[go: up one dir, main page]

WO2024096642A1 - Dna aptamer for diagnosis or treatment of colorectal cancer - Google Patents

Dna aptamer for diagnosis or treatment of colorectal cancer Download PDF

Info

Publication number
WO2024096642A1
WO2024096642A1 PCT/KR2023/017433 KR2023017433W WO2024096642A1 WO 2024096642 A1 WO2024096642 A1 WO 2024096642A1 KR 2023017433 W KR2023017433 W KR 2023017433W WO 2024096642 A1 WO2024096642 A1 WO 2024096642A1
Authority
WO
WIPO (PCT)
Prior art keywords
aptamer
sev
sevs
dna aptamer
dna
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/KR2023/017433
Other languages
French (fr)
Korean (ko)
Inventor
박기수
차병석
이은성
장영준
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.)
University Industry Cooperation Corporation of Konkuk University
Original Assignee
University Industry Cooperation Corporation of Konkuk University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from KR1020220144790A external-priority patent/KR20240066514A/en
Application filed by University Industry Cooperation Corporation of Konkuk University filed Critical University Industry Cooperation Corporation of Konkuk University
Priority claimed from KR1020230150082A external-priority patent/KR20250064751A/en
Publication of WO2024096642A1 publication Critical patent/WO2024096642A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • 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
    • C12N15/115Aptamers, i.e. nucleic acids binding a target molecule specifically and with high affinity without hybridising therewith ; Nucleic acids binding to non-nucleic acids, e.g. aptamers
    • 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

Definitions

  • the present invention is a technology related to a DNA aptamer for colon cancer diagnosis and treatment.
  • colon cancer can be diagnosed with high affinity and specificity, and it specifically binds to colon cancer-derived sEV to prevent sEV-mediated cancer metastasis. It is a technology that has been confirmed to have therapeutic potential as it has the potential to suppress it.
  • the present invention also provides a technology related to a DNA aptamer that can diagnose colon cancer with high heterogeneity characteristics, and a technology that confirms the possibility of colon cancer diagnosis with high affinity and specificity using an aptamer.
  • This invention is as part of the individual basic research of the Ministry of Science and ICT (Ministry of Science and ICT) [Project identification number: 1711157003, task number: 2020R1C1C1012275, research project name: Exosome SELEX technology (E-SELEX) and cancer diagnosis bio using the same] It is derived from research conducted.
  • Colorectal cancer a threat to global health care, is the third most diagnosed cancer and the second most common cause of cancer death worldwide. In 2020, colorectal cancer accounted for 10% of cancer diagnoses and 9.4% of cancer deaths worldwide. The neoplastic progression of colorectal cancer is initiated by a combination of factors, including genetic mutations, epigenetic modifications, and environmental factors, ultimately promoting recurrence and metastasis in various organs, especially the liver. Colon cancer is known to be a carcinoma that is particularly difficult to diagnose and treat because it is characterized by high tumor heterogeneity.
  • small extracellular vesicles also known as exosomes
  • sEVs are nano-sized lipid membrane-enclosed endoplasmic reticulum (50-200 nm in diameter) that are secreted extracellularly by all cell types.
  • sEVs are stable in various body fluids such as blood, urine, sweat, and saliva.
  • sEVs transport active biomolecules such as DNA, RNA, and proteins to locally and systematically mediate intercellular signaling between cells of origin and recipient cells.
  • sEVs derived from tumor cells can act as regulators of cancer development, progression, invasion, and metastasis. More and more studies have demonstrated the potential applications of CRC-derived sEVs.
  • sEVs microRNAs
  • lncRNAs long non-coding RNAs
  • mRNAs messenger RNAs
  • candidate sEVs biomarkers for CRC prognosis including metastasis, chemoresistance, and recurrence, were identified. Since sEVs circulating in human body fluids are considered mini versions of parent cells, non-invasive diagnosis of colorectal cancer is possible. Therefore, there is increasing research interest in using sEVs as a promising tool for diagnostic and therapeutic applications. In particular, it is very important to distinguish between colon cancer-derived sEVs and normal human serum-derived sEVs, whose origin is difficult to accurately determine.
  • aptamers which are single-stranded DNA or RNA that can bind to a specific target, are widely used in diagnostic and therapeutic development and biotechnology due to their outstanding advantages such as high stability, small size, ease of synthesis and modification, low immunogenicity, and high binding affinity and specificity. It is receiving great attention when it comes to marker discovery.
  • Aptamers are typically screened through an iterative in vitro selection process called Systematic Evolution of Ligands by EXponential Enrichment (SELEX).
  • SELEX Systematic Evolution of Ligands by EXponential Enrichment
  • the present inventors found a stable DNA aptamer that can diagnose colon cancer through sEVs, revealed its potential to treat colon cancer by inhibiting sEVs-mediated cancer metastasis, and demonstrated various properties that can be used to diagnose colon cancer.
  • the present invention was completed by finding a DNA aptamer with high binding to colon cancer sEVs and using it to develop a highly sensitive biosensor.
  • the purpose of the present invention is to provide at least one DNA aptamer for colon cancer diagnosis selected from the group consisting of base sequences of SEQ ID NOs: 4 to 17.
  • Another object of the present invention is to provide a composition for diagnosing colon cancer, a kit, and a colon cancer-specific drug delivery composition containing the aptamer.
  • Another object of the present invention is to provide a method for producing the aptamer, a method for providing information for colon cancer diagnosis using the aptamer, and a method for detecting surface biomarkers specific to colon cancer sEVs.
  • Another object of the present invention is to provide at least one DNA aptamer for colon cancer diagnosis selected from the group consisting of base sequences of SEQ ID NOs: 18 to 35.
  • Another object of the present invention is to provide a composition for diagnosing colon cancer, a kit, and a colon cancer-specific drug delivery composition containing the aptamer.
  • Another object of the present invention is to provide a method for producing the aptamer, a method for providing information for colon cancer diagnosis using the aptamer, and a method for detecting surface biomarkers specific to colon cancer sEVs.
  • the present inventors used colon cancer sEV SELEX; E-SELEX was built. After proceeding with a total of three E-SELEX loops, where each loop consists of four consecutive positive selections and one counter selection, the top 10 aptamer candidates are selected and subjected to enzyme-linked oligonucleotide assay (ELONA). ) method was used to evaluate binding affinity. Finally, the best aptamer was obtained through post-SELEX optimization, which resulted in a K d value of 3.41 nM and a detection limit of 1.0 It was confirmed that it had sufficient sensitivity for detection.
