ANTI-4-1BB ANTIBODY, ANTIGEN-BINDING FRAGMENT THEREOF AND MEDICAL USE THEREOF

Abstract
Provided are an anti-4-1BB antibody, an antigen-binding fragment thereof and medical use thereof. Further, provided are a chimeric antibody and a humanized antibody comprising a CDR region of the anti-4-1BB antibody, as well as a pharmaceutical composition comprising a human anti-4-1BB antibody or an antigen-binding fragments thereof, and use thereof as anti-cancer medicaments. In particular, provided is use of a humanized anti-4-1BB antibody in the preparation of a medicament for the treatment of diseases or conditions mediated by 4-1BB.
Description
FIELD OF THE INVENTION

The present application relates to an anti-4-1BB antibody and antigen-binding fragments, a chimeric antibody, a humanized antibody comprising the CDR regions of the anti-4-1BB antibody, and a pharmaceutical composition comprising the human anti-4-1BB antibody or antigen-binding fragment thereof, and its use as an anticancer agent.


BACKGROUND OF THE INVENTION

Cancer is a huge health challenge for human society for a long time.


Traditional therapies (such as surgery, chemotherapy, and radiotherapy) have shown little effect on treating disseminated solid tumors. Tumor immunotherapy is a hot spot in the field of cancer therapy, in which T cell immunotherapy is in a central position. Tumor immunotherapy fully utilizes and recruits killer T cells in tumor patients to kill tumors. Tumor immunotherapy may be the most effective and safest way for the treatment of tumors. Tumor immunotherapy currently has good prospects for the treatment of several different types of cancer, including disseminated metastatic tumors.


Two signaling pathways are involved in activation of T cells in human body. In addition to a first signal provided by presenting MHC-antigen peptides to T cells via antigen presenting cells (APCs), a second signal, provided by a series of costimulatory molecules, is also necessary for T cells to produce a normal immune response. This dual signaling pathway system plays a vital role in the balance of the immune system in vivo. It strictly regulates the immune responses to autoantigens and non-autoantigens. In the absence of a second signal provided by costimulatory molecules, non-response or sustained specific immune response of T cells will result in tolerance.


4-1BB (also known as CD137, TNFRSF9) is a transmembrane protein of the tumor necrosis factor receptor superfamily (TNFRS). Human 4-1BB has a protein of 255 amino acids, including a signal sequence (amino acid residues 1-17), an extracellular domain (169 amino acids), a transmembrane domain (27 amino acids), and an intracellular domain (42 amino acid). 4-1BB is expressed as a monomer or a dimer on the cell surface, and it mediates signal transduction via trimerization upon binding to its ligand (4-1BBL).


4-1BB is usually activation-dependent and is widely present in immune cells, including activated NK and NKT cells, regulatory T cells, dendritic cells (DC), stimulated mast cells, differentiated myeloid cells, monocytes, neutrophils, and eosinophils. Studies have shown that initiation of 4-1BB can enhance cell proliferation, survival, and cytokine production.


Agonistic antibodies against 4-1BB increase the expression of co-stimulatory molecules in many models and significantly enhance the response of cytolytic T lymphocytes, resulting in anti-tumor efficacy; whereas durable anti-tumor protective T cell memory responses have also been observed in tumor models with monotherapy and combination therapy targeting 4-1BB.


For the still unmet medical needs associated with 4-1BB, several international pharmaceutical companies are actively developing monoclonal antibodies against 4-1BB. Related patents such as WO2000029445, WO2005035584, WO2012032433, WO2003049755, WO2017205745A and the like. To date, anti-4-1BB antibodies provided by Bristol-Myers Squibb (BMS) and Pfizer's have entered Phase I/II clinical trials, and the similar products from Pieris have also entered Phase I clinical trials.


The present application is directed to providing anti-4-1BB antibodies with high affinity, high selectivity and high biological activity, and medicaments, compositions and methods thereof, for use in cancer therapy by stimulating 4-1BB and its pathway to lead to immune activation.


SUMMARY OF THE INVENTION

The application provides an anti-4-1BB antibody or antigen-binding fragment thereof, comprising:


an antibody light chain variable region comprising at least one LCDR selected from the group consisting of SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8; SEQ ID NO: 14, SEQ ID NO: 15 and SEQ ID NO: 16; and


an antibody heavy chain variable region comprising at least one HCDR selected from the group consisting of SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO:11, SEQ ID NO: 12, SEQ ID NO: 13 and SEQ ID NO: 45.


In a specific embodiment of the above anti-4-1BB antibody or antigen-binding fragment thereof according to present application, wherein the antibody light chain variable region comprises LCDR1, LCDR2 and LCDR3 as set forth in SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8, respectively.


In a specific embodiment of the above anti-4-1BB antibody or antigen-binding fragment thereof according to present application, wherein the antibody light chain variable region comprises LCDR1, LCDR2 and LCDR3 as set forth in SEQ ID NO: 14, SEQ ID NO: 15 and SEQ ID NO: 16, respectively.


In a specific embodiment of the above anti-4-1BB antibody or antigen-binding fragment thereof according to present application, wherein the antibody heavy chain variable region comprises HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 3, SEQ ID NO: 45 and SEQ ID NO: 5, respectively, or comprises HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 3, SEQ ID NO: 4 and SEQ ID NO: 5, respectively.


In a specific embodiment of the above anti-4-1BB antibody or antigen-binding fragment thereof according to present application, wherein the antibody heavy chain variable region comprises HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 11, SEQ ID NO: 12 and SEQ ID NO: 13, respectively.


In a specific embodiment of the above anti-4-1BB antibody or antigen-binding fragment thereof according to present application, wherein the antibody light chain variable region comprises:


LCDR1, LCDR2, and LCDR3 as set forth in SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8, respectively; or


LCDR1, LCDR2, and LCDR3 as set forth in SEQ ID NO: 14, SEQ ID NO: 15 and SEQ ID NO: 16, respectively; and


the antibody heavy chain variable region comprises:


HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 3, SEQ ID NO: 45 and SEQ ID NO: 5, respectively; or


HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 3, SEQ ID NO: 4 and SEQ ID NO: 5, respectively; or


HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 11, SEQ ID NO: 12 and SEQ ID NO: 13, respectively.


In a specific embodiment of the present application, the anti-4-1BB antibody or antigen-binding fragment thereof may be selected from any one of the following:


(1) the antibody light chain variable region comprises LCDR1, LCDR2 and LCDR3 as set forth in SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively; and the antibody heavy chain variable region comprises HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 3, SEQ ID NO: 45 and SEQ ID NO: 5, respectively; or


(2) the antibody light chain variable region comprises LCDR1, LCDR2 and LCDR3 as set forth in SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16, respectively; and the antibody heavy chain variable region comprises HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 11, SEQ ID NO: 12 and SEQ ID NO: 13, respectively.


In a specific embodiment of the present application, the antibody light chain variable region sequence is set forth in SEQ ID NO: 2; and the heavy chain variable region sequence is set forth in SEQ ID NO: 1.


In a specific embodiment of the present application, the antibody light chain variable region sequence is set forth in SEQ ID NO: 10; and the heavy chain variable region sequence is set forth in SEQ ID NO: 9.


The application also provides an anti-4-1BB antibody or antigen-binding fragment thereof, comprising:


antibody light chain variable region comprising LCDR1, LCDR2 and LCDR3 as set forth in SEQ ID NO: 42, SEQ ID NO: 43 and SEQ ID NO: 44, respectively; and


antibody heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 39, SEQ ID NO: 40 and SEQ ID NO: 41, respectively.


The present application also provides a 4-1BB antibody or antigen-binding fragment, wherein the light chain variable region LCVR of the antibody or antigen fragment is set forth in SEQ ID NO: 38, and the heavy chain variable region HCVR is set forth in SEQ ID NO: 37.


In a specific embodiment of the present application, the anti-4-1BB antibody or antigen-binding fragment thereof as described above is a murine antibody or fragment thereof.


In a specific embodiment of the present application, the antibody light chain variable region of the murine antibody or fragment thereof as described above, further comprises light chain FR regions of murine κ, λ chain or a variant thereof.


In a specific embodiment of the present application, the murine antibody or fragment thereof as described above, further comprises a light chain constant region of murine κ, λ chain or a variant thereof.


In a specific embodiment of the present application, the heavy chain variable region of the murine antibody or fragment thereof as described above further comprises heavy chain FR regions of murine IgG1, IgG2, IgG3, IgG4 or a variant thereof.


In a specific embodiment of the present application, the murine antibody or fragment thereof as described above, further comprises a heavy chain constant region of murine IgG1, IgG2, IgG3, IgG4 or a variant thereof.


In a specific embodiment of the present application, the anti-4-1BB antibody or antigen-binding fragment thereof as described above is a chimeric antibody or fragment thereof.


In a specific embodiment of the present application, the anti-4-1BB chimeric antibody or fragment thereof as described above, further comprises a light chain constant region of human κ, λ chain or a variant thereof.


In a specific embodiment of the present application, the anti-4-1BB chimeric antibody or fragment thereof as described above, further comprises a heavy chain constant region of human IgG1, IgG2, IgG3 or IgG4 or a variant thereof.


In a specific embodiment of the present application, the anti-4-1BB antibody or antigen-binding fragment thereof as described above is a human antibody or fragment thereof.


In a specific embodiment of the present application, the anti-4-1BB antibody or antigen-binding fragment thereof as described above is a humanized antibody or fragment thereof.


In a specific embodiment of the anti-4-1BB human antibody or fragment thereof as described above, wherein the light chain of the humanized antibody is set forth in SEQ ID NO: 18 or a variant thereof; the variant comprises from 0 to 10 amino acid changes in the light chain. The heavy chain sequence is set forth in SEQ ID NO: 17 or a variant thereof; the variant comprises from 0 to 10 amino acid changes in the heavy chain.


In a specific embodiment of the 4-1BB human antibody or fragment thereof as described above, wherein the light chain of the humanized antibody is set forth in SEQ ID NO: 20 or a variant thereof; the variant comprises from 0 to 10 amino acid changes in the light chain. The heavy chain sequence is set forth in SEQ ID NO: 19 or a variant thereof; the variant comprises from 0 to 10 amino acid changes in the heavy chain.


