WO2024090916A1 - Pharmaceutical composition for cancer treatment comprising anti-igsf1 antibody - Google Patents

Pharmaceutical composition for cancer treatment comprising anti-igsf1 antibody Download PDF

Info

Publication number
WO2024090916A1
WO2024090916A1 PCT/KR2023/016436 KR2023016436W WO2024090916A1 WO 2024090916 A1 WO2024090916 A1 WO 2024090916A1 KR 2023016436 W KR2023016436 W KR 2023016436W WO 2024090916 A1 WO2024090916 A1 WO 2024090916A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
antibody
igsf1
biliary tract
cells
Prior art date
Application number
PCT/KR2023/016436
Other languages
French (fr)
Korean (ko)
Inventor
이민기
손혜진
이미소
배승건
이원준
Original Assignee
웰마커바이오 주식회사
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 웰마커바이오 주식회사 filed Critical 웰마커바이오 주식회사
Publication of WO2024090916A1 publication Critical patent/WO2024090916A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to a pharmaceutical composition for preventing or treating cancer comprising an anti-IGSF1 antibody or fragment thereof as an active ingredient.
  • Cancer is one of the greatest diseases threatening human health. It is a disease that occurs when cells undergo a series of mutations, proliferate in an unlimited and uncontrolled manner, and become immortal. Causes of cancer include environmental or external factors such as chemicals, viruses, bacteria, and ionizing radiation, and internal factors such as congenital genetic mutations. For cancer discovered in the early stages, there are treatments such as surgery, radiation therapy, and chemotherapy, but their side effects are becoming a major problem, and for terminal cancer or metastatic cancer, life is terminal and ends without any special treatment.
  • epidermal growth factor receptor such as FOLFIRI (Folinic acid, Fluorouracil and Irinotecan), FOLFOX (Folinic acid, Fluorouracil and Oxaliplatin), Erbitux, Vectibix, etc.
  • FOLFIRI Fluorouracil and Irinotecan
  • FOLFOX Fluorouracil and Oxaliplatin
  • Erbitux Vectibix
  • Receptor-targeted anticancer antibody treatments and angiogenesis inhibitors such as Avastin or Zaltrap are mainly used as anticancer treatments.
  • the treatment response rate in patients who have received a prescription is very low at about 20%, or the recurrence rate within 3 years is high in patients who have shown treatment effects, and the drug is showing limited effectiveness, such as lack of response to the drug or resistance occurring after treatment. .
  • Biliary tract cancer has the characteristic that surgical resection is not effective due to its high recurrence rate and that it does not respond well to conventional chemotherapy or radiotherapy.
  • Biliary tract cancer is treated as a first-line treatment with Gemcitabine, Capecitabine, or 5-Fluorouracil (5-FU) as single agents, or with Oxaliplatin or Cisplatin. It is recommended to use it in combination with platinum compounds such as ), or in combination with gemcitabine and capecitabine.
  • multi-agent chemotherapy does not show lasting effects in patients with metastatic and/or recurrent biliary tract cancer, and in the case of advanced biliary tract cancer, the 5-year survival rate after diagnosis is less than 5%.
  • cisplatin a representative anticancer drug for the treatment of head and neck cancer, has excellent anticancer effects, but in the case of patients with cisplatin resistance or patients who develop resistance during the treatment process, problems to overcome resistance are needed. am.
  • the present inventors studied a method for effectively treating cancer and completed the present invention by confirming that tumor growth was inhibited by administration of an anti-IGSF1 antibody or fragment thereof.
  • one aspect of the present invention is to prevent or treat any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer, comprising an anti-IGSF1 antibody or fragment thereof as an active ingredient.
  • a pharmaceutical composition is provided.
  • Another aspect of the present invention provides a method for preventing or treating any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer, comprising administering an anti-IGSF1 antibody or fragment thereof to a subject.
  • Another aspect of the present invention provides an anti-IGSF1 antibody or fragment thereof for use in the prevention or treatment of any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer.
  • Another aspect of the present invention provides the use of an anti-IGSF1 antibody or fragment thereof for the manufacture of a medicament for preventing or treating any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer.
  • the anti-IGSF1 antibody according to the present invention When the anti-IGSF1 antibody according to the present invention was treated with colon cancer cells, biliary tract cancer cells, or head and neck cancer cells and immune cells, the secretion of Granzyme B and the cytokines IFN- ⁇ and TNF- ⁇ increased. . It was confirmed that the growth of colon cancer or biliary tract cancer was inhibited when the antibody was administered to a xenograft or allogeneic colon cancer or biliary tract cancer implanted mouse model. Therefore, the anti-IGSF1 antibody of the present invention can be usefully used in the treatment of colon cancer, biliary tract cancer, and head and neck cancer.
  • Figure 1 is a diagram showing the results of confirming the expression of IGSF1 in human colon cancer cell line HT29 cells through flow cytometry.
  • Figure 2 is a graph showing the results of confirming changes in the secretion amount of Granzyme B, IFN- ⁇ , and TNF- ⁇ by treatment with WM-A1-3389 antibody in a co-culture system of HT29 cells and human peripheral blood cells (PBMC).
  • PBMC peripheral blood cells
  • Figure 3 is a graph showing the results of measuring the degree of tumor growth by administration of WM-A1-3389 antibody in a mouse model implanted with the human colon cancer cell line HT29.
  • Figure 4 is a diagram showing the results of confirming the expression of IGSF1 in the mouse colon cancer cell line MC38 through immunofluorescence staining (top) and flow cytometry (bottom).
  • Figure 5 is a graph showing the results of measuring the degree of tumor growth by administration of WM-A1-3389 antibody in a mouse model implanted with the mouse colon cancer cell line MC38.
  • Figure 6 is a diagram showing the results of confirming the expression of IGSF1 in the mouse colon cancer cell line CT26 through immunofluorescence staining (top) and flow cytometry (bottom).
  • Figure 7 is a graph showing the results of measuring the degree of tumor growth by administration of WM-A1-3389 antibody in a mouse model implanted with the mouse colon cancer cell line CT26.
  • Figure 8 is a diagram showing the results of confirming the expression of IGSF1 in human biliary tract cancer cell line Choi-CK cells through flow cytometry.
  • Figure 9 is a graph showing the results of confirming changes in the secretion amount of Granzyme B, IFN- ⁇ , and TNF- ⁇ by treatment with WM-A1-3389 antibody in a co-culture system of Choi-CK cells and human peripheral blood cells (hPBMC).
  • Figure 10 is a graph showing the results of measuring the degree of tumor growth by administration of WM-A1-3389 antibody in a mouse model implanted with the human biliary tract cancer cell line Choi-CK.
  • Figure 11 is a diagram showing the results of confirming the expression of IGSF1 in the human head and neck cancer cell line FaDu cells through flow cytometry.
  • Figure 12 is a graph showing the results confirming changes in the secretion amounts of Granzyme B, IFN- ⁇ , and TNF- ⁇ by treatment with WM-A1-3389 antibody in the FaDu cell and human peripheral blood cell (hPBMC) co-culture system.
  • IGSF1 is a membrane protein encoded by the IGSF1 gene found on the X chromosome of humans and other mammalian species. Although the function of IGSF1 in normal cells is not well known, IGSF1 mutations are known to cause diseases such as IGSF1 deficiency syndrome or central hypothyroidism.
  • the IGSF1 may be included without limitation as long as it is mammalian IGSF1, but preferably refers to human IGSF1. Additionally, in the present invention, the IGSF1 protein includes, but is not limited to, both native and mutant IGSF1 proteins.
  • the native IGSF1 protein generally refers to a polypeptide containing the amino acid sequence of the native IGSF1 protein, and the amino acid sequence of the native IGSF1 protein generally refers to the amino acid sequence found in naturally occurring IGSF1.
  • Information about IGSF1 can be obtained from known databases such as GenBank of the National Institutes of Health, and may have, for example, an amino acid sequence (SEQ ID NO: 19) with Genbank accession number NP_001164433.1, but is not limited thereto.
  • antibody refers to an immunoglobulin molecule that reacts immunologically with a specific antigen, and refers to a protein molecule that specifically recognizes the antigen.
  • the antibodies include whole antibodies, monoclonal antibodies, polyclonal antibodies, single domain antibodies, single chain antibodies, multispecific antibodies, human antibodies, humanized antibodies, chimeric antibodies, intrabodies, scFvs, Fab fragments, F (ab') fragment, disulfide bond-linked Fv (sdFv), and epitope-binding fragments of any of the above.
  • anti-IGSF1 antibody refers to an antibody capable of specifically binding to IGSF1, and may be used interchangeably with “IGSF1-specific antibody” in the present invention.
  • anti-IGSF1 antibodies can specifically bind to the C terminus of IGSF1.
  • the form of the antibody may include both whole antibodies and antibody fragments thereof.
  • the heavy and light chains of immunoglobulins may each include a constant region and a variable region.
  • the light and heavy chain variable regions of immunoglobulins include three variable regions called complementarity determining regions (CDRs) and four framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • the CDR mainly functions to bind to the epitope of the antigen.
  • the CDRs of each chain are typically called CDR1, CDR2, and CDR3 sequentially, starting from the N terminus, and are identified by the chain on which the specific CDR is located.
  • the anti-IGSF1 antibody or fragment thereof of the present invention includes H-CDR1 containing the amino acid sequence of SEQ ID NO: 1, H-CDR2 containing the amino acid sequence of SEQ ID NO: 2, and H-CDR3 containing the amino acid sequence of SEQ ID NO: 3. It may include a heavy chain variable region (VH).
  • the anti-IGSF1 antibody or fragment thereof of the present invention includes L-CDR1 containing the amino acid sequence of SEQ ID NO: 4, L-CDR2 containing the amino acid sequence of SEQ ID NO: 5, and L- CDR2 containing the amino acid sequence of SEQ ID NO: 6. It may include a light chain variable region (VL) including CDR3.
  • the heavy chain variable region may include the amino acid sequence of SEQ ID NO: 7, and the light chain variable region may include the amino acid sequence of SEQ ID NO: 8.
  • the antibody may be referred to as WM-A1-3389.
  • the heavy chain variable region of the antibody consists of the amino acid sequence of SEQ ID NO: 7 and about 90% or more, about 91% or more, about 92% or more, about 93% or more, about 94% or more, about 95% or more, about 96% or more, about It may comprise or consist of an amino acid sequence having at least 97%, at least about 98%, at least about 99%, or 100% identity.
  • the light chain variable region of the antibody is about 90% or more, about 91% or more, about 92% or more, about 93% or more, about 94% or more, about 95% or more, and about 96% or more of the amino acid sequence of SEQ ID NO: 8. , may include or consist of an amino acid sequence having an identity of at least about 97%, at least about 98%, at least about 99%, or 100%.
  • Immunoglobulin heavy chain constant regions exhibit different amino acid compositions and sequences and therefore possess different types of antigenicity. Accordingly, immunoglobulins can be divided into five categories and referred to as immunoglobulin isotypes, namely IgM, IgD, IgG, IgA and IgE.
  • the corresponding heavy chains are ⁇ chain, ⁇ chain, ⁇ chain, ⁇ chain and ⁇ chain, respectively.
  • the same type of Ig can be classified into different subtypes. For example, IgG can be classified as IgG1, IgG2, IgG3, and IgG4.
  • Light chains can be classified as ⁇ or ⁇ chains depending on their different constant regions. Each of the five types of IgG can have either a ⁇ or ⁇ chain.
  • the anti-IGSF1 antibody or fragment thereof of the present invention may include a constant region derived from IgG, IgA, IgD, IgE, IgM, or a partial hybrid thereof.
  • hybrid means that sequences corresponding to immunoglobulin heavy chain constant regions of two or more different origins exist within the single-chain immunoglobulin heavy chain constant region.
  • a domain hybrid consisting of 1 to 4 domains selected from the group consisting of CH1, CH2, and CH3 of IgG, IgA, IgD, IgE, and IgM is possible.
  • the anti-IGSF1 antibody or fragment thereof of the present invention includes a light chain constant region (LC)
  • the light chain constant region may be derived from a ⁇ or ⁇ light chain.
  • antibody fragment refers to an scFv fragment, which is an Fv fragment that binds to IGSF1, as well as a Fab fragment, Fab' fragment, and F(ab') 2 fragment having antigen-binding activity, and is used in the present invention. Contains the CDR regions of the antibodies described in.
  • the Fv fragment is the smallest antibody fragment that contains the heavy and light chain variable regions, without constant regions, and retains all antigen-binding sites.
  • the pharmaceutical composition containing the anti-IGSF1 antibody or fragment thereof of the present invention as an active ingredient exhibits preventive or therapeutic efficacy against any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer.
  • the colon cancer, biliary tract cancer, and head and neck cancer may be cancers in which IGSF1 is overexpressed.
  • colon cancer used in the present invention refers to a malignant tumor composed of cancer cells that arise in the large intestine (cecum, appendix, colon, rectum, or anal canal). Pathologically, colon cancer is mostly adenocarcinoma, but it also includes lymphoma, sarcoma, squamous cell carcinoma, and metastatic lesions of other cancers. By site, it is largely divided into colon cancer and rectal cancer, with the highest incidence of cancer occurring in the lower colon, or rectum, at about 50%. The cause of colon cancer is still unclear, but genetic factors, dietary habits related to the consumption of high-fat and low-fiber foods, and inflammatory bowel disease are being considered. Colon cancer can occur at any age, but the incidence increases with age and occurs frequently in people in their 50s or 60s. The incidence ratio between men and women is somewhat higher for colon cancer in women and for rectal cancer in men.
  • biliary tract cancer used in this specification is also called cholangiocarcinoma and refers to cancer that occurs in the bile ducts.
  • the bile duct is a tube that transports bile produced in the liver to the duodenum. Cancers originating in the bile duct are divided into intrahepatic bile duct cancer (about 20% to about 25%), hilar bile duct cancer (about 50% to about 60%), and distal liver cancer, depending on the anatomical location. It can be classified into bile duct cancer (about 20% to about 25%). Adenocarcinoma arising from cholangiocytes accounts for the majority of cholangiocarcinomas.
  • cholangiocarcinoma The cause of cholangiocarcinoma is not yet clearly known, and it is caused by chronic inflammation in the bile duct cells lining the bile ducts, bile duct stones, sclerosing cholangitis, liver dystomatosis, inflammatory bowel disease, and bile duct cysts caused by congenital dilatation of the bile ducts. It is known to be related to etc.
  • head and neck cancer refers to malignant tumors that occur in the face, nose, throat, mouth, larynx, pharynx, salivary glands, and thyroid gland, excluding tumors that occur in the brain and eyes.
  • cancer that occurs in the oral cavity is called oral cancer
  • cancer that occurs in the larynx, the organ that produces sound is called laryngeal cancer
  • cancer that occurs in the pharynx is called pharyngeal cancer.
  • the pharyngeal cancer is divided into nasopharyngeal cancer, oropharyngeal cancer, and hypopharyngeal cancer depending on the location of occurrence.
  • head and neck cancer includes cervical esophageal cancer, salivary gland cancer, salivary gland cancer, malignant lymphoma, malignant melanoma, and various soft tissue cancers, and thyroid cancer is included in head and neck cancer in a comprehensive sense.
  • Smoking and drinking are known to be traditional causes of head and neck cancer
  • nasopharyngeal cancer is known to be closely related to viral infection (Epstein Barr virus).
  • gastric acid reflux disease, esophageal disease, radiation or ultraviolet rays, vitamin or iron deficiency, etc. are known to be factors that increase the incidence of head and neck cancer.
  • prevention refers to all actions that suppress or delay the onset of a disease by administering the pharmaceutical composition.
  • treatment refers to any action that improves or beneficially changes the symptoms of a disease by administering the pharmaceutical composition.
  • the anti-IGSF1 antibody or fragment thereof may be included in any amount (effective amount) depending on use, formulation, formulation purpose, etc., as long as it can exhibit anti-cancer activity.
  • “effective amount” refers to the amount of active ingredient that can induce an anticancer effect. Such effective amounts can be determined experimentally within the scope of the ordinary ability of those skilled in the art.
  • the pharmaceutical composition of the present invention contains the antibody as an active ingredient in an amount of about 0.1% to about 90% by weight, specifically about 0.5% by weight to about 75% by weight, and more specifically about 1% by weight to about 1% by weight, based on the total weight of the composition. It can be contained at 50% by weight.
  • “enhanced efficacy” e.g., improvement in efficacy
  • the term "efficacy” means survival over a period of time, such as 1 year, 5 years, or 10 years, or disease-free survival, which can be determined by one or more parameters. You can. Additionally, the parameter may include that the size of at least one tumor in the subject is suppressed.
  • the pharmaceutical composition of the present invention may contain a conventional, non-toxic pharmaceutically acceptable carrier that is formulated into a preparation according to a conventional method.
  • the pharmaceutically acceptable carrier may be any carrier that is a non-toxic material suitable for delivery to a patient. Distilled water, alcohol, fats, waxes and inert solids may be included as carriers. Pharmacologically acceptable adjuvants (buffers, dispersants) may also be included in the pharmacological composition.
  • the term “pharmaceutically acceptable carrier” refers to a carrier or diluent that does not irritate living organisms and does not inhibit the biological activity and properties of the administered compound.
  • Acceptable pharmaceutical carriers in compositions formulated as liquid solutions include those that are sterile and biocompatible, such as saline solution, sterile water, Ringer's solution, buffered saline solution, albumin injection solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and One or more of these ingredients can be mixed and used, and other common additives such as sweeteners, solubilizers, wetting agents, emulsifiers, isotonic agents, absorbents, antioxidants, preservatives, lubricants, fillers, buffers, and bacteriostatic agents are added as needed. can do.
  • compositions of the present invention can be prepared in a variety of formulations for parenteral administration (e.g., intramuscular, intravenous, or subcutaneous injection).
  • parenteral administration e.g., intramuscular, intravenous, or subcutaneous injection.
  • the pharmaceutical composition of the present invention can be formulated in the form of injections, transdermal administration, nasal inhalation, and suppositories along with a suitable carrier according to methods known in the art.
  • injectable preparations include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized preparations, and suppositories.
  • Non-aqueous solvents and suspensions include propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable esters such as ethyl oleate.
  • injectables may contain conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifiers, stabilizers, preservatives, etc.
  • the pharmaceutical composition of the present invention can be administered to a patient in a therapeutically effective amount or a pharmaceutically effective amount.
  • the term "administration” means introducing a predetermined substance into an individual by an appropriate method, and the composition may be administered through any general route as long as it can reach the target tissue. It may be administered intraperitoneally, intravenously, intramuscularly, subcutaneously, intradermally, locally, intranasally, or rectally, but is not limited thereto.
  • therapeutically effective amount refers to the amount of a compound or composition effective in preventing or treating the target disease, which is sufficient to treat the disease with a reasonable benefit/risk ratio applicable to medical treatment. This refers to an amount that does not cause side effects.
  • the level of the effective amount is determined by factors including the patient's health status, type and severity of the disease, drug activity, sensitivity to the drug, administration method, administration time, administration route and excretion rate, treatment period, combination or concurrent use of drugs, and It may be determined based on other factors well known in the medical field.
  • a therapeutically effective amount refers to an amount of drug effective in treating the disease.
  • the dosage of the composition of the present invention may vary depending on the patient's age, gender, and weight, and is generally administered from about 0.1 mg to about 1,000 mg per kg of body weight, or from about 5 mg to about 200 mg per kg of body weight every day or every other day. Alternatively, it can be administered at intervals of 2 to 3 weeks, or divided into 1 to 3 times a day. However, since it may increase or decrease depending on the route of administration, severity of disease, gender, weight, age, etc., the scope of the present invention is not limited thereto.
  • subject refers to an object to which the composition of the present invention can be applied (prescribed), including humans, rats, mice, guinea pigs, hamsters, rabbits, dogs, cats, chickens (including eggs), pigs, monkeys, It may be a mammal such as a goat. Preferably, it may be a human, but is not limited thereto.
  • the pharmaceutical composition of the present invention may be administered as an individual therapeutic agent or in combination with other therapeutic agents, may be administered sequentially or simultaneously with conventional therapeutic agents, and may be administered singly or multiple times.
  • the other therapeutic agent may additionally include any compound or natural extract whose safety has already been verified and which is known to have anticancer activity in order to increase or reinforce the anticancer activity.
  • Another aspect of the present invention is to treat any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer, comprising administering to an individual a pharmaceutical composition containing an anti-IGSF1 antibody or fragment thereof as an active ingredient.
  • a pharmaceutical composition containing an anti-IGSF1 antibody or fragment thereof as an active ingredient comprising administering to an individual a pharmaceutical composition containing an anti-IGSF1 antibody or fragment thereof as an active ingredient.
  • anti-IGSF1 antibody and fragment thereof, pharmaceutical composition, colorectal cancer, biliary tract cancer, head and neck cancer, administration, treatment and prevention are the same as described above.
  • the subject may be a mammal, preferably a human. Additionally, the individual may be a patient with the disease or an individual who is likely to suffer from the disease.
  • the administration route, dosage, and frequency of administration of the pharmaceutical composition may be administered to the subject in various methods and amounts depending on the patient's condition and the presence or absence of side effects, and the optimal administration method, dosage, and frequency of administration can be determined by a person skilled in the art. You can select by range. Additionally, the anti-IGSF1 antibody or fragment thereof may be administered in combination with other drugs or biochemically active substances known to have therapeutic effects on the disease, or may be formulated in the form of a combination preparation with other drugs.
  • Another aspect of the present invention is the prevention or treatment of any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer using a pharmaceutical composition containing the anti-IGSF1 antibody or fragment thereof of the present invention as an active ingredient.
  • a pharmaceutical composition containing the anti-IGSF1 antibody or fragment thereof of the present invention as an active ingredient Provides a purpose.
  • the anti-IGSF1 antibody and fragment thereof, pharmaceutical composition, colon cancer, biliary tract cancer, head and neck cancer, treatment and prevention are the same as described above.
  • Another aspect of the present invention provides an anti-IGSF1 antibody or fragment thereof for use in the prevention or treatment of any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer.
  • the anti-IGSF1 antibody and fragment thereof, pharmaceutical composition, colon cancer, biliary tract cancer, head and neck cancer, treatment and prevention are the same as described above.
  • Another aspect of the present invention provides the use of an anti-IGSF1 antibody or fragment thereof for the manufacture of a medicament for preventing or treating any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer.
  • the anti-IGSF1 antibody and fragment thereof, pharmaceutical composition, colon cancer, biliary tract cancer, head and neck cancer, prevention and treatment are the same as described above.
  • IGSF1 protein expression vector was created by fusing a polynucleotide encoding hig tag).
  • HEK293F cells were transfected with the prepared IGSF1 expression vector and then cultured for 6 days in medium supplemented with 1 mM valporic acid (valproate). Then, after primary purification of the IGSF1 extracellular domain using protein A agarose, secondary purification of the IGSF1 extracellular domain using Superdex 200 gel filtration chromatography, It was used for antibody selection.
  • bio-panning bio-panning, Y Biologics Co., Ltd.
  • bio-panning Y Biologics Co., Ltd.
  • human antibody library phage Y Biologics Co., Ltd.
  • the second and third rounds of biopanning were performed using the phage amplified in the first round of biopanning.
  • ELISA was performed to confirm the antigen specificity of the positive phage antibody pool obtained through each round of biopanning. In addition, it was confirmed that anti-IGSF1 antibody was enriched in the phage pool obtained through the third round.
  • the most optimized antibody was selected and named “WM-A1-3389”.
  • the CDR sequence of the WM-A1-3389 antibody is shown in Table 1 below.
  • a polynucleotide (SEQ ID NO: 23) encoding the heavy chain (SEQ ID NO: 21) was loaded into the N293F vector (Y Biologics Co., Ltd.) (hereinafter referred to as HC DNA).
  • a polynucleotide (SEQ ID NO: 24) encoding the light chain (SEQ ID NO: 22) was loaded into the N293F vector (Y Biologics Co., Ltd.) (hereinafter referred to as LC DNA).
  • LC DNA N293F vector
  • IGSF1 was confirmed in HT29 cells, a human colon cancer cell line.
  • the medium of HT29 cells was removed, washed once with PBS, and then treated with 2 ml of Accutase to separate the cells.
  • the separated cells were diluted in 8 ml of PBS (hereinafter referred to as FACS buffer) containing 2% (v/v) FBS and 0.05% (w/v) sodium azide, then centrifuged at 1,200 rpm for 1 minute to remove the supernatant. did.
  • the cell pellet was released with a vortex and resuspended with an appropriate amount of FACS buffer, and the number of cells was counted using trypan blue and distributed to each tube to reach 1 ⁇ 10 5 cells.
  • FACS buffer 200 ⁇ l was added to the remaining cell pellet, resuspended, and analyzed by FACS.
  • fluorescence value of Alexa 647 labeled in each cell was measured using BD LSRFortessa Flow cytometry (BD Bioscience), and the results were analyzed using FLowJo software.
  • Example 2.2 Confirmation of changes in cytokine secretion by co-culture of human colon cancer cell lines and peripheral blood cells
  • hPBMC human-derived peripheral blood cells
  • HT29 colon cancer cells and hPBMC were inoculated at a 1:1 ratio, and 1 ⁇ g/ml SEB (Sigma), 10 ⁇ g/ml IgG (Bio The cells were treated and cultured in a carbon dioxide incubator at 37°C for 24 hours. At this time, IgG was used as a negative control.
  • the supernatant of the co-cultured cells was collected in a 1.5 ml Eppendorf tube and the amount of cytokine secretion was measured through Cytometric bead array (CBA) assay.
  • CBA Cytometric bead array
  • the CBA assay used a BD Biosciences product and was performed according to the BD Biosciences protocol. Standard (standard sample) and co-cultured cell supernatant were added to each tube, capture beads were added, and reaction was performed at room temperature for 1 to 2 hours. After reaction, PE detection reagent was added and further reaction was performed at room temperature for 2 hours. Each tube was washed with washing buffer and measured using LSRFortessaTM Flow Cytometry equipment (BD Bioscience).
  • the secretion amount of cytokines IFN- ⁇ and TNF- ⁇ , including Granzyme B, in HT29 colon cancer cells expressing IGSF1 was statistically significantly lower in the group treated with the WM-A1-3389 antibody compared to the control group. It was confirmed that there was a significant increase.
  • Example 2.3 Confirmation of anticancer activity of WM-A1-3389 antibody in xenogeneic colon cancer cell line transplantation mouse model
  • the anticancer activity of the WM-A1-3389 antibody was confirmed in a colon cancer mouse model transplanted with a human colon cancer cell line.
  • mice 6-week-old female peripheral blood mononuclear cell humanized mice (Gem biosciences) were purchased and acclimatized for 1 week, and then HT29 cells, a human colon cancer cell line, were diluted in PBS and instilled on the right ventral side to a concentration of 5 ⁇ 10 6 cells/mice. It was injected subcutaneously (100 ⁇ l). Human IgG isotype was used as a negative control for the WM-A1-3389 antibody. When the tumor size reached an average of 140 mm 3 to 160 mm 3 , human IgG isotype (negative control) was administered intraperitoneally at a dose of 10 mg/kg or WM-A1-3389 antibody at a dose of 30 mg/kg. Administration was conducted once every three days for four weeks, and the tumor size and body weight of the mice were measured twice a week. On the 28th day of administration, the experimental animals were euthanized and the tumors were extracted and weighed.
  • HT29 cells a human colon cancer cell line
  • the WM-A1-3389 antibody administration group showed higher tumor growth inhibition efficacy than the negative control group.
  • TGI tumor growth inhibition
  • TGI tumor growth inhibition
  • Example 2.4 Confirmation of anticancer activity of WM-A1-3389 antibody in allogeneic colon cancer cell line transplantation mouse model
  • IGSF1 in MC38 cells was confirmed through immunofluorescence staining and flow cytometry. Immunofluorescence staining was performed as follows. Cells with the supernatant removed were fixed with 4% (v/v) formaldehyde (PFA, T&I) at room temperature for 15 minutes. The fixed cells were washed twice with PBS, blocking buffer was added to prevent non-specific binding, and reacted at room temperature for 1 hour. After the reaction, the primary IGSF1 antibody (sc-393786, Santa Cruz Biotechnology) was treated at a ratio of 1:100 and reacted at 4°C for more than 12 hours, protected from light.
  • PFA formaldehyde
  • the cells were washed twice with PBS, treated with anti-mouse IgG H&L (Alexa 488) (ab150113, Abcam) antibody as a secondary antibody at a ratio of 1:100, and incubated for 1 hour at room temperature, shielded from light.
  • the stained cells were washed twice with PBS and observed under a fluorescence microscope (Olympus). Flow cytometry was performed in the same manner as Example 2.1.
  • Example 2.4.2 Confirmation of anticancer activity of WM-A1-3389 antibody in mouse colon cancer cell line MC38 implanted mouse model
  • the anticancer activity of the WM-A1-3389 antibody was confirmed in a colon cancer mouse model (MC38 syngeneic mouse model) transplanted with MC38 cells, a mouse colon cancer cell line.
  • mice 5-week-old female C57BL/6N mice were purchased and acclimatized for 1 week.
  • Mouse colon cancer cells MC38 cells were diluted in PBS to prepare a concentration of 2.5 ⁇ 10 5 cells/mice, and MC38 cells were injected subcutaneously (100 ⁇ l) into the right ventral side of the mouse.
  • Mouse IgG isotype was used as a negative control for the WM-A1-3389 antibody.
  • mouse IgG isotype negative control
  • mice was administered intraperitoneally at doses of 10 mg/kg and WM-A1-3389 antibody at doses of 3, 10, and 30 mg/kg, respectively.
  • Administration was performed once every three days for two weeks, and the tumor size and body weight of the mice were measured twice a week. On the 14th day of administration, the experimental animals were euthanized and the tumors were extracted and weighed.
  • the WM-A1-3389 antibody administered group showed higher tumor growth inhibition efficacy than the negative control group, and a correlation was confirmed in which the tumor growth inhibition rate (TGI) increased in a dose-dependent manner.
  • TGI tumor growth inhibition rate
  • the WM-A1-3389 antibody dosage and tumor growth inhibition rate (TGI) are listed in Table 4 below.
  • WM-A1-3389 antibody administration group Dose(mg/kg) 3 10 30 TGI(%) 50.8 ⁇ 4.3 58.2 ⁇ 4.8 68.7 ⁇ 3.8
  • IGSF1 in CT26 cells, a mouse colon cancer cell line, was confirmed through immunofluorescence staining and flow cytometry. Immunofluorescence staining was performed in the same manner as in Example 2.4.1, and flow cytometry was performed in the same manner as in Example 2.1.
  • Example 2.4.4 Confirmation of anticancer activity of WM-A1-3389 antibody in mouse colon cancer cell line CT26 implanted mouse model
  • the anticancer activity of the WM-A1-3389 antibody was confirmed in a colon cancer mouse model (CT26 syngeneic mouse model) transplanted with CT26 cells, a mouse colon cancer cell line.
  • mice 5-week-old female C57BL/6N mice were purchased, acclimatized for 1 week, and mouse colon cancer CT26 cells were diluted in PBS and injected subcutaneously (100 ⁇ l) into the right ventral side to a concentration of 2.5 ⁇ 10 5 cells/mice. did.
  • Human IgG isotype was used as a negative control for the WM-A1-3389 antibody.
  • 10 mg/kg of mouse IgG isotype (negative control) and WM-A1-3389 antibody were administered intraperitoneally at doses of 3, 10, and 50 mg/kg, respectively.
  • Administration was performed once every three days for two weeks, and the tumor size and body weight of the mice were measured twice a week. On the 14th day of administration, the experimental animals were euthanized, the tumors were extracted, and their weight was measured.
  • the WM-A1-3389 antibody administered group showed higher tumor growth inhibition efficacy than the negative control group, and a correlation was confirmed in which the tumor growth inhibition rate (TGI) increased in a dose-dependent manner.
  • TGI tumor growth inhibition rate
  • the WM-A1-3389 antibody dosage and tumor growth inhibition rate (TGI) are listed in Table 5 below.
  • WM-A1-3389 antibody administration group Dose (mg/kg) 3 10 50 TGI (%) 44.9 ⁇ 3.8 57.0 ⁇ 3.5 66.0 ⁇ 3.5
  • the proportion of cells expressing IGSF1 in the Choi-CK cell line was found to be about 57.5%.
  • Example 3.2 Confirmation of changes in cytokine secretion by co-culture of human biliary tract cancer cell lines and peripheral blood cells
  • Example 3.3 Confirmation of anticancer activity of WM-A1-3389 antibody in xenograft biliary tract cancer cell line transplantation mouse model
  • the anticancer activity of the WM-A1-3389 antibody was confirmed in a biliary tract cancer mouse model transplanted with a human biliary tract cancer cell line.
  • mice 6-week-old female peripheral blood mononuclear cell humanized mice (Gem biosciences) were purchased and acclimatized for 1 week, and then Choi-CK cells, a human biliary tract cancer cell line, were diluted in PBS to a concentration of 5 ⁇ 10 6 cells/mice. It was injected subcutaneously (100 ⁇ l) on the dorsal side. Human IgG isotype was used as a negative control for the WM-A1-3389 antibody. When the tumor size reached an average of 80 mm 3 to 100 mm 3 , human IgG isotype (negative control) was administered intraperitoneally at a dose of 10 mg/kg or WM-A1-3389 antibody at a dose of 30 mg/kg. Administration was conducted once every three days for three weeks, and the tumor size and body weight of the mice were measured twice a week. On the 22nd day of administration, the experimental animals were euthanized and the tumors were extracted and weighed.
  • Choi-CK cells a human bil
  • the WM-A1-3389 antibody administration group showed higher tumor growth inhibition efficacy than the negative control group.
  • TGI tumor growth inhibition rate
  • TGI tumor growth inhibition
  • the proportion of cells expressing IGSF1 in the FaDu cell line was found to be about 37.7%.
  • Example 4.2 Confirmation of changes in cytokine secretion by co-culture of human head and neck cancer cell lines and peripheral blood cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to a pharmaceutical composition for treating colon cancer, biliary tract cancer, or head and neck cancer, comprising an anti-IGSF1 antibody as an active ingredient. When the anti-IGSF1 antibody according to the present invention was treated while co-culturing colon cancer cells, biliary tract cancer cells, or head and neck cancer cells with immune cells, the secretion of Granzyme B, and cytokines IFN-γ and TNF-α was increased. It was confirmed that the growth of colon cancer or biliary tract cancer was inhibited when the antibody was administered to a xenogeneic or allogeneic colon cancer or biliary tract cancer implantation mouse model. Therefore, the anti-IGSF1 antibody of the present invention can be helpful in the treatment of colon cancer, biliary tract cancer, and head and neck cancer.

