ANTIBODY OR CHIMERIC ANTIGEN RECEPTOR WHICH TARGETS CLAUDIN 18.2

Abstract
Disclosed are an antibody or an antigen binding fragment or chimeric antigen receptor thereof which binds to Claudin18.2, and a preparation method and a use. The chimeric antigen receptor sequentially comprises the antibody or the antigen binding fragment thereof which binds to the Claudin18.2 antigen, an extracellular hinge region, a transmembrane region and an intracellular signaling region. The antibody or the antigen binding fragment or chimeric antigen receptor thereof has stronger affinity and killing capability for cells secreting Claudin18.2, and a better tumor inhibiting effects.
Description
TECHNICAL FIELD

The present invention relates to the technical field of biomedicine or biopharmaceuticals, and specifically, relates to an antibody or an antigen-binding fragment thereof, or a chimeric antigen receptor that targets Claudin18.2, as well as a preparation method and use of the same for preparing a pharmaceutical composition, treating, preventing, detecting or diagnosing a disease.


BACKGROUND

Claudin18.2 is transiently expressed in gastric epithelial cells only, and is seldom expressed in other normal tissues. However, the expression of Claudin18.2 is abnormally elevated in many cancerous tissues (Niimi, Mol. Cell Biol., 21:7380-90, 2001). Claudin18.2 is expressed in gastric cancer, esophageal cancer, pancreatic cancer, lung cancer, ovarian cancer and other tumors. Antibodies targeting Claudin18.2 can mediate specific lysis of tumor cells through ADCC, CDC, inducing apoptosis and direct inhibition of proliferation. Therefore, Claudin18.2 is currently the most promising target in treatment of gastric cancer, esophageal cancer, pancreatic cancer, lung cancer and ovarian cancer.


Claudin18.1 is selectively expressed in epithelial cells of normal lungs and stomachs. The presence of two different variants introduces more complexity to Claudin18 molecules (Niimi, Mol. Cell Biol., 21:7380-90, 2001). How to further improve the effectiveness and safety is an issue to be considered in the field.


IMAB362 developed by Ganymed (Chinese Patent No. CN201380026898.3) is one of the first Claudin18.2 antibodies put into clinical trials. In its Phase II clinical trials for gastric cancer, the antibody used in combination with chemotherapy significantly prolonged the survival (13.2 vs. 8.4 months) compared with standard chemotherapy, and had more significant efficacy and a longer median survival time (16.7 months) in patients with high expression of Claudin18.2 (NCT01630083).


The present invention provides an antibody or an antigen-binding fragment thereof, or a chimeric antigen receptor T cell that targets Claudin18.2 with good efficacy for Claudin18.2-positive tumors, bringing new hope to patients with advanced gastric cancer, pancreatic cancer, and the like.


SUMMARY

All embodiments concerning VL (light chain variable region), VH (heavy chain variable region), LCDR (light chain complementarity determining region), HCDR (heavy chain complementarity determining region), LCDR1, LCDR2, LCDR3, HCDR1, HCDR2, and HCDR3 throughout the present invention may be implemented alone or in any combination.


In one aspect, the present invention provides an antibody or an antigen-binding fragment thereof, wherein:


in some embodiments, the antibody or the antigen-binding fragment thereof comprises any of the following combinations:


(1) three light chain complementarity determining regions comprising an LCDR1 amino acid sequence set forth in SEQ ID NO: 5, an LCDR2 amino acid sequence set forth in SEQ ID NO: 6 and an LCDR3 amino acid sequence set forth in SEQ ID NO: 7, and/or three heavy chain complementarity determining regions comprising an HCDR1 amino acid sequence set forth in SEQ ID NO: 8, an HCDR2 amino acid sequence set forth in SEQ ID NO: 9 and an HCDR3 amino acid sequence set forth in SEQ ID NO: 10;


(2) three light chain complementarity determining regions comprising an LCDR1 amino acid sequence set forth in SEQ ID NO: 11, an LCDR2 amino acid sequence set forth in SEQ ID NO: 12 and an LCDR3 amino acid sequence set forth in SEQ ID NO: 13, and/or three heavy chain complementarity determining regions comprising an HCDR1 amino acid sequence set forth in SEQ ID NO: 14, an HCDR2 amino acid sequence set forth in SEQ ID NO: 15 and an HCDR3 amino acid sequence set forth in SEQ ID NO: 16;


in some embodiments, the antibody or the antigen-binding fragment thereof comprises any of the following combinations:


(1) a light chain variable region of an amino acid sequence set forth in SEQ ID NO: 1 and/or a heavy chain variable region of an amino acid sequence set forth in SEQ ID NO: 2;


(2) a light chain variable region of an amino acid sequence set forth in SEQ ID NO: 3 and/or a heavy chain variable region of an amino acid sequence set forth in SEQ ID NO: 4.


In one aspect of the present invention, the antibody or the antigen-binding fragment thereof includes monoclonal antibodies, polyclonal antibodies, chimeric antibodies, humanized antibodies, Fab, Fab′, F(ab′)2, Fv, scFv or dsFv fragments.


In one aspect of the present invention, the antibody or the antigen-binding fragment thereof comprises a heavy chain constant region of an amino acid sequence set forth in SEQ ID NO: 17.


In one aspect of the present invention, the antibody or the antigen-binding fragment thereof comprises a heavy chain constant region of an amino acid sequence set forth in SEQ ID NO: 18.


In one aspect of the present invention, the antibody or the antigen-binding fragment thereof comprises a light chain constant region of an amino acid sequence set forth in SEQ ID NO: 28.


The antibody or the antigen-binding fragment disclosed herein has one or more of the following advantages: higher affinity to cells expressing Claudin18.2, enhanced ability to mediate ADCC, and better tumor inhibitory effect.


In one aspect of the present invention, any of the above antibodies or the antigen-binding fragments thereof binds to Claudin18.2.


The present invention also relates to a chimeric antigen receptor comprising the antibody or the antigen-binding fragment thereof, a related CAR-T cell, and a preparation method and use of the same.


Specifically, in one aspect, the present invention relates to a chimeric antigen receptor (CAR) comprising any of the above antibodies or the antigen-binding fragments thereof, wherein three light chain complementarity determining regions of the antibody or the antigen-binding fragment thereof comprise an LCDR1 amino acid sequence set forth in SEQ ID NO: 5, an LCDR2 amino acid sequence set forth in SEQ ID NO: 6 and an LCDR3 amino acid sequence set forth in SEQ ID NO: 7; and three heavy chain complementarity determining regions of the antibody or the antigen-binding fragment thereof comprise an HCDR1 amino acid sequence set forth in SEQ ID NO: 8, an HCDR2 amino acid sequence set forth in SEQ ID NO: 9 and an HCDR3 amino acid sequence set forth in SEQ ID NO: 10.


In another aspect, the present invention relates to a chimeric antigen receptor (CAR) comprising an antibody or an antigen-binding fragment thereof, wherein three light chain complementarity determining regions of the antibody or the antigen-binding fragment thereof comprise an LCDR1 amino acid sequence set forth in SEQ ID NO: 11, an LCDR2 amino acid sequence set forth in SEQ ID NO: 12 and an LCDR3 amino acid sequence set forth in SEQ ID NO: 13; and three heavy chain complementarity determining regions of the antibody or the antigen-binding fragment thereof comprise an HCDR1 amino acid sequence set forth in SEQ ID NO: 14, an HCDR2 amino acid sequence set forth in SEQ ID NO: 15 and an HCDR3 amino acid sequence set forth in SEQ ID NO: 16.


In another aspect, the present invention relates to a chimeric antigen receptor, wherein the sequence of the VL of the antibody or the antigen-binding fragment thereof is SEQ ID NO: 1, and the sequence of the VH is SEQ ID NO: 2.


In another aspect, the present invention relates to a chimeric antigen receptor, wherein the sequence of the VL of the antibody or the antigen-binding fragment thereof is SEQ ID NO: 3, and the sequence of the VH is SEQ ID NO: 4.


In another aspect, the VH and the VL of the antibody or the antigen-binding fragment thereof are linked through a linker; preferably, through a GGGGSGGGGSGGGGS linker; preferably, in the order of VH-GGGGSGGGGSGGGGS-VL from N terminus to C terminus.


In another aspect, the present invention relates to a chimeric antigen receptor which sequentially comprises the antibody or the antigen-binding fragment thereof according to any of the preceding aspects, an extracellular hinge region, a transmembrane region and an intracellular signaling region.


In another aspect, the present invention relates to a chimeric antigen receptor, of which the antibody or the antigen-binding fragment thereof is directed by a signal peptide.


In another aspect, the present invention relates to a chimeric antigen receptor, wherein the signal peptide may be a CD8α signal peptide, a VH3 signal peptide, an IL2 signal peptide or the like, the extracellular hinge region may be a CD8 hinge region, a CD28 hinge region or the like, the transmembrane region may be a CD8 transmembrane region, a CD28 transmembrane region, a 4-1BB transmembrane region or the like, and the intracellular signaling region may be a CD28 signaling region, a 4-1BB signaling region, an OX40 signaling region, a CD3ζ signaling region or the like.


In another aspect, the present invention relates to a chimeric antigen receptor, wherein the extracellular hinge region is a CD8 hinge region, the transmembrane region is a CD8 transmembrane region, the intracellular signaling region is 4-1BB and CD3ζ, and the antibody or the antigen-binding fragment thereof is directed by a CD8α signal peptide. Preferably, the CD8α signal peptide is a CD8α signal peptide set forth in SEQ ID NO: 21, the extracellular hinge region is a CD8 hinge region set forth in SEQ ID NO: 22, the transmembrane region is a CD8 transmembrane region set forth in SEQ ID NO: 23, and the intracellular signaling region is 4-1BB set forth in SEQ ID NO: 24 and CD3ζ set forth in SEQ ID NO: 25.


In another aspect, the present invention relates to a nucleic acid encoding the antibody or the antigen-binding fragment thereof, or the chimeric antigen receptor according to any of the preceding aspects.


In another aspect, the present invention relates to a vector comprising the nucleic acid according to the previous aspect, or expressing the antibody or the antigen-binding fragment thereof or the chimeric antigen receptor according to any of the preceding aspects. Preferably, the vector may be a viral vector; preferably, the viral vector includes, but is not limited to, a lentivirus vector, an adenovirus vector, an adeno-associated virus vector or a retrovirus vector; preferably, the vector may be a non-viral vector; preferably, the non-viral vector may be a transposon vector; preferably, the transposon vector may be a Sleeping Beauty vector, a PiggyBac vector, or the like; preferably, the vector may be a mammalian expression vector; preferably, the expression vector may be a bacterial expression vector; preferably, the expression vector may be a fungal expression vector.