In a specific embodiment of the present application, the anti-4-1BB human antibody or fragment thereof as described above, further comprises a constant region of human IgG1, IgG2, IgG3 or IgG4 or a variant thereof, preferably comprises human IgG1 or IgG2 constant region.


In a specific embodiment of the present application, the anti-4-1BB antibody or antigen-binding fragment thereof as described above is a humanized antibody or a fragment thereof.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, the humanized antibody light chain variable region further comprises light chain FR regions of human κ, λ chain or a variant thereof.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, the light chain FR region sequence of the humanized antibody light chain variable region is derived from human germline light chain IGkV3-11 sequence as set forth in SEQ ID NO: 22; or derived from human germline light chain IGkV4-1 sequence as set forth in SEQ ID NO: 24.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, wherein the humanized antibody light chain variable region sequence is set forth in SEQ ID NO: 30 or SEQ ID NO: 36, or a variant thereof.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, the humanized antibody light chain sequence is set forth in SEQ ID NO: 18 or SEQ ID NO: 20, or a variant thereof.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, the variant of the humanized antibody light chain variable region preferably has from 0 to 10 (1, 2, 3, 4, 5, 6, 7, 8, 9, 10) amino acid changes in the light chain variable region; more preferably on amino acid positions 4, 47 and 62 or any combination thereof; more preferably M4L, P47A, V62I mutation and any combination thereof; more preferably P47A and V62I mutations, M4L and V62I mutations.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, further comprises a light chain constant region of a human λ chain or a variant thereof.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, the humanized antibody heavy chain variable region further comprises heavy chain FR regions of human IgG1, IgG2, IgG3, IgG4 or a variant thereof.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, wherein the heavy chain FR region of humanized antibody heavy chain variable region is derived from human germline heavy chain IGHV3-30 sequence as set forth in SEQ ID NO: 21; or derived from human germline heavy chain IGHV 1-46 sequence as set forth in SEQ ID NO: 23.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, the humanized antibody heavy chain variable region is set forth in SEQ ID NO: 27 or SEQ ID NO: 33, or a variant thereof.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, the humanized antibody heavy chain variable region is set forth in SEQ ID NO: 17 or SEQ ID NO:19, or a variant thereof, the variant preferably has from 0 to 10 amino acid changes in the heavy chain variable region; more preferably has mutation at amino acid position 48, 53, 75, 95 or 112 or any combination thereof; more preferably has mutation of M48I, E53D, A75S, Y95F, Q112T/R112Q or any combination thereof; more preferably E53D, A75S and R112Q; more preferably M48I, Y95F and Q112T.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, further comprises a heavy chain constant region of human IgG1, IgG2, IgG3 or IgG4 or a variant thereof, preferably, comprises heavy chain FR regions of human IgG1, IgG2 or IgG4, more preferably comprises heavy chain FR regions of human IgG1 or IgG2.


In a specific embodiment of the present application, provided an anti-4-1BB humanized antibody or fragment thereof, comprising a light chain and a heavy chain:


the heavy chain comprising a heavy chain variable region selected from the group consisting of SEQ ID No. 25, 26, 27, 31, 32 and 33; and


the light chain comprising a light chain variable region selected from the group consisting of SEQ ID No. 28, 29, 30, 34, 35 and 36.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, the light chain variable region is set forth in SEQ ID NO: 36, and the heavy chain variable region is set forth in SEQ ID NO:33.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, the light chain variable region is set forth in SEQ ID NO: 30, and the heavy chain variable region is set forth in SEQ ID NO:27.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, the light chain is set forth in SEQ ID NO: 20, and the heavy chain is set forth in SEQ ID NO: 19.


In a specific embodiment of the anti-4-1BB humanized antibody or fragment thereof as described above, the light chain is set forth in SEQ ID NO: 18, and the heavy chain is set forth in SEQ ID NO: 17.


In a specific embodiment of the anti-4-1BB antibody or antigen-binding fragment thereof as described above, wherein the antigen-binding fragment is Fab, Fv, sFv, F(ab′)2, linear antibody, single chain antibody, scFv, sdAb, sdFv, Nanobody, peptibody, domain antibody and multispecific antibody (bispecific antibody, diabody, triabody and tetrabody, tandem di-scFv, tandem tri-scFv).


The application further provides an isolated monoclonal antibody or antigen-binding fragment thereof that competes for binding to 4-1BB or epitope thereof with the monoclonal antibody or antigen-binding fragment thereof described above.


The application further provides a polynucleotide encoding the anti-4-1BB antibody or antigen-binding fragment thereof as described above, the polynucleotide may be DNA or RNA.


The application further provides an expression vector comprising the polynucleotide as described above, the expression vector may be a viral vector, and the virus may be an oncolytic virus.


The application further provides a host cell transformed with the expression vector as described above.


In a specific embodiment of the present application, the host cell as described above, characterized in that the host cell is a bacterium, preferably Escherichia coli.


In a specific embodiment of the present application, the host cell as described above is yeast, preferably Pichia pastoris.


In a specific embodiment of the present application, the host cell as described above is mammalian cell, preferably Chinese hamster ovary (CHO) cell or human embryonic kidney (HEK) 293 cell.


The present application also provides a multispecific antibody comprising the light chain variable region and the heavy chain variable region as described above.


The application also provides a single chain antibody comprising the light chain variable region and the heavy chain variable region as described above.


The application also provides an antibody-drug conjugate, comprising the light chain variable region and the heavy chain variable region as described above. The antibody-drug conjugate is formed by linking the antibody-linker-drug (toxin). Well-known linkers include cleavable linker, non-cleavable linker, for example, including but not limited to, SMCC, SPDP, and the like; the toxins are also well known in the art, such as DM1, DM4, MMAE, MMAF, and the like.


The present application also provides a method for producing the anti-4-1BB antibody or antigen-binding fragment thereof, comprising the steps of expressing the antibody or antigen-binding fragment thereof in the host cell as described above, and isolating the antibody or antigen-binding fragment thereof from the host cell.


The application further provides a pharmaceutical composition comprising the anti-4-1BB antibody or antigen-binding fragment thereof as described above and a pharmaceutically acceptable excipient, diluent or carrier.


The present application further provides use of any one or combination of the following in the preparation of a medicament: the anti-4-1BB antibody or antigen-binding fragment thereof according to the present application, the pharmaceutical composition according to the present application and the antibody-drug conjugate according to the present application; wherein the medicament is for treating or preventing a 4-1BB or 4-1BBL mediated disease or condition; the disease is preferably cancer; and the cancer is most preferably selected from the group consisting of melanoma, breast cancer, ovarian cancer, prostate cancer, pancreatic cancer, kidney cancer, lung cancer, liver cancer, stomach cancer, colorectal cancer, bladder cancer, head and neck cancer, thyroid cancer, esophageal cancer, cervical cancer, sarcoma, multiple myeloma, leukemia, lymphoma, gallbladder cancer and glioblastoma.


The present application further provides a method of treating or preventing a 4-1BB or 4-1BBL mediated disease or condition, comprising administering to a subject a therapeutically or prophylactically effective amount of the anti-4-1BB antibody or antigen-binding fragment thereof according to the present application, or the pharmaceutical composition according to the present application, or the antibody-drug conjugate according to the present application; wherein the disease is preferably cancer; and the cancer is most preferably selected from the group consisting of melanoma, breast cancer, ovarian cancer, prostate cancer, pancreatic cancer, kidney cancer, lung cancer, liver cancer, stomach cancer, colorectal cancer, bladder cancer, head and neck cancer, thyroid cancer, esophageal cancer, cervical cancer, sarcoma, multiple myeloma, leukemia, lymphoma, gallbladder cancer and glioblastoma.





DESCRIPTION OF THE DRAWINGS


FIG. 1: In vitro capture ELISA assay of the murine antibodies, indicating that all the anti-4-1BB murine antibodies to be tested from each hybridomas efficiently bind to human 4-1BB-his.



FIG. 2: ELISA assay of the murine antibodies in blocking 4-1BBL ligand. The results showed that B1E7 and B1A7 antibodies were ligand-blocking types, while B1E10 and the two reference antibodies were non-blocking types.



FIG. 3: 4-1BB reporter gene activation assay of the murine antibodies, indicating that the activation intensity was ranked in the order of B1E10>MOR7480 reference>B1E7>B1A7.



FIG. 4: PBMC cell proliferation function activation assay of the two humanized antibodies. The results showed that the activation intensity of huB1E7 and huB1E10 was higher than that of reference MOR7480, wherein huB1E7 was relatively stronger.



FIG. 5: 4-1BB reporter gene activation assay of the humanized antibodies. The results showed that the activation intensity was ranked in an order consistent with that for murine antibodies, which is B1E10>MOR7480 reference>B1E7.



FIG. 6: Tumor growth curve in hu4-1BB transgenic C57BL/6 mice transplanted with MC38 intestinal cancer cells after administration of each anti-4-1BB antibody to be tested.



FIG. 7: Body weight change curve of hu4-1BB transgenic C57BL/6 mice transplanted with MC38 intestinal cancer cells after administration of each anti-4-1BB antibody to be tested.





DETAILED DESCRIPTION OF THE DISCLOSURE
1. Terminology

In order to more readily understand the invention, certain technical and scientific terms are specifically defined below. Unless specifically defined elsewhere in this document, all other technical and scientific terms used herein have the meaning commonly understood by one of ordinary skill in the art to which this invention pertains.


As used herein, the single-letter code and the three-letter code for amino acids are as described in J. Biol. Chem, 243, (1968) p 3558.


As used herein, the term “antibody” refers to immunoglobulin, which is a four-peptide chain structure formed by linking two identical heavy chains and two identical light chains by disulfide bonds. Different immunoglobulin heavy chain constant regions exhibit different amino acid compositions and sequence orders, thereby presenting different kinds of antigenicity. Accordingly, immunoglobulins can be divided into five categories, or called immunoglobulin isotypes, namely IgM, IgD, IgG, IgA and IgE, their heavy chains are μ chain, δ chain, γ chain, α chain and ε chain, respectively. According to its amino acid composition of hinge region and the number and location of heavy chain disulfide bonds, the same type of Ig can be divided into different sub-categories, for example, IgG can be divided into IgG1, IgG2, IgG3, and IgG4. Light chain can be divided into κ or λ chain considering of different constant regions. Each of the five Igs can have κ or λ chain.


