CN117843776A - Antibody molecule, nucleic acid, pharmaceutical use and method for treating inflammatory disease - Google Patents

Antibody molecule, nucleic acid, pharmaceutical use and method for treating inflammatory disease Download PDF

Info

Publication number
CN117843776A
CN117843776A CN202311774070.8A CN202311774070A CN117843776A CN 117843776 A CN117843776 A CN 117843776A CN 202311774070 A CN202311774070 A CN 202311774070A CN 117843776 A CN117843776 A CN 117843776A
Authority
CN
China
Prior art keywords
antibody
amino acid
seq
cancer
acid sequence
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.)
Pending
Application number
CN202311774070.8A
Other languages
Chinese (zh)
Inventor
刘拥军
刘广洋
王道辉
米一
陈瑶瑶
李果
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.)
Beijing Beilai Pharmaceutical Co ltd
Original Assignee
Beijing Beilai Pharmaceutical Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beijing Beilai Pharmaceutical Co ltd filed Critical Beijing Beilai Pharmaceutical Co ltd
Priority to CN202311774070.8A priority Critical patent/CN117843776A/en
Publication of CN117843776A publication Critical patent/CN117843776A/en
Pending legal-status Critical Current

Links

Abstract

The invention provides an antibody molecule, nucleic acid, pharmaceutical application and an inflammatory disease treatment method, belonging to the technical field of cell immunology. The antibodies provided by the invention include amino acid sequences of HCDR1, HCDR2 and HCDR 3; the amino acid sequence of the HCDR1 is shown as SEQ ID NO. 1, the amino acid sequence of the HCDR2 is shown as SEQ ID NO. 2, and the amino acid sequence of the HCDR3 is shown as SEQ ID NO. 3; and/or an amino acid sequence having at least 80% sequence similarity with SEQ ID NO. 1, SEQ ID NO. 2 and SEQ ID NO. 3, respectively. The antibody of the invention has higher affinity, strong blocking capability and stability, wide biological application value and clinical application value, and lays a material foundation for the development of therapeutic or diagnostic drugs for autoimmune diseases.

