CN111978402B - Novel CLDN18.2 binding molecules - Google Patents

Novel CLDN18.2 binding molecules Download PDF

Info

Publication number
CN111978402B
CN111978402B CN201910437800.2A CN201910437800A CN111978402B CN 111978402 B CN111978402 B CN 111978402B CN 201910437800 A CN201910437800 A CN 201910437800A CN 111978402 B CN111978402 B CN 111978402B
Authority
CN
China
Prior art keywords
antibody
seq
antigen
gly
ala
Prior art date
Legal status (The legal status 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 status listed.)
Active
Application number
CN201910437800.2A
Other languages
Chinese (zh)
Other versions
CN111978402A (en
Inventor
谭永聪
郎国竣
孔超
刘婵娟
邓敏
吴琪
张静
张文海
范宝国
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sanyou Biopharmaceuticals Co Ltd
Original Assignee
Sanyou Biopharmaceuticals Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CN201910437800.2A priority Critical patent/CN111978402B/en
Application filed by Sanyou Biopharmaceuticals Co Ltd filed Critical Sanyou Biopharmaceuticals Co Ltd
Priority to KR1020217039993A priority patent/KR20220012262A/en
Priority to TW109116820A priority patent/TW202108627A/en
Priority to US17/613,440 priority patent/US20220396616A1/en
Priority to EP20815629.9A priority patent/EP3978532A4/en
Priority to JP2022516255A priority patent/JP2022533804A/en
Priority to CN202080038303.6A priority patent/CN114206935B/en
Priority to CA3141504A priority patent/CA3141504A1/en
Priority to PCT/CN2020/091441 priority patent/WO2020238730A1/en
Publication of CN111978402A publication Critical patent/CN111978402A/en
Application granted granted Critical
Publication of CN111978402B publication Critical patent/CN111978402B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • C12N15/81Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/22Immunoglobulins specific features characterized by taxonomic origin from camelids, e.g. camel, llama or dromedary
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/60Fusion polypeptide containing spectroscopic/fluorescent detection, e.g. green fluorescent protein [GFP]

Abstract

The present invention provides novel CLDN18.2 binding molecules. The invention also provides nucleic acid molecules encoding said CLDN18.2 binding molecules, expression vectors and host cells for expressing CLDN18.2 binding molecules. The invention further provides methods of producing said CLDN18.2 binding molecules and uses thereof.

