WO2020149613A1 - Anticorps ayant une meilleure aptitude à pénétrer le cytosol - Google Patents

Anticorps ayant une meilleure aptitude à pénétrer le cytosol Download PDF

Info

Publication number
WO2020149613A1
WO2020149613A1 PCT/KR2020/000679 KR2020000679W WO2020149613A1 WO 2020149613 A1 WO2020149613 A1 WO 2020149613A1 KR 2020000679 W KR2020000679 W KR 2020000679W WO 2020149613 A1 WO2020149613 A1 WO 2020149613A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
chain variable
light chain
variable region
amino acid
Prior art date
Application number
PCT/KR2020/000679
Other languages
English (en)
Korean (ko)
Inventor
김동명
박유진
Original Assignee
오름테라퓨틱 주식회사
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 오름테라퓨틱 주식회사 filed Critical 오름테라퓨틱 주식회사
Publication of WO2020149613A1 publication Critical patent/WO2020149613A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates to an antibody with improved cytoplasmic permeability, and to a cytoplasmic permeable antibody or antigen-binding fragment thereof. After cell-free protein synthesis, it was adopted as a screening method capable of evaluating cytoplasmic permeability without purification, and it was confirmed that the endosomal escape ability was improved and the antigen targeting ability was maintained.
  • therapeutic antibodies Due to the high specificity and strong binding affinity for specific ligands, therapeutic antibodies are rapidly replacing chemical drugs and have become the center of treatment in the treatment of cancer and autoimmune diseases.
  • Target specific binding of an antibody relieves or reverses symptoms through a variety of mechanisms including receptor blockade, complement dependent cytotoxicity, antibody dependent cytotoxicity and regulation of T cell function.
  • the present inventors have developed a light chain variable region (VL) that exhibits cytoplasmic penetration ability, and the cytoplasmic penetration antibody TMab4 in the form of a complete IgG introduced therein, unlike the general properties of these antibodies (Choi et al., 2014).
  • VL light chain variable region
  • TMab4 the cytoplasmic penetration antibody
  • a heavy chain variable region targeting the active Ras which is a major tumor inducer inside the cytoplasm
  • a light chain variable region showing cytoplasmic penetration ability are constructed in IgG form to develop the cytoplasmic penetration antibody RT11 targeting the active Ras inside the cytoplasm.
  • the light chain variable region of the cytoplasmic infiltrating antibody detects the acidic pH of the endosome, thereby inducing structural changes, thereby confirming the ability to escape the endosome, and the endosome escape ability is manipulated by manipulating the sequence of the ring region where structural changes occur.
  • An improved TMab4-WYW (TMab4-3) antibody was developed (Kim et al., 2016).
  • the RT11-3 antibody was constructed by combining the heavy chain variable region targeting the active Ras, which is the main tumor inducer inside the cytoplasm, and the light chain variable region with improved endosomal escape ability.
  • Korean Patent No. 1602870 As an experimental method for confirming the degree of location in the cytoplasm, Korean Patent No. 1790669 discloses an improved split green fluorescent protein complementary system and a technique for its use.
  • Korean Patent Registration No. 1667023 discloses a technique for easily analyzing the inflow of an antibody produced into a cytoplasm using a cell-free protein synthesis method, but a technique for an antibody having improved cytoplasmic permeability of the present invention Has not been disclosed.
  • the present invention has been derived by the above-mentioned needs, and the present invention has completed the present invention by providing an antibody with improved cytoplasmic permeability, and confirming that the antibody has improved permeability to the cytoplasm.
  • the present invention is an antibody with enhanced cytoplasmic permeability, comprising an amino acid variation in at least one selected from the group consisting of CDR1, FR and CDR3 among the light chain variable regions of SEQ ID NO: 2, or a cytoplasmic penetration antibody or Antigen-binding fragments are provided.
  • the present invention provides a nucleic acid encoding the cytoplasmic penetration antibody or antigen-binding fragment thereof.
  • the present invention provides a composition for the delivery of an active substance in the cytoplasm comprising the cytoplasmic penetration antibody or antigen-binding fragment thereof.
  • the present invention relates to a conjugate in which the cytoplasmic penetration antibody or antigen-binding fragment thereof and a bioactive molecule are fused.
  • the present invention is a method for placing a cytoplasmic penetration antibody or an antigen-binding fragment thereof through the cell membrane and placing it in the cytoplasm, wherein the antibody has amino acid mutations in one or more selected from the group consisting of CDR1, FR and CDR3 among the light chain variable regions of SEQ ID NO:2. How to include.
  • the present invention is a light chain variable region (VL) having a cytoplasmic penetration ability, which penetrates the cell membrane and induces it to be located in the cytoplasm, wherein at least one amino acid mutation selected from the group consisting of CDR1, FR and CDR3 among the light chain variable regions of SEQ ID NO:2 It relates to a light chain variable region (VL) comprising a.
  • RT11-3 scFv antibody 1 is a confocal microscopy image confirming the cytoplasmic-infiltrated RT11-3 scFv antibody.
  • soluble RT11-3 scFv antibody synthesized by fusion and in situ cleavage was purified with Ni-NTA agarose beads, the purified RT11-3scFv antibody was treated to reporter cells at concentrations of 0.25, 0.5, 0.75 and 1.0 ⁇ M. And cultured in a 5% (v/v) CO 2 incubator at 37° C. for 12 hours, fixed the cultured reporter cells with 4% paraformaldehyde for 15 minutes, and analyzed GFP fluorescence of reporter cells, Cell nuclei were stained with Hoechst 33342.
  • (A) is a fusion of the nucleotide sequence of ubiquitin (UCE1) to the RT11-3 scFv gene, and the amount of protein synthesized as a cell-free protein in a standard reaction mixture was confirmed by TCA-precipitated radioactivity.
  • Ubiquitin (UCE1) was synthesized using a gene without a sequence
  • UCE1-RT11-3 was synthesized using a gene fused with a ubiquitin sequence
  • the empty rod is the total protein synthesis amount
  • the filled rod is the amount of soluble protein. to be.
  • (B) is a result of comparing the expression level of the ubiquitin-fusion, using a standard S12 extract and a S12 extract rich in UBP1, confirmed by TCA-precipitated radioactivity measurement. The western blot result at the top confirms the cleavage of the ubiquitin tag.
  • C After expressing UCE1 fused RT11-3 scFv in the presence of UBP1 S12 extract, the reaction mixture was diluted 4-fold in DMEM medium, and reporter cells (1 ⁇ 10 4 ) were diluted with 400 ⁇ l of the reaction mixture. Cultured in the middle. After fixing the treated reporter cells with 4% paraformaldehyde, GFP fluorescence was confirmed under confocal microscopy.
  • Figure 5 shows the process of manufacturing the mutation location and mutation library RT11-3 scFv.
  • the wild-type residues indicated in black in (A) were designed to be replaced by amino acids in red, and the primers shown in (B) were used to construct a library through a combination of PCR-amplified DNA fragments.
  • the PCR-amplified mutant gene construct is expressed in a reaction mixture for cell-free protein synthesis, diluted 4 times in DMEM and added to reporter cells for 12 hours after incubation, the reporter cells are washed with PBS, and in 100 ⁇ l PBS. It is a result of measuring GFP fluorescence (excitation wavelength: 485 nm/emission wavelength: 528 nm).
