[go: up one dir, main page]

WO2023146370A1 - Composition and method for detecting nucleic acid based on fluorescence signal capable of specifically detecting egfr mutation - Google Patents

Composition and method for detecting nucleic acid based on fluorescence signal capable of specifically detecting egfr mutation Download PDF

Info

Publication number
WO2023146370A1
WO2023146370A1 PCT/KR2023/001357 KR2023001357W WO2023146370A1 WO 2023146370 A1 WO2023146370 A1 WO 2023146370A1 KR 2023001357 W KR2023001357 W KR 2023001357W WO 2023146370 A1 WO2023146370 A1 WO 2023146370A1
Authority
WO
WIPO (PCT)
Prior art keywords
mutation
atto
nucleic acid
dna
crispr
Prior art date
Legal status (The legal status 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 status listed.)
Ceased
Application number
PCT/KR2023/001357
Other languages
French (fr)
Korean (ko)
Inventor
배태근
이재준
이효민
강승훈
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Feroka Inc
Original Assignee
Feroka Inc
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 Feroka Inc filed Critical Feroka Inc
Publication of WO2023146370A1 publication Critical patent/WO2023146370A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6827Hybridisation assays for detection of mutation or polymorphism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases [RNase]; Deoxyribonucleases [DNase]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPR]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2563/00Nucleic acid detection characterized by the use of physical, structural and functional properties
    • C12Q2563/107Nucleic acid detection characterized by the use of physical, structural and functional properties fluorescence
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2565/00Nucleic acid analysis characterised by mode or means of detection
    • C12Q2565/10Detection mode being characterised by the assay principle
    • C12Q2565/101Interaction between at least two labels
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • It relates to a composition and method for detecting a nucleic acid based on a fluorescence signal capable of specifically detecting an EGFR mutation.
  • NSCLC non-small lung cancer
  • a tyrosine kinase inhibitor a targeted cancer treatment for non-small lung cancer (NSCLC) has been shown to have a strong therapeutic effect on EGFR mutation-positive NSCLC, L858R, a major biomarker for NSCLC, and early and stable detection of EGFR mutations of exon 19 deletion is very important for effective treatment.
  • NGS Next Generation Sequencing
  • CRISPR Clustered regular interspaced short palindromic repeats
  • Cas CRISPR Associated Protein
  • One aspect hybridizes to a target sequence and includes a guide polynucleotide comprising consecutive bases complementary to an epidermal growth factor receptor (EGFR) mutant gene sequence or a nucleic acid encoding the guide polynucleotide; A CRISPR/Cas effector protein, or a nucleic acid encoding the protein; and a single-stranded, EGFR mutation-detecting composition comprising a labeled detection nucleic acid that hybridizes with the guide polynucleotide.
  • EGFR epidermal growth factor receptor
  • Another aspect is to provide a kit for detecting an EGFR mutation comprising the composition.
  • Another aspect includes contacting a sample with the composition; And detecting a target sequence in the sample by measuring a detectable signal generated by cleavage of the single-stranded detection nucleic acid by the CRISPR / Cas effector protein.
  • Another aspect is to provide a cancer diagnosis kit including the composition.
  • Another aspect is to provide a guide polynucleotide comprising a polynucleotide comprising any one of SEQ ID NOs: 2-6 or a polynucleotide having at least 95% homology to a polynucleotide of any one of SEQ ID NOs: 2-6.
  • Another aspect is to provide a method of diagnosing cancer or providing information related to cancer diagnosis, including contacting the composition with a sample.
  • One aspect hybridizes to a target sequence and includes a guide polynucleotide comprising consecutive bases complementary to an epidermal growth factor receptor (EGFR) mutant gene sequence or a nucleic acid encoding the guide polynucleotide; A CRISPR/Cas effector protein, or a nucleic acid encoding the protein; and a single-stranded, labeled detection nucleic acid that hybridizes with the guide polynucleotide.
  • EGFR epidermal growth factor receptor
  • Another aspect is to provide a kit for detecting an EGFR mutation comprising the composition.
  • the term "protospacer adjacent motif (PAM)” is a sequence recognized by the Cas protein of the CRISPR/Cas system.
  • the protospacer sequence is a sequence located at the 5' or 3' end of the PAM sequence and directs binding of the PAM effector protein complex to the target locus of interest.
  • the PAM may include a 5' T-rich motif.
  • the PAM may consist of a 2 to 10 bp DNA sequence, preferably a 2 to 6 bp DNA sequence.
  • guide polynucleotide is a short synthetic RNA molecule used for genome editing based on the CRISPR system.
  • a "guide polynucleotide” is a spacer sequence that binds to a target DNA.
  • the gRNA molecule consists of a scaffold sequence for Cas binding, an endonuclease.
  • the guide nucleotide includes guide RNA, gRNA, or guide RNA.
  • the guide polynucleotide includes crRNA.
  • the crRNA is a partial sequence present within the crRNA that binds and/or interacts with tracrRNA and/or effector proteins.
  • the crRNA may be a wild-type crRNA or an engineered crRNA.
  • the crRNA may include a direct repeat sequence and a spacer, and the direct repeat sequence may be located at the 5' end of the spacer.
  • the crRNA may be located at the 3' end of tracrRNA.
  • the guide polynucleotide includes tracrRNA.
  • the tracrRNA scaffold sequence is all or part of a tracrRNA sequence that binds and/or interacts with a crRNA and/or an effector protein.
  • the tracrRNA may be a wild-type tracrRNA or an engineered tracrRNA.
  • the engineered crRNA or tracrRNA may be a sequence in which a part of the nucleotide sequence of the wild-type crRNA or tracrRNA is artificially modified (substituted, deleted, or inserted) or modified to be shorter than the wild-type crRNA or tracrRNA sequence.
  • the guide polynucleotide may further include a linker.
  • the linker is a sequence serving to connect the tracrRNA and crRNA.
  • the linker may be a sequence of 1 to 30 nucleotides. In one embodiment, the linker may be 1 to 5, 5 to 10, 10 to 15, 15 to 20, 20 to 25, or 25 to 30 nucleotide sequences.
  • the linker may be a 5'-GAAA-3' sequence, but is not limited thereto.
  • the guide polynucleotide may be an engineered guide RNA in which one or more nucleotide sequences are deleted, substituted, or added from the wild-type guide polynucleotide.
  • the guide sequence can be any polynucleotide sequence that has sufficient complementarity with the target polynucleotide sequence to hybridize with the target sequence and direct sequence-specific binding of the CRISPR complex to the target sequence.
  • the gRNA forms a complex with the Cas protein to bind to a protospacer adjacent motif (PAM) and a target polynucleotide sequence having a protospacer (sequence complementary to a portion of the gRNA).
  • Guide RNA is the element that confers the target specificity of the gRNA:Cas protein complex.
  • EGFR can be used interchangeably with epidermal growth factor receptor as an abbreviation of Epidermal Growth Factor Receptor.
  • EGFR is part of a group of tyrosine kinase receptors, also called HER or the erbB family, which includes EGFR (HER1/ErbB1), HER2 (ErbB2), HER3 (ErbB3) and HER4 (ErbB4).
  • the EGFR mutant gene sequence may be a guide polynucleotide linked to the 3' end of a protospacer adjacent motif (PAM). More specifically, the protospacer adjacent motif (PAM) may include a 5' T-rich motif.
  • PAM protospacer adjacent motif
  • mutation refers to a case in which there is a permanent change in the nucleotide sequence of an existing gene. Depending on the effect on the structure of genetic material, mutations can occur when there is an abnormality in the structure or number of chromosomes (chromosomal abnormality) and when a change occurs on a small scale in the nucleotide sequence of a gene. Examples of abnormalities in the structure of chromosomes include deletion, duplication, inversion, and translocation. As used herein, the term “deletion mutation” refers to a case in which a portion of a chromosome is lost. As used herein, the term “point mutation” refers to a nucleotide sequence constituting a locus that occurs when one or more thousands of pairs are deleted.
  • EGFR mutation herein means G719S mutation, G179C mutation, G719A mutation, S720F mutation, T790M mutation, D761Y mutation, exon 19 deletion mutation, D770_N771 insertion mutation, V765A mutation, T783A mutation, S761I mutation, T790M mutation, V769L mutation , N771I mutation, L858R mutation, L861Q mutation, L861R mutation.
  • Representative EGFR mutation L858R is a point mutation in which the 858th amino acid is changed from Leusine (L) to Arginine (Arginine, R), and is a mutation belonging to exon 21.
  • EGFR Exon 19 deletion mutant means that exon 19 of the gene encoding EGFR is deleted. Unlike the L858R point mutation, EGFR Exon19 deletion mutants appear in various deletion sizes, such as 9bp to 18bp of exon19.
  • the cancer may have an EGFR mutation.
  • the cancer may be a solid cancer.
  • the solid cancer refers to an abnormal tissue mass originating from an organ. Solid cancers can be malignant. Different types of solid cancers are named according to the type of cells that form them. Types of solid cancer include sarcomas, carcinomas and lymphomas.
  • solid cancers include adrenal cancer, anal cancer, anaplastic large cell lymphoma, angioimmunoblastic T cell lymphoma, B cell lymphoma, cholangiocarcinoma, bladder cancer, brain/CNS tumor, breast cancer, cervical cancer, colon cancer, endometrial cancer, esophageal cancer, Tumors of the Ewing family, eye cancer, gallbladder cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (gist), gestational trophoblast disease, hepatosplenic T-cell lymphoma, Hodgkin's lymphoma, intravascular large B-cell lymphoma, renal cancer, laryngeal cancer and hypopharyngeal cancer, liver cancer, lung cancer (non-small cell and small cell), lung carcinoid lymphomatous granulomatosis, malignant mesothelioma, nasal and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, no
  • EGFR mutations can cause colorectal cancer, pancreatic cancer, glioma, head and neck cancer, and lung cancer, particularly non-small cell lung cancer (NSCLC), among others.
  • NSCLC non-small cell lung cancer
  • it is particularly effective for diagnosis of solid cancer selected from the group consisting of colorectal cancer, pancreatic cancer, glioma, head and neck cancer and lung cancer, and even more for diagnosis of non-small cell lung cancer (NSCLC).
  • the guide polynucleotide may be a guide polynucleotide having a length of 20 bp to 50 bp consisting of a scafold sequence and a target sequence in order from the 5' end to the 3' end.
  • the guide sequence is about 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 in length. , 29, 30, 35, 40, 45, 50, 75 bp or more.
  • the guide sequence is less than about 75, 50, 45, 40, 35, 30, 25, 20, 15, 12 or less nucleotides in length.
  • the guide nucleotide sequence preferably has bases of 20 bp to 50 bp, more preferably 30 bp to 50 bp in length.
  • the guide polynucleotide may further include a sequence that increases cleavage efficiency.
  • the sequence increasing the cleavage efficiency may be a T-rich tailing sequence.
  • the guide polynucleotide may include nucleotide sequences represented by SEQ ID NOs: 2 to 6. In the nucleotides represented by SEQ ID NOs: 2 to 6, T (Thymine) may be U (Uracil).
  • the method comprises the step of measuring (e.g., a V-type CRISPR/Cas effector protein (e.g., a Cas12 protein such as Cas12a, Cas12b, Cas12c, Cas12d, Cas12e)-mediated ssDNA cleavage measuring a detectable signal that becomes).
  • a V-type CRISPR/Cas effector protein e.g., a Cas12 protein such as Cas12a, Cas12b, Cas12c, Cas12d, Cas12e
  • a V-type CRISPR/Cas effector protein e.g., a Cas12 protein such as Cas12a, Cas12b, Cas12c, Cas12d, Cas12e
  • the guide RNA is a V-type CRISPR/Cas effector protein
  • the detectable signal can be any signal generated when ssDNA is cleaved.
  • the measuring step may include gold nanoparticle based detection, fluorescence polarization, colloidal phase transition/dispersion, electrochemical detection, semiconductor-based sensing;
  • phosphatase can be used to generate a pH change after ssDNA cleavage reaction by releasing inorganic phosphatase into solution by opening 2'-3' cyclic phosphatase), and detection of labeled detector ssDNA.
  • the readout of this detection method can be any convenient readout.
  • Examples of possible readouts include a measurable amount of detectable fluorescence signal; Visual analysis of bands on the gel (e.g., bands representing cleaved product relative to uncleaved substrates), visual or sensor-based detection of the presence or absence of color (i.e., color detection methods), and electrical signal (or specific positive) presence or absence, but is not limited thereto.
  • the step of measuring can in some cases be quantitative in the sense that, for example, the amount of signal detected can be used to determine the amount of target DNA present in a sample.
  • the step of measuring can in some cases be qualitative, for example in the sense that the presence or absence of a detectable signal can indicate the presence or absence of targeted DNA. In some cases, no detectable signal will be present (eg, above a given threshold level) unless the otherwise targeted DNA(s) are present above a certain threshold concentration.
  • the detection threshold can be titrated by varying the amount of CRISPR/Cas effector protein, guide RNA, sample volume and/or detector ssDNA (if one is used).
  • the methods of the present disclosure can be used to determine the amount of a target nucleic acid (eg, a sample comprising a target DNA and a plurality of non-target DNAs). Determining the amount of target DNA in the sample may include comparing the amount of detectable signal produced from the test sample to the amount of detectable signal produced from a reference sample. Determining the amount of target DNA in a sample includes measuring a detectable signal to generate a test measurement; measuring a detectable signal produced by the reference sample to produce a reference measurement; and comparing the test measurement to a reference measurement to determine the amount of target DNA present in the sample.
  • a target nucleic acid eg, a sample comprising a target DNA and a plurality of non-target DNAs. Determining the amount of target DNA in the sample may include comparing the amount of detectable signal produced from the test sample to the amount of detectable signal produced from a reference sample. Determining the amount of target DNA in a sample includes measuring a detectable signal to generate a test measurement; measuring
  • methods of the present disclosure for determining the amount of target DNA in a sample may include: a) a sample (eg, a sample comprising a target DNA and a plurality of non-target DNAs): i) a guide RNA that hybridizes with the target DNA, (ii) a CRISPR/Cas effector protein that cleave nucleic acids present in the sample, and (iii) a detection ssDNA; b) measuring a detectable signal generated by CRISPR/Cas effector protein-mediated ssDNA cleavage (eg, cleavage of the detector ssDNA) to generate a test measurement; c) measuring a detectable signal produced by the reference sample to produce a reference measurement; and d) comparing the test measurement to a reference measurement to determine the amount of target DNA present in the sample.
  • a sample eg, a sample comprising a target DNA and a plurality of non-target DNAs
  • a guide RNA that hybridizes
  • the sensitivity of a subject composition and/or method can be increased by combining detection with nucleic acid amplification.
  • nucleic acids in the sample are amplified before, simultaneously with, or after contact with a CRISPR/Cas effector protein (eg, a Cas12 protein) that cuts the ssDNA.
  • nucleic acids in the sample are amplified concurrently with contact with a CRISPR/Cas effector protein (eg, Cas12 protein).
  • CRISPR/Cas effector protein eg, Cas12 protein
  • amplification component and detection component such as CRISPR/Cas effector protein, eg, Cas12 protein, guide RNA, and detector DNA.
  • -cleavage activity is capable of starting to degrade the nucleic acid of the sample at the same time as the nucleic acid undergoes amplification.
  • Nucleic acid amplification includes polymerase chain reaction (PCR), reverse transcription PCR (RT-PCR), quantitative PCR (qPCR), reverse transcription qPCR (RT-qPCR), nested PCR, multiplex PCR, asymmetric PCR, touchdown PCR, random primer PCR, hemi-nested PCR, polymerase cycling assembly (PCA), colony PCR, ligase chain reaction (LCR), digital PCR, methylation specific-PCR (MSP), co-at low denaturation temperature Amplification-PCR (COLD-PCR), allele-specific PCR, intersequence-specific PCR (ISS-PCR), whole genome amplification (WGA), inverse PCR, and thermal asymmetric interlaced PCR (TAIL- PCR) may be included.
  • PCR polymerase chain reaction
  • RT-PCR reverse transcription PCR
  • qPCR quantitative PCR
  • RT-qPCR reverse transcription qPCR
  • nested PCR multiplex PCR
  • asymmetric PCR touchdown PCR
  • amplification is isothermal amplification.
  • the term “isothermal amplification” refers to a method of amplifying nucleic acids (eg, DNA) (eg, using an enzymatic chain reaction) that can use a single temperature incubation to eliminate the need for a thermal cycler.
  • Isothermal amplification is a form of nucleic acid amplification that does not rely on thermal denaturation of target nucleic acids during the amplification reaction and thus may not require multiple rapid changes in temperature.
  • isothermal nucleic acid amplification methods can be performed either within or outside of a laboratory environment. In combination with a reverse transcription step, these amplification methods can be used to amplify RNA isothermally.
  • isothermal amplification methods include loop-mediated isothermal amplification (LAMP), helicase-dependent amplification (HDA), recombinase polymerase amplification (RPA), strand displacement amplification (SDA), nucleic acid sequence- based amplification (NASBA), transcription-mediated amplification (TMA), nickase amplification reaction (NEAR), rolling circle amplification (RCA), multiple displacement amplification (MDA), ramification (RAM), circular helicase -Dependent Amplification (cHDA), Single Primer Isothermal Amplification (SPIA), Signal-Mediated Amplification of RNA Technology (SMART), Self-Sustained Sequence Replication (3SR), Genomic Exponential Amplification Reaction (GEAR) and Isothermal Multiple Displacement Amplification (IMDA) Including, but not limited to.
  • LAMP loop-mediated isothermal amplification
  • HDA helicase-dependent amplification
  • RPA recombinase polymerase
  • the amplification is recombinase polymerase amplification (RPA).
  • RPA recombinase polymerase amplification
  • SSB single-stranded DNA-binding protein
  • SSB strand-displacement polymerase
  • a subject method comprises a sample (eg, a sample comprising a target DNA and a plurality of non-target ssDNAs) comprising: i) a CRISPR/Cas effector protein; ii) guide RNA (or precursor guide RNA array); and iii) a detection DNA that is single-stranded and does not hybridize with the guide sequence of the guide RNA.
  • a subject method comprises contacting a sample with a labeled single-stranded detection nucleic acid (detection ssDNA) comprising a fluorescence-emitting dye pair;
  • a CRISPR/Cas effector protein e.g., a Cas12 protein such as Cas12a, Cas12b, Cas12c, Cas12d, Cas12e
  • a detectable signal to be measured is produced by the fluorescence-emitting dye pair.
  • the subject methods include contacting the sample with labeled detection ssDNA comprising a fluorescence resonance energy transfer (FRET) pair or a quencher/fluor (quencher/fluor) pair or both.
  • a subject method comprises contacting a sample with a labeled detection ssDNA comprising a FRET pair.
  • the subject methods include contacting the sample with a labeled detection ssDNA comprising a fluorine/quencher pair.
  • Fluorescence-emitting dye pairs include FRET pairs or quencher/fluor pairs. In both the case of the FRET pair and the quencher/fluorine pair, the emission spectrum of one of the dyes overlaps the region of the absorption spectrum of the other dye in the pair.
  • fluorescence-emitting dye pair is a generic term used to include both a "fluorescence resonance energy transfer (FRET) pair” and a "quencher/fluorine pair", both of which are further hereinafter. discuss in detail
  • FRET fluorescence resonance energy transfer
  • quencher/fluorine pair both of which are further hereinafter. discuss in detail
  • fluorescence-emitting dye pair is used interchangeably with the phrase “FRET pair and/or quencher/fluorine pair”.
  • the labeled detection ssDNA produces an amount of detectable signal before being cleaved, and the amount of detectable signal measured is the amount of the detected signal when the labeled detection ssDNA is cleaved. is reduced
  • the labeled detection ssDNA generates a first detectable signal before being cleaved (e.g., from a FRET pair) and the first detectable signal is generated when the labeled detection ssDNA is cleaved (e.g., from a quencher/fluor pair). 2 Generates a detectable signal.
  • the labeled detection ssDNA includes a FRET pair and a quencher/fluor pair.
  • a donor-acceptor pair (a FRET donor moiety and a FRET acceptor moiety) is referred to herein as a "FRET pair” or a "signal FRET pair".
  • FRET pair a FRET donor moiety and a FRET acceptor moiety
  • signal FRET pair a signal partner
  • the target labeled detection ssDNA contains two signal partners (signal pairs).
  • a target labeled detector ssDNA containing such a FRET pair (a FRET donor moiety and a FRET acceptor moiety) produces a detectable signal (the FRET signal) when the signal counterpart is very proximal (e.g., while on the same RNA molecule).
  • RNA molecule of a CRISPR/Cas effector protein e.g., a Cas12 protein such as Cas12a, Cas12b, Cas12c, Cas12d, Cas12e
  • a Cas12 protein such as Cas12a, Cas12b, Cas12c, Cas12d, Cas12e
  • FRET donor and acceptor moieties will be known to those skilled in the art, and any convenient FRET pair (eg, any convenient donor and acceptor moiety pair) may be used.
  • a detectable signal is generated when the labeled detector ssDNA is cleaved (eg, in some cases, the labeled detector ssDNA includes a quencher/fluor pair).
  • One signal partner of a signal quenching pair produces a detectable signal
  • the other signal partner is a quencher moiety that quenches the detectable signal of the first signal partner (i.e. when the signal partners are proximal to each other, e.g.
  • the quencher moiety quenches the signal of the signal moiety such that the signal from the signal moiety is reduced (quenched) when the signal counterpart of the signal pair is proximal).
  • the amount of detectable signal is increased when the labeled detection ssDNA is cleaved.
  • the signal exhibited by one signal partner is the signal exhibited by the other signal partner.
  • quencher signal moiety Such signal pairs are referred to herein as “quencher/fluor pairs", “quenching pairs” or “signal quenching pairs”.
  • one signal counterpart e.g., a first signal counterpart
  • a second signal counterpart e.g., a quencher moiety
  • the signal partners of this quencher/fluor pair will produce a detectable signal when the partners are separated (e.g., after cleavage of the detector ssDNA by the V-type CRISPR/Cas effector protein), but when the partners are very proximal ( eg, before cleavage of the detection ssDNA by the CRISPR/Cas effector protein) the signal will be quenched.
  • the quencher moiety can quench the signal to varying degrees from the signal moiety (eg, prior to cleavage of the detection ssDNA by the CRISPR/Cas effector protein).
  • a quencher moiety is a signal if the signal detected in the presence of the quencher moiety (when the signal counterparts are proximal to each other) is no more than 95% of the signal detected in the absence of the quencher moiety (when the signal counterparts are separated). Quench the signal from the moiety.
  • the signal detected in the presence of the quencher moiety is 90% or less, 80% or less, 70% or less, 60% or less, 50% or less, 40% or less of the signal detected in the absence of the quencher moiety. or less, 30% or less, 20% or less, 15% or less, 10% or less, or 5% or less. In some cases, no signal (eg, above background) is detected in the presence of the quencher moiety.
