AU2016346595B2 - Antibody to be cross-linked to human and mouse Sema3A, and use thereof - Google Patents

Antibody to be cross-linked to human and mouse Sema3A, and use thereof Download PDF

Info

Publication number
AU2016346595B2
AU2016346595B2 AU2016346595A AU2016346595A AU2016346595B2 AU 2016346595 B2 AU2016346595 B2 AU 2016346595B2 AU 2016346595 A AU2016346595 A AU 2016346595A AU 2016346595 A AU2016346595 A AU 2016346595A AU 2016346595 B2 AU2016346595 B2 AU 2016346595B2
Authority
AU
Australia
Prior art keywords
seq
amino acid
antibody
sema3a
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.)
Active
Application number
AU2016346595A
Other versions
AU2016346595A1 (en
Inventor
Jae Hyun Lee
Do Hyun Nam
Yong Jae Shin
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.)
Pangen Biotech Inc
Samsung Life Public Welfare Foundation
Original Assignee
Pangen Biotech Inc
Samsung Life Public Welfare Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from KR1020160123233A external-priority patent/KR101854529B1/en
Application filed by Pangen Biotech Inc, Samsung Life Public Welfare Foundation filed Critical Pangen Biotech Inc
Publication of AU2016346595A1 publication Critical patent/AU2016346595A1/en
Application granted granted Critical
Publication of AU2016346595B2 publication Critical patent/AU2016346595B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans

Abstract

The present invention provides an antibody having the ability to be cross-linked to human Sema3A and mouse Sema3A. The antibody of the present invention can be used as a therapeutic antibody for inhibiting Sema3A in various carcinomas in which Sema3A expression is high, such as glioblastoma, pancreatic cancer and liver cancer. Since Sema3A is considered to be a therapeutic target of diabetic retinopathy, autoimmune arthritis, neuropathic pain and osteoporosis, the antibody of the present invention or an antigen binding fragment thereof can be used as an agent, in addition to being used as an anticancer drug, for treating associated diseases. The antibody of the present invention inhibits the growth of cancer cells derived from various carcinomas, by using high anti-Sema3A binding and Sema3A function inhibition caused thereby, and inhibits the migration of cancer cells by inhibiting the phosphorylation of ERK among the downstream signaling materials of Sema3A, thereby being very effective in cancer prevention and treatment.