  • Wild type (WT) and selected variant RT11-3 scFv (#1-60, #5-10, #6-32 and 6-91).
  • the reporter cells (1 ⁇ 10 4 cells/confocal dish) were diluted 4 times in DMEM and then purified in the same amount (1.5 ⁇ M). Treated with antibodies. After fixing with 4% paraformaldehyde for 15 minutes, GFP fluorescence of reporter cells was confirmed. The nucleus was stained with Hoechst 33342 dye.
  • 10(A) is a quantitative comparison of the number of cells obtained with trypan blue of pH7.4 and pH5.5 by an anti-Ras?GTP cytoplasmic penetration antibody comprising a light chain variable region with improved endosomal escape ability It is a graph.
  • Figure 10 (B) is a result of observation with a confocal microscope using calcein (calcein) for the migration to the cytoplasm according to the anti-Ras?GTP cytoplasmic penetration antibody containing a light chain variable region with improved endosomal escape ability .
  • Figure 10 (C) is a result of confirming the GFP fluorescence by confocal microscopy by complementary binding of the improved split green fluorescent protein of the GFP11-SBP2 fused cytoplasmic penetration antibody wild type and endosome escape enhancing mutant.
  • Figure 11 (A) is to confirm the specific binding of the GppNHp-associated KRasG12D of anti-Ras?GTP cytoplasmic penetration antibodies containing the light chain variable region with improved endosomal escape ability, 10 or 100nM of KRasG12D?GppNHp and 100nM It is the result of analyzing the binding capacity of KRasG12D?GDP by ELISA.
  • Figure 11 (B) is a result of confirming the ability to inhibit non-adherent cell growth in human colon cancer cell lines of anti-Ras?GTP cytoplasmic infiltrating antibodies comprising a light chain variable region with improved endosomal escape ability by spheroid proliferation method.
  • the present invention relates to a cytoplasmic penetration antibody or antigen-binding fragment thereof, comprising an amino acid variation in at least one selected from the group consisting of CDR1, FR and CDR3 among the light chain variable regions of SEQ ID NO: 2.
  • the target of the antibody is preferably an antibody characterized by KRas, but is not limited thereto.
  • the cytoplasmic penetration antibody or antigen-binding fragment thereof may include an amino acid substitution selected from the group consisting of the light chain variable region of SEQ ID NO: 2:
  • Amino acid A at position 34 is replaced by D or E;
  • Amino acid Y at position 36 is replaced by F;
  • Amino acid L at position 46 is substituted with K, M, I or R;
  • 89th Q is replaced by E, M, L, I or N;
  • 96th Y is replaced by T, W, F, I or K.
  • the cytoplasmic penetration antibody or antigen-binding fragment thereof may include a light chain variable region selected from the group consisting of SEQ ID NOs: 28 to 31. In some cases, the heavy chain variable region of SEQ ID NO: 1 may be further included.
  • Fv fragment is an antibody fragment that contains a complete antibody recognition and binding site. This region consists of a dimer in which one heavy chain variable domain and one light chain variable domain are tightly virtually covalently associated with, for example, scFv.
  • the antibody may be in the form of, for example, scFv.
  • the scFv includes the VH and VL domains of the antibody, and may further include a polypeptide linker between the VH domain and the VL domain, which enables to form a desired structure for antigen binding.
  • Variable region refers to the light and heavy chain portions of an antibody molecule comprising the amino acid sequences of complementarity determining regions (CDRs; ie CDR1, CDR2, and CDR3), and framework regions (FR).
  • CDRs complementarity determining regions
  • FR framework regions
  • VH refers to the variable region of the heavy chain.
  • VL refers to the variable region of the light chain.
  • CDR complementarity determining regions
  • Framework region is a variable region residue other than the CDR residues.
  • Each variable region typically has four FRs identified as FR1, FR2, FR3 and FR4.
  • the antibody or antigen-binding fragment thereof according to the present invention is preferably adopted by the following screening method, but is not limited thereto.
  • step (2) diluting the cell-free protein synthesis reaction solution containing the target antibody synthesized in step (1) without purification, to determine a cytotoxicity and cytoplasmic permeable concentration;
  • step (3) (4) using the PCR amplification product obtained in step (3) as a synthetic template to produce mutant cytoplasmic permeable antibody by cell-free protein synthesis;
  • step (4) treating the variant cytoplasmic permeable antibodies produced in step (4) to reporter cells at a concentration determined in step (2), and selecting the variant cytoplasmic permeability antibody with enhanced cell permeability;
  • step (6) identifying the affinity for the target antigen of the variant cytoplasmic permeable antibody with enhanced permeability of the cells selected in step (5); a method for screening for a permeable enhanced cytoplasmic permeable antibody.
  • step (4) the PCR amplification products are sequentially sequenced from 5'to 3'in the order of promoter, fusion partner, 6 ⁇ His tag, heavy chain region, linker 1, light chain region, linker 2, GFP 11 , SBP2 and terminator sequence.
  • the linked gene is preferred, but is not limited to this, and can be modified as necessary.
  • the target antibody is preferably a RT11-3 single chain varible fragment (SCFv) antibody or a variant thereof including the cytoplasmic permeable light chain variable region, but is not limited thereto.
  • SCFv single chain varible fragment
  • the reporter cells are characterized in cells expressing the GFP 1-10, 1-10 expressing GFP in the cytoplasm of the reporter cells is characterized in that color development of GFP fluorescence in combination with GFP 11 connected to the penetration antibody.
  • the reporter cells are preferably HeLa cells, but are not limited thereto.
  • the fusion partner may be used as long as it is a sequence capable of increasing the expression of downstream genes and the solubility and activity of the expressed antibody, and is preferably a ubiquitin sequence having a ubiquitin or nucleotide sequence variation, but is not limited thereto.
  • the present invention relates to a conjugate in which a cytoplasmic penetration antibody or an antigen-binding fragment thereof and a bioactive molecule are fused.
  • the present invention relates to a composition for bioactive molecule delivery in the cytoplasm comprising a cytoplasmic penetration antibody or antigen-binding fragment thereof.
  • the bioactive molecule may be a form fused or bound to an antibody, for example, one or more selected from the group consisting of peptides, proteins, toxins, antibodies, antibody fragments, RNA, siRNA, DNA, small molecule drugs, nanoparticles and liposomes However, it is not limited thereto.
  • the proteins include antibodies, antibody fragments, immunoglobulins, peptides, enzymes, growth factors, cytokines, transcription factors, toxins, antigenic peptides, hormones, transport proteins, motor function proteins, receptors, signals ( signaling protein, storage protein, membrane protein, transmembrane protein, internal protein, external protein, secreted protein, viral protein, sugar protein, truncated protein, protein complex, or chemically modified Protein and the like.
  • the RNA or ribonucleic acid is a type of nucleic acid constituting a nucleotide of a chain structure based on ribose, which is a kind of pentose sugar, and has a structure in which a single helix is long twisted, and a part of DNA is transcribed.
  • the RNA may be selected from rRNA, mRNA, tRNA, miRNA, snRNA, snoRNA, aRNA, but is not limited thereto.