  • the signal detected in the absence of the quencher moiety is at least 1.2-fold greater than the signal detected in the presence of the quencher moiety (when the signal partners are proximal to each other) (e.g., at least 1.3x, at least 1.5x, at least 1.7x, at least 2x, at least 2.5x, at least 3x, at least 3.5x, at least 4x, at least 5x, at least 7x, at least 10x, at least 20x, or at least 50 times higher).
  • the signal moiety is a fluorescent label.
  • the quencher moiety quenches the signal from the fluorescent label (light signal) (eg, by absorbing energy in the label's emission spectrum).
  • emission (signal) from the fluorescent label is detectable because no signal is absorbed by the quencher moiety.
  • Any convenient donor acceptor pair (signal moiety/quencher moiety pair) may be used, and many suitable pairs are known in the art.
  • the quencher moiety absorbs energy from the signal moiety (also referred to herein as a "detectable label") and then emits a signal (eg, light at a different wavelength).
  • the quencher moiety is the signal moiety itself (e.g., the signal moiety can be 6-carboxyfluorescein, while the quencher moiety can be 6-carboxy-tetramethylrhodamine ), in some cases the pair may also be a FRET pair.
  • the quencher moiety is a dark quencher. The dark quencher absorbs the excitation energy and dissipates the energy in a different way (eg as heat). Thus, the dark quencher has minimal or no fluorescence of its own (it emits no fluorescence).
  • fluorescent labels include Alexa Fluor® dye, ATTO dye (e.g., ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532, ATTO Rho6G , ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725, ATTO 740), a DyLight dye, a cyanine dye (eg, Cy2, Cy3, Cy3.5, Cy3b, Cy5, Cy5.
  • ATTO dye e.g., ATTO 390, ATTO 425, ATTO 465, AT
  • FluoProbes dye Sulfo Cy dye, Seta dye, IRIS dye, SeTau dye, SRfluor dye, Square dye, Fluorescein isothiocyanate ( FITC), tetramethylrhodamine (TRITC), Texas Red, Oregon Green, Pacific Blue, Pacific Green, Pacific Orange, Quantum Dots and Tethered (tethered) fluorescent proteins, but are not limited thereto.
  • the detectable label is an Alexa Fluor® dye, an ATTO dye (e.g., ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532 , ATTO Rho6G, ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725, ATTO 740), a DyLight dye, a cyanine dye (e.g., ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, AT
  • fluorescein FITC
  • TRITC tetramethylrhodamine
  • the detectable label is an Alexa Fluor® dye, an ATTO dye (e.g., ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532 , ATTO Rho6G, ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725, ATTO 740), a DyLight dye, a cyanine dye (e.g., ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, AT
  • fluorescein FITC
  • TRITC tetramethylrhodamine
  • ATTO dyes include, but are not limited to: ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532, ATTO Rho6G, ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725 and ATTO 740.
  • Alexa Fluor dyes include, but are not limited to: Alexa Fluor® 350, Alexa Fluor® 405, Alexa Fluor® 430, Alexa Fluor® 488, Alexa Fluor® 500, Alexa Fluor® 514, Alexa Fluor® 532, Alexa Fluor® 546, Alexa Fluor® 555, Alexa Fluor® 568, Alexa Fluor ( Alexa Fluor (registered trademark) 610, Alexa Fluor (registered trademark) 633, Alexa Fluor (registered trademark) 635, Alexa Fluor (registered trademark) 647, Alexa Fluor (registered trademark) 660, Alexa Fluor (registered trademark) ) 680, Alexa Fluor (registered trademark) 700, Alexa Fluor (registered trademark) 750, Alexa Fluor (registered trademark) 790, etc.
  • quencher moieties include dark quencher, black hole quencher (registered trademark) (BHQ®) (e.g., BHQ-0, BHQ-1, BHQ-2, BHQ -3), Qxl Quencher, ATTO Quencher (eg ATTO 540Q, ATTO 580Q, and ATTO 612Q), Dimethylaminoazobenzenesulfonic Acid (Dabsyl), Iowa Black RQ, Iowa Black FQ, IRDye QC-1, QSY dyes (e.g., QSY 7, QSY 9, QSY 21), AbsoluteQuencher, Eclipse, and metal clusters such as gold nanoparticles, etc. don't
  • the quencher moiety is a dark quencher, black hole quencher® (BHQ®) (e.g., BHQ-0, BHQ-1, BHQ-2, BHQ-3), Qxl quencher, ATTO quencher (e.g. ATTO 540Q, ATTO 580Q and ATTO 612Q), dimethylaminoazobenzenesulfonic acid (dansyl), Iowa Black RQ, Iowa Black FQ, IRDye QC-1, QSY dyes (e.g. QSY 7, QSY 9, QSY 21), Absolute Quencher, Eclipse and Metal Cluster.
  • BHQ® black hole quencher
  • ATTO quencher e.g. ATTO 540Q, ATTO 580Q and ATTO 612Q
  • dimethylaminoazobenzenesulfonic acid dansyl
  • Iowa Black RQ Iowa Black FQ
  • IRDye QC-1 IRDye QC-1
  • QSY dyes e.g. QS
  • ATTO quenchers include, but are not limited to, ATTO 540Q, ATTO 580Q, and ATTO 612Q.
  • ATTO quenchers include, but are not limited to, ATTO 540Q, ATTO 580Q, and ATTO 612Q.
  • BHQ® Black Hole Quencher®
  • BHQ-3 BHQ-3
  • cleavage of the labeled detector ssDNA can be detected by measuring the colorimetric readout.
  • liberation of a fluorophore eg, liberation from a FRET pair, liberation from a quencher/fluor pair, etc.
  • liberation of a fluorophore can result in a wavelength shift (and thus color shift) of the detectable signal.
  • cleavage of the detector labeled ssDNA of interest can be detected by color-shift. This shift can be expressed as loss of the amount of one color signal (wavelength), gain of the amount of another color, change in the assignment of one color to another, and the like.
  • the term "vector” is a DNA delivery vehicle essential for recombinant DNA technology used to obtain, propagate (amplify), or express a protein so that a gene can be conveniently used.
  • the vector may be a viral vector or a non-viral vector.
  • the viral vector may be a retroviral (retrovirus) vector, a lentiviral (lentivirus) vector, an adenoviral (adenovirus vector), or an adeno-associated viral vector.
  • AAV vaccinia viral vector
  • poxviral vector vaccinia virus vector
  • herpes simplex viral vector herpes simplex virus vector
  • the non-viral vector may be a plasmid, phage, naked DNA, DNA complex, mRNA (transcript) or PCR amplicon.
  • the plasmid consists of the pcDNA series, pSC101, pGV1106, pACYC177, ColE1, pKT230, pME290, pBR322, pUC8/9, pUC6, pBD9, pHC79, pIJ61, pLAFR1, pHV14, pGEX series, pET series, and pUC19. It may be selected from the group.
  • the phage may be selected from the group consisting of ⁇ gt4 ⁇ B, ⁇ -Charon, ⁇ z1, and M13.
  • the vectors may optionally further contain regulatory/control elements, promoters and/or additional expression elements.
  • the regulation/control element may be operably linked to a sequence encoding each element included in the vector (ie, a nucleic acid encoding a guide RNA and/or a nucleic acid encoding an effector protein).
  • the regulatory/control elements include enhancers, introns, termination signals, polyadenylation signals, Kozak consensus sequences, Internal Ribosome Entry Sites (IRES), splice acceptors, 2A sequences, and/or It may be a replication origin (replication origin), but is not limited thereto.
  • the origin of replication may be the f1 origin of replication, the SV40 origin of replication, the pMB1 origin of replication, the adeno origin of replication, the AAV origin of replication, and/or the BBV origin of replication.
  • the vector may optionally contain a promoter.
  • the promoter may be operably linked to a sequence encoding each component included in the vector (ie, a nucleic acid encoding a guide RNA and/or a nucleic acid encoding an effector protein).
  • the promoter is not limited as long as it can appropriately express a sequence encoding each component included in the vector (ie, a nucleic acid encoding a guide RNA and/or a nucleic acid encoding an effector protein).
  • the promoter sequence may be a promoter that promotes transcription of RNA polymerase (eg, pol I, pol II, or pol III).
  • the promoters include the SV40 early promoter, mouse mammary tumor virus long terminal repeat (LTR) promoter, adenovirus major late promoter (Ad MLP), herpes simplex virus (HSV) promoter, CMV immediate early promoter region (CMVIE), and the like.
  • CMV cytomegalovirus
  • CBA chicken b-actin
  • RSV rous sarcoma virus
  • U6 small nuclear promoter U6 (Miyagishi et al., Nature Biotechnology 20, 497 - 500 (2002)), enhanced It may be one of the U6 promoter (e.g., Xia et al., Nucleic Acids Res. 2003 Sep 1;31(17)), the human H1 promoter (H1) and the 7SK promoter (7SK).
  • the vector may optionally contain additional expression elements.
  • the vector may include a nucleic acid sequence encoding additional expression elements that a person skilled in the art would like to express as needed.
  • the additional expression elements include herbicide resistance genes such as glyphosate, glufosinate ammonium, or phosphinothricin; or an antibiotic resistance gene such as ampicillin, kanamycin, G418, bleomycin, hygromycin or chloramphenicol.
  • the guide polynucleotide; and a Cas protein or a polynucleotide encoding the same and provides a CRISPR/Cas complex capable of cleaving a nucleic acid containing an EGFR mutant gene sequence.
  • CRISPR/Cas system is derived from the bacterial adaptive immune system for bacteriophages, which are particularly used as powerful tools for DNA cleavage. All CRISPR-Cas systems have target specificity through CRISPR RNA (crRNA), which can be slightly modified into guide RNA (gRNA) when used experimentally.
  • crRNA CRISPR RNA
  • gRNA guide RNA
  • a spacer of crRNA recognizes and binds to a target sequence, and at this time, the target sequence must be adjacent to a sequence called Protospacer Adjacent Motif (PAM).
  • PAM Protospacer Adjacent Motif
  • the Cas protein cuts the target DNA. Therefore, the Cas protein can be engineered to operate on a desired target simply by changing the sequence of the spacer portion of the crRNA or gRNA.
  • the Cas protein may correspond to Class 2.
  • the Cas protein corresponding to Class 2 may correspond to type V.
  • the Cas protein corresponding to the type V may be an effector protein selected from the group consisting of Cas12a, mgCas12a, Cas12b, Cas12c, Cas12d, Cas12e, Cas12g, Cas12h, and Cas12i.
  • Another aspect includes contacting a sample with the composition; And detecting a target sequence in the sample by measuring a detectable signal generated by cleavage of the single-stranded detection nucleic acid by the CRISPR / Cas effector protein. .
  • Another aspect provides a method of diagnosing cancer or providing information related to cancer diagnosis, comprising contacting the composition with a sample.
  • the isolated patient sample may include cell free DNA (cfDNA).
  • cfDNA cell free DNA
  • cfDNA is an abbreviation for circulating free DNA, and means a piece of DNA that is not present in the cell nucleus but is suspended in the blood. "Circulating free DNA” or “cell free DNA” and the like can be used interchangeably. Circulating free DNA (cfDNA) is a DNA fragment (50-200 bp) released into plasma. cfDNA can be used to describe the various forms of DNA that circulate freely in the bloodstream, including circulating tumor DNA (ctDNA), cell-free mitochondrial DNA (ccf mtDNA), and cell-free fetal DNA (cffDNA). Elevated levels of cfDNA are observed when cancer is advanced. Through cfDNA analysis, it can be used for cancer diagnosis and follow-up by confirming whether tumor-related mutations are included in the plasma DNA of cancer patients.
  • Another aspect provides a guide polynucleotide comprising a polynucleotide comprising any one of SEQ ID NOs: 2-6 or a polynucleotide having at least 95% homology to a polynucleotide of any one of SEQ ID NOs: 2-6.
  • Mutant DNA derived from a blood sample is specifically amplified through in-vitro digestion and PCR amplification using the CRISPR/Cas system using Cas nuclease according to one aspect, resulting in a 0.5 sequencing error that can be confused with sequencing errors. There is an effect that can identify mutations with a ratio of less than %.
  • 1 is a schematic diagram explaining that exon19 deletion mutant DNA can be distinguished through crRNA matching normal DNA.
  • Figure 2 is a photograph of the results of in vitro cleavage analysis of normal DNA and Exon 19-deficient DNA using crRNA and LbCas12a matching normal DNA.
  • 3 is a graph showing the results of performing CRISPR/Cas12a amplification three times on a DNA mixture in which Exon19 deletion mutant DNA was serially diluted to 1/100,000 of normal DNA.
  • Figure 5 is a schematic diagram of a fluorescence signal-based nucleic acid detection method through the side effect of LbCas12a.
  • 6 is a diagram explaining the mechanism by which crRNA for detecting exon19 deletion mutant DNA causes side effects.
  • FIG. 7 is a graph showing results of detection of exon19 deletion mutant DNA synthesized through fluorescence signals induced from the LbCas12a/crRNA complex.
  • Figure 8 is a schematic diagram of Exon19 deletion patterns validated by clinical lung cancer patient samples.
  • Figure 12 shows two positive samples (P-8) identified as containing a subtype 4 (subtype4) deletion by NGS and two negative samples (N-25, N-26) identified as not containing the mutation. This is the result of measuring the fluorescence signal of
  • E. coli BL21(DE3) cells were transformed with the pET28a-LbCas12a (addgene No. #114070) bacterial expression vector.
  • Cells were cultured in Luria Broth (LB) at 37 °C until the culture reached an O.D of 0.6 and the recombinant protein was 18 with the addition of isopropyl ⁇ -D-thiogalactoside (IPTG, GenDEPOT, Texas, USA) to a final concentration of 1 mM. Induction at °C for 18 hours.
  • the medium was removed by centrifugation (4000 rpm, 30 min) of the cell culture, and the pelleted cells were lysed in lysis buffer (20 mM Tris-HCl (pH 8.0), 300 mM NaCl, 10 mM ⁇ -mercaptoethanol (BioRad, California, U.S.A.). ), 1% Triton X-100 (Sigma) and 1 mM phenylmethylsulfonyl fluoride (Sigma, St. Louis, USA) were lysed by sonication (Qsonica, Newtown, USA).
  • the cell lysate obtained by the sonication was centrifuged at 20,000 x g for 10 minutes to remove cell debris, and the harvested soluble fraction was purified by mixing with Ni-NTA resin (Takara, Nojihigashi, Japan).
  • the mixture was incubated in binding buffer (20 mM Tris-HCl, pH 8.0, 300 nM NaCl) for 1 hour at 4 °C with agitation. After incubation, the Ni-NTA resin was washed with 10 volumes of wash buffer. Next, the protein bound to the Ni-NTA resin was eluted with an elution buffer (20 mM Tris-HCl (pH 8.0), 300 nM NaCl, and 200 mM imidazole).
  • Eluted proteins were filtered through a Centricon filter (Amicon Ultra, Millipore, Burlington, USA). The purity of the recombinant protein was confirmed by SDS-PAGE (10%, Biorad, California, USA) and Coomassie blue staining (Biorad, California, USA).
  • a DNA oligo containing the crRNA corresponding to the target DNA and the T7 promoter sequence was purchased from COSMO Genentech.
  • DNA oligo was prepared in T7 RNA polymerase (NEB, Massachusetts, U.S.A), 50 mM MgCl2, 100 mM NTPs (ATP, GTP, UTP, CTP), 10X RNA polymerase reaction buffer, Murine RNase inhibitor, 100 mM DTT and DEPC at 37°C for 8 hours. mixed while. To completely remove DNA oligos, the mixture was incubated with DNase at 37 °C for 1 hour, and RNA was purified using an RNA purification kit (RBC, New Taipei City, Taipei). Purity and concentration of purified RNA were measured using a NanodropTM 2000 Spectrophotometer (Thermo-Fisher, Massachusetts, USA). The purified RNA was aliquoted and stored at -80 °C.
  • a PCR amplicon with a partial sequence deletion in exon 19 was obtained from HEK293T genomic DNA by overlapping PCR using a DNA primer containing the mutation.
  • the purified recombinant Cas12a and crRNA designed to remove DNA other than mutant DNA were pre-mixed and incubated with the PCR amplicons for 1 hour at 37 °C. Then, the Cas12a/crRNA ribonucleoprotein complex was inactivated at 90 °C for 1 minute.
  • the cleaved mixture was amplified by PCR amplification to uncleaved mutant DNA (denaturation at 98 ° C for 30 seconds). ), primer annealing at 58 °C for 30 sec, extension at 72 °C for 30 sec, 30 cycles).
  • nested PCR denaturation at 98°C for 30 seconds, primer annealing at 58°C for 30 seconds, and extension at 72°C for 30 seconds was used to link the enriched PCR production with the barcode sequence.
  • High throughput sequencing was performed with an Illumina I-seq 100 sequencing machine (Illumina, California, USA).
  • the mutant DNA rate was calculated by analyzing the sequencing raw data through the CRISPR analyzer web tool.
  • cell-free circulating DNA (cfDNA) was extracted from the plasma of lung cancer patients, and the extracted cfDNA was artificially synthesized with purified recombinant LbCas12a protein to specifically amplify mutant DNA.
  • CrRNA was mixed to cleavage normal DNA (nolmal DNA).
  • Uncleaved mutant DNA was amplified from the cleaved mixture by PCR amplification.
  • High throughput sequencing was performed and sequencing raw data was analyzed through the CRISPR analyzer web tool.
  • Fluorescent signals using collateral effects of LbCas12a were confirmed through a fluorophore-quencher reporter assay.
  • the purified LbCas12a recombinant protein (50 nM) and the crRNA corresponding to the mutated DNA sequence (50 nM) were pre-mixed in NEBuffer 2.1 and allowed to stand at room temperature for 5 minutes.
  • the FQ reporter 5 ⁇ /6-FAM/TTATT/BHQ1/3 ⁇
  • a 96 well black plate was prepared. plate), and the fluorescence signal was measured for 1 hour (FAM) at 1 minute intervals.
  • FluoroskanTM Microplate Fluorometer Thermo-Fisher, Massachusetts, U.S.A).
  • Example 1.1 Design of crRNA to specifically amplify EGFR exon19 deletion mutants
  • a crRNA was designed to specifically amplify the EGFR exon19 deletion mutant.
  • exon19 deletion patterns appear in various deletion sizes, such as 9bp to 18bp. Accordingly, as shown in FIG. 1 , in the case of EGFR exon19 deletion mutants, normal DNA and exon19 deletion mutant DNA can be effectively distinguished without additionally introducing incorrect pairing.
  • the composition of crRNA for specifically amplifying EGFR exon19 deletion mutants is shown in Table 1.
  • WT crRNA construct (SEQ ID NO: 1) AA TTTCT ACTAA GTGTA GAT GGAGA TGTTG CTTCT CTTAA TTT AA TTTCT ACTAA GTGTA GAT direct repeat sequence GGAGA TGTTG CTTCT CTTAA target sequence TTT Sequences that increase cleavage efficiency
  • Example 1.2 Confirmation of in vitro cleavage activity of crRNA for specific amplification of EGFR exon19 deletion mutant
  • the normal DNA-specific crRNA significantly cleaved the normal DNA, but the mutant DNA containing the 15bp deletion did not cut at all.
  • Example 2 CRISPR/Cas system designed to specifically amplify EGFR exon19 deletion mutant and confirmation of EGFR exon19 deletion mutant detection sensitivity
  • PCR is performed after the normal DNA is specifically digested in the same manner as in Reference Example 3 using the crRNA of Example 1 and LbCas12a. amplified.
  • mutant DNA at a 1:100,000 dilution was not detected using conventional liquid biopsy.
  • Exon19 deletion mutant DNA could be amplified up to 12.6% through 3 amplification through the CRISPR/Cas system according to an embodiment of the present invention.
  • the above result means that the measurement sensitivity can be secured through the CRISPR/Cas12a system according to one embodiment of the present invention for mutant DNA that cannot be measured using a general liquid biopsy (No amplification, N/A).
  • cfDNA was extracted from the blood of 11 patients with Exon 19 deletion (+) and Exon 19 deletion (-) confirmed by tissue biopsy, and CRISPR/Cas12a amplification was performed. The results are shown in FIG. 4 .
  • CRISPR/Cas12a amplification significantly reduced the deleted mutant DNA to 99.1% in six Exon 19 deletion (+) samples identified through tissue biopsies (P1, P4, P8, P9, P10 and P11). amplification was confirmed. Two of these samples (P4 and P9) showed less than 1% deletion rate before amplification, but were amplified to 91.4% and 93.4%, respectively, with CRISPR/Cas12a amplification.
  • the CRISPR/Cas12a amplification system corresponding to one embodiment can effectively amplify mutant DNA derived from a blood sample and can be used as a useful tool for detecting EGFR mutant DNA.
  • CRISPR/Cas12a amplification system to solve the problem of not being able to distinguish between a fluorescent signal and a background signal induced by a side effect of Cas12a protein when detecting a small amount of target DNA using a side effect of Cas12a protein
  • the fluorescence signal amplification of the EGFR exon19 deletion mutant DNA synthesized using was confirmed.
  • LbCas12a/crRNA recognizes a target DNA
  • non-specific ssDNase which is a side effect of LbCas12a
  • the activated LbCas12a/crRNA ribonucleoprotein non-specifically cuts the single-stranded DNA containing the FQ ssDNA reporter mixed according to Reference Example 5.
  • the amount of target DNA was quantified by the fluorescence signal derived from the cleaved FQ reporter.
  • the fluorescence signal of the EGFR exon19 deletion mutant DNA was detected using crRNA matching the exon19 deletion DNA sequence at 37 °C for 20 minutes.
  • Tables 2 and 3 show the constructs of crRNAs corresponding to various types of exon19 deletion DNA sequences, and among them, the fluorescence signal detection results of EGFR exon19 deletion mutant DNA with the crRNA corresponding to SEQ ID NO: 2 are shown in FIG. .
  • "Blank” indicates that only Cas12a/crRNA ribonucleic acid protein was cultured without DNA.
  • the exon19 deletion mutant DNA synthesized as a result of detecting a fluorescent signal using crRNA matching the exon19 deletion DNA sequence without an amplification procedure using the CRISPR/Cas12a system according to one embodiment of the present invention is converted into normal DNA. It was confirmed that the signal was indistinguishable from the background signal except for the DNA mixture diluted 1/10 in . On the other hand, when CRISPR/Cas12a amplification was used, a clear fluorescence signal was observed from the background signal in all mixtures, including mixtures in which the synthesized exon19 deletion mutant DNA was diluted 1/100,000 with normal DNA.
  • the CRISPR/Cas12a system can effectively increase a low fluorescence signal that is indistinguishable from a background signal.
  • exon 19 deletion mutant subtypes 1 to 4 represent forms in which 18 bp, 15 bp, 16+2 bp, and 9 bp of exon 19 are deleted, respectively.
  • FIGS. 9 to 12 Fluorescence signals were measured at 37 °C for 40 minutes using LbCas12a and crRNA corresponding DNA sequences containing deletion subtypes of deletion mutants, and the results are shown in FIGS. 9 to 12 .
  • P represents a positive sample previously confirmed by tissue biopsy
  • N represents a negative sample previously confirmed by tissue biopsy.
  • Subtype_1 crRNA construction (SEQ ID NO: 3) AA TTTCT ACTAA GTGTA GAT GATTC CTTGA TAGCG ACGGG TTT AA TTTCT ACTAA GTGTA GAT direct repeat sequence GATTC CTTGA TAGCG ACGGG target sequence TTT Sequences that increase cleavage efficiency
  • Subtype_2 crRNA construction (SEQ ID NO: 4) AA TTTCT ACTAA GTGTA GAT GGAGA TGTTT TGATA GCGAC TTT AA TTTCT ACTAA GTGTA GAT direct repeat sequence GGAGA TGTTT TGATA GCGAC target sequence TTT Sequences that increase cleavage efficiency
  • Subtype_3 crRNA construction (SEQ ID NO: 5) AA TTTCT ACTAA GTGTA GAT GGAAT CTTGA TAGCG ACGGG TTT AA TTTCT ACTAA GTGTA GAT direct repeat sequence GGAAT CTTGA TAGCG ACGGG target sequence TTT Sequences that increase clea
  • FIGS. 11 and 12 show two positive samples (P-4) identified as containing a subtype 3 deletion by NGS and two negative samples (N-25, N-26) identified as not containing the mutation.
  • 12 shows two positive samples (P-8) confirmed to contain a subtype 4 (subtype4) deletion by NGS and two negative samples confirmed to not contain a mutation. It is the result of measuring the fluorescence signal of (N-25, N-26).
  • the fluorescence signals of P4 and P8 samples in which subtype 3 and 4 deletion type mutations were positively confirmed were indistinguishable from negative samples (N25 and N26) before amplification with the CRISPR/Cas12a system.
  • the CRISPR/Cas12a amplification system of one embodiment can effectively amplify mutant DNA derived from a blood sample. Therefore, it can be used for diagnosis of various diseases, and in addition to diagnosis, the distinction between normal DNA and mutant DNA can be applied to various fields such as cancer treatment through genome editing specific to pathogenic alleles.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Plant Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Endocrinology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present invention relates to a CRISPR/Cas system capable of specifically amplifying EGFR mutant DNA, and a use thereof for diagnosing cancer. A CRISPR/Cas12a amplification system can be used to specifically and effectively amplify mutant DNA derived from a blood sample and thereby confirm mutations having a ratio of less than 0.5% that can be confused for sequencing errors.

