WO2018124851A1 - Pharmaceutical composition for preventing or treating cancer, containing antibody specifically binding to l1cam protein and pyrimidine analog and/or platin-based anticancer drug - Google Patents

Pharmaceutical composition for preventing or treating cancer, containing antibody specifically binding to l1cam protein and pyrimidine analog and/or platin-based anticancer drug Download PDF

Info

Publication number
WO2018124851A1
WO2018124851A1 PCT/KR2018/000056 KR2018000056W WO2018124851A1 WO 2018124851 A1 WO2018124851 A1 WO 2018124851A1 KR 2018000056 W KR2018000056 W KR 2018000056W WO 2018124851 A1 WO2018124851 A1 WO 2018124851A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
variable region
chain variable
l1cam
Prior art date
Application number
PCT/KR2018/000056
Other languages
French (fr)
Korean (ko)
Inventor
홍효정
Original Assignee
강원대학교 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
Application filed by 강원대학교 산학협력단 filed Critical 강원대학교 산학협력단
Publication of WO2018124851A1 publication Critical patent/WO2018124851A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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

Definitions

  • the present invention provides an antibody that specifically binds to L1CAM protein; And pyrimidine analogues and / or platinum-based anticancer agents.
  • Biliary tract cancer is a malignant tumor of the ductal epithelium of the bile tract, which is anatomically anatomical of intrahepatic cholangiocarcinoma (ICC) or extrahepatic cholangiocarcinoma (ECC).
  • ICC intrahepatic cholangiocarcinoma
  • ECC extrahepatic cholangiocarcinoma
  • Biliary cancer is difficult to diagnose with recent clinical symptoms and has a poor prognosis because it is resistant to conventional chemotherapy and radiation therapy.
  • the overall 5-year survival rate for patients with biliary tract cancer is 5 to 10% and includes curable surgery. In all cases, the reported 5-year survival rate is 25-30%. Thus, there is a need for new and effective treatment regimens that improve the survival of biliary tract cancer patients.
  • gemcitabine As a chemotherapy drug for biliary tract cancer, gemcitabine, a pyrimidine analog that inhibits DNA synthesis, has been extensively studied. In stage 2 clinical trials, it has been known that monotherapy of gemcitabine has shown efficacy in patients with advanced or metastatic biliary tract cancer (Okusaka T, et al, Cancer Chemother Pharmacol. 2006; 57 (5): 647-53.). In addition, a drug for biliary cancer comprising gemcitabine, 5-fluorouracil, and amino acids has been developed (Korean Patent Publication No. 2014-0079831), and the combination therapy of gemcitabine and cisplatin is used for patients with locally advanced or metastatic biliary tract cancer.
  • EGFR epidermal growth factor receptor
  • cetuximab an anti-EGFR antibody
  • gemcitabine and oxaliplatin have recently been attempted, but efficacy in patients with advanced biliary tract cancer has not improved in phase II trials.
  • combination therapy of gemcitabine and oxaliplatin with EGFR tyrosine kinase inhibitor erlotinib has been tried, but the recently released Phase III clinical trials did not show an improved effect compared to chemotherapy. .
  • Phase III clinical trials did not show an improved effect compared to chemotherapy.
  • L1 cell adhesion molecule is a 200-220 kDa transmembrane glycoprotein, including six immunoglobulin-like domains, five fibronectin-type III domains, transmembrane stretch and It consists of a short cellular tail.
  • L1CAM is a neuronal adhesion molecule of the central nervous system that plays an important role in the movement of cerebellar cells and the growth of neurites in many cancers, including melanoma, neuroblastoma, ovarian cancer, colon cancer, cholangiocarcinoma and gallbladder cancer. has been reported to play an important role in the growth and metastasis of cancer cells, and that L1CAM overexpression is associated with poor prognosis of cancer (Raveh et al., Cancer Letters 282: 137-145, 2009).
  • the present inventors have diligently studied to develop a method for effectively treating cancer, specifically, biliary tract cancer.
  • the present invention is administered with a combination of Ab417, an antibody targeting L1CAM, and gemcitabine and / or cisplatin, the biliary tract cancer, It confirmed that the synergistic effect was shown with respect to the growth inhibition of, and completed this invention.
  • One object of the invention is an antibody that specifically binds to an L1 cell adhesion molecule (L1CAM) protein; And it provides a pharmaceutical composition for the prevention or treatment of cancer, including an anticancer agent, specifically pyrimidine analog, a platinum-based anticancer agent or a combination thereof.
  • L1CAM L1 cell adhesion molecule
  • Another object of the present invention is to provide a kit comprising the composition.
  • Another object of the present invention is the antibody; And administering a pyrimidine analog, a platinum anticancer agent, or a combination thereof to a subject in need thereof.
  • the pharmaceutical composition of the present invention can be usefully used for the treatment of biliary tract cancer.
  • A is tumor volume
  • B is tumor weight
  • C is body weight.
  • Each point p by "mean ⁇ standard deviation" a represents, Dunnett t- test (Dunnett's t-test) p ⁇ 0.05 (*) and p ⁇ 0.01 (**), and Steel test (Steel's test) according to the ⁇ 0.05 ( # ) means significant difference compared to the homologous control.
  • D represents the area stained with anti-human Ki-67 antibody in the tumor section. The Ki-67 index is expressed as a percentage of the positive staining site relative to the total area.
  • FIG. 3 shows the appearance of Ab417 bound to Choi-CK expressed L1CAM, and the effect of reducing the membrane-L1CAM level of Ab417.
  • A shows confocal microscopy analysis of Ab417 internalization. SCK-L1 cells were pretreated with Ab417 (10 ⁇ g / mL) for the indicated time, Ab417 bound to membrane L1CAM was anti-human IgG (H + L) Cross Adsorbed DyLight 594 (red), and internalized Ab417 was anti-human Detection was done using IgG (Fc-specific) -FITC (green).
  • B represents Choi-CK cells treated with Ab417 (10 ⁇ g / mL) for the indicated time.
  • Membrane L1CAM was detected by Western blot analysis using A10-A3, and pan-cadherin was used as loading control. Cells not treated with Ab417 were used as a control (C).
  • C is a 400x magnified confocal microscopy image stained with L1CAM for Choi-CK tumors treated with PBS (top) or Ab417 (bottom). It is expressed as a percentage of the positive staining site relative to the total area.
  • D represents the region stained with an antibody specific for the N-terminal region of human L1CAM in the tumor section. It is expressed as a percentage of the positive staining area relative to the total area.
  • FIG. 4 shows the anticancer effects of gemcitabine or cisplatin in Choi-CK heterogeneous model. Intraperitoneal injections of the indicated doses of the drug twice a week for three or four weeks. A is the tumor volume, B is the tumor weight. Each point represents 'mean ⁇ standard deviation', p ⁇ 0.05 (*) and p ⁇ 0.01 (**) by Dunnett test, and p ⁇ 0.05 ( # ) and p ⁇ 0.01 ( ## ) by Steel test Means a significant difference compared to the control (saline).
  • Figure 5 shows the anticancer effect of the combination of Ab417 and gemcitabine, or cisplatin in Choi-CK xenograft model.
  • a and C represent tumor volume
  • B and D represent tumor weight.
  • Each point represents 'mean ⁇ standard deviation', p ⁇ 0.05 (*) and p ⁇ 0.01 (**) by Dunnett test, and p ⁇ 0.05 ( # ) and p ⁇ 0.01 ( ## ) by Steel test Means a significant difference compared to the control (saline).
  • FIG. 6 shows the degree of weight change of Ab417 and gemcitabine, or cisplatin combined administration in Choi-CK xenograft model.
  • A relates to administration of gemcitabine or cisplatin alone, B to administration of Ab417 or gemcitabine alone, and combination of Ab417 and gemcitabine, C to administration of Ab417 or cisplatin alone, and the combination of Ab417 and cisplatin.
  • Each point represents a mean ⁇ standard deviation.
  • A is the tumor volume and B is the tumor weight.
  • Each point represents 'mean ⁇ standard deviation', and p ⁇ 0.01 (**) by Dunnett's t-test indicates a significant difference compared to the homologous control (hFc).
  • A is the tumor volume and B is the tumor weight.
  • Each point represents 'mean ⁇ standard deviation', meaning that p ⁇ 0.05 (*) and p ⁇ 0.01 (**) by Dunnett's test indicate a significant difference compared to the saline.
  • FIG. 10 shows the anticancer effect of the combination of Ab417 and gemcitabine, or cisplatin in TFK-1 xenograft model.
  • A is the tumor volume and B is the weight.
  • Each point represents 'mean ⁇ standard deviation', meaning that p ⁇ 0.05 (*) and p ⁇ 0.01 (**) by Dunnett's test indicate a significant difference compared to the saline.
  • one embodiment of the present invention comprises an antibody that specifically binds to L1 cell adhesion molecule (L1CAM) protein; And it provides a pharmaceutical composition for the prevention or treatment of cancer, including an anticancer agent, specifically pyrimidine analog (pyrimidine analog), a platinum-based anticancer agent or a combination thereof.
  • L1CAM L1 cell adhesion molecule
  • the treatment efficiency of biliary tract cancer is significantly increased as compared with the administration of the antibody and drug alone. Confirmed.
  • L1 CAM refers to one of the intrinsic membrane glycoproteins belonging to immunoglobulin superfamily cell adhesion molecules (CAMs).
  • CAMs immunoglobulin superfamily cell adhesion molecules
  • the L1CAM is expressed in a variety of cancers, including melanoma, neuroblastoma, ovarian cancer, colorectal cancer, cholangiocarcinoma and gallbladder cancer, and plays an important role in the growth and metastasis of cancer cells. Relevance has been reported (Raveh et al., Cancer Letters 282: 137-145, 2009).
  • the antibody of the present invention binds to both human and mouse L1CAM proteins with high affinity and exhibits significant cancer growth inhibitory effect, it can be usefully used in the field of prevention or treatment of cancer in which L1CAM is overexpressed.
  • antibody refers to a protein molecule that acts as a receptor that specifically recognizes an antigen, including an immunoglobulin molecule that is immunologically reactive with a specific antigen, including polyclonal antibodies, monoclonal antibodies, It includes both whole antibodies and antibody fragments.
  • the term also includes chimeric antibodies (eg, humanized murine antibodies) and bivalent or bispecific molecules (eg, bispecific antibodies), diabodies, triabodies, and tetrabodies.
  • the term further includes artificial antibodies based on protein scaffolds and single chain antibodies, caps, analogs of antibody constant regions that possess binding functions for FcRn.
  • the total antibody is a structure having two full length light chains and two full length heavy chains, each of which is linked by heavy and disulfide bonds.
  • the total antibody includes IgA, IgD, IgE, IgM and IgG, and IgG is a subtype, including IgG1, IgG2, IgG3 and IgG4.
  • the antibody fragment refers to a fragment having an antigen binding function, and includes Fd, Fab, Fab ', F (ab') 2 and Fv.
  • Fd means the heavy chain portion included in the Fab fragment.
  • the Fab has one antigen binding site in a structure having a variable region of the light and heavy chains, a constant region of the light chain, and a first constant region of the heavy chain (CH1 domain).
  • F (ab ') 2 antibodies are produced when the cysteine residues of the hinge region of Fab' form disulfide bonds.
  • Fv (variable fragment) means a minimum antibody fragment having only the heavy chain variable region and light chain variable region.
  • Double disulfide Fv (dsFv) is a disulfide bond is linked to the heavy chain variable region and the light chain variable region, and short chain Fv (scFv) is generally covalently linked to the variable region of the heavy chain and the light chain through a peptide linker.
  • Such antibody fragments can be obtained using proteolytic enzymes (e.g., the entire antibody can be restricted to papain and Fab can be obtained, and pepsin can yield F (ab ') 2 fragment). Can be produced through genetic recombination techniques.
  • the 'immunoglobulin' has a heavy chain and a light chain, each heavy and light chain comprises a constant region and a variable region (the region is also known as a domain).
  • the variable regions of the light and heavy chains comprise three multivariable regions and four framework regions called complementarity-determining regions (hereinafter referred to as "CDRs").
  • the CDRs mainly serve to bind epitopes of antigens.
  • the CDRs of each chain are typically called CDR1, CDR2, CDR3, starting sequentially from the N-terminus and also identified by the chain in which the particular CDR is located.
  • an antibody that specifically binds to an L1 cell adhesion molecule (L1CAM) protein refers to an antibody that binds to an L1CAM protein and inhibits the activity of the L1CAM protein.
  • the antibody that specifically binds to L1CAM is an antibody that binds to the Ig5 region of L1CAM, but is not limited thereto.
  • the antibody specifically binding to the L1CAM protein may be an antibody or a functional variant thereof comprising the heavy chain variable region of SEQ ID NO: 1 and the light chain variable region of SEQ ID NO: 5.
  • the term "functional variant of an antibody comprising a heavy chain variable region of SEQ ID NO: 1 and a light chain variable region of SEQ ID NO: 5" refers to a parent antibody comprising a heavy chain variable region of SEQ ID NO: 1 and a light chain variable region of SEQ ID NO: 5 Compared with the antibody having a difference in at least one amino acid of the heavy chain variable region and / or light chain variable region, it may be an antibody having improved physical properties compared to the parent antibody. In addition, the functional variant may retain biological activity equivalent to that of the parent antibody.
  • the antibody is an antibody having a difference in 1 to 10 amino acids of the heavy chain variable region and / or the light chain variable region as compared to the parent antibody comprising the heavy chain variable region of SEQ ID NO: 1 and the light chain variable region of SEQ ID NO: 5, Specifically, at least one amino acid, in particular 1 to 10, more specifically 1 in the heavy chain variable region and / or light chain variable region of the parent antibody comprising a heavy chain variable region of SEQ ID NO: 1 and a light chain variable region of SEQ ID NO: To 6 amino acids may be substituted.
  • Such antibodies may be monoclonal antibodies, and may be in the form of full length antibodies or antibody fragments thereof.
  • such antibodies include heavy chain CDR1 as set forth in SEQ ID NO: 2; Heavy chain CDR2 set forth in SEQ ID NO: 3 or 10; And a heavy chain variable region comprising a heavy chain CDR3 as set out in SEQ ID NO: 4 and a light chain CDR1 as set out in SEQ ID NO: 6 or 12; Light chain CDR2 set forth in SEQ ID NO: 7; And a light chain variable region comprising the light chain CDR3 set forth in SEQ ID NO: 8, but is not limited thereto.
  • the antibody comprises a heavy chain variable region selected from the group consisting of SEQ ID NOs: 1, 9 and 13; And a light chain variable region selected from the group consisting of SEQ ID NOs: 5, 11, 14, and 15, but is not limited thereto.
  • the antibody specifically binding to L1CAM may be an antibody disclosed in Korean Patent Application Laid-Open No. 10-2014-0066101 A, WO2014-077648 A1, or US 2015-0344571 Al.
  • the entire specification of the patent is incorporated herein by reference in its entirety.
  • the antibody specifically binding to L1CAM may be as follows, but is not particularly limited thereto:
  • Ab417 An antibody comprising the heavy chain variable region of SEQ ID NO: 1 and the light chain variable region of SEQ ID NO: 5 is named Ab417 in the present invention, and the antibody comprising the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 15 is provided herein. Is designated Ab417-H5L1.
  • the antibody comprising the heavy chain variable region of SEQ ID NO: 9 and the light chain variable region of SEQ ID NO: 11 is named Ab417-H6L2 in the present invention
  • the antibody comprising the heavy chain variable region of SEQ ID NO: 9 and the light chain variable region of SEQ ID NO: 14 It is named Ab417-H6L6 in the present invention.
  • Ab417-H5L1, -H6L2, and -H6L6 described above are all properties of the applicant compared to Ab417 in the prior patents of the applicant, that is, Korean Patent Application Publication No. 10-2014-0066101A, WO2014-077648 A1, or US 2015-0344571 Al. It was developed as an improved antibody and can show an effect equivalent to Ab417.
  • sequences used in the present invention are to be construed to include sequences that exhibit substantial identity with the sequences listed in the Sequence Listing, in view of variations that are biologically equivalent.
  • the term 'substantial identity' means that if the alignment of the sequence of the present invention with any other sequence is maximally matched, the aligned sequence is analyzed using algorithms commonly used in the art. By 60% homology, more specifically 70% homology, even more specifically 80% homology, most specifically 90% homology.
  • amino acid sequence having high homology with the sequence represented by SEQ ID NOS: 1 to 15, for example, an amino acid having a high homology of 70% or more, specifically 80% or more, more specifically 90% or more. Sequences should also be construed as falling within the scope of the present invention.
  • pyrimidine analog refers to a nucleoside analog compound that mimics the structure of metabolic pyrimidine.
  • the pyrimidine analogs can be used as intracellular chemotherapeutic agents, intracellular anticancer agents, anti-viral agents or nucleic acid components for DNA viruses, and the like.
  • the pyrimidine analogue may be used as an anticancer agent, and may be, for example, gemcitabine, but is not limited thereto.
  • platinum-based anticancer drugs refers to anti-cancer drugs used as drugs that damage DNA as a heavy metal compound containing platinum. It is mainly used to treat various kinds of cancers such as breast cancer, bladder cancer, gastric cancer and cervical cancer, and combines with two adjacent guanines on DNA strands to form an interstrand crosslink to inhibit DNA synthesis. Known.
  • the platinum-based anticancer agent is cisplatin (cisplatin), oxaliplatin (oxaliplatin), tetraplatin (tetraplatin), heptaplatin (heptaplatin), paraplatin (carplatin) (carboplatin) or nanoplatin ( nanoplatin), more specifically cisplatin, but is not limited thereto.
  • the type of cancer is not particularly limited, but may be a cancer in which L1CAM is overexpressed. Specifically, melanoma, neuroblastoma, ovarian cancer, colorectal cancer, biliary tract cancer (biliary duct cancer or gallbladder cancer), but is not limited thereto.
  • Biliary tract cancer refers to “biliary duct cancer” and “gallbladder cancer” and refers to a malignant tumor occurring in epithelial cells surrounding the inside of the gallbladder.
  • Bile duct cancer is a cancer that occurs in the bile duct that is the passage of bile discharged from the liver
  • gallbladder cancer refers to a cancer that occurs in the gallbladder, which is a place where the bile is first stored.
  • Biliary cancers can be divided into intrahepatic biliary and extrahepatic biliary tract cancers, depending on the location of the tumor, which may be divided into peripheral cholangiocarcinoma and hepatic cholangiocarcinoma; Extrahepatic biliary tract cancer may be classified into upper (proximal), middle, and lower (distal) biliary tract cancers according to the occurrence site.
  • Biliary cancers are difficult to diagnose early because they do not show any characteristic symptoms until they progress significantly.
  • various anatomical structures in the biliary tract and vasculature and it is difficult to determine the exact extent of tumor invasion before or even during surgery, resulting in curative resection (to remove all areas where cancer exists or is likely to exist).
  • curative resection to remove all areas where cancer exists or is likely to exist.
  • Chemotherapy is used to prevent the growth of cancer cells that may remain after surgery or metastases due to cancer metastasis.
  • prevention refers to the antibody; And all actions of inhibiting or delaying cancer by administration of a composition comprising a pyrimidine analog, a platinum anticancer agent, or a combination thereof.
  • treatment refers to the antibody; And any action in which the symptoms of cancer improve or benefit altered by administration of a composition comprising a pyrimidine analog, a platinum anticancer agent, or a combination thereof.
  • the administration of Ab417 or gemcitabine alone showed 35.3% or 44.4% cancer growth inhibition, respectively, against Choi-CK cells, which are ICC cells, but 88.8% It was confirmed that the inhibition rate of (Fig. 5).
  • TFK-1 cells which are ECC cells
  • cancer growth inhibition was 10.7% or 21.7%, respectively, but when they were used in combination, they showed an inhibition rate of 52.2%. (FIG. 10).
  • the pharmaceutical composition may further include a pharmaceutically acceptable carrier, excipient or diluent commonly used in the manufacture of the pharmaceutical composition, and the carrier may comprise a non-naturally occuring carrier. have.
  • composition means to exhibit properties that are not toxic to cells or humans exposed to the composition.
  • the pharmaceutical composition may be used in the form of oral dosage forms, external preparations, suppositories, and sterile injectable solutions, such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, and aerosols, respectively, according to conventional methods.
  • sterile injectable solutions such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, and aerosols, respectively, according to conventional methods.
  • the formulation is used in the art for the prevention or treatment of cancer, it is not limited thereto.
  • Examples of carriers, excipients, and diluents that may be included in the pharmaceutical composition include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate , Calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate, polycaprolactone (PCL; polycaprolactone), polylac Polylactic acid (PLA), poly-L-lactic acid (PLLA), Mineral oil and the like.
  • diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrating agents, surfactants, etc. which are commonly used.
  • Solid preparations for oral administration include tablets, pills, powders, granules, capsules, and the like, which may comprise at least one excipient such as starch, calcium carbonate, It may be prepared by mixing sucrose or lactose, gelatin and the like. In addition to simple excipients, lubricants such as magnesium styrate and talc may also be used.
  • Liquid preparations for oral administration include suspensions, solutions, emulsions, and syrups, and may include various excipients, such as wetting agents, sweeteners, fragrances, and preservatives, in addition to commonly used simple diluents such as water and liquid paraffin. have.
  • Formulations for parenteral administration may include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized formulations, suppositories, and the like.
  • non-aqueous solvent and suspending agent propylene glycol, polyethylene glycol, vegetable oil such as olive oil, injectable ester such as ethyl oleate and the like can be used.
  • base of the suppository witepsol, macrogol, tween 61, cacao butter, laurin butter, glycerogelatin and the like can be used.
  • the antibody included in the pharmaceutical composition of the present invention may be included in an amount of 0.0001 to 50% by weight, more specifically 0.01 to 20% by weight, based on the total weight of the final composition, but is not particularly limited thereto.
  • the pharmaceutical composition of the present invention may be administered in a pharmaceutically effective amount.
  • ⁇ pharmaceutically effective amount '' means an amount sufficient to treat or prevent a disease at a reasonable benefit / risk ratio applicable to medical treatment or prevention, and an effective dose level refers to the severity of the disease, the activity of the drug, the patient Age, body weight, health, sex, sensitivity of the patient to the drug, time of administration of the composition of the invention used, route of administration and rate of release, duration of treatment, drugs comprising the drug in combination or concurrently with the composition of the invention used and It may be determined according to factors well known in other medical fields.
  • the pharmaceutical composition of the present invention may be administered alone or in combination with known biliary tract cancer therapeutics. In consideration of all the above factors, it is important to administer an amount that can obtain the maximum effect in a minimum amount without side effects.
  • the dosage of the pharmaceutical composition of the present invention can be determined by those skilled in the art in consideration of the purpose of use, the degree of addiction of the disease, the age, weight, sex, history, or type of substance used as an active ingredient.
  • the pharmaceutical composition of the present invention may be administered at about 0.1 ng / kg to about 100 mg / kg, specifically about 1 ng / kg to about 10 mg / kg, per adult Frequency of administration is not particularly limited, but may be administered once a day or several times in divided doses. The dosage does not limit the scope of the invention in any aspect.
  • Another embodiment is the antibody; And administering to the individual a pharmaceutically effective amount of a pyrimidine analog, a platinum based anticancer agent, or a combination thereof.
  • the term "individual” may include, without limitation, mammals, farmed fish, and the like, including rats, livestock, humans, and the like, which may or may not develop cancer.
  • the route of administration and mode of administration for administering the composition is not particularly limited and may be in accordance with any route of administration and mode of administration so long as the composition can reach the desired site of interest. Can be.
  • composition may be administered through various routes of oral or parenteral, non-limiting examples of the route of administration, eye, mouth, rectal, topical, intravenous, intraperitoneal, intramuscular, intraarterial, transdermal And intranasal administration or inhalation.
  • composition may be administered by any device in which the active substance may migrate to the target cell.
  • the antibody; And pyrimidine analogs, platinum anticancer agents or combinations thereof may be administered in combination.
  • the term "combination” should be understood to denote simultaneous, separate or sequential administration. If the administration is sequential or separate, the interval of administration of the secondary components should be such that the beneficial effect of the combination is not lost.
  • Each of the agents may be administered simultaneously, sequentially, or in reverse order, and may be administered simultaneously in any combination of appropriate effective amounts, but is not limited to any particular method or order of administration.
  • the method of the present invention may comprise administering the active ingredient in a pharmaceutically effective amount.
  • Suitable total daily doses may be determined by the treating physician within the scope of good medical judgment and may be administered once or in divided doses.
  • the specific therapeutically effective amount for a particular patient is determined by the specific composition, including the type and extent of the reaction to be achieved, whether or not other agents are used in some cases, the age, weight, general health of the patient, It is desirable to apply differently depending on various factors and similar factors well known in the medical field, including sex and diet, time of administration, route of administration and rate of composition, duration of treatment, drugs used with or co-specific with the specific composition.
  • Another aspect provides a pharmaceutical kit comprising the composition.
  • the pharmaceutical kit may be in tablet, capsule or injection form, but is not limited thereto.
  • kit may comprise a pharmaceutically effective dosage of the antibody; And pyrimidine analogs, platinum-based anticancer agents, or combinations thereof.
  • the kit can also include instructions for the administration of the components.
  • ICC human intrahepatic cholangiocarcinoma
  • Choi-CK and SCK-L1 were cultured in DMEM (Welgene, Republic of Korea) supplemented with 10% FBS (Atlas Biologics, USA).
  • Choi-CK was provided by Dr. Dae-Gon Kim of Chonbuk National University.
  • TFK-1 an extrahepatic cholangiocarcinoma (ECC) cell line, was purchased from German Collection of Microorganisms and Cell Cultures, Human and Animal Cell Lines, Braunschweig, Germany, and cultured in RPMI (Thermo Fisher Scientific, USA). .
  • CHO-DG44 cell line # 9-20 expressing Ab417 antibody (Cho S, et al., MAbs. 2016; 8 (2): 414-25.) was 5% (v / v) dialyzed FBS (Thermo Fisher Scientific) ) was supplemented with MEM- ⁇ (Welgene) supplemented and replaced with medium without serum (SFM4CHO, GE Lifesciences, USA) for the production of antibodies. All cell lines were cultured in a wet incubator at 37 ° C. with 5% CO 2 .
  • Ab417 antibody (hereinafter referred to as 'Ab417') was produced in CHO-DG44 cell line (# 9-20). The culture supernatant was centrifuged and filtered using a bottle top filter (0.22 ⁇ m PES, Sartorius) and affinity chromatography was performed on Protein A-Agarose (GenScript, USA) for purification. Antibodies bound to protein A were eluted with 0.1 M sodium citrate containing 150 mM sodium chloride, pH 3.6. The eluted antibody was replaced with 25 mM sodium citrate buffer (pH 6.4) containing 150 mM sodium chloride and 0.007% Tween-20 (pH 6.4) and stored. Protein concentration was measured with a NanoDrop 2000 UV-Vis spectrophotometer (Thermo Fisher Scientific).
  • mice (BALB / c Slc- nu , 5 weeks old) were purchased from SLC, Inc., Japan and stored in a specific pathogen-free environment for 7 days according to the guidelines of the Animal Control Committee of Biotoxtech (Korea).
  • Choi-CK cells (1 ⁇ 10 6 ) were inoculated subcutaneously in the right flank of each mouse.
  • Choi-CK tumor tissue (3 ⁇ 3 ⁇ 3 mm 3 ) was inoculated subcutaneously in the back of new nude mice.
  • Gemcitabine (Gemcitabine hydrochloride, Sigma Aldrich) and cisplatin (cis-Diammineplatinum (II) dichloride, Sigma Aldrich) were dissolved in an appropriate concentration using sodium chloride (Choongwae, Republic of Korea). Additional animals not used were euthanized using CO 2 gas and fatal bleeding.
  • nude mice (BALB / c-nude, 6 weeks old) were purchased from NARA Biotech (Republic of Korea) and followed for 7 days according to the instructions of Kangwon National University IACUC (KW-160429-1). Stored in an environment free of specific pathogens.
  • Paraffin-embedding tissue sections were de-paraffinized with xylene and 95%, 90%, 70% and 50% ethanol and washed with PBS.
  • the de-paraffinized slides were heated in 0.1 mol / L citric acid buffer (pH 6.0) for 30 minutes and incubated for 15 minutes in 0.3% (v / v) hydrogen peroxide contained in methanol. Sections were blocked in normal horse serum at room temperature and blocked, and immunostaining was performed overnight at 4 ° C. with primary antibodies against Ki-67 (C-term) (1: 200, Acris, Germany).
  • Target proteins were visualized using ABC and DAB kits (Vector Laboratories, USA) and cross-stained with hematoxylin.
  • immunostaining was performed overnight at 4 ° C. with anti-NCAM-L1 (N-14) mAb (1:50, Santa Cruz Biotechnology, USA), and donkey anti-goat Incubated with Alexa 488-conjugated secondary antibody (1: 200, Thermo Fisher Scientific) for 1 hour at room temperature.
  • Cell nuclei were labeled with DAPI (5 ⁇ , Sigma Aldrich). Images were taken using a Zeiss confocal microscope and the degree of staining was quantitatively assessed using Image J software (version 1.45).
  • Ab417 Internalization of Ab417 was measured using a confocal laser scanning microscope. Medium containing Ab417 (10 ⁇ g / mL) was added to L1CAM-expressing SCK-L1 cells loaded on slide glass. After incubation at 37 ° C. for 0.5 or 4 hours, the cells were placed on ice, rinsed with 4 ° C. PBS and fixed with 4% formaldehyde (Sigma Aldrich).
  • Pan-cadherin to Pan-cadherin Rabbit mAb (1: 2,500, v / v, Cell Signaling Technology, USA) and anti-rabbit IgG-HRP conjugate (1: 5,000, v / v, Cell Signaling Technology's loading control.
  • the immunoreactive bands were then visualized using SuperSignal TM West Femto Maximum Sensitive Substrate (Thermo Fisher Scientific).
  • Results are expressed as mean ⁇ standard deviation, and the statistical comparison between groups
  • Example 1-3 Inhibition of cancer cell proliferation by gemcitabine or cisplatin
  • gemcitabine or cisplatin inhibited the proliferation of Choi-CK cells in a dose-dependent manner, and the IC 50 value of gemcitabine or cisplatin was 1.5 ⁇ g / mL or 3.0 ⁇ g / mL, respectively.
  • Example 1-5 Confirmation of anticancer effect of Ab417 and gemcitabine or cisplatin combination
  • Ab417, and the maximum dose of gemcitabine or cisplatin were injected into the Choi-CK xenograft model, according to Experiment 3, Ab417 (10 mg / kg) alone; Gemcitabine (10 mg / kg) alone; Or cisplatin (0.5 mg / kg) alone, or Ab417 (10 mg / kg) and gemcitabine (10 mg / kg); Ab417 (10 mg / kg) and cisplatin (0.5 mg / kg); Or Ab417 (10 mg / kg) and saline as a control were injected into mice with Choi-CK xenografts twice a week for 3 weeks.
  • TFK-1 cells were incubated with the antibody for a designated time (1, 2, 4, 6 hours), and the membrane protein fraction of the treated cells was bound to the Ig1 domain of human L1CAM. Analysis was by Western blot using murine anti-L1CAM mAb (A10-A3).
  • Example 2-3 Identify anticancer effects of gemcitabine or cisplatin
  • Example 2-4 Confirmation of anticancer effect of Ab417 and gemcitabine or cisplatin combination
  • Ab417 (5 mg / kg) alone; Gemcitabine (5 mg / kg) alone; Or cisplatin (0.5 mg / kg) alone, or Ab417 (5 mg / kg) and gemcitabine (5 mg / kg); Ab417 (5 mg / kg) and cisplatin (0.5 mg / kg); Or Ab417 (5 mg / kg) and saline as a control was injected into NOD / SCID mice ( n 8) with TFK-1 xenografts twice a week for 3 weeks.