Description

Novel CLDN18.2 binding molecules
Sequence listing
The present application contains a sequence listing and is incorporated herein by reference in its entirety.
Technical Field
The present application relates generally to antibodies. More specifically, the present application relates to single domain antibodies specifically recognizing CLDN18.2, methods of making the same, and uses thereof.
Background
Cell connection is an intercellular connection structure, and is an important basis for the mutual connection and synergistic effect between adjacent cells in a multicellular organism. In general, there are four types of junctions, tight junctions, adhesive junctions, gap junctions and desmosomes/hemidesmosomes, in animal cells.
Tight junctions, also known as closed junctions, are structures formed between endothelial cells or epithelial cells that prevent diffusion of materials between tissues from the intercellular space and thus can only enter the cell by active transport. The tightly connected structure is formed by dozens of Claudin proteins through protein interaction in and among cells, and the expression of the proteins has certain tissue specificity. CLDN18 is an important protein among Claudin proteins present in tight junctions.
CLDN18 is a membrane protein with a quartic transmembrane region, containing two extracellular domains. There are two CLDN18 variants in humans, CLDN18.1 and CLDN18.2, respectively. The two are distributed in different tissues, the former is mainly expressed in lung epithelial cells, the latter is specifically expressed in stomach epithelial cells and is not expressed in stomach stem cells. On the protein sequence, CLDN18.1 and CLDN18.2 have very high sequence similarity, the main difference between them is at the N-terminus, and there is only 8 amino acid difference between them in the first extracellular domain; the C-terminal sequence is identical except for the N-terminal.
Antibody therapy is becoming one of the most promising approaches to treat cancer patients. CLDN18.2 has become a very potential target for antibody drug action due to its expression specificity in tumor cells and normal tissues. However, since the sequence similarity of CLDN18.1 and CLDN18.2 is very high, it is very difficult to develop an antibody directed to CLDN18.2 only but not to CLDN18.1, and there are few reports worldwide. IMAB362, developed by Ganymed, germany, is by far the only clinically under-developed antibody directed against CLDN18.2 target. IMAB362 is a monoclonal antibody consisting of two protein heavy chains and two protein light chains. Currently, there are no single domain antibodies targeting CLDN 18.2.
Single domain antibodies, abbreviated as single domain antibodies (sdabs), are antibodies that consist of a single monomeric variable antibody domain (e.g., an antibody heavy chain variable domain). Like whole antibodies (e.g., IgG), it is capable of selectively binding to a particular antigen. But the molecular weight of single domain antibodies is much smaller than that of common antibodies consisting of two protein heavy chains and two protein light chains. The first single domain antibody was engineered from the heavy chain antibody found in camelids (Hamers-Casterman C, Atarhouch T, Muyledermans S, Robinson G, Hamers C, Songa EB, Bendahman N, Hamers R (1993) Natural encapsulation antibodies revolute their derivative of light chains 363(6428): 446-; the heavy chain antibodies found in these camelids are also referred to as VHH fragments. Currently, most studies on single domain antibodies are based on heavy chain variable domains.
Single domain antibodies have many advantages. For example, they typically exhibit high solubility, good thermostability and tissue permeability, single domain antibodies are also resistant to degradation by papain and the like due to the presence of a second pair of intramolecular disulfide bonds; furthermore, single domain antibodies can be expressed in a variety of host cells, such as yeast, plant and mammalian cells, and are expressed in high amounts, making them extremely cost effective. The advantages of single domain antibodies make them suitable for use in a variety of biotechnological and therapeutic applications. For example, they may be used to treat diseases including, but not limited to, cancer, infectious diseases, inflammatory diseases, and neurodegenerative diseases.
Although there are currently clinically under investigation monoclonal antibody drugs targeting the CLDN18.2 target, there remains an urgent need to continue to develop antibodies targeting CLDN18.2 targets as therapeutic agents. It would be desirable in the art to develop new antibodies targeting CLDN18.2, in particular single domain antibodies that specifically recognize CLDN18.2 only and not CLDN 18.1.
Disclosure of Invention
Broadly, the present disclosure provides novel compounds, methods of making, compositions, and articles of manufacture for antibodies. The benefits provided by the present disclosure are broadly applicable in the field of antibody therapy and diagnostics. More specifically, the present disclosure provides single domain antibodies targeting CLDN18.2, as well as methods of making the antibodies, expression vectors and host cells for expressing the antibodies, and the like. The antibodies of the present disclosure provide methods of treating or preventing disorders associated with the Claudin protein, particularly CLDN18.2, and uses thereof.
The inventors have for the first time found CLDN18.2 binding molecules (e.g. single domain antibodies targeting CLDN 18.2) capable of specifically binding to extracellular domain 1(ECD1) of human CLDN 18.2.
The present disclosure includes at least the following embodiments, which are respectively ordered and enumerated in the manner of "N" (where "N" denotes a number). The following list is non-exhaustive and the skilled person can combine different solutions.
A CLDN18.2 binding molecule comprising at least one immunoglobulin single variable domain, wherein said immunoglobulin single variable domain comprises CDR1, CDR2 and CDR3, and wherein
CDR1 comprises a CDR identical to SEQ ID NO: 1 amino acid sequence that is at least 80%, 85%, 90%, or 95% identical;
CDR2 comprises a CDR sequence identical to SEQ ID NO: 2 an amino acid sequence that is at least 80%, 85%, 90%, or 95% identical; and
CDR3 comprises a CDR sequence identical to SEQ ID NO: 3 amino acid sequence that is at least 80%, 85%, 90% or 95% identical.
2. The CLDN18.2 binding molecule of embodiment 1, wherein CDR1 is identical in amino acid sequence to SEQ ID NO: 1 there is a difference in amino acid addition, deletion or substitution of no more than 2 amino acids; CDR2 is identical in amino acid sequence to SEQ ID NO: 2 there is a difference in amino acid addition, deletion or substitution of no more than 2 amino acids; and/or CDR3 is identical in amino acid sequence to SEQ ID NO: 3 there is a difference in amino acid addition, deletion or substitution of no more than 2 amino acids.
3. The CLDN18.2 binding molecule of embodiment 1, wherein CDR1 is identical in amino acid sequence to SEQ ID NO: 1 there is a difference in amino acid addition, deletion or substitution of 1 amino acid; CDR2 is identical in amino acid sequence to SEQ ID NO: 2 there is a difference in amino acid addition, deletion or substitution of 1 amino acid; and/or CDR3 is identical in amino acid sequence to SEQ ID NO: 3 there is a difference in amino acid addition, deletion or substitution of 1 amino acid.
4. The CLDN18.2 binding molecule of any one of the preceding embodiments, wherein the CLDN18.2 binding molecule is an antibody or antigen-binding fragment thereof directed to CLDN 18.2.
5. The CLDN18.2 binding molecule of any one of the preceding embodiments, wherein the immunoglobulin single variable domain is a VHH; for example VHH from a camelid (e.g. alpaca).
6. The CLDN18.2 binding molecule of any one of the preceding embodiments, wherein the immunoglobulin single variable domain comprises:
i) as shown in SEQ ID NO: 1, CDR1 of the amino acid sequence set forth in seq id no;
ii) of the formula ISRGGX1T CDR2 of the amino acid sequence shown in, wherein X1Is T or S; and
iii) formula NAQWDX2GTX3CDR3 of the amino acid sequence shown by RYLEV, wherein X2Is P or V, X3Is F or I.
7. The CLDN18.2 binding molecule of any one of the preceding embodiments, wherein the immunoglobulin single variable domain comprises:
(a) as shown in SEQ ID NO: 1, as shown in SEQ ID NO: 2 and the CDR2 of the amino acid sequence shown in SEQ ID NO: 3, CDR3 of the amino acid sequence set forth in seq id no;
(b) as shown in SEQ ID NO: 1, as shown in SEQ ID NO: 4 and the CDR2 of the amino acid sequence shown in SEQ ID NO: 5 CDR3 of the amino acid sequence set forth in seq id no; or
(c) As shown in SEQ ID NO: 1, as shown in SEQ ID NO: 2 and the CDR2 of the amino acid sequence shown in SEQ ID NO: 6, CDR3 of the amino acid sequence set forth in seq id no.
8. The CLDN18.2 binding molecule of any one of the preceding embodiments, wherein the immunoglobulin single variable domain comprises
(A) SEQ ID NO: 7;
(B) and SEQ ID NO:7 amino acid sequence that is at least 80%, 85%, 90% or 95% identical; or
(C) And SEQ ID NO:7 has one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions, and/or substitutions as compared to 7.
9. The CLDN18.2 binding molecule of any one of the preceding embodiments, wherein the immunoglobulin single variable domain is modified (e.g., substituted with amino acids) at one or more of the following positions of SEQ ID No. 7: amino acid 1, 4, 5, 14, 16, 35, 47, 56, 58, 65, 92, 102, 105 or 121.
10. The CLDN18.2 binding molecule of any one of the preceding embodiments, wherein the immunoglobulin single variable domain comprises or consists of the sequence: any one of SEQ ID NOS 7-14.
11. The CLDN18.2 binding molecule of any one of the preceding embodiments, wherein the CLDN18.2 binding molecule is a single domain antibody, e.g., a heavy chain single domain antibody; a chimeric antibody; or a humanized antibody.
12. The CLDN18.2 binding molecule of any one of the preceding embodiments, wherein the immunoglobulin single variable domain is fused to another molecule, such as an Fc domain of an immunoglobulin (e.g., IgG) or a fluorescent protein.
13. The CLDN18.2 binding molecule of embodiment 12, wherein the CLDN18.2 binding molecule is a chimeric antibody comprising a VHH from a camelid and an Fc domain of a human IgG (e.g., human IgG1 or IgG 4).
14. The CLDN18.2 binding molecule of embodiment 13, wherein the CLDN18.2 binding molecule is a chimeric antibody comprising a VHH from alpaca and an Fc domain of human IgG 1.
15. The CLDN18.2 binding molecule of any one of the preceding embodiments, wherein the CLDN18.2 binding molecule binds to extracellular domain 1(ECD1) of human CLDN 18.2.
A CLDN18.2 binding molecule which competes for the same epitope as a CLDN18.2 binding molecule of any one of the preceding embodiments.
17. A CLDN18.2 binding molecule of any one of the preceding embodiments, which specifically binds to CLDN18.2 but not to CLDN 18.1.
18. An isolated nucleic acid molecule comprising a nucleic acid sequence encoding a CLDN18.2 binding molecule as defined in any one of the preceding embodiments.
19. The isolated nucleic acid molecule of embodiment 18 comprising or consisting of the sequence of seq id no: any one of SEQ ID NOs: 22-29.
20. An expression vector comprising the isolated nucleic acid molecule of embodiment 18 or 19.
21. A host cell comprising the expression vector of embodiment 20.
22. The host cell of embodiment 21, which is a bacterial cell (e.g., e.coli), a fungal cell (e.g., yeast) or a mammalian cell.
23. A pharmaceutical composition comprising at least one CLDN18.2 binding molecule as defined in any one of embodiments 1 to 17 and a pharmaceutically acceptable carrier.
24. A method of making a CLDN18.2 binding molecule as defined in any one of embodiments 1 to 17, comprising the steps of:
-expressing a CLDN18.2 binding molecule as defined in any one of embodiments 1 to 17 in a host cell of embodiment 21 or 22; and
-isolating the CLDN18.2 binding molecule from the host cell.
25. A method of treating a disorder associated with CLDN18.2 in a subject, the method comprising: administering to the subject a therapeutically effective amount of a CLDN18.2 binding molecule as defined in any one of embodiments 1-17.
26. The method of embodiment 25, wherein the disorder related to CLDN18.2 comprises a disease involving cells expressing CLDN18.2 or a disease related to cells expressing CLDN 18.2.
27. The method of embodiment 25 or 26, wherein the disorder related to CLDN18.2 comprises cancer.
28. The method of embodiment 27, wherein said cancer comprises bone cancer, blood cancer, lung cancer, liver cancer, pancreatic cancer, skin cancer, head and neck cancer, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, prostate cancer, uterine cancer, cancer of the sexual and reproductive organs, hodgkin's disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the bladder, cancer of the kidney, renal cell carcinoma, cancer of the renal pelvis, tumors of the Central Nervous System (CNS), cancer of the neuroectodermal, spinal axis, glioma, meningioma and pituitary adenoma.
29. The method of embodiment 28, wherein the cancer is gastric cancer.
30. Use of a CLDN18.2 binding molecule as defined in any one of embodiments 1 to 17 in the preparation of a medicament for the treatment or prevention of a disorder associated with CLDN 18.2.
31. A kit for the treatment or diagnosis of a disorder associated with CLDN18.2, comprising a container comprising a CLDN18.2 binding molecule as defined in any one of embodiments 1 to 17.
The foregoing is a general description and may include simplifications, generalizations, and omissions of detail as necessary. Consequently, those skilled in the art will realize that this general description is illustrative only and is not intended to be in any way limiting. Other aspects, features and advantages of the methods, compositions and/or devices and/or other subject matter described herein will become apparent in the teachings presented herein. This summary is provided to introduce a selection of concepts in a simplified form that are further described below in the detailed description. The above summary is not intended to identify key features or essential features of the claimed subject matter, nor is it intended to be used as an aid in determining the scope of the claimed subject matter. In addition, the contents of all references, patents, and published patent applications cited throughout this application are incorporated by reference herein in their entirety.
Drawings
FIGS. 1a and 1b show the flow cytometry identification of the over-expressed cell lines and tumor cell lines in example 1. Wherein, FIG. 1a shows the results of flow cytometric identification of human CLDN18.2-HEK293T, human CLDN18.1-HEK293, murine CLDN18.2-HEK293 and murine CLDN18.1-HEK293 overexpressing cell lines; FIG. 1b shows the flow cytometry identification of human CLDN18.2-KATOIII tumor cell line.
Figure 2 shows a sequence comparison between a plurality of candidate antibodies. Therein, the full length sequences of the variable regions of the candidate antibodies and the CDR sequences therein are shown, and the amino acid sequence differences between the candidate antibodies are indicated.
FIG. 3 shows the results of cross-binding experiments of candidate antibody NA1-S on human CLDN18.2-HEK293T, human CLDN18.1-HEK293, murine CLDN18.2-HEK293 and murine CLDN18.1-HEK293 overexpressing cell lines.
FIGS. 4a and 4b show the results of a comparative experiment of binding of the candidate antibody NA1-S and the control antibody at the cellular level. Wherein, FIG. 4a shows the results of a flow-through level comparison experiment of antibody NA1-S on human CLDN18.2-HEK293T overexpressing cells; FIG. 4b shows the results of a flow-level comparison experiment of antibody NA1-S and a control antibody on a human CLDN18.2-KATOIII tumor cell line.
FIGS. 5a and 5b show Complement Dependent Cytotoxicity (CDC) mediated by candidate antibody NA1-S on human CLDN18.2-HEK293T overexpressing cell line and human CLDN18.2-KATOIII tumor cell line. Wherein, FIG. 5a shows CDC mediated by antibody NA1-S on human CLDN18.2-HEK 293T; FIG. 5b shows CDC mediated by antibody NA1-S on human CLDN18.2-KATOIII tumor cell line.
FIGS. 6a and 6b show antibody dependent cell-mediated cytotoxicity (ADCC) mediated by candidate antibody NA1-S on human CLDN18.2-HEK293T and human CLDN18.2-KATOIII tumor cell lines. Wherein, FIG. 6a shows ADCC mediated by antibody NA1-S on human CLDN18.2-HEK293T cell line; FIG. 6b shows ADCC mediated by antibody NA1-S on a human CLDN18.2-KATOIII tumor cell line.
Detailed Description
While this invention is susceptible of embodiment in many different forms, there are disclosed herein specific illustrative embodiments thereof which are indicative of the principles of the invention. It should be emphasized that the invention is not limited to the specific embodiments illustrated. Moreover, any section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
Unless otherwise defined herein, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by one of ordinary skill in the art. Furthermore, unless the context requires otherwise, singular terms shall include the plural and plural terms shall include the singular. More specifically, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a protein" includes a plurality of proteins; reference to "a cell" includes mixtures of cells and the like. In this application, the use of "or" means "and/or" unless stated otherwise. Furthermore, the use of the term "including" as well as other forms (such as "includes" and "including") is not limiting. Furthermore, the ranges provided in the specification and the appended claims include the endpoints and all values between the endpoints.
Generally, the nomenclature used in connection with, and the techniques of, cell and tissue culture, molecular biology, immunology, microbiology, genetics and protein and nucleic acid chemistry and hybridization described herein are those well known and commonly used in the art. Unless otherwise indicated, the methods and techniques of the present invention are generally performed according to conventional methods well known in the art, and as referenced and discussed throughout the present specificationThe discussion is made as described in various general and more specific references. See, e.g., Abbas et al, Cellular and Molecular Immunology, 6th ed.,W.B.Saunders Company(2010);Sambrook J.&Molecular Cloning, A Laboratory Manual, 3rd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2000); ausubel et al, Short Protocols in Molecular Biology A Complex of Methods from Current Protocols in Molecular Biology, Wiley, John&Sons, inc. (2002); harlow and Lane use Antibodies A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1998); and Coligan et al, Short Protocols in Protein Science, Wiley, John&Sons, inc. (2003). The nomenclature associated with the analytical chemistry, synthetic organic chemistry, and pharmaceutical chemistry described herein, and the laboratory procedures and techniques, are those well known and commonly employed in the art. Moreover, any section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
Definition of
For a better understanding of the present invention, the definitions and explanations of the relevant terms are provided below.
As used herein, the term "antibody" or "Ab" generally refers to any form of antibody that exhibits a desired biological or binding activity. It includes, but is not limited to, humanized antibodies, fully human antibodies, chimeric antibodies and single domain antibodies. The antibody may comprise a heavy chain and a light chain. Heavy chains can be divided into μ, δ, γ, α and ε, which define the antibody isotype as IgM, IgD, IgG, IgA and IgE, respectively. Each heavy chain consists of a heavy chain variable region (VH) and a heavy chain constant region (CH). The heavy chain constant region consists of 3 domains (CH1, CH2 and CH 3). Each light chain consists of a light chain variable region (VL) and a light chain constant region (CL). The VH and VL regions can be further divided into relatively conserved regions, termed Framework Regions (FRs), and hypervariable regions, termed Complementarity Determining Regions (CDRs), separated by FRs. Each VH and VL consists of 3 CDRs and 4 FRs in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4 from N-terminus to C-terminus. The distribution of amino acids in various regions or domains follows Kabat Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987and 1991)) or Chothia & Lesk (1987) J.mol.biol.196: 901-917; chothia et al, (1989) Nature 342: 878-883. The antibodies may be of different antibody isotypes, such as IgG (including for example IgG1, IgG2, IgG3 or IgG4 subtypes), IgA1, IgA2, IgD, IgE or IgM antibodies.
The term "humanized antibody" as used herein refers to an antibody in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. Additional framework region modifications can be made within the human framework sequences.
The term "chimeric antibody" as used herein broadly refers to an engineered antibody containing one or more regions from one antibody and one or more regions from one or more other antibodies. Specifically, the chimeric antibody comprises a variable region derived from a non-human animal antibody and a constant region of another antibody, for example, a variable region of mouse origin and a constant region of human origin. A chimeric antibody may also refer to a multispecific antibody having specificity for at least two different antigens.
The term "CLDN 18.2 binding molecule" as used herein means a molecule that specifically binds to CLDN 18.2.
The terms "CLDN 18.2 antibody", "antibody directed to CLDN 18.2", "antibody specifically binding to CLDN 18.2", "antibody specifically targeting CLDN 18.2", "antibody specifically recognizing CLDN 18.2" as used herein are used interchangeably and mean an antibody capable of specifically binding to the Claudin protein CLDN 18.2. In particular, in particular embodiments, antibodies that specifically bind to human CLDN18.2, in particular antibodies that specifically bind to human CLDN18.2 but not to human CLDN18.1 are meant. The amino acid sequences of human CLDN18.2 and human CLDN18.1 are shown in SEQ ID NO. 15 and SEQ ID NO. 16, respectively; the amino acid sequences of mouse CLDN18.2 and mouse CLDN18.1 are shown in SEQ ID NO 17 and SEQ ID NO 18, respectively.
The term "immunoglobulin single variable domain" or "ISV" as used herein is generally defined herein as an amino acid sequence: which comprises an immunoglobulin fold or is capable of forming an immunoglobulin fold (i.e., by folding) under suitable conditions (e.g., physiological conditions), i.e., thereby forming an immunoglobulin variable domain (e.g., a VH, VL or VHH domain); and forming (or under suitable conditions being capable of forming) an immunoglobulin variable domain that comprises a functional antigen binding site (in the sense that it does not require interaction with another immunoglobulin variable domain (such as a VH-VL interaction) to form a functional antigen binding site).
The term "Ka" as used herein is intended to refer to the association rate of a particular antibody-antigen interaction, while the term "Kd" as used herein is intended to refer to the dissociation rate of a particular antibody-antigen interaction. As used herein, the term "KD'OR' KDThe value "is intended to represent the dissociation constant for a particular antibody-antigen interaction, which is obtained from the ratio of Kd to Ka (i.e., Kd/Ka) and expressed as molar concentration (M). K of antibodyDThe values may be determined using methods well established in the art. Determination of antibody K DA preferred method of value is by using surface plasmon resonance, preferably using a biosensor system such as
Figure BDA0002071082450000111
And (4) a system.
The term "specific binding" or "specifically binds to" as used herein refers to a non-random binding reaction between two molecules, e.g., between an antibody and an antigen.
As used herein, the ability to "inhibit binding," "block binding," or "compete for the same epitope" refers to the ability of an antibody to inhibit the binding of two molecules to any detectable degree. In some embodiments, an antibody that blocks binding between two molecules inhibits the binding interaction between the two molecules by at least 50%. In some embodiments, the inhibition may be greater than 60%, greater than 70%, greater than 80%, or greater than 90%.
The term "high affinity" antibody as used herein refers to an antibody having 1 x 10 to the target antigen-7M or less, more preferably 5X 10-8M or less, even more preferably 1X 10-8M or less, even more preferably 5X 10-9M or less, and even more preferably 1X 10-9K of M or lessDAntibodies of value.
The term "EC" as used herein50", also referred to as" half effective concentration ", refers to the concentration of drug, antibody or toxin agent that induces a response of 50% between the baseline and maximum values after a particular exposure time. In the context of the present application, EC 50In units of "nM".
As used herein, the term "epitope" refers to the portion of an antigen to which an immunoglobulin or antibody specifically binds. An "epitope" is also referred to as an "antigenic determinant". Epitopes or antigenic determinants usually consist of chemically active surface groups of molecules such as amino acids, carbohydrates or sugar side chains and usually have a specific three-dimensional structure and specific charge characteristics. For example, an epitope typically comprises at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 contiguous or non-contiguous amino acids in a unique stereo conformation, which can be a "linear epitope" or a "conformational epitope". See, e.g., epitopic Mapping Protocols in Methods in Molecular Biology, Vol.66, G.E.Morris, Ed. (1996). In a linear epitope, all the interaction sites between a protein and an interacting molecule (e.g., an antibody) are linearly present along the primary amino acid sequence of the protein. In conformational epitopes, the interaction sites span amino acid residues that are separated from each other in the protein. Antibodies can be screened depending on the competition for binding to the same epitope as detected by conventional techniques known to those skilled in the art. For example, competition or cross-competition studies may be performed to obtain antibodies that compete or cross-compete with each other for binding to an antigen (e.g., CLDN 18.2). In international patent application WO 03/048731, a high throughput method for obtaining antibodies binding to the same epitope is described, which is based on their cross-competition.
As used herein, the term "isolated" refers to the state of a substance or component that is obtained by artificial means from a natural state. An "isolated" substance or component, if it occurs in nature, may be due to a change in the natural environment in which it is found, or the separation of the substance or component from its natural environment, or both. For example, an un-isolated polynucleotide or polypeptide naturally occurs in a living animal, and the same high purity polynucleotide or polypeptide isolated from that natural state is referred to as an isolated polynucleotide or polypeptide. The term "isolated" does not exclude mixed artificial or synthetic substances nor other impurities which do not affect the activity of the isolated substance.
As used herein, the term "isolated antibody" is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities. Furthermore, the isolated antibody may be substantially free of other cellular material and/or chemicals.
As used herein, the term "vector" refers to a nucleic acid vehicle into which a polynucleotide can be inserted. When a vector allows the expression of a protein encoded by a polynucleotide inserted therein, the vector is referred to as an expression vector. The vector may be used to express the carried genetic material element in a host cell by transformation, transduction, or transfection into the host cell. Vectors are well known to those skilled in the art and include, but are not limited to, plasmids, bacteriophages, cosmids, artificial chromosomes such as Yeast Artificial Chromosomes (YACs), Bacterial Artificial Chromosomes (BACs) or P1-derived artificial chromosomes (PACs), bacteriophages such as lambda or M13 bacteriophages, and animal viruses. Animal viruses that may be used as vectors include, but are not limited to, retroviruses (including lentiviruses), adenoviruses, adeno-associated viruses, herpes viruses (e.g., herpes simplex virus), poxviruses, baculoviruses, papilloma viruses, papovaviruses (e.g., SV 40). The vector may contain a number of elements for controlling expression, including but not limited to promoter sequences, transcription initiation sequences, enhancer sequences, selection elements and reporter genes. In addition, the vector may comprise an origin of replication.
As used herein, the term "host cell" refers to any kind of cell system into which a vector can be introduced, including, but not limited to, prokaryotic cells such as e.coli (e.coli) or Bacillus subtilis (Bacillus subtilis), fungal cells such as yeast cells or Aspergillus (Aspergillus), insect cells such as S2 drosophila cells or Sf9, and animal cells such as fibroblasts, CHO cells, COS cells, NSO cells, HeLa cells, BHK cells, HEK293 cells, or human cells.
