WO2010047509A9 - Anti-dr5 antibody with improved affinity and stability, and composition for cancer prevention or treatment including same - Google Patents

Anti-dr5 antibody with improved affinity and stability, and composition for cancer prevention or treatment including same Download PDF

Info

Publication number
WO2010047509A9
WO2010047509A9 PCT/KR2009/006036 KR2009006036W WO2010047509A9 WO 2010047509 A9 WO2010047509 A9 WO 2010047509A9 KR 2009006036 W KR2009006036 W KR 2009006036W WO 2010047509 A9 WO2010047509 A9 WO 2010047509A9
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
cancer
seq
amino acid
chain variable
Prior art date
Application number
PCT/KR2009/006036
Other languages
French (fr)
Korean (ko)
Other versions
WO2010047509A2 (en
WO2010047509A3 (en
Inventor
김용성
권명희
이승현
박경진
Original Assignee
아주대학교 산학협력단
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 KR1020090066518A external-priority patent/KR101117070B1/en
Application filed by 아주대학교 산학협력단 filed Critical 아주대학교 산학협력단
Publication of WO2010047509A2 publication Critical patent/WO2010047509A2/en
Publication of WO2010047509A9 publication Critical patent/WO2010047509A9/en
Publication of WO2010047509A3 publication Critical patent/WO2010047509A3/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • 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]

Definitions

  • the present invention specifically binds to a death receptor 5 (hereinafter referred to as "DR5"), anti-DR5 antibody having improved affinity and stability for effectively killing various cancer cells, and a composition for preventing or treating cancer comprising the same. It is about.
  • DR5 death receptor 5
  • anti-DR5 antibody having improved affinity and stability for effectively killing various cancer cells
  • composition for preventing or treating cancer comprising the same. It is about.
  • TRAIL TNF-related apoptosis inducing ligand
  • TNFR protein p53-independent tumor necrosis factor receptor
  • TRAIL recombinant TRAIL
  • TRAIL-sensitive cancer cells cancer cells killed by TRAIL
  • TRAIL-resistant cancer cells cancer cells that do not die by TRAIL are called TRAIL-resistant cancer cells.
  • Antibodies having affinity specific for DR5 include TRA-8 (mouse-derived IgG), a humanized antibody developed from a mouse-derived monoclonal antibody (Walczak et al., Nat. Med., 5: 157-161) and AD5-10 (rat IgG) (Guo et al., J. Biol. Chem., 280: 41940-41952, 2005), HGS-ETR2 (human IgG1), a human-derived monoclonal antibody (Georgakis et al., Br. J. Haematol. , 130: 501-510, 2005) and KMTR2 (human IgG4) (Motoki et al. , Clin. Cancer Res. , 11: 3126-3135, 2005).
  • TRA-8 mouse-derived IgG
  • AD5-10 rat IgG
  • HGS-ETR2 human IgG1
  • KMTR2 human IgG4
  • Apoptosis of eukaryotic cells is characterized by three mechanisms, namely apoptosis, autophagy and necrosis, depending on morphological and biochemical characteristics (Kromer G et al. (2005), Cell Death Differ. 12 Suppl 2: 1463-1467). Among these, apoptosis is classified as type II programmed cell death and is increased by nutrient starvation, environmental stress, or various compounds, and the process is called endoplasmic.
  • reticulum or a newly synthesized double-membrane structure is created, which forms autophagosomes while isolating intracellular organelles (such as mitochondria) or cytoplasmic proteins, ultimately lysosomes and Fusion and destruction (Kondo Y et al. (2005). Nat. Rev. Cancer. 5: 726-734). If the child action is above a certain level, it causes apoptosis, and the development of cancer treatments using such apoptosis is attracting attention (Martinet W et al. (2009), Clinical Science. 116: 697-712).
  • the V fragment of the variable region of the antibody is a fragment that occupies most of the antibody sequence, and there are various subtypes.
  • VH heavy chain variable region
  • VH1a, VH1b, and VH5 are intermediate. Position, and VH2, VH4 and VH6 fall in terms of stability.
  • VL light chain variable region
  • Vk3 and Vk1 are known to be the most stable forms.
  • the CDR grafting method is remarkable, as it shows the results of the studies that improved the thermodynamic stability of antibodies by grafting CDRs of less stable antibodies to subtypes having relatively higher thermodynamic stability and yield of antibody production (Jung et al. , Protein.Eng . , 10: 959-966, 1997).
  • the improvement of the affinity of the therapeutic antibody can be expected to reduce the single dose, it can lead to a comprehensive treatment efficiency, such as cost reduction and increased efficacy, reduced side effects.
  • an increase in ligand-receptor binding affinity in agonists is not necessarily associated with biological activity (Jones et al . , Trends. Biotechnol. , 26: 498-505, 2008).
  • the inventors of the present invention while studying to develop an anti-DR5 antibody with improved affinity and stability than the existing anti-DR5 antibody, and produced a variant of the existing anti-DR5 human antibody, HW1, and the affinity and The stability was improved more than the existing HW1 and confirmed that effectively induce apoptosis by the progeny action on TRAIL-sensitive cancer cells expressing DR5 or TRAIL-resistant cancer cells expressing DR5, and completed the present invention.
  • the present invention is to provide an anti-DR5 antibody with improved affinity and stability.
  • the present invention is to provide a DNA encoding the anti-DR5 antibody.
  • the present invention is to provide a cell transformed with the DNA or an expression vector comprising the same.
  • the present invention is to provide a composition for preventing or treating cancer containing the anti-DR5 antibody as an active ingredient.
  • FIG. 1 is a diagram illustrating primers of VH-CDR2, VH-CDR3, and VL-CDR3 for improving affinity of HW1, an anti-DR5 human antibody.
  • Figure 2 is a diagram showing the manufacturing process of a variant library for improving the affinity of the anti-DR5 human antibody HW1.
  • 3 is a diagram analyzing the process of selecting a high affinity variant using the FACS from the variant library of HW1.
  • FIG. 4 is a diagram analyzing the affinity of the finally selected variants from the variants library of HW1 and HW1 again using FACS.
  • 5 is a diagram showing the entire nucleotide and amino acid sequence of the antibody AU11 (where the underlined portions are in turn VH-CDR1, VH-CDR2, VH-CDR3, linker, VL-CDR1, VL-CDR2, VL-CDR3).
  • Figure 6 is a view showing a result of comparing the amino acid sequence of the AU11 finally improved the affinity and stability of the existing antibody HW1 (* indicates amino acid residues are changed according to affinity improvement in CDR).
  • FIG. 7 is a diagram showing the entire nucleotide and amino acid sequence of the antibody AU12 (where the underlined portions are in turn VH-CDR1, VH-CDR2, VH-CDR3, linker, VL-CDR1, VL-CDR2, VL-CDR3).
  • FIG. 8 is a diagram showing the results of comparative analysis of the amino acid sequences of AU11 and AU12 which finally improved the affinity and stability of the existing antibody HW1.
  • FIG. 9 is a diagram showing the results of analyzing the size and purity of purified antibodies (AU11 and AU12) by reducing SDS-PAGE and non-reducing SDS-PAGE.
  • FIG. 10 is a diagram showing the results of analyzing purified antibodies (AU11 and AU12) by size exclusion chromatography.
  • FIG. 11 is a diagram showing the result of quantifying the affinity of AU11 and AU12 for DR5 using SPR and comparing the affinity with HW1.
  • Figure 13 is a diagram showing the result of measuring the same antigen binding site between HW1 and AU11 or AU12 using a competitive ELISA.
  • FIG. 14 is a diagram showing the results of measuring the cross-reactivity of AU11 and AU12 against the target antigen DR5 antigen and other analogues (DR4, DcR1, DcR2) by ELISA.
  • FIG. 15 shows the binding affinity between total DR5 and DR5 fragments CRD2 (residues 97-137) and CRD3 (residues 139-180) and HW1, AU11, AU12, TRAIL having binding capacity to them using ELISA.
  • Figure 1 shows the results.
  • FIG. 16 is a diagram showing the results of comparing and evaluating the degree of apoptosis of HW1, AU11, and AU12 in HCT116, a human colorectal cancer cell line, and U87MG, a human neurotumor cell line, by MTT-assay.
  • FIG. 17 shows MTT-assay of apoptosis ability of HW1, AU11 and AU12 in the presence of various inhibitors (Z-VAD, SP600125, SB203580, 3-MA, Chloroguine) in human colon cancer cell line HCT116 and human neurotumor cell line U87MG. It is a figure which shows the result of analysis.
  • Fig. 18 shows the results of fluorescence microscopy observation of LC3-GFP aggregates by treatment of AU11 and AU12 to U87MG cancer cell lines expressing LC3-GFP.
  • the present invention provides an anti-DR5 antibody that specifically binds to DR5, which has the amino acid sequence of SEQ ID NO: 7 or 39.
  • the present invention also provides a DNA encoding the anti-DR5 antibody.
  • the present invention also provides a cell transformed with the DNA or an expression vector comprising the same.
  • the present invention also provides a composition for preventing or treating cancer containing the anti-DR5 antibody as an active ingredient.
  • Anti-DR5 antibody according to the present invention, the amino acid residues and light chain variable VH-CDR2, VH-CDR3 in the heavy chain variable region (heavy chain variable region) of the anti-DR5 human antibody HW1 (Korean Patent No. 10-0847010) Mutating amino acid residues of VL-CDR3 in the light chain variable region to enhance affinity for DR5, as well as replacing the framework in the heavy chain variable region from the VH6 subtype to the VH3 subtype, and light chain variable It is characterized by improving the stability by substituting the structure in the region with the structural part of Vk1 or Vk3.
  • apoptotic receptor-5 (DR5) protein means a member of the tumor necrosis factor (TNF) receptor family and binds TRAIL and has an intracellular death domain at the C-terminus ( Pan et al., Science, 277: 815-818, 1997).
  • TNF tumor necrosis factor
  • DR5 binds to TRAIL, it is known to induce apoptosis in TRAIL-sensitive cancer cells and to increase apoptosis when DR5 is overexpressed, but not to induce apoptosis in normal cells.
  • DR5 includes any protein having the above characteristics, but may be, for example, having an amino acid sequence described in US Pat. No. 6,872,568, but is not limited thereto.
  • an "antibody” may be an entire form of an antibody ("antibody”) or a functional fragment thereof.
  • the whole antibody may be in the form of a monomer or a multimer in which two or more whole antibodies are bound.
  • the functional fragment of the antibody is an antibody having the heavy and light chain variable regions of the whole antibody, which means to recognize substantially the same epitope that the whole antibody recognizes.
  • Functional fragments of the antibody include, but are not limited to, single chain variable region fragments (scFv), (scFv) 2 , Fab, Fab 'and F (ab') 2 and the like, and scFv is preferred in the present invention.
  • the single chain variable region (scFv) refers to an antibody fragment in which a heavy chain variable region and a light chain variable region are linked through a linker peptide to take the form of a single chain polypeptide.
  • the antibody can be generated using methods known in the art, such as phage display methods or yeast cell surface expression systems.
  • Antibodies of the invention may be derived from any animal, including mammals, birds, and the like, including humans.
  • the antibody may be a human, mouse, donkey, sheep, rabbit, goat, guinea pig, camel, horse or chicken antibody.
  • the human antibody is an antibody having an amino acid sequence of human immunoglobulin, which includes an antibody isolated from a human immunoglobulin library or an antibody isolated from an animal transfected against one or more human immunoglobulins and not expressing an endogenous immunoglobulin ( US Pat. No. 5,939,598).
  • Antibodies of the present invention may be conjugated to enzymes, fluorescent materials, radioactive materials and proteins, but are not limited thereto. In addition, methods of conjugating such materials to antibodies are well known in the art.
  • Anti-DR5 antibody according to the present invention, the heavy chain variable region having complementary determining region CDR1, CDR2 and CDR3 having the amino acid sequence of SEQ ID NO: 1, 2 and 3 and the complementarity determining region CDR1, CDR2 and CDR3 are SEQ ID NO: 4, 5, respectively And a light chain variable region having the amino acid sequence of 6, specifically binding to DR5 only.
  • “specifically binds” means that the antibody of the present invention binds only to the DR5 antigen and does not substantially bind to DR5 (death receptor 4), death decoy receptor 1 (DcR1) and DcR2, which are DR5-like antigens. it means.
  • the light chain variable region has an amino acid sequence of SEQ ID NO: 27 or 37
  • the heavy chain variable region has an amino acid sequence of SEQ ID NO: 28 or 38.
  • the anti-DR5 antibodies of the present invention are AU11 or AU12, which are scFv antibodies having an amino acid sequence of SEQ ID NO: 7 or 39, which in turn comprise CDR1 to CDR3 of the heavy chain variable region, linker oligopeptide and CDR1 to CDR3 of the light chain variable region, respectively.
  • the antibody is preferably a monoclonal antibody.
  • Anti-Dr5 antibodies of the present invention bind to DR5 with higher affinity than HW1, and the equilibrium dissociation constant ( K D ) shows 1.17 ⁇ 10 ⁇ 8 M and 9.17 ⁇ 10 ⁇ 9 M, respectively.
  • the anti-Dr5 antibody AU11 or AU12 is relatively higher in stability than HW1, and similarly to HW1, a single molecule scFv form induces apoptosis by progeny.
  • the present invention provides DNA encoding AU11 or AU12, which is an anti-DR5 antibody.
  • the DNA may be DNA encoding an scFv having an amino acid sequence of SEQ ID NO: 7 or 39, preferably DNA having a nucleotide sequence of SEQ ID NO: 8 or 40.
  • DNA sequences encoding the antibodies of the invention can be obtained by methods well known in the art. For example, based on DNA sequences or corresponding amino acid sequences encoding portions or all of the heavy and light chains of the antibody, oligonucleotide synthesis techniques well known in the art, for example site-directed mutagenesis And the polymerase chain reaction (PCR) method.
  • PCR polymerase chain reaction
  • the present invention also provides a cell transformed with the DNA or an expression vector comprising the same.
  • the DNA or expression vector comprising the same can be delivered to an appropriate host cell using methods known in the art, such as viral transfection or non-viral based techniques.
  • the introduction of the DNA or expression vector includes, but is not limited to, adenovirus transformation, gene gun, liposome-mediated transformation and retrovirus or lentiviral-mediated transformation, plasmid, adeno-associated virus, and the like. It may be performed by any known technique.
  • the cells can be transplanted with a suitable carrier material capable of releasing or delivering the gene to the cells for a long time.
  • the cells transformed with the DNA or the expression vector including the same may be cultured under appropriate conditions to express and isolate the antibody to produce antibody molecules.
  • the antibody molecule may accumulate in the cytoplasm of the cell, be secreted from the cell, or may be targeted to periplasm or extracellular medium by an appropriate signal sequence, among which is targeted to periplasm or extracellular medium. desirable. It is also desirable to refold the produced antibody molecules and to have functional conformation using methods well known to those skilled in the art.
  • a single vector comprising a sequence encoding the heavy or light chain polypeptide is transduced into the host cell, and in order to produce an antibody including both the heavy and light chain polypeptides.
  • a first vector encoding the light chain polypeptide and a second vector encoding the heavy chain polypeptide or a single vector comprising both the light chain and heavy chain polypeptide sequences. It may be.
  • the present invention also provides a composition for preventing or treating cancer containing the anti-DR5 antibody as an active ingredient.
  • the anti-DR5 antibodies (AU11 and AU12) according to the present invention bind TRDR-sensitive cancer cells or DR5 that specifically bind to the DR5 antigen with a higher affinity than the existing anti-DR5 antibodies (HW1), and have relatively high stability and express DR5.
  • HW1 existing anti-DR5 antibodies
  • Cancer caused by the DR5 expression may include both TRAIL-sensitive cancer cells and TRAIL-resistant cancer cells, specifically, hematologic cancer, lung cancer, stomach cancer, liver cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, skin melanoma, Uterine cancer, ovarian cancer, rectal cancer, colon cancer, colon cancer, breast cancer, uterine sarcoma, fallopian tube carcinoma, endometrial carcinoma, cervical carcinoma, vaginal carcinoma, vulvar carcinoma, esophageal cancer, laryngeal cancer, small intestine cancer, thyroid cancer, parathyroid cancer, soft tissue sarcoma, Urethral cancer, penile cancer, prostate cancer, chronic or acute leukemia, solid tumors of childhood, differentiated lymphoma, bladder cancer, kidney cancer, renal cell carcinoma, renal pelvic carcinoma, primary central nervous system lymphoma, spinal contraction tumor, brain stem glioma or pituitary gland Adenomas and the like, but is not limited thereto.
  • composition of the present invention may contain one or more known active ingredients having an anticancer effect together with the anti-DR5 antibody.
  • composition of the present invention may be prepared by including one or more pharmaceutically acceptable carriers in addition to the above-described active ingredients for administration.
  • Pharmaceutically acceptable carriers may be used in combination with saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol and one or more of these components, if necessary, as an antioxidant, buffer And other conventional additives such as bacteriostatic agents can be added.
  • Diluents, dispersants, surfactants, binders and lubricants may also be added in addition to formulate into injectable formulations, pills, capsules, granules or tablets such as aqueous solutions, suspensions, emulsions and the like.
  • it may be preferably formulated according to each disease or component by a suitable method in the art or using a method disclosed in Remington's Pharmaceutical Science (Recent Edition), Mack Publishing Company, Easton PA.
  • composition of the present invention can be administered orally or parenterally (eg, applied intravenously, subcutaneously, intraperitoneally or topically) according to the desired method, and the dosage is based on the weight, age, sex and health of the patient. The range varies depending on the diet, the time of administration, the method of administration, the rate of excretion and the severity of the disease.
  • the daily dose of the anti-DR5 antibody is about 0.01 to 50 mg / kg, preferably about 0.1 to 20 mg / kg, and more preferably administered once to several times a day.
  • composition of the present invention can be used alone or in combination with methods using surgery, hormonal therapy, drug therapy and biological response modifiers for the prevention and treatment of cancer.
  • Example 1 Construction of variant library for improving affinity of HW1
  • CDR Complementarity Determining Region
  • Mutant library was designed by giving priority to CDR2, and the vector for expressing the variant on the yeast surface was used pCTCON, a yeast surface expression vector, and random nucleotide sequences were assigned to CDR2 of the heavy chain variable region as shown in FIG. .
  • the ratio of wild-type bases is 70% instead of the base mixtures of 25% each of the four bases generally used.
  • a base mixture was used (for example, adenine 70% adenine, guanine, cytosine, thymine each 10% instead of the base mixture containing 25% adenine, guanine, cytosine and thymine at the site where the wild type base is adenine (A)). Variants were constructed using the base mixture contained thick). In the same manner as described above, a primer (fragment 1-SEQ ID NO: 9, SEQ ID NO: 12; fragment 2-SEQ ID NO: 11, SEQ ID NO: 10) (Gennotek, Korea) was finally prepared so that the retention rate of each amino acid was higher than a predetermined level.
  • variants with improved affinity were selected from the mutant library of heavy chain variable CDR2, and the primers (heavy chain variable CDR3: fragment) were selected in the heavy chain variable CDR3 and light chain variable CDR3 sites, respectively, using the selected antibodies as templates.
  • Two libraries have been built to further enhance affinity. Primer sequence numbers and sequences used to construct variants for improving affinity are shown in Table 1 below.
  • the first fragment is 5'-GGT GGT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3 '(SEQ ID NO: 9) and 5'-CCT TCC CAG CCA CTC AAG GCC-3 as reverse primer.
  • SEQ ID NO: 12 was used to perform a polymerase chain reaction (PCR).
  • the second fragment is 5'-GGC CTT GAG TGG CTG GGA AGG 135 415 415 325 435 115 425 415 115 215 TAT GCA GTA TCT GTG AAA AGT-3 '(SEQ ID NO: 11) with forward primer and 5'-GAT CTC with reverse primer.
  • GAG CTA TTA CAA GTC CTC TTC AGA AAT AAG CTT TTG TTC GGA TCC-3 '(SEQ ID NO: 10) was prepared by PCR method. PCR was performed using pfu polymerase (Intron, Korea), and the reaction of 40 seconds at 94 ° C, 30 seconds at 55 ° C, and 40 seconds at 72 ° C was repeated 30 times.
  • fragments for the variants of the heavy chain variable CDR3 and light chain variable CDR3 sites, respectively, were prepared in the same manner as above.
  • the first fragment of heavy chain variable CDR3 was 5'-GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3 '(SEQ ID NO: 9) as the forward primer, and 5'-ACA GTA ATA GAC GGC as the reverse primer.
  • CGT GTC-3 '(SEQ ID NO: 14) was used, and the second fragment was a 5'-GAC ACG GCC GTC TAT TAC TGT 235 125 215 335 215 235 225 125 225 235 445 215 145 TGG GGC CAA GGG ACC ACG GTC -3 '(SEQ ID NO: 13), 5'-GAT CTC GAG CTA TTA CAA GTC CTC TTC AGA AAT AAG CTT TTG TTC GGA TCC-3' (SEQ ID NO: 10) as a reverse primer was prepared by the PCR method.
  • the first fragment of the light chain variable CDR3 was 5'-GGT GGT GGT GGT TCT GGT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3 '(SEQ ID NO: 9) as the forward primer, 5'-ACA GTA ATA AAC TGC as the reverse primer AAA ATC-3 '(SEQ ID NO: 16) was used, and the second fragment was a 5'-GAT TTT GCA GTT TAT TAC TGT 315 315 325 125 115 425 335 335 325 235 245 TTC GGC CAA GGG ACA CGA CTG-3 '(SEQ ID NO: 15), 5'-GAT CTC GAG CTA TTA CAA GTC CTC TTC AGA AAT AAG CTT TTG TTC GGA TCC-3' (SEQ ID NO: 10) as a reverse primer was prepared by the PCR method.
  • Each of the two fragments prepared was electrophoresed on a 1% agarose gel and purified using an agarose gel purification kit (Intron, Korea).
  • the purified two fragments were mixed in the same amount by 10 ⁇ M each, and then, as shown in FIG. 2, overlap extension PCR was performed to prepare a mutated whole scFv gene product.
  • the PCR was performed using pfu polymerase (Intron, Korea), and the reaction of 40 seconds at 94 ° C, 30 seconds at 55 ° C, and 1 minute at 72 ° C was repeated 30 times.
  • Mutant-inserted scFv antibody gene library (10 ⁇ g / ⁇ l) and scFv yeast surface expression vector pCTCON (1 ⁇ g / ⁇ l) were mixed, and then, electroporation was performed to the yeast EBY100 strain (Invitrogen, USA). Transformed.
  • the transformed yeast EBY100 strain was selected as SD-CAA (-ura, -trp; 20 g / l glucose, YNB without 6.7 g / l amino acid (BD, USA), 5.4 g / l Na 2 Library inducing cell surface expression of scFv in SG-CAA medium substituted with glucose to galactose after inoculation with HPO 4 , 8.56 g / L NaH 2 PO 4 H 2 O and 5 g / L casamino acid) Were screened. Cells containing the library were diluted 10-fold in SD-CAA medium stepwise to determine library size.
  • the scFv antibody library size was each constructed at about 2 ⁇ 10 7 .
  • Example 2 Screening for Improved Affinity Variants from Constructed HW1 Variant Library
  • Anti-c-myc mAb (Ig therapy, Korea) and biotin label diluted 1: 100 to select scFv variants showing high affinity to DR5 from the variant library of HW1 constructed in Example 1 above Biotin-labeled DR5 was added to PBS (PBSB, pH 7.4) with 0.2 mg of 1 mg / mL BSA using the kit (EZ-LINK TM Sulfo-NHS-LC-Biotinylation kit, Pierce, USA). Then, incubated for 30 minutes at 25 °C.
  • nucleotide sequences of the selected variants were forward primer 5'-GTT CCA GAC TAC GCT CTG CAG G-3 '(SEQ ID NO: 17) and reverse primer 5'-GAT TTT GTT ACA TCT ACA CTG TTG-3' (SEQ ID NO: 18) After analysis using the amino acid sequence was determined and compared with the amino acid sequence of the wild type.
  • the affinity of the HW1 variant was increased for each selection step.
  • the amino acid sequence of the variants selected from each library was determined and the changed CDRs of the selected wildtypes were respectively combined to finally produce new variants with the most affinity.
  • a variant derived from heavy chain variable CDR3 was used as a template, and 5'-GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3 '(SEQ ID NO: 9) and 5'- as reverse primer. Fragments were prepared by PCR using ACA GTA ATA AAC TGC AAA ATC-3 ′ (SEQ ID NO: 16).
  • 5'-GAT TTT GCA GTT TAT TAC TGT-3 '(SEQ ID NO: 19) as the forward primer 5'-GAT CTC GAG CTA TTA CAA GTC CTC TTC AGA as the reverse primer A fragment was prepared by PCR using AAT AAG CTT TTG TTC GGA TCC-3 ′ (SEQ ID NO: 10), and the reaction was repeated 30 times at 40 ° C. at 94 ° C., 30 seconds at 55 ° C., and 50 seconds at 72 ° C.
  • Each of the two fragments prepared was electrophoresed on a 1% agarose gel and purified using an agarose gel purification kit (Intron, Korea), and the two purified fragments were mixed in equal amounts by 10 ⁇ M each, and then superimposed. Extended PCR was performed to finally produce a variant whole scFv gene product with improved affinity. The PCR was repeated 30 times 40 minutes at 94 °C, 30 seconds at 55 °C, 1 minute at 72 °C.
  • the mutant scFv product thus prepared was introduced into the yeast surface expression vector pCTCON (Colby et al., Methods Enzymol., 388: 348-358, 2004) using the restriction enzyme Nhe I / BamH I (NEB, USA). The plasmid was transformed into yeast EBY100 strain (Invitrogen).
  • the final variant to which each variant was bound showed the highest affinity than the other variants.
  • HW1 an anti-DR5 human antibody
  • VH6 subtype for the heavy chain variable region and Vk3 for the light chain variable region
  • VH6 subtype is compared to other subtypes, particularly VH3. It is known that the stability is low.
  • the VH6 subtype was substituted with the VH3 subtype.
  • the whole gene was divided into three fragments and each fragment was prepared.
  • the first fragment is a forward primer 5'-TTC GCT AGC GAG GTG CAG CTG GTG GAG TCT GGG GGA GGC CTG GTA CAG CCT GGA GGG TCC CTG AGA CTC TCC TGT GCA GCC TCT GGA GAC AGT GTC TCT AGC ACC ACT GTT GCC ATG AGC TGG GTC CGC CAG GCT CCA GGG AAG GGG CTG GAG TGG GTC-3 '(SEQ ID NO: 20), 5'-ACA GTA ATA CAC AGC CGT GTC CTC GGC TCT CAG GCT GTT CAT TTG CAG ATA CAG GGT GTT CTT GGA ATT GTC TCT GGA GAT GGT GAA CCG GCC CTT CAC GGA GTC CGC ATA GTA ATT ATA CCA CTT CGA CCT ATA ATA GAT CGC TGA GAC CCA CTC CAG CCC CTT CCC-3 '(SEQ ID NO: 21).
  • the second fragment is 5'-GAG GAC ACG GCT GTG TAT TAC TGT-3 '(SEQ ID NO: 22) with forward primer, 5'-TTC CCC TGG AGA CAA AGA CAG GGT GGC TGG AGA CTG GGT CAA TAC AAT ATC CGA with reverse primer Produced using CCC GCC ACC GCC GCT GCC ACC-3 '(SEQ ID NO: 23).
  • the third fragment is 5'-ACC CTG TTG TCT TTG TCT CCA GGG GAA-3 '(SEQ ID NO: 24) with forward primer, 5'-CGT GGA TCC ACG TTT GAT TTC CAC CTT TGT CCC TTG GCC GAA GAC CGC CCG- It was produced using 3 '(SEQ ID NO: 25).
  • 4 glycine and 1 serine is repeated three times, using a (G 4 S) 3 linker consisting of a total of 15 amino acids (SEQ ID NO: 26).
  • Each fragment thus prepared was subjected to overlap extension PCR to finally prepare a variant whole scFv gene product having improved stability.
  • PCR was repeated 30 times 40 minutes at 94 °C, 30 seconds at 55 °C, 1 minute at 72 °C. Subsequently, valine # 101 and # 102 serine in the CDR3 region of the heavy chain variable region were replaced with aspartic acid and isoleucine to finally prepare AU11, an antibody having improved affinity and stability. Primer sequence numbers and sequences used to improve stability with antibody AU11 are shown in Table 2 below.
  • FIG. 5 Schematic representation of the entire nucleotide and amino acid sequence of antibody AU11 is shown in FIG. 5 (wherein the underlined portions are in turn VH-CDR1, VH-CDR2, VH-CDR3, linker, VL-CDR1, VL-CDR2,
  • FIG. 6 A comparison of the amino acid sequence of AU11 with improved affinity and stability with the existing antibody HW1 is shown in FIG. 6 (* is an amino acid residue which is changed according to affinity improvement in CDR). ).
  • Vk3 and Vk1 are known to be the most stable, and many Vk1 subtypes exist in various mouse-derived humanized antibodies commercially approved by the US FDA (Carter et al., Proc Natl Acad). Sci USA , 10; 89: 4285-4289, 1992; Werther et al ., J Immunol , 11; 157: 4986-4995, 1996; Presta et al ., J Immunol 5; 151: 2623-2632, 1993).
  • vernier zone residues contribute to thermodynamic stability in antigen binding, and are known as sites that modulate the structural entropy changes of antibodies and consequently have a significant effect on antigen binding (Makabe et al.
  • the first fragment was 5'-TTC GCT AGC GAG GTG CAG CTG GTG GAG TCT GGG-3 '(SEQ ID NO: 29) with forward primer, 5'-TGG AGA CTG GGT CAT CTG AGA GTC with reverse primer -3 '(SEQ ID NO: 30) was used, and the second fragment was a 5'-GAC TCT CAG ATG ACC CAG TCT CCA TCC TCC CTG TCT GCA TCT GTA GGA GAC AGA GTC ACC ATC ACT TGC CGG GCA AGT CAG AGC-3 '(SEQ ID NO: 31), 5'-CTT AGG GGC TTT CCC TGG TTT CTG CTG ATA CCA GGC TAA GTG GCT-3' (SEQ ID NO: 32) was used as the reverse primer, and the third fragment was a forward primer 5'-AAA GCC CCT AAG CTC CTG ATC TAT GGT GCA TCC AGC
  • AU11 As a linker linking the heavy chain variable region and the light chain variable region, (G 4 S) 3 linker in which 4 glycine and 1 serine was repeated three times was used. Each fragment thus prepared was subjected to overlap extension PCR to finally prepare a variant whole scFv gene product having improved stability. In the PCR, AU12 was finally produced by repeating a reaction of 40 seconds at 94 ° C, 30 seconds at 55 ° C, and 1 minute at 72 ° C for 30 times. Primer sequence numbers and sequences used in the preparation of AU12 are shown in Table 3 below.
  • FIG. 7 A schematic of the entire nucleotide and amino acid sequence of antibody AU12 is shown in FIG. 7 (wherein the underlined portions are in turn VH-CDR1, VH-CDR2, VH-CDR3, linker, VL-CDR1, VL-CDR2, VL-CDR3), and comparing the amino acid sequence of AU11 and AU12 finally improved affinity and stability with the existing antibody HW1 is shown in FIG. 7 (wherein the underlined portions are in turn VH-CDR1, VH-CDR2, VH-CDR3, linker, VL-CDR1, VL-CDR2, VL-CDR3), and comparing the amino acid sequence of AU11 and AU12 finally improved affinity and stability with the existing antibody HW1 is shown in FIG.
  • Antibodies with improved affinity and stability were introduced into the E. coli expression vector pKJ1 in-frame using Nhe I / BamH I. At this time, the E. coli expression vector was designed to have a T7 promoter-pelB targeting sequence-AU11 or AU12 scFv-flag tag-6X his tag.
  • antibodies AU11 and AU12 have a molecular weight of about 29 kDa. It was confirmed that the purity was 99% or more.
  • the elution buffer is PBS (50 mM phosphate, pH 7.4, 150 mM NaCl), flow rate 0.5 ml / min, column 280 nm using SuperdexTM 200 10/300 GC (GE, USA) and Agilent 1100 HPLC instrument Absorbance was measured at. Reducing SDS-PAGE was performed using 15% acrylamide gel with 1 mM DTT added to the sample buffer and non-reducing SDS-PAGE was performed with 15% acrylamide gel without 1 mM DTT added to the sample buffer, 150 volts. Analyze by loading.
  • the purified AU11 and AU12 scFv antibodies were free of multimolecular forms with non-natural disulfide bonds in reducing SDS-PAGE and non-reducing SDS-PAGE.
  • AU11 and AU12 were measured using a Biacore 2000 surface Plasmon resonance (SPR) biosensor (GE, USA). About 0.5 mg / ml of antigen and negative control group BSA (bovine serum albumin) were immobilized on a carboxymethylated dextran surface chip (CM5) at about 2,000 response units, and then the antibodies AU11 and AU12 diluted at various concentrations were added. Interaction with antigen was quantified by injection into the chip for 2 minutes at a rate of 30 ⁇ l / min.
  • SPR surface Plasmon resonance
  • HW1 was prepared at a concentration of 50nM, AU11 and AU12 at 25nM, respectively, and injected at the same time.
  • the surface of the chip was regenerated with 1M NaCl / 20mM NaOH, BIA evaluation ver. Using the 3.2 software to obtain the affinity to the movement rate constants (k on and k off) and the equilibrium dissociation constant (K D).
  • AU11 and AU12 showed affinity about 17-22 times higher than HW1 (K D : about 200nM).
  • simultaneous analysis of HW1, AU11, and AU12 at the same concentration showed that AU11 and AU12 bind more strongly in the dissociation interval than HW1.
  • HW1 The stability of AU11 and AU12 against HW1 was measured using a fluorescence spectrometer (JASCO, Japan).
  • HW1, AU11 scFv, and AU12 scFv were prepared by expression and purification in E. coli, and the prepared proteins were dissociated into guanidine-HCl (guanidine-HCl) in 0.3M units from 0M to 4.2M, respectively, to give a final concentration of 10 ⁇ g / ml.
  • an excitation wave length of 280 nm, an emission wave length of 290 to 400 nm, and a measurement rate of 250 nm / min were repeated three times.
  • the relative maximum wavelength (normalized l max ) was plotted by normalizing the wavelength at the highest value for each guanidine-HCl concentration.
  • HW1 has a median value at a concentration of about 1.64M guanidine-HCl
  • AU1 and AU12 have a median value at a concentration of about 2.53M and 2.45M, respectively.
  • Ewert S et al., Biochemistry , 42: 1517-1528, 2003 the relative maximum wavelength increases as the protein is denatured by a protein denaturation sample such as guanidine-HCl.
  • the biotin-labeled HW1 was immobilized at 30 ⁇ g / ml, the concentration of AU11 or AU12 was 5 to 300 ⁇ g / ml, mixed and added to each well, followed by incubation at 37 ° C for 1 hour, and the plate was After washing, an anti-biotin antibody conjugated with alkaline phosphatase (anti-biotin mAb AP conjugated, Sigma) was added and reacted at 37 ° C. for 1 hour. After washing three times, 50 ⁇ l of pNPP (Sigma.) was added to the substrate, and the absorbance was measured at 405 nm.
  • ELISA was performed to confirm the cross-reactivity of AU11 and AU12 against DR5, the target antigen, and DR4, DcR1, DcR2, the other antigens.
  • antigens namely DR5, DR4, DcR1 and DcR2
  • DR5 antigens
  • DcR1 antigens
  • DcR2 antigens
  • the anti-DR5 antibody of the present invention is an antibody that specifically binds to the DR5 antigen.
  • CRD2 cysteine-rich domains
  • CRD3 cysteine-rich domains
  • TRAIL showed a stronger binding force to CRD2 than CRD3, but HW1, AU11 and AU12 bound to each antigen with similar binding force for both CRD2 and CRD3. Therefore, it can be seen that the degree of effect of CRD2 and CRD3 on TRAIL or antibodies differs in the binding between TRAIL-DR5 and the binding between HW1, AU11, and AU12-DR5.
  • HCT116 As cancer cells, HCT116, a human colon cancer cell line, and U87MG, a human glioma cancer cell line, were used.
  • the cell lines stored in liquid nitrogen were taken out, dissolved rapidly at 37 ° C., and centrifuged to remove the cryopreservation medium.
  • the cells obtained from the culture medium were cultured with DMEM medium containing 10% FBS, 100unit / ml penicillin and 100 ⁇ g / ml streptomycin. (Welgene) was used to cultivate the culture flask.
  • TE buffer was treated for 3 to 5 minutes, and then the reaction of TE buffer was stopped with 5 ml of DMEM medium containing 10% FBS, followed by centrifugation to recover the cells.
  • the recovered cells were each resuspended in the same culture medium, and the cells per well were dispensed in 96-well plates by 1 ⁇ 10 4 (100 ⁇ l), incubated for 24 hours, and used for MTT analysis.
  • Human colon cancer cell line HCT116 and human neural tumor cell line U87MG to a 96-well plate at 1 ⁇ 10 4 gae added and for 10% FBS DMEM medium is put 100 ⁇ l is added 5% CO 2, at 37 °C 2 il Culture was stabilized.
  • 10 ⁇ M of Z-VAD Pan-caspase inhibitor, Santacruz
  • 10 ⁇ M of SP600125 JNK inhibitor, calbiochem
  • 10 ⁇ M of SB203580 p38 inhibitor, calbiochem
  • 10 ⁇ M of 3-MA 100 Autophagic cell death inhibitor, sigma
  • Chloroguine Autophagic cell death inhibitor (sigma) 10 ⁇ M was added and pretreated for 1 hour.
  • AU11 and AU12 were added to 30 ⁇ g / ml, respectively, and then cultured at 5% CO 2 and 37 ° C for 40 hours to measure the degree of cell death through MTT-assay.
  • AU11 and AU12 significantly reduced apoptosis in the presence of SP600125 (JNK inhibitor), 3-MA and Chloroquine in human colon cancer cell line HCT116 and human neuronal tumor cell line U87MG, Z-VAD
  • SP600125 JNK inhibitor
  • 3-MA Chloroquine
  • HCT116 human colon cancer cell line
  • human neuronal tumor cell line U87MG Z-VAD
  • SB203580 p38 inhibitor
  • a typical feature of apoptosis by apoptosis is the accumulation of intracellular LC3 proteins in or on the vacuole membrane.
  • U87MG cancer cell lines TRAIL-resistant, human glioma cell lines
  • LC3-GFP expressing LC3-GFP
  • AU11 and AU12 were added to 30 ⁇ g / ml, respectively, and incubated at 5% CO 2 , 37 ° C for 30 hours, and observed with a fluorescence microscope (Axiovert 200M, Carl Zeiss, Germany).
  • the above ingredients were mixed and filled in an airtight cloth to prepare a powder.
  • a tablet was prepared by a direct tableting method.
  • the powder was prepared by mixing the above components, the powder was filled in a hard capsule according to a conventional method for preparing a capsule to prepare a capsule.
  • the amount of the above-mentioned ingredient was prepared per ampoule (2 ml).
  • Each component was added to and dissolved in purified water according to the conventional method for preparing a liquid, and lemon flavor was added appropriately, followed by mixing the above components. Then, purified water was added thereto to adjust the total volume to 100 ml, and filled into a brown bottle and sterilized to prepare a liquid.
  • the anti-DR5 antibody according to the present invention has a higher affinity than a conventional anti-DR5 antibody, specifically binds to a DR5 protein, has a relatively high stability, and is a TRAIL-sensitive cancer cell expressing DR5 or a TRAIL-resistant cancer cell expressing DR5.
  • a conventional anti-DR5 antibody specifically binds to a DR5 protein
  • has a relatively high stability and is a TRAIL-sensitive cancer cell expressing DR5 or a TRAIL-resistant cancer cell expressing DR5.
  • the anti-DR5 antibody according to the present invention can be expected to reduce costs and improve efficiency by reducing the single dose.
  • acagtaatac acagccgtgt cctcggctct caggctgttc atttgcagat acagggtgtt 60