Description

Antibody molecule, nucleic acid, pharmaceutical use and method for treating inflammatory disease
Technical Field
The invention belongs to the technical field of cell immunology, and particularly relates to an antibody molecule, nucleic acid, pharmaceutical application and an inflammatory disease treatment method.
Background
The IL-17 cytokine family consists of 6 members, IL-17A, IL-17B, IL-17C, IL-17D, IL-17E, IL-17F, respectively, the most prominent members being IL-17A and IL-17F, which form functional homodimers or heterodimers and are adjacent in batches on the same chromosome, with a large overlapping pro-inflammatory effect. The IL-17 receptor family is a multimeric protein comprising five molecules, IL-17RA, IL-17RB, IL-17RC, IL-17RD, and IL-17RE. IL-17A may stimulate the secretion of a range of pro-inflammatory cytokines, such as IL-6, tumor necrosis factor-alpha (TNF-alpha), IL-1β, and leukocyte-mobilized cytokines, such as catalytic factor ligand 1 (CXCL 1) and ligand 8 (CXCL 8). Effector functions of IL-17A are thought to play a critical role in host responses against extracellular and intracellular pathogens and are involved in the pathogenesis of many autoimmune inflammatory diseases.
Single-domain antibodies (sdabs), also commonly referred to as nanobodies (Nb), refer to the smallest antibody fragment that naturally lacks a light chain, contains only a heavy chain, has an antigen binding capacity, and has a molecular weight of 15kDa, and the structure of a single-domain antibody consists of 4 framework regions mainly responsible for protein architecture and folding and 3 antigen-binding loops or complementarity determining regions responsible for highly variable interactions with antigens. Single domain antibodies, while simple in structure and small in size, possess a high affinity for recognizing certain smaller or more cryptic antigens on the cell surface. The smaller size of single domain antibodies is also an advantage of gene therapy vector expression because it reduces the size of the transgene, which increases the titer of the viral gene therapy vector. The single domain antibody has long storage time, good stability and resistance to chemical denaturation and thermal denaturation. The economic production cost of single domain antibodies is low. Single domain antibodies are increasingly known as alternatives to disease treatment and diagnosis due to their unique properties.
A novel trivalent nanobody, sonelokimab, is disclosed in the prior art (IL 17A/F nanobody Sonelokimab in patients with plaque psoriasis: a multicentric, randomised, displabo-controlled, phase 2b study,Kim A Papp,Mark A Weinberg,Alun Morris,Clinical Trial,2021;397 (10284): 1564-1575.) and consists of monovalent camelidae nanobodies specific for human interleukin-17A, IL-17F and human serum albumin. In the experimental process, the performances of the nano antibody, such as the therapeutic effect, safety and the like on plaque psoriasis patients are evaluated, and in the experimental process, secukinumab is adopted as a positive control, and experimental results show that compared with a placebo group, four doses of sonelokimab can be applied to effectively treat the psoriasis patients, and the nano antibody has better safety and tolerance.
In the prior art, less research is carried out on the anti-IL-17A single domain antibody, and the field still needs to develop a novel anti-IL-17A single domain antibody which can be combined with IL-17A with high affinity, and has stronger stability and good blocking effect.
The present invention is a patent application filed by the inventors for an anti-IL-17A antibody developed autonomously.
In addition to the antibodies claimed in the present invention, the inventors developed 8 additional antibodies at the same time, each of which was claimed for 9 different antibodies based on the relevant regulations of patent law singleness.
The inventors have also developed 12 tandem antibodies from these 9 antibodies, and separately claimed 12 different tandem antibodies based on the relevant regulations of patent law singleness.
The inventors have also developed a genetically modified stem cell technology based on these 12 tandem antibodies, and separately claimed 3 different genetically modified stem cells and 3 different applications based on the relevant regulations of patent law singleness.
For the convenience of understanding the technical scheme of the present invention, reference is optionally made to other series of application texts of the present project.
Disclosure of Invention
Terminology and statement of the invention:
1. as used herein, the term "amino acid": comprising natural amino acids, synthetic amino acids, amino acid analogs and amino acid mimics that function in a manner similar to natural amino acids. Natural amino acids are amino acids encoded by the genetic code. Amino acid analogs refer to those compounds that have the same basic chemical structure as a naturally occurring amino acid. Amino acids may be referred to herein by their commonly known three-letter symbols or by the one-letter symbols recommended by the IUPAC-IUB biochemical nomenclature committee (Biochemical Nomenclature Commission).
2. As used herein, the term "antibody": refers to a polypeptide or fragment thereof that specifically binds to and recognizes an antigen, including the framework regions of immunoglobulin genes. The use of the term antibody is meant to include whole antibodies and antigen-binding fragments thereof. The term antibody encompasses monospecific antibodies, bispecific antibodies, and multispecific antibodies so long as they exhibit the desired biological activity or function. In the present invention, an antibody refers to a polypeptide having IL-17A binding activity, comprising a CDR1 region as set forth in SEQ ID NO. 1, a CDR2 region as set forth in SEQ ID NO. 2 and a CDR3 region as set forth in SEQ ID NO. 3, and also includes a variant of a polypeptide having the same function as the antibody of the present invention comprising the above-mentioned CDR regions. The variant forms comprise: deletion, insertion and/or substitution of one or more amino acids, and addition of one or more amino acids at the C-terminal and/or terminal end.
3. As used herein, the term "monoclonal antibody": refers to antibodies obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific for a single antigenic site. Furthermore, each monoclonal antibody is directed against a single determinant on the antigen, as compared to conventional (polyclonal) antibody preparations, which typically comprise different antibodies directed against different determinants (epitopes). The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
4. As used herein, the terms "single domain antibody (VHH)", "single domain antibody" (single domain antibody, sdAb, or nanobody) have the same meaning, referring to the variable region of a cloned antibody heavy chain, a single domain antibody consisting of only one heavy chain variable region is constructed, which is the smallest antigen-binding fragment with complete function. Typically, after an antibody is obtained which naturally lacks the light and heavy chain constant region 1 (CH 1), the variable region of the heavy chain of the antibody is cloned, and a single domain antibody (VHH) consisting of only one heavy chain variable region is constructed. "Single-domain antibody of the invention", "anti-IL-17A single-domain antibody of the invention" are used interchangeably and refer to a single-domain antibody that specifically recognizes and binds to IL-17A.
5. As used herein, the term "homology" or "identity" refers to sequence similarity between two peptides or two nucleic acid molecules. Homology may be determined by comparing positions in each sequence, which may be aligned for comparison purposes. When a position in the comparison sequence is occupied by the same base or amino acid, then the molecules are homologous at that position.
6. As used herein, the term "epitope" refers to any antigenic determinant on an antigen to which the paratope of an antibody binds. An epitope typically comprises a chemically active surface group of a molecule, such as an amino acid or sugar side chain, and typically has specific three-dimensional structural features as well as specific charge features. 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 spatial conformation, which may be a "linear" epitope or a "conformational" epitope.
7. As used herein, the term "amino acid sequence" refers to the order in which amino acids are linked to each other to form a peptide chain (or polypeptide), and the amino acid sequence can only be read in one direction.
8. As used herein, the term "nucleic acid molecule" may be in the form of DNA or RNA. DNA forms include cDNA, genomic DNA, or synthetic DNA. The DNA may be single-stranded or double-stranded. The DNA may be a coding strand or a non-coding strand. The full-length sequence of the nucleotide molecule or a fragment thereof in the present invention can be generally obtained by a PCR amplification method or an artificial synthesis method.
9. As used herein, the term "nucleotide sequence" refers to the order of bases in DNA or RNA, i.e., A, T, G, C in DNA, or A, U, G, C in mRNA, including rRNA, tRNA, mRNA.
10. As used herein, the term "framework region" is a region of the framework, which varies widely about 110 amino acid sequences near the N-terminus of the H and L chains of an immunoglobulin, and the amino acid sequences of the other parts are relatively constant, whereby the light and heavy chains can be distinguished as variable (V) and constant (C) regions. The variable region comprises the hypervariable region HVR or complementarity determining region CDR and FR framework regions.
11. As used herein, the term "humanized" antibody refers to the antibody in which the variable region (VH or VHH) Fr region portion, the constant region portion (i.e., CH and CL regions) or all of the antibody is encoded by a human antibody gene. Humanized antibodies include chimeric antibodies, diabodies, and fully humanized antibodies.
12. As used herein, the term "Fc region" refers to the C-terminal region of an immunoglobulin, which is a functional building block consisting of only CH2 and CH3 in the heavy chain constant domain. Fc has no ability to bind antigen, however it has the property of having an extended half-life and has a constant amino acid sequence.
13. As used herein, the term "variable" means that certain portions of the variable regions in an antibody differ in sequence, which results in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the antibody variable region. It is concentrated in three segments of the light and heavy chain variable regions, known as complementarity determining regions or hypervariable regions. Herein, "variable region" is used interchangeably with "complementarity determining region".
14. The terms "hypervariable region," "complementarity determining region," "HVR," or "CDR," as used herein, refer to regions that are hypervariable in sequence and/or form structurally defined loops ("hypervariable loops") in an antibody variable domain region. Typically, a natural four-chain antibody comprises six HVRs or CDRs: three are present in VH (H1, H2, H3) and three are present in VL (L1, L2, L3). Based on the Chothia definition rules, exemplary CDRs (LCDR 1, LCDR2, LCDR3, HCDR1, HCDR2, and HCDR 3) are located at amino acid residues L26-L32 (L1), L50-L52 (L2), L91-L96 (L3), H26-H32 (H1), H52-H56 (H2), and H96-H101 (H3) (Chothia et al, J.mol.biol.196:901-917 (1987)). Based on the Kabat definition rules, exemplary CDRs (LCDR 1, LCDR2, LCDR3, HCDR1, HCDR2, and HCDR 3) are located at amino acid residues L24-L34 (L1), L50-L56 (L2), L89-L97 (L3), H31-H35 (H1), H50-H65 (H2), and H95-H102 (H3) (Kabat et al, sequences of Proteins ofImmunological Interest, the fifth edition, public Health Service, national Institutes of Health, bethesda, MD (1991)). Based on IMGT definition rules, exemplary CDRs (LCDR 1, LCDR2, LCDR3, HCDR1, HCDR2, and HCDR 3) are located at amino acid residues L27-L32 (L1), L50-L51 (L2), L89-L97 (L3), H26-H33 (H1), H51-H56 (H2), and H93-H102 (H3) (Honjo, T. And Alt, F.W. (1995) Immunoglobulin genes.academic Press pp.3-443). It is well known to those skilled in the art that the CDRs of an antibody can be defined in a variety of ways, such as Kabat definition rules based on sequence variability, chothia definition rules based on structural loop region positions, and reference tools for humanized design of antibodies based on CDR grafting (see J Mol biol.273:927-48, 1997). It will be appreciated by those skilled in the art that unless otherwise specified, the terms "CDR" and "complementarity determining region" of a given antibody or region thereof (e.g., variable region) are to be understood as encompassing complementarity determining regions defined in any of the above known schemes as described by the present invention. Although the scope of the present disclosure is based on the sequences shown by IMGT definition rules, amino acid sequences corresponding to other CDR definition rules shall also fall within the scope of the present invention.
15. The terms "include" or "comprising" as used herein mean "including but not limited to". The term is intended to be open ended to specify the presence of any stated features, elements, integers, steps, or components, but does not preclude the presence or addition of one or more other features, elements, integers, steps, components, or groups thereof. Thus, the term "comprising" includes the more limiting terms "consisting of … …" and "consisting essentially of … …". In one embodiment, the term "comprising" as used throughout the application, and in particular in the claims, may be replaced by the term "consisting of … …". The amino acid three-letter codes and one-letter codes used herein are as known to those skilled in the art, or as described in J biol. Chem,243, p3558 (1968).
16. The terms "optional," "any," or "any" used herein mean that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs or does not. For example, "optionally comprising 1 antibody heavy chain variable region" means that an antibody heavy chain variable region of a particular sequence may be, but is not required to be, present.
17. The term "about" as used herein means a range of + -20% of the numerical values thereafter. In some embodiments, the term "about" means a range of ±10% of the numerical value following that. In some embodiments, the term "about" means a range of ±5% of the numerical value following that.
18. The term "and/or" as used herein is understood to mean any one of the selectable items or a combination of any two or more of the selectable items.
19. The term "IL-17A" or "interleukin-17A" as used herein refers to a cytokine, belonging to the interleukin 17 family, produced by T cells and other types of immune cells, and playing an important role in the immune system. IL-17A is produced primarily by Th17 cells, and other cells including CD8+ T cells, γδ T cells, NK cells and neutrophils, mast cells and macrophages also express IL-17A. It acts primarily on immune cells, such as macrophages, neutrophils and endothelial cells, inducing an inflammatory response. In some examples, the term includes variants, homologs, orthologs, and paralogs. For example, antibodies specific for human IL-17A may in some cases cross-react with IL-17A protein of another species, such as a monkey. In other embodiments, antibodies specific for human IL-17A protein may be completely specific for human IL-17A protein without cross-reacting with other species or other types of proteins, or may cross-react with IL-17A proteins of some other species but not all other species.
20. The term "anti-IL-17A (single domain or nanobody)" or "IL-17A (single domain or nanobody)" as used herein refers to an antibody that specifically binds IL-17A and partially or completely neutralizes, inhibits or attenuates IL-17A activity, and/or inactivates IL-17A, prevents IL-17A responses, or downstream pathways mediated by IL-17A, or other IL-17A mediated functions.
21. The term "antibody" as used herein refers to a glycoprotein comprising a heavy chain (H) and a light chain (L) interconnected by disulfide bonds (S S). Each heavy chain consists of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region (abbreviated herein as 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 (abbreviated herein as VL) and a light chain constant region (abbreviated herein as VH). The light chain constant region consists of one domain CL. Light chains fall into two categories, kappa-type light chains and lambda-type light chains, respectively (e.g., light chain constant regions Ckappa/lambda in the present invention mean that the light chain constant regions are kappa-type light chains or lambda-type light chains). The VH and VL regions may be further subdivided into hypervariable regions (also known as Complementarity Determining Regions (CDRs)) with more conserved framework or Framework Regions (FR) interposed therebetween. Each VH and VL consists of three CDRs and 4 FRs, arranged from amino-terminus to carboxyl-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. Antibodies include monospecific antibodies, bispecific antibodies, and multispecific antibodies so long as they exhibit the desired biological activity or function.