Description

[title of invention]
ANTIBODY TO BE CROSS-LINKED TO HUMAN AND MOUSE SEMA3A AND USES THEREOF [technical field]
This application claims the priority to Korean Patent Application No. 10-2015-0149272 filed on October 27, 2015 and Korean Patent Application No. 10-2016-0123233 filed on September 26, 2016 with the Korean Intellectual Property
Office, the disclosures of which are incorporated herein by reference in their entirety.
The present invention relates to antibody to be crosslinked to human and mouse Sema3A, and uses thereof.
[background of art]
Sema3A is a secretory protein that is composed of an Iglike(immunoglobulin-like) C2-type domain, a PSI domain and a Serna domain, and it has been known to induce associated signaling by binding to NRP1 and PLXNA1.
Also, it has been reported that a high level of Sema3A specific carcinoma has a high growth rate of cancer cells, increases cancer cell migration, promotes cancer metastasis, and has poor prognosis.
Currently, no anti-cancer agent that inhibits Sema3A has been reported as an anti-cancer target, thus an anti-Sema3A antibody that inhibits the associated signaling by neutralizing the Sema3A may be a new anti-cancer treatment strategy.
Antibodies that inhibit Sema3A can be used as therapeutic agents for anti-cancer therapy such as glioblastoma, pancreatic cancer and liver cancer which are highly expressed in Sema3A.
Further, Sema3A is a factor which plays an important role in the migration of tumor-associated macrophage (AM) that is involved in the growth of cancer, and it is expected that the antibodies against Sema3A would exhibit anti-tumor effects in a variety of cancers.
Sema3A is considered as a therapeutic target for diabetic retinopathy, autoimmune arthritis, neuropathic pain or osteoporosis, and it can be used as therapeutic agents in many associated diseases in addition to anti-cancer therapeutic agents .
[DETAILED DESCRIPTION OF THE invention] [Technical Problem]
The present inventors have endeavored to develop antibodies that can bind to Sema3A, i.e., a factor involved in the growth of cancer cells, and prevent and treat cancers.
As a result, the present inventors have discovered antibody that has the ability to be cross-linked to human Sema3A and mouse Sema3A, and exhibits the ability to inhibit cancer cell growth and migration, thereby having excellent effects of preventing and treating cancers, and completed the present invention.
Provided herein is an antibody against human Sema3A or an antigen binding fragment thereof.
Provided herein is a nucleic acid molecule encoding a heavy chain variable region of an antibody against the human Sema3A.
Provided herein is a nucleic acid molecule encoding a light chain variable region of an antibody against the human Sema3A.
Also provided herein is a recombinant vector composing the nucleic acid molecule.
Also provided herein is a host cell transformed with the recombinant vector.
Provided herein is a pharmaceutical composition for preventing or treating cancer.
[Technical Solution]
According to an aspect of the present invention, there is provided an antibody (clone name AO8) to human Sema3A or its antigen binding fragment comprising:
(a) a heavy chain variable region comprising the following heavy chain complementarity determining region(CDR) amino acid sequences :
CDRH1 consisting of the amino acid sequence of SEQ ID NO: 1,
CDRH2 consisting of the amino acid sequence of SEQ ID NO: 2, and
CDRH3 consisting of the amino acid sequence of SEQ ID NO: 3; and (b) a light chain variable region comprising the following light chain CDR amino acid sequences:
CDRL1 consisting of the amino acid sequence of SEQ ID NO
4,
CDRL2 consisting of the amino acid sequence of SEQ ID NO
5, and
CDRL3 consisting of the amino acid sequence of SEQ ID NO
.
According to another aspect of the present invention, there is provided an antibody (clone name CIO) to human Sema3A or its antigen binding fragment comprising:
(a) a heavy chain variable region comprising the following heavy chain CDR amino acid sequences:
CDRH1 consisting of the amino acid sequence of SEQ ID NO:
7,
CDRH2 consisting of the amino acid sequence of SEQ ID NO:
8, and
CDRH3 consisting of the amino acid sequence of SEQ ID NO:
9; and (b) a light chain variable region comprising the following light chain CDR amino acid sequences:
CDRL1 consisting of the amino acid sequence of SEQ ID NO
io,
CDRL2 consisting of the amino acid sequence of SEQ ID NO
11, and
CDRL3 consisting of the amino acid sequence of SEQ ID NO
12.
According to still another aspect of the present invention, there is provided an antibody (clone name Fll) to human Sema3A or its antigen binding fragment comprising:
(a) a heavy chain variable region comprising the following heavy chain CDR amino acid sequences:
CDRH1 consisting of the amino acid sequence of SEQ ID NO
13,
CDRH2 consisting of the amino acid sequence of SEQ ID NO
14, and
CDRH3 consisting of the amino acid sequence of SEQ ID NO
15; and (b) a light chain variable region comprising the following light chain CDR amino acid sequences:
CDRL1 consisting of the amino acid sequence of SEQ ID NO:
16,
CDRL2 consisting of the amino acid sequence of SEQ ID NO:
17, and
CDRL3 consisting of the amino acid sequence of SEQ ID NO:
18,
In another aspect provided herein is a nucleic acid molecule encoding a heavy chain variable region of an antibody comprising the amino acid sequence of SEQ ID NO: 19, SEQ ID NO:21 or SEQ ID NO:23.
In another aspect provided herein is a nucleic acid molecule encoding a light chain variable region of an antibody comprising the amino acid sequence of SEQ ID NO: 20, SEQ ID NO:22 or SEQ ID NO:24.
In a related aspect provided herein is a recombinant vector comprising:
a nucleic acid molecule encoding a heavy chain variable region of an antibody comprising the amino acid sequence of SEQ ID NO:19, SEQ ID NO:21 or SEQ ID NO:23; and a nucleic acid molecule encoding a light chain variable region of an antibody comprising the amino acid sequence of SEQ ID NQ:20, SEQ ID NO:22 or SEQ ID NO:24.
In a further aspect provided herein is a composition comprising:
(a) one of the foregoing antibodies or its binding fragment against a human Sema3A; and (b) an acceptable carrier.
In yet another aspect provided herein is a method for preventing or treating a cancer in which Sema3A is highly expressed, the method comprising administering the abovementioned composition to a subject.
In a further aspect provided herein is the use of one of the foregoing antibodies or its binding fragment against a human Sema3A for the manufacture of a medicament for treating or preventing a cancer in which Sema3A is highly expressed.
2016346595 14 Aug 2019
The present inventors have endeavored to develop antibodies that can bind to Sema3A, i.e., a factor involved in the growth of cancer cells, and prevent and treat cancers.
As a result, the present inventors have discovered antibody that has the ability to be cross-linked to human Sema3A and mouse Sema3A, and exhibits the ability to inhibit cancer cell growth and migration, thereby having excellent effects of preventing and treating cancers.
The antibody of the present invention has a specific 10 binding ability to human Sema3A. In Particular, the antibody of the present invention has cross-linking ability against human Sema3A and mouse Sema3A.
As used herein, the term antibody relating to an antibody to human Sema3A refers to a specific antibody to 15 human Sema3A which specifically binds to human Sema3A. The antibody is meant to include complete antibody forms as well as antigen binding fragments of antibody molecules.
The complete antibody includes two full-length light chains and two full-length heavy chains, and each light chain
6A is linked to the heavy chain by disulfide bond.
The heavy chain constant region includes a gamma (γ) , mu (μ) , alpha (a) , delta (δ) and epsilon (c) type, which is classified into sub-classes such as gamma 1 (γΐ), gamma2 (γ2), gamma3 (γ3), gamma4 (γ4), alphal (al) and alpha2 (a2).
The light chain constant region includes a kappa (k) and ramda (λ) type (Cellular and Molecular Immunology, Wonsiewicz, M. J., Ed., Chapter 45, pp. 41-50, W. B. Saunders Co. Philadelphia, PA(1991); Nisonoff, A., Introduction to Molecular Immunology, 2nd Ed., Chapter 4,pp. 45-65, sinauer Associates, Inc., Sunderland, MA (1984)).
As used herein, the term antigen binding fragments refers to fragments retaining an antigen binding function, and include Fab, F(ab'), F(ab')2 Fv and the like.
Among antibody fragments, Fab has one antigen binding site which is composed of one variable domain from each heavy and light chain of the antibody, one constant region of light chain and the first constant region (CHi) of heavy chain.
Fab' is different to Fab in the senses that there is a hinge region containing one or more cysteine residues at Cterminal of CHi domain of heavy chain.
F(ab')2 antibody is produced by forming a disulfide bond between cysteine residues of hinge region of Fab'.
Fv is a minimal antibody fragment including only variable region from each heavy and light chain. And recombinant technique to prepare a Fv fragment is disclosed in PCT
International Publications WO 88/10649, WO 88/106630, WO
88/07085, WO 88/07086 and WO 88/09344.
Two-chain Fv is linked by non-covalent bond between variable regions of each heavy and light chain, and singlechain Fv is generally linked by covalent bond via a peptide linker between variable regions of each heavy and light chain, or is directly linked to each other at C-terminal, forming a dimer-like structure such as two-chain Fv.
Such antibody fragments may be obtained using a proteolytic enzymes (e.g., Fabs can be obtained by restriction-cleaved of whole antibodies to papain, The F (ab ' ) fragment can be obtained by restriction-cleaved of the whole antibody to pepsin), and may be preferably prepared by genetic recombination techniques.
In one embodiment, the antibody of the present invention is a scFv form or a complete antibody form.
In addition, the heavy chain constant region may be selected from the isotypes consisting of gamma (y) , mu (μ), alpha (a), delta (δ) and epsilon (c).
As used herein, the term heavy chain refers to both a full-length heavy chain and its fragment, which includes variable domain (VH) containing the amino acid sequence with a variable region sequence for imparting a specificity to antigen and three constant domains (CHi, CH2 and CH3) .