  • E-SELEX enzyme-linked oligonucleotide assay
  • sEVs derived from healthy serum and sEVs from normal cells, and sEVs derived from other cancer cells
  • the selected optimal aptamers were further investigated to predict biomarkers of sEVs derived from colon cancer cells, and were demonstrated to inhibit cell migration and proliferation by regulating sEV-mediated intercellular signaling.
  • TEV-SELEX Colorectal Cancer
  • SW620, LS 174T, HT29 colon cancer cells
  • TEV-SELEX Colorectal Cancer
  • SW620, LS 174T, HT29 three types of colon cancer cells
  • HT29 colon cancer cells
  • TEV-SELEX was constructed with a total of eight loops, with each loop consisting of three consecutive positive selections and one counter selection. Based on the NGS results, the top 10 aptamer candidates by frequency (%) were selected and enzyme-linked. Binding affinity was evaluated using an oligonucleotide assay (Enzyme-linked oligonucleotide assay, ELONA) method.
  • ELONA Endome-linked oligonucleotide assay
  • the present invention provides a DNA aptamer for diagnosing or treating colon cancer selected from the group consisting of the base sequences of SEQ ID NOs: 4 to 17, and one or more colon cancers selected from the group consisting of the base sequences of SEQ ID NOs: 18 to 35 Provides DNA aptamers for cancer diagnosis.
  • the present invention relates to the treatment or diagnosis of colon cancer of a DNA aptamer containing at least one sequence selected from the group consisting of the base sequences of SEQ ID NOs: 4 to 17, and the use of a DNA aptamer selected from the group consisting of the base sequences of SEQ ID NOs: 18 to 35.
  • a DNA aptamer containing at least one sequence is provided for use in colon cancer diagnosis.
  • colon cancer is used with the same meaning as “colorectal cancer,” “Colorectal cancer,” and “CRC.”
  • DNA aptamer refers to a single-stranded oligonucleotide that has the characteristic of binding to a target with high affinity and specificity and each has a unique three-dimensional structure. Through repeated in vitro selection and enrichment processes, it is possible to select DNA molecules that specifically bind to a specific target, that is, DNA aptamers, from a DNA aptamer library.
  • Diagnosis means confirming the presence or characteristics of a pathological condition. Diagnosis in the present invention is to confirm the presence or occurrence of colon cancer using a DNA aptamer.
  • treatment means any beneficial action, such as improving the symptoms or suppressing the progression of colon cancer that has already been caused by using the DNA aptamer of the present invention.
  • it may include a DNA aptamer having 90% or more sequence homology to the nucleotide sequences of SEQ ID NOs. 4 to 35, and the term “base sequence having 90% or more sequence homology” ranges from 1 to 10. It refers to a nucleotide sequence that shows similar cancer-specific binding ability by adding, deleting, or substituting 90% or more but less than 100% of the sequences with nucleotides added, deleted, or substituted.
  • the aptamer can specifically bind to sEVs derived from colon cancer cells.
  • sEVs were isolated from positive and counter targets, and when the DNA aptamer of the present invention was used, it did not have binding specificity to sEVs derived from normal colon cells, human breast cancer cells, or human glioblastoma. , it was confirmed that it specifically bound only to sEV derived from colon cancer cells (Figure 8).
  • the aptamer of the present invention may further include one or more selected from the group consisting of forward primer and reverse primer sequences.
  • a "primer” is a nucleic acid sequence with a short free 3' terminal hydroxyl group that can form a base pair with a complementary template and serves as a starting point for copying the template. It refers to a short nucleic acid sequence that performs a function.
  • colon cancer can be diagnosed by performing PCR amplification using the forward and reverse primers of the DNA aptamer described above. PCR conditions and lengths of forward and reverse primers can be modified based on those known in the art.
  • the sequence of the forward primer may consist of the base sequence of SEQ ID NO: 2
  • the sequence of the reverse primer may consist of the base sequence of SEQ ID NO: 3, but are not limited thereto.
  • the primer region was used for PCR amplification by including a forward primer in the 5' portion of the aptamer sequence and a reverse primer region in the 3' portion of the aptamer sequence in an ssDNA library during the E-SELEX or TEV-SELEX process. It is obvious to those skilled in the art that primer regions can be combined at the 5' and 3' portions for PCR amplification.
  • the DNA aptamer may consist of the base sequence of SEQ ID NO: 13.
  • the top 10 candidate aptamers by frequency (%) after SELEX were selected, and aptamers with binding affinity and specificity were selected.
  • CCE-10 had the highest affinity for sEV.
  • the DNA aptamer consisting of the base sequence of SEQ ID NO: 13 may be additionally linked to one or more of the forward primer base sequence and the reverse primer base sequence.
  • the DNA aptamer consisting of the base sequence of sequence number 15 with the forward primer removed showed the highest affinity. Therefore, the DNA aptamer may be composed of the base sequence of sequence number 15.
  • the binding dissociation constant may be 3 to 4 nM, preferably 3.2 to 3.6.
  • the DNA aptamer may consist of the base sequence of SEQ ID NO: 32.
  • the top 10 candidate aptamers by frequency (%) after SELEX were selected, and aptamers with high binding affinity and specificity were selected.
  • T6 had the highest affinity for sEVs.
  • optimization of the T6 aptamer sequence was performed. Therefore, as shown in [Table 8], the T6F T6R T6FR T6RA T6RB T6RB1 T6RB2 T6RB3 sequence was used.
  • the present invention also provides a composition for diagnosing or treating colon cancer containing the aptamer above.
  • the composition may further include physiologically acceptable excipients, carriers, or additives, including starch, gelatinized starch, microcrystalline cellulose, lactose, povidone, colloidal silicon dioxide, calcium hydrogen phosphate, Lactose, mannitol, taffy, gum arabic, pregelatinized starch, corn starch, powdered cellulose, hydroxypropyl cellulose, Opadry, sodium starch glycolate, carnauba lead, synthetic aluminum silicate, stearic acid, magnesium stearate, aluminum stearate, calcium stearate. , white sugar, dextrose, sorbitol, and talc may be used, but are not limited thereto.
  • physiologically acceptable excipients, carriers, or additives including starch, gelatinized starch, microcrystalline cellulose, lactose, povidone, colloidal silicon dioxide, calcium hydrogen phosphate, Lactose, mannitol, taffy, gum arabic, pregelatinized starch, corn star
  • composition containing the DNA aptamer when using a composition containing the DNA aptamer, it hardly binds to normal serum, normal colon cell lines, or other human cancer cell lines other than colon cancer cell lines, so it can be used to specifically target (various) colon cancer cell lines. Detection was confirmed.