Abstract

The present invention relates to a pharmaceutical composition for preventing or treating cancer, containing: an antibody specifically binding to an L1CAM protein; and a pyrimidine analogue, a platin-based anticancer drug, or a combination thereof. Compared to when administrating each ingredient individually, when co-administering an antibody specifically binding to an L1CAM protein with gemcitabine, which is a type of pyrimidine analogue, and/or cisplatin, which is a type of platin-based anticancer drug, synergistic effects are exhibited on the inhibition of bile duct cancer growth, and thus the pharmaceutical composition, of the present invention, containing the antibody and the drug can be usable in the treatment of biliary tract cancer.

Description

L1CAM 단백질에 특이적으로 결합하는 항체; 및 피리미딘 유사체 및/또는 플라틴계 항암제를 포함하는 암의 예방 또는 치료용 약학적 조성물Antibodies that specifically bind to L1CAM protein; And pyrimidine analogues and / or platinum-based anticancer agents
본 발명은 L1CAM 단백질에 특이적으로 결합하는 항체; 및 피리미딘 유사체 및/또는 플라틴계 항암제를 포함하는 암의 예방 또는 치료용 약학적 조성물에 관한 것이다.The present invention provides an antibody that specifically binds to L1CAM protein; And pyrimidine analogues and / or platinum-based anticancer agents.
담도암은(biliary tract cancer)는 담즙 기관의 관 상피(ductal epithelium)에서 발생하는 악성종양으로서, 담도암은 해부학적으로 간내 담도암(ICC; intrahepatic cholangiocarcinoma) 또는 간외 담도암(ECC; extrahepatic cholangiocarcinoma)으로 분류할 수 있다(Khan SA, et al., Cholangiocarcinoma. Lancet. 2005;366(9493):1303-14.). 담도암은 최근의 임상 증상으로 진단하기가 어렵고, 기존의 화학 요법 및 방사선 요법에 내성이 있어 예후가 좋지 않은데, 담도암 환자의 전체 5년 생존율은 5 ~ 10%이며, 치유 가능한 수술을 포함하는 경우에도 보고된 5년 생존율은 25 ~ 30%이다. 따라서, 담도암 환자의 생존율을 향상시키는 새롭고 효과적인 치료 요법이 요구되고 있다.Biliary tract cancer is a malignant tumor of the ductal epithelium of the bile tract, which is anatomically anatomical of intrahepatic cholangiocarcinoma (ICC) or extrahepatic cholangiocarcinoma (ECC). (Khan SA, et al., Cholangiocarcinoma. Lancet. 2005; 366 (9493): 1303-14.). Biliary cancer is difficult to diagnose with recent clinical symptoms and has a poor prognosis because it is resistant to conventional chemotherapy and radiation therapy. The overall 5-year survival rate for patients with biliary tract cancer is 5 to 10% and includes curable surgery. In all cases, the reported 5-year survival rate is 25-30%. Thus, there is a need for new and effective treatment regimens that improve the survival of biliary tract cancer patients.
담도암에 대한 화학 요법 약물로서, DNA 합성을 억제하는 피리미딘 유사체(pyrimidine analog)인 젬시타빈(gemcitabine)은 광범위하게 연구되어왔다. 2기 임상 시험에서, 젬시타빈의 단일 요법은 진행성 또는 전이성 담도암 환자에게 효능을 나타냄이 공지되었다(Okusaka T, et al,. Cancer Chemother Pharmacol. 2006;57(5):647-53.). 또한, 젬시타빈과 5-플루오루우라실 및 아미노산을 포함하는 담도암 치료약이 개발된바 있으며(한국 공개특허공보 제2014-0079831호), 젬시타빈 및 시스플라틴의 병용 요법은 국소 진행성 또는 전이성 담도암 환자에서 젬시타빈 단독 요법에 비해 생존율이 유의하게 높음이 보고된바 있지만, 담도암 환자의 화학 요법에 대한 반응은 제한적이며 여전히 5년 생존율은 낮다(Rizvi S, et al., Semin Liver Dis. 2014;34(4):456-64.). As a chemotherapy drug for biliary tract cancer, gemcitabine, a pyrimidine analog that inhibits DNA synthesis, has been extensively studied. In stage 2 clinical trials, it has been known that monotherapy of gemcitabine has shown efficacy in patients with advanced or metastatic biliary tract cancer (Okusaka T, et al, Cancer Chemother Pharmacol. 2006; 57 (5): 647-53.). In addition, a drug for biliary cancer comprising gemcitabine, 5-fluorouracil, and amino acids has been developed (Korean Patent Publication No. 2014-0079831), and the combination therapy of gemcitabine and cisplatin is used for patients with locally advanced or metastatic biliary tract cancer. Although survival has been reported to be significantly higher than gemcitabine monotherapy in, the response to chemotherapy in patients with biliary tract cancer is limited and still has a low 5-year survival rate (Rizvi S, et al., Semin Liver Dis. 2014; 34 (4): 456-64.
이와 관련하여, 담즙 상피 세포 성장을 조절하며, 담도암에서 과발현되는 표피 성장 인자 수용체(Epidermal growth factor receptor; EGFR)를 타겟으로한 약물의 사용이 증가하고 있다. 예로, 젬시타빈 및 옥살리플라틴(oxaliplatin)에 항-EGFR 항체인 세툭시맙(cetuximab)을 추가 투여하는 방법이 최근 시도되었으나, 임상 2상 시험에서 진행성 담도암 환자에 대한 효능이 향상되지 않았다. 또한, 젬시타빈 및 옥살리플라틴과 EGFR 티로신 인산화효소 억제제인 에를로티닙(erlotinib)의 병용 요법이 시도되었으나, 최근에 발표된 임상 3상 시험 결과에서는 화학 요법과 비교하는 경우에 개선된 효과를 나타내지 못하였다. 이에, 환자의 생존율을 향상시키는 화학 요법과 함께 새로운 표적 치료법이 필요한 실정이다.In this regard, the use of drugs targeting epidermal growth factor receptor (EGFR), which regulates bile epithelial cell growth and is overexpressed in biliary tract cancer, is increasing. For example, additional administration of cetuximab, an anti-EGFR antibody, to gemcitabine and oxaliplatin has recently been attempted, but efficacy in patients with advanced biliary tract cancer has not improved in phase II trials. In addition, combination therapy of gemcitabine and oxaliplatin with EGFR tyrosine kinase inhibitor erlotinib has been tried, but the recently released Phase III clinical trials did not show an improved effect compared to chemotherapy. . Thus, there is a need for new targeted therapies along with chemotherapy that improves patient survival.
한편, L1 세포 부착 분자(L1CAM; L1 cell adhesion molecule, CD171)는 200-220 kDa 막관통 당단백질로서, 6개의 면역 글로불린 유사 도메인, 5개의 피브로넥틴-타입 III 도메인, 막관통 스트레치(transmembrane stretch) 및 짧은 세포질 꼬리로 구성된다. L1CAM은 소뇌 세포의 이동 및 신경 돌기의 성장에 중요한 역할을 수행하는 중추 신경계의 신경 세포 접착 분자로서 흑색종(melanoma), 신경아세포종(neuroblastoma), 난소암, 대장암, 담관암 및 담낭암 등 여러 암에서 발현되고, 암세포의 성장과 전이에 중요한 역할을 하며, L1CAM 과발현이 암의 불량 예후와 관련이 있음이 보고되고 있다(Raveh et al., Cancer Letters 282: 137-145, 2009).On the other hand, L1 cell adhesion molecule (L1CAM) is a 200-220 kDa transmembrane glycoprotein, including six immunoglobulin-like domains, five fibronectin-type III domains, transmembrane stretch and It consists of a short cellular tail. L1CAM is a neuronal adhesion molecule of the central nervous system that plays an important role in the movement of cerebellar cells and the growth of neurites in many cancers, including melanoma, neuroblastoma, ovarian cancer, colon cancer, cholangiocarcinoma and gallbladder cancer. Has been reported to play an important role in the growth and metastasis of cancer cells, and that L1CAM overexpression is associated with poor prognosis of cancer (Raveh et al., Cancer Letters 282: 137-145, 2009).
본 발명자들은 암, 구체적으로 담도암을 효과적으로 치료하는 방법을 개발하고자 예의 노력 연구한 결과, L1CAM을 타겟하는 항체인 Ab417와 담도암 치료 약물인 젬시타빈 및/또는 시스플라틴을 병용 투여하는 경우, 담도암의 성장 억제에 대하여 상승효과를 나타냄을 확인하여, 본 발명을 완성하였다.The present inventors have diligently studied to develop a method for effectively treating cancer, specifically, biliary tract cancer. As a result, when the present invention is administered with a combination of Ab417, an antibody targeting L1CAM, and gemcitabine and / or cisplatin, the biliary tract cancer, It confirmed that the synergistic effect was shown with respect to the growth inhibition of, and completed this invention.
본 발명의 하나의 목적은 L1CAM(L1 cell adhesion molecule) 단백질에 특이적으로 결합하는 항체; 및 항암제, 구체적으로 피리미딘 유사체(pyrimidine analog), 플라틴계 항암제 또는 이들의 조합을 포함하는, 암의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.One object of the invention is an antibody that specifically binds to an L1 cell adhesion molecule (L1CAM) protein; And it provides a pharmaceutical composition for the prevention or treatment of cancer, including an anticancer agent, specifically pyrimidine analog, a platinum-based anticancer agent or a combination thereof.
본 발명의 다른 하나의 목적은 상기 조성물을 포함하는 키트를 제공하는 것이다.Another object of the present invention is to provide a kit comprising the composition.
본 발명의 다른 하나의 목적은 상기 항체; 및 피리미딘 유사체, 플라틴계 항암제 또는 이들의 조합을 이를 필요로 하는 개체에 투여하는 단계를 포함하는, 암의 예방 또는 치료 방법을 제공하는 것이다.Another object of the present invention is the antibody; And administering a pyrimidine analog, a platinum anticancer agent, or a combination thereof to a subject in need thereof.
L1CAM 단백질에 특이적으로 결합하는 항체 및 젬시타빈 및/또는 시스플라틴을 병용 투여하는 경우에는, 이들을 단독 투여하는 경우에 비하여 담도암의 성장 억제에 대하여 현저한 효과를 나타내므로, 상기 항체 및 약물을 포함하는 본 발명의 약학적 조성물은 담도암의 치료에 유용하게 활용될 수 있다.In the case of co-administration of an antibody and gemcitabine and / or cisplatin that specifically bind to L1CAM protein, it shows a remarkable effect on the growth inhibition of biliary tract cancer as compared to the case of administration alone, thus including the antibody and the drug. The pharmaceutical composition of the present invention can be usefully used for the treatment of biliary tract cancer.
도 1은 Choi-CK 이종이식 모델에서 Ab417의 항암 효과를 보여준다. Ab417(10 mg/kg), 비교군으로서 항-L1CAM 항체인 Hz2.2(10 mg/kg) 또는 동종형 대조군(hFc, 3.3 mg/kg)을 Choi-CK 세포를 갖는 누드 마우스(n=8)에 일주일에 세 번 정맥 주사하였다. A는 종양 부피 및 B는 종양 무게를 나타낸다. 각 지점은 '평균 ± 표준편차'를 나타내며, Dunnett t-검정(Dunnett's t-test)에 의한 p < 0.05 (*) 및 p < 0.01 (**)는 동종형 대조군에 비하여 유의적인 차이를 나타냄을 의미한다.1 shows the anticancer effect of Ab417 in Choi-CK xenograft model. Ab417 (10 mg / kg), as a comparison group anti-L1CAM antibody Hz2.2 (10 mg / kg) or isotype control (hFc, 3.3 mg / kg) in nude mice with Choi-CK cells ( n = 8 ) Intravenously three times a week. A is the tumor volume and B is the tumor weight. Each point represents 'mean ± standard deviation', and p <0.05 (*) and p <0.01 (**) by Dunnett's t-test show significant differences compared to homologous controls. it means.
도 2는 Choi-CK 이종이식 모델에서 Ab417의 항암 효과를 보여준다. Ab417(10 mg/kg) 또는 동종형 대조군(hFc, 3.3 mg/kg)을 Choi-CK 세포를 갖는 누드 마우스(n=8)에 일주일에 두 번 정맥 주사하였다. A는 종양 부피, B는 종양 무게 및 C는 몸무게를 나타낸다. 각 지점은 '평균 ± 표준편차'를 나타내며, Dunnett t-검정(Dunnett's t-test)에 의한 p < 0.05 (*) 및 p < 0.01 (**), 및 Steel 검정(Steel's test)에 의한 p < 0.05 (#)는 동종형 대조군에 비하여 유의적인 차이를 나타냄을 의미한다. D는 종양 섹션에서 항-인간 Ki-67 항체로 염색된 영역을 나타낸다. Ki-67 지수(index)는 전체 면적에 대한 양성 염색부위의 백분율로 나타내었다.2 shows the anticancer effect of Ab417 in Choi-CK xenograft model. Ab417 (10 mg / kg) or isotype control (hFc, 3.3 mg / kg) was injected intravenously twice a week in nude mice with Choi-CK cells ( n = 8). A is tumor volume, B is tumor weight and C is body weight. Each point p by "mean ± standard deviation" a represents, Dunnett t- test (Dunnett's t-test) p <0.05 (*) and p <0.01 (**), and Steel test (Steel's test) according to the < 0.05 ( # ) means significant difference compared to the homologous control. D represents the area stained with anti-human Ki-67 antibody in the tumor section. The Ki-67 index is expressed as a percentage of the positive staining site relative to the total area.
도 3은 L1CAM가 발현된 Choi-CK에 결합한 Ab417의 모습, 및 상기 Ab417의 막-L1CAM 수준 감소 효과를 보여준다. A는 Ab417 내면화의 공초점 현미경 분석을 보여준다. SCK-L1 세포를 지정된 시간 동안 Ab417(10 ㎍/mL)로 전처리하였고, 막 L1CAM에 결합된 Ab417는 항-인간 IgG(H+L) Cross Adsorbed DyLight 594 (적색), 및 내면화 Ab417는 항-인간IgG (Fc-특이적)-FITC(녹색)을 이용하여 탐지하였다. B는 지정된 시간 동안 Ab417(10 ㎍/mL)로 처리된 Choi-CK 세포를 나타낸다. 막 L1CAM는 A10-A3을 이용하여 웨스턴블롯 분석으로 탐지하였고, 판-카드헤린(pan-cadherin)은 로딩 컨트롤로 사용하였다. Ab417로 처리되지 않은 세포는 대조군(C)으로 사용하였다. C는 PBS(위) 또는 Ab417(아래)로 처리된 Choi-CK 종양에 대하여 L1CAM으로 염색한, 400배로 확대된 공초점 현미경 이미지이다. 전체 면적에 대한 양성 염색 부위의 퍼센트로 나타내었다. D는 종양 섹션에서 인간 L1CAM의 N-말단 부위에 특이적인 항체로 염색된 영역을 나타낸다. 전체 면적에 대한 양성 염색부위의 백분율로 나타내었다.3 shows the appearance of Ab417 bound to Choi-CK expressed L1CAM, and the effect of reducing the membrane-L1CAM level of Ab417. A shows confocal microscopy analysis of Ab417 internalization. SCK-L1 cells were pretreated with Ab417 (10 μg / mL) for the indicated time, Ab417 bound to membrane L1CAM was anti-human IgG (H + L) Cross Adsorbed DyLight 594 (red), and internalized Ab417 was anti-human Detection was done using IgG (Fc-specific) -FITC (green). B represents Choi-CK cells treated with Ab417 (10 μg / mL) for the indicated time. Membrane L1CAM was detected by Western blot analysis using A10-A3, and pan-cadherin was used as loading control. Cells not treated with Ab417 were used as a control (C). C is a 400x magnified confocal microscopy image stained with L1CAM for Choi-CK tumors treated with PBS (top) or Ab417 (bottom). It is expressed as a percentage of the positive staining site relative to the total area. D represents the region stained with an antibody specific for the N-terminal region of human L1CAM in the tumor section. It is expressed as a percentage of the positive staining area relative to the total area.
도 4는 Choi-CK 이종 모델에서 젬시타빈 또는 시스플라틴의 항암 효과를 보여준다. 3주 또는 4주 동안 일주일에 두 번씩 지정된 용량의 약물을 복강 내 주사하였다. A는 종양 부피, B는 종양 무게를 나타낸다. 각 지점은 '평균 ± 표준편차'를 나타내며, Dunnett 검정에 의한 p < 0.05 (*) 및 p < 0.01 (**), 및 Steel 검정에 의한 p < 0.05 (#) 및 p < 0.01 (##)는 대조군(saline)에 비하여 유의적인 차이를 나타냄을 의미한다.4 shows the anticancer effects of gemcitabine or cisplatin in Choi-CK heterogeneous model. Intraperitoneal injections of the indicated doses of the drug twice a week for three or four weeks. A is the tumor volume, B is the tumor weight. Each point represents 'mean ± standard deviation', p <0.05 (*) and p <0.01 (**) by Dunnett test, and p <0.05 ( # ) and p <0.01 ( ## ) by Steel test Means a significant difference compared to the control (saline).
도 5는 Choi-CK 이종이식 모델에서 Ab417 및 젬시타빈, 또는 시스플라틴 병용 투여의 항암효과를 보여준다. A 및 C는 종양 부피, B 및 D는 종양 무게를 나타낸다. 각 지점은 '평균 ± 표준편차'를 나타내며, Dunnett 검정에 의한 p < 0.05 (*) 및 p < 0.01 (**), 및 Steel 검정에 의한 p < 0.05 (#) 및 p < 0.01 (##)는 대조군(saline)에 비하여 유의적인 차이를 나타냄을 의미한다.Figure 5 shows the anticancer effect of the combination of Ab417 and gemcitabine, or cisplatin in Choi-CK xenograft model. A and C represent tumor volume, B and D represent tumor weight. Each point represents 'mean ± standard deviation', p <0.05 (*) and p <0.01 (**) by Dunnett test, and p <0.05 ( # ) and p <0.01 ( ## ) by Steel test Means a significant difference compared to the control (saline).
도 6은 Choi-CK 이종이식 모델에서 Ab417 및 젬시타빈, 또는 시스플라틴 병용 투여의 몸무게 변화 정도를 보여준다. A는 젬시타빈 또는 시스플라틴 단독 투여, B는 Ab417 또는 젬시타빈 단독 투여, 및 Ab417과 젬시타빈의 병용 투여, C는 Ab417 또는 시스플라틴 단독 투여, 및 Ab417과 시스플라틴의 병용 투여에 관한 것이다. 각 지점은 '평균 ± 표준편차'를 나타낸다.FIG. 6 shows the degree of weight change of Ab417 and gemcitabine, or cisplatin combined administration in Choi-CK xenograft model. A relates to administration of gemcitabine or cisplatin alone, B to administration of Ab417 or gemcitabine alone, and combination of Ab417 and gemcitabine, C to administration of Ab417 or cisplatin alone, and the combination of Ab417 and cisplatin. Each point represents a mean ± standard deviation.
도 7은 TFK-1 세포에서 Ab417에 의해 발현이 억제된 막 L1CAM 수준을 보여준다.7 shows membrane L1CAM levels suppressed by Ab417 in TFK-1 cells.
도 8 TFK-1 이종이식 모델에서 Ab417의 항암 효과를 보여준다. A는 종양 부피 및 B는 종양 무게를 나타낸다. 각 지점은 '평균 ± 표준편차'를 나타내며, Dunnett t-검정(Dunnett's t-test)에 의한 p < 0.01 (**)는 동종형 대조군(hFc)에 비하여 유의적인 차이를 나타냄을 의미한다.8 shows the anticancer effect of Ab417 in the TFK-1 xenograft model. A is the tumor volume and B is the tumor weight. Each point represents 'mean ± standard deviation', and p <0.01 (**) by Dunnett's t-test indicates a significant difference compared to the homologous control (hFc).
도 9는 TFK-1 이종이식 모델에서 젬시타빈 또는 시스플라틴의 항암 효과를 보여준다. A는 종양 부피 및 B는 종양 무게를 나타낸다. 각 지점은 '평균 ± 표준편차'를 나타내며, Dunnett 검정에 의한 p < 0.05 (*) 및 p < 0.01 (**)는 대조군(saline)에 비하여 유의적인 차이를 나타냄을 의미한다.9 shows the anticancer effect of gemcitabine or cisplatin in TFK-1 xenograft model. A is the tumor volume and B is the tumor weight. Each point represents 'mean ± standard deviation', meaning that p <0.05 (*) and p <0.01 (**) by Dunnett's test indicate a significant difference compared to the saline.
도 10은 TFK-1 이종이식 모델에서 Ab417 및 젬시타빈, 또는 시스플라틴 병용 투여의 항암효과를 보여준다. A는 종양 부피 및 B는 몸무게를 나타낸다. 각 지점은 '평균 ± 표준편차'를 나타내며, Dunnett 검정에 의한 p < 0.05 (*) 및 p < 0.01 (**)는 대조군(saline)에 비하여 유의적인 차이를 나타냄을 의미한다.10 shows the anticancer effect of the combination of Ab417 and gemcitabine, or cisplatin in TFK-1 xenograft model. A is the tumor volume and B is the weight. Each point represents 'mean ± standard deviation', meaning that p <0.05 (*) and p <0.01 (**) by Dunnett's test indicate a significant difference compared to the saline.
상기 목적을 달성하기 위하여, 본 발명의 하나의 양태는 L1CAM(L1 cell adhesion molecule) 단백질에 특이적으로 결합하는 항체; 및 항암제, 구체적으로 피리미딘 유사체(pyrimidine analog), 플라틴계 항암제 또는 이들의 조합을 포함하는, 암의 예방 또는 치료용 약학적 조성물을 제공한다.In order to achieve the above object, one embodiment of the present invention comprises an antibody that specifically binds to L1 cell adhesion molecule (L1CAM) protein; And it provides a pharmaceutical composition for the prevention or treatment of cancer, including an anticancer agent, specifically pyrimidine analog (pyrimidine analog), a platinum-based anticancer agent or a combination thereof.
