WO2019132517A1 - Method and apparatus for analyzing intracellular or intercellular protein-protein interaction - Google Patents

Method and apparatus for analyzing intracellular or intercellular protein-protein interaction Download PDF

Info

Publication number
WO2019132517A1
WO2019132517A1 PCT/KR2018/016675 KR2018016675W WO2019132517A1 WO 2019132517 A1 WO2019132517 A1 WO 2019132517A1 KR 2018016675 W KR2018016675 W KR 2018016675W WO 2019132517 A1 WO2019132517 A1 WO 2019132517A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
signal
cell
test sample
interaction
Prior art date
Application number
PCT/KR2018/016675
Other languages
French (fr)
Korean (ko)
Inventor
이홍원
나유진
최병산
이대희
Original Assignee
주식회사 프로티나
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from KR1020180034749A external-priority patent/KR20180117529A/en
Application filed by 주식회사 프로티나 filed Critical 주식회사 프로티나
Publication of WO2019132517A1 publication Critical patent/WO2019132517A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids

Definitions

  • One example includes measuring protein-protein interactions between a first protein and a second protein
  • the first protein is a protein involved in a signaling pathway between cells or tissues and / or between cells or tissues
  • the drug targeting the first protein may be a drug targeting at least one of the two or more first proteins
  • the target treatment may be a treatment targeting one or more of the two or more kinds of the first protein.
  • the treatment may include prescribing and / or administering a drug targeting the first protein. (E.g., cancer) associated with the first protein-mediated signal transduction or interactions between the first protein and the second protein (i. E., An individual that is at risk of contracting An individual requiring the prevention and / or treatment of diseases associated with signal transduction or interactions between the first protein and the second protein).
  • An individual suitable for treatment targeting the first protein is one in which the drug targeting the first protein has a desired effect (e. G., Signal transduction mediated by the first protein or interaction between the first protein and the second protein, Or prevention and / or treatment and / or relief of related diseases (anticancer effect when the disease is cancer), and the like.
  • Another example is a first protein as a target suitable for application to the cell or tissue, or to an individual from which the cell or tissue is derived, wherein the step of measuring the protein-protein interaction is performed on two or more first proteins, A method for screening a drug that targets the drug, or a method for providing information to screening.
  • Another example is a cell or tissue treated with a drug that targets the first protein or a cell or tissue isolated from a subject to which the drug is administered and / - measuring the protein or protein interaction, or a drug targeting said first protein of said subject and / or a signal transduction or first signal transduction involving said first protein and low 12 protein,
  • a candidate protein targeted to a first protein is isolated from a separate cell or tissue that has been treated, or within a cell or tissue separated from the subject to which the drug is administered, and / or between cells or tissue, wherein the first protein-mediated drug and / or the first protein-second protein-mediated signal transduction involving the step of measuring liver protein-protein interactions or the interaction between the first protein and the second protein
  • a method for screening a therapeutic agent for a disease associated with the action comprising the steps of:
  • step (3) immediately determining a signal from the reactant obtained in step (2).
  • the value for the unit amount of the first protein contained in the test sample obtained in the step (4) or (4-2) may be a value obtained by summing the values obtained for the respective first protein and the respective second proteins.
  • the reference sample may be a normal cell, a cell in which the degree of activation of the signal transduction pathway involved in the first protein is confirmed, or a signal transduction pathway in which the first protein is involved 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the degree of activation may be that of cells isolated from the identified individual.
  • Another embodiment is a method of manufacturing a semiconductor device comprising steps (1), (2) and (3), or (1), (2), (3), and (4) , (4-1) and (4-2), and in addition thereto,
  • the first protein is at least two selected from among proteins involved in a cell or tissue and / or a signal transduction pathway between cells or tissues,
  • a protein is a protein that interacts with the aforementioned protein
  • Step (5) comprises comparing the results obtained for the two or more first proteins with each other,
  • the test sample may be a lysate, a lysate, an extract, or a body fluid of a cell, a tissue, a cell or a tissue isolated from a mammalian body, and the lysate of the cancer cell, the cancer tissue, the cancer cell, , Lysates, or extracts.
  • the labeling substance may be at least one selected from the group consisting of small molecule compounds, proteins, peptides, and nucleic acid molecules that generate signals measurable through enzyme reaction, fluorescence, luminescence, or radiation detection.
  • the labeling substance of step (2) is at least one selected from the group consisting of small molecule compounds, proteins, peptides, and nucleic acid molecules that generate fluorescence, and the step of measuring the signal of step (3) A microscope or a fluorescence camera, or both.
  • the exposure time per frame of the fluorescent camera may be about 0.0 () 1 second to about 1 second.
  • the candidate compound may be further selected as a candidate drug targeting the first protein.
  • a multi-well comprising a substance for capturing a first protein that specifically binds to a first protein
  • the multiwell includes a plurality of tubes whose one surface is open or a plurality of non-through holes spaced apart from the support plate,
  • a sample injecting portion located on one open side of the tube or non-through hole, a reaction portion where protein-protein interaction occurs in the tube or non-through hole, and
  • the surface of the capturing portion of the first protein is surface-treated with a compound containing a functional group selected from the group consisting of an aldehyde group, a carboxyl group and an amine group.
  • the surface treatment may be performed using biotin, biotin-conjugated bovine serum albumin, polyethylene glycol (PEG), polyethylene glycol-biotin, or polysorbate It can be done.
  • the surface treatment may further include at least one selected from the group consisting of neutravidin, streptavidin, and avidin.
  • the multiwall may further comprise at least one second protein that interacts with the first protein on the signal transduction pathway, labeled with a labeling substance that generates a detectable signal.
  • the multi-well may be a well-known method in which interaction occurs between the first protein and the second protein
  • One or more wells including a reaction part (first reaction part) and a reaction part (second reaction part) in which a first protein and a detection substance binding to the low U protein bind to each other .
  • the first protein and the second protein are independently in cells or tissues and /
  • the second protein is a protein that interacts with the first protein in the signal transduction pathway (for example, a protein involved in a lower pathway than the first protein, A ligand of a protein, a protein which is located on the same cell as the first protein, on the surface of allogeneic cells or xenogeneic cells, or secreted in a different cell and interacts with the first protein, etc.).
  • the signal transfer may be understood to mean a signal transfer within the cell, a signal transfer from the outside of the cell to the inside of the cell, or a signal transfer between two or more cells of the same or different type (for example, immune signal transmission between immune checkpoints) .
  • the first protein may be a plasma membrane protein.
  • the first protein may be selected from the group consisting of receptor tyrosine kinase, toll-like receptors, G-protein coupled receptors (GPCR), transferrin receptors, low density lipoprotein , R0S1; BCR-Abl l fusion protein; Non-drinking body type kaineise; GTPases (GTPases); hormone receptors; Anti-apoptotic protein; And immune checkpoint proteins, and the like.
  • one of PD-L1 and egg-1 as the first protein, and the other as the second protein may be selected and applied to the method and apparatus.
  • a heterodimer of HER2 and HER3 is selected as the first protein and a sub-protein of HER2 and / or HER3 is selected as the second protein in the cell or tissue and / Applicable to devices.
  • An example includes measuring protein-protein interactions between a first protein and a second protein, wherein said first protein is HER2 and HER3, said HER2 and HER3 forming a heterodimer, HER2, HER3, or HER2 in a cell or tissue, which is a sub-protein of HER2, HER3, or both in a tissue and / or in a signal transduction pathway between cells or tissues. (Or confirmation or determination or analysis) of the signal transduction pathway in which both (e. G., HER2-HER3 heterodimer forms) are involved.
  • Another example provides a method of measuring (or identifying or determining or determining) the degree of activation of the HER2-HER3 heterodimer, comprising measuring the degree of phosphorylation of HER2 and / or HER3 in the HER2-HER3 heterodimer.
  • the second protein may be selected from proteins that bind to the phosphorylating group of the HER2-HER3 heterodimer (e.g., phosphorylated tyrosine). .
  • Another example provides an apparatus for use in the method described above.
  • the method may further include the step of supplying PC-L1 to a substrate to which a substance binding to the PD-L1 is attached to the surface, and attaching the PD-L1 to the substrate before the step of reacting.
  • (A) value i.e., [signal (A) value] / [signal (B) value] for the signal
  • quantifying the signal i.e., [signal (A) value] / [signal (B) value] for the signal.
  • a step of adding a substance competing with PD-L1 or PD-1 to the binding between PD-L1 and PD-1 is performed to perform a step of determining a reaction Chicago signal.
  • the signal (A) or () may be measured in the near-field region.
  • the substance binding to the PD-L1 attached to the substrate may be a substance binding to the end of the PD-L1.
  • a PC-L1 having a label attached to a substrate to which a substance binding to PD-L1 is attached is supplied, and a signal (produced by the reaction between PD-L1 and the substance on the surface of the substrate is measured
  • the signal A may be measured in the near-field region.
  • the method may include measuring the value of the signal A for the signal (i.e., (A) value] / [signal (B) value]) of the candidate substance, and quantifying the candidate substance.
  • the candidate substance is selected as an inhibitor of interaction between PD-L1 and PD- Step may be further included.
  • the first receptor tyrosine phosphorylase and the second receptor tyrosine phosphorylase are different kinds,
  • the prepared receptor tyrosine kinase duplex may be in an activated form, as compared to the case without the method.
  • the first receptor tyrosine phosphorylase may be HER3 and the second receptor tyrosine phosphorylase may be HER2.
  • the cholesterol-like detergent may be at least one selected from 0.05 to 5% (DGTN) and 0.003 to 2% (GDNO) of glycolic acid.
  • the term " protein-protein interaction (PPI) " may refer to a physical and / or chemical association or complex formation between a low U protein and a second protein, , Binding frequency, binding strength (intensity), and binding time.
  • the interaction (binding) between the first protein and the second protein includes not only direct interaction (binding) between them, but also other proteins (proteins located between the first protein and the second protein in the signal transduction pathway) It can be applied indirectly to the impression (bond) for indirect impression.
  • the protein-protein interaction in the present specification may be a single-molecule reaction (reaction between one molecule of the first protein and one molecule of the second protein).
  • Cell or tissue interactions may refer to all interactions taking place between two or more homologous cells or allogeneic tissues, or surface proteins of the same / homogeneous / heterogeneous cell or homologous / heterologous / heterogeneous tissue.
  • the first protein and the second protein are each independently at least one selected from proteins involved in signal transduction pathways of cells or tissues of eukaryotic organisms (for example, multicellular animals, multicellular plants, etc.) Receptor and the other is a ligand, or may be one or more selected from the surface proteins of xenogeneic cells .
  • one or more of the first and second proteins may be phosphorylated (e.g., one or more of the first and second proteins may comprise phosphorylated tyrosine (p-Tyr) ).
  • p-Tyr phosphorylated tyrosine
  • the interaction between the first protein and the second protein may be dependent on the degree of phosphorylation (p-Tyr level).
  • one or more of the first and second proteins may have a surface variation that affects interactions between them. The mutation is not dependent on the presence or level of the protein of interest to interact with on the signal path (e.g., if the first protein is a receptor, irrespective of the presence or level of the interacting ligand or subprotein) (E. G., Exon 19 and / or exon 21 mutation (deletion) of EGFR, etc.).
  • the first protein may refer to one or more (e.g., one to ten or two or ten) proteins involved in the signal transduction pathway.
  • the second protein is a protein that interacts with (binds to) the first protein.
  • the second protein may be one or more proteins selected from the proteins involved in the lower pathway of the signal transduction pathway involved in the first protein, Ten to ten or two to ten species) proteins.
  • the first protein is two or more kinds, the second protein may be independently selected for each of the first proteins, and the second protein selected for each first protein may be different, partially or completely It can be duplicated.
  • the first protein is selected from among proteins associated with pathological conditions (e.g., cancer, inflammation, other immune diseases, etc.), so that the pathological conditions (e.g., cancer, inflammation, (And / or < / RTI > improve and / or alleviate) the disease.
  • the first protein may be a protein that is a therapeutic target of the disease to be treated.
  • the first protein may be a target therapeutic agent for a disease to be treated or a target protein of a therapeutic agent to be tested. Accordingly, the first protein can be appropriately selected depending on the therapeutic agent to be sought for the disease or effect to be treated.
  • the first protein is a protein involved in the mediator pathway in the biological signal transduction pathway of a cell or a tissue, and a cell surface protein or a protein secreted (released) by xenogeneic cells (for example, exo And the like), and can be advantageously used as a therapeutic target of the drug
  • xenogeneic cells for example, exo And the like
  • one or more of the cell membrane proteins having a domain exposed to an extracellular environment (for example, an aqueous environment) and present in the cell membrane may be selected.
  • the first protein may be various receptors present in cell membranes, structural proteins associated with microfilaments, cell adhesion molecules, membrane enzymes, membrane receptors, And at least one selected from the group consisting of all kinds of cell membrane proteins, such as lipid-anchored proteins, carrier proteins, chamel proteins, transport proteins, lipid-anchored proteins, .
  • the first protein is selected from the group consisting of receptor tyrosine kinase (RTK) (e.g., epidermal growth factor receptor (EGFR; ErbBl), human epidermal growth factor receptor 2 protein (HER2) ErbB2), HER3 (Human Epidermal Growth Factor Receptor 3 protein: ErbB3), hepatocyte growth factor receptor (HGFR): MET, platelet-derived growth factor receptors (PDGFR, VGFR1, VEGFR2, VEGFR3, etc.), Insulin-like Growth Factor 1 Receptor (IGF1R, VEGFR, EGFR receptors, fibroblast growth factor receptors (FGFRs such as FGFR1 and FGFR2), insulin-like growth factor receptors (e.g., 1 ike Growth Factor Receptor; IGFR, such as IGF1R), c-KIT, RET receptor tyrosine kinase, Anaplastic lymphoma kinase (ALK), etc.
  • the c-Met protein may be derived from any species and includes, for example, a human c-Met (c-Met) (E. G., NP_000236.2), a c-Met (e. G., Macaca mulatta,
  • NPJ301162100 rodent-derived ones such as mouse c-Met (e.g., NP_032617.2), rat c-Met (e.g., NP_113705.1), and the like.
  • a protein includes, for example, a polypeptide encoded by the nucleotide sequence provided in GenBank Ace Session Number __00 (crime 45.3, or a protein comprising the amino acid sequence provided in GenBank Aceession Number NP_000236.2, or an extracellular domain thereof.
  • the receptor tyrosine kinase c-Met is involved in various mechanisms such as, for example, cancer development, cancer metastasis, cancer cell migration, cancer cell infiltration, and neovascularization process.
  • RTKs receptor tyrosine kinases
  • these EGFR, HER2, and / or HER3 EGFR, HER2, or HER3 may be a primate such as human, monkey, mouse, And may be derived from mammals such as rodents such as rats.
  • the EGFR can be obtained from GenBank Accession Nos. JQ739160, JQ739161, JQ739162, JQ739163, JQ739164, JQ739165, JQ739166, JQ739167, NM_005228.3, NM_201284.1,
  • NM_201282.1 or a polypeptide encoded by the nucleotide sequence (A) provided in NM_201283.1 or the like.
  • the HER2 may be derived from GenBank Accession No. < / RTI > Or may be a polypeptide encoded by a nucleotide sequence (mRNA) provided in X03363.1 or the like.
  • the HER3 can be obtained from GenBank Accession No. < / RTI > And may be a polypeptide encoded by a nucleotide sequence (mRNA) provided in NM_001982 or the like.
  • VEGFR Growth Factor Receptor
  • VEGF vascular endothelial growth factor
  • Overexpression of VEGFR is a cause of various diseases, and it is involved not only in the development of cancer but also after bad cases such as invasion and metastasis. For this reason, VEGF is an important target in chemotherapy.
  • the VEGFR may be derived from mammals such as primates such as humans and monkeys, and rodents such as mice and rats.
  • the VEGFR may be a polypeptide encoded by a nucleotide sequence (mRNA) provided in GenBank Accession Number AF063657.2.
  • PDGFR platelet-derived growth factor receptors
  • the above-mentioned “ platelet-derived growth factor receptors (PDGFR) is one of the surface receptor tyrosine kinases, and is associated with many diseases that cause cell growth, cell differentiation, cell growth.
  • the PDGFR may be derived from mammals such as primates such as humans and monkeys, and rodents such as mice and rats.
  • the PDGFR can be obtained from GenBank Accession Nos. NM_006206.4 (PDGFR-A), NM_002609.3 (PDGFR-B),
  • SEQ ID NO: 1 SEQ ID NO: 1
  • mRNA nucleotide sequence provided in SEQ ID NO.
  • the IGF1R may be derived from a mammal such as a primate such as a human, a monkey, a rodent such as a mouse or a rat, etc.
  • the IGF1R may be derived from a nucleotide sequence encoded by nucleotide sequence (A) provided in GenBank Accession No. NM - 2019/132517 1 »(: 1/10/06 018/016675
  • ephrins receptors are one of the surface receptor tyrosine kinases, which regulate the axon guidance, the format ion of the ssue boundaries, the cell migrat ion, and the segmentation ion.
  • the ephrin receptor may be derived from mammals such as primates such as humans and monkeys, and rodents such as mice and rats.
  • the ephrin receptor may be derived from GenBank Accession Nos. NM_004440.3, NM_004438.3, NM_004431.3,
  • Or may be a polypeptide encoded by a nucleotide sequence (mRNA) provided in NM_182690.2, NM_020526.3, NM_001406.3, NM_005227.2, NM_182685.1, and the like.
  • mRNA nucleotide sequence
  • the "transferrin receptors” are carrier proteins of transferrin, which participate in the intracellular transport of iron through receptor-mediated endocytosis and regulate intracellular iron concentrations.
  • the transferrin receptor may be derived from a mammal such as a rat, a primate such as a monkey, or a rodent such as a mouse or a rat.
  • the transferrin receptor may be derived from GenBank Accession Nos. NM_001128148.1, NM_003234.2, NM_001206855.1,
  • the " low-density lipoprotein (LDL) receptor is a carrier protein of transferrin, which participates in the intracellular transport of iron through receptor-mediated endocytosis and regulates intracellular iron concentration.
  • the LDL receptor may be derived from mammals such as primates such as humans and monkeys, and rodents such as mice and rats.
  • the transferrin receptor may be a GenBank
  • Or may be a polypeptide encoded by a nucleotide sequence (mRNA) provided in NM_001195803.1, NM_001195800.1, NM_001195798.1, and the like.
  • mRNA nucleotide sequence
  • cluster of diatoms is a protein that functions as a receptor or a ligand in various manners, and about 350 kinds of proteins are known in humans. Cel l signal ing, and cell adhesion.
  • the surface-differentiating antigens may be derived from primates such as humans and monkeys, and mammals such as rodents such as mice and rats.
  • the surface-differentiating antigens may be of any CD sequence, and in particular may be CD44, CD147, or their antigens associated with cancer metastasis, and more particularly GenBank Accession Nos.
  • RNA provided in the nucleotide sequences (NM_000610.3, NM_001728.3, X55150.1) and the like.
  • GPCR G-protein coupled receptor
  • the GPCR may be derived from mammals such as primates such as human, primate, rodent such as mouse and rat.
  • GPCRs can be chemokine receptors (Rhodopsin-1 ike receptor subfamily) associated with cancer metastasis, such as CXC chemokine receptors, CC chemokine receptors, CX3C chemokine receptors, etc., and more specifically GenBank Accession Nos.
  • chemokine receptors Rhodopsin-1 ike receptor subfamily
  • CXC chemokine receptors CXC chemokine receptors
  • CX3C chemokine receptors CX3C chemokine receptors
  • mRNA nucleotide sequence
  • the second protein may be any protein that interacts with the first protein selected as described above, for example, a protein involved in a sub-pathway of a biological signal transduction pathway of a cell or tissue involved in the first protein, A ligand, a protein which is present on the surface of a xenogeneic cell or released from a xenogeneic cell, and the like, or may be selected from two or more (for example, one to ten or two to ten) species.
  • Cells or tissues such as the signaling pathways and the proteins involved in human cells or tissues, ligands for specific receptors, xenogeneic cell surface proteins interacting with specific proteins present on the cell surface, Relatively well established (Untangling the ErbB signaling network, Nat. Rev. Mol. Cell Biol. 2, 127 (2001); Cel l 141, 1117 (2010)).
  • the selection of a protein involved in the downstream signal transduction pathway is not limited to the conventional method of the present invention. It is clear to those who have knowledge.
  • one protein participates in various bio-signal transmission pathways, and a plurality of signal transmission pathways can form a network.
  • one or more of the second proteins for any one first protein may overlap one or more of the second proteins for the other first protein (s) (i.e., 1 < / RTI > proteins, the second proteins for these two or more first proteins may be different, or some or all, the same).
  • the first protein may be a complex of two or more different proteins (first protein-first protein complex).
  • first protein-first protein complex two or more first protein-second protein complexes formed by interaction between a first protein and a second protein are linked (e.g., a bond between a second protein of a neighboring first protein-second protein complex) , And can be a cluster.
  • the first protein may be at least one selected from the group consisting of EGFR, MET, HER2, and HER3, and the second proteins for each first protein may be the same or different, , Phospholipase C (PLC) (e.g., PLC-gamma (PLC-gamma 1) (e.g., GenBank Accession No.
  • PLC Phospholipase C
  • PLC-gamma PLC-gamma 1
  • NP_002651.2 NP_877963.1, NP_037319.1, etc.
  • its SH2 domain including at least one of Src homology 2 domain: N-terminal SH2 domain and C-terminal SH domain;
  • Src homology 2 domain N-terminal SH2 domain and C-terminal SH domain;
  • amino acid sequence regions 545 to 765 in NP_037319.1 or amino acid sequence regions 540 or 545 to 765 in NP_002651.2 growth factor receptor-binding protein Grb (e.g., Grb2 (see, for example, GenBank Accession No. 2).
  • Grb growth factor receptor-binding protein
  • NP_002077.1 e.g., Grb2 (see, for example, GenBank Accession No. 2).
  • NP_002077.1, NP_987102.1, etc. or a portion thereof (e.g., the SH2 domain (NP_002077.1
  • SH3_N-SH2 domain (Amino acid sequence positions 57 to 155), SH3_N-SH2 domain (the first to 154th amino acid sequence regions of NP_002077.1), SH2- SH3_C domain (the 57th to 217th amino acids of NPJ302077.1 Amino acid sequence Site)), phosphatidylinositol 3-kinase modulating subunit
  • NP_001229395.1, NP_852556.2, NP_852664.1, NP_852665.1, P26450.2, etc. or its SH2 (SEQ ID NO: (Amino acid sequence region from 333rd to 428th), SH2_C domain (amino acid sequence region from 624th to 718th), or tandem SH2 domain (from the 333rd to the 428th amino acid sequence region) of NP_852664.1 (human p85a) (Amino acid sequence region from position 624 to position 718) or SH2_N domain (from position 333 to position 428) of P26450 (mouse p85a), SH2_C domain (amino acid sequence region from position 624 to 718), or tandem SH2 domain Amino acid sequence region from position 333 to position 718)), and the like, but the present invention is not limited thereto.
  • the combination of EGFR, MET, HER2, and HER3 as the first protein and PLC-gamma 1, Grb2, and p85_alpha as the second common proteins of these first proteins In the case of breast cancer, a combination of HER2 and HER3 is used as the first protein and PLC-gamma 1, Grb2, and p85-alpha are used as common second proteins of these first proteins.
  • the present invention is not limited thereto and can be appropriately selected according to the therapeutic agent to be sought based on the above-described diseases or effects to be treated, and it is obvious to those skilled in the art to which the present invention belongs.
  • the first protein may be at least one selected from the group consisting of EGFR, MET, HER2, and HER3, and the second protein to the first protein may be selected from among ligands binding to the respective first protein (Eg, EGFR epidermal growth factor (EGF), MET hepatocyte growth factor (HGF), HER2 and HER3 Neuregul ins (NRGs)).
  • EGF epidermal growth factor
  • HGF MET hepatocyte growth factor
  • NSGs Neuregul ins
  • the first protein is any one selected from PD-L1 and egg-1, and the second protein is the other.
  • the first protein is egg-1 and the second protein is PD-Ll.
  • the first protein is PD-L1 and the second protein is PD-1.
  • PD-L1 Programmed death-l igand 1 is a protein present on the surface of cancer cells. CD274 (cluster of diiferent ion 274) or B7_H1 (B7 homolog 1) and is a 40 kDa type 1 transmembrane protein. PD-L1 is present in subtypes of immune cells (Macrophage, etc.) as well as cancer cells. PD-L1 is known to play a major role in suppressing the immune system in various diseases such as tissue graft, immune diseases and hepatitis. The PD-L1 described in the present specification may be derived from a mammal such as a human, and examples thereof include human PD-L1 (eg, GenBank Accession No. NP_001254635.1 (gene: NM_014143.3), NP_001300958.1 NM_001314029.1),
  • NP_054862.1 (gene: NM_001267706.1), etc.).
  • PD-1 Programmed cell death protein 1
  • CD279 cluster of di fi erent iat ion 279
  • CD279 cluster of di fi erent iat ion 279
  • NP_005009.2 (gene: NM_005018.2), etc.).
  • T cells lose their ability to attack cancer cells.
  • PD-L1 and / or egg-1 may be important targets in the development of anti-cancer drugs.
  • the first protein and / or the second protein may be used in a form contained in a cell, a tissue, or a lysate or an extract thereof, or in the form of a purified protein.
  • the first protein is HER2 (Human Epidermal Growth Factor Receptor 2 protein; ErbB2) and Human Epidermal Growth Factor
  • Receptor 3 protein ErbB3
  • HER2 and HER3 may be heterodimers (HER2-HER3 heterodimers)
  • HER2 and HER3 are members of the HER family receptor tyrosine kinases (RTKs), respectively. Binding of a ligand to the extracellular domain of HER2 or HER3 induces receptor homo- or hetero-dimerization with other ErbB receptors, which leads to intracellular autophosphorylation of specific tyrosine residues.
  • HER2 and / or Autophosphorylation of HER3 leads to downstream signaling networks including MAPK and PI3K / Akt activation that affect cell proliferation, angiogenesis and metastasis.
  • HER2 overexpression and gene amplification are frequently observed in breast and stomach cancer and are associated with poor prognosis of cancer treatment and poor clinical outcome.
  • HER3 on the other hand, is well known as a resistance mechanism for several target drug therapies. For these reasons, these HER2 and / or HER3 are important targets in chemotherapy.
  • HER2 and HER3 may be derived from mammals such as humans.
  • HER3 was obtained from GenBank Accession No. NP_001005915.1, NP_001973.2, and the like, but are not limited thereto.
  • the second protein is a sub-protein of HER2 and / or HER3 (e.g., HER2-HER3 heterodimer) in various vital signal transduction pathways and interacts with HER2 and / or HER3 (e.g., HER2-HER3 heterodimer) ). ≪ / RTI > In one example, the second protein may bind to a phosphorylating group (e.g., phosphorylated tyrosine) of HER2 and / or HER3 (e.g., HER2-HER3 heterodimer).
  • a phosphorylating group e.g., phosphorylated tyrosine
  • Measuring the protein-protein interaction between the first and second proteins may comprise the following steps:
  • step (3) immediately determining a signal from the reactant obtained in step (2)
  • measuring the protein-protein interaction between the first protein and the second protein comprises, in addition to the steps (1) to (3), (4) 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the step (4) of measuring the activation level of the first protein using the signal measured in the step (3) includes the step of measuring the activation level of the first protein in the test sample added in step (1) And obtaining a signal value for the unit amount.
  • the first protein is any one selected from? - 11 and egg-1, the second protein is And? ⁇ ⁇ 1.
  • the first protein may be a quadrivalent 2 - 1, 3 heterodimer.
  • the step of measuring the degree of tyrosine kinase activation (e.g., phosphorylation of the tyrosine residue) of the other 1? 2 and / or 4? 3 in the 4 1? 2? 4 thymine 3 heterodimer may include the following steps :
  • the phosphorylating step may include treating the phosphorylating agent to the HER2-HER3 heterodimer immobilized on the substrate or the substrate.
  • the phosphate reagent may be a HER2 and / or can be at least one selected from every substances capable of HER3 phosphorylation, e.g., ATP and magnesium (e.g., MgCl 2, etc.), without being limited thereto.
  • step (3-1) at least one selected from cholesterol-like detergents including DGTN, GDN and the like is added to the HER2-
  • the detergent may be treated with a CMC (cortical microcrystalline) concentration or higher to maintain the structural and / or functional activity of the HER2-HER3 heterodimer.
  • the first protein is a HER2-HER3 heterodimer
  • the following step (4) and / in addition to the above-described step (1) to (3) or (1-1) to (3-1), the following step (4) and / ).
  • Step (1) or (1-1) Substrate preparation step in which the first protein is immobilized
  • the step (1) is a step of preparing a substrate to which a first protein is immobilized by adding a test sample containing a first protein to a substrate containing a substance that specifically binds to the first protein on the surface.
  • the first protein is as described above.
  • test sample may be any biological sample that can be used to test the response to a drug targeting the activating first protein in the cell or tissue and / or the signal transduction pathway between cells or tissues.
  • the test sample may be a lysate, a lysate, or an extract of a cell, a tissue, a cell or a tissue isolated from an individual, a body fluid (e.g., blood (whole blood, plasma or serum), saliva, etc.).
  • the subject is preferably selected from the group consisting of an activation test of a signal transduction pathway in a cell or tissue and / or a cell or tissue involved in the first protein, a reactivity test for a drug targeting the first protein, of the crystal, the first protein target therapeutic effect (For example, primates such as human, monkey, rodents such as mice, rats, etc.) which are to be used for monitoring and / or selecting effective first protein target therapeutic agents.
  • the subject may be a patient having a disease associated with the first protein.
  • the disease associated with the first protein may be a disease associated with overexpression of the first protein or activation of a signal transduction pathway in the cell or tissue and / or between the cell or tissue involved in the first protein, for example, the cancer.
  • the test sample is used to determine the degree of activation of the signal transduction pathway in a particular cancer patient (e. G., The cell or tissue involved by the first protein and / or between the cell or tissue, For example, a cancer cell, to determine whether it is an appropriate subject for a first protein target treatment, or to isolate a specific entity from which an effective first protein target therapeutic agent is to be selected).
  • tissue In the case of tissue, a size of at least 125 mm 3 is sufficient for grinding, and the amount of tissue sample required for one measurement may be in the range of about 1/50 to about 1/75 of the amount of tissues mentioned above (at least 125 mm 3 ) But is not limited thereto.
  • the amount of cellular sample required for one-time measurement of about 10 cel ls to 10 10 cel ls, of about 10 cel ls to 10 7 cel ls, of about 10 cel ls to 10 5 cel ls, of about 10 3 cel ls to 10 10 cel ls, of about 10 3 cel ls to 10 7 cel ls, of about 10 3 cel ls to 10 5 cel ls, for example, may be an order of about 10 4 + 50 cel ls-, it is not limited to, cell lines Which is a value that can be appropriately determined according to the type of the film.
  • the test sample may comprise a treatment that targets the first protein (e.g., Administration of a drug targeting the protein) may be a cell or tissue that has not been subjected to an action, or a cell or tissue that has been separated from a subject in which the treatment (or administration of the drug) has not been performed,
  • the substrate may be of any material and / or of any structure capable of immobilizing the first protein on its surface (both crystalline and amorphous 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the substrate may be a material having a refractive index of water equal to or higher than that of water (about 1.3), which occupies a large portion of the biomaterial, taking into consideration the easiness of detecting a beacon signal.
  • the substrate thickness reducible 0.1 to about 1 _ from about 0.1 to about 0.5 ⁇ , 0.1 to about 0.25 1®, or about 0.13 to about 0.21, and ⁇ be on the order of a refractive index of about 1.3 to about 2, about 1.3 About
  • the substrate may be any material that satisfies the refractive index range and may be, for example, from a material selected from the group consisting of glass (refractive index: about 1.52), quartz, and the like, but is not limited thereto.
  • the substrate may be of any type conventionally used for biological specimen observation and may be, for example, a well type, a slide type, a channel type, an array type, a microfluidic chip, a microtubule (capillary) It is not. When observing a fluorescent microscope, a cover glass can be observed on a substrate to which the sample is applied.
  • the material of the cover glass is as described above for the substrate, and the thickness may be the range described in the substrate or may be thinner (for example, a refractive index of 1.52, 0.0 > 0.17, < / RTI > but not limited thereto).
  • Substance specifically binding to the first protein can be selected from any substance capable of binding to the first protein and specifically, for example, a specific antibody or anti thereof that binds to the first protein: One-binding fragment (E.g., 1, () acid-! 7 , Fab, 1 and (2) 2), platamer (protein or nucleic acid molecule), small molecule compound, and the like.
  • the substance specifically binding to the first protein is a site that does not interfere with the interaction between the first protein and the second protein, that is, a site where the first protein and the second protein interact (bind) And may bind to the first protein at a non-specific site.
  • the substrate is suitably surface-modified to include (immobilize) a biological substance (for example, an antibody or the like) that specifically binds to the first protein on the surface, or may be surface- The binding material may be fixed.
  • the substrate may be treated (e.g., coated) with all of the compounds having a functional group capable of purifying a biological substance (for example, an antibody or the like) that specifically binds to the first protein on one surface,
  • the compound containing a functional group selected from the group consisting of an aldehyde group, a carboxyl group and an amine group may be selected from the group consisting of biotin, bovine serum albumin (BSA), bovine serum albumin bound with biotin, polyethylene glycol (PEG) Biotin-conjugated PEG (polyethylene glycol-biotin), polysorbate (e.g., Tween 20), and the like, but the present invention is not limited thereto.
  • the surface-treated substrate may be further treated (e.g., coated) with at least one selected from the group consisting of neutravidin, streptavidin, avidin, and the like.
  • the first protein may be PD-L1 or egg-1.
  • the first protein may be PD-L1.
  • a substance such as an antibody
  • PD- This clustered spot is formed, which has the advantageous effect of qualitative and / or quantitative analysis.
  • the first protein may be PD-I.
  • a substance for example, an antibody
  • test sample is used to test the level of interaction between PD-L1 and egg-1, the activation of the signal transduction pathway involved in the first protein, and the reactivity to the drug targeting the first protein, among cells or tissues It may be all possible biological samples.
  • complexes formed by binding between PD-L1 and PD-1 are clusters, and the in vivo environment can be simulated more similarly.
  • a substance that binds to the first protein may be a specific antibody or an antigen-binding fragment that specifically binds to PD-L1.
  • the antibody specifically binding to PD-L1 or The antigen-binding fragment may specifically bind to a non-binding site (for example, does not overlap with the binding site) of the PD-L1 binding site, for example, the C-terminal site of PD-L1.
  • the site where PD-L1 binds to PD-1 is generally the N-terminal region (extracellular domain site) of PD-L1, and specifically, the 20th to 150th, 20th to 130th, To 125th,
  • 25 to 150, 25 to 130, or 25 to 125 amino acid residues examples thereof include PD-L1 At least one amino acid residue selected from the group consisting of D26, Y56, E58, R113, A121, D122, Y123, and R125.
  • PD-L1 At least one amino acid residue selected from the group consisting of D26, Y56, E58, R113, A121, D122, Y123, and R125.
  • Human PD-L1 reference such as NP_054862.1) or a corresponding site (see Fig. 32).
  • the non-binding site except for the binding site of PD-L1 with PD-1 is a PD-L1 site in which the N-terminal PD-L1 of the PD-L1 described above does not overlap with the site binding to PD- And the antibody or antigen-binding fragment thereof that specifically binds to PD-L1 binds to the PD-
  • an antibody or an antigen-binding fragment which binds to 1 to 10 amino acids.
  • the antibody or antigen-binding fragment thereof that specifically binds to PD-L1 binds to the PD-L1 protein in the N-terminal to C-terminal direction, after the 250th, after the 260th
  • the amino acid sequence may be one that specifically binds to one or more amino acids selected from the amino acid positions after the 280th position, for example, from the 250th position to the 300th position, or from the 280th position to the 290th position.
  • the substance specifically binding to the first protein may be an antibody or an antigen-binding fragment thereof that specifically binds to egg-1.
  • the antibody or antigen-binding fragment that specifically binds to PD-1 binds to the binding site of PD- Binding site at the C-terminal site of the repressor-1, i.e., not overlapping the C-terminal site of the repressor-1.
  • the site where PD-1 binds to PD-L1 is generally the N-terminal region (extracellular domain site) of egg-1, and specifically the 60th to 200th, 60th to 150th, 60th From the 135th to the 135th, 65 to 150, or 65 to 135 amino acid residues, and examples thereof include a region containing at least one amino acid residue selected from the group consisting of Y68, Q75, K78, T76, 1126, 1134, and the like.
  • the non-binding site other than the binding site of PD-L with PD-L1, for example, the C-terminal site of PD-1 is a site in which the PD-1 at the N- And the antibody or antigen-binding fragment thereof that specifically binds to PD-1 may be a non-binding site other than the binding site of PD-1 with PD-L1 (for example, 1 to 30, 1 to 25, 1 to 20, 1 to 15, or 1 to 10 amino acids in the C-terminal region of PD-1) Can be.
  • the first protein may be a HER2-HER3 heterodimer with HER2 and HER3 dimerized.
  • the sample containing the first protein may comprise HER2 and / or HER3, or a HER2-HER3 heterodimer,
  • a cell isolated from a patient to be tested comprising recombinant cells expressing HER2 and HER3, a cell lysate, or a cell lysate, a recombinant cell engineered to express HER2 and HER3 simultaneously (for example, a recombinant cell expressing HER2 A recombinant cell into which a recombinant vector containing a HER3 gene is introduced into a cell, a recombinant cell into which a recombinant vector containing a HER2 gene is introduced into a cell expressing HER3, etc.), or a lysate or a lysate of the cell.
  • the heterodimer of HER2 and HER3 may be characterized by treating the dimerization agent with cells that simultaneously express or express HER2 and HER3.
  • the dimerization agent may be a ligand of HER3 (e.g., Neuregulin pi (NRGbl);
  • NP_039250 NP_039250.2, etc.
  • cholesterol-like detergent e.g., DGTN (digitonin; CAS Number: 11024-24-1)
  • GDN cholesterol-like detergent
  • NRGbl when NRGbl is treated (injected) into HER2 and HER3 expressing cells to form a HER2-HER3 heterodimer, the NRGbl may be injected into the cell prior to lysis after HER2 and HER3 expression have.
  • a cell that simultaneously expresses HER2 and HER3 (including) a cholesterol-like detergent to treat HER2-HER3
  • the cholesterol-like cleansing agent may be treated (added) after cesiolysis and prior to measurement of activation of HER2 and / or HER3 (in one example, the cell has an NRGbl of between 10 ng / mL, 50 ng / mL to 200 ng / mL, or 10 ng / mL to 100 ng / mL).
  • the concentration of the cholesterol-like detergent to be used may be in excess of the concentration of the cleaning agent used, for example, the concentration of the cholesterol-like detergent may be, for example, for DGTN 0.05% (w / v) or more, 0.05% (w / v) in excess, or 0.06% (w / v) or more, e.g., 0.05 to 2% (w / v) or 0.06 to 23 ⁇ 4> (w ⁇ ), 0.003% (w or more, 0.005% (w) or more, 0.007% (w or more), or 0.01% (w / v) or more, such as 0.003 to 2% v), 0.005 to 2% (w / v), 0.007 to 2% (w / v), or 0.01 to 23 ⁇ 4 > ( w 8 0) It is difficult to maintain the membrane permeability of the protein and maintain the tyrosine kinase activity, and the activity of the HER2-HER3 heterodimer (for
  • Digi tonin is highly unstable at dissolution and precipitates within a few hours, so it is recommended that 10 to 20% (w / v) or 8 to 12% (w / v) GDN is stable for several days even when dissolved GDN is stable for several days All solutions can be maintained at pH 7 to 8.7, 2 to 7.6, or 7.4 using HEPES buffer, and NaCl
  • protease inhibi tor cocktail can be added (eg, Sigma-Aldrich P8340, approximately 2% (w / v)).
  • the substance specifically binding to the low U protein may be selected from the first protein, i.e., HER2, HER3, or any substance capable of specifically binding to all of them (ScFv) 2, scFv-Fc, Fab, Fab 'and F (ab') 2 (SEQ ID NO: 2) of an antibody that specifically binds to HER2, HER3, Etc.), platamer (protein or nucleic acid molecule), small molecule compound, and the like.
  • a substance that specifically binds to the first protein is a region that does not interfere with the interaction between the first protein and the second protein, that is, In the region where the second protein does not interact (bind), the first It can be associated with proteins.
  • Step (2) Step of reacting the first protein task 2 protein
  • the labeled second protein is added to the substrate on which the prepared first protein is immobilized and reacted.
  • the second protein is as described above.
  • the labeled second protein may be labeled with a labeling substance that generates a detectable signal of the second protein (for example, the labeling substance is chemically (e.g., covalent or noncovalent), recombinantly, or physically bound) , And a tag-attached form to which a labeling substance can be bound.
  • the detectable signal may be selected from any signal (e.g., light, radiation, etc.) that can be measured through conventional enzymatic reactions, fluorescence, luminescence, and / or radiation detection.
  • the labeling substance may be at least one selected from the group consisting of all small molecule compounds capable of generating the label signal, proteins, peptides, and nucleic acid molecules.
  • the labeling substance examples include fluorescent dyes (small molecule compounds: Cyanine, Alex, Dylight, Fluoprobes Etc.), fluorescent proteins (e.g., green fluorescent protein (GFP), enhanced GFP), yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), blue fluorescent protein (BPF), red fluorescent protein Lt; / RTI >
  • the tag may be at least one selected from all kinds commonly used such as Hi s-tag / Ni-NTA.
  • the concentration of the labeling substance to be used may be appropriately determined in the range of about luM or less, for example, in order to allow accurate and easy detection without generating noise.
  • the signal generated from such a labeling substance can be measured by any signal detecting means (for example, a conventional fluorescence microscope, a fluorescence camera, a fluorescence intensity measuring apparatus, etc.) commonly used for detecting or measuring the signal.
  • the substrate May be further washed in a conventional manner.
  • the second protein may be PD-L1 or PD-1. In one embodiment, when the first protein is PD-L1, the second protein may be PD-1. In another embodiment, when the first protein is PD-1, the second protein may be PD-L1.
  • the step of reacting the first protein task 2 protein comprises (2-1) phosphorylating the HER2-HER3 heterodimer and contacting the phosphorylated tyrosine- And the like.
  • the step (2) or (2-1) may be carried out by adding the labeled second protein to the substrate on which the prepared first protein is immobilized, And adding the labeled phosphorylated tyrosine specific binding substance to react.
  • the labeled phosphorylated tyrosine specific binding material may be an antibody that specifically binds to phosphorylated tyrosine of HER2 or HER3.
  • the second protein is a sub-protein of HER2 and / or HER3 (e.g., HER2-HER3 heterodimer) in various biological signaling pathways, And / or proteins capable of interacting (binding) with HER3 (e.g., HER2-HER3 heterodimer).
  • the phosphorylation step may comprise treating the substrate or the HER2-HER3 heterodimer immobilized on the substrate with a phosphorylating agent .
  • the phosphate reagent may be a HER2 and / or can be at least one selected from every substances capable of HER3 phosphorylation, e.g., ATP and magnesium (e.g., MgCl 2, etc.), without being limited thereto.
  • HER2-HER3 heterodimer immobilized on the substrate or substrate before, after, or simultaneously with the phosphorylation, and prior to the activation measurement of step (3-1), as described in step (1) or , HER2-HER3 heterodimers at a concentration exceeding one or more detergents selected from cholesterol-like detergents including DGTN, GDN, etc., in excess of the CMC crest micellar concentrate, Or functional activity thereof.
  • Step (3) or (3-1) Measurement of protein-protein interaction step
  • the step (3) is a step of measuring a signal from the reactant obtained in the step (2).
  • the signal measurement may be carried out using a conventional signal (e. G., An enzyme reaction, fluorescence, luminescence, or radiation detection) 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • a signal e.g., a signal measurable via a method.
  • the measurement of protein-protein interaction in step (3) may be by real-time analysis.
  • the label signal is a fluorescence signal
  • the signal detection is performed by supplying a fluorescence signal generated by supplying a light source absorbed by a labeling substance for generating the fluorescence signal to a fluorescence microscope, a fluorescence camera, and / And may be imaged, imaged, and / or quantified using, for example, an intensity measuring device.
  • the fluorescence signal when the signal is a fluorescence signal, the fluorescence signal can be imaged and / or quantified using a fluorescence camera.
  • step (3) protein-protein interaction measurement step
  • the step of supplying the light source of the step 0) is a step of supplying a light source to the reactant of the first protein task 2 protein obtained in the step (2). If the above purpose can be achieved, There is no.
  • the light source may be continuously supplied before step (1), simultaneously or after step (2), or may be supplied for a predetermined time immediately before, simultaneously with, or immediately after step (2) no.
  • the light source may be any light source having a wavelength corresponding to a fluorescent signal, for example, a laser, a halogen lamp, or the like.
  • Wavelength of the light source can be adjusted depending on the fluorescence signal using, for example, it can be selected from about 300 to about 600 ⁇ 1 ⁇ , or about 350 to about 560 TM TM range. More specifically, the green fluorescent protein absorbs about 480 11111 , the yellow fluorescent protein absorbs about 540, the blue fluorescent protein absorbs about 375 < 1 > , and the blue fluorescent protein absorbs about 425,
  • the wavelength of the light source is about 460 to about 500
  • the wavelength of the light source is about 520 to about 560 when the yellow fluorescence is used as the fluorescence signal
  • the blue fluorescence is used as the fluorescence signal
  • the wavelength of the light source is 1 to about 350 to about 400, and when the cyan fluorescence is used as the fluorescence signal,
  • the wavelength can be selected in the range of about 400 to about 450 nm.
  • TIRF Total Internal Ref lect ion Fluorescence
  • a fluorescence camera for signal imaging such as an EMC suppressor
  • CMOS Complementary metal oxide semi conductor
  • step (3) the steps of measuring the protein-protein interaction in step (3) are carried out using a total internal reflection microscope and a fluorescent camera, for example, in more detail:
  • step (1) or step (2) is mounted on the total reflection microscope 5 .
  • the supply position of the light source in the total reflection microscope is normally downward and the fluorescence signal is emitted from above the substrate (in this case, from the lower side to the upper side, the light source supply portion, the substrate, the lens or the substrate, under that it can position in the sequence) or (observed in this case, the upward direction from below, may be located in the lens, the light source supply, substrate, or the light source supply unit, lenses, substrate, and 0, or the lens, substrate, order of the light source supply unit) can do.
  • the light source may be a laser and the intensity of the light source may be from about 0.5 mW to about 5 mW, from about 0.5 mW to about 4.5 mW, from about 0.5 mW to about 4 mW, from about 0.5 mW to about 3.5 mW, mW, about 0.5 mW to about 2.5 mW, about 0.5 mW to about 2 mff, about 1 mff to about 5 mW, about 1 mW to about 4.5 mW, about 1 mW 5 to about 4 mW, about 1 mW to about 3.5 mW From about 1 mW to about 4 mW, from about 1.5 mW to about 4 mW, from about 1 mW to about 3 mW, from about 1 mW to about 2.5 mW, from about 1 mW to about 2 mW, from about 1.5 mW to about 5 mW, 1.5 mff to about 3.5 mW,
  • the fluorescence signal generated by the light source supply is transmitted to the fluorescent camera Imaging and imaging and / or quantification.
  • the imaging (or imaging) of the fluorescence signal can be performed simultaneously with the light source supply or within the signal generation holding time, taking into account the fluorescence signal generation holding time (luminescence time) of the labeling material.
  • the exposure time per one frame, the laser power, the camera gain value, and the total shooting frame can be appropriately adjusted.
  • the laser power can be increased or the sensitivity of the fluorescent camera can be increased.
  • the exposure time per frame is from about 0.0 () 1 second to about 5 seconds, from about 0.001 second to about 3 seconds, from about 0.0 second to about 2 seconds, from about 0.0 () second to about 1 second, from about 0.001 From about 0.01 second to about 5 seconds, from about 0.
  • 1 second to about 3 seconds from about 0.01 second to about 2 seconds, from about 0.01 second to about 0.5 second, from about 0.001 second to about 0.3 second, from about 0.001 second to about 0.1 second, From about 0.01 second to about 1 second, from about 0.01 second to about 0.5 second, from about 0.01 second to about 0.3 second, from about 1 second to about 0.1 second, from about 0.05 second to about 5 seconds, From about 0.05 second to about 2 seconds, from about 0.05 second to about 1 second, from about 0.05 second to about 0.5 second, from about 0.05 second to about 0.3 second, from about 0.05 second to about 0.1 second, from about 0.07 second to about 5 seconds About 0. 07 second to about 3 seconds, about 0. 07 second to about 2 seconds, about 0.
  • 07 second to about 1 second about 0. 07 second to about 0.5 second, about 0. 07 second to about 0.3 second From about 0.07 seconds to about 0.1 seconds, from about 0.1 seconds to about 5 seconds, from about 0.1 seconds to about 3 seconds, from about 0.1 seconds To about 2 seconds, from about 0.1 second to about 1 second, from about 0.1 second to about 0.5 second, or from about 0.1 second to about 0.3 second, such as about 0.1 second.
  • the gain value can be adjusted to a value in the range of from about 10 to about 100, from about 10 to about 80, from about 10 to about 60, from about 10 to about 50, from about 20 to about 100, From about 20 to about 80, from about 20 to about 60, from about 20 to about 50, from about 30 to about 100, from about 30 to about 80, 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the imaging is performed by using one or more, for example, two or more, three or more, four or more, five or more, seven or more, performed on a substrate including one or more channels (each channel is at least two substrates comprising the same according to the determined size and the imaging area as possible)), and the signal appears 31 be
  • the fluorescence intensity measured in step (3) may be quantified using conventional equipment to quantify protein-protein interactions.
  • steps (1) to (3) can be performed for each first protein task 2 protein combination (that is, steps (1) to 3) can be repeated as many times as the number of combinations of the first protein and the second protein.
  • the step (3) of measuring the protein-protein interaction may be applied to the step (3-1) of measuring the phosphorylation of the tyrosine residue.
  • the step (3) or (3-1) is a step of measuring a signal from the reactant obtained in the step (2) or (2-1).
  • the signal measurement can be used to detect (or measure or verify) the label signal used in step (2) or (2-1) (e.g., a signal measurable via conventional means such as enzyme reaction, fluorescence, luminescence, ), which can be performed using any means available.
  • the step of measuring the phosphorylation of the tyrosine residue in step (3-1) comprises the steps of: And detecting and / or quantifying a signal generated in a reaction between a substance (e. G., An antibody) that specifically binds to a phosphorylated tyrosine residue of 1 to 3.
  • a substance e.g., An antibody
  • a fluorescent material e.g., fluorescent Protein
  • phosphorylation of the tyrosine residue can be measured by measuring and quantifying the fluorescence intensity detected in the reactant as described above.
  • Step (4) measuring the activation level of the first protein
  • the step of measuring the activation level of the first protein of the step (4) may include the step of measuring the activation level of the first protein in the test sample added in step (1) using the signal measured in step (3) ).
  • activation of a first protein means interaction (binding) of a first protein with a second protein, and" activation level of a first protein " (Binding), and the "activated first protein " may mean a first protein that interacts with (binds to) the second protein.
  • the signal value for the unit amount of the first protein refers to the unit weight or concentration (for example, lug / ml) of the first protein or the signal value measured in the step (3) Value)
  • step (3) (a) dividing the signal value measured in step (3) by the weight, concentration, or fluorescence signal value of the first protein in the test sample,
  • the increment of the signal value measured in step (3) as the first protein weight, concentration, or fluorescence signal value in the test sample increases i.e., the first protein weight, concentration, or fluorescence signal value in the test sample is x Axis, and the slope of the graph obtained by taking the signal value measured in step (3) as the y value).
  • the present specification suggests that the ratio of the activated first protein to the amount of the first protein in the sample shows a more significant correlation with the drug reactivity targeting the first protein in the sample from which the sample is derived. That is, when the level of the first protein in the sample is low (that is, the amount is at least) and the ratio of the activated U protein is high (activation level: act iatv ion score), the amount of the first protein The drug reactivity is remarkably superior as compared with the case where the ratio is low (see FIGS. 19 and 20). Therefore, the present invention is characterized in that the protein-protein interaction is measured and calculated as a value for the first protein unit amount (divided by the amount of the first protein), thereby providing more accurate information on drug reactivity determination.
  • step of measuring the activation level of the first protein of step (4) (i) directly measuring the signal value measured in step (3) by dividing the signal value by the weight , concentration , or fluorescence signal value of the first protein in the test sample added in step (1)
  • step (1) obtaining a signal value for the unit amount of the test sample by dividing the signal value measured in step (3) by the weight, concentration, or fluorescence signal value of the test sample added in step (1)
  • the step of measuring the protein-protein interaction level in the step (4-1) is a step of obtaining a signal value of the unit amount of the test sample added in the step (1) using the signal measured in the step (3).
  • the protein-protein interaction level is determined by the protein-protein interaction intensity
  • PPI strength PPI strength
  • the signal value of the unit amount of the test sample added in the step (1) by using the signal measured in the step (3) is determined by the test sample condition such as the amount of the test sample Error can be reduced.
  • the step of obtaining the signal value for the unit amount of the test sample added in the step (1) using the signal measured in the step (3) is the step of measuring the signal measured in the step (3) by the amount , Or by calculating the slope of the graph obtained by taking the signal measured in step (3) as the y-axis and the amount (concentration or weight) of the test sample added in step (1) as the x-axis.
  • the step of measuring the protein-protein interaction level may be performed for each protein combination.
  • the sum of the PPI strengths obtained for each second protein can be determined as the protein-protein interaction level (PPI score), as expressed in the following equation :
  • PPI score protein-protein interaction level
  • the obtained protein-protein interaction level (PPI strength or PPI score) is normal izat ion such that the protein-protein interaction level of the reference sample described below is 1, and the PPI strength of the test sample is set as a reference sample Of the PPI strength.
  • the activation level measurement step (step 4 or 4-2)
  • step (4-2) (the protein-protein interaction level of step (4-1) described above) is directly measured without the measurement step) or step (4-2) Value or the signal value of the unit amount of the test sample obtained in the step (4-1) to obtain the value for the unit amount of the first protein contained in the test sample.
  • the result obtained in the step (4) or the step (4-2) is referred to as an activation level (or an act ivat ion score).
  • the activation level is determined by dividing the signal value obtained in step (3) or the protein-protein interaction level obtained in step (4-1) by the amount of the first protein contained in the test sample, It is possible to more accurately measure the degree of activation of the first protein by reducing the error due to the amount and / or distribution.
  • the method provided herein may further comprise the step of measuring the amount of the first protein in the test sample.
  • the step of measuring the amount of the first protein in the test sample may be carried out before or simultaneously with the step (4) or the step (4-2).
  • the amount of the first protein can be measured by any conventional method.
  • the amount of the first protein can be measured by a conventional immunoblotting method (for example, quantitative assay, western blotting), ELISA (enzyme-linked immunosorbent assay; sandwich assay, etc.), but the present invention is not limited thereto.
  • the first protein may be quantified by adding a detection antibody to the substrate to which the first protein is immobilized, and measuring a signal generated from the label, but the present invention is not limited thereto.
  • the step of measuring the activation level may be performed for each combination of the first protein task 2 protein Respectively.
  • the protein-protein interaction level score obtained from the sum of the PPI strengths obtained for each second protein or the sum of the PPI strengths as described above) (act ivat ion score), or the sum of the act iat ion scores obtained for each second protein, and determine the act iat ion score for the two or more second proteins.
  • the protein level (PPI score) obtained from the sum of the PPI strengths obtained for each first protein or from the sum of the PPI strengths is used to determine the activation level act ivat ion score) or the sum of the act iat ion scores obtained for each first protein, and determine the act iat ion score for the two or more first proteins.
  • the activity level obtained can be normalized to an activation level of the reference sample described below, such that the activation level of the test sample is relative to the activation level of the reference sample.
  • step (4) may comprise comparing the results (protein-protein interaction level or degree of tyrosine kinase activation) obtained in step (3) To the result obtained in the sample (protein-protein interaction level or degree of tyrosine kinase activation).
  • the reference sample can be appropriately selected according to the purpose of the invention.
  • the reference sample may be (1) And / or (2) cells whose known degree of activation of the signal transduction pathway involved in the first protein (HER2, HER3, or both (e.g., HER2-HER3 heterodimer forms) Cancer cells), and / or (3) the degree of activation of the signal transduction pathway involving the first protein (HER2, HER3, or both (e.g., HER2-HER3 heterodimer forms) Cells (e. G., Normal cells or cancer cells).
  • the test sample is from a subject In the case of including isolated cancer cells, it may include normal cells of the same tissue or the same organ as the cancer cells isolated from the individual.
  • the methods further comprise, before step (4), measuring the degree of protein-protein interaction or tyrosine kinase activation between the first protein and the second protein with respect to the reference sample
  • steps can be performed with reference to the steps (1), (2), and (3) described above, or (1-1), (2-1), and (3-1).
  • a step of confirming (determining) a desired item from the comparison result obtained in step (4) may be further included. More specifically, it is as follows:
  • the step of determining may comprise the following steps:
  • the signal that the first protein participates in the test sample or the individual from which the test sample originates Confirming (determining) the degree of activation of the delivery path is higher than the degree of activation of normal cells or the level of activation (known) recognized in the reference sample;
  • the signal that the first protein participates in the test sample or the individual from which the test sample originates Confirming (determining) the degree of activation of the delivery path is equivalent to the degree of activation of normal cells or the degree of activation (known) recognized in the reference sample;
  • the protein-protein interaction level of the test sample measured in step (3) is lower than the protein-protein interaction level measured in the reference sample, the test sample or the first protein from the test sample- (Determination) that the degree of activation of the involved signaling pathway is lower than the degree of activation of normal cells or the level of activation (known) known in the reference sample.
  • Step (5) Step of comparing with reference sample
  • Step 5 is a step of comparing the results obtained in step (4) or step (4-1) or step (4-2) (protein-protein interaction level (PPI score or Activat ion score) (Protein-protein interaction level (PPI score) or activation level (Act iat ion score)) obtained from the sample 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the reference sample can be appropriately selected according to the purpose of the invention.
  • a method for measuring the activation (or confirmation or determination or analysis) of a signal transduction pathway in a cell or tissue and / or between a cell or tissue, or a method for providing information on a 5 activation measurement And / or (2) a cell in which the degree of activation of the signal transduction pathway involved in the first protein is known (confirmed) (e.g., normal or cancer cells), and / or (3) the degree of activation can 10 may comprise a known (identified), the cells (e.g., normal cells or cancer cells) isolated from the object.
  • the test sample comprises cancer cells isolated from an individual, it may include normal cells of the same tissue or the same organ as cancer cells isolated from the test specimen.
  • normal cell may refer to any cell in a non-pathological state, wherein the "non-pathological condition” refers to a condition in which the disease state is not 15 , Functional, and / or morphological abnormality, or the like.
  • a normal cell may be a disease associated with the first protein or a cell that does not have a disease to be treated of the drug to be tested, and may be derived from the same individual or tissue as the individual or tissue from which the test sample is derived.
  • the reference sample may be a normal cell or a cell having known (confirmed) , Cancer cells), or when the test sample contains cancer cells isolated from an individual, 25 normal cells of the same tissue or the same organ as the cancer cells may be included.
  • the reference sample may be a normal cell or a cell which has known the effect of treatment targeting the first protein (Eg, cancer cells), or when the test sample contains 30 cancer cells isolated from an individual, normal cells of the same tissue or the same organ as the cancer cells may be included.
  • the methods comprise the following steps (1 '), (2), (3) and (4) ), 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • step (5) determine (determine) the locations of interest from the comparison results obtained in a 20 added.
  • the test sample or the test sample (Determining) the degree of activation of the signal transduction pathway involved in the first protein in the individual from which the first protein is derived is equal to the degree of activation of the normal cell or the degree of activation (known) known in the reference sample; And / or
  • test sample or the test sample (Determining) the degree of activation of the signal transduction pathway involved in the first protein in the individual from which the first protein is derived is lower than the degree of activation of the normal cell or the known (confirmed) activation level in the reference sample.
  • Step (6) may include the following steps:
  • test sample or the test sample (Determining) the reactivity of the first protein of the individual derived from the first sample to the drug that targets the first protein is higher than the drug reactivity of the reference sample;
  • test sample or the test sample (Determining) the reactivity of the first protein of the individual derived from the target sample to the drug that targets the drug is equivalent to the drug response of the reference sample; And / or
  • test sample or the test sample (Determination) that the reactivity of the first protein of the individual derived from the target is lower than the drug reactivity of the reference sample.
  • the reference sample is selected so as to include cells having reactivity to the drug targeting the first protein to a degree required for the subject to be tested, 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • step (6) is performed in step (4) or
  • the test sample or the sample Determining whether the drug is more reactive to the drug targeting the first protein and / or that the drug is effective on the test sample or on the individual from which the test sample is derived.
  • a method for predicting reactivity to a drug or a method for predicting reactivity to a drug that targets the first protein comprising the steps of: (a) providing a test sample or a first protein of the individual from which the test sample is derived Administering a drug targeting the first protein to the subject, if it is determined that the drug has an excellent response to the drug, and / or that the drug has an effect on the test sample or an individual from which the test sample is derived You can include it.
  • individual customized treatment means suitable for each individual are provided based on the above decision content.
  • the drug is excellent in reactivity to a drug targeting the first protein in step (6), which comprises a drug targeting the first protein as an active ingredient, and / or the drug has an effect
  • a pharmaceutical composition for the treatment of a disease associated with a first protein in a determined individual is also provided.
  • Another example is the use of a drug targeting the first protein, an agent having an excellent reactivity to the drug targeting the first protein in step (6), and / or an agent in the subject determined to be effective 1 < / RTI > protein-related diseases .
  • Another example is the step of administering a drug targeting the first protein to an individual having an excellent response to the drug targeting the first protein in step (6) and / or determining that the drug is effective and it provides a method of treating diseases which are associated with the low-i 1 protein in the object containing the. 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the reference sample includes normal cells or cells (for example, cancer cells) whose effects of treatment targeting the first protein are known (confirmed), or when the fifth test sample contains cancer cells isolated from the subject, And may include normal cells of the same tissue or the same organ as the cancer cells.
  • Step (6) is a step (6) wherein the protein-protein interaction level or activation level of the test sample measured in step (4) or (4-2) is compared with the protein- For example, 0, the test sample or the individual from which the test sample is derived may be identified (determined) as a patient suitable for treatment targeting the first protein.
  • Treatment targeting the first protein may mean prescribing and / or administering a drug targeting the first protein.
  • the reference sample may include a cell having a therapeutic effect that targets the first protein.
  • test sample or the individual from which the test sample is derived is selected from the group consisting of 0 < / RTI > identified as a patient suitable for treatment, the subject is treated with a first protein
  • Perform the treatment of the target can contain the further step of (e. G., A drug for the first protein with the target prescription and / or dosage).
  • individual personalized treatment means suitable for each individual are provided based on the above-mentioned confirmation (decision) contents.
  • One example is a method for treating a disease associated with a first protein in an individual identified as being suitable for treatment targeting the first protein in step (6), comprising the step of administering a drug that targets the first protein as an active ingredient To provide a pharmacological ancestor for.
  • Another example provides the use of a drug targeting said first protein for the treatment of a disease associated with a first protein in said individual identified as being suitable for treatment targeting said s1 protein in said step (6) do.
  • Another example is to perform treatment targeting the first protein to a subject identified as suitable for treatment targeting the first protein in step (6) (e.g., administering a drug targeting the first protein and / Or administration) of the first protein in the subject, 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the reference sample includes normal cells or cells (for example, cancer cells) for which the effect of the treatment targeting the first protein is known (confirmed), or when the test sample contains cancer cells isolated from an individual, And normal cells of the same organ or the same organ.
  • Step (6) is a step (6) wherein the protein-protein interaction level or activation level of the test sample measured in step (4) or (4-2) is compared with the protein- , For example, when the test sample or an object to which the test sample is derived is subjected to treatment with a low protein targeting effect (for example, after administration of a drug targeting the first protein, The response to the drug is maintained or the drug does not develop resistance to the drug).
  • Treatment targeting the first protein may mean prescribing and / or administering a drug targeting the first protein.
  • the reference sample may include a cell having a therapeutic effect that targets the first protein.
  • a method for monitoring the effect of treatment targeting the first protein or a method for providing information to monitoring comprising the steps of: (a) administering a test sample or a first protein to a subject in a step (6) (For example, prescribing and / or administering a drug targeting a low protein) to the subject when it is confirmed that the effect of the low protein is exerted Or in the step (6), the test sample or an object to which the test sample is derived does not exhibit the effect of treatment targeting the first protein (for example, reduction in therapeutic effect (drug reactivity) or acquisition of resistance, etc.)
  • the method comprising: stopping treatment to target the first protein to the subject, and / or stopping the first protein Administering another drug to be targeted, or performing a treatment targeting a different first protein.
  • Another example is protein-protein interaction between a first protein and a second protein 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the first protein is a protein involved in a signal transduction pathway in a cell or tissue and / or between a cell or tissue
  • the second protein is a protein in the cell or tissue and /
  • the step of measuring the protein-protein interaction is performed on two or more first proteins, wherein the cells or tissues or the individual from which the cells or tissues are derived
  • the method comprises:
  • step (4) or step (4- 2) comparing the results obtained in step (4) or step (4- 2) with respect to two or more first proteins
  • the first protein two or more proteins selected from proteins involved in signal transduction pathways of cells or tissues are used.
  • the comparing step of step (5) is performed on two or more species of the first protein 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the results can be compared to each other.
  • a first protein having a high protein-protein interaction level or activation level is selected as a therapeutic target of the test sample or a test sample derived from the test sample, (Step (6)) of selecting the drug as a candidate drug for the treatment of a test sample or an individual from which the test sample is derived.
  • Another example includes measuring a protein-protein interaction between a first protein and a second protein, wherein the first protein is a protein involved in a signal transduction pathway in a cell or tissue and / or between cells or tissue, 2 protein is a downstream protein of the first protein in the cell or tissue and / or in a signal transduction pathway between cells or tissues, and the step of measuring the protein-protein interaction is performed before and after the candidate substance treatment.
  • the method comprises:
  • step (3) measuring a signal from the reactant obtained in step (2) (protein-protein interaction measurement).
  • step (4-1) Using the signal values for the unit amount of the test sample obtained in step (4-1) and the step of obtaining the signal value for the unit amount of the test sample (measurement of protein-protein interaction level) and (4-2) Obtaining a value for a unit amount of the first protein contained in the sample (measurement of the activation level); And
  • step (3) The respective results obtained in step (3), step (4), step (4-1), or step (4- 2) for the test sample not treated with the candidate compound and the treated test sample Comparing them to each other.
  • test sample not treated with the candidate compound and the treated test sample means the test sample before treatment and the test sample after treatment of the candidate compound, respectively, or a part of the test sample treated with the candidate compound and the candidate May refer to some other test sample that has not been treated with the compound.
  • the candidate compound may be selected as a candidate candidate drug, or the candidate compound may be effective as a drug targeting the first protein.
  • the method for selecting a candidate drug targeting the first protein or the method for confirming the efficacy of a candidate drug targeting the first protein can be performed after the step (5), as a result of the comparison in the step (5)
  • the candidate compound is selected as a candidate drug targeting the first protein, (Step (6)) of confirming that the compound is effective as a drug targeting the first protein.
  • One example includes measuring protein-protein interactions between a first protein and a second protein, wherein the first protein comprises And? 0-1, the second protein is another one, and the step of measuring the protein-protein interaction is carried out before and after the candidate substance treatment, wherein the first protein and / or the second protein It provides the first protein and / or the second method confirmed the efficacy of candidate drugs which target proteins: the candidate drug to target screening methods, or.
  • Another example comprises measuring a protein-protein interaction between a first protein and a second protein, 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the second protein is another one, and the step of measuring the protein-protein interaction is performed before and after the candidate substance treatment, wherein the first protein and / or the second protein
  • the present invention provides a method for screening candidate drugs for the prevention or treatment of diseases (e.g., diseases associated with the expression and / or activation of the first protein and / or the second protein; cancer, immune diseases, etc.).
  • diseases e.g., diseases associated with the expression and / or activation of the first protein and / or the second protein; cancer, immune diseases, etc.
  • the method comprises:
  • step (3) measuring a signal from the reactant obtained in step (2) (protein-protein interaction measurement).
  • step (1) obtaining a value for the unit amount of the first protein included in the test sample added in step (1) (measuring the activation level) using the signal measured in step (3); or
  • step (3) The respective results obtained in step (3), step (4), step (4-1), or step (4- 2) for the test sample not treated with the candidate compound and the treated test sample Comparing them to each other.
  • the candidate compound may be selected as a candidate drug targeting the first protein or the candidate compound may be found to be effective as a drug targeting the first protein.
  • the method for screening a candidate drug targeting the first protein or the method for confirming the efficacy of a candidate drug targeting the first protein is characterized in that after the step (5), the result of the comparison in the step (5) When the protein-protein interaction level or the activation level of the treated test sample is lower than that of the test sample in which the candidate compound is not treated, the candidate compound is selected as a candidate drug targeting the first protein or the second protein, (Step (6)) of confirming that the candidate compound is effective as a drug targeting the first protein or the second protein.
  • the candidate compound may be selected from among all biocompatible materials that can be used as a target drug of the first protein, for example, small molecule chemicals, proteins (e.g., antibodies, antibody fragments, ), Peptides, nucleic acid molecules (such as DNA, RNA (e.g., siRNA, miroRNA, shRNA, etc.), peptid nucleic acid (PNA), aptamer, And the like.
  • small molecule chemicals proteins (e.g., antibodies, antibody fragments, ), Peptides, nucleic acid molecules (such as DNA, RNA (e.g., siRNA, miroRNA, shRNA, etc.), peptid nucleic acid (PNA), aptamer, And the like.
  • proteins e.g., antibodies, antibody fragments, )
  • Peptides e.g., amino acid molecules
  • nucleic acid molecules such as DNA, RNA (e.g., siRNA, miroRNA, shRNA, etc.), peptid nucleic acid (PNA), apta
  • the test sample can be used for a disease associated with a first protein, or a disease associated with a first protein to be screened or a first protein to be screened, such as a cell (for example, a cell lysate or the like) (E.g., cell lysate, etc.) or tissue (e. G., Tissue lysate, etc.) associated with the application (treatment) target disease of the protein-targeting drug, and in one example, (E.g., cancer).
  • the test sample may be a cell or tissue isolated from an established cancer cell line or cancer patient (the cancer may be associated with overexpression and / or activation of the first protein).
  • the above steps (1) to (6) may be performed in in / iro.
  • the description of the above steps (1) to (6) and the first protein, the second protein and the like are as described above.
  • the method for screening a candidate drug targeting the above-mentioned first protein can be usefully applied to the efficacy (or confirmation or testing) of a drug developed as a candidate drug in the development of a new drug targeted by the first protein.
  • the candidate compound may be selected from among all biocompatible materials that can be used as a target drug of the first protein, for example, small molecule chemicals, proteins (e.g., antibodies, antibody fragments, ), Peptides, nucleic acid molecules (e.g., DNA, RNA (e.g., siRNA, microRNA, shRNA), peptid nucleic acid (PNA), aptamer, etc.), plant extracts, animal extracts, But it is not limited thereto.
  • small molecule chemicals proteins (e.g., antibodies, antibody fragments, ), Peptides, nucleic acid molecules (e.g., DNA, RNA (e.g., siRNA, microRNA, shRNA), peptid nucleic acid (PNA), aptamer, etc.)
  • PNA peptid nucleic acid
  • the test sample can be used for a disease associated with a first protein, or a disease associated with a first protein to be screened or a first protein to be screened, such as a cell (for example, a cell lysate or the like) (E.g., cell lysate, etc.) or tissue (e. G., Tissue lysate, etc.) associated with the application (treatment) target disease of the protein-targeting drug, and in one example, (E.g., cancer).
  • the test sample may be a cell or tissue isolated from an established cancer cell line or a cancer patient . Or may be related to overexpression and / or activation of the protein).
  • the screening method of the candidate drug targeting the first protein can be usefully applied to drugs and efficacy testing (or confirmation or testing) developed as a candidate drug in the development of a new drug targeted by the first protein.
  • Another example includes measuring a protein-protein interaction between a first protein and a second protein, wherein the first protein is a protein involved in a signal transduction pathway in a cell or tissue and / or between cells or tissue, 2 protein is at least one selected from among proteins downstream of the first protein in the cell or tissue and / or signal transduction pathway between cells or tissues, a method for selecting a concurrent therapeutic target for concurrent with the target treatment of the first protein, or A method of providing information to the selection, or a method of selecting a concomitant drug to be co-administered with a target drug of the first protein, or a method of providing information to the selection.
  • the method comprises:
  • step (3) determining the signal from the reactant obtained in step (2) (protein-protein interaction measurement).
  • step (1) obtaining a value for the unit amount of the first protein included in the test sample added in step (1) (measuring the activation level) using the signal measured in step (3); or
  • step (3) comparing the results obtained in step (3), step (4), step (4-1), or step (4-2)
  • step (4) or step (4-2) obtained for two or more second proteins with each other.
  • step (4) or step (4-2) obtained for the test sample is higher than the reference sample
  • the second protein is combined with the target treatment of the first protein
  • a drug targeting the second protein is selected as a drug for use in combination with a target drug of the first protein
  • the second protein having the highest protein-protein interaction level or activation level obtained in the second or more second protein heavy chain stage (4) or step (4-2) is used in combination with the target treatment of the first protein, Or a drug targeting the second protein may be selected as a drug for administration in combination with a target drug of the first protein.
  • a method for selecting a concurrent treatment target (or a method for providing information on the selection) for concurrently with the target treatment of the first protein, or a method for selecting a concomitant drug for co-administration with the target protein of the first protein (Or a method for providing information to the selection), after the step (5)
  • step (6) The second protein having the highest protein-protein interaction level or activation level obtained in step (4) or step (4-2) among the second protein on the two or more species is administered in parallel with the target treatment of the first protein Or a drug for targeting the second protein to a drug for use in combination with a target drug of the first protein,
  • a combination therapy means based on the above decision content / RTI >
  • One example includes a drug targeting the first protein and a drug targeting (e.g., inhibiting) a second protein selected as a concurrent therapeutic target in step (6-1) or (6-2) , ≪ / RTI > a pharmaceutical composition for coadministration for the treatment of diseases associated with the first protein.
  • the pharmaceutical composition may be used for the combined administration for the treatment of diseases related to the first protein in the test sample or the individual from which the test sample is derived.
  • Another example is a pharmaceutical composition
  • a drug targeting the first protein and a drug targeting (e.g., inhibiting) a second protein selected as a concurrent therapeutic target in step (6-1) or (6-2) 1 < / RTI > protein.
  • Another example is a method for treating a disease associated with a first protein, comprising administering to a patient in need of such treatment a therapeutically effective amount of a first protein (e. G., Administering a drug targeting the first protein) (E.g., administration of a drug that targets (e. G., Inhibits) the second protein) targeted to a second protein selected as a concurrent therapeutic target in step
  • the patient may be the test sample or an individual from which the test sample is derived.
  • a method of predicting or predicting responsiveness to a drug that targets the provided first protein a method of screening an entity suitable for treatment targeting the first protein, , And methods of screening or screening for a first protein or a target thereof as a therapeutic target suitable for application to a cell or tissue or an individual (individual patient) from which the cell or tissue is derived (Step 7) of confirming whether the first protein is post-translationally modified (PTM) in addition to the protein-protein interaction measurement and activation level measurement step described above, ).
  • PTM is a common marker showing the activation status of the first protein. It is therefore used as a control against existing methods.
  • the post-translational modification of the first protein refers to a modification (a post-translational modification related to the first protein of the first protein) that occurs more frequently in a cancer or tumor patient than a normal individual (a cancer or a tumor-free individual)
  • a gene encoding a first protein or a first protein may comprise at least one nucleotide or at least one amino acid mutation (or at least one amino acid mutation A nucleotide variation), phosphorylation (e.g., phosphorylation of a Tyr residue), and the like.
  • the mutation may be intensively present in exon 19 and / or exon 21, and in one embodiment, the mutation is a deletion of one or more genes in exon 19 and exon 21 of EGFR , Deletion of exon 19 and / or exon 21), gene mutation causing L858R substitution, phosphorylation at Tyr 1068 and / or Tyr 1086, but not limited to, normal individuals (Not present) and can be selected from all known EGFR (gene / protein) variants with high incidence in cancer or tumor patients.
  • the mutation may be phosphorylation at Tyr 1221, but is not limited thereto, and may occur in a cancerous or tumor patient with a high incidence in comparison with a normal individual (a cancer or a tumor-free individual) It can be selected from all known HER2 (gene / protein) variants.
  • the mutation may be phosphorylation at Tyr 1289, but is not limited thereto, and is more frequent in cancer or tumor patients than in normal individuals (individuals without cancer or tumor) It can be selected from all known HER3 (gene / protein) variants.
  • the mutation may be phosphorylation at Tyr 1349, but is not limited thereto, and is more frequent in cancer or tumor patients than in a normal individual (a cancer or a tumor-free individual) It can be selected from all known MET (gene / protein) variants.
  • the term "targeting a first protein " may mean promoting or inhibiting the activity of a first protein, e. G., Inhibiting the activity of a first protein
  • drug refers to any substance that exhibits a pharmacological effect, such as a small molecule compound, a protein (such as an antibody, an antibody fragment, or an analogue thereof), a peptide, a nucleic acid molecule And at least one compound selected from the group consisting of RNA (for example, siRNA, miroRNA, shRNA, etc.), peptidase nucleic acid (PNA), aptamer, etc., plant extracts, animal extracts, It can be selected.
  • RNA for example, siRNA, miroRNA, shRNA, etc.
  • PNA peptidase nucleic acid
  • the first protein is a receptor tyrosine kinase (RTK), an RTK profiling kit, or a kit for predicting the efficacy of a drug targeting the first protein.
  • the drug may be, but is not limited to, a therapeutic agent for lung cancer (such as Afatinib).
  • drug targeting a first protein refers to any substance that inhibits the activity of a first protein, such as a small molecule, a protein, (E.g., small interfering RNA (siRNA), small hairpin RNA, miRNA (microRNA), etc.), PNAs (such as antibodies, antibody fragments or analogs thereof), peptides, nucleic acid molecules a peptide nucleic acid, an aptamer, etc.), a plant stem, an animal stem, a cell stem, and the like.
  • siRNA small interfering RNA
  • miRNA miRNA
  • PNAs such as antibodies, antibody fragments or analogs thereof
  • peptides such as antibodies, antibody fragments or analogs thereof
  • nucleic acid molecules such as a peptide nucleic acid molecules a peptide nucleic acid, an aptamer, etc.
  • plant stem such as a plant stem, an animal stem, a cell stem, and the like.
  • a drug targeting a first protein &quot is a substance that binds to a first protein and / or degrades and / or structurally modifies a first protein to produce a unique function,
  • E. G. Antibodies, antibody fragments, or analogs thereof
  • peptides e. G., DNA < / RTI > , RNA (such as siRNA, microRNA, shRNA), PNA (peptide nucleic acid), aptamer, etc.), plant extracts, animal extracts, cell extracts and the like.
  • the "drug targeting a first protein " may refer to a therapeutic agent targeting a first protein, such as a target inhibitor of a first protein.
  • Targeted drugs include antiretroviral drugs such as cetuximab, gefitinib, erlotinib, afatinib, osimertinib (AZD9291), various anti-EGFR antibodies, MET target therapeutics (various anti-MET antibodies, Crizotinib, Cabozant inib etc.) Therapeutic agents (trastuzumab, Trastuzumab, Pertuzumab, Lapatinib, etc.), HER3 target therapy (Eg, various anti-HER3 antibodies), FGFR1, 2) Target therapeutic agents (Lenvatinib, Nintedanib, Regorafenib etc.), VEGFR (1,2,3) Target therapeutic agents (Bevacizumab, Axitinib, Lenvat inib etc.), PDG
  • PD-1 target therapeutic agent (Nivolumab etc.), and the like, which are selected from the group consisting of anti-inflammatory drugs (Bosutinib, Dasatinib, Imatinib, Ni lotinib etc.), AR target therapeutic agents (Abiraterone, Enzalut amide etc.) Or more, but is not limited thereto.
  • the "drug targeting the first protein” competes with the second protein in binding to the first protein such that the first protein task 2 protein (E.g., a small molecule, a protein (e.g., an antibody, an antibody fragment, or an analogue thereof), a peptide, a nucleic acid molecule ), shRNA (small hairpin RNA), miRNA (microRNA), etc.), PNA (peptide nucleic acid), aptamer, etc.), plant trunk, animal trunk, Or more).
  • the first protein task 2 protein E.g., a small molecule, a protein (e.g., an antibody, an antibody fragment, or an analogue thereof), a peptide, a nucleic acid molecule ), shRNA (small hairpin RNA), miRNA (microRNA), etc.), PNA (peptide nucleic acid), aptamer, etc.), plant trunk, animal trunk, Or more).
  • treatment targeting a first protein may refer to any medical and / or pharmaceutical activity that inhibits the activity of a first protein, for example, Quot; first protein-targeting treatment " may include administering a first protein and / or a second protein to a subject in need of inhibiting the activity of the first protein, To a subject in need thereof a drug that reduces or eliminates the inherent functions of, for example, inherent bio-signal transduction functions in cells and / or tissues, by binding and / or degrading and / or structurally modifying the first protein Prescription and / or administration.
  • Another example is protein-protein interaction between a first protein and a second protein 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the first protein is a first class 2 and the third class 3
  • the first class 2 and the third class 3 form a heterodimer
  • the second protein is a cell or a tissue and / or a cell or a cell
  • the protein-protein interaction is a sub-protein of the other of the inter-tissue signal transduction pathways
  • the step of measuring the protein-protein interaction is performed in the sample treated with the candidate substance and the untreated sample, A method for confirming the efficacy of a candidate drug targeting a protein or a method for confirming the efficacy of a candidate drug for the prevention or treatment of the disease.
  • Another example involves measuring the activation level of the inhibitor 2 and / or 1-3 of the quadrivalent 2-thymine 3 heterodimer (e.g., phosphorylation of the tyrosine residue), wherein the step of measuring activation is referred to as the candidate substance
  • the step of measuring activation is referred to as the candidate substance
  • a sample containing a heterodimer or a heterodimer of the 2-tetradecane heterodimer and an untreated heterodimer or an untreated heterodimer of 2- A method for confirming the efficacy of a candidate drug targeting the first protein or a method for confirming the efficacy of a candidate drug for prevention or treatment of the disease.
  • the method comprises:
  • the phosphorylated tyrosine specific binding substance may be an antibody specifically binding to the phosphorylated tyrosine of the first class II and / or the third class. 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the candidate material may be treated in the step (e. G., Before, after, or simultaneously with the treatment of the phosphorylated or phosphorylated tyrosine specific binding material of step (a). Phosphorylation of this step can be carried out by treating the phosphorylating agent.
  • the phosphorus- And / or all substances capable of phosphorylating urine 3, for example, may be selected from the group consisting of: 3, and magnesium (e.g., 1, 2 , 1, 2, etc.).
  • the candidate substance is subjected to the test sample not treated with the candidate substance and the treated test sample, respectively .
  • test sample not treated with the candidate substance and the treated test sample mean a test sample before treatment and a test sample after treatment of the candidate substance, respectively, or a part of the test sample treated with the candidate substance and the candidate May refer to some other test sample that has not been treated with the substance.
  • the method for confirming the efficacy of the candidate drug is a method for determining the efficacy of the candidate drug by comparing the protein-protein interaction level of the test sample treated with the candidate substance or the tyrosine 4 of tetrahedral 2 and / When the degree of kinase activation is lower than that of the test sample in which this candidate substance is not treated (i.e., the candidate substance inhibits the interaction between the first protein and the second protein or the activation of tyrosine kinase of quadruplex 2 and / or 1-3) ),
  • the candidate material is targeted to four quadrants 2, 4 quadrature 3, or both (e.
  • step (i) In the form of a 4 1 2-quadrature 3 heterodimer) Or inhibiting tyrosine kinase activation of 1? 2 and / or 1? 3) (step (i) .
  • the level of protein-protein interaction level or tyrosine kinase activation level of HER2 and / or HER3 of the test sample treated with the candidate drug is lower than that of the test sample not treated with the candidate substance.
  • the protein- Mutual levels or degree of tyrosine kinase activation of HER2 and / or HER3 is less than or equal to 75% (i.e., greater than or equal to 25% inhibition) of the protein-protein interfering level of the test sample to which the candidate agent has not been treated or the degree of tyrosine kinase activation of HER2 and / ), 50% or less (i.e., 50% or more inhibition), 45% or less, 40% or less, 35% or less, 30% or less, 25% or less Inhibition), 15% or less, or 10% or less (i.e., 90% or more inhibition).
  • the candidate agent may be selected from any biocompatible material available as a target drug for the first protein (HER2, HER3, or both (e.g., HER2-HER3 heterodimer form)), such as small molecular (eg, DNA, RNA (eg, siRNA, microRNA, shRNA, etc.), PNA (peptide nucleic acid), aptamer, etc.), proteins (eg, antibodies, antibody fragments or analogs thereof) ), Plant extracts, animal extracts, cell extracts, and the like, but the present invention is not limited thereto.
  • HER2 HER3 heterodimer form such as small molecular (eg, DNA, RNA (eg, siRNA, microRNA, shRNA, etc.), PNA (peptide nucleic acid), aptamer, etc.), proteins (eg, antibodies, antibody fragments or analogs thereof) ), Plant extracts, animal extracts, cell extracts, and the like, but the present invention is not limited thereto.
  • small molecular eg, DNA,
  • the test sample may be a cell (e.g., a cell lysate, etc.) or a tissue (e.g., a cell lysate, etc.) overexpressing and / or activating a first protein (HER2, HER3, or both (in particular, a HER2-HER3 heterodimer form) (E.g., cell lysate) or tissue (e.g., cell lysate) associated with the disease (s) involved in the application (treatment) of a drug targeting the first protein for which the disease or efficacy associated with the first protein is to be identified Tissue lysate, etc.), and in one example, may be a cell or tissue isolated from an established cell line or a patient suffering from the disease (e.g., cancer).
  • the test sample may be a cell or tissue isolated from an established cancer cell line or cancer patient (the cancer may be associated with overexpression and / or activation of the first protein).
  • the steps (a) to () may be performed in F / ZTO.
  • the terminology of each step is as described above.
  • targeting a first protein refers to promoting or inhibiting the activity of a first protein (HER2, HER3, or both (eg, a HER2-HER3 heterodimer form) You can, 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the first protein may be inhibiting the activity (tyrosine kinase activity) of the first protein, e.g., the first protein, the second protein, the second protein, the third protein, or both (e.g., other 1? .
  • Inhibition of the activity of the first protein may be associated with the first protein and / or by degradation and / or structural modification of the first protein to provide a unique function, e. G., A unique bio signaling function in 5 cells and / It can be reduced or eliminated.
  • the administration can be by oral or parenteral routes.
  • parenteral administration it can be carried out by 10 routes such as intravenous infusion, subcutaneous infusion, muscle infusion, intraperitoneal injection, endothelial administration, local administration of lesion site, intranasal administration, intrapulmonary administration or rectal administration.
  • the subject, patient or subject may be any mammal such as a primate such as a human, a monkey, a rodent such as a mouse, a rat, or the like, and may be, for example, a patient having a disease associated with the first protein .
  • the disease associated with the said protein may be a disease associated with overexpression of the first protein or activation of a signal transduction pathway in a cell or tissue 15 and / or between cells or tissues in which the first protein participates, such as cancer, inflammation, Immune diseases, and the like.
  • the subject, patient or subject may be a cancer patient.
  • the cancer may be selected from all solid tumors and blood cancers and may be, for example, a cancer associated with overexpression of a first protein or activation of a signal transduction pathway between a cell or tissue 20 and / or a cell or tissue involved in the first protein.
  • the cancer is selected from the group consisting of squamous cell carcinoma, small cell lung cancer, non-small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, peritoneal cancer, skin cancer, skin or intraocular melanoma, rectal cancer, adenocarcinoma, part thyroid cancer, adrenal cancer, soft tissue sarcoma, uterine cancer, chronic or acute leukemia, lymphocytic lymphomas, gastric cancer, 25 pancreatic cancer, schools subspecies, cervical cancer, ovarian cancer, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial or uterine cancer, salivary gland Cancer, kidney cancer, prostate cancer, mucin cancer, thyroid cancer, head and neck cancer, brain cancer, and the like, but the present invention is not limited thereto.
  • the cancer may include metastatic cancer as well as primary cancer.
  • the cancer may be resistant to conventional chemotherapy or obtained 30 times .
  • the term "responsiveness to a drug " may refer to the degree to which the drug exerts an effect on an individual receiving the drug.
  • the term & Medication or treatment cured Means a medical and / or pharmaceutical effect intended to be achieved by the subject, which may mean prevention and / or treatment of the disease to be treated, and / or alleviation and / or amelioration of symptoms.
  • the effect when the drug is an anticancer agent and the treatment is chemotherapy and the subject is a cancer patient, the effect may be an anticancer effect (prevention and / or therapeutic effect of cancer), and the anticancer effect may inhibit the growth of cancer cells As well as the effects of inhibiting migration, invasion, and / or metastasis, and / or inhibiting the deterioration of cancer and / or reducing or eliminating resistance, and the like. have.
  • a method for measuring activation of a signal transduction pathway in a cell or tissue and / or between cells or tissues as described above, a method for predicting responsiveness to a drug targeting the first protein, a method for monitoring reactivity for a drug targeting the first protein There is provided a method for measuring protein-protein interactions for use in a method for screening a first protein, a method for selecting an individual suitable for treatment targeting the first protein, and / or a method for screening a drug.
  • the apparatus for measuring protein-protein interactions may be a device for measuring activation of a signal transduction pathway in a tissue, an apparatus for predicting and / or monitoring reactivity to a drug targeting the first protein, a device suitable for treatment targeting the first protein An apparatus for screening an individual, and / or an apparatus for confirming the efficacy of a drug targeting the first protein.
  • the first protein in the device may be PD-L1 or PD-1.
  • the first protein in the device may be HER2, HER3, or both (e. G., A HER2-HER3 heterodimer form).
  • the device for measuring protein-protein interactions can be used for measuring the activation of a signal transduction pathway in cells or tissues and / or between cells or tissues, a method for predicting the reactivity of a drug targeting a first protein
  • the present invention can be applied to a method for monitoring the reactivity of a drug to a drug, a method for screening an individual suitable for treatment targeting the first protein, and / or a screening method for a drug, so that the interaction between biomolecules in a small amount of sample can be accurately and efficiently Can be observed, analyzed, detected, and / or measured.
  • An apparatus for measuring protein-protein interactions or an analytical method using the same may be usefully and effectively applied to very small amounts of samples such as biopsy (e.g., needle biopsy) samples.
  • the interaction measurement device or analysis method using the same can accurately and efficiently observe, analyze, detect, and / or measure interaction between various biomolecules (e.g., protein, nucleic acid, etc.) using a small amount of sample.
  • biomolecules e.g., protein, nucleic acid, etc.
  • the apparatus for measuring protein-protein interactions includes:
  • the apparatus for measuring protein-protein interaction includes a protein (second protein) that interacts with the first protein (e.g., participates in a lower pathway of a biological signal transduction pathway of a cell or tissue involved in the first protein)
  • the second protein is labeled with a labeling substance that generates a detectable signal
  • the labeling substance may be chemically (e.g., covalent or noncovalent), recombinant, or physical , Or it may be in the form in which 13 sugars to which the labeling substance can be attached are attached.
  • the protein-protein interaction measuring devices for the order to (11 1 1 011 11) level with the normalization of the first protein, the signal measured by the signal detecting means, detecting substances and the for binding to the first protein, A labeling substance that binds to the detection substance may be further included.
  • the detection substance binding to the first protein may be a biomolecule (for example, an antibody) binding to the first protein at a site different from the capturing substance for the first protein contained in the multiwell described above, (The labeling substance is chemically (e . G., Covalently or non-covalently bound)) to generate a detectable signal . (E. G., An antibody) capable of binding to a detection substance that binds to the first protein (see Fig. 6).
  • the apparatus for measuring protein-protein interactions includes a reaction unit comprising a substance capable of capturing a first protein that specifically binds to a first protein And may additionally include signal detection means.
  • the apparatus for measuring protein-protein interactions can be used to measure the presence and / or the degree of interaction (binding) between 1) - [the and 1) -1.
  • the reaction unit may include a substrate on which a substance for capturing a first protein is immobilized, 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the capturing material may be a substance that specifically binds to the first protein described above in the substrate preparation step in which the first protein is immobilized, for example, an antibody or an antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof as described above, binds to a non-binding site (for example, (: -terminal site) other than the binding site between 1) -1 term Antibody or antigen-binding fragment thereof, Linkage 1 antibody or an antigen-binding fragment thereof which binds to a non-binding site (for example, the 0-terminal site) except for the terminal region of the antibody.
  • the reaction unit may be, but is not limited to, a well (e.g., multiwell) type, a slide type, a channel type, an array type, a microfluidic chip, a microtubule (capillary), and the like.
  • the apparatus for measuring protein-protein interaction may further comprise a second protein interacting with the first protein.
  • the second protein is labeled with a labeling substance that generates a detectable signal (the labeling substance is chemically (e. G., Covalent or noncovalent), recombinantly or physically coupled) It may be in the form of a 3- sided attachment to which a labeling substance can be attached.
  • the first protein is? - 11 or suppress-1
  • the first protein is any one selected from the group consisting of suppression-1 and suppression-1
  • the second protein is the other.
  • the first protein is 1) -1
  • the second protein is 1) -1
  • the first protein is suppressed 1
  • the second protein is 1) -1.
  • the analyzing apparatus is capable of capturing a first protein specifically binding to the first protein A substance or a first protein, or may additionally include signal detection means.
  • the first protein comprises a cell that comprises (expresses) a 2-quaternary 3-heterodimer or a 1-12 (3) heterodimer on a cell surface or a lysate of the cell
  • the reaction part can be used in the form of (1) a cell that contains (expresses) a 2-quaternary 3-heterodimer or a 2-quaternary heterodimer on the cell surface or a cell Seaweed, or (2) a substrate on which they are immobilized.
  • Cells expressing (expressing) the quadrature 2-quadrant 3 heterodimer on the cell surface express the quadruplex and inhibitor 3 and are capable of expressing a dimerizing agent (eg, a ligand of other moieties (eg, 31 (NRGbl), etc.), cholesterol-like cleanser (Cholesterol-1 ike detergent;
  • a dimerizing agent eg, a ligand of other moieties (eg, 31 (NRGbl), etc.
  • cholesterol-like cleanser cholesterol-like cleanser
  • DGTN digi tonin; CAS Number: 11024-24-1
  • GDN glyco-diosgenin; CAS Number: 1402423-29-3
  • DGTN digi tonin
  • GDN glyco-diosgenin
  • CAS Number: 1402423-29-3 cholesterol-like cleanser
  • DGTN digi tonin
  • GDN glyco-diosgenin
  • the analyzer is capable of detecting the presence and / or degree of interaction (binding) of HER2 and HER3, particularly HER2-HER3 heterodimers and their downstream proteins in the signal transduction pathway, or the degree of activation of HER2 and / or HER3 ≪ / RTI >
  • the reaction unit may include a substrate on which a substance for capturing a first protein is immobilized on a surface, or a substrate on which the first protein is immobilized (directly) without passing through a capturing substance.
  • the first protein is HER2 and HER3, in particular HER2-HER3 heterodimer
  • the first protein capturing substance is a protein having the first protein (HER2 and / or HER3) described in the substrate preparation step in which the first protein is immobilized Specific binding material, such as an antibody or an antigen-binding fragment thereof.
  • the reaction unit may be, but is not limited to, a well (e.g., multi-well) type, a slide type, a channel type, an array type, a microfluid chip, and a microtubule (capillary).
  • the analyzer may be a device for measuring protein-protein interaction between a first protein and a second protein, and may further comprise a second protein that interacts with the first protein.
  • the analyzer may be a device for measuring the degree of activation of HER2 and / or HER3 (e.g., phosphorylation of a Tyr residue), and for this purpose, a substance (e.g., an antibody) that specifically binds phosphorylated Tyr May be further included.
  • the substance that specifically binds to the second protein or phosphorylated Tyr is labeled with a labeling substance that generates a detectable signal (the labeling substance may be labeled, for example, chemically (e.g., covalently or noncovalently) Or physically), or may be attached to a tag to which a labeling substance can be attached.
  • the labeling substance may be labeled, for example, chemically (e.g., covalently or noncovalently) Or physically), or may be attached to a tag to which a labeling substance can be attached.
  • the multiwells included in the apparatus for measuring protein-protein interactions may include a plurality of tubes whose one surface is open or a plurality of non-through holes (for example, grooves formed in the support plate )of Wherein the one or more non-through holes are defined as wells, and wherein the wells are arranged in a first direction in a second direction in which two or more arrangement tube array structures intersect the first direction, Or a structure having two or more layouts (grid structure) (see FIG. 30).
  • the multi-well may include a sample injecting part located on one opened surface of the tube or the non-through hole, a protein-protein interaction inside the tube or the non-penetrating hole (for example, (For example, a substance that specifically binds to a first protein (for example, a substance that binds to an antibody ) May comprise a capture portion (or substrate) of the immobilized or immobilizable first protein.
  • the multi-well comprises one or more wells comprising a reaction portion (a reaction portion of the first protein and the second protein: the first reaction portion) where interaction between the first protein and the second protein takes place, And a reaction unit (first protein detection unit: second reaction unit) to which a detection substance binding to the first protein binds.
  • the detection substance binding to the first protein is as described above.
  • the first protein detection unit may be used to measure the first protein level in the test sample and to normalize the signal value measured by the signal detection unit included in the device to the level of the first protein.
  • the signal detecting means may be any signal detecting means normally used according to a signal generated from the used labeling material.
  • the signal detecting means may include a signal stimulating portion and a signal detecting portion, and may further include a signal analyzing portion for analyzing (e.g., quantifying or imaging) the measured signal.
  • signal stimulation, signal detection, and signal analysis may be performed at different sites, respectively, or at least two of them may be performed simultaneously or sequentially in one site.
  • the signal detecting means may be selected from among all means capable of generating and detecting a fluorescent signal, and may include, for example, a fluorescent signal stimulating portion (e.g., a light source) , And / or a fluorescence signal analyzer.
  • a fluorescent signal stimulating portion e.g., a light source
  • a fluorescence signal analyzer e.g., a fluorescence signal analyzer
  • the signal detection means comprises a Total Internal Reflection Fluorescence (TIRF) microscope or a confocal microscope (for detecting a light source and a fluorescence signal), or additionally a fluorescence camera, such as an EMCCD (Electron-multiplying charge-coupled device)
  • TIRF Total Internal Reflection Fluorescence
  • EMCCD Electro-multiplying charge-coupled device
  • a camera or a CMOS (complementary metal oxide semi conductor) camera may be further included to perform illumination and / or quantification of the light source and the fluorescent signal.
  • the wavelength, intensity, and fluorescent camera measurement conditions e.g., exposure time per frame, laser power, camera gain value, total shooting frame, etc.) of the light source are as described above.
  • the signal detecting means according to the first protein, the substrate, the substance specifically binding to the first protein, the second protein, the labeling substance, and the labeling substance are as described above.
  • the apparatus for measuring protein-protein interactions is capable of observing, analyzing, detecting , and / or measuring interactions (e.g., protein-protein interactions) between various biomolecules Can be usefully applied.
  • another example is a method for measuring a protein-protein interaction, comprising contacting a sample containing a biomolecule (for example, a first protein) to be analyzed with a device for measuring the protein-protein interaction or a multi- (E. G., Protein-protein interaction).
  • the analysis method may further include a step of measuring a signal generated in the sample after the contacting step.
  • the signal can be appropriately selected from among all signals (for example, fluorescence, luminescence, etc.) conventionally used for biomolecule analysis, and the measurement of the signal is typically used according to the kind of the used signal All of these methods can be selected appropriately.
  • the biomolecule may be at least one selected from the group consisting of proteins, nucleic acids, and cells isolated from a living body, and may be, for example, a protein.
  • a multi-well contained in a device for measuring protein-protein interaction used for protein-protein interaction analysis is a molecule that specifically binds to one of the proteins to be analyzed on the surface (for example, ) May be fixed.
  • kits for measuring protein-protein interactions comprising a multi-well comprising a substance capable of capturing a first protein that specifically binds to a first protein as described above.
  • the kits for measuring protein-protein interactions include kits for measuring the activation of signal transduction pathways in tissues, kits for predicting and / or monitoring the response to a drug targeting the first protein, kits suitable for treatment targeting the first protein For screening individuals Kit, and / or kit for confirming the efficacy of a drug targeting the first protein.
  • the apparatus for measuring protein-protein interactions has the advantage of being able to accurately and efficiently observe, analyze, detect, and / or measure biomolecular interactions in small amounts of sample.
  • the multiwells or analytical methods using them can be usefully and effectively applied to very small samples, such as biopsy (e.g., needle biopsy) samples.
  • biopsy e.g., needle biopsy
  • Another example provides a method for preparing heterodimers of HER2 and HER3. The method may comprise treating the dimerizing agent to cells that simultaneously express or express HER2 and HER3.
  • the dimerization agent may be a ligand of HER3 (for example, Neuregul in pi (NRGbl), NP_039250, NP_039250.2, etc.), a cholesterol-like detergent (e.g., DGTNCdigi tonin; CAS Number : 11024-24-1), GDN (glycol-diosgenin; CAS Number: 1402423-29-3, etc.), and the like.
  • the method comprises contacting HER2 with
  • the NRGbl When the step of treating (injecting) NRGbl into cells simultaneously expressing HER3, the NRGbl may be injected into cells before expression after HER2 and HER3 lysis.
  • the cholesterol-like cleansing agent can be treated (added) to the washing step after cell lysis if it comprises the step of treating the cholesterol-like cleansing agent to cells simultaneously expressing HER2 and HER3 ,
  • the cell may be NRGbl injected in an amount of 10 ng / mL to 1000 ng / mL or 50 ng / mL to 200 ng / mL).
  • the concentration of the cholesterol-like detergent to be used may be a concentration exceeding that of the cleaning agent used.
  • the concentration of the cholesterol-like detergent may be DGTN (W / v), 0.05% (w / v), or 0.06% (w / v) or more, such as 0.05 to 5% ), 0.05 to 3% (w / v), 0.05 to 2% (w / v) or 0.06 to 2% (W / v), 0.005 to 2% (w / v), 0.007 to 2% (w / v) ), Or 0.01 to 2% (w8).
  • the techniques provided herein measure protein-protein interactions intracellularly and / or intercellularly, thereby preventing the disease that the protein is involved in And / or screening or selecting effective drugs for treatment.
  • Fig. 1 is a schematic diagram of a method for measuring short-term molecular protein interactions.
  • FIG. 2 is a graph showing the result of identification of a target protein (first protein) immobilized on a substrate .
  • Figure 3 is a fluorescence image showing protein interactions after infusion of the fluorescently labeled interacting protein (second protein).
  • FIG. 4 is a graph quantifying the number of PPI compexes observed in FIG.
  • FIG. 5 is a graph showing changes in the number of PPI complexes according to the amount of injected cell lysate.
  • FIG. 6 is a schematic diagram showing a process of quantifying a first protein through a monoclonal sandwich ELISA.
  • FIG. 7 is a graph showing the specificity obtained by the single-molecule sandwich ELISA method.
  • FIG. 8 is a graph showing changes in the number of PPI complexes according to the cell line type (red 1 vs light blue 3) and the state (red 1 vs black 2).
  • FIG. 9 is a graph showing changes in the number of PPI complexes according to various target RTKs (first proteins) according to cell conditions.
  • FIG. 10 is a graph showing the change in the PPI complex according to the EGFR mutation state and the calculated ratio of EGFR activated per cell based on the change.
  • FIG. 11 is a graph showing the results of performing the same method for HER2 and HER3 performed on EGFR in FIG.
  • Fig. 12 shows the result of measuring the interactions between EGFR, MET, HER2, HER3 (first protein) and the lower signal transduction protein (second protein) for each cell line and displaying them in a heatmap format.
  • FIG. 13 is a graph (left side and middle) graphically showing the results of FIG. 12 (left side) and a graph showing the reactivity results of AZD9291 (right side) as an EGFR target anticancer drug.
  • FIG. 14 is a graph showing the correlation between the reactivity (left, y axis) of the EGFR target anticancer agent (AZD9291) and Act iat ion score (left, x axis) and the diversity of the target anticancer response (right side) according to the gene type.
  • FIG. 16 shows the results of measurement of biomarker HER2 (upper) and HER3 (intermediate) expression levels used for predicting the reactivity of trastuzumab anticancer drugs in breast cancer cell lines and the degree of suppression of cell growth by trastuzumab (lower)
  • FIG. 16 shows the results of measurement of biomarker HER2 (upper) and HER3 (intermediate) expression levels used for predicting the reactivity of trastuzumab anticancer drugs in breast cancer cell lines and the degree of suppression of cell growth by trastuzumab (lower)
  • FIG. 17 is a graph showing the correlation between the result of measuring the PPI score using the HER2 or HER3 signal and the reactivity of trastuzumab (logGI50).
  • Figure 18 is a heatmap showing the results of the PPI complex signal with the three sub-signal proteins of EGFR, MET, HER2, and HER3 measured in the PDTX mouse model, respectively.
  • FIG. 19 is a graph showing the result of calculating the act iat ion score (bottom) using the expression level (top) of EGFR and the expression level of EGFR in the PDTX mouse model.
  • 20 is a graph showing the results of measurement of changes in tumor size by administering gefitinib to a PDTX mouse model.
  • Figure 21 shows tumor suppression by gef i t inib in a PDTX mouse model
  • FIG. 22 is a graph showing the results of measurements of EGFR PPI complexes measured in tissues before and after the administration of gef i t inib in a PDTX mouse model, respectively.
  • Figures 23A-23I show EGFR target inhibitor reactivity in a lung cancer PDTX model
  • 23a and 23b are graphs showing tumor volume changes in PDTX during the treatment of the vehicle or the indicated EGFR-specific inhibitor, and PDTXs (PDTX-A1 A3) Osimert inib. (PDTX-S1 S5) was treated with 5 mg per kg of weight of dairy (PDTX). 50 mg per kg of weight dairy), showing the size of the tumors obtained (test population per 3 PDTX)
  • 23c is the labeled receptor tyrosine kinase (RTK; EGFR, HER2, HER3, and MET), wherein the PPI complex counts (number of PPI complexes) 23d is a graph showing the results of normalization of EGFR expression levels in 8 PDTX (A1-A3 and S1-S5) individuals to EGFR expression levels in A549 cells (control group)
  • RTK receptor tyrosine kinase
  • 23e and 23f are graphs showing the tumor growth inhibition rate (%) as the y-axis and the value obtained by dividing the EGFR PPI sum obtained from the PDTX model (E) and the SQCC PDTX model (0) by the EGFR level as the x axis.
  • 23g is a graph showing changes in PPI complex counts (the number of PPI complexes between the second protein expressed on the EGFR and the x-axis) when the gefitinib was treated daily for 15 days,
  • 23i is a graph showing the value obtained by dividing the PPI sum by the EGFR level obtained from all 8 PDTX (A1-A3 and S1-S5) individuals as the x axis and the tumor growth inhibition rate as the y axis (Error bars: sd).
  • Figures 24a to 24d show an example of applying single-molecule co-IP and singlenolecule immunolabeling to human tumor samples
  • 24b is the amount of 1o protein produced by monoclonal immuno-labeling
  • PTM Immunolabel 1 ing level
  • PC9 cells for EGFR
  • HCC827 cells for MET
  • SKBR3 cells for HER2 and HER3
  • 24c is a graph showing PPI complex counts for the indicated RTKs at P1 and P2,
  • 24d is a graph showing the change in count PPI complex PLC ga s H2 and Grb2 in the case of the EGFR on the surface treatment hanhu pul l ing down PTPN1 (Error bars :. S d.).
  • 25d is an image showing the immunohistochemical staining (IHC) of EGFR measured in five SQCC PDTX.
  • EGFR expression was calculated by calculating the EGFR H-score as a magnification rule,
  • 25e is a scatter plot showing the correlation between EGFR level and EGFR H-score determined by monomolecular immuno-labeling, and IHC H-score has a complete linear correlation with total EGFR expression level determined by monomolecular immunodiffusion ,
  • 25f and 25g are scatter diagrams showing the correlation between EGFR level (g) and PPI sum (h) of SQCC egg TX and tumor growth inhibition,
  • 25h shows Immunoblot analysis of vehicle or gefitinib-treated PDTX-S2.
  • phosphorylation (pEGFR) of 1068th residue of EGFR is completely disappeared and phosphorylation of AKT and S6K by gefitinib treatment pAkt and pS6K), and these results show that the inhibition of tumor growth in PDTX-S2 is obtained by inhibiting the EGFR / AKT / mT0R / S6K signaling pathway by treatment with gefitinib.
  • 26A and 26B show the effects of gefitinib treatment on PDTX-S1 and PDTX-S2,
  • 26a is a graph showing the change in EGFR level when treated with gefitinib for 15 days (Error bars: s.d .; TF5)
  • FIG. 27A is a schematic diagram showing a process of acquiring a protein, analyzing protein-protein interaction, and quantifying using a cell lysate expressing GFP-PD-1 and mcherry PD-L1, respectively.
  • FIG. 27B is a fluorescence image of the protein-protein interaction obtained by using 10 nM of GFP-egg-1 and mcherry PD-L1.
  • FIG. 27C shows the result of quantitative analysis (number of PPI complexes: fluorescence- counts).
  • FIG. 28A schematically shows a process of screening for an inhibitor candidate drug.
  • FIG. 28B is a graph showing test results for optimizing the reaction conditions (reaction time) of one-step reaction in a tube for drug screening.
  • FIG. Figure 28c shows the number of PPI complexes (F-counts) between PD-1 / PD-L1 obtained using two commercially available egg-1 / PD-L1 inhibitors BMS202 and S7911 (purchased from Sel leck Chem) It is a graph showing.
  • FIG. 28d shows the PPI librarians screening (http: / /. (F-counts) between egg-1 / PD-L1 obtained using a non-macrocyclic ic PPI drug libbrary compound provided by Asinex.com/ppi/.
  • FIG. 28E is a graph showing the results obtained by conducting t i tratation on four drugs (Bl, B2, B3, and E2) selected to exhibit excellent inhibitory effect in FIG. 28D.
  • 29 is a graph showing the number of PPI complexes (F-counts) between inhib-1 / PD-L1 obtained by immobilizing various antibodies on a substrate and using GFP-PD-1 and mcherry PD-L1.
  • FIG. 30A shows Total GFP-1 abe 1 ed count (left) and fluorescent image (right) when GFP PD-L1 is immobilized on a substrate and when GFP-egg-1 is immobilized.
  • FIG. 30B is a graph showing the relationship between the cluster rat io (%) (left) and fluorescence image (right) of GFP PD-L1 and GFP suppression-1 when GFP PD-L1 was immobilized on a substrate, Lt; / RTI >
  • 31A schematically shows a design process of a PD-L1 mutant construct for epitope mapping.
  • FIG. 31B shows the degree of binding of PD-L1 antibody (9A11) to PD-L1 WT (wild-type), d280, or d270 variant.
  • Figure 31c shows the degree of protein-protein interaction between egg-1 and PD-L1 WT, d280, or d270 variants.
  • Figure 33 shows the structures of drugs Bl, B2, B3, and E2.
  • Figure 34A depicts expression of HER3-eGFP in SKBR3, followed by treatment with NRGbl to generate HER2-
  • FIG. 34B shows the HER2-eGFP concentration in the cell extract according to the amount of HER3- HER3-eGFP heterodimer. ≪ / RTI >
  • FIG. 34C is a graph showing the amount of HER2-HER3 heterodimer detected when NRGbl was treated before and after cell lysis.
  • FIG. 34D is a schematic diagram illustrating a process of inducing, extracting, and detecting heterodimers of HER2 and HER3 in cells by treating NRBbl with SKBR3 cells.
  • Figure 34E is a graph showing the amount of HER2-HER3 heterodimer detected according to the amount of cell extract added to the substrate.
  • FIG. 34f is a graph showing that HER2-HER3 heterodimer is mainly present when a cell extract extracted according to the procedure of 34d is attached to a substrate using another kind of antibody (when the ligand is not treated, EGFR-HER3, When EGFR-HER2, HER2-HER3 heterodimer, and ligand were treated, the EGFR-HER3 heterodimer was much less than the amount of HER2-HER3 heterodimer when treated with ligand)
  • Figure 34g illustrates the process of overexpressing HER2-eGFP in four K293T cells to form a HER2-eGFP-HER2-eGFP homodimer and attaching it to a substrate (PEG, neutravidin, biot in, and ant i -HER2 ant ibody surface treated) It is a schematic diagram showing the enemy.
  • FIG. 34h shows the results of measuring the fluorescence signal from the HER2_eGFP-HER2-eGFP homodimer or HER2-eGFP monomer attached to the substrate at 34g.
  • FIG. 34I is a graph showing the ratio of the dimer to the fluorescence signal obtained from the homodimer or monomer at 34h, and the ratio of the fluorescence signal to the fluorescence signal at the second step and the olimeric state.
  • FIG. It can be seen that the ratio of HER2-eGFP to that of step 2 and step 2 is higher than that of eGFP or HER3_eGFP.
  • FIG. 35A is a schematic diagram showing an example of a process for measuring the Tyr phosphorylation level of the HER2-HER3 heterodimer.
  • FIG. 35B is a graph showing the results of measuring the degree of phosphorylation (HER3 pTyr counts) of HER3 Tyr residues after treating ATP and 3 ⁇ 4 2+ on a substrate immobilized with HER2-HER3 heterodimer.
  • Figures 35c and 35d show the degree of activation of HER3 obtained by washing the HER2-HER3 heterodimer-forming cell lysate with various concentrations of digi tonin as the degree of phosphorylation of HER3 Y1289 according to digi tonin concentration Graph.
  • FIG. 35E is a graph showing the degree of HER3 activation according to the kind of detergent used for washing the HER2-HER3 heterodimer-forming cell lysate through the degree of phosphorylation of HER3 Y1289.
  • Figure 35f shows that the degree of activation of the HER2-HER3 heterodimer is decreased when HER2-HER3 heterodimers are subjected to HER2 and HER3, respectively, corresponding to EGFR, in order to show that they follow the same activation mechanism as previously known by the EGFR homodimer beam "gourd is a graph that.
  • FIG. 35G is a schematic diagram showing an example of a process for measuring the Tyr phosphorylation level of the HER2-HER2 homodimer.
  • FIG. 35h is a graph showing the results of measuring the degree of phosphorylation (HER2 pTyr counts) of HER2 Tyr residues after treating ATP and sugar 2+ on a substrate immobilized with a suppressor R2-HER2 homodimer.
  • FIG. 35I is a graph showing the degree of HER2 activation according to the type of detergent used for washing HER2-HER2 homodimer-forming cell lysates through the degree of phosphorylation of HER2 Y1196.
  • 36A is a schematic diagram showing that a HER2-HER3 heterodimer attached to a substrate induces an interaction with an eGFP-labeled lower signaling protein when subjected to a phosphorylation process.
  • FIG. 36B is a graph showing the magnitude of the fluorescence signal as to how much amount of the lower signaling protein (PLC gamma 1) interacts with one HER2-HER3 heterodimer.
  • Figure 36c is a graph showing the number of single molecular off events from a fluorescence signal to see how much of the HER2-HER3, heterodimer, interacts with the lower signal transduction protein (PLC gamma 1).
  • Figure 36d is a graph showing the magnitude of the fluorescence signal as to how much of the HER2-HER3 heterodimer interacts with the amount of the lower signal transduction protein (p85 alpha).
  • Figure 36E is a graph showing how much of the HER2-HER3 heterodimer interacts with the amount of the lower signaling protein (p85 alpha) as the number of single molecule off events from the fluorescence signal.
  • Figure 36f shows how much amount of the lower signaling protein (PI3K: p85 alpha-pi 10 alpha complex) is associated with one HER2-HER3 heterodimer And the intensity of the fluorescence signal.
  • PI3K p85 alpha-pi 10 alpha complex
  • FIG. 36g is a schematic diagram showing that the HER2 homodimer attached to the substrate induces an interaction with the eGFP-labeled lower signal transduction protein upon oxidation.
  • Figure 36h is a graph showing the magnitude of the fluorescence signal as to how much of the HER2 homodimer interacts with the amount of the lower signaling protein (PLC gamma 1).
  • Figure 36i is a graph showing how much of the HER2 homodimer interacts with PLC signaling protein (PLC gamma 1) as the number of single molecular off events from the fluorescence signal.
  • PLC signaling protein PLC gamma 1
  • Figure 36j is a graph showing the magnitude of the fluorescence signal as to how much of the HER2 homodimer interacts with the amount of the lower signaling protein (p85 alpha).
  • Figure 36k is a graph showing how much of the HER2 homodimer interacts with the amount of the lower signaling protein (p85 alpha) as the number of single molecular off events from the fluorescence signal.
  • Figure 37A is a graph showing the phosphorylation rate of the HER3 Tyr residues during ATP and Mg2 + treatment of the HER2-HER3 heterodimer (x-axis is the time taken to phosphorylate and y-axis is the amount of phosphorylated Tyr during that time).
  • Figure 37b shows the results of increasing the ATP treatment concentration in the HER2-HER3 heterodimer
  • FIG. 37C is a graph showing the rate of phosphorylation of HER2 Tyr residues measured while increasing the ATP treatment concentration using a HER2-HER2 homodimer.
  • Figure 37d shows the results of pretreatment of HER2-HER3 heterodimer and HER2 homodimer at various concentrations of Lapat inib followed by treatment with ATP and magnesium chloride ide
  • the activity of HER2 is shown by the concentration of Lapat inib.
  • FIG. 37E is a graph showing the degree of activation of HER2 obtained by treating LER2-HER3 heterodimer and HER2 homodimer with Lapat inib, ATP, and magnesium chlorid ide according to Lapat inib concentration.
  • Figure 37f is a graph showing the HER3 Y1289 phosphorylation level obtained by treating Lapat inib, ATP, and magnesium chloride ide with various concentrations of PTPN1 (Protein Tyrosine Phosphatase, Nonreceptor type 1) in the HER2-HER3 heterodimer according to Lapat inib concentration .
  • Figure 38 is a graph showing the results of application of single-molecule co-IP and single-molecule immunolabeling on human tumor samples.
  • Figures 39a-39g show SH3 domain-mediated interaction (pTyr-independent manner) between Grb2 and mutant EGFR ((a) Suppression of EGFR_Grb2 interaction by gef itinib (100 nM for 24 h) treatment during cell culture.
  • Osimertinib dose as gefitinib) was treated to H1975 for further suppression of EGFR_Grb2 interaction due to the presence of the gatekeeper mutation, T790M, in H1975 cell (bg) single-molecule immunolabeling assay (b), MIG6 (c), GAPDH (d), Shcl (e), EGFR (f) or c-Cbl
  • H1975 cell bg
  • single-molecule immunolabeling assay b
  • MIG6 c
  • GAPDH d
  • Shcl e
  • EGFR f
  • c-Cbl cell extracts used are shown in the following table, and the amount of cell extracts used are as follows: shown at bottom. Error bars in Fig. 40 show sd)
  • Example 1 Preparation of first protein (EGFR, MET, HER2, and HER3)
  • EGFR, MET, HER2, and HER3 were selected as the first protein, and the first protein was obtained from a lysate obtained by dissolving a cell line (for example, cancer cell line) or cancer cell tissue containing the same. This process is described in more detail as follows:
  • the cell line sample prepared in Example 1.1.1 was pipetted and the cells were collected by pipetting.
  • the prepared cell lysis buffer was added to the pipetted cell lysate in an amount of 200 ul per tube, and the cells were reacted for 30 minutes in a cold block (0-4 ° C) placed on ice. At this time, every 10 minutes The physically mixing process was repeated by cyclic pipetting so that the cell lysis reaction by the surfactant was actively performed.
  • Total protein concentration in the reaction product was measured using a total protein measurement method (Bradford, BCA, DC protein assay, etc.), and the protein concentration was measured to be about 5-10 mg / ml.
  • Tumor xenografts from patients with Lung squamous cell carcinoma were obtained from Yonsei University research team.
  • a brief review of the production process of Patient's tumor xenograft (PDTXs) is as follows: 6 to 8 weeks old female severe combined immunodeficient mice (NOG) and nude mice (nu / nu mi ce; Or ientBio). All animal studies were carried out in accordance with guidelines approved by the Insti- tional Animal Care and Use Committee (IACUC).
  • a patient-derived clinical tumor sample was cut into sections of 3 mm or less in size and subcutaneously transplanted into the side of the prepared NOG mouse. Tumor size was measured twice weekly with calipers to determine the growth rate in subcutaneous tissue.
  • mice mice having subsequent tumors continuously derived therefrom were named as mice, and mice having subsequent tumors continuously derived therefrom were referred to as FI, F2, F3, F4 and the like with serial numbers attached thereto.
  • Mice (F3) with third generation subsequent tumor were injected with vehicle (PBS) or gefitinib Xgef it inib).
  • the prepared PDTXs were intraperitoneally administered 50 mg / kg of gefitinib or vehicle once a day. Tumor tissues were collected from PDTX 15 days after the administration of gefitinib and used to monitor PPI and expression level changes described below. 1.2.2 Preparation of tissue solution
  • the tumor tissue obtained in Example 1.2.1 was prepared in an amount of about 20 mm 3 , but it may be larger.
  • Example 1.1.2 Approximately 300 ⁇ L of the lysis buffer prepared in Example 1.1.2 was added to 20 mm 3 of the prepared tumor tissue, and the mixture was reacted while continuing to rotate in a 4 ° C refrigerator for 1 hour. At this time, the size of the tissue was made as small as possible using surgical scissors, and the surface area per unit volume was widened so that the chemical reaction by the surfactant in the dissolution buffer occurred as efficiently as possible. After 1 hour of reaction as described above, centrifugation was performed (10 min, 15,000 g, 4 ° C). Thereafter, the pellet is discarded, the supernatant is removed, the membrane is filtered using a membrane having a pore size of 0.2 mu, and the portion of the membrane passed through the membrane is transferred to a new tube and stored Respectively.
  • Total protein concentration in the reaction product was measured using a total protein measurement method (Bradford, BCA, DC protein assay, etc.), and the protein concentration was measured to be about 5-10 mg / ml.
  • HEK293 cells ATCC
  • HeLa cells ATCC
  • the cells were lysed in a solution (50 mM Tris-HCl (pH 7.4), 1% Triton X-100, 150 mM NaCl, 1 mM EDTA, 10% glycerol, protease inhibitor cocktail (Sigma, P8340) 100X, tyrosine phosphatase inhibitor cocktail (Sigma, P5726) 100X).
  • High concentrations of surfactant (Triton X-100) was added 60 uL of the solution with respect to the first 5x10 s cel Is, since it can interfere with the protein interactions.
  • the cells in the obtained reaction mixture were released by pipetting and reacted for 30 minutes in a cold block (0-4 ° C) placed on ice. At this time, physical mixing was performed by periodic pipetting at intervals of 10 minutes so that dissolution reaction by the surfactant was actively performed.
  • aqueous solution 60 was added to 140 uL of PBS. Finally, a solution of the second protein containing 0.3% Triton X-100 can be obtained. Fluoremeter was used to measure the concentration of fluorescent protein attached to the second protein. As a result, the concentrations of the three second proteins used were measured within the range of about 400 to 1000 nM.
  • the cells were washed with 3 rd distilled water and stored at -20 ° C until used without contact with the PEG-coated surface.
  • a quartz channel type substrate or an acrylic type well type substrate was prepared.
  • cleaning and PEG coating processes were performed with reference to the cover si ip process described above.
  • the substrate was immersed in the third distilled water and sonicated for washing.
  • the washed acrylic substrate was immersed in a 5% BSA solution and reacted for 2 hours to prevent nonspecific protein adsorption and stored at -20 C until use.
  • the prepared cover lip and acrylic substrate were used. Before the test, the coverslip and the acrylic substrate were thawed or assembled, or they were pre-assembled after the PEG coating and stored in -20C And thawed before use.
  • Example 4 Protein-protein interaction imaging between a first protein and a second protein
  • Neutravidin (Thermo, A2666), an Avidin family protein, was added to the prepared substrate at a concentration of 0.1 mg / ml. After 5 minutes of reaction at room temperature, the substrate was washed twice with 30 ⁇ l of PBS buffer.
  • an antibody against the first protein to be a target was added to the prepared substrate.
  • the antibody to be used was prepared as a biotin-conjugated form.
  • the concentration of the antibody can be appropriately adjusted according to the affinity (dissociation constant, KD) of the antibody-antigen. In this experiment, the antibody concentration was about 2 ug / ml and the reaction time was about 5 minutes. If a biotin-conjugated antibody is used, the antibody of the first protein can be attached using a secondary antibody.
  • Antibodies to the first protein used at this time are summarized in the following Table 3:
  • the substrate treated with the antibody was washed twice with 30 ul of PBS buf fer.
  • a cell lysis solution or a tissue lysis solution containing the first protein well prepared in Example 1 was added to the prepared substrate.
  • the antigen-antibody reaction efficiency could be lowered to 15 minutes and the reaction time could be lowered to 15 minutes or more.
  • the substrate was washed with PBS containing 0.05% (v / v) of tween 20.
  • Tween 20 0.05% reduces nonspecic binding and helps prevent the hydrophobic c region of membrane proteins from breaking down.
  • the concentration of the second protein in the first protein lysis solution used was between 1 and 50 nM (about 30 nM) based on the fluorescent protein. If the concentration of the second protein is more than 100 nM, the background noise increases in the fluorescence microscope, which hinders accurate fluorescence signal measurement.
  • the substrate was immobilized on a fluorescence microscope and imaged to obtain data for each first protein / second protein.
  • Protein complexes were analyzed based on the toolkits provided in the Mat 1 ab program (provided by MathWorks).
  • the fluorescence image obtained in Example 4 was stored in a 16-bit unsigned integer format.
  • the fluorescence signal was obtained from eGFP (enhanced green fluorescence protein).
  • the wavelength of the laser was set to 488 nm and the laser power was adjusted to 2 mW to maintain the emission time of eGFP for about 11 seconds.
  • the first frame is discarded, and the image of the frame is averaged to generate one image. This process is repeatedly carried out while shifting the position in the wel l, Respectively.
  • the reason for discarding the early frame is to selectively use the section where the autofluorescence on the surface of the substrate disappears and the eGFP signal is maintained.
  • the selection interval may vary depending on the imaging condition / equipment construction status.
  • the exposure time per frame is set to 0.1 second by using an EMCCD (Electron-Multi loading charging-coupled device; Andor iXon Ul tra 897 EX2 (DU-897U-CS0-EXF) A fluorescence image was obtained with an EMCCD gain value of 40.
  • EMCCD Electro-Multi loading charging-coupled device
  • iXon Ul tra 897 EX2 DU-897U-CS0-EXF
  • the initial start begins at the top left corner.
  • One frame consists of 512x512 pixels. 11 pixels to the right based on the base pixel, down
  • the median value is obtained from 11 11x11 individual pixels, and this median value is subtracted from the reference pixel value [(Intensi_pixel) _
  • Threshold is the threshold value that makes the pixel intensities below the threshold in the whole image (using the algorithm to find the local maximum in the Matlab toolkit). This eliminates the local maximum that was not produced by the fluorescence signal in the image.
  • the threshold value used in the imaging conditions of this embodiment is 70.
  • the signal from the fluorescent protein occurs as a localized spreading func- tion (PSF).
  • PSF localized spreading func- tion
  • the PSF value was converted to the biological value PPI complex value through the following procedure:
  • the number of PSFs was calculated, and the mean and standard deviation were calculated. This value finally represents the number of PPI complexes under certain conditions (indicated as "Number of single PPI complexes" in the figure).
  • the concentration of the cell lysate (see Example 1.1.2) is plotted on the x-axis and the slope of the graph obtained by plotting the PPI complex value measured in each cell lysate (see Example 5), that is, [(PPI complex ) / (Number of PPI complexes per unit concentration of cell lysate (Wml))] was defined as the PPI intensity (or PPI slope) between the first protein and the second protein in the cells. All PPI intensities obtained for each cell line were combined to obtain the total PPI intensity, which was defined as the PPI score for the cell line.
  • the PPI intensity sum (or PPI score) represents the total PPI level of the tested first and low 12 proteins at the cell lysate unit concentration of each cell line.
  • RTK (EGFR, MET, HER2, or HER3 for lung cancer; HER2 and HER3 for breast cancer);
  • Second protein downstream protein (PLC-ga_a_SH2, Grb2, p85_alpha)).
  • PPI score of the cell line (Except for the reference cells), so that the PPI score of the cell line (hereinafter referred to as reference cell, in this test example, PC9 cells among lung cancer cell lines and SKBR3 cells among breast cancer cell lines, respectively)
  • test cells PC9 cells among lung cancer cell lines and SKBR3 cells among breast cancer cell lines, respectively
  • the total amount of the first protein e.g., RTK (EGFR, MET, HER2, or HER3 for lung cancer; HER2 and R4 for breast cancer
  • the total amount of the first protein was determined by sandwich ELISA or quantitative western blot using each of the antibodies described above (see Table 3), and then divided by the weight of the cell lysate (total protein weight in the cell lysate) Respectively.
  • a value obtained by dividing the obtained PPI score or normalized PPI score by the total amount of the first protein was defined as an activation score.
  • activation scores obtained from the test cells were normalized so that the activation score of the reference cells (lung cancer cell line: PC9 cell, breast cancer cell line: SKBR3 cell) was 1, The value obtained for the cell line was defined as the normalized activation score for that cell line.
  • the negative backgromid can use normal tissue of the same patient or cancer cell lysate whose test protein (for example, EGFR) is normal.
  • test protein for example, EGFR
  • the degree of interaction between the EGFR and the sub-protein is measured to obtain the PPI score.
  • Heatmap was created to add supplemental judgment to its analysis. Heatmap is only one way to show data and is not intended to limit data interpretation.
  • one axis is a second protein (lower signal protein)
  • the other axis is the number of cell lines (15 kinds (lung cancer cell line) or 11 (breast cancer cell line): 3x16 (number of the second protein (total 3: see Table 2 (p85_alpha, Grb2, PLC-ga_a_SH2) (EGFR, MET, HER2, and HER3; lung cancer), or a total of two (HER2 and HER3; breast cancer) by the first protein (see Table 1)
  • HER3 only p85-alpha is used as the second protein).
  • a heatmap was prepared by varying the color and brightness according to the PPI intensity between the first protein and the second protein obtained from the corresponding cells (for example, as the PPI intensity is increased, dark, black, , Which is not the value determined by the tester for each test). Regardless of which cell is the reference, the relative differences between the cell lines remain unchanged.
  • FIG. 1 is a schematic diagram of a method for measuring monomolecular protein interactions.
  • the Neutravidin, RTK antibody, and the cell lysis solution or tissue lysis solution to be analyzed are sequentially injected into the substrate coated with PolyeUlyene glycol, and then the sample is washed.
  • the target RTK protein It shows how it is fixed to the substrate.
  • the fluorescence signal is observed and quantified by injecting fluorescently labeled interacting protein to the substrate, and the degree of monomolecular protein interaction is measured.
  • FIG. 2 is a graph showing the result of identification of a target protein (first protein) immobilized on a substrate. With reference to the methods described in Examples 4 and 5. EGF was treated for 3 minutes in an amount of 100 ng 8 il, and the graph on the left side of Fig. 2
  • H antigens were used to attach to the substrate via an ant ibody (MAS-13266, Themof i sher) that binds to the extracellular domain of EGFR using H1666 and an ant ibody for the intracellular domain of EGFR (# 4267, Cel l signaling technology) 2 shows the result of confirming whether or not EGFR forms a dimer with HER2.
  • the graph on the right side of FIG. 2 shows whether or not EGFR forms a dimer with Shcl.
  • Shcl The results are shown by inserting the antibody.
  • the target RTK protein (first protein) It is fixed (+), not fixed (-).
  • various target proteins can be attached to the substrate through appropriate antibody screening.
  • EGFR-HER2 or EGFR-Shcl it can be confirmed that not only a single target protein but also a protein conjugate existing in vivo can be immobilized on a substrate.
  • FIG. 3 is an image showing protein interactions after the injection of fluorescently labeled interacting protein (second protein) onto a substrate to which the first protein is immobilized.
  • the PPI complexes identified in the method described in Example 5 appear in the form of a point spread function (PSF), and the PPI complexes were selected through computer algorithms. It can be seen that the fluorescence signal occurs only when the lower signaling protein is injected. Green circles represent the observed PPI complexes.
  • FIG. 4 is a graph quantifying the number of PPI compexes observed in FIG. High PPI complexes were selectively observed only when the lower signaling protein (the second protein) was injected (designated PLCgammaSH2, Grb2, and p85-alpha in the x-axis).
  • the target RTK protein the first protein (EGFR) antibody is absent (black bar) and the injected lower signaling protein is absent (buf fer of the x axis)
  • the observed signal is very small. In both cases, it can be interpreted as background noise.
  • FIG. 5 is a graph showing the increase in the number of PPI complexes according to the amount of injected cell lysate. It can be seen that the amount of observed PPI complex (y axis) increases linearly as the amount of cell lysate containing the target RTK protein (first protein: EGFR) increases. This allows a quantitative comparison of the PPI compex between samples in the amount of specific cell lysate.
  • FIG. 6 is a schematic diagram showing a process of quantifying a first protein through a monomolecular sandwich ELISA.
  • the process of attaching the target RTK protein (first protein) to the surface of the substrate is the same as in Fig.
  • a second antibody recognizing the target RTK protein can be injected to measure the amount of target RTK protein.
  • the second antibody used here is a first antibody (pull down ant ibody) used to immobilize the target RTK protein on the surface of the substrate and a second antibody (epi toe) on the target RTK protein ).
  • FIG. 7 is a graph showing the specificity obtained by the single-molecule sandwich ELISA method. It can be seen that the single molecule sandwich ELISA signal result is suppressed even if only one component out of the components shown in the schematic diagram of FIG. 6 is omitted.
  • FIG. 8 is a graph showing changes in the number of PPI complexes according to the cell line type (red 1 vs light blue 3) and the state (red 1 vs black 2).
  • the target RTK protein first protein; EGFR
  • EGF + corresponding ligand
  • PC9, light blue active mutations in the target RTK indicates that the number of PPI complexes in the observed target RTK increases.
  • FIG. 9 is a graph showing the PPI complex number change (PPI slope) per unit concentration of a sample for various target RTKs (first protein) according to cell conditions.
  • PPI slope PPI complex number change
  • FIG. 10 is a graph showing the change in the PPI complex according to the EGFR mutation state and the calculated ratio of EGFR activated per cell based on the change.
  • FIG. The upper part shows the result of measuring the interaction between the EGFR and the lower signal transduction protein for each individual cell by the PPI complex measurement method and the lower part shows the EGFR expression per cell through the single molecule sandwicz ELISA (see FIG. 6) After the two values were divided, the amount of activated EGFR per cell was measured (Absolute occupancy (%)).
  • FIG. 11 is a graph showing absolute occupancy (%) results obtained by performing the same method for HER2 and HER3 performed on EGFR in FIG. In the case of HER2, the activity is very low, while HER3 shows a very high activity ratio.
  • FIG. 12 is a graph showing the interactions (signal intensity) between EGFR, MET, HER2, HER3 (first protein) and the lower signal transduction protein (second protein) Show one result.
  • the color indicators for each signal strength are shown below.
  • FIG. 13 is a graph (left and middle) showing values obtained by adding numerical values obtained by quantifying the signal intensities between EGFR (first protein) and 3 kinds of second proteins among the results of FIG. 12, and AZD9291 0.0 >(IC50;< / RTI > And the cell survival rate is 50% as compared with that before treatment) (right side).
  • the color of each bar is distinguished by the group shown on the right depending on the EGFR gene mutation status.
  • the Act iat ion score showed a more significant correlation with the drug response (IC50) compared to the PPI score. (The higher the Act iat ion score, the lower the IC50 value (the higher the drug reactivity)).
  • 14 is a graph showing the correlation between the reactivity (y axis) and the Act iat ion score (x axis) of the EGFR target anticancer agent (AZD9291) (left) and the diversity of the target anticancer response (right) depending on the gene type.
  • FIG. 15 is a diagram showing the intensity (interaction) of signals between HER2 and HER3 (first protein) and a lower signal transduction protein (second protein) in a breast cancer cell line in the heatmap format (Example 7).
  • FIG. 16 shows the results of measurement of biomarker HER2 (upper) and HER3 (intermediate) expression levels used for predicting the reactivity of trastuzumab anticancer drugs in breast cancer cell lines and the degree of suppression of cell growth by trastuzumab (lower)
  • FIG. 16 shows the results of measurement of biomarker HER2 (upper) and HER3 (intermediate) expression levels used for predicting the reactivity of trastuzumab anticancer drugs in breast cancer cell lines and the degree of suppression of cell growth by trastuzumab (lower)
  • FIG 17 is a graph showing the correlation between the result of measuring the PPI score using the HER2 or HER3 signal and the reactivity of trastuzumab (logGI50).
  • FIG. 19 shows the expression levels (top) of EGFR and the expression levels of EGFR (results of FIG. 18) in the tissue lysate (Example 1.2) obtained in the PDTX mouse model This is a graph showing the result of calculating the activation score (bottom).
  • FIG. 20 is a graph showing the result of measuring the change in tumor size by administering gefitinib (50 mg / kg) to a PDTX mouse model (Example 1.2.1) in comparison with the results in Vehicle (PBS) administration group.
  • PBS Vehicle
  • the expression level of EGFR was not high, but the activation score was high (see FIG. 19) and the antitumor effect was also excellent (see FIG. 20).
  • the activated activation score ie, the activated EGFR ratio
  • FIG. 22 is a graph showing the results of measuring the number of EGFR PPI complexes per unit concentration of the samples measured in the tissue lysate samples before and after the administration of gefit inib (50 mg / kg) in the PDTX mouse model (Example 1.2.1) to be.
  • the EGFR PPI complexes were significantly reduced. This may be evidence that the EGFR signal is suppressed by gefitinib.
  • anti-EGFR antibody MS-378-B0 ThermoFisher
  • anti-MET antibody anti-MET antibody
  • anti- HER2 antibody BMS120BT ThermoFisher
  • HER3 antibody BAM348 R & D systems
  • mCherry ab34771 Abeam
  • anti-KRas antibody sc-521
  • EGFR antibody 4267 Cell signaling
  • anti-EGFR (pTyr 1068) antibody (ab32430 Abeam) was used as the detection antibody for each of the corresponding proteins and PTMs (post-translational modi fi cat ions)
  • Anti-EGFR (pTyr 1086) antibody (ab32086 Abeam)
  • anti-EGFR (pTyr 1173) antibody (4407 cell signaling)
  • anti-MET antibody 8494 Cell signaling
  • HER2 (pTyr 1221/1222) antibody (2243 Cell signaling), anti-HER3 antibody (ab32121 Abeam), anti-HER3 (pTyr 1289) technology, cat. No. (Ab32037 Abeam), anti-Shcl antibody (ab33770 Abeam), anti-She pTyr 239/240) antibody (abl09455 Abeam), anti-HSP90 antibody (PA3-013 ThermoF i sher), anti- MIG6 antibody (11630-1-AP Proteintech), anti-GAPDH antibody (3906 cell signaling), and anti-c-Cbl antibody (2179 Cell signaling).
  • ImmunoResearch antibodies were used as secondary antibodies.
  • Western blotting was performed using the following antibodies: anti-EGFR (pTyr 1068) antibody (2234 Cell signaling), anti-EGFR antibody (2232 Cell signaling), anti-Erk (pThr202 / Tyr204) ), Anti-Erk antibody (4696 cell signaling), anti-Akt (pSer473) antibody (4060 cell signaling), anti-Akt antibody (4691 cell signaling), anti- S6K (pSer235 / 236) Anti-S6K antibody (2217 Cell signaling), and anti-actin antibody (ab8227 Abeam) were used.
  • EGFR was stimulated using lOOng / ml EGF (PHG0311L Life technologies) (3 min).
  • the changes in PPI and lung cancer cells in lung adenocarcinoma cells were compared with that of Gefitinib (S1025 Sel leckchem), Osimert inib (S7297 Sel leckchem), BKM120 (S2247 Sel leckchem), Dabrafenib (S2807 Sel leckchem), and Trastuzumab HER2- / HER3-PPI assay, MTT assay, and tumor growth assay in the PDTX model.
  • Shcl human Shcl, Addgene 73255
  • Eat2 human Eat 2, Addgene 46423
  • APCS human APCS, Addgene 46477
  • Nckl human
  • Grb2 human Grb2; Addgene 46442
  • p85a mouse p85a Addgene 1399
  • S0S1 human S0S1, Addgene 32920
  • the eGFP-tagged CARM1 human CARM1
  • EGFR genes were obtained from Seoul National University (Korea) and KAIST (Korea), respectively. All cDNAs were cloned into pEGFP-Cl (Clontech Laboratories) to produce corresponding eGFP-1 abe 1 ed prey proteins. Mutations of W36K, R86M and W193K were introduced into the Grb2 gene, respectively, to produce N * -, SH2 * -, and C * - constructs of Grb2 variants. EGFR mutants were prepared by either deleting E746-A750 from the EGFR gene or replacing the 858th residue, lysine, with arginine.
  • Plasmid DNA 30 Were mixed with 100 M of HEK293 cell suspension containing 2x10 6 cells. Two 950V electric pulses (with a duration of 35 ms for each pulse) were applied to HEK293 cells. Transfected cells were harvested 24 hours after transfection and stored at -80 ° C.
  • PBS Phosphata buffered saline
  • osmate inib osmate inib
  • gefitinib osmate inib
  • PDTX patient-derived tumor xenograft
  • NeutrAvidin (10 jd of 0.1 mg / ml: A2666 Life technologies) was placed in each individual reaction chamber. After 10 minutes of incubation, unbound NeutrAvidin was removed. A miniaturized imaging chamber was filled with PBS After PBS was completely removed, biotinylated pul 1 down antibodies were incubated on a NeutrAvidin coated surface for 10 min, made. For MET antibodies, the primary antibody was conjugated using a biot inylated secondary antibody (alpha- mouse IgG). After washing with PBS, cancer cells or tumor tissue extracts were applied to the antibody coated surface. After 15 minutes, the unbound extract was removed and the chamber was soaked in a reservoir filled with PBS supplemented with 0.05% (v / v) Tween 20.
  • transformed HEK293 cell extracts were diluted with 30 nM (eGFP-tagged probe protein) and loaded into the imaging chamber. Implants were placed on a TIRF microscope and eGFP fluorescence was recorded with EMC suppression (20 frames; 100-ms exposure).
  • dye-labeled detection antibody was used instead of eGFP-labeled probe protein for 5 frames.
  • the detection antibody was chosen to have an epitope within the tyrosine residue on the cytoplasmic kinase site or end (tai l).
  • Detection antibodies were directly labeled with Alexa488 (MET antibody) or indirectly with Cy3-labeled secondary antibodies (EGFR, HER2, HER3 and pTyr antibodies).
  • EGFR, HER2, HER3 and pTyr antibodies Cy3-labeled secondary antibodies
  • Example 9.6 The TIFF file obtained by counting fluorescence imaging of the counting PPI complex and immuno-labeled proteins was analyzed with a custom GUI (Matlab 2016a, MathWorks). Three frames (17-19 for eGFP, 3_5 for Cy3 and Alexa488) were used to confirm the local maxima of intensities representing a single PPI complex or immunoreactive protein. For the background correction, the images obtained by spatial aliasing (11 ⁇ 11 pixels) were subtracted frame by frame from the original image. The obtained images were averaged and used to detect thresholding to local maxima (using custom Mat 1 ab GUI).
  • the PPI measurement index of HER receptor is closely related to the drug reactivity of cancer And whether single-molecule immuno-labeling or co-IP analyses can be applied to screening for specific cancers that are reactive to HER receptor target therapy (HER receptor-targeted therapies have anticancer effects) Respectively.
  • HER receptor-targeted therapies have anticancer effects
  • three types of lung cancer xenograft-derived tumor xenografted mice PDTXs; PDTX-
  • PDTX-A1-A3 Treatment of ovarian carcinoma PDTXs (PDTX-A1-A3; each population of 3 or more, the results of which are shown as the mean value) for 30 days with osmate inib (5 mg per kg of weight dairy) And compared with the control group (vehicle-treated group), the results are shown on the left side of FIG. 23B.
  • PDTX-A1 A3 showed a significant decrease in tumor size by osimert inib treatment (A1> A2> A3).
  • the PPI complex (represented by PPI count in Figure 23c) between each of the EGFR, HER2, HER3 and MET receptors and the lower signal proteins PLCgammaSH2, Grb2 and p85-alpha was measured in each egg TX (PDTX-A1 to A3) , And the left side of Fig. 23C.
  • the PPI complex counts between EGFR and the three sub-signal proteins were in the order of A1> A2> A3. This is due to the effect of the EGFR inhibitor osimert inib on tumor size reduction The same can be confirmed.
  • lung cancer While 29% of lung cancer is associated with sensitization mutations of EGFR, only 0.5% of lung SQCC have EGFR sensitization mutations. Therefore, at present, there is no appropriate biomarker for EGFR target treatment in lung SQCC.
  • PDTX_S1 S5 five suppressed TX (PDTX_S1 S5) were constructed from lung SQCC patient tissues and monolayer immuno-labeling and co-IP profiling (Example 9.5) were performed.
  • PDTX-S1-S5 Five suppressive TXs (PDTX-S1-S5) were treated with gefitinib for 15 days, The degree of tumor growth was measured and shown on the right side of FIG. 23B. PDTX-S1-S5 showed significant tumor suppression effects in S1 and S2. Of the various PDTX individuals tested, it was once again confirmed that the PPI complex count (activation score) normalized to EGFR level had a very high correlation (Spearman correlation of 0.9) with tumor growth inhibition ( Figure 23 f and Figure 25f- h).
  • PPI complex counts in PDTX-S1 and PDTX-S2 were measured before and 15 days after gefitinib treatment and are shown in Figures 23g and 26a_b.
  • PDTX-S1 maintained a detectable level of EGFR PPI complex counts, especially in the regulatory p85 a subunit.
  • PDTX-S2 on the other hand, exhibited reduced or indistinguishable levels of EGFR PPI complex counts compared to negative control with A549 cells ( Figure 23g and Figure 26).
  • PDTX-S1 exhibits increased EGFR-p85a binding
  • PDTX-S1 was treated with PI3K inhibitor BKM120 (50 mg / kg) ( Figure 23h).
  • BKM120 showed strong tumor growth inhibitory effect at the same dose (50 mg / kg) than gefitinib.
  • combination therapy of gefitinib and BKM120 (gefitinib (50 mpk (mg per lkg weight (mpk)) / BKM120 (50 mpk) atrophied the tumor.
  • the data points displayed a consistent pattern and formed a shape that closely matched the tumor growth inhibition with a Spearman correlation of 0.95.
  • Tumor tissues of human patients were characterized using the micro chamber and high-throughput single-molecule imaging system of the present invention (Fig. 24).
  • the protocol (cryogenic lysi s protocol) was applied to two lung cancer patient tissues (Yonsei University Severance Hospital) obtained by surgical resection from a lung cancer patient (Fig. 24A, P1 and P2).
  • the prepared patient tissue is pulverized ( ⁇ 0.6 cm) and immersed in liquid nitrogen. After further pulverization, PBS was added to dissolve completely, centrifuged, and pellets were taken. PBS was then added and incubated at 4 ° C with continued mixing. Then, the supernatant was taken by centrifugation.
  • tissues of 15 mm 3 (P1) and 18 mm 3 (P2) in size we obtained 10 different PPI levels for each tissue lysate obtained by the method described above and 1.0 for the PPI complex When P1 and
  • Patient P1 had a partial response to gefitinib treatment for approximately one and a half years prior to the diagnosis of progressive disease (PD), a partial response (PR) to the response evaluation criteria in solid tumors (RECIST) While Patient P2 maintained a stable disease (SD) diagnosis for one year before PD designation.
  • PD progressive disease
  • PR partial response
  • SD stable disease
  • Sup-1 is labeled with GFP and labeled with GFP labeled egg-1, while PD-L1 is labeled with mcherry Labeled and prepared in the form of mcherry labeled PD-L1.
  • Egg-1-GFP has a protein sequence from 1 to 288 including the unique signal sequence of human egg-1 (NP_005009.2) and GFP linked to the carboxy terminal (C-teminal) of the amino acid sequence (Amino acid sequence: MVSKGEELFTGWPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTLTYGVQ
  • CFSRYPDHMKQHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGH KLEYNYNSHNVYIMADKQKNGIKANFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNHYLSTQSALSK consists DPNEKRDHMVLLEFVTMGITLGMDELYK), the two proteins were prepared to be connected by a peptide linker consisting of 19 amino acid (N terminal side: PD-1, C terminal side: GFP).
  • amino acid sequence of the linker used between PD-1-GFP and the nucleic acid sequence encoding it is as follows:
  • the linker may include, but is not limited to, one or more G (glycine) and / or one or more S (serine). Glycine and serine are relatively flexible compared to other amino acids, which can be advantageous for protein folding and PPI measurement.
  • PD-Ll-mcherry has a protein sequence from 1 to 290 including a signal sequence unique to human PD-L1 (NP_054862.1) and a mcherry terminal linked to the carboxy terminal (C-terminal) of the protein sequence (Amino acid sequence: MVSKGEEDNMAIIKEFMRFKVHMEGSVNGHEFEIEGEGEGRPYEGTQTAKLKVTKGGPLPFAffDILSPQF
  • amino acid sequence of the linker used between PD-L1-mcherry and the nucleic acid sequence encoding it is as follows:
  • the linker used is a structure consisting of 16 amino acids and connects the N-terminal PD-L1 and the C terminal mcherry. If the length of the linker is shortened to a certain number, e. G., Less than 8 amino acids by amino acid number, or if it consists of rigid and / or bulky amino acids, Clashes occur between GFP1 and the GFP at the back (C-terminal portion), which can result in mi-sfolding of proteins or degradation, which can affect PPI measurement.
  • the linker may include, but is not limited to, one or more G (glycine) and / or one or more S (serine). Glycine and serine are relatively lexible compared to other amino acids, which can be advantageous for protein folding and PPI measurement.
  • the linker becomes too short, the binding with the PD_L1 antibody using the C-terminus as epi tope (for example, 9A11 (Cell Signaling Technology, # 29122S)) is seriously impaired and pul 1-down The efficiency may drop significantly, which may affect the PPI measurement.
  • 9A11 Cell Signaling Technology, # 29122S
  • the label (mcherry in this embodiment) bound to the PD-L1 immobilized on the substrate (i.e., used as the first protein) is not indispensable for the PPI measurement and can be suppressed without attaching mcherry to the PD- -L1 interactions can be measured.
  • PD-L1 when a label (for example, mcherry) is attached to PD-L1 used as the first protein, PD-L1 attached to the substrate can be quantified.
  • fluorescence quantification using a spectrophotometer is possible, so that the desired concentration of PD-L1 can be coated on the substrate, and conversely, the number of PD-L1 coated on the substrate under the corresponding conditions can be quantitatively tracked.
  • Tracking the number of PD-L1 coatings on the substrate can aid in the negativity of the inhibi tor when performing drug screening.
  • 1) -1 * (* indicates fluorescence labeling) / PD-L1 / inhibi tor mixture is incubated with Tube reactant and the PD-1 * counts to inhibit the interaction of PEKL and PD-L1 , But drugs that sometimes interfere with the binding of PD-L1 and PD-L1 antibodies may also be misdiagnosed. At this time, the number of PD-L1 bound to the substrate can be observed to select the wrongly selected drug.
  • Egg-1-GFP and soybean oil encoding egg Ll-mcherry were inserted into pCDNA 3.1 backbone, respectively, and introduced into HEK293T cells, which were expressed as proteins.
  • any means generally used can be used such as transfection using PEI (polyethyleneimine), 1 ipofect ion using 1 ipofectamine, electroporat ion, and the like.
  • transfect ion was exemplarily performed using PEI.
  • PEI is a highly positively charged substance that penetrates DNA into the cell and expresses it as a protein.
  • the specific procedure of transfect ion using PEI is as follows:
  • 500,000 HEK293T cells (ATCC [293T (ATCC® CRL-3216 TM)] were prepared on the basis of 100 mm di sh. 2 ml of Opti -MEM buf fer (Thermo Fi sher Scientif ic) was prepared, 1 ml of DNA (vector containing the previously prepared egg-1-GFP and PDLl-mcherry gene) and 1 ml of the remaining 50 ml of PEI . After 5 minutes, the DNA and PEI were mixed together and incubated for an additional 20 minutes. The DNA and PEI mixture was treated with the prepared HEK293T cells, cultured for 48 hours, and then the cells were collected.
  • ATCC [293T ATCC® CRL-3216 TM
  • the composition of the lysis buffer (Lysi s buf fer) used in the cell lysis procedure is as follows.
  • Re-suspension was added to dissolve buffer to the amount of cells collected, incubated in ice for 30 min, and then centrifuged at 14000 g for 15 min. At this time, protease inhibitor (Sigma-Aldrich (11836170001)) and phosphatase inhibitor (Sigma-Aldrich (P5726)) were added. The supernatant was taken and the fluorescence intensity was measured using a Fluoremeter and stored.
  • washed with a aminopropylsi against the covers 1 ip surface was washed lane and PEG (polyethylene glycol) were sequentially treated to perform coating.
  • the cells were washed with 3 rd distilled water and stored at -20 ° C until used without contact with the PEG-coated surface.
  • a quartz channel type substrate or an acrylic type well type substrate was prepared.
  • cleaning and PEG coating processes were performed with reference to the cover si ip process described above.
  • the substrate was immersed in the third distilled water and sonicated for washing.
  • the washed acrylic substrate was immersed in a 5% BSA solution and reacted for 2 hours to prevent nonspecific protein adsorption and stored at -20 C until use.
  • the prepared cover lip and acrylic substrate were used. Before the test, the coverslip and the acrylic substrate were thawed or assembled. After the PEG coating was completed, the assembly was pre-assembled and stored at -20 ° C And thawed before use .
  • the substrate prepared in Example 13 was added with the Avidin series protein Neutravidin (Thermo, A2666) was added at a concentration of 0.1 mg / ml. After 5 minutes of reaction at room temperature, the substrate was washed twice with 30 ⁇ l of PBS buf fer. To illustrate the use of PD-L1 or egg-1 as the first protein, the prepared substrate was treated with the antibody against the first protein. At this time, the antibody used was prepared as a biotin-conjugated form. The concentration of the antibody can be appropriately adjusted according to the affinity of the antibody-antigen (KD). In this experiment, the antibody concentration was about 2 ⁇ g / ml, and the reaction time was about 5 minutes. If a biotin-conjugated antibody is used, the antibody of the first protein can be attached using a secondary antibody.
  • KD affinity of the antibody-antigen
  • Antibodies to the first protein used in this example are summarized in Table 6 below:
  • the substrate treated with the anti-PD-L1 antibody (9A11 or 8T4X) shown in Table 6 was washed twice with PBS buf fer 30.
  • PD-L1 protein solubilization solution (supernatant after centrifugation of mCherry-PD-L1 expressing cell lysis solution; used as the first protein) prepared in Example 12 was added to the prepared substrate, and PD-L1 was immobilized on the substrate .
  • antigen-antibody reaction it is possible to continue to increase until 15 minutes, and the efficiency of antigen-antibody reaction may be lowered over 15 minutes and the reaction time is set to about 15 minutes.
  • the substrate was washed with PBS containing 0.05% (v / v) of tween 20. Tween 20 0.05% reduces nonspecic binding and helps prevent the hydrophobic region of the membrane protein from breaking down.
  • PD-1 protein solution (supernatant after centrifugation of GFP-PD-1 expressing cell lysate; used as the second protein) obtained in Example 12 was added.
  • the PD-L1 protein contained 10 nM And egg-1 protein was used at a concentration of 10 nM, 30 nM, or 50 nM.
  • the test procedure is schematically shown in Fig. 27A.
  • the PPI complex imaging process is as follows:
  • the prepared substrate (either a well type or a quartz slide can be used) was fixed on a total internal reflection fluorescence (TIRF) microscope.
  • TIRF total internal reflection fluorescence
  • a wavelength of 488 nm was used.
  • the present invention is not limited thereto, and any wavelength laser capable of emitting eGFP can be used (laser wavelength range: 460 to 500 nm). Fluorescence images were obtained using an EMCCD (Electron-multiplying charge-coupled device; Andor iXon Ultra 897 EX2 (DU-897U-CS0-EXF)) camera.
  • the laser power range is a range that maintains the light-emitting time (lifetime) of e GFP about 11 seconds.
  • the photons generated in eGFP are converted to electrons through the EMCCD element (photoelectric effect, photoelectric effect).
  • the number of electrons generated per photon can be changed through the gain value.
  • the higher the set gain the greater the number of electrons generated per photon, and the higher the sensitivity of the EMCCD.
  • background noise also increases together, so the signal-to-noise ratio is important. Therefore, although the gain of the EMC suppression is adjusted to 40 in the present embodiment, the present invention is not limited to this, and can be appropriately changed based on the foregoing description.
  • d. Total shooting frame 10 frames
  • the intensity of the signal observed in the imaging is extremely stable since the imaging is performed in succession of the incubation for 30 minutes, the incubation in the substrate for 15 minutes, and the washing step. Therefore, to shorten the experiment time, it is fixed to 10 frames, and the analysis mainly proceeds with the frame (5-7). However, a similar value can be obtained even if the initial frame 1-3 and the second half frame 8-10 of 10 frames are measured.
  • the PPI signal when the PD-L1 is fixed to the substrate, the PPI signal can form a cluster.
  • the laser power is reduced or the gain number is reduced, the fluorescence intensity of the spot is reduced, the single GFP spot is almost invisible, only the cluster signal can be left, only the cluster is analyzed, , Or if you want to analyze the correlation, you can proceed with the step further.
  • the imaged protein complex (PPI complex) was analyzed by the following procedure:
  • the protein complexes were analyzed based on the toolkits provided in the Matlab program (provided by MathWorks).
  • the fluorescence images obtained were stored in a 16-bit unsigned integer format.
  • PD-1-PDL1 analysis the following procedure was performed using a total of three frames of 5-7 frames. As described above, since the washing process is included, the intensity and stability of the signal seen in the imaging are excellent. As a result of analyzing 1-3 frames of 10 frames and 8-10 frames, There was no significant difference in the results of the analysis of all the frames. this The interval may vary depending on the imaging condition / equipment construction status.
  • the initial start begins at the top left corner.
  • One frame consists of 512x512 pixels. 11 pixels to the right based on the base pixel, down
  • the median value was obtained from 11 11x11 individual pixels, and this median value was subtracted from the reference pixel value [(Intensi_pixel) -
  • Threshold is the threshold value of the pixel that the pixel intensities become below the threshold in the whole image. This can remove local maximums not created by the fluorescence signal in the image.
  • the threshold value used in the imaging condition is 70.
  • PSF localized spread function
  • the local maximum is obtained (for example, i-th row, j-th column pixel).
  • PSF can be selected. This can be obtained using the toolkit provided in the mat lab.
  • the process of determining whether the local maximum obtained from the actual PSF is performed is performed.
  • the minimum value of the local maximum is defined (minimum intensities), and only the maximum of the obtained local maximums is used for the analysis.
  • the minimum value is defined as 75, which may vary depending on the laser power / exposure time / equipment construction situation.
  • 5x5 pixel centered on the finally obtained local maximum coordinate
  • the information was retrieved, and the centroid of intensities was obtained for this 5x5 pixel.
  • the brightness center deviates by more than 0.5 pixels from the existing local maximum coordinates (when the 2D symmetry about the PSF shape disappears), it is judged to be a non-normal fluorescence signal and removed from the analysis.
  • the number of PPI complexes was determined for all the files by performing steps 1-3 above. The information of each file taken under the same conditions was collected and the mean and standard deviation were obtained. This value finally represents the number of PPI complexes under certain conditions.
  • the number of the obtained PPI complexes can be obtained by obtaining the count of the PPI signal or by calculating the sum of the intensities of the PPI signals or by measuring the rat io of the spot in the measurement region.
  • the laser power and the gain number are adjusted to reduce the size of the cluster signal, that is, the spot where the intensity of the fluorescence is too low, (The local maximum may be reduced or overlapped spots may be distinguished).
  • cluster intensities we will be able to more closely characterize the drug.
  • drugs that inhibit PD-1 and PD-1 clustering In the case of the drug, the number of clusters will be reduced, although the number of single spot counts may be rather increased or similar, and effectively inhibiting the PD-1-PDL1 interaction in the body with cluster ing. Therefore, it would be meaningful to analyze the distribution and number of c luster.
  • intensities of only single molecules could be derived.
  • cluster rat io When accessing a specific cluster, it can be analyzed as count or sum of signals with cluster intensities above a certain threshold, or cluster rat io can be analyzed. The same is true for single molecule identification.
  • the fluorescence signal a formed at a certain threshold or higher among the signals formed by the m- .
  • the fluorescence signal value may be PPI count, sum, rat io, and the like.
  • the fluorescence signal b value for the fluorescence signal a value By analyzing the fluorescence signal b value for the fluorescence signal a value, more qualitative data can be confirmed than when analyzing only the fluorescence signal b value.
  • the measurement of the fluorescence signal a value can also be taken into account for the influence of PDL1 *, which is injected onto the substrate but not attached to the substrate, at a specific concentration of mcherry-PDL1.
  • the accuracy can be improved by quantifying the interaction of PD-L1 and PD-1 *.
  • the PD-1 mutant containing the PD-1 mutation K78A which inhibits the binding of the suppression-1 and PD-L1, or the PD-L1 mutant D122A +
  • the same test as above was carried out using an egg-1 mutant containing the PD-L1 mutant containing K124A, or A132L, a PD-1 mutation that enhances the binding of egg-1 to PD-L1.
  • FIG. 27B A fluorescence image obtained by using the anti-PD-L1 antibody 9A11 (# CST-29122; Cell Signaling Technology) is shown in FIG. 27B, and the result of quantitation (PPI complex number: F-counts) Respectively. As shown in FIGS.
  • PD-L1 antibody was immobilized on a substrate using PD-L1 antibody in the physiologic conformation of PD-1 / PD-L1. It was predicted that it would be more advantageous to approach the analysis.
  • the antibody used for immobilizing the PD-L1 on the substrate may be appropriately selected so as to expose a specific domain of PD-L1 (binding site with egg-1) have.
  • FIG. 29 shows the number of PPI complexes (F-counts) obtained by fixing the anti-PD-L1 antibody on the substrate and performing the above test.
  • the concentration of PD-L1 was fixed at 5 nM and the PD-1 concentration was tested with 0 nM, 5 nM, -1 antibody, the concentration of PD-1 was fixed at 5 nM and the PD-L1 concentration was tested while increasing to 0 nM, 5 nM, and 15 nM.
  • Neutravidin (Thermo, A2666), an Avidin series protein, was added to the substrate prepared in Example 13 at a concentration of 0.1 mg / ml. After 5 minutes of reaction at room temperature, the substrate was washed twice with PBS buffer 30. The prepared substrate was treated with the anti-PD-L1 antibody 9A11 (CST # 29122) confirmed to have excellent effect in Example 14 and FIG. 29 in the amount of about 2 ug / ml for about 5 minutes, A substrate coated with anti-PD-L1 antibody (9A11) was prepared. The prepared substrate was washed twice with 30 ul of PBS buf fer and used for further testing.
  • Inhibitors inhibit the interaction of PD-L1 / PD-1 * in three ways: 1) when the inhibitor binds to the binding interface of PD-L1 and PD-1 with strong intensity; 2) inhibitors (PD-1 independent ly) bind to specific sites of PD-L1 to induce structural changes of PD-L1; And 3) inhibitors (PD-
  • step 1) a stepwise reaction ion (PD-L1, inhibi tor, PD_1) which is not one-step react ion [ 0 ], the above step 1) can be effectively screened
  • cases 2) and 3 since the coli lion itself of the drug and the target may not be induced, a missed populat ion is generated, so that there is a limitation in testing the correct inhibitory effect.
  • the one-step reaction method in the tube (Standard 1.5 ml ep-tube) is adopted in this embodiment.
  • 28A schematically shows the inhibitor candidate drug screening process of the present embodiment Show.
  • FIG. 28B shows the results of a test for optimizing the reaction conditions of one-step reaction in a tube.
  • the GFP-egg-1 and mcherry-PD-Ll prepared in Example 12 were mixed in a tube for 30 minutes or 2 Incubation time, and fluorescence counts (F-counts).
  • F-counts fluorescence counts
  • 'Negat ive' indicates a reaction product in which mch-PDLl was not added to the tube
  • 'Negat ived means reactants without GFP-PD1 added to the tube, respectively.
  • the number of PPI complexes did not show a significant difference between the incubation time of 30 minutes and the incubation time of 2 hours.
  • the number of PPI complexes obtained using two commercially available PD-1 / PD-L1 inhibitors BMS202 and S7911 (purchased from Sel leck Chem) as candidate agents for egg-1 / PD-L1 interaction inhibition ) Is shown in Fig. In Fig. 28C, it can be seen that the PD-1 / PD-L1 complex is reduced in dependence on the inhibitor treatment concentration. Also, in the case of BMS202, it was confirmed that the IC50 value (18 nM) similar to the IC50 value calculated using the purified protein was shown. The results show that the PD-1 / PD-L1 inhibitor can be accurately determined by the PD-1 / PD-L1 interaction assay performed in this example.
  • the four scales displayed on each drug are lmM, 10 uM (in order from left), 10 uM , 100 nM, and InM.
  • P indicates the result of confirming the normal PPI level by treating DMSO instead of the drug, and shows the result when the mch-PDLl is not coated.
  • the drug A1 used as the reference drug showed no effect at all, while the four selected drugs (Bl, B2, B3, and E2) Dependent inhibition of PD-1 / PD-L1 interaction.
  • B2 showed the best efficacy, which was found to be equal to or better than the efficacy of BMS202, a commercially available drug.
  • FIG. 30A Total GFP-labeled counts and fluorescence images are shown in FIG. 30A (PSF cut of f> 150). As shown in Fig. 30 (a), it can be confirmed that GFP PD-L1 is fixed on the substrate and GFP-suppress-1 is fixed.
  • the cluster rat io (%) of GFP PD-L1 and GFP PD-1 (calculated by [Count @ PSF cut of f> 350 / Count @ PSF cut of f> 150]) is shown in FIG.
  • the cluster rat io% is a value obtained by dividing a cluster population defined as a point spread function (PSF) cutoff of 350 or more by a total count of a cutoff of 150 or more as a percentage.
  • PSF point spread function
  • a PD-L1 mutant construct For epitope mapping of anti-PD-L1 antibodies suitable for immobilizing PD-L1 on a substrate, a PD-L1 mutant construct was designed (see FIG. 31A). Specifically, the PD-L1 cytosol ic domain deletion mutant construct was designed to identify the epi tope of the PD-L1 cytosol ic domain targeting antibody. The mutant was designed as a PD-L1 amino acid sequence (NP_001300958.1) The cytosol ic part corresponding to amino acids 260-290 was divided into 10 units to produce a protein containing a mutation of d280 (amino acid deletion after 281) or d270 (deletion of amino acid deletion after the 2nd) .
  • PD of the PD-L1 ant ibody (9A11) shown to be effective for measuring the PD-L1 / PD-1 interaction see FIG. 29) previously, through the PD-L1 pul 1-down assay, The degree of binding (number of fluorescent complexes due to binding between antibody and PD-L1: F-counts) to the L1-WT (wild type), d280, or d270 mutant was measured and shown in Fig. 31B, In the case of the d280 or d270 variant of PD-L1, it does not bind to the antibody at all, while in the case of the antibody it binds sensitively. It is predicted that the sequences corresponding to amino acids 280-290 of PD-L1 will play an important role in the reaction with antibodies used to immobilize PD-L1 on the substrate in the suppression-L1 / egg-1 interaction assay have.
  • HER3_eGFP recombinant gene in which eGFP is connected to the intracellular domain termini of HER3 (R4 R3: NM_001982.3 BC082992.1, eGFP: MH087225 linker:
  • PEGFP-Nl-FLAG is injected into the HER2-ove rexpressing SKBR3 breast cancer cell line (Korean Cell Line Bank or ATCC) through electroporation to induce overexpression of HER3.
  • 30 ug of HER3_eGFP pi asm ids were transfected in 6X1CT6 SKBR3 cells using electroporat ion (electroporation condition:
  • NRGbl (lOOng / ml) was added to the prepared SKBR3 and incubated for 10 minutes to induce formation of a heterodimer of HER2 protein and HER3_eGFP protein on the cell surface (see Fig. 34A).
  • the level of HER2-HER3_eGFP heterodimer production according to the expression level of HER3-eGFP was determined by a single-molecule Total Internal Reflection Fluorescence Microscope, and the result is shown in FIG. 34B.
  • HER2-HER3 heterodimer was not produced when NRGbl was not treated after introduction of HER3-eGFP plasmid into SKBR3 as shown in FIG. 34B, whereas HER2-HER3 heterodimer was not produced when HER2-HER3 heterodimer was treated with NRGbl And increased in proportion to the expression level.
  • the results show that NRGbl should be injected to generate the HER2-HER3 heterodimer.
  • Fig. 34C Cell lysis was carried out in lysis buffer (1% Digi tonin or 1% GDNO, 10% glycerol, 50 mM HEPES-NaOH [pH 7.4], 150 mM NaCl, 2% protease inhibitor cocktail
  • DGTN digi tonin
  • GDN 0.01%
  • CHAPS0 3- (3-Cho 1am (w / v)
  • OG n-octyl P-D-glucoside
  • DDM n-Dodecyl P-D-maltose ide
  • TX100 0.10% Fluorescence microscope
  • the HER2-HER3 heterodimer thus obtained was attached to the substrate.
  • the surface of the substrate was treated with PEG, neutravidin, biotin, and anti-HER3 antibody (see below) and the cell lysate was injected to the HER2-
  • the slides were coated with a solution of PEG (3 mg biotin-PEG, lOOmg mPEG, dissolved in at lml of PEG buffer), covered with a coverslip and allowed to react with the PEG solution evenly between the slide and coverslip for 2 hours.
  • PEG 3 mg biotin-PEG, lOOmg mPEG, dissolved in at lml of PEG buffer
  • the coverslip and slide 18 Washed with distilled water and dried with compressed nitrogen gas.
  • the completed coverslip and slide were vacuum packed in a 50 ml tube (-20 ° C) to prevent contact with the treated surface of the PEG solution.
  • the Coverslip and the slide PEG solution were kept free of any contact with the treated surface.
  • the frozen coverslips and slides were dissolved in a 37 ° C incubator to prevent water droplets from forming.
  • a reaction chamber was made between the coverslip and the slide using a double-sided tape and epoxy, with the PEG-treated coverslip and the sides of the slide facing each other.
  • the neutravidin solution (lOOug / ml neutravidin,
  • the neutravidin solution remaining in the chamber was washed with PBS and incubated with biotin conjugated anti-HER2 antibody (eBioscience, Cat. No. BMS120BT, clone 2G11) or anti-HER3 antibody (R & D systems, Cat. No. BAM348, clone: # 66201 5 ⁇ g / ml, PBS or 0.1% Tr iton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) was added and reacted for 5 minutes.
  • biotin conjugated anti-HER2 antibody eBioscience, Cat. No. BMS120BT, clone 2G11
  • anti-HER3 antibody R & D systems, Cat. No. BAM348, clone: # 66201 5 ⁇ g / ml, PBS or 0.1% Tr iton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl
  • the antibody remaining in the chamber was washed with PBS, and the above cell lysate was injected, followed by reaction for 15 minutes.
  • the lysates remaining in the chamber were washed with washing buffer (0.1% digitonin, or 0.01% GDN, 50 mM HEPES [pH 7.4], 150 mM NaCl, 1% glycerol).
  • HER2-eGFP recombinant gene (HER2: NMJ304448, linker PTFLYKWDPVPVAT or GGGSGGGT, eGFP: MH087225, vector: pEGFP-Nl addgene pEGFP-Nl-FLAG) in which eGFP is connected to the intracellular domain termini of HER2, was injected into HEK293T cell line (Korean Cell Line Bank, or ATCC) through electroporation to induce HER2 overexpression and induce the formation of HER2-HER2 homodimer (also referred to as HER2 homodimer) on the cell surface ).
  • HEK293T cell line Kerean Cell Line Bank, or ATCC
  • the prepared HER2 homodimer-forming cells are lysed and then treated with a HER2 homodimer.
  • the attached HER2 homodimer is washed with detergent.
  • the phosphorylation level (degree of activation) of HER2 Y1196 obtained as a result of washing the attached HER2 homodimer using various detergents at various concentrations is shown in Fig. 35i.
  • the amount of HER2 adhered using more than 0.05% (w / v) or 0.10% (w / v) of DGTN (digitonin) or 0.01% When the homodimer was washed, the degree of HER2 activation was high.
  • the cell lysate was injected to adhere the HER2 homodimer to the substrate surface, and the DGTN (digitonin 0.1%).
  • a decrease in fluorescence intensity in one step corresponds to a monomer
  • a decrease in fluorescence signal in a two-step corresponds to a dimer.
  • HER2_eGFP has a high 2-step ratio due to a high ratio of eGFP and HER3_eGFP, which are likely to be present as relatively monomers, thus demonstrating the production of the HER2-eGFP homodimer.
  • the most probable step (recursive calculation convergence step) from raw data was determined and analyzed.
  • ATP and 3 ⁇ 4 2+ were added to the reaction chamber of the immunoprecipitated HER2-HER3 heterodimer (HER2-HER3 heterodimer bound to the substrate treated antibody in Example 18) and incubated for 5 minutes to induce Tyr phosphorylation. Afterwards, changes in HER3 pTyr levels were observed by single-molecule immunolabeling with pTyr-specific antibodies.
  • the test procedure is schematically shown in FIG. 35A (1. cell lysis, 2. dimer attachment to the substrate, 3. dimer washing attached, 4. phosphorylation by treating ATP and Mg2 +, 5. phosphorylation of HER3 or HER2 Antibody treatment, 6. fluorescence signal detection), the specific test method is as follows:
  • HER2-HER3 heterodimer was pulled down from the cell lysate (1 to 4 mg / ml) onto the surface of the sample using the biotin-conjugated HER3 antibody in Example 2 (2)
  • the remaining cell lysates were washed with DGTN or 0.01% GDN, 50 mM HEPES [pH 7.4], 150 mM NaCl, 1% glycerol and the kinase assay solution (0.1% DGTN or 0.01 ⁇ »GDN, 200 ⁇ M ATP, 10 mM magnesium chloride, 50 mM HEPES [pH 7.4], 150 mM NaCl, 1% glycerol) and reacted for 5 minutes.
  • kinase assay solution was washed with PBS (or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl), and the HER3 pTyr antibody solution (HER3 pTyr antibody (monoclonal rabbit host) Cat. No. # 479 IS, clone: 21D3) to 2 ug / ml, PBS or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) and reacted for 5 minutes.
  • PBS or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl
  • the HER3 pTyr levels thus obtained are shown in FIG. 35B.
  • the Tyr residues of the HER3 tail are nine, and in this example a total of five Tyr residues of Y1197, Y1222, Y1276, Y1289, and Y1328 were tested.
  • the degree of phosphorylation was increased by culturing both ATP and Mg2 + in pTyr residues.
  • HER2 homodimer was pulled down from the cell lysate to the surface using a biotin-conjugated HER2 antibody in the preparation and use of Example 18 (2), and then 0.1% DGTN or 0.01% GDN 50 mM HEPES [pH (0.1% DGTN or 0.01% GDN, 200 uM ATP, lOmM magnesium chloride, 50 mM HEPES [pH 7.4], 150 mM NaCl, 1% glycerol) was used to wash out the remaining cell lysate using a buffer solution glycerol) and reacted for 5 minutes.
  • kinase assay solution was washed with PBS (or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl), and the HER2 pTyr antibody solution (HER2 (PBS) or 0.1% Tr iton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) in the presence or absence of pTyr antibody (monoclonal rabbit host (Cell Signaling Technology, Cat. No. 6942S, clone: D66B7) Treated and reacted for 5 minutes.
  • PBS or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl
  • HER2 pTyr antibody solution was washed with PBS (or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) and incubated with a cy3 conjugated anti-rabbit FC antibody solution Treated with 0.1% Triton-X-100, 50 mM HEPEStpH 7.4, 150 mM NaCl) and reacted for 5 minutes.
  • HER2 pTyr level level is shown in Fig. 35H.
  • phosphorylation was increased by ATP and Mg2 + cultivation in almost all of the Tyr residues Y1139, Y1196, Y1121 / 1222 and Y1248 of HER2 tail (degree of phosphorylation increase: Y1139, control (ATP + EDTA ), About 13 times in the case of Y1196, about 10 times in the case of Y1121 / 1222 and about 13 times in the case of Y1248 in the case of Y1121 / 1222).
  • FIG. 36A schematically shows that the HER2-HER3 heterodimer attached to the substrate induces an interaction with the eGFP-labeled lower signal transduction protein when it undergoes a phosphorylation process.
  • the degree of signal transduction of the HER2-HER3 heterodimer or HER2 homodimer was measured.
  • Figure 36b shows how much of the HER2-HER3 heterodimer interacts with PLC signaling protein (PLC gamma 1) in terms of the size of the fluorescence signal.
  • Figure 36c shows the number of single molecular off events obtained from the fluorescence signal to determine how much of the HER2-HER3 heterodimer interacts with PLC signaling protein (PLC gamma 1) (black: ATP + EDTA, white: ATP + MgCh).
  • Figure 36e shows the number of single molecular off events from the fluorescence signal as to how much of the HER2-HER3 heterodimer interacts with the amount of the lower signaling protein (p85 alpha) (black: ATP + EDTA, whi te : ATP + MgCh).
  • FIG. 36f shows how much of the HER2-HER3 heterodimer interacts with the amount of the lower signaling protein (PI3K: p85 alpha -pllO alpha complex) in terms of the magnitude of the fluorescence signal.
  • FIG. 36g schematically shows that the HER2 homodimer attached to the substrate induces an interaction with the eGFP-labeled lower signal transduction protein when oxidized.
  • Figure 36j shows how much of the HER2 homodimer interacts with the amount of the lower signaling protein (p85 alpha) in terms of the size of the fluorescence signal.
  • Figure 37A is a measure of the enzymatic performance of the HER2-HER3 heterodimer (the HER2-HER3 heterodimer at a specific concentration of phosphorus used for phosphorylation 2019/132517 1 »(: 1 ⁇ ⁇ 2018/016675
  • the substrate is attached to the substrate at the inhibitor 18
  • Activation of the HER2-HER3 heterodimer was measured using HER3 pY1289 antibody, which was normalized to 100% in the absence of Lapat inib.
  • Activation of the HER2 homodimer was measured using HER2 pY1196 antibody. 100% And the standardized value. 37D,
  • Lapat inib has been shown to exhibit similar inhibition curves to HER2-HOM3 and HER2-HER3 heterodimers that are not resistant to Lapat inib.
  • HER2-HER3 heterodimer expressing cell lysates were incubated with Lapat inib, ATP (lOOuM), and magnesium chlor ide (10 mM) (again, DGTN was added to the HER2-HER3 heterodimer expressing cell lysate to confirm the correlation between phosphorylation rate and inhibitor resistance
  • PTPN1 Protein Tyrosine Phosphatase, Non-receptor type 1; ProSpecbio, Cat. PKA-219
  • PTPN1 is a dephosphorylation enzyme that forms an equilibrium between Tyr phosphorylation and dephosphorylation and effectively slows down the rate of substantial phosphorylation without changing the HER2-HER3 heterodimer into its structure.
  • FIG. 37F As shown in FIG. 37F, as the treatment concentration of PTPN1 increases (from the red graph (rightmost) to the purple graph (leftmost) in FIG. 37F) Phosphorylation is slowed down and the inhibition of phosphorylation by lapatinib is increased (ie, phosphorylation is inhibited at lower concentrations of lapatinib).
  • Example 25 Single-molecule immunolabelin and co-IP profiling for xenograft models and human patient samples.
  • mice The severed tissue was re-implanted in another group of mice to obtain subsequent tumors.
  • Mouse generations with patient-derived tumors were named F0, and subsequent generations were numbered sequentially (FI, F2, F3, etc.).
  • F3 mouse was used in the vehicle (Phosphata buffered saline, PBS), osimert inib, or gefitinib treatment test (PDTX-A1).
  • Patient PI was assessed for partial response (PR) to gefitinib therapy for about a year and a half before diagnosis of progressive disease (PD). Partial response to the response evaluation criteria in solid tumors (RECIST) While Patient P2 maintained a stable disease (SD) diagnosis for one year before PD designation.
  • PR partial response
  • PD progressive disease
  • Example 26 Effect of EGFR mutation on interaction with Grb2
  • gefitinib a TKI specific for EGFR
  • EGFR tyrosine kinase inhibition by gefitinib almost completely inhibited Grb2 binding to three cells with exon 19 deletion (no gatekeeper mutations) (Fig. 39A).
  • Some residual EGFR-Grb2 PPI counts were observed in H1975 cells with L858R and T790M gatekeeper mutations. It was also confirmed to completely inhibit Grb2 binding in H1975 cells by treating osimert inib (third generation TKI that equally well binds EGFR with gatekeeper mutations) (Fig. 39a, blue dot).
  • mutant EGFR utilizes fundamentally different mechanisms that are least dependent on surface exposed pTyr residues.
  • mutant EGFRs form a larger signaling complex than WT EGFR.
  • Some components of these signaling complexes engage GrB2 in a pTyr-independent manner. This non-canonical pathway is highly resistant to dephosphorylation of phosphatase
  • HSP90a is remarkably abundant with EGFR. This suggests that the cells depend on EGFR signaling as well as accessory proteins called HSP90a. Therefore, it is expected that ESPR inhibitor and HSP90a may be used at the same time, or the desired effect may be obtained to some extent even when the HSP90a inhibitor alone is used. On the other hand, it can be confirmed that EGFR does not work at all in the normal H1666 + cell line. 2019/132517 1 »(: 1/10/06 018/016675

