WO2016133374A1 - Method of selecting uterine contraction inhibiting agent based on protein damage information on each individual to prevent side effects of uterine contraction inhibiting agent - Google Patents

Method of selecting uterine contraction inhibiting agent based on protein damage information on each individual to prevent side effects of uterine contraction inhibiting agent Download PDF

Info

Publication number
WO2016133374A1
WO2016133374A1 PCT/KR2016/001631 KR2016001631W WO2016133374A1 WO 2016133374 A1 WO2016133374 A1 WO 2016133374A1 KR 2016001631 W KR2016001631 W KR 2016001631W WO 2016133374 A1 WO2016133374 A1 WO 2016133374A1
Authority
WO
WIPO (PCT)
Prior art keywords
drug
score
uterine contraction
sequence variation
protein
Prior art date
Application number
PCT/KR2016/001631
Other languages
French (fr)
Korean (ko)
Inventor
김주한
Original Assignee
싸이퍼롬, 인코퍼레이티드
김주한
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 싸이퍼롬, 인코퍼레이티드, 김주한 filed Critical 싸이퍼롬, 인코퍼레이티드
Publication of WO2016133374A1 publication Critical patent/WO2016133374A1/en

Links

Images

Classifications

    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B50/00ICT programming tools or database systems specially adapted for bioinformatics
    • GPHYSICS
    • G09EDUCATION; CRYPTOGRAPHY; DISPLAY; ADVERTISING; SEALS
    • G09FDISPLAYING; ADVERTISING; SIGNS; LABELS OR NAME-PLATES; SEALS
    • G09F19/00Advertising or display means not otherwise provided for
    • G09F19/12Advertising or display means not otherwise provided for using special optical effects
    • G09F19/18Advertising or display means not otherwise provided for using special optical effects involving the use of optical projection means, e.g. projection of images on clouds
    • GPHYSICS
    • G09EDUCATION; CRYPTOGRAPHY; DISPLAY; ADVERTISING; SEALS
    • G09FDISPLAYING; ADVERTISING; SIGNS; LABELS OR NAME-PLATES; SEALS
    • G09F19/00Advertising or display means not otherwise provided for
    • G09F19/22Advertising or display means on roads, walls or similar surfaces, e.g. illuminated

Definitions

  • the present invention relates to a method for selecting uterine contraction inhibitors based on individual protein damage information using individual genome sequencing to prevent adverse effects.
  • Pharmacogenetics predicts genetic differences in metabolism and reactions of drugs or chemicals in the general population or in individuals. In some individuals, reactions other than the expected drug response to the drug may be present. These drug side effects may be related to the severity of the disease being treated, drug interactions, the patient's age, nutritional status, liver and kidney function, climate, or food. Although it may be due to factors, genetic differences related to drug metabolism, for example, polymorphism of drug enzyme genes, may be affected.
  • ritodrine a type of uterine contraction inhibitor
  • ritodrine is a beta-adrenergic receptor agonist, which is used between 22 and 37 weeks of pregnancy.
  • Ritodrin is a class of beta-adrenergic receptor agonists, as well as calcium channel blockers, oxytonin antagonists, NSAIDs (non-steroidal anti-inflammatory drugs), nitrates, and MgSO4.
  • Progesterone progesterone
  • Ritodrin is known to have side effects such as tachycardia, hypotension, anxiety, chest pain, abnormal ECG, pulmonary edema, hyperglycemia, hypokalemia, arrhythmia and myocardial infarction.
  • treatment with ritodrine should include an appropriate assessment of the patient's cardiovascular status along with cardiopulmonary function monitoring and electrocardiogram monitoring throughout the treatment period. If complications occur, medications should be discontinued immediately and, in some cases, switched to other medications, leading to failure of early analgesia.
  • the pharmacogenomics mentioned above may be used as a method for predicting side effects of drugs before drug administration. It has also been found that drug dynamics are associated with genotypes for ritodrine. Previous studies have shown that the time to deliver after the administration of ritodrine is related to rs1042719 of the ADRB2 (Adrenoceptor, beta 2) gene.
  • ADRB2 Adrenoceptor, beta 2
  • the present invention was devised in view of the above, and after analyzing individual genome sequence variation information and calculating individual protein damage scores from gene sequence variation information related to pharmacodynamics or pharmacokinetics of uterine contraction inhibitors, By calculating the individual drug scores in association with the interaction between the drug and the protein, it is intended to provide a method for predicting the possibility of adverse effects of uterine contraction inhibitors and providing information for selecting the uterine contraction inhibitors.
  • the present invention comprises the steps of determining one or more gene sequence variation information involved in pharmaco-dynamics or pharmaco-kinetics of a uterine contraction inhibitor from personal genome sequence information; Calculating an individual protein damage score using the gene sequence variation information; And correlating the individual protein damage scores with the correlation between the drug and the protein to calculate the individual drug scores, thereby providing a method for providing information for selecting the uterine contraction inhibitor using the personal genome sequence variation.
  • the present invention provides a uterine contraction inhibitor that can be applied to an individual, the database for retrieving or extracting information related to the gene or protein associated with the uterine contraction inhibitor; A communication unit accessible to the database; A first calculation module configured to calculate one or more gene sequence variation information related to pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor based on the information; A second calculation module for calculating an individual protein damage score using the gene sequence variation information; A third calculating module that calculates an individual drug score by associating the individual protein damage score with a correlation between a drug and a protein; And it provides a uterine contraction inhibitor selection system using a personal genome sequence variation comprising a display unit for displaying the calculated value calculated by the calculation module.
  • the present invention comprises the steps of obtaining genetic sequence variation information involved in the pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor from the individual genome sequence information; Calculating an individual protein damage score using the gene sequence variation information; And correlating the individual protein damage score with a correlation between the drug and the protein to calculate the individual drug score, wherein the execution module executes a processor to perform the operation. .
  • Individual uterine contraction inhibitor selection method and system based on the individual genome sequence variation information of the present invention through the sequence analysis of the exon region of the gene encoding various proteins involved in the pharmacodynamics or pharmacokinetics of the uterine contraction inhibitors In other words, the reliability of the uterine contraction inhibitors is high.
  • NGS Next Generation Sequencing
  • ritodrine uterine contraction inhibitors
  • it is useful to determine whether and how to use the uterine contraction inhibitors applied to the individual by predicting the side effects or risks of uterine contraction inhibitors, for example, ritodrine in advance. This may lead to improved success rate of early analgesic suppression therapy by administering an appropriate dose of ritodrine in patients with early analgesia.
  • it may be possible to introduce a new treatment method by increasing the safety of ritodrine administration in a patient group that could not withstand the drug or had to stop the drug due to side effects.
  • 1 is a flow chart showing each step of the method for providing information for the selection of uterine contraction inhibitors using individual genome sequence variation according to an embodiment of the present invention.
  • DB database
  • 3A and 3B are flowcharts illustrating a receiver operating curve (ROC) curve for verification of a method for selecting a uterine contraction inhibitor using a genome sequence variation according to an embodiment of the present invention.
  • ROC receiver operating curve
  • the present invention is based on the discovery that individual drugs can be selected for high-safety drugs and doses / uses in the treatment of uterine contraction inhibitors by analyzing individual genome sequence variation information.
  • the present invention comprises the steps of determining one or more gene sequence variation information involved in pharmaco-dynamics or pharmaco-kinetics of a uterine contraction inhibitor from personal genome sequence information; Calculating an individual protein damage score using the gene sequence variation information; And calculating the individual drug score by associating the individual protein damage score with the correlation between the drug and the protein, thereby providing information for selecting the uterine contraction inhibitor using the personal genome sequence variation.
  • uterine contraction inhibitors include, but are not limited to, all substances that exhibit the same, similar pharmacological activity, such as drugs belonging to the drug class disclosed in Table 1, derivatives thereof, and pharmaceutically acceptable salts thereof.
  • Beta-adrenergic receptors Terbutaline, Ritodrine, Fenoterol, Albuterol Calcium channel blockers Nicardipine, Nifedipine Oxytonin antagonists Atosiban Nonsteroidal Anti-inflammatory Drugs (NSAIDs) Indomethacin, Ketorolac, Sulindac Myosin light chain inhibitor MgSO4 nitrate Nitroglycerin Progesterone Hydroxyprogesterone caproate, Micronized progesterone ethyl alcohol
  • DrugBank http://www.drugbank.ca/
  • KEGG Drug http://www.genome.jp/kegg/drug/
  • PharmGKB https://www.pharmgkb.org/
  • BLK BLK
  • SPTA1 IFT74 RSPH3
  • CYP8B1 ICE1, NKAIN3, AASDH
  • the genes involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitors are BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXG1, , HHATL, SERPINA7, TNKS, PSMD9, ZNF273, FAT4, GALNT10, OR6B1, RBBP8NL, KNDC1, UGT1A10, and ARL13B, and one selected from the group consisting of SLC15A2, SPINK6, C10orf113, and TP53, TP53. , MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT, LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EFCAB4A, CPOX, and VARS2.
  • the genes / proteins are expressed according to the nomenclature of the HUGO Gene Nomenclature Committee (HGNC) (Gray KA, Daugherty LC, Gordon SM, Seal RL, Wright MW, Bruford EA.genenames.org: the HGNC resources in 2013. Nucleic Acids Res. 2013 Jan; 41 (Database issue): D545-52. Doi: 10.1093 / nar / gks1066. Epub 2012 Nov 17 PMID: 23161694).
  • HGNC HUGO Gene Nomenclature Committee
  • Gene sequence variation used as one information in the present invention refers to a variation or polymorphism of the individual gene sequence.
  • the gene sequence mutation or polymorphism occurs at a gene region, particularly an exon region, which encodes a protein associated with pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor, but is not limited thereto.
  • base sequence variation information used in the present invention means information about the substitution, addition or deletion of the base constituting the exon of the gene. Substitution, addition, or deletion of such bases can occur for a variety of reasons, for example, by structural differences including mutations, truncation, deletions, duplications, inversions and / or translocations of chromosomes.
  • nucleotide polymorphism refers to differences between individuals of nucleotide sequences present in the genome, and the largest number of nucleotide polymorphisms is Single Nucleotide Polymorphism (SNP), and A, T, C, There is a difference between individuals in one base of the base sequence consisting of G.
  • Sequence polymorphism is a form of polyalleic variation including single nucleotide variation (SNV), short tandem repeat polymorphism (STRP), or variable number of tandem repeat (VNTR) and copy number variation (CNV), including SNPs. May appear.
  • sequence variation or polymorphism information found in an individual's genome is collected in association with proteins associated with pharmacodynamics or pharmacokinetics of uterine contraction inhibitors. That is, the nucleotide sequence information used in the method of the present invention is one or more genes involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitors, such as a target protein associated with the drug, Mutation information found in the exon region, in particular but not limited to, genes encoding enzyme proteins, transporter proteins or carrier proteins involved in drug metabolism.
  • the genome sequence information of an individual used in the present invention may be determined using a known sequence decoding method, and services such as Complete Genomics, BGI (Beijing Genome Institute), Knome, Macrogen, DNALink, etc., which provide commercially available services. May be used, but is not limited thereto.
  • Gene sequence variation information present in the genome sequence of an individual in the present invention can be extracted using a variety of methods, a sequence comparison program with a genomic sequence of a reference group, for example HG19, for example, ANNOVAR ( Wang et al., Nucleic Acids Research, 2010; 38 (16): e164), Sequence Variant Analyzer (SVA) (Ge et al., Bioinformatics. 2011; 27 (14): 19982000), Break Dancer (Chen et al., Nat Methods.2009 Sep; 6 (9): 677-81) and the like.
  • the gene sequence variation information may be received / obtained through a computer system, and in this aspect, the method of the present invention may further include receiving the genetic variation information into a computer system.
  • the computer system used in the present invention is a gene involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitors, for example, a gene encoding a target protein associated with a drug, an enzyme protein involved in drug metabolism, a transporter protein or a carrier protein. Include or have access to one or more databases containing information about them.
  • Such databases include, for example, DrugBank (http://www.drugbank.ca/), KEGG Drug (http://www.genome.jp/kegg/drug/), PharmGKB (http://www.pharmgkb.org /) May include, but is not limited to, public or private databases or knowledge bases that provide information about genes / proteins / drug-protein interactions, and the like.
  • the uterine contraction inhibitor may be information input by a user, information input from a prescription (pre), or information input from a database including information on the uterine contraction inhibitor.
  • the prescription includes, but is not limited to, electronic prescription.
  • pk pharmaco-kinetics
  • pk pharmacokinetic parameters
  • Distribution volume (Vd), clearance (CL), bioavailability (F), absorption rate coefficient (ka), maximum plasma concentration (Cmax), and time point of maximum plasma concentration, Tmax) area under the curve (AUC) measurements of changes in blood drug concentration over time.
  • pharmacodynamics or pharmacodynamic parameters refers to the characteristics related to the physiological and biochemical action of the drug on the living body and its mechanism of action, ie the reaction or effect of the living body of the drug.
  • the term “gene sequence variation score” refers to an amino acid sequence variation (substitution or addition) of a protein encoded by the gene when the genome sequence variation is found in the exon region of the gene encoding the protein. Or deletion) or a score that quantifies the extent to which transcriptional control mutations result, thereby causing significant changes or damage to the structure and / or function of the protein, wherein the gene sequence variation score is the evolution of amino acids on the genome sequence It can be calculated in consideration of the degree of preservation and the degree of change in the structure or function of the protein according to the physical properties of the modified amino acid.
  • the method of calculating the gene sequence variation score may be performed using a method known in the art. See, eg, SIFT (Sorting Intolerant From Tolerant, Pauline C et al., Genome Res. 2001 May; 11 (5): 863874; Pauline C et al., Genome Res. 2002 March; 12 (3): 436446; Jing Hul et al., Genome Biol. 2012; 13 (2): R9), PolyPhen, PolyPhen-2 (Polymorphism Phenotyping, Ramensky V et al., Nucleic Acids Res.
  • fathmm (Shihab et al., Functional Analysis through Hidden Markov Models, Hum Mutat 2013; 34 : 57-65, http://fathmm.biocompute.org.uk/) may be used to calculate the gene sequence variation score from the gene sequence variation information, but is not limited thereto.
  • the purpose of the algorithms described above is to determine how each gene sequence mutation affects protein function, how this damage damages the protein, or whether there is little effect. They have a common point in that they determine the impact of individual gene sequence variations on the structure and / or function of the protein in consideration of the changes that will result in the amino acid sequence of the protein encoded by the gene.
  • a Sorting Intolerant From Tolerant (SIFT) algorithm was used to calculate an individual gene sequence variation score.
  • SIFT Sorting Intolerant From Tolerant
  • gene sequence variation information is input to a VCF (Variant Call Format) format file, and each gene sequence variation is scored for damaging the gene.
  • VCF Variant Call Format
  • the method of the present invention calculates individual protein damage scores based on the gene sequence variation scores described above in the next step.
  • protein damage score means that two or more significant sequence mutations are found in a gene region encoding one protein, so that one protein has two or more gene sequence mutation scores. Refers to a score calculated by combining the gene sequence variation score. If there is a significant sequence variation in a gene region encoding a protein, the gene sequence variation score and the protein damage score are the same. In this case, when there are two or more gene sequence mutations encoding a protein, the protein damage score is calculated as an average value of the gene sequence variation scores calculated for each variation, and the average value is, for example, a geometric mean, an arithmetic mean, or a harmonic mean.
  • Arithmetic geometric mean, arithmetic harmonic mean, geometric harmonic mean, Pythagorean mean, quadrant mean, quadratic mean, cutting mean, windsorized mean, weighted mean, weighted geometric mean, weighted arithmetic mean, weighted harmonic mean, function mean, ⁇ average Can be computed as a generalized f-means, percentiles, maximums, minimums, modes, medians, median ranges, measures of central tendency, simple products or weighted products, or as a function of these calculations. This is not restrictive.
  • the protein damage score was calculated by Equation 1 below, and Equation 1 may be variously modified and is not limited thereto.
  • Equation 1 Sg is the protein damage score of the protein encoded by the gene g, n is the number of the nucleotide sequence analysis of the nucleotide variation of the gene g, vi is the gene sequence variation score of the i-th gene sequence variation And p is a nonzero real number.
  • p when the value of p is 1, it is an arithmetic mean, and when the value of p is -1, it is a harmonic mean, and in the extreme case where the value of p is close to 0, it is a geometric mean.
  • the protein damage score was calculated by Equation 2 below, and Equation 2 may be variously modified and is not limited thereto.
  • Equation 2 Sg is the protein damage score of the protein encoded by the gene g, n is the number of the nucleotide sequence analysis of the nucleotide sequence variation of the gene g, vi is the gene sequence variation score of the i-th gene sequence variation , wi is a weight given to vi. When all weights wi have the same value, the protein damage score Sg becomes the geometric mean value of the gene sequence variation score vi.
  • the weight may be given in consideration of the type of the protein, the pharmacokinetic or pharmacodynamic classification of the protein, the pharmacokinetic parameters of the drug enzyme protein, and the population or race distribution.
  • Vmax is the maximum enzyme reaction rate when the substrate concentration is very high
  • Km is the concentration of the substrate that causes the reaction to reach 1/2 Vmax.
  • Km can be seen as an affinity between the enzyme and the substrate. The smaller the Km, the stronger the bond between the enzyme and the substrate.
  • Kcat also called the enzyme's metabolic rate, refers to the number of substrate molecules that are metabolized at one second per enzyme active site when the enzyme is active at its maximum rate, and how fast the enzyme reaction actually occurs.
  • the method of the present invention correlates the protein damage score described above in the next step with the correlation between the drug and the protein to yield an individual drug score.
  • drug score refers to a given protein, eg, a target protein involved in the pharmacodynamics or pharmacokinetics of a given drug, an enzyme protein involved in drug metabolism, a transporter protein or Carrier proteins are found, the protein damage scores of the proteins are calculated, and then summed again to refer to the values calculated for one drug.
  • the drug score is calculated as an average value of the protein damage scores when the damage of the protein involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitors, for example, geometric mean, arithmetic mean, harmonic mean, Arithmetic geometric mean, Arithmetic harmonic mean, Geometric harmonic mean, Pythagorean mean, Quarter mean, Secondary mean, Cutting mean, Windsorized mean, Weighted mean, Weighted geometric mean, Weighted arithmetic mean, Weighted harmonic mean, Function mean, ⁇ mean, Generalized f-means, percentiles, maximums, minimums, modes, medians, median ranges, measures of central tendency, or simple products or weighted products, or arithmetic operations of these calculations This is not restrictive.
  • the drug score may be calculated by adjusting the weights of the drug, that is, the target protein involved in the pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor, the enzyme protein involved in the drug metabolism, the transporter protein, and the carrier protein in consideration of pharmacological properties.
  • the weight may be given in consideration of the pharmacokinetic parameters of the corresponding drug metabolizing enzyme, the population group or the race distribution.
  • drug scores are also taken into account when considering protein damage scores of proteins that do not interact directly with the drug but interact with precursors or metabolites of the drug, such as proteins that make up the pharmacological pathway. Can be calculated.
  • the drug score may be calculated by considering the protein damage scores of the proteins that significantly interact with the proteins involved in the pharmacodynamics or pharmacokinetics of the drug.
  • PharmGKB Wang-Carrillo et al., Clinical Pharmacology & Therapeutics 2012; 92 (4): 414-4171
  • the MIPS Mammalian Protein-Protein Interaction Database Panetl al., Bioinformatics 2005; 21 (6): 832-834
  • BIND Boder et al., Biomolecular Interaction Network Database, Nucleic Acids Res. 2003 Jan 1; 31 (1): 248-50
  • Reactome Joshi-Tope et al., Nucleic Acids Res. 2005 Jan 1; 33 (Database issue): D428-32
  • the drug score was calculated by the following Equation 3, and the following Equation 3 may be variously modified, but is not limited thereto.
  • Equation 3 Sd is a drug score of drug d, n is a protein directly involved in the pharmacodynamics or pharmacokinetics of drug d or interacts with a precursor of the drug or metabolites of the drug, for example, a pharmacological pass.
  • the number of proteins encoded by one or more genes selected from the group of genes participating in the way, gi is a protein that directly participates in the pharmacodynamics or pharmacokinetics of drug d or interacts with precursors or metabolites of the drug, eg
  • p when the value of p is 1, it is an arithmetic mean, and when the value of p is -1, it is a harmonic mean, and in the extreme case where the value of p is close to 0, it is a geometric mean.
  • the drug score was calculated by Equation 4 below, and Equation 4 may be variously modified, and is not limited thereto.
  • Sd is a drug score of drug d
  • n is a protein directly involved in the pharmacodynamics or pharmacokinetics of drug d or interacts with a precursor of the drug or metabolites of the drug, eg, a pharmacological pass.
  • the number of proteins encoded by one or more genes selected from the group of genes constituting the way, gi are proteins that directly participate in the pharmacodynamics or pharmacokinetics of drug d or interact with precursors or metabolites of the drug, eg
  • the protein damage score of the protein encoded by one or more genes selected from the gene group constituting the pharmacological pathway, wi is the weight given to the gi.
  • the drug score Sd becomes the geometric mean value of the protein damage score gi when all weights wi have the same value.
  • the weight may be given in consideration of the type of the protein, the pharmacokinetic or pharmacodynamic classification of the protein, the pharmacokinetic parameters of the drug enzyme protein, and the population or race distribution.
  • the weights were equally given regardless of the drug-protein association characteristics, but the weight was given by considering the respective characteristics of drug-protein association. It is possible to calculate the drug score. For example, different scores may be assigned to the drug's target protein and the drug's transporter protein.
  • the drug metabolizing enzyme may be weighted by the pharmacokinetic parameters Km, Vmax, and Kcat / Km to calculate the drug score.
  • the target protein may be given a higher weight by judging that the target protein is more important in pharmacological action than the transporter protein, and the transporter protein or the carrier protein may be given a high weight for the concentration-sensitive drug. This is not restrictive.
  • Weights can be closely adjusted according to the correlation between drug and drug-related proteins, and the nature of drug-protein interactions. For example, a sophisticated algorithm can be used that assigns a weight to the nature of the drug-protein interaction, such as giving the target protein two points and the transporter protein one point.
  • the drug that is, the protein that directly interacts with the uterine contraction inhibitor
  • the protein interacts with the precursor of the drug or its metabolites, and the proteins involved in the pharmacodynamics or pharmacokinetics of the drug. It is possible to improve the predictive power of the formula by utilizing protein information that interacts easily, and related protein information that participates in the signaling pathway. In other words, the protein-protein interaction network or pharmacological pathway information can be utilized to use the information of various proteins involved in it.
  • the mean value (eg, geometric mean) of the protein damage scores of related proteins participating in the same signaling pathway may be used as a drug score calculation instead of the protein damage score of the protein.
  • the individual drug scores may be calculated for all drugs for which information on one or more related proteins can be obtained or for some selected drugs.
  • individual drug scores can be converted into rank (rank).
  • the method of the present invention may further include determining whether to use a drug applied to the individual, ie, a uterine contraction inhibitor, by using the aforementioned individual drug score.
  • the individual drug scores of the present invention can be applied individually to all drugs, but are more useful if they are classified by disease, clinical characteristics, or mode of action, or applied to drugs for medical comparison.
  • Drug classification systems that can be used in the present invention are, for example, the ATC (Anatomical Therapeutic Chemical Classification System) code, the top 15 frequently prescribed drug classes during 2005-2008 in the United States (Health, United States, 2011, Centers for Disease Control and Prevention), a list of drugs that have been released from the market due to known pharmacogenomic markers that may affect drug action information on the drug label, or side effects. It may include.
  • the method of the invention may further comprise calculating a prescription score.
  • the term “prescription score” refers to the sum of the drug scores determined for each drug when two or more drugs are administered at the same time or at intervals short enough to significantly affect the pharmacological action of each other. Say your score is calculated.
  • the prescription score may be calculated by combining the drug scores determined for each drug when two or more drugs determined by the priority between the drugs and simultaneous administration are required. The calculation of the prescription score can be calculated by simply averaging, summing or multiplying the drug scores of the plurality of drugs, for example, when no protein interacts in common with the plurality of drugs. If there are proteins that interact in common with the plurality of drugs, the drug damage scores of the common interacting protein are weighted twice, for example, to calculate each drug score and then the corresponding drug scores are calculated. By summing up, the prescription score can be calculated.
  • Prescription scores are intended to determine the adequacy or risk of a multi-drug prescription that is included in the prescription applied to an individual, beyond the effects of individual drugs.
  • the method of the present invention may further comprise determining the appropriateness or risk of the prescription (pre) applied to the individual.
  • the method of the present invention includes, but is not limited to, being performed for the purpose of preventing adverse effects of uterine contraction inhibitors.
  • FIG. 1 is a flow chart showing each step of the method for providing information for the selection of uterine contraction inhibitors using individual genome sequence variation according to an embodiment of the present invention.
  • S150 Individual drug score calculation for uterine contraction inhibitors
  • S150 Individual drug score calculation for uterine contraction inhibitors
  • Drug score notation by drug score ranking
  • the step of providing a pharmacogenomic calculation process and the basis for calculating the drug scores by providing information such as pictures, charts and explanations to help the prescriber in making decisions It may further include. That is, the method according to the present invention may further include providing one or more information of gene sequence variation information, gene sequence variation score, protein damage score, drug score, and information used to calculate the same.
  • the present invention provides a uterine contraction inhibitor that can be applied to an individual, the database for retrieving or extracting information related to the gene or protein associated with the uterine contraction inhibitor; A communication unit accessible to the database; A first calculation module configured to calculate one or more gene sequence variation information related to pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor based on the information; A second calculation module for calculating an individual protein damage score using the gene sequence variation information; A third calculating module that calculates an individual drug score by associating the individual protein damage score with a correlation between a drug and a protein; And it relates to a uterine contraction inhibitor selection system using a personal genome sequence variation comprising a display unit for displaying the calculated value calculated by the calculation module.
  • the module may mean a functional and structural combination of hardware for performing the technical idea according to the present invention and software for driving the hardware.
  • the module may mean a logical unit of a predetermined code and a hardware resource for performing the predetermined code, and does not necessarily mean a physically connected code or a kind of hardware. Will be apparent to those skilled in the art.
  • output module refers to the gene sequence variation score, protein damage score, drug score, and information on which the calculation is made for the uterine contraction inhibitor and gene to be analyzed according to the method of the present invention.
  • a predetermined code for calculating each score based on the information and a logical unit of a hardware resource for performing the predetermined code and means a physically connected code or a kind of code. It doesn't mean hardware.
  • the system according to the present invention may further include a fourth calculation module for determining whether to use the uterine contraction inhibitor applied to the individual by using the individual drug score calculated in the third calculation module.
  • the system according to the present invention further includes a fifth calculation module for calculating the prescription score by combining the drug scores determined for each drug when two or more drugs determined by the priority between drugs are required. It may include.
  • the system according to the present invention inputs a list of uterine contraction inhibitors or accesses a database including information on uterine contraction inhibitors, extracts relevant information, and calculates and provides a drug score of the drug accordingly. It may further include a user interface.
  • the system according to the present invention may further include a display unit for further displaying a calculation process in which a value calculated in each calculation module or a priority between drugs is determined, and information on which the calculation or calculation is based.
  • the server including the database or its access information, the calculated information and the user interface device connected thereto may be used in conjunction with each other.
  • the system according to the invention can be updated immediately when new pharmacological / biochemical information on the drug-protein correlation is produced and can be used for further improved uterine contraction inhibitor selection.
  • the gene sequence variation information, gene sequence variation score, protein damage score, drug score and information on which the basis of the calculation stored in each calculation module Is updated.
  • System 10 of the present invention is a database (DB) 100, communication unit 200, user interface or terminal 300, the calculation unit 400 capable of searching or extracting information related to genes or proteins related to the uterine contraction inhibitors And a display unit 500.
  • DB database
  • the calculation unit 400 capable of searching or extracting information related to genes or proteins related to the uterine contraction inhibitors
  • a display unit 500 a display unit 500.
  • the user interface or the terminal 300 may request, receive and / or store uterine contraction inhibitor selection process using a personal genome sequence variation from a server, and may be a smartphone, a personal computer (PC), or a tablet. It may be configured as a terminal having a mobile communication function having a computing capability by mounting a microprocessor such as a PC, a personal digital assistant (PDA), a web pad, or the like.
  • a server may be a smartphone, a personal computer (PC), or a tablet. It may be configured as a terminal having a mobile communication function having a computing capability by mounting a microprocessor such as a PC, a personal digital assistant (PDA), a web pad, or the like.
  • a microprocessor such as a PC, a personal digital assistant (PDA), a web pad, or the like.
  • the server is a means for providing access to the database 100 for uterine contraction inhibitors, genetic variation or drug-protein correlations, and is connected to the user interface or the terminal 300 through the communication unit 200. It is configured to exchange various information.
  • the communication unit 200 may communicate in the same hardware, as well as a local area network (LAN), a metropolitan area network (MAN), a wide area network (WAN), the Internet, 2G, 3G, 4G mobile communication network, Wi-Fi (Wi-Fi), Wibro (Wibro) and the like can be included, and the communication method is wired, wireless, any communication method.
  • the database 100 may also be connected to various life science databases accessible through the Internet as well as being installed directly on the server.
  • the calculation unit 400 may include the first calculation module 410 for calculating one or more gene mutation information related to pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor using the collected / input information as described above, A second calculation module 420 for calculating an individual protein damage score and a third calculation module 430 for calculating an individual drug score may be included.
  • a storage medium includes any medium for storage or delivery in a form readable by a device such as a computer.
  • a computer readable medium may include read only memory (ROM); Random access memory (RAM); Magnetic disk storage media; Optical storage media; Flash memory devices and other electrical, optical or acoustic signaling media, and the like.
  • the present invention comprises the steps of obtaining genetic sequence variation information involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitor from the individual genome sequence information; Calculating an individual protein damage score using the gene sequence variation information; And an execution module for associating the individual protein damage score with the correlation between the drug and the protein to execute a processor to perform an operation comprising calculating the individual drug score.
  • the processor may further include determining whether to use a uterine contraction inhibitor applied to the individual by using the individual drug score.
  • the present invention provides a biomarker composition for predicting adverse effects of uterine contraction inhibitors.
  • Genes that may be included in the biomarker composition according to the present invention include BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7 TNKS, PSMD9, ZNF273, FAT4, GALNT10, OR6B1, RBBP8NL, KNDC1, UGT1A10, ARL13B, SLC15A2, SPINK6, C10orf113, TP53, TRIML2, MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT3 LRRC3C, EFCAB4A, CPOX, VARS2, and the like, but are not limited to these.
  • By analyzing the mutation of the gene or its protein it is possible to predict the occurrence of adverse effects of the uterine contraction inhibitor, it can be used as a marker using the agent that
  • Preterm labor refers to regular uterine contraction prior to 37 weeks of gestation and thus to gradual cervical dilatation and maturation, which is one of the leading causes of premature birth.
  • the birth rate is decreasing and the premature birth rate is gradually increasing due to the increase of elderly mothers (4.3% in 1995, 7% in 1998, 8.3% in 2000, 10% in 2003), which is becoming a big social problem.
  • An increase in the rate of premature births is a problem that cannot be overlooked in terms of maternal and child health. This is why the development of treatment technology for early labor, which is the main cause of premature birth, is essential.
  • uterine contraction inhibitors that have been widely used in Korea include ritodrine, magnesium (MgSO 4 ), nifedipine (Nifedipine), and atoshiban (Atosiban).
  • ritodrine, magnesium and nifedipine which are used most frequently, are adversely affected by the side effects of these drugs.
  • complications such as tachycardia, hypotension, anxiety, chest pain, abnormal ECG, pulmonary edema, etc. of pregnant women is a problem
  • the frequency and severity of side effects are very different for each pregnant woman.
  • the responsiveness to a particular drug is different for each patient, so appropriate drug administration cannot be made to each patient and treatment failure or delay is indicated.
  • sequence fragments analyzed were sequence alignment map (SAM) and binary alignment map (BAM) files aligned with human reference sequence (eg, HG19) through data cleaning and quality check.
  • SAM sequence alignment map
  • BAM binary alignment map
  • the output was in format.
  • the clean alignment result is detected using a software tool such as SAMTools: pileup, SAMTools: mpileup, GATK: recalibration, GATK: realignment, and the like to detect mutations such as single nucleotide variations (SNVs, Single Nucleotide Variants) and InDels. It was output to a file in VCF (Variant Calling Format) format.
  • VCF Variariant Calling Format
  • the above-described gene sequence variation score vi value was calculated for each variation by using the SIFT algorithm, and the individual protein damage score Sg was calculated using Equation 2. Subsequently, the individual protein damage scores were compared for each gene in 13 cases and 30 health control controls. 47 genes showing statistically significant differences in the adverse event group and the control group were selected as follows, and 28 genes with higher statistical significance were selected as the first group based on the p-value. The following genes are genes of high relevance to the pharmacokinetics or pharmacokinetics of ritodrine and its metabolites.
  • BLK BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7, TNKS, PSMD9, ZBF6AL1NT, ZBF673N10 KNDC1, UGT1A10, ARL13B
  • the drug score for each individual gene group was calculated using the method of the present invention. More specifically, after calculating the gene sequence variation score using the SIFT algorithm from each individual gene sequence variation information, the individual protein damage score for the 47 genes was calculated using Equation 2, Equation 4 Using to calculate the individual drug score for ritordlin, statistical analysis for each group. The results are shown in Table 3.
  • the protein damage score and the individual drug score were calculated from the gene sequence variation information using the 28 selected group 1 genes, and statistically significant between the normal control group and the ritodrine side effects group. It was confirmed that a difference appeared.
  • the method of the present invention can be used to predict a group of patients with high risk of side effects in the future patients with early analgesia, and induce high-risk patients to adjust the concentration of the drug or to use other alternative or interventional therapy.
  • the method of the present invention can be used to predict a group of patients with high risk of side effects in the future patients with early analgesia, and induce high-risk patients to adjust the concentration of the drug or to use other alternative or interventional therapy.
  • the personalized drug selection method according to the present invention not only targets all gene sequence mutations, but also does not require expensive case-control design observation studies, and damages individual proteins by pure calculation of genome sequence mutations. Since we propose a method of calculating the score and the individual drug score and applying it, it has the great advantage that it is possible to infer a personalized drug selection for the combination of all genome sequence variations and all drugs.
  • Validity criteria data were extracted from the established knowledge of 987 gene sequence mutation-drug interaction pairs provided by PharmGKB with 650 (65.9%) having at least one link with the 497 drugs.
  • the present invention targets the nucleotide sequence variation of the exon region, overlapping portions between the verification target data and the evaluation reference data are removed for fair evaluation. More specifically, all 36 nucleotide sequence mutations located in the exon region were removed from the 650 pairs, and only a nucleotide sequence variation of the non-coding region was selected to perform a more fair evaluation. In conclusion, we selected 614 pairs as the final gold standard for evaluation.
  • the sensitivity, specificity and area under the receiver operating curve were used. After ranking 497 drugs based on individual drug scores and setting thresholds by rank at 496 splits between each rank, (1) the drug score rank of the drug is above the threshold and the PharmGKB mutation is in the personal genome.
  • D is a set of 497 total drugs
  • GS is a set of personalized PharmGKB drugs that are used as individual gold standards by matching individual genetic sequence mutations with the risk allele of PharmGKB
  • D L is a set of top-ranking drugs The vertical bar brackets indicate the number of elements in the set.
  • ROC curve was calculated by calculating the specificity and and the AUC was calculated. More specifically, first, gene sequence variation scores were calculated using a SIFT algorithm for a total population of 1092 people, and then protein damage scores and drug scores were calculated by applying Equations 2 and 4, respectively.
  • Protein group distribution and average protein damage score calculation Protein family Protein count Related drug count Number of protein-drug pairs Average protein damage score Target protein 440 486 2357 0.798 Carrier Protein 10 50 65 0.728 Metabolic protein 74 330 1347 0.733 Transporter protein 54 176 457 0.733 system 545 497 4201 0.783
  • Table 4 shows the distribution of the protein groups for the 497 drugs used in this example, and the number of protein-drug pairs and the average protein damage score were displayed together in each group.
  • each group of proteins such as target protein, carrier protein, metabolic protein, and transporter protein
  • a result of performing an AUC analysis of individual drug scores by applying weights according to the number of individual races is considered, considering race specificity (bold line).
  • the total AUC of the population was 0.666 (African 0.744, American 0.650, Asian 0.631, European 0.653), without considering race specificity (dotted line).
  • the overall population AUC was 0.633 (African 0.623, American 0.629, Asian 0.64, European 0.636), indicating that the validity of drug score calculation considering race specificity was improved compared to that of the other population.
  • the individual drug score calculation validity AUC of the individual of the present invention is 0.634, and also applied to the weight of each protein group with race specificity ( Thick line), the individual drug score calculation validity AUC of the present invention is 0.667, indicating that the different weights are useful.