Description

EGFR 돌연변이를 특이적으로 검출할 수 있는 형광 신호 기반 핵산 검출 조성물 및 방법 Fluorescent signal-based nucleic acid detection composition and method capable of specifically detecting EGFR mutation

EGFR 돌연변이를 특이적으로 검출할 수 있는 형광 신호 기반 핵산 검출 조성물 및 방법에 관한 것이다. It relates to a composition and method for detecting a nucleic acid based on a fluorescence signal capable of specifically detecting an EGFR mutation.

다른 암에 비해 사망률이 높은 것으로 알려진 비소폐암(non-small lung cancer, NSCLC) 환자는 효과적인 치료를 위해 조기 진단이 요구된다. 비소폐암(non-small lung cancer, NSCLC)의 표적 암 치료제인 티로신 키나아제 억제제가 EGFR 돌연변이 양성 비소세포폐암에 강력한 치료 효과가 있는 것으로 나타났음을 감안할 때, 비소세포폐암(NSCLC)의 주요 바이오마커인 L858R 및 엑손19 결실의 EGFR 돌연변이를 조기에 안정적으로 검출하는 것이 효과적인 치료를 위해 매우 중요하다.Patients with non-small lung cancer (NSCLC), known to have a higher mortality rate than other cancers, require early diagnosis for effective treatment. Given that a tyrosine kinase inhibitor, a targeted cancer treatment for non-small lung cancer (NSCLC), has been shown to have a strong therapeutic effect on EGFR mutation-positive NSCLC, L858R, a major biomarker for NSCLC, and early and stable detection of EGFR mutations of exon 19 deletion is very important for effective treatment.

최근 종양 조직에서 방출되는 세포유리 순환 DNA(cell-free circulating DNA, cfDNA)의 DNA 돌연변이 검출을 통한 암 진단 가능성이 입증되었는데, 혈액 샘플을 사용하는 액체 생검 기반 진단은 조직 생검보다 훨씬 덜 침습적이고 빠르고 간단한 장점이 있다. Recently, the possibility of cancer diagnosis through the detection of DNA mutations in cell-free circulating DNA (cfDNA) released from tumor tissue has been demonstrated. Liquid biopsy-based diagnosis using blood samples is much less invasive and faster than tissue biopsy. There are simple advantages.

그러나 cfDNA(cell-free circulating DNA)에서 암을 진단하는 데는 몇 가지 장애물이 있다.However, there are several obstacles to diagnosing cancer from cell-free circulating DNA (cfDNA).

첫번째로 혈액 내 소량의 cfDNA는 정확한 진단을 방해하므로 PCR과 같은 추가 절차가 필요하는 것이고, 두번째로 차세대 시퀀싱(Next Generation Sequencing, NGS)은 여전히 0.5%의 오류율을 보여 희귀 돌연변이 DNA와 구별할 수 없다는 것이다. 특히, 혈액 샘플에서 정상 DNA의 우세한 농도는 희귀 돌연변이 DNA를 민감하게 검출하는 데 주요 장애물이다.First, a small amount of cfDNA in blood interferes with accurate diagnosis, requiring additional procedures such as PCR. Second, Next Generation Sequencing (NGS) still has an error rate of 0.5%, making it indistinguishable from rare mutant DNA. will be. In particular, the predominant concentration of normal DNA in blood samples is a major obstacle to sensitive detection of rare mutant DNA.

이러한 문제를 해결하기 위해 CRISPR(Clustered regular interspaced short palindromic repeats) 및 CRISPR 관련 단백질(CRISPR Associated Protein,Cas) 시스템을 사용하여 발암성 돌연변이 진단의 민감도를 향상시키려는 여러 연구가 시도되었다.To address these issues, several studies have attempted to improve the sensitivity of oncogenic mutation diagnosis using CRISPR (Clustered regular interspaced short palindromic repeats) and CRISPR Associated Protein (Cas) systems.

기존의 방법은 PAM 모티프 서열 내에서 점 돌연변이를 찾아 PAM을 파괴하는 전략을 기반으로 하나, 점 돌연변이가 PAM 모티프 서열에 위치해야 하며, 돌연변이 근처에 PAM 서열(NGG)이 없는 EGFR L858R 돌연변이와 같은 일부 돌연변이에 적용하는 데 방해가 된다는 한계를 가지고 있다.Existing methods are based on the strategy of finding point mutations within the PAM motif sequence to disrupt PAM, but point mutations must be located in the PAM motif sequence, and some such as the EGFR L858R mutation without a PAM sequence (NGG) near the mutation. It has limitations that hinder its application to mutations.

이에, 돌연변이를 타겟 DNA에 해당하는 프로토스페이서에 위치시켜 돌연변이 DNA를 고민감도로 검출할 수 있는 CRISPR 시스템에 대한 기술 개발이 필요한 실정이다. Accordingly, it is necessary to develop a technology for a CRISPR system capable of detecting mutant DNA with high sensitivity by locating the mutant in a protospacer corresponding to the target DNA.

일 양상은 표적 서열과 혼성화하고, EGFR(Epidermal growth factor receptor) 돌연변이 유전자 서열에 상보적인 연속적인 염기를 포함하는 가이드 폴리뉴클레오티드 또는 상기 가이드 폴리뉴클레오티드를 암호화하는 핵산; CRISPR/Cas 이펙터 단백질(effector protein), 또는 상기 단백질을 암호화하는 핵산; 및 단일 가닥이고, 상기 가이드 폴리뉴클레오티드와 혼성화 되는 표지된 검출 핵산을 포함하는 EGFR 돌연변이를 검출하기 위한 조성물을 제공하는 것이다. One aspect hybridizes to a target sequence and includes a guide polynucleotide comprising consecutive bases complementary to an epidermal growth factor receptor (EGFR) mutant gene sequence or a nucleic acid encoding the guide polynucleotide; A CRISPR/Cas effector protein, or a nucleic acid encoding the protein; and a single-stranded, EGFR mutation-detecting composition comprising a labeled detection nucleic acid that hybridizes with the guide polynucleotide.

다른 양상은 상기 조성물을 포함하는 EGFR 돌연변이를 검출하기 위한 키트를 제공하는 것이다. Another aspect is to provide a kit for detecting an EGFR mutation comprising the composition.

또 다른 양상은 시료를 상기 조성물과 접촉시키는 단계; 및 상기 CRISPR/Cas 이펙터 단백질에 의한 상기 단일 가닥의 검출 핵산의 절단에 의해 생성된 검출 가능한 신호를 측정하여, 상기 시료 내 표적 서열을 검출하는 단계를 포함하는, EGFR 돌연변이를 검출하는 방법을 제공하는 것이다. Another aspect includes contacting a sample with the composition; And detecting a target sequence in the sample by measuring a detectable signal generated by cleavage of the single-stranded detection nucleic acid by the CRISPR / Cas effector protein. will be.

또 다른 양상은 상기 조성물을 포함하는 암 진단 키트를 제공하는 것이다.Another aspect is to provide a cancer diagnosis kit including the composition.

또 다른 양상은 서열번호 2 내지 6 중 어느 하나를 포함하는 폴리뉴클레오티드 또는 서열번호 2 내지 6 중 어느 하나의 폴리뉴클레오티드와 적어도 95% 상동성을 갖는 폴리뉴클레오티드를 포함하는 가이드 폴리뉴클레오티드를 제공하는 것이다.Another aspect is to provide a guide polynucleotide comprising a polynucleotide comprising any one of SEQ ID NOs: 2-6 or a polynucleotide having at least 95% homology to a polynucleotide of any one of SEQ ID NOs: 2-6.

또 다른 양상은 상기 조성물을 시료와 접촉시키는 단계를 포함하는, 암을 진단하는 방법 또는 암 진단에 관한 정보를 제공하는 방법을 제공하는 것이다.Another aspect is to provide a method of diagnosing cancer or providing information related to cancer diagnosis, including contacting the composition with a sample.

일 양상은 표적 서열과 혼성화하고, EGFR(Epidermal growth factor receptor) 돌연변이 유전자 서열에 상보적인 연속적인 염기를 포함하는 가이드 폴리뉴클레오티드 또는 상기 가이드 폴리뉴클레오티드를 암호화하는 핵산; CRISPR/Cas 이펙터 단백질(effector protein), 또는 상기 단백질을 암호화하는 핵산; 및 단일 가닥이고, 상기 가이드 폴리뉴클레오티드와 혼성화 되는 표지된 검출 핵산을 포함하는 EGFR 돌연변이를 검출하기 위한 조성물을 제공한다. One aspect hybridizes to a target sequence and includes a guide polynucleotide comprising consecutive bases complementary to an epidermal growth factor receptor (EGFR) mutant gene sequence or a nucleic acid encoding the guide polynucleotide; A CRISPR/Cas effector protein, or a nucleic acid encoding the protein; and a single-stranded, labeled detection nucleic acid that hybridizes with the guide polynucleotide.

다른 양상은 상기 조성물을 포함하는 EGFR 돌연변이를 검출하기 위한 키트를 제공하는 것이다. Another aspect is to provide a kit for detecting an EGFR mutation comprising the composition.

본 명세서에서 용어 "프로토스페이서 인접 모티프(protospacer adjacent motif, PAM)"는 CRISPR/Cas 시스템의 Cas 단백질이 인식하는 서열이다. 상기 프로토스페이서 서열은 PAM 서열의 5' 말단 또는 3' 말단에 위치하는 서열로, PAM 이펙터 단백질 복합체가 관심이 있는 표적 유전자좌에 결합하는 것을 지시한다. 구체적으로 상기 PAM은 5' T-풍부 모티프를 포함할 수 있다. 상기 PAM은 2 내지 10 bp DNA 서열로 이루어질 수 있고, 바람직하게는 2내지 6 bp DNA서열로 이루어질 수 있다.In the present specification, the term "protospacer adjacent motif (PAM)" is a sequence recognized by the Cas protein of the CRISPR/Cas system. The protospacer sequence is a sequence located at the 5' or 3' end of the PAM sequence and directs binding of the PAM effector protein complex to the target locus of interest. Specifically, the PAM may include a 5' T-rich motif. The PAM may consist of a 2 to 10 bp DNA sequence, preferably a 2 to 6 bp DNA sequence.

본 명세서에서 용어 "가이드 폴리뉴클레오티드"는 CRISPR 시스템 기반 게놈 편집에 사용되는 짧은 합성 RNA 분자이다. "가이드 폴리뉴클레오티드"는 타겟 DNA에 결합하는 스페이서 시퀀스. gRNA 분자는 Cas의 결합을 위한 스캐폴드 서열, endonuclease로 구성된다. As used herein, the term "guide polynucleotide" is a short synthetic RNA molecule used for genome editing based on the CRISPR system. A "guide polynucleotide" is a spacer sequence that binds to a target DNA. The gRNA molecule consists of a scaffold sequence for Cas binding, an endonuclease.

상기 가이드 뉴클레오티드에는 가이드 RNA, gRNA, 또는 guide RNA를 포함한다. 또한 상기 가이드 폴리뉴클레오티드는 crRNA를 포함한다. 상기 crRNA는 tracrRNA 및/또는 이펙터 단백질과 결합 및/또는 상호작용하는 crRNA 내에 존재하는 일부 서열이다. 상기 crRNA는 야생형 crRNA 또는 엔지니어링된 crRNA 일 수 있다. 이때, 상기 crRNA은 직접반복부서열(direct repeat sequence) 및 스페이서를 포함할 수 있고, 직접반복부서열은 스페이서의 5'말단에 위치할 수 있다. 또한, 상기 crRNA는 tracrRNA의 3' 말단에 위치할 수 있다. 상기 가이드 폴리뉴클레오티드는 tracrRNA을 포함한다. 상기 tracrRNA 스캐폴드 서열은 crRNA 및/또는 이펙터 단백질과 결합 및/또는 상호작용하는 tracrRNA 전체 또는 일부 서열이다. The guide nucleotide includes guide RNA, gRNA, or guide RNA. In addition, the guide polynucleotide includes crRNA. The crRNA is a partial sequence present within the crRNA that binds and/or interacts with tracrRNA and/or effector proteins. The crRNA may be a wild-type crRNA or an engineered crRNA. In this case, the crRNA may include a direct repeat sequence and a spacer, and the direct repeat sequence may be located at the 5' end of the spacer. In addition, the crRNA may be located at the 3' end of tracrRNA. The guide polynucleotide includes tracrRNA. The tracrRNA scaffold sequence is all or part of a tracrRNA sequence that binds and/or interacts with a crRNA and/or an effector protein.

상기 tracrRNA은 야생형 tracrRNA 또는 엔지니어링된 tracrRNA일 수 있다. 상기 엔지니어링된 crRNA 또는 tracrRNA은 상기 야생형 crRNA 또는 tracrRNA의 일부 뉴클레오티드 서열이 인위적으로 변형(치환, 결실 또는 삽입)되거나, 야생형 crRNA 또는 tracrRNA 서열보다 길이가 짧도록 변형된 서열일 수 있다. 상기 가이드 폴리뉴클레오티드는 링커를 더 포함할 수 있다. 상기 링커는 상기 tracrRNA 및 crRNA을 연결하는 역할을 하는 서열이다. 상기 링커는 1 내지 30개의 뉴클레오티드 서열일 수 있다. 일 구현예로서, 상기 linker는 1 내지 5개, 5 내지 10개, 10 내지 15개, 15 내지 20개, 20개 내지 25개 또는 25 내지 30개의 뉴클레오티드 서열일 수 있다. 예를 들어, 상기 링커는 5'-GAAA-3' 서열일 수 있으나, 이에 제한된 것은 아니다. 상기 가이드 폴리뉴클레오티드는 야생형 가이드 폴리뉴클레오티드에서 하나 이상의 뉴클레오티드 서열이 삭제, 치환 또는 추가된 엔지니어링된 가이드 RNA(engineered guide RNA)인 것일 수 있다. The tracrRNA may be a wild-type tracrRNA or an engineered tracrRNA. The engineered crRNA or tracrRNA may be a sequence in which a part of the nucleotide sequence of the wild-type crRNA or tracrRNA is artificially modified (substituted, deleted, or inserted) or modified to be shorter than the wild-type crRNA or tracrRNA sequence. The guide polynucleotide may further include a linker. The linker is a sequence serving to connect the tracrRNA and crRNA. The linker may be a sequence of 1 to 30 nucleotides. In one embodiment, the linker may be 1 to 5, 5 to 10, 10 to 15, 15 to 20, 20 to 25, or 25 to 30 nucleotide sequences. For example, the linker may be a 5'-GAAA-3' sequence, but is not limited thereto. The guide polynucleotide may be an engineered guide RNA in which one or more nucleotide sequences are deleted, substituted, or added from the wild-type guide polynucleotide.

가이드 서열은 표적 서열과 혼성화하고, 표적 서열로의 CRISPR 복합체의 서열-특이적 결합을 지시하기에, 표적 폴리뉴클레오티드 서열과의 충분한 상보성을 갖는 임의의 폴리뉴클레오티드 서열일 수 있다. 상기 gRNA는 Cas 단백질과 복합체를 형성하여 프로토스페이서(protospacer) 인접 모티브("protospacer adjacent motif, PAM") 및 프로토스페이서를 갖는 표적 폴리뉴클레오티드 서열(상기 gRNA의 일부에 상보성인 서열)에 결합한다. 가이드 RNA (gRNA)는 gRNA:Cas 단백질 복합체의 표적 특이성을 부여하는 요소이다. The guide sequence can be any polynucleotide sequence that has sufficient complementarity with the target polynucleotide sequence to hybridize with the target sequence and direct sequence-specific binding of the CRISPR complex to the target sequence. The gRNA forms a complex with the Cas protein to bind to a protospacer adjacent motif (PAM) and a target polynucleotide sequence having a protospacer (sequence complementary to a portion of the gRNA). Guide RNA (gRNA) is the element that confers the target specificity of the gRNA:Cas protein complex.

본 명세서에서 용어 "EGFR"은 Epidermal Growth Factor Receptor의 약어로써 상피세포 성장인자 수용체와 상호 교환적으로 사용할 수 있다. EGFR은 HER 또는 erbB family라고도 불리는 티로신 키나아제 수용체 그룹의 일부로, erbB family에는 EGFR (HER1/ErbB1), HER2 (ErbB2), HER3 (ErbB3) 그리고 HER4 (ErbB4)가 포함된다. In the present specification, the term "EGFR" can be used interchangeably with epidermal growth factor receptor as an abbreviation of Epidermal Growth Factor Receptor. EGFR is part of a group of tyrosine kinase receptors, also called HER or the erbB family, which includes EGFR (HER1/ErbB1), HER2 (ErbB2), HER3 (ErbB3) and HER4 (ErbB4).

일 구체예에 있어서, 상기 EGFR 돌연변이 유전자 서열은 프로토스페이서 인접 모티프(PAM)의 3' 말단에 연결된 것인 가이드 폴리뉴클레오티드일 수 있다. 더욱 구체적으로 상기 프로토스페이서 인접 모티프(PAM)는 5' T-풍부 모티프를 포함하는 것일 수 있다.In one embodiment, the EGFR mutant gene sequence may be a guide polynucleotide linked to the 3' end of a protospacer adjacent motif (PAM). More specifically, the protospacer adjacent motif (PAM) may include a 5' T-rich motif.

본 명세서의 용어 "돌연변이"는 기존 유전자의 염기 순서에 영구적인 변화가 있는 경우를 의미한다. 돌연변이는 유전물질의 구조에 미치는 영향에 따라 염색체 구조나 수에 이상이 있는 경우(염색체 이상) 및 유전자의 염기서열에 작은 규모로 변화가 일어나는 경우가 있다. 염색체의 구조에 이상이 있는 경우로는 결실(deletion), 중복(duplication), 역위(inversion), 전좌(translocation)가 있다. 본 명세서의 용어 "결실 돌연변이 (deletion mutation)"는 염색체의 일부가 손실된 경우를 말한다. 본 명세서의 용어 "점 돌연변이 (point mutation)"는 유전자자리를 구성하는 뉴클레오티드 배열이 1쌍 이상 수천 쌍에 걸쳐 결실되어 일어나는 것을 말한다.As used herein, the term "mutation" refers to a case in which there is a permanent change in the nucleotide sequence of an existing gene. Depending on the effect on the structure of genetic material, mutations can occur when there is an abnormality in the structure or number of chromosomes (chromosomal abnormality) and when a change occurs on a small scale in the nucleotide sequence of a gene. Examples of abnormalities in the structure of chromosomes include deletion, duplication, inversion, and translocation. As used herein, the term "deletion mutation" refers to a case in which a portion of a chromosome is lost. As used herein, the term "point mutation" refers to a nucleotide sequence constituting a locus that occurs when one or more thousands of pairs are deleted.

본 명세서의 용어 "EGFR 돌연변이"는 G719S 돌연변이, G179C 돌연변이, G719A 돌연변이, S720F 돌연변이, T790M 돌연변이, D761Y 돌연변이, exon 19 결실 돌연변이, D770_N771 삽입 돌연변이, V765A 돌연변이, T783A 돌연변이, S761I 돌연변이, T790M 돌연변이, V769L 돌연변이, N771I 돌연변이, L858R 돌연변이, L861Q 돌연변이, L861R 돌연변이를 포함한다. 대표적인 EGFR 변이인 L858R은 858번째 아미노산을 루신(Leusine,L)에서 아르기닌 (Arginine,R)으로 바뀐 점 돌연변이 형태로서, 엑손 21번에 속하는 변이이다. 또한 비소세포폐암의 대표적인 바이오마커로서, EGFR Exon 19 결실 돌연변이는 EGFR을 암호화하는 유전자의 exon 19가 결실(deletion)된 것을 의미한다. EGFR Exon19 결실 돌연변이는 L858R 점 돌연변이와 달리 exon19의 9bp~18bp 등 다양한 결실 크기로 나타난다.The term "EGFR mutation" herein means G719S mutation, G179C mutation, G719A mutation, S720F mutation, T790M mutation, D761Y mutation, exon 19 deletion mutation, D770_N771 insertion mutation, V765A mutation, T783A mutation, S761I mutation, T790M mutation, V769L mutation , N771I mutation, L858R mutation, L861Q mutation, L861R mutation. Representative EGFR mutation L858R is a point mutation in which the 858th amino acid is changed from Leusine (L) to Arginine (Arginine, R), and is a mutation belonging to exon 21. Also, as a representative biomarker of non-small cell lung cancer, EGFR Exon 19 deletion mutant means that exon 19 of the gene encoding EGFR is deleted. Unlike the L858R point mutation, EGFR Exon19 deletion mutants appear in various deletion sizes, such as 9bp to 18bp of exon19.

일 구체예에 있어서, 암은 EGFR 돌연변이를 갖는 것일 수 있다. 상기 암은 고형암일 수 있다. 상기 고형암은 기관에서 유래하는 비정상적인 조직 덩어리를 의미한다. 고형암은 악성일 수 있다. 상이한 유형의 고형암은 이를 형성하는 세포 유형에 따라 명명된다. 고형암의 유형에는 육종, 암종 및 림프종이 포함된다. 고형암의 예로는 부신암, 항문암, 역형성 대세포 림프종, 혈관면역모세포 T 세포 림프종, B 세포 림프종, 담관암, 방광암, 뇌/CNS 종양, 유방암, 자궁경부암, 결장 암, 자궁 내막암, 식도암, 유잉(ewing) 계열의 종양, 안암, 담낭암, 위장 유암종, 위장관 간질 종양(gist), 임신 영양모세포 질환, 간비장 T 세포 림프종, 호지킨 림 프종, 혈관내 거대 B 세포 림프종, 신장 암, 후두암 및 하인두암, 간암, 폐암(비소 세포 및 소세포), 폐 유암종 림프종 육아종증, 악성 중피종, 비강 및 부비동 암, 비인두암, 신경모세포종, 결절 변연부 B 세포 림프종, 비-호지킨 림프종, 구강 및 구인두암, 골육종, 난소암, 췌장암, 음경암, 뇌하수체 종양, 일차 삼출 림프종, 전 립선 암, 망막모세포종, 횡문근육종, 타액선 암, 육종, 피부암(기저 및 편평 세포, 흑색종 및 머켈 세포), 소장암, 위암, 고환암, 흉선암, 갑상선암, 자궁육종, 질암, 외음부 암, 발텐스트롬 마크로글로불린혈증(Waldenstrom macroglobulinemia) 및 빌 름스(Wilms) 종양 등이 있다. EGFR 돌연변이는 특히 결장직장암, 췌장암, 신경교종, 두경부암 및 폐암, 특히 비소세포 폐암(NSCLC)을 일으킬 수 있다. 본 발명의 일 구현예에 따르면, 결장직장암, 췌장암, 신경교종, 두경부암 및 폐암으로 이루어진 그룹으로부터 선택되는 고형암의 진단에, 더욱 더 비소세포 폐암(NSCLC)의 진단에 특히 효과적이다.In one embodiment, the cancer may have an EGFR mutation. The cancer may be a solid cancer. The solid cancer refers to an abnormal tissue mass originating from an organ. Solid cancers can be malignant. Different types of solid cancers are named according to the type of cells that form them. Types of solid cancer include sarcomas, carcinomas and lymphomas. Examples of solid cancers include adrenal cancer, anal cancer, anaplastic large cell lymphoma, angioimmunoblastic T cell lymphoma, B cell lymphoma, cholangiocarcinoma, bladder cancer, brain/CNS tumor, breast cancer, cervical cancer, colon cancer, endometrial cancer, esophageal cancer, Tumors of the Ewing family, eye cancer, gallbladder cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (gist), gestational trophoblast disease, hepatosplenic T-cell lymphoma, Hodgkin's lymphoma, intravascular large B-cell lymphoma, renal cancer, laryngeal cancer and hypopharyngeal cancer, liver cancer, lung cancer (non-small cell and small cell), lung carcinoid lymphomatous granulomatosis, malignant mesothelioma, nasal and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, nodular marginal zone B-cell lymphoma, non-Hodgkin's lymphoma, oral and oropharyngeal cancer, osteosarcoma , ovarian cancer, pancreatic cancer, penile cancer, pituitary tumor, primary effusion lymphoma, prostate cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma, skin cancer (basal and squamous cell, melanoma and Merkel cell), small intestine cancer, gastric cancer, testicular cancer, thymic cancer, thyroid cancer, uterine sarcoma, vaginal cancer, vulvar cancer, Waldenstrom macroglobulinemia, and Wilms tumor. EGFR mutations can cause colorectal cancer, pancreatic cancer, glioma, head and neck cancer, and lung cancer, particularly non-small cell lung cancer (NSCLC), among others. According to one embodiment of the present invention, it is particularly effective for diagnosis of solid cancer selected from the group consisting of colorectal cancer, pancreatic cancer, glioma, head and neck cancer and lung cancer, and even more for diagnosis of non-small cell lung cancer (NSCLC).