Abstract

The present invention relates to an anti-DR5 antibody with improved affinity and stability that specifically binds to the death receptor 5 (DR5) to destroy cancer cells effectively and the composition for cancer prevention or treatment including the same. The anti-DR5 antibody of the present invention causes mutageneses on the amino acid residues of VH-CDR2 and VH-CDR3 within the heavy chain variable region and the amino acid residue of VL-CDR3 within the light chain variable region of HW1 which is the known anti-DR5 antibody, and replaces the framework within the heavy chain variable region from a VH6 subtype to a VH3 subtype and replaces the framework within the light chain variable region with the framework part of Vk1 or Vk3 so that affinity and stability are improved over the known anti-DR5 antibody, HW1. In addition, the anti-DR5 antibody of the present invention effectively induces cell death with respect to TRAIL-sensitive cancer cells that express DR5 and TRAIL-resistant cancer cells that express DR5 by autophagy so that it may be usefully employed in preventing or treating cancers caused by DR5 expression.

Description

친화도와 안정성이 향상된 항 DR5 항체, 및 이를 포함하는 암 예방 또는 치료용 조성물Anti-DR5 antibody with improved affinity and stability, and a composition for preventing or treating cancer comprising the same
본 발명은 세포사멸 수용체 5(death receptor 5; 이하 "DR5"라고 칭함)에 특이적으로 결합하여 다양한 암세포를 효과적으로 사멸하는 친화도와 안정성이 향상된 항 DR5 항체, 및 이를 포함하는 암 예방 또는 치료용 조성물에 관한 것이다.The present invention specifically binds to a death receptor 5 (hereinafter referred to as "DR5"), anti-DR5 antibody having improved affinity and stability for effectively killing various cancer cells, and a composition for preventing or treating cancer comprising the same. It is about.
단백질 p53-비의존적인(p53-independent) 종양괴사인자 수용체(tumor necrosis factor receptor; TNFR)를 통한 아폽토시스 경로 중, TNF-관련 아폽토시스 유도 리간드(TNF-related apoptosis inducing ligand; TRAIL)에 의해서 활성화되는 세포사멸 수용체 5(DR5)의 경로를 통한 세포사멸 기작은 정상세포에는 부작용이 적으면서 암세포에는 특이적으로 세포사멸을 유도하므로 암 치료제 개발의 중요 표적으로 여겨져 왔다(Ashkenazi 등 J. Clin. Invest., 104:155-162, 1999; 및 Ashkenazi 등, Nat. Rev. Cancer, 2:420-430, 2002).Cell death activated by TNF-related apoptosis inducing ligand (TRAIL) in the apoptosis pathway via protein p53-independent tumor necrosis factor receptor (TNFR). The apoptosis mechanism through the receptor 5 (DR5) pathway has been regarded as an important target for cancer drug development because it induces apoptosis specifically in cancer cells while having fewer side effects in normal cells (Ashkenazi et al. J. Clin. Invest ., 104 : 155-162, 1999; and Ashkenazi et al . , Nat. Rev. Cancer , 2: 420-430, 2002).
현재 DR5를 표적으로 암세포 특이적인 치료제를 개발하려는 연구로는, 상기 세포사멸 수용체들의 리간드인 재조합 TRAIL(예를 들면, TRAIL의 114-281번 아미노산 잔기)을 이용하는 방법과 세포사멸 수용체들에 특이적인 쥐 또는 인간 유래의 완전 항체(예를 들면, mAb 또는 IgG) 중에서 아고니스트(agonist) 항체를 개발하는 방법이 있다(Pollack 등, Clin. Cancer Res., 7:1362-1369, 2001; Jo 등, Nat. Med., 6:564-567, 2000; Ichikawa 등, Nat. Med., 7:954-960, 2001; 및 Walczak 등, Nat. Med., 5:157-161, 1999). 그러나, 재조합 TRAIL은 매우 불안정하여 가용성 올리고머(soluble oligomer)를 형성하므로 아폽토시스 활성이 20 내지 100배 정도 감소되고, 정상세포인 성상세포(astrocytes), 간세포(hepatocytes), 각질세포 (keratinocytes) 등에도 세포독성과 면역작용을 일으켜 부작용이 크다(Jo 등 Nat. Med., 6:564-567, 2000). 또한, TRAIL은 50% 이상의 악성 종양세포에는 세포사멸을 유도하지 못한다(Zhang 등, Cancer Gene Ther., 12:228-237, 2005). 일반적으로 TRAIL에 의해서 죽는 암세포들을 TRAIL-민감성(TRAIL-sensitive) 암세포라 하고, TRAIL에 의해서 죽지 않는 암세포들을 TRAIL-저항성(TRAIL-resistant) 암세포라고 한다.Research into the development of cancer cell-specific therapeutics targeting DR5 includes the use of recombinant TRAIL (eg, amino acid residues 114-281 of TRAIL), ligands of the apoptosis receptors, and specific apoptosis receptors. There is a method for developing an agonist antibody in a mouse or human complete antibody (eg mAb or IgG) (Pollack et al., Clin. Cancer Res., 7: 1362-1369, 2001; Jo et al., Nat. Med., 6: 564-567, 2000; Ichikawa et al., Nat. Med., 7: 954-960, 2001; and Walczak et al., Nat. Med., 5: 157-161, 1999). However, recombinant TRAIL is highly unstable and forms a soluble oligomer, so that apoptosis activity is reduced by 20 to 100 times, and cells such as astrocytes, hepatocytes, and keratinocytes are normal cells. Toxic and immune, causing side effects (Jo et al . Nat. Med. , 6: 564-567, 2000). TRAIL also does not induce apoptosis in more than 50% of malignant tumor cells (Zhang et al., Cancer Gene Ther., 12: 228-237, 2005). In general, cancer cells killed by TRAIL are called TRAIL-sensitive cancer cells, and cancer cells that do not die by TRAIL are called TRAIL-resistant cancer cells.
현재까지 DR5와 관련된 세포사멸을 유도하기 위해 개발된 DR5에 특이적인 친화도를 갖는 항체로는, 쥐 유래 단일클론 항체로부터 개발된 인간화 항체인 TRA-8 (쥐 유래 IgG) (Walczak 등, Nat. Med., 5:157-161) 및 AD5-10(쥐 IgG) (Guo 등, J. Biol. Chem., 280:41940-41952, 2005), 인간 유래 단일 클론 항체인 HGS-ETR2 (인간IgG1) (Georgakis 등, Br. J. Haematol., 130:501-510, 2005) 및 KMTR2 (인간 IgG4) (Motoki 등, Clin. Cancer Res., 11:3126-3135, 2005) 등이 있다.Antibodies having affinity specific for DR5, which have been developed to induce apoptosis associated with DR5 to date, include TRA-8 (mouse-derived IgG), a humanized antibody developed from a mouse-derived monoclonal antibody (Walczak et al., Nat. Med., 5: 157-161) and AD5-10 (rat IgG) (Guo et al., J. Biol. Chem., 280: 41940-41952, 2005), HGS-ETR2 (human IgG1), a human-derived monoclonal antibody (Georgakis et al., Br. J. Haematol. , 130: 501-510, 2005) and KMTR2 (human IgG4) (Motoki et al. , Clin. Cancer Res. , 11: 3126-3135, 2005).
상기 항체들은 모두 TRAIL-민감성 암세포들에서만 세포사멸을 유도하고, TRAIL-저항성 암세포에서는 세포사멸을 유도하지 못하였다. 또한, 단일항원 결합부위(monovalent)를 갖는 Fab 또는 scFv 형태로는 암세포에서 세포사멸을 유도하지 못하였고(예: KMTR2), 이중 항원 결합부위(divalent)를 갖는 IgG 형태로만 세포독성을 보이거나(예: HGS-ETR2 및 AD5-10) IgG를 가교제(cross-linker)로 사용해야만 세포사멸을 유도할 수 있었다(Chuntharapai 등, J. Immunol., 166:4891-4898, 2001; Motoki 등, Clin. Cancer Res., 11:3126-3135, 2005; 및 Wajant 등, Oncogene, 20:4101-4106, 2001).All of these antibodies induced apoptosis only in TRAIL-sensitive cancer cells and did not induce apoptosis in TRAIL-resistant cancer cells. In addition, Fab or scFv forms with a monoantigen binding site (monovalent) did not induce apoptosis in cancer cells (eg KMTR2), and showed cytotoxicity only with IgG forms with a double antigen binding site (divalent) ( Examples: HGS-ETR2 and AD5-10) Apoptosis could only be induced by using IgG as a cross-linker (Chuntharapai et al., J. Immunol., 166: 4891-4898, 2001; Motoki et al., Clin. Cancer Res., 11: 3126-3135, 2005; and Wajant et al., Oncogene, 20: 4101-4106, 2001).
따라서, 기존의 리간드인 TRAIL을 대신하여 DR5에 특이적인 친화도를 갖는 항체를 개발하여 세포사멸을 유도하는 방법에 관하여 많은 연구가 진행되고 있다 (Walczak 등, Nat. Med., 5:157-161, 1999; 및 Guo 등, J. Biol. Chem., 280:41940-41952, 2005; 및 Georgakis 등, Br. J. Haematol., 130:501-510, 2005). 상기 연구의 일 예로, DR5에 대한 TRAIL을 대체하여 기존에 개발된 항체들과는 다르게 TRAIL-민감성 세포주는 물론 TRAIL-저항성 세포주도 자식작용(autophagy)에 의한 세포사멸 경로를 통하여 정상세포에 독성없이 효과적으로 세포사멸을 유도하는 항체를 개발하였고(Park 등, Cancer Research, 1;67(15):7237-34, 2007; 대한민국 등록특허 제 10-0847010호에), 이 항체는 다양한 암의 예방 및 치료에 응용될 수 있다.Therefore, many studies have been conducted on the development of an antibody having affinity specific for DR5 in place of the existing ligand TRAIL and inducing apoptosis (Walczak et al . , Nat. Med. , 5: 157-161). , 1999; and Guo et al. , J. Biol. Chem. , 280: 41940-41952, 2005; and Georgakis et al. , Br. J. Haematol. , 130: 501-510, 2005). As an example of this study, unlike antibodies previously developed by substituting TRAIL for DR5, TRAIL-sensitive cell lines as well as TRAIL-resistant cell lines are effectively toxic to normal cells through apoptosis pathway by autophagy. An antibody that induces death has been developed (Park et al., Cancer Research , 1; 67 (15): 7237-34, 2007; in Korean Patent No. 10-0847010), and the antibody is applied to the prevention and treatment of various cancers. Can be.
진핵세포의 세포사멸은 3가지 기작, 즉 형태학적 및 생화학적 특징에 따라 크게 자가사멸(apoptosis), 자식작용(autophagy), 괴사(necrosis)로 나타난다 (Kromer G 등 (2005), Cell Death Differ. 12 Suppl 2:1463-1467). 이 중 자식작용에 의한 세포사멸은 2형 세포사멸(type Ⅱ programmed cell death)로 분류되고, 영양결핍(nutrient starvation)이나 환경적 스트레스, 또는 다양한 화합물들에 의해 증가되며, 그 과정은 소포체(endoplasmic reticulum, ER)에서 유래되거나 새롭게 합성되는 이중막 구조가 만들어지고, 여기에 세포 내 소기관(미토콘드리아 등)이나 세포질 내 단백질들을 고립시키면서 오토파고좀(autophagosome)을 형성하여, 궁극적으로 리소좀(lysosome)과 융합하여 파괴시킨다(Kondo Y 등 (2005). Nat. Rev. Cancer. 5:726-734). 자식작용이 일정수준 이상이면 세포사멸을 일으키며, 이러한 자식작용에 의한 세포사멸을 이용한 암치료제 개발이 현재 관심을 받고 있다 (Martinet W 등 (2009), Clinical Science. 116:697-712).Apoptosis of eukaryotic cells is characterized by three mechanisms, namely apoptosis, autophagy and necrosis, depending on morphological and biochemical characteristics (Kromer G et al. (2005), Cell Death Differ. 12 Suppl 2: 1463-1467). Among these, apoptosis is classified as type II programmed cell death and is increased by nutrient starvation, environmental stress, or various compounds, and the process is called endoplasmic. reticulum (ER) or a newly synthesized double-membrane structure is created, which forms autophagosomes while isolating intracellular organelles (such as mitochondria) or cytoplasmic proteins, ultimately lysosomes and Fusion and destruction (Kondo Y et al. (2005). Nat. Rev. Cancer. 5: 726-734). If the child action is above a certain level, it causes apoptosis, and the development of cancer treatments using such apoptosis is attracting attention (Martinet W et al. (2009), Clinical Science. 116: 697-712).
항체의 가변부위 중 V 단편은 항체 서열의 대부분을 차지하는 단편으로서 다양한 아류형(subtype)이 존재하며, 중쇄가변영역(VH)의 경우 VH3가 가장 안정한 아류형이고, VH1a, VH1b 및 VH5가 중간적 위치이며, VH2, VH4 및 VH6는 안정성 면에서 떨어진다. 경쇄가변영역(VL)의 경우 Vk3 및 Vk1이 가장 안정한 형태로 알려져 있다. 따라서, 각 아류형에 대한 열역학적 안정성 및 항체 생산수율 등에 대한 연구가 선행되어져 왔다(Ewert S 등, J. Mol. Biol., 325:531-553, 2003; Ewert S 등, Biochemistry, 42:1517-1528, 2003). 이들 연구 중 CDR 이식(CDR grafting)법은, 안정성이 떨어지는 항체의 CDR을 상대적으로 열역학적 안정성 및 항체 생산수율이 높은 아류형에 이식하여 항체의 열역학적 안정성을 높인 연구 결과를 나타내어 주목할 만 하다(Jung 등, Protein. Eng., 10:959-966, 1997).The V fragment of the variable region of the antibody is a fragment that occupies most of the antibody sequence, and there are various subtypes. In the heavy chain variable region (VH), VH3 is the most stable subtype, and VH1a, VH1b, and VH5 are intermediate. Position, and VH2, VH4 and VH6 fall in terms of stability. In the light chain variable region (VL), Vk3 and Vk1 are known to be the most stable forms. Thus, studies on thermodynamic stability and antibody production yield for each subtype have been preceded (Ewert S et al., J. Mol. Biol., 325: 531-553, 2003; Ewert S et al., Biochemistry, 42: 1517- 1528, 2003). Among these studies, the CDR grafting method is remarkable, as it shows the results of the studies that improved the thermodynamic stability of antibodies by grafting CDRs of less stable antibodies to subtypes having relatively higher thermodynamic stability and yield of antibody production (Jung et al. , Protein.Eng . , 10: 959-966, 1997).
또한, 치료용 항체의 친화도 향상은 일회 투여량의 감소효과를 기대할 수 있어, 비용 절감 및 효능 증대, 부작용 감소 등의 종합적인 치료 효율성 증가로 이어질 수 있다. 그러나, 아고니스트(agonist)에 있어서 리간드-수용체 결합친화도의 증가가 반드시 생물학적 활성과 연계되지는 않는다(Jones 등, Trends. Biotechnol., 26:498-505, 2008). 이의 일 예로, 야생형(wild-type) 인간 성장 호르몬(human growth hormone)에 비하여 수용체에 대한 친화도가 약 400배 증가한 변이체를 제작하였으나 정작 세포 성장에 있어서 특별히 향상된 점이 없는 경우 (Pearce 등, Biochemistry, 38:81-89, 1999), 또는 야생형에 비하여 외피성장인자 수용체(epidermal growth factor receptor, EGFR)에 결합력이 강한 외피성장인자 (epidermal growth factor, EGF)를 개발하였으나 오히려 길항능(antagonistic activity)을 보이는 경우(Coco 등, Nat. Biotechnol., 20:1246-1250, 2002) 등이 보고되었다.In addition, the improvement of the affinity of the therapeutic antibody can be expected to reduce the single dose, it can lead to a comprehensive treatment efficiency, such as cost reduction and increased efficacy, reduced side effects. However, an increase in ligand-receptor binding affinity in agonists is not necessarily associated with biological activity (Jones et al . , Trends. Biotechnol. , 26: 498-505, 2008). As an example of this, when a variant was prepared in which the affinity to the receptor was increased about 400 times compared to wild-type human growth hormone, but there was no improvement in cell growth (Pearce et al., Biochemistry , 38: 81-89, 1999), or developed an epidermal growth factor (EGF) that binds to the epidermal growth factor receptor (EGFR) in comparison to the wild type, but exhibits an antagonistic activity. Visible cases (Coco et al . , Nat. Biotechnol. , 20: 1246-1250, 2002).
따라서, 항체의 친화도를 향상시키기 위하여 다양한 방법이 연구되어 왔다. 구체적으로는, 1) 우선 전체 항체 서열에 대하여 무작위적으로 변이를 주어 친화도가 향상된 변이체를 선별하는 방법(Boder ET 등, Proc Natl Acad Sci USA, 26;97(20):10701-5, 2000), 2) 항체의 구조 및 선행 연구 성과를 바탕으로 친화도 향상에 영향을 줄 것이라 예상되는 특정 아미노산을 선택하여 변이체를 제작하는 방법(Barderas R 등, Proc Natl Acad Sci U S A, 105(26):9029-34, 2008), 및 3) 항원과의 특이적인 결합에 영향을 주는 부위인 상보성 결정 부위(Complementarity Determining Regions, CDRs) 만을 변이시켜 선별하는 방법(Garcia-Rodriguez C 등, Nat Biotechnol, 25(1):107-16, 2007) 등이 있고, 각각의 방법들은 독립적으로 뿐만 아니라 서로 복합적으로 사용되어 친화도 향상에 있어서 더 좋은 효과를 나타낼 수 있다.Therefore, various methods have been studied to improve the affinity of the antibody. Specifically, 1) a method of first selecting a variant having improved affinity by randomly changing the whole antibody sequence (Boder ET et al., Proc Natl Acad Sci USA , 26; 97 (20): 10701-5, 2000 ), 2) methods of constructing variants by selecting specific amino acids that are expected to affect affinity enhancement based on the structure of the antibody and previous studies (Barderas R et al., Proc Natl Acad Sci USA, 105 (26): 9029-34, 2008), and 3) a method of mutating and selecting only Complementarity Determining Regions (CDRs), which are sites affecting specific binding to antigens (Garcia-Rodriguez C et al., Nat Biotechnol , 25 ( 1): 107-16, 2007), and each method can be used independently or in combination with each other to have a better effect on improving affinity.
본 발명자들은 기존의 항 DR5 항체보다 친화도와 안정성이 더 향상된 항 DR5 항체를 개발하기 위하여 연구하던 중, 기존의 항 DR5 인간 항체인 HW1의 변이체를 제작하였으며, 상기 제작한 HW1의 변이체의 친화도 및 안정성이 기존의 HW1보다 더 향상되고 DR5를 발현하는 TRAIL-민감성 암세포 또는 DR5를 발현하는 TRAIL-저항성 암세포에 대해 자식작용에 의하여 세포사멸을 효과적으로 유도함을 확인하고, 본 발명을 완성하였다.The inventors of the present invention, while studying to develop an anti-DR5 antibody with improved affinity and stability than the existing anti-DR5 antibody, and produced a variant of the existing anti-DR5 human antibody, HW1, and the affinity and The stability was improved more than the existing HW1 and confirmed that effectively induce apoptosis by the progeny action on TRAIL-sensitive cancer cells expressing DR5 or TRAIL-resistant cancer cells expressing DR5, and completed the present invention.
본 발명은 친화도와 안정성이 향상된 항 DR5 항체를 제공하고자 한다.The present invention is to provide an anti-DR5 antibody with improved affinity and stability.
또한, 본 발명은 상기 항 DR5 항체를 코딩하는 DNA를 제공하고자 한다.In addition, the present invention is to provide a DNA encoding the anti-DR5 antibody.
또한, 본 발명은 상기 DNA 또는 이를 포함하는 발현벡터로 형질전환된 세포를 제공하고자 한다.In addition, the present invention is to provide a cell transformed with the DNA or an expression vector comprising the same.
또한, 본 발명은 상기 항 DR5 항체를 유효성분으로 함유하는 암 예방 또는 치료용 조성물을 제공하고자 한다.In addition, the present invention is to provide a composition for preventing or treating cancer containing the anti-DR5 antibody as an active ingredient.
도 1은 항-DR5 인간 항체인 HW1의 친화도 향상을 위한 VH-CDR2, VH-CDR3, VL-CDR3의 프라이머를 설계한 도이다.FIG. 1 is a diagram illustrating primers of VH-CDR2, VH-CDR3, and VL-CDR3 for improving affinity of HW1, an anti-DR5 human antibody.
도 2는 항-DR5 인간 항체인 HW1의 친화도 향상을 위한 변이체 라이브러리의 제작 과정을 나타낸 도이다.Figure 2 is a diagram showing the manufacturing process of a variant library for improving the affinity of the anti-DR5 human antibody HW1.
도 3은 HW1의 변이체 라이브러리로부터 FACS를 이용하여 친화도가 높은 변이체를 선별하는 과정을 분석한 도이다.3 is a diagram analyzing the process of selecting a high affinity variant using the FACS from the variant library of HW1.
도 4는 HW1 및 HW1의 변이체 라이브러리들로부터 최종 선별된 변이체들을 다시 FACS를 이용하여 친화도를 분석한 도이다.4 is a diagram analyzing the affinity of the finally selected variants from the variants library of HW1 and HW1 again using FACS.
도 5는 항체 AU11의 전체 뉴클레오티드 및 아미노산 서열을 도식화하여 나타낸 도이다(여기에서, 밑줄로 표시한 부분은 차례로 VH-CDR1, VH-CDR2, VH-CDR3, 링커, VL-CDR1, VL-CDR2, VL-CDR3를 나타낸 것임).5 is a diagram showing the entire nucleotide and amino acid sequence of the antibody AU11 (where the underlined portions are in turn VH-CDR1, VH-CDR2, VH-CDR3, linker, VL-CDR1, VL-CDR2, VL-CDR3).
도 6은 기존 항체인 HW1과 최종적으로 친화도 및 안정성이 향상된 AU11의 아미노산 서열을 비교 분석한 결과를 나타낸 도이다(*는 CDR에서 친화도 향상에 따라 변이된 아미노산 잔기를 표시한 것임).Figure 6 is a view showing a result of comparing the amino acid sequence of the AU11 finally improved the affinity and stability of the existing antibody HW1 (* indicates amino acid residues are changed according to affinity improvement in CDR).
도 7은 항체 AU12의 전체 뉴클레오티드 및 아미노산 서열을 도식화하여 나타낸 도이다(여기에서, 밑줄로 표시한 부분은 차례로 VH-CDR1, VH-CDR2, VH-CDR3, 링커, VL-CDR1, VL-CDR2, VL-CDR3를 나타낸 것임).7 is a diagram showing the entire nucleotide and amino acid sequence of the antibody AU12 (where the underlined portions are in turn VH-CDR1, VH-CDR2, VH-CDR3, linker, VL-CDR1, VL-CDR2, VL-CDR3).
도 8은 기존 항체인 HW1과 최종적으로 친화도 및 안정성이 향상된 AU11 및 AU12의 아미노산 서열을 비교 분석한 결과를 나타낸 도이다.FIG. 8 is a diagram showing the results of comparative analysis of the amino acid sequences of AU11 and AU12 which finally improved the affinity and stability of the existing antibody HW1.
도 9는 정제된 항체(AU11 및 AU12)의 크기 및 순도를 환원성 SDS-PAGE 및 비환원성 SDS-PAGE로 분석한 결과를 나타낸 도이다.9 is a diagram showing the results of analyzing the size and purity of purified antibodies (AU11 and AU12) by reducing SDS-PAGE and non-reducing SDS-PAGE.
도 10은 정제된 항체(AU11 및 AU12)를 크기 배제 크로마토그래피로 분석한 결과를 나타낸 도이다.10 is a diagram showing the results of analyzing purified antibodies (AU11 and AU12) by size exclusion chromatography.
도 11은 DR5에 대한 AU11 및 AU12의 친화도를 SPR을 이용하여 정량한 결과 및 HW1과의 친화도를 비교한 결과를 나타낸 도이다.11 is a diagram showing the result of quantifying the affinity of AU11 and AU12 for DR5 using SPR and comparing the affinity with HW1.
도 12는 AU11 및 AU12의 안정성을 측정한 결과를 나타낸 도이다.12 shows the results of measuring the stability of AU11 and AU12.
도 13은 HW1과 AU11 또는 AU12간의 동일 항원결합부위 여부를 경쟁적 ELISA를 이용하여 측정한 결과를 나타낸 도이다.Figure 13 is a diagram showing the result of measuring the same antigen binding site between HW1 and AU11 or AU12 using a competitive ELISA.
도 14는 표적항원인 DR5 항원 및 다른 유사항원(DR4, DcR1, DcR2)에 대한 AU11 및 AU12의 교차반응성을 ELISA로 측정한 결과를 나타낸 도이다.14 is a diagram showing the results of measuring the cross-reactivity of AU11 and AU12 against the target antigen DR5 antigen and other analogues (DR4, DcR1, DcR2) by ELISA.
도 15는 전체 DR5와 DR5의 단편들인 CRD2(잔기 97~137) 및 CRD3(잔기 139~180), 그리고 그들에 대한 결합능을 가진 HW1, AU11, AU12, TRAIL 간의 결합 친화도를 ELISA를 이용하여 측정한 결과를 나타낸 도이다.FIG. 15 shows the binding affinity between total DR5 and DR5 fragments CRD2 (residues 97-137) and CRD3 (residues 139-180) and HW1, AU11, AU12, TRAIL having binding capacity to them using ELISA. Figure 1 shows the results.
도 16은 인간 대장암 세포주인 HCT116과 인간 신경종양 세포주인 U87MG에서 HW1, AU11 및 AU12의 세포사멸 정도를 MTT-assay를 통해 비교 평가한 결과를 나타낸 도이다.FIG. 16 is a diagram showing the results of comparing and evaluating the degree of apoptosis of HW1, AU11, and AU12 in HCT116, a human colorectal cancer cell line, and U87MG, a human neurotumor cell line, by MTT-assay.
도 17은 인간 대장암 세포주인 HCT116과 인간 신경종양 세포주인 U87MG에서 다양한 저해제(Z-VAD, SP600125, SB203580, 3-MA, Chloroguine) 존재 하에 HW1, AU11 및 AU12의 세포사멸 능력을 MTT-assay로 분석한 결과를 나타낸 도이다.17 shows MTT-assay of apoptosis ability of HW1, AU11 and AU12 in the presence of various inhibitors (Z-VAD, SP600125, SB203580, 3-MA, Chloroguine) in human colon cancer cell line HCT116 and human neurotumor cell line U87MG. It is a figure which shows the result of analysis.
도 18은 LC3-GFP를 발현하는 U87MG 암세포주에 AU11 및 AU12를 처리하여 LC3-GFP 결집을 형광현미경으로 관찰한 결과를 나타낸 도이다.Fig. 18 shows the results of fluorescence microscopy observation of LC3-GFP aggregates by treatment of AU11 and AU12 to U87MG cancer cell lines expressing LC3-GFP.
본 발명은 서열번호 7 또는 39의 아미노산 서열을 갖는 것을 특징으로 하는 DR5에 특이적으로 결합하는 항 DR5 항체를 제공한다.The present invention provides an anti-DR5 antibody that specifically binds to DR5, which has the amino acid sequence of SEQ ID NO: 7 or 39.
또한, 본 발명은 상기 항 DR5 항체를 코딩하는 DNA를 제공한다.The present invention also provides a DNA encoding the anti-DR5 antibody.
또한, 본 발명은 상기 DNA 또는 이를 포함하는 발현벡터로 형질전환된 세포를 제공한다.The present invention also provides a cell transformed with the DNA or an expression vector comprising the same.
또한, 본 발명은 상기 항 DR5 항체를 유효성분으로 함유하는 암 예방 또는 치료용 조성물을 제공한다.The present invention also provides a composition for preventing or treating cancer containing the anti-DR5 antibody as an active ingredient.
이하, 본 발명에 대해 상세히 설명한다.Hereinafter, the present invention will be described in detail.
본 발명에 따른 항 DR5 항체는, 항-DR5 인간 항체인 HW1(대한민국 등록특허 제 10-0847010호)의 중쇄가변영역(heavy chain variable region) 내의 VH-CDR2, VH-CDR3의 아미노산 잔기와 경쇄가변영역(light chain variable region) 내의 VL-CDR3의 아미노산 잔기를 돌연변이화하여 DR5에 대한 친화도를 향상시키고, 더불어 중쇄가변영역 내의 구조(framework)를 VH6 아류형에서 VH3 아류형으로 치환하고, 경쇄가변영역 내의 구조를 Vk1 또는 Vk3의 구조 부분으로 치환하여 안정성을 향상시킨 것을 특징으로 한다.Anti-DR5 antibody according to the present invention, the amino acid residues and light chain variable VH-CDR2, VH-CDR3 in the heavy chain variable region (heavy chain variable region) of the anti-DR5 human antibody HW1 (Korean Patent No. 10-0847010) Mutating amino acid residues of VL-CDR3 in the light chain variable region to enhance affinity for DR5, as well as replacing the framework in the heavy chain variable region from the VH6 subtype to the VH3 subtype, and light chain variable It is characterized by improving the stability by substituting the structure in the region with the structural part of Vk1 or Vk3.
본 발명에서 "세포사멸 수용체-5 (DR5) 단백질"은 종양괴사인자(TNF) 수용체 패밀리의 일원이고 TRAIL에 결합하며, C-말단에 세포내 죽음 도메인(death domain)을 갖는 수용체를 의미한다(Pan 등, Science, 277:815-818, 1997). DR5가 TRAIL에 결합하는 경우 TRAIL-민감성 암세포에서 아폽토시스를 유도하고, DR5가 과발현되는 경우에는 아폽토시스가 증가하지만 정상세포에서는 아폽토시스를 유도하지 않는 것으로 알려져 있다. DR5는 상기한 바와 같은 특성을 갖는 단백질이면 어느 것이나 포함되며, 예를 들어 미국특허 제 6,872,568호에 기재된 아미노산 서열을 갖는 것일 수 있으나, 이에 한정되지 않는다.In the present invention, "apoptotic receptor-5 (DR5) protein" means a member of the tumor necrosis factor (TNF) receptor family and binds TRAIL and has an intracellular death domain at the C-terminus ( Pan et al., Science, 277: 815-818, 1997). When DR5 binds to TRAIL, it is known to induce apoptosis in TRAIL-sensitive cancer cells and to increase apoptosis when DR5 is overexpressed, but not to induce apoptosis in normal cells. DR5 includes any protein having the above characteristics, but may be, for example, having an amino acid sequence described in US Pat. No. 6,872,568, but is not limited thereto.