22. The terms "activity", "functional activity" or "biological activity", or the terms "biological property" or "biological feature" as used herein are used interchangeably and include, but are not limited to, epitope/antigen affinity and specificity, the ability to neutralize or antagonize IL-17A activity in vivo or in vitro, IC50, in vivo stability of antibodies, and immunogenic properties of antibodies. Other identifiable biological properties or characteristics of antibodies known in the art include, for example, cross-reactivity (i.e., cross-reactivity with non-human homologs of the targeting peptide in general, or with other proteins or tissues), and the ability to maintain high levels of expression of the protein in mammalian cells. The aforementioned properties or characteristics are observed, assayed or assessed using techniques well known in the art, including but not limited to ELISA, FACS or BIACORE plasma resonance analysis, unrestricted in vitro or in vivo neutralization assays, receptor binding, cytokine or growth factor production and/or secretion, signal transduction, and immunohistochemistry of tissue sections of different origin (including human, primate or any other source).
23. The term "affinity" or "binding affinity" as used herein refers to an inherent binding affinity that reflects interactions between members of a binding pair. The affinity of a molecule X for its partner Y can be generally represented by the equilibrium dissociation constant (KD), which is the ratio of the dissociation rate constant and the binding rate constant (Koff and Kon, respectively). Affinity can be measured by common methods known in the art. One specific method for measuring affinity is the ForteBio kinetic binding assay herein.
24. The term "high affinity" or "high affinity" as used herein, with respect to IgG antibodies, refers to KD of 1.0 x 10 for antigen -6 M or less, preferably 5.0X10-8M or less, more preferably 1.0X10 -8 M or less, 5.0X10 s -9 M or less, more preferably 1.0X10 -9 M or lower. For other antibody subtypes, "high affinity" binding may vary. For example, "high affinity" binding of IgM subtype refers to KD of 10 -6 M or less, preferably 10 -7 M or less, more preferably 10 -8 M or lower.
25. The term "antibody drug conjugate" refers to a substance obtained by linking a biologically active compound fragment to an antibody or antigen binding fragment portion thereof. The biologically active compound fragment and the targeting moiety may be linked by a linker. The linker is capable of cleaving in a specific environment (e.g., an intracellular low pH environment) or under a specific action (e.g., action of a lysosomal protease) to thereby separate the biologically active compound fragment from the targeting moiety or antibody or antigen binding fragment thereof. The linker comprises a cleavable or non-cleavable unit, such as a peptide or disulfide bond. The biologically active compound fragment is directly linked to the targeting moiety or antibody or antigen binding fragment thereof by a covalent bond that is capable of cleavage under a specific environment or action, thereby separating the biologically active compound fragment from the antibody or antigen binding fragment portion thereof.
26. The term "nucleic acid" or "polynucleotide" as used herein refers to deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) and polymers thereof in single or double stranded form. Unless specifically limited, the term includes nucleic acids containing known analogues of natural nucleotides that have similar binding properties to the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides (see, U.S. Pat. No.8278036 to Kariko et al, which discloses mRNA molecules with uridine replaced by pseudouridine, methods of synthesizing the mRNA molecules, and methods for delivering therapeutic proteins in vivo). Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences, as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed bases and/or deoxyinosine residues (Batzer, nucleic Acid Res.19:5081 (1991); ohtsuka, J.biol.chem.260:2605-2608 (1985); rossolini, mol.cell.probes 8:91 98 (1994)).
27. The term "vector" as used herein refers to any recombinant polynucleotide construct that can be used for transformation purposes (i.e., the introduction of heterologous DNA into a host cell). One type of vector is a "plasmid," which refers to a circular double-stranded DNA loop into which additional DNA segments may be ligated. Another type of vector is a viral vector, in which additional DNA segments can be ligated into the viral genome. Some vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Upon introduction into a host cell, other vectors (e.g., non-episomal mammalian vectors) integrate into the genome of the host cell and thereby replicate together with the host genome. In addition, certain vectors are capable of directing the expression of genes that are operably linked. Such vectors are referred to herein as "expression vectors".
28. The term "expression vector" as used herein refers to a nucleic acid molecule capable of replicating and expressing a gene of interest when transformed, transfected or transduced into a host cell. The expression vector contains one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to provide amplification in the host if desired.
29. The term "pharmaceutical composition" as used herein generally refers to a formulation that exists in a form that allows for the biological activity of the active ingredient to be effective and that does not contain additional ingredients that have unacceptable toxicity to the subject to which the composition is to be administered. The composition is sterile.
30. The term "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
31. The term "subject" as used herein includes any human or non-human animal. The term "non-human animal" includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, and the like.
32. The terms "therapeutically effective amount," "therapeutically effective dose," and "effective amount" as used herein refer to an amount of an anti-IL-17A antibody or antigen-binding fragment thereof of the invention that is effective to prevent or ameliorate a symptom of one or more diseases or conditions, or the development of the disease or condition, when administered to a cell, tissue, or subject, alone or in combination with other therapeutic agents. A therapeutically effective dose also refers to an amount of an antibody or antigen binding fragment thereof sufficient to result in an improvement in symptoms, e.g., to treat, cure, prevent, or ameliorate a related medical condition, or to increase the rate of treatment, cure, prevention, or amelioration of such a condition. When an active ingredient is administered to an individual, a therapeutically effective dose refers to that ingredient alone. When administered in combination, a therapeutically effective dose refers to the combined amount of the active ingredients that results in a therapeutic effect, whether administered in combination, sequentially or simultaneously. An effective amount of the therapeutic agent will result in an increase in the diagnostic criteria or parameter of at least 10%; typically at least 20%; preferably at least about 30%; more preferably at least 40%, most preferably at least 50%.
33. The term "EC50" as used herein refers to the half maximal effect concentration (concentration for 50%of maximal effect), i.e. the concentration that causes 50% of the maximal effect.
34. The term "cancer" or "tumor" as used herein refers to a physiological condition characterized by unregulated cell growth in a mammal. Included within this definition are benign and malignant cancers, and dormant tumors or micrometastases.
35. The term "autoimmune disease" or "autoimmune disease" as used herein refers to a disease caused by the fact that the immune system has a reduced, decreased and/or destroyed immune tolerance to its own components for some reason, resulting in damage of the tissues of the organs by autoantibodies and/or sensitized lymphocytes, and is manifested as dysfunction of the corresponding tissues and organs.
The technical scheme of the invention is as follows:
in a first aspect, the invention provides an antibody comprising the amino acid sequences of HCDR1, HCDR2 and HCDR 3; the amino acid sequence of the HCDR1 is shown as SEQ ID NO. 1, the amino acid sequence of the HCDR2 is shown as SEQ ID NO. 2, and the amino acid sequence of the HCDR3 is shown as SEQ ID NO. 3; and/or an amino acid sequence having at least 80% sequence similarity with SEQ ID NO. 1, SEQ ID NO. 2 and SEQ ID NO. 3, respectively.
Preferably, the amino acid sequence of the antibody comprises: an amino acid sequence obtained by at least one of substitution, deletion, addition and/or modification on the amino acid sequence shown in SEQ ID NO. 1-3.
Further preferably, the amino acid sequence of the antibody comprises: amino acid sequences having a difference of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11 amino acids compared to the amino acid sequences shown in SEQ ID NO. 1-3.
SEQ ID NO. 1 is: ESLLRLYA.
SEQ ID NO. 2 is: HTTSDTT.
SEQ ID NO. 3 is: HVTSMRDSQNY.
In particular, the substitution of one or more amino acids may be conservative substitutions of one or more amino acids. Such conservative substitutions are preferably substitutions in which one amino acid in the following groups (a) to (e) is substituted by another amino acid residue in the same group: (a) small aliphatic, non-polar or weakly polar residues: ala, ser, thr, pro and Gly; (b) Polar, negatively charged residues and (uncharged) amides: asp, asn, glu and Gln; (c) polar, positively charged residues: his, arg and Lys; (d) large aliphatic, nonpolar residues: met, leu, he, val and Cys; and (e) an aromatic residue: phe, tyr and Trp.
Further, conservative substitutions are as follows: ala to Gly or to Ser; arg to Lys; asn to Gln or to His; asp to Glu; cys to Ser; gln to Asn; glu to Asp; gly to Ala or to Pro; his to Asn or to Gln; lie to Leu or to Val; leu to Ile or to Val; lys to Arg, to gin, or to Glu; met to Leu, to Tyr or to Ile; phe to Met, to Leu, or to Tyr; ser to Thr; thr to Ser; trp to Tyr; tyr to Trp; and/or Phe to Val, to Ile or to Leu.
According to some embodiments of the invention, the functionally active variant is a single domain antibody variant (mutant) having the same or similar affinity or function as the anti-IL-17A single domain antibody of (1), e.g. a variant (mutant) that specifically binds IL-17A and blocks IL-17A binding to its receptor.
In a second aspect, the invention provides an antibody, the amino acid sequence of which comprises the framework region FR; the frame region FR includes: FR1 shown in SEQ ID NO. 4, FR2 shown in SEQ ID NO. 5, FR6 shown in SEQ ID NO. 8 and FR4 shown in SEQ ID NO. 7; and/or; an amino acid sequence having at least 80% sequence identity to the amino acid sequence set forth in SEQ ID NO. 4-7.
Preferably, the amino acid sequence of the antibody comprises: an amino acid sequence obtained by at least one of substitution, deletion, addition and/or modification of the amino acid sequence shown in SEQ ID NO. 4-7.
Preferably, the amino acid sequence of the antibody comprises: amino acid sequences having 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37 or 38 amino acid differences compared to the amino acid sequences shown in SEQ ID NOs 4-7.
SEQ ID NO. 4 is: DVQLVESGGGLVQPGGSLRLSCAAS.
SEQ ID NO. 5 is: MGWYRQLPGQEREWVAI.
SEQ ID NO. 6 is:
NYRDSVKGRFTLSRDVATNTIYLQMTSLKPEDTAVYYC。
SEQ ID NO. 7 is: WGQGTQVTVSS.
In a third aspect, the invention provides an antibody, the amino acid sequence of which comprises: the amino acid sequence of the antibody according to the first aspect and the amino acid sequence of the antibody according to the second aspect.
In a fourth aspect, the invention provides an antibody, the amino acid sequence of which comprises: FR1-HCDR1-FR2-HCDR2-FR3-HCDR3-FR4;
the amino acid sequences of HDR1, HDR2 and HDR3 are shown as SEQ ID NO. 1-3 or functional active variants of the amino acid sequences with amino acid differences of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11 compared with the amino acid sequences shown as SEQ ID NO. 1-3;
the amino acid sequences of the FR1, the FR2, the FR3 and the FR4 are shown as SEQ ID NO. 4-7 or functional active variants of the amino acid sequences with at least 80% sequence similarity compared with the amino acid sequences shown as SEQ ID NO. 4-7.
Preferably, the amino acid sequence of the HCDR1 is shown as SEQ ID NO. 1, the amino acid sequence of the HCDR2 is shown as SEQ ID NO. 2, and the amino acid sequence of the HCDR3 is shown as SEQ ID NO. 3.
Preferably, the amino acid sequence of the FR1 is shown as SEQ ID NO. 4, the amino acid sequence of the FR2 is shown as SEQ ID NO. 5, the amino acid sequence of the FR3 is shown as SEQ ID NO. 6, and the amino acid sequence of the FR4 is shown as SEQ ID NO. 7.
Preferably, the amino acid sequence of the antibody is shown as SEQ ID NO. 8.
SEQ ID NO. 8 is:
DVQLVESGGGLVQPGGSLRLSCAASESLLRLYAMGWYRQLPGQEREWVAIHTTSDTTNYRDSVKGRFTLSRDVATNTIYLQMTSLKPEDTAVYYCHVTSMRDSQNYWGQGTQVTVSS。
further preferably, the antibody is a single domain antibody.
Still further, the antibody is an anti-IL-17A antibody.
Preferably, the antibody comprises part or all of an antibody heavy chain framework region selected from human, murine, primate or camelid origin or a variant thereof;
preferably, the antibody heavy chain framework regions or variants thereof comprise part or all of a source selected from camelids;
more preferably, the heavy chain framework regions of the antibodies or variants thereof are comprised in part or in whole selected from alpaca sources.
In a fifth aspect, the present invention provides a polyclonal antibody comprising an antibody according to any one of the first to fourth aspects above.
In a sixth aspect, the present invention provides an antibody preparation comprising an antibody according to any one of the first to fourth aspects above and a pharmaceutically acceptable carrier.
In a seventh aspect, the present invention provides a recombinant protein comprising an antibody according to any one of the first to fourth aspects above.
Preferably, the recombinant protein further comprises a biologically active protein or functional fragment thereof that assists in its expression and/or secretion, or that extends its half-life in vivo;
further preferably, the biologically active protein or functional fragment thereof is selected from at least one of His tag, GST tag, MBP tag, FLAG tag and SUMO tag, immunoglobulin Fc domain, serum albumin, albumin binding polypeptide, prealbumin, carboxy terminal peptide, elastin-like polypeptide.
Still further, the biologically active protein or functional fragment thereof is a human immunoglobulin Fc domain, preferably a human IgG Fc domain, such as a human IgG1 Fc domain, an IgG2 Fc domain, an IgG3 Fc domain, an IgG4 Fc domain, more preferably a human IgG1 Fc domain.
According to some embodiments of the invention, the amino acid sequence of the human IgG1 Fc is shown in SEQ ID NO. 10.
SEQ ID NO:10:
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK。
Specifically, the nucleotide sequence encoding SEQ ID NO. 10 is shown as SEQ ID NO. 11.
SEQ ID NO:11:
GACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAACTCCTGGGGGGACCGTCAGTCTTCCTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCCCAGCCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGAGGAGATGACCAAGAACCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCACGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGGTAAA。
According to some embodiments of the invention, one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby producing an Fc region variant. An Fc region variant may comprise a human Fc region sequence (e.g., a human IgG1, igG2, igG3, or IgG4 Fc region) comprising amino acid modifications (e.g., substitutions, deletions, and insertions) at one or more amino acid positions.
In an eighth aspect, the present invention provides a kit comprising:
(1) The antibody, the polyclonal antibody, the antibody preparation or the recombinant protein of any one of the first to fourth aspects above; and;
(2) A container for holding the antibody preparation.
Preferably, the kit may further comprise a container, a buffer, an antibody recognizing the IL-17A protein, a detection substrate, etc.
In a ninth aspect, the present invention provides an antibody drug conjugate comprising:
(1) The antibody, the polyclonal antibody, the antibody preparation or the recombinant protein of any one of the first to fourth aspects above; and;
(2) A coupling moiety that binds to (1).
According to some embodiments of the invention, the coupling moiety comprises a detectable label, a drug, a toxin, a cytokine, a radionuclide, and/or an enzyme.
In a tenth aspect, the present invention provides a nucleic acid molecule encoding the antibody, polyclonal antibody or recombinant protein of any one of the first to fourth aspects above.
According to some embodiments of the invention, the nucleic acid may be RNA, DNA or cDNA.
According to some embodiments of the invention, the nucleic acid of the invention may also be in the form of a vector, may be present in and/or may be part of a vector, such as a plasmid, cosmid, or YAC. The vector may be an expression vector, which typically comprises at least one nucleic acid of the invention operably linked to one or more suitable expression control elements (e.g., promoters, enhancers, terminators, etc.).
Specifically, the sequence of the nucleic acid molecule encoding the amino acid sequence shown in SEQ ID NO. 8 is shown in SEQ ID NO. 9.
SEQ ID NO. 9 is:
GATGTGCAGCTGGTGGAGTCTGGGGGAGGCTTGGTGCAGCCTGGGGGGTCTCTGAGACTCTCCTGTGCAGCCTCTGAAAGCCTCCTCAGGTTGTATGCCATGGGCTGGTACCGCCAACTTCCAGGGCAGGAGCGCGAGTGGGTCGCAATACACACTACTAGTGACACCACTAATTATAGAGACTCCGTGAAGGGCCGATTCACGCTCTCCAGAGACGTCGCCACGAACACGATTTATCTCCAAATGACCAGCCTCAAACCTGAAGACACGGCCGTCTATTATTGTCATGTTACTTCCATGAGAGATTCACAAAACTACTGGGGCCAGGGGACCCAGGTCACCGTCTCCTCG。
in an eleventh aspect, the invention provides a biological expression vector comprising the nucleic acid molecule.