  • the siRNA is an RNA interference material having a small size composed of dsRNA, and serves to decompose by binding to an mRNA having a target sequence, and to treat a target mRNA as a therapeutic agent for a disease or experimentally It is used extensively herein, with the activity of inhibiting the expression of proteins that are degraded and translated by the target mRNA.
  • the DNA or deoxyribonucleic acid is a type of nucleic acid, and has two types of a monosaccharide, deoxyribose, with a phosphate group bound to a backbone chain, purine, and pyrimidine. It is composed of nucleobases and is a substance that stores genetic information of cells.
  • the small molecule drug has a molecular weight of less than about 1000 Daltons and is widely used herein to refer to an organic compound, an inorganic compound, or an organometallic compound having activity as a therapeutic agent for a disease.
  • Small molecule drugs used in the present invention include oligopeptides and other biomolecules having molecular weights less than about 1000 Daltons.
  • the nanoparticle means a particle made of materials having a size of 1 to 1000 nm in diameter, and the nanoparticle is a metal/metal core composed of a metal nanoparticle, a metal nanoparticle core, and a metal shell surrounding the core It may be a metal/non-metallic coreshell composed of a shell composite, a metal nanoparticle core and a non-metallic shell surrounding the core, or a non-metallic/metallic coreshell composite composed of a non-metallic nanoparticle core and a metal shell surrounding the core.
  • the metal may be selected from gold, silver, copper, aluminum, nickel, palladium, platinum, magnetic iron and oxides thereof, but is not limited thereto, and the non-metal is silica, polystyrene, latex and acrylic It may be selected from a rate-based material, but is not limited thereto.
  • the liposome consists of one or more lipid bilayer membranes surrounding the aqueous inner compartment, which are capable of associating themselves.
  • Liposomes can be characterized by membrane type and size.
  • Small unilamellar vesicles SUVs
  • Large unilamellar vesicles LUV
  • Oligolamellar large vesicles and multilamellar large vesicles have multiple, generally concentric, membrane layers and may be 100 nm or more in diameter. Liposomes with multiple non-concentric membranes, ie several small vesicles contained within larger vesicles, are called multivesicular vesicles.
  • the "fusion” or “binding” is to integrate two molecules with different or identical functions or structures, and any physical, chemical or biological method capable of binding the tumor-penetrating peptide to the protein, small molecule drug, nanoparticle or liposome. It may be a fusion by.
  • the fusion may preferably be by a linker peptide, and the linker peptide may relay fusion with the active material at various positions of the antibody light chain variable region, antibody, or fragment thereof of the present invention.
  • the present invention is a method for placing a cytoplasmic penetration antibody or an antigen-binding fragment thereof through the cell membrane and placing it in the cytoplasm, wherein the antibody has amino acid mutations in one or more selected from the group consisting of CDR1, FR and CDR3 among the light chain variable regions of SEQ ID NO:2. How to include.
  • the present invention is a light chain variable region (VL) having a cytoplasmic penetration ability, which penetrates the cell membrane and induces it to be located in the cytoplasm, wherein at least one amino acid mutation selected from the group consisting of CDR1, FR and CDR3 among the light chain variable regions of SEQ ID NO:2 It relates to a light chain variable region (VL) comprising a.
  • the antibody may include an amino acid substitution selected from the group consisting of light chain variable region of SEQ ID NO: 2 (position is according to Kabat numbering).
  • Amino acid A at position 34 is replaced by D or E;
  • Amino acid Y at position 36 is replaced by F;
  • Amino acid L at position 46 is substituted with K, M, I or R;
  • 89th Q is replaced by E, M, L, I or N;
  • 96th Y is replaced by T, W, F, I or K.
  • the antibody may include a light chain variable region selected from the group consisting of SEQ ID NOs: 28 to 31.
  • the present invention provides a pharmaceutical composition for the prevention or treatment of cancer, comprising the cytoplasmic penetration antibody or antigen-binding fragment thereof and an active substance in the cytoplasm delivered thereby.
  • the active material By using the active material, it is possible to impart to the antibody the characteristics of remaining in the cytoplasm by penetrating into the cell without affecting the antigen's high specificity and high affinity. Through this, it is present in the cytoplasm, and a wide flat surface between the protein and the protein Through this, it is possible to expect a high effect in treatment and diagnosis related to tumor- and disease-related factors constituting a structure-composite interaction.
  • KRas mutant which is a major drug resistance-related factor of various existing tumor therapeutic agents
  • effective anticancer activity can be expected through concurrent treatment with existing therapeutic agents.
  • the cancer is squamous cell carcinoma, small cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous cell carcinoma of the lung, peritoneal cancer, skin cancer, melanoma of the skin or eye, rectal cancer, anal cancer, esophageal cancer, small intestine cancer, endocrine adenocarcinoma, Parathyroid cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, chronic or acute leukemia, lymphocyte lymphoma, hepatocellular carcinoma, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, liver tumor, breast cancer, colon cancer, colon cancer, Endometrial or uterine cancer, salivary gland cancer, kidney cancer, liver cancer, prostate cancer, vulva cancer, thyroid cancer, liver cancer and head and neck cancer may be selected from the group consisting of.
  • the composition may include a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier included in the composition is commonly used in the formulation, lactose, dextrose, sucrose, sorbitol, mannitol, starch, acacia rubber, calcium phosphate, alginate, gelatin, calcium silicate, fine Crystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil, and the like, but is not limited thereto.
  • the pharmaceutical composition may further include a lubricant, a wetting agent, a sweetener, a flavoring agent, an emulsifying agent, a suspending agent, a preservative, etc. in addition to the above components.
  • the pharmaceutical composition for the prevention or treatment of cancer may be administered orally or parenterally.
  • parenteral administration intravenous injection, subcutaneous injection, intramuscular injection, intraperitoneal injection, endothelial administration, topical administration, intranasal administration, intrapulmonary administration, and rectal administration may be administered.
  • proteins or peptides are digested, so oral compositions must be formulated to coat the active agent or to protect it from degradation in the stomach.
  • the composition can be administered by any device capable of transporting the active substance to the target cell.
  • Suitable dosages of the pharmaceutical composition for the prevention or treatment of cancer are factors such as formulation method, mode of administration, patient's age, weight, sex, morbidity, food, time of administration, route of administration, rate of excretion and response sensitivity. It can be variously prescribed by.
  • the preferred dosage of the composition is in the range of 0.001-100 mg/kg on an adult basis.
  • pharmaceutically effective amount means an amount sufficient to prevent or treat cancer, or to prevent or treat diseases caused by angiogenesis.