본 발명에서 "항체"는 전체 형태의 항체("전항체") 또는 그의 기능적인 단편일 수 있다. 상기 전항체는 단량체 또는 2 이상의 전항체가 결합되어 있는 다량체의 형태일 수 있다. 상기 항체의 기능적인 단편은 전항체의 중쇄 및 경쇄 가변영역을 갖는 항체로서, 실질적으로 전항체가 인식하는 것과 동일한 항원결합부위 (epitope)를 인식하는 것을 의미한다. 상기 항체의 기능적인 단편에는 단일쇄 가변영역 단편 (scFv), (scFv)2, Fab, Fab' 및 F(ab')2 등이 포함되나 이에 한정되지 않으며, 본 발명에서는 scFv가 바람직하다. 상기 단일쇄 가변영역(scFv)은 중쇄 가변영역과 경쇄 가변영역이 링커 펩타이드를 통하여 연결되어 단일쇄 폴리펩티드 형태를 취하는 항체 단편을 의미한다.In the present invention, an "antibody" may be an entire form of an antibody ("antibody") or a functional fragment thereof. The whole antibody may be in the form of a monomer or a multimer in which two or more whole antibodies are bound. The functional fragment of the antibody is an antibody having the heavy and light chain variable regions of the whole antibody, which means to recognize substantially the same epitope that the whole antibody recognizes. Functional fragments of the antibody include, but are not limited to, single chain variable region fragments (scFv), (scFv) 2 , Fab, Fab 'and F (ab') 2 and the like, and scFv is preferred in the present invention. The single chain variable region (scFv) refers to an antibody fragment in which a heavy chain variable region and a light chain variable region are linked through a linker peptide to take the form of a single chain polypeptide.
상기 항체는 당업계에 알려져 있는 방법, 예를 들어, 파지 디스플레이 방법 또는 효모 세포 표면 발현 시스템을 사용하여 생성될 수 있다.The antibody can be generated using methods known in the art, such as phage display methods or yeast cell surface expression systems.
scFv를 제조하는 방법으로는 미국특허 제 4,946,778호 및 제 5,258,498호에 기재된 방법이 사용될 수 있으며, Fab, Fab' 및 F(ab')2 단편을 재조합적으로 생성하기 위한 방법으로는 WO 92/22324 등에 기재된 방법이 사용될 수 있다.The method described in US Pat. Nos. 4,946,778 and 5,258,498 can be used as a method for preparing scFv, and WO 92/22324 is a method for recombinantly generating Fab, Fab 'and F (ab') 2 fragments. The method described in the above can be used.
본 발명의 항체는 인간을 포함하는 포유동물, 조류 등을 포함한 임의의 동물로부터 유래한 것일 수 있다. 바람직하게는, 상기 항체는 인간, 생쥐, 당나귀, 양, 토끼, 염소, 기니피그, 낙타, 말 또는 닭의 항체일 수 있다. 여기서 인간 항체는 인간 면역글로불린의 아미노산 서열을 가진 항체로서, 인간 면역글로불린 라이브러리로부터 분리된 항체 또는 하나 이상의 인간 면역글로불린에 대하여 형질 이식되고 내재적 면역글로불린은 발현하지 않는 동물로부터 분리된 항체가 포함된다(미국특허 제 5,939,598호 참조).Antibodies of the invention may be derived from any animal, including mammals, birds, and the like, including humans. Preferably, the antibody may be a human, mouse, donkey, sheep, rabbit, goat, guinea pig, camel, horse or chicken antibody. Wherein the human antibody is an antibody having an amino acid sequence of human immunoglobulin, which includes an antibody isolated from a human immunoglobulin library or an antibody isolated from an animal transfected against one or more human immunoglobulins and not expressing an endogenous immunoglobulin ( US Pat. No. 5,939,598).
본 발명의 항체는 효소, 형광 물질, 방사선 물질 및 단백질 등과 접합된 것일 수 있으나, 이에 한정되지 않는다. 또한, 항체에 상기 물질을 접합하는 방법은 당업계에 잘 알려져 있다.Antibodies of the present invention may be conjugated to enzymes, fluorescent materials, radioactive materials and proteins, but are not limited thereto. In addition, methods of conjugating such materials to antibodies are well known in the art.
본 발명에 따른 항 DR5 항체는, 상보성 결정부위 CDR1, CDR2 및 CDR3가 각각 서열번호 1, 2 및 3의 아미노산 서열을 갖는 중쇄 가변영역과 상보성 결정부위 CDR1, CDR2 및 CDR3가 각각 서열번호 4, 5 및 6의 아미노산 서열을 갖는 경쇄 가변영역을 포함하며, DR5에만 특이적으로 결합한다.Anti-DR5 antibody according to the present invention, the heavy chain variable region having complementary determining region CDR1, CDR2 and CDR3 having the amino acid sequence of SEQ ID NO: 1, 2 and 3 and the complementarity determining region CDR1, CDR2 and CDR3 are SEQ ID NO: 4, 5, respectively And a light chain variable region having the amino acid sequence of 6, specifically binding to DR5 only.
본 발명에서 "특이적으로 결합한다"는 본 발명의 항체가 DR5 항원에만 결합하고, DR5와 유사한 항원인 DR4(death receptor 4), DcR1(death decoy receptor 1) 및 DcR2에는 실질적으로 결합하지 않는다는 것을 의미한다.In the present invention, "specifically binds" means that the antibody of the present invention binds only to the DR5 antigen and does not substantially bind to DR5 (death receptor 4), death decoy receptor 1 (DcR1) and DcR2, which are DR5-like antigens. it means.
상기 경쇄 가변영역은 서열번호 27 또는 37의 아미노산 서열을 가지며, 상기 중쇄 가변영역은 서열번호 28 또는 38의 아미노산 서열을 갖는다.The light chain variable region has an amino acid sequence of SEQ ID NO: 27 or 37, and the heavy chain variable region has an amino acid sequence of SEQ ID NO: 28 or 38.
본 발명의 항 DR5 항체는 서열번호 7 또는 39의 아미노산 서열을 갖는 scFv 항체인 AU11 또는 AU12이며, 이들은 각각 중쇄 가변영역의 CDR1 내지 CDR3, 링커 올리고펩티드 및 경쇄 가변영역의 CDR1 내지 CDR3을 차례로 포함한다. 상기 항체는 단일클론항체가 바람직하다.The anti-DR5 antibodies of the present invention are AU11 or AU12, which are scFv antibodies having an amino acid sequence of SEQ ID NO: 7 or 39, which in turn comprise CDR1 to CDR3 of the heavy chain variable region, linker oligopeptide and CDR1 to CDR3 of the light chain variable region, respectively. . The antibody is preferably a monoclonal antibody.
본 발명의 항 DR5 항체인 AU11 또는 AU12는 HW1에 비하여 DR5에 더 높은 친화도로 결합하며, 평형해리상수(K D)는 각각 1.17×10-8 M 및 9.17×10-9 M 을 나타낸다. 또한, 상기 항 DR5 항체인 AU11 또는 AU12는 HW1에 비하여 그 안정성이 상대적으로 더 높게 나타나고, HW1과 마찬가지로 단일분자인 scFv 형태로 자식작용에 의하여 세포사멸을 유도한다.Anti-Dr5 antibodies of the present invention, AU11 or AU12, bind to DR5 with higher affinity than HW1, and the equilibrium dissociation constant ( K D ) shows 1.17 × 10 −8 M and 9.17 × 10 −9 M, respectively. In addition, the anti-Dr5 antibody AU11 or AU12 is relatively higher in stability than HW1, and similarly to HW1, a single molecule scFv form induces apoptosis by progeny.
본 발명은 항 DR5 항체인 AU11 또는 AU12를 코딩하는 DNA를 제공한다. 상기 DNA는 서열번호 7 또는 39의 아미노산 서열을 갖는 scFv를 코딩하는 DNA일 수 있으며, 바람직하게는 서열번호 8 또는 40의 염기서열을 갖는 DNA일 수 있다.The present invention provides DNA encoding AU11 or AU12, which is an anti-DR5 antibody. The DNA may be DNA encoding an scFv having an amino acid sequence of SEQ ID NO: 7 or 39, preferably DNA having a nucleotide sequence of SEQ ID NO: 8 or 40.
본 발명의 항체를 코딩하는 DNA 서열은 당업계에 잘 알려진 방법에 의하여 얻어질 수 있다. 예를 들면, 상기 항체의 중쇄 및 경쇄의 일부분 또는 전부를 코딩하는 DNA 서열 또는 해당 아미노산 서열에 근거하여, 당분야에 잘 알려진 올리고뉴클레오티드 합성기법, 예를 들어 부위 지향적 돌연변이 발생법(site-directed mutagenesis) 및 중합효소 연쇄 반응(PCR)법 등을 사용하여 원하는 대로 합성할 수 있다.DNA sequences encoding the antibodies of the invention can be obtained by methods well known in the art. For example, based on DNA sequences or corresponding amino acid sequences encoding portions or all of the heavy and light chains of the antibody, oligonucleotide synthesis techniques well known in the art, for example site-directed mutagenesis And the polymerase chain reaction (PCR) method.
또한, 본 발명은 상기 DNA 또는 이를 포함하는 발현벡터로 형질전환된 세포를 제공한다.The present invention also provides a cell transformed with the DNA or an expression vector comprising the same.
상기 DNA 또는 이를 포함하는 발현벡터는 당업계에 공지된 방법, 예를 들어 바이러스성 형질감염 또는 비-바이러스 기반 기법 등을 이용하여 적절한 숙주세포에 전달될 수 있다. 이때, DNA 또는 발현벡터의 도입은 아데노바이러스성 형질전환, 유전자 총, 리포좀-매개 형질전환 및 레트로바이러스 또는 렌티바이러스-매개 형질전환, 플라스미드, 아데노-부속 바이러스를 포함하나 이에 한정되지 않으며, 당업자에게 공지된 임의의 기법에 의해 수행될 수 있다. 또한, 상기 세포는 유전자를 장시간에 걸쳐 세포에 방출 또는 전달할 수 있는 적절한 담체 물질과 함께 이식될 수 있다.The DNA or expression vector comprising the same can be delivered to an appropriate host cell using methods known in the art, such as viral transfection or non-viral based techniques. At this time, the introduction of the DNA or expression vector includes, but is not limited to, adenovirus transformation, gene gun, liposome-mediated transformation and retrovirus or lentiviral-mediated transformation, plasmid, adeno-associated virus, and the like. It may be performed by any known technique. In addition, the cells can be transplanted with a suitable carrier material capable of releasing or delivering the gene to the cells for a long time.
상기 DNA 또는 이를 포함하는 발현벡터로 형질전환된 세포를 적절한 조건 하에서 배양하여 항체를 발현시키고 분리하여 항체 분자를 생산할 수 있다.The cells transformed with the DNA or the expression vector including the same may be cultured under appropriate conditions to express and isolate the antibody to produce antibody molecules.
상기 항체 분자는 세포의 세포질 내에 축적되거나, 세포로부터 분비되거나, 적절한 신호 서열에 의하여 페리플라즘 또는 세포외 배지(supernatant)로 표적화될 수 있으며, 이들 중 페리플라즘 또는 세포외 배지로 표적화되는 것이 바람직하다. 또한, 생산된 항체 분자를 당분야의 통상의 기술자에게 잘 알려져 있는 방법을 이용하여 리폴딩(refolding)시키고 기능적 형태(conformation)를 갖도록 하는 것이 바람직하다.The antibody molecule may accumulate in the cytoplasm of the cell, be secreted from the cell, or may be targeted to periplasm or extracellular medium by an appropriate signal sequence, among which is targeted to periplasm or extracellular medium. desirable. It is also desirable to refold the produced antibody molecules and to have functional conformation using methods well known to those skilled in the art.
상기 항체 분자의 중쇄 또는 경쇄 폴리펩티드만을 생산할 필요가 있는 경우에는, 중쇄 또는 경쇄 폴리펩티드를 코딩하는 서열을 포함하는 단일 벡터를 숙주세포에 형질도입시키고, 중쇄 및 경쇄 폴리펩티드 모두를 포함하는 항체를 생산하기 위해서는, 경쇄 폴리펩티드를 코딩하는 제 1벡터 및 중쇄 폴리펩티드를 코딩하는 제 2벡터의 2개 벡터를 숙주세포에 도입하거나, 경쇄 및 중쇄 폴리펩티드를 코딩하는 서열을 모두 포함하는 단일 벡터를 상기 숙주세포에 도입시킬 수도 있다.When only the heavy or light chain polypeptide of the antibody molecule needs to be produced, a single vector comprising a sequence encoding the heavy or light chain polypeptide is transduced into the host cell, and in order to produce an antibody including both the heavy and light chain polypeptides. To introduce into the host cell two vectors, a first vector encoding the light chain polypeptide and a second vector encoding the heavy chain polypeptide, or a single vector comprising both the light chain and heavy chain polypeptide sequences. It may be.
또한, 본 발명은 상기 항 DR5 항체를 유효성분으로 함유하는 암 예방 또는 치료용 조성물을 제공한다.The present invention also provides a composition for preventing or treating cancer containing the anti-DR5 antibody as an active ingredient.
본 발명에 따른 항 DR5 항체(AU11 및 AU12)는 기존의 항 DR5 항체(HW1)보다 더 높은 친화도로 DR5 항원에 특이적으로 결합하고 안정성이 상대적으로 높으며 DR5를 발현하는 TRAIL-민감성 암세포 또는 DR5를 발현하는 TRAIL-저항성 암세포에 대해 자식작용에 의하여 세포사멸을 효과적으로 유도함으로써, 일회 투여량의 감소에 의한 비용 절감 및 효율성 향상 등의 효과를 기대할 수 있다. 따라서, 본 발명의 항 DR5 항체는 DR5 발현에 의해 야기되는 암의 예방 또는 치료에 유용하게 사용될 수 있다.The anti-DR5 antibodies (AU11 and AU12) according to the present invention bind TRDR-sensitive cancer cells or DR5 that specifically bind to the DR5 antigen with a higher affinity than the existing anti-DR5 antibodies (HW1), and have relatively high stability and express DR5. By effectively inducing apoptosis by the progeny effect on the expressing TRAIL-resistant cancer cells, it is possible to expect the effect of reducing the cost and improving efficiency by reducing the single dose. Therefore, the anti-DR5 antibody of the present invention can be usefully used for the prevention or treatment of cancer caused by the expression of DR5.
상기 DR5 발현에 의해 야기되는 암은 TRAIL-민감성 암세포 및 TRAIL-저항성 암세포를 모두 포함할 수 있으며, 구체적으로는 혈액암, 폐암, 위암, 간암, 골암, 췌장암, 피부암, 두경부암, 피부 흑색종, 자궁암, 난소암, 직장암, 대장암, 결장암, 유방암, 자궁 육종, 나팔관 암종, 자궁내막 암종, 자궁경부 암종, 질 암종, 외음부 암종, 식도암, 후두암, 소장암, 갑상선암, 부갑상선암, 연조직의 육종, 요도암, 음경암, 전립선암, 만성 또는 급성 백혈병, 유년기의 고상 종양, 분화 림프종, 방광암, 신장암, 신장 세포 암종, 신장 골반 암종, 제 1 중추신경계 림프종, 척수축 종양, 뇌간 신경교종 또는 뇌하수체 아데노마 등을 포함하나, 이에 한정되지 않는다.Cancer caused by the DR5 expression may include both TRAIL-sensitive cancer cells and TRAIL-resistant cancer cells, specifically, hematologic cancer, lung cancer, stomach cancer, liver cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, skin melanoma, Uterine cancer, ovarian cancer, rectal cancer, colon cancer, colon cancer, breast cancer, uterine sarcoma, fallopian tube carcinoma, endometrial carcinoma, cervical carcinoma, vaginal carcinoma, vulvar carcinoma, esophageal cancer, laryngeal cancer, small intestine cancer, thyroid cancer, parathyroid cancer, soft tissue sarcoma, Urethral cancer, penile cancer, prostate cancer, chronic or acute leukemia, solid tumors of childhood, differentiated lymphoma, bladder cancer, kidney cancer, renal cell carcinoma, renal pelvic carcinoma, primary central nervous system lymphoma, spinal contraction tumor, brain stem glioma or pituitary gland Adenomas and the like, but is not limited thereto.
본 발명의 조성물은 항 DR5 항체와 함께 항암효과를 갖는 공지의 유효성분을 1종 이상 함유할 수 있다.The composition of the present invention may contain one or more known active ingredients having an anticancer effect together with the anti-DR5 antibody.
본 발명의 조성물은, 투여를 위해서 상기 기재한 유효성분 이외에 추가로 약학적으로 허용가능한 담체를 1종 이상 포함하여 제조할 수 있다. 약학적으로 허용가능한 담체는 식염수, 멸균수, 링거액, 완충 식염수, 덱스트로오스 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 1 성분 이상을 혼합하여 사용할 수 있으며, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. 또한 희석제, 분산제, 계면활성제, 결합제 및 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주사용 제형, 환약, 캡슐, 과립 또는 정제로 제제화할 수 있다. 더 나아가 당분야의 적정한 방법으로 또는 Remington's Pharmaceutical Science(최근판), Mack Publishing Company, Easton PA에 개시되어 있는 방법을 이용하여 각 질환에 따라 또는 성분에 따라 바람직하게 제제화할 수 있다.The composition of the present invention may be prepared by including one or more pharmaceutically acceptable carriers in addition to the above-described active ingredients for administration. Pharmaceutically acceptable carriers may be used in combination with saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol and one or more of these components, if necessary, as an antioxidant, buffer And other conventional additives such as bacteriostatic agents can be added. Diluents, dispersants, surfactants, binders and lubricants may also be added in addition to formulate into injectable formulations, pills, capsules, granules or tablets such as aqueous solutions, suspensions, emulsions and the like. Furthermore, it may be preferably formulated according to each disease or component by a suitable method in the art or using a method disclosed in Remington's Pharmaceutical Science (Recent Edition), Mack Publishing Company, Easton PA.
본 발명의 조성물은 목적하는 방법에 따라 경구 투여하거나 비경구 투여(예를 들어, 정맥 내, 피하, 복강 내 또는 국소에 적용)할 수 있으며, 투여량은 환자의 체중, 연령, 성별, 건강상태, 식이, 투여시간, 투여방법, 배설율 및 질환의 중증도 등에 따라 그 범위가 다양하다. 상기 항 DR5 항체의 일일 투여량은 약 0.01~50㎎/㎏, 바람직하게는 약 0.1~20㎎/㎏이며, 하루 일회 내지 수회에 나누어 투여하는 것이 더욱 바람직하다.The composition of the present invention can be administered orally or parenterally (eg, applied intravenously, subcutaneously, intraperitoneally or topically) according to the desired method, and the dosage is based on the weight, age, sex and health of the patient. The range varies depending on the diet, the time of administration, the method of administration, the rate of excretion and the severity of the disease. The daily dose of the anti-DR5 antibody is about 0.01 to 50 mg / kg, preferably about 0.1 to 20 mg / kg, and more preferably administered once to several times a day.
본 발명의 조성물은 암의 예방 및 치료를 위하여 단독으로, 또는 수술, 호르몬 치료, 약물 치료 및 생물학적 반응 조절제를 사용하는 방법들과 병용하여 사용할 수 있다.The composition of the present invention can be used alone or in combination with methods using surgery, hormonal therapy, drug therapy and biological response modifiers for the prevention and treatment of cancer.
이하, 본 발명의 이해를 돕기 위하여 바람직한 실시예를 제시한다. 그러나 하기의 실시예는 본 발명을 보다 쉽게 이해하기 위하여 제공되는 것일 뿐, 실시예에 의해 본 발명의 내용이 한정되는 것은 아니다.Hereinafter, preferred examples are provided to aid in understanding the present invention. However, the following examples are merely provided to more easily understand the present invention, and the contents of the present invention are not limited by the examples.
실시예 1Example 1 : HW1의 친화도 향상을 위한 변이체 라이브러리의 구축 : Construction of variant library for improving affinity of HW1
1. HW1의 변이체 라이브러리 구축을 위한 주형과 프라이머의 설계 및 제작1. Design and fabrication of template and primer for constructing variant library of HW1
항-DR5 인간 항체인 HW1(대한민국 등록특허 제 10-0847010호)의 친화도를 향상시키기 위하여 항체부위 중 항원과의 결합에 중요하다고 알려진 CDR (Complementarity Determining Region) 부위들, 그 중에서도 중쇄가변 부위의 CDR2를 우선적으로 하여 변이체(mutant) 라이브러리를 설계하였으며, 효모 표면에 변이체를 발현하기 위한 벡터는 효모표면 발현 벡터인 pCTCON을 사용하였고, 도 1과 같이 중쇄가변 부위의 CDR2에 무작위 염기서열을 부여하였다. 이때, 중쇄가변 CDR2를 구성하는 각 야생형(wild-type) 아미노산의 비율을 일정 수준 이상으로 보존하기 위하여, 일반적으로 사용되는 4개의 염기 비율이 각각 25%인 염기 혼합체 대신 야생형 염기 비율이 70%인 염기 혼합체를 사용하였다(예를 들어, 야생형 염기가 아데닌(A)인 부위에는 아데닌, 구아닌, 시토신, 티민이 각각 25%씩 함유된 염기 혼합체 대신 아데닌 70%, 구아닌, 시토신, 티민이 각 10%씩 함유된 염기 혼합체를 사용하여 변이체를 구축하였다.). 위와 같은 방법으로 최종적으로 각 아미노산의 보존율이 일정 수준 이상이 되도록 프라이머(단편1 - 서열번호 9, 서열번호 12; 단편2 - 서열번호 11, 서열번호 10)(제노텍, 한국)를 제작하였다. 이후, 중쇄가변 CDR2의 변이체 라이브러리에서 친화도가 향상된 변이체를 선별하였고, 선별된 항체를 주형으로 하여 다시 각각 중쇄가변 CDR3 및 경쇄가변 CDR3 부위에 위에서 기술한 방법과 동일하게 프라이머(중쇄가변 CDR3: 단편1 - 서열번호 9, 서열번호 14, 단편2 - 서열번호 13, 서열번호 10; 경쇄가변 CDR3: 단편1 - 서열번호 9, 서열번호 16, 단편2: 서열번호 15, 서열번호 10)를 디자인하여 추가적인 친화도 향상을 위한 두 개의 라이브러리를 구축하였다. 친화도 향상을 위한 변이체 구축에 사용된 프라이머 서열번호 및 서열은 하기 표 1에 나타내었다.In order to enhance the affinity of the anti-DR5 human antibody HW1 (Korean Patent No. 10-0847010), CDR (Complementarity Determining Region) sites, which are known to be important for binding to the antigen, among the heavy chain variable regions Mutant library was designed by giving priority to CDR2, and the vector for expressing the variant on the yeast surface was used pCTCON, a yeast surface expression vector, and random nucleotide sequences were assigned to CDR2 of the heavy chain variable region as shown in FIG. . In this case, in order to preserve the ratio of each wild-type amino acid constituting the heavy chain variable CDR2 to a predetermined level or more, the ratio of wild-type bases is 70% instead of the base mixtures of 25% each of the four bases generally used. A base mixture was used (for example, adenine 70% adenine, guanine, cytosine, thymine each 10% instead of the base mixture containing 25% adenine, guanine, cytosine and thymine at the site where the wild type base is adenine (A)). Variants were constructed using the base mixture contained thick). In the same manner as described above, a primer (fragment 1-SEQ ID NO: 9, SEQ ID NO: 12; fragment 2-SEQ ID NO: 11, SEQ ID NO: 10) (Gennotek, Korea) was finally prepared so that the retention rate of each amino acid was higher than a predetermined level. Subsequently, variants with improved affinity were selected from the mutant library of heavy chain variable CDR2, and the primers (heavy chain variable CDR3: fragment) were selected in the heavy chain variable CDR3 and light chain variable CDR3 sites, respectively, using the selected antibodies as templates. 1-SEQ ID NO: 9, SEQ ID NO: 14, fragment 2-SEQ ID NO: 13, SEQ ID NO: 10; light chain variable CDR3: fragment 1-SEQ ID NO: 9, SEQ ID NO: 16, fragment 2: SEQ ID NO: 15, SEQ ID NO: 10) Two libraries have been built to further enhance affinity. Primer sequence numbers and sequences used to construct variants for improving affinity are shown in Table 1 below.
[표 1]TABLE 1
Figure PCTKR2009006036-appb-I000001
Figure PCTKR2009006036-appb-I000001
※ 상기 서열 중 서열번호 11, 13, 15 내의 숫자는 다음을 의미한다(1: 70%A+10%G+10%C+10%T, 2: 10%A+70%G+10%C+10%T, 3: 10%A+10%G+70%C+10%T, 4: 10%A+10%G+10%C+70%T, 5: 33%C+33%G+33%T).※ The numbers in SEQ ID NOs: 11, 13, and 15 in the sequence mean the following (1: 70% A + 10% G + 10% C + 10% T, 2: 10% A + 70% G + 10% C + 10% T, 3: 10% A + 10% G + 70% C + 10% T, 4: 10% A + 10% G + 10% C + 70% T, 5: 33% C + 33% G + 33% T).
2. 항체 유전자 각 단편의 증폭 및 결합2. Amplification and binding of each fragment of antibody gene
항-DR5 항체의 중쇄가변 CDR2에 돌연변이를 도입하기 위하여, 도 2와 같이 2개의 각 단편을 제작하였다. 첫번째 단편은 정방향 프라이머로 5'-GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3'(서열번호 9), 역방향 프라이머로 5'-CCT TCC CAG CCA CTC AAG GCC-3'(서열번호 12)를 사용하여 중합효소 연쇄반응(PCR)을 수행하여 제작하였다. 두번째 단편은 정방향 프라이머로 5'-GGC CTT GAG TGG CTG GGA AGG 135 415 415 325 435 115 425 415 115 215 TAT GCA GTA TCT GTG AAA AGT-3'(서열번호 11), 역방향 프라이머로 5'-GAT CTC GAG CTA TTA CAA GTC CTC TTC AGA AAT AAG CTT TTG TTC GGA TCC-3'(서열번호 10)를 사용하여 PCR 방법으로 제작하였다. PCR은 pfu 중합효소(인트론, 한국)를 이용하여 수행하였으며, 94℃에서 40초, 55℃에서 30초, 72℃에서 40초의 반응을 30회 반복하였다.In order to introduce mutations into the heavy chain variable CDR2 of the anti-DR5 antibody, two fragments were prepared as shown in FIG. 2. The first fragment is 5'-GGT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3 '(SEQ ID NO: 9) and 5'-CCT TCC CAG CCA CTC AAG GCC-3 as reverse primer. (SEQ ID NO: 12) was used to perform a polymerase chain reaction (PCR). The second fragment is 5'-GGC CTT GAG TGG CTG GGA AGG 135 415 415 325 435 115 425 415 115 215 TAT GCA GTA TCT GTG AAA AGT-3 '(SEQ ID NO: 11) with forward primer and 5'-GAT CTC with reverse primer. GAG CTA TTA CAA GTC CTC TTC AGA AAT AAG CTT TTG TTC GGA TCC-3 '(SEQ ID NO: 10) was prepared by PCR method. PCR was performed using pfu polymerase (Intron, Korea), and the reaction of 40 seconds at 94 ° C, 30 seconds at 55 ° C, and 40 seconds at 72 ° C was repeated 30 times.
마찬가지로, 각각 중쇄가변 CDR3 및 경쇄가변 CDR3 부위의 변이체에 대한 단편들도 상기와 같은 방법으로 제작되었다. 중쇄가변 CDR3의 첫번째 단편은 정방향 프라이머로 5'-GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3'(서열번호 9), 역방향 프라이머로 5'-ACA GTA ATA GAC GGC CGT GTC-3'(서열번호 14)를 사용하였고, 두번째 단편은 정방향 프라이머로 5'-GAC ACG GCC GTC TAT TAC TGT 235 125 215 335 215 235 225 125 225 235 445 215 145 TGG GGC CAA GGG ACC ACG GTC-3'(서열번호 13), 역방향 프라이머로 5'-GAT CTC GAG CTA TTA CAA GTC CTC TTC AGA AAT AAG CTT TTG TTC GGA TCC-3'(서열번호 10)를 사용하여 PCR 방법으로 제작하였다. 경쇄가변 CDR3의 첫번째 단편은 정방향 프라이머로 5'-GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3'(서열번호 9), 역방향 프라이머로 5'-ACA GTA ATA AAC TGC AAA ATC-3'(서열번호 16)을 사용하였고, 두번째 단편은 정방향 프라이머로 5'-GAT TTT GCA GTT TAT TAC TGT 315 315 325 125 115 425 335 335 325 235 245 TTC GGC CAA GGG ACA CGA CTG-3'(서열번호 15), 역방향 프라이머로 5'-GAT CTC GAG CTA TTA CAA GTC CTC TTC AGA AAT AAG CTT TTG TTC GGA TCC-3'(서열번호 10)를 사용하여 PCR 방법으로 제작하였다.Similarly, fragments for the variants of the heavy chain variable CDR3 and light chain variable CDR3 sites, respectively, were prepared in the same manner as above. The first fragment of heavy chain variable CDR3 was 5'-GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3 '(SEQ ID NO: 9) as the forward primer, and 5'-ACA GTA ATA GAC GGC as the reverse primer. CGT GTC-3 '(SEQ ID NO: 14) was used, and the second fragment was a 5'-GAC ACG GCC GTC TAT TAC TGT 235 125 215 335 215 235 225 125 225 235 445 215 145 TGG GGC CAA GGG ACC ACG GTC -3 '(SEQ ID NO: 13), 5'-GAT CTC GAG CTA TTA CAA GTC CTC TTC AGA AAT AAG CTT TTG TTC GGA TCC-3' (SEQ ID NO: 10) as a reverse primer was prepared by the PCR method. The first fragment of the light chain variable CDR3 was 5'-GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3 '(SEQ ID NO: 9) as the forward primer, 5'-ACA GTA ATA AAC TGC as the reverse primer AAA ATC-3 '(SEQ ID NO: 16) was used, and the second fragment was a 5'-GAT TTT GCA GTT TAT TAC TGT 315 315 325 125 115 425 335 335 325 235 245 TTC GGC CAA GGG ACA CGA CTG-3 '(SEQ ID NO: 15), 5'-GAT CTC GAG CTA TTA CAA GTC CTC TTC AGA AAT AAG CTT TTG TTC GGA TCC-3' (SEQ ID NO: 10) as a reverse primer was prepared by the PCR method.