Methods for producing the antibodies of the invention using host cells are conventional in the art and include expressing the antibody in prokaryotic or eukaryotic cells, then isolating the antibody, and typically purifying to a pharmaceutically acceptable purity. For example, in some embodiments, a nucleic acid encoding an antibody is inserted into an expression vector and the expression vector is introduced into a suitable prokaryotic or eukaryotic host cell by standard techniques known in the art, the host cell, e.g., CHO cell, NSO cell, SP2/0 cell, HEK293 cell, COS cell, per.c6(R) cell, yeast or e.coli cell is cultured under conditions and for a time sufficient to produce the antibody or functional fragment thereof of the invention, and the antibody is recovered from the cell (supernatant or cells after lysis) and purified. Conventional methods for producing antibodies are known in the art and are described, for example, in Makrides, S.C., Protein Expr. Purif.17(1999) 183-202; geisse, S. et al, Protein Expr. Purif.8(1996) 271-282; kaufman, R.J., mol.Biotechnol.16(2000) 151-160; werner, R.G., Drug Res.48(1998) 870-.
The term "identity," as used herein, refers to the relationship between the sequences of two or more polypeptide molecules or two or more nucleic acid molecules as determined by aligning and comparing the sequences. "percent identity" refers to the percentage of identical residues between amino acids or nucleotides in a comparison molecule and is calculated based on the size of the smallest molecule being compared. For these calculations, the gaps in the alignment (if any) are preferably addressed by a specific mathematical model or computer program (i.e., an "algorithm"). Methods that can be used to calculate the identity of aligned nucleic acids or polypeptides include those described in comparative Molecular Biology, (Lesk, A.M., ed.), 1988, New York: Oxford University Press; biocomputing information and Genome Projects, (Smith, D.W., ed.), 1993, New York: Academic Press; computer Analysis of Sequence Data, Part I, (Griffin, A.M., and Griffin, H.G., eds.), 1994, New Jersey: Humana Press; von Heinje, G., 1987, Sequence Analysis in Molecular Biology, New York: Academic Press; sequence Analysis Primer, (Gribskov, m.and deveux, j., eds.), 1991, New York, m.stockton Press; and those described in Carillo et al, 1988, SIAMJ. applied Math.48: 1073.
As used herein, the term "immunogenicity" refers to the ability to stimulate the formation of specific antibodies or sensitized lymphocytes in an organism. It refers not only to the property of an antigen to stimulate the activation, proliferation and differentiation of specific immune cells to eventually produce immune effector substances such as antibodies and sensitized lymphocytes, but also to the specific immune response of antibodies or sensitized T lymphocytes that can be developed in the immune system of an organism after stimulating the organism with the antigen. Immunogenicity is the most important property of an antigen. Whether an antigen is able to successfully induce the generation of an immune response in a host depends on three factors: the nature of the antigen, the reactivity of the host and the means of immunization.
As used herein, the term "transfection" refers to the process of introducing nucleic acids into eukaryotic cells, particularly mammalian cells. Protocols and techniques for transfection include, but are not limited to, lipofection, chemical and physical methods of transfection such as electroporation. Many transfection techniques are well known in the art, see, e.g., Graham et al, 1973, Virology52: 456; sambrook et al, 2001, Molecular Cloning: A Laboratory Manual; davis et al, 1986, Basic Methods in Molecular Biology, Elsevier; chu et al, 1981, Gene13: 197.
As used herein, the term "fluorescence activated cell sorting" or "FACS" refers to a specialized type of flow cytometry. It provides a method of Sorting a heterogeneous mixture of biological cells into two or more containers, one Cell at a time, based on the specific light scattering and Fluorescence characteristics of each Cell (flowmetric. Instruments for performing FACS are known to those skilled in the art and are commercially available to the public. Examples of such instruments include FACS Star Plus, FACScan and FACScat instruments from Becton Dickinson (Foster City, CA), Epics C from Coulter Epics Division (Hialeah, FL) and MoFlo from Cytomation (Colorado Springs, Colorado).
The term "subject" includes any human or non-human animal, preferably a human.
As used herein, the term "a disorder associated with CLDN 18.2" refers to any disorder caused, exacerbated or otherwise associated with increased or decreased expression or activity of CLDN18.2 (such as human CLDN 18.2).
As used herein, the term "cancer" refers to any tumor or malignant cell growth, proliferation or metastasis mediated solid or non-solid tumor such as leukemia that causes a medical condition.
The term "treatment" as used herein in the context of treating a condition generally relates to the treatment and therapy of a human or animal in which some desired therapeutic effect is achieved, for example, inhibition of disease progression, including decreased rate of progression, arrested rate of progression, regression of the condition, amelioration of the condition, and cure of the condition. Treatment as a prophylactic measure (i.e., prophylaxis) is also included. For cancer, "treatment" may refer to inhibiting or slowing the growth, proliferation, or metastasis of a tumor or malignant cell, or some combination thereof. For a tumor, "treatment" includes removal of all or part of the tumor, inhibition or slowing of tumor growth and metastasis, prevention or delay of tumor development, or some combination thereof.
As used herein, the term "therapeutically effective amount" refers to the amount of active compound or material, composition or dosage form containing the active compound that, when administered in accordance with a desired treatment regimen, is effective to produce some desired therapeutic effect commensurate with a reasonable benefit/risk ratio. In particular, a "therapeutically effective amount" means an amount or concentration of an antibody, or antigen-binding portion thereof, that is effective to treat a disorder associated with CLDN 18.2.
The term "pharmaceutically acceptable" as used herein means that the carrier, diluent, excipient, and/or salt thereof is chemically and/or physically compatible with the other ingredients of the formulation, and physiologically compatible with the recipient.
As used herein, the term "pharmaceutically acceptable carrier and/or excipient" refers to carriers and/or excipients that are pharmacologically and/or physiologically compatible with the subject and the active agent, which are well known in the art (see, e.g., Remington's Pharmaceutical sciences. edited by geno AR, 19th ed. pennsylvania: machine Publishing Company, 1995), and include, but are not limited to, pH adjusting agents, surfactants, adjuvants, and ionic strength enhancing agents. For example, pH adjusting agents include, but are not limited to, phosphate buffers; surfactants include, but are not limited to, cationic, anionic, or nonionic surfactants, such as Tween-80; ionic strength enhancers include, but are not limited to, sodium chloride.
As used herein, the term "adjuvant" refers to a non-specific immunopotentiator that, when delivered to an organism with an antigen or delivered to an organism in advance, can enhance the immune response to an antigen or alter the type of immune response in an organism. A variety of adjuvants exist in the art, including, but not limited to, aluminum adjuvants (e.g., aluminum hydroxide), Freund's adjuvants (e.g., Freund's complete adjuvant and Freund's incomplete adjuvant), Corynebacterium parvum, lipopolysaccharide, cytokines, and the like. Freund's adjuvant is currently the most commonly used adjuvant in animal experiments. Aluminum hydroxide adjuvants are more commonly used in clinical trials.
CLDN18.2 binding molecules
In some aspects, the disclosure comprises CLDN18.2 binding molecules.
In general, CLDN18.2 binding molecules may include any molecule that specifically binds to CLDN 18.2. In some cases, a "CLDN 18.2 binding molecule" may include a "CLDN 18.2 antagonist. A CLDN18.2 binding molecule or CLDN18.2 antagonist may be a polypeptide or protein, such as an antibody, more particularly an antibody that specifically binds to CLDN18.2 (e.g., human CLDN 18.2).
Antibodies include, but are not limited to, chimeric, humanized, or single domain antibodies and the like. In specific embodiments, the CLDN18.2 binding molecule is a single domain antibody, which generally refers to an antibody consisting of a single monomeric variable antibody domain. Like whole antibodies, single domain antibodies are capable of selectively binding to a particular antigen.
More particularly, a CLDN18.2 binding molecule is a heavy chain single domain antibody which may be used with the terms "VHH", "VHH antibody", "VHH domain", "VHH antibody fragment", "VHH"or" Nanobody "and the like are used interchangeably. VHH molecules from camelid antibodies are one of the smallest known intact antigen binding domains (about 15KDa, or 1/10 for conventional IgG) and are therefore well suited for delivery to dense tissues and into the confined space between macromolecules.
The single domain antibodies disclosed herein may be prepared by one of skill in the art according to methods known in the art or any future method. For example, VHHs may be obtained using methods known in the art, for example by immunising a camel and obtaining VHHs therefrom which bind to and neutralise a target antigen, or by cloning a library of VHHs of the invention using molecular biology techniques known in the art, followed by selection using phage display. In some embodiments, the single domain antibodies of the invention are naturally produced in camelids, i.e. camelids immunized with CLDN18.2 or fragments thereof using the techniques described herein for other antibodies.
In some embodiments, the single domain antibody is obtained by immunizing a llama or alpaca with the desired antigen and then isolating the mRNA encoding the heavy chain antibody. By reverse transcription and polymerase chain reaction, a gene library containing millions of cloned single domain antibodies is generated. Screening techniques such as phage display and ribosome display help identify clones that bind the antigen. Wherein phage display is the synthesis of antibody libraries on phage, screening of the libraries with the antigen of interest or antibody-binding portion thereof, and isolating the antigen-binding phage from which immunoreactive fragments can be obtained. Methods for preparing and screening such libraries are well known in the art, and kits for generating phage display libraries are commercially available (e.g., Pharmacia recombinant phage antibody System, catalog No. 27-9400-01; and Stratagene SurfZAP TMPhage display kit, catalog No. 240612). Still other methods and reagents are useful for generating and screening antibody display libraries (see, e.g., Barbas et al, Proc. Natl. Acad. Sci. USA 88: 7978-.
When the most effective clone is identified, its DNA sequence can be optimized, for example by affinity maturation or humanization, to prevent the immune response of the human body against the antibody.
Thus, a single domain antibody of the invention may be obtained by: (1) isolating the VHH domain of a naturally occurring heavy chain antibody; (2) by expressing a nucleotide sequence encoding a naturally occurring VHH domain; (3) by "humanization" of naturally occurring VHH domains or by expression of nucleic acids encoding such humanized VHH domains; (4) by "camelising" a naturally occurring VH domain from any animal species, particularly mammalian species, for example from a human, or by expressing a nucleic acid encoding such a camelised VH domain; (5) "camelization" by "domain antibodies" or "dAbs" (see, e.g., Ward et al, 1989, Nature 341: 544-546), or by expression of nucleic acids encoding such camelized VH domains; (6) preparing a protein, polypeptide or other amino acid sequence by using synthetic or semi-synthetic techniques; (7) preparing a nucleic acid encoding a VHH by using a technique for nucleic acid synthesis and then expressing the nucleic acid thus obtained; and/or (8) by any combination of the foregoing. Suitable methods and techniques for performing the foregoing will be apparent to those skilled in the art based on the disclosure herein and include, for example, those described in more detail below.
Single domain antibodies are typically generated by PCR cloning a library of variable domains into a phage display vector via cDNA from blood, lymph nodes or splenic lymphocytes obtained from an immunized animal. Antigen-specific single domain antibodies are typically selected by panning the corresponding library over an immobilized antigen (e.g., an antigen coated on the surface of a test tube plastic, a biotinylated antigen immobilized on streptavidin beads, or a membrane protein expressed on the surface of a cell). The affinity of the sdAb can be increased by modeling this strategy in vitro, for example by site-directed mutagenesis of the CDR regions and further panning of the immobilized antigen under increased stringency conditions (higher temperature, high or low salt concentration, high or low pH and low antigen concentration) (Wesolowski et al, Single domain antibodies: conditioning experimental and therapeutic tools in infection and immunity. med microbial Immunol (2009)198: 157-.
Methods for the preparation of VHHs that specifically bind to an antigen or epitope are described in the literature, see for example: van der Linden et al, Journal of Immunological Methods, 240(2000) 185-195; li et al, J Biol chem, 287(2012) 13713-13721; deffar et al, African Journal of Biotechnology Vol.8(12), pp.2645, 17 June, 2009 and WO 94/04678.
In some embodiments, the VHH of the CLDN18.2 binding molecule is fused to an Fc domain of an antibody (e.g., an Fc domain of an IgG (e.g., IgG1 or IgG 4)). By fusing VHH to an Fc domain, effector functions such as ADCC and CDC can be more effectively recruited. Furthermore, fusion of VHH to Fc domain can help CLDN18.2 binding molecules form dimers and can also help extend the in vivo half-life of CLDN18.2 binding molecules.
As used herein, the term "antibody-dependent cell-mediated cytotoxicity" or "ADCC" refers to a cytotoxic form of an antibody that binds to Fc receptors (fcrs) present on certain cytotoxic effector cells (e.g., Natural Killer (NK) cells, neutrophils, and macrophages) to enable these cytotoxic effector cells to specifically bind to antigen-bearing target cells and subsequently kill the target cells with cytotoxins. Antibody "armed" cytotoxic cells are absolutely required for such killing. The major cells mediating ADCC, NK cells, express Fc γ RIII only, whereas monocytes express Fc γ RI, Fc γ RII and Fc γ RIII. FcR expression on hematopoietic cells is summarized in Table 3 at page 464 of ravech and Kinet, Annu.Rev.Immunol9:457-92 (1991). To assess ADCC activity of a molecule of interest, an in vitro ADCC assay may be performed, for example the assay described in US patent 5500362 or US 5821337. Effector cells that can be used in such assays include Peripheral Blood Mononuclear Cells (PBMCs) and Natural Killer (NK) cells. Alternatively or additionally, the ADCC activity of a molecule of interest may be assessed in vivo, for example in an animal model as disclosed by Clynes et al PNAS (USA)95: 652-.
The term "complement-dependent cytotoxicity" or "CDC" refers to the lysis of a target cell in the presence of complement. Activation of the classical complement pathway is initiated by the binding of the first component of the complement system (C1q) to antibodies (of the appropriate subclass) that bind their cognate antigen. To assess complement activation, CDC assays can be performed, for example, by methods described in Gazzano-Santoro et al, J.Immunol.methods 202:163 (1996).
For ease of description, CLDN18.2 binding molecules are described hereinafter as CLDN18.2 antibodies.
CLDN18.2 antibodies capable of specifically binding to a specific epitope of CLDN18.2
In one aspect, the invention relates to a single domain antibody having the ability of specifically binding to CLDN18.2, but not or not substantially binding to CLDN 18.1.
The inventors have for the first time found CLDN18.2 binding molecules (e.g. single domain antibodies targeting CLDN 18.2) capable of specifically binding to extracellular domain 1(ECD1) of human CLDN 18.2.
As previously described, CLDN18.2 has two extracellular domains (ECDs), wherein the full-length sequence of human CLDN18.2 is shown in SEQ ID NO: 15. Wherein, the ECD1 is the amino acids 28-80 of SEQ ID NO. 15, as shown in SEQ ID NO. 19.
SEQ ID NO:15:
MAVTACQGLGFVVSLIGIAGIIAATCMDQWSTQDLYNNPVTAVFNYQGLWRSCVRESSGFTECRGYFTLLGLPAMLQAVRALMIVGIVLGAIGLLVSIFALKCIRIGSMEDSAKANMTLTSGIMFIVSGLCAIAGVSVFANMLVTNFWMSTANMYTGMGGMVQTVQTRYTFGAALFVGWVAGGLTLIGGVMMCIACRGLAPEETNYKAVSYHASGHSVAYKPGGFKASTGFGSNTKNKKIYDGGARTEDEVQSYPSKHDYV
SEQ ID NO:19:
DQWSTQDLYNNPVTAVFNYQGLWRSCVRESSGFTECRGYFTLLGLPAMLQAVR
In addition, mouse CLDN18.2 is shown as SEQ ID NO 17.
SEQ ID NO:17:
MSVTACQGLGFVVSLIGFAGIIAATCMDQWSTQDLYNNPVTAVFNYQGLWRSCVRESSGFTECRGYFTLLGLPAMLQAVRALMIVGIVLGVIGILVSIFALKCIRIGSMDDSAKAKMTLTSGILFIISGICAIIGVSVFANMLVTNFWMSTANMYSGMGGMGGMVQTVQTRYTFGAALFVGWVAGGLTLIGGVMMCIACRGLTPDDSNFKAVSYHASGQNVAYRPGGFKASTGFGSNTRNKKIYDGGARTEDDEQSHPTKYDYV
For human and mouse CLDN18.1, the sequences are shown in SEQ ID NO 16 and SEQ ID NO 18, respectively.
SEQ ID NO:16:
MSTTTCQVVAFLLSILGLAGCIAATGMDMWSTQDLYDNPVTSVFQYEGLWRSCVRQSSGFTECRPYFTILGLPAMLQAVRALMIVGIVLGAIGLLVSIFALKCIRIGSMEDSAKANMTLTSGIMFIVSGLCAIAGVSVFANMLVTNFWMSTANMYTGMGGMVQTVQTRYTFGAALFVGWVAGGLTLIGGVMMCIACRGLAPEETNYKAVSYHASGHSVAYKPGGFKASTGFGSNTKNKKIYDGGARTEDEVQSYPSKHDYV
SEQ ID NO:18:
MATTTCQVVGLLLSLLGLAGCIAATGMDMWSTQDLYDNPVTAVFQYEGLWRSCVQQSSGFTECRPYFTILGLPAMLQAVRALMIVGIVLGVIGILVSIFALKCIRIGSMDDSAKAKMTLTSGILFIISGICAIIGVSVFANMLVTNFWMSTANMYSGMGGMGGMVQTVQTRYTFGAALFVGWVAGGLTLIGGVMMCIACRGLTPDDSNFKAVSYHASGQNVAYRPGGFKASTGFGSNTRNKKIYDGGARTEDDEQSHPTKYDYV
CLDN18.2 antibody comprising CDRs with sequence identity to a specific sequence
In some embodiments, a CLDN18.2 antibody of the present disclosure comprises at least one immunoglobulin single variable domain (e.g., a VHH), wherein the VHH comprises CDR1, CDR2 and CDR3, and wherein CDR1 comprises an amino acid sequence identical to SEQ ID NO:1, CDR2 comprises an amino acid sequence at least 80% identical to SEQ ID NO: 2, and CDR3 comprises an amino acid sequence at least 80% identical to SEQ ID NO: 3 amino acid sequence which is at least 80% identical.
Unless otherwise indicated, the assignment of amino acids to each CDR can be according to one of the numbering schemes provided below: kabat et al (1991) Sequences of Proteins of Immunological Interest (5)thEd.), US depth.of Health and Human Services, PHS, NIH, NIH Publication No. 91-3242; chothia et al, 1987, PMID 3681981; chothia et al, 1989, PMID 2687698; MacCallum et al, 1996, PMID: 8876650; or Dubel, Ed. (2007) Handbook of Therapeutic Antibodies,3rd Ed.,Wily-VCH Verlag GmbH and Co.。
The variable regions and CDRs in an antibody sequence can be identified according to general rules that have been developed in the art (as described above, e.g., the Kabat numbering system) or by aligning the sequence to a database of known variable regions. Methods for identifying these regions are described in Kontermann and Dubel, eds., Antibody Engineering, Springer, New York, NY,2001 and Dinarello et al, Current Protocols in Immunology, John Wiley and Sons Inc., Hoboken, NJ, 2000. Exemplary databases of antibody sequences are described in and available from the "Absysis" website (maintained by Department of Biochemistry & Molecular Biology University College London, London, A.C. Martin of England) and the VBASE2 website www.vbase2.org, as described in Retter et al, Nucl. acids Res.,33(Database issue): D671-D674(2005), on www.bioinf.org.uk/abs. The sequences are preferably analyzed using the Abysis database, which integrates Sequence data from the Kabat, IMGT, and Protein Database (PDB) with structural data from the PDB, see Protein Sequence and Structure Analysis of Antibody Variable Domains in the book by Dr.Andrew C.R.Martin (Ed.: Duebel, S.and Kontermann, R., Springer-Verlag, Heidelberg, ISBN-13:978-3540413547, also available on the website bio for g.uk/abs). The Abysis database website also includes general rules that have been developed for identifying CDRs that can be used in accordance with the teachings herein.
The percent identity between two amino acid sequences can be determined using the algorithm of e.meyers and w.miller (comput.appl.biosci.,4:11-17(1988)), which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4. In addition, percent identity between two amino acid sequences can be determined by the algorithm of Needleman and Wunsch (J.mol. biol.48:444-453(1970)), which has been incorporated into the GAP program in the GCG software package (available from http:// www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, with a GAP weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
Additionally or alternatively, the protein sequences of the invention may further be used as "query sequences" to perform searches against public databases, for example to identify related sequences. Such a search may be performed using the XBLAST program (version 2.0) of Altschul, et al (1990) J.MoI.biol.215: 403-10. BLAST protein searches using the XBLAST program can be performed with a score of 50 and a word length of 3 to obtain amino acid sequences homologous to the antibody molecules of the present invention. To obtain gap alignments for comparison purposes, gap BLAST can be used, as described in Altschul et al, (1997) Nucleic Acids Res.25(17): 3389-3402. When using BLAST and gapped BLAST programs, the default parameters for each program (e.g., XBLAST and NBLAST) can be used, see www.ncbi.nlm.nih.gov.
In other embodiments, the amino acid sequence of a CDR can be at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to each of the sequences given above.
CLDN18.2 antibody comprising CDRs with amino acid additions, deletions or substitutions
In some embodiments, a CLDN18.2 antibody of the present disclosure comprises at least one immunoglobulin single variable domain (e.g., a VHH), wherein the VHH comprises CDR1, CDR2 and CDR3, and wherein CDR1 is identical in amino acid sequence to SEQ ID NO: 1 there is a difference in amino acid addition, deletion or substitution of no more than 2 amino acids; CDR2 is identical in amino acid sequence to SEQ ID NO: 2 there is a difference in amino acid addition, deletion or substitution of no more than 2 amino acids; and/or CDR3 is identical in amino acid sequence to SEQ ID NO: 3 there is a difference in amino acid addition, deletion or substitution of no more than 2 amino acids. For example, CDR1, CDR2, and CDR3 are identical to SEQ ID NOs: 1. SEQ ID NO: 2 and SEQ ID NO: 3, there is a difference in amino acid addition, deletion or substitution of only one amino acid.
Preferably, the CDRs of the isolated antibody or antigen binding portion thereof contain conservative substitutions of no more than 2 amino acids or no more than 1 amino acid. As used herein, the term "conservative substitution" refers to an amino acid substitution that does not adversely affect or alter the basic properties of the protein/polypeptide comprising the amino acid sequence. For example, conservative substitutions may be introduced by standard techniques known in the art (e.g., site-directed mutagenesis and PCR-mediated mutagenesis). Conservative amino acid substitutions include those in which an amino acid residue is substituted with another amino acid residue having a similar side chain, e.g., a physically or functionally similar residue (e.g., of similar size, shape, charge, chemical properties including the ability to form covalent or hydrogen bonds, etc.) is substituted with the corresponding amino acid residue. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, and histidine), acidic side chains (e.g., aspartic acid and glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, the corresponding amino acid residue is preferably substituted with another amino acid residue from the same side chain family. Methods for identifying conservative substitutions of amino acids are well known in the art (see, e.g., Brummell et al, biochem.32:1180-1187 (1993); Kobayashi et al, Protein Eng.12(10):879-884 (1999); and Burks et al, Proc. Natl. Acad. Sci. USA94:412-417(1997), which are incorporated herein by reference).
In certain embodiments, the immunoglobulin single variable domain of a CLDN18.2 antibody comprises:
i) as shown in SEQ ID NO: 1, CDR1 of the amino acid sequence set forth in seq id no;
ii) of the formula ISRGGX1T CDR2 of the amino acid sequence shown in, wherein X1Is T or S; and
iii) formula NAQWDX2GTX3CDR3 of the amino acid sequence shown by RYLEV, wherein X2Is P or V, X3Is F or I.
CDR-containing CLDN18.2 antibodies
In some embodiments, a CLDN18.2 antibody of the present disclosure comprises at least one immunoglobulin single variable domain (e.g., a VHH), wherein the VHH comprises CDR1, CDR2 and CDR3, and wherein CDR1, CDR2 and CDR3 are selected from the group consisting of:
(a) comprises the amino acid sequence of SEQ ID NO: 1, comprising the amino acid sequence set forth in SEQ ID NO: 2 and a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 3, CDR3 of the amino acid sequence set forth in seq id no;
(b) comprises the amino acid sequence of SEQ ID NO: 1, comprising the amino acid sequence set forth in SEQ ID NO: 4 and a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 5 CDR3 of the amino acid sequence set forth in seq id no; or
(c) Comprises the amino acid sequence of SEQ ID NO: 1, comprising the amino acid sequence set forth in SEQ ID NO: 2 and a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 6, CDR3 of the amino acid sequence set forth in seq id no.
In some embodiments, a CLDN18.2 antibody of the present disclosure comprises at least one immunoglobulin single variable domain (e.g., a VHH), wherein the VHH comprises CDR1, CDR2 and CDR3, and wherein CDR1, CDR2 and CDR3 are selected from the group consisting of:
(a) As shown in SEQ ID NO: 1, as shown in SEQ ID NO: 2 and the CDR2 of the amino acid sequence shown in SEQ ID NO: 3, CDR3 of the amino acid sequence set forth in seq id no;
(b) as shown in SEQ ID NO: 1, as shown in SEQ ID NO: 4 and the CDR2 of the amino acid sequence shown in SEQ ID NO: 5 CDR3 of the amino acid sequence set forth in seq id no; or
(c) As shown in SEQ ID NO: 1, as shown in SEQ ID NO: 2 and the CDR2 of the amino acid sequence shown in SEQ ID NO: 6, CDR3 of the amino acid sequence set forth in seq id no.
CLDN18.2 antibody defined by VHH sequence
In some embodiments, a CLDN18.2 antibody of the present disclosure comprises at least one (e.g., one) immunoglobulin single variable domain (e.g., VHH), wherein the VHH comprises:
(A) SEQ ID NO: 7;
(B) and SEQ ID NO: 7 amino acid sequence that is at least 80%, 85%, 90% or 95% identical; or
(C) And SEQ ID NO: 7 has one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions, and/or substitutions as compared to 7.
In some embodiments, a CLDN18.2 antibody of the present disclosure comprises at least one (e.g., one) immunoglobulin single variable domain (e.g., VHH), wherein the VHH:
(A) Consisting of SEQ ID NO: 7;
(B) consisting of a nucleotide sequence identical to SEQ ID NO: 7 amino acid sequence that is at least 80%, 85%, 90% or 95% identical; or
(C) Consists of a nucleotide sequence identical to SEQ ID NO: 7 has one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions, and/or substitutions.
In other embodiments, the amino acid sequence of the VHH may be at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to each of the sequences described above. As an illustrative example, an antibody may comprise an amino acid sequence identical to SEQ ID NO: 7 VHHs having at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity.
In some further embodiments, CLDN18.2 antibodies of the present disclosure may comprise conservative substitutions or modifications of amino acids in the heavy chain variable region. It is understood in the art that certain conservative sequence modifications may be made that do not eliminate the antigen-binding properties. See, e.g., Brummell et al (1993) Biochem32: 1180-8; de Wildt et al (1997) prot.Eng.10: 835-41; komissarov et al (1997) J.biol.chem.272: 26864-26870; hall et al (1992) J.Immunol.149: 1605-12; kelley and O' Connell (1993) biochem.32: 6862-35; Adib-Conquy et al (1998) int. Immunol.10:341-6 and Beers et al (2000) Clin. Can. Res.6: 2835-43.
In certain embodiments, single domain CLDN18.2 antibodies of the present disclosure are modified (e.g., amino acid substitutions) at one or more of the following positions of SEQ ID NO: 7: amino acid 1, 4, 5, 14, 16, 35, 47, 56, 58, 65, 92, 102, 105 or 121.
In certain embodiments, the variable region of a single domain CLDN18.2 antibody of the present disclosure comprises any one of SEQ ID NOs 7-14.
In certain embodiments, the variable region of a single domain CLDN18.2 antibody of the present disclosure consists of any one of SEQ ID NOs 7-14.
Nucleic acid molecules encoding the antibodies of the disclosure
In some aspects, the invention relates to an isolated nucleic acid molecule comprising a nucleic acid sequence encoding a CLDN18.2 antibody of the present disclosure.
The nucleic acids of the invention can be obtained using standard molecular biology techniques. For antibodies obtained from immunoglobulin gene libraries (e.g., using phage display technology), nucleic acids encoding such antibodies can be recovered from the gene library.
Exemplary nucleic acid molecules of the invention have the sequence shown in SEQ ID NO: 22-29, respectively.
SEQ ID NO:22:
GAGGTGCAGGTGCAGGAGTCTGGGGGAGGCCTGGTACAGGCTGGGACCTCTCTGAGACTCTCCTGTGCAGCCTCTGGCAACATCTTCCGTATCGATACCATGGGCTGGTACCGCCAGGCTCCAGGAAAGCAGCGCGAGTTGGTCGCAGGTATTTCTCGTGGTGGTACAACAACCTATGCACACTCCGTGAAGGAACGATTCACCATCTCCAGAGACAACGCCAAGAACACGATGTATCTGCAAATGAACAGCCTGAAATCTGAGGACACGGCCGGCTATTATTGTAATGCACAGGCGTGGGATCCTGGTACATTTCGGTATCTCGAAGTTTGGGGCCAGGGCACCCTGGTCACTGTCTCA
SEQ ID NO:23:
GAGGTGCAGTTGGTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCTCTGAGACTCTCCTGTGCAGCCTCTGGCAACATCTTCCGTATCGATACCATGGGCTGGTACCGCCAGGCTCCAGGAAAGCAGCGCGAGTTCGTCGCAGGTATTAGTCGTGGTGGTAGCACAAACTATGCACACTCCGTGAAGGAACGATTCACCATCTCCAGAGACAACGCCAAGAACACGATGTATCTGCAAATGAACAGCCTGAAATCTGAGGACACGGCCGGCTATTATTGTAATGCACAGGCGTGGGATCCTGGTACAATCCGGTATCTCGAAGTTTGGGGCCAGGGCACCCTGGTCACTGTCTCA
SEQ ID NO:24:
GAGGTGCAGGTGCAGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCTCTGAGACTCTCCTGTGCAGCCTCTGGCAACATCTTCCGTATCGATACCATGGGCTGGTACCGCCAGGCTCCAGGAAAGCAGCGCGAGTTGGTCGCAGGTATTTCTCGTGGTGGTACAACAACCTATGCACACTCCGTGAAGGAACGATTCACCATCTCCAGAGACAACGCCAAGAACACGATGTATCTGCAAATGAACAGCCTGAAATCTGAGGACACGGCCGGCTATTATTGTAATGCACAGGCGTGGGATCCTGGTACATTTCGGTATCTCGAAGTTTGGGGCCAGGGCACCCTGGTCACTGTCTCA
SEQ ID NO:25:
CAGGTGCAGCTCGTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCTCTGAGACTCTCCTGTGCAGCCTCTGGCAACATCTTCCGTATCGATACCATGGGCTGGTACCGCCAGGCTCCAGGAAAGCAGCGCGAGTTCGTCGCAGGTATTAGTCGTGGTGGTAGCACAAACTATGCACACTCCGTGAAGGAACGATTCACCATCTCCAGAGACAACGCCAAGAACACGATGTATCTGCAAATGAACAGCCTGAAATCTGAGGACACGGCCGGCTATTATTGTAATGCACAGGCGTGGGATCCTGGTACAATCCGGTATCTCGAAGTTTGGGGCCAGGGCACCCTGGTCACCGTGTCCTCA
SEQ ID NO:26:
CAGGTGCAGCTCGTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCTCTGAGACTCTCCTGTGCAGCCTCTGGCAACATCTTCCGTATCGATACCATGGGCTGGTACCGCCAGGCTCCAGGAAAGCAGCGCGAGTTGGTCGCAGGTATTTCTCGTGGTGGTACAACAACCTATGCACACTCCGTGAAGGAACGATTCACCATCTCCAGAGACAACGCCAAGAACACGATGTATCTGCAAATGAACAGCCTGAAATCTGAGGACACGGCCGGCTATTATTGTAATGCACAGGCGTGGGATCCTGGTACATTTCGGTATCTCGAAGTTTGGGGCCAGGGCACCCTGGTCACTGTCTCA
SEQ ID NO:27:
GAGGTGCAGTTGGTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCTCTGAGACTCTCCTGTGCAGCCTCTGGCAACATCTTCCGTATCGATACCATGGTGTGGTACCGCCAGGCTCCAGGAAAGCAGCGCGAGTTGGTCGCAGGTATTTCTCGTGGTGGTACCACAAACTATGCACACTCCGTGAAGGGCCGATTCACCATCTCCAGAGACAACGCCAAGAACACGATGTATCTGCAAATGAACAGCCTGAAATCTGAGGACACGGCCACGTATTATTGTAATGCACAGGCGTGGGATGTTGGTACAATCCGGTATCTCGAAGTTTGGGGCCAGGGCACCCTGGTCACTGTCTCA
SEQ ID NO:28:
GAGGTGCAGCTGCAGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCTCTGAGACTCTCCTGTGCAGCCTCTGGCAACATCTTCCGTATCGATACCATGGGCTGGTACCGCCAGGCTCCAGGAAAGCAGCGCGAGTTCGTCGCAGGTATTAGTCGTGGTGGTAGCACAAACTATGCACACTCCGTGAAGGAACGATTCACCATCTCCAGAGACAACGCCAAGAACACGATGTATCTGCAAATGAACAGCCTGAAATCTGAGGACACGGCCGGCTATTATTGTAATGCACAGGCGTGGGATCCTGGTACAATCCGGTATCTCGAAGTTTGGGGCCAGGGCACCCTGGTCACTGTCTCA
SEQ ID NO:29:
GAGGTGCAGGTCGTGGAGTCTGGGGGAGGCTTGGTACAGCCTGGGGGGTCTCTGAGACTCTCCTGTGCAGCCTCTGGCAACATCTTCCGTATCGATACCATGGTGTGGTACCGCCAGGCTCCAGGAAAGCAGCGCGAGTTGGTCGCAGGTATTTCTCGTGGTGGTACCACAAACTATGCACACTCCGTGAAGGGCCGATTCACCATCTCCAGAGACAACGCCAAGAACACGATGTATCTGCAAATGAACAGCCTGAAATCTGAGGACACGGCCACGTATTATTGTAATGCACAGGCGTGGGATGTTGGTACAATCCGGTATCTCGAAGTTTGGGGCCAGGGCACCCTGGTCACTGTCTCA
In some embodiments, the nucleic acids are identical to SEQ ID NOs: 22-29 has at least 80% (e.g., at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) sequence identity. In some embodiments, the percentage of identity results from the degeneracy of the genetic code, and the encoded protein sequence remains unchanged.
Nucleic acid molecules encoding the CLDN18.2 antibodies of the present disclosure can be inserted into vectors for further cloning (amplification of DNA) or for expression using recombinant techniques known in the art. Many vectors are available. The carrier or carrier component typically includes, but is not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter (e.g., SV40, CMV, EF-1. alpha.), and a transcription termination sequence. The selectable marker gene facilitates selection of host cells into which the vector has been introduced (see, e.g., U.S. Pat. Nos. 4,399,216; 4,634,665 and 5,179,017). For example, typically a selectable marker gene confers resistance to a drug (e.g., G418, hygromycin or methotrexate) on a host cell into which the vector has been introduced. In one embodiment, the selectable marker genes may include the dihydrofolate reductase (DHFR) gene (for DHFR-host cells with methotrexate selection/amplification) and the neo gene (for G418 selection). In another embodiment, the antibody may be produced by homologous recombination as known in the art. DNA encoding a monoclonal antibody is readily isolated and sequenced using conventional methods (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy chain of the antibody).
In some embodiments, vector systems include mammalian, bacterial, yeast systems and the like, and include plasmids such as, but not limited to, pALTER, pBAD, pcDNA, pCal, pL, pET, pGEMEX, pGEX, pCI, pCMV, pEGFP, pEGFT, pSV2, pFUSE, pVITRO, pVIVO, pMAL, pMONO, pSELECT, pUNO, pUO, Psg5L, pBABE, pWPXL, pBI, p15TV-L, pPro18, pTD, pRS420, pLexA, pACT2.2 and the like, as well as other laboratory and commercially available vectors. Suitable vectors may include plasmids or viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses). In one embodiment of the invention, the vector may be pET, for example pETbac containing a hexahistidine tag and a c-Myc-tag gene.
A vector comprising a nucleic acid sequence encoding a CLDN18.2 antibody of the present disclosure may be introduced into a host cell for cloning or gene expression. Suitable host cells for cloning or expressing the DNA in the vectors herein are prokaryotes, yeast or higher eukaryote cells. Suitable prokaryotes for this purpose include eubacteria, for example gram-negative or gram-positive organisms, for example Enterobacteriaceae such as Escherichia, for example Escherichia coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, for example Salmonella typhimurium, Serratia, for example Serratia marcescens and Shigella, and Bacillus such as Bacillus subtilis and Bacillus licheniformis, Pseudomonas such as Pseudomonas aeruginosa and Streptomyces.
In addition to prokaryotes, eukaryotic microorganisms such as filamentous fungi or yeast are suitable host cells for expression of CLDN18.2 antibodies of the present disclosure. Saccharomyces cerevisiae or common baker's yeast are the most commonly used among lower eukaryotic host microorganisms. However, many other genera, species and strains are commonly available and useful in the present invention, such as Schizosaccharomyces pombe (Schizosaccharomyces pombe); kluyveromyces (Kluyveromyces) hosts, such as Kluyveromyces lactis (K.lactis), Kluyveromyces fragilis (K.fragilis) (ATCC12,424), Kluyveromyces bulgaricus (K.bulgaricus) (ATCC16,045), Kluyveromyces wilsonii (K.wickramii) (ATCC 24,178), K.wallidii (ATCC56,500), Kluyveromyces drosophilus (K.drosophilarum) (ATCC36,906), Kluyveromyces thermotolerans (K.thermotolerans), and Kluyveromyces marxianus (K.marxianus); yarrowia (EP 402,226); pichia pastoris (EP 183,070); candida (Candida); trichoderma reesia (EP 244,234); neurospora crassa (Neurospora crassa); schwanniomyces such as Schwanniomyces occidentalis (schwanniomyces occidentalis); and filamentous fungi, such as Neurospora (Neurospora), Penicillium (Penicillium), Tolypocladium, and aspergillus hosts such as aspergillus nidulans (a. nidulans) and aspergillus niger (a. niger).
Other suitable host cells for expression of the CLDN18.2 antibodies of the present disclosure are derived from multicellular organisms. Examples of invertebrate cells include plant and insect cells. A large number of baculovirus strains and variants and corresponding permissive insect host cells have been identified to date from: spodoptera Frugiperda (Spodoptera Frugiperda), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruit fly), and Bombycis mori (Bombyx mori). Various viral strains for transfection are publicly available, for example the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and these viruses can be used in transfection procedures for expression of CLDN18.2 antibodies in suitable host cells, in particular for transfection of Spodoptera frugiperda cells, according to the present disclosure. Plant cell cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco may also be used as hosts.
Host cells were transformed with the above-described vectors for CLDN18.2 antibody production of the present disclosure and cultured in conventional nutrient media modified as necessary for inducing promoters, selecting transformants, or amplifying genes encoding desired sequences.
Host cells for producing CLDN18.2 antibodies of the present disclosure can be cultured in various media. Commercially available media such as Ham's F10(Sigma), Minimum Essential Medium (MEM), (Sigma), RPMI-1640(Sigma) and Dulbecco's Modified Eagle's Medium (DMEM, Sigma) are suitable for culturing the host cells. In addition, Ham et al, meth.Enz.58:44(1979), Barnes et al, anal. biochem.102:255(1980), U.S. Pat. No.4,767, 704; 4,657,866; 4,927,762; 4,560,655, respectively; or 5,122,469; WO 90/03430; WO 87/00195; or any of the media described in U.S. Pat. re.30,985 may be used as the medium for the host cell. Hormones and/or other growth factors (such as insulin, transferrin or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymine), antibiotics (such as GENTAMYCIN drugs), trace elements (defined as inorganic compounds, usually present in final concentrations in the micromolar range) and glucose or equivalent energy sources may be added to any of these media as necessary. Any other necessary supplements may also be included in appropriate concentrations known to those skilled in the art. Culture conditions such as temperature, pH, and the like are those used with the host cell previously selected for expression, and will be apparent to the ordinarily skilled artisan.
When recombinant techniques are used, the antibodies can be produced intracellularly, in the periplasmic space, or directly secreted into the culture medium. If the antibody is produced intracellularly, as a first step, particulate debris (host cells or lysed fragments) is removed, for example by centrifugation or ultrafiltration. Carter et al, Bio/Technology 10: 163-167(1992) describes a method for isolating antibodies secreted into the periplasmic space of E.coli. Briefly, cells were thawed in the presence of sodium acetate (pH3.5), EDTA, and phenylmethylsulfonyl fluoride (PMSF) within about 30 minutes. Cell debris can be removed by centrifugation. In the case of secretion of the antibody into the culture medium, the supernatant from such an expression system is usually first concentrated using a commercially available protein concentration filter, such as an Amicon or Millipore Pellicon ultrafiltration unit. Protease inhibitors such as PMSF may be included in any of the foregoing steps to inhibit proteolysis, and antibiotics may be included to prevent the growth of adventitious contaminants.
Antibodies prepared from cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, DEAE-cellulose ion exchange chromatography, ammonium sulfate precipitation, salting out, and affinity chromatography, with affinity chromatography being a preferred purification technique.
After any preliminary purification step, the mixture comprising the target antibody and contaminants may be subjected to low pH hydrophobic interaction chromatography using an elution buffer having a pH between about 2.5-4.5, preferably at low salt concentrations (e.g., about 0-0.25M salt).
Pharmaceutical composition
In some aspects, the present invention relates to a pharmaceutical composition comprising at least one CLDN18.2 binding molecule as disclosed herein (e.g., a CLDN18.2 antibody of the present disclosure) and a pharmaceutically acceptable carrier.
Components of pharmaceutical compositions
The pharmaceutical composition may optionally contain one or more additional pharmaceutically active ingredients, such as another antibody or drug. The pharmaceutical composition of the present invention may also be administered in combination with, for example, another immunostimulant, an anti-cancer agent, an anti-viral agent or a vaccine, such that the anti-CLDN 18.2 antibody enhances the immune response to the vaccine. The pharmaceutically acceptable carrier may include, for example, a pharmaceutically acceptable liquid, gel or solid carrier, aqueous medium, non-aqueous medium, antimicrobial agent, isotonic agent, buffer, antioxidant, anesthetic, suspending/dispersing agent, chelating agent, diluent, adjuvant, excipient or nontoxic auxiliary substance, a combination of various components known in the art or more.
Suitable components may include, for example, antioxidants, fillers, binders, disintegrants, buffers, preservatives, lubricants, flavorants, thickening agents, colorants, emulsifiers, or stabilizers such as sugars and cyclodextrins. Suitable antioxidants may include, for example, methionine, ascorbic acid, EDTA, sodium thiosulfate, platinum, catalase, citric acid, cysteine, thioglycerol, thioglycolic acid, mercaptosorbitol, butyl methyl anisole, butylated hydroxytoluene, and/or propyl arsenate. As disclosed herein, the antibody or antigen-binding fragment of the disclosed compositions may be oxidized in a solvent containing the antibody or antigen-binding fragment of the disclosed composition comprising one or more antioxidants, such as methionine, that reduce the antibody or antigen-binding fragment thereof. Redox can prevent or reduce the decrease in binding affinity, thereby enhancing antibody stability and extending shelf life. Thus, in some embodiments, the present invention provides compositions comprising one or more antibodies or antigen-binding fragments thereof and one or more antioxidants, such as methionine. The invention further provides methods wherein the antibody or antigen-binding fragment thereof is mixed with one or more antioxidants, such as methionine. Thus, the antibody or antigen-binding fragment thereof may be prevented from oxidation, to extend its shelf-life and/or increase activity.
To further illustrate, pharmaceutically acceptable carriers can include, for example, aqueous vehicles such as sodium chloride injection, ringer's injection, isotonic dextrose injection, sterile water injection or dextrose and lactate ringer's injection, non-aqueous vehicles such as fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil or peanut oil, antimicrobials at bacteriostatic or fungistatic concentrations, isotonicity agents such as sodium chloride or glucose, buffering agents such as phosphate or citrate buffering agents, antioxidants such as sodium bisulfate, local anesthetics such as procaine hydrochloride, suspending and dispersing agents such as sodium carboxymethylcellulose, hydroxypropylmethylcellulose or polyvinylpyrrolidone, emulsifying agents such as polysorbate 80(TWEEN-80), chelating agents such as EDTA (ethylenediaminetetraacetic acid) or EGTA (ethylene glycol tetraacetic acid), ethanol, polyethylene glycols, propylene glycol, sodium hydroxide, hydrochloric acid, citric acid or lactic acid. Antimicrobial agents used as carriers may be added to the pharmaceutical composition in a multi-dose container containing phenol or cresol, mercurial, benzyl alcohol, chlorobutanol, methyl and propyl parabens, thimerosal, benzalkonium chloride and benzethonium chloride. Suitable excipients may include, for example, water, saline, dextrose, glycerol or ethanol. Suitable non-toxic auxiliary substances may include, for example, wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers or agents such as sodium acetate, sorbitan monolaurate, triethanolamine oleate or cyclodextrins.
Administration, formulation and dosage
The pharmaceutical compositions of the present invention may be administered in vivo to a subject in need thereof by various routes including, but not limited to, oral, intravenous, intraarterial, subcutaneous, parenteral, intranasal, intramuscular, intracranial, intracardiac, intraventricular, intratracheal, buccal, rectal, intraperitoneal, intradermal, topical, transdermal and intrathecal, or by implantation or inhalation. The pharmaceutical composition of the present invention may be formulated into solid, semisolid, liquid or gaseous form; including but not limited to tablets, capsules, powders, granules, ointments, solutions, suppositories, enemas, injections, inhalants and aerosols. The appropriate formulation and route of administration may be selected according to the intended application and treatment regimen.
Suitable formulations for enteral administration include hard or soft gelatin capsules, pills, tablets, including coated tablets, elixirs, suspensions, syrups or inhalants and controlled release dosage forms thereof.
Formulations suitable for parenteral administration (e.g., by injection) include aqueous or non-aqueous, isotonic, pyrogen-free, sterile liquids (e.g., solutions, suspensions) in which the active ingredient is dissolved, suspended or otherwise provided (e.g., in liposomes or other microparticles). These liquids may additionally contain other pharmaceutically acceptable ingredients such as antioxidants, buffers, preservatives, stabilizers, bacteriostats, suspending agents, thickening agents, and solutes that render the formulation isotonic with the blood (or other relevant bodily fluids) of the intended recipient. Examples of excipients include, for example, water, alcohols, polyols, glycerol, vegetable oils, and the like. Examples of isotonic carriers suitable for use in such formulations include sodium chloride injection, ringer's solution or lactated ringer's injection. Similarly, the particular dosage regimen (i.e., dose, time and repetition) will depend on the particular individual and the individual's medical history as well as empirical considerations such as pharmacokinetics (e.g., half-life, clearance, etc.).
The frequency of administration can be determined and adjusted during the course of treatment and is based on reducing the number of proliferating or tumorigenic cells, maintaining such a reduction in tumor cells, reducing proliferation of tumor cells or delaying the development of metastases. In some embodiments, the dose administered may be adjusted or reduced to control potential side effects and/or toxicity. Alternatively, sustained continuous release formulations of the pharmaceutical compositions of the invention for use in therapy may be suitable.
One skilled in the art will appreciate that the appropriate dosage may vary from patient to patient. Determining the optimal dosage typically involves balancing the level of therapeutic benefit with any risk or deleterious side effects. The selected dosage level will depend upon a variety of factors including, but not limited to, the activity of the particular compound, the administration, the time of administration, the rate of clearance of the compound, the duration of the treatment, other drugs, compounds and/or materials used in combination, the severity of the condition, and the species, sex, age, weight, condition, general health and prior medical history of the patient. The amount of the compound and the route of administration are ultimately at the discretion of the physician, veterinarian, or clinician, but the dosage is generally selected to achieve a local concentration at the site of action that achieves the desired effect, without causing substantial deleterious or adverse side effects.
Generally, CLDN18.2 binding molecules may be administered in various dosage ranges. In some embodiments, a CLDN18.2 binding molecule provided herein may be administered at a therapeutically effective dose of about 0.01mg/kg to about 100mg/kg (e.g., about 0.01mg/kg, about 0.5mg/kg, about 1mg/kg, about 2mg/kg, about 5mg/kg, about 10mg/kg, about 15mg/kg, about 20mg/kg, about 25mg/kg, about 30mg/kg, about 35mg/kg, about 40mg/kg, about 45mg/kg, about 50mg/kg, about 55mg/kg, about 60mg/kg, about 65mg/kg, about 70mg/kg, about 75mg/kg, about 80mg/kg, about 85mg/kg, about 90mg/kg, about 95mg/kg, or about 100 mg/kg). In certain of these embodiments, the antibody is administered at a dose of about 50mg/kg or less, and in certain of these embodiments, the dose is 10mg/kg or less, 5mg/kg or less, 1mg/kg or less, 0.5mg/kg or less, or 0.1mg/kg or less. In certain embodiments, the dosage administered may vary over the course of treatment. For example, in certain embodiments, the initial administered dose may be higher than the subsequently administered dose. In certain embodiments, the dosage administered may vary over the course of treatment depending on the subject's response.
In any event, the antibodies of the invention or antigen-binding portions thereof are preferably administered to a subject in need thereof as needed. The frequency of administration can be determined by one skilled in the art, for example, by the attending physician based on considerations of the condition being treated, the age of the subject being treated, the severity of the condition being treated, the general health of the subject being treated, and the like.
In certain preferred embodiments, a course of treatment involving an antibody, or antigen-binding portion thereof, of the invention will comprise multiple doses of the selected drug product administered over a period of weeks or months. More specifically, the antibody, or antigen-binding portion thereof, of the invention may be administered daily, every two days, every four days, weekly, every ten days, every two weeks, every three weeks, monthly, every six weeks, every two months, every ten weeks, or every three months. In this regard, it will be appreciated that the dose may be varied or the interval adjusted based on patient response and clinical practice.
The dosage and regimen of the disclosed pharmaceutical compositions for treatment may also be determined empirically in individuals given one or more administrations. For example, an individual may be administered a incremental dose of a pharmaceutical composition as described herein. In selected embodiments, the dosage may be determined empirically, or may be increased or decreased in steps depending on the side effects or toxicity observed, respectively. To assess the efficacy of the selected composition, markers of a particular disease, disorder, or condition can be tracked. For cancer, these include direct measurement of tumor size by palpation or visual observation, indirect measurement of tumor size by X-ray or other imaging techniques; improvement assessed by direct tumor biopsy and microscopy of tumor samples; measuring the reduction in pain or paralysis of an indirect tumor marker (e.g., PSA for prostate cancer) or tumorigenic antigen identified according to the methods described herein; improvement in speech, vision, respiration or other disability associated with the tumor; appetite increase; or an increase in quality of life or an increase in survival as measured by accepted tests. Those skilled in the art will appreciate that the dosage will vary depending on the individual, the type of neoplastic condition, the stage of the neoplastic condition, whether the neoplastic condition has begun to metastasize to other locations in the individual, and the treatment used in the past and the treatment used concurrently.
A compatible formulation for parenteral administration (e.g., intravenous injection) may comprise a CLDN18.2 binding molecule as provided herein at a concentration of about 10 μ g/ml to about 100 mg/ml. In some embodiments, the concentration of a CLDN18.2 binding molecule may comprise 20. mu.g/ml, 40. mu.g/ml, 60. mu.g/ml, 80. mu.g/ml, 100. mu.g/ml, 200. mu.g/ml, 300. mu.g/ml, 400. mu.g/ml, 500. mu.g/ml, 600. mu.g/ml, 700. mu.g/ml, 800. mu.g/ml, 900. mu.g/ml or 1 mg/ml. In other preferred embodiments, the concentration of a CLDN18.2 binding molecule will comprise 2mg/ml, 3mg/ml, 4mg/ml, 5mg/ml, 6mg/ml, 8mg/ml, 10mg/ml, 12mg/ml, 14mg ml, 16mg/ml, 18mg/ml, 20mg/ml, 25mg/ml, 30mg/ml, 35mg/ml, 40mg/ml, 45mg/ml, 50mg/ml, 60mg/ml, 70mg/ml, 80mg/ml, 90mg/ml or 100 mg/ml. If calculated as molarity, in some embodiments, the concentration of a CLDN18.2 binding molecule may include, for example, 133nM, 266nM, 400nM, 533nM, 667nM, 1.3 μ M, 2 μ M, 2.67 μ M, 3.33 μ M, 4 μ M, 4.67 μ M, 5.33 μ M, 6 μ M, or 6.67 μ M.
Application of the invention
The CLDN18.2 binding molecules of the present invention have a number of in vitro and in vivo uses.
Treating diseases
The CLDN 18.2-associated conditions and disorders may be immune-related diseases or disorders including, but not limited to, "diseases involving cells expressing CLDN 18.2" or "diseases associated with cells expressing CLDN 18.2" or similar expressions, meaning that CLDN18.2 is expressed in cells of diseased tissues or organs. In one embodiment, expression of CLDN18.2 is increased in cells of a diseased tissue or organ as compared to the state in a corresponding healthy tissue or organ. By increasing is meant an increase of at least 10%, in particular at least 20%, at least 50%, at least 100%, at least 200%, at least 500%, at least 1000%, at least 10000% or even more. In one embodiment, expression is seen only in diseased tissue, while expression in corresponding healthy tissue is inhibited. According to the present invention, diseases associated with cells expressing CLDN18.2 include cancer diseases. Furthermore, according to the present invention, cancer diseases are preferably those in which cancer cells express CLDN 18.2.
The term "cancer disease" or "cancer" refers to or describes a physiological condition in an individual that is typically characterized by unregulated cell growth. Examples of cancer include, but are not limited to, epithelial cancer, lymphoma, blastoma, sarcoma, and leukemia. More particularly, examples of such cancers include bone cancer, blood cancer, lung cancer, liver cancer, pancreatic cancer, skin cancer, head and neck cancer, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, prostate cancer, uterine cancer, cancer of the sexual and reproductive organs, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the bladder, cancer of the kidney, cancer of the renal cell, cancer of the renal pelvis, tumors of the Central Nervous System (CNS), cancer of the neuroectodermal layer, tumor of the spinal axis, glioma, meningioma and pituitary adenoma. The term "cancer" according to the present invention also includes cancer metastasis. Preferably, a "cancer disease" is characterized by cells expressing CLDN18.2, and cancer cells express CLDN 18.2. Preferably, the cell expressing CLDN18.2 is a cancer cell, preferably a cancer cell of a cancer as described herein.
According to the present disclosure, the term "tumor" or "tumor disease" refers to an abnormal growth of cells (referred to as neoplastic cells, tumor-initiating cells or tumor cells), preferably forming a swelling or lesion. By "tumor cell" is meant an abnormal cell that grows by rapid uncontrolled cell proliferation and continues to grow after the stimulus that initiated the new growth ceases. Tumors show a partial or complete loss of structural tissue and functional coordination with normal tissue, and often form distinct tissue masses, which may be benign, premalignant, or malignant. According to the present invention, a "cancer disease" is preferably a "tumor disease". However, in general, the terms "cancer" and "tumor" are used interchangeably herein.
In one embodiment, the cancer according to the present disclosure relates to cancer cells expressing CLDN 18.2. In one embodiment, the cancer is CLDN18.2 positive. In one embodiment, expression of CLDN18.2 is at the surface of a cell. In one embodiment, at least 50%, preferably 60%, 70%, 80% or 90% of the cancer cells are CLDN18.2 positive and/or at least 40%, preferably at least 50% of the cancer cells are positive for surface expression of CLDN 18.2. In one embodiment, at least 95% or at least 98% of the cancer cells are CLDN18.2 positive. In one embodiment, at least 60%, at least 70%, at least 80% or at least 90% of the cancer cells are positive for surface expression of CLDN 18.2.
In one embodiment, the CLDN18.2 expressing cancer, cancer involving CLDN18.2 expressing cancer cells, or CLDN18.2 positive cancer is selected from the group consisting of: gastric cancer, esophageal cancer, pancreatic cancer, lung cancer (e.g., non-small cell lung cancer (NSCLC)), ovarian cancer, colon cancer, liver cancer, head-neck cancer, and cancer of the gallbladder, and metastases thereof, particularly gastric cancer metastasis (e.g., krukenberg tumor), peritoneal metastasis, and lymph node metastasis. In one embodiment, the cancer is an adenocarcinoma, in particular an advanced adenocarcinoma. Particularly preferred cancer diseases are adenocarcinoma of the stomach, esophagus, pancreatic duct, bile duct, lung and ovary. In one embodiment, the cancer is selected from gastric cancer, esophageal cancer (particularly lower esophageal cancer), cancer of the esophageal-gastric junction, and gastroesophageal cancer. In a particularly preferred embodiment, the cancer is gastroesophageal cancer, such as metastatic, refractory or recurrent advanced gastroesophageal cancer.
Furthermore, the antibodies or antigen binding portions thereof of the present disclosure can be used in combination with chemotherapy or radiotherapy.
Used in combination with chemotherapy
The antibody or antigen binding portion thereof can be used in combination with an anti-cancer agent, cytotoxic agent, or chemotherapeutic agent.
The term "anti-cancer agent" or "anti-proliferative agent" means any agent useful in the treatment of cell proliferative disorders such as cancer, and includes, but is not limited to, cytotoxic agents, cytostatic agents, anti-angiogenic agents, radiation therapy and radiotherapeutic agents, targeted anti-cancer agents, BRMs, therapeutic antibodies, cancer vaccines, cytokines, hormonal therapy, radiation therapy and anti-metastatic agents, and immunotherapeutic agents. It is to be understood that in selected embodiments as described above, such anti-cancer agents may comprise conjugates and may be conjugated to the disclosed site-specific antibodies prior to administration. More specifically, in some embodiments, a selected anticancer agent is linked to an unpaired cysteine of an engineered antibody to provide an engineered conjugate as described herein. Accordingly, such engineered conjugates are expressly contemplated within the scope of the present invention. In other embodiments, the disclosed anti-cancer agents will be administered in combination with site-specific conjugates comprising different therapeutic agents as described above.