In the present application, the antibody light chain variable region mentioned herein further comprises a light chain constant region, which comprises a human or murine κ, λ chain or a variant thereof.


In the present invention, the antibody heavy chain variable region mentioned herein further comprises a heavy chain constant region, which comprises human or murine IgG1, 2, 3, 4 or a variant thereof.


Near the N-terminal of the antibody heavy chains and light chains sequence, about 110 amino acid sequence varies largely, known as the variable region (V region); the rest of the amino acid sequence near the C-terminus is relative stable, known as the constant region (C region). Variable region comprises three hypervariable regions (HVR) and four framework region (FR) having relatively conserved sequence. The three hypervariable regions determine the specificity of the antibody, also known as a complementarity determining region (CDR). Each light chain variable region (LCVR) and each heavy chain variable region (HCVR) is composed of three CDRs and four FRs, with order from the amino terminal to the carboxyl terminal being: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. Three light chain CDRs refer to LCDR1, LCDR2, and LCDR3; three heavy chain CDRs refer to HCDR1, HCDR2 and HCDR3. The numbers and locations of CDR amino acid residues in LCVR and HCVR of the antibody or the antigen-binding fragment herein correspond with known Kabat numbering criteria (LCDR1-3, HCDR2-3), or correspond with kabat and chothia (ABM) numbering criteria (HCDR1).


The term “recombinant human antibody” includes human antibodies prepared, expressed, created or isolated by recombinant methods, and the techniques and methods involved are well known in the art, such as:


(1) an antibody isolated from a human immunoglobulin gene transgenic or transchromosomal animal (e.g., a mouse), or a hybridoma prepared therefrom;


(2) an antibody isolated from transformed host cells expressing the antibody, such as a transfectoma;


(3) an antibody isolated from a recombinant combinatorial human antibody library; and


(4) an antibody prepared, expressed, created or isolated by splicing human immunoglobulin gene sequences to another DNA sequences or the like.


Such recombinant human antibody comprises a variable region and a constant region, such regions involve specific human germline immunoglobulin sequences encoded by germline genes, but also involve subsequent rearrangements and mutations such as those occur during the antibody maturation.


The term “murine antibody” in the present application refers to monoclonal antibody against human-4-1BB or epitope thereof, which is prepared according to the knowledge and skills in the art. During the preparation, a test object is injected with 4-1BB antigen, and then hybridoma expressing antibody which possesses desired sequence or functional characteristics is separated. In a particular embodiment of the present application, the murine 4-1BB antibody or the antigen-binding fragment thereof further comprises a light chain constant region of murine κ, λ chain or a variant thereof, or further comprises a heavy chain constant region of murine IgG1, IgG2, IgG3 or IgG4, or a variant thereof.


The term “human antibody” includes antibodies having variable and constant regions with human germline immunoglobulin sequences. Human antibodies of the present application may include amino acid residues that are not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). However, the term “human antibody” does not include an antibody in which CDR sequences derived from other mammalian species germline (such as mouse germline) have been grafted onto a human framework sequence (i.e., “humanized antibody”).


The term “humanized antibody”, also known as CDR-grafted antibody, refers to an antibody generated by grafting murine CDR sequences into a variable region framework of a human antibody. Humanized antibody overcomes the strong antibody response induced by the chimeric antibody which carries a large amount of murine protein components. To avoid the decrease of activity along with the decrease of immunogenicity, the variable region of the human antibody is subjected to a minimum back mutation to maintain the activity.


The term “chimeric antibody”, is an antibody which is formed by fusing the variable region of a murine antibody with the constant region of a human antibody, the chimeric antibody can alleviate the murine antibody-induced immune response. To establish a chimeric antibody, hybridoma secreting specific murine monoclonal antibody is firstly established, a variable region gene is cloned from mouse hybridoma cells, then a constant region gene of a human antibody is cloned as desired, the mouse variable region gene is ligated with the human constant region gene to form a chimeric gene which can be inserted into a human vector, and finally the chimeric antibody molecule is expressed in the eukaryotic or prokaryotic industrial system. The constant region of a human antibody is selected from the heavy chain constant region of human IgG1, IgG2, IgG3 or IgG4 or a variant thereof, preferably the heavy chain constant region of human IgG2 or IgG4, or that of IgG1 which has no ADCC (antibody-dependent cell-mediated cytotoxicity) after amino acid mutation.


As used herein, “antigen-binding fragment” refers to a Fab fragment, a Fab′ fragment, a F(ab′)2 fragment with antigen-binding activity, as well as a Fv fragment sFv fragment binding to human 4-1BB. Fv fragment is a minimum antibody fragment carrying all antigen-binding sites, it comprises a heavy chain variable region, a light chain variable region, but without constant region. Generally, Fv antibody further comprises a polypeptide linker between the VH and VL domains, and is capable of forming a structure necessary for antigen binding. Also, different linkers can be used to connect the variable regions of two antibodies to form a polypeptide, named as single chain antibody or single chain Fv (sFv).


As used herein, “binding to 4-1BB”, refers to the ability to interact with 4-1BB or an epitope thereof, which may be of human origin. The term “antigen binding site” as used herein refers to discontinuous three-dimensional sites on the antigen, recognized by the antibody or the antigen-binding fragment of the present application.


The term “epitope” refers to the sites on an antigen that specifically bind to an immunoglobulin or antibody. The epitope can be formed by adjacent amino acids, or by non-adjacent amino acids which have been brought to be closer due totertiary folding of a protein. The Epitope formed by adjacent amino acids is typically retained after exposure to denaturing solvents, whereas the epitope formed by tertiary folding is typically lost after treatment with denaturing solvents. Epitopes typically include at least 3-15 amino acids in a unique spatial conformation. Methods for determining epitope bound by a given antibody are well known in the art, including immunoblotting and immuno precipitation assays, and the like. Methods for determining the spatial conformation of an epitope include techniques in the art and techniques described herein, such as X-ray crystallography and two-dimensional nuclear magnetic resonance.


The term “specifically binds to”, “selectively binds to” as used herein, refers to the binding of an antibody to an epitope on a predetermined antigen. Typically, the antibody binds to a predetermined antigen or the epitope thereof at approximately less than 10−7 M or even less equilibrium dissociation constant (KD), and the affinity of the antibody for binding to the predetermined antigen or epitope thereof is at least two times higher than that for non-specific antigens (such as BSA) other than the predetermined antigen (or epitope thereof) or than that for closely related antigens, as measured in an instrument via surface plasmon resonance (SPR) techniques, wherein the recombinant human 4-1BB or its epitope is used as an analyte and the antibody is used as a ligand. The term “an antibody recognizing the antigen” can be used interchangeably herein with the term “specifically binding antibody”.


The term “cross reaction” refers to the ability of the antibody of the present application to bind to 4-1BB from a different species. For example, an antibody of the present application that binds to human 4-1BB can also bind to 4-1BB from another species. Cross-reactivity is measured by detecting the specific reactivity with purified antigens in binding assays (e.g., SPR and ELISA), or by detecting the binding or functional interaction with cells physiologically expressing 4-1BB. Methods for determining cross-reactivity include standard binding assays as described herein, such as surface plasmon resonance analysis, or flow cytometry.


The terms “inhibition” or “blockade” are used interchangeably and encompass both partial and complete inhibition/blockade. Inhibition/blockade of 4-1BB preferably reduces or alters the normal level or type of activity that is generated upon binding to 4-1BB without inhibition or blockade. Inhibition and blockade are also intended to include a measurable decrease of binding affinity for 4-1BB when contacted with an anti-4-1BB antibody, compared to the binding affinity for 4-1BB in absence of anti-4-1BB antibody.


The term “inhibiting the growth” (e.g., involving cells) is intended to include any measurable reduction in cell growth.


The terms “inducing an immune response” and “enhancing an immune response” are used interchangeably and refer to the immune response to the stimulation of a particular antigen (i.e., passive or adaptive). The term “inducing”, with respect to CDC or ADCC, refers to stimulating a specific mechanism to directly kill cells.


As used herein, the term “ADCC”, namely antibody-dependent cell-mediated cytotoxicity, refers to cells expressing Fc receptors directly kill target cells coated by an antibody through recognizing the Fc segment of the antibody. ADCC effector function of the antibody can be reduced or eliminated via modification of the Fc segment in IgG. The modification refers to mutations of the antibody heavy chain constant region, such as mutations selected from N297A, L234A, L235A in IgG1; IgG2/4 chimera; F235E, or L234A/E235A mutations in IgG4.


Methods for producing and purifying antibodies and antigen-binding fragments are well known in the art and can be found, for example, in Antibodies, A Laboratory Manual, Cold Spring Harbor, Chapter 5-8 and 15. For example, mice can be immunized with human 4-1BB, or fragments thereof, and the resulting antibodies can then be renatured, purified and sequenced using conventional methods well known in the art. Antigen-binding fragments can also be prepared by conventional methods. The antibody or the antigen-binding fragment of the present invention is genetically engineered to add one or more human framework regions (FRs) to non-human derived CDRs. Human FR germline sequences can be obtained from ImMunoGeneTics(IMGT) via their website http://imgt.cines.fr, or from The Immunoglobulin FactsBook, 20011SBN012441351.


The engineered antibody or antigen-binding fragment of the present invention may be prepared and purified using conventional methods. For example, cDNA sequences encoding a heavy chain (SEQ ID NO: 17) and a light chain (SEQ ID NO: 18) may be cloned and recombined into a GS expression vector. The recombined immunoglobulin expression vector may then stably transfect CHO cells. As a more recommended method well known in the art, mammalian expression system may result in glycosylation of antibodies, typically at the highly conserved N-terminus in the FC region. Stable clones may be obtained through expression of an antibody specifically binding to human antigen. Positive clones may be expanded in a serum-free culture medium for antibody production in bioreactors. Culture medium, into which an antibody has been secreted, may be collected and purified by conventional techniques. The antibody may be subjected to filtration and concentration using common techniques. Soluble mixtures and multimers may be effectively removed by common techniques, including molecular sieve or ion exchange. The obtained product may be immediately frozen, for example at −70° C., or may be lyophilized.