According to some embodiments of the invention, the biological expression vector may be a eukaryotic expression vector or a prokaryotic expression vector, preferably a eukaryotic expression vector.
Preferably, the eukaryotic expression vector is selected from a yeast expression vector, an insect expression vector or a mammalian expression vector;
more preferably, the mammalian expression vector is selected from the group consisting of a retroviral expression vector, a lentiviral expression vector, an adenoviral expression vector, and an adeno-associated viral expression vector.
In a twelfth aspect, the invention provides a host cell, the genome of which has the nucleic acid molecule integrated therein; or; comprising said biological expression vector.
Preferably, the host cell is a bacterial cell, a fungal cell or a mammalian cell.
Further, the bacterial cells include cells of gram-negative bacterial strains and gram-positive bacterial strains.
Still further, the gram-negative bacterial strains include E.coli strains, pseudomonas strains, and Proteus strains.
Still further, the gram-positive bacterial strains include Streptomyces strains, bacillus strains, and Staphylococcus strains.
Further, the fungal cells include cells of the species Trichoderma, neurospora, and Aspergillus.
Further, the mammalian cells include HEK293 cells, heLa cells, CHO cells and COS cells.
Still further, the mammalian cells are HEK293 cells.
In a thirteenth aspect, the invention provides the use of an antibody, polyclonal antibody, antibody preparation, recombinant protein, kit, antibody drug conjugate, nucleic acid molecule, biological expression vector or host cell according to any one of the first to fourth aspects, wherein the use is selected from at least one of the following:
(1) Preparing a detection reagent or a kit;
(2) Preparing a medicament for preventing and/or treating autoimmune diseases;
(3) Preparing medicine for preventing and/or treating cancer.
In particular, the method comprises the steps of, the autoimmune diseases include psoriasis, systemic lupus erythematosus, malignant rheumatoid arthritis, adult stell disease, basse Du Bing, scleroderma, hashimoto disease, allergic vasculitis, juvenile idiopathic arthritis, temporal arteritis, myasthenia gravis, rheumatoid vasculitis, autoimmune optic neuropathy, polymyalgia rheumatica, polymyositis, multiple sclerosis, fibromyalgia, systemic scleroderma, dermatomyositis, acute glomerulonephritis, behcet's disease, progressive systemic sclerosis, sjogren's syndrome, white spot, malignant anemia, vasculitis, macrovasculitis, bullous pemphigoid, adje's disease, kaschmann's disease, giant cell arteritis, idiopathic Adje's disease, psoriasis arthritis, multifocal motor neuropathy, chronic inflammatory demyelinating polyneuropathy, chronic inflammatory demyelinating disease perinodular arteritis, amyotrophic lateral sclerosis, idiopathic azoospermia, herpes gestation, aortic inflammatory syndrome, autoimmune hemolytic anemia, eosinophilic fasciitis, pemphigoid, spondyloarthritis, leukoplakia, primary biliary cirrhosis, mixed connective tissue disease, chronic atrophic gastritis, antiphospholipid antibody syndrome, allergic granulomatous vasculitis, good-pasture syndrome, cogan syndrome, ANCA-related vasculitis, linear IgA bullous dermatoses, RS3PE syndrome, chronic discoid lupus erythematosus, igG 4-related diseases, guillain Barre syndrome, autoimmune hepatitis, acquired bullous epidermolysis, non-alcoholic steatohepatitis, lupus nephritis, type I diabetes, megaloblastic anemia, autoimmune neutropenia, idiopathic thrombocytopenic purpura, autoimmune diabetes mellitus, autoimmune hypothyroidism, primary hypothyroidism, slowly progressing type I diabetes, focal scleroderma, pemphigus, alopecia areata, neuromyelitis optica, sarcoidosis, origin-field disease, habitual abortion, inflammatory bowel disease, celiac disease, ankylosing spondylitis, severe asthma, chronic urticaria transplantation immunity, familial mediterranean fever, eosinophilic chronic sinusitis, dilated cardiomyopathy, systemic mastocytosis or inclusion body myositis.
In particular, the cancer comprises oral cancer, lymphoma, respiratory system cancer, prostate cancer, leukemia, retinoblastoma, choriocarcinoma, peritoneal cancer, eye cancer, urinary system cancer, bone cancer, endometrial cancer, uterine cancer, bladder cancer, thyroid cancer, esophageal cancer, laryngeal cancer, head and neck cancer, acute lymphoblastic leukemia, melanoma, hairy cell leukemia, neuroblastoma, salivary gland cancer, digestive system cancer, myeloma, gastric cancer, liver cancer, skin cancer, lung cancer, rectal cancer, ovarian cancer, kidney cancer, pancreatic cancer, liver cancer, sarcoma, glioblastoma, basal cell carcinoma, cholangiocarcinoma, breast cancer, cervical cancer, cholangiocarcinoma, colorectal cancer, connective tissue cancer, rhabdomyosarcoma, squamous cell carcinoma, testicular cancer, B cell lymphoma, chronic lymphocytic leukemia, chronic myeloblastic leukemia.
In a fourteenth aspect, the present invention provides a pharmaceutical composition comprising: the antibody, the polyclonal antibody, the antibody preparation, the recombinant protein, the antibody drug conjugate, the nucleic acid molecule, the biological expression vector, or the host cell of any one of the first to fourth aspects above.
Specifically, the pharmaceutically acceptable auxiliary materials are selected from at least one of solvents, diluents, disintegrants, precipitation inhibitors, surfactants, glidants, binders, lubricants, dispersants, suspending agents, isotonic agents, thickening agents, emulsifying agents, preservatives, stabilizers, hydration agents, emulsifying accelerators, buffering agents, absorbents, colorants, flavoring agents, sweeteners, ion exchangers, mold release agents, coating agents, flavoring agents and antioxidants.
In a fifteenth aspect, the present invention provides a method for detecting IL-17A in a sample in vitro for non-diagnostic purposes, said method comprising the steps of:
(1) Binding the antibody, the polyclonal antibody, the antibody preparation, the recombinant protein, the antibody drug conjugate of any one of the first to fourth aspects to a test sample;
(2) The antigen-antibody complex is detected and the results are interpreted.
In a sixteenth aspect, the present invention provides a method for preventing and/or treating an autoimmune disease, the method comprising: administering to a subject a therapeutically effective amount of the antibody, the polyclonal antibody, the antibody preparation, the recombinant protein, the antibody drug conjugate, the nucleic acid molecule, the biological expression vector, the host cell, or the pharmaceutical composition of any one of the first to fourth aspects above.
In a seventeenth aspect, the present invention provides a method for preventing and/or treating cancer, the method comprising:
administering to a subject a therapeutically effective amount of the antibody, the polyclonal antibody, the antibody preparation, the recombinant protein, the antibody drug conjugate, the nucleic acid molecule, the biological expression vector, the host cell, or the pharmaceutical composition of any one of the first to fourth aspects above.
The beneficial effects of the invention include:
compared with the traditional monoclonal antibody, the affinity of the single domain antibody 1-C11 is higher, the EC50 value of the single domain antibody 1-C11 is 29.99, and the EC 50=10.06 of the positive antibody (Ixekizumab), the single domain antibody 1-C11 has better binding activity with the Human IL-17A protein.
The IL-17A single domain antibody 1-C11 has a good blocking effect, and can block Human IL-17A protein from activating 293F-IL-17RA-IL-17Rc-ACT1-NF kappa B-Luc.
The stability of the single domain antibodies 1-C11 was strong, tm=64.25, tagg= 81.13.
Drawings
FIG. 1 is a diagram showing the SDS-PAGE detection result of IL-17A recombinant protein.
FIG. 2 is a graph showing the experimental results of activating NIH-3T3 cells with purchased IL-17A (Acro) protein.
FIG. 3 is a graph showing the experimental results of the activation of NIH-3T3 cells by recombinant IL-17A (TEST) protein.
FIG. 4 is a graph showing the results of experiments on binding IL-17A to reporter cell lines.
FIG. 5 shows the results of an IL-17A activating cell assay
FIG. 6 shows agarose gel electrophoresis of alpaca # 2.
FIG. 7 is an agarose gel electrophoresis of a 2++ 3# alpaca.
Fig. 8 is a graph of the results of a yeast pool flow assay for alpaca # 2, a: NC group: an antibody: without addition, the secondary antibody: PE-streptavidine; original library experimental group: an antibody: IL-17A-His-Biotin, secondary antibody: PE-streptavidine, alexa Fluor 647-V5; c: MACS experimental group 1: an antibody: IL-17A-His-Biotin, secondary antibody: PE-streptavidine, alexa Fluor 647-V5; d:2MACS experimental group: an antibody: IL-17A-His-Biotin, secondary antibody: PE-strepitavidin, alexa Fluor 647-V5.
FIG. 9 is a graph showing the results of a yeast pool flow assay for 2++ 3# alpaca, A: NC group: an antibody: without addition, the secondary antibody: PE-streptavidine; original library experimental group: an antibody: IL-17A-His-Biotin, secondary antibody: PE-streptavidine, alexa Fluor 647-V5; c: MACS experimental group 1: an antibody: IL-17A-His-Biotin, secondary antibody: PE-streptavidine, alexa Fluor 647-V5; d:2MACS experimental group: an antibody: IL-17A-His-Biotin, secondary antibody: PE-strepitavidin, alexa Fluor 647-V5.
FIG. 10 shows FACS detection of IL-17A target monoclonal binding to target in alpaca # 2.
FIG. 11 shows FACS detection of IL-17A target monoclonal binding to target in 2++3 llama.
FIG. 12 shows the result of SDS-PAGE of purified antibodies, wherein the left lane is marker and the right lane is 1-C11.
FIG. 13 is a graph showing the results of ELISA Binding experiments for candidate antibodies.
FIG. 14 shows the results of candidate antibody blocking function experiments.
FIG. 15 shows the results of the thermostability test of the single domain antibodies 1-C11.
FIG. 16 shows the results of the thermal stability test of the positive control antibody.
Detailed Description
In order to make the technical means, the creation features, the achievement of the purpose and the effect of the present invention easy to understand, the present invention will be further elucidated with reference to the specific embodiments, but the following embodiments are only preferred embodiments of the present invention, not all of them. Based on the examples in the embodiments, those skilled in the art can obtain other examples without making any inventive effort, which fall within the scope of the invention. In the following examples, unless otherwise specified, the methods of operation used were conventional, the equipment used was conventional, and the materials used in the examples were the same.
Experimental reagent:
agar (Sigma, cat#a1296); peptone (Sigma, cat# 93926); yeast extract (OXOID, cat#: LP 0021); sodium chloride (Allatin, CAT#: C111533); potassium chloride (aladine, cat#: P112133); magnesium sulfate (national medicine, cat#: 10013018); magnesium chloride (national medicine, cat#: 10012818); glucose (Innovative, CAT#: GT 1991); sfiI (NEB, CAT#: R0123L); t4 DNA library (TaKaRa, CAT#: 2011A); primeScript TM II 1st Strand cDNA Synthesis Kit (TaKaRa, CAT#: 6210B); nuHi power mix (new sea creature, cat#: NH 9303); 3M sodium acetate (pH 5.2-6) (Sigma, CAT#: 126-96-5); DNA fragment recovery kit (TakaRa, CAT#: 9761); glue recovery kit (Qiagen, CAT#: 28706); the Tiangen plasmid large drawing kit (Tiangen, CAT#: DP 117); HRP-Anti-M13 (iCarTab); PE-anti-Human IgG (eBioscience, cat#:124998-82);PE-Streptavidin(Biolegend,405204);Rabbit anti-Llama IgG(H+L)Secondary Antibody[HRP](Novus, cat#nbp1-75095); SS320 competence (iCarTab); BL21 competence (Biomed, BC 201-02); pComF phage display vector (iCarTab); NHS-biotin (APExBIO, CAT#: A8002); HRP-strepitavidin (Boster, CAT#: BA 1088); HRP-protein a (Boster, BA 1080);ProA Biosensors(Sartorius,CAT#:18-5010);PBS(Gbico,CAT#14190-250);DMEM(Gbico,CAT#41965-062);RPMI1640(Gbico,CAT#61870044);FBS(VivaCell,CAT#C04001-500);Genomic DNA Purification Kit(Lifetech,CAT#K0512);Mouse-IL-17A-His(ACRO,CT8-M5240);Bright-Lite Luciferase Assay System(Vazyme,CAT#DD1204-01);NHS-biotin(APExBIO,CAT#:A8002)。
experiment consumable:
50mL Falcon centrifuge tube (Corning, CAT # 352070); electric stun cup (Bio-Rad 0.2 cm); RNase free 1.5mL EP tube (QSP, CAT#: 509-GRD-Q); 200. Mu.L RNase free PCR tube (Axygen, PCR-02D-C); t125 shake flask (Corning, CAT# 431143); 15mL Falcon centrifuge tube (Corning, CAT # 430052); 6-well plate (Corning, cat#3516); 96-well plates (Corning, cat#3365); 96 Kong Heiban (F-BOTTOM (CHIMNEY WELL) BLACK).
Experimental facilities:
an electrotransport device (Eppendorf Multiporator); centrifuge (Thermo FRESCO-17); constant temperature incubator (Shanghai extract macro DNP-9052); constant temperature shake incubator (refined Qi CO-O6U); ultra clean bench (Sujing Antai SW-CJ-1 FD); PCR instrument (Applied Biosystems ABI 2720); biosafety cabinet (halr, HR40-IIA 2); a flow cytometer (Thermo Attune Nxt flow cytometer); thermo 3111CO 2 An incubator; forteBio OCTET R2.
The primers used for screening, cloning VHH fragments, and constructing nanobodies in the following examples were designed with reference to the following references:
Maass DR,Sepulveda J,Pernthaner A,Shoemaker CB.Alpaca(Lama pacos)as a convenient source of recombinant camelid heavy chain antibodies(VHHs).JImmunol Methods.2007;324(1-2):13-25.
Lin,J,Gu,Y,Xu,Y et al.Characterization and applications of nanobodies against Pseudomonas aeruginosa exotoxin a selected from single alpaca B cells.Biotechnol Biotechnol Equip 2020;34:1028-37.
Studies on design of singledomain antibodies by AlpacaVHH phage library and high throughput sequencing toconstruct Fab antibody purification system(http://hdl.handle.net/10232/00030916)。
example 1
1. IL-17A (Human) recombinant protein preparation
Retrieving Human IL-17A (Q16552-1) sequence information (SEQ ID NO: 11) from a UniProt database, adding a 6xHis tag at the C end, performing gene synthesis according to prokaryotic codon optimization, and subcloning into a pET28a vector; after verification of Sanger sequencing, plasmid extraction was performed.
Transforming the recombinant plasmid into BL21 competent, inducing overnight with 0.5mM IPTG, and collecting bacterial liquid for cleavage; the recombinant protein was purified using a nickel column.
SDS-PAGE detects the purity of the target protein, and the result shows that the purity of the IL-17A antigen protein is more than 90% after being refined.
SEQ ID NO:12:
MTPGKTSLVSLLLLLSLEAIVKAGITIPRNPGCPNSEDKNFPRTVMVNLNIHNRNTNTNPKRSSDYYNRSTSPWNLHRNEDPERYPSVIWEAKCRHLGCINADGNVDYHMNSVPIQQEILVLRREPPHCPNSFRLEKILVSVGCTCVTPIVHHVA。
2. Preparation of positive control antibody Ixekizumab
(1) Gene synthesis of Ixekizumab heavy chain and light chain variable region (the amino acid sequence of the heavy chain variable region is shown as SEQ ID NO:13, the amino acid sequence of the light chain variable region is shown as SEQ ID NO: 14), subcloning the heavy chain variable region into pcDNA3.4-hIgG4 (the amino acid sequence of IgG4 is shown as SEQ ID NO: 15) vector, subcloning the light chain variable region into pcDNA3.4-hIgKc vector (the amino acid sequence of IgG KC is shown as SEQ ID NO: 16); after verification by Sanger sequencing, the plasmid megapump kit is used for preparing the endotoxin-removing plasmid for standby.
(2) And taking out the LVTransm transfection reagent, the heavy chain expression vector and the light chain expression vector from the refrigerator, thawing at room temperature, and blowing up and down by a pipetting gun to be completely and uniformly mixed. The PBS buffer was removed and warmed to room temperature. Taking 2mL of PBS to one hole of a 6-hole plate, respectively adding 50 mug of heavy chain expression vector and light chain expression vector, fully and uniformly mixing by up-and-down blowing of a pipette, adding 300 mug of LVTransm, immediately uniformly mixing by up-and-down blowing of the pipette, and standing for 10 minutes at room temperature.
(3) Adding the above DNA/LVTransm complex into 100mL of 293F cells, gently shaking, mixing, and placing the cells at 37deg.C and 5% CO 2 Incubator, 130RPM continued to culture.
(4) After continuous cultivation for 5-7 days, the culture supernatant was collected by centrifugation, filtered with a 0.45 μm filter membrane, and the filtrate was transferred to a sterile centrifuge tube and the antibody was purified using a Protein A column.
SDS-PAGE detects purity of target antibody protein, purity >95%.
SEQ ID NO:13:
QVQLVQSGAEVKKPGSSVKVSCKASGYSFTDYHIHWVRQAPGQGLEWMGVINPMYGTTDYNQRFKGRVTITADESTSTAYMELSSLRSEDTAVYYCARYDYFTGTGVYWGQGTLVTVSS。
SEQ ID NO:14:
DIVMTQTPLSLSVTPGQPASISCRSSRSLVHSRGNTYLHWYLQKPGQSPQLLIYKVSNRFIGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQSTHLPFTFGQGTKLEIK。
SEQ ID NO:15:
ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK。
SEQ ID NO:16:
RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC。
3. ELISA detection of binding Activity of Human IL-17A recombinant protein and control antibody
(1) The IL-17A recombinant protein was diluted with sterile CBS to a final concentration of 2. Mu.g/mL. A new 96-well plate was taken and 100. Mu.L/well coated overnight at 4 ℃.
(2) The antigen coating was removed and washed 3 times with PBST (0.5% tween).
(3) 200. Mu.L/well of 3% MPBS was added and blocked for 2 hours at 37 ℃.
(4) After removal of the blocking buffer, the well plate was washed 3 times with PBST.