Abstract

The present invention relates to a method for analyzing an activation state of a signaling pathway in a cell or tissue and/or between cells or tissues through protein-protein interaction analysis, and medicine selection using the same, and/or a method for monitoring efficacy of a medicine or a method for drug candidate screening, and an apparatus for use therein.

Description

【명세서】  【Specification】
【발명의 명칭】  Title of the Invention
세포내또는세포간단백질-단백질상호작용분석 방법 및장치 【기술분야】  Method and apparatus for analyzing intracellular or intercellular protein-protein interactions
단백질-단백질 상호작용 분석을 통하여 세포 또는조직 내 및/또는 세포 또는 조직 간의 신호전달경로 (signal ing pathway)의 활성화 상태를 분석하는 방법, 이를 이용한 치료제 선정 및/또는 치료제 효능 모니터링 방법, 후보약물스크리닝 방법 및 이에 사용하기 위한장치에 관한것이다. 【배경기술】  A method for analyzing the activation state of a signaling pathway between a cell or tissue and / or a cell or tissue through analysis of protein-protein interaction, a method for selecting a therapeutic agent and / or a method for monitoring the efficacy of a therapeutic agent, And a device for use therein. BACKGROUND ART [0002]
최근 질병 진단, 예후 예측 및 치료는 유전자 분석 (genomi c prof i l ing)에 집중되고 있다. 하지만, 암을비롯한특정 질병의 발병원인은 신체를 구성하는 세포의 비정상적인 활동, 더욱 자세하게는 세포를 구성하고 조절하는 다양한 단백질간의 비정상적인 상호작용에 의한 것이으로, 유전자 분석을 통한 개별적 단백질 관찰만으로는 이해하기 어려운 문제가 있다. 실제로 이러한유전자돌연변이의 프로파일링을 통해 동일한 유전적 성질을 지닌 환자들에 대해서도 표적 항암제에 대한 감수성이 다르고, 예후 역시 다양하게 나타난다. 따라서, 개별적 단백질 분석이 아닌 단백질-단백질 간의 상호작용의 정확한 분석은 세포 내 및/또는 세포 간 신호전달 네트워크 (signal ing network)가 어떻게 변화하는 지를 이해할수 있도록 할뿐 아니라, 암과 같은 질병의 진행과 특성 및 그 치료제 선정 및 치료방법에 대한 정보까지도 제공할 수 있을 것으로기대된다.  Recently, disease diagnosis, prognosis prediction and treatment have been focused on gene analysis (genomics profiling). However, the cause of a specific disease, including cancer, is due to the abnormal activity of the cells constituting the body, more specifically, the abnormal interaction between various proteins that constitute and regulate the cell. There is a difficult problem. Indeed, profiling of these gene mutations also leads to different susceptibility to target chemotherapy and to prognosis for patients with the same genetic characteristics. Thus, an accurate analysis of protein-protein interactions, rather than individual protein analyzes, will help to understand how the intracellular and / or intercellular signaling networks change, And information on the characteristics and methods of selecting and treating the therapeutic agents.
【발명의 상세한설명】  DETAILED DESCRIPTION OF THE INVENTION
【기술적 과제】  [Technical Problem]
일 예는 제 1 단백질과 제 2 단백질 간 단백질-단백질 상호작용을 측정하는단계를포함하고,  One example includes measuring protein-protein interactions between a first protein and a second protein,
상기 제 1단백질은세포또는조직 내 및/또는세포또는조직 간의 신호전달경로 (signal ing pathway)에 관여하는 단백질이고, 제 2 단백질은 상기 세포또는조직 내 및/또는세포또는조직 간의 신호전달경로중에서 상기 제 1 단백질과 상호작용하는 단백질 (예컨대, 제 1 단백질의 하위 단백질, 제 1 단백질의 리간드, 제 1 단백질과 동일세포, 동종세포 또는 이종세포 立면에 위치하거나 이종세포에서 분비되고 제 1 단백질과 상호작용하는단백질등)인, 2019/132517 1»(:1^1{2018/016675 Wherein the first protein is a protein involved in a signaling pathway between cells or tissues and / or between cells or tissues, and the second protein is a protein involved in the signal transduction pathway in the cell or tissue and / A protein that interacts with the first protein (e.g., a sub protein of the first protein, a ligand of the first protein, the same cell as the first protein, an allogeneic cell or a heterologous cell, ≪ RTI ID = 0.0 > and / or < 2019/132517 1 »(: 1 ^ {2018/016675
세포 또는 조직 내 및/또는 세포 또는 조직 간의 신호전달경로의 활성화측정 (또는확인또는결정 또는분석) 방법 ; 상기 세포또는조직, 또는상기 세포또는조직이 유래하는 개체의 상기 제 1 단백질을표적으로 하는 약물에 대한 반응성을 예즉하는 방법 또는 예즉에 정보를 제공하는 방법 ; 및상기 제 1단백질을표적으로하는치료에 적합한개체를선별하는 방법 또는선별에 정보를제공하는방법을제공한다. Measuring (or identifying or determining or determining) the signal transduction pathway in a cell or tissue and / or between a cell or tissue; A method for predicting the reactivity of said cell or tissue, or an individual from which said cell or tissue is derived, to a drug targeting said first protein, And a method for screening a subject suitable for treatment targeting the first protein or a method for providing information to screening.
상기 제 1 단백질이 2종 이상 사용되는 경우, 상기 제 1 단백질을 표적으로하는 약물은상기 2종 이상의 제 1단백질 중어느하나또는 2종 이상을 표적으로 하는 약물일 수 있고, 상기 제 1 단백질을 표적으로 하는 치료는 상기 2종 이상의 제 1 단백질 충 어느 하나 또는 2종 이상을 표적으로하는치료일 수 있다. 상기 치료는 제 1단백질을표적으로하는 약물을처방및/또는투여하는것을포함할수 있다. 상기 개체는상기 제 1 단백질이 관여하는신호전달또는제 1단빡잘및 제 2단백질 간상호작용과 관련된 질병 (예컨대, 암)에 걸리거나 걸릴 위험이 있는 개체 (즉, 상기 제 1 단백질이 관여하는 신호전달 또는 제 1 단백질 및 제 2 단백질 간 상호작용과 관련된 질병의 예방 및/또는 치료를 필요로 하는 개체)일 수 있다. 상기 제 1 단백질을 표적으로 하는 치료에 적합한 개체는 상기 제 1 단백질을 표적으로 하는 약물이 소망하는 효과(예컨대, 상기 제 1 단백질이 관여하는 신호전달 또는 제 1 단백질 및 제 2 단백질 간상호작용과 관련된 질병의 예방및/또는치료및/또는경감효과(상기 질병이 암인 경우항암 효과)등)를발휘할수있는개체를의미할수있다.  When two or more of the first proteins are used, the drug targeting the first protein may be a drug targeting at least one of the two or more first proteins, The target treatment may be a treatment targeting one or more of the two or more kinds of the first protein. The treatment may include prescribing and / or administering a drug targeting the first protein. (E.g., cancer) associated with the first protein-mediated signal transduction or interactions between the first protein and the second protein (i. E., An individual that is at risk of contracting An individual requiring the prevention and / or treatment of diseases associated with signal transduction or interactions between the first protein and the second protein). An individual suitable for treatment targeting the first protein is one in which the drug targeting the first protein has a desired effect (e. G., Signal transduction mediated by the first protein or interaction between the first protein and the second protein, Or prevention and / or treatment and / or relief of related diseases (anticancer effect when the disease is cancer), and the like.
다른 예는 상기 단백질-단백질 상호작용을 측정하는 단계를 2개 이상의 제 1 단백질에 대하여 수행하는, 상기 세포 또는 조직, 또는 상기 세포 또는 조직이 유래하는 개체에 적용하기에 적합한 표적으로서의 제 1 단백질또는이를표적으로하는 약물을선별하는방법 또는선별에 정보를 제공하는방법을제공한다.  Another example is a first protein as a target suitable for application to the cell or tissue, or to an individual from which the cell or tissue is derived, wherein the step of measuring the protein-protein interaction is performed on two or more first proteins, A method for screening a drug that targets the drug, or a method for providing information to screening.
다른 예는, 제 1 단백질을 표적으로 하는 약물이 처리된 세포 또는 조직, 또는 상기 약물을 투여한 개체로부터 분리된 세포 또는 조직 내, 및/또는 세포 또는 조직 간의 제 1 단백질과 제 2 단백질 간 단백질-단백질 상호작용을 측정하는 단계를 포함하는, 상기 세포 또는 조직, 또는 상기 개체의 상기 제 1단백질을표적으로 하는 약물 및/또는상기 제 1단백질과 저 12단백질이 관여하는신호전달또는상기 제 1 단백질과제 2 단백질 간의 상호작용과관련된 질병의 치료제에 대한반응성을모니터링하는방법 또는 2019/132517 1»(:1^1{2018/016675 Another example is a cell or tissue treated with a drug that targets the first protein or a cell or tissue isolated from a subject to which the drug is administered and / - measuring the protein or protein interaction, or a drug targeting said first protein of said subject and / or a signal transduction or first signal transduction involving said first protein and low 12 protein, A method for monitoring the response to a therapeutic agent of a disease associated with protein task 2 protein interactions or 2019/132517 1 »(: 1 ^ {2018/016675
모니터링에 정보를 제공하는 방법을 제공한다. 다른 예는, 제 1 단백질을 표적으로 하는 후보 약물이 처리된 분리된 세포 또는 조직, 또는 상기 약물을 투여한 개체로부터 분리된 세포 또는 조직 내, 및/또는 세포 또는 조직 간의 제 1단백질과제 2단백질 간단백질-단백질상호작용을측정하는 단계를포함하는, 상기 제 1단백질을표적으로하는약물및/또는상기 제 1 단백질과 제 2 단백질이 관여하는 신호전달 또는 상기 제 1 단백질과 제 2 단백질 간의 상호작용과관련된 질병의 치료제의 스크리닝 방법을제공한다. 일구체예에서, 다음의 단계를포함하는, 세포또는조직 내 및/또는 세포또는조직 간의 신호전달경로의 활성화측정 방법이 제공된다: Provides a way to provide information to monitoring. In another example, a candidate protein targeted to a first protein is isolated from a separate cell or tissue that has been treated, or within a cell or tissue separated from the subject to which the drug is administered, and / or between cells or tissue, Wherein the first protein-mediated drug and / or the first protein-second protein-mediated signal transduction involving the step of measuring liver protein-protein interactions or the interaction between the first protein and the second protein A method for screening a therapeutic agent for a disease associated with the action. In one embodiment, there is provided a method of measuring activation of a signal transduction pathway in a cell or tissue and / or between a cell or tissue, comprising the steps of:
(1) 제 1 단백질을포함하는 시험 시료를 표면에 상기 제 1 단백질에 특이적으로 결합하는 물질을포함하는 기판에 가하여 제 1 단백질이 고정된 기판을준비하는단계;  (1) preparing a substrate to which a first protein is immobilized by adding a test sample containing a first protein to a substrate including a substance that specifically binds to the first protein on a surface thereof;
(2) 상기 준비된 제 1단백질이 고정된 기판에 표지 물질이 결합된 저 12단백질을첨가하여 반응시키는단계 ; 및  (2) adding a low-level protein to which a labeling substance is bound to a substrate on which the prepared first protein is immobilized; And
(3)단계 (2)에서 얻어진반응물로부터 신호를즉정하는단계.  (3) immediately determining a signal from the reactant obtained in step (2).
상기 활성화측정 방법은단계 (1)내지 (3)에 더하여,  In addition to steps (1) to (3) above,
(4)단계 (3)에서 측정된신호를이용하여 단계 (1)에서 첨가한시험 시료내의 제 1단백질의 단위량에 대한신호값을구하는단계, 또는  (4) obtaining a signal value of the unit amount of the first protein in the test sample added in step (1) using the signal measured in step (3), or
(4-1) 단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한 시험 시료의 단위량에 대한신호값을구하는단계 ; 및  (4-1) obtaining a signal value of a unit amount of the test sample added in step (1) using the signal measured in step (3); And
(4-2) 상기 단계 (4-1)에서 얻어진 시험 시료의 단위량에 대한 신호값을 이용하여 시험 시료에 포함된 제 1 단백질의 단위량에 대한 값을 구하는단계  (4-2) obtaining a value for the unit amount of the first protein contained in the test sample using the signal value of the unit amount of the test sample obtained in the step (4-1)
를추가로포함할수있다.  May be further included.
상기  remind
단계 (4) 또는 (4-2)에서 얻어진 시험 시료에 포함된 제 1 단백질의 단위량에 대한 값은 각각의 제 1 단백질과 각각의 제 2 단백질에 대하여 얻어진값을모두합한값일수있다.  The value for the unit amount of the first protein contained in the test sample obtained in the step (4) or (4-2) may be a value obtained by summing the values obtained for the respective first protein and the respective second proteins.
상기 단계 (4)또는 (4-2) 이후에,  After step (4) or (4-2) above,
(5) 단계 (4) 또는 (4-2)에서 얻어진 결과를 기준 시료에서 얻어진 결과와비교하는단계를추가로포함하고,  (5) comparing the result obtained in the step (4) or (4-2) with the result obtained in the reference sample,
상기 기준시료는정상세포, 제 1단백질이 관여하는신호전달경로의 활성화 정도가 확인된 세포, 또는 제 1 단백질이 관여하는 신호전달경로의 2019/132517 1»(:1^1{2018/016675 The reference sample may be a normal cell, a cell in which the degree of activation of the signal transduction pathway involved in the first protein is confirmed, or a signal transduction pathway in which the first protein is involved 2019/132517 1 »(: 1 ^ {2018/016675
활성화정도가확인된개체로부터 분리된세포를포함하는것일수있다. 다른구체예는, 상기한단계 (1), (2), 및 (3), 또는 (1) , (2) , (3) , 및 (4) , 또는 (1), (2), (3) , (4-1) 및 (4-2)를포함하고, 여기에 추가하여,The degree of activation may be that of cells isolated from the identified individual. Another embodiment is a method of manufacturing a semiconductor device comprising steps (1), (2) and (3), or (1), (2), (3), and (4) , (4-1) and (4-2), and in addition thereto,
(5)상기 단계에서 얻어진결과를비교하는단계를포함하고, 상기 제 1단백질은세포또는조직 내 및/또는세포또는조직 간의 신호전달경로에 관여하는 단백질 중에서 선택되는 2종 이상이고, 상기 제 2 단백질은상기 제 단백질과상호작용하는단백질이며, (5) comparing the results obtained in the above step, wherein the first protein is at least two selected from among proteins involved in a cell or tissue and / or a signal transduction pathway between cells or tissues, A protein is a protein that interacts with the aforementioned protein,
단계 (5)는 상기 2종 이상의 제 1 단백질에 대하여 얻어진 결과를 서로비교하는것인,  Step (5) comprises comparing the results obtained for the two or more first proteins with each other,
개체에 적용하기에 적합한 표적 약물의 선별에 정보를 제공하는 방법을제공한다.  Provides a method of providing information to the screening of a target drug suitable for application to an individual.
상기 방법에 있어서, 상기 시험 시료는 포유류 개체로부터 분리된 세포, 조직, 세포또는조직의 용해물, 파쇄물, 또는추출물, 또는체액일 수 있으며, 예컨대, 암세포, 암조직, 암세포또는암조직의 용해물, 파쇄물, 또는추출물일수있다.  In the above method, the test sample may be a lysate, a lysate, an extract, or a body fluid of a cell, a tissue, a cell or a tissue isolated from a mammalian body, and the lysate of the cancer cell, the cancer tissue, the cancer cell, , Lysates, or extracts.
상기 방법에 있어서, 상기 표지 물질은효소반응, 형광, 발광, 또는 방사선 검출을 통하여 측정 가능한 신호를 발생시키는 소분자 화합물, 단백질, 펩타이드, 및 핵산분자로 이루어진 군에서 선택된 1종 이상일 수 있다.  In the above method, the labeling substance may be at least one selected from the group consisting of small molecule compounds, proteins, peptides, and nucleic acid molecules that generate signals measurable through enzyme reaction, fluorescence, luminescence, or radiation detection.
상기 방법에 있어서, 단계 (2)의 표지 물질은 형광을 발생시키는 소분자 화합물, 단백질, 펩타이드, 및 핵산분자로 이루어진 군에서 선택된 1종 이상이고, 단계 (3)의 신호를 측정하는 단계는 전반사 형광 현미경 또는형광카메라또는이들모두를사용하여 수행되는것일수있다.  In this method, the labeling substance of step (2) is at least one selected from the group consisting of small molecule compounds, proteins, peptides, and nucleic acid molecules that generate fluorescence, and the step of measuring the signal of step (3) A microscope or a fluorescence camera, or both.
상기 방법에 있어서, 상기 형광카메라의 1프레임 당노출시간이 약 0.0()1초내지 약 1초일수있다.  In the above method, the exposure time per frame of the fluorescent camera may be about 0.0 () 1 second to about 1 second.
다른구체예는,  In another embodiment,
(I)시험 시료에 후보화합물을처리하는단계, 및  (I) treating the candidate compound with the test sample, and
(I I) 상기 후보 화합물이 처리된 시험 시료 및 처리되지 않은 시험 시료 각각에 대하여 상기 단백질-단백질 상호작용, 상호작용 수준, 활성화 수준 측정을 수행한 후 상호 비교하여 제 1 단백질을 표적으로 하는 후보 약물의 선별방법을제공한다:  (II) the protein-protein interaction, the interaction level, and the activation level are measured for each of the test sample treated with the candidate compound and the untreated test sample, and then the candidate compound Lt; RTI ID = 0.0 > of:
상기 비교 결과, 후보 화합물이 처리된 시험 시료의 단백질-단백질 상호작용, 단백질-단백질 상호작용 수준, 또는 활성화 수준이 후보 화합물이 처리되지 않은시험 시료에서보다낮은경우, 상기 후보화합물을 상기 제 1 단백질을 표적으로 하는 후보 약물로 선택하는 단계를 추가로 포함할수있다. As a result of the comparison, the protein-protein interaction, the protein-protein interaction level, or the activation level of the test sample to which the candidate compound has been treated, If the compound is lower than in the untreated test sample, the candidate compound may be further selected as a candidate drug targeting the first protein.
다른구체예는,  In another embodiment,
제 1 단백질에 특이적으로 결합하는 제 1 단백질의 포획용 물질을 포함하는멀티 웰, 및  A multi-well comprising a substance for capturing a first protein that specifically binds to a first protein, and
임의의 신호검출수단  Any signal detection means
을포함하고,  / RTI >
상기 멀티 웰은 일면이 개방된 복수의 관을 포함하거나, 지지 플레이트에 이격 형성된복수의 비관통형 홀을포함하는것으로,  Wherein the multiwell includes a plurality of tubes whose one surface is open or a plurality of non-through holes spaced apart from the support plate,
상기 관또는비관통형 홀의 개방된 일면에 위치하는시료주입부, 상기 관 또는 비관통형 홀의 내부의 단백질-단백질 상호작용이 일어나는반응부, 및  A sample injecting portion located on one open side of the tube or non-through hole, a reaction portion where protein-protein interaction occurs in the tube or non-through hole, and
상기 관또는비관통형 홀의 내부와접촉하는적어도 일부의 내벽의 표면에 제 1 단백질의 포획용 물질이 고정화되거나 고정화 가능한 제 1 단백질의 포획부를포함하며,  A capturing portion of a first protein capable of immobilizing or immobilizing a substance capable of capturing a first protein on a surface of at least a part of the inner wall in contact with the inside of the tube or the non-penetrating hole,
상기 제 1 단백질의 포획부의 표면은 알데히드기, 카르복실기 및 아민기로이루어진 군에서 선택된 작용기를포함하는화합물로표면 처리된 것인,  Wherein the surface of the capturing portion of the first protein is surface-treated with a compound containing a functional group selected from the group consisting of an aldehyde group, a carboxyl group and an amine group.
세포 또는 조직 내 및/또는 세포 또는 조직 간 단백질-단백질 상호작용분석 장치를제공한다.  To provide an apparatus for analyzing protein-protein interactions within cells or tissues and / or between cells or tissues.
상기 분석 장치에 있어서,  In the analyzer,
상기 표면 처리는, 바이오틴 (biot in) , 바이오틴이 결합된 소혈청알부민 (Bovine serum albumin) , 폴리에틸렌글리콜 (PEG, polyethylene glycol ) , 폴리에틸렌글리콜-바이오틴 (PEG-biot in) , 또는 폴리소르베이트를사용하는것일수있다.  The surface treatment may be performed using biotin, biotin-conjugated bovine serum albumin, polyethylene glycol (PEG), polyethylene glycol-biotin, or polysorbate It can be done.
상기 표면 처리는 뉴트라비딘 (neutravidin) , 스트렙타비딘 (streptavidin) , 및 아비딘 (avidin)으로 이루어진 군에서 선택된 1종 이상을 추가로 도포하는것일수있다. The surface treatment may further include at least one selected from the group consisting of neutravidin, streptavidin, and avidin.
상기 멀티월은검출가능한신호를발생시키는표지 물질로표지된, 신호전달 경로 상에서 상기 제 1 단백질과 상호작용 하는 1종 이상의 제 2 단백질을추가로포함하는것일수있다.  The multiwall may further comprise at least one second protein that interacts with the first protein on the signal transduction pathway, labeled with a labeling substance that generates a detectable signal.
상기 멀티 웰은, 제 1 단백질과 제 2 단백질 간상호작용이 일어나는 반응부 (제 1 반응부)를 포함하는 하나 이상의 웰 및 제 1 단백질과 저 U 단백질에 결합하는탐지용물질이 결합하는반응부 (제 2반응부)를포함하는 하나이상의 웰을포함하는것일수있다. The multi-well may be a well-known method in which interaction occurs between the first protein and the second protein One or more wells including a reaction part (first reaction part) and a reaction part (second reaction part) in which a first protein and a detection substance binding to the low U protein bind to each other .
상기 방법 및 장치에 있어서, 상기 세포또는조직 내 및/또는세포 또는 조직 간의 신호전달경로의 활성화 측정 방법에 있어서, 상기 제 1 단백질과 제 2 단백질은 각각 독립적으로 세포 또는 조직 내 및/또는 세포 또는 조직 간의 신호전달경로에 관여하는 단백질 중에서 선택되는 1종 이상이고, 상기 제 2 단백질은 신호전달경로 상에서 제 1 단백질과 상호작용하는 단백질 (예컨대, 제 1 단백질보다 하위 경로에 관여하는 단백질, 제 1 단백질의 리간드, 제 1 단백질과 동일세포, 동종세포 또는 이종세포 표면에 위치하거나 이종세포에서 분비되고 제 1 단백질과 상호작용하는 단백질 등) 일 수있다. 상기 신호전탈은 세포 내부에서의 신호전달, 세포외부에서 세포 내부로의 신호전달, 또는동종또는 이종의 2이상의 세포간의 신호전달 (예컨대, 면역체크포인트간의 면역신호전달 등)을 총괄하여 의미하는 것일 수 있다. 상기 제 1 단백질은 세포막 단백질일 수 있다. 상기 제 1 단백질은수용체 티로신 카이네이즈, 톨-유사 수용체, G-단백질 연결 수용체 (G-protein-coupled receptors; GPCR), 트랜스페린 수용체 (transferr in receptors) , 저밀도지질단백질 (Low_ Dens ity Lipoprotein; LDL) 수용체, R0S1; BCR-Abl l 융합 단백질; 비수용체형 카이네이즈; GTP가수분해효소 (GTPases); 호르몬 수용체; 항- 아팝토시스 단백질; 및 면역 체크포인트 단백질 등으로 이루어진 군에서 선택된 1종이상일수있다.  In the above method and apparatus, it is preferable that, in the method for measuring activation of a signal transduction pathway in the cell or tissue and / or between cells or tissues, the first protein and the second protein are independently in cells or tissues and / And the second protein is a protein that interacts with the first protein in the signal transduction pathway (for example, a protein involved in a lower pathway than the first protein, A ligand of a protein, a protein which is located on the same cell as the first protein, on the surface of allogeneic cells or xenogeneic cells, or secreted in a different cell and interacts with the first protein, etc.). The signal transfer may be understood to mean a signal transfer within the cell, a signal transfer from the outside of the cell to the inside of the cell, or a signal transfer between two or more cells of the same or different type (for example, immune signal transmission between immune checkpoints) . The first protein may be a plasma membrane protein. The first protein may be selected from the group consisting of receptor tyrosine kinase, toll-like receptors, G-protein coupled receptors (GPCR), transferrin receptors, low density lipoprotein , R0S1; BCR-Abl l fusion protein; Non-drinking body type kaineise; GTPases (GTPases); hormone receptors; Anti-apoptotic protein; And immune checkpoint proteins, and the like.
일 예에서, 제 1 단백질로서 PD-L1 및 卵 -1 중 어느 하나, 제 2 단백질로서 다른하나를선택하여 상기 방법 및장치에 적용할수있다. 다른 예에서, 제 1 단백질로서 HER2와 HER3의 헤테로다이머를 선택, 제 2 단백질로서 세포 또는 조직 내 및/또는 세포 또는 조직 간의 신호전달경로 중에서 HER2 및/또는 HER3의 하위 단백질을 선택하여 상기 방법 및 장치에 적용할수있다.  In one example, one of PD-L1 and egg-1 as the first protein, and the other as the second protein may be selected and applied to the method and apparatus. In another example, a heterodimer of HER2 and HER3 is selected as the first protein and a sub-protein of HER2 and / or HER3 is selected as the second protein in the cell or tissue and / Applicable to devices.
일 예는 제 1 단백질과 제 2 단백질 간 단백질-단백질 상호작용을 측정하는 단계를 포함하고, 상기 제 1 단백질은 HER2 및 HER3이고, 상기 HER2및 HER3는헤테로다이머를형성하며, 제 2단백질은세포또는조직 내 및/또는세포또는조직 간의 신호전달경로중에서 HER2 , HER3 , 또는 이들 모두의 하위 단백질인, 세포 또는 조직 내에서의 HER2 , HER3 , 또는 이들 모두(예컨대, HER2-HER3 헤테로다이머 형태)가 관여하는 신호전달경로의 활성화 측정 (또는 확인 또는 결정 또는 분석) 방법을 제공한다. 다른 예는 HER2-HER3 헤테로다이머에서의 HER2 및/또는 HER3의 인산화 정도를 측정하는 단계를 포함하는, HER2-HER3 헤테로다이머의 활성화 정도를 측정 (또는 확인 또는 결정 또는분석) 방법을 제공한다. 이 경우 제 2단백질은 HER2-HER3 헤테로다이머의 인산화기 (예컨대, 인산화된 타이로신)에 결합하는단백질들중에서 선택된것일수있다. . An example includes measuring protein-protein interactions between a first protein and a second protein, wherein said first protein is HER2 and HER3, said HER2 and HER3 forming a heterodimer, HER2, HER3, or HER2 in a cell or tissue, which is a sub-protein of HER2, HER3, or both in a tissue and / or in a signal transduction pathway between cells or tissues. (Or confirmation or determination or analysis) of the signal transduction pathway in which both (e. G., HER2-HER3 heterodimer forms) are involved. Another example provides a method of measuring (or identifying or determining or determining) the degree of activation of the HER2-HER3 heterodimer, comprising measuring the degree of phosphorylation of HER2 and / or HER3 in the HER2-HER3 heterodimer. In this case, the second protein may be selected from proteins that bind to the phosphorylating group of the HER2-HER3 heterodimer (e.g., phosphorylated tyrosine). .
다른예는상기 기재된방법에사용하기 위한장치를제공한다.  Another example provides an apparatus for use in the method described above.
다른예는,  In another example,
PD-L1이 부착된 기판에 표지물질이 결합된 卵 -1을 공급하여 반응시키는단계; 및  1 by supplying a labeled substance-bound egg-1 to a substrate to which PD-L1 is attached; And
상기 반응시키는단계에서 발생한신호(A)를즉정하는단계 를포함하고,  Determining a signal (A) generated in the reacting step,
상기 PD-L1과 PD-1의 반응물은클러스터를형성하는것인,  The reactants of PD-L1 and PD-1 form clusters,
PD-L1과 PD-1의 상호작용측정 방법을제공한다. 상기 방법은, 상기 반응시키는단계 이전에, PD-L1에 결합하는물질이 표면에 부착된 기판에 PC-L1을 공급하여, PD-L1을 기판에 부착시키는 단계를 추가로 포함하는 것일 수 있다. PD-L1에 결합하는 물질이 표면에 부착된 기판에 표지 물질이 결합된 PC-L1을 공급하고, PD-L1과 기판 표면의 물질 간 반응에 의하여 발생하는 신호 ( 를 측정하는 단계를 추가로 포함하는 것일 수 있다. 상기 신호 ( 에 대한신호 (A) 값 (즉, [신호 (A) 값]/[신호 (B) 값])을 구하여 정량화하는 단계를 추가로 포함하는 것일 수 있다. 상기 반응시키는 단계에서, PD-L1 및 PD-1 간 결합에 대하여 PD-L1 또는 PD- 1과 경쟁하는물질을 첨가하여 반응시카고, 신호 ( 를즉정하는 단계를 수행하는 것을 특징으로 하는 것일 수 있다. 상기 방법에서, 상기 신호 (A) 또는 ( 는 근접장 (near-f i eld) 영역에서 측정되는 것일 수 있다. 상기 방법에 있어서, 상기 기판에 부착된 PD-L1에 결합하는 물질은 PD- L1의 말단부위에 결합하는물질일수있다.  Provides a method for measuring the interaction between PD-L1 and PD-1. The method may further include the step of supplying PC-L1 to a substrate to which a substance binding to the PD-L1 is attached to the surface, and attaching the PD-L1 to the substrate before the step of reacting. A step of supplying a PC-L1 in which a labeling substance is bound to a substrate to which a substance binding to the PD-L1 has been attached, and a step of measuring a signal (generated by the PD-L1 and the substance- . (A) value (i.e., [signal (A) value] / [signal (B) value] for the signal) and quantifying the signal. In the above-mentioned step of reacting, a step of adding a substance competing with PD-L1 or PD-1 to the binding between PD-L1 and PD-1 is performed to perform a step of determining a reaction Chicago signal. In the method, the signal (A) or () may be measured in the near-field region. In the above method, the substance binding to the PD-L1 attached to the substrate may be a substance binding to the end of the PD-L1.
다른예는,  In another example,
후보 물질, 卵 -L1, 및 표지 물질이 결합된 抑- 1가혼합된 혼합물을 1 mixture of the candidate substance, egg-Ll, and the labeled substance bound thereto
PD-L1과 결합하는 물질이 표면에 부착된 기판에 공급하여 반응시키는 단계; 및 Supplying and reacting a substance that binds with PD-L1 to a substrate having a surface attached thereto; And
상기 반응시키는단계에서 얻어진신호(시를측정하는단계 를 포함하는, 抑- L1과 PD-1 간 상호작용 억제제의 선정 방법을 제공한다. 상기 방법은 PD-L1에 결합하는물질이 표면에 부착된 기판에 표지 물질이 결합된 PC-L1을 공급하고, PD-L1과 기판 표면의 물질 간 반응에 의하여 발생하는 신호 ( 를 측정하는 단계를 추가로 포함하는 것일 수 있다. 상기 신호 (A) 또는 ( 는 근접장 (near-f ield) 영역에서 측정되는 것일 수 있다. 상기 방법은상기 신호 ( 에 대한신호 (A) 값 (즉, [신호 (A) 값]/[신호 (B) 값])을구하여 정량화하는 단계를추가로 포함하는 것일 수 있다. 상기 방법에서, 후보물질 무처리군 (예컨대, 후보물질 처리 전)과비교하여, 후보물질 처리군 (예컨대, 후보물질 처리 후)에서의 신호 (A) 값 또는 신호 ( 에 대한 신호 (A) 값이 감소하는 경우, 상기 후보물질을 PD-L1과 PD-1 간 상호작용 억제제로 선정하는 단계를추가로포함할수있다. The signal obtained in the step of reacting (measuring the time Lt; RTI ID = 0.0 > PD-1. ≪ / RTI > In this method, a PC-L1 having a label attached to a substrate to which a substance binding to PD-L1 is attached is supplied, and a signal (produced by the reaction between PD-L1 and the substance on the surface of the substrate is measured The signal A may be measured in the near-field region. The method may include measuring the value of the signal A for the signal (i.e., (A) value] / [signal (B) value]) of the candidate substance, and quantifying the candidate substance. When the signal (A) value or the signal (A) value for the signal (A) value at the treatment group (for example, after the candidate substance treatment) decreases, the candidate substance is selected as an inhibitor of interaction between PD-L1 and PD- Step may be further included.
다른예는,  In another example,
제 1수용체 타이로신 인산화효소또는이를암호화하는유전자를제 2 수용체타이로신 인산화효소발현세포에 도입시키는단계;  Introducing a first receptor tyrosine kinase or a gene encoding the first receptor tyrosine kinase into a second receptor tyrosine kinase-expressing cell;
상기 세포에 제 1수요에 타이로신 인산화효소리간드를투입하여 제 1 수용체 타이로신 인산화효소와 제 2 수용체 타이로신 인산화효소 간 이합체를형성시키는단계; 및  Introducing a tyrosine phosphorylase ligand into the cell at a first demand to form a duplex between the first receptor tyrosine phosphorylase and the second receptor tyrosine phosphorylase; And
상기 이합체가 형성된 세포에 콜레스테롤-유사 세정제를 처리하는 단계  Treating the cell on which the dimer is formed with a cholesterol-like detergent
를포함하고,  Lt; / RTI >
제 1 수용체 타이로신 인산화효소과 제 2 수용체 타이로신 인산화효소는서로다른종류이고,  The first receptor tyrosine phosphorylase and the second receptor tyrosine phosphorylase are different kinds,
생체외에서 수행되는것을특징으로하는,  Lt; RTI ID = 0.0 > in vitro, < / RTI >
활성화된 수용체 타이로신 인산화효소 이합체의 제조 방법을 제공한다. 상기 제조된 수용체 타이로신 인산화효소 이합체는 상기 방법에 의하지 않은 경우와 비교하여, 활성화된 형태일 수 있다. 상기 방법에서, 상기 제 1수용체 타이로신 인산화효소는 HER3이고, 제 2수용체 타이로신 인산화효소는 HER2일 수 있다. 또한, 상기 콜레스테롤-유사 세정제는 0.05내지 5%(w八 0의 DGTN(digi tonin) 및 0.003내지 2%(w八 0의 GDN (glycol-diosgenin)중에서 선택된 1종이상일수있다.  And a method for producing an activated receptor tyrosine kinase dimer. The prepared receptor tyrosine kinase duplex may be in an activated form, as compared to the case without the method. In this method, the first receptor tyrosine phosphorylase may be HER3 and the second receptor tyrosine phosphorylase may be HER2. In addition, the cholesterol-like detergent may be at least one selected from 0.05 to 5% (DGTN) and 0.003 to 2% (GDNO) of glycolic acid.
【기술적 해결방법】  [Technical Solution]
이하, 보다상세히 설명한다. 본 명세서에서 사용된 바로서, 용어 ’’단백질-단백질 상호작용 (protein-protein interact ion: PPI)”은 저 U 단백질과 제 2 단백질 간의 물리적 및/또는화학적 결합또는복합체 형성을의미할수 있으며, 예컨대, 결합 빈도, 결합 세기 (강도) , 및 결합 시간 등의 인자들 중에서 하나 이상으로 측정될 수 있다. 또한, 상기 제 1 단백질과 제 2 단백질 간의 상호작용 (결합)은 이들 간의 직접적인 상호작용 (결합)뿐 아니라 중간에 다른 단백질 (신호전달경로 중에서 제 1 단백질 및 제 2 단백질 사이에 위치하는단백질)을매개로한간접적인상호작용 (결합)도포함할수 있다. 본명세서에서의 단백질-단백질상호작용은단분자 (s ingle molecule) 반응 (1분자의 제 1단백질과 1분자의 제 2단백질 간의 반응)일 수 있다. 세포 또는 조직 간의 상호작용은 2 이상의 동종 세포 또는 동종 조직, 또는 동일/동종/이종 세포 또는 동일/동종/이종 조직의 표면 단백질 사이에서 일어나는모든상호작용을의미하는것일수있다. This will be described in more detail below. As used herein, the term " protein-protein interaction (PPI) " may refer to a physical and / or chemical association or complex formation between a low U protein and a second protein, , Binding frequency, binding strength (intensity), and binding time. In addition, the interaction (binding) between the first protein and the second protein includes not only direct interaction (binding) between them, but also other proteins (proteins located between the first protein and the second protein in the signal transduction pathway) It can be applied indirectly to the impression (bond) for indirect impression. The protein-protein interaction in the present specification may be a single-molecule reaction (reaction between one molecule of the first protein and one molecule of the second protein). Cell or tissue interactions may refer to all interactions taking place between two or more homologous cells or allogeneic tissues, or surface proteins of the same / homogeneous / heterogeneous cell or homologous / heterologous / heterogeneous tissue.
본 명세서에서, 제 1 단백질과 제 2 단백질은 각각 독립적으로 진핵 유기체 (예컨대, 다세포 동물, 다세포 식물 등)의 세포 또는 조직의 신호전달경로에 관여하는 단백질 중에서 선택되는 1종 이상이거나, 어느 하나는수용체이고 다른 하나는 리간드이거나, 이종 세포의 표면 단백질들 중에서 선택되는 1종이상일수있다. In the present specification, the first protein and the second protein are each independently at least one selected from proteins involved in signal transduction pathways of cells or tissues of eukaryotic organisms (for example, multicellular animals, multicellular plants, etc.) Receptor and the other is a ligand, or may be one or more selected from the surface proteins of xenogeneic cells .
일 예에서, 상기 제 1 단백질 및 제 2 단백질 중에서 하나 이상은 인산화된 것일 수 있다 (예컨대, 상기 제 1 단백질 및 제 2 단백질 중에서 하나 이상은 인산화된 타이로신 (p-Tyr)을포함하는 것일 수 있다) . 이와 같이 제 1 단백질 및 제 2 단백질 중에서 하나 이상은 인산화된 것인 경우, 상기 제1 단백질과 제2 단백질 간의 상호작용은 인산화 정도 (p-Tyr 수준)에 의존적인 것일 수 있다. 다른 예에서, 상기 제 1 단백질 및 제 2 단백질 중에서 하나 이상은 이들 간상호작용에 영향을 미치는돌면변이를 갖는것일 수 있다. 상기 돌연변이는신호경로상에서 서로상호작용할대상 단백질의 존재 또는수준에 의존하지 않고 (예컨대, 제 1단백질이 수용체인 경우, 이와 상호작용하는 리간드 또는 하위 단백질의 존재 또는 수준에 무관하게) 신호전달이 가능하도록하는것일수 있다 (예컨대, EGFR의 exon 19및/또는 exon 21변이 (결실)등) .  In one example, one or more of the first and second proteins may be phosphorylated (e.g., one or more of the first and second proteins may comprise phosphorylated tyrosine (p-Tyr) ). Thus, when at least one of the first protein and the second protein is phosphorylated, the interaction between the first protein and the second protein may be dependent on the degree of phosphorylation (p-Tyr level). In another example, one or more of the first and second proteins may have a surface variation that affects interactions between them. The mutation is not dependent on the presence or level of the protein of interest to interact with on the signal path (e.g., if the first protein is a receptor, irrespective of the presence or level of the interacting ligand or subprotein) (E. G., Exon 19 and / or exon 21 mutation (deletion) of EGFR, etc.).
본 명세서에서 제공되는 모든 방법에서의 단백질-단백질 상호작용 측정에서의 단계 (1), (2), 및 (3), 또는 (1) , (2) , (3) , 및 (4) , 또는 (1), (2), (3), 및 (4-1), 또는 (1), (2) , (3) , (4-1) , 및 (4-2)는, 2019/132517 1»(:1^1{2018/016675 (1), (2), and (3), or (1), (2), (3), and (4) in the protein-protein interaction measurement in all methods provided herein, or (1), (2), (3) and (4-1), or (1), (2), (3), (4-1) 2019/132517 1 »(: 1 ^ {2018/016675
3)수용체 (제 1단백질) 및 리간드(제 2단백질)간의 상호작용(이 경우,
Figure imgf000011_0001
첨가하여 ??1변화측정 가능),
( 3 ) interaction between the receptor (first protein) and the ligand (second protein) (in this case,
Figure imgf000011_0001
1 change measurement possible),
( 1단백질 및 제 2단백질 중에서 하나이상이 인산화된 타이로신 잔기를 갖는 경우의 제 1 단백질 및 제 2 단백질 간의 상호작용 (이 경우, 타이로신의 인산화 정도로 제 1 단백질 및/또는 제 2 단백질의 활성화 정도 예측가능), 및  (1) the interaction between the first protein and the second protein when at least one of the first protein and the second protein has a phosphorylated tyrosine residue (in this case, the degree of phosphorylation of the tyrosine predicts the degree of activation of the first protein and / Available), and
0)제 1단백질 및상기 제 1단백질이 관여하는신호전달경로중에서 저 단백질의 하위 단백질인제 2단백질간의 상호작용 ( 0 ) Interaction between the first protein and the second protein of the low protein in the signal transduction pathway involved in the first protein
중에서 선택된 하나 이상, 두 개 이상, 또는 세 개 모두에 대하여 수행되는것일수있다.  , ≪ / RTI > two or more, or all three.
본 명세서에 사용된 바로서 , 제 1단백질은신호전달경로에 관여하는 1종 이상 또는 2종 이상 (예컨대, 1종 내지 10종 또는 2종 내지 10종)의 단백질을 의미할 수 있다. 또한, 제 2 단백질은 상기 제 1 단백질과 상호작용(결합)하는 단백질로서, 상기 제 1 단백질이 관여하는 신호전달경로의 하위 경로에 관여하는단백질 중에서 선택된 1종 이상또는 2종 이상 (예컨대, 1종내지 10종또는 2종 내지 10종)의 단백질을의미할 수 있다. 제 1 단백질이 2종 이상인 경우, 상기 제 2 단백질은 제 1 단백질 각각에 대하여 독립적으로 1종 이상 선택될 수 있으며, 각각의 제 1 단백질에 대하여 선택된 제 2 단백질은 서로 다르거나, 부분적으로 또는 전부중복될수있다.  As used herein, the first protein may refer to one or more (e.g., one to ten or two or ten) proteins involved in the signal transduction pathway. The second protein is a protein that interacts with (binds to) the first protein. The second protein may be one or more proteins selected from the proteins involved in the lower pathway of the signal transduction pathway involved in the first protein, Ten to ten or two to ten species) proteins. When the first protein is two or more kinds, the second protein may be independently selected for each of the first proteins, and the second protein selected for each first protein may be different, partially or completely It can be duplicated.
본 명세서에 있어서, 상기 제 1 단백질은 병적 상태 (예컨대, 암, 염증, 그 외 면역 질환 등)와 관련 있는 단백질들 중에서 선택됨으로써, 이들이 관련된 병적 상태 (예컨대, 암, 염증, 그외 면역 질환등)의 치료 (및/또는 개선 및/또는 경감)에 유용한 정보를 제공할 수 있다. 이러한 측면에서, 상기 제 1 단백질은 치료하고자 하는 질병의 치료 타겟이 되는 단백질일 수 있다. 구체적으로, 상기 제 1 단백질은 치료하고자 하는 질병에 대한 표적 치료제 또는 효과를 시험하고자 하는 치료제의 표적이 되는단백질일수 있다. 따라서, 상기 제 1단백질은치료하고자하는질병 또는효과를알고자하는치료제에 따라서 적절히 선택될수있다.  In the present specification, the first protein is selected from among proteins associated with pathological conditions (e.g., cancer, inflammation, other immune diseases, etc.), so that the pathological conditions (e.g., cancer, inflammation, (And / or < / RTI > improve and / or alleviate) the disease. In this respect, the first protein may be a protein that is a therapeutic target of the disease to be treated. Specifically, the first protein may be a target therapeutic agent for a disease to be treated or a target protein of a therapeutic agent to be tested. Accordingly, the first protein can be appropriately selected depending on the therapeutic agent to be sought for the disease or effect to be treated.
일 예에서, 상기 제 1단백질은세포또는조직의 생체 신호전달경로 중상위 경로에 관여하는단백질, 및 이종세포간상호작용관여하는세포 표면 단백질 또는 이종세포가 분비(방출)하는 단백질 (예컨대, 엑소좀 등)들 중에서 선택될 수 있으며, 약물의 치료 타겟으로서 유리하게 작용하기 위하여, 세포외 (extracel lular) 환경 (예컨대, 수성 환경)에 노출된 도메인을 갖고 세포막에 존재하는 세포막 단백질 중에서 1종 이상 선택될 수 있다. 예컨대, 상기 제 1 단백질은 세포막에 존재하는 다양한 수용체, 마이크로필라멘트 (microfilaments)와 결합된 구조단백질 (structural proteins) , 세포부착분자 (cell adhesion molecules) , 세포막 효소 (membrane enzymes ) , 세포막 수용체 (membrane receptors) , 캐리어 단백질 (carrier proteins) , 채널 단백질 (chamel proteins) , 운반 단백질 (transport proteins) , 지질 고정 단백질 (lipid-anchored proteins) 등모든종류의 세포막단백질로 이루어진 군에서 선택된 1종 이상, 일 수 있다. In one example, the first protein is a protein involved in the mediator pathway in the biological signal transduction pathway of a cell or a tissue, and a cell surface protein or a protein secreted (released) by xenogeneic cells (for example, exo And the like), and can be advantageously used as a therapeutic target of the drug In order to function, one or more of the cell membrane proteins having a domain exposed to an extracelular environment (for example, an aqueous environment) and present in the cell membrane may be selected. For example, the first protein may be various receptors present in cell membranes, structural proteins associated with microfilaments, cell adhesion molecules, membrane enzymes, membrane receptors, And at least one selected from the group consisting of all kinds of cell membrane proteins, such as lipid-anchored proteins, carrier proteins, chamel proteins, transport proteins, lipid-anchored proteins, .
일 구체예에서, 상기 제 1 단백질은 수용체 티로신 카이네이즈류 (Receptor Tyrosine Kinase; RTK) (예컨대, 상피세포 성장인자 수용체 (epidermal growth factor receptor; EGFR; ErbBl) , HER2( Human Epidermal growth factor Receptor 2 protein; ErbB2) , HER3( Human Epidermal growth factor Receptor 3 protein; ErbB3) , 간세포 성장인자 수용체 (hepatocyte growth factor receptor (HGFR) : MET) , 혈소판유래 성장인자 수용체 (platelet-derived growth factor receptors; PDGFR , 예컨대, PDGFR-alpha, PDGFR-beta 등), 혈관내피세포 성장인자 수용체 (vascular endothelial growth factors; VEGFR, 예컨대, VEGFR1 , VEGFR2 , VEGFR3등), 인슐린-유사성장인자 1 수용체 (Insulin-like Growth Factor 1 Receptor; IGF1R) , 에프린 수용체 (ephrin receptors) , 섬유아세포 성장인자 수용체 (Fibroblast growth factor receptor; FGFR, 예컨대, FGFR1, FGFR2 등), 인슐린-유사 성장인자 수용체 (Insulin-1 ike Growth Factor Receptor; IGFR, 예컨대, IGF1R등), c-KIT, RET receptor tyrosine kinase, Anaplastic lymphoma kinase (ALK) , 등); 톨-유사수용체 (Toll like receptor; 예컨대, TLR1, TLR2, TLR3, TLR4, TLR5,· TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12, TLR13) : G_단백질 연결 수용체류 (G-protein- coupled receptors; GPCR); 트랜스페린 수용체 (transferrin receptors); 저밀도지질단백질 (Low-Density Lipoprotein; LDL) 수용체; R0S1; BCR-Abll 융합 단백질; 비수용체형 카이네이즈류 (예컨대, BRAF, MEK (Mitogen- act ivated protein kinase kinase) , Src, PI3K ( Phosphoi nos i tide 3- kinase) , CDK Cycl in-dependent kinases; 예컨대, CDK4, CDK6등) 등); GTP 가수분해효소류 (GTPases) (예컨대, KRAS 등); 호르몬 수용체류 (예컨대, 에스트로겐수용체 (ER) , 프로게스테론수용체 (PR) , 안드로겐수용체 (AR) 등) ; 항-아팝토시스 단백질류 (Ant i-apoptot i c proteins) (예컨대, BCL2 (B-cel 1 lymphoma 2) , BIM (Bcl-2-1 ike protein 11) ; 면역 체크포인트 단백질류 ( Immune checkpoint proteins) (예컨대, CTLA-4 (cytotoxic T- lymphocyte -associated ant igen 4), PD-1 (programmed death 1), PD-L1 (programmed death- l igand 1) 등) 등으로 이루어진 군에서 선택된 1종 이상일수있으나, 이에 제한되는것은아니다. In one embodiment, the first protein is selected from the group consisting of receptor tyrosine kinase (RTK) (e.g., epidermal growth factor receptor (EGFR; ErbBl), human epidermal growth factor receptor 2 protein (HER2) ErbB2), HER3 (Human Epidermal Growth Factor Receptor 3 protein: ErbB3), hepatocyte growth factor receptor (HGFR): MET, platelet-derived growth factor receptors (PDGFR, VGFR1, VEGFR2, VEGFR3, etc.), Insulin-like Growth Factor 1 Receptor (IGF1R, VEGFR, EGFR receptors, fibroblast growth factor receptors (FGFRs such as FGFR1 and FGFR2), insulin-like growth factor receptors (e.g., 1 ike Growth Factor Receptor; IGFR, such as IGF1R), c-KIT, RET receptor tyrosine kinase, Anaplastic lymphoma kinase (ALK), etc.); Toll-like receptors (Toll like receptor; e. G., TLR1, TLR2, TLR3, TLR4 , TLR5, · TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12, TLR13): G_ protein connection receiving stay (G-protein- coupled receptors (GPCRs); transferrin receptors; Low-Density Lipoprotein (LDL) receptors; R0S1; BCR-Abll fusion protein; (Eg, BRAF, mitogen-activated protein kinase kinase (MEK), Src, PI3K (Phosphoinositide 3-kinase), CDK cyclin-dependent kinases such as CDK4, CDK6, etc.); GTPases (GTPases) (e.g., KRAS and the like); The hormone receptor (e.g., Estrogen receptor (ER), progesterone receptor (PR), androgen receptor (AR), etc.); Anti- apoptotic proteins (such as BCL2 (B-cel 1 lymphoma 2), BIM (Bcl-2-1 ike protein 11), immune checkpoint proteins, (For example, CTLA-4 (cytotoxic T-lymphocyte-associated antigen 4), PD-1 (programmed death 1), PD-L1 (programmed death ligand 1) But is not limited thereto.
상기 "간세포 성장인자 수용체 (MET 또는 c-Met)’’은 간세포 성장인자와 결합하는 수용체 티로신 카이네이즈를 의미한다. 상기 c-Met 단백질은 모든 종에서 유래하는 것일 수 있으며, 예컨대, 인간 c-Met (예컨대 , NP_000236.2) , 원승이 c-Met (예컨대, Macaca mulatta, The c-Met protein may be derived from any species and includes, for example, a human c-Met (c-Met) (E. G., NP_000236.2), a c-Met (e. G., Macaca mulatta,
NPJ301162100) 등과 같은 영장류 유래의 것, 또는 마우스 c-Met (예컨대, NP_032617.2) , 래트 c-Met (예컨대, NP_113705.1) 등과같은설치류유래의 것 등일 수 있다. 상기 단백질은 예를 들면, GenBank Aceession Number ■_00(犯45.3에 제공된 뉴클레오티드 서열에 의해 암호화된 폴리펩티드, 또는 GenBank Aceession Number NP_000236.2에 제공된 아미노산 서열을 포함하는 단백질, 또는 그의 세포외 도메인을 포함한다. 수용체 티로신 키나제 c-Met은 예를 들면, 암발생, 암전이, 암세포 이동, 암세포 침투, 신생혈관생성 과정 등의 여러 가지 기작에 관여한다. NPJ301162100), or rodent-derived ones such as mouse c-Met (e.g., NP_032617.2), rat c-Met (e.g., NP_113705.1), and the like. Such a protein includes, for example, a polypeptide encoded by the nucleotide sequence provided in GenBank Ace Session Number __00 (crime 45.3, or a protein comprising the amino acid sequence provided in GenBank Aceession Number NP_000236.2, or an extracellular domain thereof. The receptor tyrosine kinase c-Met is involved in various mechanisms such as, for example, cancer development, cancer metastasis, cancer cell migration, cancer cell infiltration, and neovascularization process.
상기 "상피세포성장인자수용체 (Epidermal growth factor receptor; The "Epidermal growth factor receptor "
EGFR)" , HER2 (human epidermal growth factor receptor 2 protein) , 및 HER3 (human Epidermal growth factor receptor 3 protein)는 각각 EGFR (HERD , HER2, HER3 및 HER4으로 구성되어 있는 HER 패밀리와 수용체 티로신 키나아제 (RTKs)의 일원이다. EGFR, HER2, 또는 HER3의 세포외 도메인에 대한 리간드의 결합은 다른 ErbB 수용체와의 리셉터 호모- 또는 헤테로-이량체화를 유도하며, 이는 특이적인 티로신 잔기의 세포내 자가인산화를유발한다. EGFR자가인산화는세포증식, 혈관신생 및 전이에 영향을 미치는 MAPK 및 PI3K/Akt 활성화를 포함한 다운스트림 신호전달 네트워크를 이끈다. EGFR, HER2, 및/또는 HER3의 과발현, 유전자 증폭, 돌연변이, 또는재배열은여러 종류의 인간악성 종양에서 빈번히 관찰되며, 암치료의 불량한예후및 나쁜 임상적 결과와관련 있다. 이러한이유로, 이들 EGFR, HER2, 및/또는 HER3는항암요법에 있어서 중요한표적이 된다. 상기 EGFR, HER2, 또는 HER3는 인간, 원숭이 등의 영장류, 마우스, 래트등의 설치류등의 포유류로부터 유래된 것일 수 있다. 예컨대, 상기 EGFR은 GenBank Accession Nos . JQ739160, JQ739161, JQ739162, JQ739163, JQ739164, JQ739165, JQ739166, JQ739167, NM_005228.3, NM_201284.1,EGFR), HER2 (human epidermal growth factor receptor 2 protein), and HER3 (human epidermal growth factor receptor 3 protein) are HER family members and receptor tyrosine kinases (RTKs) composed of EGFR (HERD, HER2, HER3 and HER4) Binding of the ligand to the extracellular domain of EGFR, HER2, or HER3 induces receptor homo- or hetero-dimerization with other ErbB receptors, leading to intracellular autophosphorylation of specific tyrosine residues EGFR autophosphorylation leads to downstream signaling networks, including MAPK and PI3K / Akt activation, that affect cell proliferation, angiogenesis and metastasis EGFR, HER2, and / or HER3 overexpression, gene amplification, The sequences are frequently observed in many types of human malignant tumors and are associated with poor prognosis of cancer treatment and poor clinical outcome. For this reason, these EGFR, HER2, and / or HER3 EGFR, HER2, or HER3 may be a primate such as human, monkey, mouse, And may be derived from mammals such as rodents such as rats. For example, the EGFR can be obtained from GenBank Accession Nos. JQ739160, JQ739161, JQ739162, JQ739163, JQ739164, JQ739165, JQ739166, JQ739167, NM_005228.3, NM_201284.1,
NM_201282.1, 또는 NM_201283.1 등에 제공된 뉴클레오타이드 서열 (_A)에 의하여 암호화된 폴리펩타이드일 수 있다. 예컨대, 상기 HER2는 GenBank Accession No. X03363.1 등에 제공되는 뉴클레오타이드 서열 (mRNA)에 의하여 암호화된 폴리펩타이드일 수 있다. 예컨대, 상기 HER3는 GenBank Accession No. NM_001982 등에 제공되는 뉴클레오타이드 서열 (mRNA)에 의하여 암호화된폴리펩타이드일수있다. NM_201282.1, or a polypeptide encoded by the nucleotide sequence (A) provided in NM_201283.1 or the like. For example, the HER2 may be derived from GenBank Accession No. < / RTI > Or may be a polypeptide encoded by a nucleotide sequence (mRNA) provided in X03363.1 or the like. For example, the HER3 can be obtained from GenBank Accession No. < / RTI > And may be a polypeptide encoded by a nucleotide sequence (mRNA) provided in NM_001982 or the like.
상기 "혈관내피세포 성장 인자 수용체 (Vascular Endothel i al Cel l The "Vascular Endothelial Growth Factor Receptor "
Growth Factor Receptor : VEGFR)”는 혈관내피세포 성장 인자 정상세포에서도 존재하며, 특히 암세포에서 분비되는 혈관내피세포 성장인자 (VEGF)와 결합하여 혈관신생을 일으키며, 암세포에 필요한 양분을 공급한다. VEGFR의 과발현은다양한질병의 원인이되며, 특히 암의 발생뿐 아니라, 침습, 전이 등의 나쁜예후에도관여한다. 이러한이유로, VEGF는 항암요법에 있어서 중요한표적이 된다. 상기 VEGFR은인간, 원숭이 등의 영장류, 마우스, 래트등의 설치류등의 포유류로부터 유래된 것일 수 있다. 예컨대, 상기 VEGFR은 GenBank Accession Number AF063657.2 등에 제공된 뉴클레오타이드서열 (mRNA)에 의해 암호화된폴리펩타이드일수있다. Growth Factor Receptor (VEGFR) "exists in normal vascular endothelial growth factor cells. In particular, it binds to vascular endothelial growth factor (VEGF) secreted from cancer cells to induce angiogenesis and supply necessary nutrients to cancer cells. Overexpression of VEGFR is a cause of various diseases, and it is involved not only in the development of cancer but also after bad cases such as invasion and metastasis. For this reason, VEGF is an important target in chemotherapy. The VEGFR may be derived from mammals such as primates such as humans and monkeys, and rodents such as mice and rats. For example, the VEGFR may be a polypeptide encoded by a nucleotide sequence (mRNA) provided in GenBank Accession Number AF063657.2.
상기 "혈소판유래성장인자수용체 (platelet-der ived growth factor receptors; PDGFR)"는표면수용체 티로신 카이네이즈중하나로, 세포증식, 세포분화, 세포성장 등의 조절 및 암을 일으키는 많은 질병과 관련 있다. 상기 PDGFR은 인간, 원숭이 등의 영장류, 마우스, 래트 등의 설치류 등의 포유류로부터 유래된 것일 수 있다. 예컨대, 상기 PDGFR은 GenBank Accession Nos. NM_006206.4(PDGFR-A) , NM_002609.3(PDGFR-B), The above-mentioned " platelet-derived growth factor receptors (PDGFR) "is one of the surface receptor tyrosine kinases, and is associated with many diseases that cause cell growth, cell differentiation, cell growth, The PDGFR may be derived from mammals such as primates such as humans and monkeys, and rodents such as mice and rats. For example, the PDGFR can be obtained from GenBank Accession Nos. NM_006206.4 (PDGFR-A), NM_002609.3 (PDGFR-B),
■_016205.2(卵 GFR-C) 등에 제공된 뉴클레오타이드 서열 (mRNA)에 의해 암호화된폴리펩타이드일수있다. (SEQ ID NO: 1), or a nucleotide sequence (mRNA) provided in SEQ ID NO.
상기 "인슐린-유사성장인자 1수용체 (Insul in-l ike Growth Factor 1 Receptor; IGF1R)1’는수용체 티로신 카이네이즈 중 하나로, 인슐린-유사 성장인자 l(IGF-l)에 의하여 활성화되는 막통과 수용체이다. 상기 IGF1R은 인간, 원숭이 등의 영장류, 마우스, 래트 등의 설치류 등의 포유류로부터 유래된 것일 수 있다. 예컨대, 상기 IGF1R은 GenBank Accession No. NM_000875.3 등에 제공된 뉴클레오타이드 서열 (_A)에 의해 암호화된 2019/132517 1»(:1/10公018/016675 The "Insulin-i Ike Growth Factor 1 Receptor (IGF1R) 1 " is one of the receptor tyrosine kinases and is a transmembrane receptor that is activated by insulin-like growth factor 1 (IGF-1) . The IGF1R may be derived from a mammal such as a primate such as a human, a monkey, a rodent such as a mouse or a rat, etc. For example, the IGF1R may be derived from a nucleotide sequence encoded by nucleotide sequence (A) provided in GenBank Accession No. NM - 2019/132517 1 »(: 1/10/06 018/016675
폴리펩타이드일수있다. Lt; / RTI > polypeptide.
상기 "에프린 수용체 (ephr in receptors) "는 표면 수용체 티로신 카이네이즈 중 하나로, axon guidance, format ion of t i ssue boundar i es , cel l migrat ion, segmentat ion등의 배발생 과정을 조절한다. 상기 에프린 수용체는 인간, 원숭이 등의 영장류, 마우스, 래트 등의 설치류 등의 포유류로부터 유래된 것일 수 있다. 예컨대, 상기 에프린 수용체는 GenBank Accession Nos . NM_004440.3 , NM_004438.3 , NM_004431.3, The above-mentioned "ephrins receptors" are one of the surface receptor tyrosine kinases, which regulate the axon guidance, the format ion of the ssue boundaries, the cell migrat ion, and the segmentation ion. The ephrin receptor may be derived from mammals such as primates such as humans and monkeys, and rodents such as mice and rats. For example, the ephrin receptor may be derived from GenBank Accession Nos. NM_004440.3, NM_004438.3, NM_004431.3,
NM_004442.6, NM_017449.3, NM_004093.3, NM_004441.4, NM_182472.2,NM_004442.6, NM_017449.3, NM_004093.3, NM_004441.4, NM_182472.2,
NM-005232.4, NM_005233.5, NM_173641.2, NM_001099439.1, NM_001080448.2, NM_001080448.2, NM_004443.3, NM_182689.1, NM_004428.2, NM_004439.5,NM-005232.4, NM_005233.5, NM_173641.2, NM_001099439.1, NM_001080448.2, NM_001080448.2, NM_004443.3, NM_182689.1, NM_004428.2, NM_004439.5,
NH001962.2, NM_004429.4, NM_182644.2, NM_004952.4, NM_173655.2,NH001962.2, NM_004429.4, NM_182644.2, NM_004952.4, NM_173655.2,
NM_182690.2, NM_020526.3, NM_001406.3, NM_005227.2, NM_182685.1 등에 제공된 뉴클레오타이드 서열 (mRNA)에 의해 암호화된 폴리펩타이드일 수 있다. Or may be a polypeptide encoded by a nucleotide sequence (mRNA) provided in NM_182690.2, NM_020526.3, NM_001406.3, NM_005227.2, NM_182685.1, and the like.
상기 "트랜스페린 수용체 (transferr in receptors)"는 트랜스페린의 캐리어 단백질로, 수용체-매개 엔도사이토시스를 통하여 철의 세포내 이동에 관여하며, 세포내 철 농도를 조절한다. 상기 트랜스페린 수용체는 안간, 원숭이 등의 영장류, 마우스, 래트 등의 설치류 등의 포유류로부터 유래된 것일 수 있다. 예컨대, 상기 트랜스페린 수용체는 GenBank Accession Nos . NM_001128148.1, NM_003234.2, NM_001206855.1, The "transferrin receptors" are carrier proteins of transferrin, which participate in the intracellular transport of iron through receptor-mediated endocytosis and regulate intracellular iron concentrations. The transferrin receptor may be derived from a mammal such as a rat, a primate such as a monkey, or a rodent such as a mouse or a rat. For example, the transferrin receptor may be derived from GenBank Accession Nos. NM_001128148.1, NM_003234.2, NM_001206855.1,
NM_003227.3, B⑶ 01188.1, M11507.1 등에 제공된 뉴클레오타이드 서열 (mRNA)에 의해 암호화된폴리펩타이드일수있다. NM_003227.3, B₃ 01188.1, M11507.1, and the like.
상기 "저밀도지질단백질 (Low-Densi ty Lipoprotein; LDL) 수용체"는 트랜스페린의 캐리어 단백질로, 수용체-매개 엔도사이토시스를통하여 철의 세포내 이동에 관여하며, 세포내 철 농도를 조절한다. 상기 LDL 수용체는 인간, 원숭이 등의 영장류, 마우스, 래트 등의 설치류 등의 포유류로부터 유래된 것일 수 있다. 예컨대, 상기 트랜스페린 수용체는 GenBank The " low-density lipoprotein (LDL) receptor "is a carrier protein of transferrin, which participates in the intracellular transport of iron through receptor-mediated endocytosis and regulates intracellular iron concentration. The LDL receptor may be derived from mammals such as primates such as humans and monkeys, and rodents such as mice and rats. For example, the transferrin receptor may be a GenBank
Accession Nos . NM_000527.4, NM_001195802.1, NM_001195799.1,Accession Nos. NM_000527.4, NM_001195802.1, NM_001195799.1,
NM_001195803.1, NM_001195800.1, NM_001195798.1 등에 제공된 뉴클레오타아드서열 (mRNA)에 의해 암호화된폴리펩타이드일수있다. Or may be a polypeptide encoded by a nucleotide sequence (mRNA) provided in NM_001195803.1, NM_001195800.1, NM_001195798.1, and the like.
상기 "표면 분화 항원류 (c luster of di f ferent iat ion; cluster of designat ion; CD)’'는 수용체 또는 리간드로서 다양하게 작용하는 단백질로서, 인간의 경우 약 350 가지가 알려져 있고,·세포 신호화 (cel l signal ing) , 세포부착등의 다양한세포과정에 관여한다. 상기 표면분화 항원류는 인간, 원숭이 등의 영장류, 마우스, 래트 등의 설치류 등의 포유류로부터 유래된 것일 수 있다. 예컨대, 상기 표면 분화 항원류는 모든 CD계열일 수 있으며, 특히 암전이와관련된 CD44, CD147 , 또는이의 var i ant들일 수 있고, 보다 구체적으로 GenBank Accession Nos .The above-mentioned "cluster of diatoms (CD)" is a protein that functions as a receptor or a ligand in various manners, and about 350 kinds of proteins are known in humans. Cel l signal ing, and cell adhesion. The surface-differentiating antigens may be derived from primates such as humans and monkeys, and mammals such as rodents such as mice and rats. For example, the surface-differentiating antigens may be of any CD sequence, and in particular may be CD44, CD147, or their antigens associated with cancer metastasis, and more particularly GenBank Accession Nos.
(NM_000610.3 , NM_001728.3 , X55150.1) 등에 제공된 뉴클레오타이드 서열 (mRNA)에 의해 암호화된폴리펩타이드일수있다. (MRNA) provided in the nucleotide sequences (NM_000610.3, NM_001728.3, X55150.1) and the like.
상기 nG-단백질 연결 수용체 (G-prote in-coup led receptors; GPCR)”는 막통과 수용체 단백질로, 신호 도입 경로 (signal transduct ion pathway) 및 세포 반응을 활성화하며, 다양한 질병에 관여한다. GPCR은 거대한 단백질군으로 서열 상동성 및 기능적 유사성을 기초로 6개의 클래스로 분류될 수 있다: 클래스 A 또는 클래스 1 (Rhodops in-1 ike receptors); 클래스 B또는클래스 2 (Secret in receptor fami ly) ; 클래스 C 또는 클래스 3 (Metabotropi c glutamate/pheromone); 클래스 D 또는 클래스 4 (Fungal mat ing pheromone receptors) ; 클래스 E또는 클래스 5The n G-protein coupled receptor (GPCR) is a transmembrane receptor protein that activates signal transduct ion pathway and cell response, and is involved in various diseases. GPCRs can be divided into six classes based on sequence homology and functional similarity as a large family of proteins: Class A or Class 1 (Rhodops in-1 ike receptors); Class B or Class 2 (Secret in receptor familys) ; Class C or Class 3 (Metabotropic glutamate / pheromone); Class D or Class 4 (Fungal matting pheromone receptors); Class E or Class 5
(Cycl ic AMP receptors); 및 클래스 F 또는 클래스 6(Cycl ic AMP receptors); and Class F or Class 6
(Fr zled/Smoothened) . 상기 GPCR은 인간, 원승이 등의 영장류, 마우스, 래트 등의 설치류 등의 포유류로부터 유래된 것일 수 있다. 예컨대, GPCR은 암 전이에 관련되는 케모카인 수용체 (chemokine receptors; Rhodopsin-1 ike receptor subfami ly) , 예컨대, CXC 케모카인 수용체, CC 케모카인수용체, CX3C케모카인수용체, 등일 수 있으며, 보다구체적으로 GenBank Accession Nos. NM_001123041.2, NM_005508.4 NM_005201.3, NM_016602.2 등에 제공된 뉴클레오타이드 서열 (mRNA)에 의해 암호화된 폴리펩타이드일수있다. (Fr zled / Smoothened). The GPCR may be derived from mammals such as primates such as human, primate, rodent such as mouse and rat. For example, GPCRs can be chemokine receptors (Rhodopsin-1 ike receptor subfamily) associated with cancer metastasis, such as CXC chemokine receptors, CC chemokine receptors, CX3C chemokine receptors, etc., and more specifically GenBank Accession Nos. And may be a polypeptide encoded by a nucleotide sequence (mRNA) provided in NM_001123041.2, NM_005508.4, NM_005201.3, NM_016602.2, and the like.
상기 제 2 단백질은 앞서 설명한 바와 같이 선택된 제 1 단백질과 상호작용하는 모든 단백질들, 예컨대, 상기 제 1 단백질이 관여하는 세포 또는 조직의 생체 신호전달경로의 하위 경로에 관여하는 단백질, 제 1 단백질의 리간드, 이종 세포 표면에 존재하거나 이종세포에서 방출되는 단백질등으로 이루어진군에서 1종이상또는 2종이상 (예컨대, 1종내지 10종 또는 2종 내지 10종) 선택될 수 있다. 세포 또는 조직, 예컨대, 인간의 세포또는조직의 신호전달경로및 여기에 관여하는단백질들, 특정 수용체에 대한 리간드, 세포 표면에 존재하는특정 단백질과상호작용하는 이종 세포 표면 단백질들, 세포 방출 단백질들은 비교적 잘 정립되어 있으므로 (Untangl ing the ErbB signal l ing network, Nat . Rev. Mol . Cel l Biol . 2, 127(2001); Cel l signal ing by receptor tyrosine kinases, Cel l 141, 1117 (2010) 참조), 제 1 단백질이 선정되면, 그 하류의 신호전달경로에 관여하는 단백질을 선정하는 것은 이 발명이 속하는 기술 분야의 통상의 지식을가진자에게 명확한사항이라할수있다. The second protein may be any protein that interacts with the first protein selected as described above, for example, a protein involved in a sub-pathway of a biological signal transduction pathway of a cell or tissue involved in the first protein, A ligand, a protein which is present on the surface of a xenogeneic cell or released from a xenogeneic cell, and the like, or may be selected from two or more (for example, one to ten or two to ten) species. Cells or tissues, such as the signaling pathways and the proteins involved in human cells or tissues, ligands for specific receptors, xenogeneic cell surface proteins interacting with specific proteins present on the cell surface, Relatively well established (Untangling the ErbB signaling network, Nat. Rev. Mol. Cell Biol. 2, 127 (2001); Cel l 141, 1117 (2010)). When the first protein is selected, the selection of a protein involved in the downstream signal transduction pathway is not limited to the conventional method of the present invention. It is clear to those who have knowledge.
또한, 하나의 단백질이 다양한 생체 신호전달경로에 관여하여, 다수의 신호전달경로가 네트워크를 형성할 수 있음도 잘 알려져 있다. 따라서, 제 1 단백질이 2종 이상인 경우, 어느 하나의 제 1 단백질에 대한 제 2 단백질 중 하나 이상은 다른 제 1 단백질(들)에 대한 제 2 단백질들 중 하나이상과중복될수 있다(즉, 제 1단백질이 2개 이상인 경우, 이들 2개 이상의 제 1 단백질들에 대한 제 2 단백질들은 서로 다르거나, 일부 또는 전부가동일할수있다).  Also, it is well known that one protein participates in various bio-signal transmission pathways, and a plurality of signal transmission pathways can form a network. Thus, when two or more first proteins are present, one or more of the second proteins for any one first protein may overlap one or more of the second proteins for the other first protein (s) (i.e., 1 < / RTI > proteins, the second proteins for these two or more first proteins may be different, or some or all, the same).
일 예에서, 상기 제 1 단백질은 서로 다른 2 종 이상의 단백질이 복합체 (제 1 단백질-제 1 단백질 복합체)를 이루는 것일 수있다. 다른 예에서, 제 1 단백질과 제 2 단백질 간의 상호작용에 의하여 형성된 제 1 단백질-제 2 단백질 복합체가 2 이상 연결되어 (예컨대, 이웃하는 제 1 단백질-제 2 단백질 복합체의 제 2 단백질 간의 결합), 클러스터를 이루는 것일수있다.  In one example, the first protein may be a complex of two or more different proteins (first protein-first protein complex). In another example, two or more first protein-second protein complexes formed by interaction between a first protein and a second protein are linked (e.g., a bond between a second protein of a neighboring first protein-second protein complex) , And can be a cluster.
일 구체예에서, 상기 제 1 단백질은 EGFR, MET, HER2, 및 HER3로 이루어진 군에서 선택된 1종 이상일 수 있고, 각각의 제 1 단백질에 대한 제 2 단백질은 서로 같거나 다를 수 있으며, 각각 독립적으로, 포스포리파아제 C (PLC) (예컨대, PLC-gamma (PLC-gamma 1) (예컨대, GenBank Accession No. NP_002651.2, NP_877963.1, NP_037319.1등), 또는 이의 SH2 도메인(Src homology 2 domain: N-terminal SH2 domain 및 C- terminal SH domain 중 하나 이상을 포함; 예컨대, NP_037319.1 중의 545번째부터 765번째까지의 아미노산 서열 부위 또는 NP_002651.2 중의 540번째 또는 545번째부터 765번째까지의 아미노산 서열 부위 등), 성장인자 수용체 결합 단백질 (Growth factor receptor-binding protein: Grb; 예컨대, Grb2 (예컨대, GenBank Accession No. NP_002077.1 , NP_987102.1 등) 또는 이의 일부 (예컨대, SH2 도메인 (NP_002077.1의 In one embodiment, the first protein may be at least one selected from the group consisting of EGFR, MET, HER2, and HER3, and the second proteins for each first protein may be the same or different, , Phospholipase C (PLC) (e.g., PLC-gamma (PLC-gamma 1) (e.g., GenBank Accession No. NP_002651.2, NP_877963.1, NP_037319.1, etc.) or its SH2 domain (including at least one of Src homology 2 domain: N-terminal SH2 domain and C-terminal SH domain; For example, amino acid sequence regions 545 to 765 in NP_037319.1 or amino acid sequence regions 540 or 545 to 765 in NP_002651.2), growth factor receptor-binding protein Grb (e.g., Grb2 (see, for example, GenBank Accession No. 2). NP_002077.1, NP_987102.1, etc.) or a portion thereof (e.g., the SH2 domain (NP_002077.1
57번째부터 155번째까지의 아미노산 서열 부위), SH3_N-SH2 도메인 (NP_002077.1의 첫 번째부터 154번째까지의 아미노사 서열 부위), SH2- SH3_C 도메인 (NPJ302077.1의 57번째부터 217번째까지의 아미노산 서열 부위)), 포스파티딜이노시톨 3 -카이네이즈 조절 서브유닛(Amino acid sequence positions 57 to 155), SH3_N-SH2 domain (the first to 154th amino acid sequence regions of NP_002077.1), SH2- SH3_C domain (the 57th to 217th amino acids of NPJ302077.1 Amino acid sequence Site)), phosphatidylinositol 3-kinase modulating subunit
(Phosphat idyl inosi tol 3-kinase regul atory subuni t alpha; PIK3R1; p85- alpha; 예컨대, GenBank Accession No. NP_001229395.1, NP_852556.2, NP_852664.1, NP_852665.1, P26450.2 등) 또는 이의 SH2 도메인 (예컨대, NP_852664.1 (human p85a)의 SH2_N 도메인 (333번째부터 428번째까지의 아미노산서열 부위) , SH2_C도메인 (624번째부터 718번째까지의 아미노산 서열부위) , 또는 tandem SH2도메인 (333번째부터 718번째까지의 아미노산 서열 부위) ; 또는 P26450 (mouse p85a)의 SH2_N 도메인 (333번째부터 428번째까지의 아미노산 서열 부위) , SH2_C 도메인 (624번째부터 718번째까지의 아미노산서열 부위) 또는 tandem SH2도메인 (333번째부터 718번째까지의 아미노산 서열 부위)) 등으로 이루어진 군에서 선택된 1종 이상일수있으나, 이에 제한되는것은아니다. (For example, GenBank Accession No. NP_001229395.1, NP_852556.2, NP_852664.1, NP_852665.1, P26450.2, etc.) or its SH2 (SEQ ID NO: (Amino acid sequence region from 333rd to 428th), SH2_C domain (amino acid sequence region from 624th to 718th), or tandem SH2 domain (from the 333rd to the 428th amino acid sequence region) of NP_852664.1 (human p85a) (Amino acid sequence region from position 624 to position 718) or SH2_N domain (from position 333 to position 428) of P26450 (mouse p85a), SH2_C domain (amino acid sequence region from position 624 to 718), or tandem SH2 domain Amino acid sequence region from position 333 to position 718)), and the like, but the present invention is not limited thereto.
본명세서에 제공된구체예에서는, 폐암의 경우, 제 1단백질로 EGFR, MET, HER2, 및 HER3의 조합을 사용하고, 이들 제 1 단백질의 공통된 제 2 단백질로서 PLC-gamma 1, Grb2, 및 p85_alpha를 사용하였으며, 유방암의 경우, 제 1 단백질로 HER2 및 HER3의 조합을 사용하고, 이들 제 1 단백질의 공통된 제 2단백질로서 PLC-gamma 1, Grb2, 및 p85-alpha를사용하는 것을 예시하고 있으나, 이에 제한되는 것은 아니며, 앞서 설명한 내용에 기초하여 치료하고자하는 질병 또는 효과를 알고자하는 치료제에 따라서 적절하게 선택될 수 있고, 이는 이 발명이 속하는 기술 분야의 통상의 지식을가진자에게 명확한사항이다.  In the examples provided herein, the combination of EGFR, MET, HER2, and HER3 as the first protein and PLC-gamma 1, Grb2, and p85_alpha as the second common proteins of these first proteins In the case of breast cancer, a combination of HER2 and HER3 is used as the first protein and PLC-gamma 1, Grb2, and p85-alpha are used as common second proteins of these first proteins. The present invention is not limited thereto and can be appropriately selected according to the therapeutic agent to be sought based on the above-described diseases or effects to be treated, and it is obvious to those skilled in the art to which the present invention belongs.
다른 구체예에서, 상기 제 1 단백질은 EGFR, MET, HER2, 및 HER3로 이루어진 군에서 선택된 1종 이상일 수 있고、 제 1 단백질에 대한 제 2 단백질은 각각의 제 1 단백질에 결합하는 리간드들 중에서 선택된 것일 수 있다 (예컨대, EGFR의 경우 epidermal growth factor (EGF) , MET의 경우 hepatocyte growth factor (HGF) , HER2 및 HER3의 경우 Neuregul ins (NRGs)) .  In another embodiment, the first protein may be at least one selected from the group consisting of EGFR, MET, HER2, and HER3, and the second protein to the first protein may be selected from among ligands binding to the respective first protein (Eg, EGFR epidermal growth factor (EGF), MET hepatocyte growth factor (HGF), HER2 and HER3 Neuregul ins (NRGs)).
일 예에서, 제 1 단백질은 PD-L1 및 卵 -1 중에서 선택된 어느 하나이고, 제 2 단백질은 나머지 하나인 것을 특징으로 한다. 일 구체예에서, 제 1 단백질은卵 -1이고, 제 2 단백질은 PD-L1일 수 있다. 다른 구체예에서, 제 1단백질은 PD-L1이고, 제 2단백질은 PD-1일수있다.  In one example, the first protein is any one selected from PD-L1 and egg-1, and the second protein is the other. In one embodiment, the first protein is egg-1 and the second protein is PD-Ll. In another embodiment, the first protein is PD-L1 and the second protein is PD-1.
PD-L1 (Programmed death-l igand 1)은주로 암세포표면에 존재하는 단백질로, CD274 (cluster of di f ferent i at ion 274) 또는 B7_H1(B7 homolog 1)라고도 칭해지며, 40kDa의 타입 1 막통과 단백질 (type 1 transmembrane protein)이다. PD-L1은 암세포 이외에도 면역세포 (Macrophage 등)의 subtype에도 존재한다. PD-L1은 조직 이식, 면역 질환, 간염 등과 같은 다양한 질병에서 면역계를 억제하는데 주된 역할을 하는 것으로 알려져 있다. 본 명세서에 기재된 PD-L1은 인간 등의 포유 동물 유래의 것일 수 있으며 , 예컨대, 인간 PD-L1 (e.g. , GenBank Access ion No. NP_001254635.1 (유전자: NM_014143.3) , NP_001300958.1 (유전자: NM_001314029.1),PD-L1 (Programmed death-l igand 1) is a protein present on the surface of cancer cells. CD274 (cluster of diiferent ion 274) or B7_H1 (B7 homolog 1) and is a 40 kDa type 1 transmembrane protein. PD-L1 is present in subtypes of immune cells (Macrophage, etc.) as well as cancer cells. PD-L1 is known to play a major role in suppressing the immune system in various diseases such as tissue graft, immune diseases and hepatitis. The PD-L1 described in the present specification may be derived from a mammal such as a human, and examples thereof include human PD-L1 (eg, GenBank Accession No. NP_001254635.1 (gene: NM_014143.3), NP_001300958.1 NM_001314029.1),
NP_054862.1 (유전자: NM_001267706.1)등)일수있다. NP_054862.1 (gene: NM_001267706.1), etc.).
PD-1 (Programmed cel l death protein 1)은 CD279 (cluster of di f ferent iat ion 279)라고도 칭해지며, 활성화된 T세포 (면역세포)의 표면에 주로존재하는단백질로, 면역 체크포인트중하나이며, 림프절에서의 항원 특이적 T 세포 (ant i gen-speci f ic T_cel l)의 아팝토시스 (apoptos i s )를 촉진하고, 조절 T 세포 (regulatory T cel Is)의 아팝토시스는 감소시키는 역할을 한다. 또한, 암세포 표면에서 발현되어 mTOR 등의 신호전달 (signal ing pathway)을 통하여 암 증식 (tumor prol i ferat ion)을 촉진하기도한다. 본 명세서에 기재된 PD-1은 인간등의 포유동물유래의 것일 수 있으며, 예컨대, 인간 PD-1 (e.g. , GenBank Accession No. PD-1 (Programmed cell death protein 1), also called CD279 (cluster of di fi erent iat ion 279), is a protein that is mainly present on the surface of activated T cells (immune cells) and is one of the immune checkpoints , Apoptosis in antigen-specific T cells (lymphocytes) in the lymph nodes, and apoptosis in regulatory T cells (lymphocytes) do. In addition, it is expressed on the surface of cancer cells and promotes tumor proliferation through signaling pathway such as mTOR. PD-1 described herein may be of mammalian origin, such as human, and may be, for example, human PD-1 (e.g., GenBank Accession Nos.
NP_005009.2 (유전자: NM_005018.2)등)일수있다. NP_005009.2 (gene: NM_005018.2), etc.).
PD-L1이 T 세포 표면에 있는 PD-1 단백질과 결합하면, T 세포는 암세포에 대한공격능력을상실한다.  When PD-L1 binds to the PD-1 protein on the T cell surface, T cells lose their ability to attack cancer cells.
PD-L1 및/또는 卵 -1는 항암제 개발에 있어서 중요한 표적이 될 수 있다.  PD-L1 and / or egg-1 may be important targets in the development of anti-cancer drugs.
본 명세서에서 사용되는 제 1 단백질 및/또는 제 2 단백질은 세포, 조직, 또는 이들의 용해물 또는 추출물에 포함된 형태로 사용되거나, 정제된단백질형태로사용될수있다. As used herein, the first protein and / or the second protein may be used in a form contained in a cell, a tissue, or a lysate or an extract thereof, or in the form of a purified protein.
다른 예에서, 제 1 단백질은 HER2 (Human Epidermal growth factor Receptor 2 protein; ErbB2) 및 HER3 (Human Epidermal growth factor In another example, the first protein is HER2 (Human Epidermal Growth Factor Receptor 2 protein; ErbB2) and Human Epidermal Growth Factor
Receptor 3 protein; ErbB3)이며, 상기 HER2 및 HER3는 헤테로다이머를 이루는것 (HER2-HER3헤테로다이머)일수있다, Receptor 3 protein; ErbB3), and HER2 and HER3 may be heterodimers (HER2-HER3 heterodimers)
HER2 및 HER3는각각 HER패밀리의 수용체 티로신 키나아제 (RTKs)의 일원이다. HER2 또는 HER3의 세포외 도메인에 대한 리간드의 결합은 다른 ErbB 수용체와의 리셉터 호모- 또는 헤테로-이량체화를 유도하며, 이는 특이적인 타이로신 잔기의 세포내 자가인산화를 유발한다. HER2 및/또는 HER3의 자가인산화는 세포 증식, 혈관신생 및 전이에 영향을 미치는 MAPK 및 PI3K/Akt 활성화를 포함한 다운스트림 신호전달 네트워크를 이끈다. HER2 의 과발현, 유전자 증폭은 유방암과 위암에서 빈번히 관찰되며, 암 치료의 불량한 예후 및 나쁜 임상적 결과 등과 관련 있다. 한편 HER3는 여러 표적 약물 치료의 저항 기제로 잘알려져 있다. 이러한 이유로, 이들 HER2 및/또는 HER3는 항암 요법에 있어서 중요한 표적이 된다. HER2 및 HER3는 인간 등의 포유류로부터 유래된 것일 수 있다. 예컨대, HER2는 GenBank Accession No. NP_001005862.1, NP_001276865.1, NP_001276867.1, NP_004439.2 등에 제공되는 아미노산 서열을 갖는 폴리펩타이드일 수 있으나, 이에 제한되는 것은 아니다. HER3는 GenBank Accession No. NP_001005915.1, NP_001973.2 등에 제공되는 아미노산 서열을 갖는 폴리펩타이드일수있으나, 이에 제한되는것은아니다. HER2 and HER3 are members of the HER family receptor tyrosine kinases (RTKs), respectively. Binding of a ligand to the extracellular domain of HER2 or HER3 induces receptor homo- or hetero-dimerization with other ErbB receptors, which leads to intracellular autophosphorylation of specific tyrosine residues. HER2 and / or Autophosphorylation of HER3 leads to downstream signaling networks including MAPK and PI3K / Akt activation that affect cell proliferation, angiogenesis and metastasis. HER2 overexpression and gene amplification are frequently observed in breast and stomach cancer and are associated with poor prognosis of cancer treatment and poor clinical outcome. HER3, on the other hand, is well known as a resistance mechanism for several target drug therapies. For these reasons, these HER2 and / or HER3 are important targets in chemotherapy. HER2 and HER3 may be derived from mammals such as humans. For example, HER2 is GenBank Accession No. < RTI ID = 0.0 > NP_001005862.1, NP_001276865.1, NP_001276867.1, NP_004439.2, and the like, but is not limited thereto. HER3 was obtained from GenBank Accession No. NP_001005915.1, NP_001973.2, and the like, but are not limited thereto.
상기 제 2단백질은다양한생체신호전달경로에 있어서 HER2및/또는 HER3 (예컨대, HER2-HER3 헤테로다이머)의 하위 단백질로서, HER2 및/또는 HER3 (예컨대, HER2-HER3 헤테로다이머)와 상호작용 (결합) 가능한 단백질 중에서 선택된 1종 이상일 수 있다. 일 예에서, 상기 제 2 단백질은 HER2 및/또는 HER3 (예컨대, HER2-HER3 헤테로다이머)의 인산화기 (예컨대, 인산화된타이로신)에 결합하는것일수있다.  The second protein is a sub-protein of HER2 and / or HER3 (e.g., HER2-HER3 heterodimer) in various vital signal transduction pathways and interacts with HER2 and / or HER3 (e.g., HER2-HER3 heterodimer) ). ≪ / RTI > In one example, the second protein may bind to a phosphorylating group (e.g., phosphorylated tyrosine) of HER2 and / or HER3 (e.g., HER2-HER3 heterodimer).
본명세서에서 제공되는방법에서 수행되는제 1단백질과제 2단백질 간 단백질-단백질 상호작용을 측정하는 단계, 제 1단백질 또는 제 1단백질 효소의 활성화 정도를 측정하는 단계는 분리된 세포 또는 조직에 대하여 생체외 또는세포외 (in F/iro)에서 수행되는것일수있다. Measuring the protein-protein interaction between the first protein task 2 protein and the first protein task performed in the method provided in the present invention, measuring the degree of activation of the first protein or the first protein enzyme, Or in vitro or in vivo (in F / i ro).
상기 제 1 단백질과 제 2 단백질 간 단백질-단백질 상호작용을 측정하는단계는다음의 단계를포함할수있다:  Measuring the protein-protein interaction between the first and second proteins may comprise the following steps:
(1)제 1단백질을포함하는시험 시료를, 표면에 상기 제 1단백질에 특이적으로 결합하는물질을포함하는 기판에 가하여 제 1 단백질이 고정된 기판을준비하는단계;  (1) preparing a substrate to which a first protein is immobilized by adding a test sample containing a first protein to a substrate containing a substance that specifically binds to the first protein on the surface;
(2) 상기 준비된 제 1 단백질이 고정된 기판에 표지된 (표지 물질이 결합된)제 2단백질을첨가하여 반응시키는단계 ; 및  (2) adding a second protein labeled with the labeling substance to the substrate on which the prepared first protein is immobilized, and reacting; And
(3) 단계 (2)에서 얻어진 반응물로부터 신호를 즉정하는 단계 (3) immediately determining a signal from the reactant obtained in step (2)
(단백질-단백질상호작용측정) . ( Protein-protein interaction measurement ).
상기 제 1 단백질과 제 2 단백질 간 단백질-단백질 상호작용을 측정하는 단계는, 단계 (1) 내지 (3)에 더하여, (4) 단계 (3)에서 측정된 2019/132517 1»(:1^1{2018/016675 Wherein measuring the protein-protein interaction between the first protein and the second protein comprises, in addition to the steps (1) to (3), (4) 2019/132517 1 »(: 1 ^ {2018/016675
신호를 이용하여 제 1 단백질의 활성화 수준을 측정하는 단계를 추가로 포함할수있다. Signal to measure the level of activation of the first protein.
상기 단계 (3)에서 측정된 신호를 이용하여 제 1 단백질의 활성화 수준을 측정하는 단계 (4)는 단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한시험 시료내의 제 1단백질 단위량에 대한신호값을구하는 단계일수있다.  The step (4) of measuring the activation level of the first protein using the signal measured in the step (3) includes the step of measuring the activation level of the first protein in the test sample added in step (1) And obtaining a signal value for the unit amount.
일 예에서, 상기 단계 (4)는,  In one example, the step (4)
단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한 시험 시료 내의 제 1 단백질 단위량에 대한 신호값을 구하는 단계를 통하여 수행하거나,  A step of obtaining a signal value for the first protein unit amount in the test sample added in step (1) using the signal measured in step (3)
(4-1) 단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한 시험 시료의 단위량에 대한신호값을구하는단계 (단백질-단백질 상호작용 수준측정) ; 및  (4-1) obtaining a signal value for the unit amount of the test sample added in the step (1) (measuring the protein-protein interaction level) using the signal measured in the step (3); And
(4-2) 상기 단계 (4-1)에서 얻어진 시험 시료의 단위량에 대한 신호값을 이용하여 시험 시료에 포함된 제 1 단백질의 단위량에 대한 값을 구하는단계 (활성화수준측정)  (4-2) obtaining a value for the unit amount of the first protein contained in the test sample (signal level of activation level) using the signal value of the unit amount of the test sample obtained in the step (4-1)
를포함할수있다.  .
상기 측정된 제 1단백질과제 2단백질 간단백질-단백질 상호작용을 이용하여 소망하는 사항을 측정 (확인, 결정 또는 분석)하기 위하여, 상기 단계 ⑷ 이후에,  In order to measure (confirm, determine or analyze) a desired substance by using the measured protein-protein interaction between the first protein task 2 protein, after the step (4)
(5) 단계 (4)에서 얻어진 결과를 기준 시료에서 얻어진 결과와 비교하는단계를추가로포함할수있다.  (5) comparing the result obtained in the step (4) with the result obtained in the reference sample.
일 예에서, 상기 제 1 단백질은 ?^~11 및 卵 -1 중에서 선택된 어느 하나이고, 제 2단백질은
Figure imgf000021_0001
및 ?^~1중에서 선택된 다른하나일 수 있다. 또 다른 예에서 , 상기 제 1 단백질은 四묘2 - 1¾ 헤테로다이머일 수 있다. 이 경우, 四1?2 -四묘3 헤테로다이머에서의 他1?2 및/또는 四요3의 타이로신 카이네이즈 활성화 정도 (예컨대, 타이로신 잔기의 인산화)를 측정하는단계는다음의 단계를포함할수있다:
In one example, the first protein is any one selected from? - 11 and egg-1, the second protein is
Figure imgf000021_0001
And? ^ ~ 1. In another example, the first protein may be a quadrivalent 2 - 1, 3 heterodimer. In this case, the step of measuring the degree of tyrosine kinase activation (e.g., phosphorylation of the tyrosine residue) of the other 1? 2 and / or 4? 3 in the 4 1? 2? 4 thymine 3 heterodimer may include the following steps :
(1-1) 四요2 -他요3 헤테로다이머 또는 四1?2 -四묘3 헤테로다이머를 포함하는시료를, 표면에 四묘2및/또는他요3에 특이적으로결합하는물질을 포함하는기판에 가하여
Figure imgf000021_0002
헤테로다이머가고정된 기판을준비하는 단계;
(1-1) quaternary 2-hetero 3 dimer, or a sample comprising a 4 1 2-quaternary 3 heterodimer on a surface of a quartet 2 and / or other quaternary 3 To the included substrate
Figure imgf000021_0002
Preparing a substrate on which the heterodimer is immobilized;
(2-1) 상기 抑요2 -抑: 1?3 헤테로다이머를 인산화시키고, 표지된 (표지 물질이 결합된) 인산화된 Tyr특이적 결합물질 (예컨대, 항체)를 첨가하여 반응시키는단계 ; 및 (2-1) Suppression of the 2-depressant: 1? 3 heterodimer is phosphorylated and labeled Adding a phosphorylated Tyr-specific binding substance (e.g., an antibody) to which the substance is bound); And
(3-1) 단계 (2-1)에서 얻어진 반응물로부터 신호를 즉정하는 단계 (타이로신카이네이즈활성화측정) .  (3-1) Step (tyrosine kinase activation assay) of immediately determining a signal from the reaction product obtained in the step (2-1).
상기 단계 (2-1)에서, 인산화시키는 단계는 상기 기판 또는 상기 기판에 고정된 HER2-HER3 헤테로다이머에 인산화제를 처리하는 단계를 포함할 수 있다. 상기 인산화제는 HER2 및/또는 HER3를 인산화시킬 수 있는 모든 물질들 중에서 선택된 1종 이상일 수 있으며, 예컨대, ATP와 마그네슘 (예컨대, MgCl2 등)일 수 있으나, 이에 제한되는 것은 아니다. 상기 인산화전, 후, 또는동시, 및 단계 (3-1)의 활성화측정 전에, 상기 기판 또는 기판에 고정된 HER2-HER3 헤테로다이머에 DGTN, GDN 등을 포함하는 콜레스테롤 유사 세정제들 중에서 선택된 1종 이상의 세정제 (detergent)를 CMC(cri t ical mi cel l ar concentrat ion) 이상 또는 이를 초과하는 농도로 처리하여 HER2-HER3 헤테로다이머의 구조적 및/또는 기능적 활성을유지시키는것을특징으로할수있다. In the step (2-1), the phosphorylating step may include treating the phosphorylating agent to the HER2-HER3 heterodimer immobilized on the substrate or the substrate. The phosphate reagent may be a HER2 and / or can be at least one selected from every substances capable of HER3 phosphorylation, e.g., ATP and magnesium (e.g., MgCl 2, etc.), without being limited thereto. The method of claim 1, wherein, before, after, or simultaneously with the phosphorylation, and before the activation measurement of step (3-1), at least one selected from cholesterol-like detergents including DGTN, GDN and the like is added to the HER2- The detergent may be treated with a CMC (cortical microcrystalline) concentration or higher to maintain the structural and / or functional activity of the HER2-HER3 heterodimer.
제 1단백질이 HER2-HER3헤테로다이머인경우, 앞서 설명한단계 (1) 내지 (3)또는 (1-1)내지 (3-1)에 더하여, 다음의 단계 (4) , 및/또는단계 (5)를추가로포함할수있다.  In the case where the first protein is a HER2-HER3 heterodimer, in addition to the above-described step (1) to (3) or (1-1) to (3-1), the following step (4) and / ).
이하, 상기 단계를보다상세히 설명한다:  Hereinafter, the above steps will be described in more detail:
단계 (1)또는 (1-1): 제 1단백질이 고정화된기판준비 단계  Step (1) or (1-1): Substrate preparation step in which the first protein is immobilized
상기 단계 (1)은 제 1 단백질을 포함하는 시험 시료를 표면에 상기 제 1 단백질에 특이적으로 결합하는 물질을 포함하는 기판에 가하여 제 1 단백질이 고정된기판을준비하는단계이다.  The step (1) is a step of preparing a substrate to which a first protein is immobilized by adding a test sample containing a first protein to a substrate containing a substance that specifically binds to the first protein on the surface.
상기 제 1단백질은앞서 설명한바와같다.  The first protein is as described above.
상기 시험 시료는세포또는조직 내 및/또는세포또는조직 간의 신호전달경로의 활성화 제 1 단백질을 표적으로 하는 약물에 대한 반응성 등을시험하는데사용가능한모든생물시료일수있다.  The test sample may be any biological sample that can be used to test the response to a drug targeting the activating first protein in the cell or tissue and / or the signal transduction pathway between cells or tissues.