As used herein, the term "cytotoxic agent" refers to a substance that is toxic to cells and reduces or inhibits cell function and/or causes cell destruction. In some embodiments, the agent is a naturally occurring molecule derived from a living organism. Examples of cytotoxic agents include, but are not limited to, small molecule toxins or enzymatically active toxins of bacteria (e.g., diphtheria toxin, pseudomonas endotoxin and exotoxin, staphylococcal enterotoxin a), fungi (e.g., alpha-sarcin, restrictocin), plants (abrin, ricin, gelonin, mistletoe, pokeweed antiviral protein, saporin, gelonin, momoridin, trichosanthin, barley toxin, Aleurites fordii protein, dianthin protein, phytacca merica protein (PAPI, PAPII and PAP-S), momordica charantia inhibitors, leprosy toxin, croton toxin, alkannin inhibitors, gelonin, mitegellin, restrictocin, phenomycin, neomycin and trichothecene family compounds) or animals (e.g., cytotoxic rnases, such as extracellular pancreatic rnases; dnase I, including fragments and/or variants thereof).
For purposes of the present invention, "chemotherapeutic agents" include chemical compounds (e.g., cytotoxic or cytostatic agents) that non-specifically reduce or inhibit the growth, proliferation, and/or survival of cancer cells. These chemical agents are generally directed to intracellular processes required for cell growth or division and are therefore particularly effective for cancer cells which generally grow and divide rapidly. For example, vincristine depolymerizes microtubules, thereby inhibiting the cells from entering mitosis. In general, a chemotherapeutic agent may include any chemical agent that inhibits or is designed to inhibit a cancer cell or a cell that may become sexually or produce tumorigenic progeny (e.g., TIC). These agents are often used in combination and are often most effective, for example, in regimens such as CHOP or FOLFIRI.
Examples of anticancer agents that can be used in combination with the site-specific constructs of the invention (either as a component of the site-specific conjugate or in an unconjugated state) include, but are not limited to, alkylating agents, alkyl sulfonates, aziridines, ethyleneimines, and methyl melamines, polyacetyls (acetogenins), haplotypesArboline, bryostatin, calicheastatin (calalysitin), CC-1065, kretoxin (cryptophycins), dolastatin, duocarmycin, auristatin (eleutherobin), coprinum, saxodidin (sarcodictyin), spongistatin (spongistatin), mechlorethamine, antibiotics, enediynes, dynemicin, bisphosphonates, epothilones, chromoproteenediyne antibiotic chromophores, aclacinomycin (aclacinomysins), actinomycin, apramycin, azathionine, bleomycin, actinomycin C, carabine (carabicin), carminomycin, carvachromycin, chromomycins (chromomycins), dactinomycin, dirotoxin, 6-diazo-5-oxo-L-norleucine,
Figure BDA0002071082450000382
Doxorubicin, epirubicin, esorubicin, idarubicin, sisomicin, mitomycin, mycophenolic acid, nogomycin, olivomycin, pelomycin, bodhimycin (potfiromycin), puromycin, triiron doxorubicin, roxobicin, streptonigrin, streptozotocin, tubercidin, ubenimex, setastatin, zorubicin; anti-metabolites, erlotinib, vemurafenib, crizotinib, sorafenib, ibrutinib, enzalutamide, folic acid analogues, purine analogues, androgens, anti-epinephrine, folic acid supplements such as furinic acid (frillinic acid), acetoglucuronide, aldphosphoramide glycoside, aminoacetylpropionic acid, eniluracil, amsacrine, bestirubucil, bisantrene, edatrexate, diffamine, colchicine, diaquazone, efonicine (elfornithine), ethacridine, epothilonone, etoglutacol, gallium nitrate, hydroxyurea, lentinan, lonidamine, maytansinoids (maytansinoids), mitoguazone, mitoxantrone, motdan (mopidanmol), nitrene (nitine), staudine, mechlorethamine, pirimidone, hydrazine, carbohydrazide, 2-ethyl carbazone, etoposide, foscarnitinib, gecin, gefitinib, gefinicin, gefitinib, gevazide, gefitinib, gevazide, gevafloxacin, gevazide, valbutine, gevazide, gevafloxacin, valbutine, valbutrine, valdecoxidone, gevazide, valbenvabenvazide, valdecoxidone, valdecoxine, valbutraline, valbutrine, valbutine, valdecoxidone, valbutine, valdecoxidone, valbutraline, valbutine, valbutraline, valdecoxidone, valnemadene, valdecoxidone, valbutine, valbutraline, valdecoxidone, valdecoxi,
Figure BDA0002071082450000383
Polysaccharide complexes (JHS Natural Products, Eugene, OR), Razoxan;rhizomycin; a texaphyrin; a germanium spiroamine; (ii) zonecanoic acid; a tri-imine quinone; 2,2' -trichlorotriethylamine; trichothecenes (especially T-2 toxin, Verlucurin A (verracurin A), bacillocin A and snakeheaded; uratan; vindesine; dacarbazine; mannomustine; dibromomannitol; dibromodulcitol; pipobroman; cassitoxin (gacytosine); arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxanes; chlorambucil (chlorenbucil);
Figure BDA0002071082450000381
gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; a platinum analog; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; the concentration of the vincristine is controlled by the concentration of the vincristine,
Figure BDA0002071082450000394
vinorelbine; noxiaoling; (ii) teniposide; edatrexae; daunorubicin; aminopterin; (ii) Hirodad; ibandronate; irinotecan (Camptosar, CPT-11); topoisomerase inhibitor RFS 2000; difluoromethyl ornithine; a retinoid; capecitabine; combretastatin; leucovorin; oxaliplatin; an inhibitor of PKC-alpha, Raf, H-Ras, EGFR, and VEGF-A (which reduces cell proliferation), and a pharmaceutically acceptable salt, acid, or derivative of any of the foregoing. Also included in this definition are anti-hormonal agents used to modulate or inhibit hormonal effects on tumors, such as anti-estrogens and selective estrogen receptor modulators, aromatase inhibitors that inhibit aromatase that regulates estrogen production in the adrenal glands, and anti-androgens; and troxacitabine (1, 3-dioxolane nucleoside cytosine analogues); antisense oligonucleotides, ribozymes such as inhibitors of VEGF expression and inhibitors of HER2 expression; a vaccine is provided which comprises a vaccine,
Figure BDA0002071082450000391
rIL-2;
Figure BDA0002071082450000392
A topoisomerase 1 inhibitor;
Figure BDA0002071082450000393
rmRH; vinorelbine and epsipromycin, and a pharmaceutically acceptable salt, acid or derivative of any of the foregoing.
Used in combination with radiotherapy
The invention also provides the combination of an antibody or antigen-binding portion thereof with radiotherapy (i.e., any mechanism used to induce DNA damage locally within tumor cells, such as gamma-irradiation, X-ray, UV-irradiation, microwaves, electron emission, etc.). Combination therapies using targeted delivery of radioisotopes to tumor cells are also contemplated, and the disclosed conjugates may be used in conjunction with targeted anti-cancer agents or other targeting means. Typically, radiation therapy is administered in pulses over a period of about 1 to about 2 weeks. Radiation therapy may be administered to a subject with head and neck cancer for about 6 to 7 weeks. Optionally, the radiation therapy may be administered as a single dose or as multiple sequential doses.
Diagnosis of
The invention provides in vitro and in vivo methods for detecting, diagnosing or monitoring proliferative disorders and methods of screening cells from patients to identify tumor cells, including tumorigenic cells. Such methods include identifying an individual having cancer for treatment or monitoring the progression of cancer, comprising contacting the patient or a sample obtained from the patient (in vivo or in vitro) with an antibody described herein, and detecting the presence or absence or level of binding of the bound antibody to bound or free target molecule in the sample. In some embodiments, the antibody will comprise a detectable label or reporter.
In some embodiments, binding of an antibody to a particular cell in a sample can indicate that the sample is likely to contain a tumorigenic cell, thereby indicating that an individual having cancer can be effectively treated with an antibody described herein.
Samples can be analyzed by a variety of assays, such as radioimmunoassays, enzyme immunoassays (e.g., ELISA), competitive binding assays, fluorescent immunoassays, immunoblot assays, Western blot analysis, and flow cytometry assays. Compatible in vivo diagnostic or diagnostic assays may include imaging or monitoring techniques known in the art, such as magnetic resonance imaging, computerized tomography (e.g., CAT scans), positron emission tomography (e.g., PET scans), radiography, ultrasound, and the like, as known to those skilled in the art.
Pharmaceutical pack and kit
The invention also provides pharmaceutical packages and kits comprising one or more containers of one or more doses of an antibody, or antigen-binding portion thereof. In some embodiments, a unit dose is provided, wherein the unit dose contains a predetermined amount of a composition comprising, for example, an antibody or antigen-binding portion thereof, with or without one or more additional agents. For other embodiments, such unit doses are supplied in single use prefilled syringe injections. In other embodiments, the composition contained in a unit dose may comprise saline, sucrose, or the like; buffers such as phosphate and the like; and/or formulated at a stable and effective pH range. Alternatively, in some embodiments, the composition is a conjugate composition, which can be provided as a lyophilized powder and reconstituted after addition of a suitable liquid (e.g., sterile water or saline solution). Any label on or associated with the container indicates that the encapsulated conjugate composition is used to treat the selected neoplastic disease condition. In some preferred embodiments, the composition comprises one or more substances that inhibit protein aggregation, including but not limited to sucrose and arginine.
The invention also provides kits for producing single-dose or multi-dose administration units of the site-specific conjugate and optionally one or more anti-cancer agents. The kit includes a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, and the like. The container can be formed of a variety of materials, such as glass or plastic, and contain a pharmaceutically effective amount of the disclosed conjugates in conjugated or unconjugated form. In other preferred embodiments, the container includes a sterile access port (e.g., the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). Such kits typically comprise a pharmaceutically acceptable formulation of the engineered conjugate in a suitable container, and optionally one or more anti-cancer agents in the same or different containers. The kit may also contain other pharmaceutically acceptable preparations for use in diagnosis or combination therapy. For example, such kits may contain, in addition to an antibody or antigen-binding portion thereof of the invention, any one or more anti-cancer agents, such as chemotherapeutic agents or radiotherapeutic agents; an anti-angiogenic agent; an anti-transfer agent; targeted anti-cancer agents; a cytotoxic agent; and/or other anti-cancer agents.
More specifically, kits can have a single container containing the disclosed antibodies or antigen-binding portions thereof, with or without additional components, or they can have different containers for each desired reagent. Where a combination therapeutic agent is provided for conjugation, the single solutions may be premixed in molar equivalent combinations or in a manner such that one component is more than the other. Alternatively, the conjugate and any optional anti-cancer agent of the kit may be stored separately in separate containers prior to administration to a patient. The kit may also comprise a second/third container means for holding sterile pharmaceutically acceptable buffers or other diluents such as bacteriostatic water for injection (BWFI), Phosphate Buffered Saline (PBS), ringer's solution and dextrose solution.
When the components of the kit are provided as one or more liquid solutions, the liquid solution is preferably an aqueous solution, particularly preferably a sterile aqueous or saline solution. However, the components of the kit may be provided as a dry powder. When the agent or component is provided in dry powder form, the powder may be reconstituted by the addition of a suitable solvent. It is contemplated that the solvent may also be provided in another container.
As briefly mentioned above, the kit may also contain means for administering the antibody, or antigen-binding portion thereof, and any optional components to the patient, such as one or more needles, i.v. bags or syringes, or even eye droppers, pipettes, or other similar devices, through which the formulation may be injected or introduced into the animal or administered to the affected area of the body. The kits of the invention will also typically include a means for holding vials or the like, as well as other tightly closed components for commercial sale, such as injection or blow molded plastic containers, in which the desired vials and other devices are placed and held.
Advantages of the invention
The development of antibodies against CLDN18.2, in particular human CLDN18.2, has at least the following advantages:
firstly, human CLDN18.2 is highly expressed in cancer cells, and is only specifically expressed in gastric epithelial cells in normal cells, so that toxic and side effects are low, and the possibility of drug formation is higher;
second, CLDN18.2 is expressed in the tight junctions of gastric epithelial cells, and antibodies are less likely to act against the looser cancer cells; even if the epithelial cells are killed by the antibody, stem cells below the epithelial cells can complement the damaged gastric epithelial cells by differentiation because they do not express CLDN 18.2;
Third, an antibody against CLDN18.2 can not only kill cells by ADCC and CDC, but also mediate apoptosis of cells by crosslinking CLDN18.2 through the antibody, and can inhibit proliferation of cells to some extent.
To date, no nanobody against CLND18.2 exists, and as a novel antibody, the nanobody has already an antibody caplatizumab sold on the market, which fully explains the druggability of the nanobody. Compared with other antibodies, the antibody has the remarkable advantages that the half-life length can be adjusted through chemical modification or protein fusion transformation, the permeability is strong, hidden epitopes which are inaccessible to common antibodies can be identified, the antibody is resistant to pepsin, acid and heat, is easy to produce, and can be easily assembled into bivalent antibodies and multivalent antibodies with other types of antibodies because of being single-chain.
In some embodiments of the invention, antibodies to CLDN18.2 are screened by an alpaca immune repertoire, based on which the C-terminus is fused to an Fc fragment of IgG1, and the results of the experiments with CDC and ADCC at the cellular level of some of the current candidate antibodies show better or at least comparable activity to the control antibodies. The antibody combines the characteristics of the CLDN18.2 target spot, is used for the immunotherapy of cancer, has lower toxic and side effects and better clinical efficacy, and provides more drug choices for patients.
Examples
The invention generally described herein will be more readily understood by reference to the following examples, which are provided by way of illustration and are not intended to be limiting of the present invention. These examples are not intended to be representative of the experiments that follow being all or only experiments that were conducted.
Example 1
Construction and identification of over-expression cell strain and tumor cell strain
In this example, different types of over-expression cell lines and tumor cell lines were constructed and identified by flow.
1. Construction and identification of over-expressed cell lines
The nucleic acid sequences of full-length human CLDN18.1(SEQ ID NO:16), murine CLDN18.2(SEQ ID NO:17), and murine CLDN18.1(SEQ ID NO:18) were constructed onto the pLVX-puro plasmid (Clontech, Cat # 632164). Then, the resulting plasmid was transformed into HEK293 cells by electroporation (
Figure BDA0002071082450000431
CRL-1573TM) In (1). Overexpression cell lines expressing full-length human CLDN18.1 (human CLDN18.1-HEK293), overexpression cell lines expressing murine CLDN18.2 (murine CLDN18.2-HEK293), and overexpression cell lines expressing murine CLDN18.1 (murine CLDN18.1-HEK293) were obtained by screening. Thereafter, an anti-CLDN 18 antibody [34H14L15 ] recognizing the C-terminal CLDN18 intracellular fragment (GFKASTGFGSNTKN, SEQ ID NO:21) was combined with an IMAB362 antibody (expressed and purified according to the sequence information disclosed in patent US20180127489A 1) ](Abcam, ab203563) were identified by flow cytometry to obtain successfully transformed over-expressed cell lines, as follows.
1.1 electrotransformation
First, HEK293 cells were recovered and cultured for 2-3 serial passages, and the cells were plated at 3X 10 cells the day before transfection5The cells were seeded into cell culture dishes at a density of one/mL and used the next day until the cell confluence reached about 70%. Using Trypsin with EDTA at 0.25% by volume (Gibco,25200-072) digesting the cells for 2min, collecting the cells, centrifuging the cells at room temperature for 5min at 100g, discarding the supernatant, adding 1 XDPBS (source culture, B210) to resuspend the cells, counting, and taking 5 × 106The cells were centrifuged and harvested with 250. mu.L Buffer R (Invitrogen, Neon)TMKit, PK10096) buffer, and 25 μ g of the plasmid of interest was added thereto and gently mixed by a pipette to homogeneity. The suspension was then subjected to electrotransformation in an electrotransfer apparatus (Invitrogen, Neon transfer System, MP922947) under reaction conditions of 1100V/20 ms/2.
1.2 cell culture
After the electrotransformation, the resulting cells were transferred to DMEM medium (Gibco, 11995065) containing 10% FBS (Gibco, 15140-141) by volume and containing no antibiotics, respectively, and then the cells were seeded into 10cm × 10cm cell culture dishes and cultured for 48 hours, and then the cells were dispensed into 96 well cell culture plates at an average density of 0.5/well, puromycin (Gibco, A111138-03) at a final concentration of 2 μ g/mL was added as a selection pressure, the clonal growth of the cell strain was observed for about 2 weeks, and the cell strain forming the clone was picked up and identified.
1.3 flow assay of over-expressed cell lines
The cell line obtained in section 1.2 above was identified by flow cytometry, as follows.
1.3.1 flow identification of human CLDN18.2-HEK293T and murine CLDN18.2-HEK293 cell lines
Human CLDN18.2-HEK293T (Congress Bocreation, KC-0986) and murine CLDN18.2-HEK293 cell lines were identified directly using the IMAB362 antibody.
Take 1X 105Individual cells were centrifuged at low speed (300g) to remove supernatant. The cells at the bottom of the centrifuge tube were rinsed once with a prepared FACS buffer (1 XPBS buffer containing 2% FBS by volume), and then 12.5. mu.g/mL of antibody IMAB362 was added to the rinsed cells, followed by incubation at 4 ℃ for 1 hour, and then rinsed three times with the above FACS buffer, and 0.5. mu.g of PE-labeled goat anti-human IgG Fc antibody (Abcam, ab98596) was added, followed by incubation at 4 ℃ for 1 hour. Thereafter, the cells were rinsed three times with FACS buffer, and 200. mu.L of FACS buffer was added to the cells to resuspend the cells, and finallyDetection was then performed by flow cytometry (Beckman, CytoFLEX AOO-1-1102).
1.3.2 flow identification of human CLDN18.1-HEK293 and murine CLDN18.1-HEK293 cell lines
Since the IMAB362 antibody recognizes only CLDN18.2 but not CLDN18.1, it is necessary to identify human CLDN18.1-HEK293, murine CLDN18.1-HEK293 cell lines in combination with other methods. In view of the sequence identity of human CLDN18.1 and murine CLDN18.1, cells were first fixed and ruptured as described in the cell rupture kit (eBioscience, 88-8824-00) and then identified by flow cytometry based on the recognition of the C-terminal CLDN18 intracellular segment by anti-CLDN 18 antibody [34H14L15] and specific binding to IMAB362 antibody.
The specific method comprises the following steps: take 1X 105Individual cells were centrifuged at low speed (300g) and the supernatant removed. The cells at the bottom of the centrifuge tube were rinsed once with FACS buffer, and then 200 μ L of IC fixative (eBioscience, 00-8222) was added to the rinsed cells, followed by incubation at 4 ℃ for 1 hour, followed by rinsing twice with rupture buffer (eBioscience, 00-8333), followed by addition of the aforementioned anti-CLDN 18 antibody, and incubation at 4 ℃ for 1 hour. After rinsing again three times with the above-mentioned membrane-rupturing buffer, Alexa was added
Figure BDA0002071082450000441
488 fluorescence-labeled donkey anti-rabbit IgG H&L (abcam, ab150073) 0.5. mu.g, incubated at 4 ℃ for 1h and finally detected by flow cytometry (Beckman, Cytoflex AOO-1-1102).
Simultaneously, the binding of human CLDN18.1-HEK293 and murine CLDN18.1-HEK293 cell lines was determined by the IMAB362 antibody using the method described in 1.3.1 above.
The results of the flow assay are shown in FIG. 1 a. As can be seen from fig. 1 a: the anti-CLDN 18 antibody can recognize human CLDN18.2-HEK293T, human CLDN18.1-HEK293, murine CLDN18.2-HEK293, murine CLDN18.1-HEK293, and HEK293T cells; whereas antibody IMAB362 recognizes only murine CLDN18.2-HEK293, human CLDN18.2-HEK293T, and does not recognize murine CLDN18.1-HEK293, human CLDN18.1-HEK 293. This indicates that four cell lines, human CLDN18.2-HEK293T, human CLDN18.1-HEK293, murine CLDN18.2-HEK293, and murine CLDN18.1-HEK293, all successfully expressed the corresponding CLDN18 protein.
2. Construction and identification of over-expression tumor cell strain
The gastric cancer cell line KATOIII overexpressing human CLDN18.2 (human CLDN18.2-KATOIII tumor cell line) was constructed by lentivirus transfection and identified by antibody IMAB 362. The specific method comprises the following steps:
collecting well-conditioned human gastric cancer cell KATOIII
Figure BDA0002071082450000451
HTB-103TM)5×104And (3) the ratio of 30: 1, packaged lentivirus containing the human CLDN18.2 sequence (SEQ ID NO:15), mixed well, then IMDM complete medium (Gibco, 12440061) containing 8 μ g/mL Polybrene (Polybrene, Sigma, 107689) was added, mixed well, 5% CO at 37 ℃2The constant temperature incubator of (1) was incubated for 20 hours, the medium was removed, fresh IMDM complete medium was replaced and incubation was continued for 24 hours, then transfected KATOIII cells were seeded into 96-well plates at an average density of 0.5 cells/well and puromycin was added to a final concentration of 2. mu.g/mL for resistance pressure screening at 37 ℃ with 5% CO2Culturing for 2-3 weeks in a constant temperature incubator, and selecting clones for identification.
The resistance-selected cell lines were flow-identified by the antibody IMAB362 in the same manner as described above in section 1.3.1.
The results of the flow assay are shown in FIG. 1 b. As can be seen from fig. 1b, KATOIII cells that were not transfected with CLDN18.2 were hardly recognized by antibody IMAB362, indicating that the KATOIII cell line expressed little or very little CLDN 18.2; the successfully constructed human CLDN18.2-KATOIII tumor cell line transfected by lentivirus can be recognized by the antibody IMAB362, which indicates that the cell line is successfully constructed.
Example 2
Animal immunization and serum immune titer detection
1. Immunization
In this example, alpaca immunization was used. The specific operation is as follows: the immunogen was cell line human CLDN18.2-HEK293T (Kangyuan)BO-KONG, KC-0986) and hCLDN18.2 ECD1(SEQ ID NO:19) -containing hCLDN18.2 ECD-puro plasmid. Respectively using 2X 107Human CLDN18.2-HEK293T cells (subcutaneously injected in multiple spots) and 2mg plasmid (intramuscularly injected in multiple spots) were immunized alternately in weeks to alpaca (fed by Nanchang Dajia organisms) with alpaca code number NSY002, for a total of 8 immunizations. Finally, use 2X 107Human CLDN18.2-HEK293T cells were immunized.
2. Serum immune titer determination
The measurement of the immune titer is carried out by ELISA method based on the signal of immune serum on antigen recombinant protein CLDN18.2 (Kinseri, CP 0007). The specific method is as follows.
The day before the measurement of the immune titer, the antigen recombinant protein CLDN18.2 was diluted to 1 μ g/mL with PBS to obtain a dilution. mu.L of the dilution was added to an Elisa plate and coated overnight at 4 ℃. On the day of immunotiter determination, the coated plates were rinsed twice with PBS, then blocked with PBST containing 5% skim milk powder by volume at room temperature for two hours, and rinsed twice with PBS. The non-immunized negative serum and post-immunization serum were diluted in PBS on another 96-well dilution plate, 1000-fold dilution in the first well, and 2-fold gradient dilution in the next 7 wells. The diluted serum is correspondingly added into a first Elisa plate coated with antigen recombinant protein CLDN18.2, incubated for 1h at 37 ℃, washed twice by PBS and added with a second antibody MonoRab at a ratio of 1:5000 TMRabbit anti-Camelidae VHH antibodies (King-Share, A01862-200) and finally OD values were read by a microplate reader (Molecular Devices, SpectreMax 190) at a wavelength of 450 nm. As shown in table 1, the alpaca immune titer was approximately 1: about 8000 a.
TABLE 1
Figure BDA0002071082450000471
Example 3
Construction and screening of alpaca immune bank
After completion of the immunization of the animals, 80mL of alpaca blood was collected, and PBMCs were separated by Ficoll-Paque density gradient (GE, 17144003S) for alpaca immune bank construction. The specific method comprises the following steps:
15mL of Ficoll-Paque density gradient separation solution is slowly added into a 50mL centrifuge tube, and then 15mL of collected alpaca blood is slowly added, so that a clear separation interface is kept between the two liquids. Centrifugation was carried out at around 15 ℃ under the following conditions: 400g, 20min, acceleration of 3, deceleration of 0. After centrifugation, the whole liquid surface is divided into four layers, the upper layer is a plasma mixture, the lower layer is red blood cells and granulocytes, the middle layer is a Ficoll-Paque liquid, and a white cloud layer narrow band mainly containing PBMC, namely a PBMC cell layer, is arranged at the junction of the upper layer and the middle layer. Isolated PBMCs were obtained by carefully aspirating the supernatant plasma mixture with a sterile pasteur pipette and then aspirating the PBMCs with a fresh sterile pasteur pipette. The cells were rinsed twice with PBS, centrifuged horizontally at 1500rpm for 10min at 4 ℃ and finally resuspended in 1.5mL PBS and counted by a cell counter (CountStar, CountStar Altair).
The RNA was extracted from the isolated PBMC and reverse-transcribed into cDNA by means of a reverse transcription kit (TaKaRa, 6210A). Because the molecular form of the alpaca antibody is different from that of a common antibody, the alpaca antibody does not contain a light chain and a heavy chain does not contain CH1, two fragments with different sizes are obtained by PCR on the front end of a VH germline gene and CH2 through designing primers, and a smaller target fragment is recovered through tapping; a degenerate primer containing NcoI and NotI enzyme cutting sites is designed by comparing all sequences of all V genes and J genes of a VHH antibody, all VHH genes are amplified by taking a recovered DNA fragment product as a template, and finally a target antibody gene fragment is inserted into a phage display carrier through double enzyme cutting and connection, wherein the VHH fuses GIII genes at the C end. The ligation product was recovered by using a recovery kit (Omega, D6492-02), transformed into competent E.coli SS320(Lucigen, MC 1061F) by an electrotransfer instrument (Bio-Rad, MicroPulser), and plated on 2-YT solid plates (prepared from tryptone 1.5%, yeast extract 1%, NaCl0.5%, agar 1.5%, in terms of mass volume g/mL) having ampicillin resistance. For the calculation of the storage capacity, the total number of clones formed by all the electrotransformations, i.e., the storage capacity, was calculated from the clones formed on the plate after dilution with 1. mu.L of the bacterial suspension. The immune pool has a size of 1 × 10 9cfu。
Based on storage capacityCapacity, 50 OD (5X 10 OD in 1)8cfu) was added to fresh 2-YT broth to give an initial OD of 0.05. Culturing at 37 deg.C and 220rpm to logarithmic phase, adding VSCM13 helper phage in an amount 5 times of bacteria number, mixing well, standing for 30min, culturing at 220rpm for 1h, centrifuging at 10000rpm for 5min, discarding supernatant, replacing to C +/K +2-YT culture medium, and culturing at 30 deg.C and 220rpm overnight. The next day, 13000g was centrifuged for 10min, and the supernatant was precipitated by adding 20% PEG/NaCl (prepared from 20% PEG6000 and 2.5M NaCl), and washed once with PBS to obtain phage corresponding to alpaca library for phage screening.