The antibody of the present invention is a monoclonal antibody. Monoclonal antibody or mAb, as used herein, refers to an antibody that is derived from a single clone including but not limited to any eukaryotic, prokaryotic, or phage clone. Monoclonal antibodies and antigen-binding fragments thereof can be recombined, for example, by hybridoma technologies, recombinant technologies, phage display technologies, synthetic technologies (e.g., CDR-grafting), or other technologies known in the art.


“administration” and “treatment,” as it applies to an animal, human, experimental subject, cell, tissue, organ, or biological fluid, refers to contacting an exogenous pharmaceutical, therapeutic, diagnostic agent, or composition with the animal, human, subject, cell, tissue, organ, or biological fluid. “administration” and “treatment” can refer, e.g., to therapeutic, pharmacokinetic, diagnostic, research, and experimental methods. Treatment of a cell encompasses contacting a reagent with the cell, as well as contacting a reagent with a fluid, where the fluid is in contact with the cell. “administration” and “treatment” also mean in vitro and ex vivo treatments, e.g., of a cell, by a reagent, diagnostic, binding composition, or by another cell. “Treatment,” as it applies to a human, veterinary, or a research subject, refers to therapeutic treatment, prophylactic or preventative measures, to research and diagnostic applications.


“Treat” means to the administration of a therapeutic agent (such as a composition comprising any of the antibodies of antigen-binding fragments thereof of the present invention) internally or externally to a subject suffering from, suspected to suffer from or having tendency to suffer from one or more disease symptoms for which the agent has known therapeutic activity. Typically, the therapeutic agent is administered in an amount effective to alleviate one or more disease symptoms in the treated subject or population, either by inducing the regression of or inhibiting the progression of such symptom(s) to any clinically measurable degree. The amount of a therapeutic agent that is effective to alleviate any particular disease symptom (also referred to “therapeutically effective amount”) may vary according to factors such as the disease state, age, and weight of the subject, and the ability of the drug to elicit a desired response in the subject. Whether a disease symptom has been alleviated can be assessed by any clinical measurement typically used by physicians or other skilled healthcare providers to assess the severity or progression status of that symptom. While an embodiment of the present application (e.g., a treatment method or article of manufacture) may not be effective in alleviating the disease symptom(s) of interest in every subject, it should alleviate the target disease symptom(s) of interest in a statistically significant number of subjects as determined by any statistical test known in the art such as the Student's t-test, the chi-square test, the U-test according to Mann and Whitney, the Kruskal-Wallis test (H-test), Jonckheere-Terpstra-test and the Wilcoxon-test.


“Effective amount” encompasses an amount sufficiently to ameliorate or prevent a symptom or sign of a medical condition. Effective amount also means an amount sufficiently to allow or facilitate diagnosis. An effective amount for a particular subject or veterinary subject may vary depending on factors such as the condition being treated, the general health of the subject, the route and dose of administration and the severity of side effects. An effective amount can be the maximal dose or dosing protocol that avoids significant side effects or toxic effects.


“Exogenous” refers to substances that are produced outside an organism, cell, or human body, depending on the context. “Endogenous” refers to substances that are produced within a cell, organism, or human body, depending on the context.


“Identity” refers to sequence similarity between two polynucleotide sequences or between two polypeptides. When a position in the two sequences to be compared is occupied by the same base or amino acid monomer subunit, e.g., if a position in each of two DNA molecules is occupied by adenine, then the molecules are homologous at that position. The percent of identity between two sequences is a function of the number of matched or homologous positions shared by the two sequences divided by the number of positions to be compared×100%. For example, if 6 of 10 positions in two sequences are matched or homologous when the sequences are optimally aligned, then the two sequences share 60% identity. Generally, the comparison is made when two sequences are aligned to give maximum percent identity.


As used herein, the expressions “cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny thereof. Thus, the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without considering the number of passages. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or random mutations. Mutant progeny obtained by screening, which have the same function or biological activity as that of originally transformed cell, are also contemplated. Where distinct designations are intended, it will be clear from the context.


“Optional” or “optionally” means that the event or situation that follows may but does not necessarily occur, and the description includes the instances in which the event or circumstance does or does not occur. For example, “optionally comprises 1-3 antibody heavy chain variable regions” means that the antibody heavy chain variable region with specific sequence can be, but not necessarily, be present.


“Pharmaceutical composition” refers to one containing a mixture of one or more antibodies or antigen-binding fragments thereof according to the present invention or a physiologically/pharmaceutically acceptable salt or prodrug thereof with other chemical components, as well as additional components such as physiologically/pharmaceutically acceptable carriers and excipients. The pharmaceutical composition aims at promoting the administration to an organism, facilitating the absorption of the active ingredient and thereby exerting a biological effect.


Hereinafter, the present application is further described with reference to examples; however, the scope of the present application is not limited thereto. In the examples of the present invention, where specific conditions are not described, the experiments are generally conducted under conventional conditions as described in Antibodies, A Laboratory Manual and Molecular Cloning Manual, by Cold Spring Harbor, or under conditions proposed by the material or product manufacturers. Where the source of the reagents is not specifically given, the reagents are commercially available conventional reagents.


Example 1: Preparation of Antibodies

The anti-human 4-1BB monoclonal antibody library was produced by immunizing mice. Eight-week-old BalB/C and A/J strain female mice (Comparative Medical Center of Yangzhou University, animal production license number: SCXK (Su) 2017-007) were used in the experiments. Feeding environment: SPF level. After the mice were purchased, the animals were kept in the laboratory for 1 week, with 12/12-hour light/dark cycle, at temperature of 20-25° C., and with a humidity of 40-60%.


The antigen for immunization was human 4-1BB recombinant protein with Fc tag (human 4-1BB-Fc), purchased from Acro Biosystems, Cat No #41B-H5258: expressed in HEK293, AA Gln 25-Gln 186 (http://www.acrobiosystems.cn/P150-Human_4-1BB_%7CTNFRSF9_ProteinC-Fc_Tag HEK293_expressed.html; Accession number #NP_001552.2). The antigen was emulsified with Freund's adjuvant (Sigma, Cat No: F5881/F5506): the first immunization was performed with Freund's complete adjuvant, and the rest booster immunizations were performed with Freund's incomplete adjuvant. The ratio of antigen to adjuvant was 1:1, and 25 μg protein/200 μl/mouse was injected for each immunization. The emulsified antigen was inoculated via subcutaneous injections at multiple sites (generally 6-8 sites on the back) and an injection at a single site on the toe palm, on day 0, 21, 35, and 49 (first immunization and 3 booster immunizations). The last booster immunization was performed 3 days before the splenocyte fusion, via intraperitoneal (IP) injection of 50 μg protein/mice of the antigen solution formulated with physiological saline.


On day 42 and 56, the blood samples were collected from the immunized mice and the antisera were isolated. The sera of the mice were subjected to ELISA method described in Example 2, to determine the mouse serum titer of the antibody and to determine the neutralizing activity in blocking 4-1BB/4-1BBLbinding.


After the fourth immunization, the mice with higher serum titer of the antibody and having higher activity in blocking 4-1BB/4-1BBL were selected for splenocyte fusion. Spleen lymphocytes were fused with myeloma cell Sp2/0 cells (ATCC® CRL-8287™) using optimized PEG-mediated fusion procedure, and then the fused cells were plated into 96-well plates and cultured in HAT medium for 10 to 14 days until the growth of hybridoma cells were observed. About 2 weeks after the cell fusion, the binding reactivity of the hybridoma culture medium to 4-1BB recombinant protein was detected by indirect ELISA assay described in Example 2, and the positive hybridoma cell supernatant was able to recognize and bind to 4-1BB recombinant protein.


The 4-1BB positive hybridoma cells identified by the primary screening were picked out, transferred to a new 96-well plate, and numbered according to the position of the fusion plate. After further culture for 3-4 days, the culture supernatants were collected and were subjected to indirect ELISA assay described in Test Example 1 to confirm the positive wells again. Once the culture medium was collected, the medium was replenished. For the positive wells, the supernatants were collected again for cell-based functional screening 3-4 days later, to select hybridoma cells secreting activated anti-4-1BB antibody. Brief procedures for functional screening were as follows: collecting cell strains with 4-1BB reporter (HEK293F-h4-1BB-NFkb) which were in logarithmic growth phase and seeding into 96-well plates at volume of 20 μl/well (comprising approximately 10,000 cells). Adding mouse Fc-specific secondary antibody (final concentration of 15 nM, for cross-linking the supernatant antibody) and 60 μl cell supernatant or antibody solution to be tested, incubating for 5 hours, measuring the activity of luciferase with ONE-Glo™ Luciferase Assay System (Promega, E6120), and selecting the primary hybridoma cells in which the anti-4-1BB antibody was activated according to the measured values. In the experiment, the antibody Urelumab (BMS) and MOR7480 (Pfizer) were used as positive references, and hIgG was used as a negative reference. The screening data of the top 17 hybridoma were shown in the table below.









TABLE 1







Hybridoma screening data









No.
Hybridoma No
luciferase activity












1
B1E10
2519.06


2
B1E7
1097.93


3
B1A7
964.85


4
B1H9
803.25


5
B1C8
779.48


6
3C1
769.14


7
B1H1
760.47


8
3F5
674.77


9
5G1
673.10


10
B1B9
670.17


11
B1H2
665.41


12
8D5
660.61


13
B1G1
655.91


14
B1C9
632.14


15
6A1
626.03


16
B1G12
622.64


17
2H11
607.20


PC
Urelumab (10 nM)
2193.01


PC
MOR 7480 (10 nM)
1001.57


NC
hIgG (10 nM)
289.55









The three most preferred original hybridoma cells were limiting diluted by two rounds of subcloning to obtain the monoclonal cell lines (B1E10, B1E7 and B1A7), which were then cloned and sequenced. Total cellular RNAs were obtained by conventional RNA extraction techniques, and then the PCR products of the monoclonal antibody variable regions were obtained by reverse transcription polymerase chain reaction (RT-PCR). The PCR products were separated and recovered on agarose gel, and then were cloned into a gene vector and transformed into Escherichia coli. Several transformed colonies were randomly selected and were subjected to PCR amplification to amplify the monoclonal antibody variable region for gene sequencing.