일 구체예에 있어서, 상기 가이드 폴리뉴클레오티드는 5' 말단에서 3' 말단 순서로, scafold 서열, 표적서열로 구성되는 20 bp 내지 50bp 길이인 것인, 가이드 폴리뉴클레오티드일 수 있다. 일부 구현예에서, 가이드 서열은 길이가 약 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 75 bp 이상이거나 이를 초과한다. 일부 구현예에서, 가이드 서열은 길이가 약 75, 50, 45, 40, 35, 30, 25, 20, 15, 12 개 이하의 뉴클레오티드 미만이다. 바람직하게 가이드 뉴클레오티드 서열은 길이가 20 bp 내지 50 bp, 더욱 바람직하게는 30 bp 내지 50 bp의 염기를 가진다.In one embodiment, the guide polynucleotide may be a guide polynucleotide having a length of 20 bp to 50 bp consisting of a scafold sequence and a target sequence in order from the 5' end to the 3' end. In some embodiments, the guide sequence is about 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 in length. , 29, 30, 35, 40, 45, 50, 75 bp or more. In some embodiments, the guide sequence is less than about 75, 50, 45, 40, 35, 30, 25, 20, 15, 12 or less nucleotides in length. The guide nucleotide sequence preferably has bases of 20 bp to 50 bp, more preferably 30 bp to 50 bp in length.

일 구체예에 있어서, 상기 가이드 폴리뉴클레오티드는 cleavage 효율을 높이는 서열을 더 포함할 수 있다. 일부 구현예에서, 상기 cleavage 효율을 높이는 서열은 T-풍부 테일링(T-rich tailing) 서열일 수 있다. 일부 구현예에서, 상기 가이드 폴리뉴클레오티드는 서열번호 2 내지 6으로 표시되는 뉴클레오티드 서열을 포함할 수 있다. 서열번호 2 내지 6으로 표시되는 뉴클레오티드에서 T(Thymine)은 U(Uracil)인 것일 수 있다.In one embodiment, the guide polynucleotide may further include a sequence that increases cleavage efficiency. In some embodiments, the sequence increasing the cleavage efficiency may be a T-rich tailing sequence. In some embodiments, the guide polynucleotide may include nucleotide sequences represented by SEQ ID NOs: 2 to 6. In the nucleotides represented by SEQ ID NOs: 2 to 6, T (Thymine) may be U (Uracil).

일 구체예에 있어서, 상기 방법은 측정하는 단계(예를 들어, V형 CRISPR/Cas 효과기 단백질(예를 들어, Cas12 단백질, 예컨대 Cas12a, Cas12b, Cas12c, Cas12d, Cas12e)-매개 ssDNA 절단에 의해 생성되는 검출 가능한 신호를 측정하는 단계)를 포함한다. V형 CRISPR/Cas 효과기 단백질(예를 들어, Cas12 단백질, 예컨대 Cas12a, Cas12b, Cas12c, Cas12d, Cas12e)은, 가이드 RNA가 V형 CRISPR/Cas 효과기 단백질(예를 들어, Cas12 단백질, 예컨대 Cas12a, Cas12b, Cas12c, Cas12d, Cas12e)의 존재 하에 표적 DNA와 혼성화할 때 생기는 일단 활성화된 비표적화 ssDNA를 절단하기 때문에, 검출 가능한 신호는 ssDNA가 절단될 때 생긴 임의의 신호일 수 있다. 예를 들어, 일부 경우에 측정하는 단계는, 금 나노입자 기반 검출, 형광 편광, 콜로이드 상 전이/분산, 전기화학적 검출, 반도체-기반 센싱; 예를 들어, 2'-3' 환식 포스파타제를 개방함으로써, 무기 포스파타제를 용액 내로 방출함으로써 ssDNA 절단 반응 후에 pH 변화를 생성하기 위해 포스파타제를 사용할 수 있음), 및 표지된 검출기 ssDNA의 검출 중 하나 이상을 포함할 수 있다. 이러한 검출 방법의 판독은 임의의 편리한 판독일 수 있다. 가능한 판독의 예는 측정 가능한 양의 검출 가능한 형광 신호; 겔 상에서 밴드의 시각적 분석(예를 들어, 비절단 기질에 대해 절단된 생성물을 나타내는 밴드), 색의 존재 또는 부재의 시각적 또는 센서 기반 검출(즉, 색 검출 방법), 및 전기 신호의 (또는 특정 양의) 존재 또는 부재를 포함하지만, 이들로 제한되지 않는다.In one embodiment, the method comprises the step of measuring (e.g., a V-type CRISPR/Cas effector protein (e.g., a Cas12 protein such as Cas12a, Cas12b, Cas12c, Cas12d, Cas12e)-mediated ssDNA cleavage measuring a detectable signal that becomes). A V-type CRISPR/Cas effector protein (e.g., a Cas12 protein such as Cas12a, Cas12b, Cas12c, Cas12d, Cas12e), wherein the guide RNA is a V-type CRISPR/Cas effector protein (e.g., a Cas12 protein such as Cas12a, Cas12b , Cas12c, Cas12d, Cas12e), the detectable signal can be any signal generated when ssDNA is cleaved. For example, in some cases the measuring step may include gold nanoparticle based detection, fluorescence polarization, colloidal phase transition/dispersion, electrochemical detection, semiconductor-based sensing; For example, phosphatase can be used to generate a pH change after ssDNA cleavage reaction by releasing inorganic phosphatase into solution by opening 2'-3' cyclic phosphatase), and detection of labeled detector ssDNA. can include The readout of this detection method can be any convenient readout. Examples of possible readouts include a measurable amount of detectable fluorescence signal; Visual analysis of bands on the gel (e.g., bands representing cleaved product relative to uncleaved substrates), visual or sensor-based detection of the presence or absence of color (i.e., color detection methods), and electrical signal (or specific positive) presence or absence, but is not limited thereto.

측정하는 단계는 일부 경우에, 예를 들어, 검출되는 신호의 양이 샘플 내 존재하는 표적 DNA의 양을 결정하는 데 사용될 수 있다는 의미에서 정량적일 수 있다. 측정하는 단계는 일부 경우에, 예를 들어, 검출 가능한 신호의 존재 또는 부재가 표적화된 DNA의 존재 또는 부재를 나타낼 수 있다는 의미에서 정성적일 수 있다. 일부 경우에, 달리 표적화된 DNA(들) 가 특정 역치 농도 초과에서 존재하지 않는 한, 검출 가능한 신호는 (예를 들어, 주어진 역치 수준 초과에서) 존재하지 않을 것이다. 일부 경우에, 검출 역치는 CRISPR/Cas 효과기 단백질, 가이드 RNA, 샘플 용적 및/또는 검출기 ssDNA의 양을 (하나가 사용된다면) 변형시킴으로써 적정될 수 있다. 이렇게 해서, 예를 들어, 당업자에 의해 이해될 바와 같이, 한 가지 이상의 반응을 셋업하기 위해 요망된다면, 다수의 대조군이 사용될 수 있으며, 각각은 표적 DNA의 상이한 역치 수준을 검출하도록 셋업하며, 따라서 이러한 일련의 반응은 샘플 내 존재하는 표적 DNA의 양을 결정하는 데 사용될 수 있다.The step of measuring can in some cases be quantitative in the sense that, for example, the amount of signal detected can be used to determine the amount of target DNA present in a sample. The step of measuring can in some cases be qualitative, for example in the sense that the presence or absence of a detectable signal can indicate the presence or absence of targeted DNA. In some cases, no detectable signal will be present (eg, above a given threshold level) unless the otherwise targeted DNA(s) are present above a certain threshold concentration. In some cases, the detection threshold can be titrated by varying the amount of CRISPR/Cas effector protein, guide RNA, sample volume and/or detector ssDNA (if one is used). Thus, for example, if it is desired to set up more than one response, as will be appreciated by those skilled in the art, multiple controls may be used, each set up to detect a different threshold level of target DNA, and thus such A series of reactions can be used to determine the amount of target DNA present in a sample.

일 구체예에 있어서, 본 개시내용의 방법은 표적 핵산(예를 들어, 표적 DNA 및 복수의 비-표적 DNA를 포함하는 샘플)의 양을 결정하는 데 사용될 수 있다. 샘플 내 표적 DNA 양을 결정하는 것은 시험 샘플로부터 생성된 검출 가능한 신호의 양을 기준 샘플로부터 생성된 검출 가능한 신호의 양과 비교하는 것을 포함할 수 있다. 샘플 내 표적 DNA 양을 결정하는 것은 검출 가능한 신호를 측정하여 시험 측정치를 생성하는 단계; 기준 샘플에 의해 생성된 검출 가능한 신호를 측정하여 기준 측정치를 생성하는 단계; 및 시험 측정치를 기준 측정치와 비교하여 샘플 내 존재하는 표적 DNA의 양을 결정하는 단계를 포함할 수 있다.In one embodiment, the methods of the present disclosure can be used to determine the amount of a target nucleic acid (eg, a sample comprising a target DNA and a plurality of non-target DNAs). Determining the amount of target DNA in the sample may include comparing the amount of detectable signal produced from the test sample to the amount of detectable signal produced from a reference sample. Determining the amount of target DNA in a sample includes measuring a detectable signal to generate a test measurement; measuring a detectable signal produced by the reference sample to produce a reference measurement; and comparing the test measurement to a reference measurement to determine the amount of target DNA present in the sample.

예를 들어, 일부 경우에, 샘플 내 표적 DNA의 양을 결정하기 위한 본 개시내용의 방법은: a) 샘플(예를 들어, 표적 DNA 및 복수의 비-표적 DNA를 포함하는 샘플)을: (i) 표적 DNA와 혼성화하는 가이드 RNA, (ii) 샘플 내 존재하는 핵산을 절단하는 CRISPR/Cas 효과기 단백질, 및 (iii) 검출 ssDNA와 접촉시키는 단계; b) CRISPR/Cas 효과기 단백질-매개 ssDNA 절단(예를 들어, 검출기 ssDNA의 절단)에 의해 생성된 검출 가능한 신호를 측정하여, 시험 측정치를 생성하는 단계; c) 기준 샘플에 의해 생성된 검출 가능한 신호를 측정하여 기준 측정치를 생성하는 단계; 및 d) 시험 측정치를 기준 측정치와 비교하여 샘플 내 존재하는 표적 DNA의 양을 결정하는 단계를 포함한다.For example, in some cases, methods of the present disclosure for determining the amount of target DNA in a sample may include: a) a sample (eg, a sample comprising a target DNA and a plurality of non-target DNAs): i) a guide RNA that hybridizes with the target DNA, (ii) a CRISPR/Cas effector protein that cleave nucleic acids present in the sample, and (iii) a detection ssDNA; b) measuring a detectable signal generated by CRISPR/Cas effector protein-mediated ssDNA cleavage (eg, cleavage of the detector ssDNA) to generate a test measurement; c) measuring a detectable signal produced by the reference sample to produce a reference measurement; and d) comparing the test measurement to a reference measurement to determine the amount of target DNA present in the sample.

일부 실시형태에서, 대상 조성물 및/또는 방법의 민감도는 검출을 핵산 증폭과 결합함으로써 증가될 수 있다. 일부 경우에, 샘플 내 핵산은 ssDNA를 절단한 CRISPR/Cas 효과기 단백질(예를 들어, Cas12 단백질)과의 접촉 전, 동시에 또는 후에 증폭된다. 일부 경우에, 샘플 내 핵산은 CRISPR/Cas 효과기 단백질(예를 들어, Cas12 단백질)과의 접촉과 동시에 증폭된다. 모든 성분이 동시에 첨가된다면(증폭 성분 및 검출 성분, 예컨대 CRISPR/Cas 효과기 단백질, 예를 들어, Cas12 단백질, 가이드 RNA, 및 검출기 DNA), CRISPR/Cas 효과기 단백질(예를 들어, Cas12 단백질)의 트랜스-절단 활성은 핵산이 증폭을 겪는 것과 동시에 샘플의 핵산을 분해하기 시작하는 것이 가능하다. In some embodiments, the sensitivity of a subject composition and/or method can be increased by combining detection with nucleic acid amplification. In some cases, nucleic acids in the sample are amplified before, simultaneously with, or after contact with a CRISPR/Cas effector protein (eg, a Cas12 protein) that cuts the ssDNA. In some cases, nucleic acids in the sample are amplified concurrently with contact with a CRISPR/Cas effector protein (eg, Cas12 protein). Transfection of a CRISPR/Cas effector protein (eg, Cas12 protein) if all components are added simultaneously (amplification component and detection component, such as CRISPR/Cas effector protein, eg, Cas12 protein, guide RNA, and detector DNA). -cleavage activity is capable of starting to degrade the nucleic acid of the sample at the same time as the nucleic acid undergoes amplification.

다양한 증폭 방법 및 성분은 당업자에게 공지되어 있고, 임의의 편리한 방법이 사용될 수 있다. 핵산 증폭은 중합효소 연쇄 반응(PCR), 역전사 PCR(RT-PCR), 정량적 PCR(qPCR), 역전사 qPCR(RT-qPCR), 네스티드 PCR(nested PCR), 다중복합 PCR, 비대칭 PCR, 터치다운 PCR, 무작위 프라이머 PCR, 헤미-네스티드 PCR, 중합효소 사이클링 어셈블리(PCA), 콜로니 PCR, 리가제 연쇄 반응(LCR), 디지털 PCR, 메틸화 특이적-PCR(MSP), 낮은 변성 온도에서의 공동-증폭-PCR(COLD-PCR), 대립유전자-특이적 PCR, 서열간-특이적 PCR(ISS-PCR), 전체 게놈 증폭(WGA), 역 PCR, 및 열 비대칭 엇갈림(thermal asymmetric interlaced PCR: TAIL-PCR)을 포함할 수 있다.A variety of amplification methods and components are known to those skilled in the art, and any convenient method may be used. Nucleic acid amplification includes polymerase chain reaction (PCR), reverse transcription PCR (RT-PCR), quantitative PCR (qPCR), reverse transcription qPCR (RT-qPCR), nested PCR, multiplex PCR, asymmetric PCR, touchdown PCR, random primer PCR, hemi-nested PCR, polymerase cycling assembly (PCA), colony PCR, ligase chain reaction (LCR), digital PCR, methylation specific-PCR (MSP), co-at low denaturation temperature Amplification-PCR (COLD-PCR), allele-specific PCR, intersequence-specific PCR (ISS-PCR), whole genome amplification (WGA), inverse PCR, and thermal asymmetric interlaced PCR (TAIL- PCR) may be included.

일부 경우에, 증폭은 등온 증폭이다. 용어 "등온 증폭"은 단일 온도 인큐베이션을 사용하여 열 순환기에 대한 필요를 제거할 수 있는 핵산(예를 들어, DNA) 증폭(예를 들어, 효소 연쇄 반응을 이용) 방법을 나타낸다. 등온 증폭은 증폭 반응 동안 표적 핵산의 열 변성에 의존하지 않으며 따라서 온도의 다중의 빠른 변화를 필요로 하지 않을 수 있는 핵산 증폭 형태이다. 따라서 등온 핵산 증폭 방법은 실험실 환경 내에서 또는 밖에서 수행될 수 있다. 역 전사 단계와 조합함으로써, 이들 증폭 방법은 RNA를 등온으로 증폭시키는 데 사용될 수 있다.In some cases, amplification is isothermal amplification. The term “isothermal amplification” refers to a method of amplifying nucleic acids (eg, DNA) (eg, using an enzymatic chain reaction) that can use a single temperature incubation to eliminate the need for a thermal cycler. Isothermal amplification is a form of nucleic acid amplification that does not rely on thermal denaturation of target nucleic acids during the amplification reaction and thus may not require multiple rapid changes in temperature. Thus, isothermal nucleic acid amplification methods can be performed either within or outside of a laboratory environment. In combination with a reverse transcription step, these amplification methods can be used to amplify RNA isothermally.

등온 증폭 방법의 예는 루프-매개 등온 증폭(LAMP), 헬리카제-의존적 증폭(HDA), 재조합효소 중합효소 증폭(recombinase polymerase amplification: RPA), 가닥 변위(strand displacement amplification: SDA), 핵산 서열-기반 증폭 (NASBA), 전사 매개 증폭(TMA), 틈내기 효소 증폭 반응(NEAR), 구름원 증폭(rolling circle amplification: RCA), 다중 변위 증폭(MDA), 분지(Ramification: RAM), 원형 헬리카제-의존적 증폭(cHDA), 단일 프라이머 등온 증폭(SPIA), RNA 기술의 신호 매개 증폭(SMART), 자기-지속 서열 복제(3SR), 게놈 기하급수적 증폭 반응(GEAR) 및 등온 다중 변위 증폭(IMDA)을 포함하지만, 이들로 제한되지 않는다.Examples of isothermal amplification methods include loop-mediated isothermal amplification (LAMP), helicase-dependent amplification (HDA), recombinase polymerase amplification (RPA), strand displacement amplification (SDA), nucleic acid sequence- based amplification (NASBA), transcription-mediated amplification (TMA), nickase amplification reaction (NEAR), rolling circle amplification (RCA), multiple displacement amplification (MDA), ramification (RAM), circular helicase -Dependent Amplification (cHDA), Single Primer Isothermal Amplification (SPIA), Signal-Mediated Amplification of RNA Technology (SMART), Self-Sustained Sequence Replication (3SR), Genomic Exponential Amplification Reaction (GEAR) and Isothermal Multiple Displacement Amplification (IMDA) Including, but not limited to.

일부 경우에, 증폭은 재조합효소 중합효소 증폭(RPA)이다. 재조합효소 중합효소 증폭(RPA)은 2개의 마주보는 프라이머(PCR와 매우 유사)를 사용하고, 3종의 효소, 즉, 재조합효소, 단일-가닥 DNA-결합 단백질(SSB) 및 가닥-변위 중합효소를 사용한다. 재조합효소는 이중나선 DNA에서 상동성 서열을 갖는 올리고뉴클레오티드 프라이머와 쌍을 이루고, SSB는 변위된 DNA 가닥에 결합하여 프라이머가 변위되는 것을 방지하고, 가닥 변위 중합효소는 DNA 합성을 시작하며, 여기서 프라이머는 표적 DNA에 결합된다. In some cases, the amplification is recombinase polymerase amplification (RPA). Recombinase polymerase amplification (RPA) uses two opposing primers (much like PCR) and uses three enzymes: recombinase, single-stranded DNA-binding protein (SSB) and strand-displacement polymerase. Use Recombinase pairs with oligonucleotide primers having homologous sequences in the double-stranded DNA, SSB binds to the displaced DNA strand to prevent displacement of the primer, and strand displacement polymerase initiates DNA synthesis, where the primer is bound to the target DNA.

일부 경우에, 대상 방법은 샘플(예를 들어, 표적 DNA 및 복수의 비-표적 ssDNA를 포함하는 샘플)을: i) CRISPR/Cas 효과기 단백질; ii) 가이드 RNA(또는 전구체 가이드 RNA 어레이); 및 iii) 단일 가닥이며 가이드 RNA의 가이드 서열과 혼성화하지 않는 검출 DNA와 접촉시키는 단계를 포함한다. 예를 들어, 일부 경우에, 대상 방법은 샘플을 형광-방출 염료쌍을 포함하는 표지된 단일 가닥 검출 핵산(검출 ssDNA)와 접촉시키는 단계를 포함하며; CRISPR/Cas 효과기 단백질(예를 들어, Cas12 단백질, 예컨대 Cas12a, Cas12b, Cas12c, Cas12d, Cas12e)은 (표적 DNA에 혼성화하는 가이드 RNA와 관련하여 가이드 RNA에 결합함으로써) 활성화된 후에 표지된 검출기 ssDNA를 절단하고; 측정되는 검출 가능한 신호는 형광-방출 염료쌍에 의해 생성된다. 예를 들어, 일부 경우에, 대상 방법은 형광 공명 에너지 전달(FRET) 쌍 또는 퀀처/플루오르(퀀처/플루오르) 쌍 또는 둘 다를 포함하는 표지된 검출 ssDNA와 샘플을 접촉시키는 단계를 포함한다. 일부 경우에, 대상 방법은 FRET 쌍을 포함하는 표지된 검출 ssDNA와 샘플을 접촉시키는 단계를 포함한다. 일부 경우에, 대상 방법은 플루오르/퀀처 쌍을 포함하는 표지된 검출 ssDNA와 샘플을 접촉시키는 단계를 포함한다.In some cases, a subject method comprises a sample (eg, a sample comprising a target DNA and a plurality of non-target ssDNAs) comprising: i) a CRISPR/Cas effector protein; ii) guide RNA (or precursor guide RNA array); and iii) a detection DNA that is single-stranded and does not hybridize with the guide sequence of the guide RNA. For example, in some cases, a subject method comprises contacting a sample with a labeled single-stranded detection nucleic acid (detection ssDNA) comprising a fluorescence-emitting dye pair; A CRISPR/Cas effector protein (e.g., a Cas12 protein such as Cas12a, Cas12b, Cas12c, Cas12d, Cas12e) is activated (by binding to a guide RNA in the context of a guide RNA that hybridizes to a target DNA), followed by a labeled detector ssDNA. cut; A detectable signal to be measured is produced by the fluorescence-emitting dye pair. For example, in some cases, the subject methods include contacting the sample with labeled detection ssDNA comprising a fluorescence resonance energy transfer (FRET) pair or a quencher/fluor (quencher/fluor) pair or both. In some cases, a subject method comprises contacting a sample with a labeled detection ssDNA comprising a FRET pair. In some cases, the subject methods include contacting the sample with a labeled detection ssDNA comprising a fluorine/quencher pair.

형광-방출 염료쌍은 FRET 쌍 또는 퀀처/플루오르 쌍을 포함한다. FRET 쌍과 퀀처/플루오르 쌍의 경우 둘 다에서, 염료 중 하나의 방출 스펙트럼은 쌍에서 다른 염료의 흡수 스펙트럼 영역과 중복된다. 본 명세서에서 사용되는 용어 "형광-방출 염료 쌍"은 "형광 공명 에너지 전달(FRET) 쌍"과 "퀀처/플루오르 쌍"을 둘 다 포함하기 위해 사용되는 일반적 용어이며, 이들 용어 둘 다 이하에 더 상세하게 논의한다. 용어 "형광-방출 염료 쌍"은 "FRET 쌍 및/또는 퀀처/플루오르 쌍"이라는 어구와 상호 호환적으로 사용된다.Fluorescence-emitting dye pairs include FRET pairs or quencher/fluor pairs. In both the case of the FRET pair and the quencher/fluorine pair, the emission spectrum of one of the dyes overlaps the region of the absorption spectrum of the other dye in the pair. As used herein, the term "fluorescence-emitting dye pair" is a generic term used to include both a "fluorescence resonance energy transfer (FRET) pair" and a "quencher/fluorine pair", both of which are further hereinafter. discuss in detail The term “fluorescence-emitting dye pair” is used interchangeably with the phrase “FRET pair and/or quencher/fluorine pair”.

일부 경우에 (예를 들어, 검출 ssDNA가 FRET 쌍을 포함할 때) 표지된 검출 ssDNA는 절단되기 전에 검출 가능한 신호의 양을 생성하고, 측정되는 검출 가능한 신호의 양은 표지된 검출 ssDNA가 절단될 때 감소된다. 일부 경우에, 표지된 검출 ssDNA는 (예를 들어, FRET 쌍으로부터) 절단되기 전에 제1 검출 가능한 신호를 생성하고, 표지된 검출 ssDNA가 (예를 들어, 퀀처/플루오르 쌍으로부터) 절단될 때 제2 검출 가능한 신호를 생성한다. 이렇게 해서, 일부 경우에, 표지된 검출 ssDNA는 FRET 쌍 및 퀀처/플루오르 쌍을 포함한다.In some cases (e.g., when the detection ssDNA contains a FRET pair) the labeled detection ssDNA produces an amount of detectable signal before being cleaved, and the amount of detectable signal measured is the amount of the detected signal when the labeled detection ssDNA is cleaved. is reduced In some cases, the labeled detection ssDNA generates a first detectable signal before being cleaved (e.g., from a FRET pair) and the first detectable signal is generated when the labeled detection ssDNA is cleaved (e.g., from a quencher/fluor pair). 2 Generates a detectable signal. Thus, in some cases, the labeled detection ssDNA includes a FRET pair and a quencher/fluor pair.