각각 제작된 두 개의 단편들은 1% 아가로오스 겔에서 전기영동하여 아가로오스 겔 정제 키트(인트론, 한국)를 이용하여 정제하였다. 정제된 두 개의 단편을 각 10μM씩 동량으로 혼합한 다음, 도 2와 같이 중첩 연장 PCR(overlap extension PCR)을 수행하여 변이된 전체 scFv 유전자 산물을 제조하였다. 상기 PCR은 pfu 중합효소(인트론, 한국)를 이용하여 수행하였고, 94℃에서 40초, 55℃에서 30초, 72℃에서 1분의 반응을 30회 반복하였다.Each of the two fragments prepared was electrophoresed on a 1% agarose gel and purified using an agarose gel purification kit (Intron, Korea). The purified two fragments were mixed in the same amount by 10 μM each, and then, as shown in FIG. 2, overlap extension PCR was performed to prepare a mutated whole scFv gene product. The PCR was performed using pfu polymerase (Intron, Korea), and the reaction of 40 seconds at 94 ° C, 30 seconds at 55 ° C, and 1 minute at 72 ° C was repeated 30 times.
3. 변이된 scFv 항체 유전자 라이브러리의 구축3. Construction of Mutated scFv Antibody Gene Library
돌연변이가 삽입된 scFv 항체 유전자 라이브러리(10㎍/㎕)와 scFv 효모 표면 발현벡터인 pCTCON(1㎍/㎕)를 혼합한 후, 전기천공(electroporation)을 이용하여 효모 EBY100 균주(Invitrogen, 미국)에 형질전환시켰다. 형질전환된 효모 EBY100 균주를 선택배지인 SD-CAA(-ura, -trp; 20g/ℓ의 글루코오스, 6.7g/ℓ의 아미노산이 첨가되지 않은 YNB(BD, 미국), 5.4g/ℓ의 Na2HPO4, 8.56g/ℓ의 NaH2PO4H2O 및 5g/ℓ의 카사미노산)에서 접종하여 배양한 후, 글루코오스에서 갈락토오스로 치환한 SG-CAA 배지에서 scFv의 세포 표면 발현을 유도하는 라이브러리를 선별하였다. 상기 라이브러리를 포함하는 세포를 SD-CAA 배지로 단계적으로 10배씩 희석하여 라이브러리 크기를 결정하였다.Mutant-inserted scFv antibody gene library (10 µg / µl) and scFv yeast surface expression vector pCTCON (1 µg / µl) were mixed, and then, electroporation was performed to the yeast EBY100 strain (Invitrogen, USA). Transformed. The transformed yeast EBY100 strain was selected as SD-CAA (-ura, -trp; 20 g / l glucose, YNB without 6.7 g / l amino acid (BD, USA), 5.4 g / l Na 2 Library inducing cell surface expression of scFv in SG-CAA medium substituted with glucose to galactose after inoculation with HPO 4 , 8.56 g / L NaH 2 PO 4 H 2 O and 5 g / L casamino acid) Were screened. Cells containing the library were diluted 10-fold in SD-CAA medium stepwise to determine library size.
그 결과, scFv 항체 라이브러리 크기는 각각 약 2×107 으로 구축되었음을 확인하였다.As a result, it was confirmed that the scFv antibody library size was each constructed at about 2 × 10 7 .
실시예 2Example 2 : 구축된 HW1의 변이체 라이브러리로부터 친화도가 향상된 변이체들의 선별 : Screening for Improved Affinity Variants from Constructed HW1 Variant Library
상기 실시예 1에서 구축된 HW1의 변이체 라이브러리로부터 DR5에 특이적으로 높은 친화도를 나타내는 scFv 변이체를 선별하기 위하여, 1:100으로 희석된 항-c-myc mAb(Ig therapy, 한국) 및 비오틴 표지 키트(EZ-LINK Sulfo-NHS-LC-Biotinylation kit, Pierce, 미국)를 이용하여 비오틴이 표지된 DR5 1μM을 1㎎/㎖의 BSA 0.2㎖가 첨가된 PBS(PBSB, pH 7.4)에 첨가한 후, 25℃에서 30분 동안 배양하였다. 상기 세포를 냉장된 PBSB로 세척한 후, 2차 항체로서 FITC-표지된 항-마우스 IgG(1:25로 희석) 및 피코에리쓰린이 접합된 스트렙타비딘(streptavidin-R-phycoerythrin conjugate (SA-PE), Molecular probes사, 미국; 1:100으로 희석)으로 표지하였다. 표지된 세포들을 세척하고 다시 PBSB에 재현탁한 후, FACS (fluorescence activated cell sorting)를 순차적으로 수행하여 친화도가 높은 변이체들을 선별하였다. 선별된 변이체들은 선택배지인 SD-CAA에서 배양하고, 위에 기술된 방법과 동일하게 다시 표지한 후, 선별 과정을 4회 반복하였다. 이때, 선별 과정시 사용된 비오틴이 표지된 DR5의 농도를 초기 1μM에서 점점 농도를 낮추어 마지막 4차 선별시에는 10nM의 농도를 사용하였다. 선별된 변이체들의 뉴클레오티드 서열을 정방향 프라이머 5'-GTT CCA GAC TAC GCT CTG CAG G-3'(서열번호 17) 및 역방향 프라이머 5'-GAT TTT GTT ACA TCT ACA CTG TTG-3'(서열번호 18)을 이용하여 분석한 후, 아미노산 서열을 결정하여 야생형의 아미노산 서열과 비교하였다.Anti-c-myc mAb (Ig therapy, Korea) and biotin label diluted 1: 100 to select scFv variants showing high affinity to DR5 from the variant library of HW1 constructed in Example 1 above Biotin-labeled DR5 was added to PBS (PBSB, pH 7.4) with 0.2 mg of 1 mg / mL BSA using the kit (EZ-LINK Sulfo-NHS-LC-Biotinylation kit, Pierce, USA). Then, incubated for 30 minutes at 25 ℃. After washing the cells with chilled PBSB, the streptavidin conjugated with FITC-labeled anti-mouse IgG (diluted at 1:25) and phycoerythrin as a secondary antibody (SA- PE), Molecular probes, USA; diluted 1: 100). After the labeled cells were washed and resuspended in PBSB, fluorescence activated cell sorting (FACS) was performed sequentially to select high affinity variants. Selected variants were incubated in SD-CAA, a selection medium, relabeled in the same manner as described above, and the selection process was repeated four times. At this time, the concentration of biotin-labeled DR5 used in the selection process was gradually lowered from the initial 1 μM, and the concentration of 10 nM was used for the final 4th screening. Nucleotide sequences of the selected variants were forward primer 5'-GTT CCA GAC TAC GCT CTG CAG G-3 '(SEQ ID NO: 17) and reverse primer 5'-GAT TTT GTT ACA TCT ACA CTG TTG-3' (SEQ ID NO: 18) After analysis using the amino acid sequence was determined and compared with the amino acid sequence of the wild type.
결과는 도 3에 나타내었다.The results are shown in FIG.
도 3에 나타난 바와 같이, 각 선별 단계별로 HW1 변이체의 친화도가 증가하는 경향을 나타내었다.As shown in FIG. 3, the affinity of the HW1 variant was increased for each selection step.
실시예 3Example 3 : 선별된 항체들의 친화도 결합 : Affinity binding of selected antibodies
각각의 라이브러리에서 선별된 변이체들의 아미노산 서열을 결정함과 동시에 선별된 야생형들의 변화된 CDR들을 각각 결합하여 최종적으로 가장 친화도가 향상된 새로운 변이체를 제작하였다. 중쇄가변 CDR3로부터 유래된 변이체를 주형으로 하여, 정방향 프라이머로 5'-GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3'(서열번호 9), 역방향 프라이머로 5'-ACA GTA ATA AAC TGC AAA ATC-3'(서열번호 16)를 사용하여 PCR 방법으로 단편을 제작하였다. 경쇄가변 CDR3로부터 유래된 변이체를 주형으로 하여, 정방향 프라이머로 5'-GAT TTT GCA GTT TAT TAC TGT-3'(서열번호 19), 역방향 프라이머로 5'-GAT CTC GAG CTA TTA CAA GTC CTC TTC AGA AAT AAG CTT TTG TTC GGA TCC-3'(서열번호 10)를 사용하여 PCR 방법으로 단편을 제작하였으며, 94℃에서 40초, 55℃에서 30초, 72℃에서 50초의 반응을 30회 반복하였다. 각각 제작된 두 개의 단편들을 1% 아가로오스 겔에서 전기영동하여 아가로오스 겔 정제 키트(인트론, 한국)를 이용하여 정제하고, 정제된 두 개의 단편을 각 10μM씩 동량으로 혼합한 다음, 중첩 연장 PCR을 수행하여 최종적으로 친화도가 향상된 변이체 전체 scFv 유전자 산물을 제조하였다. 상기 PCR은 94℃에서 40초, 55℃에서 30초, 72℃에서 1분의 반응을 30회 반복하였다. 이렇게 제작된 변이체 scFv 산물은 제한효소 Nhe I/BamH I (NEB, 미국)을 이용하여 효모 표면 발현 벡터인 pCTCON(Colby 등, Methods Enzymol., 388:348-358, 2004)에 도입하였고, 구축된 플라스미드를 효모 EBY100 균주(Invitrogen 사)에 형질전환하였다.The amino acid sequence of the variants selected from each library was determined and the changed CDRs of the selected wildtypes were respectively combined to finally produce new variants with the most affinity. A variant derived from heavy chain variable CDR3 was used as a template, and 5'-GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GGT GGT GGT GGT TCT GCT AGC-3 '(SEQ ID NO: 9) and 5'- as reverse primer. Fragments were prepared by PCR using ACA GTA ATA AAC TGC AAA ATC-3 ′ (SEQ ID NO: 16). Using a variant derived from the light chain variable CDR3 as a template, 5'-GAT TTT GCA GTT TAT TAC TGT-3 '(SEQ ID NO: 19) as the forward primer, 5'-GAT CTC GAG CTA TTA CAA GTC CTC TTC AGA as the reverse primer A fragment was prepared by PCR using AAT AAG CTT TTG TTC GGA TCC-3 ′ (SEQ ID NO: 10), and the reaction was repeated 30 times at 40 ° C. at 94 ° C., 30 seconds at 55 ° C., and 50 seconds at 72 ° C. Each of the two fragments prepared was electrophoresed on a 1% agarose gel and purified using an agarose gel purification kit (Intron, Korea), and the two purified fragments were mixed in equal amounts by 10 μM each, and then superimposed. Extended PCR was performed to finally produce a variant whole scFv gene product with improved affinity. The PCR was repeated 30 times 40 minutes at 94 ℃, 30 seconds at 55 ℃, 1 minute at 72 ℃. The mutant scFv product thus prepared was introduced into the yeast surface expression vector pCTCON (Colby et al., Methods Enzymol., 388: 348-358, 2004) using the restriction enzyme Nhe I / BamH I (NEB, USA). The plasmid was transformed into yeast EBY100 strain (Invitrogen).
선별된 변이체들과 위에서 제작된 최종적으로 친화도가 향상된 변이체를 실시예 2에서 기술된 방법과 동일하게 다시 FACS를 이용하여 친화도를 분석하였다. 대조군으로서 HW1을 동일한 조건으로 표지분석하여 비교하였다.The affinity was analyzed again using FACS in the same manner as described in Example 2 were selected variants and finally improved affinity variants prepared above. As a control, HW1 was compared by labeling under the same conditions.
결과는 도 4에 나타내었다.The results are shown in FIG.
도 4에 나타난 바와 같이, 각 변이체가 결합된 최종 변이체는 다른 변이체들보다 가장 높은 친화도를 나타내었다.As shown in FIG. 4, the final variant to which each variant was bound showed the highest affinity than the other variants.
실시예 4Example 4 : 친화도가 향상된 변이체의 항체 안정성 향상 : Improved antibody stability of affinity variants
1. 친화도와 안정성이 향상된 항체인 AU11의 제작1. Preparation of AU11, an antibody with improved affinity and stability
항-DR5 인간 항체인 HW1은 인간 항체를 구성하는 중쇄가변 영역에 대하여 VH6 아류형(subtype)으로, 경쇄가변 영역에 대하여 Vk3으로 구성되어 있으며, VH6 아류형은 다른 아류형, 특히 VH3에 비하여 그 안정성이 낮다고 알려져 있다. 따라서, 친화도가 향상된 변이체의 항체 안정성을 향상시키기 위하여, VH6 아류형을 VH3 아류형으로 치환하였다. 먼저, VH3-Vk3 형태의 항체를 제작하기 위하여, 전체 유전자를 크게 3개의 단편으로 나누고 각 단편을 제작하였다. 첫번째 단편은 정방향 프라이머로 5'-TTC GCT AGC GAG GTG CAG CTG GTG GAG TCT GGG GGA GGC CTG GTA CAG CCT GGA GGG TCC CTG AGA CTC TCC TGT GCA GCC TCT GGA GAC AGT GTC TCT AGC ACC ACT GTT GCC ATG AGC TGG GTC CGC CAG GCT CCA GGG AAG GGG CTG GAG TGG GTC-3'(서열번호 20), 역방향 프라이머로 5'-ACA GTA ATA CAC AGC CGT GTC CTC GGC TCT CAG GCT GTT CAT TTG CAG ATA CAG GGT GTT CTT GGA ATT GTC TCT GGA GAT GGT GAA CCG GCC CTT CAC GGA GTC CGC ATA GTA ATT ATA CCA CTT CGA CCT ATA ATA GAT CGC TGA GAC CCA CTC CAG CCC CTT CCC-3'(서열번호 21)을 이용하여 제작하였다. 두번째 단편은 정방향 프라이머로 5'-GAG GAC ACG GCT GTG TAT TAC TGT-3'(서열번호 22), 역방향 프라이머로 5'-TTC CCC TGG AGA CAA AGA CAG GGT GGC TGG AGA CTG GGT CAA TAC AAT ATC CGA CCC GCC ACC GCC GCT GCC ACC-3'(서열번호 23)을 이용하여 제작하였다. 세번째 단편은 정방향 프라이머로 5'-ACC CTG TCT TTG TCT CCA GGG GAA-3'(서열번호 24), 역방향 프라이머로 5'-CGT GGA TCC ACG TTT GAT TTC CAC CTT TGT CCC TTG GCC GAA GAC CGC CCG-3'(서열번호 25)을 이용하여 제작하였다. 또한, 중쇄 가변영역과 경쇄 가변영역을 연결하는 링커로서, 글리신(Glycine) 4개와 세린 (Serine) 1개가 3번 반복되는 구조를 가지며, 총 15개의 아미노산으로 구성된 (G4S)3 링커를 이용하였다(서열번호 26). 이렇게 제작된 각 단편은 중첩 연장 PCR을 수행하여 최종적으로 안정성이 향상된 변이체 전체 scFv 유전자 산물을 제조하였다. 상기 PCR은 94℃에서 40초, 55℃에서 30초, 72℃에서 1분의 반응을 30회 반복하였다. 이후, 중쇄가변 영역의 CDR3 지역의 101번 발린(valine), 102번 세린을 다시 아스파라긴산(aspartic acid)과 이소루신(isoleucine)으로 치환하여, 최종적으로 친화도와 안정성이 향상된 항체인 AU11을 제작하였다. 항체 AU11로의 안정성 향상에 사용된 프라이머 서열번호 및 서열은 하기 표 2에 나타내었다.HW1, an anti-DR5 human antibody, is composed of the VH6 subtype for the heavy chain variable region and Vk3 for the light chain variable region, and the VH6 subtype is compared to other subtypes, particularly VH3. It is known that the stability is low. Thus, in order to improve the antibody stability of the variants with improved affinity, the VH6 subtype was substituted with the VH3 subtype. First, in order to prepare the antibody of the VH3-Vk3 form, the whole gene was divided into three fragments and each fragment was prepared. The first fragment is a forward primer 5'-TTC GCT AGC GAG GTG CAG CTG GTG GAG TCT GGG GGA GGC CTG GTA CAG CCT GGA GGG TCC CTG AGA CTC TCC TGT GCA GCC TCT GGA GAC AGT GTC TCT AGC ACC ACT GTT GCC ATG AGC TGG GTC CGC CAG GCT CCA GGG AAG GGG CTG GAG TGG GTC-3 '(SEQ ID NO: 20), 5'-ACA GTA ATA CAC AGC CGT GTC CTC GGC TCT CAG GCT GTT CAT TTG CAG ATA CAG GGT GTT CTT GGA ATT GTC TCT GGA GAT GGT GAA CCG GCC CTT CAC GGA GTC CGC ATA GTA ATT ATA CCA CTT CGA CCT ATA ATA GAT CGC TGA GAC CCA CTC CAG CCC CTT CCC-3 '(SEQ ID NO: 21). The second fragment is 5'-GAG GAC ACG GCT GTG TAT TAC TGT-3 '(SEQ ID NO: 22) with forward primer, 5'-TTC CCC TGG AGA CAA AGA CAG GGT GGC TGG AGA CTG GGT CAA TAC AAT ATC CGA with reverse primer Produced using CCC GCC ACC GCC GCT GCC ACC-3 '(SEQ ID NO: 23). The third fragment is 5'-ACC CTG TTG TCT TTG TCT CCA GGG GAA-3 '(SEQ ID NO: 24) with forward primer, 5'-CGT GGA TCC ACG TTT GAT TTC CAC CTT TGT CCC TTG GCC GAA GAC CGC CCG- It was produced using 3 '(SEQ ID NO: 25). In addition, as a linker linking the heavy chain variable region and the light chain variable region, 4 glycine and 1 serine is repeated three times, using a (G 4 S) 3 linker consisting of a total of 15 amino acids (SEQ ID NO: 26). Each fragment thus prepared was subjected to overlap extension PCR to finally prepare a variant whole scFv gene product having improved stability. The PCR was repeated 30 times 40 minutes at 94 ℃, 30 seconds at 55 ℃, 1 minute at 72 ℃. Subsequently, valine # 101 and # 102 serine in the CDR3 region of the heavy chain variable region were replaced with aspartic acid and isoleucine to finally prepare AU11, an antibody having improved affinity and stability. Primer sequence numbers and sequences used to improve stability with antibody AU11 are shown in Table 2 below.
항체 AU11의 전체 뉴클레오티드 및 아미노산 서열을 도식화한 것은 도 5에 나타내었고(여기에서, 밑줄로 표시한 부분은 차례로 VH-CDR1, VH-CDR2, VH-CDR3, 링커, VL-CDR1, VL-CDR2, VL-CDR3를 나타낸 것임), 기존 항체인 HW1과 최종적으로 친화도 및 안정성이 향상된 AU11의 아미노산 서열을 비교 분석한 결과는 도 6에 나타내었다(*는 CDR에서 친화도 향상에 따라 변이된 아미노산 잔기를 표시한 것임).Schematic representation of the entire nucleotide and amino acid sequence of antibody AU11 is shown in FIG. 5 (wherein the underlined portions are in turn VH-CDR1, VH-CDR2, VH-CDR3, linker, VL-CDR1, VL-CDR2, A comparison of the amino acid sequence of AU11 with improved affinity and stability with the existing antibody HW1 is shown in FIG. 6 (* is an amino acid residue which is changed according to affinity improvement in CDR). ).
[표 2]TABLE 2
Figure PCTKR2009006036-appb-I000002
Figure PCTKR2009006036-appb-I000002
2. 친화도와 안정성이 향상된 항체인 AU12의 제작2. Preparation of AU12, an antibody with improved affinity and stability
경쇄가변영역은 Vk3 및 Vk1이 가장 안정하다고 알려져 있으며, 미국 FDA의 승인을 받아 시판되고 있는 다양한 쥐 유래의 인간화 항체(humanized antibody)들에서 Vk1 아류형이 다수 존재하고 있다(Carter 등, Proc Natl Acad Sci U S A, 10;89:4285-4289, 1992; Werther 등, J Immunol, 11;157:4986-4995, 1996; Presta 등, J Immunol 5;151:2623-2632, 1993). 또한, 베르니에 구역 잔기들(vernier zone residues)은 항원 결합에 있어서 열역학적 안정성에 공헌하며, 항체의 구조적인 엔트로피 변화를 조절하여 결과적으로 항원과의 결합에 중요한 영향을 미치는 부위로 알려져 있는데(Makabe 등, J Biol Chem, 11;283(2):1156-1166, 2008), Vk1 아류형을 구성하는 아미노산 중에 추가적으로 베르니에 구역에 해당하는 잔기들 및 그 외 항체 구조의 3차원 시뮬레이션 분석(http://antibody.bath.ac.uk)을 검토하여 구조적으로 중요하다고 생각되는 잔기를 추가로 선정하였으며, 가변경쇄의 2번 아미노산인 이소루신을 세린으로 치환하여, VH3-Vk3 형태의 항체인 AU11 이외에 추가적으로 VH3-Vk1 아류형을 가지는 항체를 제작하였다. AU11을 주형으로 하여, 첫 번째 단편은 정방향 프라이머로 5'-TTC GCT AGC GAG GTG CAG CTG GTG GAG TCT GGG-3'(서열번호 29), 역방향 프라이머로 5'-TGG AGA CTG GGT CAT CTG AGA GTC-3'(서열번호 30)을 이용하였고, 두 번째 단편은 정방향 프라이머로 5'-GAC TCT CAG ATG ACC CAG TCT CCA TCC TCC CTG TCT GCA TCT GTA GGA GAC AGA GTC ACC ATC ACT TGC CGG GCA AGT CAG AGC GTT AGC-3'(서열번호 31), 역방향 프라이머로 5'-CTT AGG GGC TTT CCC TGG TTT CTG CTG ATA CCA GGC TAA GTG GCT-3'(서열번호 32)을 이용하였고, 세 번째 단편은 정방향 프라이머로 5'-AAA GCC CCT AAG CTC CTG ATC TAT GGT GCA TCC AGC-3'(서열번호 33), 역방향 프라이머로 5'-GAT GGT GAG AGT GAA ATC TGT CCC AGA TCC ACT GCC ACT GAA CCT TGA TGG GAC CCC AGT GGC CCT-3'(서열번호 34)을 이용하였고, 네 번째 단편은 정방향 프라이머로 5'-TTC ACT CTC ACC ATC AGC AGT CTG CAA CCT GAA GAT TTT GCA ACT TAC TAC TGT CAA CAG CGT GCA AAC GAT TTC CCG CGG GCG GTC-3'(서열번호 35), 역방향 프라이머로 5'-CGT GGA TCC ACG TTT GAT TTC CAC-3'(서열번호 36)을 이용하여 제작하였다. AU11과 마찬가지로, 중쇄가변 영역과 경쇄가변 영역을 연결하는 링커로서 글리신 4개 및 세린 1개가 세 번 반복되는 형태인 (G4S)3 링커를 사용하였다. 이렇게 제작된 각 단편은 중첩 연장 PCR을 수행하여 최종적으로 안정성이 향상된 변이체 전체 scFv 유전자 산물을 제조하였다. 상기 PCR은 94℃에서 40초, 55℃에서 30초, 72℃에서 1분의 반응을 30회 반복하여 최종적으로 AU12를 제작하였다. AU12의 제작에 사용된 프라이머 서열 번호 및 서열은 하기 표 3에 나타내었다.In the light chain variable region, Vk3 and Vk1 are known to be the most stable, and many Vk1 subtypes exist in various mouse-derived humanized antibodies commercially approved by the US FDA (Carter et al., Proc Natl Acad). Sci USA , 10; 89: 4285-4289, 1992; Werther et al ., J Immunol , 11; 157: 4986-4995, 1996; Presta et al ., J Immunol 5; 151: 2623-2632, 1993). In addition, vernier zone residues contribute to thermodynamic stability in antigen binding, and are known as sites that modulate the structural entropy changes of antibodies and consequently have a significant effect on antigen binding (Makabe et al. J Biol Chem , 11; 283 (2): 1156-1166, 2008), three-dimensional simulation analysis of the residues of the Vernier region and other antibody structures among the amino acids constituting the Vk1 subtype (http: // antibody) .bath.ac.uk) was selected to further select residues considered to be structurally important. In addition to the VH3-Vk3 antibody AU11, VH3- was added to the serine by replacing isoleucine, the second amino acid of the modified chain, with serine. Antibodies having the Vk1 subtype were produced. With AU11 as template, the first fragment was 5'-TTC GCT AGC GAG GTG CAG CTG GTG GAG TCT GGG-3 '(SEQ ID NO: 29) with forward primer, 5'-TGG AGA CTG GGT CAT CTG AGA GTC with reverse primer -3 '(SEQ ID NO: 30) was used, and the second fragment was a 5'-GAC TCT CAG ATG ACC CAG TCT CCA TCC TCC CTG TCT GCA TCT GTA GGA GAC AGA GTC ACC ATC ACT TGC CGG GCA AGT CAG AGC GTT AGC-3 '(SEQ ID NO: 31), 5'-CTT AGG GGC TTT CCC TGG TTT CTG CTG ATA CCA GGC TAA GTG GCT-3' (SEQ ID NO: 32) was used as the reverse primer, and the third fragment was a forward primer 5'-AAA GCC CCT AAG CTC CTG ATC TAT GGT GCA TCC AGC-3 '(SEQ ID NO: 33), 5'-GAT GGT GAG AGT GAA ATC TGT CCC AGA TCC ACT GCC ACT GAA CCT TGA TGG GAC CCC AGT GGC CCT-3 '(SEQ ID NO: 34) was used, and the fourth fragment was 5'-TTC ACT CTC ACC ATC AGC AGT CTG CAA CCT GAA GAT TTT GCA ACT TAC TAC TGT CAA CAG CGT GCA AAC GAT TTC CCG CGG G CG GTC-3 '(SEQ ID NO: 35) and 5'-CGT GGA TCC ACG TTT GAT TTC CAC-3' (SEQ ID NO: 36) were prepared as reverse primers. Like AU11, as a linker linking the heavy chain variable region and the light chain variable region, (G 4 S) 3 linker in which 4 glycine and 1 serine was repeated three times was used. Each fragment thus prepared was subjected to overlap extension PCR to finally prepare a variant whole scFv gene product having improved stability. In the PCR, AU12 was finally produced by repeating a reaction of 40 seconds at 94 ° C, 30 seconds at 55 ° C, and 1 minute at 72 ° C for 30 times. Primer sequence numbers and sequences used in the preparation of AU12 are shown in Table 3 below.
항체 AU12의 전체 뉴클레오티드 및 아미노산 서열을 도식화한 것은 도 7에 나타내었고(여기에서, 밑줄로 표시한 부분은 차례로 VH-CDR1, VH-CDR2, VH-CDR3, 링커, VL-CDR1, VL-CDR2, VL-CDR3를 나타낸 것임), 기존 항체인 HW1과 최종적으로 친화도 및 안정성이 향상된 AU11 및 AU12의 아미노산 서열을 비교 분석한 결과는 도 8에 나타내었다.A schematic of the entire nucleotide and amino acid sequence of antibody AU12 is shown in FIG. 7 (wherein the underlined portions are in turn VH-CDR1, VH-CDR2, VH-CDR3, linker, VL-CDR1, VL-CDR2, VL-CDR3), and comparing the amino acid sequence of AU11 and AU12 finally improved affinity and stability with the existing antibody HW1 is shown in FIG.
[표 3]TABLE 3
Figure PCTKR2009006036-appb-I000003
Figure PCTKR2009006036-appb-I000003
실시예 5Example 5 : AU11 및 AU12 scFv 항체의 발현 및 정제 : Expression and Purification of AU11 and AU12 scFv Antibodies
친화도와 안정성이 향상된 항체인 AU11 및 AU12를 대장균 발현 벡터인 pKJ1에 Nhe I/BamH I을 이용하여 인 프레임(in-frame)으로 도입하였다. 이때, 대장균 발현 벡터는 T7 프로모터 - pelB 표적화 서열 - AU11 또는 AU12 scFv - flag tag - 6X his tag을 가지도록 설계되었다.Antibodies with improved affinity and stability, AU11 and AU12, were introduced into the E. coli expression vector pKJ1 in-frame using Nhe I / BamH I. At this time, the E. coli expression vector was designed to have a T7 promoter-pelB targeting sequence-AU11 or AU12 scFv-flag tag-6X his tag.
상기 발현벡터를 대장균 BL21(DE3)에 형질전환한 후, 항생제 암피실린 (ampicillin)이 50㎍/㎖ 첨가된 LB[5g/L yeast extract(BD사, 미국), 10g/L tryptone(BD사, 미국), 10g/L sodium chloride] 배지에서 37℃, 200rpm으로 OD600가 0.8~1.0이 되도록 배양한 후, 0.5mM IPTG(Isopropyl β-D-1-thiogalactopyranoside)를 첨가하고 16시간 동안 23℃, 150rpm의 조건 하에서 배양하였다. 발현 후 원심분리하여 상등액만을 취한 후 Talon-resin(Clontech사)을 사용하여 발현된 항체를 정제한 다음, 정제된 항체의 크기 및 순도를 환원성 SDS-PAGE 및 비환원성 SDS-PAGE로 분석하였다.After transforming the expression vector into Escherichia coli BL21 (DE3), LB [5 g / L yeast extract (BD company, USA), 10 g / L tryptone (BD company, USA) to which 50 μg / ml of antibiotic ampicillin was added ), 10 g / L sodium chloride] medium at 37 ° C. and 200 rpm to incubate the OD 600 at 0.8 to 1.0, followed by addition of 0.5 mM IPTG (Isopropyl β-D-1-thiogalactopyranoside) and 23 ° C., 150 rpm for 16 hours. Incubated under the conditions of. After expression, only the supernatant was taken by centrifugation, and then, the antibody expressed using Talon-resin (Clontech) was purified, and the size and purity of the purified antibody were analyzed by reducing SDS-PAGE and non-reducing SDS-PAGE.
결과는 도 9에 나타내었다.The results are shown in FIG.
도 9에 나타난 바와 같이, 항체 AU11 및 AU12는 약 29 kDa의 분자량을 가지며. 99% 이상의 순도로 정제됨을 확인하였다.As shown in FIG. 9, antibodies AU11 and AU12 have a molecular weight of about 29 kDa. It was confirmed that the purity was 99% or more.
실험예 1Experimental Example 1 : AU11 및 AU12 scFv 항체의 용액 상태에서의 형태 확인 : Morphology Identification in AU11 and AU12 scFv Antibodies
정제된 AU11 및 AU12 scFv 항체가 용액 상태에서 단일분자 형태(monomeric form)로 존재하는지 아니면 다중분자 형태(oligomeric form)로 존재하는지 확인하기 위하여, 크기 배제 HPLC, 환원성 SDS-PAGE 및 비환원성 SDS-PAGE를 수행하였다.To determine whether purified AU11 and AU12 scFv antibodies exist in monomeric or oligomeric form in solution, size-exclusion HPLC, reducible SDS-PAGE, and non-reducing SDS-PAGE Was performed.
크기 배제 HPLC의 경우, 용출 완충액은 PBS(50mM 포스페이트, pH 7.4, 150mM NaCl)이며, 유속은 0.5㎖/분, 컬럼은 SuperdexTM200 10/300GC (GE사, 미국) 및 Agilent 1100 HPLC 장비를 이용하여 280nm에서 흡광도를 측정하였다. 환원성 SDS-PAGE는 샘플 완충액에 1mM DTT가 첨가된 15% 아크릴아미드 겔을 사용하였고, 비환원성 SDS-PAGE는 샘플 완충액에 1mM DTT가 첨가되지 않은 15% 아크릴아미드 겔을 사용하여 수행하였으며, 150 볼트로 로딩하여 분석하였다.For size exclusion HPLC, the elution buffer is PBS (50 mM phosphate, pH 7.4, 150 mM NaCl), flow rate 0.5 ml / min, column 280 nm using SuperdexTM 200 10/300 GC (GE, USA) and Agilent 1100 HPLC instrument Absorbance was measured at. Reducing SDS-PAGE was performed using 15% acrylamide gel with 1 mM DTT added to the sample buffer and non-reducing SDS-PAGE was performed with 15% acrylamide gel without 1 mM DTT added to the sample buffer, 150 volts. Analyze by loading.
정제된 AU11 및 AU12 scFv 항체를 환원성 SDS-PAGE 및 비환원성 SDS-PAGE로 분석한 결과 및 크기 배제 HPLC 측정 결과는 각각 도 9 및 도 10에 나타내었다.Purified AU11 and AU12 scFv antibodies were analyzed by reducing SDS-PAGE and non-reducing SDS-PAGE and the results of size exclusion HPLC measurement are shown in FIGS. 9 and 10, respectively.
도 9에 나타난 바와 같이, 정제된 AU11 및 AU12 scFv 항체는 환원성 SDS-PAGE 및 비환원성 SDS-PAGE에서 비천연 이황화 결합(disulfide bond)이 있는 다중분자 형태가 없었다.As shown in FIG. 9, the purified AU11 and AU12 scFv antibodies were free of multimolecular forms with non-natural disulfide bonds in reducing SDS-PAGE and non-reducing SDS-PAGE.
또한 도 10에 나타난 바와 같이, 크기 배제 HPLC에서 정제된 AU11 및 AU12 scFv 항체를 500㎍/㎖로 주입하였을 때 HW1과 마찬가지로 순수하게 단일분자로 용출되었다.In addition, as shown in FIG. 10, when injected with 500 μg / ml of purified AU11 and AU12 scFv antibodies in size exclusion HPLC, the mixture was eluted as pure single molecule as HW1.
실험예 2Experimental Example 2 : 항체 AU11 및 AU12의 친화도 측정 : Affinity Measurement of Antibodies AU11 and AU12