(5) Positive control antibody Ixekizumab was diluted to 10. Mu.g/ml with PBS, 5-fold diluted to 7 spots, added to the ELISA plate at 100. Mu.L/well, incubated for 1 hour at room temperature, and the control wells were PBS.
(6) The liquid in the wells was removed and washed 3 times with PBST.
(7) Secondary antibody HRP-protein a (Boster, BA 1080) was added at 1:10000 dilution, 100 μl/well added to the elisa plate and incubated for 1 hour at room temperature.
(8) After removing the liquid from the wells, the well plate was washed 3 times with PBST.
(9) 100. Mu.L/well TMB color development solution was added.
(10) Incubate for 15 min at room temperature in the dark.
(11) 50. Mu.L/Kong Zhongzhi of liquid (2M HCl) was added.
(12) OD450 values within wells were read using a microplate reader.
The results of the binding capacity of the IL-17A recombinant protein and the positive antibodies are shown in the following table, and the positive antibodies can be well bound with the IL-17A antigen protein and can be used for immunization.
Table 1.
4. Human IL-17A recombinant protein activity detection
The experimental steps are as follows:
(1) Resuscitating NIH-3T3 cells from liquid nitrogen, serial subculturing to allow the cells to be in logarithmic growth phase, counting cells, and then culturing according to 2×10 5 Cell mass/wells were seeded into 96-well plates.
(2) IL-17A (ACRO, cat#ILA-H5118) and the prepared IL-17A recombinant protein (ACRO protein as a control) were added at different concentrations per well at a final concentration of 0. Mu.g/ml, 0.00001. Mu.g/ml, 0.0001. Mu.g/ml, 0.001. Mu.g/ml, 0.01. Mu.g/ml, 0.1. Mu.g/ml, 1. Mu.g/ml, 10. Mu.g/ml, respectively.
(3)37℃,5% CO 2 After culturing for 48 hours, the 96-well plate was gently removed, and the culture supernatant was collected by centrifugation, and IL-6 secretion was detected using a mouse IL-6ELISA kit.
(4) Data were processed using PRISM GraphPad, plotted, and EC50 values calculated.
Experimental results:
the results are shown in FIGS. 2-3, and according to the results, the IL-17A antigen protein has the activity of activating NIH-3T3 cells to express mIL-6, and the IL-17A (TEST, hereinafter referred to as IL-17A recombinant protein) activity is positively correlated with the concentration thereof, and can be used for immunization.
5. Construction of IL-17A reporter cell lines
The experimental steps are as follows:
based on the amino acid sequence information of IL-17RA (UniProtKB: Q96F46, SEQ ID NO: 17) and IL-17RC (UniProtKB: Q8NAC3, SEQ ID NO: 18), a lentiviral expression vector is constructed and lentivirus is packaged, 293 cells are co-infected, recombinant 293 cells which simultaneously overexpress the two receptors are screened, NFKB-Luciferase (the amino acid sequence of which is shown as SEQ ID NO:19, the nucleotide sequence of which is shown as SEQ ID NO: 20) and ACT1 gene (the nucleotide sequence of which is shown as SEQ ID NO: 21) are further stably transformed, and an IL-17A reporter cell strain 293F-IL-17 RA-17 Rc-ACT1-NF κB-Luc is constructed. And adding IL-17A protein for activation, adding a positive control antibody Ixekizumab for detection of a blocking experiment, calculating an EC50 value of the blocking experiment, and establishing a candidate antibody targeting IL-17A, and evaluating cell strains in vitro pharmacodynamics.
SEQ ID NO:17:
MGAARSPPSAVPGPLLGLLLLLLGVLAPGGASLRLLDHRALVCSQPGLNCTVKNSTCLDDSWIHPRNLTPSSPKDLQIQLHFAHTQQGDLFPVAHIEWTLQTDASILYLEGAELSVLQLNTNERLCVRFEFLSKLRHHHRRWRFTFSHFVVDPDQEYEVTVHHLPKPIPDGDPNHQSKNFLVPDCEHARMKVTTPCMSSGSLWDPNITVETLEAHQLRVSFTLWNESTHYQILLTSFPHMENHSCFEHMHHIPAPRPEEFHQRSNVTLTLRNLKGCCRHQVQIQPFFSSCLNDCLRHSATVSCPEMPDTPEPIPDYMPLWVYWFITGISILLVGSVILLIVCMTWRLAGPGSEKYSDDTKYTDGLPAADLIPPPLKPRKVWIIYSADHPLYVDVVLKFAQFLLTACGTEVALDLLEEQAISEAGVMTWVGRQKQEMVESNSKIIVLCSRGTRAKWQALLGRGAPVRLRCDHGKPVGDLFTAAMNMILPDFKRPACFGTYVVCYFSEVSCDGDVPDLFGAAPRYPLMDRFEEVYFRIQDLEMFQPGRMHRVGELSGDNYLRSPGGRQLRAALDRFRDWQVRCPDWFECENLYSADDQDAPSLDEEVFEEPLLPPGTGIVKRAPLVREPGSQACLAIDPLVGEEGGAAVAKLEPHLQPRGQPAPQPLHTLVLAAEEGALVAAVEPGPLADGAAVRLALAGEGEACPLLGSPGAGRNSVLFLPVDPEDSPLGSSTPMASPDLLPEDVREHLEGLMLSLFEQSLSCQAQGGCSRPAMVLTDPHTPYEEEQRQSVQSDQGYISRSSPQPPEGLTEMEEEEEEEQDPGKPALPLSPEDLESLRSLQRQLLFRQLQKNSGWDTMGSESEGPSA。
SEQ ID NO:18:
MPVPWFLLSLALGRSPVVLSLERLVGPQDATHCSPVSLEPWGDEERLRVQFLAQQSLSLAPVTAATARTALSGLSGADGRREERGRGKSWVCLSLGGSGNTEPQKKGLSCRLWDSDILCLPGDIVPAPGPVLAPTHLQTELVLRCQKETDCDLCLRVAVHLAVHGHWEEPEDEEKFGGAADSGVEEPRNASLQAQVVLSFQAYPTARCVLLEVQVPAALVQFGQSVGSVVYDCFEAALGSEVRIWSYTQPRYEKELNHTQQLPDCRGLEVWNSIPSCWALPWLNVSADGDNVHLVLNVSEEQHFGLSLYWNQVQGPPKPRWHKNLTGPQIITLNHTDLVPCLCIQVWPLEPDSVRTNICPFREDPRAHQNLWQAARLQLLTLQSWLLDAPCSLPAEAALCWRAPGGDPCQPLVPPLSWENVTVDKVLEFPLLKGHPNLCVQVNSSEKLQLQECLWADSLGPLKDDVLLLETRGPQDNRSLCALEPSGCTSLPSKASTRAARLGEYLLQDLQSGQCLQLWDDDLGALWACPMDKYIHKRWALVWLACLLFAAALSLILLLKKDHAKGWLRLLKQDVRSGAAARGRAALLLYSADDSGFERLVGALASALCQLPLRVAVDLWSRRELSAQGPVAWFHAQRRQTLQEGGVVVLLFSPGAVALCSEWLQDGVSGPGAHGPHDAFRASLSCVLPDFLQGRAPGSYVGACFDRLLHPDAVPALFRTVPVFTLPSQLPDFLGALQQPRAPRSGRLQERAEQVSRALQPALDSYFHPPGTPAPGRGVGPGAGPGAGDGT。
SEQ ID NO:19:
MEDAKNIKKGPAPFYPLEDGTAGEQLHKAMKRYALVPGTIAFTDAHIEVDITYAEYFEMSVRLAEAMKRYGLNTNHRIVVCSENSLQFFMPVLGALFIGVAVAPANDIYNERELLNSMGISQPTVVFVSKKGLQKILNVQKKLPIIQKIIIMDSKTDYQGFQSMYTFVTSHLPPGFNEYDFVPESFDRDKTIALIMNSSGSTGLPKGVALPHRTACVRFSHARDPIFGNQIIPDTAILSVVPFHHGFGMFTTLGYLICGFRVVLMYRFEEELFLRSLQDYKIQSALLVPTLFSFFAKSTLIDKYDLSNLHEIASGGAPLSKEVGEAVAKRFHLPGIRQGYGLTETTSAILITPEGDDKPGAVGKVVPFFEAKVVDLDTGKTLGVNQRGELCVRGPMIMSGYVNNPEATNALIDKDGWLHSGDIAYWDEDEHFFIVDRLKSLIKYKGYQVAPAELESILLQHPNIFDAGVAGLPDDDAGELPAAVVVLEHGKTMTEKEIVDYVASQVTTAKKLRGGVVFVDEVPKGLTGKLDARKIREILIKAKKGGKIAV。
SEQ ID NO:20:
atggaagatgccaaaaacattaagaagggcccagcgccattctacccactcgaagacgggaccgccggcgagcagctgcacaaagccatgaagcgctacgccctggtgcccggcaccatcgcctttaccgacgcacatatcgaggtggacattacctacgccgagtacttcgagatgagcgttcggctggcagaagctatgaagcgctatgggctgaatacaaaccatcggatcgtggtgtgcagcgagaatagcttgcagttcttcatgcccgtgttgggtgccctgttcatcggtgtggctgtggccccagctaacgacatctacaacgagcgcgagctgctgaacagcatgggcatcagccagcccaccgtcgtattcgtgagcaagaaagggctgcaaaagatcctcaacgtgcaaaagaagctaccgatcatacaaaagatcatcatcatggatagcaagaccgactaccagggcttccaaagcatgtacaccttcgtgacttcccatttgccacccggcttcaacgagtacgacttcgtgcccgagagcttcgaccgggacaaaaccatcgccctgatcatgaacagtagtggcagtaccggattgcccaagggcgtagccctaccgcaccgcaccgcttgtgtccgattcagtcatgcccgcgaccccatcttcggcaaccagatcatccccgacaccgctatcctcagcgtggtgccatttcaccacggcttcggcatgttcaccacgctgggctacttgatctgcggctttcgggtcgtgctcatgtaccgcttcgaggaggagctattcttgcgcagcttgcaagactataagattcaatctgccctgctggtgcccacactatttagcttcttcgctaagagcactctcatcgacaagtacgacctaagcaacttgcacgagatcgccagcggcggggcgccgctcagcaaggaggtaggtgaggccgtggccaaacgcttccacctaccaggcatccgccagggctacggcctgacagaaacaaccagcgccattctgatcacccccgaaggggacgacaagcctggcgcagtaggcaaggtggtgcccttcttcgaggctaaggtggtggacttggacaccggtaagacactgggtgtgaaccagcgcggcgagctgtgcgtccgtggccccatgatcatgagcggctacgttaacaaccccgaggctacaaacgctctcatcgacaaggacggctggctgcacagcggcgacatcgcctactgggacgaggacgagcacttcttcatcgtggaccggctgaagagcctgatcaaatacaagggctaccaggtagccccagccgaactggagagcatcctgctgcaacaccccaacatcttcgacgccggggtcgccggcctgcccgacgacgatgccggcgagctgcccgccgcagtcgtcgtgctggaacacggtaaaaccatgaccgagaaggagatcgtggactatgtggccagccaggttacaaccgccaagaagctgcgcggtggtgttgtgttcgtggacgaggtgcctaaaggactgaccggcaagttggacgcccgcaagatccgcgagattctcattaaggccaagaagggcggcaagatcgccgtg。
SEQ ID NO:21:
ATGCCACCTCAGTTGCAGGAAACTCGGATGAATAGAAGCATCCCCGTGGAAGTGGACGAGAGCGAGCCGTACCCTAGTCAGCTGCTGAAGCCGATCCCTGAGTACTCCCCGGAAGAGGAATCCGAACCACCAGCCCCCAACATTCGCAATATGGCCCCCAATAGCTTGTCCGCACCAACAATGCTGCACAACTCTTCTGGCGACTTCTCTCAGGCCCACTCCACCCTGAAACTGGCGAATCACCAGCGGCCTGTATCCCGGCAGGTGACCTGTCTGAGAACTCAGGTGCTTGAAGACTCCGAGGACTCTTTCTGTAGGCGGCATCCAGGTTTGGGCAAGGCGTTTCCGTCCGGCTGTTCCGCGGTTTCAGAGCCCGCTTCCGAAAGTGTCGTGGGCGCCCTGCCAGCCGAGCACCAGTTCTCCTTCATGGAAAAGCGGAACCAGTGGCTGGTCAGTCAGCTGAGCGCCGCGTCACCTGATACAGGTCACGATTCCGACAAGTCTGACCAGTCTCTGCCCAATGCGTCAGCCGATAGTCTCGGGGGCTCCCAGGAGATGGTGCAGAGACCACAGCCGCACAGAAACCGGGCCGGGCTTGATCTGCCCACCATTGATACAGGCTACGATTCCCAGCCCCAGGACGTCCTTGGCATTCGCCAGCTGGAAAGGCCTCTGCCCTTGACCTCCGTGTGTTACCCCCAGGACCTGCCCCGCCCTTTGAGAAGCCGGGAGTTTCCCCAGTTTGAGCCCCAACGATACCCTGCCTGTGCTCAGATGCTGCCTCCGAACCTGAGCCCACACGCTCCCTGGAACTACCACTATCACTGTCCCGGCAGCCCCGATCACCAGGTGCCTTATGGACACGACTACCCGCGGGCTGCATACCAGCAGGTCATACAGCCTGCCTTGCCGGGTCAGCCGCTGCCCGGAGCTTCTGTGCGCGGCCTGCACCCCGTTCAGAAAGTGATCCTGAACTATCCAAGCCCATGGGACCATGAAGAGAGACCAGCCCAAAGAGATTGCTCTTTTCCTGGGTTGCCTAGACACCAAGACCAGCCTCACCACCAGCCTCCCAATCGGGCAGGCGCCCCAGGCGAAAGTCTCGAGTGCCCCGCCGAACTCAGACCACAGGTCCCTCAGCCCCCTTCCCCCGCGGCAGTACCCAGACCCCCCTCTAACCCACCCGCCCGGGGAACGCTCAAGACTTCAAATCTCCCAGAAGAGCTGCGCAAAGTGTTCATAACCTACAGCATGGACACCGCTATGGAGGTGGTTAAGTTCGTCAACTTCCTGCTGGTCAATGGGTTCCAGACTGCAATCGACATTTTTGAGGATAGAATTCGGGGAATCGACATCATCAAGTGGATGGAGAGATACCTGCGGGATAAGACAGTGATGATTATCGTGGCCATTAGTCCCAAGTACAAGCAAGATGTGGAGGGCGCAGAATCACAGTTGGACGAAGACGAGCACGGACTCCATACAAAATATATCCACAGGATGATGCAGATCGAGTTCATTAAACAAGGCTCCATGAATTTCCGCTTCATACCGGTCCTGTTTCCAAACGCAAAAAAAGAGCATGTACCCACTTGGCTCCAGAATACCCATGTCTACTCCTGGCCCAAGAACAAGAAGAATATCCTGCTGCGCTTGCTCAGAGAAGAAGAGTATGTCGCCCCTCCAAGGGGGCCCCTCCCCACACTCCAAGTAGTGCCACTT。
Experimental results:
construction of IL-17A reporter cell line 293F-IL-17RA-IL-17Rc-ACT 1-NFkB-Luc, and addition of IL-17A recombinant protein for binding, FACS results found that: the constructed IL-17A receptor over-expression cell strain can be combined with IL-17A, and the positive rate is more than 90 percent (the result is shown in figure 4).
IL-17A activating cell assay: the results of activating 293F-IL-17RA-IL-17Rc-ACT 1-NFkB-Luc with IL-17A recombinant protein are shown in FIG. 5: the IL-17A recombinant protein can effectively activate luciferase expression in 293F-IL17Ra/IL17 Rc-NFkB-Luc reporter gene cell strain.
Ixekizumab blocks IL-17A functional assay: the positive control antibody Ixekizumab and the IL-17A recombinant protein are added into 293F-IL-17RA-IL-17Rc-ACT 1-NFkB-Luc cells together, and the positive control antibody Ixekizumab can inhibit the combination of IL-17A protein and a membrane receptor thereof and inhibit the signal of intracellular NFkB, and the dose effect is presented.
6. Animal immunization process
6.1 alpaca immunization
2 alpaca (2 # and 3 #) are immunized by using the prepared IL-17A recombinant antigen protein, 6 times of immunization are carried out, the immunization adjuvant is GERBU, the immunization interval is 14 days, and the last two times of immunization are Freund complete adjuvant.
The blood collection was arranged as follows:
taking 100ml of blood after five-way-free alpaca # 2, preparing a cDNA library, and constructing a yeast display library (yeast display library 1); after the sixth step, 100ml of blood was collected to prepare a cDNA library.
The alpaca # 3 was collected 100ml after six-way blood collection to prepare a cDNA library, and was mixed with the cDNA library prepared by six-way blood collection of alpaca # 2 to construct a yeast display library (yeast display library 2).
6.2 detection of immune titers
The experimental steps are as follows:
serum is separated from alpaca with different times of immunization, limiting dilution is carried out, ELISA detection is carried out on the serum and the alpaca with antigen pre-coated 96-well plates respectively, and the specific steps are as follows:
(1) Collecting 5mL of peripheral blood, placing a centrifuge tube with a blood sample collected in a 37 ℃ incubator for 1 hour; the blood samples were then transferred to 4 ℃ overnight;
(2) Placing the centrifuge tube with the collected blood sample in a centrifuge, and centrifuging at 5000rpm for 20min; the upper serum was separated, transferred to a new sterile centrifuge tube, and the immune serum was collected.
(3) The IL-17A recombinant protein was diluted to a final concentration of 1. Mu.g/mL using sterile CBS (carbonate buffer). A new 96-well ELISA plate was taken and coated overnight at 4℃with 100. Mu.L/well.
(4) The antigen coating was removed and washed 5 times with PBST (0.05% tween 20).
(5) 3% MPBS added at 200. Mu.L/well was blocked for 2 hours at 37 ℃;
(6) After removal of the blocking buffer, the well plate was washed 5 times with PBST;
(7) 100 μl of gradient diluted serum (100 μl/well) was added and incubated for 1 hour at room temperature, control wells were PBS;
(8) Remove the liquid in the wells and wash 5 times with PBST;
(9) Add 100. Mu.l HRP anti-Llama IgG (H+L) antibody (1:50000 dilution) and incubate at room temperature for 1 hour;
(10) After removing the liquid from the wells, the well plate was washed 5 times with PBST;
(11) Adding 100 mu L/hole TMB color development liquid;
(12) Incubating for 10-15 minutes at room temperature in a dark place;
(13) Adding 50 mu L/Kong Zhongzhi liquid;
(14) OD450 values within wells were read using a microplate reader.
Experimental results: the results of the detection are shown in tables 2 to 3 below. According to ELISA detection results, immune serum and IL-17A recombinant protein are well combined, and OD value is changed in gradient along with gradient dilution of immune serum.
TABLE 2 results of immunotiter assays
TABLE 3 results of immunotiter assays
7. Antibody yeast library construction process
7.1PBMC isolation and VHH antibody fragment cloning
The experimental steps are as follows:
(1) 100mL of peripheral blood anticoagulation sample was collected, and PBMC cells were isolated using lymphocyte separation fluid.
(2) The RNA is extracted and the RNA is extracted,using PrimeScript TM II 1st Strand cDNA Synthesis Kit reverse transcription was performed to prepare cDNA.
1) The following reaction mixture Mix was prepared in a 200 μl PCR tube:
table 4.
Reagent(s) Dosage of
Oligo dT Primer(50μM) 8μL
dNTP Mixture(10mM each) 8μL
Total RNA sample 20μg
RNase-free water Added to 80. Mu.L
2) After 5min of incubation at 65℃the product was cooled rapidly on ice.
3) The following reaction solutions were prepared in the above PCR tube:
table 5.
Reagent(s) Dosage of
The denatured reaction solution 80μL
5×PrimeScript II Buffer 32μL
RNase Inhibitor(40Μ/μL) 4μL
PrimeScriptⅡRTase(200Μ/μL) 8μL
RNase-free water 36μL
4) After mixing by blowing, packaging into 80. Mu.L/tube, placing in a PCR instrument at 42 deg.C for 1 hr, inactivating at 70 deg.C for 15 min, and finally placing cDNA sample on ice or storing at-20 deg.C for a long period of time.
(3) Amplification of VHH fragments
1) The first round PCR reaction system (50. Mu.L/tube) was configured:
table 6.
Composition of the components Dosage of
Upstream primer (5. Mu.M) 2μL
Downstream primer (10. Mu.M) 1μL
NuHi Power mix(2×) 25μL
cDNA template 2μL
Sterile water 20μL
2) After the PCR reaction system is configured, the PCR instrument is set according to the following procedure:
table 7.
3) Agarose electrophoresis of PCR products
The PCR product was analyzed by electrophoresis using 1% agarose, and a fragment having a molecular weight of about 750bp was isolated. The PCR products were recovered using a gel recovery kit and the concentration was determined using NanoDrop.
4) Two rounds of PCR reaction system (50. Mu.L/tube)
Table 8.
Composition of the components Dosage of
2 nd F primer 2μL
2 nd R primer 2μL
NuHi Power mix(2×) 25μL
One round of PCR recovery of the product 200ng
Sterile water Make up to 50 mu L
5) After the PCR reaction system is configured, the PCR instrument is set according to the following procedure:
table 9.
6) Agarose electrophoresis analysis of two rounds of PCR products
The PCR products were analyzed by electrophoresis using 1% agarose, and VHH fragments having a molecular weight of about 400bp were isolated. VHH PCR products were recovered using a gel recovery kit and concentration was determined using NanoDrop.
Experimental results:
peripheral blood was collected, total RNA was extracted, reverse transcribed into cDNA, and three rounds of PCR were performed using single domain antibody amplification primers, and the PCR products were detected by agarose gel electrophoresis (see FIGS. 6 to 7 for results): the first round of PCR respectively obtains about 1000bp and 750bp PCR bands, and the gel recovers 750bp fragments as templates for the second round of PCR. The second round of PCR gave a band of about 400bp as VHH fragment and the column was recovered as template for three rounds of PCR. The third round of PCR obtains a band of about 500bp, adds a homology arm, and then carries out homologous recombination into a yeast display vector pDISPLAY.
7.2 construction of Single-Domain antibody Yeast display libraries
(1) The yeast display vector pDISPLAY is linearized and the enzyme digestion system is as follows:
table 10.
1) The pDISPLAY vector was cut with SfiI, split-packed at 100. Mu.L/tube, and cut overnight at 50 ℃.
2) The pDISPLAY vector fragment was separated using a 1% agarose gel, the 5000bp vector fragment was excised for gel recovery and the concentration was determined using NanoDrop.
3) The recovered pDISPLAY enzyme-digested products were sub-packed in 200. Mu.L of each 1.5mL centrifuge tube, 1/10 volume (20. Mu.L) of 3M sodium acetate was added, 1. Mu.g/. Mu.L Glycogen (Glycogen) was blow-sucked and mixed well, 880. Mu.L of absolute ethanol was added, mixed well upside down and placed at-80 ℃.
(2) Construction of Yeast display library by electric transformation
1) Streaking the frozen yeast competent strain at-80 ℃ onto YPD solid culture medium plates, and activating at 30 ℃ for 3-5 days;
2) Inoculating single colony yeast to 50mL YPD culture medium, shaking and culturing at 30deg.C at 250rpm for 1-2 days;
3) Yeast competent strains were prepared. Mixing the linearized carrier segment and the PCR product, and then adding the mixture into a electric shock cup for electric shock; shake-culturing in a yeast competent transfection culture flask at 220rpm and 30deg.C for 1 hr;
4) Taking 20 mu L of heavy suspension, diluting with SDCAA 5000 times, sucking 100 mu L, coating an SDCAA plate, culturing for 2-3 days, calculating the storage capacity, and continuously culturing the rest bacterial liquid for 24 hours;