본 발명에서는, L1CAM 단백질에 결합하는 항체와 피리미딘 유사체인 젬시타빈 또는 플라틴계 항암제인 시스플라틴을 병용 투여하는 경우, 상기 항체 및 약물을 단독 투여하는 경우에 비하여 담도암의 치료 효율이 현저히 증가함을 확인하였다.In the present invention, when a combination of an antibody binding to L1CAM protein and a pyrimidine analog, gemcitabine or cisplatin, a platinum anticancer agent, the treatment efficiency of biliary tract cancer is significantly increased as compared with the administration of the antibody and drug alone. Confirmed.
본 발명에서 용어, "L1CAM(L1 cell adhesion molecule)"은 면역글로불린 수퍼패밀리 세포부착단백질들(immunoglobulin superfamily cell adhesion molecules, CAMs)에 속하는 내재성 막 당단백질(integral membrane glycoprotein)중 하나를 의미한다. 상기 L1CAM은 흑색종(melanoma), 신경아세포종(neuroblastoma), 난소암, 대장암, 담관암 및 담낭암 등 여러 암에서 발현되고, 암세포의 성장과 전이에 중요한 역할을 하며, L1CAM 과발현이 암의 불량 예후와 관련이 있음이 보고되고 있다(Raveh et al., Cancer Letters 282: 137-145, 2009). As used herein, the term "L1 CAM" refers to one of the intrinsic membrane glycoproteins belonging to immunoglobulin superfamily cell adhesion molecules (CAMs). The L1CAM is expressed in a variety of cancers, including melanoma, neuroblastoma, ovarian cancer, colorectal cancer, cholangiocarcinoma and gallbladder cancer, and plays an important role in the growth and metastasis of cancer cells. Relevance has been reported (Raveh et al., Cancer Letters 282: 137-145, 2009).
본 발명의 항체는 인간 및 마우스 L1CAM 단백질 모두에 고친화적으로 결합하며 현저한 암 성장 억제 효과를 나타내므로, L1CAM이 과발현되는 암의 예방 또는 치료 분야에 유용하게 사용할 수 있다.Since the antibody of the present invention binds to both human and mouse L1CAM proteins with high affinity and exhibits significant cancer growth inhibitory effect, it can be usefully used in the field of prevention or treatment of cancer in which L1CAM is overexpressed.
본 발명의 용어, "항체"는 면역학적으로 특정 항원과 반응성을 갖는 면역글로불린 분자를 포함하는, 항원을 특이적으로 인식하는 수용체 역할을 하는 단백질 분자를 의미하며, 다클론항체, 단일클론항체, 전체(whole) 항체 및 항체 단편을 모두 포함한다. 또한, 상기 용어는 키메라성 항체(예를 들면, 인간화 뮤린 항체) 및 이가(bivalent) 또는 이중특이성 분자(예를 들어, 이중특이성 항체), 디아바디, 트리아바디 및 테트라바디를 포함한다. 상기 용어는 추가로 FcRn에 대한 결합 기능을 보유한 단쇄 항체, 스캡, 항체 불변영역의 유사체 및 단백질 스캐폴드에 기초한 인공 항체를 포함한다. 전체 항체는 2개의 전체 길이의 경쇄 및 2개의 전체 길이의 중쇄를 가지는 구조이며, 각각의 경쇄는 중쇄와 다이설파이드 결합으로 연결되어 있다. 상기 전체 항체는 IgA, IgD, IgE, IgM 및 IgG를 포함하며, IgG는 아형(subtype)으로, IgG1, IgG2, IgG3 및 IgG4를 포함한다. 상기 항체 단편은 항원결합 기능을 보유하고 있는 단편을 의미하며, Fd, Fab, Fab', F(ab')2 및 Fv 등을 포함한다. 상기 Fd는 Fab 단편에 포함되어 있는 중쇄 부분을 의미한다. 상기 Fab는 경쇄 및 중쇄의 가변영역과 경쇄의 불변 영역 및 중쇄의 첫 번째 불변 영역(CH1 도메인)을 가지는 구조로 1개의 항원 결합 부위를 가진다. Fab'는 중쇄 CH1 도메인의 C 말단에 하나 이상의 시스테인 잔기를 포함하는 힌지 영역(hinge region)을 가진다는 점에서 Fab와 차이가 있다. F(ab')2 항체는 Fab'의 힌지 영역의 시스테인 잔기가 디설파이드 결합을 이루면서 생성된다. Fv(variable fragment)는 중쇄 가변부위 및 경쇄 가변부위만을 가지고 있는 최소의 항체조각을 의미한다. 이중디설파이드Fv(dsFv)는 디설파이드 결합으로 중쇄 가변부위와 경쇄 가변부위가 연결되어 있고 단쇄 Fv(scFv)는 일반적으로 펩타이드 링커를 통하여 중쇄의 가변 영역과 경쇄의 가변 영역이 공유 결합으로 연결되어 있다. 이러한 항체 단편은 단백질 가수분해 효소를 이용해서 얻을 수 있고(예를 들어, 전체 항체를 파파인으로 제한 절단하며 Fab를 얻을 수 있고 펩신으로 절단하면 F(ab')2 단편을 얻을 수 있다), 바람직하게는 유전자 재조합 기술을 통하여 제작할 수 있다.As used herein, the term “antibody” refers to a protein molecule that acts as a receptor that specifically recognizes an antigen, including an immunoglobulin molecule that is immunologically reactive with a specific antigen, including polyclonal antibodies, monoclonal antibodies, It includes both whole antibodies and antibody fragments. The term also includes chimeric antibodies (eg, humanized murine antibodies) and bivalent or bispecific molecules (eg, bispecific antibodies), diabodies, triabodies, and tetrabodies. The term further includes artificial antibodies based on protein scaffolds and single chain antibodies, caps, analogs of antibody constant regions that possess binding functions for FcRn. The total antibody is a structure having two full length light chains and two full length heavy chains, each of which is linked by heavy and disulfide bonds. The total antibody includes IgA, IgD, IgE, IgM and IgG, and IgG is a subtype, including IgG1, IgG2, IgG3 and IgG4. The antibody fragment refers to a fragment having an antigen binding function, and includes Fd, Fab, Fab ', F (ab') 2 and Fv. Fd means the heavy chain portion included in the Fab fragment. The Fab has one antigen binding site in a structure having a variable region of the light and heavy chains, a constant region of the light chain, and a first constant region of the heavy chain (CH1 domain). Fab 'differs from Fab in that it has a hinge region comprising at least one cysteine residue at the C terminus of the heavy chain CH1 domain. F (ab ') 2 antibodies are produced when the cysteine residues of the hinge region of Fab' form disulfide bonds. Fv (variable fragment) means a minimum antibody fragment having only the heavy chain variable region and light chain variable region. Double disulfide Fv (dsFv) is a disulfide bond is linked to the heavy chain variable region and the light chain variable region, and short chain Fv (scFv) is generally covalently linked to the variable region of the heavy chain and the light chain through a peptide linker. Such antibody fragments can be obtained using proteolytic enzymes (e.g., the entire antibody can be restricted to papain and Fab can be obtained, and pepsin can yield F (ab ') 2 fragment). Can be produced through genetic recombination techniques.
본 발명에서, 상기 '면역글로불린'은 중쇄 및 경쇄를 가지며, 각각의 중쇄 및 경쇄는 불변 영역 및 가변 영역(상기 부위는 도메인으로 또한 알려져 있음)을 포함한다. 경쇄 및 중쇄의 가변 영역은, 상보성 결정 영역(complementarity-determining region, 이하 "CDR"이라 함)이라 불리우는 3개의 다변가능한 영역 및 4개의 구조 영역(framework region)을 포함한다. 상기 CDR은 주로 항원의 에피토프(epitope)에 결합하는 역할을 한다. 각 사슬의 CDR은 전형적으로 N-말단으로부터 시작하여 순차적으로 CDR1, CDR2, CDR3로 불리우고, 또한 특정 CDR이 위치하고 있는 사슬에 의해서 식별된다.In the present invention, the 'immunoglobulin' has a heavy chain and a light chain, each heavy and light chain comprises a constant region and a variable region (the region is also known as a domain). The variable regions of the light and heavy chains comprise three multivariable regions and four framework regions called complementarity-determining regions (hereinafter referred to as "CDRs"). The CDRs mainly serve to bind epitopes of antigens. The CDRs of each chain are typically called CDR1, CDR2, CDR3, starting sequentially from the N-terminus and also identified by the chain in which the particular CDR is located.
본 발명에서 용어, "L1CAM(L1 cell adhesion molecule) 단백질에 특이적으로 결합하는 항체"는 L1CAM 단백질에 결합하여 L1CAM 단백질의 활성을 저해할 수 있는 항체를 의미한다. 본 발명의 목적상, 상기 L1CAM에 특이적으로 결합하는 항체는 L1CAM의 Ig5 영역에 결합하는 항체이나, 이에 제한되지 않는다. As used herein, the term "an antibody that specifically binds to an L1 cell adhesion molecule (L1CAM) protein" refers to an antibody that binds to an L1CAM protein and inhibits the activity of the L1CAM protein. For the purposes of the present invention, the antibody that specifically binds to L1CAM is an antibody that binds to the Ig5 region of L1CAM, but is not limited thereto.
구체적으로, 상기 L1CAM 단백질에 특이적으로 결합하는 항체는 서열번호 1의 중쇄 가변영역 및 서열번호 5의 경쇄 가변영역을 포함하는 항체 또는 이의 기능성 변이체일 수 있다.Specifically, the antibody specifically binding to the L1CAM protein may be an antibody or a functional variant thereof comprising the heavy chain variable region of SEQ ID NO: 1 and the light chain variable region of SEQ ID NO: 5.
본 발명에서 용어, "서열번호 1의 중쇄 가변영역 및 서열번호 5의 경쇄 가변영역을 포함하는 항체의 기능성 변이체"는 서열번호 1의 중쇄 가변영역 및 서열번호 5의 경쇄 가변영역을 포함하는 모항체와 비교하여 중쇄 가변영역 및/또는 경쇄 가변영역의 1개 이상의 아미노산에 차이가 있는 항체로서, 상기 모항체에 비해 물성이 개선된 항체일 수 있다. 또한, 상기 기능성 변이체는 모항체와 동등한 생물학적 활성을 보유할 수 있다.As used herein, the term "functional variant of an antibody comprising a heavy chain variable region of SEQ ID NO: 1 and a light chain variable region of SEQ ID NO: 5" refers to a parent antibody comprising a heavy chain variable region of SEQ ID NO: 1 and a light chain variable region of SEQ ID NO: 5 Compared with the antibody having a difference in at least one amino acid of the heavy chain variable region and / or light chain variable region, it may be an antibody having improved physical properties compared to the parent antibody. In addition, the functional variant may retain biological activity equivalent to that of the parent antibody.
특히, 상기 항체는 서열번호 1의 중쇄 가변영역 및 서열번호 5의 경쇄 가변영역을 포함하는 모항체와 비교하여 중쇄 가변영역 및/또는 경쇄 가변영역의 1개 내지 10개의 아미노산에서 차이가 있는 항체, 구체적으로 서열번호 1의 중쇄 가변영역 및 서열번호 5의 경쇄 가변영역을 포함하는 모항체의 중쇄 가변영역 및/또는 경쇄 가변영역에서 1 이상의 아미노산, 구체적으로 1 내지 10개, 보다 더 구체적으로 1개 내지 6개의 아미노산이 치환된 것일 수 있다. In particular, the antibody is an antibody having a difference in 1 to 10 amino acids of the heavy chain variable region and / or the light chain variable region as compared to the parent antibody comprising the heavy chain variable region of SEQ ID NO: 1 and the light chain variable region of SEQ ID NO: 5, Specifically, at least one amino acid, in particular 1 to 10, more specifically 1 in the heavy chain variable region and / or light chain variable region of the parent antibody comprising a heavy chain variable region of SEQ ID NO: 1 and a light chain variable region of SEQ ID NO: To 6 amino acids may be substituted.
이러한 항체는 단일클론항체일 수 있으며, 전장 항체 또는 이의 항체 단편 형태일 수 있다.Such antibodies may be monoclonal antibodies, and may be in the form of full length antibodies or antibody fragments thereof.
보다 구체적으로, 이러한 항체는 서열번호 2로 기재된 중쇄 CDR1; 서열번호 3 또는 10으로 기재된 중쇄 CDR2; 및 서열번호 4로 기재된 중쇄 CDR3를 포함하는 중쇄 가변영역과 서열번호 6 또는 12로 기재된 경쇄 CDR1; 서열번호 7로 기재된 경쇄 CDR2; 및 서열번호 8로 기재된 경쇄 CDR3를 포함하는 경쇄 가변영역을 포함하는 것일 수 있으나, 이에 제한되는 것은 아니다. 또한, 상기 항체는 서열번호 1, 9 및 13으로 이루어진 군으로부터 선택된 중쇄 가변영역; 및 서열번호 5, 11, 14 및 15로 이루어진 군으로부터 선택된 경쇄 가변영역을 포함하는 것일 수 있으나, 이에 제한되는 것은 아니다.More specifically, such antibodies include heavy chain CDR1 as set forth in SEQ ID NO: 2; Heavy chain CDR2 set forth in SEQ ID NO: 3 or 10; And a heavy chain variable region comprising a heavy chain CDR3 as set out in SEQ ID NO: 4 and a light chain CDR1 as set out in SEQ ID NO: 6 or 12; Light chain CDR2 set forth in SEQ ID NO: 7; And a light chain variable region comprising the light chain CDR3 set forth in SEQ ID NO: 8, but is not limited thereto. In addition, the antibody comprises a heavy chain variable region selected from the group consisting of SEQ ID NOs: 1, 9 and 13; And a light chain variable region selected from the group consisting of SEQ ID NOs: 5, 11, 14, and 15, but is not limited thereto.
상기 L1CAM에 특이적으로 결합하는 항체는 대한민국 공개특허 제10-2014-0066101 A호, WO2014-077648 A1, 또는 US 2015-0344571 Al에 개시되어 있는 항체일 수 있다. 상기 특허의 명세서 전문은 참고자료로서 모두 본원에 포함된다.The antibody specifically binding to L1CAM may be an antibody disclosed in Korean Patent Application Laid-Open No. 10-2014-0066101 A, WO2014-077648 A1, or US 2015-0344571 Al. The entire specification of the patent is incorporated herein by reference in its entirety.
구체적으로, 상기 L1CAM에 특이적으로 결합하는 항체는 다음과 같은 것일 수 있으나, 특별히 이에 제한되는 것은 아니다:Specifically, the antibody specifically binding to L1CAM may be as follows, but is not particularly limited thereto:
(1) 서열번호 2로 기재된 중쇄 CDR1; 서열번호 3으로 기재된 중쇄 CDR2; 및 서열번호 4로 기재된 중쇄 CDR3를 포함하는 중쇄 가변영역과 서열번호 6으로 기재된 경쇄 CDR1; 서열번호 7로 기재된 경쇄 CDR2; 및 서열번호 8로 기재된 경쇄 CDR3를 포함하는 경쇄 가변영역을 포함하는 것일 수 있으며, 보다 구체적으로 서열번호 1의 중쇄 가변영역 및 서열번호 5의 경쇄 가변영역을 포함하는 항체; 또는 서열번호 13의 중쇄 가변영역 및 서열번호 15의 경쇄 가변영역을 포함하는 항체일 수 있다. 서열번호 1의 중쇄 가변영역 및 서열번호 5의 경쇄 가변영역을 포함하는 항체는 본 발명에서 Ab417로 명명되며, 서열번호 13의 중쇄 가변영역 및 서열번호 15의 경쇄 가변영역을 포함하는 항체는 본 발명에서 Ab417-H5L1으로 명명된다. (1) a heavy chain CDR1 set forth in SEQ ID NO: 2; Heavy chain CDR2 set forth in SEQ ID NO: 3; And a heavy chain variable region comprising a heavy chain CDR3 as set out in SEQ ID NO: 4 and a light chain CDR1 as set out in SEQ ID NO: 6; Light chain CDR2 set forth in SEQ ID NO: 7; And a light chain variable region comprising the light chain CDR3 described in SEQ ID NO: 8, and more specifically, an antibody comprising a heavy chain variable region of SEQ ID NO: 1 and a light chain variable region of SEQ ID NO: 5; Or an antibody comprising a heavy chain variable region of SEQ ID NO: 13 and a light chain variable region of SEQ ID NO: 15. An antibody comprising the heavy chain variable region of SEQ ID NO: 1 and the light chain variable region of SEQ ID NO: 5 is named Ab417 in the present invention, and the antibody comprising the heavy chain variable region of SEQ ID NO: 13 and the light chain variable region of SEQ ID NO: 15 is provided herein. Is designated Ab417-H5L1.
(2) 서열번호 2로 기재된 중쇄 CDR1; 서열번호 10으로 기재된 중쇄 CDR2; 및 서열번호 4로 기재된 중쇄 CDR3를 포함하는 중쇄 가변영역과 서열번호 12로 기재된 경쇄 CDR1; 서열번호 7로 기재된 경쇄 CDR2; 및 서열번호 8로 기재된 경쇄 CDR3를 포함하는 경쇄 가변영역을 포함하는 것일 수 있으며, 보다 구체적으로 서열번호 9의 중쇄 가변영역 및 서열번호 11의 경쇄 가변영역을 포함하는 항체; 또는 서열번호 9의 중쇄 가변영역 및 서열번호 14의 경쇄 가변영역을 포함하는 항체일 수 있다. 서열번호 9의 중쇄 가변영역 및 서열번호 11의 경쇄 가변영역을 포함하는 항체는 본 발명에서 Ab417-H6L2로 명명되며, 서열번호 9의 중쇄 가변영역 및 서열번호 14의 경쇄 가변영역을 포함하는 항체는 본 발명에서 Ab417-H6L6로 명명된다.(2) a heavy chain CDR1 set forth in SEQ ID NO: 2; A heavy chain CDR2 set forth in SEQ ID NO: 10; And a heavy chain variable region comprising a heavy chain CDR3 as set out in SEQ ID NO: 4 and a light chain CDR1 as set out in SEQ ID NO: 12; Light chain CDR2 set forth in SEQ ID NO: 7; And it may include a light chain variable region comprising a light chain CDR3 described in SEQ ID NO: 8, More specifically, the antibody comprising a heavy chain variable region of SEQ ID NO: 9 and a light chain variable region of SEQ ID NO: 11; Or an antibody comprising a heavy chain variable region of SEQ ID NO: 9 and a light chain variable region of SEQ ID NO: 14. The antibody comprising the heavy chain variable region of SEQ ID NO: 9 and the light chain variable region of SEQ ID NO: 11 is named Ab417-H6L2 in the present invention, and the antibody comprising the heavy chain variable region of SEQ ID NO: 9 and the light chain variable region of SEQ ID NO: 14 It is named Ab417-H6L6 in the present invention.
상기 기술된 Ab417-H5L1, -H6L2, 및 -H6L6는 모두 본 출원인의 선행특허, 즉 대한민국 공개특허 제10-2014-0066101A호, WO2014-077648 A1, 또는 US 2015-0344571 Al 등에서 Ab417에 비해 물성이 개선된 항체로서 개발된 것으로, Ab417과 동등한 효과를 나타낼 수 있다.Ab417-H5L1, -H6L2, and -H6L6 described above are all properties of the applicant compared to Ab417 in the prior patents of the applicant, that is, Korean Patent Application Publication No. 10-2014-0066101A, WO2014-077648 A1, or US 2015-0344571 Al. It was developed as an improved antibody and can show an effect equivalent to Ab417.
본 발명에서 이용되는 서열은, 생물학적으로 균등 활성을 갖는 변이를 고려한다면, 서열목록에 기재된 서열과 실질적인 동일성(substantial identity)을 나타내는 서열도 포함하는 것으로 해석된다. 상기 용어, '실질적인 동일성'은 본 발명의 서열과 임의의 다른 서열을 최대한 대응되도록 얼라인(align)하고, 당업계에서 통상적으로 이용되는 알고리즘을 이용하여 얼라인된 서열을 분석한 경우에, 최소 60%의 상동성, 더욱 구체적으로 70%의 상동성, 더더욱 구체적으로 80%의 상동성, 가장 구체적으로 90%의 상동성을 나타내는 서열을 의미한다.The sequences used in the present invention are to be construed to include sequences that exhibit substantial identity with the sequences listed in the Sequence Listing, in view of variations that are biologically equivalent. The term 'substantial identity' means that if the alignment of the sequence of the present invention with any other sequence is maximally matched, the aligned sequence is analyzed using algorithms commonly used in the art. By 60% homology, more specifically 70% homology, even more specifically 80% homology, most specifically 90% homology.
따라서, 상기 서열번호 1 내지 15로 표시되는 서열과 높은 상동성을 갖는 아미노산 서열, 예를 들면 그 상동성이 70% 이상, 구체적으로 80% 이상, 더욱 구체적으로 90% 이상의 높은 상동성을 갖는 아미노산 서열도 본 발명의 범위에 포함되는 것으로 해석되어야 한다.Therefore, an amino acid sequence having high homology with the sequence represented by SEQ ID NOS: 1 to 15, for example, an amino acid having a high homology of 70% or more, specifically 80% or more, more specifically 90% or more. Sequences should also be construed as falling within the scope of the present invention.
본 발명의 용어, "피리미딘 유사체(pyrimidine analog)"는 대사성 피리미딘의 구조를 모방하는 뉴클레오시드(nucleoside) 유사체 화합물을 의미한다. 상기 피리미딘 유사체는 세포 내(intracellular) 화학치료제, 세포 내 항암제, DNA 바이러스에 대한 항-바이러스제 또는 핵산 성분 등으로 사용될 수 있다.As used herein, the term "pyrimidine analog" refers to a nucleoside analog compound that mimics the structure of metabolic pyrimidine. The pyrimidine analogs can be used as intracellular chemotherapeutic agents, intracellular anticancer agents, anti-viral agents or nucleic acid components for DNA viruses, and the like.
본 발명의 목적상, 상기 피리미딘 유사체는 항암제로 사용되는 것일 수 있으며, 구체적인 예로 젬시타빈(gemcitabine)일 수 있으나, 이에 제한되는 것은 아니다. For the purposes of the present invention, the pyrimidine analogue may be used as an anticancer agent, and may be, for example, gemcitabine, but is not limited thereto.