예컨대, 상기 시험 시료는개체로부터 분리된세포, 조직, 세포또는 조직의 용해물, 파쇄물, 또는 추출물, 체액 (예컨대, 혈액 (전혈, 혈장, 또는 혈청), 타액 등)일 수 있다. 상기 개체는 제 1 단백질이 관여하는 세포 또는 조직 내 및/또는 세포 또는 조직 간의 신호전달경로의 활성화 시험, 제 1 단백질을 표적으로 하는 약물에 대한 반응성 시험, 제 1 단백질 표적 치료에 적절한 대상인지 여부의 결정,1 단백질 표적 치료 효과의 모니터링, 및/또는 효과적인 제 1 단백질 표적 치료제의 선정 등을 수행하고자하는모든포유류(예컨대, 인간, 원숭이 등의 영장류, 마우스, 래트 등의 설치류 등)로 이루어진 군에서 선택될 수 있다. 일 예에서, 상기 개체는 제 1단백질과관련된 질병을 갖는환자일 수 있다. 상기 제 1 단백질과관련된 질병은 제 1 단백질의 과발현 또는 제 1 단백질이 관여하는 세포또는조직 내 및/또는세포또는조직 간의 신호전달경로의 활성화와 관련된 질병일 수 있으며, 예컨대, 암일 수 있다. 일 구체예에서, 상기 시험 시료는 개별 암환자 (예컨대, 제 1 단백질이 관여하는세포또는조직 내 및/또는 세포 또는 조직 간의 신호전달경로의 활성화 정도 또는 제 1 단백질을 표적으로 하는 약물에 대한 반응성을 시험하거나, 제 1 단백질 표적 치료에 적절한 대상인지 여부를 결정하거나, 또는 효과적인 제 1 단백질 표적 치료제의 선정하고자 하는 특정 개체)로부터 분리된 세포, 예컨대, 암 세포를 포함하는 것일 수 있다. 조직의 경우 분쇄를 위하여 최소 125 mm3이상의 크기이면족하고, 1회 측정에 필요한조직 시료의 양은 위에서 언급한 조직의 양(최소 125 mm3 이상)의 약 1/50 내지 약 1/75 범위로 할수 있으나, 이에 제한되는 것은 아니다. 세포 (암세포)의 경우, 1회 측정에 필요한세포 시료의 양은 약 10 cel l s 내지 1010 cel ls, 약 10 cel l s내지 107 cel ls, 약 10 cel l s내지 105 cel l s, 약 103 cel ls내지 1010 cel l s , 약 103 cel l s내지 107 cel ls, 약 103 cel l s내지 105 cel ls, 예컨대 약 104 + 50 cel ls- 정도일 일 수 있으나, 이에 제한되는 것은 아니며, 세포주의 종류에 따라서 적절하게 정해질수있는값이다. For example, the test sample may be a lysate, a lysate, or an extract of a cell, a tissue, a cell or a tissue isolated from an individual, a body fluid (e.g., blood (whole blood, plasma or serum), saliva, etc.). The subject is preferably selected from the group consisting of an activation test of a signal transduction pathway in a cell or tissue and / or a cell or tissue involved in the first protein, a reactivity test for a drug targeting the first protein, of the crystal, the first protein target therapeutic effect (For example, primates such as human, monkey, rodents such as mice, rats, etc.) which are to be used for monitoring and / or selecting effective first protein target therapeutic agents. In one example, the subject may be a patient having a disease associated with the first protein. The disease associated with the first protein may be a disease associated with overexpression of the first protein or activation of a signal transduction pathway in the cell or tissue and / or between the cell or tissue involved in the first protein, for example, the cancer. In one embodiment, the test sample is used to determine the degree of activation of the signal transduction pathway in a particular cancer patient (e. G., The cell or tissue involved by the first protein and / or between the cell or tissue, For example, a cancer cell, to determine whether it is an appropriate subject for a first protein target treatment, or to isolate a specific entity from which an effective first protein target therapeutic agent is to be selected). In the case of tissue, a size of at least 125 mm 3 is sufficient for grinding, and the amount of tissue sample required for one measurement may be in the range of about 1/50 to about 1/75 of the amount of tissues mentioned above (at least 125 mm 3 ) But is not limited thereto. In the case of cells (cancer cells), the amount of cellular sample required for one-time measurement of about 10 cel ls to 10 10 cel ls, of about 10 cel ls to 10 7 cel ls, of about 10 cel ls to 10 5 cel ls, of about 10 3 cel ls to 10 10 cel ls, of about 10 3 cel ls to 10 7 cel ls, of about 10 3 cel ls to 10 5 cel ls, for example, may be an order of about 10 4 + 50 cel ls-, it is not limited to, cell lines Which is a value that can be appropriately determined according to the type of the film.
일 예에서, 제 1 단백질을 표적으로 하는 약물에 대한 반응성 예측 및/또는 제 1 단백질 표적 치료에 적절한 대상의 선별 방법의 경우, 상기 시험 시료는 제 1 단백질을 표적으로 하는 치료 (예컨대, 제 1 단백질을 표적으로 하는 약물의 투여)가수행되지 않은 세포 또는 조직, 또는 상기 치료 (또는 약물의 투여)가 수행되지 않은 개체로부터 분리된 세포 또는 조직일 수 있으며, 제 1 단백질을 표적으로 치료에 대한 반응성을 모니터링하는 방법의 경우, 상기 시험 시료는 제 1단백질을 표적으로 하는 치료 (예컨대, 제 1 단백질을 표적으로 하는 약물의 투여)가 수행된 세포 또는 조직, 또는 상기 치료 (또는 약물의 투여)가 수행된 개체로부터 분리된세포또는조직일수있다.  In one example, in the case of prediction of responsiveness to a drug targeting the first protein and / or selection of a subject suitable for treatment of the first protein target, the test sample may comprise a treatment that targets the first protein (e.g., Administration of a drug targeting the protein) may be a cell or tissue that has not been subjected to an action, or a cell or tissue that has been separated from a subject in which the treatment (or administration of the drug) has not been performed, In the case of a method for monitoring the reactivity, the test sample may be a cell or a tissue on which the first protein-targeting treatment (for example, the administration of the drug targeting the first protein) has been performed or the treatment (or administration of the drug) Lt; RTI ID = 0.0 > isolated < / RTI >
상기 기판은 표면에 제 1 단백질을 고정시킬 수 있는 모든 재질 및/또는모든구조를 갖는 것일 수 있다 (결정질 또는 비결정질 모두사용 2019/132517 1»(:1^1{2018/016675 The substrate may be of any material and / or of any structure capable of immobilizing the first protein on its surface (both crystalline and amorphous 2019/132517 1 »(: 1 ^ {2018/016675
가능함). 일 예에서, 상기 기판은, 표지 신호의 검출용이성을고려하여, 빛의 굴절률이 생체 물질의 많은 부분을 차지하는 물의 굴절률 (약 1.3) 이상인 재질일수있다. 일 예에서, 상기 기판은두께가약 0.1내지 약 1 _, 약 0.1내지 약 0.5■, 0.1내지 약 0.25 1®, 또는 약 0.13내지 약 0.21 정도일 수 있으며, 굴절률이 약 1.3 내지 약 2, 약 1.3 내지 약Available). In one example, the substrate may be a material having a refractive index of water equal to or higher than that of water (about 1.3), which occupies a large portion of the biomaterial, taking into consideration the easiness of detecting a beacon signal. In one example, the substrate thickness reducible 0.1 to about 1 _, from about 0.1 to about 0.5 ■, 0.1 to about 0.25 1®, or about 0.13 to about 0.21, and be on the order of a refractive index of about 1.3 to about 2, about 1.3 About
1.8, 약 1.3내지 약 1.6, 또는약 1.5내지 약 1.54정도인 것일수 있다. 상기 기판은 상기 굴절률 범위를 만족시키는 모든 재질일 수 있으며, 예컨대 유리 (굴절률: 약 1.52), 석영 등으로 이루어진 군에서 선택된 재질로부터 얻어진 것일 수 있으나, 이에 제한되는 것은 아니다. 상기 기판은 생물 시료 관찰에 통상적으로 사용되는 모든 형태를 갖는 것일 수 있으며, 예컨대, 웰 타입, 슬라이드 타입, 채널 타입, 어레이 형태, 미세유체칩, 미세관 (캐필러리) 등일 수 있으나, 이에 제한되는 것은 아니다. 형광현미경 관찰시, 상기 시료가적용된 기판위에 커버글라스를 장착하여 관찰할 수 있으며, 커버글라스의 재질은 앞서 기판에서 설명한 바와 같고, 두께는 기판에서 설명한 범위 또는 이보다 얇을 수 있다 (예컨대, 굴절률 1.52, 두께 0.17·일 수 있으나이에 제한되는것은아님). 상기 제 1 단백질에 특이적으로 결합하는 물질은 제 1 단백질과 특이적으로 결합할 수 있는 모든 물질로부터 선택된 것일 수 있으며, 예컨대, 제 1 단백질에 특이적으로 결합하는 항체 또는 이의 항:원결합단편 (예컨대, 항체의 1 , ( ) 산 -!7。, Fab , 1 및 ( ’)2 등), 압타머 (단백질 또는 핵산분자), 소분자 화합물 등으로 이루어진 군에서 선택된 1종 이상일 수 있다. 이 때, 상기 제 1 단백질에 특이적으로 결합하는 물질은 제 1 단백질과 제 2 단백질과의 상호작용을 방해하지 않는 부위, 즉, 제 1 단백질과 제 2 단백질이 상호작용(결합)하는 부위가 아닌 부위에서 제 1단백질과결합하는것일수있다. 1.8, about 1.3 to about 1.6, or about 1.5 to about 1.54. The substrate may be any material that satisfies the refractive index range and may be, for example, from a material selected from the group consisting of glass (refractive index: about 1.52), quartz, and the like, but is not limited thereto. The substrate may be of any type conventionally used for biological specimen observation and may be, for example, a well type, a slide type, a channel type, an array type, a microfluidic chip, a microtubule (capillary) It is not. When observing a fluorescent microscope, a cover glass can be observed on a substrate to which the sample is applied. The material of the cover glass is as described above for the substrate, and the thickness may be the range described in the substrate or may be thinner (for example, a refractive index of 1.52, 0.0 > 0.17, < / RTI > but not limited thereto). Substance specifically binding to the first protein can be selected from any substance capable of binding to the first protein and specifically, for example, a specific antibody or anti thereof that binds to the first protein: One-binding fragment (E.g., 1, () acid-! 7 , Fab, 1 and (2) 2), platamer (protein or nucleic acid molecule), small molecule compound, and the like. At this time, the substance specifically binding to the first protein is a site that does not interfere with the interaction between the first protein and the second protein, that is, a site where the first protein and the second protein interact (bind) And may bind to the first protein at a non-specific site.
일 예에서, 상기 기판은 상기 제 1 단백질에 특이적으로 결합하는 생물학적 물질 (예컨대, 항체 등)을 표면에 포함(고정)하기 위하여 적절히 표면개질되거나, 표면에 직접 상기 제 1 단백질에 특이적으로 결합하는 물질이 고정된 것일수 있다. 상기 기판이 표면개질된 경우, 상기 기판은 일면에 상기 제 1 단백질에 특이적으로 결합하는 생물학적 물질 (예컨대, 항체 등)을고정화시킬수 있는작용기를갖는모든화합물로처리(예컨대, 도포)될수 있으며, 예컨대, 알데히드기, 카르복실기 및 아민기로이루어진 군에서 선택된 작용기를포함하는화합물로처리될수 있다. 일 구체예에서, 상기 알데히드기, 카르복실기 및 아민기로 이루어진 군에서 선택된 작용기를 포함하는 화합물은 바이오틴 (biotin), 소혈청알부민 (Bovine serum albumin; BSA) , 바이오틴이 결합된 소혈청알부민, 폴리에틸렌글리콜 (polyethylene glycol; PEG) , 바이오틴이 결합된 PEG (폴리에틸렌글리콜-바이오틴; PEG-biot in) , 폴리소베이트 (e.g. , Tween20) 등으로 이루어진 군에서 선택된 1종 이상일 수 있으나 이에 제한되는 것은 아니다. 상기 표면 처리된 기판은 뉴트라비딘 (neutravidin) , 스트렙타비딘 (streptavidin) , 아비딘 (avidin) 등으로 이루어진 군에서 선택된 1종이상이 추가로처리 (예컨대, 도포)될수있다. In one example, the substrate is suitably surface-modified to include (immobilize) a biological substance (for example, an antibody or the like) that specifically binds to the first protein on the surface, or may be surface- The binding material may be fixed. When the substrate is surface-modified, the substrate may be treated (e.g., coated) with all of the compounds having a functional group capable of purifying a biological substance (for example, an antibody or the like) that specifically binds to the first protein on one surface, For example, a compound containing a functional group selected from the group consisting of an aldehyde group, a carboxyl group and an amine group. In one embodiment, The compound containing a functional group selected from the group consisting of an aldehyde group, a carboxyl group and an amine group may be selected from the group consisting of biotin, bovine serum albumin (BSA), bovine serum albumin bound with biotin, polyethylene glycol (PEG) Biotin-conjugated PEG (polyethylene glycol-biotin), polysorbate (e.g., Tween 20), and the like, but the present invention is not limited thereto. The surface-treated substrate may be further treated (e.g., coated) with at least one selected from the group consisting of neutravidin, streptavidin, avidin, and the like.
일 예에서, 상기 제 1 단백질은 PD-L1 또는 卵 -1일 수 있다. 구체적으로, 상기 제 1단백질은 PD-L1일수 있다. 제 1단백질로서 PD-L1를 사용하여, PD-L1와 특이적으로 결합하는 물질 (예컨대, 항체)를 이용하여 PD-L1를 기판에 고정화시켜 단백질-단백질 상호작용을 측정하는 경우, PD- 1이 cluster ing된 스팟이 형성되어, 정성적 및/또는 정량적 분석에 유리한 효과를얻을수있다.  In one example, the first protein may be PD-L1 or egg-1. Specifically, the first protein may be PD-L1. When protein-protein interactions are measured by immobilizing PD-L1 on a substrate using a substance (such as an antibody) that specifically binds to PD-L1 using PD-L1 as the first protein, PD- This clustered spot is formed, which has the advantageous effect of qualitative and / or quantitative analysis.
다른 예에서, 상기 제 1 단백질은 PD-1일 수 있다. 제 1 단백질로서 PD-1를 사용하여, 卵 -1와 특이적으로 결합하는 물질 (예컨대, 항체)를 이용하여 PD-1를 기판에 고정화시켜 단백질-단백질 상호작용을 측정하는 경우상대적으로단일 스팟으로관찰되어, 단일 스팟검출이 필요한분석에 유리한효과를얻을수있다.  In another example, the first protein may be PD-I. When protein-protein interactions are measured by immobilizing PD-1 on a substrate using a substance (for example, an antibody) that specifically binds to egg-1 using PD-1 as a first protein, So that a favorable effect can be obtained in the analysis requiring a single spot detection.
상기 시험 시료는세포또는조직 간의, PD-L1와卵 -1간의 상호작용 수준, 제 1 단백질이 관여하는 신호전달경로의 활성화 여부, 제 1 단백질을 표적으로 하는 약물에 대한 반응성 등을 시험하는데 사용 가능한 모든 생물학적 시료일수있다.  The test sample is used to test the level of interaction between PD-L1 and egg-1, the activation of the signal transduction pathway involved in the first protein, and the reactivity to the drug targeting the first protein, among cells or tissues It may be all possible biological samples.
또한, 상기 PD-L1과 PD-1 (예컨대 이종세포표면에 위치)간결합에 의하여 형성된복합체는클러스터를이루어, 생체 내 환경을보다유사하게 모사할수 있다. 생체 내 환경과보다유사한클러스터를형성하기 위하여, 기판에 고정화되는제 1단백질로서 PD-L1을선택하여 사용하는 것이 유리할 수있다.  In addition, complexes formed by binding between PD-L1 and PD-1 (for example, on the surface of xenogeneic cells) are clusters, and the in vivo environment can be simulated more similarly. In order to form clusters more similar to the in vivo environment, it may be advantageous to select PD-L1 as the first protein to be immobilized on the substrate.
일 예에서, 제 1 단백질로서 PD-L1을 사용하는 경우, 상기 제 1 단백질에 .특이적으로 결합하는 물질은 PD-L1에 특이적으로 결합하는 항체 또는 이의 항원결합단편일 수 있다. PD-L1와抑- 1 간의 상호결합에 영향이 없도록 하기 위하여, 상기 PD-L1에 특이적으로 결합하는 항체 또는 항원결합단편은 PD-L1의 PD-1과의 결합 부위를 제외한 (결합 부위와 중복하지 않는) 비결합 부위, 예컨대, PD-L1의 C-말단 부위에 특이적으로 결합하는 것일 수 있다. PD-L1이 PD-1과 결합하는부위는 대체적으로 PD- L1의 N-말단쪽 부위(세포외 도메인 부위)이며, 구체적으로, 20번째부터 150번째까지, 20번째부터 130번째까지, 20번째부터 125번째까지,In one example, the case of using the PD-L1 as a first protein, a substance that binds to the first protein may be a specific antibody or an antigen-binding fragment that specifically binds to PD-L1. In order not to affect the mutual binding between PD-L1 and suppress-1, the antibody specifically binding to PD-L1 or The antigen-binding fragment may specifically bind to a non-binding site (for example, does not overlap with the binding site) of the PD-L1 binding site, for example, the C-terminal site of PD-L1. The site where PD-L1 binds to PD-1 is generally the N-terminal region (extracellular domain site) of PD-L1, and specifically, the 20th to 150th, 20th to 130th, To 125th,
25번째부터 200번째까지, 25번째부터 150번째까지, 25번째부터 130번째까지, 또는 25번째부터 125번째까지의 아미노산 부위에서 선택된 하나 이상의 아미노산 잔기를 포함하는 부위일 수 있으며, 예컨대, PD-L1의 D26, Y56, E58, R113, A121, D122, Y123, R125 등으로 이루어진 군에서 선택된 하나 이상의 아미노산 잔기 (GenBank Accession No. NP_001300958.1,25 to 150, 25 to 130, or 25 to 125 amino acid residues, and examples thereof include PD-L1 At least one amino acid residue selected from the group consisting of D26, Y56, E58, R113, A121, D122, Y123, and R125. NP_001300958.1,
NP_054862.1 등의 인간 PD-L1 기준) 또는 이에 해당하는 잔기를 포함하는 부위일수 있다(도 32참조). 상기 PD-L1의 PD-1과의 결합부위를제외한 비결합부위는, 앞서 설명한 PD-L1의 N 말단쪽의 PD-L1가 PD-1과 결합하는 부위의 와 중복되지 않는 PD-L1 부분일 수 있으며, 상기 PD-L1에 특이적으로 결합하는 항체 또는 이의 항원결합단편은 상기 PD-L1의 PD-Human PD-L1 reference such as NP_054862.1) or a corresponding site (see Fig. 32). The non-binding site except for the binding site of PD-L1 with PD-1 is a PD-L1 site in which the N-terminal PD-L1 of the PD-L1 described above does not overlap with the site binding to PD- And the antibody or antigen-binding fragment thereof that specifically binds to PD-L1 binds to the PD-
1과의 결합 부위를 제외한 비결합 부위 (예컨대, PD-L1의 C-말단 부위) 중의 1개 이상, 예컨대, 1내지 30개, 1 내지 25개, 1내지 20개, 1 내지 15개, 또는 1내지 10개의 아미노산에 결합하는항체 또는항원결합단편일 수 있다. 일 예에서, 상기 PD-L1에 특이적으로 결합하는 항체 또는 이의 항원결합단편은 PD-L1 단백질의, N-말단에서 C-말단 방향으로, 250번째 이후, 260번째 이후, 270번째 이후, 또는 280번째 이후의 아미노산 부위, 예컨대, 250번째부터 300번째까자 또는 280번째부터 290번째까지의 아미노산 부위 중 선택된 하나 이상의 .아미노산에 특이적으로 결합하는 것일수있다. 1 to 30, 1 to 25, 1 to 20, 1 to 15, or 1 to 20 of the non-binding sites (for example, the C-terminal sites of PD-L1) An antibody or an antigen-binding fragment which binds to 1 to 10 amino acids. In one example, the antibody or antigen-binding fragment thereof that specifically binds to PD-L1 binds to the PD-L1 protein in the N-terminal to C-terminal direction, after the 250th, after the 260th, The amino acid sequence may be one that specifically binds to one or more amino acids selected from the amino acid positions after the 280th position, for example, from the 250th position to the 300th position, or from the 280th position to the 290th position.
다른 예에서, 제 1 단백질로서 PD-1을 사용하는 경우, 상기 제 1 단백질에 특이적으로 결합하는 물질은 卵 -1에 특이적으로 결합하는 항체 또는 이의 항원결합단편일 수 있다. PD-1와 PD-L1 간의 상호결합에 영향이 없도록 하기 위하여, 상기 PD-1에 특이적으로 결합하는 항체 또는 항원결합단편은 抑 -L의 PD-L1과의 결합 부위를 제외한 (결합 부위와 중복하지 않는) 비결합 부위, 예컨대, 抑- 1의 C-말단 부위에 특이적으로 결합하는 것일 수 있다. 상기 PD-1이 PD-L1과결합하는부위는 대체적으로 卵 -1의 N-말단쪽 부위(세포외 도메인 부위)이며, 구체적으로 60번째부터 200번째까지, 60번째부터 150번째까지, 60번째부터 135번째까지, 65번째부터 200번째까지, 65번째부터 150번째까지, 또는 65번째부터 135번째까지의 아미노산 부위에서 선택된 하나 이상의 아미노산 잔기를 포함하는 부위일 수 있으며, 예컨대, 卵 -1의 Y68, Q75, K78, T76, 1126, 1134 등으로 이루어진 군에서 선택된 하나 이상의 아미노산 잔기를 포함하는 부위일 수 있다. 상기 PD-L의 PD-L1과의 결합 부위를 제외한 비결합 부위, 예컨대, PD-1의 C-말단 부위는, 앞서 설명한 卵 -1의 N 말단쪽의 PD-1가 PD-L1과 결합하는 부위와 중복되지 않는 PD-1 부분일 수 있으며, 상기 PD-1에 특이적으로 결합하는 항체 또는 이의 항원결합단편은 상기 PD-1의 PD-L1과의 결합 부위를 제외한 비결합 부위 (예컨대, PD-1의 C-말단부위) 중의 1개 이상, 예컨대, 1 내지 30개, 1 내지 25개, 1내지 20개, 1 내지 15개, 또는 1 내지 10개의 아미노산에 결합하는 항체 또는 항원결합단편일수있다. In another example, when PD-1 is used as the first protein, the substance specifically binding to the first protein may be an antibody or an antigen-binding fragment thereof that specifically binds to egg-1. In order to prevent mutual binding between PD-1 and PD-L1, the antibody or antigen-binding fragment that specifically binds to PD-1 binds to the binding site of PD- Binding site at the C-terminal site of the repressor-1, i.e., not overlapping the C-terminal site of the repressor-1. The site where PD-1 binds to PD-L1 is generally the N-terminal region (extracellular domain site) of egg-1, and specifically the 60th to 200th, 60th to 150th, 60th From the 135th to the 135th, 65 to 150, or 65 to 135 amino acid residues, and examples thereof include a region containing at least one amino acid residue selected from the group consisting of Y68, Q75, K78, T76, 1126, 1134, and the like. The non-binding site other than the binding site of PD-L with PD-L1, for example, the C-terminal site of PD-1, is a site in which the PD-1 at the N- And the antibody or antigen-binding fragment thereof that specifically binds to PD-1 may be a non-binding site other than the binding site of PD-1 with PD-L1 (for example, 1 to 30, 1 to 25, 1 to 20, 1 to 15, or 1 to 10 amino acids in the C-terminal region of PD-1) Can be.
다른 예에서, 상기 제 1 단백질이 HER2 및 HER3가 이합체화된 HER2- HER3 헤테로다이머일 수 있다. 이 경우, 상기 제 1 단백질을 포함하는 시료는 HER2 및/또는 HER3, 또는 HER2-HER3 헤테로다이머를 포함하거나, In another example, the first protein may be a HER2-HER3 heterodimer with HER2 and HER3 dimerized. In this case, the sample containing the first protein may comprise HER2 and / or HER3, or a HER2-HER3 heterodimer,
HER2 및 HER3를 동시에 발현하는 세포, 세포 용해물, 또는 세포 파쇄물을 포함하는 것으로, 시험 대상 환자로부터 분리된 세포, HER2와 HER3를 동시에 발현하도록 조작된 재조합 세포 (예컨대, HER2를 (과)발현하는 세포에 HER3유전자를포함하는재조합벡터가도입된 재조합세포, HER3를 (과)발현하는세포에 HER2유전자를포함하는재조합벡터가도입된 재조합 세포등), 또는상기 세포의 용해물또는파쇄물일수있다. A cell isolated from a patient to be tested comprising recombinant cells expressing HER2 and HER3, a cell lysate, or a cell lysate, a recombinant cell engineered to express HER2 and HER3 simultaneously (for example, a recombinant cell expressing HER2 A recombinant cell into which a recombinant vector containing a HER3 gene is introduced into a cell, a recombinant cell into which a recombinant vector containing a HER2 gene is introduced into a cell expressing HER3, etc.), or a lysate or a lysate of the cell.
제 1 단백질이 HER2-HER3 헤테로다이머인 경우, HER2와 HER3의 헤테로다이머 형성을 위하여, HER2와 HER3를 동시에 포함하거나 발현하는 세포에 이합체화 제제를 처리하는 것을 특징으로 할 수 있다. 상기 이합체화 제제는 HER3의 리간드 (예컨대, Neuregulin pi (NRGbl); When the first protein is a HER2-HER3 heterodimer, the heterodimer of HER2 and HER3 may be characterized by treating the dimerization agent with cells that simultaneously express or express HER2 and HER3. The dimerization agent may be a ligand of HER3 (e.g., Neuregulin pi (NRGbl);
NP_039250, NP_039250.2, 등), 콜레스테롤-유사 세정제 (Cholesterol-like detergent; 예컨대, DGTN(digitonin; CAS Number : 11024-24-1) , GDNNP_039250, NP_039250.2, etc.), cholesterol-like detergent (e.g., DGTN (digitonin; CAS Number: 11024-24-1), GDN
(glycol-diosgenin; CAS Number: 1402423-29-3) 등) 등으로 이루어진 군에서 선택된 1종이상일수 있다. 일구체예에서 , HER2와 HER3를동시에 발현하는 세포에 NRGbl을 처리(주입)하여 HER2-HER3 헤테로다이머를 형성하는 경우, 상기 NRGbl는 HER2와 HER3를 발현 후 용해(lysis) 전에 세포에 주입될 수 있다. 다른 구체예에서, HER2와 HER3를 동시에 발현(포함)하는 세포에 콜레스테롤-유사 세정제를 처리하여 HER2-HER3 해테로다이머를 형성하는 경우, 상기 콜레스테롤-유사 세정제는 세쏘 용해 후 및 HER2 및/또는 HER3의 활성화 측정 전에 처리 (첨가)될 수 있다 (일 예에서, 상기 세포는 NRGbl가 10ng/mL 내지 lOOOng/mL, 50ng/ mL 내지 200ng/mL, 또는 10ng/mL 내지 100ng/mL 의 양으로 주입된 것일 수 있음) . 이 때, 사용되는 콜레스테롤-유사 세정제의 농도는 사용되는 세정제의 CMCCcr i t i cal mi cel lar concent rat ion)을 초과하는 농도일 수 있으며, 예컨대, 세포 용해물 (cel l lysi s) 전체를 기준으로, DGTN의 경우 0.05%(w/v) 이상, 0.05%(w/v) 초과, 또는 0.06%(w/v) 이상, 예컨대, 0.05 내지 2%(w/v) 또는 0.06내지 2¾>(w八)일 수 있고, GDN의 경우 0.003%(w八 0 이상, 0.005%(w ) 이상, 0.007%(w八 0 이상, 또는 0.01%(w/v) 이상, 예컨대, 0.003내지 2%(w/v) , 0.005내지 2%(w/v) , 0.007내지 2%(w/v) , 또는 0.01 내지 2¾>(w八 0일 수 있다. 사용되는 세정제의 농도가 CMC 이하인 경우 mi cel le 구조의 유지, 단백질의 세포막 투과 영역의 보호, 및 tyrosine kinase act ivi ty유지가어렵고, HER2-HER3헤테로다이머의 활성도 (예컨대, 인산화정도)가낮아지고복구되지 않음이 확인되어, 세정제는상기 농도로 사용할 것을 제안한다. Digi tonin은 용해시 상당히 불안정해지며, 수 시간 이내에 침전물이 생긴다. 따라서 용해시 10%(w/v) 내외 (5 내지 15%(w/v) 또는 8내지 12%(w八 0)의 glycerol을 첨가하면좀더 안정적으로유지된다. GDN은 용해해도 수 일 동안 안정하다. 모든 용액은 HEPES 버퍼를 이용해 pH를 7 내지 8. 7,2 내지 7.6, 또는 7.4로 유지할 수 있다. 또 NaCl(Glycol-diosgenin; CAS Number: 1402423-29-3)), and the like. In one embodiment, when NRGbl is treated (injected) into HER2 and HER3 expressing cells to form a HER2-HER3 heterodimer, the NRGbl may be injected into the cell prior to lysis after HER2 and HER3 expression have. In another embodiment, a cell that simultaneously expresses HER2 and HER3 (including) a cholesterol-like detergent to treat HER2-HER3 In the case of forming a heterodimer, the cholesterol-like cleansing agent may be treated (added) after cesiolysis and prior to measurement of activation of HER2 and / or HER3 (in one example, the cell has an NRGbl of between 10 ng / mL, 50 ng / mL to 200 ng / mL, or 10 ng / mL to 100 ng / mL). In this case, the concentration of the cholesterol-like detergent to be used may be in excess of the concentration of the cleaning agent used, for example, the concentration of the cholesterol-like detergent may be, for example, for DGTN 0.05% (w / v) or more, 0.05% (w / v) in excess, or 0.06% (w / v) or more, e.g., 0.05 to 2% (w / v) or 0.06 to 2¾> (w八), 0.003% (w or more, 0.005% (w) or more, 0.007% (w or more), or 0.01% (w / v) or more, such as 0.003 to 2% v), 0.005 to 2% (w / v), 0.007 to 2% (w / v), or 0.01 to 2¾ > ( w 8 0) It is difficult to maintain the membrane permeability of the protein and maintain the tyrosine kinase activity, and the activity of the HER2-HER3 heterodimer (for example, phosphorylation degree) is lowered and is not restored. Thus, suggestion Digi tonin is highly unstable at dissolution and precipitates within a few hours, so it is recommended that 10 to 20% (w / v) or 8 to 12% (w / v) GDN is stable for several days even when dissolved GDN is stable for several days All solutions can be maintained at pH 7 to 8.7, 2 to 7.6, or 7.4 using HEPES buffer, and NaCl
150mM을 첨가해 적절한 이온농도를유지해주어야 한다. 세포 용해시 세포 내 protease에 의한 단백질 분해를 막기 위해 protease inhibi tor cocktai l을첨가할수있다 (예컨대 , Sigmaaldr i ch P8340 , 약 2%(w/v)) . 150 mM should be added to maintain proper ion concentration. To inhibit protein degradation by intracellular proteases during cell lysis, protease inhibi tor cocktail can be added (eg, Sigma-Aldrich P8340, approximately 2% (w / v)).
제 1 단백질이 HER2-HER3 헤테로다이머인 경우, 상기 저 U 단백질에 특이적으로 결합하는 물질은 제 1 단백질, 즉, HER2, HER3, 또는 이들 모두와특이적으로결합할수 있는모든물질로부터 선택된 것일 수 있으며, 예컨대, HER2, HER3, 또는 이들 모두에 특이적으로 결합하는 항체 또는 이의 항원결합단편 (예컨대, 항체의 scFv, (scFv)2, scFv-Fc, Fab, Fab' 및 F(ab' )2 등), 압타머 (단백질 또는 핵산분자) , 소분자 화합물 등으로 이루어진 군에서 선택된 1종 이상일 수 있다. 、 이 때, 상기 제 1 단백질 (則: R2, HER3, 또는 이들 모두)에 특이적으로 결합하는 물질은 제 1 단백질과 제 2 단백질과의 상호작용을 방해하지 않는 부위, 즉, 제 1 단백질과 제 2 단백질이 상호작용 (결합)하는 부위가 아닌 부위에서 제 1 단백질과결합하는것일수있다. When the first protein is a HER2-HER3 heterodimer, the substance specifically binding to the low U protein may be selected from the first protein, i.e., HER2, HER3, or any substance capable of specifically binding to all of them (ScFv) 2, scFv-Fc, Fab, Fab 'and F (ab') 2 (SEQ ID NO: 2) of an antibody that specifically binds to HER2, HER3, Etc.), platamer (protein or nucleic acid molecule), small molecule compound, and the like. , A substance that specifically binds to the first protein (rule: R2, HER3, or both of them) is a region that does not interfere with the interaction between the first protein and the second protein, that is, In the region where the second protein does not interact (bind), the first It can be associated with proteins.
단계 (2): 제 1단백질과제 2단백질을반응시키는단계  Step (2): Step of reacting the first protein task 2 protein
상기 단계 (2)는, 상기 준비된 제 1단백질이 고정된 기판에, 표지된 제 2단백질을첨가하여 반응시키는단계이다.  In the step (2), the labeled second protein is added to the substrate on which the prepared first protein is immobilized and reacted.
상기 제 2단백질은앞서 설명한바와같다.  The second protein is as described above.
상기 표지된 제 2 단백질은 제 2 단백질이 검출 가능한 신호를 발생시키는표지 물질로표지되거나 (표지 물질이, 예컨대, 화학적 (예컨대, 공유적 또는 비공유적) , 재조합적, 또는 물리적으로, 결합되거나), 표지물질이 결합될 수 있는 tag이 부착된 형태를 의미할 수 있다. 상기 검출 가능한 신호는 통상적인 효소 반응, 형광, 발광, 및/또는 방사선 검출을 통하여 측정될 수 있는 모든 신호 (예컨대, 빛, 방사선 등)들 중에서 선택될수 있다. 상기 표지 물질은상기 표지 신호를발생시킬 수 있는 모든 소분자 화합물, 단백질, 펩타이드, 핵산분자 등으로 이루어진 군에서 선택된 1종 이상일 수 있으며, 예컨대, 형광 염료 (소분자화합물; Cyanine, Alex, Dyl ight , Fluoprobes 등), 형광 단백질 (예컨대, 녹색형광단백질 (GFP, enhanced GFP) , 황색형광단백질 (YFP) , 청록색형광단백질 (CFP) , 청색형광단백질 (BPF) , 적색형광단백질 (RPF) 등) 등으로 이루어진 군에서 선택된 1종 이상일 수 있다. 상기 tag은 Hi s-tag / Ni-NTA등과같이 통상적으로사용되는모든종류에서 선택된 1종이상일 수 있다. 상기 표지 물질의 사용 농도는, 노이즈를 발생시키지 않고 정확하고용이한검출이 가능하도록 하기 위하여, 약 luM이하의 범위에서 적절하게 정할수 있으며, 예컨대, InM내지 1000nM, InM내지 500nM, InM 내지 100nM, 10nM 내지 lOOOnM, lOnM 내지 500nM, 또는 lOnM 내지 lOOnM 정도일 수 있으나, 이에 제한되는 것은 아니다. 상기와 같은 표지물질로부터 발생하는 신호는 이를 검출 또는 측정하는데 통상적으로 사용되는모든신호검출수단 (예컨대, 통상의 형광현미경, 형광카메라, 형광세기 측정 (정량)장치 등)에 의하여 측정될수있다.  The labeled second protein may be labeled with a labeling substance that generates a detectable signal of the second protein (for example, the labeling substance is chemically (e.g., covalent or noncovalent), recombinantly, or physically bound) , And a tag-attached form to which a labeling substance can be bound. The detectable signal may be selected from any signal (e.g., light, radiation, etc.) that can be measured through conventional enzymatic reactions, fluorescence, luminescence, and / or radiation detection. The labeling substance may be at least one selected from the group consisting of all small molecule compounds capable of generating the label signal, proteins, peptides, and nucleic acid molecules. Examples of the labeling substance include fluorescent dyes (small molecule compounds: Cyanine, Alex, Dylight, Fluoprobes Etc.), fluorescent proteins (e.g., green fluorescent protein (GFP), enhanced GFP), yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), blue fluorescent protein (BPF), red fluorescent protein Lt; / RTI > The tag may be at least one selected from all kinds commonly used such as Hi s-tag / Ni-NTA. The concentration of the labeling substance to be used may be appropriately determined in the range of about luM or less, for example, in order to allow accurate and easy detection without generating noise. For example, InM to 1000 nM, InM to 500 nM, InM to 100 nM, To about 100 nM, from about 10 nM to about 500 nM, or from about 10 nM to about 100 nM, but are not limited thereto. The signal generated from such a labeling substance can be measured by any signal detecting means (for example, a conventional fluorescence microscope, a fluorescence camera, a fluorescence intensity measuring apparatus, etc.) commonly used for detecting or measuring the signal.
제 1 단백질과 제 2 단백질 간의 상호작용을 보다 정확하게 측정하기 위하여, 반응시키는 단계 (단계 (2))와 후속하는 단백질-단백질 상호작용 측정 단계 (단계 (3)) 사이에, 상기 반응이 일어난 기판을 통상적인 방법으로세척하는단계를추가로포함할수있다.  Between the step of reacting (step (2)) and the step of measuring the subsequent protein-protein interaction (step (3)), the substrate May be further washed in a conventional manner.
일 예에서, 상기 제 2단백질은 PD-L1또는 PD-1일 일 수 있다. 일 구체예에서, 제 1 단백질이 PD-L1인 경우, 제 2 단백질은 PD-1일 수 있다. 다른 구체예에서, 제 1 단백질이 PD-1인 경우, 제 2 단백질은 PD-L1일 수 있다. In one example, the second protein may be PD-L1 or PD-1. In one embodiment, when the first protein is PD-L1, the second protein may be PD-1. In another embodiment, when the first protein is PD-1, the second protein may be PD-L1.
다른예에서, 제 1단백질이 HER2-HER3헤테로다이머인경우, (2)제 1 단백질과제 2단백질을반응시키는단계는 (2-1) HER2-HER3헤테로다이머을 인산화시키고, 인산화된 타이로신 특이적 결합 물질과 반응시키는 단계에 적용될수있다.  In another example, when the first protein is a HER2-HER3 heterodimer, (2) the step of reacting the first protein task 2 protein comprises (2-1) phosphorylating the HER2-HER3 heterodimer and contacting the phosphorylated tyrosine- And the like.
제 1 단백질이 HER2-HER3 헤테로다이머인 경우, 상기 단계 (2) 또는 (2-1)는, 상기 준비된 제 1 단백질이 고정된 기판에, 표지된 제 2 단백질을 첨가하거나 또는 제 1 단백질을 인산화시키고, 표지된 인산화된 타이로신 특이적 결합물질을 첨가하여 반응시키는단계이다. 상기 표지된 인산화된 타이로신 특이적 결합 물질은 HER2 또는 HER3의 인산화된 타이로신에 특이적으로결합하는항체일수있다.  When the first protein is a HER2-HER3 heterodimer, the step (2) or (2-1) may be carried out by adding the labeled second protein to the substrate on which the prepared first protein is immobilized, And adding the labeled phosphorylated tyrosine specific binding substance to react. The labeled phosphorylated tyrosine specific binding material may be an antibody that specifically binds to phosphorylated tyrosine of HER2 or HER3.
제 1 단백질이 HER2-HER3 헤테로다이머인 경우, 상기 제 2 단백질은, 앞서 설명한바와 같이, 다양한 생체 신호전달경로에 있어서 HER2 및/또는 HER3 (예컨대, HER2-HER3 헤테로다이머)의 하위 단백질로서, HER2 및/또는 HER3 (예컨대, HER2-HER3 헤테로다이머)와 상호작용 (결합) 가능한 단백질 중에서 선택된 1종이상일수있다.  When the first protein is a HER2-HER3 heterodimer, the second protein is a sub-protein of HER2 and / or HER3 (e.g., HER2-HER3 heterodimer) in various biological signaling pathways, And / or proteins capable of interacting (binding) with HER3 (e.g., HER2-HER3 heterodimer).
제 1단백질이 HER2-HER3헤테로다이머인경우, 상기 단계 (2-1)에서, 인산화시키는 단계는 상기 기판 또는 상기 기판에 고정된 HER2-HER3 헤테로다이머에 인산화제를 처리하는 단계를 포함할 수 있다. 상기 인산화제는 HER2 및/또는 HER3를 인산화시킬 수 있는 모든 물질들 중에서 선택된 1종 이상일 수 있으며, 예컨대, ATP와 마그네슘 (예컨대, MgCl2 등)일 수 있으나, 이에 제한되는 것은 아니다. 상기 인산화 전, 후, 또는 동시, 및 단계 (3-1)의 활성화 측정 전에, 상기 기판또는 기판에 고정된 HER2-HER3 헤테로다이머에, 앞서 단계 (1) 또는 (1-1)에서 설명한 바와 같은, DGTN, GDN 등을 포함하는 콜레스테롤-유사 세정제들 중에서 선택된 1종 이상의 세정제 (detergent )를 CMC cr i t i cal mi cel lar concent rat ion)를 초과하는 농도로 처리하여 HER2-HER3 헤테로다이머의 구조적 및/또는 기능적 활성을유지시키는것을특징으로할수있다. When the first protein is a HER2-HER3 heterodimer, in the step (2-1), the phosphorylation step may comprise treating the substrate or the HER2-HER3 heterodimer immobilized on the substrate with a phosphorylating agent . The phosphate reagent may be a HER2 and / or can be at least one selected from every substances capable of HER3 phosphorylation, e.g., ATP and magnesium (e.g., MgCl 2, etc.), without being limited thereto. HER2-HER3 heterodimer immobilized on the substrate or substrate before, after, or simultaneously with the phosphorylation, and prior to the activation measurement of step (3-1), as described in step (1) or , HER2-HER3 heterodimers at a concentration exceeding one or more detergents selected from cholesterol-like detergents including DGTN, GDN, etc., in excess of the CMC crest micellar concentrate, Or functional activity thereof.
단계 (3)또는 (3-1): 단백질-단백질상호작용측정 단계  Step (3) or (3-1): Measurement of protein-protein interaction step
상기 단계 (3)은 상기 단계 (2)에서 얻어진 반응물로부터 신호를 측정하는 단계이다. 상기 신호 측정은 단계 (2)에서 사용된 표지 신호 (예컨대, 효소 반응, 형광, 발광, 또는 방사선 검출 등의 통상적인 2019/132517 1»(:1^1{2018/016675 The step (3) is a step of measuring a signal from the reactant obtained in the step (2). The signal measurement may be carried out using a conventional signal (e. G., An enzyme reaction, fluorescence, luminescence, or radiation detection) 2019/132517 1 »(: 1 ^ {2018/016675
방법을통하여 측정 가능한신호)를검출 (또는측정 또는확인)할수 있는 모든수단을사용하여 수행될수있다. (Or measuring or verifying) a signal (e.g., a signal measurable via a method).
일 예에서, 단계 (3)에서의 단백질-단백질 상호작용 측정은 실시간 분석에 의한것일수있다.  In one example, the measurement of protein-protein interaction in step (3) may be by real-time analysis.
일 예에서, 상기 표지 신호가 형광신호인 경우, 상기 신호 검출은 상기 형광 신호를 발생시키는 표지 물질이 흡수하는 광원을 공급하여 발생하는 형광 신호를, 예컨대, 형광 현미경, 형광 카메라, 및/또는 형광 세기 측정 (정량) 장치 등을 사용하여, 영상화하거나 및/또는 정량할 수 있다.  In one example, when the label signal is a fluorescence signal, the signal detection is performed by supplying a fluorescence signal generated by supplying a light source absorbed by a labeling substance for generating the fluorescence signal to a fluorescence microscope, a fluorescence camera, and / And may be imaged, imaged, and / or quantified using, for example, an intensity measuring device.
일 구체예에서, 상기 신호는 형광 신호인 경우, 상기 형광 신호는 형광카메라를이용하여 영상화및/또는정량할수있다.  In one embodiment, when the signal is a fluorescence signal, the fluorescence signal can be imaged and / or quantified using a fluorescence camera.
상기 신호가 형광신호인 경우, 단계 (3) (단백질-단백질 상호작용 측정 단계)은  If the signal is a fluorescent signal, step (3) (protein-protein interaction measurement step)
(0상기 단계 (2)의 반응물에 광원을공급하는단계 ; 및  (0) supplying a light source to the reactant of the step (2); and
(11 )상기 공급된광원에 의하여 발생한형광신호를검출하는단계 를포함할수있다.  (11) detecting a fluorescence signal generated by the supplied light source.
상기 단계 0 )의 광원을공급하는단계는상기 단계 (2)에서 얻어진 제 1단백질과제 2단백질의 반응물에 광원흩공급하는단계로서, 이와같은 목적을 달성할 수 있다면, 광원의 공급 시기는 특별한 제한이 없다. 예컨대, 상기 광원은 단계 (1) 이전, 동시, 또는 이후부터 단계 (2) 이후까지 지속적으로 공급되거나, 단계 (2) 직전, 동시, 또는 직후에 소정의 시간동안공급될수있으나, 이에 제한되는것은아니다.  The step of supplying the light source of the step 0) is a step of supplying a light source to the reactant of the first protein task 2 protein obtained in the step (2). If the above purpose can be achieved, There is no. For example, the light source may be continuously supplied before step (1), simultaneously or after step (2), or may be supplied for a predetermined time immediately before, simultaneously with, or immediately after step (2) no.
상기 광원은 형광 신호에 해당하는 파장을 갖는 모든 광원일 수 있으며, 예컨대, 레이저, 할로겐 램프등일수있다.  The light source may be any light source having a wavelength corresponding to a fluorescent signal, for example, a laser, a halogen lamp, or the like.
상기 광원의 파장은사용된 형광신호에 따라서 조절될 수 있으며, 예컨대, 약 300^ 내지 약 6001^또는 약 350™ 내지 약 560™ 범위에서 선택될수 있다. 보다구체적으로, 녹색형광단백질은 약 48011111를흡수하고, 황색형광단백질은 약 540™를 흡수하고, 청색형광단백질은 약 375^1를 흡수하고, 청록색형광단백질은 약 425 을 흡수하므로, 형광 신호로 녹색 형광을 사용하는 경우 상기 광원의 파장은 약 460 내지 약 500™, 형광 신호로 황색 형광을 사용하는 경우 상기 광원의 파장은 약 520 내지 약 560^, 형광신호로청색 형광을사용하는경우상기 광원의 파장은 약 350 내지 약 400에1 , 형광 신호로 청록색 형광을 사용하는 경우 상기 광원의 파장은약 400내지 약 450nm범위에서 선택될수있다. Wavelength of the light source can be adjusted depending on the fluorescence signal using, for example, it can be selected from about 300 to about 600 ^ 1 ^, or about 350 to about 560 ™ ™ range. More specifically, the green fluorescent protein absorbs about 480 11111 , the yellow fluorescent protein absorbs about 540, the blue fluorescent protein absorbs about 375 < 1 > , and the blue fluorescent protein absorbs about 425, When the green fluorescence is used, the wavelength of the light source is about 460 to about 500, the wavelength of the light source is about 520 to about 560 when the yellow fluorescence is used as the fluorescence signal, and when the blue fluorescence is used as the fluorescence signal, The wavelength of the light source is 1 to about 350 to about 400, and when the cyan fluorescence is used as the fluorescence signal, The wavelength can be selected in the range of about 400 to about 450 nm.
일 구체예에서, 단계 (3)의 단백질-단백질 상호작용 측정 단계는 전반사 형광 현미경 (Total Internal Ref lect ion Fluorescence (TIRF) mi croscope)또는공초점 현미경 등을사용하여 광원을공급하고, 통상적인 5 방법으로 형광 신호를 관찰하여 수행될 수 있다. 다른 예에서, 상기 전반사 형광 현미경에 신호 영상화를 위한 형광 카메라, 예컨대 EMC抑In one embodiment, the protein of step (3) - protein interaction measuring step, by using the total internal reflection fluorescence microscopy (Total Internal Ref lect ion Fluorescence (TIRF) mi croscope) or confocal microscopy, and supplies the light source, the typical 5 Lt; RTI ID = 0.0 > fluorescence < / RTI > signal. In another example, a fluorescence camera for signal imaging, such as an EMC suppressor
(Electron-mul t iplying charge-coupled devi ce) 카메라 또는 CMOS (Complementary metal oxide semi conductor) 카메라를 장착하여 사용함으로써, 광원공급및 형광신호의 영상화및/또는정량을수행할수0 있다. (Electron-mul t iplying charge- coupled devi ce) The use equipped with a camera or a CMOS (Complementary metal oxide semi conductor) camera can perform imaging and / or quantification of the light source and fluorescence signals are fed 0.
다음에서는단계 (3)의 단백질-단백질 상호작용측정 단계를 전반사 현미경 및 형광카메라를사용하여 수행하는경우를 예를들어 보다상세히 설명한다:  In the following, the steps of measuring the protein-protein interaction in step (3) are carried out using a total internal reflection microscope and a fluorescent camera, for example, in more detail:
가) 상기 단계 (1) 또는 단계 (2)의 기판을 전반사 현미경에5 장착시킨다. 전반사 현미경에서 광원의 공급 위치는 통상적으로 아래쪽이며, 전반사 현미경 종류에 따라서, 형광 신호를 기판의 위 (이 경우, 아래에서 위 방향으로, 광원 공급부, 기판, 렌즈, 또는 기판, 광원 공급부, 렌즈의 순서로위치할수 있음) 또는아래에서 (이 경우, 아래에서 위 방향으로, 렌즈, 광원 공급부, 기판, 또는 광원 공급부, 렌즈, 기판,0 또는렌즈, 기판, 광원공급부의 순서로위치할수있음)관찰할수있다. A) The substrate of step (1) or step (2) is mounted on the total reflection microscope 5 . The supply position of the light source in the total reflection microscope is normally downward and the fluorescence signal is emitted from above the substrate (in this case, from the lower side to the upper side, the light source supply portion, the substrate, the lens or the substrate, under that it can position in the sequence) or (observed in this case, the upward direction from below, may be located in the lens, the light source supply, substrate, or the light source supply unit, lenses, substrate, and 0, or the lens, substrate, order of the light source supply unit) can do.
나)광원은레이저일수있으며, 광원의 세기는약 0.5 mW내지 약 5 mW, 약 0.5 mW내지 약 4.5 mW, 약 0.5 mW내지 약 4 mW, 약 0.5 mW내지 약 3.5 mW, 약 0.5 mW내지 약 3 mW, 약 0.5 mW내지 약 2.5 mW, 약 0.5 mW 내지 약 2 mff, 약 1 mff 내지 약 5 mW, 약 1 mW내지 약 4.5 mW, 약 1 mW5 내지 약 4 mW, 약 1 mW내지 약 3.5 mW, 약 1 mff 내지 약 3 mW, 약 1 mW 내지 약 2.5 mW, 약 1 mW내지 약 2 mW, 약 1.5 mW내지 약 5 mW, 약 1.5 mW내지 약 4.5 mW, 약 1.5 mW내지 약 4 mff, 약 1.5 mff 내지 약 3.5 mW,
Figure imgf000032_0001
B) the light source may be a laser and the intensity of the light source may be from about 0.5 mW to about 5 mW, from about 0.5 mW to about 4.5 mW, from about 0.5 mW to about 4 mW, from about 0.5 mW to about 3.5 mW, mW, about 0.5 mW to about 2.5 mW, about 0.5 mW to about 2 mff, about 1 mff to about 5 mW, about 1 mW to about 4.5 mW, about 1 mW 5 to about 4 mW, about 1 mW to about 3.5 mW From about 1 mW to about 4 mW, from about 1.5 mW to about 4 mW, from about 1 mW to about 3 mW, from about 1 mW to about 2.5 mW, from about 1 mW to about 2 mW, from about 1.5 mW to about 5 mW, 1.5 mff to about 3.5 mW,
Figure imgf000032_0001
감쇄필터를 사용하는 경우 세기가 센 광원을 사용할 수 있음)에 따라서 적절하게선택할수있다.  And a strong light source can be used when an attenuation filter is used).
다) 상기 광원 공급에 의하여 발생한 형광 신호를 형광 카메라로 촬영하여 영상화및/또는정량할수있다. C) The fluorescence signal generated by the light source supply is transmitted to the fluorescent camera Imaging and imaging and / or quantification.
상기 형광신호의 촬영 (또는영상화)은, 표지 물질의 형광신호발생 유지 시간 (발광시간, l i fet ime)을고려하여, 광원 공급과동시 또는상기 신호발생유지 시간이내에 수행할수있다.  The imaging (or imaging) of the fluorescence signal can be performed simultaneously with the light source supply or within the signal generation holding time, taking into account the fluorescence signal generation holding time (luminescence time) of the labeling material.
형광 카메라 (예컨대, EMCCD 카메라)로 형광 신호를 촬영(또는 영상화)함에 있어서, 1 프레임당 노출시간, 레이저 파워, 카메라 gain값, 총 촬영 프레임 등을 적절하게 조절할 수 있다. 예컨대, 1 프레임 당 노줄시간이 짧을수록 1 프레임에 누적되는 신호가 줄어들게 되며, 이를 상쇄하기 위하여 레이저 파워를 높이거나, 형광 카메라의 감도를 높일 수 있다. 일 예에서, 1 프레임 당 노출시간을 약 0.0()1초 내지 약 5초, 약 0.001초 내지 약 3초, 약 0.0이초 내지 약 2초, 약 0.0()1초 내지 약 1초, 약 0.001초내지 약 0.5초, 약 0.001초내지 약 0.3초, 약 0.001초내지 약 0.1초, 약 0.01초내지 약 5초, 약 0.()1초내지 약 3초, 약 0.01초내지 약 2초, 약 0. 초내지 약 1초, 약 0.01초내지 약 0.5초, 약 0.01초내지 약 0.3초, 약 0.()1초내지 약 0.1초, 약 0.05초내지 약 5초, 약 0.05초내지 약 3초, 약 0.05초 내지 약 2초, 약 0.05초 내지 약 1초, 약 0.05초 내지 약 0.5초, 약 0.05초내지 약 0.3초, 약 0.05초내지 약 0.1초, 약 0.07초 내지 약 5초, 약 0. 07초 내지 약 3초, 약 0. 07초 내지 약 2초, 약 0. 07초내지 약 1초, 약 0. 07초내지 약 0.5초, 약 0. 07초내지 약 0.3초, 약 0.07초 내지 약 0.1초, 약 0.1초 내지 약 5초, 약 0.1초 내지 약 3초, 약 0.1초 내지 약 2초, 약 0.1초 내지 약 1초, 약 0.1초 내지 약 0.5초, 또는 약 0.1초 내지 약 0.3초, 예컨대 약 0.1초로 할 수 있으나 이에 제한되는것은아니다.  In taking a fluorescent signal (or imaging) with a fluorescent camera (for example, an EMCCD camera), the exposure time per one frame, the laser power, the camera gain value, and the total shooting frame can be appropriately adjusted. For example, the shorter the dwell time per frame is, the less the accumulated signal in one frame. In order to compensate for this, the laser power can be increased or the sensitivity of the fluorescent camera can be increased. In one example, the exposure time per frame is from about 0.0 () 1 second to about 5 seconds, from about 0.001 second to about 3 seconds, from about 0.0 second to about 2 seconds, from about 0.0 () second to about 1 second, from about 0.001 From about 0.01 second to about 5 seconds, from about 0. 1 second to about 3 seconds, from about 0.01 second to about 2 seconds, from about 0.01 second to about 0.5 second, from about 0.001 second to about 0.3 second, from about 0.001 second to about 0.1 second, From about 0.01 second to about 1 second, from about 0.01 second to about 0.5 second, from about 0.01 second to about 0.3 second, from about 1 second to about 0.1 second, from about 0.05 second to about 5 seconds, From about 0.05 second to about 2 seconds, from about 0.05 second to about 1 second, from about 0.05 second to about 0.5 second, from about 0.05 second to about 0.3 second, from about 0.05 second to about 0.1 second, from about 0.07 second to about 5 seconds About 0. 07 second to about 3 seconds, about 0. 07 second to about 2 seconds, about 0. 07 second to about 1 second, about 0. 07 second to about 0.5 second, about 0. 07 second to about 0.3 second From about 0.07 seconds to about 0.1 seconds, from about 0.1 seconds to about 5 seconds, from about 0.1 seconds to about 3 seconds, from about 0.1 seconds To about 2 seconds, from about 0.1 second to about 1 second, from about 0.1 second to about 0.5 second, or from about 0.1 second to about 0.3 second, such as about 0.1 second.
예컨대, EMCCD 카메라를 사용하는 경우, 표지 물질 (예컨대, eGFP)로부터 생성된 광자(photon)는 EMCXD의 소자를 통해 전자로 바뀌어 계즉된다 (광전효과, photoelectr i c ef fect) . 이 때, 광자 1개 당 생성되는전자의 개수를 gain값을통해 변경할수 있다. 설정된 gain값이 높을수록 광자 1개 당 생성되는 전자의 개수가 늘어나서, EMCCD의 감도가 높아지는 동시에 background noi se도 함께 증가하므로 signal-to-noi se 비율이 중요하다. 일 구체예에서, 우수한 signal-to-noi se 비율을 얻기 위하여, gain값을약 10내지 약 100, 약 10내지 약 80, 약 10내지 약 60, 약 10내지 약 50, 약 20내지 약 100, 약 20내지 약 80, 약 20내지 약 60, 약 20내지 약 50, 약 30내지 약 100, 약 30내지 약 80, 약 30내지 2019/132517 1»(:1^1{2018/016675 For example, when an EMCCD camera is used, photons generated from a labeling substance (eg, eGFP) are converted to electrons through the EMCXD element (photoelectric effect). At this time, the number of electrons generated per photon can be changed through the gain value. The higher the set gain value, the greater the number of electrons generated per photon, and the higher the sensitivity of the EMCCD and the higher the background noise, the higher the signal-to-noise ratio. In one embodiment, to obtain an excellent signal-to-noise ratio, the gain value can be adjusted to a value in the range of from about 10 to about 100, from about 10 to about 80, from about 10 to about 60, from about 10 to about 50, from about 20 to about 100, From about 20 to about 80, from about 20 to about 60, from about 20 to about 50, from about 30 to about 100, from about 30 to about 80, 2019/132517 1 »(: 1 ^ {2018/016675
약 60, 또는 약 30 내지 약 50, 예컨대, 약 40 정도로 정할 수 있지만, 이에 한정되지 않고 카메라의 감도, 수명, 장비 구축 현황, 노이즈, 시험 조건등을고려하여 적절하게선택될수있다. About 60 to about 60, or about 30 to about 50, such as about 40, but the present invention is not limited thereto, and can be appropriately selected in consideration of sensitivity, life span, equipment construction status, noise,
총촬영 프레임 개수와노출시간을곱하면총촬영 시간이 얻어진다 (노출시간 * 총 촬영 프레임 개수 =촬영 시간) . 형광물질의 발광시간 (1 6) 이후에는 형광신호가사라지므로, 상기 촬영 시간이 발광시간 내에 진행되도록총촬영 프레임 개수및/또는노출시간을조절할수 있다. 단백질-단백질 상호작용측정 결과의 정확도를높이기 위하여, 상기 영상화를 하나 이상, 예컨대, 2개 이상, 3개 이상, 4개 이상, 5개 이상, 7개 이상, 또는 10개 이상 (상한값은기판의 크기 및 영상화가능한면적에 따라서 결정됨)의 기판 또는 이를 포함하는 하나 이상의 채널 (각 채널은 2개 이상의 기판을 포함함)에서 수행하고, 신호가 나타난 31The total shooting time is obtained by multiplying the total number of shooting frames by the exposure time (exposure time * total shooting frame number = shooting time). Since the fluorescence signal disappears after the emission time of the fluorescent material ( 16) , the total number of photographed frames and / or the exposure time can be adjusted so that the photographed time progresses within the light emission time. In order to increase the accuracy of the protein-protein interaction measurement results, it is preferable that the imaging is performed by using one or more, for example, two or more, three or more, four or more, five or more, seven or more, performed on a substrate including one or more channels (each channel is at least two substrates comprising the same according to the determined size and the imaging area as possible)), and the signal appears 31 be
:에世 라고도함)의깨수를구하여, 상기 얻어진 형광신호를정량화할수 있으며, 이는단백질-단백질상호작용을정량화한것으로볼수있다. : ≪ / RTI > EIA), and quantify the fluorescence signal obtained, which can be seen as quantifying protein-protein interactions .
다른 예에서, 단계 (3)에서 측정된 형광 세기를 통상적인 장치를 이용하여 수치화함으로써 단백질-단백질상호작용을정량할수도있다.  In another example, the fluorescence intensity measured in step (3) may be quantified using conventional equipment to quantify protein-protein interactions.
제 1단백질 및/또는제 2단백질을 2종이상사용하는경우, 단계 (1) 내지 (3)은각각의 제 1단백질과제 2단백질조합에 대하여 각각수행될수 있다 (즉, 단계 (1) 내지 (3)은 제 1 단백질과 제 2 단백질 조합의 수만큼 반복수행될수있다) .  When two or more of the first protein and / or the second protein are used, steps (1) to (3) can be performed for each first protein task 2 protein combination (that is, steps (1) to 3) can be repeated as many times as the number of combinations of the first protein and the second protein.
제 1 단백질이 四묘2 - 요3 헤테로다이머인 경우, 상기 (3) 단백질- 단백질 상호작용 측정 단계는 (3-1) 타이로신 잔기의 인산화 측정 단계에 적용될수있다.  In the case where the first protein is quadrature 2-y3 heterodimer, the step (3) of measuring the protein-protein interaction may be applied to the step (3-1) of measuring the phosphorylation of the tyrosine residue.
상기 단계 (3) 또는 (3-1)은상기 단계 (2) 또는 (2-1)에서 얻어진 반응물로부터 신호를측정하는단계이다. 상기 신호측정은단계 (2)또는 (2-1)에서 사용된 표지 신호 (예컨대, 효소 반응, 형광, 발광, 또는 방사선 검출 등의 통상적인 방법을통하여 측정 가능한 신호)를 검출 (또는 측정 또는확인)할수있는모든수단을사용하여 수행될수있다.  The step (3) or (3-1) is a step of measuring a signal from the reactant obtained in the step (2) or (2-1). The signal measurement can be used to detect (or measure or verify) the label signal used in step (2) or (2-1) (e.g., a signal measurable via conventional means such as enzyme reaction, fluorescence, luminescence, ), Which can be performed using any means available.
제 1 단백질이 1世1?2 -四묘3 헤테로다이머인 경우, 단계 (3-1)의 타이로신 잔기의 인산화 측정 단계는 四1?2 - 묘3 헤테로다이머와 표지된 他1?2 또는 四1?3의 인산화된 타이로신 잔기에 특이적으로 결합하는 물질 (예컨대, 항체) 간의 반응에서 발생하는 신호를 검출 및/또는 정량하는 단계를포함할수 있다. 일 예에서, 표지 물질로 형광물질 (예컨대 형광 단백질)을 사용하는 경우, 상기 타이로신 잔기의 인산화는 앞서 설명한 바와 같이, 반응물에서 검출되는 형광 세기를 측정 및 정량화하여 측정될 수있다. In the case where the first protein is a first-order? 2-quaternary 3-heterodimer, the step of measuring the phosphorylation of the tyrosine residue in step (3-1) comprises the steps of: And detecting and / or quantifying a signal generated in a reaction between a substance (e. G., An antibody) that specifically binds to a phosphorylated tyrosine residue of 1 to 3. In one example, a fluorescent material (e.g., fluorescent Protein) is used, phosphorylation of the tyrosine residue can be measured by measuring and quantifying the fluorescence intensity detected in the reactant as described above.
단계 (4): 제 1단백질의 활성화수준측정 단계  Step (4): measuring the activation level of the first protein
단계 (4)의 제 1 단백질의 활성화 수준 측정 단계는 단계 (3)에서 측정된신호를이용하여 단계 (1)에서 첨가한시험 시료내의 제 1단백질의 단위량에 대한신호값 (act ivat ion score)을구하는단계일수있다.  The step of measuring the activation level of the first protein of the step (4) may include the step of measuring the activation level of the first protein in the test sample added in step (1) using the signal measured in step (3) ).
본 명세서에서, "제 1 단백질의 활성화”는 제 1 단백질의 제 2 단백질과의 상호작용 (결합)을 의미하고, "제 1 단백질의 활성화 수준'은 제 1 단백질의 제 2 단백질과의 상호작용 (결합)의 정도를 의미하고, "활성화된 제 1 단백질은 제 2 단백질과 상호작용 (결합)한 제 1 단백질을 의미할수있다.  As used herein, "activation of a first protein " means interaction (binding) of a first protein with a second protein, and" activation level of a first protein " (Binding), and the "activated first protein " may mean a first protein that interacts with (binds to) the second protein.
제 1 단백질의 단위량에 대한 신호값이라 함은 제 1 단백질의 단위 중량또는농도 (예컨대, lug/ml )또는신호값에 대한단계 (3)에서 측정된 신호값 (신호또는신호세기의 정량화된값)을의미하는것으로,  The signal value for the unit amount of the first protein refers to the unit weight or concentration (for example, lug / ml) of the first protein or the signal value measured in the step (3) Value)
(a) 단계 (3)에서 측정된 신호값을 시험 시료 내의 제 1 단백질의 중량, 농도, 또는형광신호값으로나누어 주거나,  (a) dividing the signal value measured in step (3) by the weight, concentration, or fluorescence signal value of the first protein in the test sample,
(b) 시험 시료 내의 제 1 단백질 중량, 농도, 또는 형광 신호값의 증가에 따른단계 (3)에서 측정된신호값의 증가분 (즉시험 시료내의 제 1 단백질 중량, 농도, 또는 형광 신호값을 x축으로 하고, 단계 (3)에서 측정된 신호값을 y값으로 하여 얻어진 그래프의 기울기)을 구하여 얻어질 수있다.  (b) the increment of the signal value measured in step (3) as the first protein weight, concentration, or fluorescence signal value in the test sample increases (i.e., the first protein weight, concentration, or fluorescence signal value in the test sample is x Axis, and the slope of the graph obtained by taking the signal value measured in step (3) as the y value).
본 명세서에서는 시료 내 제 1 단백질의 양보다, 활성화된 제 1 단백질의 비율이 상기 시료가유래하는 개체에서의 제 1 단백질을 표적으로 하는 약물 반응성에 보다 유의미한 상관관계를 나타냄을 제안한다. 즉, 시료 내 제 1 단백질의 수준이 낮아도 (즉, 양이 적어도) 활성화된 저 U 단백질의 비율 (활성화 수준: act ivat ion score)이 높은 경우에는 제 1 단백질 양이 많지만 활성화된 제 1 단백질의 비율이 낮은 경우와 비교하여 약물 반응성이 현저히 우수하다 (도 19 및 20 참조) . 따라서, 본 발명은 단백질-단백질 상호작용을측정하여 이를제 1단백질 단위량에 대한값으로 계산 (제 1 단백질 양으로 나누어줌)하여 약물 반응성 판단에 보다 정확한 정보를제공한다는점을특징으로한다.  The present specification suggests that the ratio of the activated first protein to the amount of the first protein in the sample shows a more significant correlation with the drug reactivity targeting the first protein in the sample from which the sample is derived. That is, when the level of the first protein in the sample is low (that is, the amount is at least) and the ratio of the activated U protein is high (activation level: act iatv ion score), the amount of the first protein The drug reactivity is remarkably superior as compared with the case where the ratio is low (see FIGS. 19 and 20). Therefore, the present invention is characterized in that the protein-protein interaction is measured and calculated as a value for the first protein unit amount (divided by the amount of the first protein), thereby providing more accurate information on drug reactivity determination.
단계 (4)의 제 1단백질의 활성화수준측정 단계는 (i ) 단계 (3)에서 측정된 신호값을 단계 (1)에서 첨가한 시험 시료 내의 제 1 단백질의 중량, 농도, 또는 형광 신호값으로 나누어 직접적으로 구하거나, The step of measuring the activation level of the first protein of step (4) (i) directly measuring the signal value measured in step (3) by dividing the signal value by the weight , concentration , or fluorescence signal value of the first protein in the test sample added in step (1)
(i i ) 단계 (3)에서 측정된 신호값을 단계 (1)에서 첨가한 시험 시료의 중량, 농도, 또는 형광 신호값으로 나누어 시험 시료의 단위량에 대한신호값을구하는 단계 (단백질-단백질 상호작용수준측정 단계) u- 1), 및 상기 단계 (4-1)에서 얻어진 시험 시료의 단위량에 대한신호값을 단계 (1)에서 첨가한시험 시료 내의 제 1단백질의 중량, 또는농도, 또는 형광 신호값으로 나누어 시험 시료에 포함된 제 1 단백질의 단위량에 대한 신호값을구하는단계 (활성화수준즉정) (4_2)를통하여 수행될수있다.  (ii) obtaining a signal value for the unit amount of the test sample by dividing the signal value measured in step (3) by the weight, concentration, or fluorescence signal value of the test sample added in step (1) The signal level for the unit amount of the test sample obtained in step (u-1)), and the step (4-1), the weight or concentration of the first protein in the test sample added in step (4-2) of obtaining the signal value of the unit amount of the first protein contained in the test sample (activation level immediate) by dividing the fluorescence signal value by the fluorescence signal value.
단백질-단백질상호작용수준측정 단계 (4-1)  Protein-Protein Interaction Level Measurement Step (4-1)
단계 (4-1)의 단백질-단백질 상호작용 수준 측정 단계는 단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한 시험 시료의 단위량에 대한신호값을구하는단계이다.  The step of measuring the protein-protein interaction level in the step (4-1) is a step of obtaining a signal value of the unit amount of the test sample added in the step (1) using the signal measured in the step (3).
상기 단백질-단백질 상호작용 수준은 단백질-단백질 상호작용 세기 The protein-protein interaction level is determined by the protein-protein interaction intensity
(PPI strength)로도 표현할 수 있으며, 단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한 시험 시료의 단위량에 대한 신호값을 구함으로써 시험 시료의 사용량 등의 시험 시료 조건에 의한 오차를 줄일 수있다. (PPI strength), and the signal value of the unit amount of the test sample added in the step (1) by using the signal measured in the step (3) is determined by the test sample condition such as the amount of the test sample Error can be reduced.
단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한 시험 시료의 단위량에 대한신호값을구하는단계는단계 (3)에서 측정된신호를 시험 시료의 양 (농도 또는 중량)으로 나누거나, 단계 (3)에서 측정된 신호를 y축으로 하고 단계 (1)에서 첨가한 시험 시료의 양 (농도 또는 중량)을 x축으로 하여 얻어진 그래프의 기울기를 구함으로써 수행될 수 있다.  The step of obtaining the signal value for the unit amount of the test sample added in the step (1) using the signal measured in the step (3) is the step of measuring the signal measured in the step (3) by the amount , Or by calculating the slope of the graph obtained by taking the signal measured in step (3) as the y-axis and the amount (concentration or weight) of the test sample added in step (1) as the x-axis.
제 1 단백질 및/또는 제 2 단백질을 2종 이상 사용하는 경우, 상기 단백질-단백질 상호작용 수준 측정 단계는 각각의 단백질 조합에 대하여 수행될수있다.  When two or more of the first protein and / or the second protein are used, the step of measuring the protein-protein interaction level may be performed for each protein combination.
제 2단백질 (downstream protein)이 2종 이상인 경우, 다음의 수식에 표현한 바와 같이, 각각의 제 2 단백질에 대하여 얻어진 PPI strength의 합을구하여, 단백질-단백질상호작용수준 (PPI score)으로결정할수 있다:  When two or more downstream proteins are present, the sum of the PPI strengths obtained for each second protein can be determined as the protein-protein interaction level (PPI score), as expressed in the following equation :
c DD T제 1단백질 c DD T First protein
Sum of PPI시험시료 = 2V_, ^DDT f ᄀ제 1단백질 Sum of PPI test sample = 2V_ , ^ DDT f protein 1 protein
(PPI strength)k ( PPI strength) k
k=제 2단백질 제 1단백질이 2종 이상인 경우, 각각의 제 1단백질에 대하여 얻어진 PPI score를 합한 값을 단백질-단백질 상호작용 수준 (PPI score)으로 결정할수있다. k = second protein When two or more first proteins are present, the sum of the PPI scores obtained for each first protein can be determined as the protein-protein interaction level (PPI score).
다른 예에서, 상기 얻어진 단백질-단백질 상호작용 수준 (PPI strength또는 PPI score)을 하기에 설명되는 기준 시료의 단백질-단백질 상호작용 수준이 1이 되도록 normal izat ion하여, 시험 시료의 PPI strength를기준시료의 PPI strength에 대한상대값으로나타낼수있다.  In another example, the obtained protein-protein interaction level (PPI strength or PPI score) is normal izat ion such that the protein-protein interaction level of the reference sample described below is 1, and the PPI strength of the test sample is set as a reference sample Of the PPI strength.
활성화수준측정 단계 (단계 4또는 4-2)  The activation level measurement step (step 4 or 4-2)
단계 (4) (앞서 설명한 단계 (4-1)의 단백질-단백질 상호작용 수준 라 score) 측정 단계 없이 직접 활성화수준을측정하는 경우) 또는 단계 (4-2)는 상기 단계 (3)에서 얻어진 신호값 또는 단계 (4-1)에서 얻어진 시험 시료의 단위량에 대한신호값을 이용하여 시험 시료에 포함된 제 1단백질의 단위량에 대한값을구하는 단계이다. 본 명세서에서, 상기 단계 (4) 또는 단계 (4-2)에서 얻어진 결과를 활성화 수준 (또는 act ivat ion score)이라고칭한다.  (4) (the protein-protein interaction level of step (4-1) described above) is directly measured without the measurement step) or step (4-2) Value or the signal value of the unit amount of the test sample obtained in the step (4-1) to obtain the value for the unit amount of the first protein contained in the test sample. In this specification, the result obtained in the step (4) or the step (4-2) is referred to as an activation level (or an act ivat ion score).
상기 활성화 수준은 단계 (3)에서 얻어진 신호값 또는 단계 (4 - 1)에서 얻어진 단백질-단백질 상호작용 수준을 시험 시료에 포함된 제 1 단백질의 양으로 나누어줌으로써 시험 시료에 존재하는 제 1 단백질의 양 및/또는 분포 등에 의한오차를 줄여서 보다 제 1 단백질의 활성화 정도를 보다정확하게측정할수있다.  The activation level is determined by dividing the signal value obtained in step (3) or the protein-protein interaction level obtained in step (4-1) by the amount of the first protein contained in the test sample, It is possible to more accurately measure the degree of activation of the first protein by reducing the error due to the amount and / or distribution.
본 명세서에서 제공되는 방법은 시험 시료 내의 제 1 단백질의 양을 측정하는 단계를 추가로 포함할 수 있다. 시험 시료 내의 제 1 단백질의 양을 측정하는 단계는 단계 (4) 또는 단계 (4-2) 수행 전 또는 동시에 진행될수있다.  The method provided herein may further comprise the step of measuring the amount of the first protein in the test sample. The step of measuring the amount of the first protein in the test sample may be carried out before or simultaneously with the step (4) or the step (4-2).
상기 제 1단백질의 양은통상적인 모든방법으로측정할수 있으며, 예컨대, 통상적인 면역블라팅법 (예컨대, quant i tat ive western blott ing) , ELISA (enzyme- l inked immunosorbent assay; direct assay, indi rect assay, sandwich assay등) 등을사용하여 측정할수 있으나, 이에 제한되는 것은 아니다. 일 예에서, 제 1 단백질이 고정화된 기판에 표지된 검출 항체 (detect ing ant ibody)를 첨가하고 표지에서 발생하는 신호를 측정함으로써 제 1단백질을정량할수있으나, 이에 제한되는것은아니다.  The amount of the first protein can be measured by any conventional method. For example, the amount of the first protein can be measured by a conventional immunoblotting method (for example, quantitative assay, western blotting), ELISA (enzyme-linked immunosorbent assay; sandwich assay, etc.), but the present invention is not limited thereto. In one example, the first protein may be quantified by adding a detection antibody to the substrate to which the first protein is immobilized, and measuring a signal generated from the label, but the present invention is not limited thereto.
제 1 단백질 및/또는 제 2 단백질을 2종 이상 사용하는 경우, 상기 활성화수준측정 단계는각각의 제 1단백질과제 2단백질의 조합에 대하여 각각수행될수있다. When two or more of the first protein and / or the second protein are used, the step of measuring the activation level may be performed for each combination of the first protein task 2 protein Respectively.
제 2 단백질 (downstream protein)이 2종 이상인 경우, 앞서 설명한 바와 같이 각각의 제 2 단백질에 대하여 얻어진 PPI strength의 합 또는 상기 PPI strength의 합으로부터 얻어진 단백질-단백질 상호작용 수준 score)을 사용하여 활성화 수준 (act ivat ion score)을 결정하거나, 각각의 제 2 단백질에 대하여 얻어진 act ivat ion score의 합을 구하여, 상기 2종 이상의 제 2단백질에 대한활성화수준 (act ivat ion score)으로 결정할수 있다.  When there are two or more downstream proteins, the protein-protein interaction level score obtained from the sum of the PPI strengths obtained for each second protein or the sum of the PPI strengths as described above) (act ivat ion score), or the sum of the act iat ion scores obtained for each second protein, and determine the act iat ion score for the two or more second proteins.
제 1단백질이 2종이상인 경우, 앞서 설명한바와같이, 각각의 제 1 단백질에 대하여 얻어진 PPI strength의 합 또는 상기 PPI strength의 합으로부터 얻어진 단백질-단백질 상호작용 수준 (PPI score)을 사용하여 활성화수준 (act ivat ion score)을결정하거나, 각각의 제 1단백질에 대하여 얻어진 act ivat ion score의 합을 구하여, 상기 2종 이상의 제 1 단백질에 대한활성화수준 (act ivat ion score)으로결정할수있다.  When the first protein is in the form of two proteins, as described above, the protein level (PPI score) obtained from the sum of the PPI strengths obtained for each first protein or from the sum of the PPI strengths is used to determine the activation level act ivat ion score) or the sum of the act iat ion scores obtained for each first protein, and determine the act iat ion score for the two or more first proteins.
다른 예에서, 상기 얻어진 활성화 수준을 하기에 설명되는 기준 시료의 활성화수준이 1이 되도록 normal izat ion하여, 시험 시료의 활성화 수준을기준시료의 활성화수준에 대한상대값으로나타낼수있다.  In another example, the activity level obtained can be normalized to an activation level of the reference sample described below, such that the activation level of the test sample is relative to the activation level of the reference sample.
일 예에서, 제 1 단백질이 HER2-HER3 헤테로다이머인 경우, 단계 (4)는 상기 단계 (3) 또는 (3-1)에서 얻어진 결과 (단백질-단백질 상호작용 수준 또는 타이로신 카이네이즈 활성화 정도)을 기준 시료에서 얻어진 결과 (단백질-단백질 상호작용 수준 또는 타이로신 카이네이즈 활성화 정도)과비교하는단계일수있다.  In one example, when the first protein is a HER2-HER3 heterodimer, step (4) may comprise comparing the results (protein-protein interaction level or degree of tyrosine kinase activation) obtained in step (3) To the result obtained in the sample (protein-protein interaction level or degree of tyrosine kinase activation).
제 1 단백질이 HER2-HER3 헤테로다이머인 경우, 상기 기준 시료는 발명의 목적에 따라서 적절하게 선택될 수 있다. 예컨대, 세포또는조직 내 및/또는세포또는조직 간의 신호전달경로의 활성화측정 (또는 확인 또는 결정 또는 분석) 방법 또는 활성화 측정에 정보를 제공하는 방법의 경우, 상기 기준 시료는 (1) 정상 세포, 및/또는 (2) 제 1 단백질 (HER2, HER3, 또는 이들 모두 (예컨대, HER2-HER3 헤테로다이머 형태))이 관여하는 신호전달경로의 활성화 정도가 알려진 (확인된) 세포 (예컨대, 정상 세포 또는 암세포) , 및/또는 (3) 제 1단백질 (HER2, HER3, 또는이들모두 (예컨대, HER2-HER3 헤테로다이머 형태))이 관여하는 신호전달경로의 활성화 정도가 알려진 (확인된) 개체로부터 분리된 세포 (예컨대, 정상 세포 또는 암세포)를포함할수 있다. 일 구체예에서, 상기 시험 시료가개체로부터 분리된 암 세포를 포함하는 경우, 그 개체로부터 분리된 암세포와 동일 조직또는동일기관의 정상세포를포함하는것일수있다. When the first protein is a HER2-HER3 heterodimer, the reference sample can be appropriately selected according to the purpose of the invention. For example, in the case of a method for measuring the activation (or confirmation or determination or analysis) of a signal transduction pathway in a cell or tissue and / or between cells or tissues, or a method for providing information on the activity measurement, the reference sample may be (1) And / or (2) cells whose known degree of activation of the signal transduction pathway involved in the first protein (HER2, HER3, or both (e.g., HER2-HER3 heterodimer forms) Cancer cells), and / or (3) the degree of activation of the signal transduction pathway involving the first protein (HER2, HER3, or both (e.g., HER2-HER3 heterodimer forms) Cells (e. G., Normal cells or cancer cells). In one embodiment, the test sample is from a subject In the case of including isolated cancer cells, it may include normal cells of the same tissue or the same organ as the cancer cells isolated from the individual.
단계 (4)를수행하기 위하여, 상기 방법들은, 단계 (4) 이전에, 상기 기준 시료에 대하여 제 1 단백질 및 제 2 단백질 간의 단백질-단백질 상호작용또는타이로신 카이네이즈활성화정도를측정하는단계를추가로 포함할수 있으며, 이들단계는 앞서 설명한단계 (1), (2), 및 (3), 또는 (1-1), (2-1), 및 (3-1)을참조하여 수행할수있다.  In order to carry out the step (4), the methods further comprise, before step (4), measuring the degree of protein-protein interaction or tyrosine kinase activation between the first protein and the second protein with respect to the reference sample These steps can be performed with reference to the steps (1), (2), and (3) described above, or (1-1), (2-1), and (3-1).
제 1단백질이 HER2-HER3헤테로다이머인경우, 상기 단계 (4) 이후에, 단계 (4)에서 얻어진 비교 결과로부터 목적하는 사항을 확인 (결정)하는 단계를 추가로 포함할 수 있다. 이를 보다 구체적으로 살펴보면 다음과 같다:  In the case where the first protein is a HER2-HER3 heterodimer, after the step (4), a step of confirming (determining) a desired item from the comparison result obtained in step (4) may be further included. More specifically, it is as follows:
제 1단백질이 HER2-HER3헤테로다이머인경우, 상기 확인 (결정)하는 단계는다음의 단계를포함하는것일수있다:  If the first protein is a HER2-HER3 heterodimer, the step of determining may comprise the following steps:
단계 (3)에서 측정된 시험 시료의 단백질-단백질 상호작용 수준이 기준 시료에서 측정된 단백질-단백질 상호작용 수준보다 높은 경우, 시험 시료 또는 상기 시험 시료가 유래하는 개체에서 제 1 단백질이 관여하는 신호전달경로의 활성화정도가정상세포의 활성화정도또는기준시료에서 알려진 (확인된)활성화정도보다높다고확인 (결정)하는단계;  When the protein-protein interaction level of the test sample measured in step (3) is higher than the protein-protein interaction level measured in the reference sample, the signal that the first protein participates in the test sample or the individual from which the test sample originates Confirming (determining) the degree of activation of the delivery path is higher than the degree of activation of normal cells or the level of activation (known) recognized in the reference sample;
단계 (3)에서 측정된 시험 시료의 단백질-단백질 상호작용 수준이 기준 시료에서 측정된 단백질-단백질 상호작용 수준과 동등한 경우, 시험 시료 또는 상기 시험 시료가 유래하는 개체에서 제 1 단백질이 관여하는 신호전달경로의 활성화정도가정상세포의 활성화정도또는기준시료에서 알려진 (확인된)활성화정도와동등하다고확인 (결정)하는단계; 및/또는 단계 (3)에서 측정된 시험 시료의 단백질-단백질 상호작용 수준이 기준 시료에서 측정된 단백질-단백질 상호작용 수준보다 낮은 경우, 시험 시료 또는 상기 시험 시료가 유래하는 개체에서 제 1 단백질이 관여하는 신호전달경로의 활성화정도가정상세포의 활성화정도또는기준시료에서 알려진 (확인된)활성화정도보다낮다고확인 (결정)하는단계.  When the protein-protein interaction level of the test sample measured in step (3) is equal to the protein-protein interaction level measured in the reference sample, the signal that the first protein participates in the test sample or the individual from which the test sample originates Confirming (determining) the degree of activation of the delivery path is equivalent to the degree of activation of normal cells or the degree of activation (known) recognized in the reference sample; And / or the protein-protein interaction level of the test sample measured in step (3) is lower than the protein-protein interaction level measured in the reference sample, the test sample or the first protein from the test sample- (Determination) that the degree of activation of the involved signaling pathway is lower than the degree of activation of normal cells or the level of activation (known) known in the reference sample.
단계 (5): 기준시료와비교하는단계  Step (5): Step of comparing with reference sample
단계 (5)는 상기 단계 (4) 또는 단계 (4-1) 또는 단계 (4-2)에서 얻어진 결과 (단백질-단백질 상호작용 수준 (PPI score) 또는 활성화 수준 (Act ivat ion score))을기준시료에서 얻어진 결과 (단백질-단백질 상호작용 수준 (PPI score) 또는 활성화 수준 (Act ivat ion score))과 비교하는 2019/132517 1»(:1^1{2018/016675 Step 5 is a step of comparing the results obtained in step (4) or step (4-1) or step (4-2) (protein-protein interaction level (PPI score or Activat ion score) (Protein-protein interaction level (PPI score) or activation level (Act iat ion score)) obtained from the sample 2019/132517 1 »(: 1 ^ {2018/016675
단계이다. .
상기 기준시료는발명의 목적에 따라서 적절하게선택될수있다. 예컨대, 세포 또는 조직 내 및/또는 세포 또는 조직 간의 신호전달경로의 활성화측정 (또는 확인 또는 결정 또는 분석) 방법 또는 5 활성화 측정에 정보를 제공하는 방법의 경우, 상기 기준 시료는 (1) 정상 세포, 및/또는 (2) 제 1 단백질이 관여하는 신호전달경로의 활성화 정도가 알려진 (확인된) 세포 (예컨대, 정상 세포 또는 암세포), 및/또는 (3) 제 1 단백질이 관여하는 신호전달경로의 활성화 정도가 알려진 (확인된) 개체로부터 분리된 세포 (예컨대, 정상 세포 또는 암세포)를 포함할 수 10 있다. 일 구체예에서, 상기 시험 시료가 개체로부터 분리된 암 세포를 포함하는 경우, 그 개체로부터 분리된 암세포와 동일 조직 또는 동일 기관의 정상세포를포함하는것일수있다. The reference sample can be appropriately selected according to the purpose of the invention. For example, in the case of a method for measuring the activation (or confirmation or determination or analysis) of a signal transduction pathway in a cell or tissue and / or between a cell or tissue, or a method for providing information on a 5 activation measurement, And / or (2) a cell in which the degree of activation of the signal transduction pathway involved in the first protein is known (confirmed) (e.g., normal or cancer cells), and / or (3) the degree of activation can 10 may comprise a known (identified), the cells (e.g., normal cells or cancer cells) isolated from the object. In one embodiment, when the test sample comprises cancer cells isolated from an individual, it may include normal cells of the same tissue or the same organ as cancer cells isolated from the test specimen.
본 명세서에 사용된 바로서, 용어 ’’정상세포’는 비-병리적 상태의 모든 세포를 의미할 수 있으며, 상기 "비-병리적 상태"는 질병 상태가 15 아니거나, 돌연변이, 종양 형성, 기능적 및/또는 형태적 이상 등을 갖는 질병을유발할수 있는 상태가 아닌 것을 의미할수 있다. 예컨대, 정상 세포는제 1단백질과관련된 질병 또는시험 대상약물의 치료대상질병을 갖지 않는 세포일 수 있으며, 시험 시료가유래한 개체 또는 조직과 동종 또는동일한개체또는조직으로부터유래한것일수있다. As used herein, the term "normal cell" may refer to any cell in a non-pathological state, wherein the "non-pathological condition" refers to a condition in which the disease state is not 15 , Functional, and / or morphological abnormality, or the like. For example, a normal cell may be a disease associated with the first protein or a cell that does not have a disease to be treated of the drug to be tested, and may be derived from the same individual or tissue as the individual or tissue from which the test sample is derived.
20 또한, 제 1 단백질을 표적으로 하는 약물에 대한 반응성을 예측하는 방법 또는 예즉에 정보를 제공하는 방법의 경우, 상기 기준 시료는 정상 세포 또는 상기 약물에 대한 반응성이 알려진 (확인된) 세포 (예컨대, 암세포)를 포함하거나, 상기 시험 시료가 개체로부터 분리된 암 세포를 포함하는 경우, 암세포와 동일 조직 또는 동일 기관의 정상 세포를 25 포함하는것일수있다. 20 Also, in the case of a method for predicting the reactivity to a drug targeting the first protein or a method for providing information on the prediction, the reference sample may be a normal cell or a cell having known (confirmed) , Cancer cells), or when the test sample contains cancer cells isolated from an individual, 25 normal cells of the same tissue or the same organ as the cancer cells may be included.
또한, 제 1 단백질을 표적으로 하는 치료에 적합한 개체를 선별하는 방법 또는 선별에 정보를 제공하는 방법의 경우, 상기 기준 시료는 정상 세포또는상기 제 1단백질을표적으로하는치료의 효과가알려진 (확인된) 세포 (예컨대, 암세포)를포함하거나, 상기 시험 시료가개체로부터 분리된 30 암 세포를 포함하는 경우, 암세포와 동일 조직 또는 동일 기관의 정상 세포를포함하는것일수있다.  In addition, in the case of selecting the individual suitable for treatment targeting the first protein or providing the information to the selection, the reference sample may be a normal cell or a cell which has known the effect of treatment targeting the first protein (Eg, cancer cells), or when the test sample contains 30 cancer cells isolated from an individual, normal cells of the same tissue or the same organ as the cancer cells may be included.
단계 (5)를수행하기 위하여, 상기 방법들은, 단계 (5) 이전에, 상기 기준 시료에 대하여 다음의 단계 (1’), (2 ), (3 ), 및 (4 ) , 또는 (1’) , 2019/132517 1»(:1^1{2018/016675 To perform step (5), the methods comprise the following steps (1 '), (2), (3) and (4) ), 2019/132517 1 »(: 1 ^ {2018/016675
(2’), (3’) , (4-1 ) , 및 (4-2 )를추가로포함할수있다: (2 '), (3'), (4-1), and (4-2)
(1' ) 제 1단백질을포함하는기준시료를, 표면에 상기 제 1단백질에 특이적으로 결합하는 물질을포함하는 고정된 기판에 가하여 제 1 단백질이 고정된기판을준비하는단계 ;  (1 ') preparing a substrate to which a first protein is immobilized by applying a reference sample containing a first protein to a fixed substrate comprising a substance that specifically binds to the first protein on a surface thereof;
5 (2 ) 상기 준비된 제 1단백질이 고정된 기판에 표지된 제 2단백질을 첨가하여 반응시키는단계; 5 (2) adding the labeled second protein to the substrate on which the prepared first protein is immobilized and reacting;
(3' )상기 (2’)단계에서 얻어진반응물로부터 신호를측정하는단계 (단백질-단백질상호작용측정 ) ; 및  (3 ') measuring a signal from the reaction product obtained in the step (2') (measurement of protein-protein interaction); And
(4' ) 상기 측정된신호를 이용하여 상기 (1' ) 단계에서 첨가한기준 10 시료에 포함된제 1단백질의 양으로나누는단계 (활성화수준측정) (Measurement of the level of activation) by the amount of the first protein contained in the reference 10 sample added in the step (1 ') using the measured signal,
(또는 (4-1 ' ) 상기 측정된 신호를 이용하여 상기 (1’) 단계에서 첨가한 기준 시료의 중량 또는 농도로 나누는 단계 (단백질-단백질 상호작용수준측정 단계) 및 (4-2' ) 상기 단계 (4-1’)에서 얻어진 결과를 기준시료에 포함된 제 1단백질의 양으로나누는단계 (활성화수준측정)) . 15 각단계의 상세한사항은앞서 시험 시료에 대한각단계에서 설명한 바와같다. (4-1 ') dividing the measured signal by the weight or concentration of the reference sample added in the step (1') (measuring the protein-protein interaction level) and (4-2 ' Dividing the result obtained in the step (4-1 ') by the amount of the first protein contained in the reference sample (measurement of the activation level)). 15 Details of each step are as described in the previous sections for the test sample.
단계 (6)  Step (6)
본 명세서에서 제공되는 방법은, 상기 단계 (5) 이후에, 단계 (5)에서 얻어진 비교 결과로부터 목적하는 사항을 확인 (결정)하는 단계를 20 추가로포함할수있다. Methods provided herein, may be included the step of: after the step (5), step (5) determine (determine) the locations of interest from the comparison results obtained in a 20 added.
이를보다구체적으로살펴보면다음과같다:  More specifically, it is as follows:
Figure imgf000041_0001
Figure imgf000041_0001
30 상호작용 수준 또는 활성화 수준이 기준 시료에서 측정된 단백질-단백질 상호작용수준또는활성화수준보다높은경우, 시험 시료또는상기 시험 시료가 유래하는 개체에서 제 1 단백질이 관여하는 신호전달경로의 활성화 정도가정상세포의 활성화정도또는기준시료에서 알려진 (확인된) 활성화 2019/132517 1»(:1^1{2018/016675 3 0 The degree of activation of the signal transduction pathway involved in the first protein in the test sample or the individual from which the test sample is derived when the interaction level or activation level is higher than the protein-protein interaction level or activation level measured in the reference sample The degree of activation of the home specimen or the known (confirmed) activation in the reference sample 2019/132517 1 »(: 1 ^ {2018/016675
정도보다높다고확인 (결정)하는단계; (Determination) that the current time is higher than the predetermined time;
단계 (4) 또는 (4-2)에서 측정된 시험 시료의 단백질-단백질 상호작용 수준 또는 활성화 수준이 기준 시료에서 측정된 단백질-단백질 상호작용수준또는활성화수준과동등한경우, 시험 시료또는상기 시험 시료가 유래하는 개체에서 제 1 단백질이 관여하는 신호전달경로의 활성화 정도가정상세포의 활성화정도또는기준시료에서 알려진 (확인된) 활성화 정도와동등하다고확인 (결정)하는단계; 및/또는  When the protein-protein interaction level or activation level of the test sample measured in step (4) or (4-2) is equivalent to the protein-protein interaction level or activation level measured in the reference sample, the test sample or the test sample (Determining) the degree of activation of the signal transduction pathway involved in the first protein in the individual from which the first protein is derived is equal to the degree of activation of the normal cell or the degree of activation (known) known in the reference sample; And / or
단계 (4) 또는 (4-2)에서 측정된 시험 시료의 단백질-단백질 상호작용 수준 또는 활성화 수준이 기준 시료에서 측정된 단백질-단백질 상호작용수준또는활성화수준보다낮은경우, 시험 시료또는상기 시험 시료가 유래하는 개체에서 제 1 단백질이 관여하는 신호전달경로의 활성화 정도가정상세포의 활성화정도또는기준시료에서 알려진 (확인된) 활성화 정도보다낮다고확인 (결정)하는단계 .  When the protein-protein interaction level or activation level of the test sample measured in step (4) or (4-2) is lower than the protein-protein interaction level or activation level measured in the reference sample, the test sample or the test sample (Determining) the degree of activation of the signal transduction pathway involved in the first protein in the individual from which the first protein is derived is lower than the degree of activation of the normal cell or the known (confirmed) activation level in the reference sample.
( { [ ) 제 1 단백질을 표적으로 하는 약물에 대한 반응성을 예측하는 방법 또는예측에 정보를제공하는방법  ({≪ - >) method for predicting the response to a drug targeting a first protein or providing information on the prediction
단계 (6)은다음의 단계를포함하는것일수있다:  Step (6) may include the following steps:
단계 (4) 또는 (4-2)에서 측정된 시험 시료의 단백질-단백질 상호작용 수준 또는 활성화 수준이 기준 시료에서 측정된 단백질-단백질 상호작용수준또는활성화수준보다높은경우, 시험 시료또는상기 시험 시료가유래하는개체의 제 1단백질을표적으로하는 약물에 대한반응성이 기준시료의 약물반응성 보다높다고확인 (결정)하는단계;  When the protein-protein interaction level or activation level of the test sample measured in step (4) or (4-2) is higher than the protein-protein interaction level or activation level measured in the reference sample, the test sample or the test sample (Determining) the reactivity of the first protein of the individual derived from the first sample to the drug that targets the first protein is higher than the drug reactivity of the reference sample;
단계 (4) 또는 (4-2)에서 측정된 시험 시료의 단백질-단백질 상호작용 수준 또는 활성화 수준이 기준 시료에서 측정된 단백질-단백질 상호작용수준또는활성화수준과동등한경우, 시험 시료또는상기 시험 시료가유래하는개체의 제 1단백질을표적으로하는약물에 대한반응성이 기준시료의 약물반응성과동등하다고확인 (결정)하는단계 ; 및/또는  When the protein-protein interaction level or activation level of the test sample measured in step (4) or (4-2) is equivalent to the protein-protein interaction level or activation level measured in the reference sample, the test sample or the test sample (Determining) the reactivity of the first protein of the individual derived from the target sample to the drug that targets the drug is equivalent to the drug response of the reference sample; And / or
단계 (4) 또는 (4-2)에서 측정된 시험 시료의 단백질-단백질 상호작용 수준 또는 활성화 수준이 기준 시료에서 측정된 단백질-단백질 상호작용수준또는활성화수준보다낮은경우, 시험 시료또는상기 시험 시료가유래하는개체의 제 1단백질을표적으로하는 약물에 대한반응성이 기준시료의 약물반응성보다낮다고확인 (결정)하는단게.  When the protein-protein interaction level or activation level of the test sample measured in step (4) or (4-2) is lower than the protein-protein interaction level or activation level measured in the reference sample, the test sample or the test sample (Determination) that the reactivity of the first protein of the individual derived from the target is lower than the drug reactivity of the reference sample.
기준 시료를 시험 대상 개체에 요구되는 정도의 제 1 단백질을 표적으로 하는 약물에 대한 반응성을 갖는 세포를 포함하도록 선정하여, 2019/132517 1»(:1^1{2018/016675 The reference sample is selected so as to include cells having reactivity to the drug targeting the first protein to a degree required for the subject to be tested, 2019/132517 1 »(: 1 ^ {2018/016675
