WO2024012513A1 - Antibody, antigen-binding fragment thereof, and pharmaceutical use thereof - Google Patents

Antibody, antigen-binding fragment thereof, and pharmaceutical use thereof Download PDF

Info

Publication number
WO2024012513A1
WO2024012513A1 PCT/CN2023/107132 CN2023107132W WO2024012513A1 WO 2024012513 A1 WO2024012513 A1 WO 2024012513A1 CN 2023107132 W CN2023107132 W CN 2023107132W WO 2024012513 A1 WO2024012513 A1 WO 2024012513A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
variable region
chain variable
light chain
Prior art date
Application number
PCT/CN2023/107132
Other languages
French (fr)
Inventor
Min Ren
Yifan Zhang
Larry Lo
Original Assignee
Hansoh Bio Llc
Shanghai Hansoh Biomedical Co., Ltd.
Changzhou Hansoh Pharmaceutical Co., Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Hansoh Bio Llc, Shanghai Hansoh Biomedical Co., Ltd., Changzhou Hansoh Pharmaceutical Co., Ltd. filed Critical Hansoh Bio Llc
Publication of WO2024012513A1 publication Critical patent/WO2024012513A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants

Definitions

  • the present invention relates to novel antibodies or antigen-binding fragments that are specific for human CD3 expressed on T cell.
  • the body's immune system serves as a defense against infection, injury and cancer.
  • the humoral system is mediated by soluble factors, named antibodies, which neutralize products recognized as being foreign by the body.
  • the cellular system involves cells, such as T cells and macrophages, which remove and neutralize foreign invaders.
  • CD3 Cluster of differentiation 3
  • TCR T cell receptor complex
  • Antibodies against CD3 have been shown to cluster CD3 on T cells, thereby causing T cell activation in a manner similar to the engagement of the TCR by peptide-loaded MHC molecules.
  • Anti-CD3 antibodies have been proposed for therapies involving the activation of T cells.
  • Anti-CD3 antibodies have been used for the treatment of proliferative disorders such as cancer and for the treatment of autoimmune diseases.
  • OKT3 reacts with the chimpanzees CD3, but not with CD3 homologs of other primates such as rhesus monkeys.
  • the species specificity of CD3 monoclonal antibody is a significant obstacle to its development as an antibody drug for the treatment of human diseases. Any new drug candidate must undergo rigorous preclinical verification before it can be used in human patients for clinical trials.
  • the pre-clinical safety test is to administer the candidate drug to related species, preferably non-human primates.
  • chimpanzees are considered endangered species, and the use of such animals for drug safety testing is highly restricted.
  • the species described in the art that are suitable for safety evaluation testing may be rhesus monkeys, particularly cynomolgus monkeys.
  • CD3 antibodies that lack primate species-specific cross-reactivity are difficult to provide effective preclinical safety assessment data.
  • SP34 is one of the very few antibodies that can bind to a variety of primate CD3 (such as human and cynomolgus CD3) .
  • SP-34 recognizes an epitope present on solely the ⁇ chain of CD3 while the antibody UCHT1 recognize epitopes formed by ⁇ and ⁇ chains. In vivo, the ⁇ chain can also form a CD3 ⁇ / ⁇ heterodimer.
  • Several antibodies directed against human CD3 ⁇ / ⁇ have been developed in the past, but the success of these attempts so far has been limited.
  • novel CD3 binding molecules in particular novel anti-CD3 antibodies, which exhibit the desired affinity and potency profile, but which additionally are cross-reactive with other species, in particular with non-human primates such as cynomolgus monkeys, both in vitro and in a cellular context.
  • An object of the present invention is to provide a novel antibody or an antigen-binding fragment of the antibody (hereinafter, also referred to as an antibody, etc. ) which binds to human CD3 and to cynomolgus monkey CD3, a molecule comprising the antibody, and a pharmaceutical composition having cytotoxic activity, etc. which includes the antibody, etc. or the molecule as an active ingredient.
  • an antibody also referred to as an antibody, etc.
  • the present inventors have conducted extensive research to achieve this object and have realized the present invention by developing a novel anti-CD3 antibody and a molecule comprising this antibody.
  • the present invention encompasses the following aspects:
  • the present disclosure provides an anti-CD3 antibody or an antigen-binding fragment thereof, comprising one or more the CDR region sequences selected from the following or a mutant sequence thereof:
  • An antibody heavy chain variable region comprising one or more HCDR as shown in the sequences selected from SEQ ID NO: 01, SEQ ID NO: 02, SEQ ID NO: 03, SEQ ID NO: 04, SEQ ID NO: 05 and SEQ ID NO: 06; and an antibody light chain variable region comprising one or more LCDR as shown in the sequences selected from SEQ ID NO: 07, SEQ ID NO: 08, SEQ ID NO: 09 (X 1 QX 2 TX 3 FPYT) , SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12; wherein X1 is M, Q, T or A, X2 is L or A, X3 is H or Q.
  • the antibody heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 regions and the antibody light chain variable region comprising LCDR1, LCDR2 and LCDR3 regions, wherein: a) HCDR1 as shown in SEQ ID NO: 01 or SEQ ID NO: 04; b) HCDR2 as shown in SEQ ID NO: 02 or SEQ ID NO: 05; c) HCDR3 as shown in SEQ ID NO: 03 or SEQ ID NO: 06; d) LCDR1 as shown in SEQ ID NO: 07 or SEQ ID NO: 10; e) LCDR2 as shown in SEQ ID NO: 08 or SEQ ID NO: 11; f) LCDR3 as shown in SEQ ID NO: 09 or SEQ ID NO: 12.
  • an anti-CD3 antibody or antigen-binding fragment comprises HCDR1 as shown in SEQ ID NO: 01, HCDR2 as shown in SEQ ID NO: 02, and HCDR3 as shown in SEQ ID NO: 03, respectively; or HCDR1 as shown in SEQ ID NO: 04, HCDR2 as shown in SEQ ID NO: 05, and HCDR3 as shown in SEQ ID NO: 06 respectively.
  • an anti-CD3 antibody or antigen-binding fragment comprises LCDR1 as shown in SEQ ID NO: 07, LCDR2 as shown in SEQ ID NO: 08, and LCDR3 as shown in SEQ ID NO: 09, respectively; or LCDR1 as shown in SEQ ID NO: 10, LCDR2 as shown in SEQ ID NO: 11, and LCDR3 as shown in SEQ ID NO: 12, respectively.
  • the anti-CD3 antibody or antigen-binding fragment thereof comprises: a) a heavy chain variable region sequence comprises HCDR1, HCDR2 and HCDR3 as shown in SEQ ID NO: 01, SEQ ID NO: 02 and SEQ ID NO: 03, respectively; and a light chain variable region sequence comprises LCDR1, LCDR2 and LCDR3 as shown in SEQ ID NO: 07, SEQ ID NO: 08 and SEQ ID NO: 09, respectively; or b) a heavy chain variable region sequence comprises HCDR1, HCDR2 and HCDR3 as shown in SEQ ID NO: 04, SEQ ID NO: 05 and SEQ ID NO: 06, respectively; and a light chain variable region sequence comprises LCDR1, LCDR2 and LCDR3 as shown in SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12, respectively.
  • the anti-CD3 antibody or antigen-binding fragment is selected from murine antibody, chimeric antibody, humanized antibody, human antibody or the antigen-binding fragment thereof.
  • the anti-CD3 antibody or antigen-binding fragment comprises: a) the heavy chain variable region as shown in SEQ ID NO: 13, or having at least 80%, 85%, 90%, 95% or 99%sequence identity therewith; and/or the light chain variable region as shown in SEQ ID NO: 18, or having at least 80%, 85%, 90%, 95%or 99%sequence identity therewith; or b) the heavy chain variable region as shown in SEQ ID NO: 17, or having at least 80%, 85%, 90%, 95%or 99%sequence identity therewith; and/or the light chain variable region as shown in SEQ ID NO: 31, or having at least 80%, 85%, 90%, 95%or 99%sequence identity therewith.
  • the anti-CD3 antibody or antigen-binding fragment comprises: a) the heavy chain variable region having the sequence selected from any one of SEQ ID NO: 13, 14, 15 and 16; and/or the light chain variable region having the sequence selected from any one of SEQ ID NO: 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 and 30; or b) the heavy chain variable region as shown in SEQ ID NO: 17; and/or the light chain variable region as shown in SEQ ID NO: 31.
  • the anti-CD3 antibody or antigen-binding fragment comprises: a) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 18; or b) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 31; or c) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 19; or d) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 20; or e) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 21; or f) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 22; or g) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 23; or h) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ
  • the anti-CD3 antibody or antigen-binding fragment comprises: a) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 38; or b) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 51; or c) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 39; or d) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 40; or e) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 41; or f) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 42; or g) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 43; or h) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 44; or i) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 45; or
  • the anti-CD3 antibody is a full-length antibody, further comprising human antibody constant regions; preferably, the heavy chain constant region of the human antibody constant regions is selected from constant regions of human IgG1, IgG2, IgG3 and IgG4 and conventional variants thereof, and the light chain constant region of the human antibody constant regions is selected from ⁇ and ⁇ chain constant regions of human antibody and conventional variants thereof; more preferably the full-length antibody comprises a human antibody heavy chain constant region of SEQ ID NO: 32 and a human light chain constant region of SEQ ID NO: 33.
  • the CD3 antigen-binding fragment is selected from the group consisting of Fab, Fab', F (ab') 2, variable fragment (Fv) , single chain variable fragment (scFv) , dimerized domain V (diabody) , disulfide stabilized Fv (dsFv) , and any CDR-containing peptides.
  • the anti-CD3 antibody or antigen-binding fragment binds to human CD3 with an affinity of a K D value of 1 ⁇ 10 -5 M to 1 ⁇ 10 -12 M.
  • the present disclosure provides an isolated antibody or the antigen-binding fragment thereof that competing with the above-mentioned antibody or the antigen-binding fragment to bind to human CD3.
  • the above-mentioned antibody or the antigen-binding fragment thereof having at least one of the following characteristics: a) binding to human CD3 with an affinity of a K D value of 1 ⁇ 10 -7 M to 1 ⁇ 10 -12 M; b) cross-reacting with CD3 of cynomolgus monkey or rhesus monkey; c) increased activation of T cells.
  • the present disclosure provides an isolated nucleic acid molecule encoding any antibody or the antigen-binding fragment.
  • the present disclosure also provides a recombinant vector comprising the above-mentioned isolated nucleic acid molecule.
  • the present disclosure also provides a host cell transformed with the above-mentioned recombinant vector, wherein the host cell is selected from the group consisting of a prokaryotic cell and a eukaryotic cell, preferably a eukaryotic cell, more preferably a mammalian cell.
  • the present disclosure also provides a method for producing the above-mentioned antibody or the antigen-binding fragment in a medium to produce and accumulate the antibody or the antigen-binding fragment thereof, and harvesting the antibody or the antigen-binding fragment thereof from the culture.
  • the present disclosure also provides a method for immunologically detecting or measuring CD3, wherein the method comprises detecting the CD3 by contacting with the above-mentioned antibody or the antigen-binding fragment.
  • the present disclosure also provides a method for diagnosing a disease related to a human CD3 positive cell, wherein the method comprises a detecting or measuring the CD3 or CD3 positive cell by contacting with the above-mentioned antibody or the antigen-binding fragment.
  • the present disclosure provides a pharmaceutical composition, which comprises a therapeutically effective amount of the above-mentioned antibody or the antigen-binding fragment, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the present disclosure also provides a method of treating a CD3-mediated disease or disorder in a subject in need thereof, comprising administering to the subject the above-mentioned antibody or the antigen-binding fragment, or the above-mentioned pharmaceutical composition, wherein the disease or disorder is selected from the group consisting of cancer, autoimmune and inflammatory diseases