본 발명의 용어, "플라틴계 항암제"(Platinum-based anticancer drugs)는 백금을 함유한 중금속 화합물로서 DNA를 손상시키는 약물로 사용되는 항암제를 의미한다. 주로 유방암, 방광암, 위암 및 자궁경부암 등과 같은 다양한 종류의 암을 치료하는데 사용되고 있으며, DNA 가닥상의 인접한 2개의 구아닌과 결합하여 사슬 내 가교(interstrand crosslink)를 형성하여 DNA 합성을 억제함으로써 항암효과를 나타낸다고 알려져 있다. 구체적으로, 상기 플라틴계 항암제는 시스플라틴(cisplatin), 옥살리플라틴(oxaliplatin), 테트라플라틴(tetraplatin), 헵타플라틴(heptaplatin), 파라플라틴(paraplatin), 카보플라틴(carboplatin) 또는 나노플라틴(nanoplatin)일 수 있으며, 더욱 구체적으로 시스플라틴일 수 있으나, 이에 제한되는 것은 아니다.As used herein, the term "platinum-based anticancer drugs" refers to anti-cancer drugs used as drugs that damage DNA as a heavy metal compound containing platinum. It is mainly used to treat various kinds of cancers such as breast cancer, bladder cancer, gastric cancer and cervical cancer, and combines with two adjacent guanines on DNA strands to form an interstrand crosslink to inhibit DNA synthesis. Known. Specifically, the platinum-based anticancer agent is cisplatin (cisplatin), oxaliplatin (oxaliplatin), tetraplatin (tetraplatin), heptaplatin (heptaplatin), paraplatin (carplatin) (carboplatin) or nanoplatin ( nanoplatin), more specifically cisplatin, but is not limited thereto.
본 발명에서, 암의 종류는 특별히 이에 제한되지 않으나, L1CAM이 과발현되는 암일 수 있다. 구체적으로, 흑색종(melanoma), 신경아세포종(neuroblastoma), 난소암, 대장암, 담도암(담관암 또는 담낭암)일 수 있으나, 이에 제한되지 않는다.In the present invention, the type of cancer is not particularly limited, but may be a cancer in which L1CAM is overexpressed. Specifically, melanoma, neuroblastoma, ovarian cancer, colorectal cancer, biliary tract cancer (biliary duct cancer or gallbladder cancer), but is not limited thereto.
본 발명의 용어, "담도암"(biliary tract cancer)은 '담관암' 및 '담낭암'을 총칭하는 것으로서, 담낭 내부를 둘러싸고 있는 상피세포에서 발생하는 악성종양을 의미한다. 담관암은 간에서 배출하는 담즙의 통로인 담관에서 생기는 암이며, 담낭암은 상기 담즙을 1차적으로 저류하는 장소인 담낭에서 생기는 암을 의미한다. 담도암은 종양의 위치에 따라 간내 담도암과 간외 담도암으로 나눌 수 있는데, 간내 담도암은 주변부 담도암(peripheral cholangiocarcinoma)과 간문부 담도암(hilar cholangiocarcinoma)으로; 간외 담도암은 발생 부위에 따라 상부(근위부), 중부, 하부(원위부) 담도암으로 구분될 수 있다.As used herein, the term "biliary tract cancer" refers to "biliary duct cancer" and "gallbladder cancer" and refers to a malignant tumor occurring in epithelial cells surrounding the inside of the gallbladder. Bile duct cancer is a cancer that occurs in the bile duct that is the passage of bile discharged from the liver, and gallbladder cancer refers to a cancer that occurs in the gallbladder, which is a place where the bile is first stored. Biliary cancers can be divided into intrahepatic biliary and extrahepatic biliary tract cancers, depending on the location of the tumor, which may be divided into peripheral cholangiocarcinoma and hepatic cholangiocarcinoma; Extrahepatic biliary tract cancer may be classified into upper (proximal), middle, and lower (distal) biliary tract cancers according to the occurrence site.
담도암은 상당히 진행되기 전까지는 특징적인 증상이 나타나지 않아 조기 진단이 매우 어렵다. 또한, 담도, 혈관계에 해부학적 구조가 다양하고, 수술 전, 심지어는 수술 중에도 정확한 종양 침습 범위를 판단하기가 어려워 근치적 절제(암이 존재하거나 존재할 가능성이 있는 모든 부위를 최대한 제거하는 것)가 불가능한 경우도 있다. 항암 화학요법은 암이 전이되어 수술이 힘든 경우나 수술 후에 남아 있을 수 있는 암세포들의 성장을 막기 위해 시행되는데, 담도암의 치료에 있어 많은 발전이 있었으나 아직은 그 효과가 확실히 증명되지는 않은 상태이다.Biliary cancers are difficult to diagnose early because they do not show any characteristic symptoms until they progress significantly. In addition, various anatomical structures in the biliary tract and vasculature, and it is difficult to determine the exact extent of tumor invasion before or even during surgery, resulting in curative resection (to remove all areas where cancer exists or is likely to exist). Sometimes it is impossible. Chemotherapy is used to prevent the growth of cancer cells that may remain after surgery or metastases due to cancer metastasis. Although many advances have been made in the treatment of biliary tract cancer, the effects have not yet been proven.
본 발명의 용어, "예방"은 상기 항체; 및 피리미딘 유사체, 플라틴계 항암제 또는 이들의 조합을 포함하는 조성물의 투여로 암을 억제 또는 지연시키는 모든 행위를 의미한다. As used herein, the term "prevention" refers to the antibody; And all actions of inhibiting or delaying cancer by administration of a composition comprising a pyrimidine analog, a platinum anticancer agent, or a combination thereof.
본 발명의 용어, "치료"는 상기 항체; 및 피리미딘 유사체, 플라틴계 항암제 또는 이들의 조합을 포함하는 조성물의 투여로 암의 증세가 호전되거나 이롭게 변경되는 모든 행위를 의미한다.As used herein, the term “treatment” refers to the antibody; And any action in which the symptoms of cancer improve or benefit altered by administration of a composition comprising a pyrimidine analog, a platinum anticancer agent, or a combination thereof.
본 발명의 일 실시예에서, ICC 세포인 Choi-CK 세포에 대하여, Ab417 또는 젬시타빈을 단독 투여하는 경우에는 각각 35.3% 또는 44.4%의 암 성장 저해율을 나타내었지만, 이들을 병용 투여하는 경우에는 88.8%의 저해율을 나타냄을 확인하였다(도 5). 또한, ECC 세포인 TFK-1 세포에 대하여, Ab417 또는 젬시타빈을 단독 투여하는 경우에는 각각 10.7% 또는 21.7%의 암 성장 저해율을 나타내었지만, 이들을 병용 투여하는 경우에는 52.2%의 저해율을 나타냄을 확인하였다(도 10).In one embodiment of the present invention, the administration of Ab417 or gemcitabine alone showed 35.3% or 44.4% cancer growth inhibition, respectively, against Choi-CK cells, which are ICC cells, but 88.8% It was confirmed that the inhibition rate of (Fig. 5). In addition, when Ab417 or gemcitabine alone was administered to TFK-1 cells, which are ECC cells, cancer growth inhibition was 10.7% or 21.7%, respectively, but when they were used in combination, they showed an inhibition rate of 52.2%. (FIG. 10).
상기 약학적 조성물은 약학적 조성물의 제조에 통상적으로 사용하는 약학적으로 허용가능한 담체, 부형제 또는 희석제를 추가로 포함할 수 있고, 상기 담체는 비자연적 담체(non-naturally occuring carrier)를 포함할 수 있다. The pharmaceutical composition may further include a pharmaceutically acceptable carrier, excipient or diluent commonly used in the manufacture of the pharmaceutical composition, and the carrier may comprise a non-naturally occuring carrier. have.
상기 용어, '약학적으로 허용가능한'은 상기 조성물에 노출되는 세포나 인간에게 독성이 없는 특성을 나타내는 것을 의미한다.The term 'pharmaceutically acceptable' means to exhibit properties that are not toxic to cells or humans exposed to the composition.
구체적으로, 상기 약학적 조성물은 각각 통상의 방법에 따라 산제, 과립제, 정제, 캡슐제, 현탁액, 에멀젼, 시럽, 에어로졸 등의 경구형 제형, 외용제, 좌제 및 멸균 주사용액의 형태로 제형화하여 사용될 수 있으나, 당업계에서 암의 예방 또는 치료를 위하여 사용되는 제형이라면, 이에 제한되지 않는다.Specifically, the pharmaceutical composition may be used in the form of oral dosage forms, external preparations, suppositories, and sterile injectable solutions, such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, and aerosols, respectively, according to conventional methods. However, if the formulation is used in the art for the prevention or treatment of cancer, it is not limited thereto.
상기 약학적 조성물에 포함될 수 있는 담체, 부형제 및 희석제로는, 구체적인 예로, 락토즈, 덱스트로즈, 수크로스, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로즈, 메틸 셀룰로즈, 미정질 셀룰로스, 폴리비닐 피롤리돈, 물, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 탈크, 마그네슘 스테아레이트, 폴리카프로락톤(PCL; polycaprolactone), 폴리락틱액시드(PLA; Poly Lactic Acid), 폴리-L-락틱액시드(PLLA; poly- L -lactic acid), 광물유 등을 들 수 있다. Examples of carriers, excipients, and diluents that may be included in the pharmaceutical composition include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate , Calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate, polycaprolactone (PCL; polycaprolactone), polylac Polylactic acid (PLA), poly-L-lactic acid (PLLA), Mineral oil and the like.
제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제될 수 있다. When formulated, it may be prepared using diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrating agents, surfactants, etc. which are commonly used.
경구투여를 위한 고형 제제에는 정제, 환제, 산제, 과립제, 캡슐제 등이 포함되며, 이러한 고형 제제는 상기 추출물과 이의 분획물들에 적어도 하나 이상의 부형제 예를 들면, 전분, 칼슘 카보네이트(calcium carbonate), 수크로스(sucrose) 또는 락토오스(lactose), 젤라틴 등을 섞어 조제될 수 있다. 또한, 단순한 부형제 이외에 마그네슘 스티레이트, 탈크 같은 윤활제들도 사용될 수 있다. Solid preparations for oral administration include tablets, pills, powders, granules, capsules, and the like, which may comprise at least one excipient such as starch, calcium carbonate, It may be prepared by mixing sucrose or lactose, gelatin and the like. In addition to simple excipients, lubricants such as magnesium styrate and talc may also be used.
경구투여를 위한 액상 제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당 되는데 흔히 사용되는 단순 희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다. Liquid preparations for oral administration include suspensions, solutions, emulsions, and syrups, and may include various excipients, such as wetting agents, sweeteners, fragrances, and preservatives, in addition to commonly used simple diluents such as water and liquid paraffin. have.
비경구 투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조 제제, 좌제 등이 포함될 수 있다. 비수성용제, 현탁제로는 프로필렌글리콜(propylene glycol), 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔(witepsol), 마크로골, 트윈(tween) 61, 카카오지, 라우린지, 글리세로제라틴 등이 사용될 수 있다. Formulations for parenteral administration may include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized formulations, suppositories, and the like. As the non-aqueous solvent and suspending agent, propylene glycol, polyethylene glycol, vegetable oil such as olive oil, injectable ester such as ethyl oleate and the like can be used. As the base of the suppository, witepsol, macrogol, tween 61, cacao butter, laurin butter, glycerogelatin and the like can be used.
본 발명의 약학적 조성물에 포함된 상기 항체; 및 피리미딘 유사체, 플라틴계 항암제 또는 이들의 조합의 함량은 최종 조성물 총 중량을 기준으로 0.0001 내지 50 중량%, 보다 구체적으로 0.01 내지 20 중량%의 함량으로 포함될 수 있으나, 특별히 이에 제한되지 않는다.The antibody included in the pharmaceutical composition of the present invention; And the content of pyrimidine analogues, platinum anticancer agents or combinations thereof may be included in an amount of 0.0001 to 50% by weight, more specifically 0.01 to 20% by weight, based on the total weight of the final composition, but is not particularly limited thereto.
본 발명의 약학적 조성물은 약제학적으로 유효한 양으로 투여될 수 있다.The pharmaceutical composition of the present invention may be administered in a pharmaceutically effective amount.
상기 용어, '약제학적으로 유효한 양'은 의학적 치료 또는 예방에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료 또는 예방하기에 충분한 양을 의미하며, 유효 용량 수준은 질환의 중증도, 약물의 활성, 환자의 연령, 체중, 건강, 성별, 환자의 약물에 대한 민감도, 사용된 본 발명 조성물의 투여 시간, 투여 경로 및 배출 비율 치료기간, 사용된 본 발명의 조성물과 배합 또는 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다. 본 발명의 약학적 조성물은 단독으로 투여하거나 공지된 담도암 치료제와 병용하여 투여될 수 있다. 상기 요소를 모두 고려하여 부작용 없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하다.The term `` pharmaceutically effective amount '' means an amount sufficient to treat or prevent a disease at a reasonable benefit / risk ratio applicable to medical treatment or prevention, and an effective dose level refers to the severity of the disease, the activity of the drug, the patient Age, body weight, health, sex, sensitivity of the patient to the drug, time of administration of the composition of the invention used, route of administration and rate of release, duration of treatment, drugs comprising the drug in combination or concurrently with the composition of the invention used and It may be determined according to factors well known in other medical fields. The pharmaceutical composition of the present invention may be administered alone or in combination with known biliary tract cancer therapeutics. In consideration of all the above factors, it is important to administer an amount that can obtain the maximum effect in a minimum amount without side effects.
본 발명의 약학적 조성물의 투여량은 사용목적, 질환의 중독도, 환자의 연령, 체중, 성별, 기왕력, 또는 유효성분으로서 사용되는 물질의 종류 등을 고려하여 당업자가 결정할 수 있다. 예를 들어, 본 발명의 약학적 조성물은 성인 1인당 약 0.1 ng/㎏ 내지 약 100 ㎎/㎏, 구체적으로 약 1 ng/㎏ 내지 약 10 ㎎/㎏로 투여할 수 있고, 본 발명의 조성물의 투여빈도는 특별히 이에 제한되지 않으나, 1일 1회 투여하거나 또는 용량을 분할하여 수회 투여할 수 있다. 상기 투여량은 어떠한 면으로든 본 발명의 범위를 한정하는 것은 아니다.The dosage of the pharmaceutical composition of the present invention can be determined by those skilled in the art in consideration of the purpose of use, the degree of addiction of the disease, the age, weight, sex, history, or type of substance used as an active ingredient. For example, the pharmaceutical composition of the present invention may be administered at about 0.1 ng / kg to about 100 mg / kg, specifically about 1 ng / kg to about 10 mg / kg, per adult Frequency of administration is not particularly limited, but may be administered once a day or several times in divided doses. The dosage does not limit the scope of the invention in any aspect.
또 다른 하나의 양태는 상기 항체; 및 피리미딘 유사체, 플라틴계 항암제 또는 이들의 조합을 약제학적으로 유효한 양으로 개체에 투여하는 단계를 포함하는, 암의 예방 또는 치료 방법을 제공한다.Another embodiment is the antibody; And administering to the individual a pharmaceutically effective amount of a pyrimidine analog, a platinum based anticancer agent, or a combination thereof.
본 발명의 용어, "개체"는 암이 발병될 가능성이 있거나 또는 발병된 쥐, 가축, 인간 등을 포함하는 포유동물, 양식어류 등을 제한없이 포함할 수 있다.As used herein, the term "individual" may include, without limitation, mammals, farmed fish, and the like, including rats, livestock, humans, and the like, which may or may not develop cancer.
본 발명의 암의 예방 또는 치료 방법에서, 상기 조성물을 투여하는 투여 경로 및 투여 방식은 특별히 제한되지 않으며, 목적하는 해당 부위에 상기 조성물이 도달할 수 있는 한, 임의의 투여 경로 및 투여 방식에 따를 수 있다.In the method for preventing or treating cancer of the present invention, the route of administration and mode of administration for administering the composition is not particularly limited and may be in accordance with any route of administration and mode of administration so long as the composition can reach the desired site of interest. Can be.
구체적으로, 상기 조성물은 경구 또는 비경구의 다양한 경로를 통하여 투여될 수 있으며, 그 투여 경로의 비제한적인 예로는, 안구, 구강, 직장, 국소, 정맥내, 복강내, 근육내, 동맥내, 경피, 비측내 또는 흡입 등을 통하여 투여되는 것을 들 수 있다. Specifically, the composition may be administered through various routes of oral or parenteral, non-limiting examples of the route of administration, eye, mouth, rectal, topical, intravenous, intraperitoneal, intramuscular, intraarterial, transdermal And intranasal administration or inhalation.
또한, 상기 조성물은 활성 물질이 표적 세포로 이동할 수 있는 임의의 장치에 의해 투여될 수 있다.In addition, the composition may be administered by any device in which the active substance may migrate to the target cell.
또한, 상기 항체; 및 피리미딘 유사체, 플라틴계 항암제 또는 이들의 조합은 병용 투여될 수 있다.In addition, the antibody; And pyrimidine analogs, platinum anticancer agents or combinations thereof may be administered in combination.
본원에서, "병용"이란 용어가 사용될 경우 이는 동시, 개별 또는 순차 투여를 나타내는 것으로 이해되어야 한다. 상기 투여가 순차 또는 개별적인 경우, 2차 성분 투여의 간격은 상기 병용의 이로운 효과를 잃지 않도록 하는 것이어야 한다. As used herein, the term "combination" should be understood to denote simultaneous, separate or sequential administration. If the administration is sequential or separate, the interval of administration of the secondary components should be such that the beneficial effect of the combination is not lost.
상기 물질은 각각 동시, 순차적, 또는 역순으로 투여될 수 있으며, 적절한 유효량의 조합으로 동시에 투여될 수 있으나, 특정 투여 방법 또는 순서에 제한되지 않는다. Each of the agents may be administered simultaneously, sequentially, or in reverse order, and may be administered simultaneously in any combination of appropriate effective amounts, but is not limited to any particular method or order of administration.
본 발명의 방법은 상기 유효 성분들을 약학적 유효량으로 투여하는 것을 포함할 수 있다. 적합한 총 1일 사용량은 올바른 의학적 판단범위 내에서 처치의에 의해 결정될 수 있으며, 1회 또는 수회로 나누어 투여할 수 있다. 그러나 본 발명의 목적상, 특정 환자에 대한 구체적인 치료적 유효량은 달성하고자 하는 반응의 종류와 정도, 경우에 따라 다른 제제가 사용되는지의 여부를 비롯한 구체적 조성물, 환자의 연령, 체중, 일반 건강 상태, 성별 및 식이, 투여 시간, 투여 경로 및 조성물의 분비율, 치료기간, 구체적 조성물과 함께 사용되거나 동시 사용되는 약물을 비롯한 다양한 인자와 의약 분야에 잘 알려진 유사 인자에 따라 다르게 적용하는 것이 바람직하다.The method of the present invention may comprise administering the active ingredient in a pharmaceutically effective amount. Suitable total daily doses may be determined by the treating physician within the scope of good medical judgment and may be administered once or in divided doses. However, for the purposes of the present invention, the specific therapeutically effective amount for a particular patient is determined by the specific composition, including the type and extent of the reaction to be achieved, whether or not other agents are used in some cases, the age, weight, general health of the patient, It is desirable to apply differently depending on various factors and similar factors well known in the medical field, including sex and diet, time of administration, route of administration and rate of composition, duration of treatment, drugs used with or co-specific with the specific composition.
다른 하나의 양태는 상기 조성물을 포함하는 약학적 키트를 제공한다.Another aspect provides a pharmaceutical kit comprising the composition.
본 발명에서, 상기 약학적 키트는 정제, 캡슐 또는 주사 형태일 수 있으나, 이에 제한되지 않는다.In the present invention, the pharmaceutical kit may be in tablet, capsule or injection form, but is not limited thereto.