DR5 항원에 대한 AU11 및 AU12의 결합 친화도를 확인하기 위하여, Biacore 2000 SPR(surface Plasmon resonance) 바이오센서(GE사, 미국)를 이용하여 측정하였다. 약 0.5㎎/㎖의 항원 및 음성대조군인 BSA(bovine serum albumin)를 CM5 칩 (carboxymethylated dextran surface chip)에 약 2,000 반응단위(response unit)로 고정화시킨 후, 다양한 농도로 희석된 항체 AU11 및 AU12를 30㎕/분의 속도로 2분간 칩에 주입하여 항원과의 상호작용을 정량하였다. 또한, HW1과의 친화도 비교를 위하여 HW1은 50nM, AU11 및 AU12는 각각 25nM의 농도로 준비하여 같은 시간으로 주입하였다. 칩의 표면은 1M NaCl/20mM NaOH로 재생하였으며, BIA evaluation ver. 3.2 소프트웨어를 이용하여 친화도를 운동속도상수(kon 및 koff)와 평형해리상수(KD)로 얻었다.In order to confirm the binding affinity of AU11 and AU12 for the DR5 antigen, it was measured using a Biacore 2000 surface Plasmon resonance (SPR) biosensor (GE, USA). About 0.5 mg / ml of antigen and negative control group BSA (bovine serum albumin) were immobilized on a carboxymethylated dextran surface chip (CM5) at about 2,000 response units, and then the antibodies AU11 and AU12 diluted at various concentrations were added. Interaction with antigen was quantified by injection into the chip for 2 minutes at a rate of 30 μl / min. In addition, in order to compare affinity with HW1, HW1 was prepared at a concentration of 50nM, AU11 and AU12 at 25nM, respectively, and injected at the same time. The surface of the chip was regenerated with 1M NaCl / 20mM NaOH, BIA evaluation ver. Using the 3.2 software to obtain the affinity to the movement rate constants (k on and k off) and the equilibrium dissociation constant (K D).
DR5에 대한 AU11 및 AU12의 친화도를 SPR을 이용하여 정량한 결과 및 HW1과의 친화도를 비교한 결과는 도 11에 나타내었으며, DR5에 대한 AU11 및 AU12의 친화도를 운동속도상수(kon 및 koff)와 평형해리상수(KD)로 나타낸 결과는 표 4에 나타내었다.The results of quantification of the affinity of AU11 and AU12 for DR5 using SPR and the comparison of the affinity with HW1 are shown in FIG. 11, and the affinity of AU11 and AU12 for DR5 was determined by the kinematic velocity constant (k on). And k off ) and the equilibrium dissociation constant (K D ) are shown in Table 4.
[표 4]TABLE 4
Figure PCTKR2009006036-appb-I000004
Figure PCTKR2009006036-appb-I000004
도 11 및 표 4에 나타난 바와 같이, AU11과 AU12는 HW1(KD: 약 200nM)에 비하여 약 17~22배 정도 높은 친화도를 나타내었다. 또한, 같은 농도에서 HW1과 AU11 및 AU12를 동시에 분석하였을 때, AU11 및 AU12가 HW1보다 해리구간에서 더 강하게 결합하고 있음을 보여주었다.As shown in Figure 11 and Table 4, AU11 and AU12 showed affinity about 17-22 times higher than HW1 (K D : about 200nM). In addition, simultaneous analysis of HW1, AU11, and AU12 at the same concentration showed that AU11 and AU12 bind more strongly in the dissociation interval than HW1.
실험예 3Experimental Example 3 : 항체 AU11 및 AU12의 안정성 측정 : Stability Measurement of Antibodies AU11 and AU12
AU11 및 AU12의 HW1에 대한 안정성 향상 여부를 형광 분광광도계 (fluorescence spectrometer, JASCO사, 일본)를 이용하여 측정하였다. HW1, AU11 scFv, 및 AU12 scFv를 대장균에서 발현 및 정제하여 준비하고, 준비된 단백질을 각각 0M부터 4.2M까지 0.3M 단위로 하여 구아니딘·HCl(guanidine·HCl)에 해리하여 최종 10㎍/㎖의 단백질 농도로 상온에서 24시간 반응시킨 후, 여기파장 (excitation wave length) 280㎚, 방출파장(emission wave length) 290~400㎚, 측정속도는 250㎚/분으로 하여 3회 반복 측정하였다. 이렇게 측정된 값 중 각 구아니딘·HCl 농도별로 가장 높은 값을 가질 때의 파장을 표준화(normalization)하여 상대적인 최대 파장(normalized lmax)을 도식화하였다. HW1은 약 1.64 M의 구아니딘·HCl의 농도에서 그 중간값을, AU1과 AU12는 각각 약 2.53M, 2.45M의 구아니딘·HCl의 농도에서 그 중간값을 갖는다. 문헌(Ewert S 등, Biochemistry, 42:1517-1528, 2003)에 의하면, 구아니딘·HCl과 같은 단백질 변성시료에 의해 단백질이 변성될수록 상대적인 최대파장은 커진다고 알려져 있다.The stability of AU11 and AU12 against HW1 was measured using a fluorescence spectrometer (JASCO, Japan). HW1, AU11 scFv, and AU12 scFv were prepared by expression and purification in E. coli, and the prepared proteins were dissociated into guanidine-HCl (guanidine-HCl) in 0.3M units from 0M to 4.2M, respectively, to give a final concentration of 10 µg / ml. After reacting at room temperature for 24 hours at a concentration, an excitation wave length of 280 nm, an emission wave length of 290 to 400 nm, and a measurement rate of 250 nm / min were repeated three times. The relative maximum wavelength (normalized l max ) was plotted by normalizing the wavelength at the highest value for each guanidine-HCl concentration. HW1 has a median value at a concentration of about 1.64M guanidine-HCl, and AU1 and AU12 have a median value at a concentration of about 2.53M and 2.45M, respectively. According to Ewert S et al., Biochemistry , 42: 1517-1528, 2003, the relative maximum wavelength increases as the protein is denatured by a protein denaturation sample such as guanidine-HCl.
결과는 도 12에 나타내었다. The results are shown in FIG.
도 12에 나타난 바와 같이, AU11 및 AU12는 HW1에 비해 그 안정성이 상대적으로 높음을 알 수 있다.As shown in FIG. 12, it can be seen that AU11 and AU12 have relatively higher stability than HW1.
실험예 4Experimental Example 4 : DR5에 대한 HW1과 AU11, AU12 간의 동일한 항원결합부위 (epitope) 여부 규명 : Identification of the same epitope between HW1, AU11, and AU12 for DR5
DR5에 대하여 AU11 및 AU12가 HW1과 동일한 항원결합부위를 갖는지의 여부를 규명하기 위하여, AU11 또는 AU12와 HW1 간의 경쟁적 ELISA(competition ELISA)를 수행하였다.To determine whether AU11 and AU12 had the same antigen-binding site as HW1 for DR5, a competitive ELISA between AU11 or AU12 and HW1 was performed.
구체적으로는, ELISA용 96-웰 플레이트에 항원을 50㎕ (20㎍/㎖) 코팅한 후 37℃에서 1시간 동안 배양하였다. PBS(pH 7.4, with 0.05% Tween 20 및 0.02% sodium azide)로 3회 세척한 후 1% BSA가 함유된 PBS로 37℃에서 1시간 동안 배양하였다. 그 후, 비오틴을 표지한 HW1을 30㎍/㎖로 고정화하고, AU11 또는 AU12의 농도를 5~300㎍/㎖로 하여 각 웰에 혼합하여 첨가한 후 37℃에서 1시간 동안 배양하고, 플레이트를 세척한 후, 알칼라인 포스파타아제(Alkaline phosphatase)가 접합된 항-비오틴 항체(anti-biotin mAb AP conjugated, Sigma)를 첨가하여 37℃에서 1시간 동안 반응시켰다. 그 후, 3회 세척한 후 기질로 pNPP(Sigma.)를 50㎕ 첨가하여 405㎚에서 흡광도를 측정하였다.Specifically, 50 μl (20 μg / ml) of antigen was coated on a 96-well plate for ELISA, followed by incubation at 37 ° C. for 1 hour. After washing three times with PBS (pH 7.4, with 0.05% Tween 20 and 0.02% sodium azide) and incubated for 1 hour at 37 ℃ in PBS containing 1% BSA. Thereafter, the biotin-labeled HW1 was immobilized at 30 µg / ml, the concentration of AU11 or AU12 was 5 to 300 µg / ml, mixed and added to each well, followed by incubation at 37 ° C for 1 hour, and the plate was After washing, an anti-biotin antibody conjugated with alkaline phosphatase (anti-biotin mAb AP conjugated, Sigma) was added and reacted at 37 ° C. for 1 hour. After washing three times, 50 μl of pNPP (Sigma.) Was added to the substrate, and the absorbance was measured at 405 nm.
결과는 도 13에 나타내었다.The results are shown in FIG.
도 13에 나타난 바와 같이, AU11 또는 AU12의 농도가 증가할수록 HW1의 항원에 대한 결합력이 약해졌다. 따라서, AU11 또는 AU12는 HW1과 동일한 항원결합부위를 가진다는 것을 알 수 있다.As shown in FIG. 13, as the concentration of AU11 or AU12 increased, the binding strength of HW1 to the antigen was weakened. Thus, it can be seen that AU11 or AU12 has the same antigen binding site as HW1.
실험예 5Experimental Example 5 : DR5 및 유사항원에 대한 AU11과 AU12의 교차반응성 분석 : Cross-Reactivity Analysis of AU11 and AU12 for DR5 and Amusement Park
표적항원인 DR5 및 다른 유사항원인 DR4, DcR1, DcR2에 대한 AU11 및 AU12의 교차반응성을 확인하기 위하여, ELISA를 수행하였다.ELISA was performed to confirm the cross-reactivity of AU11 and AU12 against DR5, the target antigen, and DR4, DcR1, DcR2, the other antigens.
구체적으로는, ELISA용 96-웰 플레이트에 항원, 즉 DR5, DR4, DcR1 및 DcR2를 50㎕(20㎍/㎖) 씩 코팅한 후 37℃에서 1시간 동안 배양하였다. PBST(pH 7.4, with 0.1% Tween 20) 300㎕로 3회 세척한 후 5% 탈지우유(skim milk)가 함유된 PBST(pH 7.4, with 0.1% Tween 20) 300㎕를 넣어 상온에서 1시간 동안 배양하고, 각 항체 AU11, AU12, 및 양성대조군으로서 모든 수용체에 결합할 수 있는 리간드인 TRAIL를 각각 50㎕(3㎍/㎖)씩 넣어 37℃에서 1시간 동안 배양하였다. PBST 300㎕로 3번 세척한 후 쥐 유래의 항 FLAG-TAG(mouse anti-FLAG tag antibody, sigma) 항체를 첨가하여 37℃에서 1시간 동안 배양하고, 다시 PBST 200㎕로 3번 세척한 다음, 토끼 유래의 항-mouse mAb AP conjugated(pierce) 항체를 첨가하여 37℃에서 1시간 동안 배양하였다. PBST 300㎕로 3번 세척한 이후 기질로 pNPP(Sigma.)를 50㎕ 첨가하여 405㎚에서 흡광도를 측정하였다.Specifically, antigens, namely DR5, DR4, DcR1 and DcR2, were coated in 50 μl (20 μg / ml) in 96-well plates for ELISA, followed by incubation at 37 ° C. for 1 hour. After washing three times with 300 μl of PBST (pH 7.4, with 0.1% Tween 20), 300 μl of PBST (pH 7.4, with 0.1% Tween 20) containing 5% skim milk was added for 1 hour at room temperature. Each of the antibodies AU11, AU12, and TRAIL, a ligand capable of binding to all receptors, was added to each of the receptors, and 50 µl (3 µg / ml) of each antibody was incubated at 37 ° C for 1 hour. After washing three times with 300 μl of PBST, incubated at 37 ° C. for 1 hour by adding a mouse anti-FLAG-TAG (mouse anti-FLAG tag antibody, sigma) antibody derived from rats, and then washed three times with 200 μl of PBST. Rabbit-derived anti-mouse mAb AP conjugated (pierce) antibody was added and incubated at 37 ° C. for 1 hour. After washing three times with 300 μl of PBST, 50 μl of pNPP (Sigma.) Was added as a substrate, and the absorbance was measured at 405 nm.
결과는 도 14에 나타내었다.The results are shown in FIG.
도 14에 나타난 바와 같이, AU11 및 AU12는 표적항원인 DR5에만 특이적으로 결합하였으며, 양성대조군인 TRAIL은 DR5, DR4, DcR1 및 DcR2에 모두 결합함을 확인하였다. 따라서, 본 발명의 항 DR5 항체는 DR5 항원에 특이적으로 결합하는 항체임을 알 수 있다.As shown in Figure 14, AU11 and AU12 specifically bound only to the target antigen DR5, it was confirmed that the positive control TRAIL binds to all DR5, DR4, DcR1 and DcR2. Thus, it can be seen that the anti-DR5 antibody of the present invention is an antibody that specifically binds to the DR5 antigen.
실험예 6Experimental Example 6 : DR5 및 DR5 단편에 대한 TRAIL, HW1, AU11, AU12 간의 결합 여부 규명 : Determination of binding between TRAIL, HW1, AU11, and AU12 for DR5 and DR5 fragments
DR5는 구조적으로 CRD(cysteine-rich domain)라고 불리는 각각 세 개의 이황화 결합을 가지는 서브도메인(sub-domain)들로 나누어질 수 있다. 이 서브도메인들 중 특히 CRD2(잔기 97~137)와 CRD3(잔기 139~180)는 TRAIL과의 결합에 결정적 역할을 하는 50s loop와 90s loop 지역을 각각 포함하고 있다(Hymowitz 등, Mol. Cell., 4:563-571, 1999). 따라서, CRD2 및 CRD3, 그리고 DR5를 대장균 BL21(DE3)에서 정제하여 TRAIL, HW1, AU11, AU12와의 결합 여부를 ELISA를 통하여 규명하였다. 구체적으로는, ELISA용 96-웰 플레이트에 항원 즉 CRD2, CRD3, DR5를 각각 50㎕(20 ㎍/㎖) 씩 코팅한 후 37℃에서 1시간 동안 배양하였다. PBST(pH 7.4 with 0.1% Tween 20) 300㎕로 3회 세척한 후 5% 탈지우유가 함유된 PBST(pH 7.4 with 0.1% Tween 20) 200㎕를 넣어 상온에서 1시간 동안 배양하고, 각 항체 HW1, AU11, AU12, TRAIL를 각각 50㎕ (20 ㎍/㎖)씩 넣어 37℃에서 1시간 동안 배양하였다. PBST 300㎕로 3번 세척한 후 쥐 유래의 항 FLAG-TAG(mouse anti-FLAG tag antibody, sigma) 항체를 첨가하여 37℃에서 1시간 동안 배양하고, 다시 PBST 300㎕로 3번 세척한 다음, 토끼 유래의 항-mouse mAb AP conjugated(pierce) 항체를 첨가하여 37℃에서 1시간 동안 배양하였다. PBST 300㎕로 3번 세척한 이후 기질로 pNPP(Sigma.)를 50㎕ 첨가하여 405㎚에서 흡광도를 측정하였다.DR5 can be subdivided into sub-domains that each have three disulfide bonds, structurally called cysteine-rich domains (CRDs). Among these subdomains, CRD2 (residues 97-137) and CRD3 (residues 139-180) each contain 50s loop and 90s loop regions that are critical for binding to TRAIL (Hymowitz et al . , Mol. Cell. , 4: 563-571, 1999). Therefore, CRD2, CRD3, and DR5 were purified from E. coli BL21 (DE3) to determine whether they bind to TRAIL, HW1, AU11, and AU12 by ELISA. Specifically, 50 μl (20 μg / ml) of antigens, namely CRD2, CRD3, and DR5, were coated on a 96-well plate for ELISA, followed by incubation at 37 ° C. for 1 hour. After washing three times with 300 μl of PBST (pH 7.4 with 0.1% Tween 20), 200 μl of PBST (pH 7.4 with 0.1% Tween 20) containing 5% skim milk was incubated at room temperature for 1 hour, and each antibody HW1 , AU11, AU12, and TRAIL were added to 50 µl (20 µg / ml) and incubated at 37 ° C for 1 hour. After washing three times with 300 μl of PBST, incubated at 37 ° C. for 1 hour by adding an anti-FLAG-TAG (mouse anti-FLAG tag antibody, sigma) antibody derived from a mouse, and then washed three times with 300 μl of PBST. Rabbit-derived anti-mouse mAb AP conjugated (pierce) antibody was added and incubated at 37 ° C. for 1 hour. After washing three times with 300 μl PBST, 50 μl of pNPP (Sigma.) Was added as a substrate, and the absorbance was measured at 405 nm.
결과는 도 15에 나타내었다.The results are shown in FIG.
도 15에 나타난 바와 같이, TRAIL은 CRD3보다 CRD2에 상대적으로 강한 결합력을 보였으나, HW1, AU11 및 AU12는 CRD2, CRD3 모두 유사한 결합력으로 각 항원에 결합하였다. 따라서, TRAIL-DR5 간의 결합 및 HW1, AU11, AU12 - DR5 간의 결합에 있어서 CRD2 및 CRD3가 TRAIL 또는 항체에 미치는 영향 정도가 다르다는 것을 알 수 있다.As shown in FIG. 15, TRAIL showed a stronger binding force to CRD2 than CRD3, but HW1, AU11 and AU12 bound to each antigen with similar binding force for both CRD2 and CRD3. Therefore, it can be seen that the degree of effect of CRD2 and CRD3 on TRAIL or antibodies differs in the binding between TRAIL-DR5 and the binding between HW1, AU11, and AU12-DR5.
실험예 7Experimental Example 7 : 항체 AU11 및 AU12의 암세포에서의 세포사멸 유도 가능성 확인 : Confirmation of apoptosis induction of antibody AU11 and AU12 in cancer cells
항체 AU11 및 AU12의 다양한 암세포에서의 세포사멸 유도 가능성을 확인하기 위하여, 하기와 같은 실험을 수행하였다.In order to confirm the possibility of inducing apoptosis in various cancer cells of the antibodies AU11 and AU12, the following experiment was performed.
암세포로는 인간 대장암 세포주(human colon cancer cell line)인 HCT116과 인간 신경종양 세포주(human glioma cancer cell line)인 U87MG를 사용하였다. 액체 질소에 보관되어 있는 세포주들을 꺼내어 37℃에서 빠르게 녹인 후 원심분리하여 냉동보관 배지를 제거하여 얻은 세포를 배양 배지인 10% FBS, 100unit/㎖ 페니실린 및 100㎍/㎖ 스트렙토마이신이 함유된 DMEM 배지(Welgene 사)를 사용하여 배양 플라스크에 넣고 배양하였다. 3일 안정화 이후, TE 완충액 1㎖를 3~5분 처리한 후, 10% FBS가 함유된 DMEM 배지 5㎖로 TE 완충액의 반응을 정지시킨 후, 원심분리하여 세포를 회수하였다. 회수된 세포들은 동일한 배양 배지로 각각 재현탁한 후 96-웰 플레이트에 웰당 세포를 1×104 (100 ㎕)씩 분주하여 24시간 동안 배양한 후 MTT 분석에 사용하였다.As cancer cells, HCT116, a human colon cancer cell line, and U87MG, a human glioma cancer cell line, were used. The cell lines stored in liquid nitrogen were taken out, dissolved rapidly at 37 ° C., and centrifuged to remove the cryopreservation medium. The cells obtained from the culture medium were cultured with DMEM medium containing 10% FBS, 100unit / ml penicillin and 100µg / ml streptomycin. (Welgene) was used to cultivate the culture flask. After 3 days of stabilization, 1 ml of TE buffer was treated for 3 to 5 minutes, and then the reaction of TE buffer was stopped with 5 ml of DMEM medium containing 10% FBS, followed by centrifugation to recover the cells. The recovered cells were each resuspended in the same culture medium, and the cells per well were dispensed in 96-well plates by 1 × 10 4 (100 μl), incubated for 24 hours, and used for MTT analysis.
결과는 도 16에 나타내었다.The results are shown in FIG.
도 16에 나타난 바와 같이, 인간 대장암 세포주인 HCT116과 인간 신경종양 세포주인 U87MG에서 AU11, AU12 및 대조군인 HW1을 같은 농도에서 같은 시간 동안 배양하였을 때, AU11과 AU12는 모두 세포사멸효능을 보였으며, HW1의 세포사멸효능과 비교하여 세포사멸이 더 효율적으로 일어남을 확인하였다.As shown in FIG. 16, when AU11, AU12, and control HW1 were cultured at the same concentration in HCT116, a human colon cancer cell line, and U87MG, a human neurotumor cell line, at the same concentration, both AU11 and AU12 showed apoptosis effect. , Compared with the apoptosis effect of HW1 confirmed that apoptosis occurs more efficiently.
실험예 8Experimental Example 8 : 항체 AU11 및 AU12의 세포사멸 경로 기작 분석 : Apoptosis Pathway Assay of Antibodies AU11 and AU12
항체 AU11 및 AU12의 세포사멸 기작을 분석하기 위하여, 하기와 같은 실험을 수행하였다.In order to analyze the apoptosis mechanisms of the antibodies AU11 and AU12, the following experiment was performed.
인간 대장암 세포주인 HCT116과 인간 신경종양 세포주인 U87MG를 96-웰 플레이트에 1×104개 첨가하고 10% FBS가 첨가되어 있는 DMEM 배지 100㎕를 넣어 5% CO2, 37℃에서 2일 동안 배양하여 안정화시켰다. 그 다음, Z-VAD(Pan-caspase inhibitor, Santacruz) 10μM, SP600125(JNK inhibitor, calbiochem) 10μM, SB203580(p38 inhibitor, calbiochem) 10μM, 3-MA 100(Autophagic cell death inhibitor, sigma) 10μM, Chloroguine(Autophagic cell death inhibitor, sigma) 10μM을 넣고 1시간 동안 전처리하였다. AU11과 AU12를 각각 30㎍/㎖ 씩 첨가한 후, 5% CO2, 37℃에서 40시간 동안 배양하여 MTT-assay를 통하여 세포사멸 정도를 측정하였다.Human colon cancer cell line HCT116 and human neural tumor cell line, U87MG to a 96-well plate at 1 × 10 4 gae added and for 10% FBS DMEM medium is put 100㎕ is added 5% CO 2, at 37 ℃ 2 il Culture was stabilized. Next, 10 μM of Z-VAD (Pan-caspase inhibitor, Santacruz), 10 μM of SP600125 (JNK inhibitor, calbiochem), 10 μM of SB203580 (p38 inhibitor, calbiochem), 10 μM of 3-MA 100 (Autophagic cell death inhibitor, sigma), Chloroguine ( Autophagic cell death inhibitor (sigma) 10μM was added and pretreated for 1 hour. AU11 and AU12 were added to 30µg / ml, respectively, and then cultured at 5% CO 2 and 37 ° C for 40 hours to measure the degree of cell death through MTT-assay.
결과는 도 17에 나타내었다.The results are shown in FIG.
도 17에 나타난 바와 같이, AU11 및 AU12는 인간 대장암 세포주인 HCT116과 인간 신경종양 세포주인 U87MG에서 SP600125(JNK inhibitor), 3-MA 및 Chloroquine 존재 하에서는 세포사멸능력이 현저하게 감소하였으며, Z-VAD와 SB203580(p38 inhibitor) 존재 하에서는 세포사멸 정도의 변화가 없었다. 상기 결과는 scFv 항체 HW1과 특성이 동일하다. 따라서, 친화도 및 안정성이 향상된 AU11과 AU12는 자식작용에 의하여 세포사멸을 유도함을 알 수 있다.As shown in Figure 17, AU11 and AU12 significantly reduced apoptosis in the presence of SP600125 (JNK inhibitor), 3-MA and Chloroquine in human colon cancer cell line HCT116 and human neuronal tumor cell line U87MG, Z-VAD There was no change in the degree of apoptosis in the presence of and SB203580 (p38 inhibitor). The results are the same as the scFv antibody HW1. Therefore, it can be seen that AU11 and AU12 having improved affinity and stability induce apoptosis by progeny.
실험예 9Experimental Example 9 : 항체 AU11 및 AU12에 의한 LC3-GFP 결집 : LC3-GFP aggregation by antibodies AU11 and AU12
자식작용에 의한 세포사멸의 전형적인 특징은 세포 내 LC3 단백질이 자식작용 액포(Vacuole) 막 또는 내부에 모이는 것이다. AU11과 AU12의 처리에 의해서 이러한 자식작용의 특징을 보이는지 확인하기 위하여, LC3-GFP를 발현하는 U87MG 암세포주(TRAIL-resistant, human glioma cell line)를 24-웰 플레이트에 4×104개의 세포와 10% FBS가 포함되어 있는 DMEM 500㎕를 첨가하여 24시간 동안 5% CO2, 37℃에서 2일 동안 배양하여 안정화시켰다. 그 다음 AU11과 AU12를 각각 30㎍/㎖ 씩 첨가하여 30시간 동안 5% CO2, 37℃에서 배양하고, 형광현미경(Axiovert 200M, Carl Zeiss 사, 독일)으로 관찰하였다.A typical feature of apoptosis by apoptosis is the accumulation of intracellular LC3 proteins in or on the vacuole membrane. In order to determine whether these traits were characterized by treatment with AU11 and AU12, U87MG cancer cell lines (TRAIL-resistant, human glioma cell lines) expressing LC3-GFP were added to 4 × 10 4 cells in 24-well plates. 500 μl of DMEM containing 10% FBS was added thereto, followed by stabilization by incubating at 5% CO 2 for 24 hours at 37 ° C. for 2 days. Then, AU11 and AU12 were added to 30 µg / ml, respectively, and incubated at 5% CO 2 , 37 ° C for 30 hours, and observed with a fluorescence microscope (Axiovert 200M, Carl Zeiss, Germany).
결과는 도 18에 나타내었다.The results are shown in FIG.
도 18에 나타난 바와 같이, AU11과 AU12를 처리한 U87MG 암세포주에서 LC3-GFP가 자식작용 액포막 및 내부에 결집되는 것을 관찰하였다. 따라서, AU11과 AU12는 자식작용에 의하여 세포사멸을 유도함을 알 수 있다.As shown in FIG. 18, it was observed that LC3-GFP aggregated in the child acting vesicle membrane and the inside of U87MG cancer cell lines treated with AU11 and AU12. Thus, it can be seen that AU11 and AU12 induce apoptosis by child action.
하기에 본 발명의 조성물을 위한 제제예를 예시한다.Examples of preparations for the compositions of the present invention are illustrated below.
제제예 1Formulation Example 1 : 산제의 제조 : Preparation of powder
항 DR5 항체 0.1 g0.1 g of anti-DR5 antibody
유당 1.5 gLactose 1.5 g
탈크 0.5 gTalc 0.5 g
상기의 성분들을 혼합하고 기밀포에 충진하여 산제를 제조하였다.The above ingredients were mixed and filled in an airtight cloth to prepare a powder.
제제예 2Formulation Example 2 : 정제의 제조 : Preparation of Tablet
항 DR5 항체 0.1 g0.1 g of anti-DR5 antibody
락토오스 7.9 gLactose 7.9 g
결정성 셀룰로오스 1.5 g1.5 g of crystalline cellulose
마그네슘 스테아레이트 0.5 g0.5 g of magnesium stearate
상기의 성분들을 혼합한 후 직타법(direct tableting method)으로 정제를 제조하였다.After mixing the above components, a tablet was prepared by a direct tableting method.
제제예 3Formulation Example 3 : 캡슐제의 제조: Preparation of Capsule
항 DR5 항체 0.1 g0.1 g of anti-DR5 antibody
옥수수전분 5 g5 g of corn starch
카복시 셀룰로오스 4.9 g4.9 g of carboxy cellulose
상기의 성분들을 혼합하여 분말을 제조한 후, 상기 분말을 통상의 캡슐제의 제조방법에 따라 경질 캡슐에 충전하여 캡슐제를 제조하였다.After the powder was prepared by mixing the above components, the powder was filled in a hard capsule according to a conventional method for preparing a capsule to prepare a capsule.
제제예 4Formulation Example 4 : 주사제의 제조 : Preparation of Injection
항 DR5 항체 0.02~0.2 g0.02-0.2 g of anti-DR5 antibody
주사용 멸균 증류수 적량Appropriate sterile distilled water for injection
pH 조절제 적량pH adjuster
안정화제 적량Stabilizer
통상의 주사제의 제조방법에 따라 1 앰플 당(2㎖) 상기의 성분 함량으로 제조하였다.According to the conventional method for preparing an injection, the amount of the above-mentioned ingredient was prepared per ampoule (2 ml).
제제예 5Formulation Example 5 : 액제의 제조 : Manufacture of liquid
항 DR5 항체 0.1 g0.1 g of anti-DR5 antibody
이성화당 10 g10 g of isomerized sugar
만니톨 5 g5 g of mannitol
정제수 적량Purified water
통상의 액제의 제조방법에 따라 정제수에 각각의 성분을 가하여 용해시키고, 레몬향을 적량 가한 다음 상기의 성분을 혼합하였다. 그 다음 정제수를 가하여 전체 100㎖로 조절한 후 갈색병에 충전하고 멸균시켜 액제를 제조하였다.Each component was added to and dissolved in purified water according to the conventional method for preparing a liquid, and lemon flavor was added appropriately, followed by mixing the above components. Then, purified water was added thereto to adjust the total volume to 100 ml, and filled into a brown bottle and sterilized to prepare a liquid.
본 발명에 따른 항 DR5 항체는 기존의 항 DR5 항체보다 더 높은 친화도로 DR5 단백질에 특이적으로 결합하고 안정성이 상대적으로 높으며 DR5를 발현하는 TRAIL-민감성 암세포 또는 DR5를 발현하는 TRAIL-저항성 암세포에 대해 자식작용에 의하여 세포사멸을 효과적으로 유도함으로써, DR5 발현에 의해 야기되는 암의 예방 또는 치료에 유용하게 사용될 수 있다. 또한, 본 발명에 따른 항 DR5 항체는 일회 투여량의 감소에 의한 비용 절감 및 효율성 향상 등의 효과를 기대할 수 있다.The anti-DR5 antibody according to the present invention has a higher affinity than a conventional anti-DR5 antibody, specifically binds to a DR5 protein, has a relatively high stability, and is a TRAIL-sensitive cancer cell expressing DR5 or a TRAIL-resistant cancer cell expressing DR5. By effectively inducing apoptosis by progeny, it can be usefully used for the prevention or treatment of cancer caused by DR5 expression. In addition, the anti-DR5 antibody according to the present invention can be expected to reduce costs and improve efficiency by reducing the single dose.
<110> AJOU UNIVERSITY INDUSTRY-ACADEMIC COOPERATION FOUNDATION<110> AJOU UNIVERSITY INDUSTRY-ACADEMIC COOPERATION FOUNDATION
<120> Anti-death receptor 5 antibody with improved affinity and<120> Anti-death receptor 5 antibody with improved affinity and
stability, and composition for preventing or treating cancer         stability, and composition for preventing or treating cancer
comprising the same         configuring the same
<130> P09-07023<130> P09-07023
<150> KR10-2008-0104737<150> KR10-2008-0104737
<151> 2008-10-24<151> 2008-10-24
<160> 40<160> 40
<170> KopatentIn 1.71<170> KopatentIn 1.71
<210> 1<210> 1
<211> 10<211> 10
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> VH-CDR1<223> VH-CDR1
<400> 1<400> 1
Gly Asp Ser Val Ser Ser Thr Thr Val AlaGly Asp Ser Val Ser Ser Thr Thr Val Ala
1 5 10  1 5 10
<210> 2<210> 2
<211> 10<211> 10
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> VH-CDR2<223> VH-CDR2
<400> 2<400> 2
Ile Tyr His Arg Ser Tyr Trp Asp Lys TyrIle Tyr His Arg Ser Tyr Trp Asp Lys Tyr
1 5 10  1 5 10
<210> 3<210> 3
<211> 13<211> 13
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> VH-CDR3<223> VH-CDR3
<400> 3<400> 3
Ala Arg Glu Gly Ser Gly His Tyr Gly Ala Phe Asp IleAla Arg Glu Gly Ser Gly His Tyr Gly Ala Phe Asp Ile
1 5 10   1 5 10
<210> 4<210> 4
<211> 7<211> 7
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> VL-CDR1<223> VL-CDR1