  • the invention obtained fully human antibody variable region using transgenic mice with human antibody variable genes, and the obtained antibodies have a series of desired characteristics:
  • variable region sequences are different from the existing antibody
  • the obtained antibodies have capacity of binding to human CD3
  • the invention has obtained antibodies with different sequences, which can specifically bind to CD3, including to cross-reactive with other species, in particular with non-human primates such as cynomolgus monkeys.
  • FIG. 1 Mice immunized using RIMMS protocol were tail-bleeded, and serum titers were measured against Jurkat E6.1 cells by flow cytometry (BD Fortessa X20) at 1/50 dilution. The cell binding is shown as histogram of normalized count across different fluorescence intensity. UCHT1 was used as positive control. Serums from unimmunized mice were used as negative control.
  • Figure 2 Representative binding histograms from flow cytometry analysis of 13A1 hybridoma clone.
  • the solid line represents the binding to Jurkat E6.1 cell and the dash line represents the binding to J. RT-T3.5. cell.
  • F2B was used as positive control for CD3 binding and the secondary antibody-only was used as negative control.
  • FIG. 4 Graphs showing the T cell activation activity of selected hybridoma clone supernatants using NFAT reporter assay.
  • F2B and OKT3 are anti-CD3 mAbs that were used as positive controls.
  • FIG. 8 Graphs showing the T cell activation activities of indicated recombinant antibodies using NFAT reporter assay. 13A1 VH1-VL1 and 13A1 VH1-VL2 activities were shown in panel A. Activities of 13A1 VH1-VL1 and its light chain mutants or heavy chain mutants were shown in (B) , (C) and (D) .
  • Figure 9 Human PBMC activation by the anti-CD3 antibodies as measured by cell surface CD69 amount measured by flow cytometry analysis.
  • FIG. 10 T cell activation upon binding of 13A1 VH1-VL1 and its mutation, as measured by up-regulation of the activation marker CD25 on CD4 (A) or CD8 T cells (C) , respectively the activation marker CD69 on either CD4 (B) or CD8 T cells (D) .
  • CD3 and CD3 antigen are used interchangeably herein, and include any variants, isoforms and species homologs of human CD3 which are naturally expressed by cells or are expressed on cells transfected with the CD3 gene.
  • an “antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR) , interspersed with regions that are more conserved, termed framework regions (FR) .
  • CDR complementarity determining regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
  • antigen-binding fragment of an antibody, as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., CD3) . It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term "antigen-binding fragment" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F (ab') 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341: 544-546) , which consists of a VH domain; (vi) an isolated complementarity determining region (CDR) , and (vii) a combination of two or more isolated CDRs which may optionally be joined by a synthetic linker.
  • a Fab fragment a monovalent fragment consisting of the VL, VH, CL and
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv) ; see e.g., Bird et al. (1988) Science 242 : 423-426 ; and Huston et al. (1988) Proc. Natl. Acad. Sci. 85 : 5879-5883) .
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody.
  • human antibody is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • the human antibodies of the invention may include amino acid residues 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) .
  • the term “human antibody” is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • recombinant human antibody includes all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom (described further in Section I, below) , (b) antibodies isolated from a host cell transformed to express the antibody, e.g., from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial human antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of human immunoglobulin gene sequences to other DNA sequences.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences.
  • such recombinant human antibodies can be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • CDR refers to one of the six hypervariable regions within the variable domain of an antibody that primarily contributes to antigen binding.
  • One of the most commonly used definitions for the six CDRs is provided by Kabat E. A. et al. (1991) Sequences of proteins of immunological interest. NIH Publication 91-3242.
  • the Kabat definition of CDR only applies to CDR1, CDR2 and CDR3 of the light chain variable domain (LCDR1, LCDR2, LCDR3 or L1, L2, L3) , as well as CDR1, CDR2 and CDR3 of heavy chain variable domain (HCDR1, HCDR2, HCDR3 or H1, H2, H3) .
  • Methods and techniques for identifying CDRs within HCVR and LCVR amino acid sequences are well known in the art and can be used to identify CDRs within the specified HCVR and/or LCVR amino acid sequences disclosed herein.
  • Exemplary conventions that can be used to identify the boundaries of CDRs including, e.g., Chothia based on the three-dimensional structure of antibodies and the topology of the CDR loops (Chothia et al.
  • Note 1 some of these definitions (particularly for Chothia loops) vary depending on the individual publication examined; Note2: any of the numbering schemes can be used for these CDR defintions, except the contact definition uses the Chothia or Martin (enhanced Chothia) definition; Note 3: the end of the Chothia HCDR1 loop when numbered using the Kabat numbering convention varies between H32 and H34 depending on the length of the loop. This is because the Kabat numbering scheme places the insertions at H35A and H35B.
  • Constant modification or “conservative replacement or substitution” refers to substitutions of amino acids in a protein with other amino acid having similar characteristics (e.g., charge, side chain size, hydrophobicity/hydrophilicity, backbone conformation and rigidity, etc. ) , such that the changes can be frequently made without altering the biological activity of the protein. It will be appreciated by those skilled in the art that, in general, a single amino acid substitution in a non-essential region of polypeptide does not substantially alter biological activity (see, for example, Watson et al. (1987) Molecular Biology of the Gene, The Benjamin/Cummings Pub. Co., Page 224, (4th edition) ) . In addition, substitutions with structurally or functionally similar amino acids are unlikely to disrupt biological activity.
  • nucleic acid molecule refers to a DNA molecule and a RNA molecule.
  • the nucleic acid molecule may be single stranded or double stranded but is preferably a double stranded DNA.
  • a nucleic acid is “effectively linked” when it is placed into functional relationship with another nucleic acid sequence. For example, if a promoter or enhancer affects transcription of a coding sequence, the promoter or enhancer is effectively linked to the coding sequence.
  • the preparation method of the nucleic acid is a conventional preparation method in the art. Preferably, it comprises the following steps: obtaining the nucleic acid molecule encoding the above-mentioned protein by gene cloning technology, or obtaining the nucleic acid molecule encoding the above-mentioned protein by the method of artificial full-length sequence synthesis.
  • the base sequence encoding the amino acid sequence of the protein can be replaced, deleted, changed, inserted or added appropriately to provide a polynucleotide homolog.
  • the homolog of the polynucleotide of the present invention can be prepared by replacing, deleting or adding one or more bases of the gene encoding the protein sequence within the scope of maintaining the activity of the antibody.
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to that it has been linked.
  • the vector is a “plasmid” that refers to a circular double stranded DNA loop into which additional DNA segment can be ligated.
  • the vector is a viral vector, wherein an additional DNA segment can be ligated into viral genome.
  • the vectors disclosed herein are capable of self-replicating in a host cell into which they have been introduced (for example, a bacterial vector having a bacterial replication origin and a episomal mammalian vector) or can be integrated into the genome of a host cell upon introduction into host cell, thereby is replicated along with the host genome (e.g., a non-episomal mammalian vector) .
  • the recombinant expression vector can be obtained by conventional methods in the art, that is, by connecting the nucleic acid molecule of the present invention to various expression vectors, thus being constructed.
  • the expression vector is one of a variety of conventional vectors in the art, as long as it can carry the above-mentioned nucleic acid molecule.
  • the vector preferably includes: various plasmids, cosmids, phage or virus vectors and the like.
  • transfectoma includes recombinant eukaryotic host cell expressing the antibody, such as CHO cells, NS/0 cells, HEK293 cells, plant cells, or fungi, including yeast cells.
  • sequence of the DNA molecule for the antibody or a fragment thereof according to the present invention can be obtained by conventional techniques, for example, methods such as PCR amplification or genomic library screening.
  • sequences encoding light chain and heavy chain can be fused together, to form a single-chain antibody.
  • the relevant sequence can be obtained in bulk using a recombination method. This is usually carried out by cloning the sequence into a vector, transforming a cell with the vector, and then separating the relevant sequence from the proliferated host cell by conventional methods.
  • a relevant sequence can be synthesized artificially, especially when the fragment is short in length.
  • several small fragments are synthesized first, and then are linked together to obtain a fragment with a long sequence.
  • DNA sequence encoding the antibody of the present invention (or fragments thereof, or derivatives thereof) completely by chemical synthesis.
  • the DNA sequence can then be introduced into a variety of existing DNA molecules (or, for example, vectors) and cells known in the art.
  • mutations can also be introduced into the protein sequences of the present invention by chemical synthesis.
  • the host cell obtained is cultured.
  • the antibody of the present invention is purified by using conventional immunoglobulin purification steps, for example, the conventional separation and purification means well known to those skilled in the art, such as protein A-Sepharose, hydroxyapatite chromatography, gel electrophoresis, dialysis, ion exchange chromatography, hydrophobic chromatography, molecular sieve chromatography or affinity chromatography.
  • the monoclonal antibody obtained can be identified by conventional means.
  • the binding specificity of a monoclonal antibody can be determined by immunoprecipitation or an in vitro binding assay (such as radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELISA) ) .
  • the binding affinity of a monoclonal antibody can be determined by, for example, the Scatchard analysis (Munson et al., Anal. Biochem., 107: 220 (1980) ) .
  • the antibody according to the present invention can be expressed in a cell or on the cell membrane, or is secreted extracellularly. If necessary, the recombinant protein can be separated and purified by various separation methods according to its physical, chemical, and other properties. These methods are well known to those skilled in the art. The examples of these methods comprise, but are not limited to, conventional renaturation treatment, treatment by protein precipitant (such as salt precipitation) , centrifugation, cell lysis by osmosis, ultrasonic treatment, supercentrifugation, molecular sieve chromatography (gel chromatography) , adsorption chromatography, ion exchange chromatography, high performance liquid chromatography (HPLC) , and any other liquid chromatography, and the combination thereof.
  • protein precipitant such as salt precipitation
  • centrifugation such as salt precipitation
  • cell lysis by osmosis cell lysis by osmosis
  • ultrasonic treatment supercentrifugation
  • molecular sieve chromatography gel
  • k d (sec -1 )
  • k off value the term “k d” (sec -1 ) , as used herein, is intended to refer to the dissociation rate constant of a particular antibody-antigen interaction. Said value is also referred to as the k off value.
  • k a (M -1 x sec -1 ) , as used herein, is intended to refer to the association rate constant of a particular antibody-antigen interaction.