Description

항-IGSF1 항체를 포함하는 암 치료용 약학 조성물Pharmaceutical composition for cancer treatment comprising anti-IRGSF1 antibody
본 발명은 항-IGSF1 항체 또는 이의 단편을 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물에 관한 것이다.The present invention relates to a pharmaceutical composition for preventing or treating cancer comprising an anti-IGSF1 antibody or fragment thereof as an active ingredient.
암은 인류의 건강을 위협하는 최대의 질병 중의 하나로서, 세포가 일련의 돌연변이 과정을 거쳐, 무제한적이고 비조절적인 방식으로 증식하고 불사화되어 발생하는 질병이다. 암 발생의 원인으로는 화학물질, 바이러스, 세균, 전리방사선 등의 환경적 또는 외적 요인과 선천성 유전자 변이 등의 내적 요인을 들 수 있다. 초기에 발견된 암일 경우 수술, 방사선 치료, 화학적 요법 등의 치료법이 있으나 그 부작용이 큰 문제로 대두되고 있으며, 말기 암이나 전이된 암의 경우 특별한 치료법 없이 시한부 인생으로 삶을 마감하는 상황이다.Cancer is one of the greatest diseases threatening human health. It is a disease that occurs when cells undergo a series of mutations, proliferate in an unlimited and uncontrolled manner, and become immortal. Causes of cancer include environmental or external factors such as chemicals, viruses, bacteria, and ionizing radiation, and internal factors such as congenital genetic mutations. For cancer discovered in the early stages, there are treatments such as surgery, radiation therapy, and chemotherapy, but their side effects are becoming a major problem, and for terminal cancer or metastatic cancer, life is terminal and ends without any special treatment.
이러한 암 중에서 대장암의 경우, FOLFIRI(Folinic acid, Fluorouracil 및 Irinotecan), FOLFOX(Folinic acid, Fluorouracil 및 Oxaliplatin), 얼비툭스(Erbitux), 벡티빅스(Vectibix) 등과 같은 표피성장인자 수용체(EGFR, epidermal growth factor receptor) 표적 항암 항체 치료제와, 아바스틴(Avastin) 혹은 잘트랩(Zaltrap)과 같은 신생혈관 형성 억제제가 항암 치료제로 주로 사용되고 있다. 하지만 처방을 받은 환자에서의 치료 반응율은 약 20%로 매우 낮거나, 치료 효과를 나타낸 환자들의 3년 내 재발률이 높고, 약물에 대한 반응성이 없거나 치료 후 저항성이 발생하는 등 한계적 효과를 나타내고 있다.Among these cancers, in the case of colon cancer, epidermal growth factor receptor (EGFR) such as FOLFIRI (Folinic acid, Fluorouracil and Irinotecan), FOLFOX (Folinic acid, Fluorouracil and Oxaliplatin), Erbitux, Vectibix, etc. Receptor-targeted anticancer antibody treatments and angiogenesis inhibitors such as Avastin or Zaltrap are mainly used as anticancer treatments. However, the treatment response rate in patients who have received a prescription is very low at about 20%, or the recurrence rate within 3 years is high in patients who have shown treatment effects, and the drug is showing limited effectiveness, such as lack of response to the drug or resistance occurring after treatment. .
담도암은 높은 재발률 때문에 외과적 절제술이 효과적이지 않고 보통의 화학요법이나 방사선요법이 잘 듣지 않는다는 특징을 가진다. 담도암은 치료에는 1차 치료로서 젬시타빈(Gemcitabine), 카페시타빈(Capecitabine) 또는 5-플루오로우라실(5-Fluorouracil: 5-FU)을 단일 작용제로서 사용하거나 옥살리플라틴(Oxaliplatin), 시스플라틴(Cisplatin) 등과 같은 백금 화합물과 조합하여 사용하거나, 또는 젬시타빈 및 카페시타빈을 조합하여 사용하는 것을 권장하고 있다. 하지만, 다중-작용제 화학요법의 경우 전이성 및/또는 재발성 담도암 환자에서는 지속적인 효과를 나타내지 못하고 있고, 진행성 담도암의 경우 진단 후 5년 생존율이 5% 미만에 달한다.Biliary tract cancer has the characteristic that surgical resection is not effective due to its high recurrence rate and that it does not respond well to conventional chemotherapy or radiotherapy. Biliary tract cancer is treated as a first-line treatment with Gemcitabine, Capecitabine, or 5-Fluorouracil (5-FU) as single agents, or with Oxaliplatin or Cisplatin. It is recommended to use it in combination with platinum compounds such as ), or in combination with gemcitabine and capecitabine. However, multi-agent chemotherapy does not show lasting effects in patients with metastatic and/or recurrent biliary tract cancer, and in the case of advanced biliary tract cancer, the 5-year survival rate after diagnosis is less than 5%.
또한, 두경부암의 경우 수술, 방사선치료 및 항암화학요법을 주로 사용한다. 이 중, 대표적인 두경부암 치료를 위한 항암제인 시스플라틴(Cisplatin)은 뛰어난 항암효과를 가지고 있음에도 불구하고 시스플라틴 내성을 가진 환자들 혹은 치료과정에서 내성을 가지게 되는 환자의 경우 내성 극복을 위한 문제 해결이 필요한 실정이다.Additionally, for head and neck cancer, surgery, radiation therapy, and chemotherapy are mainly used. Among these, cisplatin, a representative anticancer drug for the treatment of head and neck cancer, has excellent anticancer effects, but in the case of patients with cisplatin resistance or patients who develop resistance during the treatment process, problems to overcome resistance are needed. am.
이에 본 발명자들은 암을 효과적으로 치료하는 방법을 연구한 결과, 항-IGSF1 항체 또는 이의 단편의 투여로 인해 종양 성장이 억제되는 것을 확인함으로써 본 발명을 완성하였다.Accordingly, the present inventors studied a method for effectively treating cancer and completed the present invention by confirming that tumor growth was inhibited by administration of an anti-IGSF1 antibody or fragment thereof.
상기 과제를 해결하기 위하여, 본 발명의 일 측면은, 항-IGSF1 항체 또는 이의 단편을 유효성분으로 포함하는 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암의 예방 또는 치료용 약학 조성물을 제공한다.In order to solve the above problem, one aspect of the present invention is to prevent or treat any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer, comprising an anti-IGSF1 antibody or fragment thereof as an active ingredient. A pharmaceutical composition is provided.
본 발명의 다른 측면은 항-IGSF1 항체 또는 이의 단편을 개체에게 투여하는 단계를 포함하는 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암에 대한 예방 또는 치료 방법을 제공한다.Another aspect of the present invention provides a method for preventing or treating any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer, comprising administering an anti-IGSF1 antibody or fragment thereof to a subject.
본 발명의 또 다른 측면은 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암의 예방 또는 치료에의 이용을 위한 항-IGSF1 항체 또는 이의 단편을 제공한다.Another aspect of the present invention provides an anti-IGSF1 antibody or fragment thereof for use in the prevention or treatment of any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer.
본 발명의 또 다른 측면은 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암을 예방 또는 치료하기 위한 의약의 제조를 위한 항-IGSF1 항체 또는 이의 단편의 용도를 제공한다.Another aspect of the present invention provides the use of an anti-IGSF1 antibody or fragment thereof for the manufacture of a medicament for preventing or treating any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer.
본 발명에 의한 항-IGSF1 항체를 대장암 세포, 담도암 세포 또는 두경부암 세포와 면역세포를 공동배양 시 처리한 경우, Granzyme B, 및 사이토카인인 IFN-γ 및 TNF-α의 분비가 증가하였다. 상기 항체를 이종 또는 동종의 대장암 또는 담도암 식립 마우스 모델에 투여 시 대장암 또는 담도암의 성장이 억제되는 것을 확인하였다. 따라서, 본 발명의 항-IGSF1 항체는 대장암, 담도암 및 두경부암 치료에 유용하게 사용될 수 있다.When the anti-IGSF1 antibody according to the present invention was treated with colon cancer cells, biliary tract cancer cells, or head and neck cancer cells and immune cells, the secretion of Granzyme B and the cytokines IFN-γ and TNF-α increased. . It was confirmed that the growth of colon cancer or biliary tract cancer was inhibited when the antibody was administered to a xenograft or allogeneic colon cancer or biliary tract cancer implanted mouse model. Therefore, the anti-IGSF1 antibody of the present invention can be usefully used in the treatment of colon cancer, biliary tract cancer, and head and neck cancer.
도 1은 인간 대장암 세포주 HT29 세포에서 IGSF1의 발현을 유세포 분석을 통해 확인한 결과를 나타낸 도면이다.Figure 1 is a diagram showing the results of confirming the expression of IGSF1 in human colon cancer cell line HT29 cells through flow cytometry.
도 2는 HT29 세포와 인간 말초혈액세포(PBMC)의 공동배양 시스템에서 WM-A1-3389 항체 처리에 의한 Granzyme B, IFN-γ 및 TNF-α 분비량의 변화를 확인한 결과를 나타낸 그래프이다.Figure 2 is a graph showing the results of confirming changes in the secretion amount of Granzyme B, IFN-γ, and TNF-α by treatment with WM-A1-3389 antibody in a co-culture system of HT29 cells and human peripheral blood cells (PBMC).
도 3은 인간 대장암 세포주 HT29 식립 마우스 모델에서 WM-A1-3389 항체 투여에 의한 종양 성장 정도를 측정한 결과를 나타낸 그래프이다.Figure 3 is a graph showing the results of measuring the degree of tumor growth by administration of WM-A1-3389 antibody in a mouse model implanted with the human colon cancer cell line HT29.
도 4는 마우스 대장암 세포주 MC38에서 IGSF1의 발현을 면역형광염색(상단) 및 유세포 분석(하단)을 통해 확인한 결과를 나타낸 도면이다.Figure 4 is a diagram showing the results of confirming the expression of IGSF1 in the mouse colon cancer cell line MC38 through immunofluorescence staining (top) and flow cytometry (bottom).
도 5는 마우스 대장암 세포주 MC38 식립 마우스 모델에서 WM-A1-3389 항체 투여에 의한 종양 성장 정도를 측정한 결과를 나타낸 그래프이다.Figure 5 is a graph showing the results of measuring the degree of tumor growth by administration of WM-A1-3389 antibody in a mouse model implanted with the mouse colon cancer cell line MC38.
도 6은 마우스 대장암 세포주 CT26에서 IGSF1의 발현을 면역형광염색(상단) 및 유세포 분석(하단)을 통해 확인한 결과를 나타낸 도면이다.Figure 6 is a diagram showing the results of confirming the expression of IGSF1 in the mouse colon cancer cell line CT26 through immunofluorescence staining (top) and flow cytometry (bottom).
도 7은 마우스 대장암 세포주 CT26 식립 마우스 모델에서 WM-A1-3389 항체 투여에 의한 종양 성장 정도를 측정한 결과를 나타낸 그래프이다.Figure 7 is a graph showing the results of measuring the degree of tumor growth by administration of WM-A1-3389 antibody in a mouse model implanted with the mouse colon cancer cell line CT26.
도 8은 인간 담도암 세포주 Choi-CK 세포에서 IGSF1의 발현을 유세포 분석을 통해 확인한 결과를 나타낸 도면이다.Figure 8 is a diagram showing the results of confirming the expression of IGSF1 in human biliary tract cancer cell line Choi-CK cells through flow cytometry.
도 9는 Choi-CK 세포와 인간 말초혈액세포(hPBMC)의 공동배양 시스템에서 WM-A1-3389 항체 처리에 의한 Granzyme B, IFN-γ 및 TNF-α 분비량의 변화를 확인한 결과를 나타낸 그래프이다.Figure 9 is a graph showing the results of confirming changes in the secretion amount of Granzyme B, IFN-γ, and TNF-α by treatment with WM-A1-3389 antibody in a co-culture system of Choi-CK cells and human peripheral blood cells (hPBMC).
도 10은 인간 담도암 세포주 Choi-CK 식립 마우스 모델에서 WM-A1-3389 항체 투여에 의한 종양 성장 정도를 측정한 결과를 나타낸 그래프이다.Figure 10 is a graph showing the results of measuring the degree of tumor growth by administration of WM-A1-3389 antibody in a mouse model implanted with the human biliary tract cancer cell line Choi-CK.
도 11은 인간 두경부암 세포주 FaDu 세포에서 IGSF1의 발현을 유세포 분석을 통해 확인한 결과를 나타낸 도면이다.Figure 11 is a diagram showing the results of confirming the expression of IGSF1 in the human head and neck cancer cell line FaDu cells through flow cytometry.
도 12는 FaDu 세포와 인간 말초혈액세포(hPBMC) 공동배양 시스템에서 WM-A1-3389 항체 처리에 의한 Granzyme B, IFN-γ 및 TNF-α 분비량의 변화를 확인한 결과를 나타낸 그래프이다.Figure 12 is a graph showing the results confirming changes in the secretion amounts of Granzyme B, IFN-γ, and TNF-α by treatment with WM-A1-3389 antibody in the FaDu cell and human peripheral blood cell (hPBMC) co-culture system.
항-IGSF1 항체anti-IGSF1 antibody
본 명세서에서 사용하는 용어, "IGSF1"은 인간 및 다른 포유류 종의 X 염색체에서 발견되는 IGSF1 유전자에 의해 암호화된 막단백질이다. 정상세포에서 IGSF1의 기능에 대해서는 잘 알려져 있지 않으나, IGSF1 돌연변이는 IGSF1 결핍 증후군(IGSF1 deficiency syndrome) 또는 중추성 갑상선 기능 저하증 등의 질병을 유발하는 것으로 알려져 있다.As used herein, the term “IGSF1” is a membrane protein encoded by the IGSF1 gene found on the X chromosome of humans and other mammalian species. Although the function of IGSF1 in normal cells is not well known, IGSF1 mutations are known to cause diseases such as IGSF1 deficiency syndrome or central hypothyroidism.
본 발명에서 상기 IGSF1은 포유류의 IGSF1이라면 제한없이 포함될 수 있으나, 바람직하게는 인간 IGSF1을 의미할 수 있다. 또한, 본 발명에서 상기 IGSF1 단백질은 천연형 또는 변이체 IGSF1 단백질을 모두 포함하나, 이에 제한되지 않는다. 상기 천연형 IGSF1 단백질이란 천연형 IGSF1 단백질의 아미노산 서열을 포함하는 폴리펩타이드를 일반적으로 지칭하며, 상기 천연형 IGSF1 단백질의 아미노산 서열이란 천연 발생 IGSF1에서 발견되는 아미노산 서열을 일반적으로 지칭한다. 상기 IGSF1에 대한 정보는 미국국립보건원의 GenBank 등 공지의 데이터베이스로부터 얻을 수 있으며, 예를 들어 Genbank accession number NP_001164433.1의 아미노산 서열(서열번호 19)을 가질 수 있으나, 이에 제한되지 않는다. In the present invention, the IGSF1 may be included without limitation as long as it is mammalian IGSF1, but preferably refers to human IGSF1. Additionally, in the present invention, the IGSF1 protein includes, but is not limited to, both native and mutant IGSF1 proteins. The native IGSF1 protein generally refers to a polypeptide containing the amino acid sequence of the native IGSF1 protein, and the amino acid sequence of the native IGSF1 protein generally refers to the amino acid sequence found in naturally occurring IGSF1. Information about IGSF1 can be obtained from known databases such as GenBank of the National Institutes of Health, and may have, for example, an amino acid sequence (SEQ ID NO: 19) with Genbank accession number NP_001164433.1, but is not limited thereto.
본 명세서에서 사용하는 용어, "항체"는 특정 항원과 면역학적으로 반응하는 면역글로불린 분자로, 항원을 특이적으로 인식하는 단백질 분자를 의미한다. 상기 항체는 전체(whole) 항체, 단일클론항체, 다클론항체, 단일 도메인 항체, 단쇄 항체, 다중특이적 항체, 인간 항체, 인간화 항체, 키메라 항체, 인트라바디(intrabody), scFv, Fab 단편, F(ab') 단편, 이황화 결합으로 연결한 Fv(sdFv) 및 상기 중 임의의 에피토프(epitope) 결합 단편을 포함하지만, 이에 제한되지 않는다. As used herein, the term “antibody” refers to an immunoglobulin molecule that reacts immunologically with a specific antigen, and refers to a protein molecule that specifically recognizes the antigen. The antibodies include whole antibodies, monoclonal antibodies, polyclonal antibodies, single domain antibodies, single chain antibodies, multispecific antibodies, human antibodies, humanized antibodies, chimeric antibodies, intrabodies, scFvs, Fab fragments, F (ab') fragment, disulfide bond-linked Fv (sdFv), and epitope-binding fragments of any of the above.
본 명세서에서 사용하는 용어, "항-IGSF1 항체"는 IGSF1에 특이적으로 결합할 수 있는 항체를 의미하며, 본 발명에서 "IGSF1에 특이적인 항체"와 혼용되어 사용될 수 있다. 특히, 항-IGSF1 항체는 IGSF1의 C 말단에 특이적으로 결합할 수 있다. 상기 항체의 형태는 전체(whole) 항체 및 이의 항체 단편을 모두 포함할 수 있다.As used herein, the term “anti-IGSF1 antibody” refers to an antibody capable of specifically binding to IGSF1, and may be used interchangeably with “IGSF1-specific antibody” in the present invention. In particular, anti-IGSF1 antibodies can specifically bind to the C terminus of IGSF1. The form of the antibody may include both whole antibodies and antibody fragments thereof.