The term light chain refers to both a full-length light chain and its fragment, which includes variable domain (VL) containing the amino acid sequence with a variable region sequence for specifically binding to antigen and constant domain (CL) .
As used herein, the term CDR (complementarity determining region) refers to an amino acid sequence of hypervariable region of immunoglobulin heavy and light chain (Rabat et al., Sequences of Proteins of Immunological Interest, 4th Ed., U.S. Department of Health and Human Services, National Institutes of Health (1987)). Three CDRs are involved in heavy chain (CDRH1, CDRH2 and CDRH3) and light chain (CDRL1, CDRL2 and CDRL3), respectively. CDR provides a main contacting residue to combine antibody with antigen or epitope.
Human Sema3A antibody or its antigen binding fragment may include variants of amino acid sequences set forth in the appended Sequence Listing, which are capable of specifically recognizing human Sema3A.
For example, amino acid sequence of antibody may be altered to improve binding ability and/or other biological characteristics of antibody. These alterations include, for example, deletion, insertion and/or substitution of amino acid residues of antibody.
Such amino acid variations may be provided on the basis of a relative similarity of amino acid side chains, e.g., hydrophobicity, hydrophilicity, charge, size and the like. By the analysis for size, shape and type of the amino acid side chains, it could be seen that all of arginine, lysine and histidine residues are those having positive charge; alanine, glycine and serine have a similar size; phenylalanine, tryptophan and tyrosine have a similar shape.
Accordingly, based on these considerable factors, arginine, lysine and histidine; alanine, glycine and serine; and phenylalanine, tryptophan and tyrosine may be considered to be biologically functional equivalents.
When introducing variation, a hydropathic index of amino acids may be considered. Based on the hydrophobicity and the charge, the hydropathic index is given to each amino acid:
Isoleucine (+4.5); Valine (+4.2); Leucine (+3.8); Phenylalanine (+2.8); Cysteine/Cystaine (+2.5); Methionine (+1.9); Alanine (+1.8); Glycine (-0.4); Threonine (-0.7); Serine (-0.8); Tryptophan (-0.9); Tyrosine (-1.3); Proline (-1.6); Histidine (-3.2); Glutamate (-3.5); Glutamine (-3.5); Aspartate (-3.5); Asparagine (-3.5); Lysine (-3.9); and Arginine (-4.5).
When imparting an interactive biological function of proteins, the hydropathic index of the amino acid is very important. It is well known to one of skill in the art that variations can possess a similar biological activity only where proteins are replaced with amino acids having similar hydropathic index.
Where variations are intended to introduce based on the hydropathic index, the substitution is preferably performed between amino acid residues having no more than +2 difference in hydropathic index values, more preferably within +1, still more preferably within +0.5.
On the other hand, it is well-known that substitutions between amino acids having similar hydrophilicity values may result in the generation of proteins having biologically eguivalent activities .
As disclosed in U.S. Patent No. 4,554,101, each amino acid residue is assigned the following hydrophilicity values:
Arginine (+3.0); Lysine (+3.0); Aspartate (+3.0 + 1); Glutamate (+3.0+1); Serine (+0.3); Asparagine (+0.2); Glutamine (+0.2); Glycine (0); Threonine (-0.4); Proline (-0.5+1); Alanine (-0.5); Histidine (-0.5); Cysteine (-1.0);
Methionine (-1.3); Valine (-1.5); Leucine (-1.8); Isoleucine (-1.8); Tyrosine (-2.3); Phenylalanine (-2.5); and Tryptophan (-3.4).
Where variations are intended to introduce based on the hydrophilicity values, the substitution is preferably performed between amino acid residues having no more than +2 difference in hydropathic index values, more preferably within +1, still more preferably within +0.5.
The amino acid exchanges in proteins that do not substantially change the activity of the molecule are well known to one skilled in the art (H. Neurath, R. L. Hill, The
Proteins, Academic Press, New York, 1979).
The most commonly occurring exchanges include exchanges between amino acid residues:
Ala/Ser, Val/Ile, Asp/Glu, Thr/Ser, Ala/Gly, Ala/Thr, Ser/Asn, Ala/Val, Ser/Gly, Thy/Phe, Ala/Pro, Lys/Arg, Asp/Asn, Leu/Ile, Leu/Val, Ala/Glu and Asp/Gly.
Considering the afore-mentioned variations having biologically equivalent activities, it would be understood that either antibody of the present invention or the nucleic acid encoding the same includes sequences that are substantially identical to the sequences set forth in the appended Sequence Listing.
The substantially identical sequences refers to those showing preferably at least 61%, more preferably at least 70%, still more preferably at least 80%, most preferably at least 90% nucleotide similarity to the sequences of the appended Sequence Listing, as measured using one of the sequence comparison algorithms known to those ordinarily skilled in the art, by which the nucleotide sequence of this invention is maximally aligned corresponding on random other nucleotide sequences .
Methods of alignment of sequences for comparison are well-known in the art. Various methods and algorithms of alignment are described in:
Smith and Waterman, Adv. Appl. Math. 2:482(1981); Needleman and Wunsch, J. Mol. Bio. 48:443(1970); Pearson and Lipman, Methods in Mol. Biol. 24: 307-31(1988); Higgins and Sharp, Gene 73:237-44(1988); Higgins and Sharp, CABIOS 5:1513(1989); Corpet et al., Nuc. Acids Res. 16:10881-90(1988); Huang et al., Comp. Appl. BioSci. 8:155-65(1992) and Pearson et al., Meth. Mol. Biol. 24:307-31(1994).
The NCBI Basic Local Alignment Search Tool (BLAST) (Altschul et al., J. Mol. Biol. 215: 403-10 (1990)) is available from several sources, including the National Center for Biological Information (NBCI, Bethesda, Md.) and on the Internet, for use in connection with the sequence analysis programs blastp, blasm, blastx, tblastn and tblastx. It can be accessed at www.ncbi.nlm.nih.qov/BLAST/. A description of how to determine sequence identity using this program is available at www.ncbi.nlm.nih.gov/BI-AST/blast help.html. In addition, sequencing of framework region (FR) and CDRs in antibody variable regions may be indicated based on the sequences of IMGT (www.imgt.org/) generally accessible in the art.
According to an embodiment of the invention, the heavy chain variable region of A08 antibody comprises the amino acid sequence of SEQ ID NO :19.
According to an embodiment of the invention, the light chain variable region of A08 antibody comprises the amino acid sequence of SEQ ID NO:20.
According to an embodiment of the invention, the heavy chain variable region of CIO antibody comprises the amino acid sequence of SEQ ID NO :21.
According to an embodiment of the invention, the light chain variable region of CIO antibody comprises the amino acid sequence of SEQ ID NO :22.
According to an embodiment of the invention, the heavy chain variable region of Fll antibody comprises the amino acid sequence of SEQ ID NO:23.
According to an embodiment of the invention, the light chain variable region of Fll antibody comprises the amino acid sequence of SEQ ID NO:24.
The antibody of the present invention includes, but not limited to, monoclonal antibody, polyclonal antibody, human antibody, humanized antibody, chimeric antibody, single-chain Fvs(scFV), single-chain antibody, Fab fragment, F(ab') fragment, disulfide-linked Fvs (sdFV) and anti-idiotype (antiid) antibody, and epitope-binding fragment thereof.
The antibody of the present invention is basically composed of heavy chain variable region (VH)-linker—light chain variable region (VL) .
In the scFv antibody of the present invention, the linker refers to an amino acid sequence having a predetermined length which artificially links the heavy chain and light chain variable regions .
The scFv antibody of the present invention may be represented by VH (SEQ ID NO: 19)-linker-VL (SEQ ID NO: 20); VH (SEQ ID NO: 21)-linker-VL (SEQ ID NO: 22); and VH (SEQ ID NO: 23)—linker—VL (SEQ ID NO: 24) .
The antibody or its antigen binding fragment of the present invention is specifically cross-linked to human Sema3A and mouse Sema3A.
Since the antibody or its antigen binding fragment of the present invention is capable of specifically binding to human Sema3A as well as mouse Sema3A, more accurate preclinical results can be confirmed in the efficacy evaluation using mouse tumor models.
In another aspect of this invention, there is provided a nucleic acid molecule encoding a heavy chain variable region of an antibody to be cross-linked to human Sema3A and mouse Sema3A comprising the amino acid sequence of SEQ ID NO:19, SEQ ID NO:21 or SEQ ID NO:23.
In another aspect of this invention, there is provided a nucleic acid molecule encoding a light chain variable region of an antibody to be cross-linked to human Sema3A and mouse Sema3A comprising the amino acid sequence of SEQ ID NO:20, SEQ ID NO:22 or SEQ ID NO:24.
As used herein, the term nucleic acid molecule collectively refers to RNA (gDNA and cDNA) and DNA molecules, and the basic nucleotides of nucleic acid molecules also include analogues with modified sugar or base as well as natural nucleotides (Scheit, Nucleotide Analogs, John Wiley, New York (1980); Uhlman and Reyman, Chemical Reviews, 90:543584 (1990)). The sequence of the present nucleic acid molecule encoding the variable region of heavy and light chain could be modified. Such modification includes addition, deletion or non-conservative or conservative substitution of nucleotide.
The nucleic acid molecule of this invention encoding a human Sema3A antibody also includes a nucleotide sequence sharing substantial homology with the above nucleotide sequence .
The substantial homology is determined by aligning the nucleotide sequence of the present invention with other random sequences as much as possible and analyzing the aligned sequence using an algorithm commonly used in the art, wherein the nucleotide sequence sharing homology is at least 80%, more preferably 90% and most preferable 95%.
In still further aspect of this invention, there is provided a recombinant vector comprising the above-described nucleic acid molecules.
As used herein, the term vector refers to a tool for expressing target gene in a host cell, including a plasmid vector; a cosmid vector; and a virus vector such as a bacteriophage vector, an adenovirus vector, a retrovirus vector and an adeno-associated virus vector.