  • K D (M) , as used herein, is intended to refer to the dissociation equilibrium constant of a particular antibody-antigen interaction.
  • binding refers to the binding of an antibody to an epitope on a predetermined antigen.
  • “Pharmaceutical composition" is intended to refer to a mixture containing one or more of the compounds or a physiological/pharmaceutically acceptable salt or prodrug thereof described herein with other chemical components, such as physiological/pharmaceutically acceptable carriers and excipients.
  • the purpose of the pharmaceutical composition is to promote the administration to the organism, which is beneficial to the absorption of the active ingredient and exerts the biological activity.
  • administering when applied to an animal, human, experimental subject, cell, tissue, organ, or biological fluid, refer to contact with an exogenous pharmaceutical, therapeutic, diagnostic reagent, or composition with the animal, human, subject, cell, tissue, organ, or biological fluid.
  • administering can refer, e.g., to therapeutic, pharmacokinetic, diagnostic, research, and experimental methods. Treatment of a cell encompasses contacting the cell with a reagent, as well as contacting a fluid with a reagent, wherein the fluid is in contact with the cell.
  • administering 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 when applied to a human, veterinary, or research subject, refers to therapeutic treatment, prophylactic or preventative measures, research and diagnostic applications.
  • the present disclosure includes a medicament for treating a disease associated with CD3, comprising an antibody or an antigen-binding fragment thereof of the present disclosure as an active ingredient.
  • the molecules of the present disclosure are very useful for those who suffer a tumor, cancer or infectious disease when in preparations and formulations suitable for therapeutic applications.
  • the present disclosure relates to a method for immunologically detecting or measuring CD3, a reagent for immunologically detecting or measuring CD3, a method for immunologically detecting or measuring cells expressing CD3, and a diagnostic reagent for diagnosis of disease related to CD3 positive cells, comprising the antibody or antigen-binding fragment of the present disclosure that specifically recognizes human CD3, as an active ingredient.
  • the method for detecting or determining the amount of CD3 may be any known method.
  • it includes immunodetection or assay.
  • the immunodetection or assay is a method of detecting or determining the amount of antibody or antigen by using labeled antigen or antibody.
  • immunodetection or assay include a radioactive substance labeled immunological antibody method (RIA) , an enzyme immunoassay (EIA or ELISA) , a fluorescent immunoassay (FIA) , a luminescent immunoassay, a western blotting method, physicochemical methods, etc.
  • the above-mentioned diseases related to CD3 positive cells can be diagnosed by detecting or measuring cells expressing CD3 by using the antibodies or antibody fragments thereof of the present invention.
  • a known immunodetection can be used, and preferably immunoprecipitation, fluorescent cell staining or immunohistochemical staining etc. can be used. Furthermore, a fluorescent antibody staining method etc. using FMAT8100HTS system (Applied Biosystem) can be used.
  • the room temperature described in the examples is a conventional room temperature in the art, and is generally 10-30°C.
  • CD3 antigen A combination of Jurkat E6.1, DNA and recombinant protein antigens were used to immunize mice.
  • the recombinant ectodomain of human CD3E and CD3D were expressed and purified as heterodimer and were commercially available at AcroBiosystems (catalog numbers CDD-H52Wa, CDD-H52W1, CDD-C52W4, CDD-C52W9) .
  • Either Fc, His Tag&Fc, Flag Tag or His Tag&Tag Free were used.
  • Anti-CD3 antibodies were obtained by immunizing genetically modified mouse encoding human immunoglobulin heavy and kappa light chain variable regions with a combination of human CD3 antigen, cynomolgus CD3 antigen, human CD3 DNA, and/or Jurkat E6.1 cells (Sigma-Aldrich, Catalog No. 88042803) which naturally express human CD3. The antibody immune response was monitored by a CD3-specific immunoassay.
  • splenocytes were harvested from each mouse and either (1) fused with mouse myeloma cells to preserve their viability and form hybridoma cells and screened for CD3 specificity, or (2) B-cell sorted using a human CD3 with a C-terminal 6-His tag as the sorting reagent that binds and identifies reactive antibodies (antigen-positive B cells) .
  • Figure 1 shows the results of the antibody titer in serum detected by Flow cytometry after immunization with CD3 protein.
  • mice after 3 times of immunization with the immunogens, the selected mice had an antigen-selective cell binding titer of more than 1: 50, indicating that mice had a better humoral immune response to the immunogen, and their spleen cells can be used for Hybridoma cell preparation.
  • the spleen lymphocytes and myeloma cells Sp2/0 were fused to obtain hybridoma cells by electrofusion or PEG fusion.
  • PEG fusion was performed using Clonacell TM HY technology (STEMCELL technologies) , following manufacturer’s instructions.
  • the primary cell: mouse myeloma cell line ratio was 1: 1 for electrofusion, and 10: 1 for PEG fusion.
  • ELISA was performed using the DuoSet ELISA Ancillary Kit (R&D System, DY008) . ELISA plates were coated with 5ug/ml of human CD3DE (AcroBio CDD-H52W1) and cyno CD3DE (AcroBio, CDD-C52W4) overnight. Excess unbound proteins were washed off by washing the plates three times with the wash buffer before blocking for 1 hour at room temperature. 50ul CD3 hybridoma supernatant was added in duplicate wells and incubated for 1 hour.
  • Hybridoma supernatants was subjected to binding tests on human T cell lines (CD3 + Jurkat cell line clone E6.1 and CD3 - clone J. RT-T3.5 (ATCC) using flow cytometry analysis.
  • the primary staining was performed using undiluted hybridoma supernatants, followed by fluorescence labeled secondary antibody (PE Goat anti-mouse IgG, Biolegend Cat#405307) .
  • Figure 2 shows examples of selected flow cytometry histograms. The clone 13A1 was identified with enhanced binding profile to Jurkat E6.1 compared to J. RT-T3.5 cells.
  • T cell activation activity of hybridoma supernatants or recombinant anti-CD3 antibodies were measured using NFAT reporter assay, using Jurkat-Lucia TM NFAT cells (InvivoGen jktl-nfat) .
  • U-bottom 96-well plates were dry coated with capturing antibody (goat anti-mouse IgG Fc ⁇ fragment specific, Jackson ImmunoResearch 115-005-071) , at 4 ⁇ g/ml in PBS and 50 ⁇ l/well.
  • Hybridoma supernatants or recombinant anti-CD3 antibodies diluted in culture medium were added to the pre-coated plate at 50 ⁇ l/well for overnight.
  • the Jurkat-Lucia TM NFAT cells were added to each well at 400,000 cells per well in 200ul culture medium. The cells were maintained at 37°C with 5%CO 2 for 24 hours, then the culture supernatants were collected. Luciferase activity in culture supernatant was measured using QUANTI-Luc assay solution (InvivoGen) according to manufacturer’s instruction. Luminescence signal was recorded by VICTOR Nivo Multimode Microplate Reader (PerkinElmer) . Purified anti-CD3 antibodies F2B and OKT3 were used as positive controls. Supernatant from another hybridoma clone was used as a negative control. Clone 13A1 was selected for their ability to active human T cells indicated by the in vitro reporter assay as shown in Figure 4.
  • the process of cloning sequences from positive hybridomas are as follows.
  • the logarithmic growth phase hybridoma cells were collected, RNA was extracted, and reverse transcription was performed then followed by VDJ region amplification.
  • Amplified cDNA library from each clones were subjected to next-generation sequencing.
  • the amino acid sequences of the heavy and light chain variable region DNA sequences corresponding to the antibodies 13A1 were obtained (the amino acid residues of the CDRs in VH/VL are underlined according to Kabat & Wu annotation:
  • VH and VL regions of selected clones were directly synthesized as DNA fragments with 5’-end in-frame leader sequence (MGWSCIILFLVATATGVHS) . These DNA fragments were cloned into selected vectors using NEBuilder DNA Assembly Cloning Kit (New England Biolabs) .
  • VH region was cloned into pFUSE-CHIg_hG1 vector (InvivoGen #pfuse-hchg1) , which in-frame with constant region of hIgG1 heavy chain in the vector.
  • VL region was cloned into pFUSE2-CLIg_hk vector (InvivoGen, #pfuse2-hclk) , which in-frame with constant region of hIg kappa light chain in the vector.
  • the heavy chain expression plasmid and light chain plasmids were co-transfected into Expi293F cells (ThermoFisher, #A14527) using ExpiFectamine 293 Transfection Kit (ThermoFisher, A14524) , or into ExpiCHO-Scells (ThermoFisher #A29127) using ExpiFectamine CHO Transfection Kit (ThermoFisher, A29129) . Based on the manufacturer’s instructions, plasmid DNA concentration reached 1.0 ug per ml of suspended cells, with LC: HC vector ratio 1: 1. The transfected cells were cultured 5 to 7 days on an orbital shaker at 37 C, 8%CO 2 .
  • the purpose of the mutation is to obtain a group of anti-CD3 antibodies with varied affinities and potentially lower immunogenicity by converting certain residues to the germline sequence.
  • I14T indicates a mutation from I to T at position 14 according to Kabat numbering system.
  • WT indicates that the sequence does not contain amino acid mutations.
  • 13A1_mut01 indicates that two kinds of mutation (13A1_VL1. A and 13A1_VH1) are present on the 13A1_mut01, and so on.
  • TEST EXAMPLE 1 Enzyme-linked immunosorbent assay (ELISA) detection of the binding of antibodies to hCD3 ⁇ and cynoCD3 ⁇
  • ELISA plates were coated with 5ug/ml human or cyno CD3 ⁇ -His (AcroBiosystems) overnight. Serial dilutions of recombinant antiCD3 antibodies 13A1 wereperformed, starting at 100nM and added to CD3 coated plates. The excess non-bound antibodies were washed away before addition of the mouse anti-human IgG Fc-HRP (Abcam 99774) . The results were measured by O. D. at 450nm on a microplate reader. Data were analyzed with GraphPad Prism 9 ( Figure 5) . EC50 values were calculated for the binding of the CD3 antibody to human and cyno CD3 ⁇ protein (Table 5) .
  • the binding curves to cell surface CD3 were determined for 13A1_VH1_VL1, 13A1_VH1_VL2 ( Figure 6) recombinant antibodies by flow cytometry analysis.
  • Jurkat E6.1 cells were aliquoted into 0.3 million/well and 60ul of serially diluted antibodies were added to each well. The staining was performed for 1 hour at 4 degrees in the dark.
  • the PE goat anti human IgG Fc Jackson ImmunoResearch was used as the secondary antibody at 1: 200 dilution.
  • 13A1_VH1_VL1 binding to primary T cell was determined by using the freshly isolated PBMCs from one healthy donor. 0.3 million PBMCs were aliquoted to each well and stained with anti-human TCR ⁇ / ⁇ -FITC (Biolegend Cat 306706) . 13A1 antibody was serially diluted and 60ul was added to each well as primary staining. The PE goat anti human IgG was used as secondary antibody as Figure 7A.
  • TEST EXAMPLE 3 Detection of the effect of CD3 antibody on T cell activation by NFAT reporter assay or human PBMCs activation.
  • 96 well U-bottom plates were dry coated with 10 ug/ml capturing antibody (Jackson ImmunoResearch’s AffiniPure goat anti-human IgG, Fc ⁇ specific, #109-005-170) , and then after PBS wash, were wet coated with 10 ug/ml anti-CD3 antibodies.
  • NFAT reporter cells InvivoGen #jktl-nfat
  • RPMI 1640 RPMI 1640
  • Reporter activity was measured using QUANTI-Luc assay (InvivoGen) as manufacture’s instruction.
  • PBMCs from single healthy donor were plated in RPMI1640 with GlutaMax 10%FBS medium at a density of 1x10 6 cells/ml and incubated overnight at 37°C.
  • Anti-CD3 antibodies OKT3, 13A1 and F2B, as well as control human IgG were serially diluted.
  • PBMCs were then added to the antibody solutions at 50,000 cells per well, and final volume of 200 ⁇ l per well. Cells were incubated at 37°C 5%CO2 for 2 days, then stained with anti-human CD69 antibody (Biolegend Catalog No. 310938) and analyzed by BD Fortessa X20. Data were ananlyzed by FlowJo and GraphPad Prism 9 software.
  • PBMCs activation by anti-CD3 antibodies indicated as the ⁇ MFI of CD69 over control were shown in Figure 9, EC 50 values were shown in Table 10.
  • CD4 + CD25 + represents the percentage of CD25 positive CD4 cells in a given group
  • T cell activation was determined as percent of CD4 T cells expressing CD25 (FIG. 10A) or CD69 (FIG. 10B) , respectively CD8 T cells expressing CD25 (FIG. 10C) and CD69 (FIG. 10D) .