Abstract

The present invention relates to a method and system for selecting a uterine contraction inhibiting agent on the basis of protein damage information on each individual, by using individual genome base sequence analysis. The method and system according to the present invention, as a technology that can predict the side effects or danger of certain drugs (i.e., uterine contraction inhibiting agents) for each individual through sequence analyses of gene exon regions that code for various proteins that participate in the pharmacodynamics or pharmacokinetics of uterine contraction inhibiting agents, is a general-purpose technology that is not only highly reliable but also has a wide range of applications.

Description

자궁수축억제제 부작용 방지를 위한 개인별 단백질 손상 정보 기반의 자궁수축억제제 선택 방법Method of selecting uterine contraction inhibitor based on individual protein damage information for preventing side effects of uterine contraction inhibitor
본 발명은 자궁수축억제제 부작용 방지를 위한 개인 유전체 염기서열 분석을 이용한, 개인별 단백질 손상 정보 기반의 자궁수축억제제 선택 방법에 관한 것이다. The present invention relates to a method for selecting uterine contraction inhibitors based on individual protein damage information using individual genome sequencing to prevent adverse effects.
생명공학 기술의 발전으로 인해 현재는 인간의 전 유전체 염기서열(whole genome sequence)을 분석하여 개개인의 질병을 예측하고 맞춤형 질병 예방 및 치료를 제공하는 단계까지 도달하였다.Advances in biotechnology have led to the analysis of human whole genome sequences to predict individual diseases and provide customized disease prevention and treatment.
최근에는 개인 유전체 염기서열을 비교한 결과, 염색체의 동일한 위치에 서로 다른 염기가 존재한다는 사실이 밝혀짐에 따라 이러한 염기서열의 차이를 의약품에 대한 개인 간 반응차이를 예측하는 데 이용하게 되었다. 예를 들어, 한 개인이 갖는 특정 유전체 염기서열 정보에 따라 약물대사가 느리거나 빠르기 때문에 약물에 대한 효과 및 부작용이 개인마다 다를 수 있다. Recently, as a result of comparing individual genome sequences, it was found that different bases exist at the same position on the chromosome, and thus, the differences of the base sequences were used to predict the difference in the reaction between individuals for the drug. For example, because of the slow or fast drug metabolism depending on the specific genomic sequence information of an individual, the effects and side effects on the drug may vary from person to person.
따라서 개인별 유전체 염기서열 차이를 이용하여 환자에 알맞은 약물과 용량을 선택할 수 있는 개인별 맞춤형 약물 선택에 대한 사회적 요구가 증가하고 있으며, 단일염기다형성(Single Nucleotide Polymorphism, SNP) 등의 유전체 정보를 마커로 활용하고, 해당 마커와 약물 반응성/약물 부작용 등의 상관관계에 대한 연구결과를 활용한 약물유전학 혹은 약물유전체학이 부상하고 있다. Therefore, there is an increasing social demand for personalized drug selection to select a drug and dose suitable for a patient by using individual genome sequence differences, and using genomic information such as single nucleotide polymorphism (SNP) as a marker. In addition, pharmacogenetics or pharmacogenomics using research results on the correlation between the markers and drug reactivity / drug side effects is emerging.
약물유전학(pharmacogenetics)에서는 일반 인구집단이나 개인에서 약물이나 화학물질의 대사와 반응 차이를 유전학적으로 분석하여 예측한다. 일부 개인에서 약물에 대해 예상했던 약물반응 이외의 반응이 나타나기도 하는데 이러한 약물 부작용은 치료하는 질환의 중증도, 약물상호작용, 환자의 나이, 영양상태, 간 및 신장 기능, 기후나 음식물과 같은 환경적 요인에 기인하기도 하지만, 약물대사에 관련된 유전적 차이, 예를 들면, 약물 효소 유전자의 다형성(polymorphism)이 영향을 미치기도 하기 때문에 이와 관련된 연구가 진행되고 있다. Pharmacogenetics predicts genetic differences in metabolism and reactions of drugs or chemicals in the general population or in individuals. In some individuals, reactions other than the expected drug response to the drug may be present. These drug side effects may be related to the severity of the disease being treated, drug interactions, the patient's age, nutritional status, liver and kidney function, climate, or food. Although it may be due to factors, genetic differences related to drug metabolism, for example, polymorphism of drug enzyme genes, may be affected.
한편, 자궁수축억제제의 일종인 리토드린(ritodrine)은 베타-아드레날린작동성 수용체 작용자(beta-adrenergic receptor agonist)로써 임신기간 22주에서 37주 사이에 사용되는 자궁수축억제제이다. 리토드린은 동일 계열의 베타-아드레날린작동성 수용체 작용자뿐만 아니라, 칼슘 채널 차단제(calcium channel blocker), 옥시토닌 길항제(Oxytocin antagonist), NSAIDs(비스테로이드성 항염증 약물), 질산염(nitrate), MgSO4, 프로게스테론(progesterone) 등 다양한 자궁수축억제제 중 가장 많이 사용되는 약물이다. 리토드린은 임산부에게 빈맥, 저혈압, 불안, 흉통, 심전도의 이상, 폐부종, 고혈당, 저칼륨혈증, 부정맥, 심근경색 등의 합병증을 유발하는 부작용을 가진 것으로 알려져있다. 선행연구에 의하면, 자궁수축억제제가 필요한 환자에서 1차 치료제로 사용된 약물의 빈도를 살펴보았을 때, 리토드린이 전체의 62%에서 사용되었다. 또한, 586명의 리토드린을 사용한 환자에서 관찰한 결과, 약물 부작용을 보인 환자는 47명으로 전체의 8%에 이르렀다. 이에 2013년 10월 25일 유럽의약품청(European Medicines Agency, EMA)은 리토드린의 부작용 때문에 사용을 제한하여 사용할 것을 권고하였으며, 국내 식품의약품안전처에서도 비슷한 권고조치가 발효된 상태이다. 따라서 리토드린으로 치료하기 위해서는 치료기간 내내 심폐기능 감시 및 심전도 모니터링과 함께 환자의 심혈관 상태에 대한 적절한 평가를 포함해야 한다. 만약 합병증이 발생하면 즉시 약제의 투여를 중단해야하고, 경우에 따라서는 다른 약제로 바꾸어야하기 때문에 조기진통 치료의 실패로 이어질 수 있다. Meanwhile, ritodrine, a type of uterine contraction inhibitor, is a beta-adrenergic receptor agonist, which is used between 22 and 37 weeks of pregnancy. Ritodrin is a class of beta-adrenergic receptor agonists, as well as calcium channel blockers, oxytonin antagonists, NSAIDs (non-steroidal anti-inflammatory drugs), nitrates, and MgSO4. , Progesterone (progesterone) is the most widely used drug among uterine contraction inhibitors. Ritodrin is known to have side effects such as tachycardia, hypotension, anxiety, chest pain, abnormal ECG, pulmonary edema, hyperglycemia, hypokalemia, arrhythmia and myocardial infarction. Previous studies have shown that lithodrin was used in 62% of patients when the frequency of drugs used as a primary treatment in patients requiring uterine contraction inhibitors was used. In addition, in 586 patients who used ritodrine, 47 patients had side effects, which accounted for 8% of the total. On October 25, 2013, the European Medicines Agency (EMA) recommended the limited use due to the side effects of ritodrine, and similar recommendations were in force in the Korean Food and Drug Administration. Therefore, treatment with ritodrine should include an appropriate assessment of the patient's cardiovascular status along with cardiopulmonary function monitoring and electrocardiogram monitoring throughout the treatment period. If complications occur, medications should be discontinued immediately and, in some cases, switched to other medications, leading to failure of early analgesia.
이에 약물 투여 전에 약물의 부작용을 예측하기 위한 방법으로 위에서 언급된 약물유전체학이 사용될 수 있다. 리토드린에 대해서도 약물 동태가 유전자형과 관련되어 있음이 밝혀져 있다. 선행연구에 따르면 리토드린의 투여 후 분만에 걸리는 시간이 ADRB2(Adrenoceptor, beta 2) 유전자의 rs1042719와 관련된 것으로 나타났다. The pharmacogenomics mentioned above may be used as a method for predicting side effects of drugs before drug administration. It has also been found that drug dynamics are associated with genotypes for ritodrine. Previous studies have shown that the time to deliver after the administration of ritodrine is related to rs1042719 of the ADRB2 (Adrenoceptor, beta 2) gene.
현재 약물의 체내 대사에 있어 수십에서 수백 개의 약물 대사 관련 유전자들의 유전 변이가 해당 단백질의 기능에 영향을 주어 약물 대사를 증가 또는 감소시킨다는 것이 알려져있고, 새로운 유전변이를 규명하는 것 역시 필요하지만 기존의 소수 유전자의 분석만으로는 다양한 약물 대사 관련 유전자들 전체를 파악하기에는 한계가 있다. 따라서 소수 유전자나 단일염기 다형성과 같은 마커를 이용한 인구 집단 관찰 연구 결과에 근거한 방법을 넘어서서, 개인 유전체 염기서열 변이 정보를 직접 활용하여 이에 수반되는 단백질 변이와 그 생물학적 영향에 대한 이론적 추론을 수행함으로써 보다 유용하고 신뢰할 수 있는 개인별 자궁수축억제제 선택을 위한 정보를 제공하는 방법론 도입의 필요성이 강하게 제기된다. It is now known that genetic variation in dozens or hundreds of drug metabolism-related genes in the body's metabolism affects the function of the protein, increasing or decreasing drug metabolism, and it is also necessary to identify new genetic variations. Analysis of minority genes alone is not enough to identify all of the genes involved in drug metabolism. Therefore, beyond the method based on population observation studies using markers such as minority genes or monobasic polymorphisms, it is possible to directly utilize personal genomic sequence variation information to perform theoretical inferences about the protein mutations and their biological effects. There is a strong need for the introduction of methodologies that provide information on the selection of useful and reliable individual contractile suppressors.
본 발명은 상기와 같은 점을 감안하여 안출된 것으로, 개인 유전체 염기서열 변이 정보를 분석하고, 자궁수축억제제의 약력학 또는 약동학에 관여하는 유전자 염기서열 변이 정보로부터 개인별 단백질 손상 점수를 산출한 후, 이를 약물과 단백질 사이의 상호 관계와 연관지어 개인별 약물 점수를 산출함으로써, 자궁수축억제제 부작용 가능성을 예측하고, 자궁수축억제제를 선택하기 위한 정보를 제공하는 방법을 제공하고자 한다.The present invention was devised in view of the above, and after analyzing individual genome sequence variation information and calculating individual protein damage scores from gene sequence variation information related to pharmacodynamics or pharmacokinetics of uterine contraction inhibitors, By calculating the individual drug scores in association with the interaction between the drug and the protein, it is intended to provide a method for predicting the possibility of adverse effects of uterine contraction inhibitors and providing information for selecting the uterine contraction inhibitors.
한 양태에서 본 발명은 개인 유전체 염기서열 정보로부터 자궁수축억제제의 약력학(pharmaco-dynamics) 또는 약동학(pharmaco-kinetics)에 관여하는 하나 이상의 유전자 염기서열 변이 정보를 결정하는 단계; 상기 유전자 염기서열 변이 정보를 이용하여 개인별 단백질 손상 점수를 산출하는 단계; 및 상기 개인별 단백질 손상 점수를 약물과 단백질 사이의 상호 관계와 연관지어, 개인별 약물 점수를 산출하는 단계를 포함하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법을 제공한다. In one aspect, the present invention comprises the steps of determining one or more gene sequence variation information involved in pharmaco-dynamics or pharmaco-kinetics of a uterine contraction inhibitor from personal genome sequence information; Calculating an individual protein damage score using the gene sequence variation information; And correlating the individual protein damage scores with the correlation between the drug and the protein to calculate the individual drug scores, thereby providing a method for providing information for selecting the uterine contraction inhibitor using the personal genome sequence variation. .
다른 양태에서 본 발명은 개인에게 적용할 수 있는 자궁수축억제제에 대하여, 상기 자궁수축억제제와 관련된 유전자 또는 단백질과 관련된 정보 검색 또는 추출이 가능한 데이터베이스; 상기 데이터베이스에 접근 가능한 통신부; 상기 정보에 기초하여 상기 자궁수축억제제의 약력학 또는 약동학에 관여하는 하나 이상의 유전자 염기서열 변이 정보를 산출하는 제1 산출모듈; 상기 유전자 염기서열 변이 정보를 이용하여 개인별 단백질 손상 점수를 산출하는 제2 산출모듈; 상기 개인별 단백질 손상 점수를 약물과 단백질 사이의 상호 관계와 연관지어, 개인별 약물 점수를 산출하는 제3 산출모듈; 및 상기 산출모듈에서 산출된 산출값을 표시하는 표시부를 포함하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 시스템을 제공한다. In another aspect, the present invention provides a uterine contraction inhibitor that can be applied to an individual, the database for retrieving or extracting information related to the gene or protein associated with the uterine contraction inhibitor; A communication unit accessible to the database; A first calculation module configured to calculate one or more gene sequence variation information related to pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor based on the information; A second calculation module for calculating an individual protein damage score using the gene sequence variation information; A third calculating module that calculates an individual drug score by associating the individual protein damage score with a correlation between a drug and a protein; And it provides a uterine contraction inhibitor selection system using a personal genome sequence variation comprising a display unit for displaying the calculated value calculated by the calculation module.
또 다른 양태에서 본 발명은 개인 유전체 염기서열 정보로부터 자궁수축억제제의 약력학 또는 약동학에 관여하는 유전자 염기서열 변이 정보를 입수하는 단계; 상기 유전자 염기서열 변이 정보를 이용하여 개인별 단백질 손상 점수를 산출하는 단계; 및 상기 개인별 단백질 손상 점수를 약물과 단백질 사이의 상호 관계와 연관지어, 개인별 약물 점수를 산출하는 단계;를 포함하는 동작을 수행하는 프로세서를 실행시키는 실행모듈을 포함하는, 컴퓨터 판독 가능한 매체를 제공한다. In another aspect, the present invention comprises the steps of obtaining genetic sequence variation information involved in the pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor from the individual genome sequence information; Calculating an individual protein damage score using the gene sequence variation information; And correlating the individual protein damage score with a correlation between the drug and the protein to calculate the individual drug score, wherein the execution module executes a processor to perform the operation. .
본 발명의 개인별 유전체 염기서열 변이 정보에 기반을 둔 개인별 자궁수축억제제 선택 방법 및 시스템은 자궁수축억제제의 약력학 또는 약동학에 관여하는 다양한 단백질을 코딩하는 유전자의 엑손 영역의 서열 분석을 통하여 개인별로 특정 약물, 즉 자궁수축억제제에 대한 반응성을 예측할 수 있는 기술로서 신뢰도가 높다. Individual uterine contraction inhibitor selection method and system based on the individual genome sequence variation information of the present invention through the sequence analysis of the exon region of the gene encoding various proteins involved in the pharmacodynamics or pharmacokinetics of the uterine contraction inhibitors In other words, the reliability of the uterine contraction inhibitors is high.
또한, 실험방법의 발전으로 유전형을 알아내기 위한 기술(PCR, SNP chip, capillary sequencing, NGS 등)을 임상적으로 사용하는 것이 용이해져 약물 유전체학에 기반을 둔 본 발명이 임상 현장에서 널리 사용될 것을 기대할 수 있다. 특히 차세대염기서열해독법(Next Generation Sequencing, NGS)의 가격이 급속하게 떨어져 환자당 수만원~수십만원 대의 가격에서 유전체 관점에서의 접근이 가능하다.In addition, the development of experimental methods will facilitate the clinical use of techniques for determining genotypes (PCR, SNP chip, capillary sequencing, NGS, etc.), and the present invention based on pharmacogenomics is expected to be widely used in clinical field. Can be. In particular, the price of Next Generation Sequencing (NGS) has fallen rapidly, allowing access from a genomic point of view at prices ranging from tens of thousands to hundreds of thousands of won per patient.
기존 약물유전체학의 인구집단간 비교를 통해 얻어진 소수 변이 또는 소수 유전자를 마커로 이용하는 방법은 연구 대상군 선정과 인구 집단 간의 차이에 따른 통계적 오류도 큰 반면, 본 발명의 방법은 수십개에서 수백개에 이르는 약물과 관련된 약동학, 약력학에 관계하는 단백질에 대하여 분자 수준의 연구 및 분석 결과를 직접 자궁수축억제제 선택에 적용하므로 인구 집단 간의 차이에 큰 영향을 받지 않고 적용할 수 있고, 소수 마커를 이용하는 것에 비해 신뢰도가 높다는 장점이 있다. The use of minor mutations or minor genes as markers in existing pharmacogenomics comparisons has a large statistical error due to the selection of the study group and the difference between the population groups, while the method of the present invention can range from tens to hundreds. Molecular-level studies and analyzes of drug-related pharmacokinetics and pharmacodynamics are applied directly to the selection of inhibitors of uterine contractions, making it possible to apply them without significantly affecting the differences between population groups. Has the advantage of being high.
특히, 본 발명에 따른 방법 및 시스템을 이용할 경우, 자궁수축억제제, 예를 들어, 리토드린에 대한 부작용 또는 위험성을 사전에 예측함으로써 개인에 적용되는 자궁수축억제제의 사용 여부 및 사용 방법을 결정하는데 유용하게 사용될 수 있으며, 이는 조기진통 환자에서 리토드린의 적정 용량 투여를 통한 조기진통 억제 치료의 성공률 향상으로 이어질 수 있다. 또한, 과거에는 약물을 견디지 못하거나 부작용으로 인해 약물을 중단해야 했던 환자군에 대해서도 리토드린 투여의 안전성을 높임으로써 새로운 치료방법을 도입할 수도 있을 것이다. In particular, when using the method and system according to the present invention, it is useful to determine whether and how to use the uterine contraction inhibitors applied to the individual by predicting the side effects or risks of uterine contraction inhibitors, for example, ritodrine in advance. This may lead to improved success rate of early analgesic suppression therapy by administering an appropriate dose of ritodrine in patients with early analgesia. In addition, it may be possible to introduce a new treatment method by increasing the safety of ritodrine administration in a patient group that could not withstand the drug or had to stop the drug due to side effects.
나아가 자궁수축억제제, 예를 들어, 리토드린의 약물-단백질 상호관계에 대한 새로운 지식이 발견되거나 제공되는 경우, 이는 본 발명의 방법에 용이하게 추가되고 적용될 수 있으므로, 향후 연구 결과 정보의 축적에 따라 보다 향상된 맞춤형 치료방법이 제공될 수 있는 장점이 있다.Further, when new knowledge is discovered or provided about the drug-protein interaction of uterine contractile inhibitors, for example, ritodrine, it can be easily added and applied to the method of the present invention, and accordingly to the accumulation of future research information There is an advantage that an improved customized treatment method can be provided.
도 1은 본 발명의 일 구현예에 따른 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법의 각 단계를 보여주는 흐름도이다. 1 is a flow chart showing each step of the method for providing information for the selection of uterine contraction inhibitors using individual genome sequence variation according to an embodiment of the present invention.
도 2는 본 발명의 일 구현예에 따른 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 시스템의 개략적 구성도이다(DB: 데이터베이스).2 is a schematic configuration diagram of a system for selecting a uterine contraction inhibitor using personal genome sequence variation according to an embodiment of the present invention (DB: database).
도 3a와 도3b는 본 발명의 일 실시예에 따른 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 방법의 검증을 위한 ROC (Receiver Operating Curve) 커브를 보여주는 흐름도이다.3A and 3B are flowcharts illustrating a receiver operating curve (ROC) curve for verification of a method for selecting a uterine contraction inhibitor using a genome sequence variation according to an embodiment of the present invention.
본 발명은 개인 유전체 염기서열 변이 정보의 분석을 통해 개인별로 자궁수축억제제를 이용한 약물치료에서 개인 맞춤형으로 안전성이 높은 약물과 용량/용법을 선택할 수 있다는 발견에 근거한 것이다. The present invention is based on the discovery that individual drugs can be selected for high-safety drugs and doses / uses in the treatment of uterine contraction inhibitors by analyzing individual genome sequence variation information.
한 양태에서 본 발명은 개인 유전체 염기서열 정보로부터 자궁수축억제제의 약력학(pharmaco-dynamics) 또는 약동학(pharmaco-kinetics)에 관여하는 하나 이상의 유전자 염기서열 변이 정보를 결정하는 단계; 상기 유전자 염기서열 변이 정보를 이용하여 개인별 단백질 손상 점수를 산출하는 단계; 및 상기 개인별 단백질 손상 점수를 약물과 단백질 사이의 상호 관계와 연관지어, 개인별 약물 점수를 산출하는 단계를 포함하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법에 관한 것이다. In one aspect, the present invention comprises the steps of determining one or more gene sequence variation information involved in pharmaco-dynamics or pharmaco-kinetics of a uterine contraction inhibitor from personal genome sequence information; Calculating an individual protein damage score using the gene sequence variation information; And calculating the individual drug score by associating the individual protein damage score with the correlation between the drug and the protein, thereby providing information for selecting the uterine contraction inhibitor using the personal genome sequence variation. .
본 발명에서 자궁수축억제제는 하기 표 1에 개시된 약물 계열에 속하는 약물, 이의 유도체, 및 이의 약학적으로 허용가능한 염 등 동일, 유사한 약리 활성을 나타내는 물질을 모두 포함하며, 이로 제한되는 것은 아니다.In the present invention, uterine contraction inhibitors include, but are not limited to, all substances that exhibit the same, similar pharmacological activity, such as drugs belonging to the drug class disclosed in Table 1, derivatives thereof, and pharmaceutically acceptable salts thereof.
약물 계열Drug class 약물drug
베타-아드레날린작동성 수용체Beta-adrenergic receptors Terbutaline, Ritodrine, Fenoterol, AlbuterolTerbutaline, Ritodrine, Fenoterol, Albuterol
칼슘 채널 차단제 Calcium channel blockers Nicardipine, NifedipineNicardipine, Nifedipine
옥시토닌 길항제Oxytonin antagonists AtosibanAtosiban
비스테로이드성 항염증 약물(NSAIDs)Nonsteroidal Anti-inflammatory Drugs (NSAIDs) Indomethacin, Ketorolac, SulindacIndomethacin, Ketorolac, Sulindac
미오신 경쇄 억제제(myosin light chain inhibitor)Myosin light chain inhibitor MgSO4MgSO4
질산염nitrate NitroglycerinNitroglycerin
프로게스테론Progesterone Hydroxyprogesterone caproate, Micronized progesteroneHydroxyprogesterone caproate, Micronized progesterone
에틸 알코올ethyl alcohol
본 발명에서 자궁수축억제제의 약력학 또는 약동학에 관여하는 유전자 정보는 DrugBank (http://www.drugbank.ca/) 또는 KEGG Drug (http://www.genome.jp/kegg/drug/) 또는 PharmGKB (https://www.pharmgkb.org/) 등과 같은 데이터베이스에서 수득할 수 있으며, 바람직하게는 BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7, TNKS, PSMD9, ZNF273, FAT4, GALNT10, OR6B1, RBBP8NL, KNDC1, UGT1A10, ARL13B, SLC15A2, SPINK6, C10orf113, TP53, TRIML2, MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT, LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EFCAB4A, CPOX, VARS2 등을 포함하나, 이로 제한되는 것은 아니다. 보다 구체적으로, 본 발명에서 자궁수축억제제의 약력학 또는 약동학에 관여하는 유전자는 BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7, TNKS, PSMD9, ZNF273, FAT4, GALNT10, OR6B1, RBBP8NL, KNDC1, UGT1A10 및 ARL13B로 이루어진 군에서 선택된 1종을 포함하며, 바람직하게는 상기 유전자에 SLC15A2, SPINK6, C10orf113, TP53, TRIML2, MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT, LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EFCAB4A, CPOX 및 VARS2로 이루어진 군에서 선택된 1종 이상을 더 포함할 수 있다. Gene information involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitor in the present invention is DrugBank (http://www.drugbank.ca/) or KEGG Drug (http://www.genome.jp/kegg/drug/) or PharmGKB (https://www.pharmgkb.org/) and the like, preferably BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7, TNKS, PSMD9, ZNF273, FAT4, GALNT10, OR6B1, RBBP8NL, KNDC1, UGT1A10, ARL13B, SLC15A2, SPINK6, C10orf1 1ML2, TPB, BA, M2N LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EFCAB4A, CPOX, VARS2 and the like, but are not limited to these. More specifically, in the present invention, the genes involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitors are BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXG1, , HHATL, SERPINA7, TNKS, PSMD9, ZNF273, FAT4, GALNT10, OR6B1, RBBP8NL, KNDC1, UGT1A10, and ARL13B, and one selected from the group consisting of SLC15A2, SPINK6, C10orf113, and TP53, TP53. , MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT, LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EFCAB4A, CPOX, and VARS2.