In another aspect, the vector is a lentivirus vector.


In another aspect, the lentivirus vector is plasmid pRRLSIN-Claudin18.2CAR-P2A-EGFRt shown in FIG. 11.


In another aspect, the vector is a PiggyBac (PB) transposon vector.


In another aspect, the PB transposon vector is plasmid PB CN02 CAR shown in FIG. 24.


In another aspect, the present invention relates to a cell expressing the antibody or the antigen-binding fragment thereof or the chimeric antigen receptor according to any of the preceding aspects. Preferably, the cell is a bacterial cell; preferably, the bacterial cell is an Escherichia coli cell or the like; preferably, the cell is a fungal cell; preferably, the fungal cell is a yeast cell; preferably, the yeast cell is a Pichia pastoris cell or the like; preferably, the cell is a mammalian cell; and preferably, the mammalian cell is a Chinese hamster ovary (CHO) cell, a human embryonic kidney cell (293), a B cell, a T cell, a DC cell, a NK cell, or the like.


In another aspect, the present invention relates to a CAR-T cell comprising the chimeric antigen receptor according to any of the preceding aspects.


In another aspect, the present invention relates to a method for preparing the CAR-T cell according to the previous aspect, comprising transfecting a T cell with a vector comprising a nucleic acid encoding the chimeric antigen receptor according to any of the preceding aspects. In a preferred embodiment, the vector is a non-viral vector. In a preferred embodiment, the vector is a PB transposon vector. In a preferred embodiment, the PB transposon vector is plasmid PB CN02 CAR shown in FIG. 24.


In another aspect, the present invention relates to a method for preparing the CAR-T cell according to the previous aspect, comprising transfecting a T cell with a vector comprising a nucleic acid encoding a transposase. In another preferred embodiment, the transposase is PB transposase.


In another aspect, the present invention relates to a method for preparing the CAR-T cell according to the previous aspect, comprising transfecting a T cell with a transposon vector comprising a nucleic acid encoding the chimeric antigen receptor according to any of the preceding aspects and a transposase vector comprising a nucleic acid encoding a transposase. In a preferred embodiment, the transposon vector is a PB transposon vector. In a preferred embodiment, the PB transposon vector is plasmid PB CN02 CAR shown in FIG. 24. In a preferred embodiment, the transposase is PB transposase.


In another aspect, the present invention relates to a method for preparing the CAR-T cell according to the previous aspect, comprising transducing a T cell with a lentivirus comprising the chimeric antigen receptor vector according to any of the preceding aspects to give the CAR-T cell.


In another aspect, the present invention relates to a pharmaceutical composition comprising the CAR-T cell according to any of the preceding aspects.


In another aspect, the present invention relates to a method for treating cancer, comprising administering the CAR-T cell according to any of the preceding aspects to a subject in need.


In another aspect, the present invention relates to use of the CAR-T cell according to any of the preceding aspects in treating cancer.


In another aspect, the present invention relates to use of the CAR-T cell according to any of the preceding aspects in preparing a pharmaceutical composition for treating cancer.


In another aspect, the present invention relates to a CAR-T cell having one or more of the following advantages: good killing ability to cells expressing Claudin18.2; and low killing ability to cells expressing Claudin18.1.


In one aspect, the present invention provides a pharmaceutical composition comprising: the antibody or the antigen-binding fragment thereof, the chimeric antigen receptor, the nucleic acid encoding the same, or the cell expressing the same disclosed herein; and a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier includes one or more of the following: pharmaceutically acceptable vehicle, disperser, additive, plasticizer, and excipient.


In one aspect, the present invention provides a kit comprising the antibody or the antigen-binding fragment thereof, a chimeric antigen receptor, or the nucleic acid encoding the same disclosed herein.


In some embodiments, the pharmaceutical composition may also comprise other therapeutic agents. In some embodiments, other therapeutic agents include chemotherapeutic agents, immunotherapeutic agents, or hormone therapeutic agents. The antibody or the antigen-binding fragment can be used in combination with other therapeutic agents to enhance the efficacy.


In some embodiments, “to enhance the efficacy” refers to enhancing the efficacy of other therapeutic agents or modalities. The antibody or the antigen-binding fragment disclosed herein can be administered alone or in combination with other therapeutic agents or modalities. In some embodiments, other therapeutic agents or modalities include chemotherapeutic agents, immunotherapeutic agents, hormone therapeutic agents, radiotherapy and surgery.


In another aspect, the present invention relates to use of the antibody or the antigen-binding fragment thereof, the chimeric antigen receptor, the nucleic acid, the vector or the cell according to any of the preceding aspects in preparing a pharmaceutical composition for treating or preventing a disease.


In another aspect, the present invention relates to use of the antibody or the antigen-binding fragment thereof, the chimeric antigen receptor, or the nucleic acid according to any of the preceding aspects in preparing a kit for diagnosis or detection.


In another aspect, a method for treating or preventing a disease is provided, comprising administering the antibody or the antigen-binding fragment, the chimeric antigen receptor, the nucleic acid, the vector, the cell, or the pharmaceutical composition disclosed herein to a subject in need.


In another aspect, a method for diagnosis or detection is provided, comprising administering the antibody or the antigen-binding fragment, the chimeric antigen receptor, the nucleic acid, or the kit disclosed herein to a subject in need or a sample.


In another aspect, the present invention provides use of the antibody or the antigen-binding fragment thereof, the chimeric antigen receptor, the nucleic acid, the vector, the cell, or the pharmaceutical composition according to any of the preceding aspects for treating or preventing a disease.


In another aspect, the present invention provides use of the antibody or the antigen-binding fragment thereof, the chimeric antigen receptor, the nucleic acid, or the kit according to any of the preceding aspects for detection or diagnosis.


In another aspect, the disease is a cancer.


In another aspect, the cancer is a Claudin18.2-positive cancer.


In another aspect, the cancer includes gastric cancer, pancreatic cancer, esophageal cancer, lung cancer, ovarian cancer, head and neck cancer, bladder cancer, cervical cancer, sarcoma, cytoma, colon cancer, kidney cancer, colorectal cancer, liver cancer, melanoma, breast cancer, myeloma, neuroglioma, leukemia, lymphoma, and the like.





DRAWINGS


FIGS. 1-2 show the binding sensitivity of candidate antibodies to Claudin18.2 protein measured by ELISA.



FIGS. 3A-3B show the binding of candidate antibodies to 293T-Claudin18.2 cells measured by flow cytometry.



FIGS. 4A-4B show the binding of candidate antibodies to 293T-Claudin18.1 cells measured by flow cytometry.



FIGS. 5A-5B show the binding of candidate antibodies to NUGC4 cells measured by flow cytometry.



FIGS. 6A-6B show the ADCC of candidate antibodies on 293T-Claudin18.2 cells.



FIGS. 7A-7B show the ADCC of candidate antibodies on NUGC4 cells.



FIG. 8 shows the ADCC of candidate antibodies on 293T-Claudin18.1 cells.



FIG. 9 shows the pharmacodynamics of candidate antibodies.



FIG. 10 shows a structural schematic of plasmid pRRLSIN-EGFRt.



FIG. 11 shows a structural schematic of recombinant plasmid pRRLSIN-Claudin18.2CAR-P2A-EGFRt.



FIG. 12 shows the lentivirus activity titers of different chimeric antigen receptors.



FIG. 13 shows the positive rates of T lymphocytes expressing different chimeric antigen receptors.



FIG. 14 shows the specific lysis assay of Claudin18.2 CAR-T cells with different scFvs in vitro.



FIG. 15 shows the release of IFN-gamma cytokines in supernatant of co-incubated Claudin18.2 CAR-T cells with different scFvs and different 293T cells.



FIG. 16 shows the specific lysis assay of Claudin18.2 CAR-T cells with different scFvs against Claudin18.2-positive tumor cells.



FIG. 17 shows the release of IFN-gamma cytokines in supernatant of co-incubated Claudin18.2 CAR-T cells with different scFvs and Claudin18.2-positive tumor cells.



FIG. 18 shows the tumor volumes in female NOG mice bearing NUGC4-Claudin18.2 tumor cells after receiving Claudin18.2 CAR-T cells.



FIG. 19 shows the body weight of female NOG mice bearing NUGC4-Claudin18.2 tumor cells after receiving Claudin18.2 CAR-T cells.



FIG. 20 shows the tumor growth inhibition (TGI) in female NOG mice bearing NUGC4-Claudin18.2 tumor cells after receiving Claudin18.2 CAR-T cells.



FIG. 21 shows the survival rate of female NOG mice bearing NUGC4-Claudin18.2 tumor cells after receiving Claudin18.2 CAR-T cells.



FIG. 22 shows the positive rates of T lymphocytes expressing different chimeric antigen receptors.



FIG. 23 shows the in vitro specific lysis assay of CN02 CAR-T cells prepared by different methods.



FIG. 24 shows the structural schematic of plasmid PB CN02 CAR





DETAILED DESCRIPTION

The present invention will be further described in conjunction with the following specific examples. The examples described herein are only some examples of the present invention, but not all examples. It should be understood that the following examples are given to provide those of ordinary skill in the art a complete disclosure and description of how to utilize the methods and the compositions, but are not intended to limit the scope of the present invention. Based on the examples of the present invention, all other examples obtained by those of ordinary skill in the art without creative work shall fall within the protection scope of the present invention.


Example 1. Production of Anti-Claudin18.2 Monoclonal Antibody
1.1 Immunization

BoAn-hMab transgenic mice from Shandong BoAn Biotechnology Co. Ltd. (prepared according to the method described in Chinese Patent No. CN103571872B) were immunized with plasmids (KYinno) containing Claudin18.2 genes and CHO cells (KYinno) stably expressing Claudin18.2 protein. Plasmids were used for the first immunization, and the second to the seventh immunizations were conducted using plasmids and cells alternately. A total of 10 mice were immunized. Five mice with higher serum concentration were selected for booster immunization, and the mice were euthanized 4 days later. Spleens were processed and frozen for later use.


1.2 Construction of Phage Library

Spleen cells of the immunized mice were added with Trizol (Thermo Scientific, catalog number: 15596-026) for complete lysis, and then added with a 1/5 volume of chloroform and mixed well. The mixture was incubated at room temperature for 20 min, and centrifuged at 12000 rpm at 4° C. for 20 min. The supernatant was added with an equal volume of isopropanol. The resulting mixture was incubated at room temperature for 20 min and then centrifuged at 12000 rpm at 4° C. for 20 min. The supernatant was discarded, and the precipitate was washed with 75% ethanol twice and then centrifuged at 12000 rpm at 4° C. for 5 min. The supernatant was discarded, and the precipitate was dried at room temperature and then resuspended with DEPC water to give RNA, which was then reversely transcribed into cDNA using a Roche reverse transcription kit Transcriptor First Strand cDNA Synthesis Kit as per the instructions (Roche Applied Science, catalog number: 4897030001).