Abstract

Provided are antibodies and antigen-binding fragments thereof, and in particular to such antibodies and antigen-binding fragments that have affinity and activity of stimulating T cell activation. Uses of such antibodies and antigen-binding fragments in the treatment of cancer, immune and infectious diseases and other conditions are presented.

Description

ANTIBODY, ANTIGEN-BINDING FRAGMENT THEREOF, AND PHARMACEUTICAL USE THEREOF FIELD OF THE INVENTION
The present invention relates to novel antibodies or antigen-binding fragments that are specific for human CD3 expressed on T cell.
BACKGROUND
The body's immune system serves as a defense against infection, injury and cancer. Two separate but interrelated systems, humoral and cellular immune systems, work together to protect the body. The humoral system is mediated by soluble factors, named antibodies, which neutralize products recognized as being foreign by the body. In contrast, the cellular system involves cells, such as T cells and macrophages, which remove and neutralize foreign invaders.
Cluster of differentiation 3 (CD3) is a homodimeric or heterodimeric antigen expressed on T cells in association with the T cell receptor complex (TCR) and is required for T cell activation. Antibodies against CD3 have been shown to cluster CD3 on T cells, thereby causing T cell activation in a manner similar to the engagement of the TCR by peptide-loaded MHC molecules. Anti-CD3 antibodies have been proposed for therapies involving the activation of T cells. Anti-CD3 antibodies have been used for the treatment of proliferative disorders such as cancer and for the treatment of autoimmune diseases.
Several antibodies that bind to human CD3 epsilon are known in the art, the antibody OKT3, the antibody UCHT1, the antibody SP34, the antibody F2B (WO 2017/223111 A1) , or the antibody 20G6 (WP2016/116626 A1) . OKT3 reacts with the chimpanzees CD3, but not with CD3 homologs of other primates such as rhesus monkeys. The species specificity of CD3 monoclonal antibody is a significant obstacle to its development as an antibody drug for the treatment of human diseases. Any new drug candidate must undergo rigorous preclinical verification before it can be used in human patients for clinical trials. The pre-clinical safety test is to administer the candidate drug to related species, preferably non-human primates. But chimpanzees are considered endangered species, and the use of such animals for drug safety testing is highly restricted. The species described in the art that are suitable for safety evaluation testing may be rhesus monkeys, particularly cynomolgus monkeys. However, CD3 antibodies that lack primate species-specific cross-reactivity are difficult to provide effective preclinical safety assessment data. Among the known antibodies that bind to human CD3, SP34 is one of the very few antibodies that can bind to a variety of primate CD3 (such as human and cynomolgus CD3) . However, SP-34 recognizes an epitope present on solely the ε chain of CD3 while the antibody UCHT1 recognize epitopes formed by ε and γ chains. In vivo, the ε chain can also form a CD3 δ/ε heterodimer. Several antibodies directed against human CD3δ/ε have been developed in the past, but the success of these attempts so far has been limited.
Thus, there remained still a large unmet need to develop novel CD3 binding molecules, in particular novel anti-CD3 antibodies, which exhibit the desired affinity and potency profile, but which additionally are cross-reactive with other species, in particular with non-human primates such as cynomolgus monkeys, both in vitro and in a cellular context.
BRIEF SUMMARY OF THE INVENTION
An object of the present invention is to provide a novel antibody or an antigen-binding  fragment of the antibody (hereinafter, also referred to as an antibody, etc. ) which binds to human CD3 and to cynomolgus monkey CD3, a molecule comprising the antibody, and a pharmaceutical composition having cytotoxic activity, etc. which includes the antibody, etc. or the molecule as an active ingredient.
The present inventors have conducted extensive research to achieve this object and have realized the present invention by developing a novel anti-CD3 antibody and a molecule comprising this antibody.
Specifically, the present invention encompasses the following aspects:
The present disclosure provides an anti-CD3 antibody or an antigen-binding fragment thereof, comprising one or more the CDR region sequences selected from the following or a mutant sequence thereof:
An antibody heavy chain variable region comprising one or more HCDR as shown in the sequences selected from SEQ ID NO: 01, SEQ ID NO: 02, SEQ ID NO: 03, SEQ ID NO: 04, SEQ ID NO: 05 and SEQ ID NO: 06; and an antibody light chain variable region comprising one or more LCDR as shown in the sequences selected from SEQ ID NO: 07, SEQ ID NO: 08, SEQ ID NO: 09 (X1QX2TX3FPYT) , SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12; wherein X1 is M, Q, T or A, X2 is L or A, X3 is H or Q.
In some embodiments, the antibody heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 regions and the antibody light chain variable region comprising LCDR1, LCDR2 and LCDR3 regions, wherein: a) HCDR1 as shown in SEQ ID NO: 01 or SEQ ID NO: 04; b) HCDR2 as shown in SEQ ID NO: 02 or SEQ ID NO: 05; c) HCDR3 as shown in SEQ ID NO: 03 or SEQ ID NO: 06; d) LCDR1 as shown in SEQ ID NO: 07 or SEQ ID NO: 10; e) LCDR2 as shown in SEQ ID NO: 08 or SEQ ID NO: 11; f) LCDR3 as shown in SEQ ID NO: 09 or SEQ ID NO: 12.
In some embodiments, the present disclosure provides an anti-CD3 antibody or antigen-binding fragment comprises HCDR1 as shown in SEQ ID NO: 01, HCDR2 as shown in SEQ ID NO: 02, and HCDR3 as shown in SEQ ID NO: 03, respectively; or HCDR1 as shown in SEQ ID NO: 04, HCDR2 as shown in SEQ ID NO: 05, and HCDR3 as shown in SEQ ID NO: 06 respectively.
In some embodiments, the present disclosure provides an anti-CD3 antibody or antigen-binding fragment comprises LCDR1 as shown in SEQ ID NO: 07, LCDR2 as shown in SEQ ID NO: 08, and LCDR3 as shown in SEQ ID NO: 09, respectively; or LCDR1 as shown in SEQ ID NO: 10, LCDR2 as shown in SEQ ID NO: 11, and LCDR3 as shown in SEQ ID NO: 12, respectively.
In a preferred embodiment, the anti-CD3 antibody or antigen-binding fragment thereof comprises: a) a heavy chain variable region sequence comprises HCDR1, HCDR2 and HCDR3 as shown in SEQ ID NO: 01, SEQ ID NO: 02 and SEQ ID NO: 03, respectively; and a light chain variable region sequence comprises LCDR1, LCDR2 and LCDR3 as shown in SEQ ID NO: 07, SEQ ID NO: 08 and SEQ ID NO: 09, respectively; or b) a heavy chain variable region sequence comprises HCDR1, HCDR2 and HCDR3 as shown in SEQ ID NO: 04, SEQ ID NO: 05 and SEQ ID NO: 06, respectively; and a light chain variable region sequence comprises LCDR1, LCDR2 and LCDR3 as shown in SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12, respectively.
In a preferred embodiment, the anti-CD3 antibody or antigen-binding fragment is selected from murine antibody, chimeric antibody, humanized antibody, human antibody or the antigen-binding fragment thereof.
In some embodiments, the anti-CD3 antibody or antigen-binding fragment comprises: a) the heavy chain variable region as shown in SEQ ID NO: 13, or having at least 80%, 85%, 90%, 95% or 99%sequence identity therewith; and/or the light chain variable region as shown in SEQ ID NO: 18, or having at least 80%, 85%, 90%, 95%or 99%sequence identity therewith; or b) the heavy chain variable region as shown in SEQ ID NO: 17, or having at least 80%, 85%, 90%, 95%or 99%sequence identity therewith; and/or the light chain variable region as shown in SEQ ID NO: 31, or having at least 80%, 85%, 90%, 95%or 99%sequence identity therewith.
In some embodiments, the anti-CD3 antibody or antigen-binding fragment comprises: a) the heavy chain variable region having the sequence selected from any one of SEQ ID NO: 13, 14, 15 and 16; and/or the light chain variable region having the sequence selected from any one of SEQ ID NO: 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 and 30; or b) the heavy chain variable region as shown in SEQ ID NO: 17; and/or the light chain variable region as shown in SEQ ID NO: 31.
In a preferred embodiment, the anti-CD3 antibody or antigen-binding fragment comprises: a) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 18; or b) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 31; or c) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 19; or d) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 20; or e) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 21; or f) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 22; or g) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 23; or h) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 24; or i) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 25; or j) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 26; or k) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 27; or l) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 28; or m) the heavy chain variable region of SEQ ID NO: 14 and the light chain variable region of SEQ ID NO: 18; or n) the heavy chain variable region of SEQ ID NO: 15 and the light chain variable region of SEQ ID NO: 18; or o) the heavy chain variable region of SEQ ID NO: 16 and the light chain variable region of SEQ ID NO: 18; or p) the heavy chain variable region of SEQ ID NO: 16and the light chain variable region as of SEQ ID NO: 29; or q) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region as of SEQ ID NO: 30.
In a preferred embodiment, the anti-CD3 antibody or antigen-binding fragment comprises: a) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 38; or b) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 51; or c) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 39; or d) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 40; or e) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 41; or f) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 42; or g) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 43; or h) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 44; or i) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 45; or j) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 46; or k) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 47; or l) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 48; or m) the heavy chain of SEQ ID NO: 35 and the light chain of SEQ ID NO: 38; or n) the heavy chain of SEQ ID NO: 36 and the light chain of SEQ ID NO: 38; or o) the heavy chain of SEQ ID NO: 37 and the light chain  of SEQ ID NO: 38; or p) the heavy chain of SEQ ID NO: 37 and the light chain of SEQ ID NO: 49; or q) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 50.
In some embodiments, the anti-CD3 antibody is a full-length antibody, further comprising human antibody constant regions; preferably, the heavy chain constant region of the human antibody constant regions is selected from constant regions of human IgG1, IgG2, IgG3 and IgG4 and conventional variants thereof, and the light chain constant region of the human antibody constant regions is selected from κ and λ chain constant regions of human antibody and conventional variants thereof; more preferably the full-length antibody comprises a human antibody heavy chain constant region of SEQ ID NO: 32 and a human light chain constant region of SEQ ID NO: 33.
In some embodiments, the CD3 antigen-binding fragment is selected from the group consisting of Fab, Fab', F (ab') 2, variable fragment (Fv) , single chain variable fragment (scFv) , dimerized domain V (diabody) , disulfide stabilized Fv (dsFv) , and any CDR-containing peptides.
In a preferred embodiment, the anti-CD3 antibody or antigen-binding fragment binds to human CD3 with an affinity of a KD value of 1×10-5 M to 1×10-12 M.
In one aspects, the present disclosure provides an isolated antibody or the antigen-binding fragment thereof that competing with the above-mentioned antibody or the antigen-binding fragment to bind to human CD3.
In another aspects, the above-mentioned antibody or the antigen-binding fragment thereof having at least one of the following characteristics: a) binding to human CD3 with an affinity of a KD value of 1×10-7 M to 1×10-12 M; b) cross-reacting with CD3 of cynomolgus monkey or rhesus monkey; c) increased activation of T cells.
In some embodiments, the present disclosure provides an isolated nucleic acid molecule encoding any antibody or the antigen-binding fragment.
In one aspect, the present disclosure also provides a recombinant vector comprising the above-mentioned isolated nucleic acid molecule.
In another aspect, the present disclosure also provides a host cell transformed with the above-mentioned recombinant vector, wherein the host cell is selected from the group consisting of a prokaryotic cell and a eukaryotic cell, preferably a eukaryotic cell, more preferably a mammalian cell.
In one aspect, the present disclosure also provides a method for producing the above-mentioned antibody or the antigen-binding fragment in a medium to produce and accumulate the antibody or the antigen-binding fragment thereof, and harvesting the antibody or the antigen-binding fragment thereof from the culture.
In one aspect, the present disclosure also provides a method for immunologically detecting or measuring CD3, wherein the method comprises detecting the CD3 by contacting with the above-mentioned antibody or the antigen-binding fragment.
In one aspect, the present disclosure also provides a method for diagnosing a disease related to a human CD3 positive cell, wherein the method comprises a detecting or measuring the CD3 or CD3 positive cell by contacting with the above-mentioned antibody or the antigen-binding fragment.
In some embodiments, the present disclosure provides a pharmaceutical composition, which comprises a therapeutically effective amount of the above-mentioned antibody or the antigen-binding fragment, and one or more pharmaceutically acceptable carriers, diluents or excipients.
In some embodiments, the present disclosure also provides a method of treating a CD3-mediated disease or disorder in a subject in need thereof, comprising administering to the subject  the above-mentioned antibody or the antigen-binding fragment, or the above-mentioned pharmaceutical composition, wherein the disease or disorder is selected from the group consisting of cancer, autoimmune and inflammatory diseases
The main advantages of the invention are:
(1) The invention obtained fully human antibody variable region using transgenic mice with human antibody variable genes, and the obtained antibodies have a series of desired characteristics:
i) the variable region sequences are different from the existing antibody;
ii) the obtained antibodies have capacity of binding to human CD3
iii) the obtained antibodies bind to CD3 on cell surface;
iv) the obtained antibodies stimulate T cell activation;
(2) The invention has obtained antibodies with different sequences, which can specifically bind to CD3, including to cross-reactive with other species, in particular with non-human primates such as cynomolgus monkeys.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1. Mice immunized using RIMMS protocol were tail-bleeded, and serum titers were measured against Jurkat E6.1 cells by flow cytometry (BD Fortessa X20) at 1/50 dilution. The cell binding is shown as histogram of normalized count across different fluorescence intensity. UCHT1 was used as positive control. Serums from unimmunized mice were used as negative control.
Figure 2. Representative binding histograms from flow cytometry analysis of 13A1 hybridoma clone. The solid line represents the binding to Jurkat E6.1 cell and the dash line represents the binding to J. RT-T3.5. cell. F2B was used as positive control for CD3 binding and the secondary antibody-only was used as negative control.
Figure 3. Cyno T cell binding of selected CD3 hybridoma supernatants were analyzed by flow cytometry and shown as histograms. SP34 was used as positive control and clone 27A5 was used as negative control.
Figure 4. Graphs showing the T cell activation activity of selected hybridoma clone supernatants using NFAT reporter assay. F2B and OKT3 are anti-CD3 mAbs that were used as positive controls.
Figure 5. Binding curves of 13A1 recombinant antibodies to human (A) and cyno CD3 δε (B) were determined by ELISA and shown as indicated.
Figure 6 Recombinant 13A1 binding to CD3+ Jurkat E6.1 (A) and human primary T cell (B) and positive control (F2B) were determined by flow cytometry and shown as dose response curves as indicated.
Figure 7 Binding curves of 13A1 VH1-VL1 mutants 1-10 (A) , mutants 32-34 (B) and mutants 36 (C) to Jurkat E6.1 cells were shown as indicated.
Figure 8. Graphs showing the T cell activation activities of indicated recombinant antibodies  using NFAT reporter assay. 13A1 VH1-VL1 and 13A1 VH1-VL2 activities were shown in panel A. Activities of 13A1 VH1-VL1 and its light chain mutants or heavy chain mutants were shown in (B) , (C) and (D) .
Figure 9. Human PBMC activation by the anti-CD3 antibodies as measured by cell surface CD69 amount measured by flow cytometry analysis.
Figure 10. T cell activation upon binding of 13A1 VH1-VL1 and its mutation, as measured by up-regulation of the activation marker CD25 on CD4 (A) or CD8 T cells (C) , respectively the activation marker CD69 on either CD4 (B) or CD8 T cells (D) .
Figure 11. Kinetic of 13A1 VH1_VL1 was determined by binding to different concentrations of antigen (human CD3δε) . The equilibrium rate constant KD was calculated by determining the dissociation rate constant, Koff, and association rate constant, Kon.
DETAILED DESCRIPTION
DEFINITIONS
Before the present invention is detailed below, it is to be understood that the present invention is not limited to the particular methodologies, protocols and reagents described herein, as those may vary. It is also to be understood that the terminology used herein is for the purpose of describing the particular embodiments only, and is not intended to limit the scope of the invention, which will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains.
For interpretation of the specification, the following definitions will be applied and wherever appropriate, terms used in the singular may also include the plural and vice versa. It is to be understood that the terminology used herein is for the purpose of describing specific embodiments only and is not intended to be limiting.
The terms "CD3" and "CD3 antigen" are used interchangeably herein, and include any variants, isoforms and species homologs of human CD3 which are naturally expressed by cells or are expressed on cells transfected with the CD3 gene.
An "antibody" refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR) , interspersed with regions that are more conserved, termed framework regions (FR) . Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement  system.
The term "antigen-binding fragment" of an antibody, as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., CD3) . It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term "antigen-binding fragment " of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F (ab') 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341: 544-546) , which consists of a VH domain; (vi) an isolated complementarity determining region (CDR) , and (vii) a combination of two or more isolated CDRs which may optionally be joined by a synthetic linker. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv) ; see e.g., Bird et al. (1988) Science 242 : 423-426 ; and Huston et al. (1988) Proc. Natl. Acad. Sci. 85 : 5879-5883) . Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody.
The term "human antibody" , as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues 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" , as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