<400> 4<400> 4
Gln Ser Val Ser Ser Ser HisGln Ser Val Ser Ser Ser His
1 5   1 5
<210> 5<210> 5
<211> 7<211> 7
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> VL-CDR2<223> VL-CDR2
<400> 5<400> 5
Gly Ala Ser Ser Arg Ala ThrGly Ala Ser Ser Arg Ala Thr
1 5   1 5
<210> 6<210> 6
<211> 11<211> 11
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> VL-CDR3<223> VL-CDR3
<400> 6<400> 6
Gln Gln Arg Ala Asn Asp Phe Pro Arg Ala ValGln Gln Arg Ala Asn Asp Phe Pro Arg Ala Val
1 5 10   1 5 10
<210> 7<210> 7
<211> 249<211> 249
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> amino acid sequence of AU11 scFv<223> amino acid sequence of AU11 scFv
<400> 7<400> 7
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly GlyGlu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15   1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asp Ser Val Ser Ser ThrSer Leu Arg Leu Ser Cys Ala Ala Ser Gly Asp Ser Val Ser Ser Thr
20 25 30              20 25 30
Thr Val Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu GluThr Val Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu
35 40 45          35 40 45
Trp Val Ser Ala Ile Tyr His Arg Ser Tyr Trp Asp Lys Tyr Tyr AlaTrp Val Ser Ala Ile Tyr His Arg Ser Tyr Trp Asp Lys Tyr Tyr Ala
50 55 60      50 55 60
Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys AsnAsp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn
65 70 75 80  65 70 75 80
Thr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala ValThr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val
85 90 95                  85 90 95
Tyr Tyr Cys Ala Arg Glu Gly Ser Gly His Tyr Gly Ala Phe Asp IleTyr Tyr Cys Ala Arg Glu Gly Ser Gly His Tyr Gly Ala Phe Asp Ile
100 105 110             100 105 110
Trp Gly Gln Gly Thr Leu Val Thr Val Ser Ser Gly Gly Gly Gly SerTrp Gly Gln Gly Thr Leu Val Thr Val Ser Ser Gly Gly Gly Gly Ser
115 120 125         115 120 125
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Asp Ile Val Leu Thr GlnGly Gly Gly Gly Ser Gly Gly Gly Gly Ser Asp Ile Val Leu Thr Gln
130 135 140     130 135 140
Ser Pro Ala Thr Leu Ser Leu Ser Pro Gly Glu Arg Ala Thr Leu SerSer Pro Ala Thr Leu Ser Leu Ser Pro Gly Glu Arg Ala Thr Leu Ser
145 150 155 160 145 150 155 160
Cys Arg Ala Ser Gln Ser Val Ser Ser Ser His Leu Ala Trp Tyr GlnCys Arg Ala Ser Gln Ser Val Ser Ser Ser His Leu Ala Trp Tyr Gln
165 170 175                 165 170 175
Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu Ile Tyr Gly Ala Ser SerGln Lys Pro Gly Gln Ala Pro Arg Leu Leu Ile Tyr Gly Ala Ser Ser
180 185 190             180 185 190
Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser Gly Ser Gly Ser Gly ThrArg Ala Thr Gly Ile Pro Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr
195 200 205         195 200 205
Asp Phe Thr Leu Thr Ile Ser Ser Leu Glu Pro Glu Asp Phe Ala ValAsp Phe Thr Leu Thr Ile Ser Ser Leu Glu Pro Glu Asp Phe Ala Val
210 215 220     210 215 220
Tyr Tyr Cys Gln Gln Arg Ala Asn Asp Phe Pro Arg Ala Val Phe GlyTyr Tyr Cys Gln Gln Arg Ala Asn Asp Phe Pro Arg Ala Val Phe Gly
225 230 235 240 225 230 235 240
Gln Gly Thr Lys Val Glu Ile Lys ArgGln Gly Thr Lys Val Glu Ile Lys Arg
245                 245
<210> 8<210> 8
<211> 747<211> 747
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> nucleotide sequence of AU11 scFv223 nucleotide sequence of AU11 scFv
<400> 8<400> 8
gaggtgcagc tggtggagtc tgggggaggc ctggtacagc ctggagggtc cctgagactc 60gaggtgcagc tggtggagtc tgggggaggc ctggtacagc ctggagggtc cctgagactc 60
tcctgtgcag cctctggaga cagtgtctct agcaccactg ttgccatgag ctgggtccgc 120tcctgtgcag cctctggaga cagtgtctct agcaccactg ttgccatgag ctgggtccgc 120
caggctccag ggaaggggct ggagtgggtc tcagcgatct atcacaggtc gtattgggac 180caggctccag ggaaggggct ggagtgggtc tcagcgatct atcacaggtc gtattgggac 180
aagtactatg cggactccgt gaagggccgg ttcaccatct ccagagacaa ttccaagaac 240aagtactatg cggactccgt gaagggccgg ttcaccatct ccagagacaa ttccaagaac 240
accctgtatc tgcaaatgaa cagcctgaga gccgaggaca cggctgtgta ttactgtgca 300accctgtatc tgcaaatgaa cagcctgaga gccgaggaca cggctgtgta ttactgtgca 300
agagagggta gtggccacta tggggctttt gatatctggg gccagggaac cctggtcacc 360agagagggta gtggccacta tggggctttt gatatctggg gccagggaac cctggtcacc 360
gtttcttcag gtggaggcgg ttcaggcgga ggtggcagcg gcggtggcgg gtcggatatt 420gtttcttcag gtggaggcgg ttcaggcgga ggtggcagcg gcggtggcgg gtcggatatt 420
gtattgaccc agtctccagc caccctgtct ttgtctccag gggaaagagc caccctctcc 480gtattgaccc agtctccagc caccctgtct ttgtctccag gggaaagagc caccctctcc 480
tgcagggcca gtcagagtgt tagcagcagc cacttagcct ggtaccagca gaaacctggc 540tgcagggcca gtcagagtgt tagcagcagc cacttagcct ggtaccagca gaaacctggc 540
caggctccca ggctcctcat ctatggtgca tccagcaggg ccactggcat cccagacagg 600caggctccca ggctcctcat ctatggtgca tccagcaggg ccactggcat cccagacagg 600
ttcagtggca gtgggtctgg gacagacttc actctcacca tcagcagcct agagcctgaa 660ttcagtggca gtgggtctgg gacagacttc actctcacca tcagcagcct agagcctgaa 660
gattttgcag tttattactg tcagcagcgt gcaaacgatt tcccgcgggc ggtcttcggc 720gattttgcag tttattactg tcagcagcgt gcaaacgatt tcccgcgggc ggtcttcggc 720
caagggacaa aggtggaaat caaacgt 747caagggacaa aggtggaaat caaacgt 747
<210> 9<210> 9
<211> 51<211> 51
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer<223> forward primer
<400> 9<400> 9
ggtggtggtg gttctggtgg tggtggttct ggtggtggtg gttctgctag c 51ggtggtggtg gttctggtgg tggtggttct ggtggtggtg gttctgctag c 51
<210> 10<210> 10
<211> 51<211> 51
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse primer<223> reverse primer
<400> 10<400> 10
gatctcgagc tattacaagt cctcttcaga aataagcttt tgttcggatc c 51gatctcgagc tattacaagt cctcttcaga aataagcttt tgttcggatc c 51
<210> 11<210> 11
<211> 72<211> 72
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer<223> forward primer
<400> 11<400> 11
ggccttgagt ggctgggaag gnnnnnnnnn nnnnnnnnnn nnnnnnnnnn ntatgcagta 60ggccttgagt ggctgggaag gnnnnnnnnn nnnnnnnnnn nnnnnnnnnn ntatgcagta 60
tctgtgaaaa gt 72tctgtgaaaa gt 72
<210> 12<210> 12
<211> 21<211> 21
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse primer<223> reverse primer
<400> 12<400> 12
ccttcccagc cactcaaggc c 21ccttcccagc cactcaaggc c 21
<210> 13<210> 13
<211> 81<211> 81
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer<223> forward primer
<400> 13<400> 13
gacacggccg tctattactg tnnnnnnnnn nnnnnnnnnn nnnnnnnnnn nnnnnnnnnn 60gacacggccg tctattactg tnnnnnnnnn nnnnnnnnnnnn nnnnnnnnnn nnnnnnnnnn 60
tggggccaag ggaccacggt c 81tggggccaag ggaccacggt c 81
<210> 14<210> 14
<211> 21<211> 21
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse primer<223> reverse primer
<400> 14<400> 14
acagtaatag acggccgtgt c 21acagtaatag acggccgtgt c 21
<210> 15<210> 15
<211> 75<211> 75
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer<223> forward primer
<400> 15<400> 15
gattttgcag tttattactg tnnnnnnnnn nnnnnnnnnn nnnnnnnnnn nnnnttcggc 60gattttgcag tttattactg tnnnnnnnnn nnnnnnnnnnnn nnnnnnnnnn nnnnttcggc 60
caagggacac gactg 75caagggacac gactg 75
<210> 16<210> 16
<211> 21<211> 21
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse primer<223> reverse primer
<400> 16<400> 16
acagtaataa actgcaaaat c 21acagtaataa actgcaaaat c 21
<210> 17<210> 17
<211> 22<211> 22
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward sequencing primer<223> forward sequencing primer
<400> 17<400> 17
gttccagact acgctctgca gg 22gttccagact acgctctgca gg 22
<210> 18<210> 18
<211> 24<211> 24
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse sequencing primer<223> reverse sequencing primer
<400> 18<400> 18
gattttgtta catctacact gttg 24gattttgtta catctacact gttg 24
<210> 19<210> 19
<211> 21<211> 21
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer<223> forward primer
<400> 19<400> 19
gattttgcag tttattactg t 21gattttgcag tttattactg t 21
<210> 20<210> 20
<211> 159<211> 159
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer for manufacturing AU11<223> forward primer for manufacturing AU11
<400> 20<400> 20
ttcgctagcg aggtgcagct ggtggagtct gggggaggcc tggtacagcc tggagggtcc 60ttcgctagcg aggtgcagct ggtggagtct gggggaggcc tggtacagcc tggagggtcc 60
ctgagactct cctgtgcagc ctctggagac agtgtctcta gcaccactgt tgccatgagc 120ctgagactct cctgtgcagc ctctggagac agtgtctcta gcaccactgt tgccatgagc 120
tgggtccgcc aggctccagg gaaggggctg gagtgggtc 159tgggtccgcc aggctccagg gaaggggctg gagtgggtc 159
<210> 21<210> 21
<211> 168<211> 168
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse primer for manufacturing AU11<223> reverse primer for manufacturing AU11
<400> 21<400> 21
acagtaatac acagccgtgt cctcggctct caggctgttc atttgcagat acagggtgtt 60acagtaatac acagccgtgt cctcggctct caggctgttc atttgcagat acagggtgtt 60
cttggaattg tctctggaga tggtgaaccg gcccttcacg gagtccgcat agtaattata 120cttggaattg tctctggaga tggtgaaccg gcccttcacg gagtccgcat agtaattata 120
ccacttcgac ctataataga tcgctgagac ccactccagc cccttccc 168ccacttcgac ctataataga tcgctgagac ccactccagc cccttccc 168
<210> 22<210> 22
<211> 24<211> 24
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer for manufacturing AU11<223> forward primer for manufacturing AU11
<400> 22<400> 22
gaggacacgg ctgtgtatta ctgt 24gaggacacgg ctgtgtatta ctgt 24
<210> 23<210> 23
<211> 75<211> 75
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse primer for manufacturing AU11<223> reverse primer for manufacturing AU11
<400> 23<400> 23
ttcccctgga gacaaagaca gggtggctgg agactgggtc aatacaatat ccgacccgcc 60ttcccctgga gacaaagaca gggtggctgg agactgggtc aatacaatat ccgacccgcc 60
accgccgctg ccacc 75accgccgctg ccacc 75
<210> 24<210> 24
<211> 24<211> 24
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer for manufacturing AU11<223> forward primer for manufacturing AU11
<400> 24<400> 24
accctgtctt tgtctccagg ggaa 24accctgtctt tgtctccagg ggaa 24
<210> 25<210> 25
<211> 51<211> 51
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse primer for manufacturing AU11<223> reverse primer for manufacturing AU11
<400> 25<400> 25
cgtggatcca cgtttgattt ccacctttgt cccttggccg aagaccgccc g 51cgtggatcca cgtttgattt ccacctttgt cccttggccg aagaccgccc g 51
<210> 26<210> 26
<211> 15<211> 15
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> amino acid sequence of (G4S)3 linker<223> amino acid sequence of (G4S) 3 linker
<400> 26<400> 26
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly SerGly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser
1 5 10 15  1 5 10 15
<210> 27<210> 27
<211> 111<211> 111
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> amino acid sequence of AU11 VL<223> amino acid sequence of AU11 VL
<400> 27<400> 27
Asp Ile Val Leu Thr Gln Ser Pro Ala Thr Leu Ser Leu Ser Pro GlyAsp Ile Val Leu Thr Gln Ser Pro Ala Thr Leu Ser Leu Ser Pro Gly
1 5 10 15   1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Val Ser Ser SerGlu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Val Ser Ser Ser
20 25 30              20 25 30
His Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu LeuHis Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu
35 40 45          35 40 45
Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe SerIle Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser
50 55 60      50 55 60
Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu GluGly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Glu
65 70 75 80  65 70 75 80
Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Arg Ala Asn Asp PhePro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Arg Ala Asn Asp Phe
85 90 95                  85 90 95
Pro Arg Ala Val Phe Gly Gln Gly Thr Lys Val Glu Ile Lys ArgPro Arg Ala Val Phe Gly Gln Gly Thr Lys Val Glu Ile Lys Arg
100 105 110             100 105 110
<210> 28<210> 28
<211> 123<211> 123
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> amino acid sequence of AU11 VH<223> amino acid sequence of AU11 VH
<400> 28<400> 28
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly GlyGlu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15   1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asp Ser Val Ser Ser ThrSer Leu Arg Leu Ser Cys Ala Ala Ser Gly Asp Ser Val Ser Ser Thr
20 25 30              20 25 30
Thr Val Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu GluThr Val Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu
35 40 45          35 40 45
Trp Val Ser Ala Ile Tyr His Arg Ser Tyr Trp Asp Lys Tyr Tyr AlaTrp Val Ser Ala Ile Tyr His Arg Ser Tyr Trp Asp Lys Tyr Tyr Ala
50 55 60      50 55 60
Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys AsnAsp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn
65 70 75 80  65 70 75 80
Thr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala ValThr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val
85 90 95                  85 90 95
Tyr Tyr Cys Ala Arg Glu Gly Ser Gly His Tyr Gly Ala Phe Asp IleTyr Tyr Cys Ala Arg Glu Gly Ser Gly His Tyr Gly Ala Phe Asp Ile
100 105 110             100 105 110
Trp Gly Gln Gly Thr Leu Val Thr Val Ser SerTrp Gly Gln Gly Thr Leu Val Thr Val Ser Ser
115 120         115 120
<210> 29<210> 29
<211> 33<211> 33
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer for manufacturing AU12<223> forward primer for manufacturing AU12
<400> 29<400> 29
ttcgctagcg aggtgcagct ggtggagtct ggg 33ttcgctagcg aggtgcagct ggtggagtct ggg 33
<210> 30<210> 30
<211> 24<211> 24
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse primer for manufacturing AU12<223> reverse primer for manufacturing AU12
<400> 30<400> 30
tggagactgg gtcatctgag agtc 24tggagactgg gtcatctgag agtc 24
<210> 31<210> 31
<211> 90<211> 90
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer for manufacturing AU12<223> forward primer for manufacturing AU12
<400> 31<400> 31
gactctcaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60gactctcaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60
atcacttgcc gggcaagtca gagcgttagc 90atcacttgcc gggcaagtca gagcgttagc 90
<210> 32<210> 32
<211> 45<211> 45
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse primer for manufacturing AU12<223> reverse primer for manufacturing AU12
<400> 32<400> 32
cttaggggct ttccctggtt tctgctgata ccaggctaag tggct 45cttaggggct ttccctggtt tctgctgata ccaggctaag tggct 45
<210> 33<210> 33
<211> 36<211> 36
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer for manufacturing AU12<223> forward primer for manufacturing AU12
<400> 33<400> 33
aaagccccta agctcctgat ctatggtgca tccagc 36aaagccccta agctcctgat ctatggtgca tccagc 36
<210> 34<210> 34
<211> 66<211> 66
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse primer for manufacturing AU12<223> reverse primer for manufacturing AU12
<400> 34<400> 34
gatggtgaga gtgaaatctg tcccagatcc actgccactg aaccttgatg ggaccccagt 60gatggtgaga gtgaaatctg tcccagatcc actgccactg aaccttgatg ggaccccagt 60
ggccct 66ggccct 66
<210> 35<210> 35
<211> 87<211> 87
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> forward primer for manufacturing AU12<223> forward primer for manufacturing AU12
<400> 35<400> 35
ttcactctca ccatcagcag tctgcaacct gaagattttg caacttacta ctgtcaacag 60ttcactctca ccatcagcag tctgcaacct gaagattttg caacttacta ctgtcaacag 60
cgtgcaaacg atttcccgcg ggcggtc 87cgtgcaaacg atttcccgcg ggcggtc 87
<210> 36<210> 36
<211> 24<211> 24
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> reverse primer for manufacturing AU12<223> reverse primer for manufacturing AU12
<400> 36<400> 36
cgtggatcca cgtttgattt ccac 24cgtggatcca cgtttgattt ccac 24
<210> 37<210> 37
<211> 111<211> 111
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> amino acid sequence of AU12 VL<223> amino acid sequence of AU12 VL
<400> 37<400> 37
Asp Ser Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val GlyAsp Ser Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly
1 5 10 15   1 5 10 15
Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Ser Val Ser Ser SerAsp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Ser Val Ser Ser Ser
20 25 30              20 25 30
His Leu Ala Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu LeuHis Leu Ala Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu
35 40 45          35 40 45
Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Val Pro Ser Arg Phe SerIle Tyr Gly Ala Ser Ser Arg Ala Thr Gly Val Pro Ser Arg Phe Ser
50 55 60      50 55 60
Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu GlnGly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln
65 70 75 80  65 70 75 80
Pro Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln Arg Ala Asn Asp PhePro Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln Arg Ala Asn Asp Phe
85 90 95                  85 90 95
Pro Arg Ala Val Phe Gly Gln Gly Thr Lys Val Glu Ile Lys ArgPro Arg Ala Val Phe Gly Gln Gly Thr Lys Val Glu Ile Lys Arg
100 105 110             100 105 110
<210> 38<210> 38
<211> 123<211> 123
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> amino acid sequence of AU12 VH<223> amino acid sequence of AU12 VH
<400> 38<400> 38
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly GlyGlu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15   1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asp Ser Val Ser Ser ThrSer Leu Arg Leu Ser Cys Ala Ala Ser Gly Asp Ser Val Ser Ser Thr
20 25 30              20 25 30
Thr Val Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu GluThr Val Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu
35 40 45          35 40 45
Trp Val Ser Ala Ile Tyr His Arg Ser Tyr Trp Asp Lys Tyr Tyr AlaTrp Val Ser Ala Ile Tyr His Arg Ser Tyr Trp Asp Lys Tyr Tyr Ala
50 55 60      50 55 60
Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys AsnAsp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn
65 70 75 80  65 70 75 80
Thr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala ValThr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val
85 90 95                  85 90 95
Tyr Tyr Cys Ala Arg Glu Gly Ser Gly His Tyr Gly Ala Phe Asp IleTyr Tyr Cys Ala Arg Glu Gly Ser Gly His Tyr Gly Ala Phe Asp Ile
100 105 110             100 105 110
Trp Gly Gln Gly Thr Leu Val Thr Val Ser SerTrp Gly Gln Gly Thr Leu Val Thr Val Ser Ser
115 120         115 120
<210> 39<210> 39
<211> 249<211> 249
<212> PRT<212> PRT
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> amino acid sequence of AU12 scFv<223> amino acid sequence of AU12 scFv
<400> 39<400> 39
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly GlyGlu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15   1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Asp Ser Val Ser Ser ThrSer Leu Arg Leu Ser Cys Ala Ala Ser Gly Asp Ser Val Ser Ser Thr
20 25 30              20 25 30
Thr Val Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu GluThr Val Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu
35 40 45          35 40 45
Trp Val Ser Ala Ile Tyr His Arg Ser Tyr Trp Asp Lys Tyr Tyr AlaTrp Val Ser Ala Ile Tyr His Arg Ser Tyr Trp Asp Lys Tyr Tyr Ala
50 55 60      50 55 60
Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys AsnAsp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn
65 70 75 80  65 70 75 80
Thr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala ValThr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val
85 90 95                  85 90 95
Tyr Tyr Cys Ala Arg Glu Gly Ser Gly His Tyr Gly Ala Phe Asp IleTyr Tyr Cys Ala Arg Glu Gly Ser Gly His Tyr Gly Ala Phe Asp Ile
100 105 110             100 105 110
Trp Gly Gln Gly Thr Leu Val Thr Val Ser Ser Gly Gly Gly Gly SerTrp Gly Gln Gly Thr Leu Val Thr Val Ser Ser Gly Gly Gly Gly Ser
115 120 125         115 120 125
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Asp Ser Gln Met Thr GlnGly Gly Gly Gly Ser Gly Gly Gly Gly Ser Asp Ser Gln Met Thr Gln
130 135 140     130 135 140
Ser Pro Ser Ser Leu Ser Ala Ser Val Gly Asp Arg Val Thr Ile ThrSer Pro Ser Ser Leu Ser Ala Ser Val Gly Asp Arg Val Thr Ile Thr
145 150 155 160 145 150 155 160
Cys Arg Ala Ser Gln Ser Val Ser Ser Ser His Leu Ala Trp Tyr GlnCys Arg Ala Ser Gln Ser Val Ser Ser Ser His Leu Ala Trp Tyr Gln
165 170 175                 165 170 175
Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Tyr Gly Ala Ser SerGln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Tyr Gly Ala Ser Ser
180 185 190             180 185 190
Arg Ala Thr Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly ThrArg Ala Thr Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr
195 200 205         195 200 205
Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro Glu Asp Phe Ala ThrAsp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro Glu Asp Phe Ala Thr
210 215 220     210 215 220
Tyr Tyr Cys Gln Gln Arg Ala Asn Asp Phe Pro Arg Ala Val Phe GlyTyr Tyr Cys Gln Gln Arg Ala Asn Asp Phe Pro Arg Ala Val Phe Gly
225 230 235 240 225 230 235 240
Gln Gly Thr Lys Val Glu Ile Lys ArgGln Gly Thr Lys Val Glu Ile Lys Arg
245                 245
<210> 40<210> 40
<211> 747<211> 747
<212> DNA<212> DNA
<213> Artificial Sequence<213> Artificial Sequence
<220><220>
<223> nucleotide sequence of AU12 scFv223 nucleotide sequence of AU12 scFv
<400> 40<400> 40
gaggtgcagc tggtggagtc tgggggaggc ctggtacagc ctggagggtc cctgagactc 60gaggtgcagc tggtggagtc tgggggaggc ctggtacagc ctggagggtc cctgagactc 60
tcctgtgcag cctctggaga cagtgtctct agcaccactg ttgccatgag ctgggtccgc 120tcctgtgcag cctctggaga cagtgtctct agcaccactg ttgccatgag ctgggtccgc 120
caggctccag ggaaggggct ggagtgggtc tcagcgatct atcacaggtc gtattgggac 180caggctccag ggaaggggct ggagtgggtc tcagcgatct atcacaggtc gtattgggac 180
aagtactatg cggactccgt gaagggccgg ttcaccatct ccagagacaa ttccaagaac 240aagtactatg cggactccgt gaagggccgg ttcaccatct ccagagacaa ttccaagaac 240
accctgtatc tgcaaatgaa cagcctgaga gccgaggaca cggctgtgta ttactgtgca 300accctgtatc tgcaaatgaa cagcctgaga gccgaggaca cggctgtgta ttactgtgca 300
agagagggta gtggccacta tggggctttt gatatctggg gccaagggac cctggtcacc 360agagagggta gtggccacta tggggctttt gatatctggg gccaagggac cctggtcacc 360
gtttcttcag gtggaggcgg ttcaggcgga ggtggcagcg gcggtggcgg gtcggactct 420gtttcttcag gtggaggcgg ttcaggcgga ggtggcagcg gcggtggcgg gtcggactct 420
cagatgaccc agtctccatc ctccctgtct gcatctgtag gagacagagt caccatcact 480cagatgaccc agtctccatc ctccctgtct gcatctgtag gagacagagt caccatcact 480
tgccgggcaa gtcagagcgt tagcagcagc cacttagcct ggtatcagca gaaaccaggg 540tgccgggcaa gtcagagcgt tagcagcagc cacttagcct ggtatcagca gaaaccaggg 540
aaagccccta agctcctgat ctatggtgca tccagcaggg ccactggggt cccatcaagg 600aaagccccta agctcctgat ctatggtgca tccagcaggg ccactggggt cccatcaagg 600
ttcagtggca gtggatctgg gacagatttc actctcacca tcagcagtct gcaacctgaa 660ttcagtggca gtggatctgg gacagatttc actctcacca tcagcagtct gcaacctgaa 660
gattttgcaa cttactactg tcaacagcgt gcaaacgatt tcccgcgggc ggtcttcggc 720gattttgcaa cttactactg tcaacagcgt gcaaacgatt tcccgcgggc ggtcttcggc 720
caagggacca aagtggaaat caaacgt 747caagggacca aagtggaaat caaacgt 747