  • the present invention also provides a kit for diagnosing or treating colon cancer, comprising the composition.
  • a colon cancer diagnosis kit containing an aptamer that specifically binds to colon cancer-derived sEV.
  • This colon cancer diagnostic kit may include a buffer solution and containers for detection and analysis as needed, such as bottles, tubs, sachets, envelopes, tubes, and ampoules. They may be formed in part or entirely from plastic, glass, paper, foil, wax, etc.
  • the container may be equipped with a completely or partially removable closure that may initially be part of the container or may be attached to the container by mechanical, adhesive, or other means.
  • the container may also be equipped with a stopper, allowing access to the contents by means of a needle.
  • the kit may include an external package, and the external package may include instructions for use of the components.
  • the kit may be a diagnostic sensor, RT-PCR kit, competitive RT-PCR kit, real-time RT-PCR kit, DNA chip kit, and protein chip kit.
  • the kit of the present invention may include a composition, solution, or device containing not only primers and probes that recognize the DNA aptamer, but also one or more other components suitable for the analysis method.
  • the DNA aptamer that specifically binds to the colon cancer-derived sEV may be used on conventional supports such as beads, particles, dipsticks, fibers, filters, membranes, and glass slides, and solid supports such as silane or silicate supports. By being fixed to and provided as a detection sensor, it can be used for colon cancer diagnosis.
  • the present invention may be a sensor for colon cancer diagnosis in which a DNA aptamer that specifically binds to the colon cancer-derived sEV is fixed.
  • the solid support includes at least one substantially rigid surface, on which the DNA aptamers can be immovably fixed.
  • the DNA aptamer can be immobilized by any conventional chemical coupling method.
  • biotin is bound to the end of the DNA aptamer to form a complex
  • streptavidin or avidin is immobilized on the surface of a substrate such as the chip, so that the interaction between the biotin and streptavidin or avidin immobilized on the substrate surface
  • the DNA aptamer can be immobilized on the surface of the substrate.
  • kits can be applied to the kit as a signal generation method to detect DNA aptamers bound to colon cancer sEVs. Examples include colorimetry, fluorescence, and electrochemistry, but are not limited thereto, and any method that can be implemented through a labeling method suitable for each signal generation can be used without limitation.
  • the DNA aptamer can be administered to the subject in various forms according to the selected administration route as understood by those skilled in the art through the above-described itself, composition, or kit. there is.
  • it may be administered by topical, enteral or parenteral application.
  • Topical applications include, but are not limited to, epidermis, inhalation, enemas, eye drops, ear drops, and application through mucous membranes within the body.
  • Enteral applications may include oral administration, rectal administration, vaginal administration, and gastric feeding tubes.
  • Parenteral administration is intravenous, intraarterial, intracapsular, intraorbital, intracardiac, intradermal, transtracheal, subcutaneous, intraarticular, subcapsular, subarachnoid, intrathecal, epidural, intrasternal, intraperitoneal, subcutaneous, Methods of administration may include intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal, and topical.
  • the DNA aptamer itself, the composition, or the kit described above may be formulated in an appropriate form depending on the route of administration, etc. When formulated, it may be prepared using diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrants, and surfactants, but is not limited thereto.
  • the present invention also provides a method for producing the DNA aptamer.
  • the method is
  • the aptamer in the method of producing a DNA aptamer, can specifically bind to sEV derived from colon cancer cells, but this description is omitted to avoid duplication as described above in the description of the DNA aptamer.
  • the present invention also provides a method of providing information for diagnosis or treatment of colon cancer using the DNA aptamer.
  • the present invention provides a method for detecting colorectal cancer sEV-specific surface biomarkers using the aptamer.
  • the method may include contacting the DNA aptamer with samples such as colon tissue, colon cells, blood, serum, plasma, saliva, sputum, and urine.
  • samples such as colon tissue, colon cells, blood, serum, plasma, saliva, sputum, and urine.
  • the sample is isolated from a mammal, preferably from the human body, and may be a sample that can be obtained with minimal invasiveness, secreted body fluid, or in vitro cell culture component sample, but preferably may be sEV derived from colon cancer cells, and may be a colon cancer marker. It is obvious that the sample is not limited to the above as long as it is a sample that may be included.
  • Colon cancer biomarkers can be separated through analysis of substances bound to the DNA aptamer in the sample, and the present invention can be used to detect colon cancer cell-specific surface biomarkers using the DNA aptamer. .
  • the present invention also provides a colon cancer-specific drug delivery composition containing the aptamer.
  • aptamers can be used to treat colon cancer by specifically binding to sEVs derived from colon cancer cells and inhibiting the progression of cancer through sEVs. Therefore, the present invention Since the aptamer will specifically bind to sEV derived from colon cancer cells and inhibit the mechanism of colon cancer progression, it is known to those skilled in the art that a composition containing this can be provided as a composition for treating colon cancer. It is self-explanatory.
  • aptamers to the surface of liposomes or nanoparticles, anticancer drugs, toxins, cancer growth inhibitory genes, siRNA (small interfering RNA), etc. loaded inside liposomes or nanoparticles can be selectively delivered to sEVs derived from colon cancer cells.
  • the aptamer according to the present invention includes known colon cancer-specific drugs, toxins and anticancer agents that induce cancer cell death, or Herpes simplex virusthymidine kinase (HSV-TK), cytosine deaminase (CD), etc.
  • HSV-TK Herpes simplex virusthymidine kinase
  • CD cytosine deaminase
  • siRNA small interfering RNA
  • the present invention provides the DNA aptamer It may be provided in the form of a colon cancer-specific drug delivery composition containing.
  • an aptamer that specifically binds only to colon cancer cell-derived cells can be used for diagnosis and treatment of colon cancer.
  • Figure 1 shows characterization of isolated sEVs.
  • Figure 1 (A) shows the NTA results for SW620 sEV (insert: cryo-TEM image), and Figure 1 (B) shows the NTA results for HS (human serum) sEV. Scale bar of cryo-TEM image: 100 nm.
  • Figure 1 (C) shows the results of Western blot analysis of CD63, CD9, Hsp90 ⁇ , and calnexin expression.
  • Figure 2 shows the workflow of (A) E-SELEX and (B) E-SELEX, respectively.