면역글로불린의 중쇄 및 경쇄는 각각 불변영역(constant region) 및 가변영역(variable region)을 포함할 수 있다. 면역글로불린의 경쇄 및 중쇄 가변영역은, 상보성 결정 영역(complementarity determining region, CDR)이라 불리는 3개의 다변가능한 영역 및 4개의 구조영역(framework region, FR)을 포함한다. 상기 CDR은 주로 항원의 에피토프에 결합하는 역할을 한다. 각각의 사슬의 CDR은 전형적으로 N 말단으로부터 시작하여 순차적으로 CDR1, CDR2, CDR3로 불리고, 특정 CDR이 위치하고 있는 사슬에 의해서 식별된다. The heavy and light chains of immunoglobulins may each include a constant region and a variable region. The light and heavy chain variable regions of immunoglobulins include three variable regions called complementarity determining regions (CDRs) and four framework regions (FRs). The CDR mainly functions to bind to the epitope of the antigen. The CDRs of each chain are typically called CDR1, CDR2, and CDR3 sequentially, starting from the N terminus, and are identified by the chain on which the specific CDR is located.
본 발명의 항-IGSF1 항체 또는 이의 단편은 서열번호 1의 아미노산 서열을 포함하는 H-CDR1, 서열번호 2의 아미노산 서열을 포함하는 H-CDR2 및 서열번호 3의 아미노산 서열을 포함하는 H-CDR3을 포함하는 중쇄 가변영역(VH)을 포함할 수 있다. 또한, 본 발명의 항-IGSF1 항체 또는 이의 단편은 서열번호 4의 아미노산 서열을 포함하는 L-CDR1, 서열번호 5의 아미노산 서열을 포함하는 L-CDR2 및 서열번호 6의 아미노산 서열을 포함하는 L-CDR3을 포함하는 경쇄 가변영역(VL)을 포함할 수 있다. 이때, 상기 중쇄 가변영역은 서열번호 7의 아미노산 서열을 포함할 수 있으며, 상기 경쇄 가변영역은 서열번호 8의 아미노산 서열을 포함할 수 있다. 본 명세서에서 상기 항체는 WM-A1-3389로 지칭될 수 있다.The anti-IGSF1 antibody or fragment thereof of the present invention includes H-CDR1 containing the amino acid sequence of SEQ ID NO: 1, H-CDR2 containing the amino acid sequence of SEQ ID NO: 2, and H-CDR3 containing the amino acid sequence of SEQ ID NO: 3. It may include a heavy chain variable region (VH). In addition, the anti-IGSF1 antibody or fragment thereof of the present invention includes L-CDR1 containing the amino acid sequence of SEQ ID NO: 4, L-CDR2 containing the amino acid sequence of SEQ ID NO: 5, and L- CDR2 containing the amino acid sequence of SEQ ID NO: 6. It may include a light chain variable region (VL) including CDR3. At this time, the heavy chain variable region may include the amino acid sequence of SEQ ID NO: 7, and the light chain variable region may include the amino acid sequence of SEQ ID NO: 8. In this specification, the antibody may be referred to as WM-A1-3389.
상기 항체의 중쇄 가변영역은 서열번호 7의 아미노산 서열과 약 90% 이상, 약 91% 이상, 약 92% 이상, 약 93% 이상, 약 94% 이상, 약 95% 이상, 약 96% 이상, 약 97% 이상, 약 98% 이상, 약 99% 이상, 또는 100%의 동일성을 갖는 아미노산 서열을 포함하거나 이들로 이루어질 수 있다. 또한, 상기 항체의 경쇄 가변영역은 서열번호 8의 아미노산 서열과 약 90% 이상, 약 91% 이상, 약 92% 이상, 약 93% 이상, 약 94% 이상, 약 95% 이상, 약 96% 이상, 약 97% 이상, 약 98% 이상, 약 99% 이상, 또는 100%의 동일성을 갖는 아미노산 서열을 포함하거나 이들로 이루어질 수 있다.The heavy chain variable region of the antibody consists of the amino acid sequence of SEQ ID NO: 7 and about 90% or more, about 91% or more, about 92% or more, about 93% or more, about 94% or more, about 95% or more, about 96% or more, about It may comprise or consist of an amino acid sequence having at least 97%, at least about 98%, at least about 99%, or 100% identity. In addition, the light chain variable region of the antibody is about 90% or more, about 91% or more, about 92% or more, about 93% or more, about 94% or more, about 95% or more, and about 96% or more of the amino acid sequence of SEQ ID NO: 8. , may include or consist of an amino acid sequence having an identity of at least about 97%, at least about 98%, at least about 99%, or 100%.
면역글로불린 중쇄 불변영역(CH)은 상이한 아미노산 조성 및 순서를 나타내므로, 상이한 유형의 항원성을 보유한다. 따라서, 면역글로불린은 다섯 가지 카테고리로 분류될 수 있으며, 면역글로불린 이소형, 즉, IgM, IgD, IgG, IgA 및 IgE로 지칭될 수 있다. 이에 상응하는 중쇄는 각각 μ 사슬, δ 사슬, γ 사슬, α 사슬 및 ε 사슬이다. 또한, 힌지 영역(hinge region)의 아미노산 조성 및 중쇄 이황화 결합의 수와 위치에 따라, 동일한 유형의 Ig는 상이한 하위 유형으로 분류될 수 있다. 예를 들어, IgG는 IgG1, IgG2, IgG3, 및 IgG4로 분류될 수 있다. 경쇄는 상이한 불변영역에 따라 κ 또는 λ 사슬로 분류될 수 있다. 다섯 가지 유형의 IgG 각각은 κ 또는 λ 사슬을 가질 수 있다.Immunoglobulin heavy chain constant regions (CH) exhibit different amino acid compositions and sequences and therefore possess different types of antigenicity. Accordingly, immunoglobulins can be divided into five categories and referred to as immunoglobulin isotypes, namely IgM, IgD, IgG, IgA and IgE. The corresponding heavy chains are μ chain, δ chain, γ chain, α chain and ε chain, respectively. Additionally, depending on the amino acid composition of the hinge region and the number and position of heavy chain disulfide bonds, the same type of Ig can be classified into different subtypes. For example, IgG can be classified as IgG1, IgG2, IgG3, and IgG4. Light chains can be classified as κ or λ chains depending on their different constant regions. Each of the five types of IgG can have either a κ or λ chain.
본 발명의 항-IGSF1 항체 또는 이의 단편이 불변영역을 포함하는 경우, IgG, IgA, IgD, IgE, IgM 유래, 또는 이들이 부분적으로 혼합(hybrid)된 불변영역을 포함할 수 있다.When the anti-IGSF1 antibody or fragment thereof of the present invention includes a constant region, it may include a constant region derived from IgG, IgA, IgD, IgE, IgM, or a partial hybrid thereof.
본 명세서에서 사용하는 용어, "혼합된(hybrid)"은 단쇄 면역 글로불린 중쇄 불변영역 내에 2개 이상의 상이한 기원의 면역글로불린 중쇄 불변영역에 해당하는 서열이 존재함을 의미한다. 예를 들어 IgG, IgA, IgD, IgE 및 IgM의 CH1, CH2 및 CH3으로 이루어진 그룹으로부터 선택되는 1개 내지 4개 도메인으로 이루어진 도메인의 하이브리드가 가능하다.As used herein, the term “hybrid” means that sequences corresponding to immunoglobulin heavy chain constant regions of two or more different origins exist within the single-chain immunoglobulin heavy chain constant region. For example, a domain hybrid consisting of 1 to 4 domains selected from the group consisting of CH1, CH2, and CH3 of IgG, IgA, IgD, IgE, and IgM is possible.
또한, 본 발명의 상기 항-IGSF1 항체 또는 이의 단편이 경쇄 불변영역(LC)을 포함하는 경우, 상기 경쇄 불변영역은 λ 또는 κ 경쇄 유래일 수 있다. Additionally, when the anti-IGSF1 antibody or fragment thereof of the present invention includes a light chain constant region (LC), the light chain constant region may be derived from a λ or κ light chain.
본 명세서에서 사용하는 용어, "항체 단편"은 항원-결합 활성을 갖는 Fab 단편, Fab' 단편, F(ab')2 단편 뿐만 아니라, IGSF1에 결합하는 Fv 단편인 scFv 단편을 지칭하며, 본 발명에 기술된 항체의 CDR 영역을 포함한다. Fv 단편은 불변 영역 없이, 중쇄 가변 영역 및 경쇄 가변 영역을 포함하며, 모든 항원-결합 자리를 보유하는 최소 항체 단편이다.As used herein, the term “antibody fragment” refers to an scFv fragment, which is an Fv fragment that binds to IGSF1, as well as a Fab fragment, Fab' fragment, and F(ab') 2 fragment having antigen-binding activity, and is used in the present invention. Contains the CDR regions of the antibodies described in. The Fv fragment is the smallest antibody fragment that contains the heavy and light chain variable regions, without constant regions, and retains all antigen-binding sites.
약학 조성물pharmaceutical composition
본 발명의 항-IGSF1 항체 또는 이의 단편을 유효성분으로 포함하는 약학 조성물은 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암에 대하여 예방 또는 치료 효능을 나타낸다.The pharmaceutical composition containing the anti-IGSF1 antibody or fragment thereof of the present invention as an active ingredient exhibits preventive or therapeutic efficacy against any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer.
이때, 상기 대장암, 담도암 및 두경부암은 IGSF1이 과발현되어 있는 암일 수 있다. At this time, the colon cancer, biliary tract cancer, and head and neck cancer may be cancers in which IGSF1 is overexpressed.
본 발명에서 사용하는 용어, "대장암"은 대장(맹장, 충수, 결장, 직장 또는 항문관)에 생긴 암세포로 이루어진 악성종양을 의미한다. 대장암은 병리학적으로는 대부분이 선암(adenocarcinoma)이며, 이외에도 림프종, 육종, 편평상피암, 다른 암의 전이성 병변 등이 포함된다. 부위별로는 크게 결장암과 직장암으로 구분되며, 하부 대장, 즉 직장에서 발생하는 경우가 약 50%로 가장 높다. 대장암의 발병 원인은 아직 불분명하지만 유전적 요인, 고지방 및 저섬유성 음식의 섭취와 관련된 식이습관 및 염증성 장질환 등이 고려되고 있다. 대장암은 모든 연령층에서 발생할 수 있으나, 연령이 증가함에 따라 발생빈도가 높아지며 50대 내지 60대에서 자주 발생한다. 남녀의 발생 비는 결장암은 여자, 직장암은 남자에서 다소 높게 나타난다. The term “colorectal cancer” used in the present invention refers to a malignant tumor composed of cancer cells that arise in the large intestine (cecum, appendix, colon, rectum, or anal canal). Pathologically, colon cancer is mostly adenocarcinoma, but it also includes lymphoma, sarcoma, squamous cell carcinoma, and metastatic lesions of other cancers. By site, it is largely divided into colon cancer and rectal cancer, with the highest incidence of cancer occurring in the lower colon, or rectum, at about 50%. The cause of colon cancer is still unclear, but genetic factors, dietary habits related to the consumption of high-fat and low-fiber foods, and inflammatory bowel disease are being considered. Colon cancer can occur at any age, but the incidence increases with age and occurs frequently in people in their 50s or 60s. The incidence ratio between men and women is somewhat higher for colon cancer in women and for rectal cancer in men.
본 명세서에서 사용하는 용어, "담도암"은 담관암이라고도 불리우며, 담관에서 발생하는 암을 일컫는다. 담관은 간에서 만들어지는 담즙을 십이지장으로 보내는 관으로, 담관에서 발생한 암은 해부학적 위치에 따라 간내 담관암(약 20% 내지 약 25%), 간문부 담관암(약 50% 내지 약 60%) 및 원위부 담관암(약 20% 내지 약 25%)으로 구분할 수 있다. 담관세포에 발생한 선암종이 담관암의 거의 대부분을 차지한다. 담관암의 원인은 아직 확실히 밝혀지지 않았으며, 담관 내부를 이루고 있는 담관세포에 생긴 만성적인 염증, 담관 결석, 경화성 담관염, 간디스토마증(간흡충증), 염증성 대장 질환 및 담관이 선천적으로 확장되어 생긴 담관 낭종 등과 관련이 있다고 알려져 있다.The term “biliary tract cancer” used in this specification is also called cholangiocarcinoma and refers to cancer that occurs in the bile ducts. The bile duct is a tube that transports bile produced in the liver to the duodenum. Cancers originating in the bile duct are divided into intrahepatic bile duct cancer (about 20% to about 25%), hilar bile duct cancer (about 50% to about 60%), and distal liver cancer, depending on the anatomical location. It can be classified into bile duct cancer (about 20% to about 25%). Adenocarcinoma arising from cholangiocytes accounts for the majority of cholangiocarcinomas. The cause of cholangiocarcinoma is not yet clearly known, and it is caused by chronic inflammation in the bile duct cells lining the bile ducts, bile duct stones, sclerosing cholangitis, liver dystomatosis, inflammatory bowel disease, and bile duct cysts caused by congenital dilatation of the bile ducts. It is known to be related to etc.
본 명세서에서 사용하는 용어, "두경부암"은 두경부암은 뇌와 안구에 발생하는 종양을 제외한 얼굴, 코, 목, 입안, 후두, 인두, 침샘 및 갑상선에 발생하는 악성 종양을 말한다. 두경부암 중 구강에 생기는 암을 구강암이라고 하고, 소리를 내는 기관인 후두에 생기는 암을 후두암이라고 하며, 인두에 생기는 암을 인두암이라고 한다. 상기 인두암은 발생 위치에 따라 비인두암, 구인두암 및 하인두암으로 나뉜다. 또한, 두경부암은 경부 식도암, 침샘암, 타액선암, 악성 림프종, 악성 흑색종 및 각종 연부조직 암을 포함하며, 갑상선암은 포괄적 의미의 두경부암에 포함된다. 두경부암의 전통적인 원인으로 흡연 및 음주가 알려져 있으며, 비인두암의 경우 바이러스 감염(Epstein barr virus)과 밀접한 관련이 있는 것으로 알려져 있다. 그 외에도 위액 역류성 질환, 식도 질환, 방사선 또는 자외선, 비타민 또는 철의 결핍 등이 두경부암의 발생을 증가시키는 요인으로 알려져 있다.As used herein, the term "head and neck cancer" refers to malignant tumors that occur in the face, nose, throat, mouth, larynx, pharynx, salivary glands, and thyroid gland, excluding tumors that occur in the brain and eyes. Among head and neck cancers, cancer that occurs in the oral cavity is called oral cancer, cancer that occurs in the larynx, the organ that produces sound, is called laryngeal cancer, and cancer that occurs in the pharynx is called pharyngeal cancer. The pharyngeal cancer is divided into nasopharyngeal cancer, oropharyngeal cancer, and hypopharyngeal cancer depending on the location of occurrence. In addition, head and neck cancer includes cervical esophageal cancer, salivary gland cancer, salivary gland cancer, malignant lymphoma, malignant melanoma, and various soft tissue cancers, and thyroid cancer is included in head and neck cancer in a comprehensive sense. Smoking and drinking are known to be traditional causes of head and neck cancer, and nasopharyngeal cancer is known to be closely related to viral infection (Epstein Barr virus). In addition, gastric acid reflux disease, esophageal disease, radiation or ultraviolet rays, vitamin or iron deficiency, etc. are known to be factors that increase the incidence of head and neck cancer.
본 명세서에서 사용하는 용어, "예방"은 상기 약학적 조성물의 투여에 의해 질환의 발생을 억제하거나 그의 발병을 지연시키는 모든 행위를 말한다. 상기 용어, "치료"는 상기 약학적 조성물의 투여에 의해 질환의 증세가 호전되거나 이롭게 변경하는 모든 행위를 말한다.As used herein, the term “prevention” refers to all actions that suppress or delay the onset of a disease by administering the pharmaceutical composition. The term “treatment” refers to any action that improves or beneficially changes the symptoms of a disease by administering the pharmaceutical composition.
본 발명의 약학 조성물에서 상기 항-IGSF1 항체 또는 이의 단편은 항암 활성을 나타낼 수 있는 한, 용도, 제형, 배합 목적 등에 따라 임의의 양(유효량)으로 포함될 수 있다. 여기서 "유효량"이란 항암 효과를 유도할 수 있는 유효성분의 양을 말한다. 이러한 유효량은 당업자의 통상의 능력 범위 내에서 실험적으로 결정될 수 있다. 본 발명의 약학 조성물은 유효성분으로서 상기 항체를 조성물의 총 중량을 기준으로 약 0.1 중량% 내지 약 90 중량%, 구체적으로 약 0.5 중량% 내지 약 75 중량%, 보다 구체적으로 약 1 중량% 내지 약 50 중량%로 함유할 수 있다.In the pharmaceutical composition of the present invention, the anti-IGSF1 antibody or fragment thereof may be included in any amount (effective amount) depending on use, formulation, formulation purpose, etc., as long as it can exhibit anti-cancer activity. Here, “effective amount” refers to the amount of active ingredient that can induce an anticancer effect. Such effective amounts can be determined experimentally within the scope of the ordinary ability of those skilled in the art. The pharmaceutical composition of the present invention contains the antibody as an active ingredient in an amount of about 0.1% to about 90% by weight, specifically about 0.5% by weight to about 75% by weight, and more specifically about 1% by weight to about 1% by weight, based on the total weight of the composition. It can be contained at 50% by weight.
생체이용률과 같은 약동학적 파라미터(pharmacokinetic parameters) 및 클리어런스율(clearance rate)과 같은 기본적인 파라미터(underlying parameters)도 효능에 영향을 줄 수 있다. 따라서, "향상된 효능"(예를 들어, 효능의 개선)은 향상된 약동학적 파라미터 및 향상된 효능에 기인할 수 있으며, 시험 동물 또는 인간 대상체에서 클리어런스율 및 종양 성장을 비교하거나, 생존, 재발률 또는 질병이 없는 상태에서 생존과 같은 파라미터를 비교하여 측정될 수 있다.Pharmacokinetic parameters such as bioavailability and underlying parameters such as clearance rate may also affect efficacy. Accordingly, “enhanced efficacy” (e.g., improvement in efficacy) may be due to improved pharmacokinetic parameters and improved efficacy, compared to clearance rates and tumor growth in test animals or human subjects, or to survival, recurrence rates, or disease progression. It can be measured by comparing parameters such as survival in the absence of it.