Claims (17)

  1. 서열번호 7의 아미노산 서열을 갖는 것을 특징으로 하는 DR5에 특이적으로 결합하는 항 DR5 항체.An anti-DR5 antibody that specifically binds to DR5, having the amino acid sequence of SEQ ID NO: 7.
  2. 서열번호 39의 아미노산 서열을 갖는 것을 특징으로 하는 DR5에 특이적으로 결합하는 항 DR5 항체.An anti-DR5 antibody that specifically binds to DR5, having the amino acid sequence of SEQ ID NO.
  3. 제 1항 또는 제 2항에 있어서, 상기 항체는 상보성 결정부위 CDR1, CDR2 및 CDR3가 각각 서열번호 1, 2 및 3의 아미노산 서열을 갖는 중쇄 가변영역과 상보성 결정부위 CDR1, CDR2 및 CDR3가 각각 서열번호 4, 5 및 6의 아미노산 서열을 갖는 경쇄 가변영역을 포함하는 것을 특징으로 하는 항 DR5 항체.3. The antibody of claim 1 or 2, wherein the antibody has a heavy chain variable region having complementary determining region CDR1, CDR2 and CDR3 having the amino acid sequence of SEQ ID NOs: 1, 2 and 3, and the complementary determining region CDR1, CDR2 and CDR3, respectively, An anti-DR5 antibody comprising a light chain variable region having the amino acid sequences of Nos. 4, 5, and 6.
  4. 제 3항에 있어서, 상기 경쇄 가변영역은 서열번호 27 또는 37의 아미노산 서열을 갖는 것을 특징으로 하는 항 DR5 항체.The anti-DR5 antibody according to claim 3, wherein the light chain variable region has an amino acid sequence of SEQ ID NO: 27 or 37.
  5. 제 3항에 있어서, 상기 중쇄 가변영역은 서열번호 28 또는 38의 아미노산 서열을 갖는 것을 특징으로 하는 항 DR5 항체.4. The anti-DR5 antibody according to claim 3, wherein the heavy chain variable region has an amino acid sequence of SEQ ID NO: 28 or 38.
  6. 제 1항 또는 제 2항에 있어서, 상기 항체는 DR5를 발현하는 TRAIL-민감성 암세포 또는 DR5를 발현하는 TRAIL-저항성 암세포에 대해 자식작용에 의한 세포사멸을 유도하는 것을 특징으로 하는 항 DR5 항체.The anti-DR5 antibody according to claim 1 or 2, wherein the antibody induces apoptosis by progeny against TRAIL-sensitive cancer cells expressing DR5 or TRAIL-resistant cancer cells expressing DR5.
  7. 제 1항 또는 제 2항에 있어서, 상기 항체는 단일쇄 가변영역 단편 (scFv), (scFv)2, Fab, Fab' 및 F(ab')2로 이루어진 군으로부터 선택되는 것을 특징으로 하는 항 DR5 항체.The anti-DR5 antibody of claim 1 or 2, wherein the antibody is selected from the group consisting of single-chain variable region fragments (scFv), (scFv) 2 , Fab, Fab ', and F (ab') 2 . Antibodies.
  8. 제 1항 또는 제 2항에 있어서, 상기 항체는 단일클론항체인 것을 특징으로 하는 항 DR5 항체.The anti-DR5 antibody according to claim 1 or 2, wherein the antibody is a monoclonal antibody.
  9. 제 1항의 항체의 서열번호 7의 아미노산 서열을 코딩하는 서열번호 8의 염기서열을 갖는 DNA.DNA having a nucleotide sequence of SEQ ID NO: 8 encoding an amino acid sequence of SEQ ID NO: 7 of the antibody of claim 1.
  10. 제 2항의 항체의 서열번호 39의 아미노산 서열을 코딩하는 서열번호 40의 염기서열을 갖는 DNA.DNA having a nucleotide sequence of SEQ ID NO: 40 encoding an amino acid sequence of SEQ ID NO: 39 of the antibody of claim 2.
  11. 제 9항의 DNA 또는 이를 포함하는 발현벡터로 형질전환된 세포.A cell transformed with the DNA of claim 9 or an expression vector comprising the same.
  12. 제 10항의 DNA 또는 이를 포함하는 발현벡터로 형질전환된 세포.Cell transformed with the DNA of claim 10 or an expression vector comprising the same.
  13. 각각 서열번호 1, 2, 3의 아미노산 서열을 갖는 CDR1, CDR2, CDR3로 구성된, 항 DR5 항체의 중쇄 가변영역 중 상보성 결정부위 CDR.Complementarity determining region CDRs of the heavy chain variable region of the anti-DR5 antibody, consisting of CDR1, CDR2, CDR3 having the amino acid sequence of SEQ ID NO: 1, 2, 3, respectively.
  14. 각각 서열번호 4, 5, 6의 아미노산 서열을 갖는 CDR1, CDR2, CDR3로 구성된, 항 DR5 항체의 경쇄 가변영역 중 상보성 결정부위 CDR.Complementarity determining region CDRs of the light chain variable region of the anti-DR5 antibody, consisting of CDR1, CDR2, CDR3 having the amino acid sequence of SEQ ID NO: 4, 5, 6, respectively.
  15. 제 1항 또는 제 2항 중 어느 한 항의 DR5 항체를 유효성분으로 함유하는 암 예방 또는 치료용 조성물.A composition for preventing or treating cancer, comprising the DDR5 antibody of claim 1 as an active ingredient.
  16. 제 15항에 있어서, 상기 암은 DR5 발현에 의해 야기되는 암인 것을 특징으로 하는 암 예방 또는 치료용 조성물.The method for preventing or treating cancer according to claim 15, wherein the cancer is cancer caused by DR5 expression.
  17. 제 16항에 있어서, 상기 DR5 발현에 의해 야기되는 암은 혈액암, 폐암, 위암, 간암, 골암, 췌장암, 피부암, 두경부암, 피부 흑색종, 자궁암, 난소암, 직장암, 대장암, 결장암, 유방암, 자궁 육종, 나팔관 암종, 자궁내막 암종, 자궁경부 암종, 질 암종, 외음부 암종, 식도암, 후두암, 소장암, 갑상선암, 부갑상선암, 연조직의 육종, 요도암, 음경암, 전립선암, 만성 또는 급성 백혈병, 유년기의 고상 종양, 분화 림프종, 방광암, 신장암, 신장 세포 암종, 신장 골반 암종, 제 1 중추신경계 림프종, 척수축 종양, 뇌간 신경교종 및 뇌하수체 아데노마로 이루어진 군으로부터 선택되는 것을 특징으로 하는 암 예방 또는 치료용 조성물.The method of claim 16, wherein the cancer caused by the DR5 expression is hematological cancer, lung cancer, stomach cancer, liver cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, skin melanoma, uterine cancer, ovarian cancer, rectal cancer, colon cancer, colon cancer, breast cancer , Uterine sarcoma, fallopian tube carcinoma, endometrial carcinoma, cervical carcinoma, vaginal carcinoma, vulvar carcinoma, esophageal cancer, laryngeal cancer, small intestine cancer, thyroid cancer, parathyroid cancer, soft tissue sarcoma, urethral cancer, penile cancer, prostate cancer, chronic or acute leukemia Cancer prevention characterized in that it is selected from the group consisting of childhood solid tumors, differentiated lymphoma, bladder cancer, kidney cancer, renal cell carcinoma, renal pelvic carcinoma, primary central nervous system lymphoma, spinal contraction tumor, brainstem glioma and pituitary adenoma Or a therapeutic composition.
PCT/KR2009/006036 2008-10-24 2009-10-20 Anti-dr5 antibody with improved affinity and stability, and composition for cancer prevention or treatment including same WO2010047509A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20080104737 2008-10-24
KR10-2008-0104737 2008-10-24
KR1020090066518A KR101117070B1 (en) 2008-10-24 2009-07-21 Anti-death receptor 5 antibody with improved affinity and stability, and composition for preventing or treating cancer comprising the same
KR10-2009-0066518 2009-07-21