  • the composition may be prepared in a unit dose form by formulating using a pharmaceutically acceptable carrier and/or excipient, or by incorporating it into a multi-dose container, according to a method easily carried out by those skilled in the art.
  • the formulation may be in the form of a solution, suspension, syrup or emulsion in an oil or aqueous medium, or may be in the form of ex-agent, powder, granule, tablet or capsule, and may further include a dispersant or stabilizer.
  • the composition may be administered as an individual therapeutic agent or in combination with other therapeutic agents, and may be administered sequentially or simultaneously with conventional therapeutic agents. Meanwhile, since the composition includes an antibody or an antigen-binding fragment, it may be formulated as an immune liposome.
  • Liposomes comprising antibodies can be prepared according to methods well known in the art.
  • the immune liposome is a lipid composition comprising phosphatidylcholine, cholesterol and polyethylene glycol-derivatized phosphatidylethanolamine, and may be prepared by reverse phase evaporation.
  • Fab' fragments of an antibody can be conjugated to liposomes through a disulfide-replacement reaction.
  • Chemotherapeutic agents, such as doxorubicin may additionally be included in the liposome.
  • the present invention provides a composition for diagnosing cancer comprising the cytoplasmic penetration antibody or an antigen-binding fragment thereof and an active substance in the cytoplasm delivered thereby.
  • Diagnosis means identifying the presence or characteristics of the pathophysiology. Diagnosis in the present invention is to confirm the onset and progress of cancer.
  • the complete immunoglobulin antibody and fragments thereof can be combined with a phosphor for molecular imaging to diagnose cancer through imaging.
  • the molecular imaging phosphor refers to all materials that generate fluorescence, and it is preferable to emit red or near-infrared fluorescence, and a phosphor having a high quantum yield is more preferable, but is not limited thereto. .
  • the molecular imaging phosphor is preferably, but is not limited to, a phosphor, a fluorescent protein, or other imaging material capable of binding to the tumor-penetrating peptide specifically binding to the complete immunoglobulin-type antibody and a fragment thereof.
  • the phosphor is preferably fluorescein, BODYPY, tetramethyl rhodamine, Alexa, cyanine, allopicocyanine or derivatives thereof. Does not work.
  • the fluorescent protein is preferably, but not limited to, Dronepa protein, fluorescent chromogenic gene (EGFP), red fluorescent protein (DsRFP), cyanine phosphor that exhibits near-infrared fluorescence, or other fluorescent protein. .
  • imaging materials are preferably iron oxide, radioactive isotopes, etc., but are not limited thereto, and may be applied to imaging equipment such as MR and PET.
  • the present invention relates to a nucleic acid encoding the cytoplasmic penetration antibody or antigen-binding fragment thereof.
  • the nucleic acid is a polynucleotide
  • the "polynucleotide” is a polymer of deoxyribonucleotides or ribonucleotides present in single-stranded or double-stranded form. It encompasses RNA genomic sequences, DNA (gDNA and cDNA) and RNA sequences transcribed therefrom, and includes analogs of natural polynucleotides unless otherwise specified.
  • the polynucleotide includes not only the nucleotide sequence encoding the light chain variable region (VL) and heavy chain variable region (VH) with improved endosomal escape ability described above, but also a complementary sequence to the sequence.
  • the complementary sequence includes not only perfectly complementary sequences, but also substantially complementary sequences.
  • the nucleic acid can be modified. Such modifications include addition, deletion or non-conservative substitutions or conservative substitutions of nucleotides.
  • the nucleic acid encoding the amino acid sequence is also interpreted to include a nucleotide sequence showing substantial identity to the nucleotide sequence. The substantial identity is at least 80% homology when aligning the nucleotide sequence with any other sequence to the maximum correspondence and analyzing the aligned sequence using an algorithm commonly used in the art. It may be a sequence that exhibits at least 90% homology or at least 95% homology.
  • the DNA encoding the antibody is readily separated or synthesized using conventional procedures (eg, by using an oligonucleotide probe capable of specifically binding DNA encoding the heavy and light chains of the antibody).
  • the vector may be a vector system that simultaneously expresses the light and heavy chains in one vector, or a system that expresses each in separate vectors. In the latter case, both vectors can be introduced into the host cell through co-transfomation and targeted transformation.
  • vector used in the present invention means a means for expressing a target gene in a host cell.
  • viral vectors such as plasmid vectors, cosmid vectors and bacteriophage vectors, adenovirus vectors, retroviral vectors and adeno-associated virus vectors.
  • Vectors that can be used as the recombinant vector are plasmids often used in the art (e.g., pSC101, pGV1106, pACYC177, ColE1, pKT230, pME290, pBR322, pUC8/9, pUC6, pBD9, pHC79, pIJ61, pLAFR1, pHV14) , pGEX series, pET series, and pUC19, etc., phages (e.g., ⁇ gt4 ⁇ B, ⁇ -Charon, ⁇ z1 and M13, etc.) or viruses (e.g., CMV, SV40, etc.).
  • plasmids often used in the art (e.g., pSC101, pGV1106, pACYC177, ColE1, pKT230, pME290, pBR322, pUC8/9, pUC6, pBD9, pHC79, pIJ61
  • the light chain variable region, heavy chain variable region and/or linker between them provided in the present invention may be operably linked to a promoter.
  • operatively linked refers to a functional bond between a nucleotide expression control sequence (eg, promoter sequence) and another nucleotide sequence.
  • the regulatory sequence can thereby regulate the transcription and/or translation of the other nucleotide sequence.
  • the recombinant vector can typically be constructed as a vector for cloning or for expression.
  • the expression vector can be used in the art, conventional ones used to express foreign proteins in plants, animals or microorganisms.
  • the recombinant vector can be constructed through various methods known in the art.
  • the recombinant vector can be constructed using prokaryotic or eukaryotic cells as hosts.
  • a prokaryotic cell is a host
  • a strong promoter capable of progressing transcription eg, pL ⁇ promoter, trp promoter, lac promoter, tac promoter, T7 promoter, etc.
  • It usually contains a ribosome binding site for initiation of translation and a transcription/detox termination sequence.
  • the origin of replication operating in eukaryotic cells included in the vector includes f1 origin of replication, SV40 origin of replication, pMB1 origin of replication, adeno origin of replication, AAV origin of replication, CMV origin of replication, and BBV origin of replication. Including, but not limited to.
  • a promoter derived from the genome of a mammalian cell eg, a metallothionine promoter
  • a promoter derived from a mammalian virus eg, adenovirus late promoter, vaccinia virus 7.5K promoter, SV40 promoter, Cytomegalovirus (CMV) promoter and HSK's tk promoter
  • CMV Cytomegalovirus
  • a host cell transformed with the recombinant vector can be provided.
  • the host cell may be any host cell known in the art, and prokaryotic cells include, for example, E. coli JM109, E. coli BL21, E. coli RR1, E. coli LE392, E. coli B, Bacillus strains such as E. coli X 1776, E. coli W3110, Bacillus subtilis, Bacillus thuringiensis, and intestinal bacteria and strains such as Salmonella typhimurium, Serratia marcesons, and various Pseudomonas species.