  • Figure 3 shows an agarose (2.5%) gel image for comparing symmetric and asymmetric PCR results.
  • M 25/100 bp DNA size marker; 1 and 2: w/o and w/ssDNA templates in symmetric PCR; Lanes 3 & 4: w/o and w/ssDNA templates from asymmetric PCR; Lane 5: ssDNA template (comparison)
  • Figure 4 shows the E-SELEX monitoring results using qPCR.
  • A Amplification curve.
  • B Melting curve analysis.
  • Figure 5 shows the ratio of F20th 20th cycle fluorescence intensity to Fmax maximum fluorescence intensity ( F20th / Fmax ) to estimate the degree of homogeneity.
  • Figure 6 shows the results of aptamer evaluation using the ELONA method.
  • Figure 6 (A) shows the binding affinity of the top 10 candidate aptamers
  • Figure 6 (B) shows the binding affinity of CCE-10 with and without the primer region.
  • the concentration of aptamer used in the binding affinity analysis was 250 nM.
  • Figure 7 shows the dissociation constant ( K d ) curves of CCE-10F (A) and CCE-10FR (B).
  • Figure 8 shows the evaluation results of the optimized aptamer CCE-10F.
  • Figure 8 (A) Two-dimensional and three-dimensional shapes of the predicted secondary structure of the aptamer,
  • Figure 8 (B) Binding affinity comparison with that in any ssDNA library,
  • Figure 8 (D) This is the specificity evaluation result, and the concentration of the aptamer used in the evaluation was 10 nM.
  • Figure 8 (E) shows the stability analysis results of aptamer (Apt) and sEV-bound aptamer (Apt-Exosomes) in human serum, showing the band intensity (left) and each stability (right) of the aptamer at each time point. This is a representative gel image shown.
  • Figure 9 shows the results of biomarker topology analysis. I: intact sEV, II: proteinase K (proK) treated, III: trypsin treated sEV, IV: RIPA treated sEV.
  • Figure 10 shows the results of cytotoxicity analysis. This is the analysis result for LS 174T in Figure 10 (A) and HUVEC in Figure 10 (B).
  • Figure 11 shows the results of sEV absorption analysis by LS 174T.
  • Figure 11 shows (A) confocal microscopy image, (B) flow cytometry result; Representative histograms (left) and fluorescence quantification (right) are shown, scale bar of microscopy images: 10 ⁇ m.
  • Figure 12 shows the results confirming the effect of sEV and Apt-sEV on the migration and invasion of LS 174T.
  • A representative images of wound healing assay at 0 h and 24 h (scale bar: 200 ⁇ m)
  • B quantitative analysis of gap closure
  • C representative image of transwell migration assay at 48 h (scale bar: 100 ⁇ m)
  • D quantitative analysis of migration area
  • E representative image of transwell invasion analysis at 48 hours (scale bar: 100 ⁇ m)
  • F quantitative analysis of invasion area.
  • Figure 13 shows the results confirming the effect of sEV and Apt-sEV on HUVEC angiogenesis.
  • Figure 13 shows (A) representative images of the tube formation assay at 4 h (scale bar: 100 ⁇ m), (B-C) quantitative analysis results of the relative number of junctions (B) and relative total branch length (C), respectively.
  • Figure 14 shows characterization of isolated sEVs.
  • Figure 14 (A) shows NTA results for SW620 sEV (insert: cryo-TEM image)
  • Figure 14 (B) shows NTA results for LS 174T sEV (insert: cryo-TEM image)
  • Figure 14 (C) shows the NTA results for HT29 sEVs (insert: cryo-TEM image)
  • Figure 14 (E) shows the results of Western blot analysis for the expression of CD63, Hsp90 ⁇ , and calnexin, and the scale bar of each cryo-TEM image is 100 nm.
  • Figure 15 shows the workflow of Colorectal Cancer Toggle sEV-SELEX (CRC TEV-SELEX) performed according to an embodiment of the present invention.
  • Figure 16 shows TEV-SELEX monitoring results.
  • Figure 16 (A) is an amplification curve
  • Figure 3 (B) is the ratio of F20th 20th cycle fluorescence intensity to Fmax maximum fluorescence intensity for estimating the degree of homogeneity of ssDNA from random library (Lib) and 1st to 8th loops. (F 20th /F max )
  • Figure 17 shows the binding affinity analysis results of the top 10 candidate aptamers (T1 to T10) selected according to the present invention.
  • Figure 18 shows the results of binding affinity analysis of aptamer candidates according to the present invention.
  • Figure 18 (A) shows the binding affinity of T6 and its derivative sequences (T6F, T6R, and T6FR), and
  • Figure 18 (B) to (D) shows the binding affinity of the positive target (SW620, LS 174T, The binding affinity is shown along with the ratio of T6 and its derived sequences to HT29).
  • Figure 19 shows the structural schematic diagram and binding affinity of the aptamer and its derived sequences according to the present invention.
  • Figures 19 (A) and (B) show the structure and binding affinity of T6R and its truncated versions of derived sequences (T6RA, T6RB), and
  • Figures 19 (C) and (D) show T6RB and its truncated versions.
  • the structure and binding affinity of the derived sequences (T6RB1, T6RB2, T6RB3) are shown.
  • Figure 20 shows the evaluation results of T6RB, the optimal aptamer.
  • Figure 20 (A) shows the T6RB dissociation constant ( K d ) curve and its value
  • Figure 20 (B) shows the circular dichroism (CD) spectrum of T6RB in the absence/presence of potassium ions (K + )
  • Figure 20 (C) shows the fluorescence intensity (FI) of NMM bound to T6RB in the absence/presence of K +
  • (D) in Figure 20 shows the results of the biomarker topology analysis.
  • Figure 21 shows parameter optimization results for TSA reaction.
  • Figure 21 (A) is aptamer concentration
  • Figure 21 (C) is H 2 O 2 concentration
  • Figure 21 (D) is TSA. Shows optimization results according to reaction time.
  • Figure 22 shows the evaluation results of T6RB aptamer.
  • ELONA and ELONA equipped with TSA are compared and analyzed.
  • Figure 23 shows the results confirming CRC sEV detection by TSA under optimized conditions.
  • Figure 23 (A) shows the sensitivity of SW620 sEV
  • Figure 23 (B) shows the sensitivity of LS 174T sEV
  • Figure 23 (C) shows the sensitivity of HT29 sEV
  • Figure 23 (D) shows CRC, HS and specificity of T6RB for sEVs derived from normal colon.