The method of cell selection is adopted, and the human CLDN18.2-HEK293T cell strain is used as a selection antigen, and the antibody of human CLDN18.2 is selected from a phage display library. Human CLDN18.2-HEK293T or human CLDN18.1-HEK293 was cultured in T25 culture flasks. When the cells are grown to about 90% density, the growth state is optimal, the culture supernatant is removed, the cells are rinsed once with PBS (source culture, B310KJ), then 5mL of 4% paraformaldehyde (raw culture, E672002-0500) is added for fixing for 1h, and finally the cells are rinsed twice with PBS, so that the cells can be used as antigen materials for phage cell screening. During screening, the phage corresponding to the alpaca library is firstly incubated with a fixed human CLDN18.1-HEK293 cell culture square bottle for 1h at room temperature, and then the adsorbed supernatant phage and the fixed human CLDN18.2-HEK293T cell culture square bottle are sucked for 2 h. After PBS rinsing twice, adding 3mL glycine-HCl (pH2.0) and mixing gently for 10min to elute phage specifically binding to target membrane protein CLDN18.2, then infecting eluted supernatant with logarithmic phase SS320 thallus (Lucigen, 60512-1), standing for 30min, then culturing for 1h under 220rpm, adding VSCM13 helper phage, standing for 30min, culturing for 1h under 220rpm, centrifuging and replacing to C +/K+2-YT medium, the resulting phage was used for the second round of screening. The above steps are repeated, 10 clones randomly selected in each round are subjected to sequence analysis, and the result shows that after 3 rounds of screening, the sequence enrichment is obvious after the third round of screening.
The clones from the third round of selection were picked to prepare phage supernatants in 96-well plates, positive clones were selected for recombinant protein of CLDN18.2 by phage ELISA, then all positive clones were picked for sequencing analysis, and subsequently the clone-prepared phage supernatants with unique sequence were further verified at the flow level to screen candidate antibodies that bind only to human CLDN18.2 and not to human CLDN 18.1.
The specific flow-type horizontal verification method comprises the following steps:
firstly, 1X 10 are respectively taken5Human CLDN18.2-HEK293T and human CLDN18.1-HEK293 cells were centrifuged at 500g low speed to remove the supernatant, the cells were rinsed with FACS buffer as described in 1.3.1, the supernatant was prepared by adding phage blocked with 10% FBS (Gibco, 15140-141) for 1h, incubated at 4 ℃ for 1h and then rinsed twice with FACS buffer at 1: anti-M13 murine monoclonal antibody (Sino Biological,11973-MM05T) was added to 50 and incubated at 4 ℃ for 1 h. Then rinsed twice with FACS buffer, an APC-labeled anti-mouse Fc secondary antibody (Jackson,115136071) was added, incubated at 4 ℃ for 1h, rinsed twice with FACS buffer, and finally detected by flow cytometry (Beckman, CytoFLEX AOO-1-1102).
The specific results are shown in table 2 below:
TABLE 2
Figure BDA0002071082450000491
Figure BDA0002071082450000501
The above results indicate that the antibodies of the present disclosure are all capable of specifically binding to human CLDN 18.2.
The corresponding candidate antibodies are named by the clone numbers, respectively. The amino acid sequences of each candidate antibody are shown in tables 3 and 4 below.
Table 3 CDR sequences of partial exemplary antibodies (candidate antibodies) of the disclosure
Figure BDA0002071082450000502
Table 4 variable region sequences of some exemplary antibodies (candidate antibodies) of the present disclosure
Figure BDA0002071082450000511
Figure BDA0002071082450000521
The amino acid sequences of the screened candidate antibodies were compared, and the results are shown in FIG. 2.
The comparison of the amino acid sequence identity of the variable regions between the candidate antibodies is shown in Table 5 below.
TABLE 5
Figure BDA0002071082450000522
Example 4
Generation and expression of chimeric VHH-Fc (hIgG1) antibodies
CLDN18.2 is highly expressed on cancer cells such as gastric cancer, and antibody drugs against such tumor-associated targets can kill tumors through complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC). In this example, chimeric antibodies were designed based on the candidate nanobodies screened in example 3, and expressed for subsequent CDC and ADCC experiments. Considering the specificity of the target, when the candidate nano-antibody gene is constructed on the transient expression plasmid pcDNA3.4 (Thermofeisher, A14697), a human IgG1 Fc fragment (EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK, SEQ ID NO:20) is fused at the C end of the candidate nano-antibody, and the fragment comprises a connecting region and a constant region of IgG1, so as to mediate ADCC, CDC and other effects. In the embodiment, a candidate nano antibody A-2-4 is selected, and a human IgG1 Fc fragment is fused on the basis of the candidate nano antibody A-2-4 to obtain a chimeric antibody NA1-S (also referred to as a "candidate antibody NA 1-S", "antibody NA 1-S" or "NA 1-S antibody" in the disclosure).
Expression of the NA1-S antibody was carried out using the ExpicHO transient expression system (Gibco, A29133) in the following manner:
on the day of transfection, the cell density was confirmed to be 7X 106To 1X 107About one living cell/mL, cell viability>98% at this time the cells were adjusted to a final concentration of 6X 10 using 25mL of fresh ExpCHO expression medium pre-warmed at 37 ℃6Individual cells/mL, OptiPRO precooled at 4 ℃TMSFM 1mL diluted the plasmid of interest (25. mu.g total) with 920. mu.L OptiPROTMSFM dilution 80. mu.L ExpifeacmineTMCHO, mixing the two solutions, gently blowing, beating and mixing to prepare ExpifeacamineTMCHO/plasmid DNA mixed solution, incubated for 1-5min at room temperature, transferred to the prepared cell suspension, slowly added while gently shaking the cell suspension, and finally placed in a cell culture shaker at 37 deg.C and 8% CO2Culturing under the condition.
ExpicCHO addition within 18-22 hours post-transfectionTMEnhancer and ExpicHOTMFeed, shake flask in 32 ℃ shaker and 5% CO2The culture was continued under the conditions that, at the fifth day after transfection, the same volume of ExpCHO was addedTMFeed, gently mix the cell suspension while slowly adding, after 12-15 days of transfection, the cells expressing the supernatant were centrifuged at high speed (15000g, 10min), the resulting supernatant was affinity purified with Protein A (Millipore, P2545), the target Protein was eluted with 100mM sodium acetate (pH3.0), followed by neutralization with 1M Tris-HCl, and finally the resulting Protein (i.e., antibody NA1-S) was replaced into PBS buffer by means of a concentration tube (Millipore, UFC 901096).
Example 5
Specific binding and species cross-specific binding assays for candidate antibody NA1-S
Well-grown human CLDN18.2-HEK293T, HEK293T, HEK293, human CLDN18.1-HEK293, murine CLDN18.2-HEK293 and mouse CLDN 072 were digested with Trypsin (Gibco, 25200-072) containing 0.25% EDTAMurine CLDN18.1-HEK293 cells, 1X 105The cells were incubated with 10. mu.g/mL of the candidate antibody NA1-S for 1h, and an antibody of the hIgG1 isotype was used as a control. The cells were rinsed twice with FACS buffer and incubated with 0.5. mu.g of a PE-labeled goat anti-human IgG-Fc secondary antibody (Abcam, ab98596) for 1h at 4 ℃. Thereafter, the cells were washed three times with FACS buffer and the binding of the candidate antibody to the cells was detected by flow cytometry (Beckman, CytoFLEX AOO-1-1102).
The results of flow assays (as shown in fig. 3) showed that, similar to the control antibody IMAB362, the candidate antibody NA1-S specifically bound to human CLDN18.2-HEK293T and murine CLDN18.2-HEK293, but not to HEK293T, HEK293, human CLDN18.1-HEK293, and murine CLDN18.1HEK293 cells. This indicates that candidate antibody NA1-S specifically binds to CLDN18.2 but not to CLDN18.1 and cross-recognizes on human and murine CLDN 18.2.
Example 6
Candidate antibody NA1-S was found in human CLDN18.2-HEK293T cell line and human
Comparison of binding Capacity on CLDN18.2-KATOIII tumor cell lines
Human CLDN18.2-HEK293T or human CLDN18.2-KATOIII cells with good culture status and candidate antibody NA1-S after three times of gradient dilution are respectively incubated for 1h at 4 ℃. After rinsing twice with FACS buffer, 0.5. mu.g of goat anti-human IgG-Fc antibody labeled with PE (Abcam, ab98596) was added thereto, and the mixture was incubated at 4 ℃ for 1 hour, rinsed twice with FACS buffer, and then detected by flow cytometry.
The flow assay results are shown in fig. 4a and 4b, and show that: both on human CLDN18.2-HEK293T cells and on human CLDN18.2-KATOIII tumor cell lines, the candidate antibody NA1-S showed comparable cell binding activity to the control antibody.
Example 7
Complement Dependent Cytotoxicity (CDC) of candidate antibody NA1-S
The CDC cell killing effect of the candidate antibody was measured by MTS method. Candidate antibody NA1-S, which contains IgG1 Fc fragment, can kill cells by CDC, while the MTS reagent can be reduced to a colored compound by NADPH or NADH produced by living cells, so the shade of color represents the killing effect of the antibody mediated CDC. The specific operation method comprises the following steps:
after digesting human CLDN18.2-HEK293T cells with good culture state by trypsin, 5 × 10 cells are taken 4The cells were mixed with 5-fold dilutions of rabbit serum, and 50. mu.L each of the candidate antibody NA1-S or the control antibody IMAB362 was added in a gradient and incubated at 37 ℃ for 3 h. Then, 30. mu.L of MTS reagent (Promega, G3580) was added to the candidate antibody NA1-S or the control antibody IMAB362, mixed well, and then incubated at 37 ℃ with 5% CO2The incubation was carried out in the incubator for 4 hours, during which time the color change of the medium was observed, and the OD value at a wavelength of 492nm was measured by means of a microplate reader. Wherein, 10% Triton X-100 plus target cells are used as a control of total lysis, only target cells are used as a blank negative control, and rabbit complement plus target cells are used as a background negative control.
The cell killing rate was calculated as follows: cell killing (%) - (candidate antibody well OD value-background well OD value)/(total lysis well OD value-blank well OD value) × 100%.
The results of CDC killing effect of candidate antibody NA1-S on human CLDN18.2-HEK293T tumor cells are shown in FIG. 5 a. As can be seen in FIG. 5a, the candidate antibody NA1-S exhibits a stronger CDC cell killing effect at equimolar concentrations relative to the control antibody IMAB362, with the EC50 of NA1-S being 0.5289nM and the EC50 of IMAB362 being 0.936 nM.
The CDC killing effect of candidate antibody NA1-S on human CLDN18.2-KATOIII tumor cells was determined in the same manner and the results are shown in FIG. 5 b. As can be seen from fig. 5b, NA1-S showed a stronger CDC cell killing effect at equimolar concentrations than the control antibody IMAB362, with EC50 for NA1-S being 1.91nM and EC50 for IMAB362 being 50.86 nM.
Example 8
Antibody-dependent cell-mediated cytotoxicity (ADCC)
ADCC effect was detected using Lactate Dehydrogenase (LDH) release. The principle is as follows: the variable region of the antibody binds to a target antigen on a target cell, when the Fc segment of the antibody binds to FcRIIa (also known as CD16a) on NK effector cells in PBMC, the NK cells release perforin, granzyme and the like to lyse the target cell, and then the release of lactate dehydrogenase in cell supernatant can be detected by an LDH lactate dehydrogenase kit (Takara, MK401), so that the degree of killing of the target cell by the NK cells can be determined.
The specific operation is as follows:
50 μ L of 2X 10 density per well in 96-well cell culture plates5Human CLDN18.2-HEK293T cells at a density/mL were incubated overnight (16-20h) in an incubator at 37 ℃. Adding candidate antibody NA1-S50 μ L with gradient dilution, mixing, incubating at 37 deg.C for 20min, and adding recovered 5 × 105Human PBMC cells (effector cells/target cells ratio 50: 1) of one well were incubated in an incubator at 37 ℃ for 4 hours, then the supernatant was centrifuged at 300g, then LDH detection reagent was added, the reaction was carried out for 60 minutes, and finally OD at 492nm was measured by a microplate reader (Molecular Devices, SpectraMax190) and the results were analyzed. The total lysis control was 10% Triton X-100 plus target cells, the blank negative control was target cells alone, and the background negative control was PBMC plus target cells.
The cell killing rate was calculated as follows: the killing rate (%) - (candidate antibody well OD value-background well OD value)/(full lysis well OD value-blank well OD value) × 100%
The results (FIG. 6a) show that the candidate antibody NA1-S has comparable cell killing activity to the control antibody IMAB362 at equimolar concentrations.
The ADCC killing effect of candidate antibody NA1-S on human CLDN18.2-KATOIII tumor cells was determined in the same manner and the results are shown in FIG. 6 b. The results in figure 6b show that NA1-S all exhibited a stronger ADCC cell killing effect than the control antibody IMAB362 at equimolar concentrations, with candidate antibodies killing up to 45% whereas IMAB362 control antibodies killing only 17%.
Those skilled in the art will further appreciate that the present invention may be embodied in other specific forms without departing from the spirit or central characteristics thereof. Since the foregoing description of the invention discloses only exemplary embodiments thereof, it should be understood that other variations are considered to be within the scope of the invention. Therefore, the present invention is not limited to the specific embodiments described in detail herein. Rather, reference should be made to the appended claims as indicating the scope and content of the invention.
Sequence listing
<110> Sanyou biomedical (Shanghai) Co., Ltd
<120> novel CLDN18.2 binding molecules
<130> IDC196010
<160> 29
<170> PatentIn version 3.5
<210> 1
<211> 8
<212> PRT
<213> Artificial sequence
<220>
<223> CDR sequences
<400> 1
Gly Asn Ile Phe Arg Ile Asp Thr
1 5
<210> 2
<211> 7
<212> PRT
<213> Artificial sequence
<220>
<223> CDR sequences
<400> 2
Ile Ser Arg Gly Gly Thr Thr
1 5
<210> 3
<211> 15
<212> PRT
<213> Artificial sequence
<220>
<223> CDR sequences
<400> 3
Asn Ala Gln Ala Trp Asp Pro Gly Thr Phe Arg Tyr Leu Glu Val
1 5 10 15
<210> 4
<211> 7
<212> PRT
<213> Artificial sequence
<220>
<223> CDR sequences
<400> 4
Ile Ser Arg Gly Gly Ser Thr
1 5
<210> 5
<211> 15
<212> PRT
<213> Artificial sequence
<220>
<223> CDR sequences
<400> 5
Asn Ala Gln Ala Trp Asp Pro Gly Thr Ile Arg Tyr Leu Glu Val
1 5 10 15
<210> 6
<211> 15
<212> PRT
<213> Artificial sequence
<220>
<223> CDR sequences
<400> 6
Asn Ala Gln Ala Trp Asp Val Gly Thr Ile Arg Tyr Leu Glu Val
1 5 10 15
<210> 7
<211> 121
<212> PRT
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 7
Glu Val Gln Val Gln Glu Ser Gly Gly Gly Leu Val Gln Ala Gly Thr
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asn Ile Phe Arg Ile Asp
20 25 30
Thr Met Gly Trp Tyr Arg Gln Ala Pro Gly Lys Gln Arg Glu Leu Val
35 40 45
Ala Gly Ile Ser Arg Gly Gly Thr Thr Thr Tyr Ala His Ser Val Lys
50 55 60
Glu Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Met Tyr Leu
65 70 75 80
Gln Met Asn Ser Leu Lys Ser Glu Asp Thr Ala Gly Tyr Tyr Cys Asn
85 90 95
Ala Gln Ala Trp Asp Pro Gly Thr Phe Arg Tyr Leu Glu Val Trp Gly
100 105 110
Gln Gly Thr Leu Val Thr Val Ser Ala
115 120
<210> 8
<211> 121
<212> PRT
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 8
Glu Val Gln Val Gln Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asn Ile Phe Arg Ile Asp
20 25 30
Thr Met Gly Trp Tyr Arg Gln Ala Pro Gly Lys Gln Arg Glu Leu Val
35 40 45
Ala Gly Ile Ser Arg Gly Gly Thr Thr Thr Tyr Ala His Ser Val Lys
50 55 60
Glu Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Met Tyr Leu
65 70 75 80
Gln Met Asn Ser Leu Lys Ser Glu Asp Thr Ala Gly Tyr Tyr Cys Asn
85 90 95
Ala Gln Ala Trp Asp Pro Gly Thr Phe Arg Tyr Leu Glu Val Trp Gly
100 105 110
Gln Gly Thr Leu Val Thr Val Ser Ala
115 120
<210> 9
<211> 121
<212> PRT
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 9
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asn Ile Phe Arg Ile Asp
20 25 30
Thr Met Gly Trp Tyr Arg Gln Ala Pro Gly Lys Gln Arg Glu Leu Val
35 40 45
Ala Gly Ile Ser Arg Gly Gly Thr Thr Thr Tyr Ala His Ser Val Lys
50 55 60
Glu Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Met Tyr Leu
65 70 75 80
Gln Met Asn Ser Leu Lys Ser Glu Asp Thr Ala Gly Tyr Tyr Cys Asn
85 90 95
Ala Gln Ala Trp Asp Pro Gly Thr Phe Arg Tyr Leu Glu Val Trp Gly
100 105 110
Gln Gly Thr Leu Val Thr Val Ser Ala
115 120
<210> 10
<211> 121
<212> PRT
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 10
Gln Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asn Ile Phe Arg Ile Asp
20 25 30
Thr Met Gly Trp Tyr Arg Gln Ala Pro Gly Lys Gln Arg Glu Phe Val
35 40 45
Ala Gly Ile Ser Arg Gly Gly Ser Thr Asn Tyr Ala His Ser Val Lys
50 55 60
Glu Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Met Tyr Leu
65 70 75 80
Gln Met Asn Ser Leu Lys Ser Glu Asp Thr Ala Gly Tyr Tyr Cys Asn
85 90 95
Ala Gln Ala Trp Asp Pro Gly Thr Ile Arg Tyr Leu Glu Val Trp Gly
100 105 110
Gln Gly Thr Leu Val Thr Val Ser Ser
115 120
<210> 11
<211> 121
<212> PRT
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 11
Glu Val Gln Leu Gln Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asn Ile Phe Arg Ile Asp
20 25 30
Thr Met Gly Trp Tyr Arg Gln Ala Pro Gly Lys Gln Arg Glu Phe Val
35 40 45
Ala Gly Ile Ser Arg Gly Gly Ser Thr Asn Tyr Ala His Ser Val Lys
50 55 60
Glu Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Met Tyr Leu
65 70 75 80
Gln Met Asn Ser Leu Lys Ser Glu Asp Thr Ala Gly Tyr Tyr Cys Asn
85 90 95
Ala Gln Ala Trp Asp Pro Gly Thr Ile Arg Tyr Leu Glu Val Trp Gly
100 105 110
Gln Gly Thr Leu Val Thr Val Ser Ala
115 120
<210> 12
<211> 121
<212> PRT
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 12
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asn Ile Phe Arg Ile Asp
20 25 30
Thr Met Gly Trp Tyr Arg Gln Ala Pro Gly Lys Gln Arg Glu Phe Val
35 40 45
Ala Gly Ile Ser Arg Gly Gly Ser Thr Asn Tyr Ala His Ser Val Lys
50 55 60
Glu Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Met Tyr Leu
65 70 75 80
Gln Met Asn Ser Leu Lys Ser Glu Asp Thr Ala Gly Tyr Tyr Cys Asn
85 90 95
Ala Gln Ala Trp Asp Pro Gly Thr Ile Arg Tyr Leu Glu Val Trp Gly
100 105 110
Gln Gly Thr Leu Val Thr Val Ser Ala
115 120
<210> 13
<211> 121
<212> PRT
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 13
Glu Val Gln Val Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asn Ile Phe Arg Ile Asp
20 25 30
Thr Met Val Trp Tyr Arg Gln Ala Pro Gly Lys Gln Arg Glu Leu Val
35 40 45
Ala Gly Ile Ser Arg Gly Gly Thr Thr Asn Tyr Ala His Ser Val Lys
50 55 60
Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Met Tyr Leu
65 70 75 80
Gln Met Asn Ser Leu Lys Ser Glu Asp Thr Ala Thr Tyr Tyr Cys Asn
85 90 95
Ala Gln Ala Trp Asp Val Gly Thr Ile Arg Tyr Leu Glu Val Trp Gly
100 105 110
Gln Gly Thr Leu Val Thr Val Ser Ala
115 120
<210> 14
<211> 121
<212> PRT
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 14
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asn Ile Phe Arg Ile Asp
20 25 30
Thr Met Val Trp Tyr Arg Gln Ala Pro Gly Lys Gln Arg Glu Leu Val
35 40 45
Ala Gly Ile Ser Arg Gly Gly Thr Thr Asn Tyr Ala His Ser Val Lys
50 55 60
Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Met Tyr Leu
65 70 75 80
Gln Met Asn Ser Leu Lys Ser Glu Asp Thr Ala Thr Tyr Tyr Cys Asn
85 90 95
Ala Gln Ala Trp Asp Val Gly Thr Ile Arg Tyr Leu Glu Val Trp Gly
100 105 110
Gln Gly Thr Leu Val Thr Val Ser Ala
115 120
<210> 15
<211> 261
<212> PRT
<213> Homo sapiens
<400> 15
Met Ala Val Thr Ala Cys Gln Gly Leu Gly Phe Val Val Ser Leu Ile
1 5 10 15
Gly Ile Ala Gly Ile Ile Ala Ala Thr Cys Met Asp Gln Trp Ser Thr
20 25 30
Gln Asp Leu Tyr Asn Asn Pro Val Thr Ala Val Phe Asn Tyr Gln Gly
35 40 45
Leu Trp Arg Ser Cys Val Arg Glu Ser Ser Gly Phe Thr Glu Cys Arg
50 55 60
Gly Tyr Phe Thr Leu Leu Gly Leu Pro Ala Met Leu Gln Ala Val Arg
65 70 75 80
Ala Leu Met Ile Val Gly Ile Val Leu Gly Ala Ile Gly Leu Leu Val
85 90 95
Ser Ile Phe Ala Leu Lys Cys Ile Arg Ile Gly Ser Met Glu Asp Ser
100 105 110
Ala Lys Ala Asn Met Thr Leu Thr Ser Gly Ile Met Phe Ile Val Ser
115 120 125
Gly Leu Cys Ala Ile Ala Gly Val Ser Val Phe Ala Asn Met Leu Val
130 135 140
Thr Asn Phe Trp Met Ser Thr Ala Asn Met Tyr Thr Gly Met Gly Gly
145 150 155 160
Met Val Gln Thr Val Gln Thr Arg Tyr Thr Phe Gly Ala Ala Leu Phe
165 170 175
Val Gly Trp Val Ala Gly Gly Leu Thr Leu Ile Gly Gly Val Met Met
180 185 190
Cys Ile Ala Cys Arg Gly Leu Ala Pro Glu Glu Thr Asn Tyr Lys Ala
195 200 205
Val Ser Tyr His Ala Ser Gly His Ser Val Ala Tyr Lys Pro Gly Gly
210 215 220
Phe Lys Ala Ser Thr Gly Phe Gly Ser Asn Thr Lys Asn Lys Lys Ile
225 230 235 240
Tyr Asp Gly Gly Ala Arg Thr Glu Asp Glu Val Gln Ser Tyr Pro Ser
245 250 255
Lys His Asp Tyr Val
260
<210> 16
<211> 261
<212> PRT
<213> Homo sapiens
<400> 16
Met Ser Thr Thr Thr Cys Gln Val Val Ala Phe Leu Leu Ser Ile Leu
1 5 10 15
Gly Leu Ala Gly Cys Ile Ala Ala Thr Gly Met Asp Met Trp Ser Thr
20 25 30
Gln Asp Leu Tyr Asp Asn Pro Val Thr Ser Val Phe Gln Tyr Glu Gly
35 40 45
Leu Trp Arg Ser Cys Val Arg Gln Ser Ser Gly Phe Thr Glu Cys Arg
50 55 60
Pro Tyr Phe Thr Ile Leu Gly Leu Pro Ala Met Leu Gln Ala Val Arg
65 70 75 80
Ala Leu Met Ile Val Gly Ile Val Leu Gly Ala Ile Gly Leu Leu Val
85 90 95
Ser Ile Phe Ala Leu Lys Cys Ile Arg Ile Gly Ser Met Glu Asp Ser
100 105 110
Ala Lys Ala Asn Met Thr Leu Thr Ser Gly Ile Met Phe Ile Val Ser
115 120 125
Gly Leu Cys Ala Ile Ala Gly Val Ser Val Phe Ala Asn Met Leu Val
130 135 140
Thr Asn Phe Trp Met Ser Thr Ala Asn Met Tyr Thr Gly Met Gly Gly
145 150 155 160
Met Val Gln Thr Val Gln Thr Arg Tyr Thr Phe Gly Ala Ala Leu Phe
165 170 175
Val Gly Trp Val Ala Gly Gly Leu Thr Leu Ile Gly Gly Val Met Met
180 185 190
Cys Ile Ala Cys Arg Gly Leu Ala Pro Glu Glu Thr Asn Tyr Lys Ala
195 200 205
Val Ser Tyr His Ala Ser Gly His Ser Val Ala Tyr Lys Pro Gly Gly
210 215 220
Phe Lys Ala Ser Thr Gly Phe Gly Ser Asn Thr Lys Asn Lys Lys Ile
225 230 235 240
Tyr Asp Gly Gly Ala Arg Thr Glu Asp Glu Val Gln Ser Tyr Pro Ser
245 250 255
Lys His Asp Tyr Val
260
<210> 17
<211> 264
<212> PRT
<213> Mus musculus
<400> 17
Met Ser Val Thr Ala Cys Gln Gly Leu Gly Phe Val Val Ser Leu Ile
1 5 10 15
Gly Phe Ala Gly Ile Ile Ala Ala Thr Cys Met Asp Gln Trp Ser Thr
20 25 30
Gln Asp Leu Tyr Asn Asn Pro Val Thr Ala Val Phe Asn Tyr Gln Gly
35 40 45
Leu Trp Arg Ser Cys Val Arg Glu Ser Ser Gly Phe Thr Glu Cys Arg
50 55 60
Gly Tyr Phe Thr Leu Leu Gly Leu Pro Ala Met Leu Gln Ala Val Arg
65 70 75 80
Ala Leu Met Ile Val Gly Ile Val Leu Gly Val Ile Gly Ile Leu Val
85 90 95
Ser Ile Phe Ala Leu Lys Cys Ile Arg Ile Gly Ser Met Asp Asp Ser
100 105 110
Ala Lys Ala Lys Met Thr Leu Thr Ser Gly Ile Leu Phe Ile Ile Ser
115 120 125
Gly Ile Cys Ala Ile Ile Gly Val Ser Val Phe Ala Asn Met Leu Val
130 135 140
Thr Asn Phe Trp Met Ser Thr Ala Asn Met Tyr Ser Gly Met Gly Gly
145 150 155 160
Met Gly Gly Met Val Gln Thr Val Gln Thr Arg Tyr Thr Phe Gly Ala
165 170 175
Ala Leu Phe Val Gly Trp Val Ala Gly Gly Leu Thr Leu Ile Gly Gly
180 185 190
Val Met Met Cys Ile Ala Cys Arg Gly Leu Thr Pro Asp Asp Ser Asn
195 200 205
Phe Lys Ala Val Ser Tyr His Ala Ser Gly Gln Asn Val Ala Tyr Arg
210 215 220
Pro Gly Gly Phe Lys Ala Ser Thr Gly Phe Gly Ser Asn Thr Arg Asn
225 230 235 240
Lys Lys Ile Tyr Asp Gly Gly Ala Arg Thr Glu Asp Asp Glu Gln Ser
245 250 255
His Pro Thr Lys Tyr Asp Tyr Val
260
<210> 18
<211> 264
<212> PRT
<213> Mus musculus
<400> 18
Met Ala Thr Thr Thr Cys Gln Val Val Gly Leu Leu Leu Ser Leu Leu
1 5 10 15
Gly Leu Ala Gly Cys Ile Ala Ala Thr Gly Met Asp Met Trp Ser Thr
20 25 30
Gln Asp Leu Tyr Asp Asn Pro Val Thr Ala Val Phe Gln Tyr Glu Gly
35 40 45
Leu Trp Arg Ser Cys Val Gln Gln Ser Ser Gly Phe Thr Glu Cys Arg
50 55 60
Pro Tyr Phe Thr Ile Leu Gly Leu Pro Ala Met Leu Gln Ala Val Arg
65 70 75 80
Ala Leu Met Ile Val Gly Ile Val Leu Gly Val Ile Gly Ile Leu Val
85 90 95
Ser Ile Phe Ala Leu Lys Cys Ile Arg Ile Gly Ser Met Asp Asp Ser
100 105 110
Ala Lys Ala Lys Met Thr Leu Thr Ser Gly Ile Leu Phe Ile Ile Ser
115 120 125
Gly Ile Cys Ala Ile Ile Gly Val Ser Val Phe Ala Asn Met Leu Val
130 135 140
Thr Asn Phe Trp Met Ser Thr Ala Asn Met Tyr Ser Gly Met Gly Gly
145 150 155 160
Met Gly Gly Met Val Gln Thr Val Gln Thr Arg Tyr Thr Phe Gly Ala
165 170 175
Ala Leu Phe Val Gly Trp Val Ala Gly Gly Leu Thr Leu Ile Gly Gly
180 185 190
Val Met Met Cys Ile Ala Cys Arg Gly Leu Thr Pro Asp Asp Ser Asn
195 200 205
Phe Lys Ala Val Ser Tyr His Ala Ser Gly Gln Asn Val Ala Tyr Arg
210 215 220
Pro Gly Gly Phe Lys Ala Ser Thr Gly Phe Gly Ser Asn Thr Arg Asn
225 230 235 240
Lys Lys Ile Tyr Asp Gly Gly Ala Arg Thr Glu Asp Asp Glu Gln Ser
245 250 255
His Pro Thr Lys Tyr Asp Tyr Val
260
<210> 19
<211> 129
<212> PRT
<213> Homo sapiens
<400> 19
Met Glu Thr Asp Thr Leu Leu Leu Trp Val Leu Leu Leu Trp Val Pro
1 5 10 15
Gly Ser Thr Gly Asp Ala Ala Gln Pro Ala Arg Arg Ala Arg Arg Thr
20 25 30
Lys Leu Gly Thr Glu Leu Gly Ser Thr Pro Val Trp Trp Asn Ser Ala
35 40 45
Asp Gly Arg Met Asp Gln Trp Ser Thr Gln Asp Leu Tyr Asn Asn Pro
50 55 60
Val Thr Ala Val Phe Asn Tyr Gln Gly Leu Trp Arg Ser Cys Val Arg
65 70 75 80
Glu Ser Ser Gly Phe Thr Glu Cys Arg Gly Tyr Phe Thr Leu Leu Gly
85 90 95
Leu Pro Ala Met Leu Gln Ala Val Arg Ala Ala Ile Gln His Ser Gly
100 105 110
Gly Arg Ser Arg Arg Ala Arg Thr Lys Thr His Leu Arg Arg Gly Ser
115 120 125
Glu
<210> 20
<211> 232
<212> PRT
<213> Homo sapiens
<400> 20
Glu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala
1 5 10 15
Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro
20 25 30
Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val
35 40 45
Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val
50 55 60
Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln
65 70 75 80
Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln
85 90 95
Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala
100 105 110
Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro
115 120 125
Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr
130 135 140
Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser
145 150 155 160
Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr
165 170 175
Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr
180 185 190
Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe
195 200 205
Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys
210 215 220
Ser Leu Ser Leu Ser Pro Gly Lys
225 230
<210> 21
<211> 14
<212> PRT
<213> Homo sapiens
<400> 21
Gly Phe Lys Ala Ser Thr Gly Phe Gly Ser Asn Thr Lys Asn
1 5 10
<210> 22
<211> 360
<212> DNA
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 22
gaggtgcagg tgcaggagtc tgggggaggc ctggtacagg ctgggacctc tctgagactc 60
tcctgtgcag cctctggcaa catcttccgt atcgatacca tgggctggta ccgccaggct 120
ccaggaaagc agcgcgagtt ggtcgcaggt atttctcgtg gtggtacaac aacctatgca 180
cactccgtga aggaacgatt caccatctcc agagacaacg ccaagaacac gatgtatctg 240
caaatgaaca gcctgaaatc tgaggacacg gccggctatt attgtaatgc acaggcgtgg 300
gatcctggta catttcggta tctcgaagtt tggggccagg gcaccctggt cactgtctca 360
<210> 23
<211> 360
<212> DNA
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 23
gaggtgcagt tggtggagtc tgggggaggc ttggtacagc ctggggggtc tctgagactc 60
tcctgtgcag cctctggcaa catcttccgt atcgatacca tgggctggta ccgccaggct 120
ccaggaaagc agcgcgagtt cgtcgcaggt attagtcgtg gtggtagcac aaactatgca 180
cactccgtga aggaacgatt caccatctcc agagacaacg ccaagaacac gatgtatctg 240
caaatgaaca gcctgaaatc tgaggacacg gccggctatt attgtaatgc acaggcgtgg 300
gatcctggta caatccggta tctcgaagtt tggggccagg gcaccctggt cactgtctca 360
<210> 24
<211> 360
<212> DNA
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 24
gaggtgcagg tgcaggagtc tgggggaggc ttggtacagc ctggggggtc tctgagactc 60
tcctgtgcag cctctggcaa catcttccgt atcgatacca tgggctggta ccgccaggct 120
ccaggaaagc agcgcgagtt ggtcgcaggt atttctcgtg gtggtacaac aacctatgca 180
cactccgtga aggaacgatt caccatctcc agagacaacg ccaagaacac gatgtatctg 240
caaatgaaca gcctgaaatc tgaggacacg gccggctatt attgtaatgc acaggcgtgg 300
gatcctggta catttcggta tctcgaagtt tggggccagg gcaccctggt cactgtctca 360
<210> 25
<211> 363
<212> DNA
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 25
caggtgcagc tcgtggagtc tgggggaggc ttggtacagc ctggggggtc tctgagactc 60
tcctgtgcag cctctggcaa catcttccgt atcgatacca tgggctggta ccgccaggct 120
ccaggaaagc agcgcgagtt cgtcgcaggt attagtcgtg gtggtagcac aaactatgca 180
cactccgtga aggaacgatt caccatctcc agagacaacg ccaagaacac gatgtatctg 240
caaatgaaca gcctgaaatc tgaggacacg gccggctatt attgtaatgc acaggcgtgg 300
gatcctggta caatccggta tctcgaagtt tggggccagg gcaccctggt caccgtgtcc 360
tca 363
<210> 26
<211> 360
<212> DNA
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 26
caggtgcagc tcgtggagtc tgggggaggc ttggtacagc ctggggggtc tctgagactc 60
tcctgtgcag cctctggcaa catcttccgt atcgatacca tgggctggta ccgccaggct 120
ccaggaaagc agcgcgagtt ggtcgcaggt atttctcgtg gtggtacaac aacctatgca 180
cactccgtga aggaacgatt caccatctcc agagacaacg ccaagaacac gatgtatctg 240
caaatgaaca gcctgaaatc tgaggacacg gccggctatt attgtaatgc acaggcgtgg 300
gatcctggta catttcggta tctcgaagtt tggggccagg gcaccctggt cactgtctca 360
<210> 27
<211> 360
<212> DNA
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 27
gaggtgcagt tggtggagtc tgggggaggc ttggtacagc ctggggggtc tctgagactc 60
tcctgtgcag cctctggcaa catcttccgt atcgatacca tggtgtggta ccgccaggct 120
ccaggaaagc agcgcgagtt ggtcgcaggt atttctcgtg gtggtaccac aaactatgca 180
cactccgtga agggccgatt caccatctcc agagacaacg ccaagaacac gatgtatctg 240
caaatgaaca gcctgaaatc tgaggacacg gccacgtatt attgtaatgc acaggcgtgg 300
gatgttggta caatccggta tctcgaagtt tggggccagg gcaccctggt cactgtctca 360
<210> 28
<211> 360
<212> DNA
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 28
gaggtgcagc tgcaggagtc tgggggaggc ttggtacagc ctggggggtc tctgagactc 60
tcctgtgcag cctctggcaa catcttccgt atcgatacca tgggctggta ccgccaggct 120
ccaggaaagc agcgcgagtt cgtcgcaggt attagtcgtg gtggtagcac aaactatgca 180
cactccgtga aggaacgatt caccatctcc agagacaacg ccaagaacac gatgtatctg 240
caaatgaaca gcctgaaatc tgaggacacg gccggctatt attgtaatgc acaggcgtgg 300
gatcctggta caatccggta tctcgaagtt tggggccagg gcaccctggt cactgtctca 360
<210> 29
<211> 360
<212> DNA
<213> Artificial sequence
<220>
<223> variable region sequences
<400> 29
gaggtgcagg tcgtggagtc tgggggaggc ttggtacagc ctggggggtc tctgagactc 60
tcctgtgcag cctctggcaa catcttccgt atcgatacca tggtgtggta ccgccaggct 120
ccaggaaagc agcgcgagtt ggtcgcaggt atttctcgtg gtggtaccac aaactatgca 180
cactccgtga agggccgatt caccatctcc agagacaacg ccaagaacac gatgtatctg 240
caaatgaaca gcctgaaatc tgaggacacg gccacgtatt attgtaatgc acaggcgtgg 300
gatgttggta caatccggta tctcgaagtt tggggccagg gcaccctggt cactgtctca 360