본 발명에서 유전자/단백질은 HGNC(HUGO Gene Nomenclature Committee)의 명명법에 따라 표시하였다(Gray KA, Daugherty LC, Gordon SM, Seal RL, Wright MW, Bruford EA. genenames.org: the HGNC resources in 2013. Nucleic Acids Res. 2013 Jan;41(Database issue):D545-52. doi: 10.1093/nar/gks1066. Epub 2012 Nov 17 PMID:23161694). In the present invention, the genes / proteins are expressed according to the nomenclature of the HUGO Gene Nomenclature Committee (HGNC) (Gray KA, Daugherty LC, Gordon SM, Seal RL, Wright MW, Bruford EA.genenames.org: the HGNC resources in 2013. Nucleic Acids Res. 2013 Jan; 41 (Database issue): D545-52. Doi: 10.1093 / nar / gks1066. Epub 2012 Nov 17 PMID: 23161694).
본 발명에서 일 정보로 사용되는 유전자 염기서열 변이는 개인의 유전자 염기서열의 변이 또는 다형성을 일컫는 것이다. 본 발명에서 유전자 염기서열 변이 또는 다형성은 자궁수축억제제의 약력학 또는 약동학과 관련된 단백질을 코딩하는 유전자 부위, 특히 엑손(exon) 부위에서 발생하는 것이나 이로 제한되는 것은 아니다. Gene sequence variation used as one information in the present invention refers to a variation or polymorphism of the individual gene sequence. In the present invention, the gene sequence mutation or polymorphism occurs at a gene region, particularly an exon region, which encodes a protein associated with pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor, but is not limited thereto.
본 발명에서 사용된 용어 “염기서열 변이 정보”는 유전자의 엑손을 구성하는 염기의 치환, 부가 또는 결실에 관한 정보를 의미한다. 이러한 염기의 치환, 부가, 또는 결실은 여러 가지 원인에 의해 발생할 수 있으며, 예를 들면 염색체의 돌연변이, 절단, 결실, 중복, 역위 및/또는 전좌를 포함하는 구조적 차이에 의할 수 있다. The term "base sequence variation information" used in the present invention means information about the substitution, addition or deletion of the base constituting the exon of the gene. Substitution, addition, or deletion of such bases can occur for a variety of reasons, for example, by structural differences including mutations, truncation, deletions, duplications, inversions and / or translocations of chromosomes.
다른 측면에서 염기서열의 다형성이란 유전체 상에 존재하는 염기서열의 개인 간 차이를 말하는 것으로, 염기서열 다형성에서 그 수가 가장 많은 것은 단일염기다형성(Single Nucleotide Polymorphism, SNP)이며, A, T, C, G로 이루어진 염기서열 중 하나의 염기에 개인 간 차이가 있는 것이다. 염기서열 다형성은 SNP를 포함하여 SNV(Single Nucleotide Variation), STRP(short tandem repeat polymorphism), 또는 VNTR(various number of tandem repeat) 및 CNV(Copy number variation)를 포함하는 다수체(polyalleic) 변이의 형태로 나타날 수 있다. In another aspect, nucleotide polymorphism refers to differences between individuals of nucleotide sequences present in the genome, and the largest number of nucleotide polymorphisms is Single Nucleotide Polymorphism (SNP), and A, T, C, There is a difference between individuals in one base of the base sequence consisting of G. Sequence polymorphism is a form of polyalleic variation including single nucleotide variation (SNV), short tandem repeat polymorphism (STRP), or variable number of tandem repeat (VNTR) and copy number variation (CNV), including SNPs. May appear.
본 발명의 방법에서 개인의 유전체에서 발견되는 염기서열 변이 또는 다형성 정보는 자궁수축억제제의 약력학 또는 약동학과 관련된 단백질과 관련되어 수집된다. 즉, 본 발명의 방법에 사용되는 염기서열 변이 정보는 수득한 개인의 유전체 염기서열 정보 중 자궁수축억제제의 약력학 또는 약동학에 관여하는 하나 이상의 유전자, 예를 들면, 약물과 관련된 표적(target) 단백질, 약물 대사에 관여하는 효소(enzyme) 단백질, 수송체 단백질(transporter) 또는 운반체(carrier) 단백질을 코딩하는 유전자의 특히 엑손 영역에서 발견되는 변이 정보이나 이로 제한되는 것은 아니다. In the method of the present invention, sequence variation or polymorphism information found in an individual's genome is collected in association with proteins associated with pharmacodynamics or pharmacokinetics of uterine contraction inhibitors. That is, the nucleotide sequence information used in the method of the present invention is one or more genes involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitors, such as a target protein associated with the drug, Mutation information found in the exon region, in particular but not limited to, genes encoding enzyme proteins, transporter proteins or carrier proteins involved in drug metabolism.
본 발명에서 사용되는 개인의 유전체 염기서열 정보는 공지된 염기서열해독법을 이용하여 결정될 수 있으며, 또한 상용화된 서비스를 제공하는 Complete Genomics, BGI (Beijing Genome Institute), Knome, Macrogen, DNALink 등의 서비스를 이용할 수 있으며, 이에 제한되지 않는다. The genome sequence information of an individual used in the present invention may be determined using a known sequence decoding method, and services such as Complete Genomics, BGI (Beijing Genome Institute), Knome, Macrogen, DNALink, etc., which provide commercially available services. May be used, but is not limited thereto.
본 발명에서 개인의 유전체 염기서열에 존재하는 유전자 염기서열 변이 정보는 다양한 방법을 이용하여 추출될 수 있으며, 참조군, 예를 들면 HG19의 유전체 염기서열과의 서열 비교 프로그램, 예를 들어, ANNOVAR(Wang et al., Nucleic Acids Research, 2010; 38(16): e164), SVA(Sequence Variant Analyzer) (Ge et al., Bioinformatics. 2011; 27(14): 19982000), BreakDancer(Chen et al., Nat Methods. 2009 Sep; 6(9):677-81) 등을 이용한 염기서열 비교 분석을 통해 수득될 수 있다. Gene sequence variation information present in the genome sequence of an individual in the present invention can be extracted using a variety of methods, a sequence comparison program with a genomic sequence of a reference group, for example HG19, for example, ANNOVAR ( Wang et al., Nucleic Acids Research, 2010; 38 (16): e164), Sequence Variant Analyzer (SVA) (Ge et al., Bioinformatics. 2011; 27 (14): 19982000), Break Dancer (Chen et al., Nat Methods.2009 Sep; 6 (9): 677-81) and the like.
상기 유전자 염기서열 변이 정보는 컴퓨터 시스템을 통하여 접수/수득될 수 있으며, 이런 측면에서 본 발명의 방법은 유전자 변이 정보를 컴퓨터 시스템으로 접수하는 단계를 추가로 포함할 수 있다. 본 발명에서 사용되는 컴퓨터 시스템은 상기 자궁수축억제제의 약력학 또는 약동학에 관여하는 유전자, 예를 들면 약물과 관련된 표적 단백질, 약물 대사에 관여하는 효소 단백질, 수송체 단백질 또는 운반체 단백질 등을 코딩하는 유전자에 관한 정보를 포함하는 하나 이상의 데이터베이스를 포함하거나 데이터베이스에 접근 가능하다. 이러한 데이터베이스는 예를 들면 DrugBank (http://www.drugbank.ca/), KEGG Drug (http://www.genome.jp/kegg/drug/), PharmGKB (http://www.pharmgkb.org/) 등을 포함하는 유전자/단백질/약물-단백질 상호작용 등에 관한 정보를 제공하는 공개 또는 비공개 데이터베이스 또는 지식베이스를 포함할 수 있으나, 이에 제한되지 않는다. The gene sequence variation information may be received / obtained through a computer system, and in this aspect, the method of the present invention may further include receiving the genetic variation information into a computer system. The computer system used in the present invention is a gene involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitors, for example, a gene encoding a target protein associated with a drug, an enzyme protein involved in drug metabolism, a transporter protein or a carrier protein. Include or have access to one or more databases containing information about them. Such databases include, for example, DrugBank (http://www.drugbank.ca/), KEGG Drug (http://www.genome.jp/kegg/drug/), PharmGKB (http://www.pharmgkb.org /) May include, but is not limited to, public or private databases or knowledge bases that provide information about genes / proteins / drug-protein interactions, and the like.
본 발명에서 자궁수축억제제는 사용자가 입력한 정보, 처방(전)으로부터 입력된 정보, 또는 자궁수축억제제에 대한 정보를 포함하는 데이터베이스로부터 입력된 정보일 수 있다. 상기 처방전은 전자처방전을 포함하며, 이에 제한되지 않는다.In the present invention, the uterine contraction inhibitor may be information input by a user, information input from a prescription (pre), or information input from a database including information on the uterine contraction inhibitor. The prescription includes, but is not limited to, electronic prescription.
본 발명에서 사용된 용어 "약동학(pharmaco-kinetics, pk)" 또는 "약동학적 파라미터"는 일정 시간동안 약물의 체내에서의 흡수, 이동, 분포, 전환, 배출과 관련된 약물의 특성을 일컫는 것으로, 약물의 분포용적(Vd), 청소율(CL), 생체이용율(F), 흡수속도계수(ka), 최대 혈중 약물 농도(maximum plasma concentration, Cmax), 최대 혈중 약물 농도의 도달시간(time point of maximum plasma concentration, Tmax), 일정 시간 동안의 혈중 약물 농도 변화에 대한 그래프 아래의 면적(AUC, Area Under the Curve) 측정 등을 포함하는 것이다. As used herein, the term "pharmaco-kinetics (pk)" or "pharmacokinetic parameters" refers to the properties of a drug associated with absorption, migration, distribution, conversion, and excretion of the drug in the body over time. Distribution volume (Vd), clearance (CL), bioavailability (F), absorption rate coefficient (ka), maximum plasma concentration (Cmax), and time point of maximum plasma concentration, Tmax), area under the curve (AUC) measurements of changes in blood drug concentration over time.
본 발명에서 사용된 용어 “약력학(pharmacodynamics) 또는 약력학적 파라미터”는 약물의 생체에 대한 생리학적 및 생화학적 작용과 그 작용기전, 즉 약물이 일으키는 생체의 반응 또는 효과와 관련된 특징을 일컫는 것이다. As used herein, the term “pharmacodynamics or pharmacodynamic parameters” refers to the characteristics related to the physiological and biochemical action of the drug on the living body and its mechanism of action, ie the reaction or effect of the living body of the drug.
본 발명에서 사용된 용어 “유전자 염기서열 변이 점수”란 유전체 염기서열 변이가 단백질을 코딩하는 유전자의 엑손 부위에서 발견되었을 때, 이러한 개별 변이가 해당 유전자가 코딩하는 단백질의 아미노산 서열 변이(치환, 부가 또는 결실) 또는 전사 조절 변이를 초래하여, 해당 단백질의 구조 및/또는 기능에 유의한 변화 혹은 손상을 유발하는 정도를 수치화한 점수를 말하며, 상기 유전자 염기서열 변이 점수는 유전체 염기서열 상에서 아미노산의 진화론적 보존 정도, 변형된 아미노산의 물리적 특성에 따른 해당 단백질의 구조나 기능의 변화에 미치는 정도 등을 고려하여 산출할 수 있다. As used herein, the term “gene sequence variation score” refers to an amino acid sequence variation (substitution or addition) of a protein encoded by the gene when the genome sequence variation is found in the exon region of the gene encoding the protein. Or deletion) or a score that quantifies the extent to which transcriptional control mutations result, thereby causing significant changes or damage to the structure and / or function of the protein, wherein the gene sequence variation score is the evolution of amino acids on the genome sequence It can be calculated in consideration of the degree of preservation and the degree of change in the structure or function of the protein according to the physical properties of the modified amino acid.
본 발명에서 개인별 단백질 손상 점수 또는 개인별 약물 점수 산출에 적용하기 위해, 유전자 염기서열 변이 점수를 산출하는 방법은 당업계에 공지된 방법을 이용하여 수행될 수 있다. 예를 들면, SIFT (Sorting Intolerant From Tolerant, Pauline C et al., Genome Res. 2001 May; 11(5): 863874; Pauline C et al., Genome Res. 2002 March; 12(3): 436446; Jing Hul et al., Genome Biol. 2012; 13(2): R9), PolyPhen, PolyPhen-2 (Polymorphism Phenotyping, Ramensky V et al., Nucleic Acids Res. 2002 September 1; 30(17): 38943900; Adzhubei IA et al., Nat Methods 7(4):248-249 (2010)), MAPP (Eric A. et al., Multivariate Analysis of Protein Polymorphism, Genome Research 2005;15:978986), Logre (Log R Pfam E-value, Clifford R.J et al., Bioinformatics 2004;20:1006-1014), Mutation Assessor (Reva B et al., Genome Biol. 2007;8:R232, http://mutationassessor.org/), Condel (Gonzalez-Perez A et al.,The American Journal of Human Genetics 2011;88:440449, http://bg.upf.edu/fannsdb/), GERP (Cooper et al., Genomic Evolutionary Rate Profiling, Genome Res. 2005;15:901-913, http://mendel.stanford.edu/SidowLab/downloads/gerp/), CADD (Combined Annotation-Dependent Depletion, http://cadd.gs.washington.edu/), MutationTaster, MutationTaster2 (Schwarz et al., MutationTaster2: mutation prediction for the deep-sequencing age. Nature Methods 2014;11:361362, http://www.mutationtaster.org/), PROVEAN (Choi et al., PLoS One. 2012;7(10):e46688), PMut (Ferrer-Costa et al., Proteins 2004;57(4):811-819, http://mmb.pcb.ub.es/PMut/), CEO (Combinatorial Entropy Optimization, Reva et al., Genome Biol 2007;8(11):R232), SNPeffect (Reumers et al., Bioinformatics. 2006;22(17):2183-2185, http://snpeffect.vib.be), fathmm (Shihab et al., Functional Analysis through Hidden Markov Models, Hum Mutat 2013;34:57-65, http://fathmm.biocompute.org.uk/) 등과 같은 알고리즘을 이용하여 유전자 염기서열 변이 정보로부터 유전자 염기서열 변이 점수를 산출할 수 있으며, 이에 제한되지 않는다. In order to apply to the calculation of individual protein damage score or individual drug score in the present invention, the method of calculating the gene sequence variation score may be performed using a method known in the art. See, eg, SIFT (Sorting Intolerant From Tolerant, Pauline C et al., Genome Res. 2001 May; 11 (5): 863874; Pauline C et al., Genome Res. 2002 March; 12 (3): 436446; Jing Hul et al., Genome Biol. 2012; 13 (2): R9), PolyPhen, PolyPhen-2 (Polymorphism Phenotyping, Ramensky V et al., Nucleic Acids Res. 2002 September 1; 30 (17): 38943900; Adzhubei IA et al., Nat Methods 7 (4): 248-249 (2010)), MAPP (Eric A. et al., Multivariate Analysis of Protein Polymorphism, Genome Research 2005; 15: 978986), Logre (Log R Pfam E- value, Clifford RJ et al., Bioinformatics 2004; 20: 1006-1014), Mutation Assessor (Reva B et al., Genome Biol. 2007; 8: R232, http://mutationassessor.org/), Condel (Gonzalez- Perez A et al., The American Journal of Human Genetics 2011; 88: 440449, http://bg.upf.edu/fannsdb/), GERP (Cooper et al., Genomic Evolutionary Rate Profiling, Genome Res. 2005; 15; : 901-913, http://mendel.stanford.edu/SidowLab/downloads/gerp/), CADD (Combined Annotation-Dependent Depletion, http://cadd.gs.washington.edu MutationTaster, MutationTaster2 (Schwarz et al., MutationTaster2: mutation prediction for the deep-sequencing age. Nature Methods 2014; 11: 361362, http://www.mutationtaster.org/), PROVEAN (Choi et al., PLoS One. 2012; 7 (10): e46688), PMut (Ferrer-Costa et al., Proteins 2004; 57 (4): 811-819, http://mmb.pcb.ub.es/PMut/), CEO (Combinatorial Entropy Optimization, Reva et al., Genome Biol 2007; 8 (11): R232), SNP effect (Reumers et al., Bioinformatics. 2006; 22 (17): 2183-2185, http://snpeffect.vib.be), fathmm (Shihab et al., Functional Analysis through Hidden Markov Models, Hum Mutat 2013; 34 : 57-65, http://fathmm.biocompute.org.uk/) may be used to calculate the gene sequence variation score from the gene sequence variation information, but is not limited thereto.
상술된 알고리즘들의 목적은 각각의 유전자 염기서열 변이가 단백질 기능에 얼마나 영향을 미치고, 이 영향이 단백질에 얼마나 손상을 주게 되는지, 혹은 별다른 영향이 없는지 가려내기 위함이다. 이들은 기본적으로 개별 유전자 염기서열 변이가 해당 유전자가 코딩하는 단백질의 아미노산 서열에 초래할 변화를 고려하여 해당 단백질의 구조 및/또는 기능에 미칠 영향을 판단한다는 점에서 공통점이 있다.The purpose of the algorithms described above is to determine how each gene sequence mutation affects protein function, how this damage damages the protein, or whether there is little effect. They have a common point in that they determine the impact of individual gene sequence variations on the structure and / or function of the protein in consideration of the changes that will result in the amino acid sequence of the protein encoded by the gene.
본 발명에 따른 일 구현예에서는 개별 유전자 염기서열 변이 점수를 산출하기 위하여, SIFT (Sorting Intolerant From Tolerant) 알고리즘을 이용하였다. SIFT 알고리즘의 경우, 예를 들면, VCF (Variant Call Format) 형식 파일로 유전자 염기서열 변이 정보를 입력받아, 각각의 유전자 염기서열 변이가 해당 유전자를 손상시키는 정도를 점수화 한다. SIFT 알고리즘의 경우 산출 점수가 0에 가까울수록 해당 유전자가 코딩하는 단백질의 손상이 심해서 해당 기능이 손상됐을 것으로 판단하고, 1에 가까울수록 해당 유전자가 코딩하는 단백질이 정상 기능을 유지하고 있을 것으로 판단한다. In one embodiment according to the present invention, a Sorting Intolerant From Tolerant (SIFT) algorithm was used to calculate an individual gene sequence variation score. In the SIFT algorithm, for example, gene sequence variation information is input to a VCF (Variant Call Format) format file, and each gene sequence variation is scored for damaging the gene. In the case of the SIFT algorithm, the closer the output score is to 0, the more likely the function of the protein encoded by the gene is impaired, and the closer the value is to 1, the more likely that the protein encoded by the gene is maintaining normal function. .
또 다른 알고리즘인 PolyPhen-2의 경우, 산출 점수가 높을수록 해당 유전자가 코딩하는 단백질의 기능적 손상 정도가 큰 것으로 판단한다. In another algorithm, PolyPhen-2, the higher the score, the greater the degree of functional damage of the protein encoded by the gene.
최근에는 SIFT, Polyphen2, MAPP, Logre, Mutation Assessor를 서로 비교하고 종합하여 Condel 알고리즘을 제시한 연구(GonzaeA. & LoN. Improving the assessment of the outcome of nonsynonymous SNVs with a consensus deleteriousness score, Condel. The American Journal of Human Genetics, 2011;88(4):440-449.)가 발표되었으며, 상기 연구에서는 단백질에 손상을 주는 유전자 염기서열 변이 및 영향이 적은 유전자 염기서열 변이와 관련하여 공지된 데이터의 집합인 HumVar와 HumDiv(Adzhubei, IAet al., A method and server for predicting damaging missense mutations. Nature methods, 2010;7(4):248-249)를 사용하여 상기 다섯 개의 알고리즘을 비교하였다. 그 결과, HumVar의 97.9%의 단백질 손상을 일으키는 유전자 염기서열 변이와 97.3%의 영향이 적은 유전자 염기서열 변이가 상기 다섯 개의 알고리즘 중 최소 세 개의 알고리즘에서 동일하게 감지되었으며, HumDiv의 99.7%의 단백질 손상을 일으키는 유전자 염기서열 변이와 98.8%의 영향이 적은 유전자 염기서열 변이가 상기 다섯 개의 알고리즘 중 최소 세 개의 알고리즘에서 동일하게 감지되었다. 또한, 상기 HumDiv와 HumVar에 대하여 상기 다섯 개의 알고리즘과 각 알고리즘을 통합하여 계산한 결과들의 정확도를 나타내는 ROC (Receiver Operating Curve) 곡선을 그려본 결과, 상당히 높은 수준(69%~88.2%)에서 AUC(Area Under the Receiver Operating Curve)의 일치도를 보이는 것을 확인하였다. 즉 상술한 다양한 알고리즘들은 그 산출 방법은 달라도 산출된 유전자 염기서열 변이 점수들은 서로 유의하게 상관된 것이다. 따라서 상술한 알고리즘들 또는 알고리즘들을 응용한 방법을 적용하여 산출된 유전자 염기서열 변이 점수를 본원 발명에 의한 개인별 단백질 손상 점수 및 개인별 약물 점수 산출 단계에 적용하는 것은 각 유전자 염기서열 변이 점수를 산출하는 서로 다른 알고리즘의 종류에 상관없이 본 발명의 범위에 속하는 것이다. Recently, a study that compared and synthesized SIFT, Polyphen2, MAPP, Logre, and Mutation Assessor to present the Condel algorithm (Gonzae A. & LoN. Improving the assessment of the outcome of nonsynonymous SNVs with a consensus deleteriousness score, Condel.The American Journal of Human Genetics, 2011; 88 (4): 440-449.), in the study HumVar, a set of known data relating to gene sequencing and low impact gene sequencing that damage proteins. And the five algorithms were compared using HumDiv (Adzhubei, IA et al., A method and server for predicting damaging missense mutations.Nature methods, 2010; 7 (4): 248-249). As a result, gene sequence mutations that cause protein damage of 97.9% of HumVar and gene sequence mutations of less than 97.3% were detected identically in at least three of the five algorithms, and 99.7% protein damage of HumDiv. Gene sequencing mutations and gene sequencing mutations with less influence of 98.8% were detected in at least three of the five algorithms. In addition, as a result of drawing a receiver operating curve (ROC) curve representing the accuracy of the results obtained by integrating the five algorithms and the respective algorithms with respect to the HumDiv and HumVar, the AUC (69% ~ 88.2%) Area Under the Receiver Operating Curve) was confirmed to show the agreement. That is, the above-described various algorithms have different correlation methods, but the calculated gene sequence variation scores are significantly correlated with each other. Therefore, applying the gene sequence variation scores calculated by applying the above-described algorithms or algorithms to the individual protein damage scores and the individual drug score calculation step according to the present invention is to calculate each gene sequence variation score. It is within the scope of the present invention regardless of the type of other algorithm.
유전자 염기서열 변이가 단백질을 코딩하는 유전자의 엑손 부위에 발생할 경우, 이는 단백질의 구조 및/또는 기능에 직접적인 영향을 미칠 수 있다. 따라서 상기 유전자 염기서열 변이 정보를 단백질 기능 손상 정도와 관련시킬 수 있다. 이런 측면에서 본 발명의 방법은 다음의 단계에서, 상술한 유전자 염기서열 변이 점수를 기반으로 개인별 단백질 손상 점수를 산출한다. When genetic sequence variation occurs in the exon region of a gene encoding a protein, it may directly affect the structure and / or function of the protein. Therefore, the gene sequence variation information may be related to the degree of impairment of protein function. In this respect, the method of the present invention calculates individual protein damage scores based on the gene sequence variation scores described above in the next step.
본 발명에서 사용된 용어“단백질 손상 점수”란 하나의 단백질을 코딩하는 유전자 부위에 두 개 이상의 유의한 염기서열 변이가 발견되어, 하나의 단백질이 두 개 이상의 유전자 염기서열 변이 점수를 갖게 되는 경우, 상기 유전자 염기서열 변이 점수를 종합하여 계산된 점수를 말하며, 만약 단백질을 코딩하는 유전자 부위에 유의한 염기서열 변이가 한 개인 경우에는 유전자 염기서열 변이 점수와 단백질 손상 점수가 동일하다. 이때, 단백질을 코딩하는 유전자 염기서열 변이가 두 개 이상인 경우, 단백질 손상 점수는 각 변이별로 계산된 유전자 염기서열 변이 점수들의 평균값으로 계산되며, 이러한 평균값은 예를 들면 기하평균, 산술평균, 조화평균, 산술기하평균, 산술조화평균, 기하조화평균, 피타고라스 평균, 사분평균, 이차평균, 절삭평균, 윈저화 평균, 가중평균, 가중기하평균, 가중산술평균, 가중조화평균, 함수의 평균, 멱평균, 일반화된 f-평균, 백분위수, 최대값, 최소값, 최빈값, 중앙값, 중앙범위, 중심경향도(measures of central tendency), 단순 곱 또는 가중곱, 또는 상기 산출값들의 함수 연산으로 계산될 수 있으나, 이에 제한되지 않는다. As used herein, the term “protein damage score” means that two or more significant sequence mutations are found in a gene region encoding one protein, so that one protein has two or more gene sequence mutation scores. Refers to a score calculated by combining the gene sequence variation score. If there is a significant sequence variation in a gene region encoding a protein, the gene sequence variation score and the protein damage score are the same. In this case, when there are two or more gene sequence mutations encoding a protein, the protein damage score is calculated as an average value of the gene sequence variation scores calculated for each variation, and the average value is, for example, a geometric mean, an arithmetic mean, or a harmonic mean. , Arithmetic geometric mean, arithmetic harmonic mean, geometric harmonic mean, Pythagorean mean, quadrant mean, quadratic mean, cutting mean, windsorized mean, weighted mean, weighted geometric mean, weighted arithmetic mean, weighted harmonic mean, function mean, 멱 average Can be computed as a generalized f-means, percentiles, maximums, minimums, modes, medians, median ranges, measures of central tendency, simple products or weighted products, or as a function of these calculations. This is not restrictive.