상기 약물이 시험 시료 또는 상기 시험 시료가 유래한 시험 대상 개체에 소망하는 효과를 갖는지 여부를 조사하거나, 기준 시료를 정상세포로 선정하여 상기 약물이 시험 시료 또는 시험 대상 개체에서 정상 세포보다 제 1단백질과관련된 질병에 특이적으로작용하는지 여부를조사할수 있다. 예컨대, 시험 대상 개체에 요구되는 정도의 제 1 단백질을 표적으로 하는 약물에 대한반응성을갖는세포를상기 기준시료로선정하는 경우, 단계 (6)은 단계 (4) 또는 (4-2)에서 측정된 시험 시료의 단백질-단백질 상호작용 수준 또는 활성화 수준이 기준 시료에서 측정된 단백질-단백질 상호작용수준또는활성화수준과비교하여 동등이상, 예컨대, 높은경우, 시험 시료또는상기 시험 시료가유래하는 개체의 제 1 단백질을표적으로 하는 약물에 대한 반응성이 우수하거나, 및/또는 상기 약물이 시험 시료 또는상기 시험 시료가유래하는 개체에 효과를 갖는다고 결정하는단계를 포함할수있다. It is possible to examine whether the drug has a desired effect on the test sample or the test subject from which the test sample is derived, or to select the reference sample as a normal cell, Specific illnesses < RTI ID = 0.0 > associated with < / RTI > For example, when a cell having a reactivity to a drug targeting a first protein to a degree required for a subject to be tested is selected as the reference sample, step (6) is performed in step (4) or For example, when the protein-protein interaction level or the activation level of the test sample is equal to or higher than the protein-protein interaction level or the activation level measured in the reference sample, the test sample or the sample Determining whether the drug is more reactive to the drug targeting the first protein and / or that the drug is effective on the test sample or on the individual from which the test sample is derived.
상기 제 1 단백질을 표적으로 하는 약물에 대한 반응성을 예측하는 방법 또는예측에 정보를제공하는방법에 있어서, 단계 (6)에서 시험 시료 또는 상기 시험 시료가 유래하는 개체의 제 1 단백질을 표적으로 하는 약물에 대한 반응성이 우수하거나, 및/또는 상기 약물이 시험 시료 또는 상기 시험 시료가 유래하는 개체에 효과를 갖는다고 결정된 경우, 상기 개체에게 제 1 단백질을 표적으로 하는 약물을 투여하는 단계를 추가로 포함할수있다.  A method for predicting reactivity to a drug or a method for predicting reactivity to a drug that targets the first protein, comprising the steps of: (a) providing a test sample or a first protein of the individual from which the test sample is derived Administering a drug targeting the first protein to the subject, if it is determined that the drug has an excellent response to the drug, and / or that the drug has an effect on the test sample or an individual from which the test sample is derived You can include it.
다른 측면에서, 상기의 결정 내용을 기초로 각 개체에 적합한 개별 맞춤 치료 수단이 제공된다. 일 예는 상기 제 1 단백질을 표적으로 하는 약물을 유효성분으로 포함하는, 상기 단계 (6)에서 제 1 단백질을 표적으로 하는 약물에 대한 반응성이 우수하거나, 및/또는 상기 약물이 효과를 갖는다고결정된 개체에서의 제 1단백질과관련된 질병의 치료를위한약학 조성물을제공한다. 다른 예는상기 제 1 단백질을표적으로 하는 약물의, 상기 단계 (6)에서 제 1 단백질을 표적으로 하는 약물에 대한 반응성이 우수하거나, 및/또는 상기 약물이 효과를 갖는다고 결정된 개체에서의 제 1 단백질과 관련된 질병의 치료를 위한 용도를 제공한다. 다른 예는 상기 단계 (6)에서 제 1 단백질을 표적으로 하는 약물에 대한 반응성이 우수하거나, 및/또는 상기 약물이 효과를 갖는다고 결정된 개체에게 상기 제 1 단백질을 표적으로 하는 약물을 투여하는 단계를 포함하는, 상기 개체에서의 저ᅵ 1단백질과관련된질병의 치료방법을제공한다. 2019/132517 1»(:1^1{2018/016675 On the other side, individual customized treatment means suitable for each individual are provided based on the above decision content. One example is that the drug is excellent in reactivity to a drug targeting the first protein in step (6), which comprises a drug targeting the first protein as an active ingredient, and / or the drug has an effect A pharmaceutical composition for the treatment of a disease associated with a first protein in a determined individual. Another example is the use of a drug targeting the first protein, an agent having an excellent reactivity to the drug targeting the first protein in step (6), and / or an agent in the subject determined to be effective 1 < / RTI > protein-related diseases . Another example is the step of administering a drug targeting the first protein to an individual having an excellent response to the drug targeting the first protein in step (6) and / or determining that the drug is effective and it provides a method of treating diseases which are associated with the low-i 1 protein in the object containing the. 2019/132517 1 »(: 1 ^ {2018/016675
0 ) 제 1 단백질을 표적으로 하는 치료에 적합한 개체를 선별하는 방법 또는선별에 정보를제공하는방법 0) a method for screening an object suitable for treatment targeting the first protein or a method for providing information to screening
상기 기준시료는정상세포또는상기 제 1단백질을표적으로하는 치료의 효과가 알려진(확인된) 세포 (예컨대, 암세포)를포함하거나, 상기 5 시험 시료가 개체로부터 분리된 암 세포를 포함하는 경우, 암세포와 동일 조직 또는동일기관의 정상세포를포함하는것일수있다.  The reference sample includes normal cells or cells (for example, cancer cells) whose effects of treatment targeting the first protein are known (confirmed), or when the fifth test sample contains cancer cells isolated from the subject, And may include normal cells of the same tissue or the same organ as the cancer cells.
단계 (6)은 단계 (4) 또는 (4-2)에서 측정된 시험 시료의 단백질- 단백질 상호작용 수준 또는 활성화 수준이 기준 시료에서 측정된 단백질- 단백질 상호작용 수준 또는 활성화 수준과 비교하여 동등 이상, 예컨대,0 높은 경우, 시험 시료 또는 상기 시험 시료가 유래하는 개체를 제 1 단백질을 표적으로 하는 치료에 적합한 환자로 확인 (결정)하는 단계를 포함할수있다.  Step (6) is a step (6) wherein the protein-protein interaction level or activation level of the test sample measured in step (4) or (4-2) is compared with the protein- For example, 0, the test sample or the individual from which the test sample is derived may be identified (determined) as a patient suitable for treatment targeting the first protein.
상기 제 1 단백질을 표적으로 하는 치료는 제 1 단백질을 표적으로 하는약물을처방및/또는투여하는것을의미할수있다. Treatment targeting the first protein may mean prescribing and / or administering a drug targeting the first protein.
5 상기 기준 시료는 제 1 단백질을 표적으로 하는 치료가효과를 갖는 세포를포함하는것일수있다. 5 The reference sample may include a cell having a therapeutic effect that targets the first protein.
상기 제 1 단백질을 표적으로 하는 치료에 적합한 개체를 선별하는 방법 또는선별에 정보를제공하는방법에 있어서, 단계 (6)에서 시험 시료 또는 상기 시험 시료가 유래하는 개체가 제 1 단백질을 표적으로 하는0 치료에 적합한 환자로 확인 (결정)된 경우, 상기 개체에게 제 1 단백질을 A method for screening a subject suitable for treatment targeting the first protein or a method for providing information to screening, wherein in step (6), the test sample or the individual from which the test sample is derived is selected from the group consisting of 0 < / RTI > identified as a patient suitable for treatment, the subject is treated with a first protein
_ 표적으로 하는 치료를 수행 (예컨대, 제 1 단백질을 표적으로 하는 약물을 처방및/또는투여)하는단계를추가로포함할수있다. _ Perform the treatment of the target can contain the further step of (e. G., A drug for the first protein with the target prescription and / or dosage).
다른측면에서, 상기의 확인 (결정)내용을기초로각개체에 적합한 개별 맞춤 치료 수단이 제공된다. 일 예는 상기 제 1 단백질을 표적으로5 하는 약물을 유효성분으로 포함하는, 상기 단계 (6)에서 제 1 단백질을 표적으로 하는 치료에 적합한 것으로 확인된 개체에서의 제 1 단백질과 관련된 질병의 치료를위한약학조상물을제공한다. 다른 예는상기 제 1 단백질을표적으로하는 약물의, 상기 단계 (6)에서 체 1단백질을표적으로 하는치료에 적합한것으로확인된 개체에서의 제 1단백질과관련된 질병의0 치료를위한용도를제공한다. 다른예는상기 단계 (6)에서 제 1단백질을 표적으로 하는 치료에 적합한 것으로 확인된 개체에게 상기 제 1 단백질을 표적으로 하는 치료를 수행 (예컨대, 제 1 단백질을 표적으로 하는 약물을 처방 및/또는 투여)하는 단계를 포함하는, 상기 개체에서의 제 1 단백질과 2019/132517 1»(:1^1{2018/016675 On the other side, individual personalized treatment means suitable for each individual are provided based on the above-mentioned confirmation (decision) contents. One example is a method for treating a disease associated with a first protein in an individual identified as being suitable for treatment targeting the first protein in step (6), comprising the step of administering a drug that targets the first protein as an active ingredient To provide a pharmacological ancestor for. Another example provides the use of a drug targeting said first protein for the treatment of a disease associated with a first protein in said individual identified as being suitable for treatment targeting said s1 protein in said step (6) do. Another example is to perform treatment targeting the first protein to a subject identified as suitable for treatment targeting the first protein in step (6) (e.g., administering a drug targeting the first protein and / Or administration) of the first protein in the subject, 2019/132517 1 »(: 1 ^ {2018/016675
관련된질병의 치료방법을제공한다. And provides a method of treating a related disease.
(^) 제 1 단백질을 표적으로 하는 치료의 효과 (제 1 단백질을 표적으로 하는 약물와 반응성)을 모니터링하는 방법 또는 모니터링에 정보를제공하는방법  (^) A method for monitoring the effect of a treatment targeting a first protein (reactivity with a drug targeting the first protein) or a method for providing information to monitoring
상기 기준시료는정상세포또는상기 제 1단백질을표적으로하는 치료의 효과가 알려진(확인된) 세포 (예컨대, 암세포)를 포함하거나, 상기 시험 시료가 개체로부터 분리된 암 세포를 포함하는 경우, 암세포와 동일 조직 또는동일기관의 정상세포를포함하는것일수있다.  The reference sample includes normal cells or cells (for example, cancer cells) for which the effect of the treatment targeting the first protein is known (confirmed), or when the test sample contains cancer cells isolated from an individual, And normal cells of the same organ or the same organ.
단계 (6)은 단계 (4) 또는 (4-2)에서 측정된 시험 시료의 단백질- 단백질 상호작용 수준 또는 활성화 수준이 기준 시료에서 측정된 단백질- 단백질 상호작용 수준 또는 활성화 수준과 비교하여 동등 이상, 예컨대, 높은 경우, 시험 시료 또는 상기 시험 시료가 유래하는 개체를 저 단백질을 표적으로 하는 치료가 효과를 발휘하고 있는 것 (예컨대, 제 1 단백질을 표적으로 하는 약물의 투여 후, 투여받은 개체에서 상기 약물에 대한반응성이 유지되고 있는것 또는투여받은개체에서 상기 약물에 대한 저항성이 발생하지 않은것)으로확인 (결정)하는단계를포함할수있다. 상기 제 1 단백질을 표적으로 하는 치료는 제 1 단백질을 표적으로 하는약물을처방및/또는투여하는것을의미할수있다.  Step (6) is a step (6) wherein the protein-protein interaction level or activation level of the test sample measured in step (4) or (4-2) is compared with the protein- , For example, when the test sample or an object to which the test sample is derived is subjected to treatment with a low protein targeting effect (for example, after administration of a drug targeting the first protein, The response to the drug is maintained or the drug does not develop resistance to the drug). Treatment targeting the first protein may mean prescribing and / or administering a drug targeting the first protein.
상기 기준 시료는 제 1 단백질을 표적으로 하는 치료가효과를 갖는 세포를포함하는것일수있다.  The reference sample may include a cell having a therapeutic effect that targets the first protein.
상기 제 1단백질을표적으로하는치료의 효과를모니터링 하는방법 또는모니터링에 정보를 제공하는 방법에 있어서, 단계 (6)에서 시험 시료 또는 상기 시험 시료가 유래하는 개체에서 제 1 단백질을 표적으로 하는 치료의 효과가 발휘되고 있는 것으로 확인 (결정)된 경우, 상기 개체에게 상기 제 1 단백질을 표적으로 하는 치료를 계속적으로 수행 (예컨대, 저 11 단백질을 표적으로 하는 약물을 처방 및/또는 투여)하는 단계 또는 단계 (6)에서 시험 시료또는상기 시험 시료가유래하는개체에서 제 1단백질을 표적으로하는치료의 효과가발휘되지 않는 것 (예컨대, 치료 효과 (약물 반응성) 감소 또는 저항성 획득 등)으로 확인 (결정)된 경우, 상기 개체에게 상기 제 1 단백질을표적으로 하는 치료를중단하는 단계 및/또는 제 1 단백질을표적으로 하는 다른 약물을투여하거나상이한 제 1단백질을 표적으로하는치료를수행하는단계를추가로포함할수있다.  A method for monitoring the effect of treatment targeting the first protein or a method for providing information to monitoring, comprising the steps of: (a) administering a test sample or a first protein to a subject in a step (6) (For example, prescribing and / or administering a drug targeting a low protein) to the subject when it is confirmed that the effect of the low protein is exerted Or in the step (6), the test sample or an object to which the test sample is derived does not exhibit the effect of treatment targeting the first protein (for example, reduction in therapeutic effect (drug reactivity) or acquisition of resistance, etc.) The method comprising: stopping treatment to target the first protein to the subject, and / or stopping the first protein Administering another drug to be targeted, or performing a treatment targeting a different first protein.
다른 예는 제 1 단백질과 제 2 단백질 간 단백질-단백질 상호작용을 2019/132517 1»(:1^1{2018/016675 Another example is protein-protein interaction between a first protein and a second protein 2019/132517 1 »(: 1 ^ {2018/016675
측정하는단계를포함하고, 상기 제 1단백질은세포또는조직 내 및/또는 세포또는조직 간의 신호전달경로에 관여하는단백질이고, 제 2 단백질은 상기 세포 또는 조직 내 및/또는 세포 또는 조직 간 의 신호전달경로 중에서 상기 제 1 단백질의 하류 단백질이고, 상기 단백질-단백질 상호작용을 측정하는 단계를 2개 이상의 제 1 단백질에 대하여 수행하는, 상기 세포 또는 조직 또는 상기 세포 또는 조직이 유래하는 개체 (개별 환자)에 적용하기에 적합한 치료 표적으로서의 제 1 단백질 또는 이를 표적으로 하는 약물을선별하는방법 또는 선별에 정보를 제공하는 방법을 제공한다. Wherein the first protein is a protein involved in a signal transduction pathway in a cell or tissue and / or between a cell or tissue, and the second protein is a protein in the cell or tissue and / Wherein the step of measuring the protein-protein interaction is performed on two or more first proteins, wherein the cells or tissues or the individual from which the cells or tissues are derived A method for screening a first protein or a drug targeting the first protein as a therapeutic target suitable for application to a subject, or providing information on screening.
상기 방법은,  The method comprises:
(1) 제 1 단백질을포함하는 시험 시료를 표면에 상기 제 1 단백질에 특이적으로 결합하는 물질을포함하는 기판에 가하여 제 1 단백질이 고정된 기판을준비하는단계;  (1) preparing a substrate to which a first protein is immobilized by adding a test sample containing a first protein to a substrate including a substance that specifically binds to the first protein on a surface thereof;
(2) 상기 준비된 제 1 단백질이 고정된 기판에 표지된 제 2 단백질을 첨가하여 반응시키는단계;  (2) adding a labeled second protein to the substrate on which the prepared first protein is immobilized and reacting;
(3) 단계 (2)에서 얻어진 반응물로부터 신호를 측정하는 단계 (3) measuring the signal from the reactant obtained in step (2)
(단백질-단백질상호작용측정) ; 및 (Protein-protein interaction measurement); And
(4)단계 (3)에서 측정된신호를이용하여 단계 (1)에서 첨가한시함 시료 포함된 제 1 단백질의 단위량에 대한 값을 구하는 단계 (활성화수준 측정) ; 또는  (4) obtaining a value for the unit amount of the first protein included in the sample added in step (1) (measuring the activation level) using the signal measured in step (3); or
(4-1) 단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한 시험 시료 의 단위량에 대한 신호값을 구하는 단계 (단백질-단백질 상호작용 수준 측정) , 및 (4-2) 단계 (4-1)에서 얻어진 시험 시료의 단위량에 대한 신호값을 이용하여 시험 시료에 포함된 제 1 단백질의 단위량에 대한값을구하는단계 (활성화수준측정) ; 및  (4-1) obtaining a signal value (measurement of protein-protein interaction level) for the unit amount of the test sample added in step (1) using the signal measured in step (3), and (4-2 Obtaining a value for a unit amount of the first protein included in the test sample (measuring the activation level) using the signal value of the unit amount of the test sample obtained in the step (4-1); And
(5) 2 이상의 제 1 단백질에 대하여 상기 단계 (4) 또는 단계 (4- 2)에서 얻어진결과를서로비교하는단계 (5) comparing the results obtained in step (4) or step (4- 2) with respect to two or more first proteins
를포함할수있다.  .
이 때, 상기 제 1 단백질로서 세포 또는 조직의 신호전달경로에 관여하는단백질중에서 선택되는 2종이상이 사용되고,  At this time, as the first protein, two or more proteins selected from proteins involved in signal transduction pathways of cells or tissues are used,
단계 ( 1), (2), (3) 및 (4) , 또는 단계 (1) , (2) , (3), (4-1), 및 (4-2)는 2종이상의 제 1단백질각각에 대하여 수행되며,  (1), (2), (3) and (4) or steps (1), (2), (3), (4-1) Respectively,
단계 (5)의 비교하는단계는 2종이상의 제 1단백질에 대하여 얻어진 2019/132517 1»(:1^1{2018/016675 The comparing step of step (5) is performed on two or more species of the first protein 2019/132517 1 »(: 1 ^ {2018/016675
결과를서로비교하는것일수있다. The results can be compared to each other.
상기 단계 (5)에서의 비교 결과, 단백질-단백질 상호작용수준또는 활성화수준이 높은제 1단백질을상기 시험 시료또는시험 시료가유래한 개체의 치료 표적으로 선택하거나, 상기 제 1 단백질을 표적으로 하는 약물을 시험 시료 또는 시험 시료가 유래한 개체의 치료를 위한 후보 약물로선택하는단계 (단계 (6))를추가로포함할수있다.  As a result of the comparison in the step (5), a first protein having a high protein-protein interaction level or activation level is selected as a therapeutic target of the test sample or a test sample derived from the test sample, (Step (6)) of selecting the drug as a candidate drug for the treatment of a test sample or an individual from which the test sample is derived.
상기 단계 (1) 내지 (6) 및, 시험 시료, 제 1 단백질, 제 2 단백질 등의 용어 설명은앞서 기재한바와같다.  The description of the steps (1) to (6) and the test sample, the first protein, the second protein and the like are as described above.
다른 예는 제 1 단백질과 제 2 단백질 간 단백질-단백질 상호작용을 측정하는단계를포함하고, 상기 제 1단백질은세포또는조직 내 및/또는 세포 또는 조직 간의 신호전달경로에 관여하는 단백질이고, 제 2 단백질은 상기 세포또는조직 내 및/또는세포또는조직 간의 신호전달경로중에서 상기 제 1 단백질의 하류 단백질이고, 상기 단백질-단백질 상호작용을 측정하는단계를후보물질 처리 전 및 처리 후에 수행하는, 제 1단백질을 표적으로 하는 후보 약물의 선별 방법 또는 제 1 단백질을 표적으로 하는 후보약물의 효능확인방법을제공한다.  Another example includes measuring a protein-protein interaction between a first protein and a second protein, wherein the first protein is a protein involved in a signal transduction pathway in a cell or tissue and / or between cells or tissue, 2 protein is a downstream protein of the first protein in the cell or tissue and / or in a signal transduction pathway between cells or tissues, and the step of measuring the protein-protein interaction is performed before and after the candidate substance treatment. 1 < / RTI > protein, or a method for confirming the efficacy of a candidate drug targeting the first protein.
상기 방법은,  The method comprises:
시험 시료에 후보화합물을처리하는 (예컨대, 접촉시키는)단계, 및 상기 후보화합물이 처리되지 않은 시험 시료 및 처리된 시험 시료 각각에 대하여 하기의 단계 (1), (2) , (3) , 및 (5) , 또는 (1) , (2), (3), (4), 및 (5) 또는 (1), (2) , (3) , (4-1) , 및 (5) , 또는 (1) , (2) , (3) , (4-1) , (4-2) , 및 (5)를수행하는단계를포함할수있다:  (1), (2), (3), and (3) for each of the test sample that has not been treated with the candidate compound and the treated test sample, (3), (4), and (5) or (1), (2) (1), (2), (3), (4-1), (4-2), and (5)
(1) 제 1 단백질을포함하는 시험 시료를 표면에 상기 제 1 단백질에 특이적으로 결합하는 물질을 포함하는 기판에 가하여 제 1 단백질이 고정된 기판을준비하는단계;  (1) preparing a substrate to which a first protein is immobilized by adding a test sample containing a first protein to a substrate including a substance that specifically binds to the first protein on a surface thereof;
(2) 상기 준비된 제 1 단백질이 고정된 기판에 표지된 제 2 단백질을 첨가하여 반응시키는단계;  (2) adding a labeled second protein to the substrate on which the prepared first protein is immobilized and reacting;
(3) 단계 (2)에서 얻어진 반응물로부터 신호를 측정하는 단계 (단백질-단백질상호작용측정) ; 및  (3) measuring a signal from the reactant obtained in step (2) (protein-protein interaction measurement); And
(4)단계 (3)에서 측정된신호를이용하여 단계 ( 1)에서 첨가한시험 시료 포함된 제 1 단백질의 단위량에 대한 값을 구하는 단계 (활성화수준 측정 ) ;또는  (4) obtaining a value for a unit amount of the first protein included in the test sample added in the step (1) (measuring the activation level) using the signal measured in the step (3); or
(4-1) 단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한 2019/132517 1»(:1^1{2018/016675 (4-1) Using the signals measured in step (3), the signals added in step (1) 2019/132517 1 »(: 1 ^ {2018/016675
시험 시료 의 단위량에 대한 신호값을 구하는 단계 (단백질-단백질 상호작용 수준 측정) , 및 (4-2) 단계 (4-1)에서 얻어진 시험 시료의 단위량에 대한 신호값을 이용하여 시험 시료에 포함된 제 1 단백질의 단위량에 대한값을구하는단계 (활성화수준측정) ; 및 Using the signal values for the unit amount of the test sample obtained in step (4-1) and the step of obtaining the signal value for the unit amount of the test sample (measurement of protein-protein interaction level) and (4-2) Obtaining a value for a unit amount of the first protein contained in the sample (measurement of the activation level); And
(5) 상기 후보 화합물이 처리되지 않은 시험 시료 및 처리된 시험 시료에 대하여 상기 단계 (3) , 단계 (4) , 단계 (4-1) , 또는 단계 (4- 2)에서 얻어진각각의 결과를서로비교하는단계.  (5) The respective results obtained in step (3), step (4), step (4-1), or step (4- 2) for the test sample not treated with the candidate compound and the treated test sample Comparing them to each other.
상기 후보화합물이 처리되지 않은시험 시료와처리된 시험 시료는 각각후보화합물의 처리 전시험 시료및 처리 후시험 시료를의미하거나, 시험 시료를 분량하여 상기 후보 화합물을 처리한 일부의 시험 시료 및 상기 후보 화합물을 처리하지 않은 다른 일부의 시험 시료를 의미할 수 있다.  The test sample not treated with the candidate compound and the treated test sample means the test sample before treatment and the test sample after treatment of the candidate compound, respectively, or a part of the test sample treated with the candidate compound and the candidate May refer to some other test sample that has not been treated with the compound.
상기 단계 (5)에서의 비교 결과, 후보화합물이 처리된 시험 시료의 단백질-단백질 상호작용 수준 또는 활성화수준이 후보 화합물이 처리되지 않은시험 시료에서보다낮은경우, 상기 후보화합물을상기 제 1단백질을 표적으로 하는후보 약물로선택하거나, 상기 후보 화합물이 제 1 단백질을 표적으로하는약물로서 효과가있는것으로확인할수있다.  If the result of the comparison in the step (5) is that the protein-protein interaction level or activation level of the test sample treated with the candidate compound is lower than that of the test sample in which the candidate compound is not treated, The candidate compound may be selected as a candidate candidate drug, or the candidate compound may be effective as a drug targeting the first protein.
따라서, 상기 제 1 단백질을 표적으로 하는 후보 약물의 선별 방법 또는 제 1 단백질을 표적으로 하는 후보 약물의 효능 확인 방법은, 상기 단계 (5) 이후에, 상기 단계 (5)에서의 비교 결과, 후보 화합물이 처리된 시험 시료의 단백질-단백질 상호작용 수준 또는 활성화 수준이 후보 화합물이 처리되지 않은시험 시료에서보다낮은경우, 상기 후보화합물을 상기 제 1 단백질을 표적으로 하는 후보 약물로 선택하거나, 상기 후보 화합물이 제 1 단백질을 표적으로 하는 약물로서 효과가 있는 것으로 확인하는단계 (단계 (6) )를추가로포함할수있다.  Therefore, the method for selecting a candidate drug targeting the first protein or the method for confirming the efficacy of a candidate drug targeting the first protein can be performed after the step (5), as a result of the comparison in the step (5) When the compound-protein interaction level or activation level of the test sample treated with the compound is lower than that of the test sample in which the candidate compound is not treated, the candidate compound is selected as a candidate drug targeting the first protein, (Step (6)) of confirming that the compound is effective as a drug targeting the first protein.
일 예는 제 1 단백질과 제 2 단백질 간 단백질-단백질 상호작용을 측정하는 단계를 포함하고, 상기 제 1 단백질은
Figure imgf000048_0001
및 ?0-1 중에서 선택된 어느 하나이고, 제 2 단백질은 다른 하나이고, 상기 단백질-단백질 상호작용을 측정하는 단계를 후보 물질 처리 전 및 처리 후에 수행하는, 제 1 단백질 및/또는 제 2 단백질을 표적으로 하는 후보 약물의 선별 방법, 또는: 제 1 단백질 및/또는 제 2 단백질을 표적으로 하는 후보 약물의 효능 확인 방법을 제공한다. 다른 예는 제 1 단백질과 제 2 단백질 간 단백질- 단백질 상호작용을측정하는단계를포함하고, 상기 제 1단백질은 나및 2019/132517 1»(:1^1{2018/016675
One example includes measuring protein-protein interactions between a first protein and a second protein, wherein the first protein comprises
Figure imgf000048_0001
And? 0-1, the second protein is another one, and the step of measuring the protein-protein interaction is carried out before and after the candidate substance treatment, wherein the first protein and / or the second protein It provides the first protein and / or the second method confirmed the efficacy of candidate drugs which target proteins: the candidate drug to target screening methods, or. Another example comprises measuring a protein-protein interaction between a first protein and a second protein, 2019/132517 1 »(: 1 ^ {2018/016675
?0-1 중에서 선택된 어느 하나이고, 제 2 단백질은 다른 하나이고, 상기 단백질-단백질 상호작용을 측정하는 단계를 후보 물질 처리 전 및 처리 후에 수행하는, 제 1 단백질 및/또는 제 2 단백질과 관련된 질병 (예컨대, 제 1 단백질 및/또는 제 2단백질의 (과)발현 및/또는 (과)활성화와 관련된 질병; 암, 면역 질환 등)의 예방 또는 치료을 위한 후보 약물의 선별 방법을제공한다. 0-1, and the second protein is another one, and the step of measuring the protein-protein interaction is performed before and after the candidate substance treatment, wherein the first protein and / or the second protein The present invention provides a method for screening candidate drugs for the prevention or treatment of diseases (e.g., diseases associated with the expression and / or activation of the first protein and / or the second protein; cancer, immune diseases, etc.).
상기 방법은,  The method comprises:
시험 시료에 후보화합물을처리하는 (예컨대, 접촉시키는)단계, 및 상기 후보 화합물이 처리되지 않은시험 시료 및 처리된 시험 시료 각각에 대하여 하기의 단계 (1), (2), (3), 및 (5), 또는 단계 (1), (2), (3), (4), 및 (5), 또는 단계 (1), (2), (3) , (4-1) , 및 (5), 또는 단계 (1), (2), (3), and (3) for each of the test sample that has not been treated with the candidate compound and the treated test sample, (1), (2), (3), (4), and (5) ), Or step
(1), (2), (3), (4-1) , (4-2) , 및 (5)를수행하는단계를포함할수있다: (1), (2), (3), (4-1), (4-2), and (5)
(1) 제 1 단백질을포함하는 시험 시료를 표면에 상기 제 1 단백질에 특이적으로 결합하는 물질을포함하는 기판에 가하여 제 1 단백질이 고정된 기판을준비하는단계 ;  (1) preparing a substrate to which a first protein is immobilized by adding a test sample containing a first protein to a substrate including a substance that specifically binds to the first protein on a surface thereof;
(2) 상기 준비된 제 1 단백질이 고정된 기판에 표지된 제 2 단백질을 첨가하여 반응시키는단계;  (2) adding a labeled second protein to the substrate on which the prepared first protein is immobilized and reacting;
(3) 단계 (2)에서 얻어진 반응물로부터 신호를 측정하는 단계 (단백질-단백질상호작용측정) ; 및  (3) measuring a signal from the reactant obtained in step (2) (protein-protein interaction measurement); And
(4)단계 (3)에서 측정된신호를이용하여 단계 (1)에서 첨가한시험 시료 포함된 제 1 단백질의 단위량에 대한 값을 구하는 단계 (활성화 수준 측정) ; 또는  (4) obtaining a value for the unit amount of the first protein included in the test sample added in step (1) (measuring the activation level) using the signal measured in step (3); or
(4-1) 단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한 시험 시료 의 단위량에 대한 신호값을 구하는 단계 (단백질-단백질 상호작용 수준 측정), 및 (4-2) 단계 (4-1)에서 얻어진 시험 시료의 단위량에 대한 신호값을 이용하여 시험 시료에 포함된 제 1 단백질의 단위량에 대한값을구하는단계 (활성화수준측정) ; 및  (4-1) obtaining a signal value (measurement of protein-protein interaction level) for the unit amount of the test sample added in step (1) using the signal measured in step (3), and (4-2 Obtaining a value for a unit amount of the first protein included in the test sample (measuring the activation level) using the signal value of the unit amount of the test sample obtained in the step (4-1); And
(5) 상기 후보 화합물이 처리되지 않은 시험 시료 및 처리된 시험 시료에 대하여 상기 단계 (3), 단계 (4), 단계 (4-1) , 또는 단계 (4- 2)에서 얻어진각각의 결과를서로비교하는단계.  (5) The respective results obtained in step (3), step (4), step (4-1), or step (4- 2) for the test sample not treated with the candidate compound and the treated test sample Comparing them to each other.
각단계의 구체적 설명은앞서 설명한바와같다.  The concrete description of each step is as described above.
상기 단계 (5)에서의 비교결과, 후보화합물이 처리된 시험 시료의 단백질-단백질 상호작용 수준 또는 활성화수준이 후보 화합물이 처리되지 않은 시험 시료에서보다 낮은 경우, 상기 후보 화합물을 제 1 단백질을 표적으로 하는후보 약물로 선택하거나, 상기 후보 화합물이 제 1 단백질을 표적으로하는 약물로서 효과가 있는 것으로 확인할수 있다. 따라서, 제 1 단백질을 표적으로 하는 후보 약물의 선별 방법 또는 제 1 단백질을 표적으로하는후보약물의 효능확인 방법은, 상기 단계 (5) 이후에, 상기 단계 (5)에서의 비교 결과, 후보 화합물이 처리된 시험 시료의 단백질- 단백질 상호작용 수준 또는 활성화 수준이 후보 화합물이 처리되지 않은 시험 시료에서보다 낮은 경우, 상기 후보 화합물을 제 1 단백질 또는 제 2단백질을 표적으로 하는 후보 약물로 선택하거나, 상기 후보 화합물이 제 1 단백질 또는 제 2단백질을표적으로 하는 약물로서 효과가 있는 것으로 확인하는단계 (단계 (6))를추가로포함할수있다. As a result of the comparison in the above step (5), the protein-protein interaction level or the activation level of the test sample to which the candidate compound has been treated, , The candidate compound may be selected as a candidate drug targeting the first protein or the candidate compound may be found to be effective as a drug targeting the first protein. Therefore, the method for screening a candidate drug targeting the first protein or the method for confirming the efficacy of a candidate drug targeting the first protein is characterized in that after the step (5), the result of the comparison in the step (5) When the protein-protein interaction level or the activation level of the treated test sample is lower than that of the test sample in which the candidate compound is not treated, the candidate compound is selected as a candidate drug targeting the first protein or the second protein, (Step (6)) of confirming that the candidate compound is effective as a drug targeting the first protein or the second protein.
상기 후보 화합물은 제 1 단백질의 표적 약물로 사용 가능한 모든 생체적합성 물질 중에서 선택될 수 있으며, 예컨대, 소분자 화학 약물 (smal l molecular chemi cal s) , 단백질 (예컨대, 항체, 항체 단편, 또는 이의 유사체 등), 펩타이드, 핵산분자(예컨대, DNA, RNA(예컨대, s iRNA, mi croRNA, shRNA 등), PNA (pept ide nuclei c acid) , 앱타머, 등), 식물 주줄물, 동물 주줄물, 세포 주줄물 등으로 이루어진 군에서 1종 이상 선택될수있으나, 이에 제한되는것은아니다.  The candidate compound may be selected from among all biocompatible materials that can be used as a target drug of the first protein, for example, small molecule chemicals, proteins (e.g., antibodies, antibody fragments, ), Peptides, nucleic acid molecules (such as DNA, RNA (e.g., siRNA, miroRNA, shRNA, etc.), peptid nucleic acid (PNA), aptamer, And the like. However, the present invention is not limited thereto.
상기 시험 시료는 제 1 단백질이 과발현 및/또는 (과)활성화된 세포 (예컨대, 세포용해액 등) 또는조직 (예컨대, 조직 용해액 등), 또는 제 1 단백질과 관련된 질병 또는 선별하고자 하는 제 1 단백질을 표적으로 하는 약물의 적용(치료) 대상 질병과관련된 분리된 세포 (예컨대, 세포 용해액 등) 또는조직 (예컨대, 조직 용해액 등)일 수 있으며, 일 예에서, 확립된 세포주또는상기 질병 (예컨대, 암)을 앓는환자로부터 분리된 세포또는 조직일 수 있다. 예컨대, 상기 시험 시료는 확립된 암 세포주 또는 암 환자로부터 분리된 세포 또는 조직일 수 있다 (상기 암은 제 1 단백질의 과발현및/또는(과)활성화와관련된것일수있다).  The test sample can be used for a disease associated with a first protein, or a disease associated with a first protein to be screened or a first protein to be screened, such as a cell (for example, a cell lysate or the like) (E.g., cell lysate, etc.) or tissue (e. G., Tissue lysate, etc.) associated with the application (treatment) target disease of the protein-targeting drug, and in one example, (E.g., cancer). For example, the test sample may be a cell or tissue isolated from an established cancer cell line or cancer patient (the cancer may be associated with overexpression and / or activation of the first protein).
제 1 단백질이 PD-L1 및 PD-1 중에서 선택된 어느 하나이고, 저 12 단백질은다른하나인 경우, 상기 약물스크리닝에 사용된시험 시료는제 1 단백질이 과발현 및/또는 (과)활성화된 세포 (예컨대, 세포 용해액 등) 또는조직 (예컨대, 조직 용해액 등), 또는제 1단백질과관련된 질병 또는 선별하고자 하는 제 1 단백질을 표적으로 하는 약물의 적용(치료) 대상 질병과관련된 분리된 세포 (예컨대, 세포용해액 등) 또는조직 (예컨대, 조직 용해액 등), 또는 정제된 제 1단백질을포함하는 것일 수 있으며, 일 예에서, 확립된 세포주또는상기 질병 (예컨대, 암, 면역질환등)을 앓는 환자로부터 분리된 세포 또는 조직, 또는 정제된 제 1 단백질을 포함하는 것일수 있다. 상기 제 2단백질은정제된 (또는세포로부터 분리된) 형태일 수있다. When the first protein is any one selected from PD-L1 and PD-1 and the low-12 protein is the other, the test sample used for the drug screening is a cell in which the first protein is overexpressed and / (Treatment) of a drug targeting a disease associated with the first protein or a first protein to be screened (for example, a cell lysis solution or the like) or a tissue (for example, a tissue lysis solution) Cell lysate, etc.) or tissue (e.g., Tissue lysate, etc.), or a purified first protein, and in one example, a cell or tissue isolated from an established cell line or a patient suffering from the disease (e.g., cancer, immune disorders, etc.) Lt; RTI ID = 0.0 > 1 < / RTI > The second protein may be in a purified (or isolated form) cell.
상기 단계 (1)내지 (6)은 in /iro에서 수행되는것일수있다. 상기 단계 (1) 내지 (6) 및 제 1 단백질, 제 2 단백질 등와 용어 설명은앞서 기재한바와같다.  The above steps (1) to (6) may be performed in in / iro. The description of the above steps (1) to (6) and the first protein, the second protein and the like are as described above.
상기한 제 1 단백질을 표적으로 하는 후보 약물의 선별 방법은 제 1 단백질의 표적으로 하는 신약 개발에 있어서, 후보 약물로 개발된 약물의 효능검정 (또는확인또는시험)에 유용하게 적용될수있다.  The method for screening a candidate drug targeting the above-mentioned first protein can be usefully applied to the efficacy (or confirmation or testing) of a drug developed as a candidate drug in the development of a new drug targeted by the first protein.
후보 화합물이 처리된 시험 시료의 단백질-단백질 상호작용 수준 또는 활성화수준이 후보 화합물이 처리되지 않은 시험 시료에서보다높은 경우, 상기 후보 화합물을 상기 제 1 단백질을 표적으로 하는 후보 약물로 선택하는단계 (단계 (6))를추가로포함할수있다.  Selecting the candidate compound as a candidate drug targeting the first protein when the protein-protein interaction level or activation level of the test sample treated with the candidate compound is higher than that of the test sample not treated with the candidate compound Step (6)).
상기 후보 화합물은 제 1 단백질의 표적 약물로 사용 가능한 모든 생체적합성 물질 중에서 선택될 수 있으며, 예컨대, 소분자 화학 약물 (smal l molecular chemi cal s) , 단백질 (예컨대 , 항체, 항체 단편 , 또는 이의 유사체 등), 펩타이드, 핵산분자(예컨대, DNA, RNA(예컨대, siRNA, microRNA, shRNA 등), PNA (pept ide nuclei c acid) , 앱타머, 등), 식물 추출물, 동물 추출물, 세포 추출물 등으로 이루어진 군에서 1종 이상 선택될수있으나, 이에 제한되는것은아니다.  The candidate compound may be selected from among all biocompatible materials that can be used as a target drug of the first protein, for example, small molecule chemicals, proteins (e.g., antibodies, antibody fragments, ), Peptides, nucleic acid molecules (e.g., DNA, RNA (e.g., siRNA, microRNA, shRNA), peptid nucleic acid (PNA), aptamer, etc.), plant extracts, animal extracts, But it is not limited thereto.
상기 시험 시료는 제 1 단백질이 과발현 및/또는 (과)활성화된 세포 (예컨대, 세포용해액 등) 또는조직 (예컨대, 조직 용해액 등), 또는제 1 단백질과 관련된 질병 또는 선별하고자 하는 제 1 단백질을 표적으로 하는 약물의 적용(치료) 대상 질병과관련된 분리된 세포 (예컨대, 세포용해액 등) 또는조직 (예컨대, 조직 용해액 등)일 수 있으며, 일 예에서, 확립된 세포주또는상기 질병 (예컨대, 암)을 앓는환자로부터 분리된 세포또는 조직일 수 있다. 예컨대, 상기 시험 시료는 확립된 암 세포주 또는암 환자로부터 분리된 세포 또는 조직일 수 있다 (상기 암은 제 1 .단백질의 과발현및/또는(과)활성화와관련된것일수있다). The test sample can be used for a disease associated with a first protein, or a disease associated with a first protein to be screened or a first protein to be screened, such as a cell (for example, a cell lysate or the like) (E.g., cell lysate, etc.) or tissue (e. G., Tissue lysate, etc.) associated with the application (treatment) target disease of the protein-targeting drug, and in one example, (E.g., cancer). For example, the test sample may be a cell or tissue isolated from an established cancer cell line or a cancer patient . Or may be related to overexpression and / or activation of the protein).
상기 단계 (1) 내지 (6) 및 제 1 단백질, 제 2 단백질 등의 용어 설명은앞서 기재한바와같다. 2019/132517 1»(:1^1{2018/016675 The description of the terms (1) to (6) and the terms of the first protein, the second protein and the like are as described above. 2019/132517 1 »(: 1 ^ {2018/016675
상기한 제 1 단백질을 표적으로 하는 후보 약물의 선별 방법은 제 1 단백질의 표적으로 하는 신약 개발에 있어서, 후보 약물로 개발된 약물와 효능검정 (또는확인또는시험)에 유용하게 적용될수있다. The screening method of the candidate drug targeting the first protein can be usefully applied to drugs and efficacy testing (or confirmation or testing) developed as a candidate drug in the development of a new drug targeted by the first protein.
다른 예는 제 1 단백질과 제 2 단백질 간 단백질-단백질 상호작용을 측정하는 단계를포함하고, 상기 제 1 단백질은세포또는조직 내 및/또는 세포 또는 조직 간의 신호전달경로에 관여하는 단백질이고, 제 2 단백질은 상기 세포또는조직 내 및/또는세포또는조직 간의 신호전달경로중에서 상기 제 1 단백질의 하류 단백질 중에서 선택된 1종 이상인, 제 1 단백질의 표적 치료와 병행하기 위한 병행 치료 표적을 선정하는 방법 또는 상기 선정에 정보를 제공하는 방법, 또는 제 1 단백질의 표적 약물과 병용 투여하기 위한 병용 약물을 선정하는 방법 또는 상기 선정에 정보를 제공하는방법을제공한다.  Another example includes measuring a protein-protein interaction between a first protein and a second protein, wherein the first protein is a protein involved in a signal transduction pathway in a cell or tissue and / or between cells or tissue, 2 protein is at least one selected from among proteins downstream of the first protein in the cell or tissue and / or signal transduction pathway between cells or tissues, a method for selecting a concurrent therapeutic target for concurrent with the target treatment of the first protein, or A method of providing information to the selection, or a method of selecting a concomitant drug to be co-administered with a target drug of the first protein, or a method of providing information to the selection.
상기 방법은,  The method comprises:
(1) 제 1단백질을포함하는시험 시료를표면에 상기 제 1단백질에 특이적으로 결합하는 물질을포함하는 기판에 가하여 제 1 단백질이 고정된 기판을준비하는단계;  (1) preparing a substrate to which a first protein is immobilized by adding a test sample containing a first protein to a substrate including a substance that specifically binds to the first protein on a surface thereof;
(2) 상기 준비된 제 1 단백질이 고정된 기판에 표지된 제 2 단백질을 첨가하여 반응시키는단계;  (2) adding a labeled second protein to the substrate on which the prepared first protein is immobilized and reacting;
(3) 단계 (2)에서 얻어진 반응물로부터 신호를 즉정하는 단계 (단백질-단백질상호작용측정) ; 및  (3) determining the signal from the reactant obtained in step (2) (protein-protein interaction measurement); And
(4)단계 (3)에서 측정된신호를이용하여 단계 (1)에서 첨가한시험 시료 포함된 제 1 단백질의 단위량에 대한 값을 구하는 단계 (활성화 수준 측정) ; 또는  (4) obtaining a value for the unit amount of the first protein included in the test sample added in step (1) (measuring the activation level) using the signal measured in step (3); or
(4-1) 단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한 시험 시료 의 단위량에 대한 신호값을 구하는 단계 (단백질-단백질 상호작용 수준 측정) , 및 (4-2) 단계 (4-1)에서 얻어진 시험 시료의 단위량에 대한 신호값을 이용하여 시험 시료에 포함된제 1 단백질의 단위량에 대한값을구하는단계 (활성화수준측정) ; 및  (4-1) obtaining a signal value (measurement of protein-protein interaction level) for the unit amount of the test sample added in step (1) using the signal measured in step (3), and (4-2 Obtaining a value for a unit amount of the first protein included in the test sample (measuring the activation level) using the signal value of the unit amount of the test sample obtained in the step (4-1); And
(5) 상기 단계 (3) , 단계 (4) , 단계 (4-1) , 또는 단계 (4-2)에서 얻어진결과를비교하는단계  (5) comparing the results obtained in step (3), step (4), step (4-1), or step (4-2)
를포함할수있다.  .
상기 단계 (5)의 비교하는단계는,  Wherein the step of comparing (5)
(a) 시험 시료에 대하여 얻어진 상기 단계 (4) 또는 단계 (4-2)의 2019/132517 1»(:1^1{2018/016675 (a) the step (4) or (4-2) obtained for the test sample 2019/132517 1 »(: 1 ^ {2018/016675
결과를 기준 시료에 대하여 얻어진 결과와 비교하거나 (이 경우, 상기 방법은 기준 시료는 앞서 설명한 바와 같고, 기준 시료에 대하여 앞서 설명한단계 (I ), (2' ) , (3’), 및 (4,) , 또는 (I ), (2' ) , (3 ) , (4-1’), 및 (4-2’)를추가로포함할수있다), (1), (2 '), (3'), and (4) described above with reference to the reference sample, and the results are compared with the results obtained for the reference sample , Or may further comprise (I), (2 '), (3), (4-1'), and (4-2 '))
( 2종 이상의 제 2 단백질에 대하여 얻어진 상기 단계 (4) 또는 단계 (4-2)의 결과를서로비교하여 수행될수있다.  (Comparing the results of step (4) or step (4-2) obtained for two or more second proteins with each other).
상기 단계 (5)의 비교결과,  As a result of the comparison in the step (5)
(3) 시험 시료에 대하여 얻어진 상기 단계 (4) 또는 단계 (4-2)의 단백질-단백질 상호작용 수준 또는 활성화 수준이 기준 시료보다 높은 경우, 상기 제 2단백질을제 1단백질의 표적 치료와 병행하기 위한 병행 치료표적으로선정하거나, 상기 제 2단백질을표적으로하는약물을제 1 단백질의 표적 약물과병용투여하기 위한약물로선정하거나; ( 3) When the protein-protein interaction level or the activation level of the step (4) or step (4-2) obtained for the test sample is higher than the reference sample, the second protein is combined with the target treatment of the first protein Or a drug targeting the second protein is selected as a drug for use in combination with a target drug of the first protein;
( 2종 이상의 제 2단백질 중상기 단계 (4) 또는 단계 (4-2)에서 얻어진 단백질-단백질 상호작용 수준 또는 활성화 수준이 가장 높은 제 2 단백질을 제 1 단백질의 표적 치료와 병행하기 병행 치료 표적으로 선정하거나, 상기 제 2 단백질을 표적으로 하는 약물을 제 1 단백질의 표적 약물과병용투여하기 위한약물로선정할수있다.  (The second protein having the highest protein-protein interaction level or activation level obtained in the second or more second protein heavy chain stage (4) or step (4-2) is used in combination with the target treatment of the first protein, Or a drug targeting the second protein may be selected as a drug for administration in combination with a target drug of the first protein.
따라서, 상기 제 1 단백질의 표적 치료와 병행하기 위한 병행 치료 표적을선정하는방법 (또는상기 선정에 정보를제공하는방법), 또는제 1 단백질의 표적 약물과 병용 투여하기 위한 병용 약물을 선정하는 방법 (또는상기 선정에 정보를제공하는방법)은, 상기 단계 (5) 이후에,  Therefore, a method for selecting a concurrent treatment target (or a method for providing information on the selection) for concurrently with the target treatment of the first protein, or a method for selecting a concomitant drug for co-administration with the target protein of the first protein (Or a method for providing information to the selection), after the step (5)
(6-1) 시험 시료에 대하여 얻어진 상기 단계 (4) 또는단계 (4-2)의 단백질-단백질 상호작용 수준 또는 활성화 수준이 기준 시료보다 높은 경우, 상기 제 2단백질을제 1단백질의 표적 치료와병행하기 위한 병행 치료표적으로선정하거나, 상기 제 2단백질을표적으로하는 약물을저 11 단백질의 표적 약물과병용투여하기 위한약물로선정하는단계; 또는  (6-1) When the protein-protein interaction level or the activation level of the step (4) or the step (4-2) obtained on the test sample is higher than the reference sample, And selecting a drug targeting the second protein as a drug to be used in combination with a target drug of low 11 protein; or
(6-2) 2종이상의 제 2단백질중상기 단계 (4)또는단계 (4-2)에서 얻어진 단백질-단백질 상호작용 수준 또는 활성화 수준이 가장 높은 제 2 단백질을 제 1 단백질의 표적 치료와 병행하기 위한 병행 치료 표적으로 선정하거나, 상기 제 2 단백질을 표적으로 하는 약물을 제 1 단백질의 표적 약물과병용투여하기 위한약물로선정하는단계  (6-2) The second protein having the highest protein-protein interaction level or activation level obtained in step (4) or step (4-2) among the second protein on the two or more species is administered in parallel with the target treatment of the first protein Or a drug for targeting the second protein to a drug for use in combination with a target drug of the first protein,
를추가로포함할수있다.  May be further included.
다른 측면에서, 상기의 결정 내용을 기초로 한 병용 치료 수단이 제공된다. 일 예는상기 제 1 단백질을 표적으로 하는 약물 및 상기 단계 (6-1) 또는 (6-2)에서 병행 치료 표적으로 선정된 제 2 단백질을 표적으로 하는 (예컨대, 저해하는) 약물을 포함하는, 제 1 단백질과 관련된 질병의 치료를 위한 병용 투여용 약학 조성물을 제공한다. 상기 약학 조성물은 상기 시험 시료또는상기 시험 시료가유래하는 개체에서의 제 1단백질과 관련된 질병의 치료를위한 병용투여 용도로사용될 수 있다. 다른 예는 상기 제 1단백질을표적으로하는약물및 상기 단계 (6-1)또는(6-2)에서 병행 치료표적으로선정된 제 2단백질을표적으로하는(예컨대, 저해하는) 약물의, 제 1 단백질과 관련된 질병의 치료를 위한 병용 치료에 사용하기 위한 용도를 제공한다. 다른 예는 제 1 단백질과 관련된 질병의 치료를 필요로 하는 환자에게 제 1 단백질을 표적으로 하는 치료 (예컨대, 제 1 단백질을표적으로하는 약물의 투여) 및 상기 단계 (6-1) 또는 (6-2)에서 병행 치료 표적으로 선정된 제 2 단백질을 표적으로 하는 치료 (예컨대, 상기 제 2 단백질을 표적으로 하는 (예컨대, 저해하는) 약물의 투여)를 동시에 또는 순서에 상관 없이 연속하여 수행하는 단계를 포함하는, 제 1 단백질과 관련된 질병의 치료 방법을 제공한다. 상기 환자는 상기 시험 시료또는상기 시험 시료가유래하는개체일수있다. In another aspect, a combination therapy means based on the above decision content / RTI > One example includes a drug targeting the first protein and a drug targeting (e.g., inhibiting) a second protein selected as a concurrent therapeutic target in step (6-1) or (6-2) , ≪ / RTI > a pharmaceutical composition for coadministration for the treatment of diseases associated with the first protein. The pharmaceutical composition may be used for the combined administration for the treatment of diseases related to the first protein in the test sample or the individual from which the test sample is derived. Another example is a pharmaceutical composition comprising a drug targeting the first protein and a drug targeting (e.g., inhibiting) a second protein selected as a concurrent therapeutic target in step (6-1) or (6-2) 1 < / RTI > protein. Another example is a method for treating a disease associated with a first protein, comprising administering to a patient in need of such treatment a therapeutically effective amount of a first protein (e. G., Administering a drug targeting the first protein) (E.g., administration of a drug that targets (e. G., Inhibits) the second protein) targeted to a second protein selected as a concurrent therapeutic target in step A method of treating a disease associated with a first protein. The patient may be the test sample or an individual from which the test sample is derived.
다른 예에 있어서, 상기 제공된 제 1단백질을표적으로하는 약물에 대한 반응성을 예측하는 방법 또는 예측에 정보를 제공하는 방법, 제 1 단백질을표적으로 하는 치료에 적합한 개체를 선별하는 방법 또는선별에 정보를 제공하는 방법, 및 세포 또는 조직 또는 상기 세포 또는 조직이 유래하는 개체 (개별 환자)에 적용하기에 적합한 치료 표적으로서의 제 1 단백질또는이를표적으로하는약물을선별하는방법 또는선별에 정보를 제공하는 방법은, 앞서 설명된 단백질-단백질 상호작용 측정 및 활성화 수준측정 단계에 더하여, 제 1단백질의 번역 후변형 (Post-trans lat ional modi f icat ion; PTM) 여부를 확인하는단계 (단계 (7))를추가로포함할수 있다. PTM은 제 1단백질의 활성화상태를 보여주는 일반적인 마커입니다. 그러므로현존방법과의 대조군으로서사용된다.  In another example, there is provided a method of predicting or predicting responsiveness to a drug that targets the provided first protein, a method of screening an entity suitable for treatment targeting the first protein, , And methods of screening or screening for a first protein or a target thereof as a therapeutic target suitable for application to a cell or tissue or an individual (individual patient) from which the cell or tissue is derived (Step 7) of confirming whether the first protein is post-translationally modified (PTM) in addition to the protein-protein interaction measurement and activation level measurement step described above, ). PTM is a common marker showing the activation status of the first protein. It is therefore used as a control against existing methods.
상기 제 1단백질의 번역 후변형은, 정상개체 (암또는종양을갖지 않는 개체)와 비교하여, 암 또는 종양 환자에서 발생 빈도가 높은 변형 (제 1단백질의 종양관련 번역 후 변형)을의미하며, 제 1단백질 또는 제 1 단백질을 암호화하는 유전자에서 하나 이상의 뉴클레오타이드 또는 하나 이상의 아미노산 변이 (또는 하나 이상의 아미노산 변이를 유발하는 뉴클레오타이드의 변이), 인산화 (예컨대, Tyr 잔기의 인산화) 등으로 이루어진군에서 선택된하나이상포함하는의미일 수 있다. 예컨대, 제 1 단백질이 EGFR인 경우, 상기 변이는 엑손 19및/또는엑손 21에 집중적으로 존재할수 있으며, 일 구체예에서, 상기 변이는 EGFR의 엑손 19및 엑손 21 내의 하나 이상의 유전자의 결실 (예컨대, 엑손 19 및/또는 엑손 21의 결실), L858R 치환을 유발하는 유전자 변이, Tyr 1068 및/또는 Tyr 1086에서의 인산화 중에서 선택된 하나 이상일 수 있으나, 이에 제한되는 것은아니며, 정상개체 (암또는종양을 갖지 않는 개체)와비교하여, 암 또는 종양 환자에서 발생 빈도가 높고 알려진 모든 EGFR (유전자/단백질) 변형 중에서 선택될수 있다. 제 1단백질이 HER2인 경우, 상기 변이는 Tyr 1221에서의 인산화일 수 있으나, 이에 제한되는 것은 아니며, 정상 개체 (암또는종양을갖지 않는개체)와비교하여 , 암또는종양환자에서 발생 빈도가 높고 알려진 모든 HER2 (유전자/단백질) 변형 중에서 선택될 수 있다. 제 1 단백질이 HER3인 경우, 상기 변이는 Tyr 1289에서의 인산화일 수 있으나, 이에 제한되는 것은 아니며, 정상 개체 (암또는 종양을 갖지 않는 개체)와 비교하여, 암또는 종양 환자에서 발생 빈도가 높고 알려진 모든 HER3 (유전자/단백질) 변형 중에서 선택될 수 있다. 제 1 단백질이 MET인 경우, 상기 변이는 Tyr 1349에서의 인산화일 수 있으나, 이에 제한되는 것은 아니며, 정상 개체 (암 또는 종양을 갖지 않는 개체)와 비교하여, 암 또는 종양 환자에서 발생 빈도가 높고 알려진 모든 MET (유전자/단백질)변형 중에서 선택될수있다. The post-translational modification of the first protein refers to a modification (a post-translational modification related to the first protein of the first protein) that occurs more frequently in a cancer or tumor patient than a normal individual (a cancer or a tumor-free individual) A gene encoding a first protein or a first protein may comprise at least one nucleotide or at least one amino acid mutation (or at least one amino acid mutation A nucleotide variation), phosphorylation (e.g., phosphorylation of a Tyr residue), and the like. For example, if the first protein is EGFR, the mutation may be intensively present in exon 19 and / or exon 21, and in one embodiment, the mutation is a deletion of one or more genes in exon 19 and exon 21 of EGFR , Deletion of exon 19 and / or exon 21), gene mutation causing L858R substitution, phosphorylation at Tyr 1068 and / or Tyr 1086, but not limited to, normal individuals (Not present) and can be selected from all known EGFR (gene / protein) variants with high incidence in cancer or tumor patients. When the first protein is HER2, the mutation may be phosphorylation at Tyr 1221, but is not limited thereto, and may occur in a cancerous or tumor patient with a high incidence in comparison with a normal individual (a cancer or a tumor-free individual) It can be selected from all known HER2 (gene / protein) variants. When the first protein is HER3, the mutation may be phosphorylation at Tyr 1289, but is not limited thereto, and is more frequent in cancer or tumor patients than in normal individuals (individuals without cancer or tumor) It can be selected from all known HER3 (gene / protein) variants. When the first protein is MET, the mutation may be phosphorylation at Tyr 1349, but is not limited thereto, and is more frequent in cancer or tumor patients than in a normal individual (a cancer or a tumor-free individual) It can be selected from all known MET (gene / protein) variants.
본 명세서에 사용된 바로서, 용어 "제 1 단백질을 표적으로 한다” 함은 제 1 단백질의 활성을 촉진 또는 저해하는 것을 의미할 수 있으며, 예컨대, 제 1 단백질의 활성을 저해하는 것을 의미할 수 있다. 제 1 단백질의 활성의 저해는 제 1 단백질과 결합하거나, 및/또는 제 1 단백질을 분해 및/또는 구조적 변형시켜 고유의 기능, 예컨대, 세포 및/또는 조직에서의 고유의 생체 신호전달 기능을 감소시키거나 제거하는 것일 수 있다.  As used herein, the term "targeting a first protein " may mean promoting or inhibiting the activity of a first protein, e. G., Inhibiting the activity of a first protein The inhibition of the activity of the first protein may be achieved by binding to the first protein and / or degrading and / or structurally modifying the first protein to produce a unique function, e. G., A unique bio signaling function in cells and / Lt; RTI ID = 0.0 > and / or < / RTI >
본 명세서에 사용된 바로서, 용어 "약물'’은 약리적 효과를나타내는 모든 물질, 예컨대, 소분자 화합물, 단백질 (예컨대, 항체, 항체 단편, 또는 이의 유사체 등), 펩타이드, 핵산 분자 (예컨대, DNA, RNA (예컨대, siRNA, mi croRNA, shRNA등), PNA (pept ide nucleic acid) , 앱타머, 등), 식물추출물, 동물추출물, 세포추출물등으로 이루어진 군에서 1종 이상 선택되는것일수있다. As used herein, the term "drug" refers to any substance that exhibits a pharmacological effect, such as a small molecule compound, a protein (such as an antibody, an antibody fragment, or an analogue thereof), a peptide, a nucleic acid molecule And at least one compound selected from the group consisting of RNA (for example, siRNA, miroRNA, shRNA, etc.), peptidase nucleic acid (PNA), aptamer, etc., plant extracts, animal extracts, It can be selected.
다른예는,  In another example,
(a) 제 1 단백질의 발현 수준 및/또는 번역 후 변형 (Post- translational modification; PTM)즉정부,및  (a) an expression level of the first protein and / or post-translational modification (PTM)
(b)제 1단백질과제 2단백질간의 단백질-단백질상호작용측정부 를포함하고,  (b) a first protein task 2 protein-protein interaction measurement unit,
상기 제 1단백질은수용체 티로신키나아제 (RTK)인, RTK프로파일링 키트 또는 제 1 단백질을 표적으로 하는 약물의 효능 예측용 키트를 제공한다. 일 예에서, 상기 약물은 폐암치료제 (예컨대, 아파티닙 (Afatinib)등)일수있으나,이에 제한되는것은아니다.  Wherein the first protein is a receptor tyrosine kinase (RTK), an RTK profiling kit, or a kit for predicting the efficacy of a drug targeting the first protein. In one example, the drug may be, but is not limited to, a therapeutic agent for lung cancer (such as Afatinib).
본 명세서에 사용된 바로서, 용어 "제 1 단백질을 표적으로 하는 약물"은 제 1단백질의 활성을 저해하는 모든 물질, 예컨대, 제 1단백질의 활성을 저해하는 소분자 화합물 (small molecule) , 단백질 (예컨대, 항체, 항체 단편, 또는이의 유사체 등),펩타이드,핵산분자 (예컨대, DNA, RNA (예컨대 , siRNA (small interfering RNA) , shRNA (small hairpin RNA) , miRNA (microRNA) , 등), PNA (peptide nucleic acid) , 앱타머, 등), 식물 주줄물, 동물 주줄물, 세포 주줄물 등으로 이루어진 군에서 선택되는 1종 이상일 수 있다. 보다구체적으로, "제 1단백질을표적으로 하는 약물”은 제 1 단백질과 결합하거나, 및/또는 제 1 단백질을 분해 및/또는 구조적 변형시켜 고유의 기능, 예컨대, 세포 및/또는 조직에서의 고유의 생체 신호전달 기능을 감소시키거나 제거하는 모든 물질, 예컨대, 제 1단백질의 활성을 저해하는 소분자 화합물, 단백질 (예컨대, 항체, 항체 단편, 또는 이의 유사체 등), 펩타이드, 핵산분자 (예컨대, DNA, RNA (예컨대, siRNA, microRNA, shRNA 등), PNA (peptide nucleic acid) , 앱타머, 등), 식물 추출물, 동물 추출물, 세포 추출물 등으로 이루어진 군에서 선택되는 1종 이상일수있다.  As used herein, the term "drug targeting a first protein" refers to any substance that inhibits the activity of a first protein, such as a small molecule, a protein, (E.g., small interfering RNA (siRNA), small hairpin RNA, miRNA (microRNA), etc.), PNAs (such as antibodies, antibody fragments or analogs thereof), peptides, nucleic acid molecules a peptide nucleic acid, an aptamer, etc.), a plant stem, an animal stem, a cell stem, and the like. More specifically, "a drug targeting a first protein " is a substance that binds to a first protein and / or degrades and / or structurally modifies a first protein to produce a unique function, (E. G., Antibodies, antibody fragments, or analogs thereof), peptides, nucleic acid molecules (e. G., DNA < / RTI > , RNA (such as siRNA, microRNA, shRNA), PNA (peptide nucleic acid), aptamer, etc.), plant extracts, animal extracts, cell extracts and the like.
일 구체예에서, 상기 "제 1 단백질을 표적으로 하는 약물”은 제 1 단백질을 표적으로 하는 치료제, 예컨대, 제 1 단백질의 표적 저해제를 의미하는 것일 수 있다. 일 예에서, 상기 제 1단백질을 표적으로 하는 약물은, EGFR 표적 치료제 (cetuximab, gefitinib, erlotinib, afatinib, osimertinib (AZD9291), 각종 항- EGFR 항체 등), MET 표적 치료제 (각종 항- MET 항체, Crizotinib, Cabozant inib 등), HER2 표적 치료제 (trastuzumab, Trastuzumab, Pertuzumab, Lapatinib등), HER3표적 치료제 (각종항- HER3항체 등), FGFR1, 2)표적 치료제 (Lenvatinib, Nintedanib, Regorafenib 등), VEGFR(1, 2, 3)표적 치료제 (Bevacizumab, Axitinib, Lenvat inib등), PDGFR표적 처료제 (Axitinib, Gef itinib, Imatinib등), IGF1R 표적 치료제 (Ceritinib 등), c-KIT 표적 치료제 (Axitinib, Cabozant inib, Dasatinib 등), RET 표적 치료제 (Vandetanib 등), BRAF 표적 치료제 (Vemurafenib, Dabrafenib등), MEK표적 치료제 (Trametinib 등), Src표적 치료제 (Bosutinib, Dasatinib, Ponat inib, Vandetanib등), PI3K표적 치료제 (Crizotinib, Cabozant inib등), CDK(4, 6)표적 치료제 (Palbocicl ib, Sorafenib 등), R0S1표적 치료제 (Ceritinib, Crizotinib 등), ALK표적 치료제 (Ceritinib, Crizotinib등), BCR-Abll표적 치료제In one embodiment, the "drug targeting a first protein " may refer to a therapeutic agent targeting a first protein, such as a target inhibitor of a first protein. In one example, Targeted drugs include antiretroviral drugs such as cetuximab, gefitinib, erlotinib, afatinib, osimertinib (AZD9291), various anti-EGFR antibodies, MET target therapeutics (various anti-MET antibodies, Crizotinib, Cabozant inib etc.) Therapeutic agents (trastuzumab, Trastuzumab, Pertuzumab, Lapatinib, etc.), HER3 target therapy (Eg, various anti-HER3 antibodies), FGFR1, 2) Target therapeutic agents (Lenvatinib, Nintedanib, Regorafenib etc.), VEGFR (1,2,3) Target therapeutic agents (Bevacizumab, Axitinib, Lenvat inib etc.), PDGFR target treatment agent , Vetetinib, etc.), BRAF target treatment (Vemurafenib, Dabrafenib, etc.), MEK (Gefitinib, Imatinib etc.), IGF1R target therapy (Ceritinib etc.), c-KIT target treatment (Axitinib, Cabozant inib, Dasatinib etc.) (Czototinib, Cabozant inib, etc.), CDK (4, 6) Target therapy (Palbociclib, Sorafenib, etc.), R0S1 Target Therapeutics (Ceritinib, Crizotinib, etc.), ALK Target Therapeutics (Ceritinib, Crizotinib, etc.), BCR-Abll Target Therapeutic Agents
(Bosutinib, Dasatinib, Imatinib, Ni lotinib 등), AR 표적 치료제 (Abiraterone, Enzalut amide 등), CTLA4 표적 치료제 (Ipilimumab, Tremel imumab 등), PD-1 표적 치료제 (Nivolumab 등) 등으로 이루어진 군에서 선택된 1종이상일수있으나이에 제한되는것은아니다. (Ivilimumab, Tremel imumab, etc.), PD-1 target therapeutic agent (Nivolumab etc.), and the like, which are selected from the group consisting of anti-inflammatory drugs (Bosutinib, Dasatinib, Imatinib, Ni lotinib etc.), AR target therapeutic agents (Abiraterone, Enzalut amide etc.) Or more, but is not limited thereto.
다른 구체예에서, 상기 "제 1단백질을 표적으로 하는 약물" (약물 반응성 측정 대상 또는 스크리닝 대상 약물)은 제 1 단백질과의 결합에 있어서 제 2단백질과 경쟁적으로 작용하여, 제 1단백질과제 2단백질 간의 상호작용을 억제하는 약물 (예컨대, 소분자 화합물 (small molecule) , 단백질 (예컨대, 항체, 항체 단편, 또는 이의 유사체 등), 펩타이드, 핵산 분자 (예컨대, DNA, RNA (예컨대 , siRNA (small interfering RNA) , shRNA (small hairpin RNA) , miRNA (microRNA) ,등), PNA (peptide nucleic acid) , 앱타머, 등), 식물 주줄물, 동물 주줄물, 세포 주줄물 등으로 이루어진 군에서 선택되는 1종이상)일수있다.  In another embodiment, the "drug targeting the first protein" (drug response measure or screening drug) competes with the second protein in binding to the first protein such that the first protein task 2 protein (E.g., a small molecule, a protein (e.g., an antibody, an antibody fragment, or an analogue thereof), a peptide, a nucleic acid molecule ), shRNA (small hairpin RNA), miRNA (microRNA), etc.), PNA (peptide nucleic acid), aptamer, etc.), plant trunk, animal trunk, Or more).
본 명세서에 사용된 바로서, 용어 ”제 1 단백질을 표적으로 하는 치료"는제 1단백질의 활성을저해하는모든의학적 및/또는 약학적 행위를 의미할 수 있으며, 예컨대, 앞서 설명한 바와 같은 제 1단백질의 활성을 저해하는약물을제 1단백질의 활성을저해하는것을필요로하는대상에게 처방 및/또는 투여하는 것일 수 있다. 보다 구체적으로, "제 1단백질을 표적으로 하는 치료”는 제 1단백질과 결합하거나, 및/또는 제 1단백질을 분해 및/또는 구조적 변형시켜 고유의 기능, 예컨대, 세포 및/또는 조직에서의 고유의 생체 신호전달 기능을 감소시키거나 제거하는 약물을 이를필요로하는대상에게 처방및/또는투여하는것일수있다.  As used herein, the term " treatment targeting a first protein "may refer to any medical and / or pharmaceutical activity that inhibits the activity of a first protein, for example, Quot; first protein-targeting treatment " may include administering a first protein and / or a second protein to a subject in need of inhibiting the activity of the first protein, To a subject in need thereof a drug that reduces or eliminates the inherent functions of, for example, inherent bio-signal transduction functions in cells and / or tissues, by binding and / or degrading and / or structurally modifying the first protein Prescription and / or administration.
다른 예는 제 1단백질과 제 2단백질 간 단백질-단백질 상호작용을 2019/132517 1»(:1^1{2018/016675 Another example is protein-protein interaction between a first protein and a second protein 2019/132517 1 »(: 1 ^ {2018/016675
측정하는 단계를 포함하고, 상기 제 1 단백질은 1世요2 및 抑묘3이고, 상기 1世요2및 묘3는헤테로다이머를형성하며, 제 2단백질은세포또는조직 내 및/또는세포또는조직 간의 신호전달경로중에서 他1?2, 抑1¾, 또는 이들 모두의 하위 단백질이고, 상기 단백질-단백질 상호작용을 측정하는 단계를 후보 물질이 처리된 시료와 처리되지 않은 시료에서 수행하는, 제 1 단백질을 표적으로 하는 후보 약물의 효능 확인 방법 또는 상기 질병의 예방또는치료를위한후보 약물의 효능확인 방법을제공한다. 다른예는 四묘2 - 묘3 헤테로다이머에서의 抑묘2 및/또는 1?3의 활성화 정도 (예컨대, 타이로신 잔기의 인산화)를 측정하는 단계를 포함하고, 상기 활성화를 측정하는 단계를 후보 물질이 처리된 他요2 - 요3 헤테로다이머 또는 1世묘2_ 四 1?3 헤테로다이머를 포함하는 시료와 처리되지 않은 他요2 - 요3 헤테로다이머 또는 ?世요2 - 1?3헤테로다이머를포함하는시료에서 수행하는, 제 1단백질을표적으로하는후보약물의 효능확인 방법 또는상기 질병의 예방또는치료를위한후보약물의 효능확인방법을제공한다. Wherein the first protein is a first class 2 and the third class 3, the first class 2 and the third class 3 form a heterodimer and the second protein is a cell or a tissue and / or a cell or a cell, Wherein the protein-protein interaction is a sub-protein of the other of the inter-tissue signal transduction pathways, and wherein the step of measuring the protein-protein interaction is performed in the sample treated with the candidate substance and the untreated sample, A method for confirming the efficacy of a candidate drug targeting a protein or a method for confirming the efficacy of a candidate drug for the prevention or treatment of the disease. Another example involves measuring the activation level of the inhibitor 2 and / or 1-3 of the quadrivalent 2-thymine 3 heterodimer (e.g., phosphorylation of the tyrosine residue), wherein the step of measuring activation is referred to as the candidate substance A sample containing a heterodimer or a heterodimer of the 2-tetradecane heterodimer and an untreated heterodimer or an untreated heterodimer of 2- A method for confirming the efficacy of a candidate drug targeting the first protein or a method for confirming the efficacy of a candidate drug for prevention or treatment of the disease.
상기 방법은,  The method comprises:
시험 시료에 후보물질을처리하는(예컨대, 접촉시키는)단계, 및 하기의 단계 (3), 0)), ((:), 및 ( 를 수행하는 단계를 포함할 수 있다: (For example, contacting) the candidate material with the test sample, and performing the following steps ( 3 ), (0), ((), and
3) }世1씽 - 묘3 헤테로다이머를 포함하는 시험 시료를 준비하거나, 四요2 -四요3 헤테로다이머를 포함하는 시험 시료를 표면에
Figure imgf000058_0001
및/또는 他묘3에 특이적으로 결합하는 물질을 포함하는 기판에 가하여 四요2 -四1?3 헤테로다이머가고정된기판을준비하는단계 ;
( 3 ) Prepare a test sample containing a 1-thymine 3 heterodimer or a test sample containing a tetra-quadrone-3-hetero dimer on the surface
Figure imgf000058_0001
And / or to a substrate comprising a substance that specifically binds to the other epitope 3, thereby preparing a substrate on which quaternary 2,4-heterodimer is immobilized;
( 상기 1?2 -四1씼헤테로다이머를 인산화시키고, 표지된 인산화된 타이로신특이적 결합물질을첨가하여 반응시키는단계 ;  Phosphorylating the 1,2-to 4-membered heterodimer and adding and reacting the labeled phosphorylated tyrosine specific binding substance;
0) 단계 ( 에서 얻어진 반응물로부터 신호를 측정하는 단계( 0 ) step of measuring the signal from the reactant obtained in step
(단백질-단백질 상호작용 측정 또는
Figure imgf000058_0002
및/또는 抑1?3의 타이로신 카이네이즈활성화정도측정); 및
(Protein-protein interaction measurement or
Figure imgf000058_0002
And / or the degree of activation of the tyrosine kinase activity of inhibitor 3); And
((1) 상기 후보 물질이 처리되지 않은 시험 시료 및 처리된 시험 시료에 대하여 상기 단계 ( 에서 얻어진 각각의 결과를 서로 비교하는 단계 .  (1) comparing the respective results obtained in the above step with respect to the test sample not treated with the candidate substance and the treated test sample with each other.
앞서 설명한바와같이, 상기 인산화된타이로신특이적 결합물질은 1世요2 및/또는 요3의 인산화된 타이로신에 특이적으로 결합하는 항체일 수있다. 2019/132517 1»(:1^1{2018/016675 As described above, the phosphorylated tyrosine specific binding substance may be an antibody specifically binding to the phosphorylated tyrosine of the first class II and / or the third class. 2019/132517 1 »(: 1 ^ {2018/016675
상기 후보 물질은 상기 단계 ( 에서 처리될 수 있으며, 예컨대, 단계 ( 의 인산화 또는 인산화된 타이로신 특이적 결합물질의 처리 전, 후, 또는 동시에 처리될 수 있다. 상기 단계 ( 의 인산화는 인산화제를 처리하여 수행될 수 있다. 상기 인산화제는
Figure imgf000059_0001
및/또는 요3를 인산화시킬 수 있는 모든 물질들 중에서 선택된 1종 이상일 수 있으며, 예컨대, 쇼†!3와 마그네슘 (예컨대, ¾1故12 등)일 수 있으나, 이에 제한되는 것은아니다.
The candidate material may be treated in the step (e. G., Before, after, or simultaneously with the treatment of the phosphorylated or phosphorylated tyrosine specific binding material of step (a). Phosphorylation of this step can be carried out by treating the phosphorylating agent. The phosphorus-
Figure imgf000059_0001
And / or all substances capable of phosphorylating urine 3, for example, may be selected from the group consisting of: 3, and magnesium (e.g., 1, 2 , 1, 2, etc.).
단계 ( 에서의 후보 물질이 처리되지 않은 시험 시료 및 처리된 시험 시료의 결과를 비교하기 위하여, 상기 단계 (3)-(0는 후보 물질이 처리되지 않은 시험 시료 및 처리된 시험 시료에 대하여 각각 수행될 수 있다. In order to compare the results of the untreated test sample with the treated test sample in step ( 3 ) - (0), the candidate substance is subjected to the test sample not treated with the candidate substance and the treated test sample, respectively .
상기 후보 물질이 처리되지 않은 시험 시료와 처리된 시험 시료는 각각후보물질의 처리 전 시험 시료 및 처리 후시험 시료를의미하거나, 시험 시료를분량하여 상기 후보물질을처리한 일부의 시험 시료 및 상기 후보물질을처리하지 않은다른일부의 시험 시료를의미할수있다.  The test sample not treated with the candidate substance and the treated test sample mean a test sample before treatment and a test sample after treatment of the candidate substance, respectively, or a part of the test sample treated with the candidate substance and the candidate May refer to some other test sample that has not been treated with the substance.
상기 단계 ( 에서의 비교 결과, 후보 물질이 처리된 시험 시료의 단백질-단백질 상호작용 수준 또는 四묘2 및/또는 四묘3의 타이로신 카이네이즈 활성화 정도가 후보 물질이 처리되지 않은 시험 시료에서보다 낮은경우(즉상기 후보물질이 四1?2및/또는四묘3의 타이로신 카이네이즈 활성화를 억제하는 경우), 상기 후보 물질이 }世1?2, }世요3, 또는 이들 모두(예컨대,
Figure imgf000059_0002
헤테로다이머 형태)를 표적으로 하는 (즉, 抑모2 및/또는 1?3의 타이로신 카이네이즈 활성화를 억제하는) 약물로서 효과가 있는것으로확인할수있다.
If the protein-protein interaction level of the test sample treated with the candidate substance or the tyrosine kinase activation degree of quadrature 2 and / or quadruple 3 is lower than that of the test sample in which the candidate substance is not treated, (I. E., When the candidate substance inhibits tyrosine kinase activation of < RTI ID = 0.0 > 41 < / RTI &
Figure imgf000059_0002
(I. E., Inhibiting the activation of tyrosine kinase 2, and / or < RTI ID = 0.0 > 1-3). ≪ / RTI >
따라서 , 상기 후보약물의 효능확인방법은, 상기 단계 ( 이후에, 상기 단계 ( 에서의 비교 결과, 후보물질이 처리된 시험 시료의 단백질- 단백질 상호작용 수준 또는 四묘2 및/또는 요3의 타이로신 카이네이즈 활성화 정도가 이 후보 물질이 처리되지 않은 시험 시료에서보다 낮은 경우(즉 상기 후보 물질이 제 1 단백질과 제 2 단백질 간 상호 작용 또는 四요2 및/또는 1?3의 타이로신 카이네이즈 활성화를 억제하는 경우), 상기 후보 물질이 四묘2, 四묘3, 또는 이들 모두(예컨대, 四1?2 -四묘3 헤테로다이머 형태)를 표적으로 하는(즉, 제 1 단백질과 제 2 단백질의 상호작용을 억제하거나 1?2 및/또는 1?3의 타이로신 카이네이즈 활성화를 억제하는) 약물로서 효과가 있는 것으로 확인하는 단계 (단계 (이)를 추가로 포함할 수 있다. 상기 후보약물이 처리된시험 시료의 단백질-단백질상호수준 또는 HER2및/또는 HER3의 타이로신 카이네이즈활성화정도가후보물질이 처리되지 않은 시험 시료에서보다 낮다고 함은 후보 약물이 처리된 시험 시료의 단백질-단백질 상호 수준 또는 HER2 및/또는 HER3의 타이로신 카이네이즈활성화정도가후보물질이 처리되지 않은시험 시료의 단백질- 단백질 상호 수준 또는 HER2 및/또는 HER3의 타이로신 카이네이즈 활성화 정도의 75%이하(즉, 25%이상억제), 50%이하(즉, 50%이상억제), 45% 이하, 40%이하, 35%이하, 30%이하, 25%이하(즉, 75%이상억제), 20% 이하(즉 80%이상억제), 15%이하, 또는 10%이하(즉, 90%이상억제)인 것을의미할수있다. Therefore, the method for confirming the efficacy of the candidate drug is a method for determining the efficacy of the candidate drug by comparing the protein-protein interaction level of the test sample treated with the candidate substance or the tyrosine 4 of tetrahedral 2 and / When the degree of kinase activation is lower than that of the test sample in which this candidate substance is not treated (i.e., the candidate substance inhibits the interaction between the first protein and the second protein or the activation of tyrosine kinase of quadruplex 2 and / or 1-3) ), The candidate material is targeted to four quadrants 2, 4 quadrature 3, or both (e. G., In the form of a 4 1 2-quadrature 3 heterodimer) Or inhibiting tyrosine kinase activation of 1? 2 and / or 1? 3) (step (i) . The level of protein-protein interaction level or tyrosine kinase activation level of HER2 and / or HER3 of the test sample treated with the candidate drug is lower than that of the test sample not treated with the candidate substance. This means that the protein- Mutual levels or degree of tyrosine kinase activation of HER2 and / or HER3 is less than or equal to 75% (i.e., greater than or equal to 25% inhibition) of the protein-protein interfering level of the test sample to which the candidate agent has not been treated or the degree of tyrosine kinase activation of HER2 and / ), 50% or less (i.e., 50% or more inhibition), 45% or less, 40% or less, 35% or less, 30% or less, 25% or less Inhibition), 15% or less, or 10% or less (i.e., 90% or more inhibition).
상기 후보 물질은 제 1 단백질(HER2, HER3, 또는 이들 모두(예컨대, HER2-HER3 헤테로다이머 형태))의 표적 약물로 사용 가능한 모든 생체적합성 물질 중에서 선택될 수 있으며, 예컨대, 소분자 화학 약물 (small molecular chemicals) , 단백질 (예컨대, 항체, 항체 단편, 또는 이의 유사체 등), 펩타이드, 핵산분자(예컨대, DNA, RNA(예컨대, siRNA, microRNA, shRNA 등), PNA (peptide nucleic acid) , 앱타머, 등), 식물 추출물, 동물 추출물, 세포 추출물 등으로 이루어진 군에서 1종 이상 선택될수있으나, 이에 제한되는것은아니다.  The candidate agent may be selected from any biocompatible material available as a target drug for the first protein (HER2, HER3, or both (e.g., HER2-HER3 heterodimer form)), such as small molecular (eg, DNA, RNA (eg, siRNA, microRNA, shRNA, etc.), PNA (peptide nucleic acid), aptamer, etc.), proteins (eg, antibodies, antibody fragments or analogs thereof) ), Plant extracts, animal extracts, cell extracts, and the like, but the present invention is not limited thereto.
상기 시험 시료는 제1 단백질(HER2, HER3, 또는 이들 모두(특히, HER2-HER3 헤테로다이머 형태))이 과발현 및/또는 (과)활성화된 세포 (예컨대, 세포용해액 등) 또는조직 (예컨대, 조직 용해액 등), 또는 제 1 단백질과관련된 질병 또는효능을확인하고자하는제 1단백질을표적으로 하는 약물의 적용(치료) 대상 질병과 관련된 분리된 세포 (예컨대, 세포 용해액 등) 또는조직 (예컨대, 조직 용해액 등)일 수 있으며, 일 예에서, 확립된 세포주 또는 상기 질병 (예컨대, 암)을 앓는 환자로부터 분리된 세포또는 조직일 수 있다. 예컨대, 상기 시험 시료는 확립된 암 세포주 또는 암 환자로부터 분리된 세포 또는 조직일 수 있다 (상기 암은 제 1 단백질의 과발현및/또는(과)활성화와관련된 것일수있다).  The test sample may be a cell (e.g., a cell lysate, etc.) or a tissue (e.g., a cell lysate, etc.) overexpressing and / or activating a first protein (HER2, HER3, or both (in particular, a HER2-HER3 heterodimer form) (E.g., cell lysate) or tissue (e.g., cell lysate) associated with the disease (s) involved in the application (treatment) of a drug targeting the first protein for which the disease or efficacy associated with the first protein is to be identified Tissue lysate, etc.), and in one example, may be a cell or tissue isolated from an established cell line or a patient suffering from the disease (e.g., cancer). For example, the test sample may be a cell or tissue isolated from an established cancer cell line or cancer patient (the cancer may be associated with overexpression and / or activation of the first protein).
상기 단계 (a)내지 ( 는 in F/ZTO에서 수행되는것일수있다. 각단계의 용어 설명은앞서 기재한바와같다.  The steps (a) to () may be performed in F / ZTO. The terminology of each step is as described above.
본 명세서에 사용된 바로서, 용어 제 1 단백질을 표적으로 한다'’ 함은 제1 단백질(HER2, HER3, 또는 이들 모두(예컨대, HER2-HER3 헤테로다이머 형태))의 활성을촉진또는저해하는것을의미할수 있으며, 2019/132517 1»(:1^1{2018/016675 As used herein, the term "targeting a first protein" refers to promoting or inhibiting the activity of a first protein (HER2, HER3, or both (eg, a HER2-HER3 heterodimer form) You can, 2019/132517 1 »(: 1 ^ {2018/016675
예컨대, 제 1 단백질 0恨1?2, 1抑?3, 또는 이들 모두(예컨대, 他1?2 -他묘3 헤테로다이머 형태))의 활성(타이로신 카이네이즈 활성)을 저해하는 것을 의미할 수 있다. 제 1 단백질의 활성의 저해는 제 1 단백질과 결합하거나, 및/또는제 1단백질을분해 및/또는구조적 변형시켜 고유의 기능, 예컨대, 5 세포 및/또는 조직에서의 고유의 생체 신호전달 기능을 감소시키거나 제거하는것일수있다. May mean inhibiting the activity (tyrosine kinase activity) of the first protein, e.g., the first protein, the second protein, the second protein, the third protein, or both (e.g., other 1? . Inhibition of the activity of the first protein may be associated with the first protein and / or by degradation and / or structural modification of the first protein to provide a unique function, e. G., A unique bio signaling function in 5 cells and / It can be reduced or eliminated.
상기 투여는 경구 또는 비경구 경로로 수행될 수 있다. 비경구 투여인 경우에는 정맥내 주입, 피하 주입, 근육 주입, 복강 주입, 내피 투여, 병변부위 국소투여, 비내 투여, 폐내 투여, 또는직장내 투여 등의 10 경로로수행될수있다. The administration can be by oral or parenteral routes. In the case of parenteral administration, it can be carried out by 10 routes such as intravenous infusion, subcutaneous infusion, muscle infusion, intraperitoneal injection, endothelial administration, local administration of lesion site, intranasal administration, intrapulmonary administration or rectal administration.
상기 개체, 환자 또는 대상은 모든 포유류, 예컨대 인간, 원숭이 등의 영장류, 마우스, 래트 등의 설치류 등일 수 있고, 예컨대 제 1 단백질과관련된 질병을 갖는환자일 수 있다. 상기 제 단백질과관련된 질병은 제 1 단백질의 과발현 또는 제 1 단백질이 관여하는 세포 또는 조직 15 내 및/또는 세포 또는 조직 간의 신호전달경로의 활성화와 관련된 질병일 수 있으며, 예컨대, 암, 염증, 그외 면역질환등일수 있다. 구체예에서, 상기 개체, 환자또는대상은암환자일수있다. The subject, patient or subject may be any mammal such as a primate such as a human, a monkey, a rodent such as a mouse, a rat, or the like, and may be, for example, a patient having a disease associated with the first protein . The disease associated with the said protein may be a disease associated with overexpression of the first protein or activation of a signal transduction pathway in a cell or tissue 15 and / or between cells or tissues in which the first protein participates, such as cancer, inflammation, Immune diseases, and the like. In an embodiment, the subject, patient or subject may be a cancer patient.
상기 암은모든고형암및 혈액암중에서 선택될수 있으며, 예컨대 제 1 단백질의 과발현 또는 제 1 단백질이 관여하는 세포 또는 조직 내 20 및/또는 세포 또는 조직 간의 신호전달경로의 활성화와 관련된 암일 수 있다. 예컨대, 상기 암은 편평상피세포암, 소세포폐암, 비소세포폐암, 폐의 선암, 폐의 편평상피암, 복막암, 피부암, 피부 또는 안구내 흑색종, 직장암, 항문부근암, 식도암, 소장암, 내분비선암, 부갑상선암, 부신암, 연조직 육종, 요도암, 만성 또는 급성 백혈병, 림프구 림프종, 위암, 25 췌장암, 교아종, 경부암, 난소암, 방광암, 유방암, 결장암, 대장암, 자궁내막 또는 자궁암, 침샘암, 신장암, 전립선암, 음문암, 갑상선암, 두경부암, 뇌암등으로이루어진군에서 선택된 1종이상일수 있으나이에 제한되는것은아니다. 상기 암은원발성 암뿐아니라전이성 암을포함할 수 있다. 또한, 상기 암은 기존의 항암 치료에 대하여 저항성을 갖거나 30 획득한암일수있다. The cancer may be selected from all solid tumors and blood cancers and may be, for example, a cancer associated with overexpression of a first protein or activation of a signal transduction pathway between a cell or tissue 20 and / or a cell or tissue involved in the first protein. For example, the cancer is selected from the group consisting of squamous cell carcinoma, small cell lung cancer, non-small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, peritoneal cancer, skin cancer, skin or intraocular melanoma, rectal cancer, adenocarcinoma, part thyroid cancer, adrenal cancer, soft tissue sarcoma, uterine cancer, chronic or acute leukemia, lymphocytic lymphomas, gastric cancer, 25 pancreatic cancer, schools subspecies, cervical cancer, ovarian cancer, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial or uterine cancer, salivary gland Cancer, kidney cancer, prostate cancer, mucin cancer, thyroid cancer, head and neck cancer, brain cancer, and the like, but the present invention is not limited thereto. The cancer may include metastatic cancer as well as primary cancer. In addition, the cancer may be resistant to conventional chemotherapy or obtained 30 times .
본 명세서에 사용된 바로서, 용어 "약물에 대한 반응성’’은 약물을 투여받은개체에서 상기 약물이 효과를발휘하는정도를의미할수있다. 본 명세서에 사용된 바로서, 용어 ”효과’’는 약물 또는 치료가 처치 대상에서 달성하고자하는의학적 및/또는 약학적 효과를의미하는것으로, 처치 대상이 갖는질병의 예방및/또는치료, 및/또는증상의 완화및/또는 개선 등을 의미하는 것일 수 있다. 예컨대, 상기 약물이 항암제이고 치료가항암치료이고, 상기 처치 대상이 암환자인 경우, 상기 효과는항암 효과 (암의 예방및/또는치료효과)일수 있고, 상기 항암효과는암세포의 성장을 억제하는 효과뿐 아니라, 이동 (migrat ion), 침습 ( invasion) , 및/또는 전이 (metastas is) 등을 억제, 및/또는 암의 악화를 억제, 및/또는 저항성을감소또는제거하는효과등을포함하는것일수있다. As used herein, the term "responsiveness to a drug " may refer to the degree to which the drug exerts an effect on an individual receiving the drug. As used herein, the term & Medication or treatment cured Means a medical and / or pharmaceutical effect intended to be achieved by the subject, which may mean prevention and / or treatment of the disease to be treated, and / or alleviation and / or amelioration of symptoms. For example, when the drug is an anticancer agent and the treatment is chemotherapy and the subject is a cancer patient, the effect may be an anticancer effect (prevention and / or therapeutic effect of cancer), and the anticancer effect may inhibit the growth of cancer cells As well as the effects of inhibiting migration, invasion, and / or metastasis, and / or inhibiting the deterioration of cancer and / or reducing or eliminating resistance, and the like. have.
다른 예에서, 앞서 설명한 세포 또는 조직 내 및/또는 세포 또는 조직 간의 신호전달경로의 활성화 측정, 제 1 단백질을 표적으로 하는 약물에 대한 반응성 예측 방법, 제 1 단백질을 표적으로 하는 약물에 대한 반응성 모니터링 방법, 제 1 단백질을표적으로하는 치료에 적합한 개체를 선별하는 방법, 및/또는 약물의 스크리닝 방법에 사용하기 위한 단백질- 단백질 상호작용 측정용 장치를 제공한다. 상기 단백질-단백질 상호작용 측정용 장치는 조직 내의 신호전달경로의 활성화 측정용 장치, 제 1 단백질을표적으로하는약물에 대한반응성 예측및/또는모니터링용장치, 제 1 단백질을 표적으로 하는 치료에 적합한 개체를 선별하기 위한 장치, 및/또는 제 1 단백질을 표적으로 하는 약물의 효능 확인용 장치로서 적용 가능하다.  In another example, a method for measuring activation of a signal transduction pathway in a cell or tissue and / or between cells or tissues as described above, a method for predicting responsiveness to a drug targeting the first protein, a method for monitoring reactivity for a drug targeting the first protein There is provided a method for measuring protein-protein interactions for use in a method for screening a first protein, a method for selecting an individual suitable for treatment targeting the first protein, and / or a method for screening a drug. The apparatus for measuring protein-protein interactions may be a device for measuring activation of a signal transduction pathway in a tissue, an apparatus for predicting and / or monitoring reactivity to a drug targeting the first protein, a device suitable for treatment targeting the first protein An apparatus for screening an individual, and / or an apparatus for confirming the efficacy of a drug targeting the first protein.
일 예에서, 상기 장치에서의 제 1 단백질은 PD-L1 또는 PD-1일 수 있다.  In one example, the first protein in the device may be PD-L1 or PD-1.
다른 예에서 , 상기 장치에서의 제 1단백질은 HER2, HER3, 또는 이들 모두 (예컨대, HER2-HER3헤테로다이머 형태)일수있다.  In another example, the first protein in the device may be HER2, HER3, or both (e. G., A HER2-HER3 heterodimer form).
상기 단백질-단백질 상호작용측정용장치는 앞서 설명한세포또는 조직 내 및/또는 세포 또는 조직 간의 신호전달경로의 활성화 측정, 제 1 단백질을 표적으로 하는 약물에 대한 반응성 예측 방법, 제 1 단백질을 표적으로 하는 약물에 대한 반응성 모니터링 방법, 제 1 단백질을표적으로 하는 치료에 적합한 개체를 선별하는 방법, 및/또는 약물의 스크리닝 방법에 적용됨으로써, 소량의 시료에서의 생체 분자 간 상호 작용도 정확하고효율적으로 관찰, 분석, 검출, 및/또는측정할수 있다는 이점을 갖는다. 단백질-단백질 상호작용 측정용 장치 또는 이를 이용하는 분석 방법은 생검 (예컨대, needle biopsy) 시료와 같이 매우 소량의 시료에 대해서도유용하고효과적으로적용될수 있다. 또한, 상기 단백질-단백질 2019/132517 1»(:1^1{2018/016675 The device for measuring protein-protein interactions can be used for measuring the activation of a signal transduction pathway in cells or tissues and / or between cells or tissues, a method for predicting the reactivity of a drug targeting a first protein, The present invention can be applied to a method for monitoring the reactivity of a drug to a drug, a method for screening an individual suitable for treatment targeting the first protein, and / or a screening method for a drug, so that the interaction between biomolecules in a small amount of sample can be accurately and efficiently Can be observed, analyzed, detected, and / or measured. An apparatus for measuring protein-protein interactions or an analytical method using the same may be usefully and effectively applied to very small amounts of samples such as biopsy (e.g., needle biopsy) samples. In addition, the protein-protein 2019/132517 1 »(: 1 ^ {2018/016675
상호작용 측정용 장치 또는 이를 이용하는 분석 방법은 다양한 생체 분자 (예컨대, 단백질, 핵산 등) 간의 상호작용을 소량의 시료를 이용하여 정확하고효율적으로관찰, 분석, 검출, 및/또는측정할수있다. The interaction measurement device or analysis method using the same can accurately and efficiently observe, analyze, detect, and / or measure interaction between various biomolecules (e.g., protein, nucleic acid, etc.) using a small amount of sample.
상기 단백질-단백질상호작용측정용장치는,  The apparatus for measuring protein-protein interactions includes:
제 1 단백질에 특이적으로 결합하는 제 1 단백질의 포획용 물질을 포함하는멀티 웰을포함하는것일수있고,  A multiwell containing a substance for capturing a first protein that specifically binds to the first protein,
여기에 더하여, 신호검출수단을추가로포함할수있다.  In addition, it is possible to additionally include a signal detecting means.
상기 단백질-단백질 상호작용 측정용 장치는 상기 제 1 단백질과 상호작용하는 (예컨대, 상기 제 1단백질이 관여하는세포또는조직의 생체 신호전달경로의 하위 경로에 관여하는) 단백질 (제 2 단백질)을 추가로 포함할 수 있으며, 일 예에서, 상기 제 2 단백질은 검출 가능한 신호를 발생시키는표지 물질로표지되거나(표지 물질이, 예컨대, 화학적 (예컨대, 공유적 또는 비공유적), 재조합적, 또는 물리적으로, 결합되거나), 표지물질이 결합될 수 있는 13당이 부착된 형태일 수 있다. 또한, 상기 단백질-단백질 상호작용 측정용 장치는 신호 검출 수단에서 측정된 신호값을 제 1 단백질의 수준으로 정상화 (1111 1 1011)시키기 위하여, 제 1 단백질에 결합하는 탐지용 물질 및 상기 탐지용 물질에 결합하는 표지용물질을추가로포함할수있다. The apparatus for measuring protein-protein interaction includes a protein (second protein) that interacts with the first protein (e.g., participates in a lower pathway of a biological signal transduction pathway of a cell or tissue involved in the first protein) In one example, the second protein is labeled with a labeling substance that generates a detectable signal (the labeling substance may be chemically (e.g., covalent or noncovalent), recombinant, or physical , Or it may be in the form in which 13 sugars to which the labeling substance can be attached are attached. Also, the protein-protein interaction measuring devices for the order to (11 1 1 011 11) level with the normalization of the first protein, the signal measured by the signal detecting means, detecting substances and the for binding to the first protein, A labeling substance that binds to the detection substance may be further included.
상기 제 1 단백질에 결합하는 탐지용 물질은 앞서 설명한 멀티 웰에 포함된 제 1 단백질의 포획용 물질과 상이한 부위에서 제 1 단백질과 결합하는생물분자(예컨대, 항체)일수 있으며, 상기 표지용물질은검출 가능한 신호를 발생시키는 표지 물질로 표지되고 (표지 물질이, 예컨대, 화학적 (예컨대, 공유적 또는 비공유적으로 결합). 상기 제 1 단백질에 결합하는탐지용물질에 결합가능한생물 분자 (예컨대, 항체)일 수 있다 (도 6참조). The detection substance binding to the first protein may be a biomolecule (for example, an antibody) binding to the first protein at a site different from the capturing substance for the first protein contained in the multiwell described above, (The labeling substance is chemically (e . G., Covalently or non-covalently bound)) to generate a detectable signal . (E. G., An antibody) capable of binding to a detection substance that binds to the first protein (see Fig. 6).