5) And (3) preserving bacteria: the remaining bacterial fluid was collected in a 50mL centrifuge tube, centrifuged for 5min at 3000g, the supernatant discarded, and 10mL of SDCAA was added to resuspend with 50% glycerol: resuspension = 1:1 mix, cryopreserved at-80 ℃.
8. Yeast display library panning
The experimental steps are as follows:
1) The yeast cultured in SDCAA was added to a 250mL shake flask containing 50mL SGCAA medium, and shake cultured at 30℃and 240rpm for 16h.
2) After centrifugation, the supernatant was discarded, resuspended in 1mL of 0.5% PBSA, added to a 1.5mL centrifuge tube, centrifuged for 5min at 3000g, and the supernatant discarded. The wash was repeated with 0.5% PBSA.
3) Streptavidin beads incubated with antigen were washed twice with 0.5% pbsa (5 min incubation at 4 ℃ each time) and placed on a magnetic rack for 5min, and the supernatant was discarded.
4) The yeast liquid is added into the magnetic beads combined with the antigen, and the mixture is incubated for 60 minutes at the temperature of 4 ℃ in a rotating way and placed on a magnetic rack for 15 minutes.
5) The yeast solution was discarded and the beads were left and washed three times with 0.5% PBSA (5 min incubation at 4 ℃ each).
6) Re-suspending the magnetic beads with 1mL SDCAA culture medium, sucking 0.5-5 μl of the re-suspension into 100 μLSDCAA culture medium, dividing the re-suspension into two parts, adding 500 μl of 50% glycerol ((-80deg.C) for preservation); one portion was added to a shaking tube, supplemented with 2mL SDCAA medium, 30℃at 240rpm, and cultured for 16h.
7) The bacterial solution in the shake tube was transferred to 50mL SDCAA medium (250 mL shake flask), and cultured overnight at 30℃and 240 rpm.
8) The OD600 value of the bacterial liquid is measured, a part of bacterial liquid is taken according to the OD600, centrifuged, resuspended in SGCAA, transferred to 50mL SGCAA culture medium, the final OD600 value is 1, 30 ℃, cultured overnight at 240rpm, and the rest bacterial liquid is resuspended in SDCAA with 50% glycerol=1:1, frozen at-80 ℃.
9. Yeast monoclonal flow assay
After sorting, the saccharomycete liquid is coated on an SDCAA plate, monoclonal culture is selected, after induction expression is carried out for 48 hours, the monoclonal antibody is incubated with Biotin-antigen, and after incubation is completed, flow detection is carried out by using PE-strepitavidin for the secondary antibody. Yeast clones bound to the target antigen were lysed using 0.2% SDS (10 min incubation at 95 ℃), centrifuged, and 0.5. Mu.L of the supernatant was used as a template for PCR amplification (remaining bacterial liquid-20 ℃) preservation. The flow detection result is shown in fig. 8, the yeast positive rate is 37.9% after the second magnetic separation according to the flow detection result, positive clones are remarkably enriched, the separated products are directly coated on an SDCAA plate, and single clone is selected for flow detection.
After the 2# +3# alpaca library is fully combined with the Biotin-IL-17A-His protein, 2 rounds of magnetic separation are carried out by using streptavidin magnetic beads; the sorted yeast cells were cultured, induced for expression and subjected to flow analysis. Incubation with Biotin-IL-17A-His for 1h, using PE strepitavidins for the secondary antibody, and flow-through detection after incubation was completed, the results indicated (FIG. 9): after two rounds of magnetic sorting, the ratio of Biotin-IL-17A-His combined yeast is 20.59%, positive clones are remarkably enriched, sorting products are directly coated on an SDCAA plate, and monoclonal is selected for flow detection.
10. Identification of antibody sequences
Enriching positive clones; and selecting the enriched single gram drop, performing Phage ELISA identification, and performing sequencing analysis on clones to obtain the nucleic acid and amino acid sequence information of the candidate single domain antibody. As shown in fig. 10-11, 20 monoclonals were randomly picked for sequencing analysis, with large sequence differences and good library diversity. The potential post-translational modification sites were analyzed by the In silico method against the amino acid sequence information of the CDR regions of the candidate single domain antibodies.
According to the result of the monoclonal flow detection of the yeast, positive clones combined with IL-17A-His are selected to extract genome DNA, and the antibody sequence is obtained by PCR. According to the sequencing result of the PCR product, differential clone is selected to carry out overlap PCR amplification, and the specific steps are as follows:
(1) First round PCR: amplification of CMV, VHH and FC
1) Configuration of PCR reaction System (50. Mu.L System/reaction)
Table 11.
Composition of the components Dosage of
Upstream primer (5. Mu.M) 2μL
Downstream primer (5. Mu.M) 2μL
NuHi Power mix(2×) 25μL
Stencil (fungus liquid) 1μL
Sterile water Make up to 50 mu L
2) The PCR reaction procedure was as follows:
95 ℃ for 10min; (95 ℃,15s;56 ℃,30s;68 ℃,60s;25 cycles); 68 ℃ for 10min.
3) mu.L of the PCR product was taken, 1/10 volume of 10×loading buffer was added, electrophoresis analysis was performed using 1% agarose, the CMV band size was about 750bp, the Fc band size was about 1400bp, and the VHH band size was about 500 bp.
4) The band of interest is excised from the gel and the PCR product purified and the concentration determined by NanoDrop (e.g., too high a concentration, which can be diluted for subsequent reactions).
(2) Second round PCR: overlap Extension PCR CMV, VHH and FC are connected
1) Configuring a PCR reaction system
Table 12.
Composition of the components Dosage of
CMV 1st product 50ng
VHH 1st product 50ng
Fc 1st products 50ng
NuHi Power mix(2×) 25μL
Sterile water Make up to 46. Mu.L
2) The PCR reaction procedure was as follows:
95 ℃ for 10min; (95 ℃,15s;60 ℃,30s;68 ℃,120s;15 cycles).
3) Adding 2. Mu.L of each of the upstream primer and the downstream primer;
the PCR reaction procedure was as follows:
95 ℃ for 10min; (95 ℃,15s;60 ℃,30s;68 ℃,120s;20 cycles); 68 ℃ for 10min.
4) The overlay PCR product was purified using a TakaRa DNA fragment recovery kit and the concentration was determined using NanoDrop, requiring at least 10. Mu.g of the PCR product. For subsequent cell transfection verification.
5) The transfection procedure is identical to the transfection of eukaryotic expression vectors.
Adding a signal peptide to the N end of VHH, adding IgG1-FC to the C end, and transiently transfecting HEK293 cells with the PCR product; ELISA detection was performed on the expressed antibody supernatants: 100uL of transfection supernatant was incubated in a 96-well plate pre-coated with IL-17A recombinant antibody, ELISA detection was performed using HRP-Protein A as the secondary antibody, and the results are shown in Table 13: 1-C11 is combined with IL-17A-His antigen to construct eukaryotic expression vector.
TABLE 13 ELISA Binding assay results for candidate clone transfected supernatants
11. Expression purification of candidate single domain antibodies
The experimental steps are as follows:
1) According to ELISA detection results of candidate antibodies, positive clones are selected, the obtained VHH antibody sequences are respectively subjected to gene synthesis, and subcloned into an expression vector pcDNA3.4-hIgG1-Fc in series with human IgG1Fc (SEQ ID NO: 10). After the vector is verified by sequencing, the Qiagen plasmid megapump kit is used for preparing the endotoxin-removing plasmid for standby.
2) And taking out the LVTransm transfection reagent and the single-chain antibody expression vector from the refrigerator, thawing at room temperature, and blowing up and down by a pipetting gun to completely mix uniformly. The PBS buffer was removed and warmed to room temperature. Taking 2mL of PBS to one hole of a 6-hole plate, respectively adding 130 mug antibody expression vector, blowing up and down by a pipette, fully and uniformly mixing, adding 400 mug LVTransm, immediately blowing up and down by the pipette, uniformly mixing, and standing for 10 minutes at room temperature.
3) The DNA/LVTransm complex was added to 30mL of 293F cells, and the mixture was thoroughly mixed with gentle shaking. The cells were exposed to 5% CO at 37 ℃ 2 After culturing for 6-8 hours at 130rpm in the incubator, 50mL of fresh 293 cell culture medium was added and the cells were returned to the incubator for continued culturing.
4) After 7 days of continuous culture, the culture supernatant was collected by centrifugation, filtered with a 0.45 μm filter membrane, and the filtrate was transferred to a sterile centrifuge tube and the antibody was purified using a Protein A column.
The procedure for purifying antibodies on Protein A column is as follows:
1) Samples containing the target antibodies were added to the EP tube and mixed by gently inverting the tube.
2) EP tubes were mixed at room temperature or incubated on a rotator, (1-4 hours or overnight) and 100mM PMSF was added to prevent protein degradation.
3) The magnetic beads were collected using a magnetic separation rack and the supernatant was discarded.
4) To the EP tube, 1mL of binding/washing buffer was added and thoroughly mixed, the beads were collected using a magnetic rack and the supernatant was discarded, and the washing step was repeated three times.
5) To the EP tube, 500. Mu.L of elution buffer was added, and resuspended rapidly with pipetting or vortexing, and then incubated at room temperature (about 25 ℃) for 5 minutes either in a tumble mixer or by manually gently tumbling the EP tube.
6) Magnetic beads were collected using a magnetic separation rack and the supernatant containing the eluted antibodies was transferred to a clean EP tube.
7) Steps 1) and 2) were repeated twice.
8) To each 500. Mu.l of eluate, 1/10 of a neutralization buffer was added to neutralize the pH in order to maintain the biological activity of the antibody and avoid inactivation of the antibody.
9) Binding/washing buffer: 1 XPBS, pH 7.0.
Elution buffer: (1) 0.1M glycine, pH 2-3 (2) 0.1M NaAc-HAc, pH 3.6.
Neutralization buffer: 1M Tris, pH 8.5.
Magnetic bead regeneration buffer: 0.1M NaOH.
The experimental results are shown in FIG. 12.
EXAMPLE 2ELISA detection of binding of recombinant antibodies to target proteins
The experimental steps are as follows:
1) The recombinant protein was diluted using sterile CBS to a final concentration of 2. Mu.g/mL. A new 96-well ELISA plate was taken and coated overnight at 4℃with 100. Mu.L/well.
2) The antigen coating was removed and washed 5 times with PBST (0.05% tween 20).
3) 3% MPBS added at 200. Mu.L/well was blocked for 2 hours at 37 ℃;
4) After removal of the blocking buffer, the well plate was washed 5 times with PBST;
5) After addition of the purified single domain antibody, the initial concentration was 10. Mu.g/mL and a 5-fold gradient was diluted 7, which indicated an EC50 value of 29.99 for single domain antibody 1-C11 and an EC50 for positive antibody (Ixekizumab)
=10.06, single domain antibody 1-C11 had better binding activity to the Human IL-17A protein (fig. 13). The EC50 of each candidate antibody is shown in table 14.
TABLE 14 candidate antibody EC50 values
Antibody numbering 1-C11 Ixekizumab
EC50 29.99 1.449
EXAMPLE 3FACS detection of IL-17A binding to reporter cell lines
The experimental process comprises the following steps:
1) Resuscitating 293F-IL-17RA-IL-17Rc-ACT 1-NFkB-Luc cell lines from liquid nitrogen, and adjusting the cell state to the logarithmic growth phase;
2) Dividing the cells into several parts, each cell number being 2×10 5 A cell;
3) Incubating IL-17A-His protein and target cells, and incubating for 1 hour at room temperature after fully mixing;
4) Centrifugation at 800Xg for 3 min at room temperature, removal of the supernatant containing the antibody, washing the cells 3 times with PBS;
5) Adding secondary antibody APC-His (1:500 dilution), fully mixing, and incubating for 30 minutes at room temperature in a dark place;
6) Centrifugation at 800Xg for 3 min at room temperature, removal of the supernatant containing the secondary antibody, washing the cells 3 times with PBS;
7) Flow assays were performed using 500 μl PBS to resuspend cells.
Example 4 Single Domain antibody blocking function assay
The experimental steps are as follows:
the antibody to be detected (positive antibody: ixekizumab) was diluted 10-fold in a 96-well plate, and the antibody was serially diluted 10-fold at a final concentration of 100. Mu.g/mL, 10. Mu.g/mL, 1. Mu.g/mL, 0.1. Mu.g/mL, 0.01. Mu.g/mL, 0.001. Mu.g/mL, 0.0001. Mu.g/mL, 0.00001. Mu.g/mL, 0.000001. Mu.g/mL, 0.000 0001. Mu.g/mL, 0. Mu.g/mL, 50. Mu.L of diluted gradient concentration antibody was added to a 96-well plate, 2 multiplex wells per gradient. Then 50. Mu.L of IL-17A protein (0.4. Mu.g/mL final concentration) was added to the corresponding wells. After mixing, the mixture was placed in an incubator at 37℃and incubated for 1 hour. 293F-IL-17RA-IL-17Rc-ACT 1-NFkB-Luc cells cultured to logarithmic growth phase were aspirated into 96-well plates, each well was inoculated with 2X 10 4 Individual cells. After 18h of co-cultivation, 20. Mu.L of Bright-GloTM assay reagent was added to each well and the luciferase activity values in the wells were measured using a Tecan M1000pro microplate reader.
Experimental results:
the results are shown in FIG. 14: ixekizumab positive control blocks Human IL-17A protein activation 293F-IL-17RA-IL-17Rc-ACT 1-NFkB-Luc. IL-17A single domain antibody 1-C11 in the antibody to be detected can block Human IL-17A protein from activating 293F-IL-17RA-IL-17Rc-ACT 1-NFkB-Luc, but the blocking effect is weaker than that of positive antibody.
Example 5 stability experiment
By detecting fluorescence change through a micro-differential scanning fluorescence (nanoDSF) technique, thermal denaturation and chemical denaturation of proteins can be detected under natural conditions, and the temperature (Tm) at which 50% of the proteins are in an unfolded state and the temperature (Tagg) at which aggregation begins to occur can be accurately determined; the higher the thermal denaturation Tm value and Tagg, the more stable the antibody protein.
The experimental steps are as follows:
taking 100 mu L of candidate antibody prepared in the earlier stage and Ixekizumab (the concentration of a sample is greater than 200 mu g/ml), centrifuging at 4 ℃ and 12000 Xg for 10min, sucking the sample by using a capillary tube, preparing two capillaries for each sample, taking the capillaries as parallel control, putting the capillaries into corresponding clamping grooves in sequence, ensuring that the capillaries are full of the sample, and carrying out detection analysis.
Experimental results:
as shown in fig. 15-16, the stability of the single domain antibodies 1-C11 is strong, tm=64.25, tagg= 81.13, tm=56.1, tagg=61.86 for positive control.
Application example 1 an antibody preparation
The antibody preparation comprises: an anti-IL-17A antibody, the amino acid sequence of which is shown in SEQ ID NO. 8; buffers, surfactants, amino acids, tonicity agents, and the like.
In one embodiment of the invention, the preparation of the antibody preparation comprises: weighing all the materials, adding water for dissolving and uniformly mixing, and regulating the concentrations of all the components to obtain the composition: (100-200) mg/ml of anti-IL-17A antibody (amino acid sequence shown as SEQ ID NO: 8), (1-10) mM citrate buffer, (0.1-1% w/v) Tween 80, (100-200) mM arginine and (1-10)% sucrose, the pH of the formulation being 5.0-8.0.
Application example 2A kit
The kit comprises: anti-IL-17A antibodies, recombinant proteins, antibody preparations and/or polyclonal antibodies, containers for loading antibody preparations, buffers, and the like.
In one embodiment of the invention, the kit comprises: (100-200) mg/ml of anti-IL-17A antibody (amino acid sequence shown as SEQ ID NO: 8), and a buffer solution with pH of 5.0-8.0.
Application example 3 antibody drug conjugate
The antibody drug conjugate comprises: anti-IL-17A antibodies, recombinant proteins, antibody preparations and/or polyclonal antibodies, the drug is a physiologically active substance (e.g., nucleic acid, etc.) and a linker (linker includes maleimide linker, val-Cit linker, SS linker, DMSS linker) connecting the antibody and the drug.
In one embodiment of the invention, the IL-17A antibody is connected with the drug through an SS joint, and a (100-200) mM aqueous solution is added and mixed uniformly at room temperature, and the joint reaction is terminated, so that the antibody drug conjugate is obtained.
Application example 4 pharmaceutical composition
The pharmaceutical composition comprises an anti-IL-17A antibody, a recombinant protein, an antibody preparation, a polyclonal antibody, a nucleic acid molecule, a biological expression vector and/or a host cell, and also comprises pharmaceutically acceptable auxiliary materials.
In one embodiment of the present invention, the preparation of the pharmaceutical composition comprises: preparing an anti-IL-17A antibody or an antigen binding fragment thereof with the concentration of (100-200) mg/ml, and adding (1-20 w/v) sucrose, (10-300) mM histidine and (0.1-10)% Tween 80 to obtain the pharmaceutical composition.
Finally, it should be noted that the above description is only for illustrating the technical solution of the present invention, and not for limiting the scope of the present invention, and that the simple modification and equivalent substitution of the technical solution of the present invention can be made by those skilled in the art without departing from the spirit and scope of the technical solution of the present invention.