According to a preferable embodiment, the nucleic acid molecules encoding the variable regions of light and heavy chains in the vector of the present invention are operatively linked to a promoter.
As used herein, the term operatively linked refers to functional linkage between a nucleic acid expression control sequence (for example, a promoter, signal sequence or array of transcription factor binding sites) and a second nucleic acid sequence, wherein the expression control sequence affects transcription and/or translation of the nucleic acid corresponding to the second sequence.
The vector system of the present invention may be performed by various methods known to those skilled in the art and its practical method is described in Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press (2001) , which is incorporated herein by reference .
Typically, the vector of the present invention may be constructed as cloning or expression vector.
In addition, the vector of the present invention may be constructed using a prokaryotic or eukaryotic cell as a host cell.
For instance, in each a vector of the present invention and an eukaryotic cell used as the expression vector and the host cell, the promoter derived from genome of mammalian cell (example: methallothionein promoter, β-actin promoter, human hemoglobin promoter and human muscle creatine promoter) or mammalian virus (example: adenovirus late promoter, vaccinia virus 7.5K promoter, SV40 promoter, cytomegalovirus promoter, tk promoter of HSV, mouse mammary tumor virus (MMTV) promoter, LTR promoter of HIV, promoter of moloney virus, Epstein barr virus (EBV) and Rous sarcoma virus (RSV) ) might be used, and polyadenylated sequence might be commonly used as the transcription termination sequence.
The vector of the present invention could be fused with other sequences to facilitate the purification of an antibody expressed from it.
For example, a fused sequence includes glutathione-Stransferase (Pharmacia, USA), maltose-binding protein (NEB, USA) , FLAG (IBI, USA) and 6 X His (hexahistidine; Quiagen, USA) and so on.
Since the protein expressed in the vector of the present invention is antibody, expressed antibody could be also purified throughout protein A column in an easy manner without additive sequences for purification.
On the other hand, the expression vector of the present invention includes an antibiotics-resistance gene known to those ordinarily skilled in the art as a selection marker, for example, resistant genes against ampicillin, gentamycin, carbenicillin, chloramphenicol, streptomycin, kanamycin, geneticin, neomycin and tetracycline.
In still another aspect of this invention, there is provided a host cell transformed with the above-described recombinant vector.
The host cells in which the present vector is stably and successively cloned and expressed, also utilize any one known to those skilled in the art, for example, the suitable eukaryotic host cell of the above vector includes COS7(monkey kidney cell), NSO cell, SP2/0, CHO (Chinese hamster ovary) cell, W138, BHK (baby hamster kidney) cell, MDCK, myeloma cell line, HuT 78 cell and 293 cell, but not limited thereto.
In another aspect of this invention, there is provided a pharmaceutical composition for preventing or treating cancer comprising:
(a) a pharmaceutically effective amount of an antibody or its binding fragment against a human Sema3A; and (b) a pharmaceutically acceptable carrier.
A pharmaceutical composition of the present invention uses, as an active ingredient, the antibody to human Sema3A or its antigen binding fragment of the present invention. Therefore, the overlapping descriptions therebetween are omitted to avoid excessive complication of the specification due to repetitive descriptions thereof.
As can be verified by the following examples, the antibody to human Sema3A of the present invention inhibits the growth of cancer cells derived from various cancers by a considerable binding ability to anti-Sema3A and the suppression of Sema3A function therefrom, inhibits the ERK phosphorylation of downstream signaling molecules of Sema3A and thus suppress the migration of cancer cells, thereby being very efficient in the prevention and treatment of cancers.
The cancers that can be prevented or treated by the composition of the invention may include various cancers known in the art, and examples thereof may include breast cancer, colon cancer, lung cancer, stomach cancer, liver cancer, blood cancer, bone cancer, pancreatic cancer, skin cancer, brain cancer, cervical cancer, nasopharyngeal cancer, laryngeal cancer, colon cancer, ovarian cancer, rectal cancer, colorectal cancer, vaginal cancer, small intestine cancer, endocrine cancer, thyroid cancer, parathyroid cancer, ureter cancer, urinary tract cancer, prostate cancer, bronchial cancer, bladder cancer, kidney cancer and marrow cancer.
Specifically, the cancers that can be prevented or treated by the composition of the present invention are Sema3A expressing cancers.
In the pharmaceutical compositions of the present invention, the pharmaceutically acceptable carrier may be conventional one for formulation, including lactose, dextrose, sucrose, sorbitol, mannitol, starch, Acacia gum, potassium phosphate, alginate, gelatin, potassium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrups, methyl cellulose, methylhydroxy benzoate, propylhydroxy benzoate, talc, magnesium stearate and mineral oils, but not limited thereto.
The pharmaceutical composition according to the present invention may further include a lubricant, a humectant, a sweetener, a flavoring agent, an emulsifier, a suspending agent and a preservative.
Details of suitable pharmaceutically acceptable carriers and formulations can be found in Remington's Pharmaceutical Sciences (19th ed., 1995), which is incorporated herein by reference.
The pharmaceutical composition according to the present invention may be administered parenterally. Such parenteral administration includes, for example, intravenous injection, subcutaneous injection, intramuscular injection, intraperitoneal injection or the like.
A suitable dose of the pharmaceutical composition of the present invention may vary depending on various factors such as methods of formulating, methods of administrating, the patient's ages, weights, sex, severities of diseases, diet, administration times, administration routes, excretion rates and reaction sensitivities, and a skilled physician may determine and prescribe pharmaceutically effective dose for the required treatment or prophylaxis easily. In a preferred embodiment, proper daily dose may be 0.0001-100 mg/kg (weight) .
As used herein, the term pharmaceutically effective amount refers to an amount suitable for preventing or treating cancers.
According to the conventional techniques known to those skilled in the art, the pharmaceutical composition may be formulated with pharmaceutically acceptable carrier and/or vehicle as described above, finally providing several forms including a unit dose form and a multiple dose form.
The formulation may be a solution, a suspension or an emulsion in oily or aqueous media or may be extracts, powders, granules, tablets or capsules, and may further comprise a dispersion agent or a stabilizer.
[ADVANTAGEOUS EFFECTS]
The features and advantages of one or more embodiments of the present invention are summarized as follows:
(a) The present invention provides an antibody having the ability to be cross-linked to human Sema3A and mouse Sema3A.
(b) The antibody of the present invention can be used as therapeutic antibody drugs for inhibiting Sema3A in various cancers such as glioblastoma, pancreatic cancer and liver cancer that exhibit high Sema3A expression levels.
(c) Sema3A is considered to be a therapeutic target of diabetic retinopathy, autoimmune arthritis, neuropathic pain and osteoporosis, the antibody of the present invention or an antigen binding fragment thereof can be used as an agent for treating associated diseases in addition to being used as an anti-cancer drug.
(d) The antibody of the present invention inhibits the growth of cancer cells derived from various cancers by using high Sema3A binding and Sema3A function inhibition caused thereby, and inhibits the migration of cancer cells by inhibiting the phosphorylation of ERK among the downstream signaling materials of Sema3A, thereby being very effective in cancer prevention and treatment.
Throughout this specification the word comprise, or variations such as comprises or comprising, will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.
Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present disclosure as it existed before the priority date of each of the appended claims.
[BRIEF DESCRIPTION OF drawings]
FIG. 1 is a schematic scheme of a phage display selection process for identifying anti-Sema3A scFv antibody fragments.
FIG. 2 is a graph showing the phage display panning results .
FIG. 3 shows results analyzing binding abilities of 52 species of scFv antibody fragments cross-linked to human
Sema3A.
FIG. 4 shows results re-verifying binding abilities of 52 species of Sema3A scFv.
2016346595 14 Aug 2019
23A
FIG. 5 shows results confirming the sequences of 31 species of Sema3A scFv.
FIG. 6 is a diagram of phagemid vector for the production of scFv antibody fragments.
FIG. 7 shows Coomassie staining results of three species of purified Sema3A scFv antibody fragments.
FIG. 8 is a graph showing Indirect ELISA results according to concentrations with respect to three species of anti-Sema3A scFv antibody fragment.
FIG. 9 shows results confirming Sema3A-secreting cells using Sandwich ELISA.
FIG. 10 shows results confirming the abilities to inhibit cell growth using anti-Sema3A.
FIG. 11 shows results confirming the abilities to inhibit cell migration using anti-Sema3A scFv and U87-MG cells.
FIG. 12 shows results confirming the abilities to inhibit cell migration using anti-Sema3A scFv and 131 cells.
FIG. 13 shows results confirming the abilities to inhibit cell migration using anti-Sema3A scFv and 83 cells.
FIG. 14 shows results confirming the purity of antiSema3A IgG by HPLC analysis.
FIG. 15 shows results confirming the size of anti-Sema3A IgG by Coomassie staining.
FIG. 16 shows results confirming binding abilities of antibodies to human and mouse Sema3A.
FIG. 17 shows the results of SPR analysis on binding abilities of three species of anti-Sema3A IgG to human and mouse Sema3A.