제 1단백질이 ?^-11또는 1)-1인경우, 상기 단백질-단백질상호작용 측정용 장치는, 제 1 단백질에 특이적으로 결합하는 제 1 단백질의 포획용 물질을포함하는반응부를포함하는것일 수 있고, 여기에 더하여 신호검출 수단을추가로포함할수있다.  If the first protein is? - 11 or 1) -1, the apparatus for measuring protein-protein interactions includes a reaction unit comprising a substance capable of capturing a first protein that specifically binds to a first protein And may additionally include signal detection means.
상기 단백질-단백질 상호작용 측정용 장치는 1)-[그과 ¥1)-1 간의 상호작용(결합) 여부및/또는정도를측정하는데사용될수있다.  The apparatus for measuring protein-protein interactions can be used to measure the presence and / or the degree of interaction (binding) between 1) - [the and 1) -1.
제 1 단백질이 P^-ll 또는 므1)-1인 경우, 상기 반응부는 표면에 제 1 단백질포획용물질이 고정화된 기판을포함할수 있으며, 상기 제 1단백질 2019/132517 1»(:1^1{2018/016675 When the first protein is P? -Ll or (1) -1, the reaction unit may include a substrate on which a substance for capturing a first protein is immobilized, 2019/132517 1 »(: 1 ^ {2018/016675
포획용 물질은 앞서 제 1 단백질이 고정화된 기판 준비 단계 부분에서 설명한 제 1 단백질에 특이적으로 결합하는 물질, 예컨대, 항체 또는 이의 항원결합단편일 수 있다. 예컨대, 상기 항체 또는 이의 항원결합단편은, 앞서 설명한 바와 같이, 나과 [)-1 간의 결합부위인 1)-1그의 -말단 부위를 제외한 비결합 부위 (예컨대, (:-말단 부위)에 결합하는 항
Figure imgf000064_0001
항체 또는 이의 항원결합단편,
Figure imgf000064_0003
간의 결합부위인
Figure imgf000064_0002
1의 말단부위를 제외한비결합부위 (예컨대, 0말단부위)에 결합하는 항 ?0-1 항체 또는 이의 항원결합단편일 수 있다. 상기 반응부는 웰 (예컨대, 멀티웰) 타입, 슬라이드 타입, 채널 타입, 어레이 형태, 미세유체칩, 미세관 (캐필러리) 등일 수 있으나, 이에 제한되는 것은 아니다.
The capturing material may be a substance that specifically binds to the first protein described above in the substrate preparation step in which the first protein is immobilized, for example, an antibody or an antigen-binding fragment thereof. For example, the antibody or antigen-binding fragment thereof, as described above, binds to a non-binding site (for example, (: -terminal site) other than the binding site between 1) -1 term
Figure imgf000064_0001
Antibody or antigen-binding fragment thereof,
Figure imgf000064_0003
Linkage
Figure imgf000064_0002
1 antibody or an antigen-binding fragment thereof which binds to a non-binding site (for example, the 0-terminal site) except for the terminal region of the antibody. The reaction unit may be, but is not limited to, a well (e.g., multiwell) type, a slide type, a channel type, an array type, a microfluidic chip, a microtubule (capillary), and the like.
제 1단백질이 p^-ll또는抑- 1인경우, 상기 단백질-단백질상호작용 측정용 장치는 상기 제 1 단백질과 상호작용하는 제 2 단백질을 추가로 포함할 수 있다. 일 예에서, 상기 제 2 단백질은 검출 가능한 신호를ᄊ 발생시키는표지 물질로표지되거나(표지 물질이, 예컨대, 화학적 (예컨대, 공유적 또는 비공유적), 재조합적, 또는 물리적으로, 결합되거나), 표지물질이 결합될수있는 이 부착된형태일수있다. If the first protein is p -ll or suppress-1, the apparatus for measuring protein-protein interaction may further comprise a second protein interacting with the first protein. In one example, the second protein is labeled with a labeling substance that generates a detectable signal (the labeling substance is chemically (e. G., Covalent or noncovalent), recombinantly or physically coupled) It may be in the form of a 3- sided attachment to which a labeling substance can be attached.
제 1단백질이 ?^-11또는抑- 1인 경우, 상기 제 1단백질은抑- 1그및 ?å)-1중에서 선택된 어느하나이고, 제 2단백질은다른하나이다. 예컨대, 제 1 단백질이 므1)-1그인 경우, 상기 제 2 단백질은 ?1)-1이며, 상기 제 1 단백질이抑- 1인경우, 상기 제 2단백질은 1)-1그이다.  If the first protein is? - 11 or suppress-1, the first protein is any one selected from the group consisting of suppression-1 and suppression-1, and the second protein is the other. For example, when the first protein is 1) -1, the second protein is 1) -1, and when the first protein is suppressed 1, the second protein is 1) -1.
상기 제 1단백질이, 四1?2 및 四 1«, 특히, 1世묘2 -四묘3헤테로다이머인 경우, 상기 분석 장치는, 제 1 단백질에 특이적으로 결합하는 제 1 단백질의 포획용 물질 또는 제 1 단백질을 포함하는 반응부를 포함하거나, 여기에 더하여, 신호검출수단을추가로포함하는것일수있다.  In the case where the first protein is a 4-1,2 and 4-1 < - >, in particular a 1-thymine 2- 4-tetrahedral heterodimer, the analyzing apparatus is capable of capturing a first protein specifically binding to the first protein A substance or a first protein, or may additionally include signal detection means.
일 예에서, 상기 제 1단백질은}世요2 -四묘3헤테로다이머 , 또는 1世요2- 1世1?3 헤테로다이머를 세포 표면에 포함하는 (발현하는) 세포 또는 상기 세포의 용해물 형태로 사용될 수 있으며, 상기 반응부는 (1) }世묘2 -四요3 헤테로다이머, 또는 則及2 -四1?3 헤테로다이머를 세포 표면에 포함하는 (발현하는) 세포 또는 상기 세포의 용해물, 또는 (2) 이들이 고정화된 기판을 포함하는 것일 수 있다. 상기 四묘2 - 묘3 헤테로다이머를 세포 표면에 포함하는 (발현하는) 세포는 1世1¾ 및 抑요3를 발현하고, 하기의 설명되는 이합체화 제제 (예컨대, 他요3의 리간드 (예컨대 |31 (NRGbl) 등), 콜레스테롤-유사 세정제 (Cholesterol-1 ike detergent ; 예컨대, DGTN(digi tonin; CAS Number : 11024-24-1) , GDN(glyco卜 diosgenin; CAS Number : 1402423-29-3) 등으로 이루어진 군에서 선택된 1종 이상)이 처리된 것일수 있으며, 예컨대, HER2및 HER3를발현하고, ■(¾;[이 처리된 세포로서, 임의로 DGTN 및 GDN로 이루어진 군에서 선택된 1종 이상의 콜레스테롤-유사세정제로처리된세포또는세포용해물일수있다. In one example, the first protein comprises a cell that comprises (expresses) a 2-quaternary 3-heterodimer or a 1-12 (3) heterodimer on a cell surface or a lysate of the cell And the reaction part can be used in the form of (1) a cell that contains (expresses) a 2-quaternary 3-heterodimer or a 2-quaternary heterodimer on the cell surface or a cell Seaweed, or (2) a substrate on which they are immobilized. Cells expressing (expressing) the quadrature 2-quadrant 3 heterodimer on the cell surface express the quadruplex and inhibitor 3 and are capable of expressing a dimerizing agent (eg, a ligand of other moieties (eg, 31 (NRGbl), etc.), cholesterol-like cleanser (Cholesterol-1 ike detergent; For example, DGTN (digi tonin; CAS Number: 11024-24-1), GDN (glyco-diosgenin; CAS Number: 1402423-29-3), and may be, for example, one expressing HER2 and HER3 and expressing DGTN and GDN Or a cell lysate treated with at least one cholesterol-like detergent selected from the group consisting of < RTI ID = 0.0 >
상기 분석 장치는 HER2및 HER3, 특히, HER2-HER3헤테로다이머어와 이들의 신호전달경로상의 하위 단백질 간의 상호작용(결합) 여부 및/또는 정도, 또는 HER2 및/또는 HER3의 활성화 정도 (예컨대, Tyr 잔기의 인산화 정도)를측정하는데사용될수있다.  The analyzer is capable of detecting the presence and / or degree of interaction (binding) of HER2 and HER3, particularly HER2-HER3 heterodimers and their downstream proteins in the signal transduction pathway, or the degree of activation of HER2 and / or HER3 ≪ / RTI >
상기 반응부는표면에 제 1단백질포획용물질이 고정화되거나, 상기 제 1 단백질이 포획용 물질을 통하거나 통하지 않고(직접적으로) 고정화된 기판을 포함할 수 있다. 상기 제 1 단백질은 HER2 및 HER3, 특히 , HER2- HER3헤테로다이머이며, 상기 제 1단백질포획용물질은앞서 제 1단백질이 고정화된 기판 준비 단계 부분에서 설명한 제 1 단백질 (HER2 및/또는 HER3)에 특이적으로 결합하는 물질, 예컨대, 항체 또는 이의 항원결합단편일 수 있다. 상기 반응부는 웰 (예컨대, 멀티웰) 타입, 슬라이드 타입, 채널 타입, 어레이 형태, 미세유체칩, 미세관 (캐필러리) 등일수있으나, 이에 제한되는것은아니다.  The reaction unit may include a substrate on which a substance for capturing a first protein is immobilized on a surface, or a substrate on which the first protein is immobilized (directly) without passing through a capturing substance. The first protein is HER2 and HER3, in particular HER2-HER3 heterodimer, and the first protein capturing substance is a protein having the first protein (HER2 and / or HER3) described in the substrate preparation step in which the first protein is immobilized Specific binding material, such as an antibody or an antigen-binding fragment thereof. The reaction unit may be, but is not limited to, a well (e.g., multi-well) type, a slide type, a channel type, an array type, a microfluid chip, and a microtubule (capillary).
상기 분석 장치는 제 1 단백질과 제 2 단백질 간 단백질-단백질 상호작용을 측정하는 장치일 수 있으며, 이를 위하여, 상기 제 1 단백질과 상호작용하는 제 2 단백질을 추가로 포함할 수 있다. 다른 예에서, 상기 분석 장치는 HER2 및/또는 HER3의 활성화 정도 (예컨대, Tyr 잔기의 인산화)를 측정하는 장치일 수 있으며, 이를 위하여 인산화된 Tyr에 특이적으로 결합하는 물질 (예컨대, 항체)를 추가로 포함할 수 있다. 일 예에서, 상기 제 2단백질또는인산화된 Tyr에 특이적으로결합하는물질은 검출 가능한 신호를 발생시키는 표지 물질로 표지되거나 (표지 물질이, 예컨대, 화학적 (예컨대, 공유적 또는 비공유적), 재조합적, 또는 물리적으로, 결합되거나), 표지물질이 결합될수 있는 tag이 부착된 형태일 수있다.  The analyzer may be a device for measuring protein-protein interaction between a first protein and a second protein, and may further comprise a second protein that interacts with the first protein. In another example, the analyzer may be a device for measuring the degree of activation of HER2 and / or HER3 (e.g., phosphorylation of a Tyr residue), and for this purpose, a substance (e.g., an antibody) that specifically binds phosphorylated Tyr May be further included. In one example, the substance that specifically binds to the second protein or phosphorylated Tyr is labeled with a labeling substance that generates a detectable signal (the labeling substance may be labeled, for example, chemically (e.g., covalently or noncovalently) Or physically), or may be attached to a tag to which a labeling substance can be attached.
일 실시예에 따른 단백질-단백질 상호작용 측정용 장치에 포함된 멀티 웰은 일면이 개방된 복수의 관을 포함하거나, 지지 플레이트에 이격 형성된 복수의 비관통형 홀 (예컨대, 지지 플레이트에 형성된 홈 형태)을 포함하는 구조로서, 상기 일면이 개방된 관 또는 비관통형 홀을 웰로 정의할 수 있으며, 상기 웰이 제 1 방향으로 2개 이상 배치관 일렬 구조가 상기 제 1 방향과 교차하는 제 2 방향으로 하나 존재하거나, 2개 이상 배치(격자구조)된 구조체를 의미할수 있다 (도 30 참조) . 이 때, 상기 멀티 웰은 상기 관 또는 비관통형 홀의 개방된 일면에 위치하는 시료 주입부, 상기 관 또는 비관통형 홀의 내부의 단백질-단백질 상호작용 (예컨대, 제 1 단백질과 제 2 단백질 간 상호작용)이 일어나는 반응부, 및 상기 관또는비관통형 홀의 내부와접촉하는적어도 일부의 내벽의 표면에 제 1 단백질의 포획용 물질 (예컨대, 제 1 단백질에 특이적으로 결합하는 물질(예컨대, 항체))이 고정화되거나 고정화 가능한 제 1 단백질의 포획부(또는기판)를포함할수 있다. 일 구체예에서, 상기 멀티 웰은제 1 단백질과 제 2 단백질 간 상호작용이 일어나는 반응부 (제 1 단백질과 제 2 단백질의 반응부: 제 1반응부)를포함하는하나이상의 웰및 제 1단백질과 제 1단백질에 결합하는탐지용물질이 결합하는반응부 (제 1단백질 탐지부: 제 2 반응부)를 포함하는 하나 이상의 웰을 포함할 수 있다. 상기 제 1 단백질에 결합하는탐지용물질은앞서 설명한바와같다. 상기 제 1단백질 탐지부는시험 시료내의 제 1단백질수준을측정하여, 장치에 포함된신호 검출 수단에서 측정된 신호값을 제 1 단백질의 수준으로 정상화 (normal izat ion)하는데사용될수있다. The multiwells included in the apparatus for measuring protein-protein interactions according to an embodiment may include a plurality of tubes whose one surface is open or a plurality of non-through holes (for example, grooves formed in the support plate )of Wherein the one or more non-through holes are defined as wells, and wherein the wells are arranged in a first direction in a second direction in which two or more arrangement tube array structures intersect the first direction, Or a structure having two or more layouts (grid structure) (see FIG. 30). In this case, the multi-well may include a sample injecting part located on one opened surface of the tube or the non-through hole, a protein-protein interaction inside the tube or the non-penetrating hole (for example, (For example, a substance that specifically binds to a first protein (for example, a substance that binds to an antibody ) May comprise a capture portion (or substrate) of the immobilized or immobilizable first protein. In one embodiment, the multi-well comprises one or more wells comprising a reaction portion (a reaction portion of the first protein and the second protein: the first reaction portion) where interaction between the first protein and the second protein takes place, And a reaction unit (first protein detection unit: second reaction unit) to which a detection substance binding to the first protein binds. The detection substance binding to the first protein is as described above. The first protein detection unit may be used to measure the first protein level in the test sample and to normalize the signal value measured by the signal detection unit included in the device to the level of the first protein.
상기 신호검줄 수단은 사용된 표지 물질에서 발생시키는 신호에 따라서 통상적으로사용되는모든신호검출수단일 수 있다. 예컨대, 상기 신호검출수단은신호자극부및 신호검출부를포함할수 있으며, 여기에 더하여 측정된신호를분석 (예컨대, 정량또는영상화)하는신호분석부를 추가로 포함할 수 있다. 일 예에서, 신호 자극, 신호 검출, 및 신호 분석은 각각 다른 부위에서 수행되거나, 이들 중 적어도 두 가지 이상이 하나의 부위에서 동시에 또는 연속하여 수행될 수 있다. 일 예에서, 상기 표지가 형광물질인 경우, 상기 신호 검출 수단은 형광 신호를 발생 및 검줄할수 있는모든수단들중에서 선택될 수 있으며, 예컨대, 형광신호 자극부 (예컨대, 광원), 및 형광신호검출부, 및/또는형광신호분석부를 포함할 수 있다. 일 구체예에서, 상기 신호 검출 수단은 전반사 형광 현미경(Total Internal Ref lect ion Fluorescence (TIRF) microscope) 또는 공초점 현미경 (광원 및 형광신호 검출용)을 포함하거나, 여기에 더하여, 형광카메라, 예컨대 EMCCD(Electron-mult iplying charge-coupled device) 카메라 또는 CMOS (Complementary metal oxide semi conductor) 카메라를 추가로포함하여, 광원공급및 형광신호의 영상화및/또는정량을수행할 수 있다. 상기 광원의 파장, 세기, 형광 카메라 측정 조건 (예컨대, 1 프레임당 노출시간, 레이저 파워, 카메라 gain값, 총 촬영 프레임 등)은 앞서 설명한바와같다. The signal detecting means may be any signal detecting means normally used according to a signal generated from the used labeling material. For example, the signal detecting means may include a signal stimulating portion and a signal detecting portion, and may further include a signal analyzing portion for analyzing (e.g., quantifying or imaging) the measured signal. In one example, signal stimulation, signal detection, and signal analysis may be performed at different sites, respectively, or at least two of them may be performed simultaneously or sequentially in one site. In one example, when the label is a fluorescent material, the signal detecting means may be selected from among all means capable of generating and detecting a fluorescent signal, and may include, for example, a fluorescent signal stimulating portion (e.g., a light source) , And / or a fluorescence signal analyzer. In one embodiment, the signal detection means comprises a Total Internal Reflection Fluorescence (TIRF) microscope or a confocal microscope (for detecting a light source and a fluorescence signal), or additionally a fluorescence camera, such as an EMCCD (Electron-multiplying charge-coupled device) A camera or a CMOS (complementary metal oxide semi conductor) camera may be further included to perform illumination and / or quantification of the light source and the fluorescent signal. The wavelength, intensity, and fluorescent camera measurement conditions (e.g., exposure time per frame, laser power, camera gain value, total shooting frame, etc.) of the light source are as described above.
상기 제 1 단백질, 기판, 제 1 단백질에 특이적으로 결합하는 물질, 제 2 단백질, 표지 물질, 상기 표지 물질에 따른 신호 검출 수단은 앞서 설명한바와같다.  The signal detecting means according to the first protein, the substrate, the substance specifically binding to the first protein, the second protein, the labeling substance, and the labeling substance are as described above.
본 명세서에서 제공되는 단백질-단백질 상호작용 측정용 장치는 다양한 생체분자 (예컨대, 단백질, 핵산 등) 간의 상호작용 (예컨대, 단백질-단백질 상호작용 등)을 관찰, 분석, 검출, 및/또는 측정하는데 유용하게 적용될수있다. The apparatus for measuring protein-protein interactions provided herein is capable of observing, analyzing, detecting , and / or measuring interactions (e.g., protein-protein interactions) between various biomolecules Can be usefully applied.
따라서, 다른 예는 분석하고자 하는 생체분자 (예컨대, 제 1 단백질)를포함하는시료를상기 단백질-단백질 상호작용측정용장치 또는 상기 장치에 포함된 멀티 웰에 접촉시키는 단계를 포함하는 생체분자 간 상호작용 (예컨대, 단백질-단백질 상호작용) 분석 방법을 제공한다. 상기 분석 방법은, 상기 접촉시키는 단계 이후에, 시료에서 발생하는 신호를 측정하는 단계를 추가로 포함할 수 있다. 이 때, 상기 신호는 생체분자 분석에 통상적으로사용되는모든신호들 (예컨대, 형광, 발광, 등)중에서 적절하게 선택될 수 있고, 상기 신호의 측정은 사용된 신호의 종류에 따라서 통상적으로사용되는모든방법들중에서 적절하게 선택하여 수행할 수 있다. 상기 생체분자는 생체로부터 분리된 단백질, 핵산, 세포등으로 이루어진 군에서 선택된 1종 이상일 수 있으며, 예컨대 단백질일 수 있다. 상기 생체분자가 단백질인 경우, 단백질-단백질 상호작용 분석에 사용되는 단백질-단백질상호작용측정용장치에 포함된 멀티 웰은표면에 분석 대상 단백질 중어느하나에 특이적으로결합하는분자(예컨대, 항체)가고정된 것일수있다.  Accordingly, another example is a method for measuring a protein-protein interaction, comprising contacting a sample containing a biomolecule (for example, a first protein) to be analyzed with a device for measuring the protein-protein interaction or a multi- (E. G., Protein-protein interaction). The analysis method may further include a step of measuring a signal generated in the sample after the contacting step. At this time, the signal can be appropriately selected from among all signals (for example, fluorescence, luminescence, etc.) conventionally used for biomolecule analysis, and the measurement of the signal is typically used according to the kind of the used signal All of these methods can be selected appropriately. The biomolecule may be at least one selected from the group consisting of proteins, nucleic acids, and cells isolated from a living body, and may be, for example, a protein. When the biomolecule is a protein, a multi-well contained in a device for measuring protein-protein interaction used for protein-protein interaction analysis is a molecule that specifically binds to one of the proteins to be analyzed on the surface (for example, ) May be fixed.
다른 예에서, 앞서 설명한 바와 같은 제 1 단백질에 특이적으로 결합하는 제 1 단백질의 포획용 물질을 포함하는 멀티 웰을 포함하는 단백질-단백질 상호작용 측정용 키트가 제공된다. 상기 단백질-단백질 상호작용 측정용 키트는 조직 내의 신호전달경로의 활성화 측정용 키트, 제 1 단백질을표적으로 하는 약물에 대한 반응성 예측 및/또는모니터링용 키트, 제 1 단백질을 표적으로 하는 치료에 적합한 개체를 선별하기 위한 키트, 및/또는 제 1 단백질을 표적으로 하는 약물의 효능 확인용 키트로서 적용가능하다. In another example, there is provided a kit for measuring protein-protein interactions comprising a multi-well comprising a substance capable of capturing a first protein that specifically binds to a first protein as described above. The kits for measuring protein-protein interactions include kits for measuring the activation of signal transduction pathways in tissues, kits for predicting and / or monitoring the response to a drug targeting the first protein, kits suitable for treatment targeting the first protein For screening individuals Kit, and / or kit for confirming the efficacy of a drug targeting the first protein.
본 명세서에 제공된 단백질-단백질 상호작용 측정용 장치는 소량의 시료에서의 생체 분자 간 상호 작용도 정확하고 효율적으로 관찰, 분석, 검출, 및/또는 측정할 수 있다는 이점을 갖는다. 따라서, 상기 멀티 웰 또는 이를 이용하는 분석 방법은 생검 (예컨대, needle biopsy) 시료와 같이 매우소량의 시료에 대해서도유용하고효과적으로적용될수있다. 다른예는 HER2와 HER3의 헤테로다이머 제조방법을제공한다. 상기 방법은, HER2와 HER3를 동시에 포함하거나 발현하는 세포에 이합체화 제제를 처리하는 단계를 포함할 수 있다. 상기 이합체화 제제는, 앞서 설명한 바와 같이, HER3의 리간드 (예컨대, Neuregul in p i (NRGbl); NP_039250, NP_039250.2 등), 콜레스테롤-유사 세정제 (Cholesterol-l ike detergent; 예컨대, DGTNCdigi tonin; CAS Number : 11024-24-1), GDN (glycol-diosgenin; CAS Number : 1402423-29-3 등) , 등으로 이루어진 군에서 선택된 1종 이상일 수 있다. 일 구체예에서, 상기 방법이 HER2와 The apparatus for measuring protein-protein interactions provided herein has the advantage of being able to accurately and efficiently observe, analyze, detect, and / or measure biomolecular interactions in small amounts of sample. Thus, the multiwells or analytical methods using them can be usefully and effectively applied to very small samples, such as biopsy (e.g., needle biopsy) samples. Another example provides a method for preparing heterodimers of HER2 and HER3. The method may comprise treating the dimerizing agent to cells that simultaneously express or express HER2 and HER3. The dimerization agent may be a ligand of HER3 (for example, Neuregul in pi (NRGbl), NP_039250, NP_039250.2, etc.), a cholesterol-like detergent (e.g., DGTNCdigi tonin; CAS Number : 11024-24-1), GDN (glycol-diosgenin; CAS Number: 1402423-29-3, etc.), and the like. In one embodiment, the method comprises contacting HER2 with
HER3를 동시에 발현하는 세포에 NRGbl을 처리 (주입)하는 단계를 포함하는 경우, 상기 NRGbl는 HER2와 HER3를 발현 후 용해 (lysis) 전에 세포에 주입될수 있다. 다른구체예에서, HER2와 HER3를동시에 발현하는세포에 콜레스테롤-유사 세정제를 처라하는 단계를 포함하는 경우, 상기 콜레스테롤-유사 세정제는 세포 용해 후의 세척 단계에 처리 (첨가)될 수 있다 (일 예에서, 상기 세포는 NRGbl가 10ng/mL 내지 1000ng/mL 또는 50ng/mL내지 200ng/mL의 양으로주입된 것일 수 있음) . 이 때, 사용되는 콜레스테롤-유사 세정제의 농도는 사용되는 세정제의 CMCCcr i t ical mi cel lar concent rat ion) 를 초과하는 농도일 수 있으며, 예컨대, 세포 용해물 (cel l lysi s)을기준으로, DGTN의 경우 0.05%(w/v) 이상, 0.05%(w/v) 초과, 또는 0.06%(w/v) 이상, 예컨대, 0.05 내지 5%(w/v) , 0.05 내지 4%(w/v) , 0.05내지 3%(w/v) , 0.05내지 2%(w/v)또는 0.06내지 2%(w八 0일 수 있고, 의 경우 0.003%(w八) 이상, 0.003%(w八 0 이상, 0.007%(w八 0 이상, 또는 0.01%(w/v) 이상, 예컨대, 0.003 내지 2%(w/v) , 0.005 내지 2%(w/v) , 0.007내지 2%(w/v) , 또는 0.01내지 2%(w八)일수있다. When the step of treating (injecting) NRGbl into cells simultaneously expressing HER3, the NRGbl may be injected into cells before expression after HER2 and HER3 lysis. In another embodiment, the cholesterol-like cleansing agent can be treated (added) to the washing step after cell lysis if it comprises the step of treating the cholesterol-like cleansing agent to cells simultaneously expressing HER2 and HER3 , The cell may be NRGbl injected in an amount of 10 ng / mL to 1000 ng / mL or 50 ng / mL to 200 ng / mL). In this case, the concentration of the cholesterol-like detergent to be used may be a concentration exceeding that of the cleaning agent used. For example, the concentration of the cholesterol-like detergent may be DGTN (W / v), 0.05% (w / v), or 0.06% (w / v) or more, such as 0.05 to 5% ), 0.05 to 3% (w / v), 0.05 to 2% (w / v) or 0.06 to 2% (W / v), 0.005 to 2% (w / v), 0.007 to 2% (w / v) ), Or 0.01 to 2% (w8).
【발명의 효과】  【Effects of the Invention】
본명세서에서 제공되는기술은세포내 및/또는세포간의 단백질- 단백질 상호작용을 측정함으로써, 상기 단백질이 관여하는 질병의 예방 및/또는 치료에 있어서 효과적인 약물을 스크리닝 또는 선택하는데 유용하게 적용될수있다. The techniques provided herein measure protein-protein interactions intracellularly and / or intercellularly, thereby preventing the disease that the protein is involved in And / or screening or selecting effective drugs for treatment.
【도면의 간단한설명】  BRIEF DESCRIPTION OF THE DRAWINGS
도 1은단분자단백질상호작용측정방법에 관한모식도이다.  Brief Description of the Drawings Fig. 1 is a schematic diagram of a method for measuring short-term molecular protein interactions.
도 2는 기판에 고정된 표적단백질 (제 1 단백질) 확인 결과를 보여주는그래프이다. FIG. 2 is a graph showing the result of identification of a target protein (first protein) immobilized on a substrate .
도 3은 형광표지된 상호작용 단백질 (제 2 단백질) 주입 후 단백질 상호작용을나타낸형광이미지이다.  Figure 3 is a fluorescence image showing protein interactions after infusion of the fluorescently labeled interacting protein (second protein).
도 4는도 3에서 관측된 PPI comp lex의 개수를정량화한그래프이다. 도 5는 주입된 세포 용해액 양에 따른 PPI complex 개수 변화를 보여주는그래프이다.  FIG. 4 is a graph quantifying the number of PPI compexes observed in FIG. FIG. 5 is a graph showing changes in the number of PPI complexes according to the amount of injected cell lysate.
도 6은 단분자 sandwi ch ELISA를 통하여 제 1 단백질을 정량하는 과정을보여주는모식도이다.  6 is a schematic diagram showing a process of quantifying a first protein through a monoclonal sandwich ELISA.
도 7은 단분자 sandwich ELISA 방법을 통하여 얻어진 특이도 (speci f i ci ty)결과를보여주는그래프이다.  FIG. 7 is a graph showing the specificity obtained by the single-molecule sandwich ELISA method.
도 8은세포주 종류 (빨간색① vs 하늘색③) 및 상태 (빨간색① vs 검은색②)에 따른 PPI complex개수의 변화를보여주는그래프이다.  FIG. 8 is a graph showing changes in the number of PPI complexes according to the cell line type (red ① vs light blue ③) and the state (red ① vs black ②).
도 9는세포의 상태에 따른다양한표적 RTK(제 1단백질) 별로 PPI complex개수변화를보여주는그래프이다.  FIG. 9 is a graph showing changes in the number of PPI complexes according to various target RTKs (first proteins) according to cell conditions.
도 10은 EGFR변이 상태에 따른 PPI complex의 변화및 이를토대로 세포당활성화된 EGFR의 비율계산한결과를보여주는그래프이다.  FIG. 10 is a graph showing the change in the PPI complex according to the EGFR mutation state and the calculated ratio of EGFR activated per cell based on the change. FIG.
도 11은 도 10에서 EGFR에 대하여 수행한 방법을 HER2, HER3에 대해서 동일하게수행한결과를보여주는그래프이다.  FIG. 11 is a graph showing the results of performing the same method for HER2 and HER3 performed on EGFR in FIG.
도 12는각각의 세포주에 대해 EGFR, MET, HER2, HER3 (제 1단백질) 와 하위신호전달 단백질(제 2 단백질) 사이의 상호작용을 모두 측정하여 heatmap형식으로표시한결과를보여준다.  Fig. 12 shows the result of measuring the interactions between EGFR, MET, HER2, HER3 (first protein) and the lower signal transduction protein (second protein) for each cell line and displaying them in a heatmap format.
도 13은도 12의 결과를 정량적으로 나타낸 그래프 (좌측 및 중간) 및 EGFR 표적항암제의 일종인 AZD9291의 반응성 결과를 보여주는 그래프(우측)이다.  FIG. 13 is a graph (left side and middle) graphically showing the results of FIG. 12 (left side) and a graph showing the reactivity results of AZD9291 (right side) as an EGFR target anticancer drug.
도 14는 EGFR 표적 항암제(AZD9291)의 반응성 (좌측, y축)과 Act ivat ion score (좌측, x축) 사이의 상관관계 및 유전자 타입에 따른 표적 항암제 반응의 다양성 (우측)을보여주는그래프이다.  FIG. 14 is a graph showing the correlation between the reactivity (left, y axis) of the EGFR target anticancer agent (AZD9291) and Act iat ion score (left, x axis) and the diversity of the target anticancer response (right side) according to the gene type.
도 15는 유방암 세포주에서 HER2와 HER3 신호의 세기를 나타낸 heatmap이다. 15 shows the intensity of HER2 and HER3 signals in breast cancer cell lines It is a heatmap.
도 16은 유방암 세포주에서 기존에 trastuzumab 항암제의 반응성을 예측하기 위해 사용되는 biomarker인 HER2 (상단), HER3 (중간) 발현량을 측정한 결과 및 trastuzumab에 의해 세포 성장이 억제되는 정도 (하단)를 측정하여 나타낸그래프이다.  FIG. 16 shows the results of measurement of biomarker HER2 (upper) and HER3 (intermediate) expression levels used for predicting the reactivity of trastuzumab anticancer drugs in breast cancer cell lines and the degree of suppression of cell growth by trastuzumab (lower) FIG.
도 17은 HER2또는 HER3신호를이용하여 PPI score를측정한결과와 trastuzumab반응성 (logGI50)사이의 상관관계를보여주는그래프이다. 도 18은 PDTX마우스모델에서 측정한 EGFR, MET, HER2, 및 HER3의 3종의 하위 신호 단백질과의 PPI complex 신호 결과를 각각 보여주는 heatmap이다.  17 is a graph showing the correlation between the result of measuring the PPI score using the HER2 or HER3 signal and the reactivity of trastuzumab (logGI50). Figure 18 is a heatmap showing the results of the PPI complex signal with the three sub-signal proteins of EGFR, MET, HER2, and HER3 measured in the PDTX mouse model, respectively.
도 19는 PDTX마우스모델에서 EGFR의 발현량(상단) 및상기 EGFR의 발현량을 이용하여 act ivat ion score를 계산한 결과 (하단)를 보여주는 그래프이다.  FIG. 19 is a graph showing the result of calculating the act iat ion score (bottom) using the expression level (top) of EGFR and the expression level of EGFR in the PDTX mouse model.
도 20은 PDTX 마우스 모델에 gef i t inib을 투여하여 종양크기의 변화를측정한결과를보여주는그래프이다.  20 is a graph showing the results of measurement of changes in tumor size by administering gefitinib to a PDTX mouse model.
도 21은 PDTX 마우스 모델에서 gef i t inib에 의한 종양크기 억제 Figure 21 shows tumor suppression by gef i t inib in a PDTX mouse model
(tumor growth inhibi t ion) 정도와 EGFR act ivat ion score 사이의 상관관계를보여주는그래프이다. (Tumor growth inhibi- tion) and EGFR act iat ion score.
도 22는 PDTX 마우스 모델에서 gef i t inib을 처방하기 전, 후의 조직에서 각각 측정된 EGFR PPI complex 개수 측정 결과를 보여주는 그래프이다.  FIG. 22 is a graph showing the results of measurements of EGFR PPI complexes measured in tissues before and after the administration of gef i t inib in a PDTX mouse model, respectively.
도 23a내지 23i는폐암 PDTX모델에서의 EGFR표적 저해제 반응성을 보여주는것으로,  Figures 23A-23I show EGFR target inhibitor reactivity in a lung cancer PDTX model,
23a는 PDTX모델생성 과정을예시적으로보여주는모삭도이고, 23b는 비히클또는표시된 EGFR-특이적 억제제 처리시의 PDTX에서의 종양부피 변화를 보여주는 그래프로서, 폐선암종 PDTXs (PDTX-A1 A3)에는 오시머티닙 (Osimert inib; 5 mg per 1 kg of weight dai ly)을 처리하고, 폐편평세포암(就 ICC) PDTXs (PDTX-S1 S5)에는 게피티닙 (gef i t inib; 50 mg per 1 kg of weight dai ly)을 처리하여 얻어진 종양크기 변화를 보여주며 (각 PDTX당시험 개체수는 3이상임),  23a and 23b are graphs showing tumor volume changes in PDTX during the treatment of the vehicle or the indicated EGFR-specific inhibitor, and PDTXs (PDTX-A1 A3) Osimert inib. (PDTX-S1 S5) was treated with 5 mg per kg of weight of dairy (PDTX). 50 mg per kg of weight dairy), showing the size of the tumors obtained (test population per 3 PDTX)
23c는 표시된 수용체 타이로신 카이네이즈 (RTK; EGFR, HER2, HER3 및 MET)의 하위신호단백질에 대한 PPI complex counts (PPI complex의 수)를보여주는그래프이고, 23d는 8마리의 PDTX (A1-A3및 S1~S5) 개체에서의 EGFR발현수준을 A549 세포 (대조군)에서의 EGFR 발현 수준으로 normal i zat ion한 결과를 보여주는그래프이며, 23c is the labeled receptor tyrosine kinase (RTK; EGFR, HER2, HER3, and MET), wherein the PPI complex counts (number of PPI complexes) 23d is a graph showing the results of normalization of EGFR expression levels in 8 PDTX (A1-A3 and S1-S5) individuals to EGFR expression levels in A549 cells (control group)
23e및 23f는종양성장억제율 (%)을 y축으로하고, 폐선암종 PDTX 모델 (E) 및 SQCC PDTX 모델 (0로부터 얻어진 EGFR PPI 합계를 EGFR 수준으로나눈값을 x축으로하여 나타낸그래프이고,  23e and 23f are graphs showing the tumor growth inhibition rate (%) as the y-axis and the value obtained by dividing the EGFR PPI sum obtained from the PDTX model (E) and the SQCC PDTX model (0) by the EGFR level as the x axis.
23g는 gef i t inib을 15일 동안 매일 처리한 경우의 PPI complex counts (EGFR와 x축에 기재된 제 2 단백질 간 PPI complex의 수)의 변화를 보여주는그래프이며,  23g is a graph showing changes in PPI complex counts (the number of PPI complexes between the second protein expressed on the EGFR and the x-axis) when the gefitinib was treated daily for 15 days,
23h는 PDTX-Sl(n=2)에 gef i t inib과 BKM120을 15일간 병용 처리한 경우의 종양성장정도를단독처리시와비교하여 보여주는그래프이고, 23h is a graph showing the degree of tumor growth when PDTX-Sl (n = 2) was treated with gefitinib and BKM120 for 15 days in comparison with that in single treatment,
23i는 8마리의 PDTX (A1-A3 및 S1-S5) 개체 모두에서 얻어진 EGFR 수준으로 PPI 합계를 나눈 값을 x축으로 하고 종양 성장 억제율 (%)를 y축으로하여 나타낸그래프이다 (Error bars: s.d. ) . 23i is a graph showing the value obtained by dividing the PPI sum by the EGFR level obtained from all 8 PDTX (A1-A3 and S1-S5) individuals as the x axis and the tumor growth inhibition rate as the y axis (Error bars: sd).
도 24a 내지 24d는 단분자 (single-molecule) co-IP 및 단분자 면역표지법 (singlenolecule immunolabel ing)을 .인간 종양 시료에 적용한 예를보여주는것으로,  Figures 24a to 24d show an example of applying single-molecule co-IP and singlenolecule immunolabeling to human tumor samples,
24a는 두 종양 환자 (P1 및 P2)의 종양 절제 수술로 얻어진 인간 종양조직을보여주며,  24a shows human tumor tissues obtained by tumor resection of two tumor patients (P1 and P2)
24b는 단분자 면역표지법에 의하여 얻어진 1◦개 단백질의 발현량과 24b is the amount of 1o protein produced by monoclonal immuno-labeling
PTM 수준 ( Immunolabel 1 ing level ) 및 고효율 단분자 영상화 시스템을 사용하여 각 시료를 단분자 co-IP에 의하여 얻어진 10개의 단백질-단백질 쌍의 PPI 수준을 보여주는 그래프로서, 양성대조군으로서 PC9 세포 ( for EGFR) , HCC827세포 ( for MET) , 및 SKBR3세포 ( for HER2및 HER3)가각각 사용되었으며, A graph showing the PPI levels of 10 protein-protein pairs obtained by monomolecular co-IP using PTM (Immunolabel 1 ing level) and a high-efficiency monomolecular imaging system. As a positive control, PC9 cells (for EGFR ), HCC827 cells (for MET), and SKBR3 cells (for HER2 and HER3)
24c는 P1 및 P2에서의 표시된 RTK에 대한 PPI complex counts를 보여주는그래프이고,  24c is a graph showing PPI complex counts for the indicated RTKs at P1 and P2,
24d는 표면 상에서 EGFR을 pul l ing down한후 PTPN1 처리한 경우의 PLCg-asH2및 Grb2의 PPI complex count 변화를보여주는그래프이다 (Error bars : s . d . ) . 24d is a graph showing the change in count PPI complex PLC ga s H2 and Grb2 in the case of the EGFR on the surface treatment hanhu pul l ing down PTPN1 (Error bars :. S d.).
도 25a내지 25h는 PDTX-model (n=3)의 특성을보여주는것으로, Figs. 25A to 25H show the characteristics of the PDTX-model (n = 3)
25a내지 25c는 MET수준 (a) , HER2수준 (b) , 및 HER3수준 (c)을 HCC827세포 ( for MET) 및 SKBR3세포 ( for HER2및 HER3)에서의 MET, HER2, 및 HER3의 수준과비교한결과를보여주는그래프로서 (Error bars: s.d.; JT=5), 이들 RTK중어느것도과발현되지 않았음을확인할수있고, 25a to 25c show MET, HER2, and HER3 levels in HCC827 cells (for MET) and SKBR3 cells (for HER2 and HER3) for MET levels (a), HER2 levels And HER3 levels (Error bars: sd; JT = 5), indicating that these RTKs were not overexpressed,
25d는 대표적으로 5 마리의 SQCC PDTX에서 측정된 EGFR의 면역조직화학염색 (IHC) 결과를 보여주는 이미지이며, EGFR의 발현은 magnification rule로 EGFR H-score를계산하여산정하였고,  25d is an image showing the immunohistochemical staining (IHC) of EGFR measured in five SQCC PDTX. EGFR expression was calculated by calculating the EGFR H-score as a magnification rule,
25e는 단분자 면역표지법에 의해 결정된 EGFR수준과 EGFR H-score 간의 상관 관계를 보여주는 산포도로서, IHC H-score는 단분자 면역표지법에 의해 결정된 전체 EGFR발현수준과완전한선형 상관관계를 나타내는것으로확인되었으며,  25e is a scatter plot showing the correlation between EGFR level and EGFR H-score determined by monomolecular immuno-labeling, and IHC H-score has a complete linear correlation with total EGFR expression level determined by monomolecular immunodiffusion ,
25f 및 25g은 SQCC 卵 TX의 EGFR 수준 (g) 및 PPI 합계 (h)와 종양 성장억제의 상관관계를보여주는산포도이고,  25f and 25g are scatter diagrams showing the correlation between EGFR level (g) and PPI sum (h) of SQCC egg TX and tumor growth inhibition,
25h는 vehicle 또는 gefitinib 처리한 PDTX-S2의 Immunoblot 분석 결과를보여주는 것으로, 15일 gefitinib처리 후 EGFR의 1068번째 잔기인 티로신의 인산화 (pEGFR)가 완전히 사라지고, gefitinib 처리에 의하여 AKT 및 S6K의 인산화 (각각 pAkt와 pS6K)도 억제됨을보여주며, 이러한결과는 PDTX-S2에서의 종양 성장 억제 효과가 gefitinib 처리에 의한 EGFR/AKT/mT0R/S6K 신호전달 경로를 억제함으로써 얻어지는 것임을 보여준다.  25h shows Immunoblot analysis of vehicle or gefitinib-treated PDTX-S2. After 15 days of treatment with gefitinib, phosphorylation (pEGFR) of 1068th residue of EGFR is completely disappeared and phosphorylation of AKT and S6K by gefitinib treatment pAkt and pS6K), and these results show that the inhibition of tumor growth in PDTX-S2 is obtained by inhibiting the EGFR / AKT / mT0R / S6K signaling pathway by treatment with gefitinib.
도 26a 및 26b는 PDTX-S1및 PDTX-S2에서의 gefitinib 처리 효과를 보여주는것으로,  26A and 26B show the effects of gefitinib treatment on PDTX-S1 and PDTX-S2,
26a는 15일 동안 gefitinib 처리한 경우의 EGFR 수준 변화를 보여주는그래프이고 (Error bars: s.d.; TF5),  26a is a graph showing the change in EGFR level when treated with gefitinib for 15 days (Error bars: s.d .; TF5)
26b는 gefitinib 처리에 의한 EGFR PPI 억제 정도를 보여주는 그래프로, A549 세포의 EGFR PPI complex count를 음성대조군으로 나타내었다 (Error bars: s.d.; n=5) .  26b is a graph showing the degree of inhibition of EGFR PPI by gefitinib treatment and EGFR PPI complex count of A549 cells as a negative control group (Error bars: s.d .; n = 5).
도 27a는 GFP-PD-1과 mcherry PD-L1을각각발현시킨 세포용해액을 이용하여 단백질을 획득, 단백질-단백질 상호작용 분석, 및 정량하는 과정을예시적으로보여주는모식도이다.  FIG. 27A is a schematic diagram showing a process of acquiring a protein, analyzing protein-protein interaction, and quantifying using a cell lysate expressing GFP-PD-1 and mcherry PD-L1, respectively.
도 27b은 10nM의 GFP-卵 -1과 mcherry PD-L1을 사용하여 진행하여 얻어진 단백질-단백질 상호작용의 형광 이미지이고, 도 27c는 이를 정량한 결과 (PPI complex 개수; Fluorescence-count s; F-counts)를 보여주는 그래프이다.  27B is a fluorescence image of the protein-protein interaction obtained by using 10 nM of GFP-egg-1 and mcherry PD-L1. FIG. 27C shows the result of quantitative analysis (number of PPI complexes: fluorescence- counts).
도 28a는억제제후보약물스크리닝 과정을모식적으로보여준다. 도 28b는 약물 스크리닝을 위한 tube에서의 one-step react ion의 반응조건(반응시간)을최적화하기 위한시험 결과를보여주는그래프이다. 도 28c는 시판중인 두 종의 卵 -1/PD-L1 억제제인 BMS202 및 S7911 (이상, Sel leck Chem에서 구입)을 사용하여 얻어진 PD-1/PD-L1 간의 PPI complex개수(F-counts)를보여주는그래프이다. 28A schematically shows a process of screening for an inhibitor candidate drug. FIG. 28B is a graph showing test results for optimizing the reaction conditions (reaction time) of one-step reaction in a tube for drug screening. FIG. Figure 28c shows the number of PPI complexes (F-counts) between PD-1 / PD-L1 obtained using two commercially available egg-1 / PD-L1 inhibitors BMS202 and S7911 (purchased from Sel leck Chem) It is a graph showing.
도 28d는 PPI l ibrary screening (http:/八,. asinex.com/ppi/)에서 제공하는 non-macrocycl ic PPI drug l ibrary의 화합물을 사용하여 얻어진 卵 -1/PD-L1간의 PPI complex개수(F-counts)를보여주는그래프이다.  FIG. 28d shows the PPI librarians screening (http: / /. (F-counts) between egg-1 / PD-L1 obtained using a non-macrocyclic ic PPI drug libbrary compound provided by Asinex.com/ppi/.
도 28e는상기 도 28d에서 우수한억제 효능을보이는것으로선별된 4종의 약물 (Bl, B2, B3, 및 E2)을 대상으로 t i trat ion을진행하여 얻어진 결과를보여주는그래프이다.  FIG. 28E is a graph showing the results obtained by conducting t i tratation on four drugs (Bl, B2, B3, and E2) selected to exhibit excellent inhibitory effect in FIG. 28D.
도 29은 다양한 항체를 기판에 고정시키고 GFP-PD-1과 mcherry PD- L1을 사용하여 진행하여 얻어진 抑- 1/PD-L1 간의 PPI complex 개수(F- counts)를나타낸그래프이다.  29 is a graph showing the number of PPI complexes (F-counts) between inhib-1 / PD-L1 obtained by immobilizing various antibodies on a substrate and using GFP-PD-1 and mcherry PD-L1.
도 30a는기판에 GFP PD-L1을고정시킨 경우와, GFP-卵 -1을고정시킨 경우의 Total GFP- 1 abe 1 ed count (왼쪽) 및 형광 이미지 (오른쪽)를 보여준다.  FIG. 30A shows Total GFP-1 abe 1 ed count (left) and fluorescent image (right) when GFP PD-L1 is immobilized on a substrate and when GFP-egg-1 is immobilized.
도 30b는기판에 GFP PD-L1을고정시킨 경우와, GFP-PD-1을고정시킨 경우의 GFP PD-L1및 GFP抑- 1의 cluster rat io(%) (왼쪽) 및 형광이미지 (오른쪽)를보여준다.  FIG. 30B is a graph showing the relationship between the cluster rat io (%) (left) and fluorescence image (right) of GFP PD-L1 and GFP suppression-1 when GFP PD-L1 was immobilized on a substrate, Lt; / RTI >
도 31a는 에피토프 맵핑을 위한 PD-L1 mutant construct의 설계 과정을모식적으로보여준다.  31A schematically shows a design process of a PD-L1 mutant construct for epitope mapping.
도 31b는 PD-L1 ant ibody (9A11)의 PD-L1 WT (야생형), d280, 또는 d270변이체에 대한결합정도를보여준다.  FIG. 31B shows the degree of binding of PD-L1 antibody (9A11) to PD-L1 WT (wild-type), d280, or d270 variant.
도 31c는卵 -1와 PD-L1 WT, d280, 또는 d270 변이체 간의 단백질- 단백질상호작용정도를보여준다.  Figure 31c shows the degree of protein-protein interaction between egg-1 and PD-L1 WT, d280, or d270 variants.
도 32는 卵 -1와 PD-L1 간의 상호작용(결합) 부위를 모식적으로 보여준다.  32 schematically shows the interaction (binding) site between egg-1 and PD-L1.
도 33는약물 Bl, B2, B3, 및 E2의 구조를보여준다.  Figure 33 shows the structures of drugs Bl, B2, B3, and E2.
도 34a는 HER3-eGFP를 SKBR3에 발현한 뒤, NRGbl을 처리하여 HER2- Figure 34A depicts expression of HER3-eGFP in SKBR3, followed by treatment with NRGbl to generate HER2-
HER3-eGFP heterodimer를 형성시키는 과정을 예시적으로 보여주는 모식도이다. Lt; RTI ID = 0.0 > HER3-eGFP heterodimer. ≪ / RTI >
도 34b는상기 세포추출물에 존재하는 HER3-eGFP의 양에 따라 HER2 - HER3-eGFP heterodimer의 양이 비례함을보여주는그래프이다. FIG. 34B shows the HER2-eGFP concentration in the cell extract according to the amount of HER3- HER3-eGFP heterodimer. ≪ / RTI >
도 34c는 NRGbl를 세포 용해 전에 처리한 경우와세포 용해 이후에 처리한경우에 검출된 HER2-HER3 heterodimer의 양을보여주는그래프이다. 도 34d는 SKBR3세포에 NRGbl을처리하여 세포내에 존재하는 HER2와 HER3의 heterodimer를 유도, 추출, 및 검출하는 과정을 예시적으로 보여주는모식도이다.  FIG. 34C is a graph showing the amount of HER2-HER3 heterodimer detected when NRGbl was treated before and after cell lysis. FIG. FIG. 34D is a schematic diagram illustrating a process of inducing, extracting, and detecting heterodimers of HER2 and HER3 in cells by treating NRBbl with SKBR3 cells. FIG.
도 34e는기판에 첨가한세포추출물의 양에 따라검출된 HER2-HER3 heterodimer의 양을보여주는그래프이다.  Figure 34E is a graph showing the amount of HER2-HER3 heterodimer detected according to the amount of cell extract added to the substrate.
도 34f는 34d의 과정을 따라 추출된 세포 추출물을 다른 종류의 항체를 이용해 기판에 부착했을 때, HER2-HER3 heterodimer가 주요하게 존재함을 보여주는 그래프이다 (리간드를 처리하지 않았을 때, EGFR-HER3, EGFR-HER2, HER2-HER3 heterodimer , 리간드를 처리했을 때, EGFR-HER3 heterodimer는리간드를처리했을때 HER2-HER3 heterodimer의 양에 비하여 매우적다)  FIG. 34f is a graph showing that HER2-HER3 heterodimer is mainly present when a cell extract extracted according to the procedure of 34d is attached to a substrate using another kind of antibody (when the ligand is not treated, EGFR-HER3, When EGFR-HER2, HER2-HER3 heterodimer, and ligand were treated, the EGFR-HER3 heterodimer was much less than the amount of HER2-HER3 heterodimer when treated with ligand)
도 34g는 HER2-eGFP를四 K293T세포에 과발현시켜 HER2-eGFP - HER2- eGFP homodimer 형성하고 기판 (PEG, neutravidin, biot in, 및 ant i -HER2 ant ibody를 표면처리된) 에 부착하는 과정을 예시적으로 보여주는 모식도이다.  Figure 34g illustrates the process of overexpressing HER2-eGFP in four K293T cells to form a HER2-eGFP-HER2-eGFP homodimer and attaching it to a substrate (PEG, neutravidin, biot in, and ant i -HER2 ant ibody surface treated) It is a schematic diagram showing the enemy.
도 34h는 34g에서 기판에 부착된 HER2_eGFP - HER2-eGFP homodimer 또는 HER2-eGFP monomer로부터 나오는형광신호를측정한결과를보여준다. 도 34i는 34h에서 homodimer 또는 monomer로부터 얻은 형광신호에서 단분자 형광 신호 꺼짐 현상을 관찰해 2단계로 꺼짐 (dimer) , 과 2단계 이상으로 꺼짐 (ol igomer)의 비율을 나타내는 그래프이다. 2단계 및 2단계 이상의 비율이 HER2-eGFP의 경우가 eGFP또는 HER3_eGFP보다 많은 것을 알 수있다.  Figure 34h shows the results of measuring the fluorescence signal from the HER2_eGFP-HER2-eGFP homodimer or HER2-eGFP monomer attached to the substrate at 34g. FIG. 34I is a graph showing the ratio of the dimer to the fluorescence signal obtained from the homodimer or monomer at 34h, and the ratio of the fluorescence signal to the fluorescence signal at the second step and the olimeric state. FIG. It can be seen that the ratio of HER2-eGFP to that of step 2 and step 2 is higher than that of eGFP or HER3_eGFP.
도 35a는 HER2-HER3 heterodimer의 Tyr 인산화 수준 측정 과정을 예시적으로보여주는모식도이다.  FIG. 35A is a schematic diagram showing an example of a process for measuring the Tyr phosphorylation level of the HER2-HER3 heterodimer. FIG.
도 35b는 HER2-HER3 heterodimer가 고정화된 기판에 ATP와 ¾¾2+를 처리한 후 HER3 Tyr 잔기들의 인산화 정도 (HER3 pTyr counts)를 측정한 결과를보여주는그래프이다. FIG. 35B is a graph showing the results of measuring the degree of phosphorylation (HER3 pTyr counts) of HER3 Tyr residues after treating ATP and ¾ 2+ on a substrate immobilized with HER2-HER3 heterodimer.
도 35c 및 도 35d는 HER2-HER3 heterodimer 형성 세포 용해물을 다양한 농도의 digi tonin를 사용하여 세척하여 얻어진 HER3의 활성화 정도를 digi tonin 농도에 따른 HER3 Y1289의 인산화 정도로 보여주는 그래프이다. Figures 35c and 35d show the degree of activation of HER3 obtained by washing the HER2-HER3 heterodimer-forming cell lysate with various concentrations of digi tonin as the degree of phosphorylation of HER3 Y1289 according to digi tonin concentration Graph.
도 35e는 HER2-HER3 heterodimer 형성 세포용해물의 세척에 사용된 detergent 종류에 따른 HER3의 활성화정도를 HER3 Y1289의 인산화정도를 통하여 보여주는그래프이다.  FIG. 35E is a graph showing the degree of HER3 activation according to the kind of detergent used for washing the HER2-HER3 heterodimer-forming cell lysate through the degree of phosphorylation of HER3 Y1289.
도 35f는 HER2-HER3 heterodimer가 기존에 EGFR homodimer에 의해 알려진 것과동일한활성화 메커니즘을 따른다는 것을보이기 위해 EGFR과 상응하는 단일 위치 변이를 각각 HER2와 HER3에 시켰을 때, HER2-HER3 heterodimer의 활성화정도가감소하는것을보'여주는그래프이다. Figure 35f shows that the degree of activation of the HER2-HER3 heterodimer is decreased when HER2-HER3 heterodimers are subjected to HER2 and HER3, respectively, corresponding to EGFR, in order to show that they follow the same activation mechanism as previously known by the EGFR homodimer beam "gourd is a graph that.
도 35g는 HER2-HER2 homodimer의 Tyr 인산화 수준 측정 과정을 예시적으로보여주는모식도이다.  FIG. 35G is a schematic diagram showing an example of a process for measuring the Tyr phosphorylation level of the HER2-HER2 homodimer.
도 35h는 抑 R2-HER2 homodimer 가 고정화된 기판에 ATP와 당2+를 처리한 후 HER2 Tyr 잔기들의 인산화 정도 (HER2 pTyr counts)를 측정한 결과를보여주는그래프이다. FIG. 35h is a graph showing the results of measuring the degree of phosphorylation (HER2 pTyr counts) of HER2 Tyr residues after treating ATP and sugar 2+ on a substrate immobilized with a suppressor R2-HER2 homodimer.
도 35i는 HER2-HER2 homodimer 형성 세포 용해물의 세척에 사용된 detergent 종류에 따른 HER2의 활성화정도를 HER2 Y1196의 인산화 정도를 통하여 보여주는그래프이다.  FIG. 35I is a graph showing the degree of HER2 activation according to the type of detergent used for washing HER2-HER2 homodimer-forming cell lysates through the degree of phosphorylation of HER2 Y1196.
도 36a는 기판에 부착된 HER2-HER3 heterodimer가 인산화 과정을 거쳤을 때 eGFP 표지된 하위 신호전달 단백질과의 상호작용을 유도한다는 것을보여주는모식도이다.  36A is a schematic diagram showing that a HER2-HER3 heterodimer attached to a substrate induces an interaction with an eGFP-labeled lower signaling protein when subjected to a phosphorylation process.
도 36b는 하나의 HER2-HER3 heterodimer에 얼마나 많은 양의 하위 신호전달 단백질 (PLC gamma 1)과 상호작용하는지를 형광신호의 크기로 보여주는그래프이다.  FIG. 36B is a graph showing the magnitude of the fluorescence signal as to how much amount of the lower signaling protein (PLC gamma 1) interacts with one HER2-HER3 heterodimer.
도 36c는 하나의 HER2-HER3,heterodimer에 얼마나 많은 양의 하위 신호전달 단백질 (PLC gamma 1)과 상호작용하는지를 형광신호로부터 얻은 단일분자꺼짐 현상의 개수로보여주는그래프이다.  Figure 36c is a graph showing the number of single molecular off events from a fluorescence signal to see how much of the HER2-HER3, heterodimer, interacts with the lower signal transduction protein (PLC gamma 1).
도 36d는 하나의 HER2-HER3 heterodimer에 얼마나 많은 양의 하위 신호전달 단백질 (p85 alpha)과 상호작용하는지를 형광신호의 크기로 보여주는그래프이다.  Figure 36d is a graph showing the magnitude of the fluorescence signal as to how much of the HER2-HER3 heterodimer interacts with the amount of the lower signal transduction protein (p85 alpha).
도 36e는 하나의 HER2-HER3 heterodimer에 얼마나 많은 양의 하위 신호전달 단백질 (p85 alpha)과 상호작용하는지를 형광신호로부터 얻은 단일분자꺼짐 현상의 개수로보여주는그래프이다.  Figure 36E is a graph showing how much of the HER2-HER3 heterodimer interacts with the amount of the lower signaling protein (p85 alpha) as the number of single molecule off events from the fluorescence signal.
도 36f는 하나의 HER2-HER3 heterodimer에 얼마나 많은 양의 하위 신호전달 단백질 (PI3K : p85 alpha - pi 10 alpha complex)과 상호작용하는지를형광신호의 크기로보여주는그래프이다 . Figure 36f shows how much amount of the lower signaling protein (PI3K: p85 alpha-pi 10 alpha complex) is associated with one HER2-HER3 heterodimer And the intensity of the fluorescence signal.
도 36g는기판에 부착된 HER2 homodimer가인산화과정을거쳤을 때 eGFP 표지된 하위 신호전달 단백질과의 상호작용을 유도한다는 것을 보여주는모식도이다.  FIG. 36g is a schematic diagram showing that the HER2 homodimer attached to the substrate induces an interaction with the eGFP-labeled lower signal transduction protein upon oxidation.
도 36h는하나의 HER2 homodimer에 얼마나많은 양의 하위 신호전달 단백질 (PLC gamma 1)과 상호작용하는지를 형광신호의 크기로 보여주는 그래프이다.  Figure 36h is a graph showing the magnitude of the fluorescence signal as to how much of the HER2 homodimer interacts with the amount of the lower signaling protein (PLC gamma 1).
도 36i은하나의 HER2 homodimer에 얼마나많은양의 하위 신호전달 단백질 (PLC gamma 1)과상호작용하는지를 형광신호로부터 얻은 단일 분자 꺼짐 현상의 개수로보여주는그래프이다.  Figure 36i is a graph showing how much of the HER2 homodimer interacts with PLC signaling protein (PLC gamma 1) as the number of single molecular off events from the fluorescence signal.
도 36j은하나의 HER2 homodimer에 얼마나많은양의 하위 신호전달 단백질 (p85 alpha)과 상호작용하는지를 형광신호의 크기로 보여주는 그래프이다.  Figure 36j is a graph showing the magnitude of the fluorescence signal as to how much of the HER2 homodimer interacts with the amount of the lower signaling protein (p85 alpha).
도 36k은하나의 HER2 homodimer에 얼마나많은 양의 하위 신호전달 단백질 (p85 alpha)과 상호작용하는지를 형광신호로부터 얻은 단일 분자 꺼짐 현상의 개수로보여주는그래프이다.  Figure 36k is a graph showing how much of the HER2 homodimer interacts with the amount of the lower signaling protein (p85 alpha) as the number of single molecular off events from the fluorescence signal.
도 37a는 HER2-HER3 heterodimer에 ATP 와 Mg2 + 처리시의 HER3 Tyr 잔기의 인산화 속도를 보여주는 그래프이다 (x축은 인산화에 걸린 시간, y축은해당시간동안인산화된 Tyr의 양) .  Figure 37A is a graph showing the phosphorylation rate of the HER3 Tyr residues during ATP and Mg2 + treatment of the HER2-HER3 heterodimer (x-axis is the time taken to phosphorylate and y-axis is the amount of phosphorylated Tyr during that time).
도 37b는 HER2-HER3 heterodimer에 ATP 처리 농도를 증가시키면서 Figure 37b shows the results of increasing the ATP treatment concentration in the HER2-HER3 heterodimer
HER3 Tyr잔기의 인산화속도를측정하여 나타낸그래프이다. Lt; RTI ID = 0.0 > Tyr < / RTI >
도 37c는 HER2-HER2 homodimer를 사용하여 ATP 처리 농도를 증가시키면서 HER2 Tyr잔기의 인산화속도를측정하여 나타낸그래프이다. 도 37d는 HER2-HER3 heterodimer와 HER2 homodimer에 여러 농도의 Lapat inib을선처리 한뒤, ATP와 magnesium chlor ide를후처리하여 얻어진 FIG. 37C is a graph showing the rate of phosphorylation of HER2 Tyr residues measured while increasing the ATP treatment concentration using a HER2-HER2 homodimer. Figure 37d shows the results of pretreatment of HER2-HER3 heterodimer and HER2 homodimer at various concentrations of Lapat inib followed by treatment with ATP and magnesium chloride ide
HER2의 활성화정도를 Lapat inib농도에 따라사나타낸그래프이다.The activity of HER2 is shown by the concentration of Lapat inib.
도 37e는 HER2-HER3 heterodimer와 HER2 homodimer에 Lapat inib, ATP, 및 magnesium chlor ide를 동시에 처라하여 얻어진 HER2의 활성화 정도를 Lapat inib농도에 따라서 나타낸그래프이다.  FIG. 37E is a graph showing the degree of activation of HER2 obtained by treating LER2-HER3 heterodimer and HER2 homodimer with Lapat inib, ATP, and magnesium chlorid ide according to Lapat inib concentration.
도 37f는 HER2-HER3 heterodimer 에 Lapat inib, ATP, 및 magnesium chlor ide를 다양한 농도의 PTPN1 (Protein Tyrosine Phosphatase, Non receptor type 1)과 함께 처리하여 얻어진 HER3 Y1289 인산화 수준을 Lapat inib농도에 따라서 나타낸그래프이다. 도 38은 single-molecule co-IP및 single-molecule immunolabel ing의 인간종양표본에 대한적용결과를보여주는그래프이다. 도 39a~39g는 Grb2와 mutant EGFR 간의 SH3 도메인-매개 상호작용 (pTyr- independent manner)을 보여준다 ((a) Suppression of EGFR_Grb2 interaction by gef itinib (100 nM for 24 h) treatment during cell culture. Osimertinib (same dose as gefitinib) was treated to H1975 for further suppression of EGFR_Grb2 interaction due to the presence of gatekeeper mutation, T790M , in H1975 cell (b-g) Single-molecule immunolabel ing assay profiling the change in components of EGFR- associated proteins depending on the presence of phosphatase inhibitor during cell lysis or EGFR-TKI treatment during cell culture. After EGFR immunoprecipitat ion, HSP90 a (b) , MIG6 (c) , GAPDH (d) , Shcl (e) , EGFR (f ) or c-Cbl (g) antibody was used as the detection antibody, respectively. Inset in Fig. 40g shows the immunolabel ing count for Cbl-b. The inset x_ and y-axis are the same as for Fig. 40g. The amounts of cell extracts used are shown at bottom. Error bars in Fig. 40 show s.d. ) Figure 37f is a graph showing the HER3 Y1289 phosphorylation level obtained by treating Lapat inib, ATP, and magnesium chloride ide with various concentrations of PTPN1 (Protein Tyrosine Phosphatase, Nonreceptor type 1) in the HER2-HER3 heterodimer according to Lapat inib concentration . Figure 38 is a graph showing the results of application of single-molecule co-IP and single-molecule immunolabeling on human tumor samples. Figures 39a-39g show SH3 domain-mediated interaction (pTyr-independent manner) between Grb2 and mutant EGFR ((a) Suppression of EGFR_Grb2 interaction by gef itinib (100 nM for 24 h) treatment during cell culture. Osimertinib dose as gefitinib) was treated to H1975 for further suppression of EGFR_Grb2 interaction due to the presence of the gatekeeper mutation, T790M, in H1975 cell (bg) single-molecule immunolabeling assay (b), MIG6 (c), GAPDH (d), Shcl (e), EGFR (f) or c-Cbl The amounts of cell extracts used are shown in the following table, and the amount of cell extracts used are as follows: shown at bottom. Error bars in Fig. 40 show sd)
【발명의 실시를위한형태】  DETAILED DESCRIPTION OF THE INVENTION
본 발명을 하기 실시예를 들아 더욱 자세히 설명할 것이나, 본 발명의 권리범위가하기 실시예로한정되는의도는아니다.  The present invention will be described in more detail with reference to the following examples, but the scope of the present invention is not limited to the following examples.
실시예 1: 제 1단백질 (EGFR, MET, HER2,및 HER3)준비  Example 1: Preparation of first protein (EGFR, MET, HER2, and HER3)
제 1 단백질로서 EGFR, MET, HER2, 및 HER3를 선정하였으며, 이를 포함하는세포주 (예컨대, 암세포주)또는암세포조직을용해하여 얻어진 용해물 (lysate)로부터 상기 제 1 단백질을 얻었다. 본과정을 보다상세히 설명하면다음과같다:  EGFR, MET, HER2, and HER3 were selected as the first protein, and the first protein was obtained from a lysate obtained by dissolving a cell line (for example, cancer cell line) or cancer cell tissue containing the same. This process is described in more detail as follows:
1.1.세포용해물준비  1.1 Preparation of cell lysate
1.1.1.세포주준비  1.1.1. Cell line preparation
세포주를배지 (RPMI 1640, high glucose (Thermo 11965-092))에 2 x Cell lines were cultured in medium (RPMI 1640, high glucose (Thermo 11965-092)) at 2 x
106 cel Is의 양으로 분주하여 배양하였다. 100-pi culture dish에서 90% conf luency이상일 때 상기 세포주를수집하여 2개의 1.5 ml 튜브에 나누어 넣었다. 상기 튜브를 원심분리 (5 min x 15,000g) 하여 배양 배지를 제거한후세포만남기고 - 80°C에 얼려서 보관하였다. 10 6 cel Is. When the cell line was over 90% confluency in a 100-pi culture dish, the cell line was collected and divided into two 1.5 ml tubes. The tube centrifuged (5 min x 15,000g) to leave only the cell after removing the culture medium were kept eolryeoseo to 80 ° C.
준비된세포주를아래의 표 1에 정리하였다: [표 1] The prepared cell lines are listed in Table 1 below: [Table 1]
Figure imgf000078_0001
Figure imgf000078_0001
1.1.2.세포용해물준비 1.1.2 Preparation of cell lysate
50 mM Tris-HCl (pH 7.4), l%(v/v) Triton X-100, 150 mM NaCl, 1 mM EDTA, 10%(v/v) glycerol , protease inhibitor cocktail (Sigma, P8340) 100X, 및 tyrosine phosphatase inhibitor cocktail (Sigma, P5726) 100) (의 조성을갖는세포용해 버퍼를준비하였다.  100 mM Tris-HCl (pH 7.4), 1% (v / v) Triton X-100, 150 mM NaCl, 1 mM EDTA, 10% (v / v) glycerol, protease inhibitor cocktail tyrosine phosphatase inhibitor cocktail (Sigma, P5726) 100).
상기 실시예 1.1.1에서 준비된 세포주 시료를 피펫팅하여 뭉쳐있는 세포들을피펫으로풀어주었다. 상기 피펫팅한세포주시료에 상기 준비된 세포용해 버퍼를각튜브당 200ul의 양으로넣고ᄂ얼음위에 올려진 cold block (0-4°C)에 보관하면서 총 30분 반응시켰다. 이 때 10분 간격으로 주기적인 피펫팅을 통해 물리적으로 섞는 과정을 반복하여 계면활성제에 의한세포용해 반응이 활발하게 일어나도록하였다. The cell line sample prepared in Example 1.1.1 was pipetted and the cells were collected by pipetting. The prepared cell lysis buffer was added to the pipetted cell lysate in an amount of 200 ul per tube, and the cells were reacted for 30 minutes in a cold block (0-4 ° C) placed on ice. At this time, every 10 minutes The physically mixing process was repeated by cyclic pipetting so that the cell lysis reaction by the surfactant was actively performed.
상기한 바와 같이 30분 반응 후, 원심분리를 수행하였다 (10 min, 15,000g, 4°C ) . 그 후, 가라 앉은 부분 (pel let)은 버리고, 상청액 (supernatant )만을 취하여 0.2um 크기 기공을 갖는 멤브레인을 사용하여 여과시켜 상기 멤브레인을 통과한 부분을 새로운 튜브에 옮겨 담아, 다음시험에사용시까지 보관하였다.  After 30 min of reaction as described above, centrifugation was performed (10 min, 15,000 g, 4 ° C). Thereafter, the pellet is discarded, the supernatant is removed, the membrane is filtered using a membrane having a pore size of 0.2 mu, and the portion of the membrane passed through the membrane is transferred to a new tube and stored Respectively.
전체단백질 계량법 (Bradford, BCA, DC protein assay 등)을 이용하여 상기 반응 결과물내의 전체 단백질 농도를측정한결과, 단백질 농도가약 5-10 mg/ml 정도로측정되었다.  Total protein concentration in the reaction product was measured using a total protein measurement method (Bradford, BCA, DC protein assay, etc.), and the protein concentration was measured to be about 5-10 mg / ml.
1.2.조직용해액준비  1.2 Preparation of tissue solution
1.2.1. 환자유래종양이종이식모델준비  1.2.1. Patient-derived tumor xenograft model preparation
폐편평세포암 (Lung squamous cel l carcinoma; SQCC) 환자 유래의 tumor xenograft를 연세대학교 연구팀으로부터 입수하였다. Pat ient- der ived tumor xenograft (PDTXs) 제작과정을간략히 살피면 다음과같다: 6 내지 8주령의 암컷 중증 복합 면역 결핍증 마우스 (severe combined immunodef icient mice; NOG) 및 누드 마우스 (nu/nu mi ce; Or ientBio)를 준비하였다. 모든 동물 시험은 Inst i tut ional Animal Care and Use Commi ttee (IACUC)에 의하여 승인된 가이드라인에 따라서 수행하였다. 환자에서 _유래한 임상종양 샘플을 3mm이하의 크기의 절편으로 절단한후 상기 준비된 N0G 마우스의 옆구리에 피하 이식하였다. 종양 크기는 캘리퍼스로주 2회 측정하여 피하조직에서의 성장률을측정하였다. 종양 크기가직경 1.5 cm정도가되면, 종양조직을 적출하고작은 절편 (한면 길이가 약 5mm인 육각형)으로 절단하였다. 그 후, 절단된 조직을 다른 마우스에 재이식하여 연속적으로 subsequent tumor를 포함하는 개체들을 얻었다. 이렇게 얻어진 환자-유래 종양을갖는마우스를 라고명명하고, 이로부터 연속적으로 유래된 subsequent tumor를 갖는 마우스들을 일련번호를 붙여서 FI, F2, F3, F4 등으로 칭하였다. 3세대 subsequent tumor를 갖는 마우스 (F3)에 비히클 (PBS) 또는 게피티닙 Xgef i t inib)를 투여하여 시험에 사용하였다. 상기 준비된 PDTXs에 50 mg/kg의 게피티닙 또는 비히클을 하루 한 번 복강내 (intraper i toneal) 투여하였다. 게피티닙 투여로부터 15일 후 PDTX로부터 종양 조직을 수거하여 하기하는 PPI 및 발현수준변화모니터링에사용하였다. 1.2.2.조직용해액준비 Tumor xenografts from patients with Lung squamous cell carcinoma (SQCC) were obtained from Yonsei University research team. A brief review of the production process of Patient's tumor xenograft (PDTXs) is as follows: 6 to 8 weeks old female severe combined immunodeficient mice (NOG) and nude mice (nu / nu mi ce; Or ientBio). All animal studies were carried out in accordance with guidelines approved by the Insti- tional Animal Care and Use Committee (IACUC). A patient-derived clinical tumor sample was cut into sections of 3 mm or less in size and subcutaneously transplanted into the side of the prepared NOG mouse. Tumor size was measured twice weekly with calipers to determine the growth rate in subcutaneous tissue. When the size of the tumor reached about 1.5 cm in diameter, the tumor tissue was excised and cut into small sections (hexagon with one side length of about 5 mm). Thereafter, the excised tissue was re-implanted in another mouse to obtain consecutive individuals containing subsequent tumors. Mice with the patient-derived tumors thus obtained were named as mice, and mice having subsequent tumors continuously derived therefrom were referred to as FI, F2, F3, F4 and the like with serial numbers attached thereto. Mice (F3) with third generation subsequent tumor were injected with vehicle (PBS) or gefitinib Xgef it inib). The prepared PDTXs were intraperitoneally administered 50 mg / kg of gefitinib or vehicle once a day. Tumor tissues were collected from PDTX 15 days after the administration of gefitinib and used to monitor PPI and expression level changes described below. 1.2.2 Preparation of tissue solution
상기 실시예 1.2.1.에서 얻어진 종양조직을 20 mm3 정도 양으로 준비하였으나, 이보다큰경우라도무방하다. The tumor tissue obtained in Example 1.2.1 was prepared in an amount of about 20 mm 3 , but it may be larger.
상기 준비된종양조직 20 mm3당 앞서 실시예 1.1.2에서 준비된 용해 버퍼를 약 300 uL 정도 첨가하고, 4°C 냉장고에서 1시간 동안 계속 회전시키며 반응시켰다. 이 때, 수술용 가위를 이용하여 조직의 크기를 최대한 잘게 만들어, 단위 부피 당 표면적을 넓게 하여, 용해 버퍼 내의 계면활성제에 의한화학반응이 최대한효율적으로 일어날수 있도록하였다. 상기한 바와 같이 1시간 반응 후, 원심분리를 수행하였다 (10 min, 15,000g, 4°C ) . 그 후, 가라 앉은 부분 (pel let)은 버리고, 상청액 (supernatant )만을 취하여 0.2um 크기 기공을 갖는 멤브레인을 사용하여 여과시켜 상기 멤브레인을 통과한 부분을 새로운 튜브에 옮겨 담아, 다음시험에 사용시까지 보관하였다. Approximately 300 μL of the lysis buffer prepared in Example 1.1.2 was added to 20 mm 3 of the prepared tumor tissue, and the mixture was reacted while continuing to rotate in a 4 ° C refrigerator for 1 hour. At this time, the size of the tissue was made as small as possible using surgical scissors, and the surface area per unit volume was widened so that the chemical reaction by the surfactant in the dissolution buffer occurred as efficiently as possible. After 1 hour of reaction as described above, centrifugation was performed (10 min, 15,000 g, 4 ° C). Thereafter, the pellet is discarded, the supernatant is removed, the membrane is filtered using a membrane having a pore size of 0.2 mu, and the portion of the membrane passed through the membrane is transferred to a new tube and stored Respectively.
전체단백질 계량법 (Bradford, BCA, DC protein assay 등)을 이용하여 상기 반응 결과물내의 전체 단백질 농도를측정한 결과, 단백질 농도가약 5-10 mg/ml 정도로측정되었다.  Total protein concentration in the reaction product was measured using a total protein measurement method (Bradford, BCA, DC protein assay, etc.), and the protein concentration was measured to be about 5-10 mg / ml.
실시예 2: 제 2단백질준비  Example 2: Preparation of second protein
본 실시예에서는 상기 실시예 1에서 준비된 제 1 단백질의 하위신호전달 단백질에 형광 단백질이 부착된 형태의 제 2 단백질의 준비 과정이 예시된다. In this example, preparation of a second protein in which a fluorescent protein is attached to a lower signal transduction protein of the first protein prepared in Example 1 is exemplified.
본실시예에서 예시되는제 2단백질을아래의 표 2에 정리하였다: [표到  The second protein exemplified in this example is summarized in Table 2 below:
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000080_0001
Figure imgf000081_0001
단백질의 발현이 적은세포주인 HEK293세포 (ATCC)및 HeLa세포 (ATCC)를 준비하였다.  HEK293 cells (ATCC) and HeLa cells (ATCC), which are low cell expressing proteins, were prepared.
상기 표 2에 기재된 제 2단백질각각에 대한발현벡터를상기 준비된 HEK293 세포 또는 HeLa 세포에 주입하고 배양하여, 제 2 단백질을 발현시켰다. 24시간배양후세포를수집한후,적절한양으로분배하여 - 80°C에 보관하였다. An expression vector for each of the second proteins listed in Table 2 was injected into the prepared HEK293 cells or HeLa cells and cultured to express the second protein. After 24 h incubation, the cells were harvested, dispensed in the appropriate volume and stored at -80 ° C.
상기 세포에 용해액 ((50 mM Tris-HCl (pH 7.4), 1% Triton X-100, 150 mM NaCl , 1 mM EDTA, 10% glycerol , protease inhibitor cocktail (Sigma, P8340) 100X, tyrosine phosphatase inhibitor cocktail (Sigma, P5726) 100X))을 넣어 주었다. 높은 농도의 계면활성제 (Triton X-100)는 단백질상호작용을방해할수있으므로,우선 5x10s cel Is에 대하여 60 uL의 용해액을투입하였다. The cells were lysed in a solution (50 mM Tris-HCl (pH 7.4), 1% Triton X-100, 150 mM NaCl, 1 mM EDTA, 10% glycerol, protease inhibitor cocktail (Sigma, P8340) 100X, tyrosine phosphatase inhibitor cocktail (Sigma, P5726) 100X). High concentrations of surfactant (Triton X-100) was added 60 uL of the solution with respect to the first 5x10 s cel Is, since it can interfere with the protein interactions.
파이펫팅을 통해 뭉쳐있는 상기 얻어진 반응 혼합물 내의 세포를 모두풀어준후, 이를 얼음위에 올려진 cold block (0-4 °C)에 보관하면서 총 30분 반응시켰다. 이 때 10분 간격으로 주기적인 파이펫팅을 통해 물리적으로섞는과정을수행하여 계면활성제에 의한용해 반응이 활발하게 일어나도록하였다.  The cells in the obtained reaction mixture were released by pipetting and reacted for 30 minutes in a cold block (0-4 ° C) placed on ice. At this time, physical mixing was performed by periodic pipetting at intervals of 10 minutes so that dissolution reaction by the surfactant was actively performed.
30분 반응 후, 원심분리를 수행하였다 (10 min, 15,000g, 4°C). 가라 앉은 부분은 버리고 (pellet), 상층액만 (supernatant)실험을 위해 새로운튜브에 옮겨 담았다.  After 30 minutes of reaction, centrifugation was performed (10 min, 15,000 g, 4 ° C). The sinking was discarded (pellet) and transferred to a new tube for supernatant experiments.
상기 얻어진 수용액 60 를 취하여 140 uL의 PBS에 첨가해주었다. 이렇게 되면 최종적으로 0.3% Triton X-100이 포함된 제 2단백질의 용해액을 얻을 수 있다. Fluoremeter를 이용하여 제 2단백질에 부착된 형광단백질의 농도를 측정하였다. 그 결과 사용된 3종의 제 2 단백질의 농도가대략 400내지 1000nM범위 내로측정되었다.  The obtained aqueous solution 60 was added to 140 uL of PBS. Finally, a solution of the second protein containing 0.3% Triton X-100 can be obtained. Fluoremeter was used to measure the concentration of fluorescent protein attached to the second protein. As a result, the concentrations of the three second proteins used were measured within the range of about 400 to 1000 nM.
실시예 3: 기판의준비  Example 3: Preparation of substrate
Coverslip을 R0H 1M 용액에 침지시킨 후, sonicator에 담아서 세척하였다 (20-30 min). 그 후, 3차 증류수로 잘 씻은 후, piranha solution (황산:과산화수소 = 2:1 - 3:1 (v八 0)을 이용하여 세척하였다. 세척한 covers 1 ip 표면에 대하여 aminopropylsi lane와 PEG를 차례로 처리하여 코팅을수행하였다. Coverslip was immersed in R0H 1M solution and washed with a sonicator (20-30 min). After that, it was washed well with tertiary distilled water and then washed with piranha solution (sulfuric acid: hydrogen peroxide = 2: 1 - 3: 1 (v 八 0). The washed coverslip surface was treated with aminopropyl lane and PEG in order.
2시간 반응후, 3차증류수로 세척한뒤 PEG코팅된 면이 접촉되지 않게 하여 -20°C에서사용시까지 보관하였다. After 2 hours of reaction, the cells were washed with 3 rd distilled water and stored at -20 ° C until used without contact with the PEG-coated surface.
동시에 석영 재질의 channel 형태의 기판또는 아크릴 재질의 well 형태의 기판을 준비하였다. 석영 재질의 기판의 경우, 앞서 설명한 cover si ip처리 과정을참조로세척 및 PEG코팅 과정을수행하였다.  At the same time, a quartz channel type substrate or an acrylic type well type substrate was prepared. In the case of a quartz substrate, cleaning and PEG coating processes were performed with reference to the cover si ip process described above.
아크릴 재질의 기판의 경우, 제작 후 3차 증류수에 침지시킨 후 sonication하여 세척하였다. 세척된 아크릴 재질의 기판을 5% BSA용액에 침지시키고 2시간 반응시켜 비특이적 단백질 흡착 (nonspecific protein adsorption)을방지하고, -20C에서사용시까지 보관하였다.  In the case of an acrylic substrate, the substrate was immersed in the third distilled water and sonicated for washing. The washed acrylic substrate was immersed in a 5% BSA solution and reacted for 2 hours to prevent nonspecific protein adsorption and stored at -20 C until use.
하기 시험에서는 상기 준비된 covers lip과 아크릴 재질의 기판을 사용하였으며,시험 전에 상기 coverslip과아크릴 재질의 기판을해동하여 조립하거나, PEG 코팅이 끝난 뒤 미리 조립하고, 조립된 형태로 -20C에 보관하여사용전에 해동하여사용하였다.  In the following test, the prepared cover lip and acrylic substrate were used. Before the test, the coverslip and the acrylic substrate were thawed or assembled, or they were pre-assembled after the PEG coating and stored in -20C And thawed before use.
실시예 4: 제 1 단백질과 제 2 단백질 간 단백질-단백질 상호작용 이미징  Example 4: Protein-protein interaction imaging between a first protein and a second protein
상기 준비된 기판에 Avidin 계열의 단백질인 Neutravidin (Thermo , A2666)을 0.1 mg/ml 농도로 투입하였다. 상온에서 5분 반응 시킨 후 PBS buffer 30 ul를이용하여 2회 기판을세척하였다.  Neutravidin (Thermo, A2666), an Avidin family protein, was added to the prepared substrate at a concentration of 0.1 mg / ml. After 5 minutes of reaction at room temperature, the substrate was washed twice with 30 μl of PBS buffer.
상기 준비된 기판에 표적이 되는 제 1단백질에 대한 항체를 투입하였다. 이 때, 사용되는 항체는 biotin이 접합된 형태인 것으로 준비하였다. 항체의 농도는항체-항원의 affinity (dissociation constant , KD)에 따라 적절히 조절될 수 있으며, 본 실험예에서는 2 ug/ml 정도로 사용하였으며, 반응시간은 5분 정도로 하였다. 만약 Biotin이 접합되어 있지 않은 항체를 사용하는 경우에는 이차 항체를 이용하여 제 1단백질의 항체를부착할수있다.  An antibody against the first protein to be a target was added to the prepared substrate. At this time, the antibody to be used was prepared as a biotin-conjugated form. The concentration of the antibody can be appropriately adjusted according to the affinity (dissociation constant, KD) of the antibody-antigen. In this experiment, the antibody concentration was about 2 ug / ml and the reaction time was about 5 minutes. If a biotin-conjugated antibody is used, the antibody of the first protein can be attached using a secondary antibody.
이 때사용된제 1단백질에 대한항체를다음의 표 3에 정리하였다: Antibodies to the first protein used at this time are summarized in the following Table 3:
[표 3] [Table 3]
Figure imgf000082_0001
상기 항체가 처리된 기판을 PBS buf fer 30 ul를 이용하여 2회 세척하였다. 상기 준비된 기판에 앞서 실시예 1에서 준비한제 1단백잘을 포함하고 있는 세포 용해액 또는 조직용해액을 투입하였다. 항원-항체 반응의 경우 15분까지는 계속증가할수 있고 15분을초과하면서 시간대비 항원-항체 반응 효율이 낮아질 수 있어서, 반응시간을 15분 정도로 설정하였다.
Figure imgf000082_0001
The substrate treated with the antibody was washed twice with 30 ul of PBS buf fer. A cell lysis solution or a tissue lysis solution containing the first protein well prepared in Example 1 was added to the prepared substrate. In the case of antigen-antibody reaction, the antigen-antibody reaction efficiency could be lowered to 15 minutes and the reaction time could be lowered to 15 minutes or more.
반응 후, 기판을 PBS에 tween 20이 0.05%(v/v) 포함된 버퍼를 이용하여 세척하였다. Tween 20 0.05%는 nonspeci f ic binding을 줄여주는 동시에, 막단백질의 hydrophobi c region이 망가지지 않도록도와준다.  After the reaction, the substrate was washed with PBS containing 0.05% (v / v) of tween 20. Tween 20 0.05% reduces nonspecic binding and helps prevent the hydrophobic c region of membrane proteins from breaking down.
그 후, 상기 기판에 실시예 2에서 얻어진 제 2단백질 용해액을 투입하였다. 사용된 제 1단백질 용해액 내 제 2단백질의 농도는형광단백질 기준으로 1-50 nM사이의 값 (약 30nM)으로하였다. 제 2단백질농도가 100 nM 이상이 되면 형광현미경에서 background noi se가 커지게 되어 정확한 형광신호를측정하는데방해가된다.  Thereafter, the second protein solution obtained in Example 2 was added to the above-mentioned substrate. The concentration of the second protein in the first protein lysis solution used was between 1 and 50 nM (about 30 nM) based on the fluorescent protein. If the concentration of the second protein is more than 100 nM, the background noise increases in the fluorescence microscope, which hinders accurate fluorescence signal measurement.
형광현미경에 기판을고정시키고이미징하여, 각각의 제 1단백질/제 2 단백질에 대한데이터를얻었다.  The substrate was immobilized on a fluorescence microscope and imaged to obtain data for each first protein / second protein.
실시예 5: 단백질복합체 (PPI complex)분석  Example 5: Analysis of protein complex (PPI complex)
Mat 1 ab프로그램 (MathWorks 제공)에서 제공되는 toolki t에 기반하여 단백질복합체를분석하였다.  Protein complexes were analyzed based on the toolkits provided in the Mat 1 ab program (provided by MathWorks).
상기 실시예 4에서 얻어진 형광미지는 16bit unsigned integer 형식으로 저장하였다. 형광 신호는 eGFP (enhanced green f luorescent protein)로부터 얻었으며, 상기 신호를 관측하기 위하여 레이저의 파장을 488nm로 하였고, eGFP의 발광시간을 11초 정도 유지시키기 위하여 레이저 파워를 2mW로 조절하여 사용하였다. 전체 프레임 (30 프레임) 중, 초반 프레임을 버리고, 프레임의 이미지를 평균하여 하나의 이미지를 생성하였으며, 이와 같은 과정을 wel l 안에서 위치를 옮겨가며 반복 수행하여 총 5개의 이미지를 획득하여 아래 과정을 수행하였다. 초반 프레임을 버리는 이유는, 아무것도 없는 기판의 표면에서 발생하는 불필요한 신호 (autof luorescence)가 사라지고, eGFP 신호가 유지되는 구간을 선별하여 사용하기 위함이다. 이와 같이 선별되는 구간은 이미징 조건/장비구축 상태에 따라서 달라질 수 있다. 본 실시예에서는 EMCCD (Electron-mult iplying charge-coupled devi ce; Andor iXon Ul tra 897 EX2 (DU-897U-CS0-EXF)) 카메라를사용하여 1프레임당노출시간을 0.1초로하고, EMCCD gain값은 40으로하여 형광이미지를얻었다. The fluorescence image obtained in Example 4 was stored in a 16-bit unsigned integer format. The fluorescence signal was obtained from eGFP (enhanced green fluorescence protein). To observe the signal, the wavelength of the laser was set to 488 nm and the laser power was adjusted to 2 mW to maintain the emission time of eGFP for about 11 seconds. In the whole frame (30 frames), the first frame is discarded, and the image of the frame is averaged to generate one image. This process is repeatedly carried out while shifting the position in the wel l, Respectively. The reason for discarding the early frame is to selectively use the section where the autofluorescence on the surface of the substrate disappears and the eGFP signal is maintained. The selection interval may vary depending on the imaging condition / equipment construction status. In this embodiment, the exposure time per frame is set to 0.1 second by using an EMCCD (Electron-Multi loading charging-coupled device; Andor iXon Ul tra 897 EX2 (DU-897U-CS0-EXF) A fluorescence image was obtained with an EMCCD gain value of 40.
노이즈를 제거하기 위해, 각 프레임 마다 아래와 같은 절차를 수행하였다:  To eliminate noise, the following procedure was performed for each frame:
(a) 최초시작은왼쪽상단에서 시작한다. 1프레임은 512x512개의 픽셀로구성되어 있다. 기준픽셀을기준으로오른쪽으로 11개, 아래쪽으로 (a) The initial start begins at the top left corner. One frame consists of 512x512 pixels. 11 pixels to the right based on the base pixel, down
11개인 11x11개의 픽셀에서 medi an 값을 구하고, 이 medi an 값을 기준 픽셀의 수치에서 빼주었다 [(Intensi ty_pixel )_The median value is obtained from 11 11x11 individual pixels, and this median value is subtracted from the reference pixel value [(Intensi_pixel) _
(medi anIntensi ty_llxllneighborhood)] . 512x512의 모든 픽셀에 대해서 위 절차를 수행하였다. Medi an f i lter ing을 통해서 pepper & salt noi se를 제거하였다. (median anintensi ty_llxllneighborhood)]. The above procedure was performed for all pixels of 512x512. Median filtering was used to remove pepper & salt noi se.
(b) 위의 처리된 이미지에서 s i gma=0.7 , si ze=5x5의 Gaussian smoothing을수행하여, 이미지를부드럽게만들어 주었다.  (b) Gaussian smoothing of s i gma = 0.7 and si ze = 5x5 was performed on the processed image to smooth the image.
(c) Threshold를 설정하였다. Threshold는 전체 이미지에서 pixel intensi ty가 threshold 이하가 되는 pixel의 값을 threshold 값으로 모두 만들어 준다 (Mat l ab toolki t에서 local maximum을 찾는 알고리즘을 사용함) . 이를 통해 이미지에서 형광신호에 의해서 만들어지지 않은 국소 극대값 ( local maximum)을 제거할수 있다. 본실시예의 이미징 조건에서 사용되는 threshold값은 70이다.  (c) Threshold was set. Threshold is the threshold value that makes the pixel intensities below the threshold in the whole image (using the algorithm to find the local maximum in the Matlab toolkit). This eliminates the local maximum that was not produced by the fluorescence signal in the image. The threshold value used in the imaging conditions of this embodiment is 70.
형광단백질로부터 나오는 신호는 특정 위치에 모여있는 형태 ( local i zed point spread funct ion (PSF))로 발생하는데, 이 PSF 개수 (물리적인 값)를 측정하고자 하는 제 1 단백질과 제 2 단백질 간의 PPI complex숫자 (생물학적인 값)이다. 상기 PSF 값은 아래와 같은 과정을 거쳐서 생물학적인값인 PPI complex값으로전환시켰다:  The signal from the fluorescent protein occurs as a localized spreading func- tion (PSF). The PPI complex between the first and second proteins to measure the PSF count (physical value) It is a number (biological value). The PSF value was converted to the biological value PPI complex value through the following procedure:
(a) Local maximum의 위치를구하였다 (예를들어, i번째 row, j번째 column pixel ) . 앞서 기술한바와같이, 단분자형광신호는 512x512픽셀 중에서 특정 위치 (현재 관측장비 하에서 대략 5x5픽셀사이즈, 1 pixel = 0.167 마이크로미터)에 모여 형성되므로, local maximum을 찾으면 개별 PSF를선별할수 있다. 이는 Mat lab에서 제공되는 toolki t을이용하여 구할 수있다.  (a) Locate the local maximum (for example, i-th row, j-th column pixel). As described above, the single-molecule fluorescence signal is formed by collecting at 512x512 pixels at a specific position (approximately 5x5 pixel size under the current observation equipment, 1 pixel = 0.167 micrometer). This can be obtained using the toolkit provided in the mat lab.
(b) 상기 (a)에서 구한 local maximum이 실제 PSF로부터 발생한 것인지에 대한 판별과정을 거친다. 우선 local maximum의 최소값 (minimum intens i ty value)을 정의하고, 상기 얻어진 local maximum 중에서 이 최소값보다큰경우만분석에 사용하였다. 본실시예에서 사용된 최소값은 2019/132517 1»(:1/10公018/016675 (b) It is judged whether the local maximum obtained in the above (a) is generated from the actual PSF. First, the minimum value of the local maximum is defined, and only the case where the local maximum is greater than the minimum value is used for the analysis. The minimum value used in this embodiment is 2019/132517 1 »(: 1/10/06 018/016675
75이고, 이 값은 레이저 파워/노출시간/장비구축 상황에 따라서 달라질 수 있다. 최종적으로 얻어진 local maximum 좌표를 중심으로 5x5 픽셀의 정보를 불러온 후, 이 5x5 픽셀에서 밝기에 대한 중심을 구하였다 (centroid of intensi ty) . 이 때, 구해진 밝기 중심이 기존 local maximum 좌표에 대하여 0.5 pixel 이상 벗어나게 되면 (PSF모양에 대한 2D 대칭이 사라지면), 정상적이지 않은형광신호라고판단하고분석에서 제외시켰다. 75, and this value may vary depending on the laser power / exposure time / equipment construction situation. The 5x5 pixel information centered on the finally obtained local maximum coordinate was obtained, and the center of brightness was obtained from the 5x5 pixel (centroid of intensities). At this time, if the obtained center of brightness deviates by more than 0.5 pixels relative to the existing local maximum coordinates (when the 2D symmetry about the PSF shape disappears), it is judged to be a non-normal fluorescence signal and excluded from the analysis.
(c) 모든 조건을 통과한 PSF 만 최종적으로 선별하여 좌표와 전체 개수를 구하였다. 이 때 얻어진 PSF 전체 개수가 PPI comp lex의 개수가 된다 i  (c) Only the PSF that passed all the conditions were finally selected to obtain the coordinates and the total number. The total number of PSFs obtained at this time is the number of PPI comp lex. I
같은 조건하에서 촬영된 모든 파일에 대하여 상기 과정을 수행하여 The above procedure is performed for all files photographed under the same conditions
PSF개수를구한후, 이를 취합하여 평균과표준편차를 구하였다. 이 값이 최종적으로 특정 조건에서 PPI complex의 개수를 나타내는 값이 된다 (도면중에 "Number of single PPI complexes"로표시됨) . The number of PSFs was calculated, and the mean and standard deviation were calculated. This value finally represents the number of PPI complexes under certain conditions (indicated as "Number of single PPI complexes" in the figure).
실시예 6: PPI 세기, PPI 스코어 (PPI score) , 및 활성화 스코어 (Act ivat ion score)의 결정  Example 6 Determination of PPI Strength, PPI Score, and Activative Score
세포 용해물 (실시예 1.1.2 참조)의 농도를 x축으로 하고, 각 세포 용해물에서 측정된 PPI complex 값 (실시예 5 참조)을 축으로 하여 얻어진 그래프의 기울기, 즉, [(PPI complex)/ (세포용해액 단위 농도 ( Wml ) 당 PPI complex 개수)]를 상기 세포에서의 제 1 단백질과 제 2 단백질 간 PPI 세기 (또는 PPI slope)로 정의하였다. 각 세포주 별로 얻어진 모든 PPI 세기를합하여 PPI 세기 총합을구하였으며, 이 값을그세포주에 대한 PPI 스코어 (PPI score)로 정의하였다. 상기 PPI 세기 총합 (또는 PPI 스코어)은 각 세포주의 세포용해물 단위 농도에서의 시험된 제 1 단백질과 저 12단백질의 총 PPI 정도를나타낸다.  The concentration of the cell lysate (see Example 1.1.2) is plotted on the x-axis and the slope of the graph obtained by plotting the PPI complex value measured in each cell lysate (see Example 5), that is, [(PPI complex ) / (Number of PPI complexes per unit concentration of cell lysate (Wml))] was defined as the PPI intensity (or PPI slope) between the first protein and the second protein in the cells. All PPI intensities obtained for each cell line were combined to obtain the total PPI intensity, which was defined as the PPI score for the cell line. The PPI intensity sum (or PPI score) represents the total PPI level of the tested first and low 12 proteins at the cell lysate unit concentration of each cell line.
상기 PPI세기 총합을구하는방법을수식으로표현하면다음과같다:
Figure imgf000085_0001
The method of obtaining the PPI intensity sum is expressed as follows:
Figure imgf000085_0001
(제 1 단백질: RTK (폐암의 경우, EGFR, MET, HER2 , 또는 HER3; 유방암의 경우 HER2및 HER3) ;  (First protein: RTK (EGFR, MET, HER2, or HER3 for lung cancer; HER2 and HER3 for breast cancer);
제 2단백질: downstream protein (PLC-ga_a_SH2 , Grb2 , p85_alpha) ) . 또한, 세포주들 간 상대적인 PPI 스코어를 나타내기 위하여, 특정 세포주 (이하, '기준 세포라 칭함;본 시험예에서, 폐암세포주중에서는 PC9세포를, 유방암 세포주 중에서는 SKBR3세포를 각각 사용함)의 PPI 스코어가 1이 되도록 다른 세포(상기 기준 세포를 제외한 세포로서, 이하 '시험 세포’라 칭함)에서 얻어진 PPI 스코어들을 normalization하였으며, 이 때 각각의 세포주에 대하여 얻어진 값을 그 세포주에 대한 normalized PPI스코어로정의하였다. Second protein: downstream protein (PLC-ga_a_SH2, Grb2, p85_alpha)). In addition, in order to show the relative PPI scores among the cell lines, (Except for the reference cells), so that the PPI score of the cell line (hereinafter referred to as reference cell, in this test example, PC9 cells among lung cancer cell lines and SKBR3 cells among breast cancer cell lines, respectively) Hereinafter referred to as "test cells") were normalized, and the values obtained for each cell line were defined as normalized PPI scores for the cell lines.