The phage library was constructed by the method described in Carlos F. Barbas III, Phage display: A Laboratory Manual. Sequences of variable regions of heavy and light chains were obtained from cDNA by PCR, and then subjected to overlap extension PCR to give scFv sequence. Then, the scFv was digested with SfiI enzyme (NEB, catalog number: R0123L) for 5 h (50° C.) and ligated with plasmid pCOMB3× (Biovector Science Lab, Inc., BIOVECTOR510837) through T4 DNA ligase (Sino Biological Inc.). The ligation product was electrotransfected into competent Escherichia coli TG1 cells (Lucigen, catalog number: A96595-2), which were then cultivated on a shaker at 220 rpm at 37° C. and infected with a phage, and the supernatant of the culture was collected, concentrated and purified to give the phage library.


1.3 Screening

1.3.1 Plate screening: A plate was coated with Claudin18.2 protein (Genscript Biotech) at 0.3 μg/well, and incubated overnight at 4° C. The plate was blocked with 2% BSA for 1 h the next day, and the phage library (2×1012) was added for a 2-h incubation. After 4-10 washings, the phages specifically binding to Claudin18.2 were eluted with an eluent buffer (pH 2.2) (4.2 mL of concentrated hydrochloric acid (Tianjin Kemiou Chemical Reagent Co., Ltd.) was added to 500 mL of ultrapure water, and the mixture was adjusted to pH 2.2 with glycine powder (Biotopped, BG0617-500)).


1.3.2 Cell screening: The phage library (2×1012) was rotationally mixed with 293T-Claudin18.1 cells (3×106 cells/vial) at room temperature and incubated for 1 h. The resulting mixture was blocked with 2% BSA for 1 h, then rotationally mixed with 293T-Claudin18.2 cells (2×106 cells/vial) at room temperature and incubated for 2 h. After 4-10 washings, the phages specifically binding to Claudin18.2 were eluted with an eluent buffer (pH 2.2). Phages passing the cell screening could be further screened on plates.


Example 2. Construction and Production of Complete Antibodies

Clones CLD387-C115, CLD389-C279\CA802\CA852, CLDQMix-CA808.1\CA811\CA818, CLDQ1-CA841\CA843, CLD393-C1002\C1024 were sequenced by Invitrogen Biotechnology Co., Ltd. The amino acid sequences of the variable regions of the clones are shown in Table 1.









TABLE 1







Amino acid sequences of variable regions of active clones









Clone ID
VL sequence
VH sequence





CA808.1
DIVMTQSPSSLSASVGDRVTITCRA
EVQLVQSGGGVVQPGRSLRLSCAAS



SQGIRNDLGWYQQKPGKAPKLLIY

GFTFSSYAMHWVRQAPGKGLEWVA





AASSLQSGVPSRFSGSGSGTDFTLTI

VISYDGSNKYYADSVKGRFTISRDNS



SSLQPEDFATYYCLQDYNYPRTFG
KNTLYLQMNSLRAEDTAVYYCAREL



QGTKVEIK (SEQ ID NO: 1)

RFFDWLLGSAFDIWGQGTTVTVSS




CDR
(SEQ ID NO: 2)



LCDR1: QGIRND (SEQ ID NO: 5)
CDR



LCDR2: AAS (SEQ ID NO: 6)
HCDR1: GFTFSSYA (SEQ ID NO: 8)



LCDR3: LQDYNYPRT (SEQ ID
HCDR2: ISYDGSNK (SEQ ID NO: 9)



NO: 7)
HCDR3: ARELRFFDWLLGSAFDI




(SEQ ID NO: 10)





CA841
DIQMTQSPSTLSASVGDRVTITCRA
QVQLVQSGAEVKKPGSSVKVSCKAS



SQSINSWLAWYQQKPGKAPKLLIY

GGTFSSYAISWVRQAPGQGLEWMGR





KASSLESGVPSRFSGSGSGTEFTLTI


IIPILGIANYAQKFQGRVTVTADKSTS




SSLQPDDFATYYCQQYNSFPLTFG
TAYMELSSLRSEDTAVYYCARERDN



GGTKVEIK (SEQ ID NO: 3)

WDPYYYYYGMDVWGQGTTVTVSS




CDR
(SEQ ID NO: 4)



LCDR1: QSINSW (SEQ ID NO: 11)
CDR



LCDR2: KAS (SEQ ID NO: 12)
HCDR1: GGTFSSYA (SEQ ID NO: 14)



LCDR3: QQYNSFPLT (SEQ ID
HCDR2: IIPILGIA (SEQ ID NO: 15)



NO: 13)
HCDR3: ARERDNWDPYYYYYGMDV




(SEQ ID NO: 16)





C115
DIVMTQSPSSLSASVGDRVTITCRA
QVQLVESGGGVVQPGRSLRLSCAAS



SQGIRNDLGWYQQKPGKAPKLLIY

GFTFSSYAMHWVRQVPGKGLEWVAI





AASSLQSGVPSRFSGSGSGTDFTLTI


ISYDGSIKYYADSVKGQFTISRDNSK




SSLQPEDFATYYCLQDYNFPLTFGQ
NTLSLQMNSLRPEDTAVYYCARDPT



GTKVEIK

MVRGVRGMDVWGQGTTVTVSS






C279
EIVMTQSPPSLSASVGDRVTITCRA
EVQLVQSGGGVVQPGRSLRLSCAAS



SQGIRNDLGWYQQKPGKAPKLLIY

GFTFSNYAMHWVRQAPGKGLEWVA





AASSLQSGVPSRFSGSGSGTDFTLTI

VISYDGSNKYYADSVKGRFTISRDNS



SSLQPEDFATYYCLQDYNYPWTFG
KNTLYLQMNSLRAEDTAVYYCAREL



QGTKLEIK

RFFDWLLGRAFDIWGQGTMVTVSS






CA802
DIQMTQSPSSLSASVGDRVTITCRA
QVQLVESGGGVVQPGRSLRLSCAAS



SQGIRNDLGWYQQKPGKAPKLLIY

GFTFSSYAMHWVRQVPGKGLEWVAI





AASSLQSGVPSRFSGSGSGTDFTLTI


ISYDGSIKYYADSVKGQFTISRDNSK




SSLQPEDFATYYCLQDYNYPLTFG
NTLSLQMNSLRPEDTAVYYCARDPT



GGTKLEIK

MVRGVRGMDVWGQGTTVTVSS






CA811
DIVMTQSPSSLSASVGDRVTITCRA
QVQLVESGGGVVQPGRSLRLSCAAS



SQGIRNDLGWYQQKPGKAPKLLIY

GFTFSSYAMHWVRQAPGKGLEWVA





KASSLESGVPSRFSGSGSGTDFTLTI

VISYDGSNKYYADSVKGRFTISRDNS



SSLQPEDFATYYCLQDYNYPLTFG
KNTLYLQMNSLRAEDTAVYYCARDP



GGTKLEIK

TMVRGVRGMDVWGQGTTVTVSS






CA818
DIVMTQSPSTLSASVGDRVTITCRA
GAATAVGRRTVEASETLSLTCAVYG



SQGIRNDLGWYQQKPGKAPKLLIY

GSFSGYYWSWIRQPPGKGLEWIGEIN





DASSLQSGVPSRFSGSGYGTDFTLT


HSGSTNYNPSLKSRVTISVDTSKNQFS




ISSLQPEDFATYYCLQDYNYPWTF
LKLSSVTAADTAVYYCARGPYDILTG



GQGTKLEIK

YYPYWYFDLWGRGTLVTVSS






CA843
DIVMTQSPSSLSASVGDRVTITCRA
QVQLVESGGGVVQPGRSLRLSCAAS



SQGIRNDLGWYQQKPGKAPKLLIY

GFTFSSYAMHWVRQAPGKGLEWVA





AASSLQSGVPSRFSGSGSGTDFTLTI

VISYDGSNKYYADSVKGRFTISRDNS



SSLQPEDFATYYCLQDYNYPLTFG
KNTLYLQMNSLRAEDTAVYYCAREG



GGTKVEIK

IPVRGFDYWGQGTLVTVSS






CA852
EIVMTQSPSTLSASIGDRVTITCRAS
QVQLVESGGGVVQPGRSLRLSCAAS




QGIRNDLGWYQQKPGKAPKLLIYA


GFTFSSYAMHWVRQAPGKGLEWVA





ASSLQSGVPSRFSGSGSGTDFTLTIS

VISYDGSKKYYADSVKGRFTISRDNS



SLQPEDFATYYCLQDYNYPLTFGG
KNTLYLQMNSLRAEDTAVYYCARDP



GTKVEIK

TMVRGVRGMDVWGQGTTVTVSS






C1002
ETTLTQSPDSLAVSLGERATINCKS
QVQLVESGGGVVQPGRSLRLSCAAS



SQSVLYSSNNKNYLAWYQQKPGQ

GFTFSSHDMHWVRQAPGKGLEWVA




PPKLLIYWASTRESGVPDRFSGSGS
DISDDGSNKNYADSVKGRFTISRDNS



GTDFTLTISSLQTEDVAVYYCQQY
KNTLYLQMNSLRAEDAAVYYCARE




YSTPYTFGQGTKVDIK


GLRWFGEFYYSYGMDVWGQGTTVT




VSS






C1024
DIVMTQSPDSLAVSLGERATINCKS
EVQLVQSGGGVVQPGRSLRLSCAAS



SQSVLYSSNNKNYLAWYQQKPGQ

GFTFSSYDMHWVRQAPGKGLEWVA




PPKLLIYWASTRESGVPDRFSGSGS
DISDDGSNKNYADSVKGRFTISRDNS



GTDFTLTISSLQAEDVAVYYCQQY
KNTLYLQMNSLRAEDAAVYYCARE




DSTPYTFGQGTKLEIK


GLRWFGEFYYSYGMDVWGQGTTVT





VSS









Through methods such as variable region gene amplification (2*Phanta Max Master Mix, manufacturer: Vazyme, catalog number: P515-AA, batch number: 7E211 GB), signal peptide and variable region overlap extension, and homologous recombination (ClonExpress II One Step Cloning Kit, manufacturer: Vazyme, catalog number: C112-01, batch number: 7E211L8), nucleotide sequence fragments encoding VH or VL were respectively inserted into vectors pCDNA3.4 (Life Technology) containing a nucleotide sequence encoding a heavy chain constant region (SEQ ID NO: 17) and pCDNA3.4 (Life Technology) containing a nucleotide sequence encoding a light chain constant region (SEQ ID NO: 28). Then the vectors were transfected into HEK293 cells and incubated on a shaker at 37° C./8% CO2/125 rpm. After 6-7 days of transient expression the supernatant was purified by Protein A affinity chromatography to give Claudin18.2 antibodies, and the antibody concentration was determined by the extinction coefficient at UV280.