Publications (3)

Publication Number Publication Date
WO2010047509A2 WO2010047509A2 (en) 2010-04-29
WO2010047509A9 true WO2010047509A9 (en) 2010-09-02
WO2010047509A3 WO2010047509A3 (en) 2010-10-21

Family

ID=42119820

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2009/006036 WO2010047509A2 (en) 2008-10-24 2009-10-20 Anti-dr5 antibody with improved affinity and stability, and composition for cancer prevention or treatment including same

Country Status (1)

Country Link
WO (1) WO2010047509A2 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20140118682A (en) * 2013-03-29 2014-10-08 (주)셀트리온 Composition comprising two or more influenza A viruses neutralizing binding molecules
KR101796277B1 (en) 2016-04-12 2017-11-13 앱클론(주) Antibodies Binding Specifically to HER2 with Improved Stability
AU2017322783B2 (en) * 2016-09-08 2020-04-30 Woori Technologies Corporation Deglycosylated antibody specifically binding to clec14a and uses thereof
KR101926834B1 (en) * 2017-03-21 2018-12-07 동아에스티 주식회사 Anti-dr5 antibody and use thereof
JP7403733B2 (en) 2017-09-04 2023-12-25 国立感染症研究所長 Method for manufacturing influenza HA split vaccine
CN109837243A (en) * 2017-11-25 2019-06-04 深圳宾德生物技术有限公司 A kind of Chimeric antigen receptor T cell and its preparation method and application for the targeting DR5 knocking out PD1
CN113501876B (en) * 2021-07-16 2022-10-18 四川大学华西医院 Nano antibody, nucleic acid, expression vector, host cell and application of nano antibody specifically binding to protein kinase p38 delta

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6872568B1 (en) * 1997-03-17 2005-03-29 Human Genome Sciences, Inc. Death domain containing receptor 5 antibodies
TWI318983B (en) * 2000-05-02 2010-01-01 Uab Research Foundation An antibody selective for a tumor necrosis factor-related apoptosis-inducing ligand receptor and uses thereof
US8029783B2 (en) * 2005-02-02 2011-10-04 Genentech, Inc. DR5 antibodies and articles of manufacture containing same
KR100847010B1 (en) * 2006-07-05 2008-07-17 아주대학교산학협력단 5 5 antibody binding specifically to dr5 and composition for preventing or treating a cancer comprising the same

Also Published As

Publication number Publication date
WO2010047509A2 (en) 2010-04-29
WO2010047509A3 (en) 2010-10-21

Similar Documents

Publication Publication Date Title
WO2010047509A2 (en) Anti-dr5 antibody with improved affinity and stability, and composition for cancer prevention or treatment including same
WO2018194381A1 (en) Binding molecule specific for lrig-1 protein and use thereof
WO2019225787A1 (en) Anti-b7-h3 antibody and use thereof
WO2019107812A1 (en) Antibody inhibiting activated ras in cell by internalizing into cytosol of cell, and use thereof
WO2022039490A1 (en) Anti-b7-h4/anti-4-1bb bispecific antibodies and use thereof
WO2016137108A1 (en) Novel antibody binding to tfpi and composition comprising the same
WO2018026248A1 (en) Novel antibody against programmed cell death protein (pd-1), and use thereof
WO2019050362A2 (en) Antibody against human dlk1 and use thereof
WO2021020846A1 (en) Anti-her2/anti-4-1bb bispecific antibody and use thereof
WO2020080854A1 (en) Binding molecule specific to lrig-1 protein, and use thereof
WO2022177394A1 (en) Bispecific single domain antibody to pd-l1 and cd47 and use thereof
WO2020101365A1 (en) Anti-c-met antibody showing enhanced stability or antigen-binding fragments thereof
WO2021101346A1 (en) Anti-ror1/anti-4-1bb bispecific antibodies and uses thereof
WO2019190206A1 (en) Antibody binding specifically to ecl-2 of claudin 3, fragment thereof, and use thereof
WO2021020845A1 (en) Anti-egfr/anti-4-1bb bispecific antibody and use thereof
WO2019125070A1 (en) Antibody or antigen-binding fragment thereof that specifically recognizes b cell malignancies, chimeric antigen receptor comprising same, and uses thereof
WO2020022782A1 (en) Composition for preventing or treating immune-related diseases
WO2018026249A1 (en) Antibody against programmed death-ligand 1 (pd-l1), and use thereof
WO2019132579A2 (en) Immunotoxin comprising ribonuclease fused to cytotransmab
WO2021101349A1 (en) Antibody that binds to ror1 and b7-h3, antibody-drug conjugate containing same, and use thereof
WO2022035200A1 (en) Fusion protein comprising il-12 and anti-cd20 antibody and use thereof
WO2022124866A1 (en) Anti-pd-1 antibody and uses thereof
WO2021071319A1 (en) Multispecific fusion protein and use thereof
WO2021006714A1 (en) Composition for preventing, ameliorating or treating immune checkpoint inhibitor-resistant cancer
WO2023239228A1 (en) Fusion protein comprising anti-tigit antibody, interleukin-15, and interleukin-15 receptor alpha sushi domain, and use thereof

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: 09822185

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09822185

Country of ref document: EP

Kind code of ref document: A2