  • prokaryotic cells include, for example, E. coli JM109, E. coli BL21, E. coli RR1, E. coli LE392, E. coli B, Bacillus strains such as E. coli X 1776, E. coli W3110, Bacillus subtilis, Bacillus thuringiensis, and intestinal bacteria and strains such as Salmonella typhimurium, Serratia marcesons, and various Pseudomonas
  • yeast Sacharomyce cerevisiae
  • insect cells plant cells and animal cells
  • SP2/0 Chinese hamster ovary (CHO) K1
  • CHO DG44 Chinese hamster ovary (CHO) K1
  • CHO DG44 Chinese hamster ovary (CHO) K1
  • CHO DG44 Chinese hamster ovary (CHO) K1
  • CHO DG44 Chinese hamster ovary (CHO) K1
  • CHO DG44 CHO DG44
  • PER.C6, W138 BHK
  • COS-7 COS-7
  • HepG2 Huh7, 3T3, RIN and MDCK cell lines and the like
  • an insertion method well known in the art can be used.
  • the transport method for example, when the host cell is a prokaryotic cell, a CaCl2 method or an electroporation method can be used, and when the host cell is a eukaryotic cell, a micro-injection method, calcium phosphate precipitation method, electroporation method, liposome -Mediated transfection and gene bombardment may be used, but is not limited thereto.
  • the method for selecting the transformed host cell can be easily performed according to a method well known in the art using a phenotype expressed by a selection label.
  • the selection marker is a specific antibiotic resistance gene
  • the transformant can be easily selected by culturing the transformant in a medium containing the antibiotic.
  • HeLa cells were purchased from the American Type Culture Collection (ATCC) and supplemented with 10% fetal bovine serum (GE Healthcare, Logan, UT, USA) and 1% antibiotic-antibacterial (Thermo Fisher Scientific, Waltham, MA, USA). Cultured in one DMEM (Life Technologies, Grand Island, NY, USA).
  • ATP ATP
  • GTP GTP
  • UTP UTP
  • CTP Creatine Phosphate
  • Creatine Kinase were purchased from Roche Applied Science, Indianapolis, IN, USA.
  • L-[U- 14 C] leucine was purchased from Perkin Elmer (Waltham, MA, USA). All other chemical reagents were purchased from Sigma-Aldrich (St. Louis, MO, USA) and used without further purification.
  • the S12 extract was prepared from E. coli strain BL21Star (DE3) (Thermo Fisher Scientific) containing plasmid pTUM4 according to a method known in the art.
  • VL and VH sequences of antibodies that are inherent in the cytoplasm and bind to KRas ⁇ GTP were assembled in the scFv format with (G 4 S) 3 linkers between the two chains (RT11-3 scFv).
  • Modeling of the three-dimensional structure of the mutant RT11-3 scFvs was performed using the ABodyBuilder algorithm (http://opig.stats.ox.ac.uk/webapps/sabpred), and the three-dimensional structure image of the antibody was PyMol It was shown using the program (Schrodinger, Cambridge, MA, USA).
  • RT11 VH and hT4-3 VL constituting RT11-3 scFv are shown in Table 1.
  • PCR library was cloned into plasmid pK7 (pK7-RT11-3Lib) and used to transform the E. coli DH5 ⁇ strain.
  • the individual variant RT11-3 gene was amplified by colony PCR and used as a template in cell-free protein synthesis. According to the experiment, to enhance soluble expression, a ubiquitin sequence was inserted upstream of the modified RT11-3 scFv gene.
  • the standard reaction mixture for cell-free protein synthesis of RT11-3 scFv is 57 mM HEPES-KOH (pH 8.2); 1.2 mM ATP; 0.85 mM each of GTP, UTP and CTP; 80 mM ammonium acetate; 34 ⁇ g/ml 1-5-formyl-5,6,7,8-tetrahydro folic acid (folic acid); 1.0 amino acid each of 20 amino acids; 2% PEG (8,000); 3.2 U/ml creatine kinase; 67 mM creatine phosphate; 0.01 mM L-[U- 14 C] leucine (11.1 GBq/mmol); 27% (v/v) S12 extract; And 50 ng/ml of PCR amplified DNA component.
  • a reaction mixture was prepared using a S12 extract rich in UBP1 instead of a standard S12 extract, and the cell-free protein synthesis reaction was performed at 30°C for 1 hour. For a while.
  • Quantification of scFv was quantified by measuring the TCA insoluble radioactivity level using a Tri-Carb 2810TR liquid scintillation counter. The amount of soluble antibody was determined by measuring the level of TCA insoluble radioactivity in the supernatant after centrifugation (13,000 x g, 10 minutes). The size of the cell-free protein synthesized scFv was confirmed by electrophoresis on 16% tricine gel, followed by Coomassie blue staining or Western blotting.
  • the reaction mixture was centrifuged at 13,000 ⁇ g for 10 minutes, and 5 ml of the supernatant was equilibrated with PBS, and then mixed with 400 ⁇ l of the Ni-NTA slurry. After washing 3 times with 2 ml of wash buffer (50 mM NaH2PO4, 300 mM NaCl and 10 mM imidazole), the resin-bound scFv was dissolved in 250 ⁇ l of elution buffer (50 mM NaH 2 PO 4 , 300 mM NaCl and 100 mM) (Imidazole).
  • wash buffer 50 mM NaH2PO4, 300 mM NaCl and 10 mM imidazole
  • the eluted solution was desalted and concentrated using a centrifugal ultrafiltration device (Merck Millipore, Burlington, MA, USA) equipped with a barrier membrane of 10,000 molecular weight. 500 ⁇ l of eluent was added and centrifuged at 14,000 ⁇ g for 10 minutes. The concentrated protein (50 ⁇ l) was diluted with 450 ⁇ l of PBS and centrifuged again. This process was repeated 5 times. After the final centrifugation step, desalted, concentrated scFv was recovered and diluted with PBS to the desired concentration.
  • a centrifugal ultrafiltration device Merck Millipore, Burlington, MA, USA
  • the cytoplasmic penetration efficiency of RT11-3 scFv was evaluated by split-GFP complementation assay.
  • the cytoplasmic-penetration efficiency of a cell-free protein synthesized RT11-3 scFv variant was measured using a HeLa cell line expressing a fusion structure of streptavidin and GFP 1-10 fragment [HeLa (SA-GFP 1-10 )].
  • RT11-3 scFv antibody contains a streptavidin-binding peptide (SBP2) sequence linked to the GFP 11 fragment, and thus, when the antibody is delivered to the cytoplasm, through the interaction between streptavidin and SBP2 , From the fragmented GFP 1-10 and GFP 11 fragments fused to show GFP fluorescence, cytoplasmic penetration of the RT11-3 scFv antibody was confirmed.
  • SBP2 streptavidin-binding peptide
  • HeLa-SA-GFP 1-10 cells were inoculated in 96-well plates at a density of 1 ⁇ 10 4 cells per well in 100 ⁇ l of DMEM and cultured at 37° C. for 5 hours at 5% (v/v) CO 2 condition. .
  • reporter cells were treated with 50 ⁇ l of cell-free protein synthesis RT11-3 scFv for 12 hours. Thereafter, the reporter cells were washed twice with PBS, and resuspended in 100 ⁇ l of PBS, and then GFP fluorescence (excitation wavelength: 485 nm / emission wavelength: 528 nm) was measured. Whether or not the cytoplasmic delivery of RT11-3 scFv was confirmed through confocal microscopy image analysis of reporter cells.
  • 3 ⁇ 10 5 reporter cells were treated with 1.2 ml of reaction mixture diluted in various ratios for 12 hours, and then analyzed by flow cytometry. Cell morphology was determined based on forward scattering and lateral scattering light.
  • Heavy chain variable region (RT11 VH: SEQ ID NO: X) and heavy chain constant region of an antibody in which DNA encoding the secretion signal peptide is fused at the 5'end to construct a heavy chain expression vector for production in the form of a complete IgG monoclonal antibody DNAs encoding heavy chains containing (CH1-hinge-CH2-CH3) were cloned into NotD/HindIII into pcDNA3.4 (Invitrogen) vector, respectively.
  • a cytoplasmic penetration light chain variable region (hT4-3, #1-60, #5-10, #6-32, fused with DNA encoding a secretory signal peptide at the 5'end) 6-91) and the DNA encoding the light chain containing the light chain constant region (CL) were cloned into NotD/HindIII into the pcDNA3.4 vector, respectively.
  • the light chain and heavy chain expression vectors were expressed and purified using transient transfection.
  • HEK293-F cells suspended in serum-free FreeStyle 293 expression medium were transfected with a mixture of plasmid and Polyethylenimine (PEI) (Polyscience).
  • PEI Polyethylenimine