Antibody IMAB362 was selected as the reference antibody. In the Phase II clinical trial for gastric cancer, the antibody in combination with chemotherapy significantly prolonged the survival (13.2 vs. 8.4 months) as compared with standard chemotherapy. IMAB362 had more significant efficacy and a longer median survival time (16.7 months) in patients with high expression of Claudin18.2. IMAB362 is also one of the first Claudin18.2 antibodies put into clinical trials.


Production of reference antibody: Amino acid sequence of Claudin18.2 antibody IMAB362 of Ganymed is available in IMGT database and the Patent No. CN201380026898, with the heavy chain and light chain sequences set forth in SEQ ID NOs: 19 and 20, respectively. The whole gene sequence was synthesized, inserted in vector pCDNA3.4 and expressed in HEK293 cells to produce an antibody named IMAB362.


Example 3. Characterization of Anti-Claudin18.2 Antibodies
3.1 Binding of Antibodies to Claudin18.2 Protein by ELISA

Plates were coated with Claudin8.2 antigen (Genscript Biotech) of different concentrations (0.2 μg/mL, 0.05 μg/mL, 0.0125 μg/mL) at 100 μL/well, incubated overnight at 4° C., and blocked with 3% skimmed milk powder at 37° C. for 1 h. 100 μL of candidate antibodies was added to each well at 1 μg/mL, and incubated at 37° C. for 1 h, followed by goat anti-human IgG/HRP and a 1-h incubation at 37° C. After a 10-min color development, OD450 was measured on a microplate reader. The results are shown in FIG. 1, FIG. 2, Table 2 and Table 3.









TABLE 2







Binding sensitivity of candidate antibodies to Claudin18.2 protein measured by ELSIA











Antigen
Antigen
Antigen



concentration
concentration
concentration


Antibody ID
(0.2 μg/mL)
(0.05 μg/mL)
(0.0125 μg/mL)













CLDQ1-CA843-IgG1
1.847
0.452
0.148


CLD387-115-IgG1
2.724
1.66
0.449


CLD389-279-IgG1
3.094
2.15
0.702


CLD389-CA802-IgG1
2.618
1.622
0.579


CLDMIX-CA808.1-IgG1
2.996
1.167
0.389


CLDMIX-CA811-IgG1
2.705
1.062
0.307


CLDQ1-CA841-IgG1
2.38
0.484
0.165


CLD389-CA852-IgG1
2.809
1.17
0.332


IMAB362
2.819
1.475
0.532
















TABLE 3







Binding sensitivity of candidate antibodies to Claudin18.2 protein measured by ELSIA











Antigen
Antigen
Antigen



concentration
concentration
concentration


Antibody ID
(0.2 μg/mL)
(0.05 μg/mL)
(0.0125 μg/mL)





CLD383-C1024-IgG1
0.212
0.109
0.084


CLD387-C1002-IgG1
0.187
0.105
0.076


CLDMix-CA818-IgG1
3.189
1.281
0.492


IMAB362
3.832
3.288
1.901









3.2 Binding of Antibodies to 293T-Claudin18.1/18.2 Cells and NUGC4 Cells Measured by Flow Cytometry

To a 96-well round-bottom plate, 50 μL of 293T-Claudin18.1 or 18.2 cells (KYinno) or NUGC4 cells were added at 1×105 cells/well. Each candidate antibody was serially diluted with FACS buffer (sterile PBS, 0.2% BSA), and added to the 96-well round-bottom plate at 50 μL/well before an incubation at 4° C. for 1 h. The supernatant was discarded after centrifugation at 2000 rpm for 3 min. Then the resulting cells were washed twice with FACS buffer, added with 100 μL/well of fluorescent secondary antibody (Southern Biotech, 2040-09), with a final concentration of 1 μg/mL, and incubated at 4° C. for 1 h before a centrifugation at 2000 rpm for 3 min. The supernatant was discarded, and the resulting cells were washed twice with FACS buffer, resuspended with 100 μL/well of FACS buffer, and analyzed by a flow cytometer (ACEA Pharma, NovoCyte 2060). The results are shown in FIGS. 3A-5B and Tables 4-9.


As shown in FIG. 3A, with 293T-Claudin18.2 cells being the target, the mean fluorescence intensity of candidate antibody CLDQMIX-CA808.1-IgG1 was higher than that of IMAB362 at concentrations of 3 μg/mL, 1.5 μg/mL, 0.75 μg/mL and 0.375 μg/mL. This indicates that the candidate antibody CLDQMIX-CA808.1-IgG1 has a higher affinity to 293T-Claudin18.2 cells at concentrations of 3 μg/mL, 1.5 μg/mL, 0.75 μg/mL and 0.375 μg/mL.


As shown in FIG. 5B, with NUGC4 cells being the target, the mean fluorescence intensity of candidate antibody CLDQMIX-CA808.1-IgG1 was higher than that of IMAB362 at concentrations of 3 μg/mL, 1.5 μg/mL, and 0.75 μg/mL. This indicates that the candidate antibody CLDQMIX-CA808.1-IgG1 has a higher affinity to NUGC4 cells expressing Claudin8.2 at concentrations of 3 μg/mL, 1.5 μg/mL, and 0.75 μg/mL.


As shown in FIG. 3A, with 293T-Claudin18.2 cells being the target, the mean fluorescence intensity of the candidate antibody CLDQ1-CA841-IgG6 was higher than that of IMAB362 at concentrations of 3 μg/mL and 1.5 μg/mL. This indicates that the candidate antibody CLDQ1-CA841-IgG1 has a higher affinity to 293T-Claudin18.2 cells at concentrations of 3 μg/mL and 1.5 μg/mL.


As shown in FIG. 4A, with 293T-Claudin18.1 cells being the target, the mean fluorescence intensities of the candidate antibodies CLDQMIX-CA808.1-IgG and CLDQ1-CA841-IgG were similar to that of IMAB362, which were all at lower levels at various concentrations.


The above results show that CLDQMIX-CA808.1-IgG9 and CLDQ1-CA841-IgG3 have better ability of binding cells expressing Claudin18.2, and have weak ability of binding Claudin18.1. It indicates that Claudin18.2 cells are more prone to binding, and specific binding to targets other than Claudin18.2 is less likely to occur in clinical applications, thereby achieving better pharmaceutical effects.









TABLE 4







Binding of candidate antibodies to 293T-Claudin18.2 cells measured by flow cytometry


(corresponding to FIG. 3A)












Antibody
Antibody
Antibody
Antibody



concentration
concentration
concentration
concentration


Antibody ID
(3 μg/mL)
(1.5 μg/mL)
(0.75 μg/mL)
(0.375 μg/mL)














CLD387-115-IgG1
952384
1002923
714249
501750


CLD389-279-IgG1
1239649
1057051
775956
487518


CLD389-CA802-IgG1
1226148
1049205
773824
494669


CLDQMIX-CA808. 1-IgG1
1382494
1196101
752733
535348


CLDMIX-CA811-IgG1
806980
657103
504375
324014


CLDQ1-CA841-IgG1
1092233
854886
583477
307641


CLDQ1-CA843-IgG1
609424
603959
399820
182880


CLD389-CA852-IgG1
741760
577688
364883
196431


IMAB362
902134
849017
688504
497940
















TABLE 5







Binding of candidate antibodies to 293T-Claudin18.2 cells measured by flow cytometry


(corresponding to FIG. 3B)












Antibody
Antibody
Antibody
Antibody



concentration
concentration
concentration
concentration


Antibody ID
(3 μg/mL)
(1.5 μg/mL)
(0.75 μg/mL)
(0.375 μg/mL)














CLDMix-CA818-IgG1
33274
34210
17288
9356


CLD393-C1002-IgG1
1326330
1156050
852105
405515


CLD393-C1024-IgG1
1374048
1030045
787017
308011


IMAB362
1037843
877912
649900
407382
















TABLE 6







Binding of candidate antibodies to 293T-Claudin18.1 cells measured by flow cytometry


(corresponding to FIG. 4A)












Antibody
Antibody
Antibody
Antibody



concentration
concentration
concentration
concentration


Antibody ID
(3 μg/mL)
(1.5 μg/mL)
(0.75 μg/mL)
(0.375 μg/mL)














CLD387-115-IgG1
124974
133585
78691
63446


CLD389-279-IgG1
128702
72429
52042
36901


CLD389-CA802-IgG1
193720
162744
140589
96678


CLDQMIX-CA808. 1-IgG1
47521
28885
29387
26004


CLDMIX-CA811-IgG1
114724
93551
40496
26022


CLDQ1-CA841-IgG1
10958
37855
18119
21489


CLDQ1-CA843-IgG1
86333
59051
35542
23218


CLD389-CA852-IgG1
70114
72110
40071
18876


IMAB362
35584
24896
31961
15638
















TABLE 7







Binding of candidate antibodies to 293T-Claudin18.1 cells measured by flow cytometry


(corresponding to FIG. 4B)












Antibody
Antibody
Antibody
Antibody



concentration
concentration
concentration
concentration


Antibody ID
(3 μg/mL)
(1.5 μg/mL)
(0.75 μg/mL)
(0.375 μg/mL)














CLDMix-CA818-IgG1
34418
17935
15184
7957


CLD393-C1002-IgG1
15767
8615
12343
6963


CLD393-C1024-IgG1
10099
17142
9851
7892


IMAB362
12483
17319
20666
12817
















TABLE 8







Binding of candidate antibodies to NUGC4 cells measured by flow cytometry


(corresponding to FIG. 5A)











Antibody
Antibody
Antibody



concentration
concentration
concentration


Antibody ID
(3 μg/mL)
(1.5 μg/mL)
(0.75 μg/mL)













CLD387-115-IgG1
201377
126584
101395


CLD389-279-IgG1
158296
78264
40032


CLD389-CA802-IgG1
113085
58148
24412


CLDMIX-CA811-IgG1
43084
26853
15543


CLDMix-CA818-IgG1
42617
20432
15029


CLDQ1-CA841-IgG1
32421
13208
11119


IMAB362
130923
97087
69564
















TABLE 9







Binding of candidate antibodies to NUGC4 cells measured by


flow cytometry (corresponding to FIG. 5B)