The heavy and light chain variable region sequences of murine mAb B1E10 were as follows:









B1E10 HCVR


SEQID NO: 1


EVQLVESGGGLVEPGGSLKLSCAASGFTFGDYYMYWVRQTPEKRLEWVAT





ISDGGSYTYYPDNVKGRFTISRDNAKNSLDLQMSHLKSEDTAMYYCARYY





SKALYAMDYWGRGTSVTVSS





B1E10 LCVR


SEQID NO: 2


DIVLTQSPASLAVSLGQRATLSCRASKSVSTSGFSYIHWYQQKPGQPPKL





LIYTASNLESGVPARFSGSGSGTDFTLNIHPVEEEDAATYYCQHSRELPL





TFGAGTKLEVK






It comprises the following CDR sequences:









TABLE 2







CDR sequences











Name
sequence
SEQID NO







HCDR1
GFTFGDYYMY
SEQID NO: 3







HCDR2
TISDGGSYTYYPDNVKG
SEQID NO: 4







HCDR3
YYSKALYAMDY
SEQID NO: 5







LCDR1
RASKSVSTSGFSYIH
SEQID NO: 6







LCDR2
TASNLES
SEQID NO: 7







LCDR3
QHSRELPLT
SEQID NO: 8










The heavy and light chain variable region sequences of murine mAb B1E7 were as follows:









B1E7 HCVR


SEQID NO: 9


EVQLQQSGAELVRAGSSVEMSCKASGYTFTSYGLNWVKQRPGQGLEWIGY





INPGSGYTKYNEKFEGKTTLTVDKSSSTVYMQLRSLTSEDSAVYFCARWG





LGRNWNFAVWGTGTTVTVSS





B1E7 LCVR


SEQID NO: 10


DIVLTQSPASLAVSLGQRATMSCRASESVDSYGNTFMHWYQQKPGQPPKL





LIYRASNLESGIPARFSGSGSGTDFTLTINPVEADDVATYYCQQSNEDPL





TFGSGTKLEIK






It comprises the following CDR sequences:









TABLE 3







CDR sequences











Name
sequence
SEQID NO







HCDR1
GYTFTSYGLN
SEQID NO: 11







HCDR2
YINPGSGYTKYNEKFEG
SEQID NO: 12







HCDR3
WGLGRNWNFAV
SEQID NO: 13







LCDR1
RASESVDSYGNTFMH
SEQID NO: 14







LCDR2
RASNLES
SEQID NO: 15







LCDR3
QQSNEDPLT
SEQID NO: 16










The heavy and light chain variable region sequences of murine mAb B1A7 were as follows:









B1A7 HCVR


SEQID NO: 37


QVTLKESGPGILQPSQTLSLTCSFSGFSLSTSGMGVGWIRQPSGKGLEWL





AHIWWDDVKRYNPALKSRLTISKDTSSSQVFLKIASVDTADTATYYCAQM





TSSVFAYWGQGTLVTVSA





B1A7 LCVR


SEQID NO: 38


DIVLTQSPDSLAVSLGQRATISCRASESVDSYGHSFMHWYQQKPGQPPKL





LIYRASNLESGIPARFSGSGSRTDFTLTINVVEADDVATYYCHQSYEEPW





TFGGGTKLEIK






It comprises the following CDR sequences:









TABLE 4







CDR sequences











Name
sequence
SEQID NO







HCDR1
GFSLSTSGMGVG
SEQID NO: 39







HCDR2
HIWWDDVKRYNPALKS
SEQID NO: 40







HCDR3
MTSSVFAY
SEQID NO: 41







LCDR1
RASESVDSYGHSFMH
SEQID NO: 42







LCDR2
RASNLES
SEQID NO: 43







LCDR3
HQSYEEPWT
SEQID NO: 44










Example 2: Identification and Screening of Antibodies by ELISA Assay

1. Binding Assay Via In Vitro Indirect ELISA:


Human 4-1BB-Fc protein was diluted to a concentration of 2 μg/ml with carbonate buffer pH 9.6, and added to a 96-well microtiter plate at a volume of 100 μl/well, and placed in an incubator at 37° C. for 2 hours. The plate was washed once with PBST (PBS containing 0.05% Tween-20, pH 7.4), 5% skim milk (skim milk powder, Bright Dairy) blocking solution diluted with PBST, was added at 200 μl/well, and incubated at 37° C. for 2 hours or at 4° C. overnight (16-18 hours) for blocking. Once the blocking was finished, the blocking solution was discarded and the plate was washed 4 times with PBST buffer.


The test mouse sera (or antibodies, cell culture supernatants) were diluted to various concentrations with sample dilution solution containing 5% NHS (2.5% skim milk in PBST) (i.e., the final concentration of normal human IgG is 1 mg/ml), incubated at room temperature for 40 minutes, added into the microtiter plate, 100 μl/well, and incubated at 37° C. for 40 minutes. After the incubation, the plate was washed 4 times with PBST, 100 μl/well of HRP-labeled goat anti-mouse secondary antibody (Jackson Immuno Research, cat #115-036-071) diluted with PBST was added, and incubated at 37° C. for 40 minutes. The plate was washed 4 times with PBST, 100 μl/well of TMB chromogenic substrate (Huzhou Innoreagents Biotechnology Co., Ltd.) was added, incubated at room temperature for 10-15 min in the dark, and 50 μl/well 1M H2SO4 was added to terminate the reaction. The absorbance was read at 450 nm under a microplate reader (Beijing Prang New Technology Co., Ltd., Model DNM-9602) and the resulting data were analyzed.


2. Binding Assay Via In Vitro Capture ELISA

The goat anti-mouse IgG secondary antibody (Jackson Immuno Research, cat #115-006-071) was diluted to a concentration of 2 μg/ml with PBS buffer, pH 7.4, added into a 96-well microtiter plate at 100 μl/well, and placed in an incubator at 37° C. for 2 hours. The plate was washed once with PBST, 200 μl/well of 5% skim milk (skim milk powder, Bright Dairy) blocking solution diluted with PBST was added, and incubated at 37° C. for 2 hours or at 4° C. overnight (16-18 hours) for blocking. After the blocking was finished, the blocking solution was discarded and the plate was washed 4 times with PBST.


The test mouse sera or antibodies were diluted to various concentrations with 5% NHS sample dilution solution (2.5% skim milk in PBST), incubated at room temperature for 40 minutes, added into the plate at 100 μl/well, and incubated at 37° C. for 40 minutes. After the incubation, the plate was washed 4 times with PBST, 100 μl/well of the biotinylated 4-1BB-his protein (Sinobiological, Beijing #10041-H08H) diluted with the sample dilution solution was added, and incubated at 37° C. for 40 minutes. After the incubation, the plate was washed 4 times with PBST, 100 μl/well of HRP-labeled streptavidin (Jackson Immuno Research, cat #016-030-084) diluted with PBST was added, and incubated at 37° C. for 40 minutes. The plate was washed 4 times with PBST, 100 μl/well of TMB chromogenic substrate (Huzhou Innoreagents Biotechnology Co., Ltd.) was added, incubated at room temperature for 10-15 min in the dark, and 50 μl/well 1M H2SO4 was added to terminate the reaction. The absorbance was read at 450 nm under a microplate reader (Beijing Prang New Technology Co., Ltd., Model DNM-9602) and the resulting data were analyzed. The capture ELISA results of three hybridoma mouse antibodies and the Pfizer reference antibody MOR7480 and the BMS reference antibody Urelumab were shown in FIG. 1. The EC50 values were shown in the table below.









TABLE 5







EC50 values












Antibody
B1E10
B1E7
B1A7
Urelumab
MOR7480





EC50 (ng/ml)
880.9
332
55.95
191.9
83.97









3. Blocking the Binding of 4-1BB to 4-1BBL by the Antibodies Via ELISA Assay

The 4-1BB ligand (i.e., Tumor necrosis factor ligand superfamily member 9, TNFSF9) is capable of binding to 4-1BB on the T cell surface and can activate T cell immune function as a positive costimulatory signal. In this assay, the selected anti-human 4-1BB antibodies were tested for their ability of blocking the binding of human 4-1BB protein to human 4-1BB ligand via an in vitro blocking assay.


Specifically, the human 4-1BB-his protein was coated onto a 96-well plate, anti-human 4-1BB antibodies (or mouse sera) was added and incubated, and human 4-1BBL-Fc protein was added, incubated and washed. The amount of 4-1BBL-Fc binding to 4-1BB coated on the bottom of the plate was determined by HRP-labeled goat anti-human IgG secondary antibody (Jackson Immuno Research, cat #109-036-098), and the IC50 values of the 4-1BB antibodies in blocking the 4-1BB active epitope were calculated.


The 4-1BB-his protein was diluted to a concentration of 2 μg/ml with a carbonate buffer pH 9.6, and added into a 96-well microtiter plate at a volume of 100 μl/well, and placed in an incubator at 37° C. for 2 hours. The plate was washed once with PBST, 200 μl/well of 5% skim milk (skim milk powder, Bright Dairy) blocking solution diluted with PBST was added, and incubated at 37° C. for 2 hours or at 4° C. overnight (16-18 hours) for blocking. After the blocking was finished, the blocking solution was discarded and the plate was washed 4 times with PBST.


The test mouse sera or antibodies and internal control 4-1BBL were diluted to various concentrations with a sample diluent solution (2.5% skim milk in PBST), added into a plate at 100 μl/well, and incubated in an incubator at 37° C. for 40 minutes. After the incubation, the plate was washed 4 times with PBST, 100 μl/well of 0.8 ng/ml 4-1BBL-Fc (Acro, #41L-H5257) protein diluted with a sample dilution solution was added, and incubated at 37° C. for 40 minutes. After the incubation, the plate was washed 4 times with PBST, 100 μl/well of HRP-labeled goat anti-human IgG secondary antibody (Jackson Immuno Research, cat #109-036-098) diluted with PBST was added, and incubated at 37° C. for 40 minutes. The plate was washed 4 times with PBST, 100 μl/well TMB chromogenic substrate (Huzhou Innoreagents Biotechnology Co., Ltd.) was added, incubated at room temperature for 10-15 min in the dark, and 50 μl/well of 1M H2SO4 was added to terminate the reaction. The absorbance was read at 450 nm under a microplate reader (Beijing Prang New Technology Co., Ltd., Model DNM-9602) and the resulting data were analyzed. The results of the three hybridoma mouse antibodies and Pfizer reference antibody in blocking 4-1BBL were shown in FIG. 2, and the EC50 values were shown in the table below.