본 명세서에서 사용하는 용어, "효능(efficacy)"은 1년, 5년 또는 10년과 같이 일정 기간에 걸쳐 생존 또는 질병이 없는 상태에서 생존(disease-free survival)과 같은 하나 이상의 파라미터에 의해 결정될 수 있다. 뿐만 아니라, 상기 파라미터는 개체에서 적어도 하나의 종양의 크기가 억제되는 것을 포함할 수 있다.As used herein, the term "efficacy" means survival over a period of time, such as 1 year, 5 years, or 10 years, or disease-free survival, which can be determined by one or more parameters. You can. Additionally, the parameter may include that the size of at least one tumor in the subject is suppressed.
본 발명의 약학 조성물은 통상적인 방법에 따라 제제로 배합되는 통상적이고 무독성인 약학적으로 허용가능한 담체를 포함할 수 있다. The pharmaceutical composition of the present invention may contain a conventional, non-toxic pharmaceutically acceptable carrier that is formulated into a preparation according to a conventional method.
상기 약학적으로 허용 가능한 담체는 환자에게 전달하기에 적절한 비-독성 물질이면 어떠한 담체라도 가능하다. 증류수, 알코올, 지방, 왁스 및 비활성 고체가 담체로 포함될 수 있다. 약물학적으로 허용되는 애쥬번트(완충제, 분산제) 또한 약물학적 조성물에 포함될 수 있다.The pharmaceutically acceptable carrier may be any carrier that is a non-toxic material suitable for delivery to a patient. Distilled water, alcohol, fats, waxes and inert solids may be included as carriers. Pharmacologically acceptable adjuvants (buffers, dispersants) may also be included in the pharmacological composition.
본 명세서에서 사용하는 용어, "약학적으로 허용가능한 담체"는 생물체를 자극하지 않고 투여 화합물의 생물학적 활성 및 특성을 저해하지 않는 담체 또는 희석제를 말한다. 액상 용액으로 제제화되는 조성물에 있어서 허용되는 약학적 담체로는, 멸균 및 생체에 적합한 것으로서, 식염수, 멸균수, 링거액, 완충 식염수, 알부민 주사용액, 덱스트로즈 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 한 성분 이상을 혼합하여 사용할 수 있으며, 필요에 따라 감미제, 용해 보조제, 습윤제, 유화제, 등장화제, 흡수제, 항산화제, 보존제, 활택제, 충전제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다.As used herein, the term “pharmaceutically acceptable carrier” refers to a carrier or diluent that does not irritate living organisms and does not inhibit the biological activity and properties of the administered compound. Acceptable pharmaceutical carriers in compositions formulated as liquid solutions include those that are sterile and biocompatible, such as saline solution, sterile water, Ringer's solution, buffered saline solution, albumin injection solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and One or more of these ingredients can be mixed and used, and other common additives such as sweeteners, solubilizers, wetting agents, emulsifiers, isotonic agents, absorbents, antioxidants, preservatives, lubricants, fillers, buffers, and bacteriostatic agents are added as needed. can do.
본 발명의 조성물은 비경구 투여(예컨대, 근육내, 정맥내 또는 피하 주사)를 위한 다양한 제형으로 제조될 수 있다. 본 발명의 약학 조성물이 비경구용 제형으로 제조될 경우, 적합한 담체와 함께 당업계에 공지된 방법에 따라 주사제, 경피 투여제, 비강 흡입제 및 좌제의 형태로 제제화될 수 있다. 주사용 제제에는 멸균된 수용액제, 비수성용제, 현탁제, 유제, 동결건조 제제 및 좌제가 포함된다. 비수성용제, 현탁제로는 프로필렌글리콜, 폴리에틸렌글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔, 마크로골, 트윈61, 카카오지, 라우린지, 글리세로제라틴 등이 사용될 수 있다. 한편, 주사제에는 용해제, 등장화제, 현탁화제, 유화제, 안정화제, 방부제 등과 같은 종래의 첨가제가 포함될 수 있다.The compositions of the present invention can be prepared in a variety of formulations for parenteral administration (e.g., intramuscular, intravenous, or subcutaneous injection). When the pharmaceutical composition of the present invention is prepared as a parenteral formulation, it can be formulated in the form of injections, transdermal administration, nasal inhalation, and suppositories along with a suitable carrier according to methods known in the art. Injectable preparations include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized preparations, and suppositories. Non-aqueous solvents and suspensions include propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable esters such as ethyl oleate. As a base for suppositories, Withepsol, Macrogol, Tween 61, cacao, laurin, glycerogeratin, etc. can be used. Meanwhile, injectables may contain conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifiers, stabilizers, preservatives, etc.
약제학적 조성물의 제제화와 관련하여서는 당업계에 공지되어 있으며, 구체적으로 문헌[Remington's Pharmaceutical Sciences(19th ed., 1995)] 등을 참조할 수 있다. 상기 문헌은 본 명세서의 일부로서 간주된다.Regarding the formulation of pharmaceutical compositions, it is known in the art, and specifically, references can be made to the literature [Remington's Pharmaceutical Sciences (19th ed., 1995)]. The above documents are considered part of this specification.
본 발명의 약학 조성물은 치료학적으로 유효한 양 또는 약학적으로 유효한 양으로 환자에 투여될 수 있다.The pharmaceutical composition of the present invention can be administered to a patient in a therapeutically effective amount or a pharmaceutically effective amount.
본 명세서에서 사용하는 용어, "투여"는 적절한 방법으로 개체에게 소정의 물질을 도입하는 것을 의미하며, 상기 조성물의 투여 경로는 목적 조직에 도달할 수 있는 한 어떠한 일반적인 경로를 통하여 투여될 수 있다. 복강내 투여, 정맥내 투여, 근육내 투여, 피하 투여, 피내 투여, 국소 투여, 비내 투여, 직장내 투여될 수 있으나, 이에 한정되지는 않는다.As used herein, the term "administration" means introducing a predetermined substance into an individual by an appropriate method, and the composition may be administered through any general route as long as it can reach the target tissue. It may be administered intraperitoneally, intravenously, intramuscularly, subcutaneously, intradermally, locally, intranasally, or rectally, but is not limited thereto.
여기서 "치료학적으로 유효한 양" 또는 "약학적으로 유효한 양"이란 대상 질환을 예방 또는 치료하는데 유효한 화합물 또는 조성물의 양으로서, 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분하며 부작용을 일으키지 않을 정도의 양을 의미한다. 상기 유효량의 수준은 환자의 건강상태, 질환의 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 방법, 투여 시간, 투여 경로 및 배출 비율, 치료 기간, 배합 또는 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다. 일 구체예에서 치료학적으로 유효한 양은 상기 질환을 치료하는데 효과적인 약물의 양을 의미한다.Here, “therapeutically effective amount” or “pharmaceutically effective amount” refers to the amount of a compound or composition effective in preventing or treating the target disease, which is sufficient to treat the disease with a reasonable benefit/risk ratio applicable to medical treatment. This refers to an amount that does not cause side effects. The level of the effective amount is determined by factors including the patient's health status, type and severity of the disease, drug activity, sensitivity to the drug, administration method, administration time, administration route and excretion rate, treatment period, combination or concurrent use of drugs, and It may be determined based on other factors well known in the medical field. In one embodiment, a therapeutically effective amount refers to an amount of drug effective in treating the disease.
구체적으로, 본 발명의 조성물의 투여량은 환자의 나이, 성별, 체중에 따라 달라질 수 있으며, 일반적으로는 체중 kg 당 약 0.1 mg 내지 약 1,000 mg, 또는 약 5 mg 내지 약 200 mg을 매일, 격일 또는 2주 내지 3주 간격으로 투여하거나, 1일 1회 내지 3회로 나누어 투여할 수 있다. 그러나, 투여 경로, 질병의 중증도, 성별, 체중, 연령 등에 따라서 증감될 수 있으므로, 본 발명의 범위는 이에 한정되지 않는다.Specifically, the dosage of the composition of the present invention may vary depending on the patient's age, gender, and weight, and is generally administered from about 0.1 mg to about 1,000 mg per kg of body weight, or from about 5 mg to about 200 mg per kg of body weight every day or every other day. Alternatively, it can be administered at intervals of 2 to 3 weeks, or divided into 1 to 3 times a day. However, since it may increase or decrease depending on the route of administration, severity of disease, gender, weight, age, etc., the scope of the present invention is not limited thereto.
상기 용어 "개체"란 본 발명의 조성물이 적용(처방)될 수 있는 대상을 의미하며, 인간을 포함한 쥐, 생쥐, 기니피그, 햄스터, 토끼, 개, 고양이, 닭(알 포함), 돼지, 원숭이, 염소 등의 포유동물일 수 있다. 바람직하게는, 인간일 수 있으나 이에 제한되지 않는다. The term "subject" refers to an object to which the composition of the present invention can be applied (prescribed), including humans, rats, mice, guinea pigs, hamsters, rabbits, dogs, cats, chickens (including eggs), pigs, monkeys, It may be a mammal such as a goat. Preferably, it may be a human, but is not limited thereto.
본 발명의 약학 조성물은 개별 치료제로 투여하거나 다른 치료제와 병용하여 투여될 수 있고, 종래의 치료제와 순차적으로 또는 동시에 투여될 수 있으며, 단일 또는 다중 투여될 수 있다. 이때, 상기 다른 치료제는 항암 활성의 상승 또는 보강을 위하여 이미 안전성이 검증되고 항암 활성을 갖는 것으로 공지된 임의의 화합물이나 천연 추출물을 추가로 포함할 수 있다. The pharmaceutical composition of the present invention may be administered as an individual therapeutic agent or in combination with other therapeutic agents, may be administered sequentially or simultaneously with conventional therapeutic agents, and may be administered singly or multiple times. At this time, the other therapeutic agent may additionally include any compound or natural extract whose safety has already been verified and which is known to have anticancer activity in order to increase or reinforce the anticancer activity.
상기한 요소들을 모두 고려하여 최소한의 부작용으로 또는 부작용 없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하며, 이는 당업자에 의해 용이하게 결정될 수 있다.Taking all of the above factors into consideration, it is important to administer an amount that can achieve maximum effect with minimal or no side effects, and this can be easily determined by a person skilled in the art.
본 발명의 다른 측면은, 항-IGSF1 항체 또는 이의 단편을 유효성분으로 포함하는 약학 조성물을 개체에 투여하는 단계를 포함하는 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암에 대한 예방 또는 치료 방법을 제공한다. 여기서 항-IGSF1 항체 및 이의 단편, 약학 조성물, 대장암, 담도암, 두경부암, 투여, 치료 및 예방은 상술한 바와 동일하다.Another aspect of the present invention is to treat any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer, comprising administering to an individual a pharmaceutical composition containing an anti-IGSF1 antibody or fragment thereof as an active ingredient. Provides prevention or treatment methods for Here, anti-IGSF1 antibody and fragment thereof, pharmaceutical composition, colorectal cancer, biliary tract cancer, head and neck cancer, administration, treatment and prevention are the same as described above.
상기 개체는 포유동물일 수 있으며, 바람직하게는 인간일 수 있다. 또한, 상기 개체는 상기 질환을 갖고 있는 환자이거나 질환을 앓을 가능성이 큰 개체일 수 있다. The subject may be a mammal, preferably a human. Additionally, the individual may be a patient with the disease or an individual who is likely to suffer from the disease.
상기 약학 조성물의 투여경로, 투여량 및 투여횟수는 환자의 상태 및 부작용의 유무에 따라 다양한 방법 및 양으로 대상에게 투여될 수 있고, 최적의 투여방법, 투여량 및 투여횟수는 통상의 기술자가 적절한 범위로 선택할 수 있다. 또한, 상기 항-IGSF1 항체 또는 이의 단편은 상기 질환에 대하여 치료 효과가 공지된 다른 약물 또는 생화학적 활성물질과 병용하여 투여되거나, 다른 약물과의 조합 제제 형태로 제형화될 수 있다.The administration route, dosage, and frequency of administration of the pharmaceutical composition may be administered to the subject in various methods and amounts depending on the patient's condition and the presence or absence of side effects, and the optimal administration method, dosage, and frequency of administration can be determined by a person skilled in the art. You can select by range. Additionally, the anti-IGSF1 antibody or fragment thereof may be administered in combination with other drugs or biochemically active substances known to have therapeutic effects on the disease, or may be formulated in the form of a combination preparation with other drugs.
본 발명의 또 다른 측면은, 본 발명의 항-IGSF1 항체 또는 이의 단편을 유효성분으로 포함하는 약학 조성물의 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암에 대한 예방 또는 치료 용도를 제공한다. 상기 항-IGSF1 항체 및 이의 단편, 약학 조성물, 대장암, 담도암, 두경부암, 치료 및 예방은 상술한 바와 동일하다.Another aspect of the present invention is the prevention or treatment of any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer using a pharmaceutical composition containing the anti-IGSF1 antibody or fragment thereof of the present invention as an active ingredient. Provides a purpose. The anti-IGSF1 antibody and fragment thereof, pharmaceutical composition, colon cancer, biliary tract cancer, head and neck cancer, treatment and prevention are the same as described above.
본 발명의 또 다른 측면은 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암의 예방 또는 치료에의 이용을 위한 항-IGSF1 항체 또는 이의 단편을 제공한다. 상기 항-IGSF1 항체 및 이의 단편, 약학 조성물, 대장암, 담도암, 두경부암, 치료 및 예방은 상술한 바와 동일하다.Another aspect of the present invention provides an anti-IGSF1 antibody or fragment thereof for use in the prevention or treatment of any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer. The anti-IGSF1 antibody and fragment thereof, pharmaceutical composition, colon cancer, biliary tract cancer, head and neck cancer, treatment and prevention are the same as described above.
본 발명의 또 다른 측면은 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암을 예방 또는 치료하기 위한 의약의 제조를 위한 항-IGSF1 항체 또는 이의 단편의 용도를 제공한다. 여기서 항-IGSF1 항체 및 이의 단편, 약학 조성물, 대장암, 담도암, 두경부암, 예방 및 치료는 상술한 바와 동일하다.Another aspect of the present invention provides the use of an anti-IGSF1 antibody or fragment thereof for the manufacture of a medicament for preventing or treating any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer. Here, the anti-IGSF1 antibody and fragment thereof, pharmaceutical composition, colon cancer, biliary tract cancer, head and neck cancer, prevention and treatment are the same as described above.
이하, 본 발명을 하기 실시예에 의해 상세히 설명한다. 단, 하기 실시예는 본 발명을 예시하기 위한 것일 뿐, 본 발명이 이들에 의해 제한되는 것은 아니다.Hereinafter, the present invention will be explained in detail by the following examples. However, the following examples are only for illustrating the present invention, and the present invention is not limited thereto.
실시예 1. 항-IGSF1 항체 제작Example 1. Production of anti-IGSF1 antibody
실시예 1.1. IGSF1 항원 발현 및 정제Example 1.1. IGSF1 antigen expression and purification
Jurkat 세포 cDNA 라이브러리에서 PCR 방법을 통해 IGSF1의 세포외 도메인만을 증폭한 후, N293F 벡터(와이바이오로직스(주))를 이용하여 카복시 말단(C 말단)에 인간 Fc(fragment crystallizable region) 및 히스택(hig tag)을 코딩하는 폴리뉴클레오티드를 융합시켜 IGSF1 단백질 발현 벡터를 제작하였다. HEK293F 세포에 제작한 IGSF1 발현 벡터를 형질감염(transfection)시킨 후, 1 mM valporic acid(valproate)를 첨가한 배지에서 6일 동안 배양하였다. 그리고, protein A 아가로오즈(agarose)를 사용하여 IGSF1 세포외 도메인을 1차 정제(purification)한 후, Superdex 200 겔 여과 크로마토그래피(chratography)를 이용하여 IGSF1 세포외 도메인을 2차 정제한 후, 항체 선별에 사용하였다.After amplifying only the extracellular domain of IGSF1 from the Jurkat cell cDNA library through PCR, human Fc (fragment crystallizable region) and Histack were added to the carboxy terminus (C terminus) using the N293F vector (Y Biologics Co., Ltd.) An IGSF1 protein expression vector was created by fusing a polynucleotide encoding hig tag). HEK293F cells were transfected with the prepared IGSF1 expression vector and then cultured for 6 days in medium supplemented with 1 mM valporic acid (valproate). Then, after primary purification of the IGSF1 extracellular domain using protein A agarose, secondary purification of the IGSF1 extracellular domain using Superdex 200 gel filtration chromatography, It was used for antibody selection.