FIG. 18 shows the results confirming the abilities to inhibit cell migration using anti-Sema3A IgG and U87-MG cells.
FIG. 19 shows the results confirming the abilties to inhibit cell migration using anti-Sema3A IgG and 131 cells.
FIG. 20 shows the results confirming the abilities to inhibit cell migration using anti-Sema3A IgG and 83 cells.
FIG. 21 shows the results confirming the efficacy of anti-Sema3A antibody that inhibits ERK phosphorylation.
FIG. 22 shows the results confirming the abilities to promote the growth of glioblastoma cells in Sema3A IgG.
FIG. 23 shows the measurement results on the degree of inhibition of cell proliferation according to concentrations of anti-Sema3A IgG.
FIG. 24 shows the results confirming reductions in tumor size by anti-Sema3A IgG in animal models.
FIG. 25 shows the measurement results on tumor weight changes by anti-Sema3A IgG in animal models.
FIG. 26 shows the measurement results on body weight changes according to the administration of anti-Sema3A IgG in animal models.
FIG. 27 shows the results confirming apoptotic effects by subjecting immunofluorescence staining after administration of anti-Sema3A IgG in animal models.
FIG. 28 shows the results confirming TAM distribution after administration of anti-Sema3A IgG in animal models.
[DETAILED DESCRIPTION OF THE embodiments]
Hereinafter, the present invention will be described in detail with reference to examples. These examples are only for illustrating the present invention more specifically, and it will be apparent to those skilled in the art that the scope of the present invention is not limited by these examples.
Example 1: Panning using recombinant human Sema3A protein
The scFv antibody fragments cross-linked to human Sema3A were identified through phage display screening by using the existing constructed synthetic scFv phage library (Yang et al., Mol. Cells. 27:225-235, 2009). The phage display screening procedure was shown in FIG. 1.
Specifically, for the collection of phagemid vectors in a phage type, which were introduced into E. coli host ER2537, four sub-library samples were respectively cultured in 400 ml of media (SB/ampicillin/2% glucose) for 2 hours. When OD600 is 0.5-0.7, the host cells were centrifuged at 5,000 g for 20 minutes to remove the supernatant, and then suspended in 400 ml of secondary media (SB/ampicillin). Then, 1012 pfu (plaque forming unit) of helper phage (VCSM13) was added thereto and cultured for 1 hour.
After that, the antibiotic Kanamycin was added at a concentration of 70 pg/ml, followed by culturing overnight at 30°C, so that the phage library was secreted outside the host cell. Then, the centrifuged culture was treated with the polyethylene glycol (PEG) solution to precipitate only phage form, thereby collecting the phage library.
Using the phage library thus obtained, phage display screening was performed through repeated rounds of panning. The counted sub-libraries were collected to 2.5*1012 pfu, and then treated with the immunotube coated with rhSema3A-Fc protein diluted to 10 pg/ml in TBS for 1 hour. The immunotube and the phage particles before the treatment were treated with a blocking solution containing 3% skim milk for 1 hour, thereby preventing non-specific binding thereof. The immunotube was washed with TBST(0.1% Tween 20) solution and 100 mM TEA was added thereto and kept to stand for 10 minutes, thereby collecting phages bound to Sema3A. For the confirmation of the number of collected phages (output), after infecting the host cells, phage counting was performed in the medium. The remaining collected solution was centrifuged at 3,000 rpm for 15 minutes and then the settled ER2537 was mixed in 500 μΐ of a culture medium (SB). The mixture was plated on the 15 cm-media and then cultured, and then 5 ml of SB medium (50% glycerol) was added, followed by collection and storage (-80°C) of colonies.
For the repeated rounds of panning, 50 μΐ of aliquot was taken from the stored phage solution from the previous round of panning, and subjected to phage particle amplification.
The phage particles which were cultured in the host cell ER2537, added with helper phages, and collected, were separated by PEG precipitation, and the next round of panning using the phage particles were progressed in the same manner.
As the number of panning was increased, it was confirmed that percentages of the phage particles after panning were increased as compared to those before panning. This means that the phage particles specific to Sema3A through the panning were amplified. The results were shown in FIG. 2.
Example 2: ELISA and Sequence Analysis for selection of Anti-Sema3A scFv Candidates
The phage particles collected from the fourth round of panning were confirmed as colonies in the medium through infection of host cell ER2537. These colonies were taken and inoculated in a 96 well plate containing 200 μΐ of SB/ampicillin media and then cultured at 37°C for 2-3 hours.
After that, for the induction of scFv-pIII protein expression, the final concentration of 1 mM of IPTG (isopropyl β-D-l-thiogalactopyranoside) was added to each well, followed by culturing overnight at 30°C. The cultured plate was centrifuged at 3,000 rpm for 15 minutes to remove the supernatant.
Then, for the collection of phage particles in the periplasm of the cultured cells, the culture plate was added with 40 μΐ of TES solution (20% w/v sucrose, 50 mM Tris, 1 mM EDTA, pH 8.0) in each well and then kept to stand at 4°C for 30 minutes, so that the cells were lysed.
After that, the cells were treated with 60 μΐ of 0.2* TES solution, and then kept to stand for at 4°C for 30 minutes.
After lysing the cells under osmotic pressure, the plate was centrifuged at 3,000 rpm for 15 minutes, thereby obtaining scFv-pIII protein of the supernatant.
μΐ of the supernatant thus obtained was added to each well of a 96 well plate coated with Sema3A protein, which was previously prepared, followed by binding at room temperature for 1 hour, and subseguently, washing procedures were performed for six times using TBST and distilled water.
Then, anti-HA antibody bound to HRP capable of binding to HA-tag in scFv pIH was added, followed by binding at room temperature for 1 hour, and subsequently, washing procedures were performed for six times using TBST (0.1% Tween20) and distilled water.
After induction of a color reaction using TMB solution, the color reaction was stopped with addition of H2SO4 solution and the values thereof were measured at O.D. 450 nm. A total of 86 clones were analyzed, and 52 clones (binding affinity >2-fold) out of them showed a higher binding affinity to
Sema3A (FIG. 3).
As a control group, the BSA solution was used, and among 52 clones, 31 clones having high binding affinities reverified through ELISA were selected (FIG. 4).
After that, phagemid was collected from the 31 clones, and DNA sequence analysis thereof was performed. A total of 5 clones having different sequences were selected.
It was verified that three (3) A08 clones, twenty-one (21) Fll clones and two (2) CIO clones had identical DNA sequences, and additionally, A10 and E10 clones had different DNA sequences (FIG. 5).
In the order of increasing the number of clones with identical sequences, Fll, A08 and CIO were selected as the final Seman3A scFv candidates.
Example 3: Production of Anti-Sema3A scFv Protein and Verification on Binding Affinity to Sema3A
The basic structure of phagemid can be confirmed in FIG.
6, and in the case of the host cell ER2537 used in the above procedures, since it suppresses the transcription stop codon (amber codon (UAG)) located in front of the phage pIH, the expression of scFv alone is not possible therein.
Accordingly, by using the expression strain (TOP10F'), which is a non-suppressor strain, the phagemid was transduced into the expression strain. After that, the expression strains into which respective phagemids were introduced without mutation were confirmed through DNA sequencing. A colony was taken from the expression strains, and inoculated in 3 ml of LB/ampicillin media, followed by culturing overnight at 37°C.
After the culturing overnight, 3 ml of the culture solution was transferred to 400 ml of media (SB/ampicillin), and then further cultured until Q.D600 reached 0.5-0.7. 1 mM IPTG as final concentration was added, followed by culturing overnight at 30°C. After the culture solution was centrifuged, the expression hosts were lysed in 40 ml of TES solution and then added with 60 ml of 0.2X TES, thereby collecting the phage particles in the periplasm. The collected supernatant was filtered through an 0.45 pm filter.
For His-tag purification, the scFv proteins present in the filtered solution were added with 1 ml of Ni-NTA beads (Qiagen) and allowed to bind thereto at room temperature for 1 hour, and then Ni-NTA beads were packed in the gravity column (Bio-rad) , followed by Collecting scFv proteins using 200 mM imidazole solution.
Through SDS-PAGE and Coomassie blue staining after expression and purification of each clone, each scFv was verified to have a size of about 28 kDa, and the results were
2016346595 26 Oct 2016 shown in FIG. 7.
The DNA sequences of each clone in the form of purified scFv are set forth in Table 1 and Table 2 below.
[Table 1]
Sequence information on heavy chain FR and CDR regions of three anti-Sema3A scFv antibody fragments
Heavy chain FR1 CDR1 FR2 CDR2 FR3 CDR3 FR4
AO 8 EVQLLESGGGL VQPGGSLRLSC AAS GFTFS DYA MSWVRQAPG KGLEWVSG IYYDDS SQ YYADSVKGRFTISRDNS KNTLYLQMNSLRAEDTA VYYC AKNLG RFDY WGQGTL VTVSS
CIO EVQLLESGGGL VQPGGSLRLSC AAS GFTFS DYA MSWVRQAPG KGLEWVSG IYYDDS SQ YYADSVEGRFTISRDNS KNTLYLQMNSLRAEDTA VYYC ARYLG LFDY WGQGTL VTVSS
Fll EVQLLESGGGL VQTGGSLRLSC AAS GFTFS DYA MSWVRQAPG KGLEWVSW IYYDSG SK YYADSVKGRFTISRDNS KNTLYLQMNSLRAEDTA VYYC AKLNG DFDY WGQGTL VTVSS
[Table 2]
Sequence information on light chain FR and CDR regions of three anti-Sema3A scFv antibody fragments
Light chain FR1 CDR1 FR2 CDR2 FR3 CDR3 FR4
AO 8 QSVLTQPPSASGTPG QRVTISCTGS SSNIGS NA VTWYQQLPGT APKLLIY DDN HRPSGVPDRFSGSKSGTS ASLAISGLRSEDEADYYC GAWDDSL SAYV FGGGTK LTVL
CIO QSVLTQPPSASGTPG QRVTISCSGS SSNIGN NS VNWYQQLPGT APKLLIY SDS QRPSGVPDRFSGSKSGTS ASLAISGLRSEDEADYYC GSWDYSL SAYV FGGGTK LTVL
Fll QSVLTQPPSASGTPG QRVTISCSGS SSNIGN ND VSWYQQLPGT APKLLIY ADS HRPSGVPDRFSGSKSGTS ASLAISGLRSEDEADYYC GAWDSSL SGYV FGGGTK LTVL
To determine the binding affinity to human Sema3A according to concentrations of the respective antibody protein fragments, in each 96 well coated with 200 ng of Sema3A and BSA, each scFv was treated with concentrations of 2,000 ng/ml, 1,000 ng/ml, 500 ng/ml, 250 ng/ml, 125 ng/ml, 62.5 ng/ml, 31.25 ng/ml and 15.62 ng/ml to analyze changes in the OD values .