공여자-수용자 쌍(FRET 공여자 모이어티 및 FRET 수용자 모이어티)은 본 명세서에서 "FRET 쌍" 또는 "신호 FRET 쌍"으로서 지칭된다. 따라서, 일부 경우에, 하나의 신호 상대가 FRET 공여자 모이어티이고 다른 신호 상대가 FRET 수용자 모이어티일 때, 대상 표지된 검출 ssDNA는 2개의 신호 상대(신호 쌍)를 포함한다. 따라서 이러한 FRET 쌍(FRET 공여자 모이어티 및 FRET 수용자 모이어티)를 포함하는 대상 표지된 검출 ssDNA는 신호 상대가 매우 근위일 때(예를 들어, 동일한 RNA 분자 상에 있는 동안) 검출 가능한 신호(FRET 신호)를 나타낼 것이지만, 상대가 분리될 때(예를 들어, CRISPR/Cas 효과기 단백질(예를 들어, Cas12 단백질, 예컨대 Cas12a, Cas12b, Cas12c, Cas12d, Cas12e)의 RNA 분자의 절단 후에) 신호는 감소(또는 부재)될 것이다.A donor-acceptor pair (a FRET donor moiety and a FRET acceptor moiety) is referred to herein as a "FRET pair" or a "signal FRET pair". Thus, in some cases, when one signal partner is a FRET donor moiety and the other signal partner is a FRET acceptor moiety, the target labeled detection ssDNA contains two signal partners (signal pairs). Thus, a target labeled detector ssDNA containing such a FRET pair (a FRET donor moiety and a FRET acceptor moiety) produces a detectable signal (the FRET signal) when the signal counterpart is very proximal (e.g., while on the same RNA molecule). ), but when the counterpart is separated (e.g., after cleavage of an RNA molecule of a CRISPR/Cas effector protein (e.g., a Cas12 protein such as Cas12a, Cas12b, Cas12c, Cas12d, Cas12e)) the signal decreases ( or absent).

FRET 공여자 및 수용자 모이어티(FRET 쌍)는 당업자에게 공지될 것이며, 임의의 편리한 FRET 쌍(예를 들어, 임의의 편리한 공여자 및 수용자 모이어티 쌍)이 사용될 수 있다. FRET donor and acceptor moieties (FRET pairs) will be known to those skilled in the art, and any convenient FRET pair (eg, any convenient donor and acceptor moiety pair) may be used.

일부 경우에, 표지된 검출 ssDNA가 절단될 때 검출 가능한 신호가 생성된다(예를 들어, 일부 경우에, 표지된 검출 ssDNA는 퀀처/플루오르 쌍을 포함한다). 신호 퀀칭 쌍의 하나의 신호 상대는 검출 가능한 신호를 생성하며, 다른 신호 상대는 제1 신호 상대의 검출 가능한 신호를 퀀칭하는 퀀처 모이어티이다(즉, 신호 상대가 서로 근위일 때, 예를 들어, 신호쌍의 신호 상대가 근위일 때 신호 모이어티로부터의 신호가 감소(퀀칭)되도록 퀀처 모이어티는 신호 모이어티의 신호를 퀀칭시킨다).In some cases, a detectable signal is generated when the labeled detector ssDNA is cleaved (eg, in some cases, the labeled detector ssDNA includes a quencher/fluor pair). One signal partner of a signal quenching pair produces a detectable signal, and the other signal partner is a quencher moiety that quenches the detectable signal of the first signal partner (i.e. when the signal partners are proximal to each other, e.g. The quencher moiety quenches the signal of the signal moiety such that the signal from the signal moiety is reduced (quenched) when the signal counterpart of the signal pair is proximal).

예를 들어, 일부 경우에, 검출 가능한 신호의 양은 표지된 검출 ssDNA가 절단될 때 증가된다. 일부 경우에, 예를 들어, CRISPR/Cas 효과기 단백질에 의한 절단 전에 동일한 ssDNA 분자 상에 신호가 둘 다 존재할 때, 예를 들어, 하나의 신호 상대(신호 모이어티)에 의해 나타나는 신호는 다른 신호 상대(퀀처 신호 모이어티)에 의해 퀀칭된다. 이러한 신호 쌍은 본 명세서에서 "퀀처/플루오르 쌍", "퀀칭 쌍" 또는 "신호 퀀칭 쌍"으로서 지칭된다. 예를 들어, 일부 경우에, 하나의 신호 상대(예를 들어, 제1 신호 상대)는 제2 신호 상대(예를 들어, 퀀처 모이어티)에 의해 퀀칭되는 검출 가능한 신호를 생성하는 신호 모이어티이다. 따라서 이러한 퀀처/플루오르쌍의 신호 상대는 상대가 분리될 때(예를 들어, V형 CRISPR/Cas 효과기 단백질에 의한 검출 ssDNA의 절단 후) 검출 가능한 신호를 생성할 것이지만, 상대가 매우 근위일 때(예를 들어, CRISPR/Cas 효과기 단백질에 의한 검출 ssDNA의 절단 전에) 신호는 퀀칭될 것이다.For example, in some cases, the amount of detectable signal is increased when the labeled detection ssDNA is cleaved. In some cases, e.g., when both signals are present on the same ssDNA molecule prior to cleavage by the CRISPR/Cas effector protein, e.g., the signal exhibited by one signal partner (signal moiety) is the signal exhibited by the other signal partner. (quencher signal moiety). Such signal pairs are referred to herein as "quencher/fluor pairs", "quenching pairs" or "signal quenching pairs". For example, in some cases, one signal counterpart (e.g., a first signal counterpart) is a signal moiety that produces a detectable signal that is quenched by a second signal counterpart (e.g., a quencher moiety) . Thus, the signal partners of this quencher/fluor pair will produce a detectable signal when the partners are separated (e.g., after cleavage of the detector ssDNA by the V-type CRISPR/Cas effector protein), but when the partners are very proximal ( eg, before cleavage of the detection ssDNA by the CRISPR/Cas effector protein) the signal will be quenched.

퀀처 모이어티는 신호 모이어티로부터(예를 들어, CRISPR/Cas 효과기 단백질에 의한 검출 ssDNA의 절단 전에) 다양한 정도로 신호를 퀀칭시킬 수 있다. 일부 경우에, 퀀처 모이어티는 퀀처 모이어티의 존재 하에(신호 상대가 서로 근위일 때) 검출된 신호가 퀀처 모이어티의 부재 하에(신호 상대가 분리될 때) 검출된 신호의 95% 이하인 경우 신호 모이어티로부터 신호를 퀀칭시킨다. 예를 들어, 일부 경우에, 퀀처 모이어티의 존재에서 검출된 신호는 퀀처 모이어티의 부재 하에 검출된 신호의 90% 이하, 80% 이하, 70% 이하, 60% 이하, 50% 이하, 40% 이하, 30% 이하, 20% 이하, 15% 이하, 10% 이하, 또는 5% 이하일 수 있다. 일부 경우에, 신호 없음(예를 들어, 배경 초과)은 퀀처 모이어티의 존재 하에 검출된다.The quencher moiety can quench the signal to varying degrees from the signal moiety (eg, prior to cleavage of the detection ssDNA by the CRISPR/Cas effector protein). In some cases, a quencher moiety is a signal if the signal detected in the presence of the quencher moiety (when the signal counterparts are proximal to each other) is no more than 95% of the signal detected in the absence of the quencher moiety (when the signal counterparts are separated). Quench the signal from the moiety. For example, in some cases, the signal detected in the presence of the quencher moiety is 90% or less, 80% or less, 70% or less, 60% or less, 50% or less, 40% or less of the signal detected in the absence of the quencher moiety. or less, 30% or less, 20% or less, 15% or less, 10% or less, or 5% or less. In some cases, no signal (eg, above background) is detected in the presence of the quencher moiety.

일부 경우에, 퀀처 모이어티의 부재 하에(신호 상대가 분리될 때) 검출된 신호는 퀀처 모이어티의 존재 하에(신호 상대가 서로 근위일 때) 검출된 신호보다 적어도 1.2배 초과(예를 들어, 적어도 1.3배, 적어도 1.5 배, 적어도 1.7 배, 적어도 2 배, 적어도 2.5 배, 적어도 3 배, 적어도 3.5 배, 적어도 4 배, 적어도 5 배, 적어도 7 배, 적어도 10 배, 적어도 20배, 또는 적어도 50배 초과)이다.In some cases, the signal detected in the absence of the quencher moiety (when the signal partners are separated) is at least 1.2-fold greater than the signal detected in the presence of the quencher moiety (when the signal partners are proximal to each other) (e.g., at least 1.3x, at least 1.5x, at least 1.7x, at least 2x, at least 2.5x, at least 3x, at least 3.5x, at least 4x, at least 5x, at least 7x, at least 10x, at least 20x, or at least 50 times higher).

일부 경우에, 신호 모이어티는 형광 표지이다. 일부 이러한 경우에, 퀀처 모이어티는 (예를 들어, 표지의 방출 스펙트럼에서 에너지를 흡수함으로써) 형광 표지로부터의 신호(광 신호)를 퀀칭시킨다. 따라서, 퀀처 모이어티가 신호 모이어티에 근접하지 않을 때, 형광 표지로부터의 방출(신호)는 퀀처 모이어티에 의해 신호가 흡수되지 않기 때문에 검출 가능하다. 임의의 편리한 공여자 수용자 쌍(신호 모이어티/퀀처 모이어티 쌍)이 사용될 수 있고, 다수의 적합한 쌍은 당업계에 공지되어 있다.In some cases, the signal moiety is a fluorescent label. In some such cases, the quencher moiety quenches the signal from the fluorescent label (light signal) (eg, by absorbing energy in the label's emission spectrum). Thus, when the quencher moiety is not in proximity to the signal moiety, emission (signal) from the fluorescent label is detectable because no signal is absorbed by the quencher moiety. Any convenient donor acceptor pair (signal moiety/quencher moiety pair) may be used, and many suitable pairs are known in the art.

일부 경우에, 퀀처 모이어티는 신호 모이어티(또한 본 명세서에서 "검출 가능한 표지"로서 지칭됨)로부터 에너지를 흡수하고, 이어서, 신호(예를 들어, 상이한 파장에서의 광)를 방출한다. 따라서, 일부 경우에, 퀀처 모이어티는 신호 모이어티 그 자체이고(예를 들어, 신호 모이어티는 6-카복시플루오레세인일 수 있는 한편, 퀀처 모이어티는 6-카복시-테트라메틸로다민일 수 있음), 일부 경우에, 쌍은 또한 FRET 쌍일 수 있다. 일부 경우에, 퀀처 모이어티는 다크 퀀처이다. 다크 퀀처는 여기 에너지를 흡수하며, 상이한 방법으로 (예를 들어, 열로서) 에너지를 소멸시킨다. 따라서, 다크 퀀처는 그 자체의 형광이 최소이거나 없다(형광을 방출하지 않는다). In some cases, the quencher moiety absorbs energy from the signal moiety (also referred to herein as a "detectable label") and then emits a signal (eg, light at a different wavelength). Thus, in some cases, the quencher moiety is the signal moiety itself (e.g., the signal moiety can be 6-carboxyfluorescein, while the quencher moiety can be 6-carboxy-tetramethylrhodamine ), in some cases the pair may also be a FRET pair. In some cases, the quencher moiety is a dark quencher. The dark quencher absorbs the excitation energy and dissipates the energy in a different way (eg as heat). Thus, the dark quencher has minimal or no fluorescence of its own (it emits no fluorescence).

형광 표지의 예는 알렉사 플루오르(Alexa Fluor)(등록상표) 염료, ATTO 염료(예를 들어, ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532, ATTO Rho6G, ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725, ATTO 740), a DyLight 염료, 사이아닌 염료(예를 들어, Cy2, Cy3, Cy3.5, Cy3b, Cy5, Cy5.5, Cy7, Cy7.5), 플루오프로브(FluoProbes) 염료, 설포 Cy 염료, 세타(Seta) 염료, IRIS 염료, SeTau 염료, SRfluor 염료, 스퀘어(Square) 염료, 플루오레세인 아이소티오사이아네이트(FITC), 테트라메틸로다민(TRITC), 텍사스 레드(Texas Red), 오리건 그린(Oregon Green), 퍼시픽 블루(Pacific Blue), 퍼시픽 그린(Pacific Green), 퍼시픽 오렌지(Pacific Orange), 양자점 및 테더드(tethered) 형광 단백질을 포함하지만, 이들로 제한되지 않는다.Examples of fluorescent labels include Alexa Fluor® dye, ATTO dye (e.g., ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532, ATTO Rho6G , ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725, ATTO 740), a DyLight dye, a cyanine dye (eg, Cy2, Cy3, Cy3.5, Cy3b, Cy5, Cy5. 5, Cy7, Cy7.5), FluoProbes dye, Sulfo Cy dye, Seta dye, IRIS dye, SeTau dye, SRfluor dye, Square dye, Fluorescein isothiocyanate ( FITC), tetramethylrhodamine (TRITC), Texas Red, Oregon Green, Pacific Blue, Pacific Green, Pacific Orange, Quantum Dots and Tethered (tethered) fluorescent proteins, but are not limited thereto.

일부 경우에, 검출 가능한 표지는 알렉사 플루오르(Alexa Fluor)(등록상표) 염료, ATTO 염료(예를 들어, ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532, ATTO Rho6G, ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725, ATTO 740), a DyLight 염료, 사이아닌 염료(예를 들어, Cy2, Cy3, Cy3.5, Cy3b, Cy5, Cy5.5, Cy7, Cy7.5), 플루오프로브 염료, 설포 Cy 염료, 세타 염료, IRIS 염료, SeTau 염료, SRfluor 염료, 스퀘어 염료, 플루오레세인(FITC), 테트라메틸로다민(TRITC), 텍사스 레드, 오리건 그린, 퍼시픽 블루, 퍼시픽 그린 및 퍼시픽 오렌지로부터 선택된 형광 표지이다.In some cases, the detectable label is an Alexa Fluor® dye, an ATTO dye (e.g., ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532 , ATTO Rho6G, ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725, ATTO 740), a DyLight dye, a cyanine dye (e.g. Cy2, Cy3, Cy3.5, Cy3b, Cy5 , Cy5.5, Cy7, Cy7.5), fluoroprobe dye, sulfo Cy dye, theta dye, IRIS dye, SeTau dye, SRfluor dye, square dye, fluorescein (FITC), tetramethylrhodamine (TRITC), and a fluorescent label selected from Texas Red, Oregon Green, Pacific Blue, Pacific Green and Pacific Orange.

일부 경우에, 검출 가능한 표지는 알렉사 플루오르(Alexa Fluor)(등록상표) 염료, ATTO 염료(예를 들어, ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532, ATTO Rho6G, ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725, ATTO 740), a DyLight 염료, 사이아닌 염료(예를 들어, Cy2, Cy3, Cy3.5, Cy3b, Cy5, Cy5.5, Cy7, Cy7.5), 플루오프로브 염료, 설포 Cy 염료, 세타 염료, IRIS 염료, SeTau 염료, SRfluor 염료, 스퀘어 염료, 플루오레세인(FITC), 테트라메틸로다민(TRITC), 텍사스 레드, 오리건 그린, 퍼시픽 블루, 퍼시픽 그린, 퍼시픽 오렌지, 양자점 및 테더드 형광 단백질로부터 선택된 형광 표지이다.In some cases, the detectable label is an Alexa Fluor® dye, an ATTO dye (e.g., ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532 , ATTO Rho6G, ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725, ATTO 740), a DyLight dye, a cyanine dye (e.g. Cy2, Cy3, Cy3.5, Cy3b, Cy5 , Cy5.5, Cy7, Cy7.5), fluoroprobe dye, sulfo Cy dye, theta dye, IRIS dye, SeTau dye, SRfluor dye, square dye, fluorescein (FITC), tetramethylrhodamine (TRITC), and a fluorescent label selected from Texas Red, Oregon Green, Pacific Blue, Pacific Green, Pacific Orange, quantum dots and tethered fluorescent proteins.

ATTO 염료의 예는 하기를 포함하지만, 이들로 제한되지 않는다: ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532, ATTO Rho6G, ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725 및 ATTO 740.Examples of ATTO dyes include, but are not limited to: ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 514, ATTO 520, ATTO 532, ATTO Rho6G, ATTO 542, ATTO 550, ATTO 565, ATTO Rho3B, ATTO Rho11, ATTO Rho12, ATTO Thio12, ATTO Rho101, ATTO 590, ATTO 594, ATTO Rho13, ATTO 610, ATTO 620, ATTO Rho14, ATTO 633, ATTO 647, ATTO 647N, ATTO 655, ATTO Oxa12, ATTO 665, ATTO 680, ATTO 700, ATTO 725 and ATTO 740.

알렉사플루오르 염료의 예는 하기를 포함하지만, 이들로 제한되지 않는다: 알렉사 플루오르(등록상표) 350, 알렉사 플루오르(등록상표) 405, 알렉사 플루오르(등록상표) 430, 알렉사 플루오르(등록상표) 488, 알렉사 플루오르(등록상표) 500, 알렉사 플루오르(등록상표) 514, 알렉사 플루오르(등록상표) 532, 알렉사 플루오르(등록상표) 546, 알렉사 플루오르(등록상표) 555, 알렉사 플루오르(등록상표) 568, 알렉사 플루오르(등록상표) 594, 알렉사 플루오르(등록상표) 610, 알렉사 플루오르(등록상표) 633, 알렉사 플루오르(등록상표) 635, 알렉사 플루오르(등록상표) 647, 알렉사 플루오르(등록상표) 660, 알렉사 플루오르(등록상표) 680, 알렉사 플루오르(등록상표) 700, 알렉사 플루오르(등록상표) 750, 알렉사 플루오르(등록상표) 790 등.Examples of Alexa Fluor dyes include, but are not limited to: Alexa Fluor® 350, Alexa Fluor® 405, Alexa Fluor® 430, Alexa Fluor® 488, Alexa Fluor® 500, Alexa Fluor® 514, Alexa Fluor® 532, Alexa Fluor® 546, Alexa Fluor® 555, Alexa Fluor® 568, Alexa Fluor ( Alexa Fluor (registered trademark) 610, Alexa Fluor (registered trademark) 633, Alexa Fluor (registered trademark) 635, Alexa Fluor (registered trademark) 647, Alexa Fluor (registered trademark) 660, Alexa Fluor (registered trademark) ) 680, Alexa Fluor (registered trademark) 700, Alexa Fluor (registered trademark) 750, Alexa Fluor (registered trademark) 790, etc.

퀀처 모이어티의 예는 다크 퀀처(dark quencher), 블랙 홀 퀀처(Black Hole Quencher)(등록상표) (BHQ(등록상표)) (예를 들어, BHQ-0, BHQ-1, BHQ-2, BHQ-3), Qxl 퀀처, ATTO 퀀처(예를 들어, ATTO 540Q, ATTO 580Q, 및 ATTO 612Q), 다이메틸아미노아조벤젠설폰산(답실(Dabsyl)), 아이오와 블랙(Iowa Black) RQ, 아이오와 블랙 FQ, IRDye QC-1, QSY 염료(예를 들어, QSY 7, QSY 9, QSY 21), 앱솔루트퀀처(AbsoluteQuencher), 이클립스(Eclipse) 및 금속 클러스터, 예컨대, 금 나노입자 등을 포함하지만, 이들로 제한되지 않는다.Examples of quencher moieties include dark quencher, black hole quencher (registered trademark) (BHQ®) (e.g., BHQ-0, BHQ-1, BHQ-2, BHQ -3), Qxl Quencher, ATTO Quencher (eg ATTO 540Q, ATTO 580Q, and ATTO 612Q), Dimethylaminoazobenzenesulfonic Acid (Dabsyl), Iowa Black RQ, Iowa Black FQ, IRDye QC-1, QSY dyes (e.g., QSY 7, QSY 9, QSY 21), AbsoluteQuencher, Eclipse, and metal clusters such as gold nanoparticles, etc. don't

일부 경우에, 퀀처 모이어티는 다크 퀀처, 블랙 홀 퀀처(등록상표) (BHQ(등록상표)) (예를 들어, BHQ-0, BHQ-1, BHQ-2, BHQ-3), Qxl 퀀처, ATTO 퀀처(예를 들어, ATTO 540Q, ATTO 580Q 및 ATTO 612Q), 다이메틸아미노아조벤젠설폰산(답실), 아이오와 블랙 RQ, 아이오와 블랙 FQ, IRDye QC-1, QSY 염료(예를 들어, QSY 7, QSY 9, QSY 21), 앱솔루트퀀처, 이클립스 및 금속 클러스터로부터 선택된다.In some cases, the quencher moiety is a dark quencher, black hole quencher® (BHQ®) (e.g., BHQ-0, BHQ-1, BHQ-2, BHQ-3), Qxl quencher, ATTO quencher (e.g. ATTO 540Q, ATTO 580Q and ATTO 612Q), dimethylaminoazobenzenesulfonic acid (dansyl), Iowa Black RQ, Iowa Black FQ, IRDye QC-1, QSY dyes (e.g. QSY 7, QSY 9, QSY 21), Absolute Quencher, Eclipse and Metal Cluster.

ATTO 퀀처의 예는 ATTO 540Q, ATTO 580Q 및 ATTO 612Q를 포함하지만, 이들로 제한되지 않는다. 블랙홀 퀀처(등록상표)(BHQ(등록상표))의 예는 BHQ-0(493㎚), BHQ-1(534㎚), BHQ-2(579㎚) 및 BHQ-3(672㎚)를 포함하지만, 이들로 제한되지 않는다.Examples of ATTO quenchers include, but are not limited to, ATTO 540Q, ATTO 580Q, and ATTO 612Q. Examples of Black Hole Quencher® (BHQ®) include BHQ-0 (493 nm), BHQ-1 (534 nm), BHQ-2 (579 nm) and BHQ-3 (672 nm) , but not limited to these.

일부 경우에, 표지된 검출 ssDNA의 절단은 비색 판독을 측정함으로써 검출될 수 있다. 예를 들어, 형광단의 유리(예를 들어, FRET 쌍으로부터의 유리, 퀀처/플루오르 쌍으로부터의 유리 등)는 검출 가능한 신호의 파장 이동(따라서 색 이동)을 초래할 수 있다. 따라서, 일부 경우에, 대상 표지된 검출 ssDNA의 절단은 색-이동에 의해 검출될 수 있다. 이러한 이동은 한 가지 색 신호(파장)양의 상실, 다른 색의 양의 획득, 한 가지 색의 다른 색으로의 할당 변화 등으로서 표현될 수 있다.In some cases, cleavage of the labeled detector ssDNA can be detected by measuring the colorimetric readout. For example, liberation of a fluorophore (eg, liberation from a FRET pair, liberation from a quencher/fluor pair, etc.) can result in a wavelength shift (and thus color shift) of the detectable signal. Thus, in some cases, cleavage of the detector labeled ssDNA of interest can be detected by color-shift. This shift can be expressed as loss of the amount of one color signal (wavelength), gain of the amount of another color, change in the assignment of one color to another, and the like.

본 명세서의 용어 "벡터"는 유전자를 편리하게 활용할 수 있도록 확보, 증식(증폭)하거나, 단백질을 발현시키는 데 이용하는 유전자 재조합 기술(recombinant DNA technology)에 필수적으로 필요한 DNA 전달체이다. 상기 벡터는 바이러스 벡터 또는 비-바이러스 벡터일 수 있다. 예를 들어, 상기 바이러스 벡터는 레트로바이러스 벡터(retroviral(retrovirus) vector), 렌티바이러스 벡터(lentiviral(lentivirus) vector), 아데노바이러스 벡터(adenoviral(adenovirus vector), 아데노-연관 바이러스 벡터(adeno-associated viral (adeno-associated virus; AAV) vector), 백시니아바이러스 벡터(vaccinia viral(vaccinia virus) vector), 폭스바이러스 벡터(poxviral(poxvirus) vector) 및 단순포진 바이러스 벡터(herpes simplex viral(herpes simplex virus) vector)로 구성된 군에서 선택되는 하나 이상의 바이러스 벡터일 수 있다.As used herein, the term "vector" is a DNA delivery vehicle essential for recombinant DNA technology used to obtain, propagate (amplify), or express a protein so that a gene can be conveniently used. The vector may be a viral vector or a non-viral vector. For example, the viral vector may be a retroviral (retrovirus) vector, a lentiviral (lentivirus) vector, an adenoviral (adenovirus vector), or an adeno-associated viral vector. (adeno-associated virus; AAV) vector), vaccinia viral (vaccinia virus) vector, poxviral (poxvirus) vector and herpes simplex viral (herpes simplex virus) vector ) may be one or more viral vectors selected from the group consisting of

상기 비-바이러스 벡터는 플라스미드, 파지, 네이키드 DNA, DNA 복합체, mRNA(전사물) 또는 PCR 앰플리콘(amplicon)일 수 있다. 예를 들어, 상기 플라스미드는 pcDNA 시리즈, pSC101, pGV1106, pACYC177, ColE1, pKT230, pME290, pBR322, pUC8/9, pUC6, pBD9, pHC79, pIJ61, pLAFR1, pHV14, pGEX 시리즈, pET 시리즈, 및 pUC19으로 이뤄진 군에서 선택된 것일 수 있다. 예를 들어, 상기 파지는 λgt4λB, λ-Charon, λ△z1, 및 M13으로 이뤄진 군에서 선택된 것일 수 있다.The non-viral vector may be a plasmid, phage, naked DNA, DNA complex, mRNA (transcript) or PCR amplicon. For example, the plasmid consists of the pcDNA series, pSC101, pGV1106, pACYC177, ColE1, pKT230, pME290, pBR322, pUC8/9, pUC6, pBD9, pHC79, pIJ61, pLAFR1, pHV14, pGEX series, pET series, and pUC19. It may be selected from the group. For example, the phage may be selected from the group consisting of λgt4λB, λ-Charon, λΔz1, and M13.