또한, 상기 키트는 약학적으로 유효한 투여량으로 상기 항체; 및 피리미딘 유사체(pyrimidine analog), 플라틴계 항암제 또는 이들의 조합을 포함할 수 있으나, 이에 제한되지 않는다.In addition, the kit may comprise a pharmaceutically effective dosage of the antibody; And pyrimidine analogs, platinum-based anticancer agents, or combinations thereof.
또한, 상기 키트는 상기 성분들의 투여를 위한 설명서를 포함할 수 있다.The kit can also include instructions for the administration of the components.
이하, 본 발명을 하기 실시예에서 보다 구체적으로 설명한다. 그러나 이들 실시예는 본 발명의 이해를 돕기 위한 것일 뿐, 이들에 의해 본 발명이 한정되는 것은 아니다.Hereinafter, the present invention will be described in more detail in the following Examples. However, these examples are only for aiding the understanding of the present invention, and the present invention is not limited thereto.
실험예 1. 사용한 세포주 및 배양 방법Experimental Example 1. Cell line and culture method used
인간 ICC(intrahepatic cholangiocarcinoma; 간내 담도암) 세포주인 Choi-CK 및 SCK-L1는 10% FBS(Atlas Biologics, USA)가 보충된 DMEM(Welgene, Republic of Korea)에서 배양하였다. 상기 Choi-CK는 전북대학교 김대곤 박사님으로부터 제공받았다. ECC(extrahepatic cholangiocarcinoma; 간외 담도암) 세포주인 TFK-1는 DSMZ (German Collection of Microorganisms and Cell Cultures, Human and Animal Cell Lines, Braunschweig, Germany)에서 구매하였고, RPMI(Thermo Fisher Scientific, USA)에서 배양하였다. Ab417 항체를 발현하는 CHO-DG44 세포주 #9-20(Cho S, et al., MAbs. 2016;8(2):414-25.)는 5%(v/v) 투석된 FBS(Thermo Fisher Scientific)가 보충된 MEM-α(Welgene)에서 배양하였고, 항체의 생산을 위하여 혈청이 포함되지 않은 배지(SFM4CHO, GE Lifesciences, USA)로 교체하였다. 모든 세포주는 5% CO2, 37℃의 습식 인큐베이터에서 배양하였다.The human intrahepatic cholangiocarcinoma (ICC) cell lines Choi-CK and SCK-L1 were cultured in DMEM (Welgene, Republic of Korea) supplemented with 10% FBS (Atlas Biologics, USA). Choi-CK was provided by Dr. Dae-Gon Kim of Chonbuk National University. TFK-1, an extrahepatic cholangiocarcinoma (ECC) cell line, was purchased from German Collection of Microorganisms and Cell Cultures, Human and Animal Cell Lines, Braunschweig, Germany, and cultured in RPMI (Thermo Fisher Scientific, USA). . CHO-DG44 cell line # 9-20 expressing Ab417 antibody (Cho S, et al., MAbs. 2016; 8 (2): 414-25.) Was 5% (v / v) dialyzed FBS (Thermo Fisher Scientific) ) Was supplemented with MEM-α (Welgene) supplemented and replaced with medium without serum (SFM4CHO, GE Lifesciences, USA) for the production of antibodies. All cell lines were cultured in a wet incubator at 37 ° C. with 5% CO 2 .
실험예 2. Ab417 항체의 생산 방법Experimental Example 2. Production method of Ab417 antibody
Ab417 항체(이하, 'Ab417'로 명명)는 CHO-DG44 세포주(#9-20)에서 생산하였다. 배양 상층액을 원심분리하고, 바틀탑 필터(0.22 ㎛ PES, Sartorius)를 이용하여 여과하였으며, 정제를 위하여 단백질 A-아가로스(GenScript, USA)에서 친화 크로마토그래피를 수행하였다. 단백질 A에 결합한 항체들을 150 mM 염화나트륨(pH 3.6)이 포함된 0.1 M 구연산 나트륨을 이용하여 용출하였다. 용출된 항체를 150 mM 염화나트륨 및 0.007% Tween-20(pH 6.4)가 포함된 25 mM의 구연산 나트륨 버퍼(pH 6.4)로 교체한 뒤 보관하였다. 단백질 농도는 NanoDrop 2000 UV-Vis 분광광도계(Thermo Fisher Scientific)로 측정하였다.Ab417 antibody (hereinafter referred to as 'Ab417') was produced in CHO-DG44 cell line (# 9-20). The culture supernatant was centrifuged and filtered using a bottle top filter (0.22 μm PES, Sartorius) and affinity chromatography was performed on Protein A-Agarose (GenScript, USA) for purification. Antibodies bound to protein A were eluted with 0.1 M sodium citrate containing 150 mM sodium chloride, pH 3.6. The eluted antibody was replaced with 25 mM sodium citrate buffer (pH 6.4) containing 150 mM sodium chloride and 0.007% Tween-20 (pH 6.4) and stored. Protein concentration was measured with a NanoDrop 2000 UV-Vis spectrophotometer (Thermo Fisher Scientific).
실험예 3. 인비보(Experimental Example 3. In vivo ( In vivoIn vivo ) 항암 효과 분석Anticancer Effect Analysis
누드 마우스(BALB/c Slc-nu, 5 주령)는 일본 SLC, Inc.에서 구입하였고, Biotoxtech(한국)의 동물 관리위원회의 지침에 따라 7일 동안 특정 병원균이 없는 환경에서 보관하였다. Nude mice (BALB / c Slc- nu , 5 weeks old) were purchased from SLC, Inc., Japan and stored in a specific pathogen-free environment for 7 days according to the guidelines of the Animal Control Committee of Biotoxtech (Korea).
Ab417, 약물(젬시타빈; gemcitabine 또는 시스플라틴; cisplatin), 또는 Ab417 및 상기 약물의 조합에 대한 항암 효과를 확인하기 위하여, 위원회의 사전 승인을 받았다(각각 B13698, B13933, 또는 B13934). In order to confirm the anticancer effect on Ab417, the drug (gemcitabine; gemcitabine or cisplatin), or Ab417, and a combination of the above drugs, prior approval of the committee was obtained (B13698, B13933, or B13934, respectively).
구체적으로, Choi-CK 세포(1 × 106)를 각 마우스의 오른쪽 측복부에 피하 접종하였다. Choi-CK 종양 조직(3 × 3 × 3 mm3)을 새로운 누드 마우스의 등부에 피하 접종하였다. 종양 체적이 약 100 mm3에 이르는 경우, 마우스를 임의의 그룹으로 나누었고 Ab417 또는 약물(젬시타빈 또는 시스플라틴)을 일주일에 두 번(그룹 당 n = 8) 정맥 내(i.v.) 또는 복강 내(i.p.) 주사하였다. 젬시타빈(Gemcitabine hydrochloride, Sigma Aldrich)과 시스플라틴(cis-Diammineplatinum (II) dichloride, Sigma Aldrich)은 염화나트륨(sodium chloride; Choongwae, Republic of Korea)을 이용하여 적절한 농도로 용해하였다. 사용되지 않은 추가의 동물들은 CO2 가스 및 치명적인 출혈을 이용하여 안락사시켰다.Specifically, Choi-CK cells (1 × 10 6 ) were inoculated subcutaneously in the right flank of each mouse. Choi-CK tumor tissue (3 × 3 × 3 mm 3 ) was inoculated subcutaneously in the back of new nude mice. When the tumor volume reached about 100 mm 3 , mice were divided into arbitrary groups and Ab417 or drug (gemcitabine or cisplatin) was administered twice a week (n = 8 per group) intravenously (iv) or intraperitoneally (ip) Injection. Gemcitabine (Gemcitabine hydrochloride, Sigma Aldrich) and cisplatin (cis-Diammineplatinum (II) dichloride, Sigma Aldrich) were dissolved in an appropriate concentration using sodium chloride (Choongwae, Republic of Korea). Additional animals not used were euthanized using CO 2 gas and fatal bleeding.
종양의 성장은 캘리퍼로 길이와 넓이를 측정하였고, 다음의 공식을 이용하여 종양 부피를 계산하였다: TV (mm3) = L (mm) × W2 (mm2) × 1/2. 이때, 상기 L은 길이, W는 넓이를 의미한다. 또한, 동물의 체중도 측정하였다. 아이소플루레인(isoflurane)을 이용하여 동물을 마취시켰고, 종양 조직을 꺼낸 뒤 무게를 측정하였다. IR(Tumor growth inhibition rate; 종양 성장 저해율)은 다음과 같은 공식을 이용하여 계산하였다: IR (%) = (1 - T/C) × 100. 이때, 상기 T는 테스트 물질 그룹에 대한 종양의 평균 무게, C는 음성 대조 그룹에 대한 부피 또는 무게를 의미한다.Tumor growth was measured in length and width with a caliper and the tumor volume was calculated using the following formula: TV (mm 3 ) = L (mm) x W 2 (mm 2 ) x 1/2. In this case, L means length and W means width. Animal weights were also measured. Animals were anesthetized using isoflurane and the tumor tissue was taken out and weighed. Tumor growth inhibition rate (IR) was calculated using the following formula: IR (%) = (1-T / C) x 100, where T is the mean of the tumor for the test substance group Weight, C means volume or weight for the negative control group.
항암 활성의 메커니즘을 규명하기 위하여, 누드 마우스(BALB/c-nude, 6 주령)을 NARA Biotech (Republic of Korea)로부터 구입하였고, 강원대학교 IACUC (KW-160429-1)의 지침에 따라 7일 동안 특정 병원균이 없는 환경에서 보관하였다. In order to elucidate the mechanism of anticancer activity, nude mice (BALB / c-nude, 6 weeks old) were purchased from NARA Biotech (Republic of Korea) and followed for 7 days according to the instructions of Kangwon National University IACUC (KW-160429-1). Stored in an environment free of specific pathogens.
Choi-CK 세포(1 × 106)를 누드 마우스(n=3)의 오른쪽 측복부에 피하 접종하였다. 종양 부피가 약 110 mm3에 도달할 때, 마우스를 임의의 두 그룹으로 나누었고, 항체 또는 PBS를 일주일에 세 번 복강 내 주사하였다. 주사 10일 후, 종양을 꺼내어 Ki-67의 탐지를 위해 면역조직화학 및 L1CAM의 탐지를 위해 면역형광염색을 수행하였다. 실험 후, 동물들은 CO2 가스 및 치명적인 출혈을 이용하여 안락사시켰다.Choi-CK cells (1 × 10 6 ) were inoculated subcutaneously in the right flank of nude mice ( n = 3). When the tumor volume reached about 110 mm 3 , mice were divided into any two groups and injected intraperitoneally with antibody or PBS three times a week. Ten days after injection, tumors were removed and immunofluorescence stained for immunohistochemistry for detection of Ki-67 and detection of L1CAM. After the experiment, the animals were euthanized using CO 2 gas and fatal bleeding.
한편, TFK-1의 이종모델을 위하여, NOD/SCID 마우스(5 주령)을 동물자원센터(Animal Resources Center; ARC, Australia)에서 구입하였고, 아산의료원(한국)의 동물윤리위원회의 지침에 따라 특정 병원균이 없는 환경에서 보관하였다. TFK-1 세포(5 × 106)를 각 마우스의 우측 뒷다리에 이식하였다. 마우스를 임의의 그룹(그룹 당 n=8)으로 나누었고, Ab417 또는 약물을 일주일에 두 번 주입하였다.On the other hand, for the heterogeneous model of TFK-1, NOD / SCID mice (5 weeks old) were purchased from Animal Resources Center (ARC, Australia) and specified according to the guidelines of the Animal Ethics Committee of Asan Medical Center (Korea). Stored in a pathogen free environment. TFK-1 cells (5 × 10 6 ) were implanted into the right hind limb of each mouse. Mice were divided into arbitrary groups ( n = 8 per group) and injected Ab417 or drug twice a week.
실험예 4. 면역조직화학 분석Experimental Example 4 Immunohistochemical Analysis
마우스를 마취시키고 조직을 수득한 뒤, 파라핀 섹션(paraffin sections)을 위해 수득한 조직을 포르말린(formalin)으로 고정하였다. 파라핀-임베딩 조직 섹션을 자일렌(xylene), 및 95%, 90%, 70% 및 50% 에탄올로 탈-파라핀화시켰고, PBS로 세척하였다. 탈-파라핀화된 슬라이드를 30분 동안 0.1 mol/L 구연산 버퍼(pH 6.0)에서 가열하였고, 메탄올에 포함된 0.3%(v/v) 과산화수소에서 15분 동안 인큐베이션하였다. 섹션을 상온에서 일반 말 혈청에 두어 블로킹하였고, Ki-67(C-term) (1:200, Acris, Germany)에 대한 일차 항체로 4℃에서 하룻밤 동안 면역염색을 수행하였다. ABC 및 DAB 키트(Vector Laboratories, USA)를 이용하여 타겟 단백질을 가시화하였고, 헤마톡실린(hematoxylin)으로 교차염색하였다.After the mice were anesthetized and tissues were obtained, the tissues obtained for paraffin sections were fixed with formalin. Paraffin-embedding tissue sections were de-paraffinized with xylene and 95%, 90%, 70% and 50% ethanol and washed with PBS. The de-paraffinized slides were heated in 0.1 mol / L citric acid buffer (pH 6.0) for 30 minutes and incubated for 15 minutes in 0.3% (v / v) hydrogen peroxide contained in methanol. Sections were blocked in normal horse serum at room temperature and blocked, and immunostaining was performed overnight at 4 ° C. with primary antibodies against Ki-67 (C-term) (1: 200, Acris, Germany). Target proteins were visualized using ABC and DAB kits (Vector Laboratories, USA) and cross-stained with hematoxylin.
면역형광 염색을 위해, 항-NCAM-L1(N-14) mAb(1:50, Santa Cruz Biotechnology, USA)로 4℃에서 하룻밤 동안 면역염색을 수행하였고, 동키(donkey) 항-고트(goat) Alexa 488-접합 이차항체(1:200, Thermo Fisher Scientific)로 상온에서 1시간 동안 인큐베이션하였다. 세포핵은 DAPI(5 μM, Sigma Aldrich)로 표지하였다. Zeiss 공초점 현미경을 이용하여 이미지를 촬영하였으며, Image J 소프트웨어를(version 1.45)를 사용하여 염색 정도를 정량적으로 평가하였다.For immunofluorescence staining, immunostaining was performed overnight at 4 ° C. with anti-NCAM-L1 (N-14) mAb (1:50, Santa Cruz Biotechnology, USA), and donkey anti-goat Incubated with Alexa 488-conjugated secondary antibody (1: 200, Thermo Fisher Scientific) for 1 hour at room temperature. Cell nuclei were labeled with DAPI (5 μΜ, Sigma Aldrich). Images were taken using a Zeiss confocal microscope and the degree of staining was quantitatively assessed using Image J software (version 1.45).
실험예 5. 항체 내면화(Antibody internalization) 어세이Experimental Example 5. Antibody internalization assay
Ab417의 내면화는 공초점 레이저 스캐닝 현미경을 이용하여 측정하였다. Ab417(10 ㎍/mL)를 포함하는 배지를 슬라이드 글라스에 올려진 L1CAM-발현 SCK-L1 세포에 첨가하였다. 37℃에서 0.5 또는 4시간 동안 배양한 이후, 세포를 얼음에 두었고, 4℃ PBS로 헹군 뒤, 4% 포름알데히드(Sigma Aldrich)로 고정하였다. Internalization of Ab417 was measured using a confocal laser scanning microscope. Medium containing Ab417 (10 μg / mL) was added to L1CAM-expressing SCK-L1 cells loaded on slide glass. After incubation at 37 ° C. for 0.5 or 4 hours, the cells were placed on ice, rinsed with 4 ° C. PBS and fixed with 4% formaldehyde (Sigma Aldrich).
제로 타임(zero-time) 대조군으로서, Ab417를 포름알데히드로 고정된 세포에 첨가하였다. 세포 표면 항체를 래빗(rabbit) 항-인간 IgG(H+L) Cross Adsorbed DyLight 594 다클론성 항체(1:100, v/v, Thermo Fisher Scientific)로 탐지하였다. 세포 표면 이차 항체를 포름알데히드를 사용하여 5분 동안 고정하였다. 세척한 이후, 세포를 블로킹하였고, PBS에 포함된 10% 고트 혈청 및 0.1% Triton X-100로 상온에서 30분 동안 투과시켰다. 내면화된 L1CAM을 표지하기 위하여, 세포를 5% 고트 혈청 및 0.02% Triton X-100가 포함된 PBS에서 항-인간 IgG(Fc-specific)-FITC (1:500, v/v)로 상온에서 30분 동안 인큐베이션하였다. 슬라이드를 PBS로 3번 세척하였고, SlowFade® Antifade 키트(Thermo Fisher Scientific)를 이용하여 마운트하였다. 형광 신호를 Olympus LX70 FV300 05-LGP-193(OLYMPUS, Japan)로 탐지하였다.As a zero-time control, Ab417 was added to formaldehyde fixed cells. Cell surface antibodies were detected with a rabbit anti-human IgG (H + L) Cross Adsorbed DyLight 594 polyclonal antibody (1: 100, v / v, Thermo Fisher Scientific). Cell surface secondary antibodies were fixed for 5 minutes using formaldehyde. After washing, cells were blocked and permeabilized for 30 minutes at room temperature with 10% goth serum and 0.1% Triton X-100 contained in PBS. To label internalized L1CAM, cells were treated with anti-human IgG (Fc-specific) -FITC (1: 500, v / v) in PBS with 5% goth serum and 0.02% Triton X-100 at room temperature. Incubate for minutes. Slides were washed three times with PBS and mounted using SlowFade® Antifade kit (Thermo Fisher Scientific). Fluorescence signals were detected with Olympus LX70 FV300 05-LGP-193 (OLYMPUS, Japan).
실험예 6. Ab417-처리 세포에서의 세포막 L1CAM 수준의 분석Experimental Example 6. Analysis of cell membrane L1CAM levels in Ab417-treated cells
Choi-CK 또는 TFK-1 세포는 Ab417(10 ㎍/mL)로 지정된 시간 동안 인큐베이션하였고, 막단백질 분획을 MEM-PERTM Plus Membrane Protein Extraction 키트(Thermo Fisher Scientific)로 수득하였다. 수득한 단백질 샘플을 BCA 단백질 정량 어세이를 통해 정량하였고, SDS-PAGE를 수행하였다. 단백질을 니트로셀룰로스 막(nitrocellulose membrane)에 옮긴 후, 0.1% Tween-20 및 5% 스킴 밀크(skim milk, BD Biosciences)가 포함된 TBS(Tris-buffered saline)로 블로킹하였다. L1CAM은 인간 L1CAM에 결합한 마우스 mAb 및 고트 항-마우스 IgG-HRP 접합체(1:8,000, v/v, Thermo Fisher Scientific)를 이용하여 탐지하였다. 판-카드헤린(Pan-cadherin)을 판-카드헤린 래빗 mAb(1: 2,500, v/v, Cell Signaling Technology, USA) 및 항-래빗 IgG-HRP 접합체(1:5,000, v/v, Cell Signaling Technology)의 로딩 컨트롤(loading control)로 사용하였다. 이후, 면역반응성 밴드를 SuperSignalTM West Femto Maximum Sensitive 기질(Thermo Fisher Scientific)을 이용하여 가시화하였다.Choi-CK or TFK-1 cells were incubated for a time designated as Ab417 (10 μg / mL) and membrane protein fractions were obtained with the MEM-PER Plus Membrane Protein Extraction Kit (Thermo Fisher Scientific). The obtained protein sample was quantified via a BCA protein quantitative assay and SDS-PAGE was performed. Proteins were transferred to nitrocellulose membranes and then blocked with Tris-buffered saline (TBS) containing 0.1% Tween-20 and 5% skim milk (BD Biosciences). L1CAM was detected using mouse mAb and goth anti-mouse IgG-HRP conjugates (1: 8,000, v / v, Thermo Fisher Scientific) bound to human L1CAM. Pan-cadherin to Pan-cadherin Rabbit mAb (1: 2,500, v / v, Cell Signaling Technology, USA) and anti-rabbit IgG-HRP conjugate (1: 5,000, v / v, Cell Signaling Technology's loading control. The immunoreactive bands were then visualized using SuperSignal West Femto Maximum Sensitive Substrate (Thermo Fisher Scientific).
실험예 7. 통계적 분석Experimental Example 7. Statistical Analysis
결과값은 '평균 ± 표준 편차'로 나타내었고 그룹 간의 통계적 비교는 Results are expressed as mean ± standard deviation, and the statistical comparison between groups
Dunnett t-검정(Dunnett's t-test) 또는 Steel 검정(Steel's test)에 이어 단방향 분석을 이용하여 수행하였다. p <0.05의 값은 통계적으로 유의한 것으로 판단하였다.Dunnett t-test (Dunnett's t-test) or Steel test (Steel's test) followed by one-way analysis. The value of p <0.05 was judged to be statistically significant.
실시예 1. ICC 이종이식 마우스 모델에서 항암 활성 확인Example 1 Confirmation of Anticancer Activity in ICC Xenograft Mouse Model
실시예 1-1. Ab417의 항암 활성 확인Example 1-1. Anticancer activity of Ab417
본 발명자들의 선행연구에서는, ICC 세포인 Choi-CK 이종이식을 갖는 누드 마우스(n=8)에 Ab417 항체(10 mg/kg)를 일주일에 세 번 정맥 내 주입하였을 때의 Ab417의 항암 활성을 측정한 결과, 도 1의 A 및 B에서 볼 수 있듯이, 동종형 대조군으로서 재조합 인간 Fc(hFc)와 비교 시, 종양 무게의 평균에 기초하여 68.6%의 암 성장 저해 활성을 나타냄을 확인하였다. 또한, 상기 Ab417는 몸무게에 영향을 미치지 않았으며, 다른 부작용을 야기하지 않음을 확인하였다(Cho S, et al. MAbs. 2016;8(2):414-25.).In our previous study, we measured the anticancer activity of Ab417 when intravenous Ab417 antibody (10 mg / kg) was injected intravenously into nude mice ( n = 8) with Choi-CK xenografts as ICC cells. As a result, as shown in A and B of Figure 1, compared with recombinant human Fc (hFc) as a homologous control, it was confirmed that the cancer growth inhibitory activity of 68.6% based on the average of the tumor weight. In addition, Ab417 did not affect the weight, it was confirmed that does not cause other side effects (Cho S, et al. MAbs. 2016; 8 (2): 414-25.).
이에, 본 발명에서는, Ab417가 용량 의존적인 항암 효과를 나타내는지 확인하기 위하여, 상기 실험예 3에 따라, Ab417(10 mg/kg) 또는 hFc(3.3 mg/kg)를 동일한 모델(n=8)에 일주일에 두 번 정맥 주사하였다.Therefore, in the present invention, in order to confirm whether Ab417 has a dose-dependent anticancer effect, according to Experimental Example 3, Ab417 (10 mg / kg) or hFc (3.3 mg / kg) in the same model ( n = 8) Intravenously twice a week.