  • SW620 human colon cancer
  • LS 174T human colon cancer
  • CCD-18Co human normal colon
  • SKBR3 human breast cancer
  • U-87 MG human glioblastoma
  • cells were replaced with conditioned medium supplemented with 5% (v/v) sEV-depleted FBS (Gibco; Thermo Fisher Scientific) and 1% (v/v) penicillin-streptomycin and incubated for 48 h. Cultured in a 5% CO 2 incubator under humidified conditions at 37°C. The conditioned medium containing the secreted sEVs was recovered, and the sEVs were concentrated, isolated, and purified for further use.
  • sEV-depleted FBS Gibco; Thermo Fisher Scientific
  • penicillin-streptomycin penicillin-streptomycin
  • the conditioned medium was first subjected to a series of centrifugations at 300 xg for 5 min, 2,000 xg for 20 min, and 10,000 xg for 30 min to remove cells, cell debris, microvesicles, and apoptotic bodies, respectively. Afterwards, the supernatant was sequentially syringe filtered using filters with pore sizes of 0.45 and 0.22 ⁇ m (Sartorius, G ⁇ ttingen, Germany) to exclude extracellular vesicles larger than 200 nm in size. For concentration and purification, a tangential flow filtration system with a 300K membrane (Pall Corporation, New York, NY, USA) was used until the medium was concentrated to the desired volume.
  • sEVs were isolated from human serum (HS) (Sigma-Aldrich, St Louis, MO, USA) in the same manner as described above, except that they were diluted in 1 ⁇ PBS before serial centrifugation.
  • HS human serum
  • SW620 positive target
  • HS healthy human serum
  • NTA Nanoparticle Tracking Analysis
  • the concentration and size distribution of isolated sEVs were analyzed using a NanoSight NS300 instrument (Malvern Panalytical, Malvern, UK).
  • the laser was set at 532 nm and three 60-second videos were recorded for each sample at 25 frames per second using an sCMOS camera.
  • the camera level was set to 14 with well-adjusted camera focus and the detection threshold was set to 5. Brownian motion of particles was evaluated using NanoSight software (Malvern Panalytical). NTA settings were kept consistent between samples and samples were diluted in PBS for optimal measurements.
  • Cryo-TEM was used to determine the size of SW620 cells and HS-derived sEVs in TEM images. Before sample application, the grid was glow discharged to make it hydrophilic. Each sEV sample (3 ⁇ L) was placed on a perforated carbon-coated copper grid (200 mesh; Quantifoil, profl ⁇ bichau, Germany) and vitrified using a Thermo Scientific Vitrobot (Thermo Fisher Scientific) by placing it in liquid ethane and maintaining the temperature. liquid nitrogen. After vitrification, the samples were stored in liquid nitrogen. Before imaging, the stored samples were transferred to a cryoholder where the temperature was continuously maintained at approximately -180°C using liquid nitrogen. Cryo-TEM images were obtained at 25 kV using a JEM-2100PLUS electron microscope (JEOL, Tokyo, Japan) coupled with a CMOS camera.
  • JEOL JEM-2100PLUS electron microscope
  • Equal volumes of SW620 cells and HS-derived sEVs were concentrated using an Amicon Ultra-0.5 device with a 30K filter (Merck Millipore, Burlington, MA, USA) according to the manufacturer's instructions. Each sample was supplemented with 5x SDS-PAGE loading buffer (Biosesang, Seongnam, Republic of Korea), heated at 95°C for 10 min, and incubated at 300 V for 10 min for 20 min using the Mini-Protean tetra system (Bio-Rad Laboratories). % TGX stain-free protein gel (Bio-Rad Laboratories, Hercules, CA, USA).
  • an important step in E-SELEX is to efficiently separate the bound and unbound aptamer pools.
  • immunoplates as a facile targeted sEV immobilization platform that can not only maintain the integrity of biologically active sEVs but also facilitate the selection of high-quality aptamers with diagnostic value.
  • oligonucleotides used were synthesized by Integrated DNA Technologies (IDT; Coralville, IA, USA). The types of DNA included in the DNA library and the types of forward and reverse primers used are shown in Table 1 below.
  • a random library was designed with a central random region of 40 nucleotides (N40) between the primer regions at both ends used in the PCR amplification step.
  • SW620 cell-derived sEVs and normal human serum (HS)-derived sEVs were used as positive and counter targets, respectively, and selection was performed using maxi-binding immunoplates throughout the E-SELEX process.
  • a nascent single-stranded DNA (ssDNA) library 100 nM ) was denatured at 95°C for 5 minutes and rapidly cooled on ice for 10 minutes.
  • Negative selection was performed first to remove ssDNA likely to bind to BSA and immune plates. 1 g/L of BSA solution was inoculated into the immune plate and incubated at 37°C for 2 hours. After washing three times with washing buffer (WB; DPBS with 5 mM MgCl 2 ) on a shaker (500 rpm) for 30 seconds each, the prepared ssDNA library was added and incubated for 1 hour. Afterwards, the supernatant containing unbound ssDNA was recovered and concentrated using Nucleospin Gel and PCR clean-up (Macherey-Nagel, Duren, Germany) according to the manufacturer's instructions.
  • WB washing buffer
  • DPBS DPBS with 5 mM MgCl 2
  • the recovered ssDNA was then amplified using symmetric PCR (Asy-PCR) with different ratios of primer concentrations. Briefly, mixture buffer for Asy-PCR reactions containing 0.2 mM dNTPs, ssDNA template recovered from each selection, high-fidelity nPfu-forte DNA polymerase, and a forward-to-reverse primer ratio of 20:1 in 1x nPfu forte (Enzynomics , Daejeon, Republic of Korea).
  • the thermal cycling protocol was set up as follows: initial denaturation at 95°C for 3 min, followed by 20 cycles of 10 s at 95°C (denaturation), 1 min at 68°C (annealing and elongation), and a final extension of 5 min at 68°C. .
  • the Asy-PCR product was separated on a 2.5% agarose gel, and the ssDNA band was extracted and purified using Nucleospin gel and PCR clean-up.
  • SW620 sEV (20 ⁇ g/mL) were incubated in immunoplates at 37°C for 2 h and then washed (three times) by WB.
  • ssDNA from negative selection was added to the wells, then incubated at 37°C for 1 hour and washed (three times) with WB to remove unbound ssDNA.
  • samples from each well were suspended in BB and heated to 95°C for 10 min to recover bound ssDNA from sEVs and then processed as previously described.