본 발명에 따른 일 구현예에서는 하기 수학식 1에 의해 단백질 손상 점수를 산출하였으며, 하기 수학식 1은 다양한 변형이 가능하고 이에 제한되지 않는다. In one embodiment according to the present invention, the protein damage score was calculated by Equation 1 below, and Equation 1 may be variously modified and is not limited thereto.
Figure PCTKR2016001631-appb-M000001
Figure PCTKR2016001631-appb-M000001
상기 수학식 1에서 Sg는 유전자 g가 코딩하는 단백질의 단백질 손상 점수, n은 상기 유전자 g의 염기서열 변이 중 분석대상 염기서열 변이의 수, vi는 i 번째 유전자 염기서열 변이의 유전자 염기서열 변이 점수이고, p는 0이 아닌 실수이다. 상기 수학식 1에서 상기 p의 값이 1일 때는 산술평균, 상기 p의 값이 -1일 때는 조화평균이 되며, 상기 p의 값이 0에 가까워지는 극한의 경우에는 기하평균이 된다.In Equation 1, Sg is the protein damage score of the protein encoded by the gene g, n is the number of the nucleotide sequence analysis of the nucleotide variation of the gene g, vi is the gene sequence variation score of the i-th gene sequence variation And p is a nonzero real number. In Equation 1, when the value of p is 1, it is an arithmetic mean, and when the value of p is -1, it is a harmonic mean, and in the extreme case where the value of p is close to 0, it is a geometric mean.
본 발명에 따른 또 다른 일 구현예에서는 하기 수학식 2에 의해 단백질 손상 점수를 산출하였으며, 하기 수학식 2는 다양한 변형이 가능하고 이에 제한되지 않는다. In another embodiment according to the present invention, the protein damage score was calculated by Equation 2 below, and Equation 2 may be variously modified and is not limited thereto.
Figure PCTKR2016001631-appb-M000002
Figure PCTKR2016001631-appb-M000002
상기 수학식 2에서 Sg는 유전자 g가 코딩하는 단백질의 단백질 손상 점수, n은 상기 유전자 g의 염기서열 변이 중 분석대상 염기서열 변이의 수, vi는 i 번째 유전자 염기서열 변이의 유전자 염기서열 변이 점수, wi는 상기 vi에 부여되는 가중치이다. 모든 가중치 wi가 같은 값을 갖는 경우 상기 단백질 손상 점수 Sg는 상기 유전자 염기서열 변이 점수 vi의 기하평균 값이 된다. 상기 가중치는 해당 단백질의 종류, 해당 단백질의 약동학적 또는 약력학적 분류, 해당 약물 효소 단백질의 약동학적 파라미터, 인구 집단 또는 인종별 분포를 고려하여 부여될 수 있다. In Equation 2, Sg is the protein damage score of the protein encoded by the gene g, n is the number of the nucleotide sequence analysis of the nucleotide sequence variation of the gene g, vi is the gene sequence variation score of the i-th gene sequence variation , wi is a weight given to vi. When all weights wi have the same value, the protein damage score Sg becomes the geometric mean value of the gene sequence variation score vi. The weight may be given in consideration of the type of the protein, the pharmacokinetic or pharmacodynamic classification of the protein, the pharmacokinetic parameters of the drug enzyme protein, and the population or race distribution.
본 발명에서 사용된 용어 “약물 대사 효소의 약동학적 파라미터”는 Vmax, Km, Kcat/Km 등을 포함하는 것이다. Vmax는 기질 농도가 매우 높을 때의 최대 효소 반응 속도이고, Km은 해당 반응이 1/2 Vmax에 도달하게 하는 기질의 농도이다. Km은 해당 효소와 해당 기질 간의 친화도로 볼 수 있으며, Km이 작을수록 해당 효소와 해당 기질 간의 결합이 강하다. 효소의 대사회전수라고도 불리는 Kcat은 해당 효소가 최대 속도로 활동하고 있을 때 각 효소 활성 부위 당 1초의 시간에 대사되는 기질 분자의 개수를 의미하며, 해당 효소 반응이 실제 얼마나 빠르게 일어나는지를 의미한다.The term "pharmacokinetic parameters of drug metabolizing enzyme" as used herein includes Vmax, Km, Kcat / Km and the like. Vmax is the maximum enzyme reaction rate when the substrate concentration is very high, and Km is the concentration of the substrate that causes the reaction to reach 1/2 Vmax. Km can be seen as an affinity between the enzyme and the substrate. The smaller the Km, the stronger the bond between the enzyme and the substrate. Kcat, also called the enzyme's metabolic rate, refers to the number of substrate molecules that are metabolized at one second per enzyme active site when the enzyme is active at its maximum rate, and how fast the enzyme reaction actually occurs.
본 발명의 방법은 다음 단계에서 상술한 단백질 손상 점수를 약물과 단백질 사이의 상호 관계와 연관지어, 개인별 약물 점수를 산출한다. The method of the present invention correlates the protein damage score described above in the next step with the correlation between the drug and the protein to yield an individual drug score.
본 발명에서 사용된 용어“약물 점수”란 소정의 약물, 예를 들어 자궁수축억제제가 주어졌을 때, 해당 약물의 약력학 또는 약동학에 관여하는 표적 단백질, 약물 대사에 관여하는 효소 단백질, 수송체 단백질 또는 운반체 단백질들을 찾아내어, 해당 단백질들의 단백질 손상 점수를 계산한 후, 이를 다시 종합하여 하나의 약물에 대하여 산출된 값을 말한다. As used herein, the term “drug score” refers to a given protein, eg, a target protein involved in the pharmacodynamics or pharmacokinetics of a given drug, an enzyme protein involved in drug metabolism, a transporter protein or Carrier proteins are found, the protein damage scores of the proteins are calculated, and then summed again to refer to the values calculated for one drug.
본 발명에서 약물 점수는 자궁수축억제제의 약력학 또는 약동학에 관여하는 단백질의 손상이 두 개 이상인 경우, 상기 단백질 손상 점수들의 평균값으로 계산되며, 이러한 평균값은 예를 들면 기하평균, 산술평균, 조화평균, 산술기하평균, 산술조화평균, 기하조화평균, 피타고라스 평균, 사분평균, 이차평균, 절삭평균, 윈저화 평균, 가중평균, 가중기하평균, 가중산술평균, 가중조화평균, 함수의 평균, 멱평균, 일반화된 f-평균, 백분위수, 최대값, 최소값, 최빈값, 중앙값, 중앙범위, 중심경향도(measures of central tendency), 또는 단순 곱 또는 가중곱, 또는 상기 산출값들의 함수 연산으로 계산될 수 있으나, 이에 제한되지 않는다. In the present invention, the drug score is calculated as an average value of the protein damage scores when the damage of the protein involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitors, for example, geometric mean, arithmetic mean, harmonic mean, Arithmetic geometric mean, Arithmetic harmonic mean, Geometric harmonic mean, Pythagorean mean, Quarter mean, Secondary mean, Cutting mean, Windsorized mean, Weighted mean, Weighted geometric mean, Weighted arithmetic mean, Weighted harmonic mean, Function mean, 멱 mean, Generalized f-means, percentiles, maximums, minimums, modes, medians, median ranges, measures of central tendency, or simple products or weighted products, or arithmetic operations of these calculations This is not restrictive.
상기 약물 점수는 약물학적 특성을 고려하여 해당 약물, 즉 자궁수축억제제의 약력학 또는 약동학에 관여하는 표적 단백질, 약물 대사에 관여하는 효소 단백질, 수송체 단백질 및 운반체 단백질의 가중치를 조율하여 산출될 수 있으며, 상기 가중치는 해당 약물 대사 효소의 약동학적 파라미터, 인구 집단 또는 인종별 분포 등을 고려하여 부여될 수 있다. 또한, 해당 약물과 직접 상호작용하지는 않지만, 해당 약물의 전구체 또는 해당 약물의 대사산물들과 상호작용하는 단백질, 예를 들면, 약물학적 패스웨이를 구성하는 단백질들의 단백질 손상 점수도 함께 고려하여 약물 점수를 산출할 수 있다. 또한, 해당 약물의 약력학 또는 약동학에 관여하는 단백질들과 유의하게 상호작용하는 단백질들의 단백질 손상 점수를 함께 고려하여 약물 점수를 산출할 수 있다. 상기 해당 약물의 약물학적 패스웨이를 구성하거나, 패스웨이를 구성하는 단백질들과 유의하게 상호작용하거나, 그 신호전달경로에 참여하는 단백질에 관한 정보는 PharmGKB (Whirl-Carrillo et al., Clinical Pharmacology & Therapeutics 2012;92(4):414-4171), The MIPS Mammalian Protein-Protein Interaction Database (Pagel etl al., Bioinformatics 2005;21(6):832-834), BIND (Bader et al., Biomolecular Interaction Network Database, Nucleic Acids Res. 2003 Jan 1;31(1):248-50), Reactome (Joshi-Tope et al., Nucleic Acids Res. 2005 Jan 1;33(Database issue):D428-32) 등의 공지된 생물학적 데이터베이스에서 검색할 수 있다. The drug score may be calculated by adjusting the weights of the drug, that is, the target protein involved in the pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor, the enzyme protein involved in the drug metabolism, the transporter protein, and the carrier protein in consideration of pharmacological properties. The weight may be given in consideration of the pharmacokinetic parameters of the corresponding drug metabolizing enzyme, the population group or the race distribution. In addition, drug scores are also taken into account when considering protein damage scores of proteins that do not interact directly with the drug but interact with precursors or metabolites of the drug, such as proteins that make up the pharmacological pathway. Can be calculated. In addition, the drug score may be calculated by considering the protein damage scores of the proteins that significantly interact with the proteins involved in the pharmacodynamics or pharmacokinetics of the drug. For information on the proteins that make up the pharmacological pathway of the drug, or which significantly interact with the proteins that make up the pathway, or participate in the signaling pathway, see PharmGKB (Whirl-Carrillo et al., Clinical Pharmacology & Therapeutics 2012; 92 (4): 414-4171), The MIPS Mammalian Protein-Protein Interaction Database (Pagel etl al., Bioinformatics 2005; 21 (6): 832-834), BIND (Bader et al., Biomolecular Interaction Network Database, Nucleic Acids Res. 2003 Jan 1; 31 (1): 248-50), Reactome (Joshi-Tope et al., Nucleic Acids Res. 2005 Jan 1; 33 (Database issue): D428-32), etc. Searched biological databases.
본 발명에 따른 일 구현예에서는 하기 수학식 3에 의해 약물 점수를 산출하였으며, 하기 수학식 3은 다양한 변형이 가능하므로, 이에 제한되지 않는다. In one embodiment according to the present invention, the drug score was calculated by the following Equation 3, and the following Equation 3 may be variously modified, but is not limited thereto.
Figure PCTKR2016001631-appb-M000003
Figure PCTKR2016001631-appb-M000003
상기 수학식 3에서 Sd는 약물 d의 약물 점수, n은 상기 약물 d의 약력학 또는 약동학에 직접 관여하거나 해당 약물의 전구체 또는 해당 약물의 대사산물들과 상호작용하는 단백질, 예를 들면, 약물학적 패스웨이에 참여하는 유전자 군에서 선정된 하나 이상의 유전자가 코딩하는 단백질의 수, gi는 상기 약물 d의 약력학 또는 약동학에 직접 관여하거나 해당 약물의 전구체 또는 해당 약물의 대사산물들과 상호작용하는 단백질, 예를 들면, 약물학적 패스웨이에 참여하는 유전자 군에서 선정된 하나 이상의 유전자가 코딩하는 단백질의 단백질 손상 점수이며, p는 0이 아닌 실수이다. 상기 수학식 3에서 상기 p의 값이 1일 때는 산술평균, 상기 p의 값이 -1일 때는 조화평균이 되며, 상기 p의 값이 0에 가까워지는 극한의 경우에는 기하평균이 된다.In Equation 3, Sd is a drug score of drug d, n is a protein directly involved in the pharmacodynamics or pharmacokinetics of drug d or interacts with a precursor of the drug or metabolites of the drug, for example, a pharmacological pass. The number of proteins encoded by one or more genes selected from the group of genes participating in the way, gi is a protein that directly participates in the pharmacodynamics or pharmacokinetics of drug d or interacts with precursors or metabolites of the drug, eg For example, the protein damage score of a protein encoded by one or more genes selected from a group of genes participating in pharmacological pathways, where p is a nonzero real number. In Equation 3, when the value of p is 1, it is an arithmetic mean, and when the value of p is -1, it is a harmonic mean, and in the extreme case where the value of p is close to 0, it is a geometric mean.
본 발명에 따른 또 다른 일 구현예에서는 하기 수학식 4에 의해 약물 점수를 산출하였으며, 하기 수학식 4는 다양한 변형이 가능하므로, 이에 제한되지 않는다. In another embodiment according to the present invention, the drug score was calculated by Equation 4 below, and Equation 4 may be variously modified, and is not limited thereto.
Figure PCTKR2016001631-appb-M000004
Figure PCTKR2016001631-appb-M000004
상기 수학식 4에서 Sd는 약물 d의 약물 점수, n은 상기 약물 d의 약력학 또는 약동학에 직접 관여하거나 해당 약물의 전구체 또는 해당 약물의 대사산물들과 상호작용하는 단백질, 예를 들면, 약물학적 패스웨이를 구성하는 유전자 군에서 선정된 하나 이상의 유전자가 코딩하는 단백질의 수, gi는 상기 약물 d의 약력학 또는 약동학에 직접 관여하거나 해당 약물의 전구체 또는 해당 약물의 대사산물들과 상호작용하는 단백질, 예를 들면, 약물학적 패스웨이에 구성하는 유전자 군에서 선정된 하나 이상의 유전자가 코딩하는 단백질의 단백질 손상 점수이며, wi는 상기 gi에 부여되는 가중치이다. 모든 가중치 wi가 같은 값을 갖는 경우 상기 약물 점수 Sd는 상기 단백질 손상 점수 gi의 기하평균값이 된다. 상기 가중치는 해당 단백질의 종류, 해당 단백질의 약동학적 또는 약력학적 분류, 해당 약물 효소 단백질의 약동학적 파라미터, 인구 집단 또는 인종별 분포를 고려하여 부여될 수 있다.In Equation 4, Sd is a drug score of drug d, n is a protein directly involved in the pharmacodynamics or pharmacokinetics of drug d or interacts with a precursor of the drug or metabolites of the drug, eg, a pharmacological pass. The number of proteins encoded by one or more genes selected from the group of genes constituting the way, gi, are proteins that directly participate in the pharmacodynamics or pharmacokinetics of drug d or interact with precursors or metabolites of the drug, eg For example, the protein damage score of the protein encoded by one or more genes selected from the gene group constituting the pharmacological pathway, wi is the weight given to the gi. The drug score Sd becomes the geometric mean value of the protein damage score gi when all weights wi have the same value. The weight may be given in consideration of the type of the protein, the pharmacokinetic or pharmacodynamic classification of the protein, the pharmacokinetic parameters of the drug enzyme protein, and the population or race distribution.
본 발명에 따른 일 구현예의 방법에 사용되는 기하평균 계산법의 경우 약물과 단백질의 연관성이 갖는 특징과 상관없이 가중치를 모두 동일하게 부여하였지만, 약물과 단백질의 연관성이 갖는 각 특징을 고려한 가중치를 부여하여 약물 점수를 산출하는 것이 가능하다. 예를 들어 약물의 표적 단백질과 약물과 관련된 수송체 단백질에는 다른 점수가 부여될 수 있다. 또한, 예를 들어 해당 약물 대사 효소에는 그 약동학적 파라미터인 Km, Vmax, Kcat/Km를 가중치로 부여하여 약물 점수를 산출하는 것이 가능하다. 또한, 예를 들어 표적 단백질을 수송체 단백질과 비교하여 약리작용상 더 중요하다고 판단하여 높은 가중치를 부여할 수 있고, 수송체 단백질이나 운반체 단백질은 농도에 민감한 약물에 대해서 높은 가중치가 부여될 수 있으며, 이에 제한되지 않는다. 가중치는 약물과 약물 관련 단백질 간의 상관관계, 약물과 단백질 상호작용의 특성에 따라 면밀히 조정될 수 있다. 예를 들면 표적 단백질에는 2점을, 수송체 단백질에는 1점을 부여하는 것과 같이 약물과 단백질의 상호작용의 특성에 대한 가중치를 부여한 정교한 알고리즘을 사용할 수 있다. In the geometric mean calculation method used in the method of the embodiment according to the present invention, all weights were equally given regardless of the drug-protein association characteristics, but the weight was given by considering the respective characteristics of drug-protein association. It is possible to calculate the drug score. For example, different scores may be assigned to the drug's target protein and the drug's transporter protein. In addition, for example, the drug metabolizing enzyme may be weighted by the pharmacokinetic parameters Km, Vmax, and Kcat / Km to calculate the drug score. In addition, for example, the target protein may be given a higher weight by judging that the target protein is more important in pharmacological action than the transporter protein, and the transporter protein or the carrier protein may be given a high weight for the concentration-sensitive drug. This is not restrictive. Weights can be closely adjusted according to the correlation between drug and drug-related proteins, and the nature of drug-protein interactions. For example, a sophisticated algorithm can be used that assigns a weight to the nature of the drug-protein interaction, such as giving the target protein two points and the transporter protein one point.
상기의 서술에서는 약물, 즉 자궁수축억제제와 직접 상호작용하는 단백질만을 예로 들었지만, 해당 약물의 전구체 또는 해당 약물의 대사산물들과 상호작용하는 단백질, 해당 약물의 약력학 또는 약동학에 관여하는 단백질들과 유의하게 상호작용하는 단백질, 그 신호전달경로에 참여하는 관련 단백질 정보를 활용하여 상기 수식의 예측능력을 향상시킬 수 있다. 즉 단백질-단백질 상호작용 네트워크 혹은 약물학적 패스웨이 정보를 활용하여 이에 관여하는 다양한 단백질의 정보를 사용할 수 있다. 즉 해당 약물과 직접 상호작용하는 단백질에 유의한 변이가 발견되지 않아 해당 단백질 손상 점수 계산값이 없거나 손상 없음(예를 들면, SIFT 알고리즘을 적용한 경우 1.0점)인 경우에도, 해당 단백질과 상호작용하거나 같은 신호전달경로에 참여하는 관련 단백질들의 단백질 손상 점수의 평균값(예를 들면, 기하평균치) 등을 해당 단백질의 단백질 손상 점수로 대신 사용하여, 약물 점수 산출에 사용할 수 있다.In the above description, only the drug, that is, the protein that directly interacts with the uterine contraction inhibitor, is taken as an example, but the protein interacts with the precursor of the drug or its metabolites, and the proteins involved in the pharmacodynamics or pharmacokinetics of the drug. It is possible to improve the predictive power of the formula by utilizing protein information that interacts easily, and related protein information that participates in the signaling pathway. In other words, the protein-protein interaction network or pharmacological pathway information can be utilized to use the information of various proteins involved in it. That is, even if no significant mutation was found for a protein that directly interacts with the drug, and there is no corresponding protein damage score calculation or no damage (for example, 1.0 if SIFT algorithm is applied), The mean value (eg, geometric mean) of the protein damage scores of related proteins participating in the same signaling pathway may be used as a drug score calculation instead of the protein damage score of the protein.
상기의 개인별 약물 점수는 하나 이상의 연관 단백질에 대한 정보가 획득 가능한 모든 약물 또는 그중 선별된 일부 약물에 대해서 산출할 수 있다. 또한, 이러한 개인별 약물 점수는 순위점수(rank)로 환산 가능하다. The individual drug scores may be calculated for all drugs for which information on one or more related proteins can be obtained or for some selected drugs. In addition, such individual drug scores can be converted into rank (rank).
본 발명의 방법은 상술한 개인별 약물 점수를 이용하여 상기 개인에 적용되는 약물, 즉 자궁수축억제제의 사용 여부를 결정하는 단계를 추가로 포함할 수 있다. The method of the present invention may further include determining whether to use a drug applied to the individual, ie, a uterine contraction inhibitor, by using the aforementioned individual drug score.
본 발명의 개인별 약물 점수는 모든 약물에 개별적으로 적용할 수 있지만, 질환별, 임상적 특징, 또는 작용방식 등으로 분류하여 적용하거나 의학적 비교 대상 약물들에 적용하면 더 유용하다. 본 발명에서 사용 가능한 약물 분류체계는, 예를 들면 ATC (Anatomical Therapeutic Chemical Classification System) 코드, 미국에서 자주 처방되는 15가지 약물 목록 (top 15 frequently prescribed drug classes during 2005~2008 in the United States (Health, United States, 2011, Centers for Disease Control and Prevention)), 약물유전체학적 마커가 알려져 있어서 약물의 표시에 기재되는 약물 작용 정보에 영향을 줄 수 있는 약물, 또는 부작용 등으로 시장에서 퇴출된 약물 목록 등을 포함할 수 있다.The individual drug scores of the present invention can be applied individually to all drugs, but are more useful if they are classified by disease, clinical characteristics, or mode of action, or applied to drugs for medical comparison. Drug classification systems that can be used in the present invention are, for example, the ATC (Anatomical Therapeutic Chemical Classification System) code, the top 15 frequently prescribed drug classes during 2005-2008 in the United States (Health, United States, 2011, Centers for Disease Control and Prevention), a list of drugs that have been released from the market due to known pharmacogenomic markers that may affect drug action information on the drug label, or side effects. It may include.
본 발명의 방법은 처방 점수를 계산하는 단계를 추가로 포함할 수 있다. The method of the invention may further comprise calculating a prescription score.
본 발명에서 사용된 용어 “처방 점수”란 두 개 이상 복수의 약물이 동시에 혹은 서로 약리작용에 유의한 영향을 미칠 만큼 짧은 시간 간격을 두고 투약되는 경우, 상기 각 약물에 대하여 결정된 상기 약물 점수를 종합하여 계산되는 점수를 말한다. 본 발명에서 처방점수는 상기 약물 간의 우선순위에 의해 결정된 약물이 두 개 이상이고 동시 투약이 필요한 경우, 상기 각 약물에 대하여 결정된 약물 점수를 종합하여 산출될 수 있다. 처방 점수의 계산은, 예를 들어, 해당 복수 약물과 공통으로 상호작용하는 단백질이 존재하지 않는 경우에는, 단순히 해당 복수 약물의 약물 점수들을 평균 내거나 합산 혹은 곱함으로써 산출할 수 있다. 해당 복수 약물과 공통으로 상호작용하는 단백질이 존재하는 경우에는, 해당 공통 상호작용 단백질의 단백질 손상 점수에, 예를 들면 2배의 가중치를 부여하여 각각의 약물 점수를 산출한 후, 해당 약물 점수들을 합산함으로써 처방 점수를 산출할 수 있다. As used herein, the term “prescription score” refers to the sum of the drug scores determined for each drug when two or more drugs are administered at the same time or at intervals short enough to significantly affect the pharmacological action of each other. Say your score is calculated. In the present invention, the prescription score may be calculated by combining the drug scores determined for each drug when two or more drugs determined by the priority between the drugs and simultaneous administration are required. The calculation of the prescription score can be calculated by simply averaging, summing or multiplying the drug scores of the plurality of drugs, for example, when no protein interacts in common with the plurality of drugs. If there are proteins that interact in common with the plurality of drugs, the drug damage scores of the common interacting protein are weighted twice, for example, to calculate each drug score and then the corresponding drug scores are calculated. By summing up, the prescription score can be calculated.
처방 점수는 개별 약물의 효과를 넘어서서, 개인에 대해 적용되는 처방(전) 안에 포함된 복수 약물 처방의 적정성 혹은 위험성을 판단하기 위한 것이다. 이러한 측면에서 본 발명의 방법은 개인에 대해 적용되는 처방(전)의 적정성 혹은 위험성을 판단하는 단계를 추가로 포함할 수 있다. Prescription scores are intended to determine the adequacy or risk of a multi-drug prescription that is included in the prescription applied to an individual, beyond the effects of individual drugs. In this respect the method of the present invention may further comprise determining the appropriateness or risk of the prescription (pre) applied to the individual.
본 발명의 방법은 자궁수축억제제 부작용 방지를 목적으로 수행되는 것을 포함하며, 이에 제한되지 않는다. The method of the present invention includes, but is not limited to, being performed for the purpose of preventing adverse effects of uterine contraction inhibitors.
도 1은 본 발명의 일 구현예에 따른 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법의 각 단계를 보여주는 흐름도이다. 본 발명에 따른 일 구현예에서는 (1) 개인 사용자의 유전체 염기서열 정보 입력 또는 수신 (S100), (2) 자궁수축억제제 관련 정보 입력 또는 수신 (S110), (3) 개인 사용자의 유전자 염기서열 변이 정보 결정 (S120), (4) 자궁수축억제제에 대한 개인별 단백질 손상 점수 계산 (S130), (5) 자궁수축억제제에 대한 개인별 약물 점수 계산 (S140), (6) 약물 점수 표기, 약물 점수 순위별 정렬 또는 우선순위 결정 (S150), 및 (7) 약물 점수와 우선순위를 고려한 자궁수축억제제 선택 및 처방점수 계산 (S160)의 순서로 자궁수축억제제 선택을 위한 정보를 제공하는 방법이 진행된다.1 is a flow chart showing each step of the method for providing information for the selection of uterine contraction inhibitors using individual genome sequence variation according to an embodiment of the present invention. In one embodiment according to the present invention (1) input or reception of genome sequence information of an individual user (S100), (2) input or reception of uterine contraction inhibitor-related information (S110), (3) genetic sequence variation of the individual user Information determination (S120), (4) Individual protein damage score calculation for uterine contraction inhibitors (S130), (5) Individual drug score calculation for uterine contraction inhibitors (S140), (6) Drug score notation, by drug score ranking A method of providing information for uterine contraction inhibitor selection in order of sorting or priority determination (S150), and (7) uterine contraction inhibitor selection and prescription score calculation in consideration of drug scores and priorities is performed.