The term "recombinant human antibody" , as used herein, includes all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom (described further in Section I, below) , (b) antibodies isolated from a host cell transformed to express the antibody, e.g., from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial human antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies can be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
The term “CDR” refers to one of the six hypervariable regions within the variable domain of an antibody that primarily contributes to antigen binding. One of the most commonly used definitions for the six CDRs is provided by Kabat E. A. et al. (1991) Sequences of proteins of immunological interest. NIH Publication 91-3242. As used herein, the Kabat definition of CDR only applies to CDR1, CDR2 and CDR3 of the light chain variable domain (LCDR1, LCDR2, LCDR3  or L1, L2, L3) , as well as CDR1, CDR2 and CDR3 of heavy chain variable domain (HCDR1, HCDR2, HCDR3 or H1, H2, H3) .
Methods and techniques for identifying CDRs within HCVR and LCVR amino acid sequences are well known in the art and can be used to identify CDRs within the specified HCVR and/or LCVR amino acid sequences disclosed herein. Exemplary conventions that can be used to identify the boundaries of CDRs including, e.g., Chothia based on the three-dimensional structure of antibodies and the topology of the CDR loops (Chothia et al. (1989) Nature 342: 877-883) , Kabat based on antibody sequence variability (Kabat et al., Sequences of Proteins of Immunological Interest, 4th edition, US Department of Health and Human Services, National Institutes of Health (1987) ) , AbM (University of Bath) , Contact (University College London) , International ImMunoGeneTics database (IMGT) (imgt. cines. fr/on the World Wide Web) , and North CDR definition based on the affinity propagation clustering using a large number of crystal structures. Those skilled in the art can easily identify the CDRs defined by each numbering system.
A useful comparison of CDR numbering is as below:
Note 1: some of these definitions (particularly for Chothia loops) vary depending on the individual publication examined; Note2: any of the numbering schemes can be used for these CDR defintions, except the contact definition uses the Chothia or Martin (enhanced Chothia) definition; Note 3: the end of the Chothia HCDR1 loop when numbered using the Kabat numbering convention varies between H32 and H34 depending on the length of the loop. This is because the Kabat numbering scheme places the insertions at H35A and H35B.
“Conservative modification” or “conservative replacement or substitution” refers to substitutions of amino acids in a protein with other amino acid having similar characteristics (e.g., charge, side chain size, hydrophobicity/hydrophilicity, backbone conformation and rigidity, etc. ) , such that the changes can be frequently made without altering the biological activity of the protein. It will be appreciated by those skilled in the art that, in general, a single amino acid substitution in a non-essential region of polypeptide does not substantially alter biological activity (see, for example, Watson et al. (1987) Molecular Biology of the Gene, The Benjamin/Cummings Pub. Co., Page 224, (4th edition) ) . In addition, substitutions with structurally or functionally similar amino acids are unlikely to disrupt biological activity.
The term “nucleic acid molecule” as used herein refers to a DNA molecule and a RNA molecule. The nucleic acid molecule may be single stranded or double stranded but is preferably a double stranded DNA. A nucleic acid is “effectively linked” when it is placed into functional relationship with another nucleic acid sequence. For example, if a promoter or enhancer affects transcription of  a coding sequence, the promoter or enhancer is effectively linked to the coding sequence.
The preparation method of the nucleic acid is a conventional preparation method in the art. Preferably, it comprises the following steps: obtaining the nucleic acid molecule encoding the above-mentioned protein by gene cloning technology, or obtaining the nucleic acid molecule encoding the above-mentioned protein by the method of artificial full-length sequence synthesis.
Those skilled in the art know that the base sequence encoding the amino acid sequence of the protein can be replaced, deleted, changed, inserted or added appropriately to provide a polynucleotide homolog. The homolog of the polynucleotide of the present invention can be prepared by replacing, deleting or adding one or more bases of the gene encoding the protein sequence within the scope of maintaining the activity of the antibody.
The term “vector” refers to a nucleic acid molecule capable of transporting another nucleic acid to that it has been linked. In one embodiment, the vector is a “plasmid” that refers to a circular double stranded DNA loop into which additional DNA segment can be ligated. In another embodiment, the vector is a viral vector, wherein an additional DNA segment can be ligated into viral genome. The vectors disclosed herein are capable of self-replicating in a host cell into which they have been introduced (for example, a bacterial vector having a bacterial replication origin and a episomal mammalian vector) or can be integrated into the genome of a host cell upon introduction into host cell, thereby is replicated along with the host genome (e.g., a non-episomal mammalian vector) .
The recombinant expression vector can be obtained by conventional methods in the art, that is, by connecting the nucleic acid molecule of the present invention to various expression vectors, thus being constructed. The expression vector is one of a variety of conventional vectors in the art, as long as it can carry the above-mentioned nucleic acid molecule. The vector preferably includes: various plasmids, cosmids, phage or virus vectors and the like.
The term "transfectoma" , as used herein, includes recombinant eukaryotic host cell expressing the antibody, such as CHO cells, NS/0 cells, HEK293 cells, plant cells, or fungi, including yeast cells.
The sequence of the DNA molecule for the antibody or a fragment thereof according to the present invention can be obtained by conventional techniques, for example, methods such as PCR amplification or genomic library screening. In addition, the sequences encoding light chain and heavy chain can be fused together, to form a single-chain antibody.
Once a relevant sequence is obtained, the relevant sequence can be obtained in bulk using a recombination method. This is usually carried out by cloning the sequence into a vector, transforming a cell with the vector, and then separating the relevant sequence from the proliferated host cell by conventional methods.
In addition, a relevant sequence can be synthesized artificially, especially when the fragment is short in length. Usually, several small fragments are synthesized first, and then are linked together to obtain a fragment with a long sequence.
At present, it is possible to obtain a DNA sequence encoding the antibody of the present invention (or fragments thereof, or derivatives thereof) completely by chemical synthesis. The DNA sequence can then be introduced into a variety of existing DNA molecules (or, for example, vectors) and cells known in the art. In addition, mutations can also be introduced into the protein sequences of the present invention by chemical synthesis.
In general, under conditions suitable for expression of the antibody according to the present  invention, the host cell obtained is cultured. Then, the antibody of the present invention is purified by using conventional immunoglobulin purification steps, for example, the conventional separation and purification means well known to those skilled in the art, such as protein A-Sepharose, hydroxyapatite chromatography, gel electrophoresis, dialysis, ion exchange chromatography, hydrophobic chromatography, molecular sieve chromatography or affinity chromatography.
The monoclonal antibody obtained can be identified by conventional means. For example, the binding specificity of a monoclonal antibody can be determined by immunoprecipitation or an in vitro binding assay (such as radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELISA) ) . The binding affinity of a monoclonal antibody can be determined by, for example, the Scatchard analysis (Munson et al., Anal. Biochem., 107: 220 (1980) ) .
The antibody according to the present invention can be expressed in a cell or on the cell membrane, or is secreted extracellularly. If necessary, the recombinant protein can be separated and purified by various separation methods according to its physical, chemical, and other properties. These methods are well known to those skilled in the art. The examples of these methods comprise, but are not limited to, conventional renaturation treatment, treatment by protein precipitant (such as salt precipitation) , centrifugation, cell lysis by osmosis, ultrasonic treatment, supercentrifugation, molecular sieve chromatography (gel chromatography) , adsorption chromatography, ion exchange chromatography, high performance liquid chromatography (HPLC) , and any other liquid chromatography, and the combination thereof.
As used herein, the term "kd" (sec -1) , as used herein, is intended to refer to the dissociation rate constant of a particular antibody-antigen interaction. Said value is also referred to as the koff value.
The term "ka" (M-1 x sec -1) , as used herein, is intended to refer to the association rate constant of a particular antibody-antigen interaction.
The term "KD" (M) , as used herein, is intended to refer to the dissociation equilibrium constant of a particular antibody-antigen interaction.
The terms “specific binding, ” “selective binding, ” “selectively bind, ” and “specifically bind” refer to the binding of an antibody to an epitope on a predetermined antigen.
The term"Pharmaceutical composition " , as used herein, is intended to refer to a mixture containing one or more of the compounds or a physiological/pharmaceutically acceptable salt or prodrug thereof described herein with other chemical components, such as physiological/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to promote the administration to the organism, which is beneficial to the absorption of the active ingredient and exerts the biological activity.
“Administration” and “treatment, ” when applied to an animal, human, experimental subject, cell, tissue, organ, or biological fluid, refer to contact with an exogenous pharmaceutical, therapeutic, diagnostic reagent, 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 the cell with a reagent, as well as contacting a fluid with a reagent, wherein 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, ” when applied to a human, veterinary, or research subject, refers to therapeutic treatment, prophylactic or preventative measures, research and diagnostic applications.
In addition, the present disclosure includes a medicament for treating a disease associated with  CD3, comprising an antibody or an antigen-binding fragment thereof of the present disclosure as an active ingredient.
There is no limitation on the diseases related to CD3, as long as it is a disease associated with CD3, for example, the therapeutic response induced by the molecules disclosed in the present disclosure can be reduced by binding human CD3. Therefore, the molecules of the present disclosure are very useful for those who suffer a tumor, cancer or infectious disease when in preparations and formulations suitable for therapeutic applications.
In addition, the present disclosure relates to a method for immunologically detecting or measuring CD3, a reagent for immunologically detecting or measuring CD3, a method for immunologically detecting or measuring cells expressing CD3, and a diagnostic reagent for diagnosis of disease related to CD3 positive cells, comprising the antibody or antigen-binding fragment of the present disclosure that specifically recognizes human CD3, as an active ingredient.
In the present disclosure, the method for detecting or determining the amount of CD3 may be any known method. For example, it includes immunodetection or assay.
The immunodetection or assay is a method of detecting or determining the amount of antibody or antigen by using labeled antigen or antibody. Examples of immunodetection or assay include a radioactive substance labeled immunological antibody method (RIA) , an enzyme immunoassay (EIA or ELISA) , a fluorescent immunoassay (FIA) , a luminescent immunoassay, a western blotting method, physicochemical methods, etc.
The above-mentioned diseases related to CD3 positive cells can be diagnosed by detecting or measuring cells expressing CD3 by using the antibodies or antibody fragments thereof of the present invention.
In order to detect cells expressing the polypeptide, a known immunodetection can be used, and preferably immunoprecipitation, fluorescent cell staining or immunohistochemical staining etc. can be used. Furthermore, a fluorescent antibody staining method etc. using FMAT8100HTS system (Applied Biosystem) can be used.
EXAMPLES
The invention is further illustrated by the following specific examples. It is to be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the invention. The experimental methods without detailed conditions in the following examples are generally in accordance with the conditions described in the conventional conditions such as Sambrook. J et al. "Guide to Molecular Cloning Laboratory" (translated by Huang Peitang et al., Beijing: Science Press, 2002) , or in accordance with the conditions recommended by the manufacturer (for example, product manuals) . Percentages and parts are by weight unless otherwise stated. The experimental materials and reagents used in the following examples are commercially available unless otherwise specified.
The room temperature described in the examples is a conventional room temperature in the art, and is generally 10-30℃.
EXAMPLE 1 Immunization and screening of antibody
Preparation of immunogen
Design of CD3 antigen: A combination of Jurkat E6.1, DNA and recombinant protein antigens  were used to immunize mice. The recombinant ectodomain of human CD3E and CD3D were expressed and purified as heterodimer and were commercially available at AcroBiosystems (catalog numbers CDD-H52Wa, CDD-H52W1, CDD-C52W4, CDD-C52W9) . Either Fc, His Tag&Fc, Flag Tag or His Tag&Tag Free were used.
Immunization
Immunization protocol:
Anti-CD3 antibodies were obtained by immunizing genetically modified mouse encoding human immunoglobulin heavy and kappa light chain variable regions with a combination of human CD3 antigen, cynomolgus CD3 antigen, human CD3 DNA, and/or Jurkat E6.1 cells (Sigma-Aldrich, Catalog No. 88042803) which naturally express human CD3. The antibody immune response was monitored by a CD3-specific immunoassay. When a desired immune response was achieved splenocytes were harvested from each mouse and either (1) fused with mouse myeloma cells to preserve their viability and form hybridoma cells and screened for CD3 specificity, or (2) B-cell sorted using a human CD3 with a C-terminal 6-His tag as the sorting reagent that binds and identifies reactive antibodies (antigen-positive B cells) . Figure 1 shows the results of the antibody titer in serum detected by Flow cytometry after immunization with CD3 protein.
The results show that: after 3 times of immunization with the immunogens, the selected mice had an antigen-selective cell binding titer of more than 1: 50, indicating that mice had a better humoral immune response to the immunogen, and their spleen cells can be used for Hybridoma cell preparation.
Spleen cell fusion
The spleen lymphocytes and myeloma cells Sp2/0 (CRL-158) were fused to obtain hybridoma cells by electrofusion or PEG fusion. PEG fusion was performed using ClonacellTM HY technology (STEMCELL technologies) , following manufacturer’s instructions. The primary cell: mouse myeloma cell line ratio was 1: 1 for electrofusion, and 10: 1 for PEG fusion.
Screening of hybridoma cells by ELISA
ELISA was performed using the DuoSet ELISA Ancillary Kit (R&D System, DY008) . ELISA plates were coated with 5ug/ml of human CD3DE (AcroBio CDD-H52W1) and cyno CD3DE (AcroBio, CDD-C52W4) overnight. Excess unbound proteins were washed off by washing the plates three times with the wash buffer before blocking for 1 hour at room temperature. 50ul CD3 hybridoma supernatant was added in duplicate wells and incubated for 1 hour. Excess unbound antibodies were washed off and 50ul of 1: 1000 diluted secondary antibody Goat anti-mouse IgG Fc-HRP (Jackson ImmunoResearch 115-035-071) was added to each well for another 1 hour. Plates were washed before the addition of chemiluminescence agents (color A and color B) according to manufacturer's protocol. Optical density at 450nm of samples were measured by microplate reader (PerkinElmer) .
Table 1. The binding activity results of ELISA assay
The results show that the supernatant of hybridoma clone 13A1 antibody of the present disclosure have high affinity to the human and cynomolgus CD3.
Jurkat Binding Screening
Hybridoma supernatants was subjected to binding tests on human T cell lines (CD3+ Jurkat cell line clone E6.1 and CD3-clone J. RT-T3.5 (ATCC) using flow cytometry analysis. The primary staining was performed using undiluted hybridoma supernatants, followed by fluorescence labeled secondary antibody (PE Goat anti-mouse IgG, Biolegend Cat#405307) . Figure 2 shows examples of selected flow cytometry histograms. The clone 13A1 was identified with enhanced binding profile to Jurkat E6.1 compared to J. RT-T3.5 cells.
Cyno-T cell (HSC-F) Binding
Selected clones from Jurkat cell binding screening were further tested for cynomolgus monkey T cell binding (HSC-F) using flow cytometry analysis. Cynomolgus monkey T cell line (HSC-F) was obtained from Non-Human Primate Reagent Resource (NHPRR) . The staining was performed in a similar fashion as described above. Representative results are shown in Figure 3.
T cell activation (NFAT Reporter Assay)
T cell activation activity of hybridoma supernatants or recombinant anti-CD3 antibodies were measured using NFAT reporter assay, using Jurkat-LuciaTM NFAT cells (InvivoGen jktl-nfat) . U-bottom 96-well plates were dry coated with capturing antibody (goat anti-mouse IgG Fcγ fragment specific, Jackson ImmunoResearch 115-005-071) , at 4μg/ml in PBS and 50 μl/well. Hybridoma supernatants or recombinant anti-CD3 antibodies diluted in culture medium were added to the pre-coated plate at 50 μl/well for overnight. The Jurkat-LuciaTM NFAT cells were added to each well at 400,000 cells per well in 200ul culture medium. The cells were maintained at 37℃ with 5%CO2 for 24 hours, then the culture supernatants were collected. Luciferase activity in culture supernatant was measured using QUANTI-Luc assay solution (InvivoGen) according to manufacturer’s instruction. Luminescence signal was recorded by VICTOR Nivo Multimode Microplate Reader (PerkinElmer) . Purified anti-CD3 antibodies F2B and OKT3 were used as positive controls. Supernatant from another hybridoma clone was used as a negative control. Clone 13A1 was selected for their ability to active human T cells indicated by the in vitro reporter assay as shown in Figure 4.
EXAMPLE 2 Sequencing of positive hybridoma clones
The process of cloning sequences from positive hybridomas are as follows. The logarithmic growth phase hybridoma cells were collected, RNA was extracted, and reverse transcription was performed then followed by VDJ region amplification. Amplified cDNA library from each clones were subjected to next-generation sequencing. The amino acid sequences of the heavy and light chain variable region DNA sequences corresponding to the antibodies 13A1 were obtained (the amino acid residues of the CDRs in VH/VL are underlined according to Kabat & Wu annotation: 