  • Counter selection which relies on incubation of HS-derived sEVs with ssDNA pools, was also performed in the same manner as negative selection to eliminate ssDNA binding to normal sEVs.
  • stringent conditions were performed in continuous E-SELEX loops as described in [Table 2].
  • AC refers to each step in the E-SELEX loop cycle.
  • Negative selection in the E-SELEX procedure was performed to remove non-target single-stranded DNA (ssDNA) that binds nonspecifically to components of the buffer and/or the immune plate itself. Unbound ssDNA obtained from negative selection was enriched for further selection containing three E-SELEX loops, where each loop (denoted A-, B-, and C-loop) was used for four consecutive positive selections and one It included one counter-screening. Considering the high metastatic potential, SW620 cell-derived sEVs were selected as a positive selection target, and HS-derived sEVs were selected as a counter-selection target to establish a clinical environment for potential in vivo application. The loop was repeated three times for a total of 15 individual selection steps, during which the stringency conditions were gradually increased to improve the binding affinity and specificity of the aptamer (Table 2).
  • E-SELEX Another important consideration for the success of E-SELEX is monitoring the E-SELEX procedure to determine the number of loops performed.
  • qPCR can study DNA pools of diverse sequences and provides important information about the homogeneity of ssDNA recovered through E-SELEX in terms of ssDNA pool diversity.
  • the number of loops was selected by analyzing ssDNA obtained from each loop (A-C) by qPCR.
  • PCR was performed on a mixture containing the ssDNA template recovered from each selection and a forward-reverse primer ratio of 1:1 in 1X TOPreal qPCR premix (Enzynomics).
  • the thermal cycling protocol was set up as follows: initial denaturation at 95°C for 15 min, followed by 20 cycles of 10 s at 95°C (denaturation), 15 s at 60°C (annealing), and 15 s at 72°C (extension). This was followed by melting temperature (Tm) analysis while gradually increasing the temperature from 55°C to 95°C at an increment rate of 0.5°C.
  • the initial library of ssDNA is highly heterogeneous in sequence due to a random region of 40 nucleotides. However, heterogeneity will be reduced because an iterative process of selecting a pool of aptamers that specifically bind to the target will be continuously performed. This expected behavior was demonstrated through changes in amplification and melting curves ( Figure 4).
  • amplification of the random library red line; Lib
  • amplification curves were analyzed quantitatively by calculating the ratio of the fluorescence intensity in the last cycle ( 20th ) to the maximum fluorescence intensity in the final qPCR cycle.
  • a ratio higher than 0.95 was considered a plateau of amplification and was sufficient to ensure high homogeneity in ssDNA libraries.
  • the random ssDNA library (Lib) shows the lowest ratio, reflecting significant heterogeneity, but the ratio increases as the E-SELEX loop is continuously repeated.
  • the ratio of C-loop alone showed a value greater than 0.95, proving that C-loop reached a plateau in amplification due to the increase in ssDNA homogeneity.
  • the ssDNA obtained from the C-loop was analyzed by NGS, and the top 10 candidate aptamers were selected based on frequency (%) for further evaluation.
  • NGS Next-generation sequencing
  • the concentrated ssDNA pool was symmetrically PCR amplified with primers to generate dsDNA, resolved on an agarose gel (2.5%), and purified using Nucleospin Gel and PCR clean-up. Finally, the purified dsDNA was processed and the sequence of the candidate aptamer was analyzed through NGS (next-generation sequencing), which was performed by a sequencing company (Clinomics, Ulsan, Republic of Korea).
  • the forward primer of SEQ ID NO: 2 was included in the 5' part of the aptamer sequence
  • the reverse primer of SEQ ID NO: 3 was included in the 3' part of the ssDNA library [Table 3 ], the forward and reverse primer portions are not indicated.
  • CCE stands for colorectal cancer exosome aptamer.
  • Enzyme-linked oligonucleotide assay (ELONA)
  • the immune plate was further incubated with streptavidin-peroxidase polymer (Sigma-Aldrich; 1:1,000) diluted in 0.1% PBST for 1 hour at 37°C and then washed (three times) with 0.1% PBST.
  • streptavidin-peroxidase polymer Sigma-Aldrich; 1:1,000
  • 1x TMB substrate solution (Invitrogen, Carlsbad, CA, USA) was added to each well and incubated for 6 min in the dark and at room temperature.
  • the reaction was terminated by adding 0.5 M sulfuric acid, and the absorbance was immediately measured at 450 nm using a Spectramax iD5 multi-mode microplate reader (Molecular Devices, San Jose, CA, USA).
  • the normalized value ⁇ A was calculated based on the following equation.
  • At is the absorbance of the sample containing both sEV and aptamer
  • Ac is the absorbance of the control containing only sEV.
  • concentration of aptamer varies depending on the analysis method, and all experiments were repeated three times.
  • Primer regions were included in the ssDNA library for PCR amplification during the E-SELEX process. Because shorter length aptamers are synthesized at lower cost and are more suitable for aptamer-based biosensor development, we performed post-SELEX optimization of the CCE-10 aptamer by truncating the forward or reverse primer regions. CCE-10 aptamers lacking the forward, reverse, or both regions were designated CCE-10F, CCE-10R, and CCE-10FR, respectively.
  • the CCE-10 aptamer was indicated by SEQ ID NO: 13 in the above-mentioned Table 3, but in [Table 4] below, it was named CCE 10' including the forward and reverse primer portions, and was listed as SEQ ID NO: 14.
  • the binding affinity of cleaved CCE-10 aptamers (CCE-10F, CCE-10R, and CCE-10FR) and SW620 sEV (positive target) is similar to that of uncleaved CCE-10'.
  • the binding affinity was similar to that of the aptamer.
  • the binding affinity to HS sEV (opposite target) was significantly different. Specifically, aptamers without the forward primer region (CCE-10F and CCE-10FR) had further reduced binding affinity for HS sEVs, whereas aptamers without the reverse primer region (CCE-10R) had a reduced binding affinity. appeared to increase significantly.
  • CCE-10F The binding affinity of CCE-10F was quantitatively evaluated by estimating Kd. Specifically, CCE-10F was exposed to a certain number of SW620 and HS sEVs at various concentrations (0-100 nM) and then analyzed using the ELONA method described above in Example 3.
  • Figure 7 shows the dissociation constant ( K d ) curves of CCE-10F (A) and CCE-10FR (B).