본 발명에 따른 방법은 상기 순위별로 정렬된 약물 점수를 선택하면, 해당 약물 점수가 산출된 약물유전체학적 계산과정과 근거를 그림, 도표 및 설명 등의 정보로 제공하여 처방의사의 의사결정을 돕는 단계를 추가로 포함할 수 있다. 즉, 본 발명에 따른 방법은 유전자 염기서열 변이 정보, 유전자 염기서열 변이 점수, 단백질 손상 점수, 약물 점수 및 그 산출에 사용된 정보 중 하나 이상의 정보를 제공하는 단계를 추가로 포함할 수 있다. When the method according to the present invention selects the drug scores sorted by the ranking, the step of providing a pharmacogenomic calculation process and the basis for calculating the drug scores by providing information such as pictures, charts and explanations to help the prescriber in making decisions It may further include. That is, the method according to the present invention may further include providing one or more information of gene sequence variation information, gene sequence variation score, protein damage score, drug score, and information used to calculate the same.
다른 양태에서 본 발명은 개인에게 적용할 수 있는 자궁수축억제제에 대하여, 상기 자궁수축억제제와 관련된 유전자 또는 단백질과 관련된 정보 검색 또는 추출이 가능한 데이터베이스; 상기 데이터베이스에 접근 가능한 통신부; 상기 정보에 기초하여 상기 자궁수축억제제의 약력학 또는 약동학에 관여하는 하나 이상의 유전자 염기서열 변이 정보를 산출하는 제1 산출모듈; 상기 유전자 염기서열 변이 정보를 이용하여 개인별 단백질 손상 점수를 산출하는 제2 산출모듈; 상기 개인별 단백질 손상 점수를 약물과 단백질 사이의 상호 관계와 연관지어, 개인별 약물 점수를 산출하는 제3 산출모듈; 및 상기 산출모듈에서 산출된 산출값을 표시하는 표시부를 포함하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 시스템에 관한 것이다. In another aspect, the present invention provides a uterine contraction inhibitor that can be applied to an individual, the database for retrieving or extracting information related to the gene or protein associated with the uterine contraction inhibitor; A communication unit accessible to the database; A first calculation module configured to calculate one or more gene sequence variation information related to pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor based on the information; A second calculation module for calculating an individual protein damage score using the gene sequence variation information; A third calculating module that calculates an individual drug score by associating the individual protein damage score with a correlation between a drug and a protein; And it relates to a uterine contraction inhibitor selection system using a personal genome sequence variation comprising a display unit for displaying the calculated value calculated by the calculation module.
본 발명에서 모듈이라 함은, 본 발명에 따른 기술적 사상을 수행하기 위한 하드웨어 및 상기 하드웨어를 구동하기 위한 소프트웨어의 기능적, 구조적 결합을 의미할 수 있다. 예컨대, 상기 모듈은 소정의 코드와 상기 소정의 코드가 수행되기 위한 하드웨어 리소스(resource)의 논리적인 단위를 의미할 수 있으며, 반드시 물리적으로 연결된 코드를 의미하거나, 한 종류의 하드웨어를 의미하는 것은 아님은 본 발명 기술분야의 당업자에게 자명한 것이다. In the present invention, the module may mean a functional and structural combination of hardware for performing the technical idea according to the present invention and software for driving the hardware. For example, the module may mean a logical unit of a predetermined code and a hardware resource for performing the predetermined code, and does not necessarily mean a physically connected code or a kind of hardware. Will be apparent to those skilled in the art.
본 발명에서 사용된 용어 “산출모듈”은 본 발명의 방법에 따라 분석대상이 되는 자궁수축억제제 및 유전자에 대하여, 상기 유전자 염기서열 변이 점수, 단백질 손상 점수, 약물 점수 및 그 산출의 근거가 되는 정보 및 상기 정보를 근거로 각 점수를 계산하는 소정의 코드와 상기 소정의 코드가 수행되기 위한 하드웨어 리소스(resource)의 논리적인 단위를 의미할 수 있으며, 반드시 물리적으로 연결된 코드를 의미하거나, 한 종류의 하드웨어를 의미하는 것은 아니다. The term “output module” used in the present invention refers to the gene sequence variation score, protein damage score, drug score, and information on which the calculation is made for the uterine contraction inhibitor and gene to be analyzed according to the method of the present invention. And a predetermined code for calculating each score based on the information and a logical unit of a hardware resource for performing the predetermined code, and means a physically connected code or a kind of code. It doesn't mean hardware.
본 발명에 따른 시스템은 또한 상기 제3 산출모듈에서 산출된 상기 개인별 약물 점수를 이용하여 상기 개인에 적용되는 자궁수축억제제의 사용 여부를 결정하는 제4 산출모듈을 추가로 포함할 수 있다. The system according to the present invention may further include a fourth calculation module for determining whether to use the uterine contraction inhibitor applied to the individual by using the individual drug score calculated in the third calculation module.
본 발명에 따른 시스템은 또한 약물 간의 우선순위에 의해 결정된 약물이 두 개 이상이고 동시 투약이 필요한 경우, 상기 각 약물에 대하여 결정된 상기 약물 점수를 종합하여 처방 점수로 산출하는 제5 산출모듈을 추가로 포함할 수 있다.The system according to the present invention further includes a fifth calculation module for calculating the prescription score by combining the drug scores determined for each drug when two or more drugs determined by the priority between drugs are required. It may include.
본 발명에 따른 시스템은 상기 사용자에 의해 자궁수축억제제 목록을 입력하거나, 또는 자궁수축억제제에 대한 정보를 포함하는 데이터베이스에 접근하여 관련 정보를 추출하고, 이에 따라 상기 약물의 약물 점수를 산출하여 제공하는 사용자 인터페이스를 추가로 포함할 수 있다. The system according to the present invention inputs a list of uterine contraction inhibitors or accesses a database including information on uterine contraction inhibitors, extracts relevant information, and calculates and provides a drug score of the drug accordingly. It may further include a user interface.
본 발명에 따른 시스템은 또한 각 산출모듈에서 산출된 값 또는 약물 간의 우선순위가 결정된 계산과정 및 상기 산출 또는 계산의 기초가 된 정보를 추가로 표시하는 표시부를 추가로 포함할 수 있다. The system according to the present invention may further include a display unit for further displaying a calculation process in which a value calculated in each calculation module or a priority between drugs is determined, and information on which the calculation or calculation is based.
본 발명에 따른 시스템에서, 상기 데이터베이스 또는 그 접근 정보를 포함하는 서버, 산출된 정보 및 이와 연결된 사용자 인터페이스 장치는 서로 연계되어 사용될 수 있다. In the system according to the present invention, the server including the database or its access information, the calculated information and the user interface device connected thereto may be used in conjunction with each other.
본 발명에 따른 시스템은 약물-단백질 상호관계에 대한 약물학/생화학적 새로운 정보가 산출되는 경우 즉시 업데이트되어 더 향상된 자궁수축억제제 선택에 사용될 수 있다. 본 발명에 따른 일 구현예에서는 데이터베이스 또는 지식베이스의 갱신에 따라, 상기 각 산출모듈에 저장된 상기 유전자 염기서열 변이 정보, 유전자 염기서열 변이 점수, 단백질 손상 점수, 약물 점수 및 그 산출의 근거가 되는 정보가 갱신된다. The system according to the invention can be updated immediately when new pharmacological / biochemical information on the drug-protein correlation is produced and can be used for further improved uterine contraction inhibitor selection. In one embodiment according to the present invention, according to the update of the database or knowledge base, the gene sequence variation information, gene sequence variation score, protein damage score, drug score and information on which the basis of the calculation stored in each calculation module Is updated.
도 2는 본 발명의 일 구현예에 따른 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 시스템의 개략적 구성도이다. 본 발명의 시스템(10)은 자궁수축억제제와 관련된 유전자 또는 단백질과 관련된 정보 검색 또는 추출이 가능한 데이터베이스(DB)(100), 통신부(200), 사용자 인터페이스 또는 단말(300), 산출부(400) 및 표시부(500)를 포함하여 구성될 수 있다. 2 is a schematic configuration diagram of a system for selecting a uterine contraction inhibitor using personal genome sequence variation according to an embodiment of the present invention. System 10 of the present invention is a database (DB) 100, communication unit 200, user interface or terminal 300, the calculation unit 400 capable of searching or extracting information related to genes or proteins related to the uterine contraction inhibitors And a display unit 500.
본 발명에 따른 시스템에서 사용자 인터페이스 또는 단말(300)은 서버로부터 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 처리를 요청, 결과 수신 및/또는 저장할 수 있으며, 스마트폰, PC(Personal Computer), 태블릿 PC, 개인 휴대 정보 단말기(Personal Digital Assistant, PDA), 웹 패드 등과 같이 메모리 수단을 구비하고 마이크로프로세서를 탑재하여 연산 능력을 갖춘 이동 통신 기능을 구비한 단말기로 구성될 수 있다.In the system according to the present invention, the user interface or the terminal 300 may request, receive and / or store uterine contraction inhibitor selection process using a personal genome sequence variation from a server, and may be a smartphone, a personal computer (PC), or a tablet. It may be configured as a terminal having a mobile communication function having a computing capability by mounting a microprocessor such as a PC, a personal digital assistant (PDA), a web pad, or the like.
본 발명에 따른 시스템에서 서버는 자궁수축억제제, 유전자 변이 또는 약물-단백질 상호관계에 대한 데이터베이스(100)에 대한 접근을 제공하는 수단으로, 통신부(200)을 통해 사용자 인터페이스 또는 단말(300)과 연결되어 각종 정보를 교환할 수 있도록 구성된다. 여기서, 통신부(200)는 동일한 하드웨어에서의 통신은 물론, 구내 정보 통신망(local area network, LAN), 도시권 통신망(metropolitan area network, MAN), 광역 통신망(wide area network, WAN), 인터넷, 2G, 3G, 4G 이동 통신망, 와이파이(Wi-Fi), 와이브로(Wibro) 등을 포함할 수 있으며, 통신 방식도 유선, 무선을 가리지 않으며 어떠한 통신 방식이라도 상관없다. 데이터베이스(100) 또한 서버에 직접 설치된 것뿐 아니라 목적에 따라 인터넷 등을 통해 접근 가능한 다양한 생명과학 데이터베이스에 연결될 수 있다. In the system according to the present invention, the server is a means for providing access to the database 100 for uterine contraction inhibitors, genetic variation or drug-protein correlations, and is connected to the user interface or the terminal 300 through the communication unit 200. It is configured to exchange various information. Here, the communication unit 200 may communicate in the same hardware, as well as a local area network (LAN), a metropolitan area network (MAN), a wide area network (WAN), the Internet, 2G, 3G, 4G mobile communication network, Wi-Fi (Wi-Fi), Wibro (Wibro) and the like can be included, and the communication method is wired, wireless, any communication method. The database 100 may also be connected to various life science databases accessible through the Internet as well as being installed directly on the server.
본 발명에 따른 시스템에서 산출부(400)는 상술한 바와 같이 수집/입력된 정보를 이용하여 자궁수축억제제의 약력학 또는 약동학에 관여하는 하나 이상의 유전자 변이 정보를 산출하는 제1 산출모듈(410), 개인별 단백질 손상 점수를 산출하는 제2 산출모듈(420), 개인별 약물 점수를 산출하는 제3 산출모듈(430)을 포함하여 구성될 수 있다. In the system according to the present invention, the calculation unit 400 may include the first calculation module 410 for calculating one or more gene mutation information related to pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor using the collected / input information as described above, A second calculation module 420 for calculating an individual protein damage score and a third calculation module 430 for calculating an individual drug score may be included.
본 발명에 따른 방법은 하드웨어, 펌웨어, 또는 소프트웨어 또는 이들의 조합으로 구현될 수 있다. 소프트웨어로 구현되는 경우 저장매체는 컴퓨터와 같은 장치에 의해 판독 가능한 형태의 저장 또는 전달하는 임의의 매체를 포함한다. 예를 들면 컴퓨터 판독 가능한 매체는 ROM(read only memory); RAM(random access memory); 자기디스크 저장 매체; 광저장 매체; 플래쉬 메모리 장치 및 기타 전기적, 광학적 또는 음향적 신호 전달 매체 등을 포함한다.  The method according to the invention can be implemented in hardware, firmware, or software or a combination thereof. When implemented in software, a storage medium includes any medium for storage or delivery in a form readable by a device such as a computer. For example, a computer readable medium may include read only memory (ROM); Random access memory (RAM); Magnetic disk storage media; Optical storage media; Flash memory devices and other electrical, optical or acoustic signaling media, and the like.
이러한 측면에서 본 발명은 개인 유전체 염기서열 정보로부터 자궁수축억제제의 약력학 또는 약동학에 관여하는 유전자 염기서열 변이 정보를 입수하는 단계; 상기 유전자 염기서열 변이 정보를 이용하여 개인별 단백질 손상 점수를 산출하는 단계; 및 상기 개인별 단백질 손상 점수를 약물과 단백질 사이의 상호 관계와 연관지어, 개인별 약물 점수를 산출하는 단계를 포함하는 동작을 수행하는 프로세서를 실행시키는 실행모듈을 포함하는 컴퓨터 판독 가능한 매체를 제공한다. In this aspect, the present invention comprises the steps of obtaining genetic sequence variation information involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitor from the individual genome sequence information; Calculating an individual protein damage score using the gene sequence variation information; And an execution module for associating the individual protein damage score with the correlation between the drug and the protein to execute a processor to perform an operation comprising calculating the individual drug score.
상기 프로세서는 상기 개인별 약물 점수를 이용하여 상기 개인에 적용되는 자궁수축억제제의 사용 여부를 결정하는 단계를 추가로 포함할 수 있다.The processor may further include determining whether to use a uterine contraction inhibitor applied to the individual by using the individual drug score.
또 다른 양태에서 본 발명은 자궁수축억제제 부작용 예측용 바이오 마커 조성물을 제공한다. In another aspect, the present invention provides a biomarker composition for predicting adverse effects of uterine contraction inhibitors.
본 발명에 따른 바이오 마커 조성물에 포함될 수 있는 유전자에는, BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7, TNKS, PSMD9, ZNF273, FAT4, GALNT10, OR6B1, RBBP8NL, KNDC1, UGT1A10, ARL13B, SLC15A2, SPINK6, C10orf113, TP53, TRIML2, MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT, LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EFCAB4A, CPOX, VARS2 등이 포함되며, 이로 제한되는 것은 아니다. 상기 유전자 또는 이의 단백질의 변이를 분석함으로써 자궁수축억제제 부작용 발생을 예측할 수 있는바, 이를 검출할 수 있는 제제를 이용하여 마커로 활용할 수 있다. Genes that may be included in the biomarker composition according to the present invention include BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7 TNKS, PSMD9, ZNF273, FAT4, GALNT10, OR6B1, RBBP8NL, KNDC1, UGT1A10, ARL13B, SLC15A2, SPINK6, C10orf113, TP53, TRIML2, MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT3 LRRC3C, EFCAB4A, CPOX, VARS2, and the like, but are not limited to these. By analyzing the mutation of the gene or its protein, it is possible to predict the occurrence of adverse effects of the uterine contraction inhibitor, it can be used as a marker using the agent that can detect this.
이하, 하기 실시예를 통하여 본 발명을 보다 상세히 설명한다. 이들 실시예는 본 발명을 상세히 설명하기 위한 것으로 본 발명의 범위가 이들 실시예에 의해 제한되는 것은 아니다.Hereinafter, the present invention will be described in more detail with reference to the following examples. These examples are intended to illustrate the present invention in detail, but the scope of the present invention is not limited by these examples.
실시예Example 1. 자궁수축억제제( 1. Uterine contraction inhibitors RitodrineRitodrine ) 처치에서 심각한 부작용 경고 사인을 보인 조기 진통 환자의 개인 유전체 염기서열 변이 정보 분석 및 적용Analysis and Application of Individual Genome Sequence Variation Information in Early Analgesic Patients with Significant Side Effects Warning Signs
조기진통(preterm labor)은 임신 37주 이전에 규칙적인 자궁수축과 이에 따른 점진적 자궁경부 개대 및 숙화를 지칭하는 것으로, 조산의 주요 원인 중 하나이다. 국내에서는 출산율이 저하중인 동시에 고령산모의 증가로 조산의 비율이 점차 증가(1995년 4.3%에서 1998년 7%, 2000년 8.3%, 2003년 10%)하고 있어 큰 사회문제로 대두되고 있다. 저출산이 급격히 진행되는 상황에서 조산의 비율이 증가하는 것은 모자보건학적인 측면에서 간과할 수 없는 문제이다. 조산의 가장 주요한 원인인 조기 진통에 대한 치료기술의 개발이 반드시 필요한 이유이다. Preterm labor refers to regular uterine contraction prior to 37 weeks of gestation and thus to gradual cervical dilatation and maturation, which is one of the leading causes of premature birth. In Korea, the birth rate is decreasing and the premature birth rate is gradually increasing due to the increase of elderly mothers (4.3% in 1995, 7% in 1998, 8.3% in 2000, 10% in 2003), which is becoming a big social problem. An increase in the rate of premature births is a problem that cannot be overlooked in terms of maternal and child health. This is why the development of treatment technology for early labor, which is the main cause of premature birth, is essential.
현재까지 진행되고 있는 조산의 치료는 대부분 증상이 생긴 이후에 이루어지고 있으며, 조기 진통의 산모는 진단이 되는 시점에 입원하여 자궁수축 억제제를 우선적으로 사용하게 된다. 국내에서 그동안 많이 사용된 대표적인 자궁수축억제제로는 리토드린(ritodrine), 마그네슘(MgSO4), 니페디핀(Nifedipine), 아토시반(Atosiban) 등이 있다. Most of the premature births that have been performed to date are performed after symptoms occur, and mothers of early labor are hospitalized at the time of diagnosis and preferentially use uterine contraction inhibitors. Representative uterine contraction inhibitors that have been widely used in Korea include ritodrine, magnesium (MgSO 4 ), nifedipine (Nifedipine), and atoshiban (Atosiban).
그 중 가장 높은 빈도로 사용되는 것은 리토드린, 마그네슘, 니페디핀이나, 이들 약물의 부작용이 문제가 되고 있다. 특히, 가장 많은 빈도로 사용되는 리토드린의 경우, 임산부의 빈맥, 저혈압, 불안, 흉통, 심전도의 이상, 폐부종 등의 합병증이 문제되는데, 임산부마다 부작용의 빈도 및 중증도가 매우 다른 것으로 알려져 있다. 이와 같이 환자마다 특정 약물에 대한 반응성이 달라 각 환자에게 적합한 약물 투여가 이루어지지 못하고 치료 실패나 지연 등이 나타나게 된다.Among them, ritodrine, magnesium and nifedipine, which are used most frequently, are adversely affected by the side effects of these drugs. In particular, in the case of the most frequently used ritodrine, complications such as tachycardia, hypotension, anxiety, chest pain, abnormal ECG, pulmonary edema, etc. of pregnant women is a problem, it is known that the frequency and severity of side effects are very different for each pregnant woman. As such, the responsiveness to a particular drug is different for each patient, so appropriate drug administration cannot be made to each patient and treatment failure or delay is indicated.
이에 본 발명의 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 제공 방법을 이용하여, 자궁수축억제제의 일종인 리토드린 치료 반응 위험군을 구분할 수 있는지를 확인하기 위하여, 하기와 같은 실험을 수행하였다. Thus, the following experiment was performed to determine whether a risk response group of ritodrine treatment, which is a type of uterine contraction inhibitor, can be distinguished using the method for providing selection of a uterine contraction inhibitor using the personal genome sequence variation of the present invention.
조기 진통 발생 시 태아의 재태 기간을 연장하기 위해 자궁수축억제제 리토드린 치료를 받은 산모 중 부작용이 나타난 군 13례를 분석하였다. 객관적인 비교를 위하여 정상대조군 30례와의 유전자 염기서열 비교 분석을 수행하였다. 상기 13례에 해당하는 산모들이 겪은 부작용은 다음 표 2과 같다.We analyzed 13 cases of side effects among mothers who received uterine contraction inhibitor ritodrine to prolong fetal gestation during preterm labor. Genetic sequences were compared with 30 normal controls for objective comparison. Side effects experienced by the mothers corresponding to the 13 cases are shown in Table 2 below.
일련번호Serial Number 부작용Side Effect
1One 호흡곤란Dyspnea
22 폐부종, 폐울혈Pulmonary Edema, Pulmonary Congestion
33 빈맥(116회/분), 호흡곤란Tachycardia (116 times / minute), difficulty breathing
44 호흡곤란Dyspnea
55 호흡곤란, 양측성 늑막삼출을 동반한 폐부종, CK-MB상승 (5.6ng/mL), BNP상승 (1128pg/mL)Shortness of breath, pulmonary edema with bilateral pleural effusion, CK-MB elevation (5.6ng / mL), BNP elevation (1128pg / mL)
66 빈맥 (120회/분), 호흡곤란Tachycardia (120 beats / minute), difficulty breathing
77 빈맥 (114회/분), 호흡곤란Tachycardia (114 times / minute), difficulty breathing
88 호흡곤란Dyspnea
99 빈맥 (110회/분), 호흡곤란, 심계항진Tachycardia (110 beats / minute), dyspnea, palpitations
1010 빈맥 (108회/분), 두통, 발한Tachycardia (108 beats / minute), headache, sweating
1111 호흡곤란, 심계항진Shortness of breath, palpitations
1212 폐부종Pulmonary Edema
1313 폐부종Pulmonary Edema
상기 13례의 개인 유전체 염기서열 변이 정보 수득을 위하여 Life Technology사의 Ion AmpliSeq Exome Kit을 이용하여 80배수 전장 엑솜 염기서열 해독법 (Whole Exome Sequencing)을 수행하였다. 이때 상기 방법 외에 엑솜 외에 개인의 유전체 전체의 정보를 얻어내는 전장 유전체 염기서열 해독법 (Whole Genome Sequencing) 또는 500-1000개의 자궁수축억제제 관련 주요 유전자에 대한 표적 엑솜 염기서열 해독법 (Targeted Exome Sequencing)을 대안적으로 수행할 수 있다.In order to obtain information on the 13 individual genome sequences, 80-fold full-length exome sequencing was performed using Ion AmpliSeq Exome Kit from Life Technology. In addition to the above method, alternative to full genome sequencing that obtains the entire genome of the individual in addition to the exome or target exome sequencing of major genes related to 500-1000 uterine contraction inhibitors Can be done as
분석된 염기서열 절편은 데이터 정비(Data Cleaning)와 품질확인(Quality Check)의 과정을 거쳐 인간 참조군 서열(예, HG19)에 맞추어 정렬된 SAM (Sequence Alignment Map) 및 BAM (Binary Alignment Map) 파일 형식으로 출력되었다. 상기 클린 배열 결과(cleaned alignment result)는 SAMTools:pileup, SAMTools:mpileup, GATK:recalibration, GATK:realignment 등의 소프트웨어 도구를 활용하여 단일염기변이(SNVs, Single Nucleotide Variants), InDels 등의 변이를 검출하여 VCF(Variant Calling Format) 형식의 파일로 출력되었다.The sequence fragments analyzed were sequence alignment map (SAM) and binary alignment map (BAM) files aligned with human reference sequence (eg, HG19) through data cleaning and quality check. The output was in format. The clean alignment result is detected using a software tool such as SAMTools: pileup, SAMTools: mpileup, GATK: recalibration, GATK: realignment, and the like to detect mutations such as single nucleotide variations (SNVs, Single Nucleotide Variants) and InDels. It was output to a file in VCF (Variant Calling Format) format.
상기 유전자 염기서열 변이 정보를 담은 VCF 파일을 입력받아 전술한 유전자 염기서열 변이 점수 vi값을 SIFT 알고리즘을 활용하여 각 변이별로 계산한 후, 수학식 2를 이용하여 개인별 단백질 손상 점수 Sg를 산출하였다. 이어 상기 부작용이 나타난 13례와 건강 대조군 30례에서 각 유전자에 대하여 상기 개인별 단백질 손상점수를 비교하였다. 상기 부작용 군과 대조군에서 통계적으로 유의한 차이를 보인 유전자 47개를 하기와 같이 선별하였으며, p-값(value)을 기준으로 통계적 유의성이 더 높은 유전자 28개를 제1군으로 선별하였다. 하기 유전자들은 리토드린 및 그 대사산물의 약력학 또는 약동학에 높은 관련성이 있는 유전자이다. After receiving the VCF file containing the gene sequence variation information, the above-described gene sequence variation score vi value was calculated for each variation by using the SIFT algorithm, and the individual protein damage score Sg was calculated using Equation 2. Subsequently, the individual protein damage scores were compared for each gene in 13 cases and 30 health control controls. 47 genes showing statistically significant differences in the adverse event group and the control group were selected as follows, and 28 genes with higher statistical significance were selected as the first group based on the p-value. The following genes are genes of high relevance to the pharmacokinetics or pharmacokinetics of ritodrine and its metabolites.