Antibody
Antibody
Antibody



concentration
concentration
concentration


Antibody ID
(3 μg/mL)
(1.5 μg/mL)
(0.75 μg/mL)













CLDQMIX-CA808.1-IgG1
185377
137723
75649


CLDQ1-CA843 -IgG1
13935
10612
7234


CLD389-CA852-IgG1
18351
11955
9229


CLD393-C1002-IgG1
5170
4891
4946


CLD393-C1024-IgG1
6141
5643
5210


IMAB362
116862
93509
55403









3.3 ADCC of Antibodies

Sterile fetal bovine serum was thawed and added to an RPMI1640 medium at a ratio of 1:99 to give an ADCC buffer. PBMC cells were thawed and incubated overnight in an incubator at 37° C./5% CO2. The density of target cells (293T-Claudin18.1 or 18.2) was adjusted with the ADCC Buffer to 2×105 cells/mL, and 50 μL of the target cells was added to each well of a 96-well round-bottom plate. The antibodies to be tested were diluted by 10× with the ADCC Buffer from 10 μg/mL or 50 μg/mL, then 50 μL of the diluted antibody was added to each well of the 96-well round-bottom plate coated with the target cells, and incubated in an incubator at 37° C./5% CO2 for 30-60 min. PBMC cells were collected and diluted with the ADCC buffer to a density from 2×106 cells/mL to 5×106 cells/mL, then 100 μL of the diluted cells was added to each well of the 96-well round-bottom plate coated with the target cells and the sample to be tested, and incubated in an incubator at 37° C./5% CO2 for 4-6 h. After incubation, the cells were centrifuged at 300 g for 2-5 min, then 50 μL of supernatant was carefully pipetted to a new 96-well flat-bottom plate, and 50 μL of LDH test solution (Promega, G1780) was added. The cells were then incubated in an incubator at 37° C./5% CO2 for 30 min. A terminating buffer was added after incubation. The OD value at 490 nm was measured by a microplate reader, with a background wavelength being 650 nm. The results are shown in FIGS. 6A-6B and Tables 10-11.


As shown in FIG. 6A, with 293T-Claudin18.2 as target and PBMC as effector cell, the inhibition rate of the candidate antibody CLDQMIX-CA808.1-IgG1 to the target cells at concentrations of 1 μg/mL and 0.1 μg/mL was higher than that of the reference antibody IMAB362 at the corresponding concentrations, suggesting that the candidate antibody CLDQMIX-CA808.1-IgG1 has a better ability of mediating ADCC than the reference antibody IMAB362 at the concentrations of 1 μg/mL and 0.1 μg/mL. This indicates that CLDQMIX-CA808.1-IgG1 can kill target cells expressing Claudin18.2 better and has better pharmaceutical effects.









TABLE 10







ADCC of candidate antibodies on 293T-Claudin18.2 cells


(corresponding to FIG. 6A)









Inhibition rate, %











Antibody
Antibody
Antibody



concentration
concentration
concentration


Antibody ID
(10 μg/mL)
(1 μg/mL)
(0.1 μg/mL)













CLD387-115-IgG1
15.48077
21.48077
12.28077


CLD389-279-IgG1
40.95
16.05
7.530769


CLD389-CA802-IgG1
61.21154
61.38846
25.60769


CLDQMIX-
57.43462
63.55769
39.42692


CA808.1-IgG1





CLDMIX-CA811-IgG1
63.24208
33.91539
18.17308


CLDQ1-CA841-IgG1
58.48615
27.23462
15.64231


CLDQ1-CA843-IgG1
53.19346
18.85385
−2.088462


CLD389-CA852-IgG1
39.92692
12.28077
−0.2115385


IMAB362
60.56292
41.36427
20.82923
















TABLE 11







ADCC of candidate antibodies on 293T-Claudin18.2 cells


(corresponding to FIG. 6B)









Inhibition rate, %











Antibody
Antibody
Antibody



concentration
concentration
concentration


Antibody ID
(50 μg/mL)
(5 μg/mL)
(0.5 μg/mL)





CLDMix-CA818-IgG1
45
25
15


CLD393-C1002-IgG1
64
49
22


CLD393-C1024-IgG1
48
47
15


IMAB362
49
45
18









Cells were counted and diluted with an ADCC buffer to 4×105 cells/mL. A proper amount of the sample was taken for a serial dilution. Effector cells Jurkat (G7011, Promega) were centrifuged at 1500 rpm with the supernatant discarded, and resuspended with 1% FBS RPMI-1640 medium. The cells were counted and diluted with the ADCC buffer to 8×105 cells/mL. Then 25 μL of the target cells was added to each well of a white 96-well plate (3917, Costar), and 25 μL of the serially diluted antibodies was added to each well coated with the target cells. 25 μL of effector cells (Jurkat) was added to each well in a ratio of effector cells to target cells of 20000:10000. Then the 96-well plate was incubated in a cell incubator for 5 h, and equilibrated at room temperature. Then 75 μL of a Bio-Glo color-developing buffer (G7940, Promega) was added to each well for a 15-min reaction, and the plate was detected on a Tecan microplate reader (chemiluminescence). The results are shown in FIGS. 7A-8 and Tables 12-13.


As shown in FIG. 7A, with NUGC4 being the target, the EC50 value of CLDQMIX-CA808.1-IgG1 was 1.264 μg/mL, which was less than that of the reference antibody IMAB362 of 2.154 μg/mL, indicating that the candidate antibody CLDQMIX-CA808.1-IgG1 has a better ability of mediating ADCC than the reference antibody IMAB362. This indicates that CLDQMIX-CA808.1-IgG1 can kill target cells expressing Claudin18.2 better and has better pharmaceutical effects.









TABLE 12







ADCC of candidate antibodies on NUGC4 cells


(corresponding to FIG. 7A)










Antibody ID
EC50 (μg/mL)
Antibody ID
EC50 (μg/mL)













CLDQMIX-
1.264
CLD387-115-
14.27


CA808.1-IgG1

IgG1



CLDQ1-CA841 -IgG1
17.33
IMAB362
2.154
















TABLE 13







ADCC of candidate antibodies on NUGC4 cells


(corresponding to FIG. 7B)










Antibody ID
EC50 (μg/mL)
Antibody ID
EC50 (μg/mL)













CLD389-279-IgG1
0.81
IMAB362
2.56


CLD389-CA802-IgG1
15.73
/
/









Example 4. Modification of Fc Terminus of Anti-Claudin 18.2 Antibodies
4.1 Modification of Fc Terminus of Antibodies

In order to enhance the ADCC of antibodies and change the affinity between the antibodies and Fc receptor, the Fc terminus of the antibody was mutated. The mutated amino acid sequence of heavy chain constant region is set forth in SEQ ID NO: 18. The resulting antibody was named CLDQMix-CA808.1-IgG1-VLPLL.


4.2 Affinity of Modified Antibody to Fc Receptors
4.2.1 Affinity of Antibody to Human FcRn

The antibody binding kinetics was determined by OctetRED 96 system based on the biolayer interferometry (BLI). Human FcRn (ACROBiosystems, FCM-H82W4, 1 μg/mL) was loaded to Streptavidin (SA) Dip and Read™ Biosensors with a loading height of 0.2 nm. The antibody was serially diluted by 2× with PBST from 33.3 mM, and a blank control was set. The Association time was set to 150 s, and the dissociation time was set to 100 s. After the assay, the equilibrium dissociation constant (kD) was calculated using Steady State Analysis.


4.2.2 Affinity of Antibody to Human CD32a(H)

The antibody binding kinetics was determined by OctetRED 96 system based on the biolayer interferometry (BLI). Human CD32a(H) (Sino Biological, 10374-H27H1-B, 1 μg/mL) was loaded to Streptavidin (SA) Dip and Read™ Biosensors with a loading height of 0.2 nm. The antibody was serially diluted by 2× with PBST from 1000 mM, and a blank control was set. The Association time was set to 150 s, and the dissociation time was set to 100 s. After the assay, the equilibrium dissociation constant (kD) was calculated using Steady State Analysis.


4.2.3 Affinity of Antibody to Human CD16a(V)

The antibody binding kinetics was determined by OctetRED 96 system based on the biolayer interferometry (BLI). Human CD16a(V) (Sino Biological, 10389-H27H1-B, 0.5 μg/mL) was loaded to Streptavidin (SA) Dip and Read™ Biosensors with a loading height of 0.5 nm. The antibody was serially diluted by 2× with PBST from 166.7 mM, and a blank control was set. The Association time was set to 30 s, and the dissociation time was set to 100 s. After the assay, association constant (kon), dissociation constant (kdis) were calculated by curve fitting with a 1:1 model and equilibrium dissociation constant (kD) were calculated in the ratio of kd/ka.


4.2.4 Affinity of Antibody to Human CD16a(F)

The antibody binding kinetics was determined by OctetRED 96 system based on the biolayer interferometry (BLI). Human CD16a(F) (Sino Biological, 10389-H27H-B, 0.5 μg/mL) was loaded to Streptavidin (SA) Dip and Read™ Biosensors with a loading height of 0.5 nm. The antibody was serially diluted by 2× with PBST from 333.3 mM, and a blank control was set. The Association time was set to 30 s, and the dissociation time was set to 100 s. After the assay, association constant (kon), dissociation constant (kdis) were calculated by curve fitting with a 1:1 model and equilibrium dissociation constant (kD) were calculated in the ratio of kd/ka.


4.2.5 Affinity of Antibody to Human CD32b

The antibody binding kinetics was determined by OctetRED 96 system based on the biolayer interferometry (BLI). Human CD32a(H) (Sino Biological, 10374-H27H1-B, 1 μg/mL) was loaded to Streptavidin (SA) Dip and Read™ Biosensors with a loading height of 0.2 nm. The antibody was serially diluted by 2× with PBST from 2000 mM, and a blank control was set. The Association time was set to 40 s, and the dissociation time was set to 50 s. After the assay, the equilibrium dissociation constant (kD) was calculated using Steady State Analysis.


As shown in Table 14, the affinity of CLDQMIX-CA808.1-IgG1-VLPLL to agonistic receptors, particularly Human CD16a(F), was greatly improved, thus better promoting the ADCC in subjects.