Claims (31)

1. An antibody, characterized in that: the antibody comprises amino acid sequences of HCDR1, HCDR2 and HCDR 3; the amino acid sequence of the HCDR1 is shown as SEQ ID NO. 1, the amino acid sequence of the HCDR2 is shown as SEQ ID NO. 2, and the amino acid sequence of the HCDR3 is shown as SEQ ID NO. 3; and/or an amino acid sequence having at least 80% sequence similarity with SEQ ID NO. 1, SEQ ID NO. 2 and SEQ ID NO. 3, respectively.
2. The antibody of claim 1, wherein: the amino acid sequence of the antibody comprises: an amino acid sequence obtained by at least one of substitution, deletion, addition and/or modification on the amino acid sequence shown in SEQ ID NO. 1-3.
3. The antibody of claim 2, wherein: the amino acid sequence of the antibody comprises: amino acid sequences having a difference of 1, 2, 3, 4, 5, 6, 7 or 8 amino acids compared to the amino acid sequences shown in SEQ ID NO. 1-3.
4. An antibody, characterized in that: the amino acid sequence of the antibody comprises a framework region FR; the frame region FR includes: FR1 shown in SEQ ID NO. 4, FR2 shown in SEQ ID NO. 5, FR6 shown in SEQ ID NO. 8 and FR4 shown in SEQ ID NO. 7; and/or; an amino acid sequence having at least 80% sequence identity to the amino acid sequence set forth in SEQ ID NO. 4-7.
5. The antibody of claim 4, wherein: the amino acid sequence of the antibody comprises: an amino acid sequence obtained by at least one of substitution, deletion, addition and/or modification of the amino acid sequence shown in SEQ ID NO. 4-7.
6. The antibody of claim 5, wherein: the amino acid sequence of the antibody comprises: amino acid sequences having 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38 or 39 amino acid differences compared to the amino acid sequences shown in SEQ ID NOs 4-7.
7. An antibody, characterized in that: the amino acid sequence of the antibody comprises: an amino acid sequence of the antibody of any one of claims 1 to 3 and an amino acid sequence of the antibody of any one of claims 4 to 6.
8. An antibody, characterized in that: the amino acid sequence of the antibody comprises: FR1-HCDR1-FR2-HCDR2-FR3-HCDR3-FR4;
the amino acid sequences of HDR1, HDR2 and HDR3 are shown as SEQ ID NO. 1-3 or functional active variants of the amino acid sequences with amino acid differences of 1, 2, 3, 4, 5, 6, 7 or 8 compared with the amino acid sequences shown as SEQ ID NO. 1-3;
the amino acid sequences of the FR1, the FR2, the FR3 and the FR4 are shown as SEQ ID NO. 4-7 or functional active variants of the amino acid sequences with at least 80% sequence similarity compared with the amino acid sequences shown as SEQ ID NO. 4-7.
9. The antibody of claim 8, wherein: the amino acid sequence of the HCDR1 is shown as SEQ ID NO. 1, the amino acid sequence of the HCDR2 is shown as SEQ ID NO. 2, and the amino acid sequence of the HCDR3 is shown as SEQ ID NO. 3.
10. The antibody of claim 8 or 9, wherein: the amino acid sequence of the FR1 is shown as SEQ ID NO. 4, the amino acid sequence of the FR2 is shown as SEQ ID NO. 5, the amino acid sequence of the FR3 is shown as SEQ ID NO. 6, and the amino acid sequence of the FR4 is shown as SEQ ID NO. 7.
11. The antibody of claim 10, wherein: the amino acid sequence of the antibody is shown as SEQ ID NO. 8.
12. The antibody of any one of claims 1-11, wherein: the antibody is a single domain antibody.
13. The antibody of any one of claims 1-12, wherein: the antibody is an anti-IL-17A antibody.
14. The antibody of any one of claims 1-13, wherein: comprising part or all of an antibody heavy chain framework region selected from human, murine, primate or camelid origin or a variant thereof;
preferably, the antibody heavy chain framework regions or variants thereof comprise part or all of a source selected from camelids;
more preferably, the heavy chain framework regions of the antibodies or variants thereof are comprised in part or in whole selected from alpaca sources.
15. A polyclonal antibody, characterized in that: comprising the antibody of any one of claims 1-14.
16. An antibody preparation, characterized in that: the antibody preparation comprising the antibody of any one of claims 1-14 and a pharmaceutically acceptable carrier.
17. A recombinant protein, characterized in that: comprising the antibody of any one of claims 1-14.
18. The recombinant protein according to claim 17, wherein: also included are biologically active proteins or functional fragments thereof that assist in their expression and/or secretion, or that extend their half-life in vivo;
preferably, the biologically active protein or functional fragment thereof is selected from at least one of His tag, GST tag, MBP tag, FLAG tag and SUMO tag, immunoglobulin Fc domain, serum albumin, albumin binding polypeptide, prealbumin, carboxy terminal peptide, elastin-like polypeptide.
19. The recombinant protein according to claim 18, wherein: the biologically active protein or functional fragment thereof is a human immunoglobulin Fc domain, preferably a human IgG Fc domain, such as a human IgG1 Fc domain, an IgG2 Fc domain, an IgG3 Fc domain, an IgG4 Fc domain, more preferably a human IgG1 Fc domain.
20. A kit, characterized in that: the kit comprises:
(1) The antibody of any one of claims 1-14, the polyclonal antibody of claim 15, the antibody preparation of claim 16, or the recombinant protein of any one of claims 17-19; and;
(2) A container for holding the antibody preparation.
21. An antibody drug conjugate, characterized in that: the antibody drug conjugate comprises:
(1) The antibody of any one of claims 1-14, the polyclonal antibody of claim 15, the antibody preparation of claim 16, or the recombinant protein of any one of claims 17-19; and;
(2) A coupling moiety that binds to (1).
22. A nucleic acid molecule characterized in that: the nucleic acid molecule encodes the antibody of any one of claims 1-14, the polyclonal antibody of claim 15, or the recombinant protein of any one of claims 17-19.
23. A biological expression vector, characterized in that: the biological expression vector comprises the nucleic acid molecule of claim 22.
24. A host cell, characterized in that: the host cell genome having integrated therein the nucleic acid molecule of claim 22; or; comprising the biological expression vector of claim 23.
25. The use of the antibody of any one of claims 1-14, the polyclonal antibody of claim 15, the antibody preparation of claim 16, the recombinant protein of any one of claims 17-19, the kit of claim 20, the antibody drug conjugate of claim 21, the nucleic acid molecule of claim 22, the biological expression vector of claim 23, or the host cell of claim 24, characterized in that: the use is selected from at least one of the following:
(1) Preparing a detection reagent or a kit;
(2) Preparing a medicament for preventing and/or treating autoimmune diseases;
(3) Preparing medicine for preventing and/or treating cancer.
26. Use according to claim 25, characterized in that: the autoimmune diseases include psoriasis, systemic lupus erythematosus, malignant rheumatoid arthritis, adult stell disease, basse Du Bing, scleroderma, hashimoto disease, allergic vasculitis, juvenile idiopathic arthritis, temporal arteritis, myasthenia gravis, rheumatoid vasculitis, autoimmune optic neuropathy, polymyalgia rheumatica, polymyositis, multiple sclerosis, fibromyalgia, systemic scleroderma, dermatomyositis, acute glomerulonephritis, behcet's disease, progressive systemic sclerosis, sjogren's syndrome, white spot, malignant anemia, vasculitis, macrovasculitis, bullous pemphigoid, adje's disease, kaschmann's disease, giant cell arteritis, idiopathic Adje's disease, psoriasis arthritis, multifocal motor neuropathy, chronic inflammatory demyelinating polyneuropathy, chronic inflammatory demyelinating disease perinodular arteritis, amyotrophic lateral sclerosis, idiopathic azoospermia, herpes gestation, aortic inflammatory syndrome, autoimmune hemolytic anemia, eosinophilic fasciitis, pemphigoid, spondyloarthritis, leukoplakia, primary biliary cirrhosis, mixed connective tissue disease, chronic atrophic gastritis, antiphospholipid antibody syndrome, allergic granulomatous vasculitis, good-pasture syndrome, cogan syndrome, ANCA-related vasculitis, linear IgA bullous dermatoses, RS3PE syndrome, chronic discoid lupus erythematosus, igG 4-related diseases, guillain Barre syndrome, autoimmune hepatitis, acquired bullous epidermolysis, non-alcoholic steatohepatitis, lupus nephritis, type I diabetes, megaloblastic anemia, autoimmune neutropenia, idiopathic thrombocytopenic purpura, autoimmune diabetes mellitus, autoimmune hypothyroidism, primary hypothyroidism, slowly progressing type I diabetes, focal scleroderma, pemphigus, alopecia areata, neuromyelitis optica, sarcoidosis, origin-field disease, habitual abortion, inflammatory bowel disease, celiac disease, ankylosing spondylitis, severe asthma, chronic urticaria transplantation immunity, familial mediterranean fever, eosinophilic chronic sinusitis, dilated cardiomyopathy, systemic mastocytosis or inclusion body myositis.
27. Use according to claim 25 or 26, characterized in that: the cancer comprises oral cancer, lymphoma, respiratory system cancer, prostate cancer, leukemia, retinoblastoma, choriocarcinoma, peritoneal cancer, eye cancer, urinary system cancer, bone cancer, endometrial cancer, uterine cancer, bladder cancer, thyroid cancer, esophageal cancer, laryngeal cancer, head and neck cancer, acute lymphoblastic leukemia, melanoma, hairy cell leukemia, neuroblastoma, salivary gland cancer, digestive system cancer, myeloma, gastric cancer, liver cancer, skin cancer, lung cancer, rectal cancer, ovarian cancer, kidney cancer, pancreatic cancer, liver cancer, sarcoma, glioblastoma, basal cell carcinoma, cholangiocarcinoma, breast cancer, cervical cancer, cholangiocarcinoma, colorectal cancer, connective tissue cancer, rhabdomyosarcoma, squamous cell carcinoma, testicular cancer, B cell lymphoma, chronic lymphocytic leukemia, chronic myeloblastic leukemia.
28. A pharmaceutical composition characterized by: the pharmaceutical composition comprises: the antibody of any one of claims 1-14, the polyclonal antibody of claim 15, the antibody preparation of claim 16, the recombinant protein of any one of claims 17-19, the antibody drug conjugate of claim 21, the nucleic acid molecule of claim 22, the biological expression vector of claim 23, or the host cell of claim 24.
29. A method for in vitro detection of IL-17A in a sample for non-diagnostic purposes, characterized in that: the method comprises the following steps:
(1) Binding the antibody of any one of claims 1-14, the polyclonal antibody of claim 15, the antibody preparation of claim 16, the recombinant protein of any one of claims 17-19, the antibody drug conjugate of claim 21 to a test sample;
(2) The antigen-antibody complex is detected and the results are interpreted.
30. A method for preventing and/or treating autoimmune diseases, characterized in that: the method comprises the following steps: administering to a subject a therapeutically effective amount of the antibody of any one of claims 1-14, the polyclonal antibody of claim 15, the antibody preparation of claim 16, the recombinant protein of any one of claims 17-19, the antibody drug conjugate of claim 21, the nucleic acid molecule of claim 22, the biological expression vector of claim 23, the host cell of claim 24, or the pharmaceutical composition of claim 28.
31. A method for preventing and/or treating cancer, characterized by: the method comprises the following steps:
Administering to a subject a therapeutically effective amount of the antibody of any one of claims 1-14, the polyclonal antibody of claim 15, the antibody preparation of claim 16, the recombinant protein of any one of claims 17-19, the antibody drug conjugate of claim 21, the nucleic acid molecule of claim 22, the biological expression vector of claim 23, the host cell of claim 24, or the pharmaceutical composition of claim 28.
CN202311774070.8A 2023-12-21 2023-12-21 Antibody molecule, nucleic acid, pharmaceutical use and method for treating inflammatory disease Pending CN117843776A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311774070.8A CN117843776A (en) 2023-12-21 2023-12-21 Antibody molecule, nucleic acid, pharmaceutical use and method for treating inflammatory disease