(제1군)(Group 1)
BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7, TNKS, PSMD9, ZNF273, FAT4, GALNT10, OR6B1, RBBP8NL, KNDC1, UGT1A10, ARL13B BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7, TNKS, PSMD9, ZBF6AL1NT, ZBF673N10 KNDC1, UGT1A10, ARL13B
(제2군)(2nd group)
SLC15A2, SPINK6, C10orf113, TP53, TRIML2, MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT, LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EFCAB4A, CPOX, VARS2 SLC15A2, SPINK6, C10orf113, TP53, TRIML2, MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT, LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EFCAB4A, CPOX, VARS2
이와 같이 선별된 유전자군에 대하여 본 발명의 방법을 이용하여 개인별 약물점수를 산출하였다. 보다 구체적으로, 각 개인별 유전자 염기서열 변이 정보로부터 SIFT 알고리즘을 이용하여 유전자 염기서열 변이 점수를 산출한 후, 수학식 2를 사용하여 상기 47개 유전자에 대한 개인별 단백질 손상 점수를 산출하였고, 수학식 4를 사용하여 리토드린에 대한 개인별 약물 점수를 산출한 후, 각 군별로 통계 분석하였다. 그 결과를 표 3에 나타내었다. Thus, the drug score for each individual gene group was calculated using the method of the present invention. More specifically, after calculating the gene sequence variation score using the SIFT algorithm from each individual gene sequence variation information, the individual protein damage score for the 47 genes was calculated using Equation 2, Equation 4 Using to calculate the individual drug score for ritordlin, statistical analysis for each group. The results are shown in Table 3.
리토드린 부작용 군 (n=13)Ritodrin Side Effects Group (n = 13) 정상 대조군 (n=30)Normal control (n = 30) p-valuep-value
약물점수 평균 (1군 약물)Drug Score Average (Group 1 drugs) 0.2364386660.236438666 0.5824548060.582454806 1.55E-081.55E-08
약물점수 평균 (1,2군 약물)Drug Score Average (Group 1 and 2 Drugs) 0.1557564740.155756474 0.5429854910.542985491 7.62E-147.62E-14
1군 단백질 손상점수Group 1 protein damage score
BLKBLK 0.16000.1600 0.58000.5800 4.35E-064.35E-06
SPTA1SPTA1 0.32010.3201 0.48870.4887 6.84E-066.84E-06
IFT74IFT74 0.24890.2489 0.55810.5581 1.42E-051.42E-05
RSPH3RSPH3 0.27300.2730 0.61060.6106 1.85E-051.85E-05
CYP8B1CYP8B1 0.45370.4537 0.70530.7053 2.01E-052.01E-05
ICE1ICE1 0.48540.4854 0.70260.7026 2.56E-052.56E-05
NKAIN3NKAIN3 0.21130.2113 0.56140.5614 2.57E-052.57E-05
AASDHAASDH 0.16400.1640 0.58290.5829 4.15E-054.15E-05
FUT6FUT6 0.13850.1385 0.45670.4567 0.000150.00015
SLC12A7SLC12A7 0.36000.3600 0.59670.5967 0.000150.00015
CD1ACD1A 0.35380.3538 0.85530.8553 0.000190.00019
CYP1A1CYP1A1 0.02910.0291 0.36350.3635 0.000210.00021
CARS2CARS2 0.22910.2291 0.72800.7280 0.000230.00023
ZDHHC12ZDHHC12 0.16580.1658 0.40840.4084 0.000330.00033
CSPG5CSPG5 0.24000.2400 0.49330.4933 0.000340.00034
PXT1PXT1 0.02000.0200 0.34670.3467 0.000340.00034
HHATLHHATL 0.27090.2709 0.49720.4972 0.000370.00037
SERPINA7SERPINA7 0.31460.3146 0.90100.9010 0.000400.00040
TNKSTNKS 0.38340.3834 0.67690.6769 0.000400.00040
PSMD9PSMD9 0.43060.4306 0.90600.9060 0.000510.00051
ZNF273ZNF273 0.09750.0975 0.37710.3771 0.000530.00053
FAT4FAT4 0.22950.2295 0.43200.4320 0.000540.00054
GALNT10GALNT10 0.35230.3523 0.65800.6580 0.000550.00055
OR6B1OR6B1 0.28340.2834 0.50280.5028 0.000560.00056
RBBP8NLRBBP8NL 0.20780.2078 0.57650.5765 0.000560.00056
KNDC1KNDC1 0.33530.3353 0.52800.5280 0.000620.00062
UGT1A10UGT1A10 0.61850.6185 1.00001.0000 0.008990.00899
ARL13BARL13B 0.86460.8646 0.87970.8797 0.445180.44518
2군 단백질 손상점수Group 2 protein damage score
SLC15A2SLC15A2 0.08720.0872 0.36100.3610 0.000710.00071
SPINK6SPINK6 0.04000.0400 0.32800.3280 0.000710.00071
C10orf113C10orf113 0.00000.0000 0.30000.3000 0.000710.00071
TP53TP53 0.17000.1700 0.41900.4190 0.000710.00071
TRIML2TRIML2 0.30050.3005 0.51330.5133 0.000720.00072
MAD1L1MAD1L1 0.30860.3086 0.62320.6232 0.000750.00075
ASZ1ASZ1 0.26380.2638 0.68100.6810 0.000760.00076
MAN2B2MAN2B2 0.24950.2495 0.44490.4449 0.000800.00080
CAPN14CAPN14 0.19650.1965 0.39760.3976 0.000800.00080
BAATBAAT 0.33800.3380 0.53390.5339 0.000870.00087
LAMA4LAMA4 0.10800.1080 0.33220.3322 0.000870.00087
ADCY3ADCY3 0.39920.3992 0.73620.7362 0.000880.00088
GRM7GRM7 0.35010.3501 0.71040.7104 0.000930.00093
SNAP47SNAP47 0.22640.2264 0.45460.4546 0.001030.00103
LRIT2LRIT2 0.30790.3079 0.62320.6232 0.001040.00104
LRRC3CLRRC3C 0.17310.1731 0.62530.6253 0.001050.00105
EFCAB4AEFCAB4A 0.35050.3505 0.49960.4996 0.001060.00106
CPOXCPOX 0.35620.3562 0.84500.8450 0.001210.00121
VARS2VARS2 0.06970.0697 0.32200.3220 0.001220.00122
상기 표 3에 나타낸 바와 같이, 선별된 28개의 제1군 유전자들을 이용하여 유전자 염기서열 변이정보로부터 단백질 손상점수 및 개인별 약물 점수를 산출한 결과, 정상 대조군과 리토드린 부작용 군 사이에 통계적으로 유의하게 차이가 나타남을 확인하였다. As shown in Table 3, the protein damage score and the individual drug score were calculated from the gene sequence variation information using the 28 selected group 1 genes, and statistically significant between the normal control group and the ritodrine side effects group. It was confirmed that a difference appeared.
또한, 제2군 유전자들을 추가로 포함하여 총 47개의 유전자 염기서열 변이정보로부터 개인별 약물 점수를 산출한 결과에서도, 두 군 사이의 개인별 약물점수가 통계적으로 유의하게 차이를 보였다(p-value <0.05). In addition, even when the individual drug scores were calculated from a total of 47 gene sequence variation information including the second group genes, the individual drug scores between the two groups showed statistically significant differences (p-value <0.05). ).
상기 결과를 통하여, 본 발명에 따른 개인 유전체 염기서열 변이 정보 분석을 통한 개인별 약물 점수 산출을 이용하여 리토드린 치료 시 심각한 약물 부작용 경고 사인을 겪은 군과 그렇지 않은 군을 유의하게 구분할 수 있으며, 이를 통해 원치 않는 부작용을 사전에 예방할 수 있음을 확인하였다.Through the above results, it is possible to significantly distinguish between the group that suffered serious drug side effect warning sign and the other group that did not suffer serious drug side effect warning by using the individual drug score calculation through analysis of individual genome sequence variation information according to the present invention. It was confirmed that unwanted side effects can be prevented in advance.
따라서 본 발명의 방법을 이용하여 향후 조기 진통 환자에서 리토드린 투여시 부작용 발생 가능성이 높은 군을 예측할 수 있으며, 고 위험군에 대해서는 약물의 농도를 조절하거나 대체 가능한 다른 치료법 혹은 중재요법을 사용하도록 유도할 수 있을 것이다.Therefore, the method of the present invention can be used to predict a group of patients with high risk of side effects in the future patients with early analgesia, and induce high-risk patients to adjust the concentration of the drug or to use other alternative or interventional therapy. Could be.
실시예Example 2. 개인 유전체 염기서열 변이 정보를 기반으로 하는 맞춤형 약물 선택 방법의 타당성 검증 2. Validation of Custom Drug Selection Method Based on Personal Genomic Sequence Variation Information
아직까지 개인 유전체 염기서열 변이 정보와 약물학적 반응의 개인차에 관한 신뢰할만한 연구결과는 매우 제한적이다. 현재까지의 연구는 특정 변이가 양성인 또는 음성인 군을 약물별로 비교하여 반응성의 개인차를 연구하는 증례-대조군 관찰연구의 패러다임을 따라왔다. 이러한 연구 패러다임에서는 수많은 염기서열 변이와 수많은 약물쌍으로 이루어지는 모든 조합에 대해 각각 고비용의 증례-대조군 연구를 수행해야하지만 현실적으로는 불가능하다. 반면 본 발명에 따른 개인별 맞춤형 약물 선택 방법은 모든 유전자 염기서열 변이를 대상으로 할 뿐만 아니라, 고비용의 증례-대조군 설계의 관찰 연구를 필요로 하지 않고, 유전체 염기서열 변이에 대한 순수 계산만으로 개인별 단백질 손상 점수와 개인별 약물 점수를 산출하고 이를 적용하는 방법을 제안하므로, 모든 유전체 염기서열 변이와 모든 약물 사이의 조합에 대하여 개인별 맞춤형 약물 선택을 위한 추론이 가능하다는 큰 장점을 갖는다. To date, reliable studies of individual genome sequence variation information and individual differences in pharmacologic responses have been limited. To date, research has followed a paradigm of case-control observation studies in which individual differences in responsiveness are studied by comparing drug-positive or negative groups by drug. In this research paradigm, expensive case-control studies must be performed for all combinations of numerous sequence variations and numerous drug pairs, but this is not practical. On the other hand, the personalized drug selection method according to the present invention not only targets all gene sequence mutations, but also does not require expensive case-control design observation studies, and damages individual proteins by pure calculation of genome sequence mutations. Since we propose a method of calculating the score and the individual drug score and applying it, it has the great advantage that it is possible to infer a personalized drug selection for the combination of all genome sequence variations and all drugs.
본 발명의 방법에 따른 개인별 맞춤형 약물 선택 산출 결과의 타당성 평가를 위해 다음과 같은 기준으로 497개의 다빈도 처방 약물을 선택하였다; (1) 미국에서 가장 흔히 처방되는 15가지 약물(top 15 frequently prescribed drug classes during 2005~2008 in the United State (Health, United States, 2011, Centers for Disease Control and Prevention)의 ATC 코드에 포함되는 약물 중 적어도 한 개 이상의 약력학 또는 약동학 관련 유전자가 알려진 약물, (2) 확립된 약물유전체학적 유전체 염기서열 변이 마커의 작용이 미국 식약처의 의약품 라벨 표시에 적용된 약물, (3) 약물 부작용 등으로 시장에서 퇴출된 것으로 DrugBank 데이터베이스에 공지된 약물. 497 multi-frequency prescription drugs were selected for the feasibility of the results of personalized drug selection calculation according to the method of the present invention; (1) Among the drugs included in the ATC code of the top 15 frequently prescribed drug classes during 2005-2008 in the United State (Health, United States, 2011, Centers for Disease Control and Prevention). Drugs with known at least one pharmacodynamic or pharmacokinetic gene, (2) the action of established pharmacogenomic genome sequence markers applied to the drug labeling of the US Food and Drug Administration, and (3) drug side effects from the market. Drugs known to the DrugBank database.
타당성 평가 기준 자료로는 PharmGKB가 제공하는 987개의 유전자 염기서열 변이-약물 상호작용 쌍에 대한 확립된 지식 중 상기 497개의 약물과 적어도 하나 이상의 연결을 갖는 650개(65.9%)를 추출했다. 본 발명이 엑손 영역의 염기서열 변이를 대상으로 한 점을 고려하여, 공정한 평가를 위해서 검증 대상 자료와 평가 기준 자료 사이에 겹치는 부분은 제거하였다. 좀 더 구체적으로는 상기 650쌍 중에서 엑손 영역에 위치한 염기서열 변이 36개를 모두 제거하고 비코딩 영역의 염기서열 변이만을 선택하여 좀 더 공정한 평가를 수행하였다. 결론적으로, 평가를 위한 최종 황금표준으로 614쌍을 선택하였다. Validity criteria data were extracted from the established knowledge of 987 gene sequence mutation-drug interaction pairs provided by PharmGKB with 650 (65.9%) having at least one link with the 497 drugs. Considering that the present invention targets the nucleotide sequence variation of the exon region, overlapping portions between the verification target data and the evaluation reference data are removed for fair evaluation. More specifically, all 36 nucleotide sequence mutations located in the exon region were removed from the 650 pairs, and only a nucleotide sequence variation of the non-coding region was selected to perform a more fair evaluation. In conclusion, we selected 614 pairs as the final gold standard for evaluation.
다음으로 The 1000 Genomes Project가 제공하는 1092명의 전장 유전체 염기서열을 분석하여 1092명 각각에 대해 본 발명에 따른 방법을 적용하여, 개인별 약물유전체학적 위험성과 PharmGKB에 등록된 유전자 염기서열 변이별 약물유전체학적 위험성을 각각 계산하였다. Next, 1092 full-length genome sequences provided by The 1000 Genomes Project were analyzed, and the method according to the present invention was applied to each of 1092 persons, and the pharmacogenomic risks of individuals and the pharmacogenomics of each gene sequence registered in PharmGKB were analyzed. Risks were each calculated.
타당도 평가에는 민감도, 특이도 및 ROC 곡선하면적(Area Under the Receiver Operating Curve)를 사용했다. 개인별 약물 점수를 바탕으로 497개의 약물에 순위를 매기고 각 순위 사이의 496개의 분할 위치에 순위별로 역치를 설정한 후, (1) 해당 약물의 약물 점수 순위가 역치보다 상위에 있고 PharmGKB 변이가 개인 유전체 변이에 있을 때는 참양성, (2) 해당 약물의 약물 점수 순위가 역치보다 하위에 있고 PharmGKB 변이가 개인 유전체 변이에 없을 때는 참음성, (3) 해당 약물의 약물 점수 순위가 역치보다 상위에 있으나 PharmGKB 변이가 개인 유전체 변이에는 없을 때는 위양성, (4) 해당 약물의 약물 점수 순위가 역치보다 하위에 있으나 PharmGKB 변이가 개인 유전체 변이에 있을 때는 위음성으로 정했다, 각 개인에서 각 순위역치 L에 대해 참양성, 참음성, 위양성, 위음성의 개수를 산출하여 하기 식과 같이 민감도와 특이도를 계산하였다. For the validity evaluation, the sensitivity, specificity and area under the receiver operating curve were used. After ranking 497 drugs based on individual drug scores and setting thresholds by rank at 496 splits between each rank, (1) the drug score rank of the drug is above the threshold and the PharmGKB mutation is in the personal genome. True positive when mutant, (2) drug score ranking of the drug is below the threshold and true if the PharmGKB mutation is not in the individual genome variation, (3) drug score ranking of the drug is above the threshold but PharmGKB False positives when the mutation was not present in the individual genome variation, and (4) the drug score ranking of the drug was lower than the threshold, but when the PharmGKB mutation was in the individual genome variation, it was false negative.True positive for each rank threshold L in each individual, The number of true negatives, false positives and false negatives was calculated to calculate sensitivity and specificity as shown in the following equation.
Figure PCTKR2016001631-appb-I000001
Figure PCTKR2016001631-appb-I000001
Figure PCTKR2016001631-appb-I000002
Figure PCTKR2016001631-appb-I000002
상기 D는 497개의 전체 약물의 집합, GS는 개인별로 개인 유전자 염기서열 변이가 PharmGKB의 위험 대립유전형과 일치하여 개인별 황금표준으로 사용되는 개인화된 PharmGKB 약물의 집합, DL은 순위역치 상위 약물의 집합이며, 수직 막대 괄호는 해당 집합의 원소 개수를 의미한다. D is a set of 497 total drugs, GS is a set of personalized PharmGKB drugs that are used as individual gold standards by matching individual genetic sequence mutations with the risk allele of PharmGKB, and D L is a set of top-ranking drugs The vertical bar brackets indicate the number of elements in the set.
계산 결과, 18명의 경우 PharmGKB의 변이와 일치하는 변이를 한 개도 가지고 있지 않아서 황금표준으로 사용되는 개인화된 PharmGKB 약물의 집합을 정의할 수 없었기에, 이들은 본 타당성 분석에서 제외하였으며, 모든 역치에 대한 민감도와 특이도를 계산하여 ROC 곡선을 그리고, AUC를 계산하였다. 보다 구체적으로, 먼저 1092명의 전체 인구집단을 대상으로 SIFT 알고리즘을 이용하여 유전자 염기서열 변이 점수를 산출한 후, 이에 수학식 2와 수학식 4를 적용하여 단백질 손상 점수 및 약물 점수를 각각 산출하였다. 또한, 인종별 분포에 따른 가중치 적용의 유용성을 판단하기 위해서 인종 특이적 민감도, 특이도 및 이에 기반을 둔 AUC 값 산출을 The 1000 Genomes Project에 명시된 4개의 인종(African (AFR, n=246), American (AMR, n=181), Asian (ASN, n=286), European (EUR, n=379))별로 각각 동일하게 수행한 후, 인종 특이적 민감도, 특이도 및 AUC를 각각 구하였다. 그 결과를 표 4, 표 5 및 도 3에 나타내었다. Calculations showed that 18 patients did not have any variations that matched PharmGKB's variation and therefore could not define a set of personalized PharmGKB drugs used as gold standards, so they were excluded from this feasibility analysis and were sensitive to all thresholds. The ROC curve was calculated by calculating the specificity and and the AUC was calculated. More specifically, first, gene sequence variation scores were calculated using a SIFT algorithm for a total population of 1092 people, and then protein damage scores and drug scores were calculated by applying Equations 2 and 4, respectively. In addition, in order to determine the usefulness of weighting according to racial distribution, the calculation of racial-specific sensitivity, specificity, and AUC values based thereon is calculated using the four races (African (AFR, n = 246), American (AMR, n = 181), Asian (ASN, n = 286), and European (EUR, n = 379) were performed in the same manner, respectively, and race-specific sensitivity, specificity, and AUC were determined, respectively. The results are shown in Table 4, Table 5 and FIG.
단백질 군별 분포 및 평균 단백질 손상 점수 산출 Protein group distribution and average protein damage score calculation
단백질 군Protein family 단백질 수Protein count 관련 약물 수Related drug count 단백질-약물 쌍의 수Number of protein-drug pairs 평균 단백질 손상 점수Average protein damage score
표적 단백질Target protein 440440 486486 23572357 0.7980.798
운반체 단백질 Carrier Protein 1010 5050 6565 0.7280.728
대사효소 단백질Metabolic protein 7474 330330 13471347 0.7330.733
수송체 단백질Transporter protein 5454 176176 457457 0.7330.733
system 545545 497497 42014201 0.7830.783
The 1000 Genomes Project 데이터를 이용한 단백질 군별 및 인종별 약물 점수 산출 타당도(AUC) 산출 Calculate validity (AUC) of drug scores by protein group and race using The 1000 Genomes Project data
TotalTotal AFRAFR AMRAMR ASNASN EUREUR
약물 점수 산출 타당도(AUC)Drug Score Calculation Validity (AUC)
표적 단백질Target protein 0.6170.617 0.6340.634 0.6080.608 0.6140.614 0.6140.614
운반체 단백질Carrier Protein 0.5540.554 0.5110.511 0.5990.599 0.4850.485 0.5940.594
대사효소 단백질Metabolic protein 0.5870.587 0.6420.642 0.5800.580 0.5580.558 0.5790.579
수송체 단백질Transporter protein 0.4970.497 0.4920.492 0.4880.488 0.4890.489 0.5120.512
단백질 군별 가중치를 비적용 또는 적용한 약물 점수 산출 타당도(AUC)Drug Score Calculation Validity (AUC) with or without protein weights
단순기하평균Simple geometric mean 0.6660.666 0.7440.744 0.6500.650 0.6340.634 0.6530.653
가중기하평균Weighted geometric mean 0.6670.667 0.7420.742 0.6520.652 0.6330.633 0.6540.654
상기 표 4는 본 실시예에서 사용한 약물 497개에 대한 단백질 군별 분포를 나타낸 것으로, 각 군별로 단백질-약물 쌍의 수와 평균 단백질 손상 점수를 함께 표시하였다.Table 4 shows the distribution of the protein groups for the 497 drugs used in this example, and the number of protein-drug pairs and the average protein damage score were displayed together in each group.
상기 표 5는 수학식 4를 이용하여 약물 점수를 산출할 때, 단백질 군별 가중치를 적용하지 않은 경우(단순기하평균)와 적용한 경우(가중기하평균)에 각각 산출된 개인별 약물 점수 산출 타당도(AUC)를 각 단백질 군별, 각 인종별로 구분하여 나타낸 것이다. In Table 5, when calculating the drug score using Equation 4, the individual drug score calculation validity (AUC) calculated when the protein group weight is not applied (simple geometric mean) and when applied (weighted geometric mean), respectively It is represented by each protein group, each race.
보다 구체적으로, 전체 인구 집단을 예로 들면, 약물 점수 산출을 위하여 수학식 4에서 가중치 wi를 부여하지 않고(가중치 wi=1) 표적 단백질, 운반체 단백질, 대사효소 단백질, 수송체 단백질 등 각 단백질 군별로 산출한 AUC 값은 각각 0.617, 0.554, 0.587, 0.497이었으며, 이를 단백질 군별 가중치로 사용하여(수학식 4의 가중치 wi에 각 값을 대입) 산출한 개인별 가중기하평균 약물 점수 산출 타당도(AUC=0.667)를 구하였다(도 3b 참조). 그 결과, 상기 단백질 군별 가중치를 적용한 개인별 가중기하평균 약물 점수 산출 타당도는 가중치를 부여하지 않고(가중치 wi=1) 단순기하평균 산출식에 적용하여 산출한 개인별 단순기하평균 약물 점수 산출 타당도(AUC=0.666)보다 0.001점 향상됨을 확인하였다(도 3a 참조).More specifically, taking the entire population as an example, each group of proteins, such as target protein, carrier protein, metabolic protein, and transporter protein, is not given weight (wi = 1) in Equation 4 for drug score calculation. The calculated AUC values were 0.617, 0.554, 0.587, and 0.497, respectively, and the weighted geometric mean drug score calculation validity (AUC = 0.667) was calculated using the protein group weight (substituting each value into the weight w i of Equation 4). ) Was obtained (see FIG. 3B). As a result, the validity of the individual weighted geometric mean drug score calculation validity applied to the protein group weight was not given a weight (wi = 1), and the individual geometrical mean drug score calculation validity calculated by applying to the simple geometric mean calculation formula (AUC = 0.666), it was confirmed that the 0.001 point improvement (see Fig. 3a).
또한, 도 3a에 나타낸 바와 같이, 가중치 적용의 또 다른 예로 각 인종별 인원수에 따른 가중치를 적용하여 개인별 약물 점수 산출 타당도(AUC) 분석을 수행한 결과, 인종 특이성을 고려한 경우(굵은선), 전체 인구 집단(Total)의 AUC 값은 0.666(아프리카인(African) 0.744, 아메리카인(American) 0.650, 아시아인(Asian) 0.631, 유럽인(European) 0.653)이었으며, 인종 특이성을 고려하지 않은 경우(점선), 전체 인구집단 AUC 값은 0.633(아프리카인 0.623, 아메리카인 0.629, 아시아인 0.64, 유럽인 0.636)으로 나타나, 인종 특이성을 고려한 약물 점수 산출 타당도가 그렇지 않은 경우에 비해 더 향상되는 것을 확인하였다. In addition, as shown in FIG. 3A, as another example of weighting, a result of performing an AUC analysis of individual drug scores by applying weights according to the number of individual races is considered, considering race specificity (bold line). The total AUC of the population was 0.666 (African 0.744, American 0.650, Asian 0.631, European 0.653), without considering race specificity (dotted line). The overall population AUC was 0.633 (African 0.623, American 0.629, Asian 0.64, European 0.636), indicating that the validity of drug score calculation considering race specificity was improved compared to that of the other population.
또한, 도 3b에 나타낸 바와 같이, 인종 특이성을 고려하지 않고 단백질 군별 가중치만을 적용한 경우(점선), 본 발명의 개인별 약물 점수 산출 타당도 AUC는 0.634이고, 인종 특이성과 함께 단백질 군별 가중치도 함께 적용한 경우(굵은선), 본 발명의 개인별 약물 점수 산출 타당도 AUC는 0.667로 나타나, 서로 다른 가중치의 유용성이 있음을 알 수 있다.In addition, as shown in Figure 3b, when applying only the weight of each protein group without considering race specificity (dotted line), the individual drug score calculation validity AUC of the individual of the present invention is 0.634, and also applied to the weight of each protein group with race specificity ( Thick line), the individual drug score calculation validity AUC of the present invention is 0.667, indicating that the different weights are useful.
이상에서 본원의 예시적인 구현예에 대하여 상세하게 설명하였지만 본원의 권리범위는 이에 한정되는 것은 아니고 다음의 청구범위에서 정의하고 있는 본원의 기본 개념을 이용한 당업자의 여러 변형 및 개량 형태 또한 본원의 권리범위에 속하는 것이다.Although the exemplary embodiments of the present application have been described in detail above, the scope of the present application is not limited thereto, and various modifications and improvements of those skilled in the art using the basic concepts of the present application defined in the following claims are also provided. It belongs to.
본 발명에서 사용되는 모든 기술용어는, 달리 정의되지 않는 이상, 본 발명의 관련 분야에서 통상의 당업자가 일반적으로 이해하는 바와 같은 의미로 사용된다. 본 명세서에 참고문헌으로 기재되는 모든 간행물의 내용은 본 발명에 도입된다. All technical terms used in the present invention, unless defined otherwise, are used in the meaning as commonly understood by those skilled in the art in the related field of the present invention. The contents of all publications described herein by reference are incorporated into the present invention.