  • K d dissociation constant
  • CCE-10F secondary structure The two-dimensional and three-dimensional structures of CCE-10F secondary structure were predicted and analyzed using OligoAnalyzer Tool (IDT) and Mol * 3D Viewer (RCSB PDB), respectively.
  • IDTT OligoAnalyzer Tool
  • RCSB PDB Mol * 3D Viewer
  • FIG 8 (A) shows that CCE-10F contains two different stem-loop motifs with a minimum Gibbs free energy of -2.39 kcal/mol at 37°C.
  • the affinity of CCE-10F was compared with that of a random library (Lib) and a random library without the forward primer region (Lib-F).
  • Lib random library
  • Lib-F the forward primer region
  • FIG. 8 (D) does not show binding to sEVs derived from CCD-18Co cells (normal colon), SKBR3 cells (human breast cancer), and U-87 MG cells (human glioblastoma), but binds to sEVs derived from SW620 cells. This indicates that CCE-10F has high binding specificity.
  • CCE-10F allows it to detect sEVs derived from CCD-18Co cells, SKBR3 cells, and U-87 MG cells, even though sEVs derived from CCD-18Co cells, SKBR3 cells, and U-87 MG cells were not included as targets of counterscreening. It was confirmed that the affinity of this aptamer for is low and is similar to the binding affinity for HS-derived sEV.
  • aptamer (Apt) alone or Aptamer-SW620 sEV were cultured in 80% human serum (Sigma-Aldrich) for 0–48 h. Samples recovered at each time interval were first treated with proK (200 ⁇ g/mL) at 37°C for 1 hour to remove serum or sEV-derived proteins and stored at -80°C until use. Next, each sample was mixed with Novex TBE-urea sample buffer (Invitrogen), heated at 95°C for 10 min, and dissolved in a urea-modified polyacrylamide gel (15%).
  • Figure 8 (E) shows gel images and stability (%) of aptamers cultured in various time frames from 0 to 48 hours.
  • Aptamer alone and Apt-sEV were stable for up to 24 hours, maintaining stability of 33% and 44%, respectively.
  • no significant differences were observed for up to 3 hours, after which Apt-sEV showed higher stability than the aptamer alone. This may be because the aptamer binds to sEVs, providing more resistance to nucleases in serum. Since the aptamer used in the serum stability analysis is unmodified, modification with functional groups such as phosphothioate bond and 2' fluoro-base increases the resistance of the aptamer to nucleases and improves stability without compromising binding ability. It is expected to be secured.
  • lysates of SW620-derived sEVs were prepared using RIPA lysis buffer supplemented with 1X Halt protease inhibitor cocktail (Thermo Fisher Scientific). sEVs were mixed with lysis buffer supplemented with protease inhibitors, incubated on ice for 20 min and sonicated in a water bath sonicator (SH-2140D, SAE HAN ULTRASONIC, Seoul, Republic of Korea) with 5 cycles of 30 s on/off at 40 kHz. Processed.
  • ProK proteinase K
  • SW620 cell-derived sEVs were treated with 20 ⁇ g/mL proK (Engenomics) for 1 h at 37°C with gentle vortexing every 20 min.
  • Proteinase activity was inhibited by addition of 5 mM phenylmethylsulfonyl fluoride (PMSF) for 10 min at room temperature.
  • PMSF phenylmethylsulfonyl fluoride
  • trypsin-EDTA 0.25% trypsin-EDTA (Welgene) was added to SW620 cell-derived sEVs and incubated at 37°C for 30 min. Trypsin activity was terminated by adding an equal volume of PBS containing 10% FBS. The final composition of each treatment was stored at -80°C until use.
  • the decomposing enzyme activities of proK and trypsin only act on the extravesicular membrane protein of the vesicle, and the intravesicular protein remains unaffected, making it possible to distinguish the location of the biomarker.
  • Using RIPA lysis buffer releases proteins within vesicles from the lumen of sEVs and membrane proteins from the lipid bilayer, exposing them in their original form.
  • Figure 9 shows the results of biomarker topology analysis.
  • I intact sEV
  • II proteinase K (proK) treated
  • III trypsin treated
  • IV RIPA treated sEV results.
  • Figure 9 (I-III) shows that CCE-10F exhibits negligible binding affinity to proK and trypsin-treated sEV.
  • CCE-10F showed comparable binding affinity to RIPA-lysed sEVs compared to intact sEVs ( Figure 9, I and IV).
  • LS 174T cells The viability of LS 174T cells was first assessed after treatment with SW620 cell-derived sEVs, CCE-10F aptamer-sEV complex (Apt-sEV), and CCE-10F aptamer alone (Apt).
  • LS 174T cells The viability of LS 174T cells was tested in the presence of SW620 and aptamer-SW620 sEVs using CCK-8 (Dojindo, Rockville, MD, USA) according to the manufacturer's instructions.
  • Cells (2 x 10 4 cells/well) were inoculated into a 96-well plate. After overnight culture, cells were treated with PBS, SW620, and Aptamer-SW620 sEV for 24 hours at 37°C.
  • the concentrations of sEV and aptamer used in this analysis were 1 x 10 10 particles/mL and 200 nM, respectively.
  • CCK-8 reagent was added to each well and incubated for an additional 2 hours at 37°C. Finally, the absorbance of the sample was measured at 450 nm using a microplate reader, and the sample treated with PBS was considered to have 100% cell viability.
  • sEVs (1 ⁇ 10 particles) were labeled with 100 ⁇ M CFSE (Abcam, Cambridge, UK) and incubated at 37°C on a shaker at 400 rpm for 2 h in the dark.
  • SEC size exclusion chromatography
  • Fractions were collected and stored at -80°C until use.
  • LS 174T cells (2 x 10 5 cells/well) were seeded in a 24-well plate and cultured at 37°C overnight. Next, the cells were replaced with fresh medium and treated with SW620 and Aptamer-SW620 sEV at 37°C for 24 hours.
  • the concentrations of sEV and aptamer used in this analysis were 1 x 10 9 particles and 200 nM, respectively.
  • concentrations of sEV and aptamer used in this analysis were 1 x 10 9 particles and 200 nM, respectively.
  • cells were washed with DPBS, fixed with 4% paraformaldehyde (PFA), and treated with DAPI for nuclear staining.
  • Confocal images were collected on an LSM 900 confocal microscope (Carl Zeiss, Thornwood, NY, USA) and analyzed with Zen software (Carl Zeiss).