또한,각세포용해물내의 제 1단백질 (예컨대, RTK(폐암의 경우, EGFR, MET, HER2, 또는 HER3; 유방암의 경우 HER2 및 四 R3)의 총량을 측정하였다. 상기 제 1단백질의 총량은 앞서 기재한 각각의 항체 (표 3 참조)를 사용하는 Sandwich ELISA 또는 quantitative western blot 등을 통하여 정량한 후, 세포 용해물의 중량 (세포 용해물 내의 총 단백질 중량)으로나눈값으로정하였다.  In addition, the total amount of the first protein (e.g., RTK (EGFR, MET, HER2, or HER3 for lung cancer; HER2 and R4 for breast cancer) in each cell lysate was measured. The total amount of the first protein was determined by sandwich ELISA or quantitative western blot using each of the antibodies described above (see Table 3), and then divided by the weight of the cell lysate (total protein weight in the cell lysate) Respectively.
상기 얻어진 PPI스코어 또는 normalized PPI스코어를제 1단백질의 총량으로 나누어 얻어진 값을 활성화 스코어 (activation score)로 정의하였다. 이 때, 세포주들 간 상대적인 활성화 스코어를 나타내기 위하여, 기준세포(폐암세포주: PC9세포,유방암세포주: SKBR3세포)의 활성화 스코어가 1이 되도록 시험 세포에서 얻어진 활성화 스코어들을 normalization하였으며, 이 때 각각의 세포주에 대하여 얻어진 값을 그 세포주에 대한 normalized활성화스코어로정의하였다.  A value obtained by dividing the obtained PPI score or normalized PPI score by the total amount of the first protein was defined as an activation score. At this time, activation scores obtained from the test cells were normalized so that the activation score of the reference cells (lung cancer cell line: PC9 cell, breast cancer cell line: SKBR3 cell) was 1, The value obtained for the cell line was defined as the normalized activation score for that cell line.
PDTX (patient -derived tumor xenogrft) 마우스 모델에서 PPI 스코어를 구하는 경우,상기 방법으로 얻어진 값에서 negative background 값을측정하여 이를공제하여 background noise를줄일 수있다. 이 경우, negative backgromid는 동일 환자의 정상조직 또는 시험 단백질 (예컨대, EGFR)이 정상인 암세포 용해물을 사용할 수 있다. 일 예로 EGFR유전자가 정상상태인 A549세포에서 EGFR과각하위 단백질 사이의 상호작용 정도를 측정하여 PPI 스코어를 구한다. 이를 negative background로 설정하여 각 PDTX마우스모델에서 얻어진 PPI 스코어에서 negative background를빼서 최종 PPI스코어를산출한다.  When a PPI score is obtained in a PDTX (patient-dependent tumor xenograft) mouse model, background noise can be reduced by measuring a negative background value from the value obtained by the above method. In this case, the negative backgromid can use normal tissue of the same patient or cancer cell lysate whose test protein (for example, EGFR) is normal. For example, in the A549 cell, in which the EGFR gene is in a normal state, the degree of interaction between the EGFR and the sub-protein is measured to obtain the PPI score. We set this as a negative background and subtract the negative background from the PPI score obtained from each PDTX mouse model to yield the final PPI score.
실시예 7 : Heatmap작성  Example 7: Heatmap creation
Data를 실시예 5에서 수치화한 것에 더하여, 이의 분석에 보충적 판단을추가하기 위하여 Heatmap을작성하였다. Heatmap는 data를보여주는 한방법일뿐이며, data해석을특별히 제한하기 위한것은아니다.  In addition to quantifying the data in Example 5, a Heatmap was created to add supplemental judgment to its analysis. Heatmap is only one way to show data and is not intended to limit data interpretation.
X축과 Y축 중, 한쪽 축은 제 2단백질 (하위신호 단백질)로 하고, 다른 축은 세포 종류로 하여 3x16(제 2 단백질 개수 (총 3개: 표 2 참조(p85_alpha, Grb2, PLC-ga_a_SH2)) x 세포주 개수(종 15개 (폐암 세포주) 또는 총 11개 (유방암 세포주): 표 1 참조))의 격자체를 만들었으며, 이러한 격자체는 제 1 단백질 별로 총 4개 (EGFR, MET, HER2, 및 HER3; 폐암), 또는 총 2개 (HER2 및 HER3; 유방암)를 만들었다 (단, HER3의 경우 제 2단백질로서 p85-alpha만사용함). 그후, 해당세포에서 얻어진 제 1 단백질과 제 2 단백질 간의 PPI 세기에 따라서 색과 밝기를 변화시키며 Heatmap을 작성하였다 (예컨대, PPI 세기가높아질수록 어둡고 검은색에서 중간밝기의 붉은색 , 밝은녹색 순서로표시될수 있는데, 이는 각시험마다시험자가정하는값으로정해진 값은아니다). 어떠한세포를 기준으로삼더라도세포주사이의 상대적인차이는변하지 않는다. Among the X axis and the Y axis, one axis is a second protein (lower signal protein) The other axis is the number of cell lines (15 kinds (lung cancer cell line) or 11 (breast cancer cell line): 3x16 (number of the second protein (total 3: see Table 2 (p85_alpha, Grb2, PLC-ga_a_SH2) (EGFR, MET, HER2, and HER3; lung cancer), or a total of two (HER2 and HER3; breast cancer) by the first protein (see Table 1) However, in the case of HER3, only p85-alpha is used as the second protein). Then, a heatmap was prepared by varying the color and brightness according to the PPI intensity between the first protein and the second protein obtained from the corresponding cells (for example, as the PPI intensity is increased, dark, black, , Which is not the value determined by the tester for each test). Regardless of which cell is the reference, the relative differences between the cell lines remain unchanged.
실시예 8: 약물 반응성과, PPI 스코어 및 활성화 스코어 간의 상관관계  Example 8 Correlation Between Drug Reactivity, PPI Score and Activation Score
상기 실시예 1내지 7에 기재된방법으로시험한결과를도면을들어 아래에 설명하였다:  The results of tests conducted by the methods described in Examples 1 to 7 are described below with reference to the drawings:
도 1은 단분자 단백질 상호작용 측정방법에 관한 모식도이다. 좌측은 PolyeUiylene glycol 이 코팅된 기판에 Neutravidin, RTK 항체, 및 분석하고자 하는 세포 용해액 또는 조직 용해액을 순서대로 주입한 후, 세척하는 방법을 모식적으로 보여주고, 중간은 이를 통해 표적 RTK 단백질을 기판에 고정하는 모습을 보여주며. 우측은 기판에 형광표지된 상호작용 단백질을 주입하여 형광신호를 관측 및 계량하여 단분자 단백질 상호작용정도를측정하는모습을보여준다.  1 is a schematic diagram of a method for measuring monomolecular protein interactions. On the left side, the Neutravidin, RTK antibody, and the cell lysis solution or tissue lysis solution to be analyzed are sequentially injected into the substrate coated with PolyeUlyene glycol, and then the sample is washed. In the middle, the target RTK protein It shows how it is fixed to the substrate. On the right side, the fluorescence signal is observed and quantified by injecting fluorescently labeled interacting protein to the substrate, and the degree of monomolecular protein interaction is measured.
도 2는 기판에 고정된 표적단백질 (제 1 단백질) 확인 결과를 보여주는그래프이다. 실시예 4및 5에 기재된 방법을참조하여 시험하였다. EGF은 100ng八 il의 양으로 3분간 처리하였으며, 도 2의 왼쪽 그래프는 FIG. 2 is a graph showing the result of identification of a target protein (first protein) immobilized on a substrate. With reference to the methods described in Examples 4 and 5. EGF was treated for 3 minutes in an amount of 100 ng 8 il, and the graph on the left side of Fig. 2
H1666을사용하고 EGFR의 extracel lul ar domain에 결합하는 ant ibody(MAS- 13266, Themof i sher)를 통해 기판에 부착하고, EGFR의 intracel lular domain (#4267, Cel l signal ing technology) 대한 ant ibody를 넣어 부착 확인한 결과이고, 도 2의 가운데 그래프는 EGFR이 HER2와 dimer를 형성하는지 여부를 HER2의 ant ibody를 넣어서 확인한 결과이고, 도 2의 오른쪽그래프는 EGFR이 Shcl과 dimer를 형성하는지 여부를 Shcl의 항체를 넣어서 확인한결과를보여준다. H antigens were used to attach to the substrate via an ant ibody (MAS-13266, Themof i sher) that binds to the extracelular domain of EGFR using H1666 and an ant ibody for the intracelular domain of EGFR (# 4267, Cel l signaling technology) 2 shows the result of confirming whether or not EGFR forms a dimer with HER2. The graph on the right side of FIG. 2 shows whether or not EGFR forms a dimer with Shcl. Shcl The results are shown by inserting the antibody.
기판의 항체 주입 여부에 따라 표적 RTK 단백질 (제 1 단백질)이 고정되거나 (+로 표시), 고정되지 않게 된다 (-로 표시). 이를 통해 적절한 항체 선별을 거쳐 다양한 표적단백질을 기판에 부착할 수 있다. 또한 EGFR-HER2나 EGFR-Shcl의 경우와 같이 단일 표적 단백질뿐만 아니라 생체 내 존재하던 단백질 결합체 형태로도기판에 고정할수 있음을확인할 수있다. Depending on whether the substrate is injected with the antibody, the target RTK protein (first protein) It is fixed (+), not fixed (-). Through this, various target proteins can be attached to the substrate through appropriate antibody screening. In addition, as in the case of EGFR-HER2 or EGFR-Shcl, it can be confirmed that not only a single target protein but also a protein conjugate existing in vivo can be immobilized on a substrate.
도 3은 제 1 단백질이 고정화된 기판에 형광표지된 상호작용 단백질 (제 2 단백질) 주입 후 단백질 상호작용을 나타낸 이미지이다. 실시예 5에서 설명한방법으로관즉된 PPI complex는 Point spread funct ion(PSF) 형태로나타나며, 컴퓨터 알고리즘을통해 PPI complex를선별하였다. 하위 신호전달단백질이 주입된상태에서만형광신호가발생하는것을볼수 있다. 초록색원형은관찰된 PPI complex를나타낸것이다.  FIG. 3 is an image showing protein interactions after the injection of fluorescently labeled interacting protein (second protein) onto a substrate to which the first protein is immobilized. The PPI complexes identified in the method described in Example 5 appear in the form of a point spread function (PSF), and the PPI complexes were selected through computer algorithms. It can be seen that the fluorescence signal occurs only when the lower signaling protein is injected. Green circles represent the observed PPI complexes.
도 4는도 3에서 관측된 PPI comp lex의 개수를정량화한그래프이다. 하위 신호전달 단백질(제 2 단백질)이 주입된 경우 (x축에서 PLCgammaSH2, Grb2, 및 p85-alpha로 표시)에만 선택적으로 높은 PPI complex가 관측된 것을 확인할수 있다. 반면 표적 RTK단백질 (제 1 단백질 (EGFR)의 항체가 없거나 (검은 막대), 주입된 하위 신호전달단백질이 없는 경우 (x축의 buf fer)에는 관측되는 신호가 매우 작은 것을 볼 수 있다. 두 경우에도 관즉되는것은 background noi se로해석할수있다.  FIG. 4 is a graph quantifying the number of PPI compexes observed in FIG. High PPI complexes were selectively observed only when the lower signaling protein (the second protein) was injected (designated PLCgammaSH2, Grb2, and p85-alpha in the x-axis). On the other hand, when the target RTK protein (the first protein (EGFR) antibody is absent (black bar) and the injected lower signaling protein is absent (buf fer of the x axis), the observed signal is very small. In both cases, it can be interpreted as background noise.
도 5는 주입된 세포용해액 양에 따른 PPI complex 개수 증가를· 보여주는 그래프이다. 표적 RTK 단백질 (제 1 단백질: EGFR)을 포함하는 세포 용해액의 양이 증가함에 따라 축), 관측되는 PPI complex 의 양 (y축)도 선형적으로 증가함을 볼 수 있다. 이를 통해 특정 세포 용해액의 양에서 시료간의 PPI comp lex의 정량적 비교가가능하다.  5 is a graph showing the increase in the number of PPI complexes according to the amount of injected cell lysate. It can be seen that the amount of observed PPI complex (y axis) increases linearly as the amount of cell lysate containing the target RTK protein (first protein: EGFR) increases. This allows a quantitative comparison of the PPI compex between samples in the amount of specific cell lysate.
도 6은 단분자 sandwich ELISA를 통하여 제 1 단백질을 정량하는 과정을 보여주는 모식도이다. 기판의 표면에 표적 RTK 단백질(제 1 단백질)을부착하는 과정은 도 1과동일하다. 형광표지된 하위 신호단백질 (제 2단백질) 대신, 표적 RTK단백질을인식하는제 2항체를주입하여 표적 RTK단백질의 양을 측정할 수 있다. 이 때 사용되는 제 2 항체 (detect ion ant ibody)는표적 RTK단백질을기판의 표면에 고정시키기 위해 사용된 제 1 항체 (pul l down ant ibody)와 표적 RTK 단백질 상에서 서로 다른 항체인식부위 (epi tope)를 가지고 있어야 한다. 제 2 항체를 인식하는 형광표지된 항체 (labeled ant i body)를 통해 기판의 표면에 고정된 RTK 단백질의 양을단분자기법(실시예 5참조)을통해측정할수있다. 도 7은 단분자 sandwich ELISA 방법을 통하여 얻어진 특이도 (speci f ici ty) 결과를 보여주는그래프이다. 도 6의 모식도에 나타난구성 요소중에서 하나의 구성요소만빠져도단분자 sandwich ELISA신호결과가 억제됨을볼수있다. 6 is a schematic diagram showing a process of quantifying a first protein through a monomolecular sandwich ELISA. The process of attaching the target RTK protein (first protein) to the surface of the substrate is the same as in Fig. Instead of the fluorescently labeled lower signal protein (second protein), a second antibody recognizing the target RTK protein can be injected to measure the amount of target RTK protein. The second antibody used here is a first antibody (pull down ant ibody) used to immobilize the target RTK protein on the surface of the substrate and a second antibody (epi toe) on the target RTK protein ). The amount of RTK protein immobilized on the surface of the substrate through the labeled ant i body recognizing the second antibody can be measured via a single molecule technique (see Example 5). FIG. 7 is a graph showing the specificity obtained by the single-molecule sandwich ELISA method. It can be seen that the single molecule sandwich ELISA signal result is suppressed even if only one component out of the components shown in the schematic diagram of FIG. 6 is omitted.
도 8은 세포주 종류 (빨간색① vs 하늘색③) 및 상태 (빨간색① vs 검은색②)에 따른 PPI complex개수의 변화를보여주는그래프이다. 세포에 존재하는 표적 RTK 단백질(제 1 단백질; EGFR)이 해당 리간드에 의해 활성화되었을 때 (EGF+), 그렇지 않은 경우와 비교하여, 동일 주입량에서 높은 PPI complex가 관측됨을 볼 수 있다. 또한 표적 RTK에 활성변이가 존재하면 (PC9, 하늘색), 관측되는 표적 RTK의 PPI complex 개수가 증가함을확인할수있다.  FIG. 8 is a graph showing changes in the number of PPI complexes according to the cell line type (red ① vs light blue ③) and the state (red ① vs black ②). The target RTK protein (first protein; EGFR) was activated by the corresponding ligand (EGF +), a higher PPI complex was observed at the same dose compared to the case without EGFR. In addition, the presence of active mutations in the target RTK (PC9, light blue) indicates that the number of PPI complexes in the observed target RTK increases.
도 9는 세포의 상태에 따른 다양한 표적 RTK (제 1 단백질) 별로 시료의 단위농도 당 PPI complex 개수 변화(PPI slope)를 보여주는 그래프이다. 실시예 6에 기재된 단분자 co-IP 기술을 이용하여 EGFR, MET, HER2, HER3 각각의 표적 단백질 (제 1 단백질)에 대하여 l igand st imulat ion 하였을 때 (회색)와 그렇지 않은 상태 (검은색)에서 PPI complex 개수가 달라지는 것을 정량적으로 측정하였다. 이를 바탕으로 PPI complex정량을통해표적 RTK의 활성도를측정할수있다.  FIG. 9 is a graph showing the PPI complex number change (PPI slope) per unit concentration of a sample for various target RTKs (first protein) according to cell conditions. Using the monomolecular co-IP technique described in Example 6, when the target protein (first protein) of each of EGFR, MET, HER2 and HER3 was subjected to 1-stigm imitation (gray) and not (black) The number of PPI complexes was measured quantitatively. Based on this, the activity of the target RTK can be measured through PPI complex quantification.
도 10은 EGFR변이 상태에 따른 PPI complex의 변화및 이를토대로 세포 당 활성화된 EGFR의 비율 계산한 결과를 보여주는 그래프이다. 상단은 PPI complex 측정법을 통해 개별 세포 별로 EGFR과 하위신호전달 단백질 사이의 상호작용을측정한 결과를 보여주는 것이고, 하단은 단분자 sandwi ch ELISA (도 6 참조)를통해 세포 당 EGFR발현량을 측정한후 두 값을 나누어 각 세포 당 활성화된 EGFR의 양을 계량한 결과(Absolute occupancy(%))를보여준다.  FIG. 10 is a graph showing the change in the PPI complex according to the EGFR mutation state and the calculated ratio of EGFR activated per cell based on the change. FIG. The upper part shows the result of measuring the interaction between the EGFR and the lower signal transduction protein for each individual cell by the PPI complex measurement method and the lower part shows the EGFR expression per cell through the single molecule sandwicz ELISA (see FIG. 6) After the two values were divided, the amount of activated EGFR per cell was measured (Absolute occupancy (%)).
도 11은 도 10에서 EGFR에 대하여 수행한 방법을 HER2, HER3에 대해서 동일하게 수행하여 얻어진 Absolute occupancy(%) 결과를보여주는 그래프이다. HER2의 경우 활성도가 매우 낮은 반면, HER3는 매우 높은 활성비율을나타낸다.  FIG. 11 is a graph showing absolute occupancy (%) results obtained by performing the same method for HER2 and HER3 performed on EGFR in FIG. In the case of HER2, the activity is very low, while HER3 shows a very high activity ratio.
도 12는폐암세포주에 대해 EGFR, MET, HER2, HER3(제 1단백질)와 하위신호전달 단백질(제 2 단백질) 사이의 상호작용(신호 세기)을 모두 측정하여 heatmap 형식(실시예 7)으로 표시한 결과를 보여준다. 각 신호 세기에 대한 color indicator는아래에 표시되어 있다. 도 13은도 12의 결과중에서 EGFR(제 1단백질)과 3종의 제 2단백질 간의 신호세기를각각정량한수치를더한값을나타낸그래프(좌측및 중간) 및 각세포주의 EGFR표적항암제의 일종인 AZD9291(0smert inib)에 대한 반응성(IC50; 세포 생존률이 처리 전과 비교하여 50%가 되는 처리 농도)결과를보여주는그래프(우측)이다. FIG. 12 is a graph showing the interactions (signal intensity) between EGFR, MET, HER2, HER3 (first protein) and the lower signal transduction protein (second protein) Show one result. The color indicators for each signal strength are shown below. FIG. 13 is a graph (left and middle) showing values obtained by adding numerical values obtained by quantifying the signal intensities between EGFR (first protein) and 3 kinds of second proteins among the results of FIG. 12, and AZD9291 0.0 >(IC50;< / RTI > And the cell survival rate is 50% as compared with that before treatment) (right side).
각 막대의 색은 EGFR 유전자 변이 상태에 따라 우측에 표시된 그룹으로 구별된다. PPI score와 비교하여, Act ivat ion score가 약물 반응성 (IC50)과 보다 유의미한 상관관계를 보임을 확인할 수 있다 (Act ivat ion score가높을수록 IC50값이 낮아짐(약물반응성이 높아짐)). 도 14는 EGFR 표적 항암제(AZD9291)의 반응성 (y축)과 Act ivat ion score (x축) 사이의 상관관계 (좌측) 및 유전자 타입에 따른 표적 항암제 반응의 다양성 (우측)을 보여주는 그래프이다. Act ivat ion score는 AZD9291의 반응성에 대하여 높은 상관관계(r=0.85)를 보이며, 기존 EGFR 유전자 검사에서는 동일한 유전자형을 가지고 있다고 하더라도 약물 반응성이 다르게나타날수있음을볼수있다.  The color of each bar is distinguished by the group shown on the right depending on the EGFR gene mutation status. The Act iat ion score showed a more significant correlation with the drug response (IC50) compared to the PPI score. (The higher the Act iat ion score, the lower the IC50 value (the higher the drug reactivity)). 14 is a graph showing the correlation between the reactivity (y axis) and the Act iat ion score (x axis) of the EGFR target anticancer agent (AZD9291) (left) and the diversity of the target anticancer response (right) depending on the gene type. The Act iat ion score showed a high correlation with the reactivity of AZD9291 (r = 0.85), and the EGFR gene test can show different drug reactivity even if it has the same genotype.
도 15는 유방암 세포주에서 HER2 및 HER3(제 1 단백질)와 하위신호전달단백질(제 2단백질) 간의 신호의 세기(상호작용)를 heatmap 형식(실시예 7)으로나타낸그림이다.  FIG. 15 is a diagram showing the intensity (interaction) of signals between HER2 and HER3 (first protein) and a lower signal transduction protein (second protein) in a breast cancer cell line in the heatmap format (Example 7).
도 16은 유방암 세포주에서 기존에 trastuzumab 항암제의 반응성을 예측하기 위해 사용되는 biomarker인 HER2 (상단), HER3 (중간) 발현량을 측정한 결과 및 trastuzumab에 의해 세포 성정이 억제되는 정도 (하단)를 측정하여 나타낸그래프이다.  FIG. 16 shows the results of measurement of biomarker HER2 (upper) and HER3 (intermediate) expression levels used for predicting the reactivity of trastuzumab anticancer drugs in breast cancer cell lines and the degree of suppression of cell growth by trastuzumab (lower) FIG.
도 17은 HER2또는 HER3신호를이용하여 PPI score를측정한결과와 trastuzumab 반응성 (logGI50) 사이의 상관관계를 보여주는 그래프이다. PPI score(r=0.91)는 기존의 biomarker인 HER2(r=0.54)나 pHER2(r=0.44) 발현량 (하단)과 비교하여 trastuzumab 반응성과 보다 높은 상관관계를 보임을확인할수있다.  17 is a graph showing the correlation between the result of measuring the PPI score using the HER2 or HER3 signal and the reactivity of trastuzumab (logGI50). The PPI score (r = 0.91) showed a higher correlation with trastuzumab reactivity than the conventional biomarker HER2 (r = 0.54) or pHER2 (r = 0.44)
도 18은 PDTX 마우스 모델에서 얻어진 조직 용해액 (실시예 1.2) (n=5; PDTX-1, - 2, -3, -4, 및 -5로표시)에서 측정한 EGFR, MET, HER2, 및 HER3의 3종의 하위 신호 단백질과의 PPI complex 신호 결과를 각각 보여주는 heatmap이다.  Fig. 18 shows the results of the tissue lysis (Example 1.2) (n = 5; And METH, HER2, and HER3 as measured by the PDTX-1, -2, -3, -4, and -5, respectively.
도 19는 PDTX마우스모델에서 얻어진조직 용해액 (실시예 1.2)에서 EGFR의 발현량 (상단) 및 상기 EGFR의 발현량 (도 18의 결과)를 이용하여 activation score를계산한결과 (하단)를보여주는그래프이다. 19 shows the expression levels (top) of EGFR and the expression levels of EGFR (results of FIG. 18) in the tissue lysate (Example 1.2) obtained in the PDTX mouse model This is a graph showing the result of calculating the activation score (bottom).
도 20은 PDTX 마우스 모델 (실시예 1.2.1)에 gefitinib(50mg/kg)을 투여하여 종양크기의 변화를 측정한 결과를 Vehicle (PBS) 투여군에서의 결과와비교하여 보여주는 그래프이다. PDTX-2의 경우, EGFR발현 수준은 높지 않지만 activation score는 높게 나타났으며 (도 19 참조), 항종양 효과역시 우수하게 나타남 (도 20참조)을확인할수 있다. 이러한결과는 EGFR 발현 수준보다는 활성화된 activation score (즉 활성화된 EGFR 비율)이 약물반응성과보다밀접한상관관계를나타냄을확인시켜 준다. 도 21은 PDTX 마우스 모델 (실시예 1.2.1)에서 gefitinib에 의한 종양크기 억제 (tumor growth inhibition) 정도와 EGFR activation score 사이의 상관관계를 보여주는 그래프이다. 앞서 설명한 바와 같이 gefitinib에 의한 종양크기 억제 (tumor growth inhibition) 정도와 EGFR activation score 사이에 유의미한 상관관계 (r=0.96)가 있음을 확인할 수 있다.  FIG. 20 is a graph showing the result of measuring the change in tumor size by administering gefitinib (50 mg / kg) to a PDTX mouse model (Example 1.2.1) in comparison with the results in Vehicle (PBS) administration group. In the case of PDTX-2, the expression level of EGFR was not high, but the activation score was high (see FIG. 19) and the antitumor effect was also excellent (see FIG. 20). These results confirm that the activated activation score (ie, the activated EGFR ratio) is more closely correlated with the drug response than the EGFR expression level. 21 is a graph showing the correlation between the degree of tumor growth inhibition by gefitinib and the EGFR activation score in the PDTX mouse model (Example 1.2.1). As described above, there is a significant correlation (r = 0.96) between the extent of tumor growth inhibition by gefitinib and the EGFR activation score.
도 22는 PDTX마우스모델 (실시예 1.2.1)에서 gef it inib(50mg/kg)을 투여하기 전, 후의 조직 용해액 시료에서 각각측정된 시료의 단위 농도당 EGFR PPI complex 개수 측정 결과를 보여주는 그래프이다. Gefitinib을 처리한후 얻은조직에서는 EGFR PPI complex가현저히 줄어든 것을볼수 있다. 이는 gefitinib에 의해서 EGFR신호가억제된증거가될수있다. 실시예 9  FIG. 22 is a graph showing the results of measuring the number of EGFR PPI complexes per unit concentration of the samples measured in the tissue lysate samples before and after the administration of gefit inib (50 mg / kg) in the PDTX mouse model (Example 1.2.1) to be. In the tissues obtained after treatment with Gefitinib, the EGFR PPI complexes were significantly reduced. This may be evidence that the EGFR signal is suppressed by gefitinib. Example 9
9.1.항체및시약준비  9.1. Antibody and reagent preparation
각각의 해당 단백질의 pulling down을 위하여, 다음의 항체를 사용하였다: 항- EGFR 항체 (MS-378-B0 ThermoFisher) , 항- MET 항체 (ab89297 Abeam) , 항- HER2 항체 (BMS120BT ThermoFisher), 항- HER3 항체 (BAM348 R&D systems) mCherry (ab34771 Abeam) , 및 항- KRas 항체 (sc-521 The following antibodies were used for the pulling down of respective proteins: anti-EGFR antibody (MS-378-B0 ThermoFisher), anti-MET antibody (ab89297 Abeam), anti- HER2 antibody (BMS120BT ThermoFisher) HER3 antibody (BAM348 R & D systems) mCherry (ab34771 Abeam), and anti-KRas antibody (sc-521
Santa Cruz) . Santa Cruz).
각각의 해당 단백질 및 PTMs(post-translat ional modi f i cat ions)에 대한 검출 항체로서 다음의 항체를 사용하였다: 항- EGFR항체 (4267 Cell signaling), 항- EGFR(pTyr 1068) 항체 (ab32430 Abeam) , 항- EGFR(pTyr 1086) 항체 (ab32086 Abeam) , 항- EGFR(pTyr 1173) 항체 (4407 Cell signaling), 항- MET 항체 (8494 Cell signaling), 항- HER2 항체 (MA5-15050 EGFR antibody (4267 Cell signaling), anti-EGFR (pTyr 1068) antibody (ab32430 Abeam) was used as the detection antibody for each of the corresponding proteins and PTMs (post-translational modi fi cat ions) , Anti-EGFR (pTyr 1086) antibody (ab32086 Abeam), anti-EGFR (pTyr 1173) antibody (4407 cell signaling), anti-MET antibody (8494 Cell signaling)
ThermoFisher), 항- HER2(pTyr 1221/1222) 항체 (2243 Cell signaling), 항_ HER3 항체 (ab32121 Abeam) , 항- HER3 (pTyr 1289) 항체 (Cell signaling technology, cat . No. 4791) , 항- Grb2항체 (ab32037 Abeam) , 항- Shcl항체 (ab33770 Abeam) , 항- She pTyr 239/240) 항체 (abl09455 Abeam) , 항- HSP90 항체 (PA3-013 ThermoF i sher ) , 항- MIG6 항체 (11630-1-AP Proteintech) , 항- GAPDH 항체 (3906 Cell signaling), 및 항- c-Cbl 항체 (2179 Cell signaling). HER2 (pTyr 1221/1222) antibody (2243 Cell signaling), anti-HER3 antibody (ab32121 Abeam), anti-HER3 (pTyr 1289) technology, cat. No. (Ab32037 Abeam), anti-Shcl antibody (ab33770 Abeam), anti-She pTyr 239/240) antibody (abl09455 Abeam), anti-HSP90 antibody (PA3-013 ThermoF i sher), anti- MIG6 antibody (11630-1-AP Proteintech), anti-GAPDH antibody (3906 cell signaling), and anti-c-Cbl antibody (2179 Cell signaling).
바이오틴 (biotin) 접합된 항-마우스 면역글로불린 G (IgG) (405303 BioLegend) 및 Cy3 접합된 항-래빗 IgG (111-165-046 Jackson The biotin conjugated anti-mouse immunoglobulin G (IgG) (405303 BioLegend) and Cy3 conjugated anti-rabbit IgG (111-165-046 Jackson
ImmunoResearch)항체들을 secondary antibody로사용하였다. ImmunoResearch antibodies were used as secondary antibodies.
웨스턴블라팅은다음의 항체들을사용하여 수행하였다: 항- EGFR(pTyr 1068) 항체 (2234 Cell signaling), 항- EGFR항체 (2232 Cell signaling), 항- Erk(pThr202/Tyr204) 항체 (9106 Cell signaling), 항- Erk 항체 (4696 Cell signaling), 항- Akt(pSer473) 항체 (4060 Cell signaling), 항- Akt 항체 (4691 Cell signaling), 항 -S6K(pSer235/236) 항체 (4858 Cell signaling), 항 -S6K 항체 (2217 Cell signaling), 및 항- act in 항체 (ab8227 Abeam)를사용하였다.  Western blotting was performed using the following antibodies: anti-EGFR (pTyr 1068) antibody (2234 Cell signaling), anti-EGFR antibody (2232 Cell signaling), anti-Erk (pThr202 / Tyr204) ), Anti-Erk antibody (4696 cell signaling), anti-Akt (pSer473) antibody (4060 cell signaling), anti-Akt antibody (4691 cell signaling), anti- S6K (pSer235 / 236) Anti-S6K antibody (2217 Cell signaling), and anti-actin antibody (ab8227 Abeam) were used.
lOOng/ml EGF (PHG0311L Life technologies)를 사용 (3분간)하여 EGFR를자극하였다.  EGFR was stimulated using lOOng / ml EGF (PHG0311L Life technologies) (3 min).
Gefitinib (S1025 Sel leckchem) , Osimert inib (S7297 Sel leckchem) , BKM120 (S2247 Sel leckchem) , Dabrafenib (S2807 Sel leckchem) , 및 Trastuzumab (A1046 BioVision)를 폐선암 (lung adenocarcinoma) 세포에서의 PPI 변화 및 유방암 세포에서의 HER2-/HER3-PPI 측정, MTT assay에 의한 세포생존률측정 , 및 PDTX모델에서의 종양성장측정에 사용하였다.  The changes in PPI and lung cancer cells in lung adenocarcinoma cells were compared with that of Gefitinib (S1025 Sel leckchem), Osimert inib (S7297 Sel leckchem), BKM120 (S2247 Sel leckchem), Dabrafenib (S2807 Sel leckchem), and Trastuzumab HER2- / HER3-PPI assay, MTT assay, and tumor growth assay in the PDTX model.
9.2. 세포배양  9.2. Cell culture
모든 세포주들은 10%(w/v) 우태아혈청 (26140-079 Life technologies) , 10//g/ml gentamicin (15710-063 Life technologies) , 100 units/ml 페니실린, 및 100 促/ml 스트렙토마이신 (15140-122 Life technologies)이 보충된 RPMI1640배지 (22400-105 Life technologies)에서 배양하였다. PC9-GR (gefitinib내성 세포주; Accession No. CVCL_S706) , HCC827-GR5 (gefitinib 내성 세포주; Accession No. CVCL_V622), 및 HCC4006-ER (erlotinib내성 세포주; Accession No. CVCL_S746) 세포주들은 각각 100 의 gefitinib또는 erlotinib의 존재하에서 배양하였다. 모든 세포주들은 37 °C 및 5% C02조건의 가습배양기에서 배양하였다. 배양된 세포를 찬 phosphate buffered saline (PBS)로 행구었다. 찬 PBS 1ml와 scraper (90020 SPL Life Science)를사용하여 세포를신속하게 수집하였다. 하나의 petri dish (직경 100 mm)로부터 얻어진 세포 현탁액을 3-4 aliquots로 분량하였다. 이 분액을 4 °C에서 3,000xg로 5분간 원심분리하였다. 상등액을 버리고 얻어진 펠렛을 -80°C에서 보관하고, 이후분석에사용하였다. All cell lines were incubated with 10% (w / v) fetal bovine serum (26140-079 Life technologies), 10 // g / ml gentamicin (15710-063 Life technologies), 100 units / ml penicillin, 15140-122 Life technologies) were supplemented in RPMI1640 medium (22400-105 Life technologies). Cell lines of PC9-GR (gefitinib resistant cell line: Accession No. CVCL_S706), HCC827-GR5 (gefitinib resistant cell line; Accession No. CVCL_V622), and HCC4006-ER (erlotinib resistant cell line; Accession No. CVCL_S746) . ≪ / RTI > All cell lines were cultured in a humidified incubator at 37 ° C and 5% CO 2 . Cultured cells were washed with cold phosphate buffered saline (PBS). With 1 ml of cold PBS Cells were rapidly harvested using a scraper (90020 SPL Life Science). The cell suspension from one petri dish (100 mm diameter) was divided into 3-4 aliquots. This fraction was centrifuged for 5 minutes at 3,000xg at 4 ° C. The supernatant was discarded and the resulting pellet was stored at -80 ° C and was then used for analysis.
9.3. eGFP표지된 prey protein의 제작및형질감염 (transfections) 9.3. Preparation and transfections of eGFP-labeled prey proteins
Bglll and EcoRI를사용하여 tandem SH2도메인 (542 to 765 amino acids of NM_013187.1)을 포함하는 Rat PLC Y SH2 cDNA를 Rat cDNA library로부터 직접 분리하였다. Grb2 (인간 Grb2; Addgene 46442) , p85 a (마우스 p85 a Addgene 1399) , Shcl (인간 Shcl, Addgene 73255) , Eat2 (인간 Eat 2, Addgene 46423), APCS (인간 APCS, Addgene 46477) , Nckl (인간 Nckl, Addgene 45903) 및 S0S1 (인간 S0S1, Addgene 32920)의 cDNA들을 각각의 원래의 플라스미드에 포함된 제한부위에 대응하는 제한효소들을 사용하여 절제하였다. eGFP-tagged CARM1 (인간 CARM1) 및 EGFR유전자는각각서울대학교 (한국)와 KAIST (한국)으로부터 제공받았다. 모든 cDNA를 pEGFP-Cl (Clontech Laboratories)에 클로닝하여 상응하는 eGFP- 1 abe 1 ed prey protein들을제작하였다. Grb2유전자에 W36K, R86M및 W193K 점돌연변이를 각각 도입하여 Grb2 변이체인 N*-, SH2*-, 및 C*- construct를 각각 제작하였다. EGFR 유전자에서 E746-A750을 결실 시키거나 858번째 잔기인 라이신을 아르기닌으로 치환하여 EGFR 변이체를 제작하였다. Using the EcoRI and Bglll were isolated directly Rat PLC Y S H2 cDNA containing the tandem SH2 domain (542 to 765 amino acids of NM_013187.1 ) from Rat cDNA library. Shcl (human Shcl, Addgene 73255), Eat2 (human Eat 2, Addgene 46423), APCS (human APCS, Addgene 46477), Nckl (human), Grb2 (human Grb2; Addgene 46442), p85a (mouse p85a Addgene 1399) Nckl, Addgene 45903) and S0S1 (human S0S1, Addgene 32920) were excised using restriction enzymes corresponding to the restriction sites contained in each original plasmid. The eGFP-tagged CARM1 (human CARM1) and EGFR genes were obtained from Seoul National University (Korea) and KAIST (Korea), respectively. All cDNAs were cloned into pEGFP-Cl (Clontech Laboratories) to produce corresponding eGFP-1 abe 1 ed prey proteins. Mutations of W36K, R86M and W193K were introduced into the Grb2 gene, respectively, to produce N * -, SH2 * -, and C * - constructs of Grb2 variants. EGFR mutants were prepared by either deleting E746-A750 from the EGFR gene or replacing the 858th residue, lysine, with arginine.
상기 얻어진 플라스미드를 Neon transfection system (MPK5000 Life technologies)을 사용하여 제조사의 지시에 따라 electroporation을 통해 HEK293세포에 도입하였다. 플라스미드 DNA 30 ! 를 2xl06세포를함유한 HEK293 세포현탁액 100 M와 혼합하였다. 2번의 950V 전기펄스 (with a duration of 35 ms for each pulse)를 HEK293 세포에 적용하였다. 형질감염 후 24시간 경과 후에 형질감염된 세포를 수확한 후 -80 °C에서 보관하였다. The obtained plasmid was introduced into HEK293 cells by electroporation using a Neon transfection system (MPK5000 Life technologies) according to the manufacturer's instructions. Plasmid DNA 30! Were mixed with 100 M of HEK293 cell suspension containing 2x10 6 cells. Two 950V electric pulses (with a duration of 35 ms for each pulse) were applied to HEK293 cells. Transfected cells were harvested 24 hours after transfection and stored at -80 ° C.
9.4. 폐암환자유래의 종양이종이식 모델  9.4. Tumor xenograft model from lung cancer patient
모든 동물 연구는 Institutional Animal Care and Use Committee All animal studies were conducted by the Institutional Animal Care and Use Committee
(IA⑶ C)으로부터 승인받은 지침에 따라 수행하였다. 6 내지 8주령의 암컷 마우스 (severe combined immunodef icient (NOG) and nude (nu/nu) mice; OrientBio)를사용하였다. 임상종양샘플 (폐선암종 (lung adenocarcinoma) 환자유래 또는 폐편평상피암 (lung squamous cell carcinoma; SQCC) 환자 유래)을 ~3 mm3 크기의 조각으로 절단한 후, 상기 N0G 마우스의 옆구리 내부로 피하 이식 (subcutaneous implantation) 하였다. 이식 후 1~4개월 경과후, 이식된 부위에서 종양이 관찰되었다. caliper로 주 2회 종양의 체적을 측정하여 종양의 피하 성장률을 측정하였다. 종양 크기가 직경(IA, C). (NOG) and nude (nu / nu) mice (OrientBio) at 6 to 8 weeks of age were used. Clinical tumor samples (lung adenocarcinoma) Patient-derived or lung squamous cell carcinoma (SQCC) patients) were cut into ~ 3 mm 3 pieces and subcutaneously implanted into the lateral side of the N0G mice. After 1 to 4 months after transplantation, a tumor was observed at the transplanted site. The volume of the tumor was measured twice weekly with caliper to determine the subcutaneous growth rate of the tumor. Tumor size is diameter
1.5cm에 도달하였을 때, 종양 조직을 절제하고 작은 절편 (대략 5 mm3 크기)으로 절단하였다. 상기 절단된 조직을 다른 마우스 집단에 재이식하여 속발성 종양 (subsequent tumors)을 얻었다. 환자유래 종양을 갖는마우스세대를 이라명명하고, 이후의 후속세대는차례대로번호를 매겼다 (FI, F2, F3 등) (도 23a 참조). 3세대 (F3) 마우스를 vehicleUpon reaching 1.5 cm, the tumor tissue was excised and cut into small sections (approximately 5 mm 3 size). The severed tissue was re-implanted in another group of mice to obtain subsequent tumors. Mouse generations with patient-derived tumors were named and subsequent generations were numbered sequentially (FI, F2, F3, etc.) (see FIG. 23A). A third generation (F3)
(Phosphata buffered saline, PBS) , osimert inib, 또는 gefitinib 처리 시험에사용하였다. (Phosphata buffered saline, PBS), osmate inib, or gefitinib.
상기 얻어진 환자 유래 종양 이종이식 마우스 (patient -derived tumor xenograft; PDTX) 중에서, 폐선암종환자유래 종양이 이식된마우스 (F3; n=3)을卵 TX-A1, 卵 TX-A2, 및 卵 TX-A3으로 명명하고, lung SQCC환자 유래 종양이 이식된 마우스 F3; n=5)을 PDTX-S1, 卵 TX-S2, 卵 TX-S3, PDTX- S4, 및 PDTX-S5로명명하였다.  In the obtained patient-derived tumor xenograft (PDTX), a mouse (F3; n = 3) transplanted with a malignant tumor-derived tumor was transplanted into an egg TX-A1, an egg TX- A3, and mice with FT (n = 5) implanted with lung SQCC patient-derived tumors were named as PDTX-S1, egg TX-S2, egg TX-S3, PDTX-S4 and PDTX-S5.
상기 얻어진 환자 유래 종양 이종이식 마우스 (pat ient -derived tumor Mnograft; PDTX)에 체중 (kg) 당 5mg의 osimert inib 또는 50mg의 gefitinib또는 vehicle를 각각 1일 1회 복강내 주사로주입하였다. 상기 약물 처치 후 15일 경과 후에 PDTX로부터 종양 조직을 절제하여 PPI 및 발현수준변화를모니터링하였다.  5 mg of osimert inib or 50 mg of gefitinib or vehicle per kg body weight was injected into the obtained patient-derived tumor xenograft (PDTX) by intraperitoneal injection once a day. Tumor tissues were excised from PDTX 15 days after the drug treatment to monitor changes in PPI and expression levels.
9.5. 단분자 co-IP (Single-molecule co-IP)및 면역 표지 이미지화 (immuno labeling imaging)  9.5. Single molecule co-IP (single-molecule co-IP) and immuno-labeling imaging
단분자 co- IP및 immunolabel ing이미지화에 대한자세한프로토콜은 Detailed protocols for monomolecular co-IP and immunolabel imaging
"Lee, H. W. et al . Real-time single-molecule coimmunoprecipitat ion of weak protein-protein interactions . Nat . Pro toe. 8, 2045-2060 ,Lee, H. W. et al., Real-time single-molecule coimmunoprecipitate ion of weak protein-protein interactions., Nat. Pro. Toe. 8, 2045-2060,
(2013)"를 참조하였다. NeutrAvidin (10 jd of 0.1 mg/ml : A2666 Life technologies)을각각의 개별 반응챔버에 넣었다. 10분간인큐베이션후, 미결합 NeutrAvidin을 제거하였다. Miniaturized imaging chamber를 PBS가 채워진 reservoir에 담갔다가 100번 정도 측면방향으로 흔들어서 완전히 씻어주었다. PBS를 완전히 제거한 후, biotinylated pul 1 down antibodies를 NeutrAvidin 코팅된 표면에서 10 분간 배양하여 증을 만들었다. MET 항체의 경우, biot inylated secondary ant ibody (alpha- mouse IgG)를사용하여 1차항체를 결합시켰다. PBS로 첨버를세척한후, 암세포또는종양조직 추출물을항체가코팅된표면에 적용하였다. 15분 후, 미결합 추출물을 제거하고, 챔버를 0.05%(v/v) Tween 20이 보충된 PBS로채워진 reservoi r에 담갔다. NeutrAvidin (10 jd of 0.1 mg / ml: A2666 Life technologies) was placed in each individual reaction chamber. After 10 minutes of incubation, unbound NeutrAvidin was removed. A miniaturized imaging chamber was filled with PBS After PBS was completely removed, biotinylated pul 1 down antibodies were incubated on a NeutrAvidin coated surface for 10 min, made. For MET antibodies, the primary antibody was conjugated using a biot inylated secondary antibody (alpha- mouse IgG). After washing with PBS, cancer cells or tumor tissue extracts were applied to the antibody coated surface. After 15 minutes, the unbound extract was removed and the chamber was soaked in a reservoir filled with PBS supplemented with 0.05% (v / v) Tween 20.
single-molecule co-IP imaging을 위하여, 형질전환된 HEK293 세포 추출물을 30 nM (eGFP-tagged probe protein)으로 희석시킨 후, imaging chamber에 로딩시켰다. 첨버를 TIRF 현미경 상에 위치시키고 eGFP 형광을 EMC抑으로기록하였다 (20프레임 ; 100-ms노출) .  For single-molecule co-IP imaging, transformed HEK293 cell extracts were diluted with 30 nM (eGFP-tagged probe protein) and loaded into the imaging chamber. Implants were placed on a TIRF microscope and eGFP fluorescence was recorded with EMC suppression (20 frames; 100-ms exposure).
single-molecule immunolabel ing imaging의 경우, 5 프레임 동안 eGFP- labeled probe protein을 대신하여 dye- labeled 검출 항체를 사용하였다. 검출 및 풀다운 항체 간의 중복을 피하기 위하여, 검출 항체는 세포질 키나아제 부위 또는 말단 (tai l ) 상의 티로신 잔기 내에 에피토프를 갖는 것으로선정하였다. 검출항체는 Alexa488 (MET항체)로 직접 표지하거나, Cy3 -표지된 2 차 항체 (EGFR, HER2, HER3 및 pTyr 항체)로 간접적으로 가시화하였다. TIFF stack에서의 5또는 20프레임의 형광 (0.1초노출)을 기록한후, 형광스팟의 개수를카운팅하여, 단분자 PPI complex 또는 면역표지된 (immunolabeled) 단백질의 수를 즉정하였다. 단분자수 (single-molecule counts)의 평균 및 표준 편차는 동일한 반응 챔버 내에서 10개의 상이한위치로부터 구하였다.  For single-molecule immunolabel imaging, dye-labeled detection antibody was used instead of eGFP-labeled probe protein for 5 frames. To avoid duplication between detection and pulldown antibodies, the detection antibody was chosen to have an epitope within the tyrosine residue on the cytoplasmic kinase site or end (tai l). Detection antibodies were directly labeled with Alexa488 (MET antibody) or indirectly with Cy3-labeled secondary antibodies (EGFR, HER2, HER3 and pTyr antibodies). After recording 5 or 20 frames of fluorescence (0.1 second exposure) on a TIFF stack, the number of fluorescent spots was counted and the number of unimolecular PPI complexes or immunolabeled proteins was immediate. The mean and standard deviation of single-molecule counts were determined from ten different positions in the same reaction chamber.
실시예 9.6. Count ing PPI complex및면역표지된단백질의 counting 형광이미징하여 얻어진 TIFF파일을 custom GUI (wr i tten in Mat 1 ab (Mat lab 2016a, MathWorks))로 분석하였다. 세 개의 프레임 (eGFP의 경우 17-19, Cy3 및 Alexa488의 경우 3_5)을 사용하여 단일 PPI 복합체 또는 면역표지된 단백질을 대표하는 세기 (intensi ty)의 local maxima를 확인하였다. background보정을위하여, spat i al medi an-f i l ter ing (11x11 pixel)으로 얻은 이미지를 원본 이미지에서 프레임별로 공제 (subtracted)하였다. 얻어진 이미지는 평균화하여 thresholding 투 local maxima를검출하는데사용하였다 (custom Mat 1 ab GUI사용) .  Example 9.6. The TIFF file obtained by counting fluorescence imaging of the counting PPI complex and immuno-labeled proteins was analyzed with a custom GUI (Matlab 2016a, MathWorks). Three frames (17-19 for eGFP, 3_5 for Cy3 and Alexa488) were used to confirm the local maxima of intensities representing a single PPI complex or immunoreactive protein. For the background correction, the images obtained by spatial aliasing (11 × 11 pixels) were subtracted frame by frame from the original image. The obtained images were averaged and used to detect thresholding to local maxima (using custom Mat 1 ab GUI).
실시예 10: EGFR표적 억제제에 대한 PDTX의 반응성 예측  Example 10: Prediction of responsiveness of PDTX to EGFR target inhibitor
10.1. 폐선암 이종이식된 PDTX에서의 Osimert inib에 대한 반응성 예즉  10.1. Reactivity to Osimert inib in PDTX transplanted with lung carcinoma
HER 계열 수용체의 PPI 측정 지표가 암의 약물 반응성과 밀접하게 연관되어 있음을 확인하였으며, HER 계열 수용체 표적 치료에 반응성을 갖는 (HER 계열 수용체 표적 치료가 항암 효과를 보이는) 특정 암을 스크리닝에 single-molecule immuno label ing또는 co-IP analysi s를 적용할 수 있는지 조사하였다. 이를위하여, 상기 실시예 9에서 기술한방법으로 3 종의 폐선암종 환자유래 종양 이종이식 마우스 (PDTXs; 도 23의 PDTX-The PPI measurement index of HER receptor is closely related to the drug reactivity of cancer And whether single-molecule immuno-labeling or co-IP analyses can be applied to screening for specific cancers that are reactive to HER receptor target therapy (HER receptor-targeted therapies have anticancer effects) Respectively. For this, three types of lung cancer xenograft-derived tumor xenografted mice (PDTXs; PDTX-
A1 A3)를제작하였다. A1 A3).
이들 폐선암종 抑 TX는 EGFR 유전자에서 활성화 돌연변이 (exon 19 또는 L858R돌연변이)를갖는것으로확인되었다.  These abundant carcinosarcoma TX were found to have an activating mutation (exon 19 or L858R mutation) in the EGFR gene.
상기 폐선암종 PDTXs (PDTX-A1-A3; 각각의 개체수는 3이상이며 하기 결과는그평균값으로나타냄)에 대하여 30일동안 osimert inib(5 mg per 1 kg of weight dai ly)을처리하고종양크기를측정하여, 대조군 (vehi cle 투여군)과비교하여, 그결과를도 23b좌측에 나타내었다. 도 23b좌측 결과로부터 알 수 있는 바와 같이, PDTX-A1 A3는 osimert inib 처리에 의하여 현저한종양크기의 감소를보였다 (A1>A2>A3) .  Treatment of ovarian carcinoma PDTXs (PDTX-A1-A3; each population of 3 or more, the results of which are shown as the mean value) for 30 days with osmate inib (5 mg per kg of weight dairy) And compared with the control group (vehicle-treated group), the results are shown on the left side of FIG. 23B. As can be seen from the left-hand side of FIG. 23B, PDTX-A1 A3 showed a significant decrease in tumor size by osimert inib treatment (A1> A2> A3).
또한 각 卵 TX(PDTX-A1~A3)에서 EGFR, HER2, HER3 및 MET 수용체 각각과 하위신호단백질인 PLCgammaSH2, Grb2, 및 p85-alpha 각각 간의 PPI complex (도 23c에서 PPI count로 표시됨)를 측정하여, 도 23c 좌측에 나타내었다. 도 23c에 나타난 바와 같이, EGFR과 3종의 하위신호단백질 간의 PPI complex count는 A1>A2>A3순서로 나타났으며, 이는 도 23b에 나타난 EGFR 저해제인 osimert inib 처리시의 종양 크기 감소효과 양상과동일함을 확인할수 있다. 이러한결과는 폐선암종의 PDTX 모델에서 표적단백질과 하위신호단백질 간의 PPI complex count가 상기 표적단백질을 표적으로 하는 표적치료제의 항암 효과와 유의미한 상관관계를나타냄을보이는것이다.  The PPI complex (represented by PPI count in Figure 23c) between each of the EGFR, HER2, HER3 and MET receptors and the lower signal proteins PLCgammaSH2, Grb2 and p85-alpha was measured in each egg TX (PDTX-A1 to A3) , And the left side of Fig. 23C. As shown in FIG. 23C, the PPI complex counts between EGFR and the three sub-signal proteins were in the order of A1> A2> A3. This is due to the effect of the EGFR inhibitor osimert inib on tumor size reduction The same can be confirmed. These results show that the PPI complex count between the target protein and the lower signal protein in the PDTX model of lung cancer carcinoma shows a significant correlation with the anticancer effect of the target therapeutic agent targeting the target protein.
또한, 8마리의 PDTX (A1-A3 및 S1-S5) 개체에서의 EGFR과 다른 수용체 (MET, HER2및 HER3)의 발현수준을측정하고, 상기 각각의 수용체의 발현수준을 대조군 (EGFR: A549 세포, MET: HCC827-GR5, HER2 및 HER3: SKBR3)에서의 발현 수준으로 normal izat ion하여 그 결과를도 23d 및 도 25a_c에 나타내었다.  Expression levels of EGFR and other receptors (MET, HER2 and HER3) in 8 PDTX (A1-A3 and S1-S5) individuals were also measured and the expression levels of the respective receptors were measured in a control (EGFR: A549 cells , MET: HCC827-GR5, HER2 and HER3: SKBR3), and the results are shown in FIGS. 23d and 25a_c.
도 23c에 나타난 바와 같이, 시험된 卵 TX모델 (A1~A3)는 모두 MET, As shown in FIG. 23C, all of the tested egg TX models (A1 to A3) were MET,
HER2 및 HER3 수용체에 대해 유의미한 PPI complex 수치를 나타내지 않았지만, EGFR에 대해서는 어느 정도 유의미한 PPI complex 수치를 나타내었다. 이러한 결과는 PDTXs가 단백질과 PPI 수준에서 EGFR signaling에 종양 유전자 중독 (oncogene addict ion)을 나타냄을 의미하는 것이라할수있다. Did not show significant PPI complex levels for HER2 and HER3 receptors, but showed a somewhat significant PPI complex level for EGFR. These results suggest that PDTXs inhibit EGFR This means that the oncogene addiction is expressed on the signaling.
다음으로, normalized PPI count (제 1 단백질의 단위 농도당 PPI complex 수; activation score 해당)에 의하여, 폐선암종 세포주에서 입증된 바와 같이, osimertinib 치료에 대한 卵 TX의 반응성을 예측할 수 있는지를 조사하였다. 상기에서 PDTX 모델 (A1 A3)에 30일동안 osimert inib(5 mg per 1 kg of weight daily)을 처리하여 얻어진 각 모델에서의 종양성장억제율 (tumor growth inhibition(%)=[( h^vehicle- A Igefi t inib) / A V vehicle] x 100 : vehicle.· vehicle 처리 전후의 종양 부피 변화; Mgefitinib· gef it inib 처리 전후의 종양 부피 변화)을 y죽으로하고, PPI sum/EGFR level (PPI sum: PPI score이고, PPI sum/EGFR level: Activation score)를 x축으로하여 도 23e에 나타내었다. 도 23e에 나타난바와같이, normalized PPI count (PPI sum/EGFR level; Activation score)가 실제로 osimert inib에 의한 종양 성장 억제와 높은 상관 관계 (r=l)를나타냄을확인할수있다.  Next, we investigated whether the reactivity of egg TX to osimertinib treatment can be predicted, as evidenced by the lung cancer cell line, by normalized PPI count (PPI complex number per unit concentration of the first protein; corresponding to the activation score). The tumor growth inhibition (%) in each model obtained by treating the PDTX model (A1 A3) for 30 days with osmate inib (5 mg per kg of weight daily) EGFR level (PPI sum: PPI) was calculated by subtracting the tumor volume before and after the vehicle treatment and the tumor volume before and after treatment with Mgefitinib and gefit inib. score, and PPI sum / EGFR level: Activation score) is shown on the x-axis in FIG. 23E. As shown in FIG. 23E, it can be confirmed that the normalized PPI count (PPI sum / EGFR level: Activation score) actually shows a high correlation (r = 1) with tumor growth inhibition by osimert inib.
10.2. lung SQCCClung squamous cell carcinoma)이종이식 PDTX에서의 Osimert inib에 대한반응성 예측  10.2. lung SQCCClung squamous cell carcinoma) Reactivity prediction for Osimert inib in xenotransplantation PDTX
폐선암의 29%가 EGFR의 감작 돌연변이와 관련되어 있는 반면, lung SQCC의 0.5%만이 EGFR의 감작 돌연변이를 가지고 있다. 따라서, 현재로서는 lung SQCC에서 EGFR 표적 치료를 위한 적절한 바이오 마커가 없는실정이다.  While 29% of lung cancer is associated with sensitization mutations of EGFR, only 0.5% of lung SQCC have EGFR sensitization mutations. Therefore, at present, there is no appropriate biomarker for EGFR target treatment in lung SQCC.
본실시예에서는 lung SQCC환자조직으로부터 5마리의 抑 TX(PDTX_ S1 S5)를 제작하고 단분자 면역 표지 및 co-IP 프로파일링 (실시예 9.5)을 수행하였다. 5마리의 PDTX(PDTX-S1~S5)가모두 MET, HER2 및 HER3수용체 단백질 및 이들과관련된 PPI complex가최소수준을 나타냄을 확인하였다 (도 23c, 우측및도 25a-c).  In this example, five suppressed TX (PDTX_S1 S5) were constructed from lung SQCC patient tissues and monolayer immuno-labeling and co-IP profiling (Example 9.5) were performed. The five PDTX (PDTX-S1 to S5) mammary gland MET, HER2 and HER3 receptor proteins and their associated PPI complexes were found to exhibit minimal levels (Figure 23c, right and Figures 25a-c).
한편, 총 EGFR count (EGFR level)와 EGFR PPI complex count가 상당한수준으로검출됨을확인하였다 (도 23c및 23d, 및도 25d및 25e). 이러한 결과는 lung SQCC가 증식 신호에 있어서 종종 EGFR에 의존하기 때문인것으로예상된다.  On the other hand, it was confirmed that the total EGFR count (EGFR level) and the EGFR PPI complex count were detected at a considerable level (FIGS. 23C and 23D, and FIGS. 25D and 25E). These results are expected to be due to the fact that lung SQCC is often dependent on EGFR in the proliferative signal.
10.3. lung SQCC( lung squamous cell cm"cinoma)이종이식 PDTX에서의 gefitinib에 대한반응성 예측  10.3. lung SQCC (lung squamous cell cm "cinoma) reactivity prediction for gefitinib in xenotransplantation PDTX
5마리의 抑 TX(PDTX-S1~S5)를 모두 15일동안 gefitinib를 투여하고, 종양 성장 정도를 측정하여 도 23b 우측에 나타냈다. PDTX-S1-S5중에서 S1과 S2에서 현저한종양억제 효과를보이는것으로확인되었다. 시험된 다양한 PDTX 개체들 중에서, EGFR 수준으로 normalized된 PPI complex count (Activation score)가 종양 성장 억제와 매우 높은 상관관계 (Spearman correlation of 0.9)를 가짐을 다시 한번 확인하였다 (도 23 f 및도 25f-h) . Five suppressive TXs (PDTX-S1-S5) were treated with gefitinib for 15 days, The degree of tumor growth was measured and shown on the right side of FIG. 23B. PDTX-S1-S5 showed significant tumor suppression effects in S1 and S2. Of the various PDTX individuals tested, it was once again confirmed that the PPI complex count (activation score) normalized to EGFR level had a very high correlation (Spearman correlation of 0.9) with tumor growth inhibition (Figure 23 f and Figure 25f- h).
상기 얻어진 결과들은 normalized PPI count가 비소세포 폐암 (non- smal 1 cell lung cancer)의 EGFR표적 억제제에 대한반응성과밀접한상관 관계가 있음을보여준다. PPI 수치의 합계가아닌 normalized PPI 수치가 항종양약물에 대한반응성과보다높은관련성을갖는지 이해하기 위하여, 고유한 signaling phenotype과 gefitinib반응성을보인 2마리의 lung SQCC PDTX (PDTX-S1및 S2)를집중적으로관찰하였다.  The results obtained show that the normalized PPI count is closely related to the response of non-small cell lung cancer to EGFR target inhibitors. Two lung SQCC PDTX (PDTX-S1 and S2) with intrinsic signaling phenotype and gefitinib reactivity were studied intensively to understand whether normalized PPI values, rather than the sum of PPI values, were more relevant to response to anti-tumor drugs Respectively.
gefitinib 처리 전 (Before)과 처리후 15일 경과 후 (After), PDTX- S1과 PDTX-S2에서의 PPI complex count를 측정하여 도 23g 및 도 26a_b에 나타내었다.  The PPI complex counts in PDTX-S1 and PDTX-S2 were measured before and 15 days after gefitinib treatment and are shown in Figures 23g and 26a_b.
상기 결과에서 보여지는 바와 같이, PDTX-S1은 특히 의 조절 p85 a 서브유닛에서 검출가능한 수준의 EGFR PPI complex counts를 유지하였다. 반면, PDTX-S2는 A549세포를사용한음성 대조군과비교하여 감소하거가구별되지 않는정도의 EGFR PPI complex counts를나타냈다 (도 23g 및 도 26). 따라서, 시험에 사용된 gefitinib 투여량 (50 mg/kg)은 As shown in the above results, PDTX-S1 maintained a detectable level of EGFR PPI complex counts, especially in the regulatory p85 a subunit. PDTX-S2, on the other hand, exhibited reduced or indistinguishable levels of EGFR PPI complex counts compared to negative control with A549 cells (Figure 23g and Figure 26). Thus, the gefitinib dose (50 mg / kg) used in the study
PDTX-S2에서 EGFRs의 hyperactive but smaller pool을 완전히 억제하여 해당 암의 위축 (shrinkage)을 유도한다고 할 수 있다. 반면, 동일한 gefitinib투여량은 PDTX-S1에서 EGFR활성을억제하지 못하고상당한 EGFR 과발현을 나타내었다. 이러한 결과는 높은 normalized PPI cmint를 갖는 抑 TX-S2가,높은 EGFR수준과 total PPI count를갖는抑 TX-S1과비교하여 , gefitinib에 대하여 보다우수한반응성을보이는 이유를제안한다고할수 있다. In PDTX-S2, the hyperactive but smaller pool of EGFRs is completely inhibited, leading to the shrinkage of the cancer. On the other hand, the same gefitinib dose did not inhibit EGFR activity in PDTX-S1 and showed significant EGFR overexpression. These results suggest that TX-S2 with high normalized PPI cmint exhibits better reactivity to gefitinib than suppress TX-S1 with high EGFR level and total PPI count.
상기 결과에서 PDTX-S1이 증가된 EGFR-p85a 결합을보인다는사실에 기초하여, PDTX-S1을 PI3K inhibitor인 BKM120(50 mg/kg)으로 처리하였다 (도 23h).  Based on the above results that PDTX-S1 exhibits increased EGFR-p85a binding, PDTX-S1 was treated with PI3K inhibitor BKM120 (50 mg / kg) (Figure 23h).
상기 결과에서 보여지는 바와 같이, BKM120는 동일한투여용량 (50 mg/kg)에서 gefitinib보다 강력한 종양 성장 억제 효과를 보였다. 또한, gefitinib과 BKM120의 병용 치료 (gefitinib (50mpk(mg per lkg weight (mpk))/BKM120(50mpk))는종양을위축시켰다. 이러한결과는상이한 다운스트림 단백질을 사용하여 PPI를 검사하는 single-molecule co-IP prof i l ing이 표적 신호화경로의 선택 및 복수의 약물의 병용 치료 전략을 설계하는데유용하게사용될수있음을제안한다. As shown in the above results, BKM120 showed strong tumor growth inhibitory effect at the same dose (50 mg / kg) than gefitinib. In addition, combination therapy of gefitinib and BKM120 (gefitinib (50 mpk (mg per lkg weight (mpk)) / BKM120 (50 mpk) atrophied the tumor. These results suggest that single-molecule co-IP profiling of PPI using different downstream proteins can be useful for selecting target signaling pathways and designing a combined treatment strategy for multiple drugs.
10.4. 비소세포성 폐암이종이식 PDTX에서의 반응성 예측  10.4. Prediction of reactivity in non-small cell lung cancer xenotransplantation PDTX
두 종류의 비소세포성 폐암 이식 모델인 PDTX-A1-A3 및 PDTX- S1 S5로부터 얻은 normal ized PPI complex counts를 풀링하고 단일 플롯을 비교하였다 (도 23i ) .  Normalized PPI complex counts from PDTX-A1-A3 and PDTX-S1 S5, two non-small cell lung cancer transplantation models, were pooled and single plots compared (FIG. 23I).
유전체 변이 양상 및 암 서브타입에서의 모든 차이에도 불구하고, data points는 일정한 양상을 보이며, Spearman상관 관계가 0.95인 종양 성장억제와잘일치하는모양을형성하였다.  Despite all differences in the genetic and cancer subtypes, the data points displayed a consistent pattern and formed a shape that closely matched the tumor growth inhibition with a Spearman correlation of 0.95.
이러한 결과는 normal ized PPI complex count가 EGFR-표적 요법에 대한효능예측마커로서 작용할수 있는 EGFR신호강도의 척도 (gauge)임을 제안한다.  These results suggest that the normal ized PPI complex count is a measure of the EGFR signal intensity that can serve as an efficacy predictor for EGFR-targeted therapies.
실시예 11. 인간 환자 시료에 대한 단분자 면역 표지 (Single- molecule immunolabel in)및 co-IP프로파일링 (co-IP prof i l ing)  Example 11. Single-molecule immunolabeling and co-IP profiling for human patient samples (co-IP profiling)
본 발명에서 제시되는 마이크로 챔버와 고성능 단분자 영상 시스템 (high-throughput single-molecule imaging system)을사용하여 인간 환자의 종양조직을특성화하였다 (도 24) . Tumor tissues of human patients were characterized using the micro chamber and high-throughput single-molecule imaging system of the present invention (Fig. 24).
Figure imgf000099_0001
Figure imgf000099_0001
프로토콜 (cryogenic lysi s protocol )을폐선암종환자로부터 외과적 절제로 얻은 두 개의 폐선암종 환자조직 (연세대 세브란스병원)에 적용하였다 (도 24a, P1및 P2라고함) . The protocol (cryogenic lysi s protocol) was applied to two lung cancer patient tissues (Yonsei University Severance Hospital) obtained by surgical resection from a lung cancer patient (Fig. 24A, P1 and P2).
간략히 설명하면, 상기 준비된 환자조직을분쇄 (~0.6cm)하고, 액체 질소에 침지시킨다. 추가 분쇄 후 PBS를 투여하여 완전히 용해시키고, 원심분리하여, 펠렛을 취하였다. 이후 PBS를 투여하고, 계속적으로 섞어주면서 4°C에서 배양하였다. 그후, 원심분리하여 상청액을취하였다. 크기가 15mm3 (P1) 및 18mm3 (P2)인 조직을 사용하여, 상기 기재한 방법으로 얻어진 각 조직 용해물에 대하여 10개의 서로 다른 PPI level (posi t ive control에서의 PPI complex를 1.0으로 하였을 때의 P1 및Briefly, the prepared patient tissue is pulverized (~0.6 cm) and immersed in liquid nitrogen. After further pulverization, PBS was added to dissolve completely, centrifuged, and pellets were taken. PBS was then added and incubated at 4 ° C with continued mixing. Then, the supernatant was taken by centrifugation. Using tissues of 15 mm 3 (P1) and 18 mm 3 (P2) in size, we obtained 10 different PPI levels for each tissue lysate obtained by the method described above and 1.0 for the PPI complex When P1 and
P2에서의 PPI complex의 상대값) 및 1◦개의 서로다른단백질 및 PTM(post- translat ional modi f icat ions) 수준 (발현량)을 즉정 (실시예 9.5의 immunolabel ing참조)하였다 (도 24b) . 양성 대조군으로서 PC9세포 ( for EGFR), HCC827세포 (for MET), 및 SKBR3세포 (for HER2및 HER3)가각각 사용하였다. (Relative value of the PPI complex at P2) and the level of 1 < 0 > different proteins and PTM (expression levels) (see immunolabeling of Example 9.5) (Fig. As a positive control, PC9 cells (for EGFR), HCC827 cells (for MET), and SKBR3 cells (for HER2 and HER3).
두종양조직에및 P2)모두 EGFR유전자에 exonl9돌연변이 (exonl9 결실 변이)를 갖고 있었지만, 샘플 만이 유의미한 EGFR PPI complex counts를나타냈다 (도 24b, c및 표 4) .  Both tumor tissues and P2) had an exonl9 mutation (exonl9 deletion mutation) in the EGFR gene, but only the sample showed significant EGFR PPI complex counts (Figure 24b, c and Table 4).
[표 4]  [Table 4]
Biopsy type Deposite date EGFR genotype Treatment Response  Biopsy type Deposite date EGFR genotype Treatment Response
Figure imgf000100_0001
Figure imgf000100_0001
(PR=Partial response, PD=Pr ogress ive disease)  (PR = partial response, PD = progressive disease)
다른 수용체인 HER 수용체와 MET 수용체에 대해서는 두 샘플 모두에서 의미있는 PPI counts가관찰되지 않았다 (도 24b, c).  For the other receptors HER and MET receptors, meaningful PPI counts were not observed in both samples (Fig. 24b, c).
Patient P1은진행성 질환 (progressive disease; PD) 진단 전에 약 1년 반동안 gefitinib치료에 대한부분적 반응성 (partial response; PR; 고형 종양에서의 반응성 평가 기준 (response evaluation criteria in solid tumors; RECIST)에 따른 부분적 반응성)을 유지한 반면, Patient P2는 PD designation 전에 1년 동안 stable disease (SD) diagnosis를 유지하였다.  Patient P1 had a partial response to gefitinib treatment for approximately one and a half years prior to the diagnosis of progressive disease (PD), a partial response (PR) to the response evaluation criteria in solid tumors (RECIST) While Patient P2 maintained a stable disease (SD) diagnosis for one year before PD designation.
마지막으로, 가장높은활성의 EGFR신호를보이는 PDTX-A1및 인간 환자 샘플 P1으로부터 유래된 mutant EGFR (exon 19 결실)에 대하여 PTPN1를처리하여 in vitro탈인산화를수행하였다 (도 24d) .  Finally, in vitro dephosphorylation was performed by treating PTPNl against PDTX-A1 showing the highest active EGFR signal and mutant EGFR (exon 19 deletion) derived from human patient sample Pl (Fig. 24d).
탈인산화 후, eGFP- labeled PLCgamma 및 Grb2와, mutant EGFR 복합체와의 결합을 측정하여 도 24d에 나타내었다. 도 24d에 나타난 바와 같이, 탈인산화 (+PTPN1)에 의하여, pTyr-SH2 도메인 상호작용에 전적으로 의존하는 PLCga_asH2의 결합은거의 완전하게 중지되는반면, Grb2 binding ccxints의 경우에는 50% 이상 및 80% 이상이 탈인산화 후에도 유지되었다. 이러한 결과는 mutant EGFR의 pTyr- independent signaling median ism이 PDTX모델과외과적 종양조직에서 실제로작동함을제안한다. After dephosphorylation, the binding of eGFP-labeled PLCgamma and Grb2 to the mutant EGFR complex was measured and is shown in Figure 24d. As shown in Figure 24d, the binding of PLCga_as H 2, which is entirely dependent on the pTyr-SH2 domain interaction, is almost completely stopped by dephosphorylation (+ PTPN1), while in the case of Grb2 binding ccxints, % Were retained after dephosphorylation. These results suggest that the pTyr-independent signaling median ism of the mutant EGFR actually works in extracorporeal tumor tissue of the PDTX model.
실시예 12:면역단백질 PD-1및 PD-L1의준비  Example 12 Preparation of Immune Proteins PD-1 and PD-L1
抑- 1은 GFP로표지하여 GFP labeled卵 -1형태로, PD-L1은 mcherry로 표지하여 mcherry labeled PD-L1형태로각각준비하였다. Sup-1 is labeled with GFP and labeled with GFP labeled egg-1, while PD-L1 is labeled with mcherry Labeled and prepared in the form of mcherry labeled PD-L1.
卵-1-GFP는 human 卵-1 (NP_005009.2)고유의 신호 서열 (Signal sequence)을 포함한, 1번부터 288번까지의 단백질 서열 및 상기 아미노산 서열의 카복시 말단 (C-teminal)에 연결된 GFP (아미노산 서열: MVSKGEELFTGWPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTLTYGVQ Egg-1-GFP has a protein sequence from 1 to 288 including the unique signal sequence of human egg-1 (NP_005009.2) and GFP linked to the carboxy terminal (C-teminal) of the amino acid sequence (Amino acid sequence: MVSKGEELFTGWPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTLTYGVQ
CFSRYPDHMKQHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGH KLEYNYNSHNVYIMADKQKNGIKANFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNHYLSTQSALSK DPNEKRDHMVLLEFVTMGITLGMDELYK)로 구성되며, 상기 두 단백질은 19개 아미노산으로 구성된 펩타이드 링커에 의하여 연결되도록 준비하였다 (N terminal쪽: PD-1, C terminal쪽: GFP) . CFSRYPDHMKQHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGH KLEYNYNSHNVYIMADKQKNGIKANFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNHYLSTQSALSK consists DPNEKRDHMVLLEFVTMGITLGMDELYK), the two proteins were prepared to be connected by a peptide linker consisting of 19 amino acid (N terminal side: PD-1, C terminal side: GFP).
상기 PD-1-GFP 사이에 사용된 링커의 아미노산 서열 및 이를 암호화하는핵산서열은다음과같다:  The amino acid sequence of the linker used between PD-1-GFP and the nucleic acid sequence encoding it is as follows:
핵산서열:  Nucleic acid sequence:
51-AGAATTCTGCAGTCGACGGTACCGCGGGCCCGGGATCCACCGGTCGGATCTGGTAGT-3' ; 아미노산서열: 5 1 -AGAATTCTGCAGTCGACGGTACCGCGGGCCCGGGATCCACCGGTCGGATCTGGTAGT-3 '; Amino acid sequence:
N'-RILQSTVPRARDPPVGSGS-C .  N'-RILQSTVPRARDPPVGSGS-C.
단,상기 링커의 길이 및서열은단지 예시에 불과하다.  However, the length and sequence of the linker are merely illustrative.
링커의 길이가 특정 개수, 예를 들어 아미노산 개수 기준으로 8개 (아미노산 기준) 미만으로 짧아지게 되거나, 또는 rigid및/또는 bulky한 아미노산으로 구성될 경우, 앞부분(N-말단 부분)의 卵-1과 뒷부분(C-말단 부분)의 GFP사이에 clash가 생겨서 단백질이 misfolding이 되거나, 쉽게 degradation이 일어날수 있어 PPI즉정에 영향을미치는문제가발생할수 있다. 상기 링커는 하나 이상의 G(글라이신) 및/또는 하나 이상의 S(세린)을 포함할 수 있으나, 이에 한정되는 것은 아니다. 글라이신과 세린은 다른 아미노산들과 비교하여 상대적으로 flexible하기 때문에 단백질의 folding및 PPI측정에 유리할수있다.  If the length of the linker is shortened to a certain number, for example, less than 8 (based on the number of amino acids), or consists of a rigid and / or bulky amino acid, the egg- Clashes between the GFPs at the back (C-terminal) and the backside (C-terminal) may cause misfolding of the protein or may easily result in degradation, which may affect the PPI immediate effect. The linker may include, but is not limited to, one or more G (glycine) and / or one or more S (serine). Glycine and serine are relatively flexible compared to other amino acids, which can be advantageous for protein folding and PPI measurement.
PD-Ll-mcherry는 human PD-L1 (NP_054862.1) 고유의 신호서열 (Signal sequence)을 포함한, 1번부터 290번까지의 단백질 서열과 상기 단백질서열의 카복시 말단 (C-terminal )에 연결된 mcherry (아미노산서열: MVSKGEEDNMAIIKEFMRFKVHMEGSVNGHEFEIEGEGEGRPYEGTQTAKLKVTKGGPLPFAffDILSPQF PD-Ll-mcherry has a protein sequence from 1 to 290 including a signal sequence unique to human PD-L1 (NP_054862.1) and a mcherry terminal linked to the carboxy terminal (C-terminal) of the protein sequence (Amino acid sequence: MVSKGEEDNMAIIKEFMRFKVHMEGSVNGHEFEIEGEGEGRPYEGTQTAKLKVTKGGPLPFAffDILSPQF
MYGSKAYVKHPADIPDYLKLSFPEGFKWERVMNFEDGGWTVTQDSSLQDGEFIYKVKLRGTNFPSDGPV MQKKTMGWEASSERMYPEDGALKGEIKQRLKLKDGGHYDAEVKTTYKAKKPVQLPGAYNVNIKLDITSHN EDYTIVEQYERAEGRHSTGGMDELYK) 로 구성되며, 상기 두 단백질은 16개 아미노산으로 구성된 핍타이드 링커에 의하여 연결되도록 준비하였다 (N terminal쪽: PD-L1, C terminal쪽: mcherry) . Lt; / RTI & (N-terminal side: PD-L1, C-terminal side: mcherry).
상기 PD-Ll-mcherry 사이에 사용된 링커의 아미노산 서열 및 이를 암호화하는핵산서열은다음과같다:  The amino acid sequence of the linker used between PD-L1-mcherry and the nucleic acid sequence encoding it is as follows:
핵산서열:  Nucleic acid sequence:
5 ' -TACCCAACTTTCTTGTACAAAGTGGTTGATCCGGTACCGGTCGCCACC-3 ' ;  5 '-TACCCAACTTTCTTGTACAAAGTGGTTGATCCGGTACCGGTCGCCACC-3';
아미노산서열:  Amino acid sequence:
N'-YPTFLYKWDPVPVAT-C .  N'-YPTFLYKWDPVPVAT-C.
단, 상기 링커의 길이 및서열은단지 예시에 불과하다.  However, the length and sequence of the linker are merely illustrative.
상기 사용된 링커는 16개의 아미노산으로 구성된 구조이며, N terminal의 PD-L1과 C terminal의 mcherry를연결하고 있다. 링커의 길이가 특정 개수, 예를 들어 아미노산 개수 기준으로 8개 (아미노산 기준) 미만으로 짧아지게 되거나, 또는 r igid 및/또는 bulky한 아미노산으로 구성될 경우, 앞부분(N-말단 부분)의 PD-1과 뒷부분(C-말단 부분)의 GFP사이에 clash가 생겨서 단백질이 mi sfolding이 되거나, 쉽게 degradat ion이 일어날수 있어 PPI 측정에 영향을미치는문제가발생할수 있다. 상기 링커는 하나 이상의 G(글라이신) 및/또는 하나 이상의 S(세린)을 포함할 수 있으나, 이에 한정되는 것은 아니다. 글라이신과 세린은 다른 아미노산들과 비교하여 상대적으로 f lexible하기 때문에 단백질의 folding및 PPI 측정에 유리할수 있다. 여기에 더하여, 링커가 너무 짧아지게 될 경우 C-terminus를 epi tope로 사용하고 있는 PD_L1 항체(예컨대, 9A11 (Cel l Signal ing Technology, #29122S) 등)와의 binding이 심각하게 저해되고 pul 1-down효율이 많이 떨어질 수 있어, PPI 측정에 영향을미칠수있다.  The linker used is a structure consisting of 16 amino acids and connects the N-terminal PD-L1 and the C terminal mcherry. If the length of the linker is shortened to a certain number, e. G., Less than 8 amino acids by amino acid number, or if it consists of rigid and / or bulky amino acids, Clashes occur between GFP1 and the GFP at the back (C-terminal portion), which can result in mi-sfolding of proteins or degradation, which can affect PPI measurement. The linker may include, but is not limited to, one or more G (glycine) and / or one or more S (serine). Glycine and serine are relatively lexible compared to other amino acids, which can be advantageous for protein folding and PPI measurement. In addition, when the linker becomes too short, the binding with the PD_L1 antibody using the C-terminus as epi tope (for example, 9A11 (Cell Signaling Technology, # 29122S)) is seriously impaired and pul 1-down The efficiency may drop significantly, which may affect the PPI measurement.
단, 기판에 고정되는 (즉, 제 1 단백질로 사용되는) PD-L1에 결합된 표지 (본실시예의 경우 mcherry)는 PPI측정에 필수적인 것은아니며, PD- L1에 mcherry부착없이도抑- 1/PD-L1의 상호작용을측정할수있다.  However, the label (mcherry in this embodiment) bound to the PD-L1 immobilized on the substrate (i.e., used as the first protein) is not indispensable for the PPI measurement and can be suppressed without attaching mcherry to the PD- -L1 interactions can be measured.
한편, 제 1 단백질로 사용되는 PD-L1에 표지 (예컨대, mcherry)를 붙이면 기판에 부착된 PD-L1을 정량할수 있다는 장점이 있다. 이 경우, Spectrophotometer를 이용한 형광 정량이 가능하기 때문에, 목적하는 농도의 PD-L1을 기판에 코팅할 수 있으며, 반대로, 해당 조건에서 기판에 코팅되는 PD-L1의 개수를정량적으로추적할수도있다.  On the other hand, when a label (for example, mcherry) is attached to PD-L1 used as the first protein, PD-L1 attached to the substrate can be quantified. In this case, fluorescence quantification using a spectrophotometer is possible, so that the desired concentration of PD-L1 can be coated on the substrate, and conversely, the number of PD-L1 coated on the substrate under the corresponding conditions can be quantitatively tracked.
PD-L1의 개수를 정량추적할 경우의 장점은 다음과 같이 예시할 수 있다: The advantage of quantitative tracking of the number of PD-L1 is as follows have:
기판에 코팅되는 PD-L1의 수를 추적하면 drug screening을 할 경우 inhibi tor의 negat ive select ion에 도움이 될 수 있다. Tube react ion을 통하여 1)-1*(*는 형광표지)/ PD-L1/ inhibi tor mixture를 기판에 incubat ion하고 PD-1*를 count하게 되면 PEKL과 PD-L1의 상호작용을 저해하는 약물을선별할수 있지만, 간혹 PD-L1과항- PD-L1항체의 결합을 저해하는 약물도 잘못 선별될 수 있다. 이때 기판에 결합된 PD-L1의 수를 관찰하면 이와같이 잘못선별된 약물들을다시 골라낼수있다. Tracking the number of PD-L1 coatings on the substrate can aid in the negativity of the inhibi tor when performing drug screening. 1) -1 * (* indicates fluorescence labeling) / PD-L1 / inhibi tor mixture is incubated with Tube reactant and the PD-1 * counts to inhibit the interaction of PEKL and PD-L1 , But drugs that sometimes interfere with the binding of PD-L1 and PD-L1 antibodies may also be misdiagnosed. At this time, the number of PD-L1 bound to the substrate can be observed to select the wrongly selected drug.
한편, 표지되지 않은 PD-L1을 사용하는 경우에도 negat ive select ion이 불가능한것은아니다. PD-L1 ELISA를통하여 관찰할수 있다. 다만, 표지된 (예컨대, mcherry로 표지된) PD-L1을 사용하게 되면 해당 과정을 one-step으로진행할수있다.  On the other hand, negated ive select ion is not impossible even when using unlabeled PD-L1. PD-L1 ELISA. However, if you use a labeled PD-L1 (labeled mcherry, for example), you can proceed with the process one-step.
卵 -1-GFP와卵 Ll-mcherry를 암호화하는두유전자를각각 pCDNA 3.1 backbone에 삽입하고, HEK293T세포에 도입하여, 단백질로발현시켰다.  Egg-1-GFP and soybean oil encoding egg Ll-mcherry were inserted into pCDNA 3.1 backbone, respectively, and introduced into HEK293T cells, which were expressed as proteins.
유전자의 숙주세포내도입을위하여, 통상적으로사용가능한모든 수단, 예컨대, PEI(polyethyleneimine)을 이용하는 형질도입, 1 ipofectamine 등을 이용하는 1 ipofect ion, electroporat ion 등의 방법이 사용될수있다.  For introduction of a gene into a host cell, any means generally used can be used such as transfection using PEI (polyethyleneimine), 1 ipofect ion using 1 ipofectamine, electroporat ion, and the like.
본 실시예에서는, 예시적으로 PEI를 이용한 transfect ion을 수행하였다. PEI는 highly posi t ive charge를 갖는 물질로, DNA를 머금고 세포 안으로침투하여, 단백질로발현시킨다. PEI를 이용한 transfect ion의 구체적 과정은다음과같다:  In the present embodiment, transfect ion was exemplarily performed using PEI. PEI is a highly positively charged substance that penetrates DNA into the cell and expresses it as a protein. The specific procedure of transfect ion using PEI is as follows:
100mm di sh를 기준으로 50만개의 HEK293T세포 (ATCC [293T (ATCC® CRL-3216™)] )를준비하였다. Opt i -MEM buf fer (Thermo Fi sher Scient i f ic) 2ml을 준비하여, 1ml은 DNA (앞서 준비된 卵 -1-GFP와 PDLl-mcherry 유전자를 포함하는 벡터) 20ug과, 나머지 1ml은 PEI 50ug과 각각 혼합하였다. 5분 후, DNA와 PEI를 서로 혼합하고, 추가로 20분 동안 배양하였다. DNA와 PEI 혼합물을 상기 준비된 HEK293T 세포에 처리하고, 48시간동안배양한후, 세포를수집하였다.  500,000 HEK293T cells (ATCC [293T (ATCC® CRL-3216 ™)] were prepared on the basis of 100 mm di sh. 2 ml of Opti -MEM buf fer (Thermo Fi sher Scientif ic) was prepared, 1 ml of DNA (vector containing the previously prepared egg-1-GFP and PDLl-mcherry gene) and 1 ml of the remaining 50 ml of PEI . After 5 minutes, the DNA and PEI were mixed together and incubated for an additional 20 minutes. The DNA and PEI mixture was treated with the prepared HEK293T cells, cultured for 48 hours, and then the cells were collected.
세포용해 과정에 사용된 용해 버퍼 (Lysi s buf fer )의 조성은다음과 같다.  The composition of the lysis buffer (Lysi s buf fer) used in the cell lysis procedure is as follows.
【표 5] [Table 5]
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000103_0001
Figure imgf000104_0001
수집한 세포의 양에 맞게 용해 버퍼를 첨가한 투, re-suspension, ice에서 30분 incubation한후, 14000g이상에서 15분동안원심분리하였다. 이때 protease inhibitor (Sigma-Aldr ich (11836170001) ) , phosphatase inhibitor (Sigma-Aldr ich (P5726))를 첨가하였다. 상등액을 취하여, Fluoremeter를이용하여 형광농도를측정한후보관하였다.  Re-suspension was added to dissolve buffer to the amount of cells collected, incubated in ice for 30 min, and then centrifuged at 14000 g for 15 min. At this time, protease inhibitor (Sigma-Aldrich (11836170001)) and phosphatase inhibitor (Sigma-Aldrich (P5726)) were added. The supernatant was taken and the fluorescence intensity was measured using a Fluoremeter and stored.
실시예 13.기판의준비  Example 13. Preparation of substrate
Covers lip을 K0H 1M 용액에 침지시킨 후, sonicator에 담아서 세척하였다 (20-30 min). 그 후, 3차 증류수로 잘 씻은 후, piranha solution (황산:과산화수소 = 2:1 3:1 (\"v))을 이용하여 세척하였다. 세척한 covers 1 ip 표면에 대하여 aminopropylsi lane와 PEG(polyethylene glycol)를차례로처리하여 코팅을수행하였다. Covers lip was immersed in KOH 1M solution and washed with a sonicator (20-30 min). Then, after washing well with deionized water, piranha solution (sulfuric acid: aqueous hydrogen peroxide = 2: 1 3: 1 ( \ "v)). Was washed with a aminopropylsi against the covers 1 ip surface was washed lane and PEG (polyethylene glycol) were sequentially treated to perform coating.
2시간 반응후, 3차증류수로 세척한뒤 PEG코팅된 면이 접촉되지 않게 하여 -20°C에서사용시까지 보관하였다.  After 2 hours of reaction, the cells were washed with 3 rd distilled water and stored at -20 ° C until used without contact with the PEG-coated surface.
동시에 석영 재질의 channel 형태의 기판또는 아크릴 재질의 well 형태의 기판을 준비하였다. 석영 재질의 기판의 경우, 앞서 설명한 cover si ip처리 과정을참조로세척 및 PEG코팅 과정을수행하였다.  At the same time, a quartz channel type substrate or an acrylic type well type substrate was prepared. In the case of a quartz substrate, cleaning and PEG coating processes were performed with reference to the cover si ip process described above.
아크릴 재질의 기판의 경우, 제작 후 3차 증류수에 침지시킨 후 sonication하여 세척하였다. 세척된 아크릴 재질의 기판을 5% BSA용액에 침지시키고 2시간 반응시켜 비특이적 단백질 흡착 (nonspecific protein adsorption)을방지하고, -20C에서사용시까지 보관하였다.  In the case of an acrylic substrate, the substrate was immersed in the third distilled water and sonicated for washing. The washed acrylic substrate was immersed in a 5% BSA solution and reacted for 2 hours to prevent nonspecific protein adsorption and stored at -20 C until use.
하기 시험에서는 상기 준비된 covers lip과 아크릴 재질의 기판을 사용하였으며, 시험 전에 상기 coverslip과아크릴 재질의 기판을해동하여 조립하거나, PEG 코팅이 끝난 뒤 미리 조립하고, 조립된 형태로 -20°C에 보관하여사용전에 해동하여사용하였다. In the following test, the prepared cover lip and acrylic substrate were used. Before the test, the coverslip and the acrylic substrate were thawed or assembled. After the PEG coating was completed, the assembly was pre-assembled and stored at -20 ° C And thawed before use .
실시예 14. PD-L1과 PD-1간단백질-단백질상호작용측정  Example 14. Protein-protein interaction measurement between PD-L1 and PD-1
PD-L1과 PD-1간단백질-단백질상호작용유도  Induction of protein-protein interaction between PD-L1 and PD-1
상기 실시예 13에서 준비된 기판에 Avidin 계열의 단백질인 Neutravidin (Thermo , A2666)을 0.1 mg/ml 농도로 투입하였다. 상온에서 5분반응시킨후 PBS buf fer 30 ul를이용하여 2회 기판을세척하였다. 제 1단백질로서 PD-L1또는卵 -1을사용한경우를 예시하기 위하여, 상기 준비된 기판에 상기 제 1 단백질에 대한 항체를 처리하였다. 이 때, 사용되는 항체는 biot in이 접합된 형태인 것으로 준비하였다. 항체의 농도는 항체-항원의 af f ini ty (di ssoci at ion constant , KD)에 따라 적절히 조절될 수 있으며, 본 실험예에서는 2 ug/ml 정도로 사용하였으며, 반응시간은 5분정도로하였다. 만약 Biot in이 접합되어 있지 않은항체를 사용하는 경우에는 이차항체를 이용하여 제 1단백질의 항체를부착할수 있다. The substrate prepared in Example 13 was added with the Avidin series protein Neutravidin (Thermo, A2666) was added at a concentration of 0.1 mg / ml. After 5 minutes of reaction at room temperature, the substrate was washed twice with 30 μl of PBS buf fer. To illustrate the use of PD-L1 or egg-1 as the first protein, the prepared substrate was treated with the antibody against the first protein. At this time, the antibody used was prepared as a biotin-conjugated form. The concentration of the antibody can be appropriately adjusted according to the affinity of the antibody-antigen (KD). In this experiment, the antibody concentration was about 2 μg / ml, and the reaction time was about 5 minutes. If a biotin-conjugated antibody is used, the antibody of the first protein can be attached using a secondary antibody.
본 실시예에서 사용된 제 1 단백질에 대한 항체를 다음의 표 6에 정리하였다:  Antibodies to the first protein used in this example are summarized in Table 6 below:
【표 6】  [Table 6]
Figure imgf000105_0001
Figure imgf000105_0001
상기 표 6에 예시된 항 PD-L1 항체 (9A11 또는 湖 8T4X)가 처리된 기판을 PBS buf fer 30 를이용하여 2회 세척하였다. 상기 준비된 기판에 앞서 실시예 12에서 준비한 PD-L1단백질 용해액 (mCherry-PD-Ll발현세포 용해액의 원심분리 후의 상등액; 제 1 단백질로 사용)을 투입하여, 기판에 PD-L1을 고정화시켰다. 항원-항체 반응의 경우 15분까지는 계속 증가할 수 있고 15분을초과하면서 시간대비 항원-항체 반응효율이 낮아질수 있어서, 반응시간을 15분 정도로 설정하였다. 반응 후, 기판을 PBS에 tween 20이 0.05%(v/v) 포함된 버퍼를 이용하여 세척하였다. Tween 20 0.05%는 nonspeci f ic binding을줄여주는동시에, 막단백질의 hydrophobic region이 망가지지 않도록도와준다.  The substrate treated with the anti-PD-L1 antibody (9A11 or 8T4X) shown in Table 6 was washed twice with PBS buf fer 30. PD-L1 protein solubilization solution (supernatant after centrifugation of mCherry-PD-L1 expressing cell lysis solution; used as the first protein) prepared in Example 12 was added to the prepared substrate, and PD-L1 was immobilized on the substrate . In the case of antigen-antibody reaction, it is possible to continue to increase until 15 minutes, and the efficiency of antigen-antibody reaction may be lowered over 15 minutes and the reaction time is set to about 15 minutes. After the reaction, the substrate was washed with PBS containing 0.05% (v / v) of tween 20. Tween 20 0.05% reduces nonspecic binding and helps prevent the hydrophobic region of the membrane protein from breaking down.
그 후, 상기 얻어진 표면에 PD-L1 단백질이 고정된 기판에 실시예 Thereafter, on the substrate on which the PD-L1 protein was immobilized on the obtained surface,
12에서 얻어진 PD-1단백질용해액 (GFP-PD-1발현 세포용해액의 원심분리 후의 상등액; 제 2 단백질로 사용)을 투입하였다. PD-L1 단백질은 10nM 농도로사용하고, 卵 -1단백질은 10nM, 30nM, 또는 50nM농도로사용하였다. 상기 시험 과정을도 27a에 모식적으로나타내었다. PD-1 protein solution (supernatant after centrifugation of GFP-PD-1 expressing cell lysate; used as the second protein) obtained in Example 12 was added. The PD-L1 protein contained 10 nM And egg-1 protein was used at a concentration of 10 nM, 30 nM, or 50 nM. The test procedure is schematically shown in Fig. 27A.
단백질단백질상호작용에 의한복합체 (PPI complex)의 이미징  Imaging of the complex (PPI complex) by protein protein interaction
단백질복합체 (PPI complex) 이미징 과정은다음과같다:  The PPI complex imaging process is as follows:
상기 준비된 기판 (well type 또는 quartz slide 중 어느 것을 사용하여도 무방함)을 전반사 형광현미경 (Total Internal Reflection Fluorescence (TIRF) microscope) 위에 고정시켰다. eGFP의 형광신호를 관측하기 때문에, 488 nm 의 파장을 이용하였으나, 이에 제한되지 않고 eGFP를 발광 시킬 수 있는 어떠한 파장의 레이저를 사용해도 무방하다 (레이저 파장 범위: 460 500 nm). 형광 이미지는 EMCCD (Electron- multiplying charge-coupled device; Andor iXon Ultra 897 EX2 (DU-897U- CS0-EXF))카메라를사용하여 얻었다.  The prepared substrate (either a well type or a quartz slide can be used) was fixed on a total internal reflection fluorescence (TIRF) microscope. In order to observe the fluorescence signal of eGFP, a wavelength of 488 nm was used. However, the present invention is not limited thereto, and any wavelength laser capable of emitting eGFP can be used (laser wavelength range: 460 to 500 nm). Fluorescence images were obtained using an EMCCD (Electron-multiplying charge-coupled device; Andor iXon Ultra 897 EX2 (DU-897U-CS0-EXF)) camera.
예시적으로,이미징을위한 EMCCD카메라의 각종수치를다음과같이 조정하였다:  Illustratively, various values of the EMCCD camera for imaging were adjusted as follows:
a. 레이저 파워 : 2 mW  a. Laser power: 2 mW
레이저 파워가너무높으면 eGFP가빨리 형광신호를잃어버리게 되고 ( phot ob leaching) ,너무낮으면 EMCCD에 의해 신호를관측하기 어려워진다. 상기 레이저 파워 범위는 eGFP의 발광시간 (lifetime)을 11초 정도로 유지시켜 주는범위이다. If the laser power is too high, eGFP will lose the fluorescence signal quickly (photob leaching), and if it is too low, it will be difficult to observe the signal by the EMCCD. The laser power range is a range that maintains the light-emitting time (lifetime) of e GFP about 11 seconds.
b. EMCCD카메라의 1프레임 당노출시간 (exposure time) : 0.1초 b. Exposure time per frame of EMCCD camera: 0.1 second
1 프레임 당 노출시간이 짧을수록 1 프레임에 누적되는 신호가 줄어들게 되므로, 이를 극복하기 위해서 레이저 파워를 높이거나, EMCCD의 감도를높여야한다. The shorter the exposure time per frame is, the fewer signals accumulated in one frame. To overcome this, the laser power must be increased or the sensitivity of EMCCD must be increased.
c. EMCCD의 gain (gain이 클수록 측정된 신호가 증폭되고 동시에 노이즈도증가함) : 40  c. The gain of the EMCCD (the larger the gain, the more the measured signal is amplified and the higher the noise): 40
eGFP에서 생성된 광자 (photon)는 EMCCD의 소자를 통해 전자로 바뀌어 계측된다 (광전효과, photoelectric effect) . 이 때, 광자 1개 당 생성되는 전자의 개수를 gain 값을 통해 변경할 수 있다. 설정된 gain이 높을수록 광자 1개 당 생성되는 전자의 개수가 늘어나서, EMCCD의 감도가 높아진다. 동시에 background noise도 함께 증가하므로 signal-to-noise 비율이 중요하다. 따라서, 본 실시예에서는 EMC抑의 gain을 40으로 조정하였지만, 이에 제한되는 것은 아니고, 앞서 설명한 내용을 근거로 적절히 변경할수있다. d. 총촬영 프레임 : 10프레임 The photons generated in eGFP are converted to electrons through the EMCCD element (photoelectric effect, photoelectric effect). At this time, the number of electrons generated per photon can be changed through the gain value. The higher the set gain, the greater the number of electrons generated per photon, and the higher the sensitivity of the EMCCD. At the same time, background noise also increases together, so the signal-to-noise ratio is important. Therefore, although the gain of the EMC suppression is adjusted to 40 in the present embodiment, the present invention is not limited to this, and can be appropriately changed based on the foregoing description. d. Total shooting frame: 10 frames
10 프레임 촬영을 하게 되면 현재 노출시간을 기준으로 1초동안의 PPI 변화를관찰할수 있다. 본실시예에서는 30분 간의 tube incubat ion, 15분 간의 기판에서의 incubat ion, washing step을 차례로 진행한 투, 이미징을 진행하므로, 이미징에서 관찰되는 신호의 세기 및 강도는 굉장히 안정적이다. 따라서 실험시간의 단축을 위하여 10프레임으로 고정하여 사용하고 있으며 분석은주로 (5-7)프레임을진행한다. 다만, 10프레임 중 초기 프레임 (1-3), 후반프레임 (8-10)을 측정하더라도 비슷한 값을 확보할 수있다.  If you take a 10 frame shot, you can observe the PPI change for 1 second based on the current exposure time. In this embodiment, the intensity of the signal observed in the imaging is extremely stable since the imaging is performed in succession of the incubation for 30 minutes, the incubation in the substrate for 15 minutes, and the washing step. Therefore, to shorten the experiment time, it is fixed to 10 frames, and the analysis mainly proceeds with the frame (5-7). However, a similar value can be obtained even if the initial frame 1-3 and the second half frame 8-10 of 10 frames are measured.
한 조건에 대해 (하나의 wel l 혹은 하나의 channel ) , 위치를 옮겨가며 각각 다른 위치에서 최소 10장의 이미지를 얻고 상기 얻어진 10장의 이미지를각각분석한후, 평균과표준편차를각조건의 대표값으로 설정하였다. 10장은 최소 기준이며, 장 수가 많을수록 통계적 정확도가 높아진다. 한 조건에서 모든 이미지를 얻고 나면 다음으로 이동하여 (다음 wel l혹은다음 channel )동일한동작을반복하였다.  A minimum of 10 images were obtained at different positions, each position was shifted for one condition (one wel l or one channel), and the 10 images obtained were analyzed. The mean and the standard deviation were compared with the representative values of each condition Respectively. Chapter 10 is the minimum standard, and the higher the number, the higher the statistical accuracy. Once all the images have been acquired under one condition, move to the next one (next wel or next channel) and repeat the same operation.