TABLE 14







Affinity of candidate antibody to Fc receptors














KD (M) of





KD (M) of
CLDQMIX-
Ratio




CLDQMIX-
CA808.1-IgG1
of



FC receptor
CA808.1-IgG1
VLPLL
KD





Agonistic
Human CD32a(H)
4.10E-07
3.00E-07
1.37↑


receptors
Human CD16a(V)
1.07E-07
5.41E-08
1.98↑



Human CD16a(F)
5.66E-07
1.19E-07
4.75↑



Human FcRn
6.93E-09
5.27E-09
1.31↑


Inhibitory
Human CD32b
3.20E-06
3.00E-06



receptor









Example 5. Pharmacodynamics of Anti-Claudin18.2 Antibodies

Human gastric cancer NUGC4 cells (JCRB Cell Bank, catalog number: JCRB0834) were cultivated in an RPMI1640 medium containing 10% fetal bovine serum, 100 U/mL penicillin and 100 μg/mL streptomycin through monolayer culture in vitro in an incubator at 37° C./5% CO2. The cells were digested with trypsin-EDTA twice a week for passaging as per conventional practice. At a cell saturation of 80%-90% and a required number, the cells were harvested, counted and grafted into BALB/c nude mice (female, 6-8 weeks old, 18-22 g) (Shanghai Lingchang Biotechnology Co., Ltd.). 0.2 mL (1×106 cells) of NUGC4 cells (along with matrigel in a volume ratio of 1:1) was subcutaneously grafted on the right back of each mouse, and the mice were randomized when the mean tumor volume was approximately 60-70 mm3. The animals were weighed before administration and the tumor volume was measured. The mice were randomized by the tumor volume (randomized block design), 8 in each group. The weight was measured twice a week, and the tumor diameter was measured with a vernier caliper twice a week. The tumor volume was calculated using the following formula: V=0.5a×b2, where, a and b represent the long diameter and short diameter of the tumor respectively. The results are shown in FIG. 9 and Table 15.


As shown in FIG. 9, the tumor inhibitory effect of 5 mg/kg CLDQMIX-CA808.1-IgG1-VLPLL was comparable to that of 10 mg/kg IMAB362. Compared with 10 mg/kg IMAB362, 10 mg/kg CLDQMIX-CA808.1-IgG1-VLPLL demonstrated better tumor inhibitory effect. This indicated that the CLDQMIX-CA808.1-IgG1-VLPLL antibody has better tumor inhibitory activity than IMAB362, and the inhibitory effect of CLDQMIX-CA808.1-IgG1-VLPLL on tumors is dose-dependent. The tumor inhibitory effect increases with the dose.









TABLE 15







Pharmacodynamics of candidate antibodies


(corresponding to FIG. 9)









Final tumor


Antibody ID
volume (mm3)





Vehicle
1536.5 ± 195.8


IMAB362 10 mg/kg
1301.0 ± 177.2


CLDQMIX-CA808.1-IgG1-VLPLL 5 mg/kg
1307.8 ± 186.4


CLDQMIX-CA808.1-IgG1-VLPLL 10 mg/kg
1103.3 ± 186.5


CLDQMIX-CA808.1-IgG1-VLPLL 20 mg/kg
1006.4 ± 207.1









Example 6. Preparation of Claudin18.2-Specific Chimeric Antigen Receptor-Modified T Cells
6.1 Preparation of Gene Fragment of Chimeric Antigen Receptor

In the present invention, a fusion gene fragment was designed in the following order of coding genes: CD8α signal peptide, CA841 scFv VH-linker-CA841 scFv VL, CD8 hinge region, CD8 transmembrane region, and 4-1BB and CD3ζ intracellular signaling regions, and the fusion gene was directly synthesized by gene synthesis techniques, allowing the expressed chimeric antigen receptor to have an amino acid sequence of scFv VH-linker-scFv VL-CD8 hinge-CD8TM-4-1BB-CD3ζ. The linker had a sequence of GGGGSGGGGSGGGGS, the CD8α signal peptide had a sequence of SEQ ID NO: 21, the CD8 hinge region (CD8 hinge) had a sequence of SEQ ID NO: 22, the CD8 transmembrane region (CD8 TM) had a sequence of SEQ ID NO: 23, the 4-1BB had a sequence of SEQ ID NO: 24, and the CD3ζ had a sequence of SEQ ID NO: 25.


The whole gene of pRRLSIN lentivirus vector containing a human EF1a promoter was synthesized, and the green fluorescent protein (GFP) sequence was replaced by an EGFRt marker protein sequence to give a pRRLSIN-EGFRt vector (see FIG. 10).


6.2 Construction of Lentiviral Expression Vector of Chimeric Antigen Receptor

In the example, the vector system used to construct the lentivirus plasmid vector of the present invention was a third generation self-inactivated lentivirus vector system. The system has three plasmids: a pMDLg-pRRE packaging plasmid (Unibio, VT1449) encoding protein Gag/Pol, a pRSV-rev packaging plasmid (Unibio, VT1445) encoding Rev protein, and an envelope plasmid PMD2.G (Unibio, VT1443) encoding VSV-G protein.


In the example, a lentivirus expression vector expressing specific CAR and EGFRt (SEQ ID NO: 27) linked by P2A (SEQ ID NO: 26) was constructed, and the target gene obtained in Section 6.1 was linked to the pRRLSIN-EGFRt vector to form a recombinant plasmid named pRRLSIN-Claudin18.2CAR-P2A-EGFRt (see FIG. 11). The specific sequence was pRRLSIN-CD8α-scFv VH-linker-scFv VL-CD8hinge-CD8TM-4-1BB-CD3ζ-P2A-EGFRt. After verified by enzyme digestion and sequencing, the successfully constructed vector was ready to package. CAR-P2A-EGFRt was transcribed into a single mRNA, but finally translated into two peptide chains of EGFRt and anti-Claudin18.2 chimeric antigen receptor. Anti-Claudin18.2 CAR was located on the cell membrane under the direction of the CD8α signal peptide.


The four sequences containing the target CAR obtained in the example are as follows:


scFv CA808.1-CD8hinge-CD8TM-4-1BB-CD3ζ-P2A-EGFRt (hereinafter referred to as CN01)


scFv CA841-CD8hinge-CD8TM-4-1BB-CD3ζ-P2A-EGFRt (hereinafter referred to as CN02)


scFv C279-CD8hinge-CD8TM-4-1BB-CD3ζ-P2A-EGFRt (hereinafter referred to as CN03)


scFv C115-CD8hinge-CD8TM-4-1BB-CD3ζ-P2A-EGFRt (hereinafter referred to as CN04)


6.3 Preparation of Chimeric Antigen Receptor Lentivirus

The pRRLSIN-Claudin18.2CAR-P2A-EGFRt expression plasmid and pMDLg-pRRE, pRSV-rev and pMD2.G helper plasmids were extracted and mixed with the transfection reagent polyethyleneimine (PEI) in a certain ratio to co-transfect 293T cells. The major procedures are as follows:


(1) The 293T cells passaged to 5-8th generations (ATCC CRL-3216) were seeded at a cell density of 7×106 in a DMEM medium (purchased from GIBCO) containing 10% FBS (purchased from GIBCO) in 75 cm3 cell culture flasks. After mixing, the cells were cultivated in a CO2 incubator at 37° C./5% CO2 for 24 h before transfection. A cell aggregation of about 70-80% was observed on the next day, and the cells were transfected.


(2) 24 h later, the target expression plasmid and the pMDLg-pRRE, pRSV-rev and pMD2.G helper plasmids were mixed in a weight ratio of 4:3:2:2, and diluted with an Opti-MEM medium (purchased from GIBCO) to give a solution A. A PEI diluent was prepared in a ratio of total plasmids:PEI=3:1, and diluted with the Opti-MEM medium to obtain a solution B. The solutions A and B were mixed well and incubated at room temperature for 15 min.


(3) The 293T cells were immobilized on a plate, and slowly added with the plasmid-PEI mixture. The resulting mixture was shaken gently, and cultivated in a CO2 incubator at 37° C./5% CO2 for 4-6 h. After incubation, the medium was replaced with a fresh DMEM medium containing 10% FBS.


(4) After 48 h and 96 h of transfection, the culture supernatant containing viruses was collected and centrifuged at 3000 rpm at 4° C. for 5 min. The supernatant was filtered through a 0.45 μm filter, mixed with PEG8000/NaCl in a volume ratio of 4:1, incubated at 4° C. for 2-3 h, and centrifuged at a high speed for 30 min. The supernatant was discarded and the precipitate was resuspended with precooled T cell medium X-VIVO 15 (Lonza, 04-418Q) or PBS to give a virus concentrate which was stored at −80° C. for later use.


6.4 Lentivirus Titer Assay

In the example, the biological activity titer of lentivirus was determined by infecting cells. The 293T cells were used for lentivirus activity assay, and 1×105 cells were inoculated to each well of a 24-well culture plate. 1 mL of fresh DMEM medium containing 10% FBS was added to each well. The mixture was diluted to a final concentration of 6 μg/mL with transfection additive Polybrene. The lentivirus concentrate was serially diluted by 3× to the 5th concentration, added at 1 μL/well in duplicate, and mixed well. The cells were incubated in a CO2 incubator at 37° C./5% CO2 for 24 h. After 24 h, the cells were digested, and the positive rate of protein expression of CAR or EGFRt was detected by a flow cytometer using an anti-human IgG(Fab)2 (Jackson ImmunoResearch, 109-065-006) or anti-human EGFRt (Biolegend, 352904) flow dye. The titer was calculated by the following formula: lentivirus activity titer (TU/mL)=positive rate×dilution factor×100×105. The activity titers of lentivirus concentrates of the above CAR (CN01, CN02, CN03 and CN04) packaged by PEI transfection were greater than 1×108 TU/mL (FIG. 12).


6.5 Preparation of T Lymphocytes

Peripheral blood mononuclear cells (PBMCs) purchased from AllCells were marked with microbeads through a CD3 MicroBeads human-lyophilized Kit (purchased from Miltenyi Biotech). CD3+ T lymphocytes with high purity were selected, with a proportion of CD3 positive T cells over 95%. The purified T cells were activated and proliferated using a human CD3CD28 T cell activator (Dynabeads Human T-Activator CD3/CD28, Thermo Fisher, 11132D).


6.6 Lentivirus-Transduced T Lymphocytes

CAR-T cells were obtained by transducing T cells with the lentivirus prepared in Section 6.3. After stimulated and activated for 24-48 h, T lymphocytes from Section 6.5 were observed using microscopy for their activation. Activated T lymphocytes are larger in volume with elongated or irregular shape. The activated T lymphocytes were collected, centrifuged and resuspended in a T cell medium X-VIVO 15 (Lonza, 04-418Q) with a final concentration of 10 ng/mL IL-7 and 5 ng/mL IL-15 and a final volume of 1 mL, and added to a 12-well culture plate. The lentivirus was diluted to MOI=3-5 with the same medium and mixed with 1×106 activated T lymphocytes for infection. The mixture was incubated overnight on a 24-well plate in an incubator at 37° C./5% CO2. The next day, the cells were centrifuged again and the medium was refreshed. The cell density was measured every 2 days thereafter, and the cells were further expanded with the cell density controlled at NMT 2×106 cells/mL. After the T cells were co-incubated with the lentivirus for 48-72 h, the expression of different chimeric antigen receptors was determined by flow cytometry. With non-transduced T lymphocytes as negative control, the positive rates of T lymphocytes expressing different chimeric antigen receptors are shown in Table 16 (FIG. 13).