NOTE: The order is FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4, italic portion represents FR sequence, and underlined portion represents CDR sequence.
EXAMPLE 3 Construction and Expression of Recombinant antibody 13A1
1. Molecular cloning of recombinant antibodies
The cDNA sequences that encode VH and VL regions of selected clones were directly synthesized as DNA fragments with 5’-end in-frame leader sequence (MGWSCIILFLVATATGVHS) . These DNA fragments were cloned into selected vectors using NEBuilder DNA Assembly Cloning Kit (New England Biolabs) . VH region was cloned into pFUSE-CHIg_hG1 vector (InvivoGen #pfuse-hchg1) , which in-frame with constant region of hIgG1 heavy chain in the vector. VL region was cloned into pFUSE2-CLIg_hk vector (InvivoGen, #pfuse2-hclk) , which in-frame with constant region of hIg kappa light chain in the vector.
2. Expression and purification of recombinant antibodies
The heavy chain expression plasmid and light chain plasmids were co-transfected into Expi293F cells (ThermoFisher, #A14527) using ExpiFectamine 293 Transfection Kit (ThermoFisher, A14524) , or into ExpiCHO-Scells (ThermoFisher #A29127) using ExpiFectamine CHO Transfection Kit (ThermoFisher, A29129) . Based on the manufacturer’s instructions, plasmid DNA concentration reached 1.0 ug per ml of suspended cells, with LC: HC vector ratio 1: 1. The transfected cells were cultured 5 to 7 days on an orbital shaker at 37 C, 8%CO2. Conditioned medium was collected and antibodies were purified using HiTrap MabSelect SuRe column (Cytiva, #17549112) on AKTA Pure 25 machine (Cytiva) . Eluted antibodies were neutralized with Tris Buffer (pH 9.0) and subjected to PBS buffer exchange. Product concentration was measured by UV absorption, and quality was determined by SDS-PAGE and HPLC.
EXAMPLE 4 The mutation and germlinization of the clone 13A1
The purpose of the mutation is to obtain a group of anti-CD3 antibodies with varied affinities and potentially lower immunogenicity by converting certain residues to the germline sequence.
Selection of a template and back mutation design for hybridoma clone 13A1 (Table2 and Table3) .
Table 2. 13A1 light chain variable region and heavy chain variable region mutations
NOTE: For example, I14T indicates a mutation from I to T at position 14 according to Kabat numbering system. WT indicates that the sequence does not contain amino acid mutations.
Table 3. Sequence combinations for antibody 13A1