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311774070.8A CN117843776A (en) 2023-12-21 2023-12-21 Antibody molecule, nucleic acid, pharmaceutical use and method for treating inflammatory disease

Publications (1)

Publication Number Publication Date
CN117843776A true CN117843776A (en) 2024-04-09

Family

ID=90532256

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311774070.8A Pending CN117843776A (en) 2023-12-21 2023-12-21 Antibody molecule, nucleic acid, pharmaceutical use and method for treating inflammatory disease

Country Status (1)

Country Link
CN (1) CN117843776A (en)

Similar Documents

Publication Publication Date Title
JP7317272B2 (en) TIGIT Antibodies, Antigen-Binding Fragments Thereof, and Medical Uses Thereof This application is based on and claims priority from Application No. CN201710908565.3 filed on September 29, 2019. The disclosure of which is incorporated herein by reference in its entirety.
JP2023075294A (en) Anti-cd47 antibody and application thereof
US10150809B2 (en) Antibodies that bind peptidoglycan recognition protein 1
TW201010725A (en) Anti-IL-12/IL-23 antibodies
JP6865826B2 (en) Antibodies targeting interleukin 17A, their production methods and applications
BR112019022702A2 (en) range of anti-interferon antibodies and their uses.
EP3744734A1 (en) Anti-4-1bb antibody, antigen-binding fragment thereof and medical use thereof
CN112646032A (en) BCMA-targeted humanized monoclonal antibodies with human monkey crossover
WO2023125888A1 (en) Gprc5d antibody and application thereof
EP4192489A2 (en) Il2rb binding molecules and methods of use
EP4257605A1 (en) Anti-tslp nanobody and use thereof
EP3683234A1 (en) Il-6r antibody and antigen binding fragment thereof and medical use
WO2022037528A1 (en) Single variable domain and antigen binding molecule binding bcma
CN117843776A (en) Antibody molecule, nucleic acid, pharmaceutical use and method for treating inflammatory disease
CN109776677B (en) Humanized anti-IL-13 antibody and preparation method and application thereof
CN117820475A (en) Novel nano antibody aiming at IL-17A, medicine, preparation method and application
CN114591432B (en) anti-TNFalpha single domain antibodies and uses thereof
CN117843779A (en) Antibodies, nucleic acids, pharmaceutical formulations and methods of treatment of inflammatory diseases
WO2024017326A1 (en) Anti-gprc5d nanobody and use thereof
CN117843803A (en) Novel tandem single domain antibodies and their use in the treatment of disease
WO2022127842A1 (en) Bispecific antibody targeting il-17a and il-36r and application thereof
CN117903304A (en) Novel antibody sequence structure and application thereof
US20220411527A1 (en) Compositions and methods for transferrin receptor 1 targeting
WO2022121941A1 (en) Anti-human msln antibody and application thereof
CN117820477A (en) Novel anti-IL-17A single domain antibody concatemer and application thereof

Legal Events

Date Code Title Description
PB01 Publication
SE01 Entry into force of request for substantive examination