TABLE 6







EC50 values














B1E10
B1E7
B1A7
Urelumab
MOR7480
4-1BBL

















EC50 (ng/ml)
N/A
591.9
416.9
N/A
N/A
162.1









As can be seen from the table, both B1E7 and B1A7 effectively block the epitope of 4-1BBL, while B1E10, MOR7480 and Urelumab cannot block the binding to 4-1BBL.


Example 3: Construction and Expression of Anti-4-1BB Recombinant Chimeric Antibodies

Site-directed amino acid mutations were introduced into FR region (framework region) of the heavy chain variable region (VH) and the light chain variable region (VL) of the murine antibody B1E10 and B1E7 of the present application, respectively. According to different combinations of amino acid mutations, different humanized antibody heavy and light chains were designed, and plasmids with different light and heavy chain combinations were transfected into cells to produce humanized antibodies.


The heavy chain vector was designed as follows: signal peptide+mutated heavy chain variable region sequence+human IgG1 constant region sequence. The light chain vector was designed as follows: signal peptide+mutated light chain variable region sequence+human Kappa constant region sequence. The above sequences were inserted into pCEP4 vector, respectively. After the vector plasmids were confirmed, the plasmids were extracted and subjected to sequencing for verification. The validated plasmids were transfected into human 293F cells with PEI. The continuously cultured293F cells were cultured in serum-free medium (Shanghai Aupmaxi, OPM-293CD03) until reaching logarithmic growth phase, and used for cell transfection. 21.4 μg of the plasmid comprising the humanized antibody light chain and 23.6 μg of plasmid comprising the humanized antibody heavy chain were dissolved in 10 ml of Opti-MEM® I Reduced Serum Medium (GIBCO, 31985-070), mixed thoroughly, and then added with 200 μg of PEI, mixed, incubated at RT for 15 min, and added into 50 mL of cells. The cell culture conditions: 5% CO2, 37° C., 125 rpm/min. During culturing, supplementary material was added on day 1 and day 3, until the cell viability was less than 70%, and the cell supernatants were collected and centrifuged. The centrifuged cell culture medium was loaded onto an antibody purification affinity column, and washed with phosphate buffer, eluted with glycine-hydrochloric acid buffer (pH 2.7, 0.1M Gly-HCl), neutralized with 1 M Tris hydrochloric acid, pH9.0, and dialyzed against phosphate buffer to finally obtain the purified chimeric antibody ChB1E10 and ChB1E7.


Example 4: In Vitro Cell Reporter Assay of Anti-4-1BB Antibodies

The stable cell strain HEK293F-h4-1BB-NFkB (Shanghai ChemPartner Chemistry) was thawed with DMEM/FBS medium, and the passaged cells were seeded into a 96-well plate at 4×103 cells per well. Each antibody (including the test antibodies, the reference antibody, the IgG negative control, and so forth) was mixed with anti-F(ab′)2 at a set ratio, serially diluted, and incubated at 37° C. for 0.5 hours in a cell culture incubator. The antibodies were then added into the cell plate, incubated for 5 hours in the incubator, 100 μl of detection buffer was added to lyse the cells, and incubated at room temperature for 10 minutes in the dark. Fluorescence readings were measured with Envision and activity-concentration curves were plotted. The results of the three murine antibodies obtained from in vitro reporter assay were shown in FIG. 3. The reporter reading value for B1E10 was stronger than that for PfizerreferenceMOR7480; whereas the value for B1E7 was slightly weaker than the reference.


Example 5: The In Vitro Binding Affinity and Kinetics of the Recombinant Antibodies

Biacore is a well-known method for the objective detection of the affinity and kinetics of proteins. The present inventors analyzed the characteristic affinity and binding kinetics of the 4-1BB antibodies to be tested of this application via Biacore T200 (GE).


The recombinant anti-4-1BB antibodies to be tested of the present application were covalently linked to CMS (GE) chip by NHS standard amino coupling method with a kit available from Biacore. Then, a series of gradient concentrations of human 4-1BB His protein (Sinobiological, Beijing #10041-H08H) diluted in the same buffer were injected at a flow rate of 10 μL/min. Regeneration was performed with regeneration reagents provided in the kit. Antigen-antibody binding kinetics were followed for 3 minutes and dissociation kinetics were followed for 10 minutes. The resulting data were analyzed using 1:1 (Langmuir) binding model with BIAevaluation software available from GE. The ka (kon), kd (koff) and KD values of the chimeric antibodies determined by this method were shown in the table below.









TABLE 7







Kinetic parameters of the antibodies










Antibodies to be tested
ka (1/Ms)
kd (1/s)
KD (M)





ChB1E7
4.053E+6
5.997E−3
1.480E−9 


ChB1E10
1.282E+6
5.421E−4
4.228E−10


MOR7480 (Pfizer)
8.051E+5
3.359E−4
4.172E−10









Example 6: Humanization of the Mouse Antibodies

In the present example, two strains (B1E10 and B1E7) having the strongest functional activity among the resulting murine antibodies were humanized. Based on the typical structure of the murine antibody VH/VLCDR obtained, the heavy and light chain variable region sequences were aligned against those disclosed in the antibody Germline database to obtain a human germline template with high homology.


The human germline light chain framework region was derived from human κ light chain gene, and the human germline light chain template used in the present application were preferably IGKV3-11*01 (for B1E10) and IGKV4-1*01 (for B1E7). The human germline heavy chain framework region was derived from human heavy chain, and the human germline heavy chain templates used in the present application were preferably IGHV3-30*01 (for B1E10) and IGHV1-46*01 (for B1E7).









Human germline heavy chain template IGHV3-30*01


(SEQ ID NO: 21):


QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWVAV





ISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR





Human germline light chain template IGKV3-11*01


(SEQ ID NO: 22):


EIVLTQSPATLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLLIYD





ASNRATGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSNWP





Human germline heavy chain template IGHV1-46*01


(SEQ ID NO: 23):


QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWMGI





INPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCAR





Human germline light chain template IGKV4-1*01


(SEQ ID NO: 24):


DIVMTQSPDSLAVSLGERATINCKSSQSVLYSSNNKNYLAWYQQKPGQPP





KLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQYYST





P






The CDR regions of the murine antibody were grafted onto the selected humanized template, as a substitute for the humanized variable regions, and then recombined with IgG constant region. Then, based on the three-dimensional structure of the murine antibody, the embedding residues, the residues that directly interact with the CDR regions, and the residues that have an important influence on the conformation of VL and VH were subjected to back mutation. Further, for antibody B1E10, the chemically unstable amino acid residue in the CDR regions was optimized, i.e., HCDR2: TISDGGSYTYYPDNVKG (SEQ ID NO: 4) was optimized as: TISEGGSYTYYPDNVKG (SEQ ID NO: 45). A series of humanized molecules were obtained.


The heavy chain variable region sequences of the two antibodies are shown in SEQ ID NOs: 25-27 and SEQ ID NOs: 31-33, respectively; and the light chain variable region sequences are set forth in SEQ ID NOs: 28-30 and SEQ ID NO: 34-36, respectively.









huB1E10-VH-a (SEQ ID NO: 25):


EVQLVESGGGVVQPGRSLRLSCAASGFTFGDYYMYWVRQAPGKGLEWVAT





ISEGGSYTYYPDNVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARYY





SKALYAMDYWGRGTLVTVSS





huB1E10-VH-b (SEQ ID NO: 26):


EVQLVESGGGVVQPGRSLRLSCAASGFTFGDYYMYWVRQAPGKGLEWVAT





ISEGGSYTYYPDNVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARYY





SKALYAMDYWGQGTLVTVSS





huB1E10-VH-c (SEQ ID NO: 27):


EVQLVESGGGVVQPGRSLRLSCAASGFTFGDYYMYWVRQAPGKGLEWVAT





ISEGGSYTYYPDNVKGRFTISRDNAKNTLYLQMNSLRAEDTAVYYCARYY





SKALYAMDYWGRGTLVTVSS





huB1E10-VL-a (SEQ ID NO: 28):


EIVLTQSPATLSLSPGERATLSCRASKSVSTSGFSYIHWYQQKPGQAPRL





LIYTASNLESGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQHSRELPL





TFGQGTKLEVK





huB1E10-VL-b (SEQ ID NO: 29):


EIVLTQSPATLSLSPGERATLSCRASKSVSTSGFSYIHWYQQKPGQPPRL





LIYTASNLESGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQHSRELPL





TFGQGTKLEVK





huB1E10-VL-c (SEQ ID NO: 30):


EIVLTQSPATLSLSPGERATLSCRASKSVSTSGFSYIHWYQQKPGQPPRL





LIYTASNLESGVPARFSGSGSGTDFTLTISSLEPEDFAVYYCQHSRELPL





TFGQGTKLEVK





huB1E7-VH-a (SEQ ID NO: 31):


EVQLVQSGAEVKKPGSSVKVSCKASGYTFTSYGLNWVRQAPGQGLEWIGY





INPGSGYTKYNEKFEGRTTLTVDKSTSTVYMELSSLRSEDTAVYFCARWG





LGRNWNFAVWGTGTLVTVSS





huB1E7-VH-b (SEQ ID NO: 32):


EVQLVQSGAEVKKPGSSVKVSCKASGYTFTSYGLNWVRQAPGQGLEWMGY





INPGSGYTKYNEKFEGRTTLTVDKSTSTVYMELSSLRSEDTAVYFCARWG





LGRNWNFAVWGTGTLVTVSS





huB1E7-VH-c (SEQ ID NO: 33):


EVQLVQSGAEVKKPGSSVKVSCKASGYTFTSYGLNWVRQAPGQGLEWMGY





INPGSGYTKYNEKFEGRVTITADKSTSTAYMELSSLRSEDTAVYYCARWG





LGRNWNFAVWGQGTLVTVSS





huB1E7-VL-a (SEQ ID NO: 34):


DIVLTQSPDSLAVSLGERATINCRASESVDSYGNTFMHWYQQKPGQPPKL





LIYRASNLESGIPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQSNEDPL





TFGQGTKLEIK





huB1E7-VL-b (SEQ ID NO: 35):