상기 벡터는 선택적으로 조절/제어 구성요소, 프로모터 및/또는 부가 발현 요소를 추가로 포함할 수 있다. 상기 조절/제어 구성요소는 벡터에 포함된 각 구성요소를 암호화하는 서열(즉, 가이드 RNA를 암호화하는 핵산 및/또는 이펙터 단백질을 암호화하는 핵산)에 작동 가능하게 연결될 수 있다. 상기 조절/제어 구성요소는 인핸서, 인트론, 종결 신호, 폴리아데닐화 신호, 코작 공통(Kozak consensus) 서열, 내부 리보솜 유입 부위(IRES, Internal Ribosome Entry Site), 스플라이스 억셉터, 2A 서열 및/또는 복제원점(replication origin)일 수 있으나, 이에 제한되는 것은 아니다. 상기 복제원점은 f1 복제원점, SV40 복제원점, pMB1 복제원점, 아데노 복제원점, AAV 복제원점, 및/또는 BBV 복제원점일 수 있다. 상기 벡터는 선택적으로 프로모터를 포함할 수 있다. 상기 프로모터는 벡터에 포함된 각 구성요소를 암호화하는 서열(즉, 가이드 RNA를 암호화하는 핵산 및/또는 이펙터 단백질을 암호화하는 핵산)에 작동 가능하게 연결될 수 있다. 상기 프로모터는 벡터에 포함된 각 구성요소를 암호화하는 서열(즉, 가이드 RNA를 암호화하는 핵산 및/또는 이펙터 단백질을 암호화하는 핵산)을 적절히 발현시킬 수 있는 것이라면 제한되지 않는다. 일 구현예로서, 상기 프로모터 서열은 RNA 중합효소(예를 들어, pol I, pol II, 또는 pol III)의 전사를 촉진시키는 프로모터일 수 있다. 예를 들어, 상기 프로모터는 SV40 초기 프로모터, mouse mammary tumor virus long terminal repeat(LTR) 프로모터, adenovirus major late 프로모터 (Ad MLP), herpes simplex virus (HSV) 프로모터, CMV immediate early promoter region (CMVIE)와 같은 cytomegalovirus (CMV) 프로모터, Chicken b-actin (CBA) 프로모터, rous sarcoma virus (RSV) 프로모터, human U6 small nuclear 프로모터 (U6) (Miyagishi et al., Nature Biotechnology 20, 497 - 500 (2002)), enhanced U6 프로모터 (e.g., Xia et al., Nucleic Acids Res. 2003 Sep 1;31(17)), human H1 프로모터 (H1) 및 7SK 프로모터 (7SK) 중 하나 수 있다. The vectors may optionally further contain regulatory/control elements, promoters and/or additional expression elements. The regulation/control element may be operably linked to a sequence encoding each element included in the vector (ie, a nucleic acid encoding a guide RNA and/or a nucleic acid encoding an effector protein). The regulatory/control elements include enhancers, introns, termination signals, polyadenylation signals, Kozak consensus sequences, Internal Ribosome Entry Sites (IRES), splice acceptors, 2A sequences, and/or It may be a replication origin (replication origin), but is not limited thereto. The origin of replication may be the f1 origin of replication, the SV40 origin of replication, the pMB1 origin of replication, the adeno origin of replication, the AAV origin of replication, and/or the BBV origin of replication. The vector may optionally contain a promoter. The promoter may be operably linked to a sequence encoding each component included in the vector (ie, a nucleic acid encoding a guide RNA and/or a nucleic acid encoding an effector protein). The promoter is not limited as long as it can appropriately express a sequence encoding each component included in the vector (ie, a nucleic acid encoding a guide RNA and/or a nucleic acid encoding an effector protein). In one embodiment, the promoter sequence may be a promoter that promotes transcription of RNA polymerase (eg, pol I, pol II, or pol III). For example, the promoters include the SV40 early promoter, mouse mammary tumor virus long terminal repeat (LTR) promoter, adenovirus major late promoter (Ad MLP), herpes simplex virus (HSV) promoter, CMV immediate early promoter region (CMVIE), and the like. cytomegalovirus (CMV) promoter, chicken b-actin (CBA) promoter, rous sarcoma virus (RSV) promoter, human U6 small nuclear promoter (U6) (Miyagishi et al., Nature Biotechnology 20, 497 - 500 (2002)), enhanced It may be one of the U6 promoter (e.g., Xia et al., Nucleic Acids Res. 2003 Sep 1;31(17)), the human H1 promoter (H1) and the 7SK promoter (7SK).

상기 벡터는 선택적으로 부가 발현 요소를 포함할 수 있다. 상기 벡터는 가이드 RNA를 암호화하는 핵산 외에 통상의 기술자가 필요에 의해 발현시키고자 하는 부가 발현 요소를 암호화하는 핵산 서열을 포함하고 있을 수 있다. 예를 들어, 상기 부가 발현 요소는, 글리포세이트(glyphosate), 글루포시네이트암모늄 (glufosinate ammonium) 또는 포스피노트리신(phosphinothricin)과 같은 제초제 저항성 유전자; 또는 암피실린(ampicillin), 카나마이신(kanamycin), G418, 블레오마이신 (Bleomycin), 하이그로마이신(hygromycin) 또는 클로람페니콜(chloramphenicol)과 같은 항생제 내성 유전자일 수 있다.The vector may optionally contain additional expression elements. In addition to the nucleic acid encoding the guide RNA, the vector may include a nucleic acid sequence encoding additional expression elements that a person skilled in the art would like to express as needed. For example, the additional expression elements include herbicide resistance genes such as glyphosate, glufosinate ammonium, or phosphinothricin; or an antibiotic resistance gene such as ampicillin, kanamycin, G418, bleomycin, hygromycin or chloramphenicol.

일 양상은 상기 가이드 폴리뉴클레오티드; 및 Cas 단백질 또는 이를 암호화하는 폴리뉴클레오티드를 포함하고, EGFR 돌연변이 유전자 서열을 포함하는 핵산을 절단(cleavage)할 수 있는 CRISPR/Cas 복합체를 제공한다.In one aspect, the guide polynucleotide; and a Cas protein or a polynucleotide encoding the same, and provides a CRISPR/Cas complex capable of cleaving a nucleic acid containing an EGFR mutant gene sequence.

본 명세서의 용어 "CRISPR/Cas 시스템"은 박테리오파지에 대한 박테리아 적응 면역 시스템에서 파생된 것으로, 특히 DNA 절단을 위한 강력한 도구로 사용된다. 모든 CRISPR-Cas 시스템은 CRISPR RNA (crRNA)를 통해 타겟에 대한 특이성을 가지는데, 실험적으로 사용되는 경우에는 가이드 RNA (guide RNA, gRNA)로 약간 변형되어 사용될 수 있다. crRNA의 스페이서(spacer)가 타겟 서열을 인지하고 결합하는데, 이때 타겟 서열은 Protospacer Adjacent Motif (PAM) 라는 서열에 인접하여 존재해야 한다. crRNA와 타겟의 결합 이후, Cas 단백질이 타겟 DNA를 절단한다. 따라서 crRNA 또는 gRNA의 스페이서 부분의 서열을 바꾸는 것만으로 원하는 타겟에 Cas 단백질이 작동하도록 조작할 수 있다. The term "CRISPR/Cas system" herein is derived from the bacterial adaptive immune system for bacteriophages, which are particularly used as powerful tools for DNA cleavage. All CRISPR-Cas systems have target specificity through CRISPR RNA (crRNA), which can be slightly modified into guide RNA (gRNA) when used experimentally. A spacer of crRNA recognizes and binds to a target sequence, and at this time, the target sequence must be adjacent to a sequence called Protospacer Adjacent Motif (PAM). After binding of the crRNA to the target, the Cas protein cuts the target DNA. Therefore, the Cas protein can be engineered to operate on a desired target simply by changing the sequence of the spacer portion of the crRNA or gRNA.

일 구체예에 있어서, 상기 Cas 단백질은 Class 2에 해당하는 것 일 수 있다. 구체적으로, 상기 Class 2에 해당하는 Cas 단백질은 type Ⅴ에 해당할 수 있다. 더욱 구체적으로, 상기 type Ⅴ에 해당하는 Cas 단백질은 Cas12a, mgCas12a, Cas12b, Cas12c, Cas12d, Cas12e, Cas12g, Cas12h, 및 Cas12i로 이루어진 군으로부터 선택된 어느 하나인 이펙터 단백질(effector protein)일 수 있다. In one embodiment, the Cas protein may correspond to Class 2. Specifically, the Cas protein corresponding to Class 2 may correspond to type V. More specifically, the Cas protein corresponding to the type V may be an effector protein selected from the group consisting of Cas12a, mgCas12a, Cas12b, Cas12c, Cas12d, Cas12e, Cas12g, Cas12h, and Cas12i.

또 다른 양상은 시료를 상기 조성물과 접촉시키는 단계; 및 상기 CRISPR/Cas 이펙터 단백질에 의한 상기 단일 가닥의 검출 핵산의 절단에 의해 생성된 검출 가능한 신호를 측정하여, 상기 시료 내 표적 서열을 검출하는 단계를 포함하는, EGFR 돌연변이를 검출하는 방법을 제공한다. Another aspect includes contacting a sample with the composition; And detecting a target sequence in the sample by measuring a detectable signal generated by cleavage of the single-stranded detection nucleic acid by the CRISPR / Cas effector protein. .

또 다른 양상은 상기 조성물을 시료와 접촉시키는 단계를 포함하는, 암을 진단하는 방법 또는 암 진단에 관한 정보를 제공하는 방법을 제공한다. Another aspect provides a method of diagnosing cancer or providing information related to cancer diagnosis, comprising contacting the composition with a sample.

일 구현예에 있어서, 상기 분리된 환자 시료는 cfDNA(cell free DNA)를 포함할 수 있다.In one embodiment, the isolated patient sample may include cell free DNA (cfDNA).

본 명세서의 용어 "cfDNA"는 Circulating free DNA의 약어로 세포핵 안에 존재하지 않고 혈액속에 부유하는 DNA의 조각을 의미한다. "순환 자유 DNA" 또는 "세포 유리 DNA" 등과 상호 교환하여 사용할 수 있다. 순환 자유 DNA (cfDNA)는 혈장으로 방출된 DNA 단편 (50-200 bp)이다. cfDNA는 순환 종양 DNA(ctDNA), 무세포 미토콘드리아 DNA(ccf mtDNA) 및 무세포 태아 DNA(cffDNA)등을 포함하며, 혈류에서 자유롭게 순환하는 다양한 형태의 DNA를 설명하는 데 사용할 수 있다. cfDNA의 상승된 수준은 암이 진행된 경우 관찰된다. cfDNA의 분석을 통하여 암 환자의 혈장 DNA에 종양 관련 돌연변이가 포함되어 있는지 여부를 확인하여 암 진단 및 후속 조치에 사용될 수 있다.As used herein, the term "cfDNA" is an abbreviation for circulating free DNA, and means a piece of DNA that is not present in the cell nucleus but is suspended in the blood. "Circulating free DNA" or "cell free DNA" and the like can be used interchangeably. Circulating free DNA (cfDNA) is a DNA fragment (50-200 bp) released into plasma. cfDNA can be used to describe the various forms of DNA that circulate freely in the bloodstream, including circulating tumor DNA (ctDNA), cell-free mitochondrial DNA (ccf mtDNA), and cell-free fetal DNA (cffDNA). Elevated levels of cfDNA are observed when cancer is advanced. Through cfDNA analysis, it can be used for cancer diagnosis and follow-up by confirming whether tumor-related mutations are included in the plasma DNA of cancer patients.

또 다른 양상은 서열번호 2 내지 6 중 어느 하나를 포함하는 폴리뉴클레오티드 또는 서열번호 2 내지 6 중 어느 하나의 폴리뉴클레오티드와 적어도 95% 상동성을 갖는 폴리뉴클레오티드를 포함하는 가이드 폴리뉴클레오티드를 제공한다.Another aspect provides a guide polynucleotide comprising a polynucleotide comprising any one of SEQ ID NOs: 2-6 or a polynucleotide having at least 95% homology to a polynucleotide of any one of SEQ ID NOs: 2-6.

혈액 시료에서 유래한 변이 DNA를 일 양상에 따른 Cas 뉴클레아제를 이용한 CRISPR/Cas 시스템을 이용하여 in-vitro 절단 및 PCR 증폭을 통해 돌연변이 DNA를 특이적으로 증폭시켜 시퀀싱 오류로 혼동될 수 있는 0.5% 미만의 비율을 갖는 돌연변이를 확인할 수 있는 효과가 있다.Mutant DNA derived from a blood sample is specifically amplified through in-vitro digestion and PCR amplification using the CRISPR/Cas system using Cas nuclease according to one aspect, resulting in a 0.5 sequencing error that can be confused with sequencing errors. There is an effect that can identify mutations with a ratio of less than %.

도 1은 정상 DNA와 일치하는crRNA을 통해 exon19 결실 돌연변이 DNA를 구별할 수 있음을 설명하는 모식도 이다.1 is a schematic diagram explaining that exon19 deletion mutant DNA can be distinguished through crRNA matching normal DNA.

도 2는 정상적인 DNA와 일치하는 crRNA 및 LbCas12a를 사용하여 정상 DNA와 Exon 19 결손 DNA를 in vitro 절단 분석한 결과 사진이다.Figure 2 is a photograph of the results of in vitro cleavage analysis of normal DNA and Exon 19-deficient DNA using crRNA and LbCas12a matching normal DNA.

도 3은 Exon19 결실 돌연변이 DNA를 정상 DNA에 1/100,000까지 연속 희석한 DNA혼합물에 대해 CRISPR/Cas12a 증폭을 각각 3회씩 수행한 결과를 나타낸 그래프이다.3 is a graph showing the results of performing CRISPR/Cas12a amplification three times on a DNA mixture in which Exon19 deletion mutant DNA was serially diluted to 1/100,000 of normal DNA.

도 4은 조직 생검을 통해 결실된 Exon19 결실 여부가 확인된 검체 중 CRISPR/Cas12a 증폭을 통해 Exon19 결실 여부 돌연변이를 확인한 결과를 나타낸 그래프이다.4 is a graph showing the results of confirming Exon19 deletion mutations through CRISPR/Cas12a amplification among specimens in which Exon19 deletion was confirmed through tissue biopsy.

도 5는 LbCas12a의 부수적 효과를 통한 형광 신호 기반 핵산 검출 방법의 개략도이다.Figure 5 is a schematic diagram of a fluorescence signal-based nucleic acid detection method through the side effect of LbCas12a.

도 6은 exon19 결실 돌연변이 DNA 검출을 위한 crRNA가 부수적 효과를 일으키는 기작을 설명하는 그림이다.6 is a diagram explaining the mechanism by which crRNA for detecting exon19 deletion mutant DNA causes side effects.

도 7은 LbCas12a/crRNA 복합체로부터 유도된 형광 신호를 통해 합성된 exon19 결실 돌연변이 DNA 검출 결과를 나타낸 그래프이다.7 is a graph showing results of detection of exon19 deletion mutant DNA synthesized through fluorescence signals induced from the LbCas12a/crRNA complex.

도 8은 임상 폐암 환자 샘플에 의해 검증된 Exon19 결손 패턴의 개략도이다.Figure 8 is a schematic diagram of Exon19 deletion patterns validated by clinical lung cancer patient samples.

도 9는 NGS에 의한 하위 유형 1(subtype1) 결실을 포함하는 것으로 확인된 2개의 양성 샘플(P-1, P-11)과 돌연변이를 포함하지 않는 것으로 확인된 2개의 음성 샘플(N-25, N-26)의 형광 신호를 측정한 결과이다. 9 shows two positive samples (P-1, P-11) confirmed to contain a subtype 1 deletion by NGS and two negative samples (N-25, N-25, This is the result of measuring the fluorescence signal of N-26).

도 10은 NGS에 의한 하위 유형 2(subtype2) 결실을 포함하는 것으로 확인된 2개의 양성 샘플(P-9, P-10)과 돌연변이를 포함하지 않는 것으로 확인된 2개의 음성 샘플(N-25, N-26)의 형광 신호를 측정한 결과이다.10 shows two positive samples (P-9, P-10) confirmed to contain a subtype 2 deletion by NGS and two negative samples (N-25, N-25, This is the result of measuring the fluorescence signal of N-26).

도 11은 NGS에 의한 하위 유형 3(subtype3) 결실을 포함하는 것으로 확인된 2개의 양성 샘플(P-4)과 돌연변이를 포함하지 않는 것으로 확인된 2개의 음성 샘플(N-25, N-26)의 형광 신호를 측정한 결과이다. 11 shows two positive samples (P-4) identified as containing a subtype 3 deletion by NGS and two negative samples (N-25, N-26) identified as not containing the mutation. This is the result of measuring the fluorescence signal of

도 12는 NGS에 의한 하위 유형 4(subtype4) 결실을 포함하는 것으로 확인된 2개의 양성 샘플(P-8)과 돌연변이를 포함하지 않는 것으로 확인된 2개의 음성 샘플(N-25, N-26)의 형광 신호를 측정한 결과이다.Figure 12 shows two positive samples (P-8) identified as containing a subtype 4 (subtype4) deletion by NGS and two negative samples (N-25, N-26) identified as not containing the mutation. This is the result of measuring the fluorescence signal of

이하에서는 실시예를 들어 본 발명을 더욱 구체적으로 설명하고자 하나, 이는 예시적인 것에 불과할 뿐 본 발명의 범위를 제한하고자 함이 아니다. 아래 기재된 실시예들은 발명의 본질적인 요지를 벗어나지 않는 범위에서 변형될 수 있음은 당 업자들에게 있어 자명하다. Hereinafter, the present invention will be described in more detail with examples, but this is only illustrative and is not intended to limit the scope of the present invention. It is apparent to those skilled in the art that the embodiments described below may be modified within a range that does not deviate from the essential gist of the invention.

참조예 1. LbCas12a 재조합 단백질의 정제 및 확인Reference Example 1. Purification and confirmation of LbCas12a recombinant protein

LbCas12a 재조합 단백질을 발현하기 위해 대장균 BL21(DE3) 세포를 pET28a-LbCas12a(addgene No. #114070) 박테리아 발현 벡터로 형질전환시켰다. 배양액이 0.6의 O.D에 도달할 때까지 세포를 37°C에서 Luria Broth(LB)에서 배양하고 재조합 단백질은 isopropyl β-D-thiogalactoside (IPTG, GenDEPOT, Texas, USA)를 최종 농도 1mM로 첨가하여 18°C에서 18시간 동안 유도하였다. 세포 배양물을 원심분리(4000rpm, 30분)하여 배지를 제거하고, 펠릿화된 세포를 용해 완충액(20mM Tris-HCl(pH 8.0), 300mM NaCl, 10mM β-메르캅토에탄올(BioRad, California, U.S.A), 1% Triton X-100(Sigma) 및 1mM phenylmethylsulfonyl fluoride(Sigma, St. Louis, U.S.A) 초음파 처리(Qsonica, Newtown, U.S.A)에 의해 용해(lysis)하였다.To express the LbCas12a recombinant protein, E. coli BL21(DE3) cells were transformed with the pET28a-LbCas12a (addgene No. #114070) bacterial expression vector. Cells were cultured in Luria Broth (LB) at 37 °C until the culture reached an O.D of 0.6 and the recombinant protein was 18 with the addition of isopropyl β-D-thiogalactoside (IPTG, GenDEPOT, Texas, USA) to a final concentration of 1 mM. Induction at °C for 18 hours. The medium was removed by centrifugation (4000 rpm, 30 min) of the cell culture, and the pelleted cells were lysed in lysis buffer (20 mM Tris-HCl (pH 8.0), 300 mM NaCl, 10 mM β-mercaptoethanol (BioRad, California, U.S.A.). ), 1% Triton X-100 (Sigma) and 1 mM phenylmethylsulfonyl fluoride (Sigma, St. Louis, USA) were lysed by sonication (Qsonica, Newtown, USA).

상기 초음파 처리에 의한 세포 용해물을 20,000 x g에서 10분 동안 원심분리하여 세포 파편을 제거하고, 수확한 가용성 분획을 Ni-NTA 수지(Takara, Nojihigashi, Japan)와 혼합하여 정제하였다. 혼합물을 결합 완충액(20mM Tris-HCl(pH 8.0), 300nM NaCl)에서 1시간 동안 4°C에서 교반하면서 인큐베이션하였다. 인큐베이션 후, Ni-NTA 수지를 10 부피의 세척 완충액으로 세척하였다. 다음으로, Ni-NTA 수지에 결합된 단백질을 용출 완충액(20mM Tris-HCl(pH 8.0), 300nM NaCl 및 200mM 이미다졸)으로 용출(elution)시켰다. The cell lysate obtained by the sonication was centrifuged at 20,000 x g for 10 minutes to remove cell debris, and the harvested soluble fraction was purified by mixing with Ni-NTA resin (Takara, Nojihigashi, Japan). The mixture was incubated in binding buffer (20 mM Tris-HCl, pH 8.0, 300 nM NaCl) for 1 hour at 4 °C with agitation. After incubation, the Ni-NTA resin was washed with 10 volumes of wash buffer. Next, the protein bound to the Ni-NTA resin was eluted with an elution buffer (20 mM Tris-HCl (pH 8.0), 300 nM NaCl, and 200 mM imidazole).

용출된 단백질은 Centricon 필터(Amicon Ultra , 밀리포어, 벌링턴, 미국). 재조합 단백질의 순도는 SDS-PAGE(10%, Biorad, California, U.S.A) 및 Coomassie blue 염색(Biorad, California, U.S.A)으로 확인하였다.Eluted proteins were filtered through a Centricon filter (Amicon Ultra, Millipore, Burlington, USA). The purity of the recombinant protein was confirmed by SDS-PAGE (10%, Biorad, California, USA) and Coomassie blue staining (Biorad, California, USA).

참조예 2. crRNA의 시험관 내 전사Reference Example 2. In vitro transcription of crRNA

crRNA의 시험관 내(in vitro) 전사(transcription)를 위해, 표적 DNA에 해당하는 crRNA와 T7 프로모터 서열을 포함하는 DNA 올리고(DNA oligo)는 COSMO Genentech에서 구입하였다. For in vitro transcription of crRNA, a DNA oligo containing the crRNA corresponding to the target DNA and the T7 promoter sequence was purchased from COSMO Genentech.

DNA oligo는 T7 RNA polymerase (NEB, Massachusetts, U.S.A), 50mM MgCl2, 100mM NTPs (ATP, GTP, UTP, CTP), 10X RNA polymerase reaction buffer, Murine RNase inhibitor, 100mM DTT 및 DEPC와 37°C에서8시간 동안 혼합하였다. DNA 올리고를 완전히 제거하기 위하여 상기 혼합물을 DNase와 함께 37°C에서 1시간 동안 배양하고, RNA 정제 키트(RBC, New Taipei City, Taipei)를 사용하여 RNA를 정제하였다. 정제된 RNA의 순도(purity)와 농도(concentration)는 Nanodrop™ 2000 Spectrophotometer(Thermo-Fisher, Massachusetts, U.S.A.)로 측정하였다. 정제된 RNA를 분취하여 -80°C에 보관하였다.DNA oligo was prepared in T7 RNA polymerase (NEB, Massachusetts, U.S.A), 50 mM MgCl2, 100 mM NTPs (ATP, GTP, UTP, CTP), 10X RNA polymerase reaction buffer, Murine RNase inhibitor, 100 mM DTT and DEPC at 37°C for 8 hours. mixed while. To completely remove DNA oligos, the mixture was incubated with DNase at 37 °C for 1 hour, and RNA was purified using an RNA purification kit (RBC, New Taipei City, Taipei). Purity and concentration of purified RNA were measured using a Nanodrop™ 2000 Spectrophotometer (Thermo-Fisher, Massachusetts, USA). The purified RNA was aliquoted and stored at -80 °C.