NOTE: This table shows various sequence combinations of different mutations. For example, 13A1_mut01 indicates that two kinds of mutation (13A1_VL1. A and 13A1_VH1) are present on the 13A1_mut01, and so on.
Table 4. Light chain and heavy chain of the antibody
The performance and effect of the present disclosure were verified by the following biochemical tests or in vivo pharmacological test:
TEST EXAMPLE 1. Enzyme-linked immunosorbent assay (ELISA) detection of the binding of antibodies to hCD3δε and cynoCD3δε
1. recombinant antibodies
ELISA plates were coated with 5ug/ml human or cyno CD3δε-His (AcroBiosystems) overnight. Serial dilutions of recombinant antiCD3 antibodies 13A1 wereperformed, starting at 100nM and added to CD3 coated plates. The excess non-bound antibodies were washed away before addition of the mouse anti-human IgG Fc-HRP (Abcam 99774) . The results were measured by O. D. at 450nm on a microplate reader. Data were analyzed with GraphPad Prism 9 (Figure 5) . EC50 values were calculated for the binding of the CD3 antibody to human and cyno CD3δε protein (Table 5) .
Table 5. EC50 values of 13A1 binding to human and cyno CD3 δε
TEST EXAMPLE 2. Detection of the binding of antibodies to CD3+ Jurkat cells and human  primary T cell by Flow cytometry (FACS)
1. recombinant antibodies
The binding curves to cell surface CD3 were determined for 13A1_VH1_VL1, 13A1_VH1_VL2 (Figure 6) recombinant antibodies by flow cytometry analysis. Jurkat E6.1 cells were aliquoted into 0.3 million/well and 60ul of serially diluted antibodies were added to each well. The staining was performed for 1 hour at 4 degrees in the dark. The PE goat anti human IgG Fc (Jackson ImmunoResearch) was used as the secondary antibody at 1: 200 dilution.
13A1_VH1_VL1 binding to primary T cell was determined by using the freshly isolated PBMCs from one healthy donor. 0.3 million PBMCs were aliquoted to each well and stained with anti-human TCRα/β-FITC (Biolegend Cat 306706) . 13A1 antibody was serially diluted and 60ul was added to each well as primary staining. The PE goat anti human IgG was used as secondary antibody as Figure 7A.
Stained samples were analyzed by flow cytometer (BD LSRFortessa) . CD3 binding was measured by PE florescence intensity on JurkatE6.1 (Fig. 6A) or primary T cells (TCRα/β+) (Fig. 6B) and was analyzed by FlowJo software. Data were analyzed with GraphPad Prism 9. The binding affinity was calculated and shown as EC50 (nM) values (Table 6) .
Table 6. EC50 values of 13A1 binding to Jurkat E6.1 cell
2. 13A1 mutations
Series of single amino acid or multiple amino acids mutations were introduced to 13A1_VH1_VL1 in order to modulate the binding affinity to CD3. The sequence of 13A1_VH1_VL1 was compared to the germ-line sequence and mutations were introduced on various CDR and framework regions back to the germ-line sequence (Example 3) . Recombinant antibodies containing the mutations were generated in human IgG1 format. Serial dilutions were performed starting at 1000nM and the binding curve to Jurkat E6.1 cells for each mutant was determined as described above by flow cytometry analysis. The result of this is shown in Figure 7A-C and the EC50 of binding is shown in Table 7A-C.
Table 7A. EC50 values of 13A1 mutants binding to Jurkat E6.1 cell

Table 7B. EC50 values of 13A1 mutants binding to Jurkat E6.1 cell
Table 7C. Relative EC50 values of 13A1 mutants binding to Jurkat E6.1 cell
TEST EXAMPLE 3. Detection of the effect of CD3 antibody on T cell activation by NFAT reporter assay or human PBMCs activation.
1. NFAT reporter assay
To assess the potential of CD3 antibodies to trigger T-cell activation, 96 well U-bottom plates  were dry coated with 10 ug/ml capturing antibody (Jackson ImmunoResearch’s AffiniPure goat anti-human IgG, Fcγ specific, #109-005-170) , and then after PBS wash, were wet coated with 10 ug/ml anti-CD3 antibodies. After PBS wash, NFAT reporter cells (InvivoGen #jktl-nfat) were added to each well at 0.4 million/well in RPMI 1640, with 10%FBS. After 24 hours of incubation at 37 C, 5%CO2. Reporter activity was measured using QUANTI-Luc assay (InvivoGen) as manufacture’s instruction. luminescence was measured using Victor Nivo multimode microplate reader (PerkinElmer) , and results were analyzed by GraphPad Prism 9 software. The effect of recombinant 13A1 on T cell activations were shown in Figure 8A, and EC50 values were shown in Table8. Different light chain and/or heavy chain mutations of 13A1_VH1_VL1 exhibited difference in their T cell activation ability compared to parental 13A1 as indicated in Figure 8B-D, and EC50 values were displayed in Table 9A-C.
Table 8. EC50 values of control and candidate recombinant mAbs activating the NFAT reporter cells
Table 9A. EC50 values of control and 13A1 mutants activating the NFAT reporter cells
Table 9B. EC50 values of control and 13A1 mutants activating the NFAT reporter cells
Table 9C. Relative EC50 values of control and 13A1 mutants activating the NFAT reporter cells
2. Human PBMCs Activation
Fresh PBMCs from single healthy donor were plated in RPMI1640 with GlutaMax 10%FBS medium at a density of 1x106 cells/ml and incubated overnight at 37℃. Anti-CD3 antibodies OKT3, 13A1 and F2B, as well as control human IgG were serially diluted. PBMCs were then added to the antibody solutions at 50,000 cells per well, and final volume of 200 μl per well. Cells were incubated at 37℃ 5%CO2 for 2 days, then stained with anti-human CD69 antibody (Biolegend Catalog No. 310938) and analyzed by BD Fortessa X20. Data were ananlyzed by FlowJo and GraphPad Prism 9 software. PBMCs activation by anti-CD3 antibodies indicated as the ΔMFI of CD69 over control were shown in Figure 9, EC50 values were shown in Table 10.
Table 10. EC50 values of control and 13A1_VH1_VL1 primary human PBMCs activation
Similar experiment was performed to evaluate 13A1_Mut36 activity on primary human T cell activation, with the indication of induced expression of T cell activation markers (CD25 and CD69) . In brief, fresh isolated PBMCs were incubated a serial diluted 13A1, 13A1-Mut36, OKT3 and Control IgG, then stained with the following antibodies: anti-hCD8 (Biolegend 300906) , anti-hCD69 (Biolegend 310938) , anti-hCD4 (Biolegend 300537) , and anti-hCD25 (Biolegend 302606) . Then stained cells analyzed by flow cytometery as described above.
The results were shown in Figure 10A-D. Both 13A1 and 13A1-Mut36 present excellent T cell activation. CD4+CD25+ (%CD4+) represents the percentage of CD25 positive CD4 cells in a given group, T cell activation was determined as percent of CD4 T cells expressing CD25 (FIG. 10A) or CD69 (FIG. 10B) , respectively CD8 T cells expressing CD25 (FIG. 10C) and CD69 (FIG. 10D) .
TEST EXAMPLE 4. Antibody affinity test
The affinity between 13A1_VH1_VL1 and human CD3δε was determined by Octet (Octet Red 384) instrument. Anti-hIgG Fc Capture (AHC) Biosensors were selected, and the sensors were balanced with buffer solution for 10min. Subsequently, sensors were dipped into the wells containing 2ug/ml 13A1_VH1_VL1 to load the antibody onto the probes. The excess unbound antibody was washed off. The antigen binding was performed in the wells containing serially diluted human CD3δε-His, with concentration ranging from 300nM to 0.4nM, for 10min. Then probes were dipped into new buffer wells to initiate the dissociation for another 30min. The buffer subtracted data was input into Prism to plot the association and dissociation curves (Figure 11) . The equilibrium rate constant KD was calculated to be ~24nM
SEQUENCE LIST






Claims (23)