Claims (24)

1. An antibody or antigen binding fragment thereof that binds to CLDN18.2, comprising an immunoglobulin single variable domain, wherein the immunoglobulin single variable domain comprises:
(a) As shown in SEQ ID NO: 1, as shown in SEQ ID NO: 2 and the CDR2 of the amino acid sequence shown in SEQ ID NO: 3, CDR3 of the amino acid sequence set forth in seq id no;
(b) as shown in SEQ ID NO: 1, as shown in SEQ ID NO: 4 and the CDR2 of the amino acid sequence shown in SEQ ID NO: 5 CDR3 of the amino acid sequence set forth in seq id no; or
(c) As shown in SEQ ID NO: 1, as shown in SEQ ID NO: 2 and the CDR2 of the amino acid sequence shown in SEQ ID NO: 6, CDR3 of the amino acid sequence set forth in seq id no.
2. The antibody or antigen binding fragment thereof of claim 1, wherein the immunoglobulin single variable domain is a VHH.
3. The antibody or antigen binding fragment thereof according to claim 2, wherein the immunoglobulin single variable domain is a VHH from a camelid.
4. The antibody or antigen binding fragment thereof of claim 1, wherein the immunoglobulin single variable domain comprises
(A) SEQ ID NO: 7;
(B) and SEQ ID NO: 7 amino acid sequence that is at least 80%, 85%, 90% or 95% identical; or
(C) And SEQ ID NO: 7 compared to an amino acid sequence having one or more additions, deletions and/or substitutions of amino acids in the framework region.
5. The antibody or antigen binding fragment thereof of claim 4, wherein the immunoglobulin single variable domain is modified at one or more of the following positions of SEQ ID NO 7: amino acid 1, 4, 5, 14, 16, 35, 47, 58, 65, 92, or 121.
6. The antibody or antigen binding fragment thereof of claim 4, wherein the immunoglobulin single variable domain consists of: any one of SEQ ID NOs 7-14.
7. The antibody or antigen-binding fragment thereof of any one of claims 1-6, which is a single domain antibody, a chimeric antibody, or a humanized antibody.
8. The antibody or antigen binding fragment thereof of any one of claims 1-6, wherein the immunoglobulin single variable domain is fused to an Fc domain of an immunoglobulin or a fluorescent protein.
9. The antibody or antigen-binding fragment thereof of claim 8, wherein the immunoglobulin is an IgG.
10. The antibody or antigen-binding fragment thereof of claim 8, wherein the antibody or antigen-binding fragment thereof is a chimeric antibody comprising a VHH from a camelid and an Fc domain of a human IgG.
11. The antibody or antigen-binding fragment thereof of claim 10, wherein the IgG is human IgG1 or IgG 4.
12. The antibody or antigen-binding fragment thereof of claim 10, which is a chimeric antibody comprising a VHH from alpaca and an Fc domain of human IgG 1.
13. The antibody or antigen-binding fragment thereof of claim 1, which binds to extracellular domain 1(ECD1) of human CLDN 18.2.
14. The antibody or antigen-binding fragment thereof of claim 1, which specifically binds to CLDN18.2 but does not bind to CLDN 18.1.
15. An isolated nucleic acid molecule comprising a nucleic acid sequence encoding an antibody or antigen-binding fragment thereof as defined in any one of the preceding claims.
16. The isolated nucleic acid molecule of claim 15 comprising or consisting of the sequence of seq id no: any one of SEQ ID NOs 22-29.
17. An expression vector comprising the isolated nucleic acid molecule of claim 15 or 16.
18. A host cell comprising the expression vector of claim 17.
19. The host cell of claim 18, wherein the host cell is a bacterial cell, a fungal cell, or a mammalian cell.
20. The host cell of claim 19, wherein the bacterial cell is e.
21. A pharmaceutical composition comprising an antibody or antigen-binding fragment thereof as defined in any one of claims 1 to 14 and a pharmaceutically acceptable carrier.
22. A method of preparing an antibody or antigen-binding fragment thereof as defined in any one of claims 1 to 14, comprising the steps of:
-expressing an antibody or antigen-binding fragment thereof as defined in any one of claims 1 to 14 in a host cell according to claim 18; and
-isolating the antibody or antigen-binding fragment thereof from the host cell.
23. Use of an antibody or antigen-binding fragment thereof as defined in any one of claims 1 to 14 in the manufacture of a medicament for the treatment or prevention of a CLDN 18.2-associated cancer.
24. A kit for the treatment or diagnosis of a disorder associated with CLDN18.2, comprising a container comprising an antibody or antigen-binding fragment thereof as defined in any one of claims 1 to 14.
CN201910437800.2A 2019-05-24 2019-05-24 Novel CLDN18.2 binding molecules Active CN111978402B (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CN201910437800.2A CN111978402B (en) 2019-05-24 2019-05-24 Novel CLDN18.2 binding molecules
TW109116820A TW202108627A (en) 2019-05-24 2020-05-21 Novel cldn182 binding molecule
US17/613,440 US20220396616A1 (en) 2019-05-24 2020-05-21 Novel cldn18.2 binding molecule
EP20815629.9A EP3978532A4 (en) 2019-05-24 2020-05-21 Novel cldn18.2 binding molecule
KR1020217039993A KR20220012262A (en) 2019-05-24 2020-05-21 Novel CLDN18.2 binding molecule
JP2022516255A JP2022533804A (en) 2019-05-24 2020-05-21 A novel CLDN18.2 binding molecule
CN202080038303.6A CN114206935B (en) 2019-05-24 2020-05-21 Novel CLDN18.2 binding molecules
CA3141504A CA3141504A1 (en) 2019-05-24 2020-05-21 Novel cldn18.2 binding molecule
PCT/CN2020/091441 WO2020238730A1 (en) 2019-05-24 2020-05-21 Novel cldn18.2 binding molecule