참조예 3. PCR 앰플리콘 및 시험관 내 절단의 준비Reference Example 3. Preparation of PCR amplicons and in vitro excision

HEK293T의 genomic DNA 로부터 돌연변이를 포함하는 DNA 프라이머(primer)를 사용한 중첩 PCR(overlap PCR)에 의해 엑손 19 (exon 19)에 일부 서열이 삭제된 PCR 앰플리콘(PCR amplicon) 돌연변이 얻었다.A PCR amplicon with a partial sequence deletion in exon 19 was obtained from HEK293T genomic DNA by overlapping PCR using a DNA primer containing the mutation.

정제된 recombinant Cas12a와 돌연변이 DNA 이외의 DNA를 제거하도록 디자인된 crRNA를 미리 혼합하고 PCR 앰플리콘과 함께 37°C에서 1시간 동안 배양하였다. 이후 90°C에서 1분 동안 Cas12a/crRNA 리보핵단백질 복합체(ribonucleoprotein complex)를 비활성화(inactivation)시켰다.The purified recombinant Cas12a and crRNA designed to remove DNA other than mutant DNA were pre-mixed and incubated with the PCR amplicons for 1 hour at 37 °C. Then, the Cas12a/crRNA ribonucleoprotein complex was inactivated at 90 °C for 1 minute.

PCR 앰플리콘의 절단(cleavage)은 아가로스 겔 전기영동(agarose gel electrophoresis)(2%)에 의해 확인하였다.Cleavage of PCR amplicons was confirmed by agarose gel electrophoresis (2%).

참조예 4. CRISPR/Cas12a 증폭을 통한 돌연변이 DNA 농축Reference Example 4. Concentration of mutant DNA through CRISPR/Cas12a amplification

돌연변이 DNA(mutant DNA)를 특이적으로 증폭(specifically amplify)하기 위해 정제된 재조합 LbCas12a 단백질과 인위적으로 합성된 crRNA를 미리 혼합하고, PCR 앰플리콘(PCR amplicon)과 함께 37°C에서 1시간 동안 배양하였다. 이후, 90°C에서 1분 동안 배양하여 Cas12a/crRNA 리보핵단백질 복합체를 비활성화하였다.In order to specifically amplify mutant DNA, purified recombinant LbCas12a protein and artificially synthesized crRNA were pre-mixed and incubated with PCR amplicon for 1 hour at 37 °C did Then, the Cas12a/crRNA ribonucleoprotein complex was inactivated by incubation at 90 °C for 1 min.

상기 과정을 통해 정상 DNA(nolmal DNA)를 절단(cleavage)한 후, 절단된 혼합물(the cleaved mixture)을 PCR 증폭을 통해 절단되지 않은 돌연변이 DNA를 증폭시켰다(98°C에서 30초 동안 변성(denaturation), 58°C에서 30초 동안 프라이머 어닐링(primer annealing), 72°C에서 30초 동안 신장(extension), 30 cycles). After cleavage of normal DNA through the above process, the cleaved mixture was amplified by PCR amplification to uncleaved mutant DNA (denaturation at 98 ° C for 30 seconds). ), primer annealing at 58 °C for 30 sec, extension at 72 °C for 30 sec, 30 cycles).

높은 처리량 시퀀싱(High throughput sequencing)을 위해 nested PCR(98°C에서 30초 동안 변성(denaturation), 58°C에서 30초 동안 프라이머 어닐링(primer annealing) 및 72°C에서 30초 동안 신장(extension), 35 cycles)을 사용하여 enriched PCR production을 바코드 서열(barcode sequence)과 연결하였다. 높은 처리량 시퀀싱(High throughput sequencing)은 Illumina I-seq 100 시퀀싱 기기(Illumine, California, USA)로 수행하였다.For high throughput sequencing, nested PCR (denaturation at 98°C for 30 seconds, primer annealing at 58°C for 30 seconds, and extension at 72°C for 30 seconds) , 35 cycles) was used to link the enriched PCR production with the barcode sequence. High throughput sequencing was performed with an Illumina I-seq 100 sequencing machine (Illumina, California, USA).

CRISPR 분석기 웹 도구(CRISPR anylyzer web tool)를 통해 시퀀싱 원시 데이터(sequencing raw data)를 분석하여 돌연변이 DNA 비율(rate)을 계산하였다.The mutant DNA rate was calculated by analyzing the sequencing raw data through the CRISPR analyzer web tool.

구체적으로, 무세포 순환 DNA(cfDNA)는 폐암 환자의 혈장에서 추출였고, 추출된 cfDNA를 돌연변이 DNA(mutant DNA)를 특이적으로 증폭(specifically amplify)하기 위해 정제된 재조합 LbCas12a 단백질과 인위적으로 합성된 crRNA를 혼합하여 정상 DNA(nolmal DNA)를 절단(cleavage) 하였다. 절단된 혼합물(the cleaved mixture)을 PCR 증폭을 통해 절단되지 않은 돌연변이 DNA를 증폭시켰다. 높은 처리량 시퀀싱(High throughput sequencing)을 수행하고, CRISPR 분석기 웹 도구(CRISPR anylyzer web tool)를 통해 시퀀싱 원시 데이터(sequencing raw data)를 분석하였다.Specifically, cell-free circulating DNA (cfDNA) was extracted from the plasma of lung cancer patients, and the extracted cfDNA was artificially synthesized with purified recombinant LbCas12a protein to specifically amplify mutant DNA. CrRNA was mixed to cleavage normal DNA (nolmal DNA). Uncleaved mutant DNA was amplified from the cleaved mixture by PCR amplification. High throughput sequencing was performed and sequencing raw data was analyzed through the CRISPR analyzer web tool.

참조예 5. CRISPR/Cas12a 기반 형광 리포터 분석Reference Example 5. CRISPR/Cas12a-based fluorescent reporter assay

LbCas12a의 부수적 효과(collateral effects)을 이용한 형광 신호는 형광단-소광제 리포터 분석(fluorophore-quencher reporter assay)을 통해 확인하였다. 안정적인 복합체 형성을 위해 정제된 LbCas12a 재조합 단백질(50nM)과 돌연변이된 DNA 서열(50nM)에 해당하는 crRNA를 NEBuffer 2.1에 미리 혼합하여 실온에서 5분 동안 두었다. DNA 혼합물과 FQ 리포터(5`/6-FAM/TTATT/BHQ1/3`)를 LbCas12a/crRNA 리보뉴클레아제 어셈블리 혼합물(LbCas12a/crRNA ribonuclease assembly mixture)에 첨가한 후 96 웰 블랙 플레이트(96 well black plate)에 로딩하고, 형광 신호를 1분 간격으로 1시간(FAM) 측정하였다. (FQ (λex: 483/30 nm, λem: 530/30 nm) Fluoroskan™ Microplate Fluorometer(Thermo-Fisher, Massachusetts, U.S.A).)Fluorescent signals using collateral effects of LbCas12a were confirmed through a fluorophore-quencher reporter assay. To form a stable complex, the purified LbCas12a recombinant protein (50 nM) and the crRNA corresponding to the mutated DNA sequence (50 nM) were pre-mixed in NEBuffer 2.1 and allowed to stand at room temperature for 5 minutes. After adding the DNA mixture and the FQ reporter (5`/6-FAM/TTATT/BHQ1/3`) to the LbCas12a/crRNA ribonuclease assembly mixture, a 96 well black plate was prepared. plate), and the fluorescence signal was measured for 1 hour (FAM) at 1 minute intervals. (FQ (λex: 483/30 nm, λem: 530/30 nm) Fluoroskan™ Microplate Fluorometer (Thermo-Fisher, Massachusetts, U.S.A).)

실시예 1. EGFR exon19 결실 돌연변이를 특이적으로 증폭하기 위한 crRNA 설계 및 활성 확인Example 1. crRNA design and activity confirmation for specifically amplifying EGFR exon19 deletion mutants

실시예 1.1. EGFR exon19 결실 돌연변이를 특이적으로 증폭하기 위한 crRNA 설계Example 1.1. Design of crRNA to specifically amplify EGFR exon19 deletion mutants

EGFR exon19 결실 돌연변이를 특이적으로 증폭하기 위한crRNA를 설계하였다. A crRNA was designed to specifically amplify the EGFR exon19 deletion mutant.

구체적으로, L858R 점 돌연변이와 달리 exon19 결실 패턴은 9bp~18bp 등 다양한 결실 크기로 나타난다. 따라서 도 1에서 보는 바와 같이, EGFR exon19 결실 돌연변이의 경우 추가적으로 잘못된 페어링을 도입하지 않고도 효과적으로 정상 DNA와 exon19 결실 돌연변이 DNA를 구별할 수 있다. EGFR exon19 결실 돌연변이를 특이적으로 증폭하기 위한crRNA의 구성은 표 1에 기재하였다.Specifically, unlike the L858R point mutation, exon19 deletion patterns appear in various deletion sizes, such as 9bp to 18bp. Accordingly, as shown in FIG. 1 , in the case of EGFR exon19 deletion mutants, normal DNA and exon19 deletion mutant DNA can be effectively distinguished without additionally introducing incorrect pairing. The composition of crRNA for specifically amplifying EGFR exon19 deletion mutants is shown in Table 1.

WT crRNA 구성 (서열번호 1)WT crRNA construct (SEQ ID NO: 1) AA TTTCT ACTAA GTGTA GATAA TTTCT ACTAA GTGTA GAT GGAGA TGTTG CTTCT CTTAAGGAGA TGTTG CTTCT CTTAA TTTTTT AA TTTCT ACTAA GTGTA GATAA TTTCT ACTAA GTGTA GAT 직접반복부(direct repeat) 서열direct repeat sequence GGAGA TGTTG CTTCT CTTAAGGAGA TGTTG CTTCT CTTAA 표적 서열target sequence TTTTTT 절단(cleavage) 효율을 높이는 서열Sequences that increase cleavage efficiency

실시예 1.2. EGFR exon19 결실 돌연변이 특이적 증폭을 위한crRNA의 in vitro 절단 활성 확인Example 1.2. Confirmation of in vitro cleavage activity of crRNA for specific amplification of EGFR exon19 deletion mutant

EGFR Exon19 유전자좌에서 15 bp로 결실된 DNA 단편을 합성하고 정상 DNA 서열과 완벽하게 일치하는 crRNA 및 LbCas12a 으로 시험관 내 절단 분석을 수행하였다. 상기 결과는 도 2에 나타내었다.A DNA fragment with a 15 bp deletion in the EGFR Exon19 locus was synthesized and an in vitro cleavage assay was performed with crRNA and LbCas12a that perfectly matched the normal DNA sequence. The results are shown in Figure 2.

도 2에서 보는 바와 같이, 정상 DNA 특이적 crRNA는 정상 DNA를 상당히 절단하였으나, 15bp 결실을 포함하는 돌연변이 DNA는 전혀 절단되지 않았다. As shown in Figure 2, the normal DNA-specific crRNA significantly cleaved the normal DNA, but the mutant DNA containing the 15bp deletion did not cut at all.

상기 결과는 L858R point mutated DNA와 달리 crRNA에 mis-pairing을 도입하지 않고도 정상 DNA와 Exon19 결실 돌연변이 DNA를 구별하여 정상 DNA만을 절단할 수 있음을 의미한다. The above result means that, unlike L858R point mutated DNA, only normal DNA can be cut by distinguishing between normal DNA and Exon19 deletion mutant DNA without introducing mis-pairing into crRNA.

실시예 2. EGFR exon19 결실 돌연변이를 특이적으로 증폭하도록 설계된 CRISPR/Cas시스템 및 EGFR exon19 결실 돌연변이 검출 감도 확인Example 2. CRISPR/Cas system designed to specifically amplify EGFR exon19 deletion mutant and confirmation of EGFR exon19 deletion mutant detection sensitivity

실시예 2.1 EGFR exon19 결실 돌연변이를 특이적으로 증폭하도록 설계된 CRISPR/Cas시스템 Example 2.1 CRISPR/Cas system designed to specifically amplify EGFR exon19 deletion mutants

매우 적은 양의 EGFR exon19 결실 돌연변이 DNA를 특이적으로 증폭할 수 있는지 확인하기 위해 실시예 1의 crRNA와 LbCas12a를 이용하여 참조예 3과 같은 방법으로 정상 DNA를 특이적으로 절단한 후 PCR을 돌연변이 DNA를 증폭시켰다.In order to confirm that a very small amount of EGFR exon19 deletion mutant DNA can be specifically amplified, PCR is performed after the normal DNA is specifically digested in the same manner as in Reference Example 3 using the crRNA of Example 1 and LbCas12a. amplified.

구체적으로 15bp의 exon 19결실 DNA 단편을 정상 DNA 단편과 1:100,000이 되도록 희석하였다. 시험관 절단 및 PCR 증폭을 포함하는 CRISPR/Cas12a 증폭을 희석된 DNA 혼합물에 대해 각각 3회씩 수행하였다. Exon19 결실 돌연변이 빈도는 높은 처리량 시퀀싱으로 계산하였다. 상기 결과는 도 10에 나타내었다.Specifically, a 15 bp exon 19 deleted DNA fragment was diluted with a normal DNA fragment at a ratio of 1:100,000. CRISPR/Cas12a amplification, including in vitro digestion and PCR amplification, was performed on the diluted DNA mixture in triplicate each. Exon19 deletion mutation frequencies were calculated by high-throughput sequencing. The results are shown in FIG. 10 .

도 3에서 보는 바와 같이, 1:100,000 희석된 돌연변이 DNA는 일반적인 액체 생검을 사용하여 검출되지 않았다. 반면, 본 발명의 일 실시예에 따른 CRISPR/Cas 시스템을 통한 3회 증폭을 통해 Exon19 결실 돌연변이 DNA를 최대 12.6%까지 증폭할 수 있음을 확인하였다. As shown in Figure 3, mutant DNA at a 1:100,000 dilution was not detected using conventional liquid biopsy. On the other hand, it was confirmed that the Exon19 deletion mutant DNA could be amplified up to 12.6% through 3 amplification through the CRISPR/Cas system according to an embodiment of the present invention.

상기 결과는 일반적인 액체 생검(No amplification, N/A)을 사용하여서는 측정할 수 없는 돌연변이 DNA를 본 발명의 일 구체체예에 따른 CRISPR/Cas12a 시스템을 통하여 측정 민감도 확보할 수 있음을 의미한다.The above result means that the measurement sensitivity can be secured through the CRISPR/Cas12a system according to one embodiment of the present invention for mutant DNA that cannot be measured using a general liquid biopsy (No amplification, N/A).

실시예 2.2 폐암환자의 cfDNA에서 CRISPR/Cas12a 증폭을 통한 Exon19 결실 돌연변이 검출Example 2.2 Detection of Exon19 deletion mutation through CRISPR/Cas12a amplification in cfDNA of lung cancer patients

임상 샘플에서 Exon19 결실 돌연변이 DNA 검출 효과를 확인하기 위하여 폐암환자의 cfDNA에서 Exon19 결실 돌연변이 DNA의 CRISPR/Cas12a 증폭을 수행하였다.To confirm the effect of detecting Exon19 deletion mutant DNA in clinical samples, CRISPR/Cas12a amplification of Exon19 deletion mutant DNA was performed in cfDNA of lung cancer patients.

구체적으로 조직 생검으로 확인된 Exon 19 결실(+) 환자 11명과 Exon 19 결실(-) 환자 11명의 혈액에서 cfDNA를 추출하고, CRISPR/Cas12a 증폭을 수행하였다. 상기 결과는 도 4에 나타내었다.Specifically, cfDNA was extracted from the blood of 11 patients with Exon 19 deletion (+) and Exon 19 deletion (-) confirmed by tissue biopsy, and CRISPR/Cas12a amplification was performed. The results are shown in FIG. 4 .

도 4에서 보는 바와 같이, CRISPR/Cas12a 증폭은 조직 생검(P1, P4, P8, P9, P10 및 P11)을 통해 확인된 6개의 Exon 19 결실(+) 샘플에서 결실된 돌연변이 DNA를 99.1%로 크게 증폭함을 확인하였다. 이들 샘플 중 2개(P4 및 P9)는 증폭 전에는 1% 미만의 결실률을 보였지만, CRISPR/Cas12a 증폭을 통해 각각 91.4% 및 93.4%로 증폭되었다.As shown in FIG. 4, CRISPR/Cas12a amplification significantly reduced the deleted mutant DNA to 99.1% in six Exon 19 deletion (+) samples identified through tissue biopsies (P1, P4, P8, P9, P10 and P11). amplification was confirmed. Two of these samples (P4 and P9) showed less than 1% deletion rate before amplification, but were amplified to 91.4% and 93.4%, respectively, with CRISPR/Cas12a amplification.

상기 결과는 일 구체예에 해당하는 CRISPR/Cas12a 증폭 시스템이 혈액 시료에서 유래한 변이 DNA를 효과적으로 증폭할 수 있어 EGFR 돌연변이 DNA 검출에 유용한 도구로 활용될 수 있음을 의미한다.The above result means that the CRISPR/Cas12a amplification system corresponding to one embodiment can effectively amplify mutant DNA derived from a blood sample and can be used as a useful tool for detecting EGFR mutant DNA.

실시예 3. CRISPR/Cas12a 시스템을 통한 LbCas12a의 부수적 효과에 기반한 EGFR exon19 결실 돌연변이 DNA의 형광 신호 증폭 확인Example 3. Confirmation of fluorescent signal amplification of EGFR exon19 deletion mutant DNA based on side effect of LbCas12a through CRISPR/Cas12a system

실시예 3.1. 합성된 EGFR exon19 결실 돌연변이 DNA의 형광 신호 증폭 확인Example 3.1. Confirmation of fluorescence signal amplification of synthesized EGFR exon19 deletion mutant DNA

Cas12a 단백질의 부수적 효과를 이용하여 소량의 표적 DNA 검출 시 Cas12a 단백질의 부수적 효과로 유도된 형광 신호와 배경 신호를 구별할 수 없는 문제를 해결하기 위해 본 발명의 일 구체예에 따른 CRISPR/Cas12a 증폭 시스템을 이용하여 합성된 EGFR exon19 결실 돌연변이 DNA의 형광 신호 증폭 확인하였다.CRISPR/Cas12a amplification system according to one embodiment of the present invention to solve the problem of not being able to distinguish between a fluorescent signal and a background signal induced by a side effect of Cas12a protein when detecting a small amount of target DNA using a side effect of Cas12a protein The fluorescence signal amplification of the EGFR exon19 deletion mutant DNA synthesized using was confirmed.

구체적으로 도 5에서 나타낸 바와 같이, LbCas12a/crRNA가 표적 DNA를 인식하면 LbCas12a의 부수적 효과인 비특이적 ssDNase가 활성화된다. 활성화된 LbCas12a/crRNA 리보핵단백질은 참조예5에 따라 혼합한 FQ ssDNA 리포터를 포함한 단일 가닥 DNA를 비특이적으로 절단한다. 표적 DNA의 양은 절단된 FQ 리포터로부터 유도된 형광 신호에 의해 정량화하였다. 도 6에서 나타낸 바와 같이 exon19 결실 DNA 서열과 일치하는 crRNA를 사용하여 EGFR exon19 결실 돌연변이 DNA의 형광 신호를 검출 할 수 있는지 37°C에서 20분 동안 측정하였다. 다양한 유형의 exon19 결실 DNA 서열과 일치하는 crRNA구성은 표 2 및 표 3에 나타내었고, 이 중, 서열번호 2에 해당하는 crRNA로 EGFR exon19 결실 돌연변이 DNA의 형광 신호를 검출 결과는 도 7에 나타내었다. 도 7에서"Blank"는 DNA 없이 Cas12a/crRNA 리보핵산단백질만 배양한 것을 나타낸다.Specifically, as shown in FIG. 5 , when LbCas12a/crRNA recognizes a target DNA, non-specific ssDNase, which is a side effect of LbCas12a, is activated. The activated LbCas12a/crRNA ribonucleoprotein non-specifically cuts the single-stranded DNA containing the FQ ssDNA reporter mixed according to Reference Example 5. The amount of target DNA was quantified by the fluorescence signal derived from the cleaved FQ reporter. As shown in FIG. 6 , the fluorescence signal of the EGFR exon19 deletion mutant DNA was detected using crRNA matching the exon19 deletion DNA sequence at 37 °C for 20 minutes. Tables 2 and 3 show the constructs of crRNAs corresponding to various types of exon19 deletion DNA sequences, and among them, the fluorescence signal detection results of EGFR exon19 deletion mutant DNA with the crRNA corresponding to SEQ ID NO: 2 are shown in FIG. . In FIG. 7, "Blank" indicates that only Cas12a/crRNA ribonucleic acid protein was cultured without DNA.

exon19 결실 DNA 서열과 일치하는 crRNA구성 (서열번호 2)crRNA construction matching exon19 deletion DNA sequence (SEQ ID NO: 2) AA TTTCT ACTAA GTGTA GAT GGAGA TGTCT TGATA GCGAC TTT AA TTTCT ACTAA GTGTA GAT GGAGA TGTCT TGATA GCGAC TTT AA TTTCT ACTAA GTGTA GATAA TTTCT ACTAA GTGTA GAT 직접반복부(direct repeat) 서열direct repeat sequence GGAGA TGTCT TGATA GCGACGGAGA TGTCT TGATA GCGAC 표적 서열target sequence TTTTTT TTT 서열TTT sequence

도 7에서 보는 바와 같이, 본 발명의 일 구체예에 따른 CRISPR/Cas12a 시스템을 이용한 증폭 절차 없이 exon19 결실 DNA 서열과 일치하는 crRNA를 사용하여 형광신호를 검출한 결과 합성된 exon19 결실 돌연변이 DNA를 정상 DNA에 1/10로 희석한 DNA 혼합물을 제외하고는 배경 신호와 구별되지 않음을 확인하였다. 반면 CRISPR/Cas12a 증폭을 사용한 경우 합성된 exon19 결실 돌연변이 DNA를 정상 DNA로 1/100,000 희석된 혼합물을 포함한 모든 혼합물에서 배경 신호로부터 뚜렷한 형광 신호를 관찰하였다. As shown in FIG. 7, the exon19 deletion mutant DNA synthesized as a result of detecting a fluorescent signal using crRNA matching the exon19 deletion DNA sequence without an amplification procedure using the CRISPR/Cas12a system according to one embodiment of the present invention is converted into normal DNA. It was confirmed that the signal was indistinguishable from the background signal except for the DNA mixture diluted 1/10 in . On the other hand, when CRISPR/Cas12a amplification was used, a clear fluorescence signal was observed from the background signal in all mixtures, including mixtures in which the synthesized exon19 deletion mutant DNA was diluted 1/100,000 with normal DNA.

상기 결과는 일 구체예에 따른 CRISPR/Cas12a 시스템이 배경 신호와 구별할 수 없었던 낮은 형광 신호를 효과적으로 높일 수 있음을 의미한다. The above result means that the CRISPR/Cas12a system according to one embodiment can effectively increase a low fluorescence signal that is indistinguishable from a background signal.