TABLE 16







Positive rates of T lymphocytes expressing different


chimeric antigen receptors








Cells transfected with the following CAR
Positive rate of CAR





CN01
30.5%


CN02
19.1%


CN03
24.3%


CN04
13.2%









After being infected with lentiviruses packaging different chimeric antigen receptors, T lymphocytes were cultivated for about 9 days, reaching about 300× expansion, which indicated that T lymphocytes expressing different chimeric antigen receptors could be expanded in vitro to a certain extent, providing a guarantee for subsequent in vitro functional studies and pharmacodynamic studies in animals.


6.7 In Vitro Toxicity Assay
6.7.1 Target Specificity Assay

Claudin-18 has two splicing variants, i.e., Claudin 18.1 and Claudin 18.2, with only eight amino acid alterations in sequence. Claudin 18.1 is selectively expressed in normal lung cells, while Claudin 18.2 is highly restricted in normal cells, but is frequently ectopically activated and overexpressed in multiple tumors (e.g., gastric cancer, lung cancer, pancreatic cancer). In the example, with 293T cells (purchased from KYinno, KC-0990/KC-0986) overexpressing Claudin18.1 protein and Claudin18.2 protein being the target cells and prepared Claudin18.2 CAR-T with different scFvs being the effector cell, a co-incubation system of CAR-T cells and target cells was established using 293T cells, 293T cells overexpressing Claudin18.2 protein and 293T cells overexpressing Claudin18.1 protein in different E:T (effector cells:target cells) ratios. The specific response of CAR-T to the two proteins was evaluated by measuring the lysis rate of tumor cells. The results of in vitro assay (FIG. 14) demonstrated that, with a fixed number of tumor cells, the efficiency of killing tumor cells ranged from 20%-50% at 24 h (Table 17) when the prepared Claudin18.2 CAR-T with different scFvs (CN01, CN02, CN03, CN04) was co-incubated with 293T-hClaudin18.2 cells at E:T ratios of 1:1 and 3:1. When co-incubated with 293T-hClaudin18.1 cells at E:T ratios of 1:1 and 3:1, the prepared Claudin18.2 CAR-T cells with different scFvs were significantly different in specific lysis ability against 293T-hClaudin18.1 cells, among which the CN02 CAR-T had no significant specific lysis ability against 293T-hClaudin18.1 cells, while the CN01, CN03 and CN04 CAR-T had different degrees of specific lysis ability against 293T-hClaudin18.1 cells. The specific response of CAR-T cells was also evaluated by measuring the content of cytokines (INF-gamma) secreted into the culture supernatant. The difference in release of IFN-gamma cytokines in supernatant of the Claudin18.2 CAR-T (CN01, CN02, CN03, CN04) with different scFvs and the 293T-hClaudin18.1 cells was consistent with the results of killing assay (FIG. 15 and Table 18). The IFN-gamma cytokines in 293T-hClaudin18.1 cells co-incubation supernatant in CN02 group was significantly lower than that in CN01, CN03 and CN04 groups.


Specific lysis assay: an LDH Release Assay Kit (Dojindo, CK12) was used for assay, which is an INT chromogenic reaction catalyzed by diaphorase, and measures the activity of LDH released during cytotoxicity via colorimetry. Damage to the cell membrane structure caused by cell apoptosis or necrosis will lead to release of enzymes in cytoplasm into the cultures, including lactate dehydrogenase (LDH) with relatively stable enzymatic activity. The cytotoxicity can be quantitatively analyzed by activity assay of LDH released from lysed cells into the cultures. LDH release is considered as an important indicator of cell membrane integrity and is widely used for cytotoxicity assay.


Cytokine assay: Human IFN-gamma ELISA kit (R&D Systems, SIF50) was used for measuring cytokines, which is based on the immobilization of an antigen or antibody and enzymatic labeling of the antigen or antibody. The antigen or antibody that binds to the surface of a solid carrier retains the immunological activity, while the enzyme labeled antigen or antibody retains both immunological activity and enzymatic activity. During the assay, the test substance (the antigen or antibody) in the sample are bound to the immobilized antibody or antigen. Non-binding substances are removed by washing, and the enzyme-labeled antigen or antibody is added. In this case, the amount of enzyme immobilized is associated with the amount of the test substance in the sample. After a substrate that reacts with the enzyme is added for color development, the content of the test substance in the sample could be judged by the color for qualitative or quantitative analysis.









TABLE 17





Specific lysis assay of Claudin18.2 CAR-T cells with different


scFvs in vitro




















Specific lysis rate







(E:T = 1:1)
T
CN01
CN02
CN03
CN04





293T
1.60%
 5.90%
7.25%
 9.18%
 3.96%


293T-hClaudin18.1
2.42%
17.60%
5.75%
34.85%
15.25%


293T-hClaudin18.2
0.77%
28.31%
39.68%
51.53%
23.78%





Specific lysis rate







(E:T = 3:1)
T
CN01
CN02
CN03
CN04





293T
 1.10%
14.91%
19.21%
23.08%
 8.93%


293T-hClaudin18.1
 2.72%
30.79%
11.98%
44.53%
32.91%


293T-hClaudin18.2
−0.49%
58.02%
55.86%
65.62%
49.72%
















TABLE 18





Release of IFN-gamma cytokines in supernatant of


co-incubated Claudin18.2 CAR-T


cells with different scFvs and different 293T cells




















IFN-gamma







pg/mL (E:T = 1:1)
T
CN01
CN02
CN03
CN04





293T
0.00
0.00
0.00
578.76
0.00


293T-hClaudin18.1
0.00
1059.53
0.00
7229.32
3291.64


293T-hClaudin18.2
0.00
3818.19
7091.96
11235.69
7961.92





IFN-gamma







pg/mL (E:T = 3:1)
T
CN01
CN02
CN03
CN04





293T
0.00
17.87
0.00
899.27
0.00


293T-hClaudin18.1
0.00
1952.37
0.00
10411.52
4619.46


293T-hClaudin18.2
0.00
4791.17
10171.14
14188.95
8889.10









6.7.2 Target Toxicity Assay

In the example, an in vitro pharmacodynamic test was established by simulating the mechanism of action (MOA) of the product. The inventor constructed a plasmid overexpressing Claudin18.2 protein with a plvx vector, and prepared a lentivirus. Gastric cancer cells NUGC4 and AGS were infected with the lentivirus, and NUGC4 cells and AGS cells with high expression of Claudin18.2 protein were obtained through subsequent screening of positive cells as target cells for functional verification of CAR-T cells. Claudin18.2 CAR-T cells with different scFvs prepared above were used as effector cells. A co-incubation system of CAR-T cells and target tumor cells was established in different E:T (effector cells:target cells) ratios. The biological efficacy of the CAR-T cells was evaluated by measuring the lysis rate of tumor cells, with a co-incubation system of non-transduced T cells and tumor cells being the control.


The results of in vitro assay (FIG. 16 and Table 19) demonstrated that the efficiency of killing tumor cells was excellent at 24 h when CAR-T cells were co-incubated with Claudin18.2-positive tumor cells (AGS-claudin18.2 and NUGC-4), which was significantly higher than that of T cells, while the killing effect on the Claudin18.2-negative cell line AGS was mild. Also, the biological efficacy of CAR-T cells was evaluated by measuring the content of cytokines (INF-gamma) secreted into the culture supernatant. After Claudin18.2 CAR-T cells were co-incubated with Claudin18.2-positive tumor cells (AGS-claudin18.2 and NUGC-4), the expression of the IFN-gamma cytokines was significantly higher than that of the T cell group (FIG. 17, Table 20).









TABLE 19





Specific lysis assay of Claudin18.2 CAR-T cells with different


scFvs against Claudin18.2-positive tumor cells




















Specific lysis rate







(E:T = 1:1)
T
CN01
CN02
CN03
CN04





AGS
2.01%
 8.83%
 6.32%
10.89%
−1.12%


AGS-claudin18.2
7.61%
43.94%
41.18%
47.84%
37.02%


NUGC-4
5.05%
58.25%
52.22%
68.23%
57.64%





Specific lysis rate







(E:T = 3:1)
T
CN01
CN02
CN03
CN04





AGS
 3.11%
17.53%
14.67%
20.50%
13.18%


AGS-claudin18.2
15.92%
44.20%
43.86%
58.56%
41.87%


NUGC-4
 5.17%
64.78%
59.11%
66.50%
66.38%
















TABLE 20





Release of IFN-gamma cytokines in supernatant of


co-incubated Claudin18.2 CAR-T


cells with different scFvs and Claudin18.2-positive tumor cells




















IFN-gamma







pg/mL (E:T = 1:1)
T
CN01
CN02
CN03
CN04





AGS
6.42
 475.74
 235.36
 842.04
 304.04


AGS-claudin18.2
0.00
2787.98
4470.66
 7732.98
4962.87


NUGC-4
0.00
8580.04
7401.03
13731.08
9266.85





IFN-gamma







pg/mL (E:T = 3:1)
T
CN01
CN02
CN03
CN04





AGS
63.66
 979.40
 349.83
 613.10
  0.00


AGS-claudin18.2
 0.00
3955.55
8145.06
11968.28
6130.44


NUGC-4
 0.00
6382.26
4745.38
11624.88
7206.43









The in vitro cytotoxicity assay shows that all the T lymphocytes expressing different chimeric antigen receptors have good killing ability on Claudin18.2-positive tumor cells, which provides a basis for pharmacodynamic studies in animals.


6.8 Studies in Animals

In the example, a pharmacodynamic model of immunodeficient mouse bearing gastric cancer tumor was established. Based on in vitro studies, each of the female NOG mice (purchased from Charles River) was grafted with 1×107 NUGC4-Claudin18.2 cells on the back. The mice were administered on Day 11 after grafting (the tumor volume was about 80-100 mm3). The vehicle control group was administered with 0.9% normal saline, the Mock-T (T cells not transfected with plasmid) group was administered with 1×107 cells, and CN02 low-dose and high-dose groups (positive cells) were administered with 5.00×106 and 1.00×107 cells respectively. The dose volume was 100 μL. 6 animals were allocated in each group. Tumors were measured twice a week after administration. The tumor growth curve was plotted, TGI and T/C were calculated, and all tumors were photographed at the end of the study. Blood was sampled before CAR-T administration (Day −2), on Days 2, 9 and 28 after administration, and the vector copy number (VCN) of CAR in peripheral blood of mice was measured by qPCR, so as to confirm the expansion of CART cells. The results showed that within 36 days after CAR-T administration, the efficacy of both treatment group was significant. In the 6 mice in the Claudin18.2 CAR-T (CN02) low-dose group, the tumor regressed completely (6/6), and in 5 mice in the high-dose group, the tumor regressed completely (5/6) (FIG. 18).