  1. An anti-CD3 antibody or an antigen-binding fragment thereof comprising: an antibody heavy chain variable region comprising one or more HCDR as shown in the sequences selected from SEQ ID NOs: 01, SEQ ID NO: 02, SEQ ID NO: 03, SEQ ID NO: 04, SEQ ID NO: 05 and SEQ ID NO: 06; and an antibody light chain variable region comprising one or more LCDR as shown in the sequences selected from SEQ ID NOs: 07, SEQ ID NO: 08, SEQ ID NO: 09 (X1QX2TX3FPYT) , SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12; wherein X1 is M, Q, T or A, X2 is L or A, X3 is H or Q.
  2. An anti-CD3 antibody or antigen-binding fragment thereof according to claim 1, wherein the antibody heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 regions and the antibody light chain variable region comprising LCDR1, LCDR2 and LCDR3 regions, wherein:
    a) HCDR1 as shown in SEQ ID NO: 01 or SEQ ID NO: 04;
    b) HCDR2 as shown in SEQ ID NO: 02 or SEQ ID NO: 05;
    c) HCDR3 as shown in SEQ ID NO: 03 or SEQ ID NO: 06;
    d) LCDR1 as shown in SEQ ID NO: 07 or SEQ ID NO: 10;
    e) LCDR2 as shown in SEQ ID NO: 08 or SEQ ID NO: 11;
    f) LCDR3 as shown in SEQ ID NO: 09 or SEQ ID NO: 12.
  3. An anti-CD3 antibody or antigen-binding fragment thereof according to claim 1, wherein the heavy chain variable region sequence comprises
    HCDR1 as shown in SEQ ID NO: 01,
    HCDR2 as shown in SEQ ID NO: 02, and
    HCDR3 as shown in SEQ ID NO: 03, respectively;
    or
    HCDR1 as shown in SEQ ID NO: 04,
    HCDR2 as shown in SEQ ID NO: 05, and
    HCDR3 as shown in SEQ ID NO: 06 respectively.
  4. An anti-CD3 antibody or antigen-binding fragment thereof according to claim 1, wherein the light chain variable region sequence comprises
    LCDR1 as shown in SEQ ID NO: 07,
    LCDR2 as shown in SEQ ID NO: 08, and
    LCDR3 as shown in SEQ ID NO: 09, respectively;
    or
    LCDR1 as shown in SEQ ID NO: 10,
    LCDR2 as shown in SEQ ID NO: 11, and
    LCDR3 as shown in SEQ ID NO: 12, respectively.
  5. An anti-CD3 antibody or antigen-binding fragment thereof according to claim 1, wherein:
    a) a heavy chain variable region sequence comprises HCDR1, HCDR2 and HCDR3 as shown in SEQ ID NO: 01, SEQ ID NO: 02 and SEQ ID NO: 03, respectively; and a light chain variable region sequence comprises LCDR1, LCDR2 and LCDR3 as shown in SEQ ID NO: 07, SEQ ID NO:  08 and SEQ ID NO: 09, respectively;
    or
    b) a heavy chain variable region sequence comprises HCDR1, HCDR2 and HCDR3 as shown in SEQ ID NO: 04, SEQ ID NO: 05 and SEQ ID NO: 06, respectively; and a light chain variable region sequence comprises LCDR1, LCDR2 and LCDR3 as shown in SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12, respectively.
  6. An anti-CD3 antibody or antigen-binding fragment thereof according to any one of claims 1-5, wherein the antibody or antigen-binding fragment thereof is selected from murine antibody, chimeric antibody, humanized antibody, human antibody or the antigen-binding fragment thereof.
  7. An anti-CD3 antibody or antigen-binding fragment thereof according to claim 6, wherein the antibody or antigen-binding fragment thereof comprising:
    a) the heavy chain variable region as shown in SEQ ID NO: 13, or having at least 80%, 85%, 90%, 95%or 99%sequence identity therewith; and/or the light chain variable region as shown in SEQ ID NO: 18, or having at least 80%, 85%, 90%, 95%or 99%sequence identity therewith;
    or
    b) the heavy chain variable region as shown in SEQ ID NO: 17, or having at least 80%, 85%, 90%, 95%or 99%sequence identity therewith; and/or the light chain variable region as shown in SEQ ID NO: 31, or having at least 80%, 85%, 90%, 95%or 99%sequence identity therewith.
  8. An anti-CD3 antibody or antigen-binding fragment thereof according to claim 7, wherein the antibody or antigen-binding fragment thereof comprising:
    a) the heavy chain variable region having the sequence selected from any one of SEQ ID NO: 13, 14, 15 and 16; and/or the light chain variable region having the sequence selected from any one of SEQ ID NO: 30, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 and 29;
    or
    b) the heavy chain variable region as shown in SEQ ID NO: 17; and/or the light chain variable region as shown in SEQ ID NO: 31.
  9. An anti-CD3 antibody or antigen-binding fragment thereof according to claim 8, wherein the antibody or antigen-binding fragment thereof comprising:
    a) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 30; or
    b) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 31; or
    c) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 19; or
    d) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 20; or
    e) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 21; or
    f) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 22; or
    g) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 23; or
    h) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 24; or
    i) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 25; or
    j) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 26; or
    k) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 27; or
    l) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 28; or
    m) the heavy chain variable region of SEQ ID NO: 14 and the light chain variable region of SEQ ID NO: 18; or
    n) the heavy chain variable region of SEQ ID NO: 15 and the light chain variable region of SEQ ID NO: 18; or
    o) the heavy chain variable region of SEQ ID NO: 16 and the light chain variable region of SEQ ID NO: 18; or
    p) the heavy chain variable region of SEQ ID NO: 16 and the light chain variable region of SEQ ID NO: 29; or
    q) the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region as of SEQ ID NO: 18.
  10. An anti-CD3 antibody or antigen-binding fragment thereof according to claim 9, wherein the antibody or antigen-binding fragment thereof comprising:
    a) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 50; or
    b) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 51; or
    c) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 39; or
    d) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 40; or
    e) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 41; or
    f) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 42; or
    g) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 43; or
    h) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 44; or
    i) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 45; or
    j) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 46; or
    k) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 47; or
    l) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 48; or
    m) the heavy chain of SEQ ID NO: 35 and the light chain of SEQ ID NO: 38; or
    n) the heavy chain of SEQ ID NO: 36 and the light chain of SEQ ID NO: 38; or
    o) the heavy chain of SEQ ID NO: 37 and the light chain of SEQ ID NO: 38; or
    p) the heavy chain of SEQ ID NO: 37 and the light chain of SEQ ID NO: 49; or
    q) the heavy chain of SEQ ID NO: 34 and the light chain of SEQ ID NO: 38.
  11. An anti-CD3 antibody or antigen-binding fragment thereof according to claim 1, wherein the  antibody further comprising human antibody constant regions;
    preferably, the heavy chain constant region of the human antibody constant regions is selected from constant regions of human IgG1, IgG2, IgG3 and IgG4 and conventional variants thereof, and the light chain constant region of the human antibody constant regions is selected from κ and λ chain constant regions of human antibody and conventional variants thereof;
    more preferably the antibody comprises a human antibody heavy chain constant region of SEQ ID NO: 32 and a human light chain constant region of SEQ ID NO: 33.
  12. An anti-CD3 antibody or antigen-binding fragment thereof according to claim 1, wherein the antigen-binding fragment is selected from the group consisting of Fab, Fab', F (ab') 2, variable fragment (Fv) , single chain variable fragment (scFv) , dimerized domain V (diabody) , disulfide stabilized Fv (dsFv) and CDR-containing peptides.
  13. An anti-CD3 antibody or antigen-binding fragment thereof according to claim 8, wherein the antibody or antigen-binding fragment thereof binds to human CD3 with an affinity of a KD value of 1×10-5 M to 1×10-12 M.
  14. An isolated antibody or the antigen-binding fragment thereof, competing with the antibody or the antigen-binding fragment according to claim 1 to bind to human CD3.
  15. An anti-CD3 antibody or the antigen-binding fragment thereof according to claim 9, having at least one of the following characteristics:
    a) binding to human CD3 with an affinity of a KD value of 1×10-5 M to 1×10-12 M;
    b) cross-reacting with CD3 of cynomolgus monkey or rhesus monkey;
    c) increased activation of T cells.
  16. An isolated nucleic acid molecule encoding the antibody or the antigen-binding fragment thereof according to any one of claims 1-15.
  17. A recombinant vector comprising the isolated nucleic acid molecule according to claim 16.
  18. A host cell transformed with the recombinant vector according to claim 17, wherein the host cell is selected from the group consisting of a prokaryotic cell and a eukaryotic cell, preferably a eukaryotic cell, more preferably a mammalian cell.
  19. A method for producing the antibody or the antigen-binding fragment thereof according to any one of claims 1-15, wherein the method comprises culturing the host cell according to claim 18 in a medium to produce and accumulate the antibody or the antigen-binding fragment thereof according to any one of claims 1-15, and harvesting the antibody or the antigen-binding fragment thereof from the culture.
  20. A method for immunologically detecting or measuring CD3, wherein the method comprises detecting the CD3 by contacting with the anti-CD3 antibody or antigen-binding fragment thereof of claim 1.
  21. A method for diagnosing a disease related to a human CD3 positive cell, wherein the method comprises a detecting or measuring the CD3 or CD3 positive cell by contacting with the anti-CD3 antibody or antigen-binding fragment thereof of claim 1.
  22. A pharmaceutical composition, which comprises a therapeutically effective amount of the anti-CD3 antibody or the antigen-binding fragment thereof according to any one of claim 1-15, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
  23. A method of treating a CD3-mediated disease or disorder in a subject in need thereof, comprising administering to the subject the anti-CD3 antibody or antigen-binding fragment according to any one of claims claim 1-15 or the pharmaceutical composition of claim 22, wherein the disease or disorder is selected from the group consisting of cancer, autoimmune and/or inflammatory diseases.
PCT/CN2023/107132 2022-07-13 2023-07-13 Antibody, antigen-binding fragment thereof, and pharmaceutical use thereof WO2024012513A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263388825P 2022-07-13 2022-07-13
US63/388,825 2022-07-13

Publications (1)

Publication Number Publication Date
WO2024012513A1 true WO2024012513A1 (en) 2024-01-18

Family

ID=89535619

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/107132 WO2024012513A1 (en) 2022-07-13 2023-07-13 Antibody, antigen-binding fragment thereof, and pharmaceutical use thereof

Country Status (1)

Country Link
WO (1) WO2024012513A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014047231A1 (en) * 2012-09-21 2014-03-27 Regeneron Pharmaceuticals, Inc. Anti-cd3 antibodies, bispecific antigen-binding molecules that bind cd3 and cd20, and uses thereof
WO2014163684A1 (en) * 2013-04-03 2014-10-09 Ibc Pharmaceuticals, Inc. Combination therapy for inducing immune response to disease
WO2017053856A1 (en) * 2015-09-23 2017-03-30 Regeneron Pharmaceuticals, Inc. Optimized anti-cd3 bispecific antibodies and uses thereof
WO2019045856A1 (en) * 2017-08-28 2019-03-07 Systimmune, Inc. Anti-cd3 antibodies and methods of making and using thereof
WO2021104371A1 (en) * 2019-11-26 2021-06-03 Shanghai Epimab Biotherapeutics Co., Ltd. Antibodies to cd3 and bcma, and bispecific binding proteins made therefrom

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014047231A1 (en) * 2012-09-21 2014-03-27 Regeneron Pharmaceuticals, Inc. Anti-cd3 antibodies, bispecific antigen-binding molecules that bind cd3 and cd20, and uses thereof
WO2014163684A1 (en) * 2013-04-03 2014-10-09 Ibc Pharmaceuticals, Inc. Combination therapy for inducing immune response to disease
WO2017053856A1 (en) * 2015-09-23 2017-03-30 Regeneron Pharmaceuticals, Inc. Optimized anti-cd3 bispecific antibodies and uses thereof
WO2019045856A1 (en) * 2017-08-28 2019-03-07 Systimmune, Inc. Anti-cd3 antibodies and methods of making and using thereof
WO2021104371A1 (en) * 2019-11-26 2021-06-03 Shanghai Epimab Biotherapeutics Co., Ltd. Antibodies to cd3 and bcma, and bispecific binding proteins made therefrom

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ROOT, A.R. ET AL.: "Discovery and optimization of a novel anti-GUCY2c x CD3 bispecific antibody for the treatment of solid tumors", MABS, vol. 13, no. 1, 31 December 2021 (2021-12-31), XP055875731, DOI: 10.1080/19420862.2020.1850395 *

Similar Documents

Publication Publication Date Title
JP7317272B2 (en) TIGIT Antibodies, Antigen-Binding Fragments Thereof, and Medical Uses Thereof This application is based on and claims priority from Application No. CN201710908565.3 filed on September 29, 2019. The disclosure of which is incorporated herein by reference in its entirety.
KR102522693B1 (en) Novel monoclonal antibodies to cytotoxic t-lymphocyte-associated protein 4 (ctla-4)
CN109843927B (en) anti-B7-H3 antibodies, antigen binding fragments thereof, and medical uses thereof
JP2023503180A (en) Anti-human claudin-18.2 antibody and its application
KR20210142638A (en) CD3 antigen-binding fragment and applications thereof
CA3019740A1 (en) Administration of a bispecific construct binding to cd33 and cd3 for use in a method for the treatment of myeloid leukemia
WO2021098822A1 (en) Bispecific antibodies
WO2023125888A1 (en) Gprc5d antibody and application thereof
WO2022105811A1 (en) Humanized cd19 antibody and use thereof
CN110461874B (en) anti-GITR antibodies, antigen binding fragments thereof, and medical uses thereof
CA3231761A1 (en) Anti-human cd3 antibody and use thereof
CN117616048A (en) CD19 antibodies and uses thereof
WO2024012513A1 (en) Antibody, antigen-binding fragment thereof, and pharmaceutical use thereof
CN116761889A (en) Antigen binding proteins and uses thereof
CN109593135B (en) Anti-human PD-L1 monoclonal antibody and application thereof
CN114957468A (en) anti-Siglec 15 antibody and application thereof
WO2024012434A1 (en) Antibody, antigen-binding fragment thereof, and pharmaceutical use thereof
TW202413405A (en) Antibody, antigen-binding fragment thereof, and pharmaceutical use thereof
WO2024094151A1 (en) Multi-specific antibody and medical use thereof
CN115947855B (en) Preparation of anti-CD 24 antibodies and uses thereof
WO2024088386A1 (en) Antibody, antigen-binding fragment thereof, and pharmaceutical use thereof
WO2023036215A1 (en) Bispecific antigen binding molecule and use thereof
RU2817143C2 (en) Anti-cd79b antibody, antigen-binding fragment thereof and pharmaceutical use thereof
US20230159652A1 (en) Transferrin receptor 1 targeting for carcinogenesis prevention
WO2023179716A1 (en) Anti-cd3 antibody, preparation method therefor and use thereof

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: 23839003

Country of ref document: EP

Kind code of ref document: A1