실시예 3.2. 환자의 cfDNA에서 Exon19 결실 하위 패턴 돌연변이 DNA의 형광 신호 증폭 확인Example 3.2. Confirmation of fluorescent signal amplification of Exon19 deletion subpattern mutant DNA in patient cfDNA

cfDNA에서 희귀 돌연변이 DNA 검출에 적용될 수 있는지 확인하기 위해 NGS를 통해 돌연변이가 있는 것으로 확인된 환자의 cfDNA에서 exon19 결실 돌연변이 DNA 검출을 위한 CRISPR/Cas12a 증폭을 수행하였다. 폐암 환자 시료에서 exon19 결실 돌연변이 DNA의 형광 신호를 검출하기 위해, NSCLC(비소세포폐암) 환자에서 발견되는 4개의 exon19 결실 돌연변이 하위 패턴 각각에 해당하는 crRNA를 설계하여 도 3에 나타내었다. 도 8에서 보는 바와 같이, exon 19 결실 돌연변이 하위 유형 1~4(subtype1~4)는 exon 19 에서 각각 18bp, 15bp, 16+2bp 및 9bp가 삭제된 형태를 나타낸다. 결실 돌연변이 하위 유형의 결실을 포함하는 LbCas12a 및 crRNA 해당 DNA 서열을 사용하여 37°C에서 40분 동안 형광 신호를 측정하였고, 그 결과를 도 9 내지 도 12에 나타내였다. 도 9 내지 도 12에서 P는 이전에 조직 생검에 의해 확인된 양성 샘플을 나타내고, N은 이전에 조직 생에 의해 확인된 음성 샘플을 나타낸다.To confirm that it can be applied to detection of rare mutant DNA in cfDNA, CRISPR/Cas12a amplification was performed to detect exon19 deletion mutant DNA in cfDNA of patients confirmed to have the mutation through NGS. In order to detect fluorescence signals of exon19 deletion mutant DNA in lung cancer patient samples, crRNAs corresponding to each of the four exon19 deletion mutant subpatterns found in NSCLC (non-small cell lung cancer) patients were designed and shown in FIG. 3 . As shown in FIG. 8 , exon 19 deletion mutant subtypes 1 to 4 (subtypes 1 to 4) represent forms in which 18 bp, 15 bp, 16+2 bp, and 9 bp of exon 19 are deleted, respectively. Fluorescence signals were measured at 37 °C for 40 minutes using LbCas12a and crRNA corresponding DNA sequences containing deletion subtypes of deletion mutants, and the results are shown in FIGS. 9 to 12 . In Figures 9-12, P represents a positive sample previously confirmed by tissue biopsy, and N represents a negative sample previously confirmed by tissue biopsy.

Subtype_1 crRNA 구성 (서열번호3)Subtype_1 crRNA construction (SEQ ID NO: 3) AA TTTCT ACTAA GTGTA GAT GATTC CTTGA TAGCG ACGGG TTT AA TTTCT ACTAA GTGTA GAT GATTC CTTGA TAGCG ACGGG TTT AA TTTCT ACTAA GTGTA GATAA TTTCT ACTAA GTGTA GAT 직접반복부(direct repeat) 서열direct repeat sequence GATTC CTTGA TAGCG ACGGGGATTC CTTGA TAGCG ACGGG 표적 서열target sequence TTTTTT 절단(cleavage) 효율을 높이는 서열Sequences that increase cleavage efficiency Subtype_2 crRNA 구성 (서열번호 4)Subtype_2 crRNA construction (SEQ ID NO: 4) AA TTTCT ACTAA GTGTA GAT GGAGA TGTTT TGATA GCGAC TTT AA TTTCT ACTAA GTGTA GAT GGAGA TGTTT TGATA GCGAC TTT AA TTTCT ACTAA GTGTA GATAA TTTCT ACTAA GTGTA GAT 직접반복부(direct repeat) 서열direct repeat sequence GGAGA TGTTT TGATA GCGACGGAGA TGTTT TGATA GCGAC 표적 서열target sequence TTTTTT 절단(cleavage) 효율을 높이는 서열Sequences that increase cleavage efficiency Subtype_3 crRNA 구성 (서열번호 5)Subtype_3 crRNA construction (SEQ ID NO: 5) AA TTTCT ACTAA GTGTA GAT GGAAT CTTGA TAGCG ACGGG TTT AA TTTCT ACTAA GTGTA GAT GGAAT CTTGA TAGCG ACGGG TTT AA TTTCT ACTAA GTGTA GATAA TTTCT ACTAA GTGTA GAT 직접반복부(direct repeat) 서열direct repeat sequence GGAAT CTTGA TAGCG ACGGGGGAAT CTTGA TAGCG ACGGG 표적 서열target sequence TTTTTT 절단(cleavage) 효율을 높이는 서열Sequences that increase cleavage efficiency Subtype_4 crRNA 구성 (서열번호 6)Subtype_4 crRNA construction (SEQ ID NO: 6) AA TTTCT ACTAA GTGTA GAT GGAGA TGTTG GTTCC TTGAT TTT AA TTTCT ACTAA GTGTA GAT GGAGA TGTTG GTTCC TTGAT TTT AA TTTCT ACTAA GTGTA GATAA TTTCT ACTAA GTGTA GAT 직접반복부(direct repeat) 서열direct repeat sequence GGAGA TGTTG GTTCC TTGATGGAGA TGTTG GTTCC TTGAT 표적 서열target sequence TTTTTT 절단(cleavage) 효율을 높이는 서열Sequences that increase cleavage efficiency

도 9는 NGS에 의한 하위 유형 1(subtype1) 결실을 포함하는 것으로 확인된 2개의 양성 샘플(P-1, P-11)과 돌연변이를 포함하지 않는 것으로 확인된 2개의 음성 샘플(N-25, N-26)의 형광 신호를 측정한 결과이다. 도 9에서 보는 바와 같이, 본 발명의 일 구현예인 CRISPR/Cas12a 증폭 수행 전에 모든 샘플에서 형광 신호가 검출되지 않았다. 반면, CRISPR/Cas12a 증폭(3회)을 통해 증폭된 두 개의 양성 샘플(P1 및 P11)의 형광 신호는 유의하게 검출되었으나, 음성 샘플(N25 및 N26)의 형광 신호는 검출되지 않았다.9 shows two positive samples (P-1, P-11) confirmed to contain a subtype 1 deletion by NGS and two negative samples (N-25, N-25, This is the result of measuring the fluorescence signal of N-26). As shown in FIG. 9, no fluorescence signal was detected in all samples prior to CRISPR/Cas12a amplification, which is an embodiment of the present invention. On the other hand, the fluorescence signals of the two positive samples (P1 and P11) amplified through CRISPR/Cas12a amplification (3 times) were significantly detected, but the fluorescence signals of the negative samples (N25 and N26) were not detected.

도 10은 NGS에 의한 하위 유형 2(subtype2) 결실을 포함하는 것으로 확인된 2개의 양성 샘플(P-9, P-10)과 돌연변이를 포함하지 않는 것으로 확인된 2개의 음성 샘플(N-25, N-26)의 형광 신호를 측정한 결과이다. subtype2 결실을 가진 임상 샘플의 형광 신호는 CRISPR/Cas12a 증폭없이도 양성 샘플(P10)과 음성 샘플(N25, N26)을 구별할 수 있었다. 이는 P10 샘플이 증폭 전에 이미 결실된 돌연변이 DNA의 높은 비율(50.4%)을 포함하고 있기 때문으로 보인다. 그럼에도 불구하고, subtype2 결실이 있는 샘플의 형광 신호는 음성 샘플과 CRISPR/Cas12a 증폭에 의해 더욱 명확하게 구별되는 것을 확인하였다.10 shows two positive samples (P-9, P-10) confirmed to contain a subtype 2 deletion by NGS and two negative samples (N-25, N-25, This is the result of measuring the fluorescence signal of N-26). Fluorescent signals of clinical samples with subtype2 deletion were able to discriminate positive samples (P10) from negative samples (N25, N26) without CRISPR/Cas12a amplification. This seems to be because the P10 sample contains a high percentage (50.4%) of the mutant DNA already deleted before amplification. Nevertheless, it was confirmed that the fluorescence signal of the sample with subtype2 deletion was more clearly distinguished from the negative sample by CRISPR/Cas12a amplification.

도 11은 NGS에 의한 하위 유형 3(subtype3) 결실을 포함하는 것으로 확인된 2개의 양성 샘플(P-4)과 돌연변이를 포함하지 않는 것으로 확인된 2개의 음성 샘플(N-25, N-26)의 형광 신호를 측정한 결과이고, 도 12는 NGS에 의한 하위 유형 4(subtype4) 결실을 포함하는 것으로 확인된 2개의 양성 샘플(P-8)과 돌연변이를 포함하지 않는 것으로 확인된 2개의 음성 샘플(N-25, N-26)의 형광 신호를 측정한 결과이다. 도 11 및 도 12에서 보는 바와 같이, subtype3 및 4 결실 유형 돌연변이가 양성으로 확인된 P4 및 P8 샘플의 형광 신호는 CRISPR/Cas12a 시스템으로 증폭 전에는 음성 샘플(N25 및 N26)과 구분할 수 없었다. 반면, CRISPR/Cas12a 증폭을 통해 형광 신호가 강화되었음을 확인하였다. 11 shows two positive samples (P-4) identified as containing a subtype 3 deletion by NGS and two negative samples (N-25, N-26) identified as not containing the mutation. 12 shows two positive samples (P-8) confirmed to contain a subtype 4 (subtype4) deletion by NGS and two negative samples confirmed to not contain a mutation. It is the result of measuring the fluorescence signal of (N-25, N-26). As shown in FIGS. 11 and 12, the fluorescence signals of P4 and P8 samples in which subtype 3 and 4 deletion type mutations were positively confirmed were indistinguishable from negative samples (N25 and N26) before amplification with the CRISPR/Cas12a system. On the other hand, it was confirmed that the fluorescence signal was enhanced through CRISPR/Cas12a amplification.

상기 결과는 일 구체예의 CRISPR/Cas12a 증폭 시스템은 혈액 시료에서 유래한 변이 DNA를 효과적으로 증폭할 수 있음을 의미한다. 따라서 다양한 질병 진단에 활용될 수 있을 뿐만 아니라, 진단 분야 외에도 정상 DNA와 변이 DNA의 구분이 병원성 대립유전자 특이적 유전체 편집을 통한 암 치료 등 다양한 분야에 적용될 수 있다. The above result means that the CRISPR/Cas12a amplification system of one embodiment can effectively amplify mutant DNA derived from a blood sample. Therefore, it can be used for diagnosis of various diseases, and in addition to diagnosis, the distinction between normal DNA and mutant DNA can be applied to various fields such as cancer treatment through genome editing specific to pathogenic alleles.

Claims (20)

표적 서열과 혼성화하고, EGFR(Epidermal growth factor receptor) 돌연변이 유전자 서열에 상보적인 연속적인 염기를 포함하는 가이드 폴리뉴클레오티드 또는 상기 가이드 폴리뉴클레오티드를 암호화하는 핵산; A guide polynucleotide that hybridizes to a target sequence and includes consecutive bases complementary to an epidermal growth factor receptor (EGFR) mutant gene sequence or a nucleic acid encoding the guide polynucleotide; CRISPR/Cas 이펙터 단백질(effector protein), 또는 상기 단백질을 암호화하는 핵산; 및 A CRISPR/Cas effector protein, or a nucleic acid encoding the protein; and 단일 가닥이고, 상기 가이드 폴리뉴클레오티드와 혼성화 되는 표지된 검출 핵산을 포함하는 EGFR 돌연변이를 검출하기 위한 조성물.A composition for detecting an EGFR mutation comprising a single-stranded, labeled detection nucleic acid that hybridizes with the guide polynucleotide. 제1항에 있어서, 상기 단일 가닥 검출 핵산은 DNA는 형광-방출 염료쌍을 포함하는 것인, 조성물.The composition according to claim 1, wherein the single-stranded detection nucleic acid DNA comprises a fluorescence-emitting dye pair. 제2항에 있어서, 상기 형광-방출 염료쌍은 FRET 쌍 또는 퀸처/플루오르 쌍인 것인, 조성물.The composition of claim 2, wherein the fluorescence-emitting dye pair is a FRET pair or a quencher/fluor pair. 제1항에 있어서, 상기 EGFR 돌연변이는 G719S 돌연변이, G179C 돌연변이, G719A 돌연변이, S720F 돌연변이, T790M 돌연변이, D761Y 돌연변이, exon 19 결실 돌연변이, D770_N771 삽입 돌연변이, V765A 돌연변이, T783A 돌연변이, S761I 돌연변이, T790M 돌연변이, V769L 돌연변이, N771I 돌연변이, L858R 돌연변이, L861Q 돌연변이, L861R 돌연변이 중 어느 하나인 것인, 조성물.The method of claim 1, wherein the EGFR mutations are G719S mutation, G179C mutation, G719A mutation, S720F mutation, T790M mutation, D761Y mutation, exon 19 deletion mutation, D770_N771 insertion mutation, V765A mutation, T783A mutation, S761I mutation, T790M mutation, V769L Mutation, N771I mutation, L858R mutation, L861Q mutation, any one of the L861R mutation, the composition. 제1항에 있어서, 상기 가이드 폴리뉴클레오티드는 5' 말단에서 3' 말단 순서로, 직접 반복부 서열, 표적 서열에 혼성화할 수 있는 가이드 서열을 포함하는 것인, 조성물.The composition according to claim 1, wherein the guide polynucleotide comprises, in order from the 5' end to the 3' end, a direct repeating sequence and a guide sequence capable of hybridizing to the target sequence. 제5항에 있어서, 상기 가이드 폴리뉴클레오티드는 3'말단에 절단 효율을 높이는 서열을 더 포함하는 것인, 조성물.The composition according to claim 5, wherein the guide polynucleotide further comprises a sequence increasing cleavage efficiency at the 3' end. 제1항에 있어서, 상기 CRISPR/Cas 이펙터 단백질은 Cas12a, mgCas12a, Cas12b, Cas12c, Cas12d, Cas12e, Cas12g, Cas12h, 및 Cas12i로 이루어진 군으로부터 선택된 어느 하나인 이펙터 단백질인 것인, 조성물.The composition of claim 1, wherein the CRISPR/Cas effector protein is any one effector protein selected from the group consisting of Cas12a, mgCas12a, Cas12b, Cas12c, Cas12d, Cas12e, Cas12g, Cas12h, and Cas12i. 제1항에 있어서, 상기 가이드 폴리뉴클레오티드 또는 상기 가이드 폴리뉴클레오티드를 암호화하는 핵산은 서열번호 2 내지 6 중 어느 하나로 표시되는 뉴클레오티드 서열을 포함하는 것인, 조성물.The composition according to claim 1, wherein the guide polynucleotide or the nucleic acid encoding the guide polynucleotide comprises a nucleotide sequence represented by any one of SEQ ID NOs: 2 to 6. 제1 항의 조성물을 포함하는 EGFR 돌연변이를 검출하기 위한 키트. A kit for detecting an EGFR mutation comprising the composition of claim 1. 제9항에 있어서, 핵산 증폭 성분을 더 포함하는 것인 키트.10. The kit according to claim 9, further comprising a nucleic acid amplification component. 시료를 제1 항의 조성물과 접촉시키는 단계; 및 contacting a sample with the composition of claim 1; and CRISPR/Cas 이펙터 단백질에 의한 상기 단일 가닥의 검출 핵산의 절단에 의해 생성된 검출 가능한 신호를 측정하여, 상기 시료 내 표적 서열을 검출하는 단계를 포함하는, EGFR 돌연변이를 검출하는 방법. A method for detecting an EGFR mutation comprising detecting a target sequence in the sample by measuring a detectable signal generated by cleavage of the single-stranded detection nucleic acid by the CRISPR/Cas effector protein. 제11항에 있어서, 상기 시료는 cfDNA(cell free DNA)인 것인, EGFR 돌연변이를 검출하는 방법. The method of claim 11, wherein the sample is cell free DNA (cfDNA). 제11항에 있어서, 상기 시료 내 존재하는 상기 표적 서열의 양을 결정하는 단계를 포함하는, EGFR 돌연변이를 검출하는 방법.The method of claim 11 , comprising determining the amount of the target sequence present in the sample. 제13항에 있어서, 상기 결정하는 단계는, 상기 검출 가능한 신호를 측정하여 시험 측정치를 생성하는 단계; 기준 샘플에 의해 생성된 검출 가능한 신호를 측정하여 기준 측정치를 생성하는 단계; 및 상기 시험 측정치를 상기 기준 측정치와 비교하여 상기 시료 내 존재하는 표적 서열의 양을 결정하는 단계를 포함하는 것인, EGFR 돌연변이를 검출하는 방법.14. The method of claim 13, wherein the determining step comprises: measuring the detectable signal to generate a test measurement; measuring a detectable signal produced by the reference sample to produce a reference measurement; and comparing the test measurement to the reference measurement to determine the amount of the target sequence present in the sample. 제11항에 있어서, 상기 CRISPR/Cas 이펙터 단백질은 Cas12a, mgCas12a, Cas12b, Cas12c, Cas12d, Cas12e, Cas12g, Cas12h, 및 Cas12i로 이루어진 군으로부터 선택된 어느 하나인 이펙터 단백질인 것인, EGFR 돌연변이를 검출하는 방법.The method of claim 11, wherein the CRISPR / Cas effector protein is any one effector protein selected from the group consisting of Cas12a, mgCas12a, Cas12b, Cas12c, Cas12d, Cas12e, Cas12g, Cas12h, and Cas12i, detecting EGFR mutations method. 제11항에 있어서, 상기 검출 가능한 신호를 측정하는 단계는 금 나노입자 기반 검출, 형광 편광, 콜로이드 상 전이/분산, 전기 화학적 검출 및 반도체-기반 센싱 중 하나 이상을 포함하는 것인, EGFR 돌연변이를 검출하는 방법.12. The method of claim 11, wherein measuring the detectable signal comprises one or more of gold nanoparticle-based detection, fluorescence polarization, colloidal phase transition/dispersion, electrochemical detection, and semiconductor-based sensing, EGFR mutation. How to detect. 제11항에 있어서, 상기 검출 핵산은 형광-방출 염료쌍을 포함하는 것인, EGFR 돌연변이를 검출하는 방법. The method of claim 11, wherein the detection nucleic acid comprises a fluorescence-emitting dye pair. 제17항에 있어서, 상기 형광-방출 염료쌍은 상기 검출 핵산의 절단 전에 검출 가능한 신호의 양을 생성하고, 검출 가능한 신호의 상기 양은 상기 검출 핵산의 절단 후에 감소되는 것인, EGFR 돌연변이를 검출하는 방법.18. The method of claim 17, wherein the fluorescence-emitting dye pair generates an amount of detectable signal before cleavage of the detection nucleic acid, and wherein the amount of detectable signal is reduced after cleavage of the detection nucleic acid. method. 서열번호 2 내지 6 중 어느 하나를 포함하는 폴리뉴클레오티드 또는 서열번호 2 내지 6 중 어느 하나의 폴리뉴클레오티드와 적어도 95% 상동성을 갖는 폴리뉴클레오티드를 포함하는 가이드 폴리뉴클레오티드.A guide polynucleotide comprising a polynucleotide comprising any one of SEQ ID NOs: 2 to 6 or a polynucleotide having at least 95% homology to a polynucleotide of any one of SEQ ID NOs: 2 to 6. 시료를 제1 항의 조성물과 접촉시키는 단계; 및 contacting a sample with the composition of claim 1; and CRISPR/Cas 이펙터 단백질에 의한 상기 단일 가닥의 검출 핵산의 절단에 의해 생성된 검출 가능한 신호를 측정하여, 상기 시료 내 표적 서열을 검출하는 단계를 포함하는, 암을 진단하는 방법 또는 암 진단에 관한 정보를 제공하는 방법.A method for diagnosing cancer or information about cancer diagnosis, comprising detecting a target sequence in the sample by measuring a detectable signal generated by cleavage of the single-stranded detection nucleic acid by a CRISPR/Cas effector protein. How to provide.
PCT/KR2023/001357 2022-01-28 2023-01-30 Composition and method for detecting nucleic acid based on fluorescence signal capable of specifically detecting egfr mutation Ceased WO2023146370A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2022-0013526 2022-01-28
KR20220013526 2022-01-28

Publications (1)

Publication Number Publication Date
WO2023146370A1 true WO2023146370A1 (en) 2023-08-03

Family

ID=87472326

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/001357 Ceased WO2023146370A1 (en) 2022-01-28 2023-01-30 Composition and method for detecting nucleic acid based on fluorescence signal capable of specifically detecting egfr mutation

Country Status (2)

Country Link
KR (1) KR20230117058A (en)
WO (1) WO2023146370A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190095160A (en) * 2018-02-06 2019-08-14 주식회사 진씨커 Kit for isolation of mutant cell free DNA and method for isolation of mutant cell free DNA using the same
KR20200074102A (en) * 2017-10-04 2020-06-24 더 브로드 인스티튜트, 인코퍼레이티드 Diagnosis method based on CRISPR effector system

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20200074102A (en) * 2017-10-04 2020-06-24 더 브로드 인스티튜트, 인코퍼레이티드 Diagnosis method based on CRISPR effector system
KR20190095160A (en) * 2018-02-06 2019-08-14 주식회사 진씨커 Kit for isolation of mutant cell free DNA and method for isolation of mutant cell free DNA using the same

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
LIU YIN, CHEN YANLING, DANG LU, LIU YIXIN, HUANG SHISHENG, WU SANYUN, MA PEIXIANG, JIANG HONGQIANG, LI YI, PAN YUNBAO, WEI YONGCHA: "EasyCatch, a convenient, sensitive and specific CRISPR detection system for cancer gene mutations", MOLECULAR CANCER, vol. 20, no. 1, 1 December 2021 (2021-12-01), XP093067019, DOI: 10.1186/s12943-021-01456-x *
TSOU JEN-HUI, LENG QIXIN, JIANG FENG: "A CRISPR Test for Rapidly and Sensitively Detecting Circulating EGFR Mutations", DIAGNOSTICS, MDPI, CH, vol. 10, no. 2, 1 February 2020 (2020-02-01), CH , pages 114, XP055938699, ISSN: 2075-4418, DOI: 10.3390/diagnostics10020114 *
WANG L I, JIANG JING, LI XIANGNING, LI KAI, HE RONGZHANG, LI JIA, DUAN LILI, LUO WEIHAO, HU ZHENG, LUO DIXIAN: "Improved EGFR mutation detection sensitivity after enrichment by Cas9/sgRNA digestion and PCR amplification", ACTA BIOCHIMICA BIOPHYSICA SINICA, BLACKWELL PUBLISHING, INC., MALDEN, MA, US, vol. 52, no. 12, 1 November 2020 (2020-11-01), US , pages 1316 - 1324, XP093080500, ISSN: 1672-9145, DOI: 10.1093/abbs/gmaa123 *

Also Published As

Publication number Publication date
KR20230117058A (en) 2023-08-07

Similar Documents

Publication Publication Date Title
US6270967B1 (en) Method for detecting a nucleic acid base sequence
ES2961374T3 (en) Amplification with primers of limited nucleotide composition
US9340832B2 (en) Methods for enriching a variant nucleic acid from a nucleic acid population in a sample
US8679788B2 (en) Methods for the detection of nucleic acid differences
US7888075B2 (en) Detection of methicillin-resistant and methicillin-sensitive Staphylococcus aureus in biological samples
JP2020078324A (en) Detection of methicillin-resistant Staphylococcus aureus in biological samples
WO2010135917A1 (en) Method for detecting variations in nucleic acid sequences
CA2835283C (en) Detection of methicillin-resistant staphylococcus aureus
Lorente et al. Detection of methylation in promoter sequences by melting curve analysis-based semiquantitative real time PCR
JP6876437B2 (en) DNA amplification method based on strand invasion
US20210285039A1 (en) Looped primer and loop-de-loop method for detecting target nucleic acid
WO2012009813A1 (en) Methods of detecting genetic polymorphisms using melting temperature of probe-amplicon complexes
US20220195499A1 (en) Quantification of ngs dna by adapter sequence
WO2023146366A1 (en) Crispr/cas system capable of specifically amplifying egfr-mutant dna, and use thereof
WO2017099445A1 (en) Genetic marker for discriminating and detecting causative bacteria of fish edwardsiellosis and streptococcosis, and method for discriminating and detecting causative bacteria using same
JP5239853B2 (en) Mutant gene detection method
US20190211377A1 (en) Cobra probes to detect a marker for epidemic ribotypes of clostridium difficile
CN102822354B (en) Assay for the detection of closely related serotypes of human papillomavirus (HPV)
US20140377762A1 (en) Method for enriching and detection of variant target nucleic acids
US20120142003A1 (en) Trichomonas vaginalis testing using tv5.8s as a target
WO2023146370A1 (en) Composition and method for detecting nucleic acid based on fluorescence signal capable of specifically detecting egfr mutation
WO2010121298A1 (en) Detection of staphylococcus aureus
WO2013105774A1 (en) Method for detecting c-met gene using cuttable probe
WO2022203297A1 (en) Target nucleic acid amplification method using guide probe and clamping probe and composition for amplifying target nucleic acid comprising same
WO2021045479A1 (en) Novel pna oligomer, use thereof for detecting dna methylation, and method for detecting dna methylation using same

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: 23747411

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: 23747411

Country of ref document: EP

Kind code of ref document: A1