앞서 설명한바와같이, PD-L1을기판에 고정하는경우, PPI 신호가 cluster를 형성할수 있다. 이 경우, 레이저 파워를 줄이거나 gain수를 줄이면 spot의 형광신호 세기가 줄어들어, single GFP spot은 거의 안보이고, cluster의 신호만을 남길 수 있고, Cluster에 대한 분석만을 진행하거나, 또는 single molecule 만을 분석하고자 할 때, 또는 상관관계를 분석하고자 할 때 해당 step을 추가적으로 진행할 수도 있을 것으로생각된다.  As described above, when the PD-L1 is fixed to the substrate, the PPI signal can form a cluster. In this case, if the laser power is reduced or the gain number is reduced, the fluorescence intensity of the spot is reduced, the single GFP spot is almost invisible, only the cluster signal can be left, only the cluster is analyzed, , Or if you want to analyze the correlation, you can proceed with the step further.
PPI complex분석  PPI complex analysis
상기와 같이 이미징된 단백질 복합체 (PPI comp lex)는 다음의 과정으로분석하였다:  The imaged protein complex (PPI complex) was analyzed by the following procedure:
본 실시예에서는 Mat lab 프로그램 (MathWorks 제공)에서 제공되는 toolki t에 기반하여 단백질복합체를분석하였다.  In this example, the protein complexes were analyzed based on the toolkits provided in the Matlab program (provided by MathWorks).
1. 상기 얻어진 형광이미지는 16bi t uns i gned integer 형식으로 저장하였다. PD-1-PDL1 분석에 있어서, 5-7 프레임의 총 3개의 프레임을 사용하여 아래 과정을수행하였다. 앞서 기재한바와 같이 washing과정이 포함되기 때문에 이미징에서 보이는신호의 세기, 안정성이 매우우수하고, 10개의 프레임 중에서 1-3 프레임을 분석한 결과와 8-10 프레암을 분석한 결과, 1-10 프레임을 모두 분석한 결과를 비교해도 큰 차이가 없었다. 이 구간은이미징 조건/장비구축상태에 따라서 달라질수있다. 1. The fluorescence images obtained were stored in a 16-bit unsigned integer format. In PD-1-PDL1 analysis, the following procedure was performed using a total of three frames of 5-7 frames. As described above, since the washing process is included, the intensity and stability of the signal seen in the imaging are excellent. As a result of analyzing 1-3 frames of 10 frames and 8-10 frames, There was no significant difference in the results of the analysis of all the frames. this The interval may vary depending on the imaging condition / equipment construction status.
2. 노이즈를 제거하기 위해, 각 프레임 마다 아래와 같은 절차를 수행하였다:  2. To eliminate noise, the following procedure was performed for each frame:
a. 최초 시작은 왼쪽상단에서 시작한다. 1 프레임은 512x512 개의 픽셀로구성되어 있다. 기준픽셀을기준으로오른쪽으로 11개, 아래쪽으로 a. The initial start begins at the top left corner. One frame consists of 512x512 pixels. 11 pixels to the right based on the base pixel, down
11개인 11x11개의 픽셀에서 median 값을 구하고, 이 medi an 값을 기준 픽셀의 수치에서 빼주었다 [(Intensi ty_pixel) -The median value was obtained from 11 11x11 individual pixels, and this median value was subtracted from the reference pixel value [(Intensi_pixel) -
(medi anIntensi ty_llxllneighborhood)] . 512x512의 모든 픽셀에 대해서 위 절차를 수행하였다. Medi an f i l ter ing을 통해서 pepper & sal t noi se를 제거하였다. (median anintensi ty_llxllneighborhood)]. The above procedure was performed for all pixels of 512x512. The mediating effect of pepper & sal noi was eliminated.
b. 위의 처리된 이미지에서 sigma^O.?, si ze=5 x 5의 Gaussi an smoothing을수행하였다. 이를통해 이미지를부드럽게 만들어 주었다. c. Threshold를 설정하였다. Threshold는 전체 이미지에서 pixel intensi ty가 threshold 이하가 되는 pixel의 값을 threshold 값으로 모두 만들어 주었다. 이를 통해 이미지에서 형광신호에 의해서 만들어지지 않은 국소극대값 ( local maximum)을 제거할 수 있다. 본 실시예에서 이미징 조건에서사용되는 threshold값은 70이다.  b. In the above processed image, Gaussian smoothing of sigma ^ O.?, Si ze = 5 x 5 was performed. This helped to soften the image. c. Threshold was set. Threshold is the threshold value of the pixel that the pixel intensities become below the threshold in the whole image. This can remove local maximums not created by the fluorescence signal in the image. In this embodiment, the threshold value used in the imaging condition is 70.
3. 형광단백질로부터 나오는 신호는 특정 위치에 모여있는 형태로 발생한다 ( local i zed point spread funct ion (PSF)) . 즉 이 PSF (물리적인 값)가관측하고자하는 PPI complex (생물학적인 값)이다. 아래 단계는 이 PSF를판별하기 위한과정이다:  3. The signal from the fluorescent protein occurs in a specific location (localized spread function (PSF)). That is, this PSF (physical value) is the PPI complex (biological value) to be measured. The following steps are for determining this PSF:
a. Local maximum의 위치를구하였다 (예를들어, i번째 row, j번째 column pixel) . 앞서 기술한 바와 같아 단분자 형광신호는 512x512픽셀 중에서 특정 위치에 (현재 관측장비 하에서 대략 5x5픽셀사이즈, 1 pixel = 0.167 마이크로미터) 모여 형성되므로, local maximum을 찾으면 개별 a. The local maximum is obtained (for example, i-th row, j-th column pixel). As described above, since the monomolecular fluorescence signal is formed at a specific position among 512x512 pixels (approximately 5x5 pixel size, 1 pixel = 0.167 micrometer under the present observation equipment), when the local maximum is found,
PSF를 선별할 수 있다. 이는 Mat lab에서 제공되는 toolki t을 이용하여 구할수있다. PSF can be selected. This can be obtained using the toolkit provided in the mat lab.
b. 위에서 구한 local maximum이 실제 PSF로부터 발생한 것인지에 대한 판별과정을 수행하였다. 우선 local maximum의 최소값을 정의하고 (minimum intensi ty value) , 얻어진 local maximum 중에서 이 최소값보다 큰 경우만 분석에 사용하였다. 본 실시예에서는 최소값을 75로 정의하였으며, 이는 레이저 파워/노출시간/장비구축 상황에 따라서 달라질 수 있다. 최종적으로 얻어진 local maximum 좌표를 중심으로 5x5 픽셀의 정보를불러오고, 이 5x5픽셀에서 밝기에 대한중심을구하였다(centroid of intensi ty) . 이 때, 구해진 밝기 중심이 기존 local maximum 좌표에 대하 0.5 pixel 이상벗어나게 되면 (PSF모양에 대한 2D대칭이 사라지면), 정상적이지 않은형광신호라고판단하고분석에서 제거하였다. b. The process of determining whether the local maximum obtained from the actual PSF is performed is performed. First, the minimum value of the local maximum is defined (minimum intensities), and only the maximum of the obtained local maximums is used for the analysis. In this embodiment, the minimum value is defined as 75, which may vary depending on the laser power / exposure time / equipment construction situation. 5x5 pixel centered on the finally obtained local maximum coordinate The information was retrieved, and the centroid of intensities was obtained for this 5x5 pixel. At this time, if the brightness center deviates by more than 0.5 pixels from the existing local maximum coordinates (when the 2D symmetry about the PSF shape disappears), it is judged to be a non-normal fluorescence signal and removed from the analysis.
c. 모든 조건을 통과한 PSF 만 최종적으로 분류하여 좌표와 전체 개수를구하였다.  c. Only the PSF that passed all the conditions were finally classified and the coordinates and the total number were obtained.
모든 파일에 대해 상기 1-3의 과정을 수행하면서 PPI complex의 개수를 구하였다. 같은 조건하에서 촬영된 각 파일의 정보를 취합하여 평균과 표준편차를 구하였다. 이 값이 최종적으로 특정 조건에서 PPI complex의 개수를나타내는값이 된다.  The number of PPI complexes was determined for all the files by performing steps 1-3 above. The information of each file taken under the same conditions was collected and the mean and standard deviation were obtained. This value finally represents the number of PPI complexes under certain conditions.
상기 얻어진 PPI complex의 개수는 PPI신호의 count를구한것이나, 그 외에도 PPI 신호의 intensi ty의 합을 구하거나, 측정 영역에서 spot의 rat io등을측정하여 분석할수도있다.  The number of the obtained PPI complexes can be obtained by obtaining the count of the PPI signal or by calculating the sum of the intensities of the PPI signals or by measuring the rat io of the spot in the measurement region.
일 예에서, cluster의 신호만을 측정한 경우, 레이저 파워, gain 수를 조절하여, Cluster 신호의 크기가 줄어들면, 즉 형광의 세기가 너무 진한 spot이 줄어들게 되고, PSF의 정확한 계산에 조금 더 유리하게 된다 (local maximum이 줄어들기 때문도 있고, 겹쳐진 spot이 구분되는 경우도 있다). cluster intensi ty를 측정함으로써, 약물의 특성을좀 더 면밀하게 파악할수 있을것이다. 어떤 약물의 경우에는 PD-1과 PD-1의 cluster ing을 억제하는 약물이 있다. 해당 약물의 경우 single spot의 count는 오히려 늘어나거나 비슷하겠지만, cluster의 개수는 줄어들게 될 것이고, 실제적으로 cluster ing을 동반한 체내의 PD-1-PDL1 interact ion은 효과적으로억제할수 있을것이다. 따라서 c luster의 분포, 수를분석하는 것이 의미가있을것이다.  In one example, when only the signal of the cluster is measured, the laser power and the gain number are adjusted to reduce the size of the cluster signal, that is, the spot where the intensity of the fluorescence is too low, (The local maximum may be reduced or overlapped spots may be distinguished). By measuring cluster intensities, we will be able to more closely characterize the drug. In some drugs, there are drugs that inhibit PD-1 and PD-1 clustering. In the case of the drug, the number of clusters will be reduced, although the number of single spot counts may be rather increased or similar, and effectively inhibiting the PD-1-PDL1 interaction in the body with cluster ing. Therefore, it would be meaningful to analyze the distribution and number of c luster.
또한, 전체 intensi ty에서 cluster intensi ty를 빼면 single molecule만의 intensi ty를도출할수았다.  Also, by subtracting the cluster intensities from the total intensities, intensities of only single molecules could be derived.
cluster를 특정하여 접근하는 경우 특정 threshold 이상의 cluster intensi ty를 갖는 신호의 count 또는 sum으로 분석할 수 있고, 또는 cluster rat io를 분석할수도 있다. Single molecule을 특정하여 분석하는 경우도마찬가지이다.  When accessing a specific cluster, it can be analyzed as count or sum of signals with cluster intensities above a certain threshold, or cluster rat io can be analyzed. The same is true for single molecule identification.
PD-L1*의 형광신호대비 PD-1*의 형광신호값분석 Analysis of fluorescence signal value of PD-1 * versus fluorescence signal of PD-L1 *
1) mcherry가 부착된 PD-L1을 기판에 공급한 후, mcherry에 의해 형성되는 신호 중 특정 threshold 이상에서 형성되는 형광신호 a를 검출한다. 1) After supplying m -erry-attached PD-L1 to the substrate, the fluorescence signal a formed at a certain threshold or higher among the signals formed by the m- .
2) GFP가 부착된 抑- 1을 공급한 후 GFP에 의해 형성되는 신호 중 특정 threshold이상에서 형성되는형광신호 b를검출한다.  2) Detection of a fluorescent signal b formed at a certain threshold or higher among the signals formed by GFP after supplying suppression 1 with GFP attached thereto.
3) 형광신호 a값에 대한형광신호 b값을분석한다. 상기 형광신호 값은 PPI count , sum, rat io등일수있다.  3) Analyze the fluorescence signal b value for the fluorescence signal a value. The fluorescence signal value may be PPI count, sum, rat io, and the like.
형광신호 a값에 대한 형광신호 b값을 분석함으로써, 형광신호 b값만을 분석하는 경우보다 더 정성적인 데이터를 확인할 수 있다. 형광신호 a값을 측정하면, 특정 농도의 mcherry-PDLl을 기판에 주입하였으나 기판에 부착되지 않은 PDL1*에 대한 영향도고려할수 있다. 또한, PD-L1과 PD-1*의 상호작용을정량화함에도정확도가향상될수 있다.  By analyzing the fluorescence signal b value for the fluorescence signal a value, more qualitative data can be confirmed than when analyzing only the fluorescence signal b value. The measurement of the fluorescence signal a value can also be taken into account for the influence of PDL1 *, which is injected onto the substrate but not attached to the substrate, at a specific concentration of mcherry-PDL1. In addition, the accuracy can be improved by quantifying the interaction of PD-L1 and PD-1 *.
비교예  Comparative Example
비교를 위하여, 抑- 1과 PD-L1의 결합을 저해하는 PD-1 돌연변이인 K78A를포함하는 PD-1변이체, 또는卵 -1과 PD-L1의 결합을저해하는 PD-L1 돌연변이인 D122A+K124A를 포함하는 PD-L1 변이체, 또는 卵 -1과 PD-L1의 결합을 증대시키는 PD-1 돌연변이인 A132L을 포함하는 卵 -1 변이체를 사용하여, 상기와동일한시험을수행하였다.  For comparison, the PD-1 mutant containing the PD-1 mutation K78A, which inhibits the binding of the suppression-1 and PD-L1, or the PD-L1 mutant D122A + The same test as above was carried out using an egg-1 mutant containing the PD-L1 mutant containing K124A, or A132L, a PD-1 mutation that enhances the binding of egg-1 to PD-L1.
결과  result
상기 기판에 항 PD-L1 항체인 9A11(#CST-29122; Cel l Signal ing Technology)를사용하여 얻어진 형광이미지를도 27b에, 이를정량한결과 (PPI complex 개수; F-counts)를 도 27c에 각각 나타내었다. 도 27b 및 27c에 나타난바와같이, PD-1과 PD-L1을모두야생형 (WT)인 것으로사용할 경우, 정상적으로 PD-L1에 PD-1이 결합하면서 抑- 1의 농도 의존적으로 GFP 신호가 관찰된 반면, PD-1과 PD-L1의 결합을 저해하는 돌연변이인 卵 -1 K78A변이체 또는 PD-L1 D122A+K124A변이체를사용한경우, PD-1의 농도와 상관없이, GFP 신호를 관찰할 수 없었으며, 抑- 1과 卵 -L1의 결합력을 증대시키는 PD-1 A132L 변이체를 사용할 경우에는 WT과 비교하여 더 많은 신호를 관찰할 수 있었다. 상기 결과는 본 실시예에서 수행된 방법에 의하여 卵 -1과 PD-L1간의 단백질-단백질 상호작용 (PD-L1/PD-1상호작용)을 정확하게측정할수있음을확인시키는것이다. A fluorescence image obtained by using the anti-PD-L1 antibody 9A11 (# CST-29122; Cell Signaling Technology) is shown in FIG. 27B, and the result of quantitation (PPI complex number: F-counts) Respectively. As shown in FIGS. 27B and 27C, when PD-1 and PD-L1 are both wild type (WT), PD-1 binds normally to PD-L1 and GFP signal is observed in a concentration- On the other hand, when the mutant egg-1 K78A mutant or PD-L1 D122A + K124A mutant inhibiting the binding of PD-1 and PD-L1 was used, no GFP signal could be observed regardless of the concentration of PD-1 , And the PD-1 A132L mutant, which enhances the binding strength between Sup-1 and egg-Ll, was able to observe more signals than WT. The above results confirm that the protein-protein interaction (PD-L1 / PD-1 interaction) between egg-1 and PD-L1 can be accurately measured by the method performed in this example .
PD-L1/PD-1 상호작용 측정에 있어서, 기판에 항- PD-L1 항체를 고정하여 시험하는 경우, 형광신호에서 duster로 보이는 spot이 관찰되는 반면, 반대로, 제 1 단백질로서 卵 -1을 제 2 단백질로서 PD-L1을 각각 사용하고, 항 PDᅳ 1 항체를 기판에 고정하여, 상기와 동일한 과정으로 시험한결과, cluster가전혀 관찰되지 않고, single GFP spot으로보이는 이미지가관찰되었다. In the PD-L1 / PD-1 interaction measurement, when the anti-PD-L1 antibody was immobilized on the substrate, spot visible as a duster in the fluorescence signal was observed. On the contrary, PD-L1 was used as the second protein, the anti-PD? 1 antibody was immobilized on the substrate, As a result of the test, no images of cluster electrons were observed, and a single GFP spot image was observed.
따라서, physiologic condi t ion에서 nanocul ster를 이루는 PD-1/PD- L1의 특성상 PD-L1 항체를 사용하여 PD-L1를 기판에 고정화하여 사용하는 조건이 inhibi tor의 효능에 대한정량분석 등의 다양한분석에 접근하는데 보다유리할수있다고예측되었다.  Therefore, PD-L1 antibody was immobilized on a substrate using PD-L1 antibody in the physiologic conformation of PD-1 / PD-L1. It was predicted that it would be more advantageous to approach the analysis.
기판에 항 PD-1항체를처리하여 PD-1를고정화하여 PPI를측정하는 것을 배제하는 것은 아니다. 그런 경우, 卵 -1-mcherry 및 抑 L1-GFP로 단백질이 재조합되는 것 이외에, 상기한 PPI 측정 방법에 차이가 없다. 다만, cluster 형성에 의한 신호 발생의 차이가 있으므로, 이에 따라서 분석 방법을적절하게 변경 적용할수있다.  It is not excluded to measure the PPI by immobilizing PD-1 by treating the substrate with the anti-PD-1 antibody. In such a case, there is no difference in the above-mentioned PPI measurement method except that the protein is recombined with egg-1-mcherry and suppressed L1-GFP. However, since there is a difference in signal generation due to cluster formation, the analysis method can be appropriately changed accordingly.
또한, 기판에 PD-L1이 고정화되도록설계할때, PD-L1의 특정 도메인 (卵 -1과 binding하는 부위)이 노출되도록 하기 위하여, PD-L1을 기판에 고정화하는데사용되는항체를적절히 선택할수있다.  When the PD-L1 is immobilized on the substrate, the antibody used for immobilizing the PD-L1 on the substrate may be appropriately selected so as to expose a specific domain of PD-L1 (binding site with egg-1) have.
상기 표 6에 예시된 항- PD-L1 항체 중, 9A11(#CST-29122; Cel l Of the anti-PD-L1 antibodies exemplified in Table 6, 9A11 (# CST-29122; Cel l
Signal ing Technology)는 PD-L1의 C_말단에 결합하는 항체이고, #D8T4X (Cel l Signal ing Technology)는 PD-L1의 N_말단에 결합하는 항체이며, 항- 卵 -1 항체 중, D4W2J(Cel l Signal ing Technology)는 卵 -1의 C-말단에 결합하는항체이고, NAT105 (Abeam)는卵 -1의 N-말단에 결합하는항체이다. 이들항 PD-L1항체를기판에 고정하여 상기한시험을수행 한결과 얻어진 PPI complex 개수 (F-counts)를 도 29에 나타내었다. 도 29에서, 항- PD-L1 항체를 사용하여 시험한 경우, PD-L1의 농도는 5nM로 고정하고, PD-1 농도를 0 nM, 5 nM, 및 15 쌔로 증가시키면서 시험하고, 항- PD-1 항체를 사용하여 시험한 경우, PD-1의 농도는 5nM로 고정하고, PD-L1 농도를 0 nM, 5 nM, 및 15 nM로증가시키면서 시험하였다. Signaling technology is an antibody that binds to the C_ terminus of PD-L1, # D8T4X (Cell Signaling Technology) is an antibody that binds to the N_ terminus of PD-L1, and D4W2J (Cell Signaling Technology) is an antibody that binds to the C-terminal of egg-1, and NAT105 (Abeam) is an antibody that binds to the N-terminal of egg-1. FIG. 29 shows the number of PPI complexes (F-counts) obtained by fixing the anti-PD-L1 antibody on the substrate and performing the above test. In FIG. 29, when tested with the anti-PD-L1 antibody, the concentration of PD-L1 was fixed at 5 nM and the PD-1 concentration was tested with 0 nM, 5 nM, -1 antibody, the concentration of PD-1 was fixed at 5 nM and the PD-L1 concentration was tested while increasing to 0 nM, 5 nM, and 15 nM.
도 29에 나타난 바와 같이, PD-L1의 N-말단에 결합하는 항체인 #D8T4X (Cel l Signal ing Technology)를 사용하는 경우, C_말단에 결합하는 항체인 9A11 (CST#29122; Cel l Signal ing Technology)을 사용하는 경우와 비교하여, 卵 -1/PD-L1 상호 작용이 조건에서 적게 count ing되는 것을 확인할수있다.  29, when # D8T4X (Cell Signaling Technology), which is an antibody that binds to the N-terminal of PD-L1, is used, 9A11 (CST # 29122; 1 / PD-L1 interaction is less counted under the condition, compared to the case of using an in-vitro technology.
상기 결과는,卵 -1과 PD-L1이 상호작용을하는계면이 N-말단부위로 한정되어 있기 때문에, 기판쪽으로 PD-L1의 C-말단부위를 고정하는 것이, 안정적인 PD-1및 PDL1간상호작용을유도하는것에 유리함을보여준다. 실시예 15. PD-1/PD-L1상호작용억제 약물스크리닝에의 적용 The above results indicate that fixing the C-terminal region of PD-L1 toward the substrate is difficult because the interface between the egg-1 and the PD-L1 interacts with the N-terminal region, It is advantageous to induce the action. Example 15. Application to PD-1 / PD-L1 interaction inhibition drug screening
상기 실시예 13에서 준비된 기판에 Avidin 계열의 단백질인 Neutravidin (Thermo, A2666)을 0.1 mg/ml 농도로 투입하였다. 상온에서 5분반응시킨후 PBS buf fer 30 를이용하여 2회 기판을세척하였다. 상기 준비된 기판에 상기 실시예 14 및 도 29에서 우수한 효과를 갖는 것으로 확인된 항 PD-L1 항체 9A11 (CST#29122)를 처리하여 2 ug/ml 정도의 양으로 약 5분 간처리하여, 표면에 항 PD-L1항체 (9A11)가코팅된 기판을준비하였다. 상기 준비된 기판을 PBS buf fer 30 ul를이용하여 2회 세척하여 이후시험에사용하였다.  Neutravidin (Thermo, A2666), an Avidin series protein, was added to the substrate prepared in Example 13 at a concentration of 0.1 mg / ml. After 5 minutes of reaction at room temperature, the substrate was washed twice with PBS buffer 30. The prepared substrate was treated with the anti-PD-L1 antibody 9A11 (CST # 29122) confirmed to have excellent effect in Example 14 and FIG. 29 in the amount of about 2 ug / ml for about 5 minutes, A substrate coated with anti-PD-L1 antibody (9A11) was prepared. The prepared substrate was washed twice with 30 ul of PBS buf fer and used for further testing.
실시예 12에서 준비된 GFP-PD-l(lOnM)과 mcherry-卵 -Ll(30nM) (PD- The GFP-PD-1 (ONM) and mcherry-egg-L1 (30 nM) (PD-
L1의 경우 mcherry가 부착되지 않은 형태로도 사용 가능함)를 PD-1/PD-L1 상호작용 억제제 후보 물질과 함께 tube에서 30분동안 반응시킨 반응물을 20ul의 양으로상기 준비된기판에 가하여 15분간반응시켰다. L1 in the absence of mcherry) was reacted with PD-1 / PD-L1 interaction inhibitor candidate material in a tube for 30 minutes, and the reaction was applied to the prepared substrate in an amount of 20 ul, followed by reaction for 15 minutes .
PD-L1, PD-MGFP 표지된 PD-1) , 및 후보 약물을 tube에서 동시에 반응 (one-step react ion)시킨 반응물을 기판에 처리하는 이유 (즉, 순차적으로처리하지 않은이유)는다음과같다:  PD-L1, and PD-MGFP labeled PD-1), and the reason why the candidate drug was reacted in the tube (one-step reacted) same:
억제제가 PD-L1/PD-1*의 상호작용을 저해하는 경우는 크게 3가지로 구분할 수 있다: 1) 억제제가 PD-L1과 PD-1의 binding inter face에 강한 세기로 결합하는 경우; 2) 억제제가 (PD-1 independent ly) PD-L1의 특정 부위에 결합하여 PD-L1의 구조변화를유도하는경우; 및 3) 억제제가 (PD-Inhibitors inhibit the interaction of PD-L1 / PD-1 * in three ways: 1) when the inhibitor binds to the binding interface of PD-L1 and PD-1 with strong intensity; 2) inhibitors (PD-1 independent ly) bind to specific sites of PD-L1 to induce structural changes of PD-L1; And 3) inhibitors (PD-
L1 independent ly) PD-1의 특정 부위에 결합하여 PD-1의 구조 변화를 유도하는경우. L1 independent ly) when it binds to a specific site of PD-1 and induces a structural change of PD-1.
본 시험을 one-step react ion0] 아닌 stepwi se react ion (PD-L1 , inhibi tor , PD_1을 순차적으로 washing/binding 과정을 통하여 결합)에 의하여 수행하는 경우, 상기 1)번의 경우는 효과적으로 선별할 수 있는 반면, 2) 및 3) 번의 경우는, 약물과 target의 col l i sion자체가유도되지 않을수 있기 때문에, 검출에서 놓치는 populat ion이 생기게 되어, 정확한 억제 효과를 시험하는데 한계가 있다. 또한 one-step react ion을 하는 경우가시간적인 면에서도 효율적이므로, 본 실시예에서는 tube (Standard 1.5ml ep-tube)에서의 one-step react ion방식을채택하였다. If this test is carried out by a stepwise reaction ion (PD-L1, inhibi tor, PD_1) which is not one-step react ion [ 0 ], the above step 1) can be effectively screened On the other hand, in cases 2) and 3), since the coli lion itself of the drug and the target may not be induced, a missed populat ion is generated, so that there is a limitation in testing the correct inhibitory effect. Also, since one-step reaction is efficient in terms of time, the one-step reaction method in the tube (Standard 1.5 ml ep-tube) is adopted in this embodiment.
이후의 시험 과정은상기 실시예 16의 PPI complex의 이미징 (488 nm) 및 PPI complex분석 과정과동일하게수행하였다.  The subsequent test procedure was the same as the imaging of the PPI complex of Example 16 (488 nm) and the PPI complex analysis.
도 28a는본실시예의 억제제후보약물스크리닝 과정을모식적으로 보여준다. 28A schematically shows the inhibitor candidate drug screening process of the present embodiment Show.
도 28b는 tube에서의 one- step react ion의 반응 조건을 최적화하기 위한 시험 결과를 보여주는 것으로, 실시예 12에서 준비된 GFP-卵 -1와 mcherry-PD-Ll을 tube에서 혼합한후 30분또는 2시간동안 incubat ion하고, Fluorescence counts (F-counts)를추적한결과를보여주는그래프이다. 도 28b에서, 'Negat ive '는 tube에 mch-PDLl을 첨가하지 않은 반응물, FIG. 28B shows the results of a test for optimizing the reaction conditions of one-step reaction in a tube. The GFP-egg-1 and mcherry-PD-Ll prepared in Example 12 were mixed in a tube for 30 minutes or 2 Incubation time, and fluorescence counts (F-counts). In FIG. 28B, 'Negat ive' indicates a reaction product in which mch-PDLl was not added to the tube,
'Negat ived는 tube에 GFP-PD1을 첨가하지 않은 반응물을 각각 의미한다. 도 28b에서 확인되는 바와 같이, 30분 incubat ion을 진행한 경우와 2시간 incubat ion을 진행한 경우에서 PPI complex 개수는 큰 차이를 보이지 않았다. 'Negat ived means reactants without GFP-PD1 added to the tube, respectively. As can be seen in FIG. 28 (b), the number of PPI complexes did not show a significant difference between the incubation time of 30 minutes and the incubation time of 2 hours.
卵 -1/PD-L1 상호작용 억제제 후보 물질로서 시판중인 두 종의 PD- 1/PD-L1 억제제인 BMS202 및 S7911 (이상, Sel leck Chem에서 구입)을 사용하여 얻어진 PPI complex 개수 (F-counts)를 도 28c에 나타내었다. 도 28c에서, PD-1/PD-L1 complex가 상기 억제제 처리 농도 의존적으로 감소하는것을확인할수 있다. 또한, BMS202의 경우에는, 정제된 단백질을 사용하여 계산된 IC5◦값과 유사한 정도의 IC50 값 (18nM)을 보이는 것을 확인하였다. 상기 결과는 본 실시예에서 수행된 PD-1/PD-L1 상호작용 측정에 의하여 PD-1/PD-L1억제제를정확하게판별할수있음을보여준다.  The number of PPI complexes obtained using two commercially available PD-1 / PD-L1 inhibitors BMS202 and S7911 (purchased from Sel leck Chem) as candidate agents for egg-1 / PD-L1 interaction inhibition ) Is shown in Fig. In Fig. 28C, it can be seen that the PD-1 / PD-L1 complex is reduced in dependence on the inhibitor treatment concentration. Also, in the case of BMS202, it was confirmed that the IC50 value (18 nM) similar to the IC50 value calculated using the purified protein was shown. The results show that the PD-1 / PD-L1 inhibitor can be accurately determined by the PD-1 / PD-L1 interaction assay performed in this example.
PD-1/PD-L1 상호작용 억제제 후보 물질로서 PPI l ibrary screening (http:/八 vww. asinex.com/ppi/)에서 제공하는 non-macrocycl ic PPI drug l ibrary 중에서 샘플링한 80개의 화합물을 luM의 양으로 사용하여 얻어진 PPI complex 개수 (F-counts)를 도 28d에 나타내었다. 도 28d의 결과에서, PD-1/PD-L1 complex 개수가 Control count 150개보다 적은 (예컨대, 100개 미만인) 약물 4종을 선별하였다 (플레이트 번호: Bl, B2, B3, 및 E2; 화합물 구조는 도 33 참조) . 약물 대신 DMS0를 처리하여 Negat ive control로사용하였고, BMS202를처리하여 posi t ive control로사용하였다. 상기 선별된 4종의 약물 (Bl, B2, B3, 및 E2)을 대상으로 t i trat ion을 진행하여 그 결과를 도 28e에 나타내었다 (각 약물에 표시된 4개의 눈금은 왼쪽부터 순서대로 lmM, 10uM, 100nM, InM를 나타내며, P는 약물 대신 DMS0를 처리하여 정상적으로 존재하는 PPI level을 확인한 결과를나타내고, 은 mch-PDLl을 coat ing하지 않은경우의 결과를나타냄) . 도 28e에 나타난 바와 같이, 대조 약물로 사용된 약물 A1의 경우 전혀 효능을보이지 않은반면, 선별된 4종의 약물 (Bl, B2, B3, 및 E2)은모두 농도 의존적으로 PD-1/PD-L1 상호작용 억제 효과를 나타내었다. 특히, B2가가장좋은효능을보였으며, 이는 시판 약물인 BMS202의 효능과동등 이상인정도로확인되었다. Eighty compounds sampled in the non-macrocyclic PPI drug librium provided by PPI libris screening (http: / / vw.asinex.com/ppi/) as candidates for PD-1 / PD- The number of PPI complexes (F-counts) obtained by the use of the amount of each of the PPI complexes is shown in FIG. In the results of FIG. 28 (d), four drugs with PD-1 / PD-L1 complex number less than 150 (for example, less than 100) were selected (plate numbers: B1, B2, B3, and E2; 33). DMSO was used instead of the drug and used as a negative control. BMS202 was used as a positive control. The results are shown in FIG. 28E (the four scales displayed on each drug are lmM, 10 uM (in order from left), 10 uM , 100 nM, and InM. P indicates the result of confirming the normal PPI level by treating DMSO instead of the drug, and shows the result when the mch-PDLl is not coated. As shown in FIG. 28 (e), the drug A1 used as the reference drug showed no effect at all, while the four selected drugs (Bl, B2, B3, and E2) Dependent inhibition of PD-1 / PD-L1 interaction. In particular, B2 showed the best efficacy, which was found to be equal to or better than the efficacy of BMS202, a commercially available drug.
실시예 16. 기판에 PD-1을고정시킨경우와 PD-L1을고정시킨경우의 비교  Example 16 Comparison between PD-1 and PD-1 immobilized on a substrate
실시예 12내지 14의 과정을참조하여, 기판에 GFP PD-L1을고정시킨 경우와, GFP-PD-1을 고정시킨 경우의 PD-L1와 PD-1 간 단백질-단백질 상호작용(형광 complex개수: F counts)을측정하였다.  Protein-protein interaction between PD-L1 and PD-1 when GFP PD-L1 was immobilized on a substrate and GFP-PD-1 was immobilized on the substrate (fluorescence complex number : F counts) were measured.
Total GFP- labeled count 및 형광 이미지를 도 30a에 나타내었다 (PSF cut of f >150). 도 30a에 나타난 바와 같이, 기판에 GFP PD-L1을 고정시킨 조건과 GFP-抑- 1을 고정시킨 조건에서 유사한 결과를 나타냄을 확인할수있다.  Total GFP-labeled counts and fluorescence images are shown in FIG. 30A (PSF cut of f> 150). As shown in Fig. 30 (a), it can be confirmed that GFP PD-L1 is fixed on the substrate and GFP-suppress-1 is fixed.
GFP PD-L1및 GFP PD-1의 cluster rat io(%) (calculated by [Count @ PSF cut of f >350/Count @ PSF cut of f >150])를 도 30b에 나타내었다. 상기 Cluster rat io %)는 PSF (point spread funct ion) cut of f 350이상으로 정의한 cluster populat ion을 cut of f 150이상의 total count로 나눈 값을 백분율로 표시한 값이다. 도 30b에 나타난 바와 같이, 기판에 GFP PD-L1을 고정시킨 조건에서, GFP-卵 -1을 고정시킨 조건과 비교하여, 40%이상증가한 cluster rat io를보이는것을확인할수있다.  The cluster rat io (%) of GFP PD-L1 and GFP PD-1 (calculated by [Count @ PSF cut of f> 350 / Count @ PSF cut of f> 150]) is shown in FIG. The cluster rat io% is a value obtained by dividing a cluster population defined as a point spread function (PSF) cutoff of 350 or more by a total count of a cutoff of 150 or more as a percentage. As shown in Fig. 30B, it can be seen that the cluster rat io increased by 40% or more as compared with the condition in which GFP-O-1 was fixed under the condition that GFP PD-L1 was immobilized on the substrate.
실시예 17.항 PD-L1항체의 에피토프맵핑  Example 17. Epitope mapping of anti-PD-L1 antibodies
PD-L1를 기판에 고정시키기에 적합한 항 PD-L1 항체의 에피토프 맵핑을 위하여, PD-L1 mutant construct를 설계하였다 (도 31a 참조). 구체적으로, PD-L1 cytosol i c domain target ing ant ibody의 epi tope을 확인하기 위하여, PD-L1 cytosol ic domain delet ion mutant construct를 설계하였으며, 상기 mutant는 PD-L1 아미노산 서열 ( NP_001300958.1) 중, 아미노산 260-290번에 해당하는 cytosol ic part를 10개 단위로 나누어, 각각 d280 (281번째 이후의 아미노산 부위 결실) 또는 d270 (2기번째 이후의 아미노산부위 결실)의 변이를포함하는단백질을생산하였다.  For epitope mapping of anti-PD-L1 antibodies suitable for immobilizing PD-L1 on a substrate, a PD-L1 mutant construct was designed (see FIG. 31A). Specifically, the PD-L1 cytosol ic domain deletion mutant construct was designed to identify the epi tope of the PD-L1 cytosol ic domain targeting antibody. The mutant was designed as a PD-L1 amino acid sequence (NP_001300958.1) The cytosol ic part corresponding to amino acids 260-290 was divided into 10 units to produce a protein containing a mutation of d280 (amino acid deletion after 281) or d270 (deletion of amino acid deletion after the 2nd) .
실시예 12 내지 14를 참조하여, PD-L1 pul 1-down assay를 통하여, 앞서 (도 29 참조) PD-L1/PD-1 상호작용 측정에 효과적인 것으로 나타난 PD-L1 ant ibody (9A11)의 PD-L1 WT (야생형), d280, 또는 d270 변이체에 대한 결합 정도 (항체와 PD-L1 간 결합에 의한 형광 complex 개수: F- counts)를 측정하여, 도 31b에 나타내었다. 도 31b에 나타난 바와 같이, PD-L1의 d280또는 d270변이체의 경우, 항체와전혀 결합하지 않는 반면, 의 경우에는 항체와 민감하게 결합하는 것을 확인할 수 있다. 이를 통하여, PD-L1의 아미노산 280-290번에 해당하는 서열이 抑- L1/卵 -1 상호작용 측정에서 PD-L1을 기판에 고정화하기 위하여 사용되는 항체와의 반응에 중요하게 작용할것임을예측할수있다. PD of the PD-L1 ant ibody (9A11) shown to be effective for measuring the PD-L1 / PD-1 interaction (see FIG. 29) previously, through the PD-L1 pul 1-down assay, The degree of binding (number of fluorescent complexes due to binding between antibody and PD-L1: F-counts) to the L1-WT (wild type), d280, or d270 mutant was measured and shown in Fig. 31B, In the case of the d280 or d270 variant of PD-L1, it does not bind to the antibody at all, while in the case of the antibody it binds sensitively. It is predicted that the sequences corresponding to amino acids 280-290 of PD-L1 will play an important role in the reaction with antibodies used to immobilize PD-L1 on the substrate in the suppression-L1 / egg-1 interaction assay have.
또한, PD-L1의 d280또는 d270 변이가 PD-1과의 상호작용에 미치는 영향을시험하기 위하여, PD-1/PD-L1상호작용을기반으로한면역침강실험 (실시예 14 참조)에 의하여 PD-1 (항 PD-1 항체를 이용하여 기판에 고정시킴)와 PD-Ll WT, d280, 또는 d270 변이체 간의 단백질-단백질 상호작용 정도를 측정하여, 도 31c에 나타내었다. 도 31c에 나타난 바와 같이, PD-L1의 d270변이체 및 d280변이체의 경우, 1T대비 약 70%정도의 수준으로 PD-1과의 결합력을보임을확인할수있다.  Furthermore, in order to test the effect of the d280 or d270 mutation of PD-L1 on the interaction with PD-1, an immunoprecipitation experiment based on the PD-1 / PD-L1 interaction (see Example 14) The degree of protein-protein interaction between PD-1 (immobilized on substrate with anti-PD-1 antibody) and PD-Ll WT, d280, or d270 mutant was measured and is shown in FIG. As shown in FIG. 31C, the d270 mutant and the d280 mutant of PD-L1 showed a binding strength to PD-1 at a level of about 70% of 1T.
실시예 18 : HER2-HER3 heterodimer형성  Example 18: Formation of HER2-HER3 heterodimer
HER3의 세포내 도메인 말단에 eGFP가 연결된 HER3_eGFP 재조합 유전자 (四 R3: NM_001982.3 BC082992.1, eGFP: MH087225 링커: HER3_eGFP recombinant gene in which eGFP is connected to the intracellular domain termini of HER3 (R4 R3: NM_001982.3 BC082992.1, eGFP: MH087225 linker:
PTFLYKWDPVPVAT 또는 GGGSGGGT, 벡터: pEGFP-Nl addgene, pEGFP-Nl- FLAG))를 HER2-ove rexpressing SKBR3 breast cancer cel Is (한국세포주은행 또는 ATCC)에 electroporat ion을통해 주입하여, HER3의 과발현을유도한다. 5X1CT6개의 SKBR3 세포에 30ug의 HER3_eGFP pi asm id를 전기 천공법 (electroporat ion) 을 이용하여 transfect ion 한다 (전기 천공법 조건:PEGFP-Nl-FLAG) is injected into the HER2-ove rexpressing SKBR3 breast cancer cell line (Korean Cell Line Bank or ATCC) through electroporation to induce overexpression of HER3. 30 ug of HER3_eGFP pi asm ids were transfected in 6X1CT6 SKBR3 cells using electroporat ion (electroporation condition:
950V, 35ms , 2 pul se) . 90mm cul ture di sh에 1일동안배양하여 (RPMI1640, FBS 10%) SKBR3가 HER3_eGFP를 발현할 수 있도록 하였다. 기존의 배지를 제거하고 FBS를 넣지 않은 배지 (RPMI1640) 로 12 18시간동안 배양하여 불특정한생장인자의 영향을제거하였다. 950V, 35ms, 2 pulses). (RPMI1640, FBS 10%) in 90 mm cul ture di sh to allow SKBR3 to express HER3_eGFP. Existing medium was removed and cultured for 12-18 hours with FBS-free medium (RPMI1640) to remove the influence of unspecified growth factors.
상기 준비된 SKBR3에 NRGbl (lOOng/ml)을 투입하여 10분동안 incubat ion 하여, 세포 표면에서의 HER2 단백질과 HER3_eGFP 단백질의 헤테로다이머를형성을유도하였다 (도 34a참조) .  NRGbl (lOOng / ml) was added to the prepared SKBR3 and incubated for 10 minutes to induce formation of a heterodimer of HER2 protein and HER3_eGFP protein on the cell surface (see Fig. 34A).
HER3-eGFP 발현량에 따른 HER2-HER3_eGFP heterodimer 생성 수준을 전반사 단분자 형광현미경 (single-molecule Total Internal Ref lect ion Fluorescence Mi croscope)으로 즉정하여, 그 결과를 도 34b에 나타내었다. 도 34b에 나타난 바와 같이, SKBR3에 HER3-eGFP plasmid를 도입시킨 후 NRGbl를 처리하지 않은 경우 HER2-HER3 heterodimer가거의 생성되지 않은 것에 반하여, NRGbl를 처리한 경우, HER2-HER3 heterodimer가 HER3-eGFP 발현량에 비례하여 증가함을 확인할 수 있다. 상기 결과는 NRGbl을 투입하여야, HER2-HER3 heterodimer를생성시킬수있음을보여준다. The level of HER2-HER3_eGFP heterodimer production according to the expression level of HER3-eGFP was determined by a single-molecule Total Internal Reflection Fluorescence Microscope, and the result is shown in FIG. 34B. HER2-HER3 heterodimer was not produced when NRGbl was not treated after introduction of HER3-eGFP plasmid into SKBR3 as shown in FIG. 34B, whereas HER2-HER3 heterodimer was not produced when HER2-HER3 heterodimer was treated with NRGbl And increased in proportion to the expression level. The results show that NRGbl should be injected to generate the HER2-HER3 heterodimer.
또한, NRGbl를 세포 용해 전에 처리한 경우와 세포 용해 이후에 처리한 경우의 HER2-HER3 heterodimer 생성 수준을 전반사 단분자 형광현미경 (s ingl e-molecule Total Internal Ref lect ion Fluorescence In addition, the level of HER2-HER3 heterodimer production when NRGbl was treated before and after cell lysis was analyzed by total internal fluorescence microscopy
Mi croscope)으로측정하여, 그 결과를도 34c에 나타내었다. 세포용해는 용해 버퍼 ( 1% Digi tonin또는 1% GDN , 10% glycerol , 50mM HEPES-NaOH [pH 7.4] , 150mM NaCl , 2% protease inhibi tor cocktai l (Sigmaaldr i chMi croscope). The results are shown in Fig. 34C. Cell lysis was carried out in lysis buffer (1% Digi tonin or 1% GDNO, 10% glycerol, 50 mM HEPES-NaOH [pH 7.4], 150 mM NaCl, 2% protease inhibitor cocktail
P8340) %농도는 모두 w八 0를 사용하여 수행하였다. 도 34c에서와 바와 같이, NRGbl를 세포 용해 후에 처리한 경우 (즉, 세포 용해물에 NRGbl를 처리; Extract+NRGbl) , HER2-HER3 heterodimer가 NRGbl를 처리하지 않은 경우와유사한정도로거의 생성되지 않은 것에 반하여, ■Gbl를세포용해 전에 처리한 경우, HER2-HER3 heterodimer 생성 수준이 현저하게 높은 것으로 나타났다. 이러한 결과는 rnGbl를 세포 용해 이전에 투여하여야 HER2-HER3 heterodimer를 생성시킬 수 있으며, 세포 용해와 동시 또는 그 이후에 NRGbl를투여하는경우 HER2-HER3 heterodimer를유의미한수준으로 생성시킬수없음을보여준다. P8340)% concentrations were all performed using w0. As shown in FIG. 34C, when NRGbl was treated after cell lysis (i.e., treatment of NRGbl with cell lysate; Extract + NRGbl), the HER2-HER3 heterodimer was produced to a similar extent as that in the case where NRGbl was not treated On the contrary, when Gbl was treated before cell lysis, the level of HER2-HER3 heterodimer production was remarkably high. These results suggest that administration of rnGbl prior to cell lysis can produce a HER2-HER3 heterodimer, and that administration of NRGbl concurrently with or after cell lysis does not produce a significant level of HER2-HER3 heterodimer.
상기와 같은 세포 용해 이후에, 상기 세포 용해물을 기판에 처리해 HER2-HER3 heterodimer를 기판에 부착한 뒤 detergent로 세척하였다. detergent 종류에 따른 HER3 활성화 정도를 시험하기 위하여, detergent로서, DGTN(digi tonin) 0. 10%(w/v) , GDN 0.01%(w/v) , CHAPS0 (3- ( [ 3-Cho 1 am i dopropy "dimethyl ammon i o ) -2-hydroxy- 1-pr opanesu 1 f onat e ) 1.00%(w/v) , 0G(n-octy卜 P -D-glucos i de) 1.00%(w/v) , DDM(n-Dodecyl P -D- mal tos ide) 0.01%(w/v) , 및 TX100 0. 10%(w八 0를 각각사용하여 세척한후, HER3의 Y1289의 인산화정도를 전반사단분자형광현미경 (s ingle-mo lecule After the cell lysis, the cell lysate was treated on the substrate to attach the HER2-HER3 heterodimer to the substrate and washed with detergent. DGTN (digi tonin) 0.1% (w / v), GDN 0.01% (w / v), CHAPS0 (3- (3-Cho 1am (w / v), OG (n-octyl P-D-glucoside) 1.00% (w / v) , DDM (n-Dodecyl P-D-maltose ide) 0.01% (w / v), and TX100 0.10% Fluorescence microscope (s single-mo lecule
Total Internal Ref l ect ion Fluorescence Mi croscope)으로 즉정하여, 그 결과를도 35e에 나타내었다. Total Internal Reflectance Fluorescence Microcope), and the result is shown in Fig. 35E.
도 35e에서와같이, 기판에 부착된 HER2-HER3 heterodimer를세척시, detergent로서 digi tonin 또는 를 사용하는 경우, HER3의 Y1289의 인산화 정도가 높게 나타난 반면, CHAPS0 , 0G , DDM , 및 TX100를 사용한 경우에는낮게나타남을알수있다.  35E, when using digi tonin or detergent as a detergent when washing a HER2-HER3 heterodimer attached to a substrate, the degree of phosphorylation of Y1289 of HER3 was high, whereas when CHAPS0, 0G, DDM, and TX100 were used In the first place.
또한, detergent 농도에 따른 HER3활성화정도를시험하기 위하여 , 대표적으로 digi tonin를 0. l%(w/v) , 0.05%(w/v) , 0.01%(w/v) 및 0.0%(W八) (무처리) 등의 다양한 농도로 사용하여 세포 용해물을 세척한 경우의 HER3의 Y1289의 인산화 정도를 전반사 단분자 형광현미경 (single molecule Total Internal Reflection Fluorescence Microscope)으로 측정하여 , 그결과를도 35c및 35d에 나타내었다. 도 35c및 35d에 나타난 바와 같이, digitonin의 경우, 세포 세척시의 사용 농도를 약 0.05%(w/v) 이상으로 하는 경우에 HER3 활성화 정도가 우수하고, 이보다 낮은 경우, 활성화정도가감소함을확인할수있다. In order to test the degree of HER3 activation according to the detergent concentration, digi tonin was added in an amount of 0.1% (w / v), 0.05% (w / v), 0.01% The degree of phosphorylation of Y1289 of HER3 when the cell lysate was washed using various concentrations such as 0.0% (W8) (no treatment) was measured with a single molecule Total Internal Reflection Fluorescence Microscope, The results are shown in Figs. 35C and 35D. As shown in FIGS. 35C and 35D, in the case of digitonin, the degree of HER3 activation is excellent when the concentration used for cell washing is about 0.05% (w / v) or more, You can check.
상기와 같이 얻어진 HER2-HER3 heterodimer를 기판에 부착시켰다. 이를 위하여, 기판 (substrate)에 PEG, neutravidin, biotin, 및 anti_HER3 antibody를 표면처리 한 후 (아래 참조), 세포 용해물을 주입하여 HER2- The HER2-HER3 heterodimer thus obtained was attached to the substrate. To this end, the surface of the substrate was treated with PEG, neutravidin, biotin, and anti-HER3 antibody (see below) and the cell lysate was injected to the HER2-
HER3 heterodimer를 기판 표면에 부착한 뒤, digitonin (0.1%(w八 0)로 세척한다. After attaching the HER3 heterodimer to the substrate surface, rinse with digitonin (0.1% (w / v)).
(1) PEG표면처리  (1) PEG surface treatment
a. Coverslip과 quartz slide를 황산과 과산화수소 2:1 용액 (piranha 용액) 에 30분동안 반응시킨다. 반응이 끝나면
Figure imgf000117_0001
증류수에
a. Coverslip and quartz slide are reacted with sulfuric acid and hydrogen peroxide 2: 1 solution (piranha solution) for 30 minutes. When the reaction is over
Figure imgf000117_0001
In distilled water
10분동안중화시켰다. Neutralized for 10 minutes.
b. 중화된 coverslip과 slide를 18 M요 증류수에 5회 이상 세척하였다.  b. Neutralized coverslips and slides were rinsed in distilled water 18 times more than 5 times.
c . Coverslip과 slide를 ami nos i lane 용액 (3ml N_[3_ (Tr imethoxysi lyl )propyl ] ethyl enedi amine, 5m 1 acetic acid, 92ml methanol ) 에 20분동안 반응시키고, 반응이 끝나면 methanol로 2회 세척하고 methanol에 완전히 잠기게두었다.  c. Coverslip and slide were incubated for 20 minutes in ami nos i lane solution (3 ml N_ [3_ (Tr imethoxysilyl) propyl] ethyl enediamine, 5 ml acetic acid, 92 ml methanol) .
d. 압축질소가스를이용해 준비된 coverslip과 slide를건조시켰다. PEG버퍼 (42mg Sodium bicarbonate, 347.5mg potassium sulfate을 5m 1 18 MQ 증류수에 용해)를준비하였다.  d. The prepared coverslip and slide were dried with compressed nitrogen gas. PEG buffer (42 mg sodium bicarbonate, 347.5 mg potassium sulfate dissolved in 5 m 1 18 MQ distilled water) was prepared.
e. Slide에 PEG용액 (3mg biotin-PEG, lOOmg mPEG, at lml of PEG 버퍼에 용해)을도포한 뒤, coverslip으로 덮어 slide와 coverslip사이에 PEG용액이 골고루묻게하고, 2시간동안반응시켰다.  e. The slides were coated with a solution of PEG (3 mg biotin-PEG, lOOmg mPEG, dissolved in at lml of PEG buffer), covered with a coverslip and allowed to react with the PEG solution evenly between the slide and coverslip for 2 hours.
f . 반응이 끝난 coverslip과 slide를 18
Figure imgf000117_0002
증류수로 씻어 내고 압축 질소 가스로 건조시켰다. 완성된 coverslip과 slide는 PEG용액이 처리된 면이 접촉되지 않도록 50ml tube에 넣어 진공 포장 후 냉동하였다 (-20°C). Coverslip과 slide의 PEG용액이 처리된 면에 어떠한 접촉도 없도록하였다. (2)조립 및사용
f. The coverslip and slide 18
Figure imgf000117_0002
Washed with distilled water and dried with compressed nitrogen gas. The completed coverslip and slide were vacuum packed in a 50 ml tube (-20 ° C) to prevent contact with the treated surface of the PEG solution. The Coverslip and the slide PEG solution were kept free of any contact with the treated surface. (2) Assembly and use
a. 얼려두었던 coverslip과 slide를 37 °C 인큐베이터에서 녹여 물방울이 맺히지 않도록하였다. a. The frozen coverslips and slides were dissolved in a 37 ° C incubator to prevent water droplets from forming.
b. PEG처리된 coverslip과 slide의 면이 서로마주보도록하여 양면 테잎과 에폭시등을 이용하여 covers Lip과 slide 사이에 반응 chamber를 만들었다.  b. A reaction chamber was made between the coverslip and the slide using a double-sided tape and epoxy, with the PEG-treated coverslip and the sides of the slide facing each other.
c. 에폭시가충분히 굳으면 neutravidin용액 (lOOug/ml neutravidin, c. Once the epoxy is sufficiently cured, the neutravidin solution (lOOug / ml neutravidin,
PBS 또는 0.1% Triton-X-100, 50mM HEPES[pH 7.4], 150mM NaCl)을 chamber에 넣고 5분동안반응시켰다. PBS or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) was added to the chamber and reacted for 5 minutes.
d. Chamber에 남아있는 neutravidin용액을 PBS로 씻어내고 biotin conjugated된 항 HER2항체 (eBioscience, Cat .No. BMS120BT, clone 2G11) 또는 항 HER3 항체 (R&D systems , Cat .No. BAM348 , clone: #66201) (2~5ug/ml , PBS또는 0.1% Tr iton-X-100, 50mM HEPES[pH 7.4], 150mM NaCl) 를넣고, 5분동안반응시켰다.  d. The neutravidin solution remaining in the chamber was washed with PBS and incubated with biotin conjugated anti-HER2 antibody (eBioscience, Cat. No. BMS120BT, clone 2G11) or anti-HER3 antibody (R & D systems, Cat. No. BAM348, clone: # 66201 5 μg / ml, PBS or 0.1% Tr iton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) was added and reacted for 5 minutes.
e. Chamber에 남아있는항체를 PBS로씻어내고, 상기 ,세포용해물을 주입한후, 15분동안반응시켰다.  e. The antibody remaining in the chamber was washed with PBS, and the above cell lysate was injected, followed by reaction for 15 minutes.
f . Chamber에 남아있는 용해물을 washing buffer (0.1% digitonin, 또는 0.01% GDN, 50mM HEPES[pH 7.4], 150mM NaCl, 1% glycerol)로 세척하였다.  f. The lysates remaining in the chamber were washed with washing buffer (0.1% digitonin, or 0.01% GDN, 50 mM HEPES [pH 7.4], 150 mM NaCl, 1% glycerol).
실시예 19. HER2-HER2 homodimer형성  Example 19. Formation of HER2-HER2 homodimer
실시예 18의 방법을 참조하여, HER2의 세포내 도메인 말단에 eGFP가 연결된 HER2-eGFP 재조합 유전자 (HER2: NMJ304448, 링커 PTFLYKWDPVPVAT 또는 GGGSGGGT, eGFP: MH087225, 벡터: pEGFP-Nl addgene pEGFP-Nl-FLAG)를 HEK293T 세포주 (한국세포주은행, 또는 ATCC)에 electroporation을 통해 주입하여, HER2의 과발현을 유도하고, 상기 세포 표면에서의 HER2-HER2 homodimer (HER2 homodimer로도표시함)의 형성을유도하였다 (도 34g참조). 상기 준비된 HER2 homodimer 형성 세포를용해 (lysis)한후, 기판에 처리해 HER2 homodimer를 부착한다. 부착된 HER2 homodimer를 detergent를 이용해 세척한다. 실시예 18의 시험과정을 참조하여, 다양한 detergent를 다양한농도로사용하여 부착된 HER2 homodimer를세척한결과얻어진 HER2 Y1196의 인산화 수준 (활성화 정도)을 도 35i에 나타내었다. 도 35i에서와 같이, detergent로서 DGTN(digitonin)를 0.05%(w/v) 이상 또는 0.10%(w/v) 이상을 사용하거나, GDN 0.01%(w/v) 이상 사용하여 부착된 HER2 homodimer를세척한경우, HER2활성화정도가높은것으로나타났다. HER2-eGFP recombinant gene (HER2: NMJ304448, linker PTFLYKWDPVPVAT or GGGSGGGT, eGFP: MH087225, vector: pEGFP-Nl addgene pEGFP-Nl-FLAG) in which eGFP is connected to the intracellular domain termini of HER2, Was injected into HEK293T cell line (Korean Cell Line Bank, or ATCC) through electroporation to induce HER2 overexpression and induce the formation of HER2-HER2 homodimer (also referred to as HER2 homodimer) on the cell surface ). The prepared HER2 homodimer-forming cells are lysed and then treated with a HER2 homodimer. The attached HER2 homodimer is washed with detergent. Referring to the test procedure of Example 18, the phosphorylation level (degree of activation) of HER2 Y1196 obtained as a result of washing the attached HER2 homodimer using various detergents at various concentrations is shown in Fig. 35i. As shown in FIG. 35i, the amount of HER2 adhered using more than 0.05% (w / v) or 0.10% (w / v) of DGTN (digitonin) or 0.01% When the homodimer was washed, the degree of HER2 activation was high.
실시예 18의 과정을참조하여, 기판(substrate)에 PEG, neutravidin, biotin, 및 ant i-HER2 antibody를표면처리 한후, 세포용해물을주입하여, HER2 homodimer를 기판 표면에 부착시킨 뒤, 상기 DGTN(digitonin) 0.1%(w八0로세척했다.  After the surface of the substrate was treated with PEG, neutravidin, biotin, and ant i-HER2 antibody, the cell lysate was injected to adhere the HER2 homodimer to the substrate surface, and the DGTN (digitonin 0.1%).
이 때 얻어진 형광신호를 도 34h에 나타내었다. 도 34h에서, 1- step으로 형광 세기가 감소하는 경우는 monomer에 해당하고, 2-step으로 형광신호가감소하는 경우는 dimer에 해당한다. 도 34h에서, 상대적으로 monomer로존재할가능성이 높은 eGFP와 HER3_eGFP와비교하여 HER2_eGFP는 2-step의 비율이 높게 나왔으므로 HER2-eGFP homodimer의 생성을증명할수 있다.  The fluorescence signal thus obtained is shown in Fig. 34H. In FIG. 34H, a decrease in fluorescence intensity in one step corresponds to a monomer, and a decrease in fluorescence signal in a two-step corresponds to a dimer. In Figure 34h, HER2_eGFP has a high 2-step ratio due to a high ratio of eGFP and HER3_eGFP, which are likely to be present as relatively monomers, thus demonstrating the production of the HER2-eGFP homodimer.
Hidden Markov Model에 기반하여 raw data로부터 가장가능성이 높은 step (recursive calculation이 수렴하는 step)을정하여 분석하였다.  Based on the Hidden Markov Model, the most probable step (recursive calculation convergence step) from raw data was determined and analyzed.
실시예 20. HER2-HER3 heterodimer의 tyrosine kinase의 활성도(activity)측정  Example 20. Measurement of activity of tyrosine kinase of HER2-HER3 heterodimer
면역 침전된 HER2-HER3 heterodimer (실시예 18에서 기판에 처리된 항체에 결합한 HER2-HER3 heterodimer)의 반응 챔버에 ATP와 ¾¾2+를 추가하고 5분동안배양하여, Tyr 인산화를유도하였다. 이후 pTyr-specific antibodies로 single-molecule immunolabel ing하여 HER3 pTyr levels의 변화를 관찰하였다. 상기 시험 과정을 도 35a에 모식적으로 나타내었으며 (1. 세포용해, 2. 기판에 dimer 부착, 3. 부착된 dimer 세척 , 4. ATP와 Mg2+을처리해 인산화, 5. HER3또는 HER2의 인산화 Tyr결합항체 처리, 6. 형광신호검출), 그구체적인시험 방법은다음과같다: ATP and ¾ 2+ were added to the reaction chamber of the immunoprecipitated HER2-HER3 heterodimer (HER2-HER3 heterodimer bound to the substrate treated antibody in Example 18) and incubated for 5 minutes to induce Tyr phosphorylation. Afterwards, changes in HER3 pTyr levels were observed by single-molecule immunolabeling with pTyr-specific antibodies. The test procedure is schematically shown in FIG. 35A (1. cell lysis, 2. dimer attachment to the substrate, 3. dimer washing attached, 4. phosphorylation by treating ATP and Mg2 +, 5. phosphorylation of HER3 or HER2 Antibody treatment, 6. fluorescence signal detection), the specific test method is as follows:
실시예 18의 (2) 조립 및 사용에서, biotin conjugated HER3 antibody를 이용해 설명한 방법을 참조하여, 세포 용해물 (1~4 mg/ml)로부터 HER2-HER3 heterodimer를 표면에 pull-down한 뒤, 0.1% DGTN 또는 0.01% GDN, 50mM HEPES[pH 7.4], 150mM NaCl , 1% glycerol 를 이용해 남은 세포 용해물을 씻어내고, kinase assay용액 (0.1% DGTN또는 0.01¾» GDN, 200uM ATP, 10mM magnesium chloride, 50mM HEPES[pH 7.4] , 150mM NaCl, 1% glycerol)을처리하여 5분동안반응시켰다.  HER2-HER3 heterodimer was pulled down from the cell lysate (1 to 4 mg / ml) onto the surface of the sample using the biotin-conjugated HER3 antibody in Example 2 (2) The remaining cell lysates were washed with DGTN or 0.01% GDN, 50 mM HEPES [pH 7.4], 150 mM NaCl, 1% glycerol and the kinase assay solution (0.1% DGTN or 0.01 Â »GDN, 200 ÂM ATP, 10 mM magnesium chloride, 50 mM HEPES [pH 7.4], 150 mM NaCl, 1% glycerol) and reacted for 5 minutes.
b. 남은 kinase assay 용액을 PBS (또는 0.1% Triton-X-100, 50mM HEPES[pH 7.4], 150mM NaCl)로 씻어내고, HER3 pTyr antibody 용액 (HER3 pTyr antibody (monoclonal rabbit host) (Cell Signal ing Technology, Cat. No. #479 IS, clone:21D3) ~2ug/ml, PBS 또는 0.1% Triton-X-100, 50mM HEPES[pH 7.4], 150mM NaCl)을처리해 5분동안반응시켰다. b. The remaining kinase assay solution was washed with PBS (or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl), and the HER3 pTyr antibody solution (HER3 pTyr antibody (monoclonal rabbit host) Cat. No. # 479 IS, clone: 21D3) to 2 ug / ml, PBS or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) and reacted for 5 minutes.
c. 남은 HER3 pTyr antibody용액을 PBS (또는 0.1% Triton-X-100, 50mM HEPES[pH 7.4] , 150mM NaCl)로씻어내고, cy3 conjugated anti -rabbit FC antibody 용액 (polyclonal goat host ant i -rabbit FC antibody c. The remaining HER3 pTyr antibody solution was washed with PBS (or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) and incubated with cy3 conjugated anti-rabbit FC antibody
(Jackson ImmunoResearch Labs, Cat .No.111-165-046) at 5nM, PBS또는 0.1% Triton-X-100, 50mM HEPES[pH 7.4], 150mM NaCl)을 처리하고, 5분 동안 반응시켰다. (Jackson ImmunoResearch Labs, Cat. No. 11-165-046) at 5 nM, PBS or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) and reacted for 5 minutes.
d. 남은 cy3 conjugated anti -rabbit FC antibody용액을 PBS (또는 0.1% Triton-X-100, 50mM HEPES[pH 7.4], 150mM NaCl)로 씻어내고 Total d. The remaining cy3 conjugated anti-rabbit FC antibody solution was washed with PBS (or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl)
Internal Reflection Microscope을이용해 이미징 및 정량하였다. Imaging and quantification were performed using an Internal Reflection Microscope.
이와 같이 얻어진 HER3 pTyr levels을 도 35b에 나타내었다. HER3 tail의 Tyr residues는총 9개이고, 본실시예에서는 Y1197, Y1222, Y1276, Y1289, 및 Y1328의 총 5개의 Tyr 잔기를 시험하였다. 도 35b에 나타난 바와 같이, pTyr 잔기 모두 ATP와 Mg2+ 배양에 의해 인산화 정도가 증가하였다. The HER3 pTyr levels thus obtained are shown in FIG. 35B. The Tyr residues of the HER3 tail are nine, and in this example a total of five Tyr residues of Y1197, Y1222, Y1276, Y1289, and Y1328 were tested. As shown in FIG. 35B, the degree of phosphorylation was increased by culturing both ATP and Mg2 + in pTyr residues.
실시예 21. HER2-HER2 homodimer의 tyrosine kinase의 활성도 (activity)측정  Example 21. Measurement of activity of tyrosine kinase of HER2-HER2 homodimer
면역 침전된 HER2-HER2 heterodimer (실시예 19에서 기판에 처리된 항체에 결합한 HER2-HER2 homodimer)의 반응 챔버에 ATP와 Mf+를추가하고 5분동안 배양하여, Tyr 인산화를 유도하였다. 이후 pTyr-specif ic antibodies로 single-molecule immunolabel ing하여 HER2 pTyr levels의 변화를관찰하였다. 상기 시험 과정을도 35g에 모식적으로나타내었으며, 그구체적인시험 방법은다음과같다: ATP and Mf + were added to the reaction chamber of the immunoprecipitated HER2-HER2 heterodimer (HER2-HER2 homodimer bound to the substrate treated antibody in Example 19) and incubated for 5 minutes to induce Tyr phosphorylation. Subsequently, changes in HER2 pTyr levels were observed by single-molecule immunolabeling with pTyr-specif ic antibodies. The above test procedure is schematically shown in Figure 35G, and its specific test method is as follows:
a. 실시예 18의 (2) 조립 및 사용에서, biotin conjugated HER2 antibody를 이용해 설명한 방법을 참조하여, 세포 용해물로부터 HER2 homodimer를 표면에 pull-down한 뒤, 0.1% DGTN 또는 0.01% GDN 50mM HEPES[pH 7.4], 150mM NaCl, 1% glycerol 를 이용해 남은 세포 용해물을 씻어내고, kinase assay용액 (0.1% DGTN또는 0.01% GDN, 200uM ATP, lOmM magnesium chloride, 50mM HEPES[pH 7.4] , 150mM NaCl , 1% glycerol ) 을 처리하여 5분동안반응시켰다.  a. HER2 homodimer was pulled down from the cell lysate to the surface using a biotin-conjugated HER2 antibody in the preparation and use of Example 18 (2), and then 0.1% DGTN or 0.01% GDN 50 mM HEPES [pH (0.1% DGTN or 0.01% GDN, 200 uM ATP, lOmM magnesium chloride, 50 mM HEPES [pH 7.4], 150 mM NaCl, 1% glycerol) was used to wash out the remaining cell lysate using a buffer solution glycerol) and reacted for 5 minutes.
b. 남은 kinase assay 용액을 PBS (또는 0.1% Triton-X-100, 50mM HEPES[pH 7.4], 150mM NaCl)로 씻어내고, HER2 pTyr antibody용액 (HER2 pTyr antibody (monoclonal rabbit host) (Cell Signaling Technology, Cat.No.#6942S, clone:D66B7) ~2ug/ml , PBS또는 0.1% Tr iton-X-100, 50mM HEPES[pH 7.4], 150mM NaCl)을처리해 5분동안반응시켰다. b. The remaining kinase assay solution was washed with PBS (or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl), and the HER2 pTyr antibody solution (HER2 (PBS) or 0.1% Tr iton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) in the presence or absence of pTyr antibody (monoclonal rabbit host (Cell Signaling Technology, Cat. No. 6942S, clone: D66B7) Treated and reacted for 5 minutes.
c. 남은 HER2 pTyr antibody용액을 PBS (또는 0.1% Triton-X-100, 50mM HEPES[pH 7.4] , 150mM NaCl)로씻어내고, cy3 conjugated anti -rabbit FC antibody 용액 (polyclonal goat host anti -rabbit FC antibody (Jackson ImmunoResearch Labs , Cat .No.111-165-046) at 5nM, PBS또는 0.1% Triton-X-100, 50mM HEPEStpH 7.4], 150mM NaCl)을 처리하고, 5분 동안 반응시켰다.  c. The remaining HER2 pTyr antibody solution was washed with PBS (or 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) and incubated with a cy3 conjugated anti-rabbit FC antibody solution Treated with 0.1% Triton-X-100, 50 mM HEPEStpH 7.4, 150 mM NaCl) and reacted for 5 minutes.
d. 남은 cy3 conjugated anti -rabbit FC antibody용액을 PBS (또는 d. The remaining cy3 conjugated anti-rabbit FC antibody solution was diluted with PBS (or
0.1% Triton-X-100, 50mM HEPES[pH 7.4], 150mM NaCl) 로 씻어내고 Total Internal Reflection Microscope을이용해 이미징 및정량하였다. Washed with 0.1% Triton-X-100, 50 mM HEPES [pH 7.4], 150 mM NaCl) and imaged and quantified using a Total Internal Reflection Microscope.
이와 같이 얻어진 HER2 pTyr level크을 도 35h에 나타내었다. 도 35h에 나타난 바와 같이, HER2 tail의 Tyr residues Y1139, Y1196, Y1121/1222 및 Y1248의 거의 대부분에서 ATP와 Mg2+ 배양에 의해 인산화가 증가하였다 (인산화증가정도: Y1139의 경우, 대조군 (ATP+EDTA) 대비 약 23배; Y1196의 경우, 대조군 대비 약 13배, Y1121/1222의 경우, 대조군 대비 약 10배, 및 Y1248의 경우, 대조군대비 약 13배). The thus obtained HER2 pTyr level level is shown in Fig. 35H. As shown in FIG. 35h, phosphorylation was increased by ATP and Mg2 + cultivation in almost all of the Tyr residues Y1139, Y1196, Y1121 / 1222 and Y1248 of HER2 tail (degree of phosphorylation increase: Y1139, control (ATP + EDTA ), About 13 times in the case of Y1196, about 10 times in the case of Y1121 / 1222 and about 13 times in the case of Y1248 in the case of Y1121 / 1222).
실시예 22. HER2-HER3 heterodimer의신호전달시험  Example 22. Signal transduction test of HER2-HER3 heterodimer
도 36a에 기판에 부착된 HER2-HER3 heterodimer가 인산화 과정을 거쳤을 때 eGFP 표지된 하위 신호전달 단백질과의 상호작용을 유도한다는 것을보여주는모식적으로나타내었다.  FIG. 36A schematically shows that the HER2-HER3 heterodimer attached to the substrate induces an interaction with the eGFP-labeled lower signal transduction protein when it undergoes a phosphorylation process.
실시예 20또는 21의 방법을참조하여 , HER2-HER3 heterodimer또는 HER2 homodimer의 신호전달정도를측정하였다.  With reference to the method of Example 20 or 21, the degree of signal transduction of the HER2-HER3 heterodimer or HER2 homodimer was measured.
하나의 HER2-HER3 heterodimer에 얼마나 많은 양의 하위 신호전달 단백질 (PLC gamma 1)과상호작용하는지를 형광신호의 크기로측정하여 도 36b에 나타내었다.  Figure 36b shows how much of the HER2-HER3 heterodimer interacts with PLC signaling protein (PLC gamma 1) in terms of the size of the fluorescence signal.
하나의 HER2-HER3 heterodimer에 얼마나 많은 양의 하위 신호전달 단백질 (PLC gamma 1)과상호작용하는지를 형광신호로부터 얻은 단일 분자 꺼짐 현상의 개수로 측정하여 도 36c에 나타내었다 (black: ATP+EDTA, white: ATP+MgCh) .  Figure 36c shows the number of single molecular off events obtained from the fluorescence signal to determine how much of the HER2-HER3 heterodimer interacts with PLC signaling protein (PLC gamma 1) (black: ATP + EDTA, white: ATP + MgCh).
하나의 HER2-HER3 heterodimer에 얼마나 많은 양의 하위 신호전달 단백질 (p85 alpha)과 상호작용하는지를 형광신호의 크기로 측정하여 도 36d에 나타내었다. By measuring the magnitude of the fluorescence signal how much of the HER2-HER3 heterodimer interacts with the amount of the lower signaling protein (p85 alpha) 36d.
하나의 HER2-HER3 heterodimer에 얼마나 많은 양의 하위 신호전달 단백질 (p85 alpha)과 상호작용하는지를 형광신호로부터 얻은 단일 분자 꺼짐 현상의 개수로 측정하여 도 36e에 나타내었다 (black: ATP+EDTA, whi te: ATP+MgCh) .  Figure 36e shows the number of single molecular off events from the fluorescence signal as to how much of the HER2-HER3 heterodimer interacts with the amount of the lower signaling protein (p85 alpha) (black: ATP + EDTA, whi te : ATP + MgCh).
하나의 HER2-HER3 heterodimer에 얼마나 많은 양의 하위 신호전달 단백질 (PI3K : p85 alpha - pllO alpha complex)과 상호작용하는지를 형광신호의 크기로측정하여 도 36f에 나타내었다.  FIG. 36f shows how much of the HER2-HER3 heterodimer interacts with the amount of the lower signaling protein (PI3K: p85 alpha -pllO alpha complex) in terms of the magnitude of the fluorescence signal.
도 36g에 기판에 부착된 HER2 homodimer가인산화과정을거쳤을때 eGFP 표지된 하위 신호전달 단백질과의 상호작용을 유도한다는 것을 보여주는모식적으로나타내었다.  FIG. 36g schematically shows that the HER2 homodimer attached to the substrate induces an interaction with the eGFP-labeled lower signal transduction protein when oxidized.
하나의 HER2 homodimer에 얼마나 많은 양의 하위 신호전달 단백질 How much amount of the lower signaling protein in one HER2 homodimer
(PLC gamma 1)과상호작용하는지를형광신호의 크기로측정하여 도 36h는에 나타내었다. (PLC gamma 1) was measured by the magnitude of the fluorescence signal, and FIG. 36h shows the result.
하나의 HER2 homodimer에 얼마나 많은 양의 하위 신호전달 단백질 How much amount of the lower signaling protein in one HER2 homodimer
(PLC gamma 1)과 상호작용하는지를 형광신호로부터 얻은 단일 분자 꺼짐 현상의 개수로 측정하여 도 36i에 나타내었다 (bl ack: ATP+EDTA, whi te: ATP+MgCl2) . (ATP + EDTA, whi te: ATP + MgCl 2) as measured by the number of single molecular off events obtained from the fluorescence signal .
하나의 HER2 homodimer에 얼마나 많은 양의 하위 신호전달 단백질 (p85 alpha)과 상호작용하는지를 형광신호의 크기로 측정하여 도 36j에 나타내었다.  Figure 36j shows how much of the HER2 homodimer interacts with the amount of the lower signaling protein (p85 alpha) in terms of the size of the fluorescence signal.
하나의 HER2 homodimer에 얼마나 많은 양의 하위 신호전달 단백질 How much amount of the lower signaling protein in one HER2 homodimer
(p85 alpha)과 상호작용하는지를 형광신호로부터 얻은 단일 분자 꺼짐 현상의 개수로 측정하여 도 36k에 나타내었다 (black: ATP+EDTA, whi te: ATP+MgCl2) . (p85 alpha), as shown in Figure 36k (black: ATP + EDTA, whi te: ATP + MgCl 2) as measured by the number of single molecular off events from the fluorescence signal.
실시예 23. HER2-HER3 heterodimer와 HER2-HER2 homodimer의 인산화 속도비교  Example 23. Comparison of phosphorylation rate between HER2-HER3 heterodimer and HER2-HER2 homodimer
HER2-HER3 heterodimer를 사용한 실시예 20의 방법을 참조하여, 시간에 따른 HER3 Y1289의 인산화정도를 측정하여, 도 37a에 나타내었다. HER2-HER3 heterodimer가 세포의 prol i ferat ion을 강하게 유도한다는 것은 알려져 있었지만 효소적 특성은 알려지지 않았다. 도 37a는 HER2-HER3 heterodimer의 효소적 성능을 측정한 것이라고 할 수 있다 (인산화에 사용되는 연료인 의 특정한 농도에서 HER2-HER3 heterodimer가 얼마나 2019/132517 1»(:1^1{2018/016675 Referring to the method of Example 20 using the HER2-HER3 heterodimer, the degree of phosphorylation of HER3 Y1289 over time was measured and shown in Figure 37A. The HER2-HER3 heterodimer was known to strongly induce the prolipid ion of the cell, but the enzymatic character was unknown. Figure 37A is a measure of the enzymatic performance of the HER2-HER3 heterodimer (the HER2-HER3 heterodimer at a specific concentration of phosphorus used for phosphorylation 2019/132517 1 »(: 1 ^ {2018/016675
빨리 인산화를진행하는지 보여줌). 도 373에서 X축은 인산화에 걸린시간, V축은해당시간동안인산화 양을나타낸다. Indicating rapid phosphorylation). FIG time from 3 to 37 X-axis phosphorylation, V-axis represents the amount of phosphorylated during that time.
또한, 抑요2 -四묘3110(1 61'를사용한실시예 20의 방법을참조하여, ? 농도를 증가시키면서 }世1?3 잔기의 인산화 속도를 측정하였다. 이는 !3의 농도에 따라 11 610(1^61·가인산화하는속도가어떻게 변하는지 알아보기 위한 것이다. 다양한 꾸 농도에서 인산화 속도를 측정하면 나타내는 척도인 ¾(마이캘리스 상수)과
Figure imgf000123_0001
(최대 측정할 수 있다. ¾은 인산화 속도가 최대 인산화 속도의
Figure imgf000123_0002
농도이며, ■에 해당하는 ^TP 농도에서 인산화 속도를 측정하면 최대 인산화 속도의 절반이 얻어지고,
Figure imgf000123_0003
최대 인산화 속도로 11336가도달할수있는최대 인산화속도이다.
Also, with reference to the method of Example 20 using the constraint 2 - 4 31 1 0 (1 61 ' ,? And the phosphorylation rate of the first and third residues was measured while increasing the concentration. this is ! 3 , the rate of oxidation of 1 6110 (1 ^ 61 · g) is changed. When the rate of phosphorylation was measured at various concentrations, the ¾ (Mycalis constant) and
Figure imgf000123_0001
(You can measure up to. And the phosphorylation rate of ¾ is the maximum phosphorylation rate
Figure imgf000123_0002
Concentration, and measuring the phosphorylation rate at the ^ TP concentration corresponding to < RTI ID = 0.0 > (I) < / RTI >
Figure imgf000123_0003
It is the maximum phosphorylation rate that can reach 11336 at the maximum phosphorylation rate.
상기 얻어진 결과를 도 371)에 나타내었다 (점선은 붉은 선이 수렴하는 값을나타냄). 도 371)의 데이터는주로 1?3의 1289를측정한 것으로, 다른 (1197, 1222, 1276, 1328)을 측정했을 때에도 비슷한 수준의 인산화 속도가 측정되는 것으로 보아 측정된 값이 특정
Figure imgf000123_0004
해당하는 것이 아님을 알 수 있다. 도 371)에서와 같이,
Figure imgf000123_0005
농도가 증가함에 따라서, 四묘3
Figure imgf000123_0006
잔기의 인산화속도도증가함을확인할수 있다. 비교를 위하여, 四묘2 -四요21101110(1 161·를 사용한 실시예 21의 과정을 참조하여 , ?농도를증가시키면서 四요2 打 잔기 (凡196)의 언산화속도, ¾,
Figure imgf000123_0007
측정하여, 도 37(:에 나타내었다.
The obtained results are shown in Figure 371 (the dotted line indicates the convergence value of the red line). 371) is a measurement of 1289 of 1 to 3, and a similar level of phosphorylation rate is measured when the other (1197, 1222, 1276, 1328) is measured.
Figure imgf000123_0004
It can be seen that this does not apply. 371)
Figure imgf000123_0005
As the concentration increases,
Figure imgf000123_0006
And the phosphorylation rate of the residue is also increased. For comparison, two seedlings四-四to refer to the process of John 21 101 110 (1 in Example 21 with 161, while increasing the concentration required四frozen oxidation rate for the second打residue (凡196), ¾,
Figure imgf000123_0007
The measurement is shown in FIG. 37 (:).
도 3개 및 37(:의 비교로부터 알 수 있는 바와 같이, }恨1?2-
Figure imgf000123_0008
As can be seen from the comparison of Figures 3 and 37 (:),
Figure imgf000123_0008
,
Figure imgf000123_0009
100배 이상 빠르다는 것을 의미한다. 반면, 四묘2 -四묘3 느 근대 !"에서의 인산화의 ¾1은 1?2-]101110 11161'와 비교하여 2배 이하의
Figure imgf000123_0009
It means that it is 100 times faster. On the other hand,四two graves? - by ¾ 1 phosphorylation in the grave three四slow modernity "is compared with the first 2] 11 161 101 110, more than twice
Figure imgf000123_0012
Figure imgf000123_0012
[世1?2억제제인 18에서 기판에 부착된
Figure imgf000123_0010
The substrate is attached to the substrate at the inhibitor 18
Figure imgf000123_0010
11 61'0 11161·와 실시예
Figure imgf000123_0011
부착된 }世묘2 1101110 111아 에 각각 처리한후, 실시예 20및 21의 과정을 참조하여, (1001虛)와 163 1111 (±10:^(16(101 )를 처리하고 (이 때에도 0끄은 0.1%로, 0.01%로 유지해야함), 각 HER2 dimer들의 활성화정도를측정하였다. 상기 얻어진 결과를 도 37d에 나타내었다. 도 37d에서, x축은 사용한 Lapat inib의 농도를 나타내고, y축은 표준화된 인산화 정도를 나타낸다. HER2-HER3 heterodimer의 활성화는 HER3 pY1289 항체를 이용해 측정한 것으로 Lapat inib이 없을 때를 100%로 하여 표준화하였고, HER2 homodimer의 활성화는 HER2 pY1196 항체를 이용하여 측정한 것으로 Lapat inib이 없을 때를 100%로 하여 표준화한 값이다. 도 37d에 나타난 바와 같이,
1 1 61'0 11161 · Example
Figure imgf000123_0011
Attaching a}世seedlings 21 101 110, each processing a 111 Ah after Example 20 and with reference to the 21 courses, (100 1虛) and 1卵63 1111 (± 1 0 : ^ (1 6 (10 1) (At this time, 0.1%, 0% and 0.01% And the degree of activation of each HER2 dimer was measured. The obtained results are shown in Fig. 37D. In Figure 37d, the x-axis represents the concentration of used Lapat inib and the y-axis represents the normalized degree of phosphorylation. Activation of the HER2-HER3 heterodimer was measured using HER3 pY1289 antibody, which was normalized to 100% in the absence of Lapat inib. Activation of the HER2 homodimer was measured using HER2 pY1196 antibody. 100% And the standardized value. 37D,
Lapat inib은 Lapat inib에 저항성이 없는 HER2 -homodimer와 HER2-HER3 heterodimer에 유사한저해곡선을보이는것으로확인돠었다. Lapat inib has been shown to exhibit similar inhibition curves to HER2-HOM3 and HER2-HER3 heterodimers that are not resistant to Lapat inib.
실시예 18에서 기판에 부착된 HER2-HER3 heterodimer와 실시예 The HER2-HER3 heterodimer attached to the substrate in Example 18 and the Example
19에서 기판에 부착된 HER2 homodimer에, Lapat inib, ATP (lOOuM) , 및 magnesium chlor ide(10mM)를 함께 처리하고(이 때에도 DGTN은 0.1%로, GDN은 0.0B로 유지해야 함), HER2의 활성화 정도를 측정하여, 그 결과를 도 37e에 나타내었다. 도 37e에서와 같이, HER2 homodimer와 비교하여, HER2-HER3 heterodimer의 경우, HER2 homodimer와 비교하여, Lapat inib에 의한 저해 효과가 낮게 나타났다. 이러한 결과는 HER2-HER3 heterodimer 발현 세포가 ATP와 magnesium 존재 하에서 (Tyr 인산화 가능한 경우)19, the HER2 homodimer attached to the substrate was treated together with Lapat inib, ATP (lOOuM), and magnesium chloride ide (10 mM) (at this time DGTN should be kept at 0.1% and GDN at 0.0B) The degree of activation was measured. The result is shown in Fig. 37E. As shown in Figure 37 (e), the HER2-HER3 heterodimer had a lower inhibitory effect of Lapat inib compared to the HER2 homodimer, as compared to the HER2 homodimer. These results suggest that HER2-HER3 heterodimer-expressing cells can be activated by ATP and magnesium (when Tyr is phosphorylated)
Lapat inib에 대하여 저항성을 가짐을 보여준다. 앞서 HER2-HER3 heterodimer가 HER2 -homodimer와 비교하여 인산화 속도가 현저히 빠르다는 것이 확인되었으므로, 이러한 인산화 속도가 저해제 (Lapat inib)에 대한 저항성과 관련이 있다고 할 수 있다 (즉, 인산화 속도가 빠를수록 저해제 저항성이 잘나타난다고볼수있음) . Lt; RTI ID = 0.0 > Lapat inib. ≪ / RTI > It has been previously shown that the HER2-HER3 heterodimer is significantly faster than the HER2-homodimer in phosphorylation rate, so this phosphorylation rate may be related to the resistance to the inhibitor (Lapat inib) (ie, Resistance can be seen as well).
인산화 속도와 저해제 저항성 간의 상관 관계를 보다 명확하게 확인하기 위하여, HER2-HER3 heterodimer 발현 세포 용해액에, Lapat inib, ATP (lOOuM), 및 magnesium chlor ide(10mM)와 함께(이 때에도 DGTN은 HER2-HER3 heterodimer expressing cell lysates were incubated with Lapat inib, ATP (lOOuM), and magnesium chlor ide (10 mM) (again, DGTN was added to the HER2-HER3 heterodimer expressing cell lysate to confirm the correlation between phosphorylation rate and inhibitor resistance
0.1%로, GDN은 0.01%로 유지해야 함), PTPN1 (Protein Tyrosine Phosphatase, Non-receptor type 1; ProSpecbio, Cat .No. PKA-219)을 처리하고, HER3 Y1289의 인산화 수준을 측정하여, 도 37f에 나타내었다. PTPN1는 탈인산화 효소로서, Tyr 인산화와 탈인산화 과정 사이의 평형을 형성하고, HER2-HER3 heterodimer를 구조으로는 변화시키지 않으면서 실질적 인산화 속도를 효과적으로 늦추는 역할을 한다. 도 37f에 나타난 바와 같이, PTPN1의 처리 농도가 높아짐에 따라 (도 37f에서 붉은색 그래프(가장 오른쪽)에서 보라색 그래프(가장 왼쪽)로 갈수록), 실질적인 인산화속도가느려지며, Lapatinib에 의한인산화저해 효과는증가담 (즉, 더 낮은농도의 Lapatinib에도인산화가저해 됨)을확인할수있다. 0.1%, GDN should be maintained at 0.01%), PTPN1 (Protein Tyrosine Phosphatase, Non-receptor type 1; ProSpecbio, Cat. PKA-219) was treated and the phosphorylation level of HER3 Y1289 was measured and is shown in Figure 37f. PTPN1 is a dephosphorylation enzyme that forms an equilibrium between Tyr phosphorylation and dephosphorylation and effectively slows down the rate of substantial phosphorylation without changing the HER2-HER3 heterodimer into its structure. As shown in FIG. 37F, as the treatment concentration of PTPN1 increases (from the red graph (rightmost) to the purple graph (leftmost) in FIG. 37F) Phosphorylation is slowed down and the inhibition of phosphorylation by lapatinib is increased (ie, phosphorylation is inhibited at lower concentrations of lapatinib).
실시예 25. 이종이식동물모델및 인간환자시료에 대한단일분자 면역 표지 (Single-molecule immunolabelin) 및 co-IP 프로파일링 (co-IP profiling)  Example 25. Single-molecule immunolabelin and co-IP profiling for xenograft models and human patient samples.
모든 동물 연구는 Institutional Animal Care and Use Committee (IA⑶ C)으로부터 승인받은 지침에 따라 수행하였다. 6 내지 8주령의 암컷 마우스 (severe combined immunodef icient (NOG) and nude (nu/nu) mice; OrientBio)를사용하였다. 임상종양샘플 (폐선암종 (lung adenocarcinoma) 환자유래)을 ~3 nun3크기의 조각으로절단한후, 상기 N0G마우스의 옆구리 내부로피하 이식 (subcutaneous implantation) 하였다. 이식 투 1~4개월 경과후, 이식된 부위에서 종양이 관찰되었다. caliper로 주 2회 종양의 체적을 측정하여 종양의 피하 성장률을 측정하였다. 종양 크기가 직경 1.5cm에 도달하였을 때, 종양 조직을 절제하고 작은 절편 (대략 5 mm3 크기)으로 절단하였다. 상기 절단된 조직을 다른 마우스 집단에 재이식하여 속발성 종양 (subsequent tumors)을 얻었다. 환자유래 종양을 갖는마우스세대를 F0이라명명하고, 이후의 후속세대는차례대로번호를 매겼다 (FI, F2, F3 등). 3세대 (F3) 마우스를 vehicle (Phosphata buffered saline, PBS) , osimert inib, 또는 gefitinib 처리 시험에 사용하였다 (PDTX-A1). All animal studies were conducted in accordance with guidelines approved by the Institutional Animal Care and Use Committee (IA, C). (NOG) and nude (nu / nu) mice (OrientBio) at 6 to 8 weeks of age were used. Clinical tumor samples (derived from lung adenocarcinoma patients) were cut into ~ 3 nun 3 size pieces and subcutaneously implanted into the lateral side of the N0G mice. After 1 to 4 months of transplantation, the tumor was observed at the transplanted site. The volume of the tumor was measured twice weekly with caliper to determine the subcutaneous growth rate of the tumor. When the tumor size reached 1.5 cm in diameter, tumor tissue was excised and cut into small sections (approximately 5 mm 3 size). The severed tissue was re-implanted in another group of mice to obtain subsequent tumors. Mouse generations with patient-derived tumors were named F0, and subsequent generations were numbered sequentially (FI, F2, F3, etc.). A third generation (F3) mouse was used in the vehicle (Phosphata buffered saline, PBS), osimert inib, or gefitinib treatment test (PDTX-A1).
Patient PI은진행성 질환 (progressive disease; PD) 진단전에 약 1년 반동안 gefitinib치료에 대한부분적 반응성 (partial response; PR; 고형 종양에서의 반응성 평가 기준 (response evaluation criteria in solid tumors; RECIST) i 따른 부분적 반응성)을 유지한 반면, Patient P2는 PD designation 전에 1년 동안 stable disease (SD) diagnosis를 유지하였다.  Patient PI was assessed for partial response (PR) to gefitinib therapy for about a year and a half before diagnosis of progressive disease (PD). Partial response to the response evaluation criteria in solid tumors (RECIST) While Patient P2 maintained a stable disease (SD) diagnosis for one year before PD designation.
상기 PDTX-A1 및 인간 환자 샘플 P1 (진행성 질환 (progressive disease; PD) 진단 전에 약 1년 반 동안 gefitinib 치료에 대한 부분적 반응성 (partial response; PR; 고형 종양에서의 반응성 평가 기준 (response evaluation criteria in sol id tumors; RECIST)에 따른 부분적 반응성)을유지하는폐선암종환자유래 암조직 시료; EGFR genotype : exon 19)로부터 유래된 mutant EGFR 복합체에 대하여 in vitro 탈인산화를 수행하였다 (도 38). 탈인산화 후, eGFP- labeled PLC-gammasH2 및 Grb2와, mutant EGFR 복합체와의 결합을 측정하였다. 탈인산화에 의하여, pTyr-SH2 도메인 상호작용에 전적으로 의존하는 PLC-gammaSH2의 결합은 거의 완전하게 중지되는 반면, Grb2 binding counts의 50%이상 및 80%이상은 탈인산화 후에도 유지되었다. 이렇나 결과는 mutant EGFR의 pTyr- independent signaling mechanism이 PDTX모델과외과적 종양조직에서 실제로작동함을 제안한다. The PDTX-A1 and the human patient sample P1 (partial response: PR; response evaluation criteria in sol (PD) for the treatment of gefitinib for about a year and a half before the diagnosis of progressive disease In vitro dephosphorylation was performed on mutant EGFR complexes derived from EGFR genotype: exon 19) (Fig. 38). After dephosphorylation, the binding of eGFP-labeled PLC-gammasH2 and Grb2 to the mutant EGFR complex was measured . By dephosphorylation, binding of PLC-gamma SH2 entirely dependent on pTyr-SH2 domain interaction was almost completely stopped, while over 50% and over 80% of Grb2 binding counts were maintained after dephosphorylation. These results suggest that the pTyr-independent signaling mechanism of the mutant EGFR actually works in extracorporeal tumor tissue of the PDTX model.
실시예 26. EGFR돌연변이의 Grb2와의상호작용에 대한영향시험 돌연변이된 EGFRs가 Grb2와 상호작용하는데 티로신키나아제 활성을 필요로 하는지 여부를 시험하기 위하여, EGFR에 특이적인 TKI 인 게피티닙을 24시간동안 EGFR돌연변이(pTyr 1068및 1086)를갖는 4종의 세포 (PC9, HCC827, HCC1006, H1975)에 처리하였다. 그 후, 게피티닙 처리된 세포를 용해시키고, EGFR ᅵ11111111110-1) (^1)1131^011을 수행하였다. 게피티닙에 의한 EGFR 티로신키나아제 저해는 엑손 19결실 (gatekeeper mutations은 갖지 않음)을 갖는 3개의 세포에 대한 Grb2 결합을 거의 완전히 억제하였다(도 39a). 일부 residual EGFR-Grb2 PPI count는 L858R 및 T790M gatekeeper 돌연변이를 갖는 H1975세포에서 관찰되었다. 또한, osimert inib (gatekeeper mutations을 갖는 EGFR에 똑같이 잘 결합하는 3 세대 TKI)를 처리함으로써 H1975세포에서 Grb2결합을 완전히 억제함을 확인했다(도 39a,파란색 점). Example 26. Effect of EGFR mutation on interaction with Grb2 To test whether mutated EGFRs require tyrosine kinase activity in order to interact with Grb2, gefitinib, a TKI specific for EGFR, was incubated for 24 hours Were treated with four cells (PC9, HCC827, HCC1006, H1975) with EGFR mutations (pTyr 1068 and 1086). Then, it was dissolved in a nip Pitti treated cells, EGFR it was performed 11111111110-1) (^ 1) 11 31 ^ 011. EGFR tyrosine kinase inhibition by gefitinib almost completely inhibited Grb2 binding to three cells with exon 19 deletion (no gatekeeper mutations) (Fig. 39A). Some residual EGFR-Grb2 PPI counts were observed in H1975 cells with L858R and T790M gatekeeper mutations. It was also confirmed to completely inhibit Grb2 binding in H1975 cells by treating osimert inib (third generation TKI that equally well binds EGFR with gatekeeper mutations) (Fig. 39a, blue dot).
앞서 살펴본 바와 같이, TKI 처리로 인해 Grb2와 돌연변이 EGFR 사이의 결합이 완전히 억제되었다.이러한현상을보다더 관찰하기 위하여, 우리는 single-molecule immunolabel 1 ing scheme을 사용하여 EGFR의 다른 알려진 상호 작용 파트너의 co-IP를시험하여 ,그 결과를그림 도 39b_g에 나타내었다.  As previously noted, the binding between Grb2 and mutant EGFR was completely inhibited by TKI treatment. To further observe this phenomenon, we used a single-molecule immunolabel 1 ing scheme to identify other known interacting partners of EGFR Co-IP was tested and the results are shown in Figure 39b_g.
우선, 내인성 HSP90(알파 isoform)의 co-IP 수준을 정량화하였다. HSP90의 association은돌연변이 EGFR에 대해 선택적으로관찰되었으며 (도 39b; 청색 대 흑색 막대), 이러한 HSP90 association은 시험관내 탈인산화 처리에 내성을갖는것으로나타났다(도 39b; 흑색 대 붉은막대).그러나, 24시간동안 TKI를 처리함으로써 돌연변이 EGFR에서 HSP90을 거의 완전히 분리시킬 수 있었다 (도 39b; 검은 색 대 노란색 바). 게피티닙 처리 24 시간 후 돌연변이 EGFR과 관련된 GAPDH가 더 많았지만, MIG6(천연 EGFR 억제제) 및 GAPDH35에 대하여 유사한 패턴을 관찰하였다 (도 39c, d). 대조적으로, Shbl (Grb2 결합 20에 대한 잘 알려진 지지체)은 WT 및 돌연변이 EGFR(도 39e)에 걸쳐 유사한수준의 결합을나타냈다. First, the co-IP level of endogenous HSP90 (alpha isoform) was quantitated. The association of HSP90 was selectively observed with mutant EGFR (Figure 39b; blue-black bars), and this HSP90 association was shown to be resistant to in vitro dephosphorylation (Figure 39b, black versus red bars) (Fig. 39b; black versus yellow bars) by treating TKI for a period of time. A similar pattern was observed for MIG6 (natural EGFR inhibitor) and GAPDH35 (Figure 39c, d), although GAPDH associated with mutant EGFR was more abundant after 24 hours of gefitinib treatment. In contrast, Shbl (a well known support for Grb2 binding 20) exhibited similar levels of binding across WT and mutant EGFR (Figure 39e).
시험관내 탈인산화후 Shcl과 WT EGFR의 상호 작용이 감소되었지만, 돌연변이 EGFR과의 결합은 HSP90, MIG6 및 GAPDH에서 관찰된 것과 유사한 정도의 시험 관내 탈인산화에 대한 저항성을 보였다. 또한, 24 시간의 게피티닙 처리로 인하여 Shcl과 WT 및 돌연변이 EGFR과의 연관성(associ at ion)이 안정화되거나 심지어 증가하였다. 이러한 결과는 Shcl이 돌연변이 EGFR과선택적으로 연관된 다른단백질과다르게 행동함을 나타낸다. 마지막으로, single molecule immunolabel 1 ing technique을 사용하여, TKI 치료가 전체 EGFR수준에 영향을 미치지 않음을 확인하였다 (도 39f).  Although the interaction of Shcl and WT EGFR was reduced after in vitro dephosphorylation, the binding to the mutant EGFR showed resistance to in vitro dephosphorylation similar to that observed in HSP90, MIG6 and GAPDH. In addition, the associat ion of Shcl, WT and mutant EGFR was stabilized or even increased due to 24 hours of gefitinib treatment. These results indicate that Shcl behaves differently from other proteins selectively associated with the mutant EGFR. Finally, using the single molecule immunolabel 1 ing technique, it was found that TKI treatment did not affect the total EGFR level (Figure 39f).
상기 결과는, WT EGFR에 대한 Grb2의 결합은 일차적으로 표준 경로(canonical pathway)에 의해, 즉 pTyr 1068및 1086과 Grb2 SH2도메인 사이의 상호작용을통해, 매개된다는것을제안한다.  The results suggest that the binding of Grb2 to WT EGFR is primarily mediated by the canonical pathway, i.e. through the interaction between pTyr 1068 and 1086 and the Grb2 SH2 domain.
이와 대조적으로, 돌연변이 EGFR은 표면 노출 pTyr 잔기에 최소한 의존하는 근본적으로 다른 메카니즘을 이용한다. 돌연변이 EGFRs가 WT EGFR보다더 큰신호 전달복합체를 형성한다는 것을관찰하였다. 이러한 신호 전달복합체의 일부구성 요소는 Grb2에 대하여 pTyr 독립적인(pTyr- independent) 방식으로 인력을작용시킨다. 이러한비공식(non-canonical) 경로는 인산가수분해효소의 탈인산화 작용에 대하여 큰 저항성을 가지며 In contrast, mutant EGFR utilizes fundamentally different mechanisms that are least dependent on surface exposed pTyr residues. We observed that mutant EGFRs form a larger signaling complex than WT EGFR. Some components of these signaling complexes engage GrB2 in a pTyr-independent manner. This non-canonical pathway is highly resistant to dephosphorylation of phosphatase
EGFR 돌연변이가 있는 암이 자극성 리간드가 없을 때 EGFR 신호 전달을 유지하는 방법을 설명할 수 있다. 또한, 돌연변이 EGFR과 c-Cbl (및 Cbl- b)의 co-IP수준이 WT EGFR보다현저하게 낮다는 것을 발견했다 (도 39g). 이러한 결과는 돌연변이 EGFR에 특유의 신호 전달 복합체가 EGFR이 분해되는과정을방해한다는것을시사한다. It is possible to explain how cancer with an EGFR mutation maintains EGFR signaling in the absence of a stimulatory ligand. In addition, we found that co-IP levels of mutant EGFR and c-Cbl (and Cbl-b) were significantly lower than WT EGFR (Figure 39g). These results suggest that a signaling complex unique to the mutant EGFR hinders the degradation of EGFR.
도 39a~g의 결과를 보면, EGFR로 침강하였을 때, The results in Figures 39a-g show that when sedimented with EGFR,
PC9/HCC827/HCC4006의 경우 HSP90a가 EGFR과함께 유난히 많이 나오는것을 볼 수 있다. 이는 해당 세포들이 EGFR 신호전달에 의존하는 동시에, HSP90a라는 accessory단백질도 동시에 사용함을 제안한다. 그러므로 EGFR 억제제와 HSP90a를 동시에 사용하거나, 혹은 HSP90a 억제제를 단독으로 사용시에도 어느 정도 소망하는 효과를 얻을 수 있을 것으로 기대된다. 반면 EGFR이 정상형태인 H1666+세포주에서는전혀 작동하지 않음도확인할 수있다. 2019/132517 1»(:1/10公018/016675 In the case of PC9 / HCC827 / HCC4006, it can be seen that HSP90a is remarkably abundant with EGFR. This suggests that the cells depend on EGFR signaling as well as accessory proteins called HSP90a. Therefore, it is expected that ESPR inhibitor and HSP90a may be used at the same time, or the desired effect may be obtained to some extent even when the HSP90a inhibitor alone is used. On the other hand, it can be confirmed that EGFR does not work at all in the normal H1666 + cell line. 2019/132517 1 »(: 1/10/06 018/016675
Figure imgf000128_0001
Figure imgf000128_0001