Claims (29)

  1. 개인 유전체 염기서열 정보로부터 자궁수축억제제의 약력학(pharmaco-dynamics) 또는 약동학(pharmaco-kinetics)에 관여하는 하나 이상의 유전자 염기서열 변이 정보를 결정하는 단계; Determining one or more gene sequence variation information related to pharmaco-dynamics or pharmaco-kinetics of the uterine contraction inhibitor from the individual genome sequence information;
    상기 유전자 염기서열 변이 정보를 이용하여 개인별 단백질 손상 점수를 산출하는 단계; 및Calculating an individual protein damage score using the gene sequence variation information; And
    상기 개인별 단백질 손상 점수를 자궁수축억제제와 단백질 사이의 상호 관계와 연관지어 개인별 약물 점수를 산출하는 단계를 포함하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. Calculating a personal drug score by associating the individual protein damage score with a correlation between the uterine contraction inhibitor and the protein, providing information for selecting the uterine contraction inhibitor using the personal genome sequence variation.
  2. 제 1 항에 있어서, The method of claim 1,
    상기 자궁수축억제제는 베타-아드레날린작동성 수용체 작용자, 칼슘 채널 차단제, 옥시토신 길항제, 비스테로이드성 항염증 약물, 질산염, 프로게스테론, 에틸 알코올 및 황산마그네슘으로 이루어진 군으로부터 선택된 1종 이상의 계열에 속하는 약물인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The uterine contraction inhibitor is a drug belonging to at least one family selected from the group consisting of beta-adrenergic receptor agonists, calcium channel blockers, oxytocin antagonists, nonsteroidal anti-inflammatory drugs, nitrates, progesterone, ethyl alcohol and magnesium sulfate And providing information for selecting a uterine contraction inhibitor using personal genome sequence variation.
  3. 제 1 항에 있어서, The method of claim 1,
    상기 약력학 또는 약동학에 관여하는 유전자는 Gene involved in the pharmacodynamics or pharmacokinetics
    BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7, TNKS, PSMD9, ZNF273, FAT4, GALNT10, OR6B1, RBBP8NL, KNDC1, UGT1A10 및 ARL13B으로 이루어진 군으로부터 선택된 1종 이상인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7, TNKS, PSMD9, ZBF6AL1NT, ZBF673N10 A method of providing information for selection of a uterine contraction inhibitor using at least one individual genome sequence variation selected from the group consisting of KNDC1, UGT1A10 and ARL13B.
  4. 제 3 항에 있어서, The method of claim 3, wherein
    SLC15A2, SPINK6, C10orf113, TP53, TRIML2, MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT, LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EFCAB4A, CPOX 및 VARS2 로 이루어진 군으로부터 선택된 1종 이상의 유전자를 더 포함하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. One or more genes further selected from the group consisting of SLC15A2, SPINK6, C10orf113, TP53, TRIML2, MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT, LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EFCAB4A, CPOX and VARS2 And providing information for selecting a uterine contraction inhibitor using personal genome sequence variation.
  5. 제 1 항에 있어서, The method of claim 1,
    상기 약력학 또는 약동학에 관여하는 유전자는 Gene involved in the pharmacodynamics or pharmacokinetics
    BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7, TNKS, PSMD9, ZNF273, FAT4, GALNT10, OR6B1, RBBP8NL, KNDC1, UGT1A10, ARL13B, SLC15A2, SPINK6, C10orf113, TP53, TRIML2, MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT, LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EFCAB4A, CPOX 및 VARS2 으로 이루어진 군으로부터 선택된 1종 이상인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. BLK, SPTA1, IFT74, RSPH3, CYP8B1, ICE1, NKAIN3, AASDH, FUT6, SLC12A7, CD1A, CYP1A1, CARS2, ZDHHC12, CSPG5, PXT1, HHATL, SERPINA7, TNKS, PSMD9, ZBF6AL1NT, ZBF673N10 A group consisting of KNDC1, UGT1A10, ARL13B, SLC15A2, SPINK6, C10orf113, TP53, TRIML2, MAD1L1, ASZ1, MAN2B2, CAPN14, BAAT, LAMA4, ADCY3, GRM7, SNAP47, LRIT2, LRRC3C, EF and VA4 The above, a method for providing information for the selection of uterine contraction inhibitors using personal genome sequence variation.
  6. 제 1 항에 있어서, The method of claim 1,
    상기 유전자 염기서열 변이 정보는 유전자의 엑손(exon)을 구성하는 염기의 치환, 부가 또는 결실인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. Wherein the gene sequence variation information is a substitution, addition or deletion of the base constituting the exon (exon) of the gene, a method for providing information for the selection of the uterine contraction inhibitor using the personal genomic sequence variation.
  7. 제 1 항에 있어서, The method of claim 1,
    상기 유전자 염기서열 변이 정보는 참조군의 유전체 염기서열과의 비교 분석을 통해 수득되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법The gene sequence variation information is obtained by comparative analysis with the genome sequence of the reference group, a method for providing information for selection of the uterine contraction inhibitor using the individual genome sequence variation
  8. 제 1 항에 있어서, The method of claim 1,
    상기 단백질 손상 점수 또는 상기 약물 점수는 SIFT (Sorting Intolerant From Tolerant), PolyPhen (Polymorphism Phenotyping), PolyPhen-2, MAPP (Multivariate Analysis of Protein Polymorphism), Logre (Log R Pfam E-value), MutationAssessor, MutationTaster, MutationTaster2, PROVEAN (Protein Variation Effect Analyzer), PMut, Condel, GERP (Genomic Evolutionary Rate Profiling), GERP++, CEO (Combinatorial Entropy Optimization), SNPeffect, fathmm, 및 CADD (Combined Annotation-Dependent Depletion)로 이루어진 군에서 선택된 하나 이상의 알고리즘을 이용하여 산출된 하나 이상의 유전자 염기서열 변이 점수로부터 산출되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The protein damage score or the drug score is SIFT (Sorting Intolerant From Tolerant), PolyPhen (Polymorphism Phenotyping), PolyPhen-2, MAPP (Multivariate Analysis of Protein Polymorphism), Logre (Log R Pfam E-value), Mutation Assessor, Mutation Tester, One selected from the group consisting of MutationTaster2, PROVEAN (Protein Variation Effect Analyzer), PMut, Condel, GERP (Genomic Evolutionary Rate Profiling), GERP ++, CEO (Combinatorial Entropy Optimization), SNPeffect, fathmm, and CADD (Combined Annotation-Dependent Depletion) A method for providing information for selecting a uterine contraction inhibitor using individual genome sequence variation, which is calculated from one or more gene sequence variation scores calculated using the above algorithm.
  9. 제 1 항에 있어서, The method of claim 1,
    상기 단백질 손상 점수 또는 약물 점수는, 유전자 염기서열 변이 점수로부터 산출되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. Wherein the protein damage score or drug score is calculated from a gene sequence variation score, wherein the protein damage score or drug score is calculated from the gene sequence variation score.
  10. 제 9 항에 있어서, The method of claim 9,
    상기 유전자 염기서열 변이 점수는 SIFT (Sorting Intolerant From Tolerant), PolyPhen (Polymorphism Phenotyping), PolyPhen-2, MAPP (Multivariate Analysis of Protein Polymorphism), Logre (Log R Pfam E-value), MutationAssessor, MutationTaster, MutationTaster2, PROVEAN (Protein Variation Effect Analyzer), PMut, Condel, GERP (Genomic Evolutionary Rate Profiling), GERP++, CEO (Combinatorial Entropy Optimization), SNPeffect, fathmm, 및 CADD (Combined Annotation-Dependent Depletion)로 이루어진 군에서 선택된 하나 이상의 알고리즘을 유전자 염기서열 변이에 적용하여 산출되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The gene sequence variation score is SIFT (Sorting Intolerant From Tolerant), PolyPhen (Polymorphism Phenotyping), PolyPhen-2, MAPP (Multivariate Analysis of Protein Polymorphism), Logre (Log R Pfam E-value), MutationAssessor, MutationTaster, MutationTaster2, One or more algorithms selected from the group consisting of Protein Variation Effect Analyzer (PROVEAN), PMut, Condel, Genomic Evolutionary Rate Profiling (GERP), GERP ++, Combinatorial Entropy Optimization (CEO), SNPeffect, fathmm, and Combined Annotation-Dependent Depletion (CAD) Method of providing information for selecting the uterine contraction inhibitor using the individual genome sequence variation, which is calculated by applying to the gene sequence variation.
  11. 제 1 항에 있어서, The method of claim 1,
    상기 단백질 손상 점수는, 단백질을 코딩하는 유전자에서 발견되는 분석대상 염기서열 변이가 두 개 이상인 경우, 상기 유전자 염기서열 변이 점수들의 평균값으로 산출되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The protein damage score is calculated as an average value of the gene sequence mutation scores when two or more nucleotide sequence mutations found in the gene encoding the protein are selected. How to provide information for.
  12. 제 11 항에 있어서, The method of claim 11,
    상기 평균값은 기하평균, 산술평균, 조화평균, 산술기하평균, 산술조화평균, 기하조화평균, 피타고라스 평균, 헤론 평균, 역조화평균, 평균제곱근편차, 센트로이드 평균, 사분평균, 이차평균, 절삭평균, 윈저화 평균, 가중평균, 가중기하평균, 가중산술평균, 가중조화평균, 함수의 평균, 멱평균, 일반화된 f-평균, 백분위수, 최대값, 최소값, 최빈값, 중앙값, 중앙범위, 중심경향도(measures of central tendency), 단순 곱 및 가중 곱으로 이루어진 군으로부터 선택된 하나 이상에 의해 계산되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The mean values are geometric mean, arithmetic mean, harmonic mean, arithmetic mean, arithmetic harmonic mean, geometric harmonic mean, Pythagorean mean, heron mean, inverse harmonic mean, mean square deviation, centroid mean, quadrant mean, quadratic mean, cutting mean , Windsing Mean, Weighted Average, Weighted Geometric Mean, Weighted Arithmetic Mean, Weighted Harmonic Mean, Function Mean, Power Average, Generalized f-Mean, Percentile, Maximum, Minimum, Mode, Median, Median Range, Central Trend A method of providing information for uterine contraction inhibitor selection using personal genome sequence variation, which is calculated by one or more selected from the group consisting of measures of central tendency, simple product and weighted product.
  13. 제 1 항에 있어서, The method of claim 1,
    상기 단백질 손상 점수는 하기 수학식 1에 의해 산출되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The protein damage score is calculated by Equation 1, method for providing information for the selection of uterine contraction inhibitors using individual genome sequence variation.
    [수학식 1][Equation 1]
    Figure PCTKR2016001631-appb-I000003
    Figure PCTKR2016001631-appb-I000003
    상기 수학식 1에서 Sg는 유전자 g가 코딩하는 단백질의 단백질 손상 점수, n은 상기 유전자 g의 염기서열 변이 중 분석대상 염기서열 변이의 수, vi는 i 번째 염기서열 변이의 유전자 염기서열 변이 점수이며, p는 0이 아닌 실수임. In Equation 1, Sg is the protein damage score of the protein encoded by the gene g, n is the number of the nucleotide sequence analysis of the nucleotide sequence variation of the gene g, vi is the gene sequence variation score of the ith nucleotide sequence variation , p is a nonzero real number.
  14. 제 1 항에 있어서, The method of claim 1,
    상기 단백질 손상 점수는 하기 수학식 2에 의해 산출되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The protein damage The score is calculated by Equation 2 below, the method for providing information for the selection of the uterine contraction inhibitor using the individual genome sequence variation.
    [수학식 2][Equation 2]
    Figure PCTKR2016001631-appb-I000004
    Figure PCTKR2016001631-appb-I000004
    상기 수학식 2에서 Sg는 유전자 g가 코딩하는 단백질의 단백질 손상 점수, n은 상기 유전자 g의 염기서열 변이 중 분석대상 염기서열 변이의 수, vi는 i 번째 염기서열 변이의 유전자 염기서열 변이 점수이며, wi는 상기 i 번째 염기서열 변이의 유전자 염기서열 변이 점수 vi에 부여되는 가중치임. In Equation 2, Sg is a protein damage score of the protein encoded by the gene g, n is the number of the nucleotide sequence analysis of the nucleotide sequence variation of the gene g, vi is the gene sequence variation score of the ith nucleotide sequence variation , wi is a weight given to the gene sequence variation score vi of the i-th sequence variation.
  15. 제 1 항에 있어서, The method of claim 1,
    상기 약물 점수는, 자궁수축억제제의 약력학 또는 약동학에 관여하는 단백질의 손상이 두 개 이상인 경우, 단백질 손상 점수들의 평균값으로 산출되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The drug score may be calculated as an average value of protein damage scores when two or more proteins are involved in pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor. How to give.
  16. 제 15 항에 있어서, The method of claim 15,
    상기 평균값은 기하평균, 산술평균, 조화평균, 산술기하평균, 산술조화평균, 기하조화평균, 피타고라스 평균, 헤론 평균, 역조화평균, 평균제곱근편차, 센트로이드 평균, 사분평균, 이차평균, 절삭평균, 윈저화 평균, 가중평균, 가중기하평균, 가중산술평균, 가중조화평균, 함수의 평균, 멱평균, 일반화된 f-평균, 백분위수, 최대값, 최소값, 최빈값, 중앙값, 중앙범위, 중심경향도(measures of central tendency), 단순 곱 및 가중 곱으로 이루어진 군으로부터 선택된 하나 이상에 의해 계산되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The mean values are geometric mean, arithmetic mean, harmonic mean, arithmetic mean, arithmetic harmonic mean, geometric harmonic mean, Pythagorean mean, heron mean, inverse harmonic mean, mean square deviation, centroid mean, quadrant mean, quadratic mean, cutting mean , Windsing Mean, Weighted Average, Weighted Geometric Mean, Weighted Arithmetic Mean, Weighted Harmonic Mean, Function Mean, Power Average, Generalized f-Mean, Percentile, Maximum, Minimum, Mode, Median, Median Range, Central Trend A method of providing information for uterine contraction inhibitor selection using personal genome sequence variation, which is calculated by one or more selected from the group consisting of measures of central tendency, simple product and weighted product.
  17. 제 1 항에 있어서, The method of claim 1,
    상기 약물 점수는 하기 수학식 3에 의해 산출되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The drug score is calculated by the following Equation 3, method for providing information for the selection of uterine contraction inhibitors using individual genome sequence variation.
    [수학식 3][Equation 3]
    Figure PCTKR2016001631-appb-I000005
    Figure PCTKR2016001631-appb-I000005
    상기 수학식 3에서 Sd는 약물 d의 약물 점수, n은 상기 약물 d의 약력학 또는 약동학에 관여하는 하나 이상의 유전자가 코딩하는 단백질의 수, gi는 상기 약물 d의 약력학 또는 약동학에 관여하는 하나 이상의 유전자가 코딩하는 단백질 손상 점수이며, p는 0이 아닌 실수임.In Equation 3, Sd is a drug score of drug d, n is the number of proteins encoded by one or more genes involved in the pharmacodynamics or pharmacokinetics of drug d, and gi is one or more genes involved in the pharmacodynamics or pharmacokinetics of drug d. Is the protein damage score that p encodes, p is a nonzero real number.
  18. 제 1 항에 있어서, The method of claim 1,
    상기 약물 점수는 하기 수학식 4에 의해 산출되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. Wherein the drug score is calculated by the following equation, method for providing information for uterine contraction inhibitor selection using personal genomic sequence variation.
    [수학식 4][Equation 4]
    Figure PCTKR2016001631-appb-I000006
    Figure PCTKR2016001631-appb-I000006
    상기 수학식 4에서 Sd는 약물 d의 산출된 약물 점수, n은 상기 약물 d의 약력학 또는 약동학에 관여하는 하나 이상의 유전자가 코딩하는 단백질의 수, gi는 상기 약물 d의 약력학 또는 약동학에 관여하는 하나 이상의 유전자가 코딩하는 단백질의 단백질 손상 점수이며, wi는 상기 약물 d의 약력학 또는 약동학에 관여하는 하나 이상의 유전자가 코딩하는 단백질의 단백질 손상점수 gi에 부여되는 가중치임. In Equation 4, Sd is the calculated drug score of drug d, n is the number of proteins encoded by one or more genes involved in the pharmacodynamics or pharmacokinetics of drug d, and gi is the one involved in the pharmacodynamics or pharmacokinetics of drug d. Protein damage score of the protein encoded by the above gene, wi is the weight given to the protein damage score gi of the protein encoded by one or more genes involved in the pharmacodynamics or pharmacokinetics of the drug d.
  19. 제 1 항에 있어서, The method of claim 1,
    상기 단백질 손상 점수 또는 약물 점수는 해당 단백질의 종류, 해당 단백질의 약력학 또는 약동학적 분류, 해당 약물 대사 효소의 약동학 파라미터, 또는 인구 집단 또는 인종별 분포를 고려하여 결정된 값으로 가중치를 부여하여 산출되는 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The protein damage score or drug score is calculated by weighting to a value determined in consideration of the type of the protein, the pharmacokinetic or pharmacokinetic classification of the protein, the pharmacokinetic parameters of the drug metabolizing enzyme, or the population or race distribution. Method for providing information for the selection of inhibitors of uterine contraction using phosphorus, individual genome sequence variation.
  20. 제 1 항에 있어서, The method of claim 1,
    상기 방법은 상기 개인별 약물 점수를 이용하여 상기 개인에 적용되는 자궁수축억제제의 사용 여부 또는 사용 방법을 결정하는 단계를 추가로 포함하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The method further includes determining whether or not to use the uterine contraction inhibitor applied to the individual using the individual drug score, providing information for selecting the uterine contraction inhibitor using a personal genomic sequence variation How to.
  21. 제 1 항에 있어서, The method of claim 1,
    상기 방법은 상기 자궁수축억제제의 약력학 또는 약동학에 관여하는 유전자의 염기서열 변이 정보를 컴퓨터 시스템으로 접수하는 단계를 추가로 포함하며, The method further includes the step of receiving the sequence information of the sequence of the gene involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitor to the computer system,
    상기 컴퓨터 시스템은 상기 자궁수축억제제의 약력학 또는 약동학에 관여하는 유전자 정보를 포함하는 데이터베이스를 포함하거나 또는 상기 데이터베이스에 접근 가능한 것인, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. The computer system provides information for selecting a uterine contraction inhibitor using personal genome sequence variation, wherein the computer system includes or has access to a database containing genetic information involved in the pharmacokinetics or pharmacokinetics of the uterine contraction inhibitor. Way.
  22. 제 1 항 내지 제 21 항 중 어느 한 항에 있어서, The method according to any one of claims 1 to 21,
    상기 방법은 자궁수축억제제 부작용 방지를 목적으로 수행되는 것을 특징으로 하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택을 위한 정보를 제공하는 방법. Wherein the method is carried out for the purpose of preventing adverse effects of uterine contraction inhibitors, a method for providing information for the selection of uterine contraction inhibitors using personal genome sequence variation.
  23. 개인에게 적용할 수 있는 자궁수축억제제에 대하여, 상기 자궁수축억제제와 관련된 유전자 또는 단백질과 관련된 정보 검색 또는 추출이 가능한 데이터베이스;A database capable of searching or extracting information related to genes or proteins related to the uterine contraction inhibitors, which may be applied to an individual;
    상기 데이터베이스에 접근 가능한 통신부; A communication unit accessible to the database;
    상기 정보에 기초하여 상기 자궁수축억제제의 약력학 또는 약동학에 관여하는 하나 이상의 유전자 염기서열 변이 정보를 산출하는 제1 산출모듈; A first calculation module configured to calculate one or more gene sequence variation information related to pharmacodynamics or pharmacokinetics of the uterine contraction inhibitor based on the information;
    상기 유전자 염기서열 변이 정보를 이용하여 개인별 단백질 손상 점수를 산출하는 제2 산출모듈; A second calculation module for calculating an individual protein damage score using the gene sequence variation information;
    상기 개인별 단백질 손상 점수를 약물과 단백질 사이의 상호 관계와 연관지어, 개인별 약물 점수를 산출하는 제3 산출모듈; 및A third calculating module that calculates an individual drug score by associating the individual protein damage score with a correlation between a drug and a protein; And
    상기 산출모듈에서 산출된 산출값을 표시하는 표시부를 포함하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 시스템. And a display unit for displaying the calculated value calculated by the calculation module.
  24. 제 23 항에 있어서, The method of claim 23,
    상기 시스템은 상기 제3 산출모듈에서 산출된 상기 개인별 약물 점수를 이용하여 상기 개인에 적용되는 자궁수축억제제의 사용 여부를 결정하는 제4 산출모듈을 추가로 포함하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 시스템. The system further comprises a fourth calculation module for determining whether or not to use the uterine contraction inhibitor applied to the individual using the individual drug score calculated in the third calculation module, the uterus using a personal genome sequence variation Shrinkage Inhibitor Selection System.
  25. 제 23 항에 있어서, The method of claim 23,
    상기 시스템은 사용자에 의한 자궁수축억제제의 입력에 따라 상기 자궁수축억제제에 대한 개인별 약물 점수를 산출하여 제공하는 사용자 인터페이스를 추가로 포함하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 시스템. The system further comprises a user interface for calculating and providing a personal drug score for the uterine contraction inhibitor according to the input of the uterine contraction inhibitor, the uterine contraction inhibitor selection system using a personal genome sequence variation.
  26. 제 23 항에 있어서, The method of claim 23,
    상기 표시부는 상기 각 산출모듈에서 산출된 값, 계산 과정, 또는 상기 계산의 기초가 된 정보를 추가로 표시하는 표시부를 추가로 포함하는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 시스템. The display unit further comprises a display unit for additionally displaying a value calculated by each calculation module, a calculation process, or the information on which the calculation is based, uterine contraction inhibitor selection system using a personal genome sequence variation.
  27. 제 23 항 내지 제 26 항 중 어느 한 항에 있어서, The method according to any one of claims 23 to 26,
    상기 각 산출모듈은 상기 유전자 염기서열 변이 정보, 단백질 손상 점수, 약물 점수 및 그 산출의 근거가 되는 정보가 저장되며,Each calculation module stores the gene sequence variation information, protein damage score, drug score, and information on which the calculation is based.
    상기 데이터베이스의 갱신에 따라 상기 각 산출모듈의 정보가 갱신되는, 개인 유전체 염기서열 변이를 이용한 자궁수축억제제 선택 시스템. The information on each calculation module is updated according to the update of the database, the uterine contraction inhibitor selection system using a personal genome sequence variation.
  28. 하기 프로세서를 실행시키는 실행모듈을 포함하는 컴퓨터 판독 가능한 매체: A computer readable medium comprising an execution module for executing the processor:
    개인 유전체 염기서열 정보로부터 자궁수축억제제의 약력학 또는 약동학에 관여하는 하나 이상의 유전자 염기서열 변이 정보를 입수하는 단계; Obtaining one or more gene sequence variation information related to pharmacokinetics or pharmacokinetics of the uterine contraction inhibitor from the individual genome sequence information;
    상기 유전자 염기서열 변이 정보를 이용하여 개인별 단백질 손상 점수를 산출하는 단계; 및Calculating an individual protein damage score using the gene sequence variation information; And
    상기 개인별 단백질 손상 점수를 약물과 단백질 사이의 상호 관계와 연관지어, 개인별 약물 점수를 산출하는 단계;를 포함하는 동작을 수행하는 프로세서.Computing the individual drug score by associating the individual protein damage score with the correlation between the drug and the protein.
  29. 제 28 항에 있어서, The method of claim 28,
    상기 프로세서는 상기 개인별 약물 점수를 이용하여 상기 개인에 적용되는 자궁수축억제제의 사용 여부를 결정하는 단계;를 추가로 포함하는, 컴퓨터 판독 가능한 매체.And determining, by the processor, whether or not to use a uterine contraction inhibitor applied to the individual by using the individual drug score.
PCT/KR2016/001631 2015-02-17 2016-02-17 Method of selecting uterine contraction inhibiting agent based on protein damage information on each individual to prevent side effects of uterine contraction inhibiting agent WO2016133374A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2015-0024450 2015-02-17
KR20150024450 2015-02-17