In the case of A08, C10 and Fll scFv, it can be confirmed by changes in the OD values that the size of the binding affinities of scFv to Sema3A increases as the concentration increases, compared to BSA, and this can be confirmed in FIG. 8.
Example 4: Verification on Abilities of Anti-Sema3A scFv to inhibit cell growth and cell migration
The binding affinities to Sema3A proteins were verified by ELISA, and then the cell proliferation assay and the cell migration assay were used in order to verify the anti-cancer abilities to Sema3A, which is substantially secreted by cells.
First, the secretion of Sema3A was verified by Sandwich ELISA. As a result, it was verified that, among the patientderived cells, 559 secreted less Sema3A, whereas 131 and 83 hyper-secreted Sema3A. The media and NPC were used as Negative controls and U87-MG Cells were used as a positive control (FIG. 9).
In order to perform the cell proliferation assay, 5 * 103 cells of 559 and 131 were treated with 50 pg/ml of anti-Sema3A scFv. The cell growth rate was measured using the EZ-Cytox cell viability assay kit (Daeil Lab. Service) on day 4 after the treatment.
559 cells secreting less Sema3A showed no change in the cell growth rate after anti-Sema3A scFv treatment, whereas 131 cells hypersecreting Sema3A showed 70% of the cell growth rate after anti-Sema3A scFv treatment as compared to the control group (FIG. 10).
In order to verify the abilities to inhibit cell migration using anti-Sema3A scFv, the cell migration assay was performed using U87-MG, 131 and 83 cells which are Sema3A-hypersecreting cells .
First, PLO(Poly-L-Ornithine) was added to a transwell(Corning) and coated at room temperature for 30 minutes, followed by air-drying. For U87-MG cells, 5*104 U87-MG cells and 50 pg/ml of three species of Sema3A scFv were added to 100 ul of DMEM media without growth factor, and the solution was added to a transwell. 600 μΐ of DMEM culture solution containing 10% FBS (fatal bovine serum) was added to the bottom well and cultured overnight at 37°C . For 131 and 83 cells which are the patient-derived cells, l*105 cells and three species of Sema3A scFv were respectively added to 100 μΐ NBA culture solution not containing the growth factor (EGF and bFGF), and NBA culture solution containing the growth factor was added to the bottom well and cultured overnight at 37°C.
Then, 600 μΐ of methanol, hematoxylin and eosin were prepared for one per transwell in a 12-well, and then the transwell was kept in methanol for one minute and then allowed to stand in hematoxylin for 5 minutes to stain the nuclei.
Next, after washing with water and wiping moisture off, the resultant was kept in eosin for 30 seconds to stain cytoplasm. It was again washed with water and wiped cleanly inside the transwell with a cotton swab. It can be observed from FIG. 11 that the nucleus was stained by hematoxylin and the cytoplasm was stained by eosin.
For U87-MG cells, when cell migration of a control not treated with Sema3A scFv antibodies was regarded as 100%, cell migration of a cell with A0 8 antibody fragment was reduced to 78%, with C10 antibody fragment to 70%, with Fll antibody fragment to 74% (FIG. 11).
For patient-derived cells, 131 and 83, cell migration of a cell treated with A08 antibody fragment was reduced to 11% and 21%, with C10 antibody fragment to 19% and 44%, and Fll antibody fragment to 7% and 28%, respectively (FIGS. 12 and 13).
Three species of Sema3A antibody fragments exhibited a higher effect of inhibiting the cell migration in 131, 83 cells (patient-derived cells) than in cell line U87-MG, which showed the potential as an anti-cancer agent to inhibit cell migration of cancer cells.
Example 5: Production of IgG from Anti-Sema3A Antibody Fragment
For the conversion of anti-Sema3A antibody fragment into forms of IgG, the genes of the heavy chain sequences and light chain sequences of scFv Sema3A were transfected using Expi 293F expression system (life technologies).
In order to obtain Sema3A IgG in the culture solution, the purification was performed using AKTA protein purification system and Amicon centrifugal filter. The production amount was 118 mg/L for A08, 138 mg/L for CIO and 330 mg/L for Ell.
In order to confirm the purity of the purified anti-Sema3A antibody, the high performance liquid chromatography was introduced. Since the size of IgG is 150 kD, it corresponds to the material eluted from the marker peak at 16.388 minutes.
It was confirmed that three species of Sema3A antibodies (A08, CIO and Ell) were detected from this peak and the purity was 98%, 98.5% and 99%, respectively.
The forms of anti-IgG Sema3A according to the sizes were confirmed through SDS PAGE and Coomassie staining. Under nonreducing conditions, a band was detected at 150 kD which is the size of IgG, and under reducing conditions, the disulfide bond was broken and thus, the sizes of the heavy chain sequence and the light chain sequence were shown to be 50 kD and 25 kD, respectively (FIG. 15).
In order to confirm the binding affinity of three Sema3A antibodies to Sema3A, ELISA was performed under two concentration conditions (500 nM, 50 nM) . BSA was used as a negative control, and mouse Sema3A and human Sema3A proteins were used as an experimental group.
It was confirmed that three species of Sema3A antibodies have biding affinities to human Sema3A and mouse Sema3A, which can be seen in FIG. 16. The reason why the present antibody has a binding affinity to mouse Sema3A in addition to human Sema3A was assumed that the proteins have a low specificity between the species compared to other proteins and thus, the sequence homology between the human Sema3A and mouse Sema3A is 98% or more. Therefore, it is considered that the antibody has cross-linking abilities to human Sema3A and mouse Sema3A (FIG. 16) .
In order to measure the binding affinities of three antiSema3A antibodies to human Sema3A and mouse Sema3A, SPR analysis was performed using Biacore system.
The measurement results showed that the binding affinities to human Sema3A were A08 (KD=1.187E-9), C10 (KD=5.312E-10), and Fll (KD=5.617E-10), and the binding affinities to mouse Sema3A were A08 (KD=4.221E-9), C10 (KD=3.090E-9), and Fll (KD=3.272E-10).
Accordingly, it was confirmed that three anti-Sema3A antibodies showed cross-reactivity, and particularly Fll showed the highest binding affinities to human Sema3A and mouse Sema3A (FIG. 17).
Example 6: Verification on Abilities of Anti-Sema3A IgG to inhibit cell migration
As previously verifying abilities of anti-Sema3A scFv to inhibit cancer cell migration, the abilities of three Sema3A antibodies (A08, CIO and Fll) converted to IgG forms to inhibit cancer cell migration were re-verified. Cell migration assay was performed using U87-MG, 131 and 83 cells hypersecreting Sema3A, and 2 pg/ml of anti-Sema3A antibodies. Cell migration assay was performed by the methods such as those shown in FIG. 11 through FIG. 13 as previously described.
For U87-MG cells, A08 exhibited the highest abilities to inhibit cell migration with 50% (FIG. 18), and for 131 and 83 cells, Fll was the most effective which showed lower levels of cell migration with 74% and 52% respectively, compared to the control (FIGS. 19 and 20).
Studies that ERK signal mechanism is associated with cell migration in which Sema3A is involved in colorectal cancer (Neufeld, G et al., Cold Spring Harbor perspectives in medicine,2012) and that Sema3A is involved in Rho/ROCK signal mechanism and ERK signal mechanism in glioblastoma (Zohrabian,
V. M., Anti-cancer research , 119-123,2009) have been reported.
l*106 cells of 83 cells were treated with Fll(50 pg/ml) for 30 minutes at 37°C, followed by performing Western Blotting to confirm whether the Antibody can inhibit ERK phosphorylation or not. SDS-PAGE protein electrophoresis on 8% gel was carried out, and p-ERK, ERK and β-actin were probed with the antibodies.
The results of comparison of the control group and the experimental group with Fll treatment showed that ERK and Bactin were not changed, and ERK phosphorylation was reduced (FIG. 21).
Thus, it was confirmed that anti-Sema3A antibody inhibited cell migration by inhibiting the Phosphorylation of ERK among downstream signaling molecules of Sema3A.
Example 7: Verification on abilities of anti-Sema3A IgG to inhibit cell growth
Recombinant human Sema3A was treated with 131 and 83 cells followed by observing the changes of cell growth to find out whether Sema3A was involved in cell growth of glioblastoma. As the results of cell proliferation assay using Edu, it was confirmed that the cell growth was increased by 20 % and 15% in 131 and 83 cells, respectively (FIG. 22).
Then, As the results of Fll treatment to 131 cells, it was confirmed that the cell growth was inhibited depending on the concentration of antibody, and the inhibited cell growth to
40% compared to the control was observed at the highest concentration (2uM) of antibody (FIG. 23).
Example 8: Assessment on Efficacies of Anti-Sema3A IgG with 131 Subcutaneous Model
To confirm anti-cancer efficacy of anti-Sema3A Fll in vivo, a xenograft model was constructed using gliobalstoma 131 cells hypersecreting Sema3A.
As the results of confirmation of the sizes of the tumor after injecting with 5 mg/kg and 25 mg/kg of anti-Sema3A Fll (i.v.) for 3 weeks, it was confirmed that the tumor size was reduced to 60% in the group injected with 25 mg/kg (3 times/week) as compared to the control (FIG. 24) . Also, the changes of the tumor weight of individual groups were similarly calculated (FIG. 25).
Specific changes of the body weight by anti-Sema3A antibody injected were not confirmed (FIG. 26) . Immunofluorescence was performed in control group and Group 3 tissues (Fll 25 mg/kg, 3 times/week) which exhibited the highest efficacy, and it was confirmed that Sema3A and p-ERK were significantly reduced in the tissues of the groups treated with anti-Sema3A.
Apoptosis effects were also observed due to an increase of
2016346595 26 Oct 2016
TUNEL positive cells as compared to the control (FIG. 27) .
Many publications have reported that Sema3A is involved in TAM infiltration (Casazza A, et al. Cancer cell. 2013; 24(6):695709 / Hu ZQ, et al. Oncotarget. 2016).
Thus, to confirm this, the reduction of TAM distribution by Sema3A antibody was confirmed through staining Ibal which is a macrophage marker (FIG. 28).