실시예 1.2. IGSF1 인간 항체의 선별Example 1.2. Selection of IGSF1 human antibodies
IGSF1 항원을 코팅하고 블로킹한 후 준비된 인간 항체 라이브러리 파아지(phage)(와이바이오로직스(주))를 이용하여 바이오패닝(bio-panning, 와이바이오로직스(주))을 진행하고 항원에 특이적으로 결합한 파아지들만 용출(elution)하였다. 첫 번째 라운드의 바이오패닝에서 증폭된 파아지를 가지고 두 번째 및 세 번째 라운드의 바이오패닝을 수행하였다. 각 라운드의 바이오패닝을 통해 얻어진 양성 파아지 항체 풀(pool)에 대한 항원과의 특이성을 확인하고자 ELISA를 수행하였다. 또한, 세 번째 라운드를 통해 얻어진 파아지 풀에 항-IGSF1 항체가 인리치(enrich)된 것을 확인하였다. 각 폴리 파아지 ELISA에서 결합능이 큰 세 번째 라운드의 패닝으로부터 수백 종의 모노클론(monoclone)들을 선별하였고, 이를 이용하여 ELISA 분석을 통해 IGSF1에 특이적으로 결합하는지 여부를 확인하여 예비 항체 클론을 확보하였다. 선별된 예비 항체 클론들에 대해 DNA 염기서열 분석을 통해 염기서열이 서로 다른 파아지를 99종 선별하였다. 선별된 99종의 양성 파아지 클론들은 항원인 IGSF1에는 강하게 결합하였으나, 다른 항원들에 대해서는 결합하지 않는 것을 확인하였다. 상기 방법을 통해 추가적으로 다양한 다른 항원을 이용하여 IGSF1 항원에 특이성을 나타내는 항체를 선별한 결과 총 95종을 선별할 수 있었다.After coating and blocking the IGSF1 antigen, bio-panning (bio-panning, Y Biologics Co., Ltd.) was performed using the prepared human antibody library phage (Y Biologics Co., Ltd.), and the antigen-specific binding was performed. Only phages were eluted. The second and third rounds of biopanning were performed using the phage amplified in the first round of biopanning. ELISA was performed to confirm the antigen specificity of the positive phage antibody pool obtained through each round of biopanning. In addition, it was confirmed that anti-IGSF1 antibody was enriched in the phage pool obtained through the third round. In each polyphage ELISA, hundreds of monoclones were selected from the third round of panning with high binding capacity, and using these, preliminary antibody clones were obtained by confirming whether they specifically bind to IGSF1 through ELISA analysis. . For the selected preliminary antibody clones, 99 phages with different base sequences were selected through DNA sequence analysis. It was confirmed that the 99 selected positive phage clones strongly bound to the antigen, IGSF1, but did not bind to other antigens. Through the above method, a total of 95 types were selected as a result of selecting antibodies showing specificity for the IGSF1 antigen using various other antigens.
실시예 1.3. IGSF1 항원에 대한 특이성 확인 Example 1.3. Confirmation of specificity for IGSF1 antigen
선별한 항체에 대해 IGSF1을 비롯한 다른 항원들에 대한 특이성을 ELISA 방법을 통해 비교 분석하였다. 대조군 항원들인 mFc, hRAGE-Fc, CD58-Fc, ITGA6-Fc 등 다양한 종류의 불특정 항원에 대해서 파아지 클론들의 결합 여부를 확인하였다. 이와 같이 하여 수득한 항체들에 대해 파아지에서 IgG whole 벡터로 전환을 하였고, 전환된 95개 클론의 중쇄 서열 및 경쇄 서열이 파아지 항체의 서열과 일치하는 것을 확인하였다. 수득된 항체 중 가장 최적화된 항체를 선별하고, 이를 "WM-A1-3389"라 명명하였다. WM-A1-3389 항체의 CDR 서열은 하기 표 1과 같다. The specificity of the selected antibodies to other antigens, including IGSF1, was compared and analyzed using ELISA. The binding of phage clones to various types of unspecified antigens, including control antigens mFc, hRAGE-Fc, CD58-Fc, and ITGA6-Fc, was confirmed. The antibodies obtained in this way were converted from phage to IgG whole vectors, and it was confirmed that the heavy and light chain sequences of the converted 95 clones matched the sequences of the phage antibodies. Among the obtained antibodies, the most optimized antibody was selected and named “WM-A1-3389”. The CDR sequence of the WM-A1-3389 antibody is shown in Table 1 below.
CDRCDR 아미노산 서열amino acid sequence 서열번호sequence number
H-CDR1H-CDR1 GGTFSTYAGGTFSTYA 1One
H-CDR2H-CDR2 IIPFVGTVIIPFVGTV 22
H-CDR3H-CDR3 VRDGGRSYFDSVRDGRSYFDS 33
L-CDR1L-CDR1 TSNIGSNLTSNIGSNL 44
L-CDR2L-CDR2 DNHDNH 55
L-CDR3L-CDR3 VAWDDSLNGYVVAWDDSLNGYV 66
실시예 1.4. WM-A1-3389 항체 생산Example 1.4. WM-A1-3389 antibody production
WM-A1-3389 항체를 생산하기 위하여, 중쇄(서열번호 21)를 코딩하는 폴리뉴클레오티드(서열번호 23)를 N293F 벡터(와이바이오로직스(주))에 적재하였다(이하, HC DNA). 또한, 경쇄(서열번호 22)를 암호화하는 폴리뉴클레오티드(서열번호 24)를 N293F 벡터(와이바이오로직스(주))에 적재하였다(이하, LC DNA). 상기 벡터를 세포에 형질전환 시킨 후, WM-A1-3389 항체를 수득 및 정제하였다. 정제된 단백질은 SDS-PAGE를 통해 확인하였다.To produce the WM-A1-3389 antibody, a polynucleotide (SEQ ID NO: 23) encoding the heavy chain (SEQ ID NO: 21) was loaded into the N293F vector (Y Biologics Co., Ltd.) (hereinafter referred to as HC DNA). In addition, a polynucleotide (SEQ ID NO: 24) encoding the light chain (SEQ ID NO: 22) was loaded into the N293F vector (Y Biologics Co., Ltd.) (hereinafter referred to as LC DNA). After the vector was transformed into cells, WM-A1-3389 antibody was obtained and purified. Purified proteins were confirmed through SDS-PAGE.
아미노산 서열amino acid sequence 서열번호sequence number
중쇄(Heavy chain)Heavy chain QVQLVQSGAEVKRPGSSVKVSCKASGGTFSTYAISWVRQAPGQGLEWMGRIIPFVGTVDYAQKFQDRVTITADKSTNTAYMELSSLRSEDTAVYYCVRDGGRSYFDSWGPGILVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKQVQLVQSGAEVKRPGSSVKVSCKASGGTFSTYAISWVRQAPGQGLEWMGRIIPFVGTVDYAQKFQDRVTITADKSTNTAYMELSSLRSEDTAVYYCVRDGGRSYFDSWGPGILVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNH KPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPV LDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK 2121
경쇄(Light chain)Light chain QFVLTQPPSVSAAPGQDVIISCSGNTSNIGSNLVSWFQQFPETAPKLLIYDNHKRPSGISDRFSGTKSGTSASLAISGLQSEDEADYYCVAWDDSLNGYVFGTGTKVTVLRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECQFVLTQPPSVSAAPGQDVIISCSGNTSNIGSNLVSWFQQFPETAPKLLIYDNHKRPSGISDRFSGTKSGTSASLAISGLQSEDEADYYCVAWDDSLNGYVFGTGTKVTVLRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHK VYACEVTHQGLSSPVTKSFNRGEC 2222
실시예 2. 대장암에 대한 WM-A1-3389 항체의 항암 활성Example 2. Anticancer activity of WM-A1-3389 antibody against colon cancer
실시예 2.1. 인간 대장암 세포주에서 IGSF1의 발현 확인Example 2.1. Confirmation of IGSF1 expression in human colon cancer cell lines
먼저, 인간 대장암 세포주인 HT29 세포에서 IGSF1의 발현을 확인하였다. First, the expression of IGSF1 was confirmed in HT29 cells, a human colon cancer cell line.
구체적으로, HT29 세포의 배지를 제거하고, PBS로 1회 세척한 뒤 Accutase 2 ㎖를 처리하여 세포를 분리하였다. 분리된 세포는 2%(v/v) FBS 및 0.05%(w/v) sodium azide가 포함된 PBS(이하, FACS 버퍼) 8 ㎖에 희석한 뒤 1,200 rpm에서 1분간 원심분리하여 상층액을 제거하였다. 세포 펠렛(pellet)을 vortex로 풀어주고 적정량의 FACS 버퍼로 재부유시킨 뒤, 트리판 블루(trypan blue)로 세포수를 계수하여 각 튜브당 1Х105 cells가 되도록 분주하였다. 그 다음, 1,200 rpm에서 1분간 원심분리하여 상층액을 제거하고 세포 펠렛을 vortex를 사용하여 풀어주었다. 그리고 Alexa Fluor 647이 표지된 항-인간 IGSF1 항체(sc-393786 AF647, Santa Cruz Biotechnology)를 FACS 버퍼 200 ㎕당 0.4 ㎍씩 처리하고 차광하여 4℃에서 30분간 반응시켰다. 반응이 끝난 후, 각 튜브에 FACS 버퍼 1 ㎖씩 넣고 1,200 rpm에서 1분간 원심분리하여 상층액을 제거하였다. 상기 과정을 총 2번 진행하였다. 마지막으로 남은 세포 펠렛에 FACS 버퍼 200 ㎕를 첨가하여 재부유시키고 FACS로 분석하였다. FACS 분석은 BD LSRFortessa Flow cytometry(BD Bioscience)를 사용하여 각 세포에 표지된 Alexa 647의 형광 값을 측정한 후, FLowJo 소프트웨어를 사용하여 결과를 분석하였다.Specifically, the medium of HT29 cells was removed, washed once with PBS, and then treated with 2 ml of Accutase to separate the cells. The separated cells were diluted in 8 ml of PBS (hereinafter referred to as FACS buffer) containing 2% (v/v) FBS and 0.05% (w/v) sodium azide, then centrifuged at 1,200 rpm for 1 minute to remove the supernatant. did. The cell pellet was released with a vortex and resuspended with an appropriate amount of FACS buffer, and the number of cells was counted using trypan blue and distributed to each tube to reach 1Х10 5 cells. Then, the supernatant was removed by centrifugation at 1,200 rpm for 1 minute, and the cell pellet was released using a vortex. Then, 0.4 ㎍ of Alexa Fluor 647-labeled anti-human IGSF1 antibody (sc-393786 AF647, Santa Cruz Biotechnology) was added per 200 ㎕ of FACS buffer and incubated at 4°C for 30 minutes, protected from light. After the reaction was completed, 1 ml of FACS buffer was added to each tube and centrifuged at 1,200 rpm for 1 minute to remove the supernatant. The above process was performed a total of two times. Finally, 200 ㎕ of FACS buffer was added to the remaining cell pellet, resuspended, and analyzed by FACS. For FACS analysis, the fluorescence value of Alexa 647 labeled in each cell was measured using BD LSRFortessa Flow cytometry (BD Bioscience), and the results were analyzed using FLowJo software.
그 결과, 도 1에 나타낸 바와 같이, HT29 세포주에서 IGSF1을 발현하는 세포 비율은 약 49.8%로 나타났다.As a result, as shown in Figure 1, the proportion of cells expressing IGSF1 in the HT29 cell line was approximately 49.8%.
실시예 2.2. 인간 대장암 세포주 및 말초혈액세포의 공동배양에 의한 사이토카인 분비량의 변화 확인Example 2.2. Confirmation of changes in cytokine secretion by co-culture of human colon cancer cell lines and peripheral blood cells
대장암 세포주 및 말초혈액세포의 공동배양 시스템을 이용하여, WM-A1-3389 항체 처리에 의한 사이토카인 분비량 변화를 확인하였다.Using a co-culture system of colon cancer cell lines and peripheral blood cells, changes in cytokine secretion due to treatment with WM-A1-3389 antibody were confirmed.
구체적으로, 인간 유래의 말초혈액세포(hPBMC)를 1,200 rpm에서 10분간 원심분리하여 상층액을 제거하고, 10% RPMI에 재부유시켰다. 그 후에 HT29 대장암 세포와 hPBMC를 각각 1:1 비율로 접종하고 1 ㎍/㎖ SEB(Sigma), 10 ㎍/㎖ IgG(Bio X-cell) 또는 10 ㎍/㎖ WM-A1-3389 항체를 각각 처리하여 37℃ 이산화탄소 배양기에서 24시간동안 배양하였다. 이때, IgG는 음성 대조군으로 사용하였다. 공동배양한 세포의 상층액은 1.5 ㎖ 에펜도르프 튜브에 모아 Cytometric bead array(CBA) assay를 통해 사이토카인 분비량을 측정하였다. CBA assay는 BD Biosciences 제품을 사용하였으며, BD Biosciences의 프로토콜에 따라 수행하였다. 각 튜브에 스탠다드(표준 시료) 및 공동배양세포 상층액을 각각 넣고, capture bead를 추가하여 상온에서 1 내지 2시간 동안 반응시켰다. 반응 후, PE detection reagent를 첨가하여 실온에서 2시간동안 추가 반응시켰다. 각 튜브를 세척 버퍼로 세척하고, LSRFortessa™ Flow Cytometry 장비(BD Bioscience)를 이용하여 측정하였다.Specifically, human-derived peripheral blood cells (hPBMC) were centrifuged at 1,200 rpm for 10 minutes to remove the supernatant, and resuspended in 10% RPMI. Afterwards, HT29 colon cancer cells and hPBMC were inoculated at a 1:1 ratio, and 1 ㎍/㎖ SEB (Sigma), 10 ㎍/㎖ IgG (Bio The cells were treated and cultured in a carbon dioxide incubator at 37°C for 24 hours. At this time, IgG was used as a negative control. The supernatant of the co-cultured cells was collected in a 1.5 ml Eppendorf tube and the amount of cytokine secretion was measured through Cytometric bead array (CBA) assay. The CBA assay used a BD Biosciences product and was performed according to the BD Biosciences protocol. Standard (standard sample) and co-cultured cell supernatant were added to each tube, capture beads were added, and reaction was performed at room temperature for 1 to 2 hours. After reaction, PE detection reagent was added and further reaction was performed at room temperature for 2 hours. Each tube was washed with washing buffer and measured using LSRFortessa™ Flow Cytometry equipment (BD Bioscience).
그 결과, 도 2에 나타낸 바와 같이, IGSF1이 발현되어 있는 HT29 대장암 세포에서 Granzyme B를 비롯하여 사이토카인 IFN-γ 및 TNF-α 분비량이 대조군에 비해 WM-A1-3389 항체를 처리한 군에서 통계적으로 유의하게 증가하는 것을 확인하였다.As a result, as shown in Figure 2, the secretion amount of cytokines IFN-γ and TNF-α, including Granzyme B, in HT29 colon cancer cells expressing IGSF1 was statistically significantly lower in the group treated with the WM-A1-3389 antibody compared to the control group. It was confirmed that there was a significant increase.
실시예 2.3. 이종 대장암 세포주 식립 마우스 모델에서 WM-A1-3389 항체의 항암 활성 확인Example 2.3. Confirmation of anticancer activity of WM-A1-3389 antibody in xenogeneic colon cancer cell line transplantation mouse model
인간 대장암 세포주를 이식한 대장암 마우스 모델에서 WM-A1-3389 항체의 항암 활성을 확인하였다.The anticancer activity of the WM-A1-3389 antibody was confirmed in a colon cancer mouse model transplanted with a human colon cancer cell line.
구체적으로, 6주령 암컷 말초 혈액 단핵세포 인간화 마우스(Gem biosciences)를 구입하여 1주 동안 순화한 후, 인간 대장암 세포주인 HT29 세포를 PBS에 희석하여 5Х106 cells/mice의 농도가 되도록 오른쪽 배측면에 피하(100 ㎕) 주사하였다. WM-A1-3389 항체에 대한 음성 대조군으로 인간 IgG isotype을 사용하였다. 종양의 크기가 평균 140 mm3 내지 160 mm3가 되었을 때 인간 IgG isotype(음성 대조군) 10 mg/kg 또는 WM-A1-3389 항체 30 mg/kg 용량으로 복강 투여하였다. 투여는 3일에 한 번씩 4주 동안 실시하였으며, 주 2회 마우스의 종양 크기와 체중을 측정하였다. 투여 28일째 되는 날, 실험 동물을 안락사 시키고 종양을 적출하여 무게를 측정하였다. Specifically, 6-week-old female peripheral blood mononuclear cell humanized mice (Gem biosciences) were purchased and acclimatized for 1 week, and then HT29 cells, a human colon cancer cell line, were diluted in PBS and instilled on the right ventral side to a concentration of 5Х10 6 cells/mice. It was injected subcutaneously (100 ㎕). Human IgG isotype was used as a negative control for the WM-A1-3389 antibody. When the tumor size reached an average of 140 mm 3 to 160 mm 3 , human IgG isotype (negative control) was administered intraperitoneally at a dose of 10 mg/kg or WM-A1-3389 antibody at a dose of 30 mg/kg. Administration was conducted once every three days for four weeks, and the tumor size and body weight of the mice were measured twice a week. On the 28th day of administration, the experimental animals were euthanized and the tumors were extracted and weighed.
그 결과, 도 3에 나타낸 바와 같이, WM-A1-3389 항체 투여군은 음성 대조군에 비해 종양 성장 억제 효능이 높게 나타났다. As a result, as shown in Figure 3, the WM-A1-3389 antibody administration group showed higher tumor growth inhibition efficacy than the negative control group.
음성 대조군과 비교 시, WM-A1-3389 항체 투여량 및 종양 성장 억제율(Tumor growth inhibition: TGI)을 하기의 표 3에 기재하였다(***: P<0.001).When compared to the negative control group, the WM-A1-3389 antibody dosage and tumor growth inhibition (TGI) are listed in Table 3 below (***: P<0.001).
WM-A1-3389 항체 투여군WM-A1-3389 antibody administration group
Dose (mg/kg)Dose (mg/kg) 3030
TGI (%)TGI (%) 55.5 ± 4.455.5 ± 4.4
P-ValueP-Value 0.0002 (***)0.0002 (***)
표 3에 나타낸 바와 같이, 약 55.5%의 종양 성장 억제율(Tumor growth inhibition: TGI)을 보였다. 또한, 개별 개체들에 대해서도 종양 성장이 억제되는 것을 확인할 수 있었다.As shown in Table 3, tumor growth inhibition (TGI) was shown to be about 55.5%. In addition, it was confirmed that tumor growth was suppressed in individual subjects.