Publications (1)

Publication Number Publication Date
WO2016133374A1 true WO2016133374A1 (en) 2016-08-25

Family

ID=56692233

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2016/001631 WO2016133374A1 (en) 2015-02-17 2016-02-17 Method of selecting uterine contraction inhibiting agent based on protein damage information on each individual to prevent side effects of uterine contraction inhibiting agent

Country Status (2)

Country Link
KR (1) KR20160101706A (en)
WO (1) WO2016133374A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107144695A (en) * 2017-04-19 2017-09-08 南昌大学 Application of the Arl13b albumen in cancer diagnosis
CN108566387A (en) * 2018-03-27 2018-09-21 中国工商银行股份有限公司 Method, equipment and the system of data distribution are carried out based on udp protocol

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102403778B1 (en) * 2020-02-05 2022-05-30 서울대학교산학협력단 Biomarker for diagnosing pulmonary edema and use thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20020008111A (en) * 1998-12-28 2002-01-29 다케다 야쿠힌 고교 가부시키가이샤 Screening method
US20030096264A1 (en) * 2001-06-18 2003-05-22 Psychiatric Genomics, Inc. Multi-parameter high throughput screening assays (MPHTS)
JP2006228079A (en) * 2005-02-21 2006-08-31 Hitachi Ltd Chemical information support system
WO2012167278A1 (en) * 2011-06-02 2012-12-06 Almac Diagnostics Limited Molecular diagnostic test for cancer
KR20140048673A (en) * 2012-10-16 2014-04-24 안형준 Medicine retrieval system minimized individual side-effects based on private single nucleotide polymorphism and the method thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20020008111A (en) * 1998-12-28 2002-01-29 다케다 야쿠힌 고교 가부시키가이샤 Screening method
US20030096264A1 (en) * 2001-06-18 2003-05-22 Psychiatric Genomics, Inc. Multi-parameter high throughput screening assays (MPHTS)
JP2006228079A (en) * 2005-02-21 2006-08-31 Hitachi Ltd Chemical information support system
WO2012167278A1 (en) * 2011-06-02 2012-12-06 Almac Diagnostics Limited Molecular diagnostic test for cancer
KR20140048673A (en) * 2012-10-16 2014-04-24 안형준 Medicine retrieval system minimized individual side-effects based on private single nucleotide polymorphism and the method thereof

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107144695A (en) * 2017-04-19 2017-09-08 南昌大学 Application of the Arl13b albumen in cancer diagnosis
CN107144695B (en) * 2017-04-19 2019-02-26 南昌大学 Application of the Arl13b albumen in cancer diagnosis
CN108566387A (en) * 2018-03-27 2018-09-21 中国工商银行股份有限公司 Method, equipment and the system of data distribution are carried out based on udp protocol
CN108566387B (en) * 2018-03-27 2021-08-20 中国工商银行股份有限公司 Method, equipment and system for data distribution based on UDP protocol

Also Published As

Publication number Publication date
KR20160101706A (en) 2016-08-25

Similar Documents

Publication Publication Date Title
US20210241849A1 (en) Method for personalized selection of a drug for a subject
Lee et al. Genome-wide pathway analysis of genome-wide association studies on systemic lupus erythematosus and rheumatoid arthritis
Leung et al. Noninvasive twin zygosity assessment and aneuploidy detection by maternal plasma DNA sequencing
US8417459B2 (en) Methods of selection, reporting and analysis of genetic markers using broad-based genetic profiling applications
Sobreira et al. Patients with a Kabuki syndrome phenotype demonstrate DNA methylation abnormalities
Peters et al. Detection and phasing of single base de novo mutations in biopsies from human in vitro fertilized embryos by advanced whole-genome sequencing
Wilson et al. Analysis of IL10 haplotypic associations with severe malaria
Baye et al. Mapping genes that predict treatment outcome in admixed populations
Ning et al. Association between the polymorphisms of interleukin-4, the interleukin-4 receptor gene and asthma
Bourchany et al. Reducing diagnostic turnaround times of exome sequencing for families requiring timely diagnoses
WO2016133374A1 (en) Method of selecting uterine contraction inhibiting agent based on protein damage information on each individual to prevent side effects of uterine contraction inhibiting agent
Melton et al. Whole-exome sequencing in multiplex preeclampsia families identifies novel candidate susceptibility genes
Kumar et al. Genetic association of key Th1/Th2 pathway candidate genes, IRF2, IL6, IFNGR2, STAT4 and IL4RA, with atopic asthma in the Indian population
Mezzavilla et al. Increased rate of deleterious variants in long runs of homozygosity of an inbred population from Qatar
Mao et al. Advanced whole-genome sequencing and analysis of fetal genomes from amniotic fluid
Phani et al. Genetic association of KCNJ10 rs1130183 with seizure susceptibility and computational analysis of deleterious non-synonymous SNPs of KCNJ10 gene
Ursini et al. Prioritization of potential causative genes for schizophrenia in placenta
Liu et al. Cross-ancestry genome-wide association meta-analyses of hippocampal and subfield volumes
Ruaño et al. Physiogenomic analysis of the Puerto Rican population
KR102482819B1 (en) Method for personalized prevention of adverse drug reaction of anticancer drug based on information of individual deleterious protein sequence variation
Cali et al. Biallelic PRMT7 pathogenic variants are associated with a recognizable syndromic neurodevelopmental disorder with short stature, obesity, and craniofacial and digital abnormalities
WO2016133375A1 (en) Method for selecting osteoporosis therapeutic agent based on protein damage information of individual to prevent osteoporosis therapeutic agent side effects
WO2017074036A2 (en) Method and system for selecting customized drug using genomic nucleotide sequence variation information and survival information of cancer patient
Duconge et al. Clinical implications of genetic admixture in Hispanic Puerto Ricans: impact on the pharmacogenetics of CYP2C19 and PON1
Liedtke et al. Correlated expression analysis of genes implicated in schizophrenia: identification of putative disease-related pathways

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: 16752704

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: 16752704

Country of ref document: EP

Kind code of ref document: A1