그 결과, 도 2의 A 내지 C에서 볼 수 있듯이, Ab417는 몸무게에 영향을 미치지 않으면서 암 성장을 억제함을 확인하였다. 주입 22일 이후에, Ab417가 처리된 그룹의 평균 종양 부피 및 무게는 각각 278.3 mm3(p < 0.05) 및 0.17 g(p < 0.05), 대조군은 574.5 mm3 및 0.26 g로서, 대조군(hFc)과 비교 시 Ab417 처리된 그룹의 암 성장 억제율은 35.6%임을 확인하였다.As a result, as can be seen from A to C of Figure 2, it was confirmed that Ab417 inhibits cancer growth without affecting the weight. After 22 days of infusion, the average tumor volume and weight of the Ab417-treated group were 278.3 mm 3 ( p <0.05) and 0.17 g ( p <0.05), respectively, and the control group was 574.5 mm 3 and 0.26 g, the control group (hFc). Compared with the Ab417 treated group, the cancer growth inhibition rate was 35.6%.
상기 결과를 통해, Ab417은 Choi-CK 이종이식 모델에서 용량 의존적으로 종양 성장을 억제함을 알 수 있었다.The results show that Ab417 inhibits tumor growth in a dose-dependent manner in the Choi-CK xenograft model.
아울러, Ab417에 의한 종양 성장 억제는 암세포의 증식이 억제된 것에 기인한 결과인지 확인하고자, 상기 실험예 3 및 4에 따라, 세포 증식의 마커인 Ki-67로 면역 염색을 수행하였다.In addition, to confirm whether tumor growth inhibition by Ab417 is a result of inhibition of cancer cell proliferation, according to Experimental Examples 3 and 4, immunostaining was performed with Ki-67, a marker of cell proliferation.
그 결과, 도 2의 D에서 볼 수 있듯이, Ab417 처리된 종양은 대조군에 비하여 더 낮은 Ki-67 지수를 나타내었다. As a result, as shown in D of FIG. 2, Ab417 treated tumors showed a lower Ki-67 index than the control.
상기 결과를 통해, Ab417는 생체 내(in vivo) 암세포의 증식을 억제함으로써 종양의 성장을 저해함을 알 수 있었다.The results indicate that Ab417 inhibits tumor growth by inhibiting proliferation of cancer cells in vivo .
실시예 1-2. Ab417의 막 L1CAM에 대한 영향 분석Example 1-2. Analysis of the effect of Ab417 on membrane L1CAM
L1CAM 발현의 하향 조절은 인비트로 ICC 세포의 증식 및 이동을 저해하고, 생체 내 종양의 성장을 억제한다고 알려져 있다(Min JK, et al., Clin Cancer Res. 2010;16(14):3571-80.). 이에, Ab417에 의한 종양 세포 증식 저해 활성의 메커니즘을 확인하기 위하여, 상기 실험예 5에 따라, SCK-L1 세포를 이용하여 항체 내면화 어세이를 수행하였다. Down regulation of L1CAM expression is known to inhibit proliferation and migration of ICC cells in vitro and to inhibit tumor growth in vivo (Min JK, et al., Clin Cancer Res. 2010; 16 (14): 3571-80 .). Thus, in order to confirm the mechanism of tumor cell proliferation inhibitory activity by Ab417, according to Experimental Example 5, an antibody internalization assay was performed using SCK-L1 cells.
그 결과, 도 3의 A에서 볼 수 있듯이, Ab417의 세포질로의 내면화는 항체가 세포에 첨가된 4시간 이후에 탐지됨을 확인하였다.As a result, as shown in FIG. 3A, it was confirmed that the internalization of Ab417 into the cytoplasm was detected 4 hours after the antibody was added to the cells.
이후, 막(membrane) L1CAM 수준이 Ab417의 내면화에 의하여 감소되는지 확인하고자, 상기 실험예 6에 따라, Choi-CK 세포를 각 지정된 시간(1, 2, 4 또는 6시간) 동안 항체로 인큐베이션하였고, 처리된 세포의 막단백질 분획을 인간 L1CAM의 Ig1 도메인에 결합하는 뮤라인(murine) 항-L1CAM mAb(A10-A3)를 이용한 웨스턴블롯으로 분석하였다.Then, to confirm that membrane L1CAM levels were reduced by internalization of Ab417, according to Experiment 6, Choi-CK cells were incubated with antibodies for each designated time (1, 2, 4 or 6 hours), Membrane protein fractions of treated cells were analyzed by Western blot using murine anti-L1CAM mAb (A10-A3) that binds to the Ig1 domain of human L1CAM.
그 결과, 도 3의 B에서 볼 수 있듯이, Ab417는 농도 의존적인 방법으로 막 L1CAM 수준을 하향조절함을 확인하였다. 또한, 도 3의 C 및 D에서 볼 수 있듯이, 인간 L1CAM의 N-말단 부위에 특이적인 mAb로 수행된 Choi-CK 종양의 면역형광 염색을 통해, Ab417 처리된 종양은 대조군인 hFc가 처리된 종양에 비하여 L1CAM 수준이 상당히 감소됨을 확인하였다.As a result, as shown in B of Figure 3, Ab417 was confirmed to down-regulate the membrane L1CAM level in a concentration-dependent manner. In addition, as shown in FIGS. 3C and 3D, through immunofluorescence staining of Choi-CK tumors performed with mAbs specific for the N-terminal region of human L1CAM, Ab417-treated tumors were treated with hFc-treated tumors. It was confirmed that the L1CAM level is significantly reduced compared to.
상기 결과를 통해, Ab417는 내면화되어 막 L1CAM 수준을 감소시킴으로써 암세포의 증식을 억제함을 알 수 있었다.The results showed that Ab417 internalized to inhibit the proliferation of cancer cells by reducing membrane L1CAM levels.
실시예 1-3. 젬시타빈 또는 시스플라틴의 암세포 증식 억제 효과 확인Example 1-3. Inhibition of cancer cell proliferation by gemcitabine or cisplatin
젬시타빈 또는 시스플라틴은 진행된 담도암의 표준 치료제로 고려되고 있기 때문에, Choi-CK 세포의 생체 내 증식에 대한 상기 약물의 영향을 확인하였다.Since gemcitabine or cisplatin is considered to be the standard treatment for advanced biliary tract cancer, the effect of this drug on the in vivo proliferation of Choi-CK cells was confirmed.
그 결과, 젬시타빈 또는 시스플라틴은 Choi-CK 세포의 증식을 용량 의존적인 방법으로 저해하였고, 젬시타빈 또는 시스플라틴의 IC50 값은 각각 1.5 ㎍/mL 또는 3.0 ㎍/mL임을 확인하였다.As a result, gemcitabine or cisplatin inhibited the proliferation of Choi-CK cells in a dose-dependent manner, and the IC 50 value of gemcitabine or cisplatin was 1.5 μg / mL or 3.0 μg / mL, respectively.
아울러, 상기 약물에 의한 종양 세포 증식 저해 활성의 메커니즘을 확인하기 위하여, 유세포 분석을 수행하였다.In addition, flow cytometry was performed to confirm the mechanism of tumor cell proliferation inhibitory activity by the drug.
그 결과, IC50 값의 젬시타빈 또는 시스플라틴을 처리하는 경우, 세포 주기는 G2/M 상태에서 정지됨을 확인하였다. 또한, 젬시타빈보다 시스플라틴을 처리하는 경우에 세포 주기가 정지된 세포가 더욱 증가함을 확인하였다.As a result, it was confirmed that when treated with gemcitabine or cisplatin with an IC 50 value, the cell cycle was stopped in the G2 / M state. In addition, when treated with cisplatin than gemcitabine, it was confirmed that the cell cycle was further increased.
상기 결과를 통해, 젬시타빈 또는 시스플라틴은 암세포의 주기를 정지시킴으로써 세포의 증식을 저해함을 알 수 있었다.Through the above results, it was found that gemcitabine or cisplatin inhibits the proliferation of cells by stopping the cycle of cancer cells.
실시예 1-4. 젬시타빈 또는 시스플라틴의 항암 효과 Example 1-4. Anticancer Effects of Gemcitabine or Cisplatin
Ab417, 및 젬시타빈 또는 시스플라틴의 병용 투여를 위하여, 본 발명자들은 Choi-CK 종양 이종이식 모델에서 젬시타빈 또는 시스플라틴의 항암 효과를 확인하였다. 구체적으로, 상기 실험예 3에 따라, 젬시타빈(10, 25, 또는 50 mg/kg), 시스플라틴(0.5 또는 1.5 mg/kg) 또는 대조군으로서 살린(saline)을 Choi-CK 이종이식을 갖는 누드마우스(n=8)에 일주일에 두 번씩 복강 내 주사하였다. 주사 22일 후, 암 조직을 제거하여 무게를 측정하였다.For the combined administration of Ab417, and gemcitabine or cisplatin, we identified the anticancer effect of gemcitabine or cisplatin in the Choi-CK tumor xenograft model. Specifically, according to Experimental Example 3, nude mice having Choi-CK xenografts with gemcitabine (10, 25, or 50 mg / kg), cisplatin (0.5 or 1.5 mg / kg) or saline as a control Intraperitoneal injection twice a week at ( n = 8). Twenty two days after the injection, the cancerous tissue was removed and weighed.
그 결과, 도 4의 A 및 B에서 볼 수 있듯이, 10 또는 25 mg/kg의 젬시타빈은 각각 55.4% 또는 80.7%의 암 성장 저해율을 나타내었고, 용량 의존적인 방법으로 암 성장을 저해함을 확인하였다. 반면, 0.5 또는 1.5 mg/kg의 시스플라틴은 각각 39.8% 또는 36.7%의 암 성장 저해율을 나타내었고, 농도가 달라져도 동일한 항암 효과를 나타냄을 확인하였다.As a result, as shown in A and B of Figure 4, 10 or 25 mg / kg gemcitabine showed a growth inhibition rate of 55.4% or 80.7%, respectively, it was confirmed that inhibits cancer growth in a dose-dependent method It was. On the other hand, 0.5 or 1.5 mg / kg of cisplatin showed a cancer growth inhibition rate of 39.8% or 36.7%, respectively, it was confirmed that the same anti-cancer effect even at different concentrations.
실시예 1-5. Ab417, 및 젬시타빈 또는 시스플라틴 병용 투여의 항암 효과 확인Example 1-5. Confirmation of anticancer effect of Ab417 and gemcitabine or cisplatin combination
Ab417과 약물을 병용 투여하는 경우, 항체 또는 약물 단독을 투여하는 경우에 비하여 종양 성장 억제 효과가 월등한지 확인하였다.When Ab417 and drug were administered in combination, it was confirmed whether the tumor growth inhibitory effect was superior to that of antibody or drug alone.
구체적으로, Ab417, 및 젬시타빈 또는 시스플라틴의 최대 투여량을 Choi-CK 이종 이식 모델에 주입하였고, 상기 실험예 3에 따라, Ab417(10 mg/kg) 단독; 젬시타빈(10 mg/kg) 단독; 또는 시스플라틴(0.5 mg/kg) 단독을 투여하거나, Ab417(10 mg/kg) 및 젬시타빈(10 mg/kg); Ab417(10 mg/kg) 및 시스플라틴(0.5 mg/kg); 또는 Ab417(10 mg/kg) 및 대조군으로서 살린을 일주일에 두 번씩 3주 동안 Choi-CK 이종이식을 갖는 마우스에 주입하였다.Specifically, Ab417, and the maximum dose of gemcitabine or cisplatin were injected into the Choi-CK xenograft model, according to Experiment 3, Ab417 (10 mg / kg) alone; Gemcitabine (10 mg / kg) alone; Or cisplatin (0.5 mg / kg) alone, or Ab417 (10 mg / kg) and gemcitabine (10 mg / kg); Ab417 (10 mg / kg) and cisplatin (0.5 mg / kg); Or Ab417 (10 mg / kg) and saline as a control were injected into mice with Choi-CK xenografts twice a week for 3 weeks.
그 결과, 도 5의 A 및 B에서 볼 수 있듯이, Ab417 및 젬시타빈을 병용 투여하는 경우에 암 성장은 완전히 저해되었고, 종양 무게에 근거하여, 대조군에 비하여 88.8%의 암 성장 저해율을 나타냄을 확인하였다. 반면, Ab417 또는 젬시타빈은 각각 35.3% 또는 44.4%의 암 성장 저해율을 나타냄을 확인하였다.As a result, as shown in Figures A and B of Figure 5, when combined with Ab417 and gemcitabine cancer growth was completely inhibited, based on the tumor weight, it was confirmed that the cancer growth inhibition rate of 88.8% compared to the control group It was. On the other hand, Ab417 or gemcitabine was confirmed to exhibit cancer growth inhibition of 35.3% or 44.4%, respectively.
또한, 도 5의 C 및 D에서 볼 수 있듯이, Ab417 및 시스플라틴을 병용 투여한 경우, 종양 무게에 근거하여, 대조군에 비하여 79.2%(p < 0.01)의 암 성장 저해율을 나타냄을 확인하였다. 반면, Ab417 또는 시스플라틴은 각각 39.7% 또는 51.2%의 암 성장 저해율을 나타내었다.In addition, as shown in C and D of Figure 5, when combined with Ab417 and cisplatin, it was confirmed that the cancer growth inhibition rate of 79.2% ( p <0.01) compared to the control based on the tumor weight. Ab417 or cisplatin, on the other hand, exhibited cancer growth inhibition of 39.7% or 51.2%, respectively.
아울러, 도 6의 A 내지 C에서 볼 수 있듯이, 병용 또는 단독 투여는 몸무게에 영향을 미치지 않음을 확인하였다.In addition, as shown in Figures A to C of 6, it was confirmed that the combination or alone administration does not affect the weight.
상기 결과를 통해, Ab417, 및 시스플라틴 또는 젬시타빈의 병용 투여는 항체 또는 약물 단독 투여에 비하여, 현저히 우수한, 상승효과를 나타냄을 알 수 있었다.From the above results, it was found that the combined administration of Ab417, and cisplatin or gemcitabine showed a significantly superior synergistic effect compared to the administration of antibody or drug alone.
실시예 2. ECC 이종이식 마우스 모델에서 항암 활성 확인Example 2. Confirmation of anticancer activity in ECC xenograft mouse model
실시예 2-1. Ab417의 막 L1CAM에 대한 영향 분석Example 2-1. Analysis of the effect of Ab417 on membrane L1CAM
Ab417의 내면화에 의하여 ECC 세포인 TFK-1 세포의 막 L1CAM 수준이 감소하는지 확인하고자 하였다.Being an ECC cell by internalization of Ab417 We tried to determine if the membrane L1CAM levels of TFK-1 cells decreased.
구체적으로, 상기 실험예 6에 따라, 지정된 시간(1, 2, 4, 6 시간) 동안 TFK-1 세포를 항체와 함께 배양하였고, 처리된 세포의 막단백질 분획을 인간 L1CAM의 Ig1 도메인에 결합하는 뮤라인(murine) 항-L1CAM mAb(A10-A3)를 이용한 웨스턴블롯으로 분석하였다.Specifically, according to Experiment 6, TFK-1 cells were incubated with the antibody for a designated time (1, 2, 4, 6 hours), and the membrane protein fraction of the treated cells was bound to the Ig1 domain of human L1CAM. Analysis was by Western blot using murine anti-L1CAM mAb (A10-A3).
그 결과, 도 7에서 볼 수 있듯이, Ab417는 농도 의존적인 방법으로 막 L1CAM 수준을 하향조절함을 확인하였다.As a result, it can be seen that Ab417 downregulates membrane L1CAM levels in a concentration dependent manner.
상기 결과를 통해, Ab417는 ECC 세포에서도, 내면화되어 막 L1CAM 수준을 감소시킴으로써 암세포의 증식을 억제함을 알 수 있었다.The results indicate that Ab417 inhibits the proliferation of cancer cells by internalizing even ECC cells to reduce membrane L1CAM levels.
실시예 2-2. Ab417의 항암 활성 확인Example 2-2. Anticancer activity of Ab417
ECC 모델에서 Ab417의 항암효과를 확인하기 위하여, 상기 실험예 3에 따라, Ab417(10 mg/kg), hFc(3.3 mg/kg) 또는 대조군으로서 살린을 TFK-1 이종이식을 갖는 NOD/SCID 마우스(n=8)에 일주일에 두 번씩 4주 동안 정맥 내 주사하였다.In order to confirm the anticancer effect of Ab417 in the ECC model, according to Experimental Example 3 above, Ab417 (10 mg / kg), hFc (3.3 mg / kg) or NOD / SCID mice having TFK-1 xenografts with saline as a control group ( n = 8) twice a week intravenously for 4 weeks.
그 결과, 도 8의 A 및 B에서 볼 수 있듯이, Ab417는 대조군에 비하여, 종양 무게에 근거하여, 65.4%의 항암 활성을 나타냄을 확인하였다.As a result, as can be seen in Figures A and B of Figure 8, Ab417 compared with the control group, it was confirmed that the anti-cancer activity of 65.4% based on the weight of the tumor.
상기 결과를 통해, Ab417은 ECC 세포에서도, 우수한 종양 성장을 억제 활성을 나타냄을 알 수 있었다.The results indicate that Ab417 exhibits excellent tumor growth inhibitory activity even in ECC cells.
실시예 2-3. 젬시타빈 또는 시스플라틴의 항암 효과 확인Example 2-3. Identify anticancer effects of gemcitabine or cisplatin
Ab417, 및 젬시타빈 또는 시스플라틴 병용 투여의 항암 효과를 확인하기 위하여, 먼저 ECC 종양 모델에서 젬시타빈 또는 시스플라틴 단독 투여의 항암효과를 확인하였다. 상기 실험예 3에 따라, 젬시타빈(5 또는 10 mg/kg), 시스플라틴(0.5 또는 2 mg/kg) 또는 대조군으로서 살린(saline)을 TFK-1 이종이식을 갖는 NOD/SCID 마우스(n=8)에 일주일에 두 번씩 복강 내 주사하였다. 주사 22일 후, 암 조직을 제거하여 무게를 측정하였다.In order to confirm the anticancer effect of Ab417, and gemcitabine or cisplatin combined administration, first, the anticancer effect of gemcitabine or cisplatin alone was administered in the ECC tumor model. According to Experimental Example 3 above, gemcitabine (5 or 10 mg / kg), cisplatin (0.5 or 2 mg / kg) or saline as a control (saline) NOD / SCID mice with TFK-1 xenograft ( n = 8 ) Intraperitoneally twice a week. Twenty two days after the injection, the cancerous tissue was removed and weighed.
그 결과, 도 9의 A 및 B에서 볼 수 있듯이, 5 또는 10 mg/kg의 젬시타빈은 종양 무게에 근거하여, 각각 43.2% 또는 77.0%의 암 성장 저해율을 나타냄을 확인하였고, 용량 의존적인 방법으로 암 성장을 저해함을 확인하였다.As a result, as can be seen in Figures A and B of 5, Gemcitabine of 5 or 10 mg / kg was confirmed to show cancer growth inhibition rate of 43.2% or 77.0%, respectively, based on tumor weight, dose-dependent method It was confirmed that inhibiting cancer growth.
반면에, 0.5 또는 2.0 mg/kg의 시스플라틴은 종양 무게에 근거하여, 각각 54.1% 또는 63.5%의 암 성장 저해율을 나타내었으나, 종양 부피에 근거하여는 단지 8.3% 또는 20.4% 정도의 암 성장 저해율을 나타냄을 확인하였다.On the other hand, 0.5 or 2.0 mg / kg of cisplatin showed 54.1% or 63.5% cancer growth inhibition, respectively, based on tumor weight, but only 8.3% or 20.4% inhibition of cancer growth based on tumor volume. It confirmed that it was shown.
실시예 2-4. Ab417, 및 젬시타빈 또는 시스플라틴 병용 투여의 항암 효과 확인Example 2-4. Confirmation of anticancer effect of Ab417 and gemcitabine or cisplatin combination
Ab417과 약물을 병용 투여하는 경우, 항체 또는 약물 단독을 투여하는 경우에 비하여 종양 성장 억제 효과가 월등한지 확인하였다.When Ab417 and drug were administered in combination, it was confirmed whether the tumor growth inhibitory effect was superior to that of antibody or drug alone.
구체적으로, 10 mg/kg의 Ab417은 TFK-1 모델에서 우수한 암 성장 저해 효과를 나타내었기 때문에, 본 발명에서는 더 낮은 용량인 5 mg/kg의 Ab417를 사용하였다.Specifically, since 10 mg / kg of Ab417 showed an excellent cancer growth inhibitory effect in the TFK-1 model, a lower dose of 5 mg / kg of Ab417 was used in the present invention.
상기 실험예 3에 따라, Ab417(5 mg/kg) 단독; 젬시타빈(5 mg/kg) 단독; 또는 시스플라틴(0.5 mg/kg) 단독을 투여하거나, Ab417(5 mg/kg) 및 젬시타빈(5 mg/kg); Ab417(5 mg/kg) 및 시스플라틴(0.5 mg/kg); 또는 Ab417(5 mg/kg) 및 대조군으로서 살린을 일주일에 두 번씩 3주 동안 TFK-1 이종이식을 갖는 NOD/SCID 마우스(n=8)에 주입하였다.According to Experiment 3, Ab417 (5 mg / kg) alone; Gemcitabine (5 mg / kg) alone; Or cisplatin (0.5 mg / kg) alone, or Ab417 (5 mg / kg) and gemcitabine (5 mg / kg); Ab417 (5 mg / kg) and cisplatin (0.5 mg / kg); Or Ab417 (5 mg / kg) and saline as a control was injected into NOD / SCID mice ( n = 8) with TFK-1 xenografts twice a week for 3 weeks.
그 결과, 도 10의 A 및 B에서 볼 수 있듯이, Ab417 및 젬시타빈을 병용 투여하는 경우에 암 성장은 완전히 저해되었고, 종양 부피에 근거하여, 대조군에 비하여 52.2%의 암 성장 저해율을 나타냄을 확인하였다. 반면, Ab417 또는 젬시타빈을 단독 투여하는 경우에는 각각 10.7% 또는 21.7%의 암 성장 저해율을 나타냄을 확인하였다.As a result, as shown in Figures A and B of Figure 10, in combination with Ab417 and gemcitabine cancer growth was completely inhibited, based on the tumor volume, it was confirmed that the cancer growth inhibition rate of 52.2% compared to the control group It was. On the other hand, when Ab417 or gemcitabine alone was administered, it was confirmed that the cancer growth inhibition rate of 10.7% or 21.7%, respectively.
상기 결과를 통해, Ab417, 및 젬시타빈의 병용 투여는 항체 또는 약물 단독 투여에 비하여, 그 효과가 현저히 우수한, 상승효과를 나타냄을 알 수 있었다.From the above results, it was found that the combined administration of Ab417 and gemcitabine showed a synergistic effect, which was significantly better than the antibody or drug alone administration.
이상의 설명으로부터, 본 발명이 속하는 기술분야의 당업자는 본 발명이 그 기술적 사상이나 필수적 특징을 변경하지 않고서 다른 구체적인 형태로 실시될 수 있다는 것을 이해할 수 있을 것이다. 이와 관련하여, 이상에서 기술한 실시 예들은 모든 면에서 예시적인 것이며 한정적인 것이 아닌 것으로서 이해해야만 한다. 본 발명의 범위는 상기 상세한 설명보다는 후술하는 특허 청구범위의 의미 및 범위 그리고 그 등가 개념으로부터 도출되는 모든 변경 또는 변형된 형태가 본 발명의 범위에 포함되는 것으로 해석되어야 한다.From the above description, those skilled in the art will appreciate that the present invention can be implemented in other specific forms without changing the technical spirit or essential features. In this regard, the embodiments described above are to be understood in all respects as illustrative and not restrictive. The scope of the present invention should be construed that all changes or modifications derived from the meaning and scope of the following claims and equivalent concepts rather than the detailed description are included in the scope of the present invention.

