages 198 - 205 |
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 877 - 883 |
CHU ET AL., MOL.IMMUNOL., vol. 45, 2008, pages 3926 |
DENARDO ET AL., CLIN CANCER RES, vol. 4, no. 10, 1998, pages 2483 - 90 |
DOMINGUES ET AL., NAT. STRUCT. BIOL., vol. 6, 1999, pages 652 - 656 |
E. MEYERSW. MILLER: "Current Protocols in Molecular Biology", vol. 4, 1989, JOHN WILEY & SONS, pages: 11 - 17 |
ELIOPOULS, A.G. ET AL., CD40 INDUCES APOPTOSIS IN CANCER CELLS THROUGH CANCER ACTIVATION OF CYTOTOXICITY LIGANDS OF THE TUMOR NECROSIS FACTOR SUPERFAMILY CELL BIOL, vol. 20, no. 15, 2000, pages 5503 - 15 |
FLATMAN ET AL., J. CHROMATOGR., vol. B848, 2007, pages 79 - 87 |
FONG, L.E.G. ENGLEMAN: "Dendritic cells in cancer immunology", ANNU REV IMMUNOL, vol. 18, 2000, pages 245 - 73 |
GENTZ ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 821 - 824 |
GLENNIE: "inflammatory CD40 antibiotics and Cancer therapy", CLIN CANCER RES, vol. 19, no. 5, 2013, pages 1035 - 43 |
HASSAN, S.B. ET AL., ANTI-CD40-MEDIATED CANCER IMMUNOLOGY: AN UPDATE OF RECOVERY AND ON CLINICAL TRIALS, IMMUNOPHARMACOLOGICAL IMMUNOLOGY, vol. 36, no. 2, 2014, pages 96 - 104 |
HE ET AL., J. IMMUNOL., vol. 173, 2004, pages 4919 - 28 |
HOLLIGER, PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 |
KABAT ET AL.: "Sequences of proteins of Immunological Interest", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH |
LI, NAT. BIOTECHNOL., vol. 18, 2000, pages 1251 - 1256 |
LIRAVATCH, SCIENCE, vol. 333, 2011, pages 1030 |
LIU, Y.H. ET AL.: "High-throughput reformatting of phage-displayed antibody fragments to IgGs by one-step emulsion PCR.", PROTEIN ENGINEERING, DESIGN & SELECTION., vol. 31, no. 11,, 9 April 2019 (2019-04-09), XP055741283, DOI: 20200526100540X * |
LONBERG, CURR.OPIN.IMMUNOL, vol. 20, 2008, pages 450 - 459 |
MIMOTO ET AL., PROTEIN ENGINEERING DESIGN & SELECTION, vol. 26, 2013, pages 589 |
MONFARDINI ET AL., CURR. PHARM. DES., vol. 8, 2002, pages 2185 - 2199 |
MUYLDERMANS, J. BIOTECHNOL., vol. 74, 2001, pages 277 - 302 |
NEEDLEMAWUNSCH: "J. Mol. Biol.", vol. 48, 1970, pages: 444 - 453 |
PARDOL DM.: "The block of immune checkpoints in Cancer immunology", NAT REV CANCER, vol. 12, no. 4, 2012, pages 252 |
POLJAK, STRUCTURE, vol. 2, 1994, pages 1121 |
ROTHE, M. ET AL.: "TRAF2-mediated activation of NF-kappa B by TNF receptor 2 and CD40", SCIENCE, vol. 269, no. 5229, 1995, pages 1424 - 7 |
SATOSONE, BIOCHEM. J., vol. 371, 2003, pages 603 - 608 |
See also references of EP3954705A4 |
SHIELD ET AL., JBC, vol. 277, 2002, pages 26733 |
SMITH-GARVIN, J.E.G.A. KORETZKYM.S. JORDAN: "T cell activation", ANNU REV IMMUNOL, vol. 27, 2009, XP055761943, DOI: 10.1146/annurev.immunol.021908.132706 |