  • 200 ml was transfected into a shake flask (Corning) HEK293-F cells were seeded in 100 ml of medium at a density of 2 ⁇ 10 6 cells/ml, and cultured at 150 rpm and 8% CO 2 conditions.
  • suitable heavy and light chain plasmids were diluted in 10 ml of FreeStyle 293 expression medium with 125 ⁇ g of heavy chain and 125 ⁇ g of light chain with a total of 250 ⁇ g (2.5 ⁇ g/ml), 750 ⁇ g of PEI (7.5 ⁇ g/ml) ) was mixed with 10 ml of diluted medium and reacted at room temperature for 10 minutes. Thereafter, the reacted mixed medium was put in cells seeded with 100 ml previously, and then cultured at 150 rpm and 8% CO 2 for 4 hours, and then the remaining 100 ml of FreeStyle 293 expression medium was added and cultured for 6 days.
  • Protein was purified from the cell culture supernatant taken with reference to the standard protocol. Antibodies were applied to Protein A Sepharose column (GE healthcare) and washed with PBS (pH 7.4). After eluting the antibody at pH 3.0 with 0.1M glycine buffer, the sample was immediately neutralized with 1M Tris buffer. The eluted antibody fraction was concentrated by exchanging buffer with PBS (pH 7.4) through a dialysis method. The purified protein was quantified using the absorbance at 280 nm and the extinction coefficient.
  • the experiment was conducted to confirm the change in the non-specific binding capacity of the antibody according to the manipulation of the light chain variable region sequence.
  • a washing buffer HBSS buffer, 50MM HEPES.
  • PBS, RT11-3, #1-60, #5-10, #6-32, 6-91, cetuximab 100 were added to a blocking buffer (HBSS buffer, 50MM HEPES, 1% BSA).
  • HRP-conjugated anti-human mAb is conjugated with a labeled antibody. Absorbance was quantified at 450 nm by reacting with TMB ELISA solution.
  • a cover slip was placed and 200 ⁇ l of 0.01% poly-L-lysine solution was added to the floating cell to attach Ramos to the plate, and reacted at 25° C. for 20 minutes. After washing with PBS, 5 ⁇ 10 4 Ramos cells per well were put in 0.5 ml of medium containing 10% FBS and cultured at 37° C. for 30 minutes.
  • cytoplasmic pH 7.4 buffer (HBSS (Welgene), 50 mM HEPES pH 7.4), the initial endosomal pH pH 5.5 buffer (HBSS (Welgene), 50 mM MES pH 5.5) 200 ⁇ l, 0.5 ⁇ M and 1 ⁇ M RT11-3, #1-60, #5-10, #6-32, and 6-91 were added and incubated at 37°C for 2 hours. Then, after carefully washing with PBS, 10 ⁇ l of trypan blue was mixed with 190 ⁇ l of PBS, and 200 ⁇ l per well was dispensed and observed under a microscope.
  • a cover slip was placed in a 24-well plate, and 2.5 ⁇ 10 4 HeLa cells per well were put into 0.5 ml of a medium containing 10% FBS and cultured at 5% CO 2 and 37° C. for 12 hours. After confirming cell adhesion, RT11-3, #1-60, #5-10, #6-32, 6-91 0.1, 0.25, 0.5 ⁇ M were cultured at 37° C. for 6 hours. After 4 hours, the wells containing the antibody were treated with 150 ⁇ M calcein and incubated at 37° C. for 2 hours. After removing the medium and washing with PBS, proteins attached to the cell surface were removed with a weakly acidic solution (200 mM glycine, 150 mM NaCl pH 2.5). After PBS washing, cells were fixed for 10 minutes at 25°C after adding 4% paraformaldehyde. The nuclei were stained (blue fluorescence) using Hoechst 33342 and observed with a confocal microscope.
  • a cover slip was placed in a 24-well plate, and 2.5 ⁇ 10 4 HeLa cells per well were put in 0.5 ml of a medium containing 10% FBS and cultured at 5% CO 2 and 37° C. for 12 hours. After confirming cell adhesion, 200 ⁇ l of RT11-3, #1-60, #5-10, #6-32 and 6-91 IgG were treated and incubated for 6 hours. Thereafter, the reporter cells were washed with PBS and a weakly acidic solution, and then the cells were fixed. The nuclei were stained (blue fluorescence) using Hoechst 33342 and observed with a confocal microscope.
  • TMab4-3 which is a cytoplasmic penetration antibody containing TMab4 VH without antigenic targeting ability and cytoplasmic penetration light chain variable region, was used as a control, and an anti-Ras?GTP cytoplasmic penetration antibody RT11-3 containing a light chain variable region with improved cytoplasmic penetration ability, After #1-60, #5-10, #6-32 and 6-91 were combined at a concentration of 5 ⁇ g/ml in 96-well EIA/RIA plates for 1 hour at room temperature, 0.1% TBST (12 mM Tris, It was washed three times for 10 minutes with pH 7.4, 137 mM NaCl, 2.7 mM KCl, 0.1% Tween20, 5 mM MgCl 2 ).
  • RT11-3 scFv As a platform expressing RT11-3 scFv, a cell-free protein synthesis system derived from E. coli extract was used. The RT11-3 scFv was produced at a concentration of 6.4 ⁇ M (221 ⁇ g/ml) in an initial experiment using a standard reaction mixture, and about 25% of the synthetic protein was included in the soluble fraction. RT11-3 scFv synthesized in 5 ml reaction was purified using Ni-NTA agarose resin. As described in [Materials and Methods], after desalting and concentration, about 1.8 nmole (62 ⁇ g) of purified protein was obtained in 0.3 ml of phosphate buffered saline (PBS: 6.0 ⁇ M).
  • PBS phosphate buffered saline
  • the purified RT11-3 scFv antibody was treated in a medium containing HeLa-SA-GFP1-10 reporter cells, and then confirmed through confocal microscopy image analysis.
  • the cytoplasmic penetration of the cell-free protein-synthesized RT11-3 scFv antibody was confirmed from the GFP fluorescence intensity increasing in proportion to the amount of protein added to the medium (FIG. 1 ).
  • the cell viability of reporter cells was analyzed by diluting the cell-free protein synthesis reaction mixture by concentration in DMEM (Dulbecco's Modified Eagle's Medium) in order to confirm that the above-mentioned cytotoxicity is alleviated. .
  • DMEM Dulbecco's Modified Eagle's Medium
  • the reporter cells were not significantly affected when the reaction mixture was diluted 4 times or more (FIG. 3A), and similarly, in the flow cytometry, the proportion of living cells was diluted 4 times or more for 12 hours. It was confirmed that cells were alive without being affected by incubation with (FIG. 3B).
  • a ubiquitin (hereinafter referred to as'UCE1') base sequence has been developed that stimulates the translation of a fusion protein much more effectively than a wild-type ubiquitin sequence. Can be improved up to.
  • RT11-3 of the present invention As a result of acellular protein synthesis of RT11-3 scFv fused with UCE1, soluble RT11-3 scFv of about 2.8 ⁇ M was produced (FIG. 4A ).
  • an antibody having a wild-type RT11-3 scFv sequence was produced using S12 extract containing UBP1 (ubiquitin carboxyl-terminal hydrolase 1), and UBP1 contained in the S12 extract cleaves the UCE1 tag in situ during a cell-free protein synthesis reaction.
  • S12 extract containing UBP1 ubiquitin carboxyl-terminal hydrolase 1
  • UBP1 contained in the S12 extract cleaves the UCE1 tag in situ during a cell-free protein synthesis reaction.
  • Western blot analysis confirmed that the concentration of the soluble protein of RT11-3 scFv in which the ubiquitin sequence was removed was maintained by maintaining cell-free protein synthesis from the extract rich in UBP1 (FIG. 4B ).
  • the mutant RT11-3 gene was mutated through the process disclosed in FIG. 5, and the mutated gene library was cloned into the pK7 plasmid (pK7RT11-3).
  • the amount of cytoplasmic penetrating antibody in this assay can be influenced by the efficiency of endosomal escape as well as its expression level.
  • reporter cells of the same concentration (1.5 ⁇ M) as the selected mutant scFv were purified after purification.
  • Measurement of the intensity of GFP fluorescence in reporter cells by confocal microscopy showed that GFP fluorescence showed the strongest antibody with #6-32, and GFP fluorescence intensity in the order of antibodies with 6-91, #1-60, and #5-10. It was confirmed (Fig. 8).
  • Example 4 Fully IgG-type cytoplasmic penetration antibody mutation RT11-3 expression and purification including the selected light chain variable region (VL) with improved endosomal escape ability