실시예 2.4. 동종 대장암 세포주 식립 마우스 모델에서 WM-A1-3389 항체의 항암 활성 확인Example 2.4. Confirmation of anticancer activity of WM-A1-3389 antibody in allogeneic colon cancer cell line transplantation mouse model
실시예 2.4.1. 마우스 대장암 세포주 MC38에서 IGSF1 발현 확인Example 2.4.1. Confirmation of IGSF1 expression in mouse colon cancer cell line MC38
마우스 대장암 세포주인 MC38 세포에서 IGSF1의 발현을 면역형광염색 및 유세포 분석을 통해 확인하였다. 면역형광염색은 다음과 같이 진행하였다. 상층액을 제거한 세포를 4%(v/v) 포름알데하이드(PFA, T&I)로 상온에서 15분간 고정하였다. 고정한 세포를 PBS로 2회 세척하고 비-특이적인 결합을 막기위해 블로킹 버퍼를 추가하여 상온에서 1시간 동안 반응시켰다. 반응 후, 1차 IGSF1 항체(sc-393786, Santa Cruz Biotechnology)를 1:100 비율로 처리하고 차광하여 4℃에서 12시간 이상 반응시켰다. 반응 후, PBS로 2회 세척하고 2차 항체로 항-마우스 IgG H&L(Alexa 488)(ab150113, Abcam) 항체를 1:100 비율로 처리하고 차광하여 상온에서 1시간 동안 반응시켰다. 염색한 세포를 PBS로 2회 세척하고 형광 현미경(Olympus)으로 관찰하였다. 유세포 분석은 실시예 2.1과 동일한 방법으로 수행하였다.The expression of IGSF1 in MC38 cells, a mouse colon cancer cell line, was confirmed through immunofluorescence staining and flow cytometry. Immunofluorescence staining was performed as follows. Cells with the supernatant removed were fixed with 4% (v/v) formaldehyde (PFA, T&I) at room temperature for 15 minutes. The fixed cells were washed twice with PBS, blocking buffer was added to prevent non-specific binding, and reacted at room temperature for 1 hour. After the reaction, the primary IGSF1 antibody (sc-393786, Santa Cruz Biotechnology) was treated at a ratio of 1:100 and reacted at 4°C for more than 12 hours, protected from light. After the reaction, the cells were washed twice with PBS, treated with anti-mouse IgG H&L (Alexa 488) (ab150113, Abcam) antibody as a secondary antibody at a ratio of 1:100, and incubated for 1 hour at room temperature, shielded from light. The stained cells were washed twice with PBS and observed under a fluorescence microscope (Olympus). Flow cytometry was performed in the same manner as Example 2.1.
그 결과, 도 4에 나타낸 바와 같이, MC38 세포에서 IGSF1의 발현이 관찰되었고, IGSF1을 발현하는 세포 비율은 약 45.4%로 나타났다.As a result, as shown in Figure 4, expression of IGSF1 was observed in MC38 cells, and the proportion of cells expressing IGSF1 was approximately 45.4%.
실시예 2.4.2. 마우스 대장암 세포주 MC38 식립 마우스 모델에서 WM-A1-3389 항체의 항암 활성 확인Example 2.4.2. Confirmation of anticancer activity of WM-A1-3389 antibody in mouse colon cancer cell line MC38 implanted mouse model
마우스 대장암 세포주인 MC38 세포를 이식한 대장암 마우스 모델(MC38 syngeneic mouse model)에서 WM-A1-3389 항체의 항암 활성을 확인하였다.The anticancer activity of the WM-A1-3389 antibody was confirmed in a colon cancer mouse model (MC38 syngeneic mouse model) transplanted with MC38 cells, a mouse colon cancer cell line.
구체적으로, 5주령 암컷 C57BL/6N 마우스를 구입하여 1주 동안 순화시켰다. 마우스 대장암 세포 MC38 세포를 PBS에 희석하여 2.5Х105 cells/mice의 농도가 되도록 준비하고, MC38 세포를 마우스의 오른쪽 배측면에 피하(100 ㎕) 주사하였다. WM-A1-3389 항체에 대한 음성 대조군으로 마우스 IgG isotype을 사용하였다. 종양의 크기가 평균 50 mm3 내지 100 mm3가 되었을 때 마우스 IgG isotype(음성 대조군) 10 mg/kg, 및 WM-A1-3389 항체 3, 10 및 30 mg/kg의 용량으로 각각 복강 투여하였다. 투여는 3일에 한 번씩 2주 동안 실시하였으며, 주 2회 마우스의 종양 크기와 체중을 측정하였다. 투여 14일째 되는 날, 실험 동물을 안락사 시키고 종양을 적출하여 무게를 측정하였다.Specifically, 5-week-old female C57BL/6N mice were purchased and acclimatized for 1 week. Mouse colon cancer cells MC38 cells were diluted in PBS to prepare a concentration of 2.5Х10 5 cells/mice, and MC38 cells were injected subcutaneously (100 ㎕) into the right ventral side of the mouse. Mouse IgG isotype was used as a negative control for the WM-A1-3389 antibody. When the tumor size reached an average of 50 mm 3 to 100 mm 3 , mouse IgG isotype (negative control) was administered intraperitoneally at doses of 10 mg/kg and WM-A1-3389 antibody at doses of 3, 10, and 30 mg/kg, respectively. Administration was performed once every three days for two weeks, and the tumor size and body weight of the mice were measured twice a week. On the 14th day of administration, the experimental animals were euthanized and the tumors were extracted and weighed.
그 결과, 도 5에 나타낸 바와 같이, WM-A1-3389 항체 투여군은 음성 대조군에 비해 종양 성장 억제 효능이 높게 나타났으며, 투여 농도 의존적으로 종양 성장 억제율(TGI)이 증가하는 상관성을 확인할 수 있었다. 음성 대조군과 비교 시, WM-A1-3389 항체 투여량 및 종양 성장 억제율(TGI)을 하기의 표 4에 기재하였다.As a result, as shown in Figure 5, the WM-A1-3389 antibody administered group showed higher tumor growth inhibition efficacy than the negative control group, and a correlation was confirmed in which the tumor growth inhibition rate (TGI) increased in a dose-dependent manner. . When compared to the negative control, the WM-A1-3389 antibody dosage and tumor growth inhibition rate (TGI) are listed in Table 4 below.
WM-A1-3389 항체 투여군WM-A1-3389 antibody administration group
Dose(mg/kg)Dose(mg/kg) 33 1010 3030
TGI(%)TGI(%) 50.8 ± 4.350.8 ± 4.3 58.2 ± 4.858.2 ± 4.8 68.7 ± 3.868.7 ± 3.8
실시예 2.4.3. 마우스 대장암 세포주 CT26에서 IGSF1 발현 확인Example 2.4.3. Confirmation of IGSF1 expression in mouse colon cancer cell line CT26
마우스 대장암 세포주인 CT26 세포에서 IGSF1의 발현을 면역형광염색 및 유세포 분석을 통해 확인하였다. 면역형광염색은 실시예 2.4.1과 동일한 방법으로 수행하였고, 유세포 분석은 실시예 2.1과 동일한 방법으로 수행하였다. The expression of IGSF1 in CT26 cells, a mouse colon cancer cell line, was confirmed through immunofluorescence staining and flow cytometry. Immunofluorescence staining was performed in the same manner as in Example 2.4.1, and flow cytometry was performed in the same manner as in Example 2.1.
그 결과, 도 6에 나타낸 바와 같이, CT26 세포주에서 IGSF1의 발현이 관찰되었고, IGSF1을 발현하는 세포 비율은 약 57.2%로 나타났다.As a result, as shown in Figure 6, expression of IGSF1 was observed in the CT26 cell line, and the proportion of cells expressing IGSF1 was approximately 57.2%.
실시예 2.4.4. 마우스 대장암 세포주 CT26 식립 마우스 모델에서 WM-A1-3389 항체의 항암 활성 확인Example 2.4.4. Confirmation of anticancer activity of WM-A1-3389 antibody in mouse colon cancer cell line CT26 implanted mouse model
마우스 대장암 세포주인 CT26 세포를 이식한 대장암 마우스 모델(CT26 syngeneic mouse model)에서 WM-A1-3389 항체의 항암 활성을 확인하였다.The anticancer activity of the WM-A1-3389 antibody was confirmed in a colon cancer mouse model (CT26 syngeneic mouse model) transplanted with CT26 cells, a mouse colon cancer cell line.
구체적으로, 5주령 암컷 C57BL/6N 마우스를 구입하여 1주 동안 순화 후, 마우스 대장암 세포 CT26 세포를 PBS에 희석하여 2.5Х105 cells/mice의 농도가 되도록 오른쪽 배측면에 피하(100 ㎕) 주사하였다. WM-A1-3389 항체에 대한 음성 대조군으로 인간 IgG isotype을 사용하였다. 종양의 크기가 평균 80 mm3 내지 100 mm3가 되었을 때 마우스 IgG isotype(음성 대조군) 10 mg/kg, 및 WM-A1-3389 항체를 3, 10 및 50 mg/kg의 용량으로 각각 복강 투여하였다. 투여는 3일에 한 번씩 2주 동안 실시하였으며, 주 2회 마우스의 종양 크기와 체중을 측정하였다. 투여 14일째 되는 날, 실험 동물을 안락사 시켜 종양을 적출하여 무게를 측정하였다.Specifically, 5-week-old female C57BL/6N mice were purchased, acclimatized for 1 week, and mouse colon cancer CT26 cells were diluted in PBS and injected subcutaneously (100 ㎕) into the right ventral side to a concentration of 2.5Х10 5 cells/mice. did. Human IgG isotype was used as a negative control for the WM-A1-3389 antibody. When the tumor size reached an average of 80 mm 3 to 100 mm 3 , 10 mg/kg of mouse IgG isotype (negative control) and WM-A1-3389 antibody were administered intraperitoneally at doses of 3, 10, and 50 mg/kg, respectively. . Administration was performed once every three days for two weeks, and the tumor size and body weight of the mice were measured twice a week. On the 14th day of administration, the experimental animals were euthanized, the tumors were extracted, and their weight was measured.
그 결과, 도 7에 나타낸 바와 같이, WM-A1-3389 항체 투여군은 음성 대조군에 비해 종양 성장 억제 효능이 높게 나타났으며, 투여 농도 의존적으로 종양 성장 억제율(TGI)이 증가하는 상관성을 확인할 수 있었다. 음성 대조군과 비교 시, WM-A1-3389 항체 투여량 및 종양 성장 억제율(TGI)을 하기의 표 5에 기재하였다.As a result, as shown in Figure 7, the WM-A1-3389 antibody administered group showed higher tumor growth inhibition efficacy than the negative control group, and a correlation was confirmed in which the tumor growth inhibition rate (TGI) increased in a dose-dependent manner. . When compared to the negative control, the WM-A1-3389 antibody dosage and tumor growth inhibition rate (TGI) are listed in Table 5 below.
WM-A1-3389 항체 투여군WM-A1-3389 antibody administration group
Dose (mg/kg)Dose (mg/kg) 33 1010 5050
TGI (%)TGI (%) 44.9 ± 3.844.9 ± 3.8 57.0 ± 3.557.0 ± 3.5 66.0 ± 3.566.0 ± 3.5
실시예 3. 담도암에 대한 WM-A1-3389 항체의 항암 활성Example 3. Anticancer activity of WM-A1-3389 antibody against biliary tract cancer
실시예 3.1. 인간 담도암 세포주에서 IGSF1의 발현 확인Example 3.1. Confirmation of IGSF1 expression in human biliary tract cancer cell lines
인간 담도암 세포주인 Choi-CK 세포에서 IGSF1의 발현을 실시예 2.1과 동일한 방법으로 확인하였다. Expression of IGSF1 in Choi-CK cells, a human biliary tract cancer cell line, was confirmed in the same manner as Example 2.1.
그 결과, 도 8에 나타낸 바와 같이, Choi-CK 세포주에서 IGSF1을 발현하는 세포 비율은 약 57.5%로 나타났다.As a result, as shown in Figure 8, the proportion of cells expressing IGSF1 in the Choi-CK cell line was found to be about 57.5%.
실시예 3.2. 인간 담도암 세포주 및 말초혈액세포의 공동배양에 의한 사이토카인 분비량의 변화 확인Example 3.2. Confirmation of changes in cytokine secretion by co-culture of human biliary tract cancer cell lines and peripheral blood cells
담도암 세포주 및 말초혈액세포의 공동배양 시스템을 이용하여, WM-A1-3389 항체 처리에 의한 사이토카인 분비량 변화를 실시예 2.2와 동일한 방법으로 확인하였다.Using a co-culture system of biliary tract cancer cell lines and peripheral blood cells, changes in cytokine secretion due to treatment with WM-A1-3389 antibody were confirmed in the same manner as in Example 2.2.
그 결과, 도 9에 나타낸 바와 같이, IGSF1이 발현되어 있는 Choi-CK 담도암 세포에서 Granzyme B를 비롯하여 사이토카인 IFN-γ 및 TNF-α 분비량이 대조군에 비해 WM-A1-3389 항체를 처리한 군에서 통계적으로 유의하게 증가하는 것을 확인하였다.As a result, as shown in Figure 9, the secretion of Granzyme B, as well as cytokines IFN-γ and TNF-α, in Choi-CK biliary tract cancer cells expressing IGSF1 was higher in the group treated with the WM-A1-3389 antibody compared to the control group. A statistically significant increase was confirmed.
실시예 3.3. 이종 담도암 세포주 식립 마우스 모델에서 WM-A1-3389 항체의 항암 활성 확인Example 3.3. Confirmation of anticancer activity of WM-A1-3389 antibody in xenograft biliary tract cancer cell line transplantation mouse model
인간 담도암 세포주를 이식한 담도암 마우스 모델에서 WM-A1-3389 항체의 항암 활성을 확인하였다.The anticancer activity of the WM-A1-3389 antibody was confirmed in a biliary tract cancer mouse model transplanted with a human biliary tract cancer cell line.
구체적으로, 6주령 암컷 말초 혈액 단핵세포 인간화 마우스(Gem biosciences)를 구입하여 1주 동안 순화한 후, 인간 담도암 세포주인 Choi-CK 세포를 PBS에 희석하여 5Х106 cells/mice의 농도가 되도록 오른쪽 배측면에 피하(100 ㎕) 주사하였다. WM-A1-3389 항체에 대한 음성 대조군으로 인간 IgG isotype을 사용하였다. 종양의 크기가 평균 80 mm3 내지 100 mm3가 되었을 때 인간 IgG isotype(음성 대조군) 10 mg/kg 또는 WM-A1-3389 항체 30 mg/kg 용량으로 복강 투여하였다. 투여는 3일에 한 번씩 3주 동안 실시하였으며, 주 2회 마우스의 종양 크기와 체중을 측정하였다. 투여 22일째 되는 날, 실험 동물을 안락사 시키고 종양을 적출하여 무게를 측정하였다. Specifically, 6-week-old female peripheral blood mononuclear cell humanized mice (Gem biosciences) were purchased and acclimatized for 1 week, and then Choi-CK cells, a human biliary tract cancer cell line, were diluted in PBS to a concentration of 5Х10 6 cells/mice. It was injected subcutaneously (100 μl) on the dorsal side. Human IgG isotype was used as a negative control for the WM-A1-3389 antibody. When the tumor size reached an average of 80 mm 3 to 100 mm 3 , human IgG isotype (negative control) was administered intraperitoneally at a dose of 10 mg/kg or WM-A1-3389 antibody at a dose of 30 mg/kg. Administration was conducted once every three days for three weeks, and the tumor size and body weight of the mice were measured twice a week. On the 22nd day of administration, the experimental animals were euthanized and the tumors were extracted and weighed.
그 결과, 도 10에 나타낸 바와 같이, WM-A1-3389 항체 투여군은 음성 대조군에 비해 종양 성장 억제 효능이 높게 나타났다. As a result, as shown in Figure 10, the WM-A1-3389 antibody administration group showed higher tumor growth inhibition efficacy than the negative control group.
음성 대조군과 비교 시, WM-A1-3389 항체 투여량 및 종양 성장 억제율(TGI)을 하기의 표 6에 기재하였다(***: P<0.001).When compared to the negative control group, the WM-A1-3389 antibody dosage and tumor growth inhibition rate (TGI) are listed in Table 6 below (***: P<0.001).
WM-A1-3389 항체 투여군WM-A1-3389 antibody administration group
Dose (mg/kg)Dose (mg/kg) 3030
TGI (%)TGI (%) 58.0 ± 4.358.0 ± 4.3
P-ValueP-Value 0.0008 (***)0.0008 ( *** )
표 6에 나타낸 바와 같이, 약 58.0%의 종양 성장 억제율(Tumor growth inhibition: TGI)을 보였다. 또한, 개별 개체들에 대해서도 종양 성장이 억제되는 것을 확인할 수 있었다.As shown in Table 6, tumor growth inhibition (TGI) was shown to be about 58.0%. In addition, it was confirmed that tumor growth was suppressed in individual subjects.
실시예 4. 두경부암에 대한 WM-A1-3389 항체의 항암 활성Example 4. Anticancer activity of WM-A1-3389 antibody against head and neck cancer
실시예 4.1. 인간 두경부암 세포주에서 IGSF1의 발현 확인Example 4.1. Confirmation of IGSF1 expression in human head and neck cancer cell lines
인간 두경부암 세포주인 FaDu 세포에서 IGSF1의 발현을 실시예 2.1과 동일한 방법으로 확인하였다. The expression of IGSF1 in FaDu cells, a human head and neck cancer cell line, was confirmed in the same manner as Example 2.1.
그 결과, 도 11에 나타낸 바와 같이, FaDu 세포주에서 IGSF1을 발현하는 세포 비율은 약 37.7%로 나타났다.As a result, as shown in Figure 11, the proportion of cells expressing IGSF1 in the FaDu cell line was found to be about 37.7%.
실시예 4.2. 인간 두경부암 세포주 및 말초혈액세포의 공동배양에 의한 사이토카인 분비량의 변화 확인Example 4.2. Confirmation of changes in cytokine secretion by co-culture of human head and neck cancer cell lines and peripheral blood cells
두경부암 세포주 및 말초혈액세포의 공동배양 시스템을 이용하여, WM-A1-3389 항체 처리에 의한 사이토카인 분비량 변화를 실시예 2.2와 동일한 방법으로 확인하였다.Using a co-culture system of head and neck cancer cell lines and peripheral blood cells, changes in cytokine secretion due to treatment with WM-A1-3389 antibody were confirmed in the same manner as in Example 2.2.
그 결과, 도 12에 나타낸 바와 같이, IGSF1이 발현되어 있는 FaDu 두경부암 세포에서 Granzyme B를 비롯하여 사이토카인 IFN-γ 및 TNF-α 분비량이 대조군에 비해 WM-A1-3389 항체를 처리한 군에서 통계적으로 유의하게 증가하는 것을 확인하였다.As a result, as shown in Figure 12, the secretion of Granzyme B and cytokines IFN-γ and TNF-α from FaDu head and neck cancer cells expressing IGSF1 was statistically significantly higher in the group treated with the WM-A1-3389 antibody compared to the control group. It was confirmed that there was a significant increase.