Claims

2019/132517 1»(:1^1{2018/016675 【특허청구범위】 【청구항 1] (I )시험 시료에 후보화합물을처리하는단계, 및 (I I ) 상기 후보 화합물이 처리된 시험 시료 및 처리되지 않은 시험 시료각각에 대하여 하기의 단계 (1) , (2) , (3) , 및 (5) , 또는 (1) , (2) ,(3) , (4) , 및 (5) , 또는 (1) , (2) , (3) , (4-1) , 및 (5) , 또는 (1), (2), (3), (4-1), (4-2) , 및 (5)를 수행하는 단계를 포함하는, 제 1 단백질을 표적으로하는후보약물의 선별방법: (I) treating a candidate compound with a test sample, and (II) contacting the candidate compound with the treated test sample and the treated sample. (1), (2), (3) and (5), or (1), (2), (3), (4) and (1), (2), (3), (4-1), and (5) 5). ≪ / RTI > A method of screening candidate drugs targeting a first protein comprising:
(1) 제 1 단백질을 포함하는 시험 시료를 표면에 상기 제 1 단백질에 특이적으로 결합하는 물질을 포함하는 기판에 가하여 제 1 단백질이 고정된기판을준비하는단계;  (1) preparing a substrate to which a first protein is immobilized by adding a test sample containing a first protein to a substrate including a substance that specifically binds to the first protein on a surface thereof;
(2) 상기 준비된 제 1 단백질이 고정된 기판에 표지된 제 2 단백질을 첨가하여 반응시키는단계;  (2) adding a labeled second protein to the substrate on which the prepared first protein is immobilized and reacting;
(3) 단계 (2)에서 얻어진 반응물로부터 신호를 측정하여 단백질- 단백질상호작용을측정하는단계 ;  (3) measuring a signal from the reactant obtained in step (2) to measure protein-protein interaction;
(4)단계 (3)에서 측정된신호를이용하여 단계 (1)에서 첨가한시험 시료 포함된 제 1 단백질의 단위량에 대한 값을 구하여 활성화 수준을 측정하는단계;  (4) measuring an activation level by obtaining a value for a unit amount of the first protein included in the test sample added in step (1) using the signal measured in step (3);
(4-1) 단계 (3)에서 측정된 신호를 이용하여 단계 (1)에서 첨가한 시험 시료의 단위량에 대한 신호값을 구하여 단백질-단백질 상호작용 수준을측정하는단계;  (4-1) measuring a protein-protein interaction level by obtaining a signal value for a unit amount of the test sample added in step (1) using the signal measured in step (3);
(4-2) 단계 (4-1)에서 얻어진 시험 시료의 단위량에 대한 신호값을 이용하여 시험 시료에 포함된 제 단백질의 단위량에 대한 값을 구하여 활성화수준을측정하는단계 ;  (4-2) measuring the activation level by obtaining a value for the unit amount of the protein contained in the test sample using the signal value of the unit amount of the test sample obtained in the step (4-1);
(5) 상기 후보 화합물이 처리되지 않은 시험 시료 및 처리된 시험 시료에 대하여 상기 단계 (3), 단계 (4), 단계 (4-1) , 또는 단계 (4- 2)에서 얻어진각각의 결과를서로비교하는단계.  (5) The respective results obtained in step (3), step (4), step (4-1), or step (4- 2) for the test sample not treated with the candidate compound and the treated test sample Comparing them to each other.
【청구항 2]  [Claim 2]
제 1항에 있어서, 상기 단계 (5) 이후에  The method of claim 1, wherein after step (5)
(6) 상기 단계 (5)에서의 비교 결과, 후보 화합물이 처리된 시험 시료의 단백질-단백질 상호작용, 단백질-단백질 상호작용 수준, 또는 활성화 수준이 후보 화합물이 처리되지 않은 시험 시료에서보다 낮은 경우, 상기 후보화합물을상기 제 1단백질을표적으로하는후보약물로 2019/132517 1»(:1^1{2018/016675 (6) As a result of the comparison in the above step (5), when the protein-protein interaction, the protein-protein interaction level, or the activation level of the test sample treated with the candidate compound is lower than that of the test sample in which the candidate compound is not treated , The candidate compound is selected as a candidate drug targeting the first protein 2019/132517 1 »(: 1 ^ {2018/016675
선택하는단계를추가로포함하는, 방법 . The method further comprising the step of selecting.
【청구항 3】  [Claim 3]
제 1항또는제 2항에 있어서, 상기 단계 (1), (2), (3), 및 (5), 또는 (1) , (2) , (3) , (4) , 및 (5) , 또는 (1) , (2) , (3) , (4-1) , 및 (5) , 또는 5 (1), (2), (3), (4-1), (4-2), 및 (5)는,  3. The method of claim 1 or 2, wherein the steps (1), (2), (3) and (5), or (1), (2), (3), (4) (1), (2), (3), (4-1), and (5) , And (5)
(3)수용체 (제 1단백질)및 리간드 (제 2단백질)간의 상호작용,( 3) the interaction between the receptor (first protein) and the ligand (second protein)
( 1단백질 및 제 2단백질 중에서 하나이상이 인산화된타이로신 잔기를갖는경우의 제 1단백질및 제 2단백질간의 상호작용, 및 (Interaction between a first protein and a second protein when at least one of the first protein and the second protein has a phosphorylated tyrosine residue, and
(0)제 1단백질및상기 제 1단백질이 관여하는신호전달경로중에서 10 제 1단백질의 하위 단백질인제 2단백질간의 상호작용 (0) the first protein and the interaction between the lower protein ingest 2 protein of the 10 proteins in the first transmission path to engage the first protein
중에서 선택된하나이상에 대하여 수행되는것인, 방법. ≪ / RTI >
【청구항 4] [4]
제 1항또는제 2항에 있어서, 상기 단계 (1), (2) , (3), 및 (5), 또는 (1), (2), (3), (4), 및 (5) , 또는 (1) , (2) , (3) , (4-1) , 및 (5), 또는 15 (1), (2), (3), (4-1), (4-2), 및 (5)는, 3. The method of claim 1 or 2, wherein the steps (1), (2), (3) and (5), or (1), (2), (3), (4) , or (1), (2), (3), (4-1), and (5), or 15 (1), (2), (3), (4-1), (4-2) , And (5)
(3)수용체 (제 1단백질)및 리간드 (제 2단백질)간의 상호작용,( 3) the interaction between the receptor (first protein) and the ligand (second protein)
( 1단백질 및 제 2단백질 중에서 하나이상이 인산화된타이로신 잔기를갖는경우의 제 1단백질및제 2단백질간의 상호작용, 및 (Interaction between a first protein and a second protein when at least one of the first protein and the second protein has a phosphorylated tyrosine residue, and
(0)제 1단백질및상기 제 1단백질이 관여하는신호전달경로중에서 20 제 1단백질의 하위 단백질인제 2단백질간의 상호작용 (0) the first protein and the interaction between the lower protein ingest 2 protein of the 20 proteins in the first transmission path to engage the first protein
중에서 선택된두개 이상에 대하여 수행되는것인, 방법. ≪ / RTI >
【청구항 5] [Claim 5]
저 항또는제 2항에 있어서 , 상기 제 1단백질을표적으로하는후보 약물은 제 1 단백질의 활성 저해제, 제 1 단백질과관련된 질병의 치료제, 25 및 제 1 단백질과 제 2 단백질 간 상호작용 저해제로 이루어진 군에서 선택된 1종이상인, 방법.  4. The method according to claim 2, wherein the candidate drug targeting the first protein is selected from the group consisting of an activity inhibitor of the first protein, a therapeutic agent for a disease associated with the first protein, and an inhibitor of interaction between the first protein and the second protein Wherein one selected from the group consisting of.
【청구항 6]  [Claim 6]
下1)-1그이 부착된 기판에 표지물질이 결합된 卵 -1을 공급하여 반응시키는단계; 및  1) -1: supplying egg-1 bound with a labeling substance to the substrate to which the labeling substance is bound and reacting; And
30 상기 반응시키는단계에서 발생한신호 (쇼)를측정하는단계 30 Measuring the signal (show) generated in the reacting step
를포함하고,  Lt; / RTI >
상기 1)-1그과卵 -1의 반응물은클러스터를형성하는것인, 므1)-1그과 1)-1의 상호작용측정 방법 . Wherein the reactants of 1) -1 and egg-1 form clusters, and 1) a method of measuring the interaction of 1) and 1) -1.
【청구항 7] [7]
제 6항에 있어서, 상기 반응시키는단계 이전에,  7. The method of claim 6, wherein, prior to the step of reacting,
抑-나에 결합하는 물질이 표면에 부착된 기판에 PD-L1을 공급하여, PD-L1을기판에 부착시키는단계  A step of supplying PD-L1 to a substrate to which a substance bound to the substrate is attached and attaching the PD-L1 to the substrate
를추가로포함하는, PD-L1과 PD-1의 상호작용측정 방법 .  Wherein the PD-L1 and the PD-1 interact with each other.
【청구항 8]  [8]
제 6항에 있어서,  The method according to claim 6,
PD-L1에 결합하는물질이 표면에 부착된 기판에 표지 물질이 결합된 PD-L1을공급하고, PD-L1과기판표면의 물질 간반응에 의하여 발생하는 신호 ( 를측정하는단계  (PD-L1) to which a labeling substance binds to a substrate to which a substance binding to the PD-L1 has been attached, and a step of measuring a signal
를추가로포함하는, PD-L1과 PD-1의 상호작용측정 방법 .  Wherein the PD-L1 and the PD-1 interact with each other.
【청구항 9]  9]
제 8항에 있어서, 상기 신호 ( 에 대한 신호 (A) 값을 구하여 정량화하는단계를추가로포함하는, PD-L1과 PD-1의 상호작용측정 방법 .  The method according to claim 8, further comprising the step of obtaining and quantifying the value of the signal (A) for the signal (PD-L1 and PD-1).
【청구항 10] [Claim 10]
제 6항에 있어서, 상기 반응시키는 단계에서, PD-L1 및 PD-1 간 결합에 대하여 PD-L1또는 PD-1과경쟁하는물질을첨가하여 반응시키고, 신호 (시를측정하는단계를수행하는 것을특징으로하는, PD-L1과 PD-1의 상호작용측정 방법 .  7. The method according to claim 6, wherein in the step of reacting, a substance which competes with PD-L1 or PD-1 for the binding between PD-L1 and PD-1 is added and reacted, (PD-1) and PD-1 (PD-1).
【청구항 11】 Claim 11
제 6항 내지 제 10항 중 어느 한 항에 있어서, 상기 신호 (A) 또는 ( 는 근접장 (near-f ield) 영역에서 측정되는 것인, PD-L1과 PD-1의 상호작용측정 방법 .  11. A method according to any one of claims 6 to 10, wherein the signal (A) or () is measured in a near-field region.
【청구항 12】  Claim 12
제 6항내지 제 10항중어느한항에 있어서, 상기 기판에 부착된 PD- 11. A method according to any one of claims 6 to 10, wherein the PD-
L1에 결합하는 물질은 PD-L1의 말단 부위에 결합하는 물질인, PD-L1과 PD-1의 상호작용측정 방법 . The method for measuring the interaction between PD-L1 and PD-1, wherein the substance binding to L1 is a substance binding to the terminal region of PD-L1.
【청구항 13】  Claim 13
후보 물질, PD-L1, 및 표지 물질이 결합된 PD-1가혼합된 혼합물을 PD-L1과 결합하는 물질이 표면에 부착된 기판에 공급하여 반응시키는 단계; 및  Supplying a mixture of a candidate substance, PD-L1, and a labeled substance-bound mixture of PD-1 with a substance that binds to the PD-L1 to a surface-attached substrate; And
상기 반응시키는단계에서 얻어진신호 ( 를측정하는단계  The step of measuring the signal (obtained in the step of reacting)
를포함하는, PD-L1과 PD-1간상호작용억제제의 선정 방법 . 2019/132517 1»(:1^1{2018/016675 Lt; RTI ID = 0.0 > PD-L1 < / RTI > and PD-1. 2019/132517 1 »(: 1 ^ {2018/016675
【청구항 14】 14.
제 13항에 있어서, 14. The method of claim 13,
나에 결합하는물질이 표면에 부착된 기판에 표지 물질이 결합된 나을공급하고, 1)-나과기판표면의 물질 간반응에 의하여 발생하는 신호( 를측정하는단계  (1) measuring the signal generated by the reaction between the substance on the surface of the substrate and the substrate,
를추가로포함하는, 나과 P^-l간상호작용억제제의 선정 방법 .  Wherein the method further comprises the step of:
【청구항 15】 15.
제 14항에 있어서, 상기 신호 ( 에 대한 신호 ( 값을 구하여 정량화하는단계를추가로포함하는,
Figure imgf000132_0001
상호작용 억제제의 선정 방법.
15. The method of claim 14, further comprising: obtaining a signal (a value for the signal,
Figure imgf000132_0001
Selection method of interaction inhibitor.
【청구항 16】  Claim 16
제 1수용체 타이로신 인산화효소또는이를암호화하는유전자를제 2 수용체타이로신 인산화효소발현세포에 도입시키는단계;  Introducing a first receptor tyrosine kinase or a gene encoding the first receptor tyrosine kinase into a second receptor tyrosine kinase-expressing cell;
상기 세포에 제 1수요에 타이로신 인산화효소리간드를투입하여 제 1 수용체 타이로신 인산화효소와 제 2 수용체 타이로신 인산화효소 간 이합체를형성시키는단계; 및  Introducing a tyrosine phosphorylase ligand into the cell at a first demand to form a duplex between the first receptor tyrosine phosphorylase and the second receptor tyrosine phosphorylase; And
상기 이합체가 형성된 세포에 콜레스테롤-유사 세정제를 처리하는 단계  Treating the cell on which the dimer is formed with a cholesterol-like detergent
를포함하고,  Lt; / RTI >
제 1 수용체 타이로신 인산화효소과 제 2 수용체 타이로신 인산화효소는서로다른종류이고,  The first receptor tyrosine phosphorylase and the second receptor tyrosine phosphorylase are different kinds,
생체외에서수행되는것을특징으로하는,  Lt; RTI ID = 0.0 > in vitro, < / RTI >
활성화된수용체타이로신 인산화효소이합체의 제조방법 .  Wherein the activated receptor tyrosine kinase dimer is produced by the method.
【청구항 17】  17.
제 16항에 있어서, 상기 제 1수용체 타이로신 인산화효소는 요3이고, 제 2 수용체 타이로신 인산화효소는 묘2인, 활성화된 수용체 타이로신 인산화효소이합체의 제조방법.  17. The method of claim 16, wherein the first receptor tyrosine kinase is iodine 3 and the second receptor tyrosine kinase is Seed 2.
【청구항 18】  Claim 18
제 16항또는제 17항에 있어서,  18. The method according to claim 16 or 17,
상기 콜레스테롤-유사세정제는 0.05내지
Figure imgf000132_0002
The cholesterol-
Figure imgf000132_0002
및 0.003 내지 2%(八 0의 (용1 ¥1- 036 11) 중에서 선택된 1종 아상인, 활성화된수용체타이로신 인산화효소이합체의 제조방법 . And 0.003 to 2% (八0 of (1 ¥ 1- 03 6 11) 1 ahsang one selected from the method for the preparation of the activated receptor tyrosine kinases for dimer.
PCT/KR2018/016675 2017-12-26 2018-12-26 Method and apparatus for analyzing intracellular or intercellular protein-protein interaction WO2019132517A1 (en)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
KR20170180281 2017-12-26
KR10-2017-0180281 2017-12-26
KR20170180282 2017-12-26
KR10-2017-0180282 2017-12-26
KR20170180279 2017-12-26
KR10-2017-0180280 2017-12-26
KR20170180280 2017-12-26
KR10-2017-0180279 2017-12-26
KR1020180034749A KR20180117529A (en) 2017-04-19 2018-03-26 Method for predicting drug responsiveness by protein-protein interaction analysis
KR10-2018-0034749 2018-03-26

Publications (1)

Publication Number Publication Date
WO2019132517A1 true WO2019132517A1 (en) 2019-07-04

Family

ID=67067901

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2018/016675 WO2019132517A1 (en) 2017-12-26 2018-12-26 Method and apparatus for analyzing intracellular or intercellular protein-protein interaction

Country Status (1)

Country Link
WO (1) WO2019132517A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7659368B2 (en) * 1996-07-12 2010-02-09 Genentech, Inc. ErbB2 and ErbB3 chimeric heteromultimer adhesins
US7666606B2 (en) * 2004-12-04 2010-02-23 Los Alamos National Security, Llc Protein- protein interaction detection system using fluorescent protein microdomains
KR20120120093A (en) * 2011-04-20 2012-11-01 한국과학기술원 Apparatus for analyzing single protein-protein interactions in whole cell lysates
KR20140113543A (en) * 2013-03-14 2014-09-24 부산대학교 산학협력단 Methods for Screening Therapeutics for Cancer Using Interaction between AIMP2-DX2 and p14/ARF
KR20150064205A (en) * 2012-11-08 2015-06-10 에프. 호프만-라 로슈 아게 Her3 antigen binding proteins binding to the beta-hairpin of her3
KR20180135878A (en) * 2017-04-19 2018-12-21 주식회사 프로티나 Method and apparatus for analyzing protein-protein interactions

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7659368B2 (en) * 1996-07-12 2010-02-09 Genentech, Inc. ErbB2 and ErbB3 chimeric heteromultimer adhesins
US7666606B2 (en) * 2004-12-04 2010-02-23 Los Alamos National Security, Llc Protein- protein interaction detection system using fluorescent protein microdomains
KR20120120093A (en) * 2011-04-20 2012-11-01 한국과학기술원 Apparatus for analyzing single protein-protein interactions in whole cell lysates
KR20150064205A (en) * 2012-11-08 2015-06-10 에프. 호프만-라 로슈 아게 Her3 antigen binding proteins binding to the beta-hairpin of her3
KR20140113543A (en) * 2013-03-14 2014-09-24 부산대학교 산학협력단 Methods for Screening Therapeutics for Cancer Using Interaction between AIMP2-DX2 and p14/ARF
KR20180135878A (en) * 2017-04-19 2018-12-21 주식회사 프로티나 Method and apparatus for analyzing protein-protein interactions

Similar Documents

Publication Publication Date Title
KR102008521B1 (en) Protein-protein interaction analysis method and apparatus
Monypenny et al. ALIX regulates tumor-mediated immunosuppression by controlling EGFR activity and PD-L1 presentation
JP6619070B2 (en) ROS kinase in lung cancer
US20220267854A1 (en) Egfr and ros1 kinase in cancer
US20170275706A1 (en) Alk and ros kinase in cancer
US20100203043A1 (en) Treatment and diagnosis of metastatic prostate cancer with inhibitors of epidermal growth factor receptor (egfr)
KR20200044706A (en) Method for Predicting Recurrence of Breast Cancer
KR20130001342A (en) Identification of non-small cell lung carcinoma (nsclc) tumors expressing pdgfr-alpha
WO2018194406A1 (en) Method and apparatus for analysis of protein-protein interaction
WO2019132517A1 (en) Method and apparatus for analyzing intracellular or intercellular protein-protein interaction
KR102267593B1 (en) Method and Apparatus for Screening of Drugs Targeting Heterodimer of HER2 and HER3 and Method for Screening the Same
US20110027809A1 (en) Method for evaluation of degree of malignancy of tumor cell
Gad Characterizing the Signaling, Crosstalk, and Trafficking of Adhesion G Protein-Coupled Receptor G2 and the Calcium-Sensing Receptor in Parathyroid Adenoma Models
KR20210001095A (en) Pharmaceutical Composition for Inhibiting Interaction of PD-L1 and PD-1 and Method of Screening an Inhibitor
Tali The role of ShcA and ErbB2 (HER2) in regulating cell migration in breast cancer
Fukumoto Studies on p120-catenin: The regulatory or phosphorylation domain of p120-catenin controls E-cadherin dynamics at the plasma membrane

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: 18894722

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18894722

Country of ref document: EP

Kind code of ref document: A1