  • the cytoplasmic penetration antibody was constructed in scFv form to select a light chain variable region with improved endosomal escape ability. Improving the escape ability of the selected endosomes
  • the light chain variable region was expressed and purified in animal cells as a monoclonal antibody in the form of a complete IgG to confirm the improvement of the escape ability of endosomes.
  • a heavy chain variable region of an antibody in which DNA encoding the secretion signal peptide is fused at the 5'end (RT11 VH: SEQ ID NO: 1)
  • DNA encoding the heavy chain containing the heavy chain constant region was cloned into NotD/HindIII in pcDNA3.4 (Invitrogen) vector, respectively.
  • a cytoplasmic penetration light chain variable region (hT4-3, #1-60, #5-10, #6-32, fused with DNA encoding a secretory signal peptide at the 5'end) 6-91) and the DNA encoding the light chain containing the light chain constant region (CL) were cloned into NotD/HindIII into pcDNA3.4 (Invitrogen) vector, respectively.
  • the light chain and heavy chain expression vectors were expressed and purified using transient transfection.
  • HEK293-F cells Invitrogen
  • serum-free FreeStyle 293 expression medium Invitrogen
  • PEI Polyethylenimine
  • HEK293-F cells Upon 200 ml transfection in a shake flask (Corning), HEK293-F cells were seeded in 100 ml of medium at a density of 2 ⁇ 10 6 cells/ml, and cultured at 150 rpm, 8% CO 2 .
  • the appropriate heavy and light chain plasmids were diluted in 10 ml of FreeStyle 293 expression medium (Invitrogen) to 125 ⁇ g of heavy chain and 125 ⁇ g of light chain to a total of 250 ⁇ g (2.5 ⁇ g/ml), 750 ⁇ g (7.5 ⁇ g/ml) of PEI was mixed with diluted 10 ml of medium and reacted at room temperature for 10 minutes. Thereafter, the reacted mixed medium was put into cells seeded with 100 ml previously, and then cultured at 150 rpm and 8% CO2 for 4 hours, and then the remaining 100 ml of FreeStyle 293 expression medium was added and cultured for 6 days.
  • FreeStyle 293 expression medium Invitrogen
  • Protein was purified from the cell culture supernatant taken with reference to the standard protocol. Antibodies were applied to Protein A Sepharose column (GE healthcare) and washed with PBS (pH 7.4). After eluting the antibody at pH 3.0 with 0.1M glycine buffer, the sample was immediately neutralized with 1M Tris buffer. The eluted antibody fraction was concentrated by exchanging buffer with PBS (pH 7.4) through a dialysis method. The purified protein was quantified using absorbance and absorption coefficient at a wavelength of 280 nm.
  • Example 5 Confirmation of non-specific binding of the cytoplasmic penetration antibody in the form of a complete IgG comprising a light chain variable region (VL) with improved endosome escape capability
  • the HeLa (HSPG+), pgsD-677 (HSGP-) cell line was incubated in a 96-well plate so that the cells were completely filled at the bottom of the well, and then washed three times with a washing buffer (HBSS buffer, 50 mM HEPES). Then, PBS, RT11-3, #1-60, #5-10, #6-32, #6-91, cetuximab were added to blocking buffer (HBSS buffer, 50mM HEPES, 1% BSA) 100, 50, 25 , 12.5, 6.25 and diluted to a concentration of 3.125ng/ml, and incubated at 4°C for 2 hours.
  • HBSS buffer 50 mM HEPES
  • BSA blocking buffer
  • HRP-conjugated anti-human mAb was conjugated with a labeled antibody. Absorbance was quantified by reacting with TMB ELISA solution. The highest non-specific cell surface binding capacity was measured in #6-32 antibody, and the other antibodies confirmed the non-specific cell surface binding capacity almost similar to that of wild type RT11-3 (FIG. 9).
  • Example 5 a light chain variable region with improved endosomal escape ability was selected in the form of scFv, and the endosomes escape ability improvement of the complete IgG form containing this light chain variable region was improved.
  • a cover slip was placed in a 24-well plate, and 2.5 ⁇ 10 4 HeLa cells per well were put into 0.5 ml of a medium containing 10% FBS and cultured at 5% CO 2 at 37° C. for 12 hours. After confirming cell adhesion, RT11-3, #1-60, #5-10, #6-32, #6-91 0.1, 0.25, 0.5 ⁇ M were cultured at 37° C. for 6 hours. After 4 hours, 150 ⁇ M of calcein was treated in the wells containing the antibody and cultured at 37° C. for 2 hours. After removing the medium and washing with PBS, proteins attached to the cell surface were removed with a weakly acidic solution (200 mM glycine, 150 mM NaCl pH 2.5).
  • a weakly acidic solution 200 mM glycine, 150 mM NaCl pH 2.5.
  • Example 7 Cytoplasmic penetration through complementary binding of an improved split green fluorescent protein Confirmation of the cytoplasmic location of a monoclonal antibody
  • RT11-3-GFP11-SBP2, #1-60-GFP11-SBP2, #5-10-GFP11-SBP2, #6-32-GFP11- fused with GFP11-SBP2 peptide to the heavy chain C-terminus of RT11 SBP2, and #6-91-GFP11-SBP2 were constructed.
  • RT11-3-GFP11-SBP2, #1-60-GFP11-SBP2, # 5-10-GFP11-SBP2, #6-32-GFP11-SBP2, #6-91-GFP11-SBP2 0.2, 0.4, and 0.8 ⁇ M were cultured at 37° C. for 6 hours. After washing with PBS and a weakly acidic solution, the cells were fixed. The nuclei were stained (blue fluorescence) using Hoechst 33342 and observed with a confocal microscope.
  • Example 8 Evaluation of tumor cell growth inhibitory ability of anti-Ras?GTP cytoplasmic penetration antibody comprising light chain variable region with improved endosomal escape ability
  • TMab4-3 which is a cytoplasmic penetration antibody containing TMab4 VH without an antigen targeting ability and an endosomal escape light chain variable region, was used as a control, and an anti-Ras?GTP cytoplasmic penetration antibody RT11- comprising an endosomal escape ability enhanced light chain variable region 3, #1-60, #5-10, #6-32, #6-91 were combined at a concentration of 5 ⁇ g/ml in 96-well EIA/RIA plates for 1 hour at room temperature, and then 0.1% TBST (12 mM) Tris, pH 7.4, 137mM NaCl, 2.7mM KCl, 0.1% Tween20, 5mM MgCl 2 ) were washed 3 times for 10 minutes.
  • Antibodies #1-60, #6-32, and #6-91, except for #5-10 antibodies, show KRasG12D binding similar to RT11-3, and the Ras targeting ability of heavy chain variable regions is affected by changes in light chain variable regions. It was confirmed that it was hardly received (Fig. 11(A)).
  • the #1-60 and #6-32 antibodies which have improved endosomal escape ability and maintain antigen targeting ability, are Ras mutant cell line specific cells. It was confirmed that the growth inhibitory effect was improved.
  • the present invention relates to an antibody having improved cytoplasmic permeability, and is an antibody having a mutant sequence introduced into a light chain variable region.
  • the antibody according to the present invention has an effect of improving endosomal escape ability and maintaining antigen targeting ability.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biophysics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne un anticorps ayant meilleure aptitude à pénétrer le cytosol, et un anticorps pénétrant le cytosol comprenant une mutation d'acide aminé sur au moins un élément choisi dans le groupe constitué par CDR1, FR et CDR3 dans une région variable de chaîne légère, ou un fragment de liaison à l'antigène de celle-ci. L'anticorps a été choisi à l'aide d'un procédé de criblage permettant d'évaluer l'anticorps pour sa capacité à pénétrer le cytosol, sans purification après la synthèse de protéines acellulaires et s'est avéré être utile pour améliorer la capacité d'échappement endosomique et maintenir une capacité de ciblage d'antigène.
PCT/KR2020/000679 2019-01-15 2020-01-14 Anticorps ayant une meilleure aptitude à pénétrer le cytosol WO2020149613A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20190005299 2019-01-15
KR10-2019-0005299 2019-01-15