Claims (11)

  1. (a) L1CAM(L1 cell adhesion molecule) 단백질에 특이적으로 결합하는 항체; 및 (a) an antibody that specifically binds to an L1 cell adhesion molecule (L1CAM) protein; And
    (b) 피리미딘 유사체(pyrimidine analog), 플라틴계 항암제 또는 이들의 조합을 포함하는, 담도암의 예방 또는 치료용 약학적 조성물.(b) A pharmaceutical composition for the prevention or treatment of biliary tract cancer, comprising a pyrimidine analog, a platinum anticancer agent or a combination thereof.
  2. 제1항에 있어서, 상기 L1CAM(L1 cell adhesion molecule) 단백질에 특이적으로 결합하는 항체는 서열번호 1의 중쇄 가변영역 및 서열번호 5의 경쇄 가변영역을 포함하는 항체 또는 이의 기능성 변이체인, 약학적 조성물.The pharmaceutical composition of claim 1, wherein the antibody that specifically binds to an L1 cell adhesion molecule (L1CAM) protein is an antibody comprising a heavy chain variable region of SEQ ID NO: 1 and a light chain variable region of SEQ ID NO: 5, or a functional variant thereof. Composition.
  3. 제2항에 있어서, 상기 기능성 변이체는 서열번호 1의 중쇄 가변영역 및 서열번호 5의 경쇄 가변영역을 포함하는 항체의 중쇄 가변영역, 경쇄 가변영역, 또는 이들 둘 다에서 1개 이상의 아미노산이 치환되고, 서열번호 1의 중쇄 가변영역 및 서열번호 5의 경쇄 가변영역을 포함하는 항체에 비해 물성이 개선된 것인, 약학적 조성물.The method of claim 2, wherein the functional variant is substituted with one or more amino acids in the heavy chain variable region, light chain variable region, or both of the antibody comprising a heavy chain variable region of SEQ ID NO: 1 and a light chain variable region of SEQ ID NO: 5 The improved physical properties compared to the antibody comprising a heavy chain variable region of SEQ ID NO: 1 and a light chain variable region of SEQ ID NO: 5.
  4. 제1항에 있어서, 상기 항체는 서열번호 2로 기재된 중쇄 CDR1; 서열번호 3 또는 10으로 기재된 중쇄 CDR2; 및 서열번호 4로 기재된 중쇄 CDR3를 포함하는 중쇄 가변영역과 서열번호 6 또는 12로 기재된 경쇄 CDR1; 서열번호 7로 기재된 경쇄 CDR2; 및 서열번호 8로 기재된 경쇄 CDR3를 포함하는 경쇄 가변영역을 포함하는 것인, 약학적 조성물. The antibody of claim 1, wherein the antibody comprises a heavy chain CDR1 as set forth in SEQ ID NO: 2; Heavy chain CDR2 set forth in SEQ ID NO: 3 or 10; And a heavy chain variable region comprising a heavy chain CDR3 as set out in SEQ ID NO: 4 and a light chain CDR1 as set out in SEQ ID NO: 6 or 12; Light chain CDR2 set forth in SEQ ID NO: 7; And a light chain variable region comprising the light chain CDR3 set forth in SEQ ID NO: 8.
  5. 제4항에 있어서, 상기 항체는 서열번호 1, 9 및 13으로 이루어진 군으로부터 선택된, 중쇄 가변영역; 및The method according to claim 4, wherein the antibody is selected from the group consisting of SEQ ID NO: 1, 9 and 13, heavy chain variable region; And
    서열번호 5, 11, 14 및 15로 이루어진 군으로부터 선택된, 경쇄 가변영역을 포함하는, 약학적 조성물. A pharmaceutical composition comprising a light chain variable region selected from the group consisting of SEQ ID NOs: 5, 11, 14 and 15.
  6. 제1항에 있어서, 상기 피리미딘 유사체는 젬시타빈(gemcitabine)인 것인, 약학적 조성물.The pharmaceutical composition of claim 1, wherein the pyrimidine analog is gemcitabine.
  7. 제1항에 있어서, 상기 플라틴계 항암제는 시스플라틴(cisplatin), 옥살리플라틴(oxaliplatin), 테트라플라틴(tetraplatin), 헵타플라틴(heptaplatin), 파라플라틴(paraplatin), 카보플라틴(carboplatin) 또는 나노플라틴(nanoplatin)인 것인, 약학적 조성물.The method of claim 1, wherein the platinum-based anticancer agent is cisplatin (cisplatin), oxaliplatin (oxaliplatin), tetraplatin (heptraplatin), heptalatin (paratapin), carboplatin (carboplatin) or nano Platinum (nanoplatin), pharmaceutical composition.
  8. 제1항에 있어서, 상기 담도암은 간내 담도암 또는 간외 담도암인 것인, 약학적 조성물.The pharmaceutical composition of claim 1, wherein the biliary tract cancer is intrahepatic biliary tract cancer or extrahepatic biliary tract cancer.
  9. (a) L1CAM(L1 cell adhesion molecule) 단백질에 특이적으로 결합하는 항체; 및 (a) an antibody that specifically binds to an L1 cell adhesion molecule (L1CAM) protein; And
    (b) 피리미딘 유사체(pyrimidine analog), 플라틴계 항암제 또는 이들의 조합을 포함하는, 담도암의 예방 또는 치료용 약학적 키트.(b) A pharmaceutical kit for preventing or treating biliary tract cancer, comprising a pyrimidine analog, a platinum anticancer agent or a combination thereof.
  10. (a) L1CAM(L1 cell adhesion molecule) 단백질에 특이적으로 결합하는 항체; 및 (a) an antibody that specifically binds to an L1 cell adhesion molecule (L1CAM) protein; And
    (b) 피리미딘 유사체(pyrimidine analog), 플라틴계 항암제 또는 이들의 조합을 이를 필요로 하는 개체에게 투여하는 단계를 포함하는, 담도암의 치료 방법.(b) administering a pyrimidine analog, a platinum anticancer agent, or a combination thereof to a subject in need thereof.
  11. 제10항에 있어서, 상기 (a)의 항체와 (b)의 피리미딘 유사체(pyrimidine analog), 플라틴계 항암제 또는 이들의 조합은 동시, 개별, 또는 순차적으로 투여되는 것인, 담도암의 치료 방법.The method of claim 10, wherein the antibody of (a) and the pyrimidine analog of (b), the platinum-based anticancer agent or a combination thereof are administered simultaneously, separately or sequentially. .
PCT/KR2018/000056 2016-12-30 2018-01-02 Pharmaceutical composition for preventing or treating cancer, containing antibody specifically binding to l1cam protein and pyrimidine analog and/or platin-based anticancer drug WO2018124851A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2016-0183615 2016-12-30
KR20160183615 2016-12-30