Claims (7)

  1. 항-IGSF1 항체 또는 이의 단편을 유효성분으로 포함하는 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암에 대한 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer, comprising an anti-IGSF1 antibody or fragment thereof as an active ingredient.
  2. 제1항에 있어서,According to paragraph 1,
    상기 항-IGSF1 항체는 서열번호 1의 아미노산 서열을 포함하는 H-CDR1, 서열번호 2의 아미노산 서열을 포함하는 H-CDR2 및 서열번호 3의 아미노산 서열을 포함하는 H-CDR3을 포함하는 중쇄 가변영역; 및The anti-IGSF1 antibody has a heavy chain variable region comprising H-CDR1 including the amino acid sequence of SEQ ID NO: 1, H-CDR2 including the amino acid sequence of SEQ ID NO: 2, and H-CDR3 including the amino acid sequence of SEQ ID NO: 3. ; and
    서열번호 4의 아미노산 서열을 포함하는 L-CDR1, 서열번호 5의 아미노산 서열을 포함하는 L-CDR2 및 서열번호 6의 아미노산 서열을 포함하는 L-CDR3을 포함하는 경쇄 가변영역을 포함하는 것인, 약학 조성물.Comprising a light chain variable region comprising L-CDR1 containing the amino acid sequence of SEQ ID NO: 4, L-CDR2 containing the amino acid sequence of SEQ ID NO: 5, and L-CDR3 containing the amino acid sequence of SEQ ID NO: 6, Pharmaceutical composition.
  3. 제2항에 있어서,According to paragraph 2,
    상기 중쇄 가변영역은 서열번호 7의 아미노산 서열을 포함하며;The heavy chain variable region includes the amino acid sequence of SEQ ID NO: 7;
    상기 경쇄 가변영역은 서열번호 8의 아미노산 서열을 포함하는 것인, 약학 조성물.A pharmaceutical composition wherein the light chain variable region includes the amino acid sequence of SEQ ID NO: 8.
  4. 제1항에 있어서,According to paragraph 1,
    상기 항-IGSF1 항체는 IGSF1의 C 말단에 특이적으로 결합하는 것을 특징으로 하는 것인, 약학 조성물.The anti-IGSF1 antibody is a pharmaceutical composition characterized in that it specifically binds to the C terminus of IGSF1.
  5. 항-IGSF1 항체 또는 이의 단편을 개체에게 투여하는 단계를 포함하는 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암에 대한 예방 또는 치료 방법.A method for preventing or treating any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer, comprising administering an anti-IGSF1 antibody or fragment thereof to a subject.
  6. 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암의 예방 또는 치료에의 이용을 위한 항-IGSF1 항체 또는 이의 단편.An anti-IGSF1 antibody or fragment thereof for use in the prevention or treatment of any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer.
  7. 대장암, 담도암 및 두경부암으로 이루어진 군에서 선택되는 어느 하나의 암을 예방 또는 치료하기 위한 의약의 제조를 위한 항-IGSF1 항체 또는 이의 단편의 용도.Use of an anti-IGSF1 antibody or fragment thereof for the manufacture of a medicament for preventing or treating any one cancer selected from the group consisting of colon cancer, biliary tract cancer, and head and neck cancer.
PCT/KR2023/016436 2022-10-24 2023-10-23 Pharmaceutical composition for cancer treatment comprising anti-igsf1 antibody WO2024090916A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2022-0137536 2022-10-24
KR1020220137536A KR20240057508A (en) 2022-10-24 2022-10-24 Pharmaceutical composition comprising anti-igsf1 antibody for treating cancer