Publications (1)

Publication Number Publication Date
WO2020149613A1 true WO2020149613A1 (fr) 2020-07-23

Family

ID=71613957

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2020/000679 WO2020149613A1 (fr) 2019-01-15 2020-01-14 Anticorps ayant une meilleure aptitude à pénétrer le cytosol

Country Status (2)

Country Link
KR (1) KR20200088780A (fr)
WO (1) WO2020149613A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022138692A1 (fr) * 2020-12-23 2022-06-30 Chugai Seiyaku Kabushiki Kaisha Molécules de liaison à l'antigène présentant une activité de pénétration dans le cytosol améliorée
CN116574748A (zh) * 2023-07-10 2023-08-11 昆明医科大学 一种用于靶向KRAS高频突变肿瘤的嵌合型nTCR-T构建方法

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20160011599A (ko) * 2014-07-22 2016-02-01 아주대학교산학협력단 완전한 이뮤노글로불린 형태의 세포질 침투능을 갖는 항체를 이용하여 세포내 활성화된 ras를 억제하는 방법 및 그의 이용
US20170073429A1 (en) * 2014-03-04 2017-03-16 Yale University Cell penetrating anti-guanosine antibody based therapy for cancers with ras mutations
KR20180129514A (ko) * 2017-05-26 2018-12-05 오름테라퓨틱 주식회사 세포질 침투 항체 및 이의 용도
KR20190056340A (ko) * 2017-11-16 2019-05-24 오름테라퓨틱 주식회사 세포의 세포질에 침투하여 세포내 활성화된 Ras를 억제하는 항체 및 이의 용도

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170073429A1 (en) * 2014-03-04 2017-03-16 Yale University Cell penetrating anti-guanosine antibody based therapy for cancers with ras mutations
KR20160011599A (ko) * 2014-07-22 2016-02-01 아주대학교산학협력단 완전한 이뮤노글로불린 형태의 세포질 침투능을 갖는 항체를 이용하여 세포내 활성화된 ras를 억제하는 방법 및 그의 이용
KR20180129514A (ko) * 2017-05-26 2018-12-05 오름테라퓨틱 주식회사 세포질 침투 항체 및 이의 용도
KR20190056340A (ko) * 2017-11-16 2019-05-24 오름테라퓨틱 주식회사 세포의 세포질에 침투하여 세포내 활성화된 Ras를 억제하는 항체 및 이의 용도

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KIM, J.: "Engineering of a tumor cell -specific, cytosol-penetrating antibody with high endosomal escape efficacy", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 503, 4 July 2018 (2018-07-04), pages 2510 - 2516, XP055679694, DOI: 10.1016/j.bbrc.2018.07.008 *
SHIN, S.-M.: "Antibody targeting intracellular oncogenic Ras mutants exerts anti-tumour effects after systemic administration", NATURE COMMUNICATIONS, vol. 8, 10 May 2017 (2017-05-10), pages 1 - 14, XP055434123, DOI: 10.1038/ncomms15090 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022138692A1 (fr) * 2020-12-23 2022-06-30 Chugai Seiyaku Kabushiki Kaisha Molécules de liaison à l'antigène présentant une activité de pénétration dans le cytosol améliorée
CN116574748A (zh) * 2023-07-10 2023-08-11 昆明医科大学 一种用于靶向KRAS高频突变肿瘤的嵌合型nTCR-T构建方法
CN116574748B (zh) * 2023-07-10 2023-09-12 昆明医科大学 一种用于靶向KRAS高频突变肿瘤的嵌合型nTCR-T构建方法

Also Published As

Publication number Publication date
KR20200088780A (ko) 2020-07-23

Similar Documents

Publication Publication Date Title
KR101602870B1 (ko) 완전한 이뮤노글로불린 형태의 항체를 세포막을 투과하여 세포질에 위치시키는 방법 및 그의 이용
Funami et al. The cytoplasmic ‘linker region’in Toll-like receptor 3 controls receptor localization and signaling
KR101602876B1 (ko) 완전한 이뮤노글로불린 형태의 세포질 침투능을 갖는 항체를 이용하여 세포내 활성화된 ras를 억제하는 방법 및 그의 이용
WO2016153276A1 (fr) Peptide liant spécifiquement la neuropiline-1, protéine de fusion fusionnée à celui-ci et utilisation correspondante
WO2014189303A1 (fr) Peptide transtumoral spécifique de la neuropiline et protéine de fusion comprenant ce peptide fusionné
WO2016013870A1 (fr) Procédé pour positionner, dans le cytoplasme, un anticorps au format immunoglobuline complète par pénétration de l'anticorps à travers la membrane cellulaire, et son utilisation
Caron de Fromentel et al. Restoration of transcriptional activity of p53 mutants in human tumour cells by intracellular expression of anti-p53 single chain Fv fragments
KR100847010B1 (ko) 세포사멸 수용체 5 (dr5)에 특이적으로 결합하는 항체 및이를 포함하는 암 예방 또는 치료용 조성물
WO2020149613A1 (fr) Anticorps ayant une meilleure aptitude à pénétrer le cytosol
WO2021133036A1 (fr) Anticorps anti-lilrb1 et ses utilisations
US11155641B2 (en) Cytosol-penetrating antibody and use thereof
KR20190056340A (ko) 세포의 세포질에 침투하여 세포내 활성화된 Ras를 억제하는 항체 및 이의 용도
WO2024178772A1 (fr) Anticorps à domaine unique se liant de manière spécifique à la protéine cll1 et son utilisation
WO2022216014A1 (fr) Anticorps anti-cntn4 et son utilisation
Gellini et al. Generation of human single-chain antibody to the CD99 cell surface determinant specifically recognizing Ewing’s sarcoma tumor cells
WO2016013871A1 (fr) Procédé d'inhibition de la protéine ras activée dans une cellule utilisant un anticorps ayant la capacité de pénétrer dans le cytoplasme et se présentant sous la forme d'une immunoglobuline complète, et son utilisation
Wang et al. Potent, selective CARs as potential T-cell therapeutics for HPV-positive cancers
Ota et al. LyGDI functions in cancer metastasis by anchoring Rho proteins to the cell membrane
EP2754451A1 (fr) Utilisation d'ache comme nucléase
KR102091195B1 (ko) 세포질 침투 항체 및 이의 용도
KR20180129514A (ko) 세포질 침투 항체 및 이의 용도
WO2022025585A1 (fr) Anticorps anti-lilrb1 et ses utilisations
WO2023022271A1 (fr) Anticorps anti-igsf1 et son utilisation
WO2018226020A2 (fr) Gène d'anticorps dr4 humanisé ayant une activité inductrice d'apoptose et agent thérapeutique à double action à base de cellule t de récepteur d'antigène chimérique ou de cellule tueuse naturelle l'utilisant
KR102154177B1 (ko) 세포질 침투성이 증진된 항체의 스크리닝 방법

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20742077

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20742077

Country of ref document: EP

Kind code of ref document: A1