(1) Body weight: compared with the vehicle control and Mock groups, the Claudin18.2 CAR-T (CN02) low-dose and high-dose groups had no significant difference in body weight (FIG. 19).


(2) Tumor growth inhibition (TGI): 14 days after administration, the TGI of the Claudin18.2 CAR-T (CN02) low-dose and high-dose groups was 100% and 88.33% respectively (FIG. 20).


(3) Death rate: by 31 days after administration, one animal was found dead in the vehicle control group, and no death was observed in the Claudin18.2 CAR-T (CN02) low-dose and high-dose groups (FIG. 21).


Example 7. Preparation of CAR-T Cells by Non-Viral Method
7.1 Construction of Non-Viral PiggyBac(PB) Transposon Vector

In the example, with a pBluescript vector (synthesized by General BIOL) as the backbone, a gene insulator sequence cHS4 was found and placed at both termini of a polyclonal site, 5′ITR and 3′ITR sequences of PB transposon were found and constructed inside the cHS4 sequence of the vector, an EF1a promoter was inserted at the 5′ terminus inside the ITR, and a polyA signal was inserted at the 3′ terminus. A polyclonal sequence was retained in the middle, into which a CAR-P2A-EGFRt sequence was inserted to form a PB CN02 CAR plasmid structure, as shown in FIG. 24.


7.2 Preparation of CAR-T Cells by Non-Viral PB Transposon Vector

7.2.1 Peripheral blood mononuclear cells (PBMCs) purchased from AllCells were marked with microbeads through a CD3 MicroBeads human-lyophilized Kit (purchased from Miltenyi Biotech). CD3+ T lymphocytes with high purity were selected, with a proportion of CD3 positive T cells over 95%. The purified T cells were activated and proliferated using a human CD3CD28 T cell activator (Dynabeads Human T-Activator CD3/CD28, Thermo Fisher, 11132D).


7.2.2 Electroporation was performed on Day 3 after stimulation. The cells for electroporation were resuspended using a pipette and counted. 5×106 cells were used for each electroporation. The 5×106 cells were diluted with DPBS (GIBCO, 14190-144) to 4 mL, and centrifuged at 300 g at room temperature for 10 min. The supernatant was discarded, and the cells were resuspended and washed with 5 mL of DPBS, and centrifuged at 300 g at room temperature for 10 min and then the supernatant was discarded. The cells were then resuspended in 100 μL of electroporation buffer Entranster-E (Engreen, 98668-20), and the cell suspension was transferred to a 1.5-mL centrifuge tube.


The components in Table 21 were added to the centrifuge tube and mixed well.









TABLE 21







Electroporation system










Component
Volume (μL)














PB CNO2 CAR plasmid (1 μg/μL)
5



PB transposase plasmid (1 μg/μL)
5



Cell suspension
100



Total
110










Electroporation was performed using an electroporation instrument manufactured by Lonza. The cell/plasmid suspension was quickly transferred to the cuvette, and the cuvette was tapped to allow the cell suspension to fully form a balanced liquid level in the cuvette. The program EQ115 was used for electroporation. The cuvette was taken out carefully after electroporation. Then 500 μL of preheated T cell medium X-VIVO 15 (Lonza, 04-418Q) was added and equilibrated in an incubator at 37° C. for 5 min, and the cells were resuspended using a microporous loading tip by blowing 2-3 times. The cells were transferred to a 12-well plate containing 2 mL of preheated medium and incubated at 37° C. The medium was refreshed 4-6 h after electroporation to improve the viability. The supernatant was discarded, and preheated fresh medium was added. The cells were incubated in an incubator at 37° C./5% CO2 for 48 h before the test.


Meanwhile, a CAR-T control group using lentiviruses was set. The preparation method can be seen in Section 6.6.


48-72 h after electroporation, the expression of chimeric antigen receptors was determined by flow cytometry using an anti-human IgG(Fab)2 antibody, with non-transduced T lymphocytes as negative control. The positive rates of T lymphocytes expressing different chimeric antigen receptors are shown in Table 22 (FIG. 22).









TABLE 22







Positive rates of T lymphocytes expressing different


chimeric antigen receptors










Cell
Positive rate of CAR, %














Non-transduced T cell
0.56



Non-viral PB CN02 CAR-T
35.4



Lentiviral Lenti CN02 CAR-T
38










7.3 Target Toxicity Assay

In the example, an in vitro pharmacodynamic test was conducted by simulating the mechanism of action (MOA) of the product. The constructed gastric cancer cells NUGC4 and AGS with high expression of Claudin18.2 were used as target cells, and CN02 CAR-T cells prepared using the above non-viral (PB) and CN02 CAR-T cells prepared using lentiviruses (lenti) were used as effector cells. A co-incubation system of CAR-T cells and target tumor cells was established in different E:T (effector cells:target cells) ratios. The biological efficacy of the CAR-T cells was evaluated by measuring the lysis rate of tumor cells, with a co-incubation system of non-transduced T cells and tumor cells being the control.


The results of in vitro assay (FIG. 23 and Table 23) demonstrated that the lysis rate of tumor cells was linearly correlated with the number of effector cells for a fixed number of tumor cells; and the efficiency of killing tumor cells was excellent at 24 h when CAR-T cells were co-incubated with Claudin18.2-positive tumor cells (AGS-claudin18.2 and NUGC-4), which was significantly higher than that of T cells, while the killing effect on the Claudin18.2-negative cell line AGS was mild.









TABLE 23





In vitro specific lysis assay of CN02 CAR-T cells


prepared by different methods


















Specific lysis rate
Non-transduced
Lentiviral Lenti
Non-viral PB


(E:T = 1:1)
T cell
CN02 CAR-T
CN02 CAR-T





AGS
 0.23%
 3.30%
 1.12%


AGS-claudin18.2
−4.79%
56.49%
29.28%


NUGC-4
−5.50%
21.00%
12.00%





Specific lysis rate
Non-transduced
Lentiviral Lenti
Non-viral PB


(E:T = 3:1)
T cell
CN02 CAR-T
CN02 CAR-T





AGS
5.40%
 7.20%
 6.43%


AGS-claudin18.2
0.59%
83.83%
64.83%


NUGC-4
5.00%
42.00%
25.00%








Claims
  • 1. An antibody or an antigen-binding fragment thereof, comprising three light chain complementarity determining regions and/or three heavy chain complementarity determining regions, wherein the three light chain complementarity determining regions of the antibody or the antigen-binding fragment thereof comprise an LCDR1 set forth in SEQ ID NO: 5, an LCDR2 set forth in SEQ ID NO: 6 and an LCDR3 set forth in SEQ ID NO: 7, and/or the three heavy chain complementarity determining regions of the antibody or the antigen-binding fragment thereof comprise an HCDR1 set forth in SEQ ID NO: 8, an HCDR2 set forth in SEQ ID NO: 9 and an HCDR3 set forth in SEQ ID NO: 10,orthe three light chain complementarity determining regions of the antibody or the antigen-binding fragment thereof comprise an LCDR1 set forth in SEQ ID NO: 11, an LCDR2 set forth in SEQ ID NO: 12 and an LCDR3 set forth in SEQ ID NO: 13, and/or the three heavy chain complementarity determining regions of the antibody or the antigen-binding fragment thereof comprise an HCDR1 set forth in SEQ ID NO: 14, an HCDR2 set forth in SEQ ID NO: 15 and an HCDR3 set forth in SEQ ID NO: 16.
  • 2. An antibody or an antigen-binding fragment thereof, wherein the antibody or the antigen-binding fragment thereof comprises a light chain variable region set forth in SEQ ID NO: 1, and/or a heavy chain variable region set forth in SEQ ID NO: 2,orthe antibody or the antigen-binding fragment thereof comprises a light chain variable region set forth in SEQ ID NO: 3, and/or a heavy chain variable region set forth in SEQ ID NO: 4.
  • 3. The antibody or the antigen-binding fragment thereof according to claims 1 or 2, comprising a heavy chain constant region set forth in SEQ ID NO: 17 or a heavy chain constant region set forth in SEQ ID NO: 18.
  • 4. A chimeric antigen receptor (CAR), comprising the antibody or the antigen-binding fragment thereof according to claims 1 or 2.
  • 5. The chimeric antigen receptor according to claim 4, sequentially comprising the antibody or the antigen-binding fragment thereof, an extracellular hinge region, a transmembrane region and an intracellular signaling region, wherein, preferably, the extracellular hinge region is a CD8 hinge region, the transmembrane region is a CD8 transmembrane region, and the intracellular signaling region is 4-1BB and CD3ζ.
  • 6. A nucleic acid or a cell, wherein the nucleic acid encodes the antibody or the antigen-binding fragment thereof according to any one of claims 1 to 3, or the chimeric antigen receptor according to claims 4 or 5; and the cell expresses the antibody or the antigen-binding fragment thereof according to any one of claims 1 to 3, or the chimeric antigen receptor according to claims 4 or 5.
  • 7. A pharmaceutical composition, comprising the antibody or the antigen-binding fragment thereof according to any one of claims 1 to 3, the chimeric antigen receptor according to claims 4 or 5, or the nucleic acid or the cell according to claim 6.
  • 8. A kit, comprising the antibody or the antigen-binding fragment thereof according to any one of claims 1 to 3, or the nucleic acid according to claim 6.
  • 9. Use of the antibody or the antigen-binding fragment thereof according to any one of claims 1 to 3, the chimeric antigen receptor according to claims 4 or 5, or the nucleic acid according to claim 6 for treating, preventing, detecting or diagnosing a disease, wherein, preferably, the disease is cancer.
  • 10. The use according to claim 9, wherein the cancer is a Claudin18.2-positive cancer; preferably, the cancer includes gastric cancer, pancreatic cancer, esophageal cancer, lung cancer, ovarian cancer, head and neck cancer, bladder cancer, cervical cancer, sarcoma, cytoma, colon cancer, kidney cancer, colorectal cancer, liver cancer, melanoma, breast cancer, myeloma, neuroglioma, leukemia and lymphoma.
Priority Claims (2)
Number Date Country Kind
201910459129.1 May 2019 CN national
201910459622.3 May 2019 CN national
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2020/092849 5/28/2020 WO 00