Publications (1)

Publication Number Publication Date
WO2024090916A1 true WO2024090916A1 (en) 2024-05-02

Family

ID=90831271

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/016436 WO2024090916A1 (en) 2022-10-24 2023-10-23 Pharmaceutical composition for cancer treatment comprising anti-igsf1 antibody

Country Status (2)

Country Link
KR (1) KR20240057508A (en)
WO (1) WO2024090916A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110563845A (en) * 2019-09-12 2019-12-13 滨州医学院 anti-IGSF 9 antibody, pharmaceutical composition and application thereof
EP3176269B1 (en) * 2014-07-29 2020-12-02 Wellmarker Bio Co., Ltd. Inhibitors of met and igsf1 for treating cancer
KR20210122185A (en) * 2020-03-31 2021-10-08 웰마커바이오 주식회사 Pharmaceutical composition for preventing or treating cancer comprising antibodies against igsf1, and method for treating cancer using the same
WO2023022271A1 (en) * 2021-08-20 2023-02-23 웰마커바이오 주식회사 Anti-igsf1 antibody and use thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3176269B1 (en) * 2014-07-29 2020-12-02 Wellmarker Bio Co., Ltd. Inhibitors of met and igsf1 for treating cancer
CN110563845A (en) * 2019-09-12 2019-12-13 滨州医学院 anti-IGSF 9 antibody, pharmaceutical composition and application thereof
KR20210122185A (en) * 2020-03-31 2021-10-08 웰마커바이오 주식회사 Pharmaceutical composition for preventing or treating cancer comprising antibodies against igsf1, and method for treating cancer using the same
WO2023022271A1 (en) * 2021-08-20 2023-02-23 웰마커바이오 주식회사 Anti-igsf1 antibody and use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
YAOYAO GUAN; YINGHAO WANG; ADHEESH BHANDARI; ERJIE XIA; OUCHEN WANG: "IGSF1: A novel oncogene regulates the thyroid cancer progression", CELL BIOCHEMISTRY AND FUNCTION., BUTTERWORTH, GUILDFORD., GB, vol. 37, no. 7, 25 July 2019 (2019-07-25), GB , pages 516 - 524, XP071523442, ISSN: 0263-6484, DOI: 10.1002/cbf.3426 *

Also Published As

Publication number Publication date
KR20240057508A (en) 2024-05-03

Similar Documents

Publication Publication Date Title
JP4854912B2 (en) Antibodies against cancer
US8734799B2 (en) Unconjugated anti-TfR antibodies and compositions thereof for the treatment of cancer
US20240117045A1 (en) Treatment of head and neck cancer
US11999788B2 (en) Treatment or prevention method of radiation damage by administration of IL-5 receptor alpha chain binding antibody
KR20180008571A (en) Treatment of multiple myeloma (MM)
US20190270802A1 (en) Treating cancer with a combination of a pd-1 antagonist and an il-27 antagonist
KR20010101446A (en) Use of antibodies for anticancer vaccination
WO2024090916A1 (en) Pharmaceutical composition for cancer treatment comprising anti-igsf1 antibody
US20230416367A1 (en) Combination therapy of anti-pd-1 active agent, anti-tim-3 active agent, and anti-lag-3 active agent for treating cancer
EP4386000A1 (en) Recombinant anti-human-cd25 antibody and use thereof
WO2023022271A1 (en) Anti-igsf1 antibody and use thereof
WO2023211068A1 (en) Pharmaceutical composition for cancer treatment, comprising anti-igsf1 antibody and anti-pd-1 antibody
WO2020098785A1 (en) Humanized anti-human pd-l1 monoclonal antibody and preparation method therefor and use thereof
US20210087281A1 (en) Hgf-met agonist for use in the treatment of cancer and colorectal fibrosis
WO2022005068A1 (en) Antibody specifically binding to lgals3bp, and use thereof
WO2023027507A1 (en) Antibody drug for prevention or treatment of autoimmune diseases
WO2023182657A1 (en) Anti-fetuin-b antibody and composition comprising corresponding antibody as active ingredient for prevention, treatment, or alleviation of muscle diseases
WO2023191470A1 (en) Antibodies that specifically bind to api5 protein
WO2022005171A1 (en) Composition for treating coronavirus disease, comprising anti-ang2 antibody, and use thereof
WO2024085726A1 (en) Anti-cancer composition comprising anti-vsig4 antibody
EP4349858A1 (en) Chimeric antigen receptor specifically binding to cd300c antigen or receptor thereof
WO2024106939A1 (en) Antibody specifically binding to b7-h3
WO2022131687A2 (en) Anti-hvem antibody, and composition and method associated with same
TWI665215B (en) A monoclonal antibody inhibiting immunosuppressive functions of pathogens, antigen-binding fragments thereof, and hybridomas producing such antibody
Michieli HGF-MET AGONIST FOR USE IN THE TREATMENT OF CANCER AND COLORECTAL FIBROSIS

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: 23883022

Country of ref document: EP

Kind code of ref document: A1