Claims (15)

1. An antibody to human Sema3A or its antigen binding fragment comprising:
(a) a heavy chain variable region comprising the following heavy chain complementarity determining region(CDR) amino acid sequences:
CDRH1 consisting of the amino acid sequence of SEQ ID NO: 1,
CDRH2 consisting of the amino acid sequence of SEQ ID NO: 2, and
CDRH3 consisting of the amino acid sequence of SEQ ID NO: 3; and (b) a light chain variable region comprising the following light chain CDR amino acid sequences:
CDRL1 consisting of the amino acid sequence of SEQ ID NO: 4,
CDRL2 consisting of the amino acid sequence of SEQ ID NO: 5, and
CDRL3 consisting of the amino acid sequence of SEQ ID NO: 6.
2. The antibody or its antigen binding fragment according to claim 1, wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 19.
3. The antibody or its antigen binding fragment according to claim 1 or claim 2, wherein the light chain variable region comprises the amino acid sequence of SEQ ID NO: 20.
4 . An antibody to human Sema3A or its antigen binding fragment comprising:
(a) a heavy chain variable region comprising the following heavy chain CDR amino acid sequences:
CDRH1 consisting of the amino acid sequence of SEQ ID
NO: 7,
CDRH2 consisting of the amino
NO: 8, and
CDRH3 consisting of the amino
NO: 9; and acid sequence of SEQ ID acid sequence of SEQ ID (b) a light chain variable region comprising the
following light chain CDR . amino acid sequences: CDRL1 consisting of the amino acid sequence of SEQ ID NO: 10, CDRL2 consisting of the amino acid sequence of SEQ ID NO: 11, and CDRL3 consisting of the amino acid sequence of SEQ ID NO: 12 . 5. The antibody or its antigen binding fragment
according to claim 4, wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO:
21.
6. The antibody or its antigen binding fragment according to claim 4 or claim 5, wherein the light chain variable region comprises the amino acid sequence of SEQ ID NO: 22.
7 . An antibody to human Sema3A or its antigen binding fragment comprising:
(a) a heavy chain variable region comprising the following heavy chain CDR amino acid sequences:
CDRH1 consisting of the amino acid sequence of SEQ ID NO: 13, CDRH2 consisting of the amino acid sequence of SEQ ID NO: 14, and CDRH3 consisting of the amino acid sequence of SEQ ID NO: 15; and (b) i a light chain variable region comprising the following light chain CDR . amino acid sequences: CDRL1 consisting of the amino acid sequence of SEQ ID
2016346595 14 Oct 2019
NO: 16,
CDRL2 consisting of the amino acid sequence of SEQ ID
NO: 17, and
CDRL3 consisting of the amino acid sequence of SEQ ID
NO: 18.
8. The antibody or its antiqen bindinq fraqment accordinq to claim 7, wherein the heavy chain variable reqion comprises the amino acid sequence of SEQ ID NO: 23.
9. The antibody or its antiqen bindinq fraqment accordinq to claim 7 or claim 8, wherein the liqht chain variable reqion comprises the amino acid sequence of SEQ ID NO: 24.
10. A recombinant vector comprisinq:
a nucleic acid molecule encodinq a heavy chain variable reqion of an antibody comprisinq the amino acid sequence of SEQ ID NO: 19, SEQ ID NO:21 or SEQ ID NO:23; and a nucleic acid molecule encodinq a liqht chain variable reqion of an antibody comprisinq the amino acid sequence of SEQ ID NQ:20, SEQ ID NO:22 or SEQ ID NO:24.
11. A host cell transformed with the recombinant vector of claim 10; or a recombinant vector comprisinq a nucleic acid molecule encodinq a heavy chain variable reqion of an antibody comprisinq the amino acid sequence of SEQ ID NO: 19, SEQ ID NO:21 or SEQ ID NO:23, and a recombinant vector comprisinq a nucleic acid molecule encodinq a liqht chain variable reqion of an antibody comprisinq the amino acid sequence of SEQ ID NQ:20, SEQ ID NO:22 or SEQ ID NO:24.
2016346595 14 Oct 2019
12. A composition comprising:
(a) the antibody or its binding fragment against a human Sema3A of any one of claims 1 to 9; and (b) an acceptable carrier.
13. A method for preventing or treating a cancer in which Sema3A is highly expressed, the method comprising administering the composition of claim 12 to a subject.
14. The method according to claim 13, wherein the cancer is glioblastoma, pancreatic cancer, or liver cancer.
15. Use of the antibody or its binding fragment against a human Sem3A of any one of claims 1 to 9 for the manufacture of a medicament for treating or preventing a cancer in which Sema3A is highly expressed.
16. Use of the antibody according to claim 15, wherein the cancer is glioblastoma, pancreatic cancer, or liver cancer .
AU2016346595A 2015-10-27 2016-10-26 Antibody to be cross-linked to human and mouse Sema3A, and use thereof Active AU2016346595B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
KR20150149272 2015-10-27
KR10-2015-0149272 2015-10-27
KR1020160123233A KR101854529B1 (en) 2015-10-27 2016-09-26 Antibodies cross-reactive to Human and Mouse Sema3A and Uses thereof
KR10-2016-0123233 2016-09-26
PCT/KR2016/012072 WO2017074013A1 (en) 2015-10-27 2016-10-26 Antibody to be cross-linked to human and mouse sema3a, and use thereof