DIVLTQSPDSLAVSLGERATINCRASESVDSYGNTFMHWYQQKPGQPPKL





LIYRASNLESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQSNEDPL





TFGQGTKLEIK





huB1E7-VL-c (SEQ ID NO: 36):


DIVMTQSPDSLAVSLGERATINCRASESVDSYGNTFMHWYQQKPGQPPKL





LIYRASNLESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQSNEDPL





TFGQGTKLEIK






After a small-scale expression test for the above combinations of each light and heavy chain (as described in Example 3), a comparison of the number of the back mutations, and a comprehensive evaluation, humanized huB1E10 (with VH-c heavy chain and VL-c light chain) and huB1E7 antibody molecule (with VH-c heavy chain and VL-c light chain) were finally selected. The intact light and heavy chain sequences (IgG1 subtype) of these antibodies are set forth in SEQ ID NOs: 17-20.









huB1E10 HC


SEQID NO: 17


EVQLVESGGGVVQPGRSLRLSCAASGFTFGDYYMYWVRQAPGKGLEWVAT





ISEGGSYTYYPDNVKGRFTISRDNAKNTLYLQMNSLRAEDTAVYYCARYY





SKALYAMDYWGRGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVK





DYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSNFGTQT





YTCNVDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTL





MISRTPEVTCVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFR





VVSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTL





PPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSD





GSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK





huB1E10 LC


SEQID NO: 18


EIVLTQSPATLSLSPGERATLSCRASKSVSTSGFSYIHWYQQKPGQPPRL





LIYTASNLESGVPARFSGSGSGTDFTLTISSLEPEDFAVYYCQHSRELPL





TFGQGTKLEVKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKV





QWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEV





THQGLSSPVTKSFNRGEC





huB1E7 HC


SEQID NO: 19


EVQLVQSGAEVKKPGSSVKVSCKASGYTFTSYGLNWVRQAPGQGLEWMGY





INPGSGYTKYNEKFEGRVTITADKSTSTAYMELSSLRSEDTAVYYCARWG





LGRNWNFAVWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVK





DYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSNFGTQT





YTCNVDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSVFLFPPKPKDTL





MISRTPEVTCVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFR





VVSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTL





PPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSD





GSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK





huB1E7 LC


SEQID NO: 20


DIVMTQSPDSLAVSLGERATINCRASESVDSYGNTFMHWYQQKPGQPPKL





LIYRASNLESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQSNEDPL





TFGQGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKV





QWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEV





THQGLSSPVTKSFNRGEC






Example 7: Functional Assay of the Activation of PBMC Cells In Vitro

The cell viability in the presence of anti-4-1BB antibodies were determined via proliferation assay of the fresh human peripheral blood mononuclear cells (PBMCs).


Fresh human PBMCs were isolated from fresh blood containing anticoagulant heparin by Ficoll density gradient centrifugation. Various concentrations of anti-4-1BB antibody by gradient dilution and 3 μg/mL anti-CD3 antibody were coated in ELISA plate and incubated at 4° C. overnight. The plate was washed on the next day, peripheral blood mononuclear cells at a density of 2×105/well was added, and incubated in an incubator at 37° C., 5% CO2 for 7 days. After the incubation, the proliferation of PBMCs was measured with a CCK-8 kit. The values of 0D450 were measured, and the EC50values were calculated from the 0D450 values.


As a result, as shown in FIG. 4, the humanized antibody huB1E7 and huB1E10 (both are IgG1 subtype) showed higher efficiency on activating PBMC proliferation, compared to the reference antibody MOR7480, wherein huB1E7 showed a stronger activation function.


Example 8: Activity Data of the Humanized Antibodies

An in vitro reporter activity assay was performed for the humanized antibodies (the procedures were as described in Example 4). The results were shown in FIG. 5. Consistent with the tendency as presented in murine antibodies, huB1E10 showed an increased activity of activating the reporter, when compared to the reference antibody MOR7480, while huB1E7 showed a weaker activity than the reference. This tendency was inconsistent with what was observed for the PBMC proliferation activity.


An in vitro affinity kinetic assay was performed for the humanized antibodies (the procedures were as described in Example 5) and the results were shown in the table below:









TABLE 8







Affinity kinetics assay










Antibodies to be tested
ka (1/Ms)
kd (1/s)
KD (M)





huB1E7
3.392E+6
6.627E−3
1.954E−9 


huB1E10
9.847E+6
2.630E−3
2.671E−10


MOR7480 (Pfizer)
8.051E+5
3.359E−4
4.172E−10









The results showed that both humanized antibody huB1E7 and huB1E10 retained binding activity to 4-1BB, wherein the affinity of huB1E10 was nearly twice than that of the reference antibody MOR7480, and the affinity of huB1E7 was slightly weaker.


Example 9: Inhibition of Growth of Tumor Cells in Transgenic Mice by Theanti-4-1BB Antibodies

The hu4-1BB transgenic C57BL/6 mice (female) were housed in an SPF facility environment at the Animal Center of Beijing Biotech Co., Ltd., using an independent ventilation cage with IVC. Mice were subjected to 3-7 days of adaptive feeding before the experiment. Temperature: 20-26° C.; Humidity: 40-70%; Lighting: 12/12-hour light/dark cycle.


MC38 cells were purchased from Shanghai Shunran Biotechnology Co., Ltd., and the cells were cultured in Dulbecco's Modified Eagle Medium supplemented with 10% inactivated fetal bovine serum (Dulbecco's Modified Eagle's Medium) in an incubator at 37° C., 5% CO2.


Cells were sub-cultured every 3 to 4 days, when approaching complete confluence. The hu4-1BB humanized homozygous mice were inoculated subcutaneously on the right flank with a concentration of 5×106/ml MC38 tumor cells resuspended in PBS, 0.1 ml/mice, and a total of 50 mice were inoculated.


The tumor volumes were measured twice using a caliper, and the long and short diameters of the tumors were measured. The tumor volume was calculated as follows: the tumor volume (mm3)=0.5×(long diameter×short diameter2).


Relative tumor inhibition rate TGI (%): TGI %=(1−T/C)×100%. T/C % means the relative tumor increase rate, that is, it means the percentage of tumor volume or tumor weight of the treatment group relative to that of the control group at certain time point. T and C are the tumor volume (TV) or tumor weight (TW) of the treatment group and that of IgG1 control group at a specific time point, respectively.


When the average tumor volume reached 140 mm3, the mice with moderate tumor volume were selected and randomly assigned to each experimental group according to the tumor volume. Drug administration was started on the day of grouping. The specific dosage regimen was shown in the following table. After the last administration, the body weight and tumor growth of the animals were observed for 9 days, then the experiment was ended. During the observation, tumor volume and animal body weight were measured twice a week, and the measured values were recorded, as shown in Table 10. At the end of the experiment, the tumor was removed from the mice and weighed. The animals were euthanized and the tumors were photographed.









TABLE 9







Dosage regimen















Number of








animals
dosage
Administration
Administration
Number of


Group
Test drug
(mice)
(mg/kg)a
route
frequency
doses





1
Blank control (PBS)
6

i.p. injection
Q3d
6


2
huB1E10-IgG1
6
10
i.p. injection
Q3d
6


3
huBlE10-IgG2
6
10
i.p. injection
Q3d
6


4
MOR7480-IgG2
6
10
i.p. injection
Q3d
6


5
huBlE7-IgG1
6
10
i.p. injection
Q3d
6









The results of the pharmacologic test were shown in Table 10, FIG. 6 and FIG. 7. The efficacy of antibody huB1E10-IgG2 subtype was close to that of Pfizer reference agent MOR7480 at the same dosage, while the efficacy of huB1E10-IgG1 subtype was significantly enhanced, while without significant effect on the body weight of mice.


The antibody huB1E7-IgG1 showed significantly more potent efficacy, wherein 2 out of 6 tumor-bearing mice showed complete elimination of tumors.









TABLE 10







Tumor volume in mouse (mm3)