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910437800.2A CN111978402B (en) 2019-05-24 2019-05-24 Novel CLDN18.2 binding molecules

Publications (2)

Publication Number Publication Date
CN111978402A CN111978402A (en) 2020-11-24
CN111978402B true CN111978402B (en) 2022-06-28

Family

ID=73436850

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910437800.2A Active CN111978402B (en) 2019-05-24 2019-05-24 Novel CLDN18.2 binding molecules

Country Status (1)

Country Link
CN (1) CN111978402B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112979817B (en) * 2021-04-27 2021-08-13 宝船生物医药科技(上海)有限公司 Monoclonal antibody for recognizing anti-CLDN 18_2 antibody and preparation method and application thereof
CN115991784A (en) * 2021-10-19 2023-04-21 宝船生物医药科技(上海)有限公司 anti-CD 47-CLDN18.2 bispecific antibody and uses thereof
CN116410326A (en) * 2021-12-30 2023-07-11 三优生物医药(上海)有限公司 anti-CD 40 xCLDN 18.2 bispecific antibody and uses thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101687929A (en) * 2007-05-29 2010-03-31 加尼梅德药物公司 Monoclonal antibodies against claudin-18 for treatment of cancer
CN104321345A (en) * 2012-05-09 2015-01-28 加尼梅德药物公司 Antibodies against claudin 18.2 useful in cancer diagnosis
EP2852408A1 (en) * 2012-05-23 2015-04-01 GANYMED Pharmaceuticals AG Combination therapy involving antibodies against claudin 18.2 for treatment of cancer
WO2018006882A1 (en) * 2016-07-08 2018-01-11 科济生物医药(上海)有限公司 Antibody for anti-claudin 18a2 and use thereof
CN109762067A (en) * 2019-01-17 2019-05-17 北京天广实生物技术股份有限公司 In conjunction with the antibody and application thereof of people Claudin 18.2

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101687929A (en) * 2007-05-29 2010-03-31 加尼梅德药物公司 Monoclonal antibodies against claudin-18 for treatment of cancer
CN104321345A (en) * 2012-05-09 2015-01-28 加尼梅德药物公司 Antibodies against claudin 18.2 useful in cancer diagnosis
EP2852408A1 (en) * 2012-05-23 2015-04-01 GANYMED Pharmaceuticals AG Combination therapy involving antibodies against claudin 18.2 for treatment of cancer
WO2018006882A1 (en) * 2016-07-08 2018-01-11 科济生物医药(上海)有限公司 Antibody for anti-claudin 18a2 and use thereof
CN109762067A (en) * 2019-01-17 2019-05-17 北京天广实生物技术股份有限公司 In conjunction with the antibody and application thereof of people Claudin 18.2

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Claudin 18.2 is a target for IMAB362 antibody in pancreatic neoplasms;Stefan Wöll,等;《International Journal of Cancer》;20140201;第134卷(第3期);全文 *
Claudin-18 splice variant 2 is a pan-cancer target suitable for therapeutic antibody development;Ugur Sahin,等;《Clinical Cancer Research》;20081201;第14卷(第23期);全文 *
CLDN18.2蛋白在恶性肿瘤治疗中的研究进展;徐良额,等;《中国肿瘤临床》;20190330;第46卷(第6期);全文 *

Also Published As

Publication number Publication date
CN111978402A (en) 2020-11-24

Similar Documents

Publication Publication Date Title
CN114206935B (en) Novel CLDN18.2 binding molecules
CN112480248B (en) Molecules that specifically bind to CLD18A2
CN110204614B (en) Anti-human LAG-3 monoclonal antibody and preparation method and application thereof
CN110357961B (en) Anti-human 4-1BB monoclonal antibody, preparation method and application thereof
CN111978403B (en) Novel CLDN18.2 binding molecules
CN112236454B (en) Novel anti-PD-1 antibodies
CN111978402B (en) Novel CLDN18.2 binding molecules
US20240067725A1 (en) Monoclonal antibody against human lag-3, method for preparing same, and use thereof
JP7445752B2 (en) Novel anti-PD-L1 antibody
JP2022550364A (en) Novel anti-PD-L1/anti-LAG-3 bispecific antibodies and uses thereof
AU2022345323A1 (en) D3-binding molecules and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40032484

Country of ref document: HK

GR01 Patent grant
GR01 Patent grant
PE01 Entry into force of the registration of the contract for pledge of patent right
PE01 Entry into force of the registration of the contract for pledge of patent right

Denomination of invention: Novel CLDN18.2 Binding Molecules

Effective date of registration: 20230519

Granted publication date: 20220628

Pledgee: Industrial Bank Co.,Ltd. Shanghai Pudong Sub branch

Pledgor: Sanyou biomedical (Shanghai) Co.,Ltd.

Registration number: Y2023310000202