Publications (1)

Publication Number Publication Date
WO2018124851A1 true WO2018124851A1 (en) 2018-07-05

Family

ID=62710671

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2018/000056 WO2018124851A1 (en) 2016-12-30 2018-01-02 Pharmaceutical composition for preventing or treating cancer, containing antibody specifically binding to l1cam protein and pyrimidine analog and/or platin-based anticancer drug

Country Status (2)

Country Link
KR (1) KR20180079232A (en)
WO (1) WO2018124851A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023078224A1 (en) * 2021-11-03 2023-05-11 同润生物医药(上海)有限公司 Novel anti-l1cam antibody

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040115206A1 (en) * 2002-10-24 2004-06-17 Thomas Primiano Antibody-mediated induction of tumor cell death
KR20080018149A (en) * 2006-08-23 2008-02-27 한국생명공학연구원 A pharmaceutical composition for treating cholangiocarcinoma, a method for inhibiting growth or invasion of cholangiocarcinoma and a method for treating cholangiocarcinoma
KR20100060351A (en) * 2008-11-27 2010-06-07 한국생명공학연구원 A composition for treating l1cam-expressing cancer comprising an inhibitor of activity or expression of l1cam and anticancer agent
KR20120005412A (en) * 2010-07-08 2012-01-16 강원대학교산학협력단 A pharmaceutical composition for treating gallbladder carcinoma, a method for inhibiting growth or invasion of gallbladder carcinoma and a method for treating gallbladder carcinoma
KR20140066101A (en) * 2012-11-16 2014-05-30 강원대학교산학협력단 Antibody specifically binding to l1cam in humans and mice, and use thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040115206A1 (en) * 2002-10-24 2004-06-17 Thomas Primiano Antibody-mediated induction of tumor cell death
KR20080018149A (en) * 2006-08-23 2008-02-27 한국생명공학연구원 A pharmaceutical composition for treating cholangiocarcinoma, a method for inhibiting growth or invasion of cholangiocarcinoma and a method for treating cholangiocarcinoma
KR20100060351A (en) * 2008-11-27 2010-06-07 한국생명공학연구원 A composition for treating l1cam-expressing cancer comprising an inhibitor of activity or expression of l1cam and anticancer agent
KR20120005412A (en) * 2010-07-08 2012-01-16 강원대학교산학협력단 A pharmaceutical composition for treating gallbladder carcinoma, a method for inhibiting growth or invasion of gallbladder carcinoma and a method for treating gallbladder carcinoma
KR20140066101A (en) * 2012-11-16 2014-05-30 강원대학교산학협력단 Antibody specifically binding to l1cam in humans and mice, and use thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023078224A1 (en) * 2021-11-03 2023-05-11 同润生物医药(上海)有限公司 Novel anti-l1cam antibody

Also Published As

Publication number Publication date
KR20180079232A (en) 2018-07-10

Similar Documents

Publication Publication Date Title
AU2019229076B2 (en) Anti-TIGIT antibodies and uses thereof
WO2020076105A1 (en) Novel anti-c-kit antibody
CA3142119A1 (en) Dosage of an antibody-drug conjugate for treating cancer
JP2017114763A (en) Anti-CD98 antibody-drug conjugate
CN105979966B (en) Novel anti-neurotrophin-1 antibodies
US20240067731A1 (en) Mica/b antibodies and methods of use
WO2016085289A1 (en) Antibody binding specifically to pre-s1 of hepatitis b virus and use of the antibody
US20210253711A1 (en) Anti-mica/b antibodies that block mica/b shedding and methods of use
WO2017030370A1 (en) Chimeric antibody receptor to which anti-cotinine antibody is linked, and use thereof
WO2018174544A2 (en) Antibody binding specifically to muc1 and use thereof
FR3107648A1 (en) anti-CD56 antibody-drug conjugates and their use in therapy
WO2019212253A1 (en) Antibody specifically binding to c-met, and use thereof
WO2018124851A1 (en) Pharmaceutical composition for preventing or treating cancer, containing antibody specifically binding to l1cam protein and pyrimidine analog and/or platin-based anticancer drug
WO2018186706A1 (en) Nk cell-activating fusion protein, nk cell, and pharmaceutical composition including same
WO2020145669A1 (en) Anti-beta 1 integrin humanized antibody, and pharmaceutical composition for treating cancer, comprising same
WO2020004937A1 (en) Anti-bcma antibody-drug conjugate and use thereof
EP3999542A1 (en) Axl antibody-drug conjugates for use in treating cancer
KR20220034117A (en) Immune checkpoint blocking bispecific molecule
US11649277B2 (en) Method for treating skin diseases using a humanized antibody that binds to vimentin
WO2016084993A1 (en) Novel egfrviii antibody and composition comprising same
WO2013176516A1 (en) Antibody-drug conjugate prepared by using transglutaminase and use thereof
WO2014021693A2 (en) Novel monoclonal antibody which is specifically bound to tm4sf5 protein and use thereof
WO2020071881A1 (en) Pdgf receptor antibody and use thereof
WO2019022274A1 (en) Antibody specifically binding to pauf protein, and use thereof
WO2024063624A1 (en) Antibody-drug conjugate comprising drug and antibody specifically binding to grp94 or antigen-binding fragment 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: 18734041

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: JP

122 Ep: pct application non-entry in european phase

Ref document number: 18734041

Country of ref document: EP

Kind code of ref document: A1