Days after inoculation
















8
11
15
18
22
25
29
32










Animal
Days after grouping
















group
No.
0
3
7
10
14
17
21
24



















G1
32328
125.9
205.7
367.4
483.4
877.3
1355.7
1141.7
1458.9


Blank control-,
32329
128.0
259.4
339.2
561.4
833.4
1157.0
1146.3
1179.1


i.p., Q3DX6
32333
120.0
127.2
217.0
225.8
327.8
513.8
527.1
765.5



32338
124.5
207.5
551.6
554.0
1262.2
1736.6
1765.3
1625.0



32355
157.1
343.0
388.3
424.3
944.0
1469.5
2390.5
3526.2



32375
186.7
205.0
182.3
272.1
367.0
606.0
517.8
866.5



mean
140
225
341
420
769
1140
1248
1570



SD
26
72
133
143
359
488
728
1014



SEM
11
29
54
58
147
199
297
414



CV value
18.7%
31.9%
38.9%
33.9%
46.8%
42.8%
58.3%
64.6%


G2
32331
188.4
257.3
263.7
187.6
166.7
156.4
96.0
74.8


B1E10-IgG1
32332
159.2
268.8
193.4
189.9
143.0
93.8
35.0
34.5


10 mg/kg,
32349
144.4
337.0
253.4
158.2
188.2
122.8
98.9
144.7


i.p., Q3DX6
32354
109.1
152.1
144.5
122.4
91.1
51.2
38.4
33.0



32358
105.4
264.0
252.9
274.7
416.6
381.7
378.4
337.0



32359
134.6
294.2
485.0
429.0
546.1
610.0
816.9
729.6



mean
140
262
265
227
259
236
244
226



SD
31
61
117
111
180
217
308
272



SEM
13
25
48
45
74
88
126
111



CV value
22.4%
23.4%
44.0%
48.9%
69.7%
91.8%
126.4%
120.5%



TGI

−0.449
0.375
0.690
0.811
0.904
0.906
0.940



P value
0.991
0.352
0.320
0.026
0.011
0.002
0.011
0.011


G3
32340
140.2
226.4
296.5
283.2
292.0
612.3
934.0
1246.3


B1E10-IgG2
32344
134.9
210.1
174.2
185.1
146.7
110.1
76.8
58.9


10 mg/kg,
32347
188.1
160.4
186.3
126.8
149.1
158.8
313.9
239.7


i.p., Q3DX6
32348
150.5
170.4
219.5
270.8
329.0
308.3
616.8
961.2



32350
119.1
254.0
371.5
370.8
523.4
835.1
1272.3
1370.7



32362
109.4
292.3
481.7
424.1
571.6
712.1
1130.9
1170.3



mean
140
219
288
277
335
456
724
841



SD
28
50
120
111
181
304
471
555



SEM
11
20
49
45
74
124
192
227



CV value
19.7%
22.9%
41.8%
40.1%
53.9%
66.8%
65.0%
66.0%



TGI

0.068
0.263
0.512
0.690
0.684
0.473
0.510



P value
1.000
0.876
0.488
0.080
0.025
0.015
0.169
0.153


G4
32326
160.2
176.4
201.1
150.6
97.5
30.8
21.3
0.0


MOR7480-IgG2
32341
114.4
246.8
249.0
244.9
583.4
903.8
867.7
1211.2


10 mg/kg,
32357
188.2
440.1
520.3
453.1
452.2
548.0
586.7
760.4


i.p., Q3DX6
32360
126.5
216.9
236.9
276.4
554.6
714.2
647.2
1104.9



32361
139.0
258.8
238.8
254.1
289.8
283.8
344.1
485.1



32374
112.4
204.5
243.8
188.0
254.7
405.3
311.9
375.6



mean
140
257
282
261
372
481
463
656



SD
29
94
118
105
190
311
298
460



SEM
12
39
48
43
78
127
122
188



CV value
21.0%
36.7%
41.9%
40.1%
51.0%
64.8%
64.4%
70.1%



TGI

−0.390
0.294
0.567
0.631
0.659
0.708
0.639



P value
0.987
0.516
0.432
0.052
0.038
0.019
0.035
0.072


G5
32345
169.6
236.5
153.4
98.6
72.6
42.5
25.7
22.8


B1E7-IgG1
32351
140.5
161.6
207.2
95.3
96.3
84.3
25.7
0.0


10 mg/kg,
32352
172.9
376.7
289.9
272.4
161.7
123.9
54.8
51.1


i.p., Q3DX6
32365
130.8
188.3
210.4
137.8
58.6
42.7
0.0
0.0



32368
124.7
214.9
231.2
120.2
66.2
52.4
0.0
0.0



32371
102.8
230.3
252.8
203.1
140.0
76.3
14.7
0.0



mean
140
235
224
155
99
70
20
12



SD
27
75
46
70
42
31
20
21



SEM
11
31
19
28
17
13
8
9



CV value
19.3%
31.9%
20.6%
45.2%
42.8%
44.7%
101.8%
171.1%



TGI

−0.122
0.582
0.949
1.065
1.070
1.108
1.089



P value
0.992
0.816
0.069
0.002
0.001
0.000
0.002
0.004








Claims
  • 1. An anti-4-1BB antibody or antigen-binding fragment thereof, comprising a heavy chain and a light chain, wherein: the heavy chain comprises a heavy chain variable region comprising(I) HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 3, SEQ ID NO: 4 and SEQ ID NO: 5, respectively; or(II) HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 3, SEQ ID NO: 45 and SEQ ID NO: 5, respectively; or(III) HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 11, SEQ ID NO: 12 and SEQ ID NO: 13, respectively; or(IV) HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 39, SEQ ID NO: 40 and SEQ ID NO: 41, respectively; and/orthe light chain comprises a light chain variable region comprising:(I) LCDR1, LCDR2 and LCDR3 as set forth in SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8, respectively; or(II) LCDR1, LCDR2 and LCDR3 as set forth in SEQ ID NO: 14, SEQ ID NO: 15 and SEQ ID NO: 16, respectively; or(III) LCDR1, LCDR2 and LCDR3 as set forth in SEQ ID NO: 42, SEQ ID NO: 43 and SEQ ID NO: 44, respectively.
  • 2. The anti-4-1BB antibody or antigen-binding fragment thereof according to claim 1, which comprises any one selected from the group consisting of (I) to (IV): (I) the heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 3, SEQ ID NO: 4 and SEQ ID NO: 5, respectively; andthe light chain variable region comprising LCDR1, LCDR2 and LCDR3 as set forth in SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively;(II) the heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 3, SEQ ID NO: 45 and SEQ ID NO: 5; andthe light chain variable region comprising LCDR1, LCDR2 and LCDR3 as set forth in SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8, respectively;(III) the heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 11, SEQ ID NO: 12 and SEQ ID NO: 13, respectively; andthe light chain variable region comprising LCDR1, LCDR2 and LCDR3 as set forth in SEQ ID NO: 14, SEQ ID NO: 15 and SEQ ID NO: 16, respectively;(IV) the heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 as set forth in SEQ ID NO: 39, SEQ ID NO: 40 and SEQ ID NO: 41, respectively; andthe light chain variable region comprising LCDR1, LCDR2, and LCDR3 as set forth in SEQ ID NO: 42, SEQ ID NO: 43 and SEQ ID NO: 44, respectively.
  • 3. The anti-4-1BB antibody or antigen-binding fragment according to claim 1, wherein: the heavy chain variable region is as set forth in SEQ ID NO: 1 and the light chain variable region is as set forth in SEQ ID NO: 2; orthe heavy chain variable region is as set forth in SEQ ID NO: 9 and the light chain variable region is as set forth in SEQ ID NO: 10; orthe heavy chain variable region is as set forth in SEQ ID NO: 37 and the light chain variable region is as set forth in SEQ ID NO: 38.
  • 4. The anti-4-1BB antibody or antigen-binding fragment thereof according to claim 1, which is a murine antibody, a chimeric antibody, a human antibody, a humanized antibody or fragment thereof.
  • 5. (canceled)
  • 6. The anti-4-1BB antibody or antigen-binding fragment thereof according to claim 1, comprising: the heavy chain variable region, which is shown as the sequence selected from the group consisting of SEQ ID NOs: 25-27; and/orthe light chain variable region, which is shown as the sequence selected from the group consisting of SEQ ID NOs: 28-30.
  • 7. The anti-4-1BB antibody or antigen-binding fragment thereof according to claim 1, comprising: the heavy chain as set forth in SEQ ID NO: 17 or a variant sequence thereof, or SEQ ID NO: 19 or a variant sequence thereof; and/orthe light chain as set forth in SEQ ID NO: 18 or a variant sequence thereof, or SEQ ID NO: 20 or a variant sequence thereof;wherein each of the variant sequence of SEQ ID NO: 17, the variant sequence of SEQ ID NO: 19, the variant sequence of SEQ ID NO: 18, and/or the variant sequence of SEQ ID NO: 20 comprises 1 to 10 amino acid changes.
  • 8. The anti-4-1BB antibody or antigen-binding fragment thereof according to claim 4, wherein the heavy chain comprises a heavy chain constant region of human IgG1, IgG2, IgG3 or IgG4 or a variant thereof.
  • 9. (canceled)
  • 10. A bispecific antibody or multispecific antibody, comprising the light chain variable region and the heavy chain variable region as defined in claim 1.
  • 11. (canceled)
  • 12. (canceled)
  • 13. A polynucleotide encoding the anti-4-1BB antibody or antigen-binding fragment thereof according to claim 1.
  • 14. An expression vector comprising the polynucleotide of claim 13.
  • 15. A host cell transformed with the expression vector of claim 14.
  • 16. A method for preparing an anti-4-1BB antibody or antigen-binding fragment thereof, comprising the steps of: expressing the anti-4-1BB antibody or antigen-binding fragment thereof in the host cell of claim 15; andisolating the anti-4-1BB antibody or antigen-binding fragment thereof from the host cell.
  • 17. A pharmaceutical composition comprising: the antibody or antigen-binding fragment thereof according to claim 1, anda pharmaceutically acceptable excipient, diluent or carrier.
  • 18. (canceled)
  • 19. A method of treating or preventing cancer, comprising: administering to a subject in need thereof a therapeutically or prophylactically effective amount of the antibody or antigen-binding fragment thereof according to claim 1.
  • 20. The anti-4-1BB antibody or antigen-binding fragment thereof according to claim 1, wherein, the heavy chain variable region comprise the sequence selected from the group consisting of SEQ ID NOs: 31-33, and/orthe light chain variable region comprises the sequence selected from the group consisting of SEQ ID NOs: 34-36.
  • 21. The anti-4-1BB antibody or antigen-binding fragment thereof according to claim 1, wherein, the heavy chain variable region comprises the sequence of SEQ ID NO: 27, and/orthe light chain variable region comprises the sequence of SEQ ID NO: 30.
  • 22. The anti-4-1BB antibody or antigen-binding fragment thereof according to claim 1, wherein, the heavy chain variable region comprises the sequence of SEQ ID NO: 33, and/orthe light chain variable region comprises the sequence of SEQ ID NO: 36.
  • 23. The anti-4-1BB antibody or antigen-binding fragment thereof according to claim 7, wherein, the variant sequence of SEQ ID NO: 17 comprises any one of the mutations E53D, A75S and R112Q or any combination thereof; and/orthe variant sequence of SEQ ID NO: 18 comprises any one of the mutations P47A and V62I or a combination thereof; and/orthe variant sequence of SEQ ID NO: 19 comprises any one of the mutations M48I, Y95F and Q112T or any combination thereof; and/orthe variant sequence of SEQ ID NO: 20 comprises any one of the mutations M4L and V62I or a combination thereof.
  • 24. The anti-4-1BB antibody or antigen-binding fragment thereof according to claim 7, wherein, the variant sequence of SEQ ID NO: 17 is E53D.
  • 25. The method of claim 19, wherein, the cancer is selected from the group consisting of melanoma, breast cancer, ovarian cancer, prostate cancer, pancreatic cancer, kidney cancer, lung cancer, liver cancer, stomach cancer, colorectal cancer, bladder cancer, head and neck cancer, thyroid cancer, esophageal cancer, cervical cancer, sarcoma, multiple myeloma, leukemia, lymphoma, gallbladder cancer and glioblastoma.
Priority Claims (2)
Number Date Country Kind
201810058076.8 Jan 2018 CN national
201811472564.X Dec 2018 CN national
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2019/072484 1/21/2019 WO 00