STAHLI, METHODS IN ENZYMOLOGY, vol. 9, 1983, pages 242 - 253 |
VAN DIJKVAN DE WINKEL, CURR.OPIN.PHARMACOL, vol. 5, 2001, pages 368 - 74 |
WHITE ET AL., J. IMMUNOL., vol. 187, pages 1754 |
WILSON ET AL., CELL, vol. 37, 1984, pages 767 |
YAMADA ET AL., CANCER SCI, vol. 104, 2013, pages 14 - 21 |
YAO SZHU YCHEN L.: "Advances in targeting cell surface signaling molecules for tumor modulation", NAT REV DRUG DISCOV, vol. 12, no. 2, 2013, pages 130 - 146 |
YU ET AL., J. AM. CHEM.SOC., vol. 135, 2013, pages 9723 |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2022262099A1 (zh) * | 2021-06-17 | 2022-12-22 | 南京蓝盾生物科技有限公司 | 抗cd70内化的抗体、抗体偶联物及其应用 |
Also Published As
Publication number | Publication date |
---|---|
CA3136491A1 (en) | 2020-10-15 |
TW202103733A (zh) | 2021-02-01 |
SG11202111188VA (en) | 2021-11-29 |
JP2022527406A (ja) | 2022-06-01 |
JP7535060B2 (ja) | 2024-08-15 |
US20220324988A1 (en) | 2022-10-13 |
AU2020256828A1 (en) | 2021-11-18 |
EP3954705A1 (en) | 2022-02-16 |
CN113728008A (zh) | 2021-11-30 |
EP3954705A4 (en) | 2023-06-07 |
CN113728008B (zh) | 2024-04-09 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US11643465B2 (en) | Anti-PD-1 antibodies | |
US11365255B2 (en) | PD-1 antibody, antigen-binding fragment thereof, and medical application thereof | |
US20210269528A1 (en) | Novel anti-pd-l1 antibodies | |
RU2725811C1 (ru) | Антитела против 4-1bb человека и их применение | |
US20230348601A1 (en) | Bispecific antibody for icos and pd-l1 | |
CN109757103B (zh) | 针对tim3的抗体及其用途 | |
JP2022046651A (ja) | 4-1bb結合分子 | |
WO2018036472A1 (zh) | 一种抗pd1单克隆抗体、其药物组合物及其用途 | |
JP2021529557A (ja) | 抗腫瘍免疫チェックポイント調節因子アンタゴニスト | |
JP2020532965A (ja) | 抗cd137分子及びその使用 | |
JP7535060B2 (ja) | 抗cd40抗体およびその使用 | |
KR20150061041A (ko) | 인간 ox40 수용체에 대한 결합 분자 | |
KR20230093010A (ko) | 신규 항-lilrb2 항체 및 유도체 생성물 | |
JP2019502695A (ja) | PD−1に対する抗体分子とC−Met阻害剤との組合せおよびその使用 | |
TW201840585A (zh) | 用於白斑病的治療的抗人cxcr3抗體 | |
CN116419970B (zh) | 低毒性抗ox40抗体、其药物组合物及应用 | |
WO2024109657A1 (zh) | 抗ccr8抗体及其用途 | |
WO2023173393A1 (zh) | 结合b7-h3的抗体及其用途 | |
CN116829588A (zh) | 新型抗lilrb2抗体和衍生产物 | |
CN118852427A (zh) | 靶向cd70的单克隆抗体及其用途 | |
AU2016203429A1 (en) | Binding Molecules to the Human OX40 Receptor | |
AU2014202213A1 (en) | Binding Molecules to the Human OX40 Receptor |
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: 20786934 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2021560225 Country of ref document: JP Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 3136491 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: 2020256828 Country of ref document: AU Date of ref document: 20200410 Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 2020786934 Country of ref document: EP Effective date: 20211110 |