Publications (2)

Publication Number Publication Date
AU2016346595A1 AU2016346595A1 (en) 2018-06-14
AU2016346595B2 true AU2016346595B2 (en) 2019-11-14

Family

ID=58631728

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2016346595A Active AU2016346595B2 (en) 2015-10-27 2016-10-26 Antibody to be cross-linked to human and mouse Sema3A, and use thereof

Country Status (3)

Country Link
CN (1) CN108290942B (en)
AU (1) AU2016346595B2 (en)
WO (1) WO2017074013A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20220287A1 (en) 2019-05-09 2022-02-25 Boehringer Ingelheim Int ANTI-SEMA3A ANTIBODIES AND THEIR USES TO TREAT EYE DISEASES
US20220127344A1 (en) 2020-10-23 2022-04-28 Boehringer Ingelheim International Gmbh Anti-sema3a antibodies and their uses for treating a thrombotic disease of the retina
WO2022207554A1 (en) * 2021-03-30 2022-10-06 Bayer Aktiengesellschaft Anti-sema3a antibodies and uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003007803A2 (en) * 2001-07-20 2003-01-30 Mount Sinai School Of Medicine Of New York University Methods for diagnosing and treating alzheimer's disease and parkinson's disease
WO2014127479A1 (en) * 2013-02-21 2014-08-28 Rsem, Limited Partnership Inhibition of sema3a in the prevention and treatment of ocular hyperpermeability
WO2015083156A1 (en) * 2013-12-02 2015-06-11 Bnai Zion Medical Center Semaphorin 3a as a diagnostic marker for urothelial cancer

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011055550A1 (en) * 2009-11-05 2011-05-12 国立大学法人大阪大学 Therapeutic agent for autoimmune diseases or allergy, and method for screening for the therapeutic agent
US9879075B2 (en) * 2013-02-06 2018-01-30 Yokohama City University and Chiome Bioscience Inc. Anti-semaphorin 3A antibody and treatment of Alzheimer's disease and inflammatory immune diseases using same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003007803A2 (en) * 2001-07-20 2003-01-30 Mount Sinai School Of Medicine Of New York University Methods for diagnosing and treating alzheimer's disease and parkinson's disease
WO2014127479A1 (en) * 2013-02-21 2014-08-28 Rsem, Limited Partnership Inhibition of sema3a in the prevention and treatment of ocular hyperpermeability
WO2015083156A1 (en) * 2013-12-02 2015-06-11 Bnai Zion Medical Center Semaphorin 3a as a diagnostic marker for urothelial cancer

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SHEN, W-W et al; "Breast cancer cells promote osteoblastic differentiation via Sema 3A signaling pathway in vitro", Int J Clin Exp Pathol (2015);VOL: 8, NO: 2, PAGES:1584-1593 *

Also Published As

Publication number Publication date
AU2016346595A1 (en) 2018-06-14
CN108290942A (en) 2018-07-17
WO2017074013A1 (en) 2017-05-04
CN108290942B (en) 2021-08-27

Similar Documents

Publication Publication Date Title
KR102589136B1 (en) Anti-B7-H3 antibody and its uses
US9296817B2 (en) Antibodies cross-reactive to human and mouse c-Met and uses thereof
CN112574310B (en) anti-SIRP alpha antibodies and uses thereof
JP2023544143A (en) Anti-claudin 18.2 and anti-CD3 bispecific antibodies and uses thereof
AU2016346595B2 (en) Antibody to be cross-linked to human and mouse Sema3A, and use thereof
TW202132351A (en) Anti-cd47 / anti-pd-l1 antibodies and applications thereof
CN113227148B (en) anti-GPC 3 antibody, antigen-binding fragment thereof, and medical use thereof
US10604571B2 (en) Antibody to human and mouse SEMA3A and use thereof
CN110606892B (en) LAG-3 antibody with high affinity and high biological activity and application thereof
EP4286520A1 (en) Antigen binding protein and use thereof
KR20200063147A (en) PDL1 targeting antibodies and methods of use thereof
US20180346567A1 (en) Antibody To Be Cross-Linked To Human SEMA3A And Use Thereof
US10604572B2 (en) Antibody to human and mouse Sema3A and use thereof
US10640777B2 (en) Antibody to human and mouse SEMA3A and use thereof
CN112969715A (en) anti-CD 47 antigen binding protein and application thereof
EP4353749A1 (en) Anti-masp-2 antibody and use thereof
CN114316043B (en) TGF beta1 antigen binding molecule and application thereof
WO2024078558A1 (en) Anti-cd100 antibody and use thereof
JP2024509369A (en) Anti-PD-L1 antibody and its use
TW202313690A (en) Anti-b7-h4 antibody, and preparation method therefor and use thereof
CN117186235A (en) Bispecific antibody of anti-4-1 BB/anti-EGFRvIII, preparation method and application thereof
CN115215936A (en) CSF1R antigen binding proteins
CN116284406A (en) PD-1 binding protein and application thereof

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)