  • Figure 11 (A) shows that both sEV and Apt-sEV labeled with CFSE were taken up by LS 174T cells, as evidenced by the green fluorescence signal. Uptake of sEV and Apt-sEV was also quantitatively assessed by flow cytometry. As shown in Figure 11B (left), significant changes in CFSE intensity were observed in LS 174T cells treated with sEV (orange) and Apt-sEV (green) compared to the control (red). Additionally, in Figure 11 (B) (right), the mean fluorescence intensity (MFI) of CFSE was significantly increased in sEV and Apt-sEV, demonstrating effective uptake of sEV by LS 174T cells.
  • MFI mean fluorescence intensity
  • SW620 CRC cell line is originally derived from metastatic lymph nodes and exhibits the ability to metastasize to the liver. Therefore, sEVs derived from SW620 cells may also have metastatic potential, which leads to the development of high invasion and migration abilities in the cells.
  • CCE-10F To explore potential biomarkers for SW620 sEVs specifically targeted by CCE-10F, we selected LS 174T, a CRC cell line with a low metastatic potential, as a model system and demonstrated that CCE-10F inhibits the metastasis of SW620 cell-derived sEVs. We investigated whether it affected the possibility.
  • Wound healing assays were performed to evaluate the effect of sEVs and aptamer-conjugated sEVs on the migration and proliferation abilities of LS 174T cells. Because sEVs can specifically convey cellular information to the microenvironment surrounding the tumor, we hypothesized that sEVs derived from SW620 cells, which have a high metastatic potential, induce cell proliferation, migration, and ultimately metastasis through the sEV-mediated signaling pathway.
  • LS 174T cells (7 ⁇ 10 5 cells/well) were seeded in 24-well plates and incubated overnight under standard culture conditions. Next, a sterile 200 ⁇ L pipette tip was used to induce wounding by scraping the cell monolayer across the center of the well. Cells were then gently washed twice with DPBS and replaced with conditioned DMEM supplemented with 1% FBS. After treating cells with SW620 or aptamer-SW620 sEV, wound gap images were taken at 0 and 24 hours using an Optinity inverted microscope (MDM Instruments, Seoul, Republic of Korea). Wound gap area was analyzed using ImageJ software (NIH, Bethesda, MD, USA). Wound healing (%) was determined using Equation 2 below.
  • Y wound healing (%)
  • a f is the wound gap area at 24 hours
  • a i is the wound gap area at 0 hours.
  • the concentrations of SW620 sEV and aptamer used in this analysis were 2 x 10 10 particles/ml and 200 nM, respectively.
  • Figure 12 (A) illustrates wound gap images of control cells and cells treated with sEV or Apt-sEV at initial (0 h; upper panel) and final (24 h; lower panel) incubation times.
  • Figure 12 (B) When quantitatively analyzed in Figure 12 (B), it was found that 32% of gap closure was induced in LS 174T treated with sEV alone.
  • SW620 sEV can induce LS 174T migration and proliferation, and that LS 174T, which has a low metastatic potential, can also acquire migration ability.
  • the gap closure of LS 174T treated with Apt-sEV was only 18%, which was significantly reduced compared to the group treated with sEV alone (p ⁇ 0.005).
  • the inhibition of migration and proliferation by Apt-sEVs can be inferred that the interaction between Apt and sEVs blocks the structure of protein-based biomarkers, ultimately preventing downstream cellular responses.
  • transwell migration assay without Matrigel
  • transwell invasion assay with Matrigel
  • Transwell cell migration and invasion assays were performed in 24-well plates using inserts with an 8 ⁇ m polyethylene terephthalate (PET) membrane with a diameter of 6.5 mm (Corning Inc., Corning, NY, USA).
  • PET membranes were precoated with Matrigel (Corning Inc.) prior to cell seeding.
  • LS 174T cells resuspended in serum-free DMEM were seeded in the upper chamber of the insert (5 x 10 4 cells/insert) and treated with PBS, SW620, or aptamer-SW620 sEV.
  • DMEM supplemented with 1% FBS was added to the lower chamber as a chemoattractant.
  • Figures 12 (C) and 12 (D) show that the vertical migration of LS 174T cells was significantly inhibited when Apt-sEV was treated compared to the sEV treatment group (p ⁇ 0.0001).
  • the same inhibitory effect of Apt-sEV was also observed in the transwell invasion assay ( Figures 11E and 11F), and the invasion area (%) between sEV and Apt-sEV treatment groups showed a significant difference (p ⁇ 0.0001).
  • Various biomarkers from previous literature have been identified to be involved in migration and invasion of CRC cell lines, some of which are also involved in angiogenesis, epithelial-mesenchymal transition (EMT) and ultimately tumor metastasis. Additionally, biomarkers of CRC cell-derived sEVs were confirmed to promote CRC liver metastasis.
  • HUVEC cells The viability of HUVEC cells was initially assessed after treatment with SW620 sEV, CCE-10F aptamer-sEV complex (Apt-sEV), and CCE-10F aptamer alone (Apt).
  • Figure 10 (B) confirmed that Apt-sEV (green) and Apt (blue) did not show cytotoxicity to HUVEC cells compared to the control group.
  • statistical analysis confirmed that the cell survival rates of Apt-sEV and Apt groups were not significantly different from those of the control group (one-way ANOVA, p > 0.05).
  • HUVEC cells treated with sEV showed a significant increase in cell survival rate compared to the control group (p ⁇ 0.0001). This suggests that SW620 sEV promotes the proliferation ability of HUVEC through the sEV-mediated signaling pathway.
  • Tube formation assays were performed in 96-well plates (SPL Life Sciences). Before cell seeding, 96-well plates were pre-coated with Matrigel (Corning Inc.) HUVECs suspended in CEFOgro-HUVEC medium containing 1% FBS and seeded on Matrigel-coated plates (1 x 10 cells/well). and treated with PBS, SW620, or Aptamer-SW620 sEV. After 4 hours of incubation at 37°C, cells were photographed using an Optinity inverted microscope (MDM Instruments) and analyzed with ImageJ software. It was defined as the relative number of junctions and total branch length of samples treated with sEV and Apt-sEV for samples treated with PBS. The concentrations of SW620 sEV and aptamer used in this analysis were 1 x 10 9 particles/mL and 200 nM, respectively.
  • Figure 13 (A) shows a representative image of tube formation, showing that only sEV treated cells developed tube formation, whereas Apt-sEV treated cells were not effective in forming tubes comparable to the control group. clearly shown.
  • Figures 13 (B) and 13 (C) the relative junction number and total branch length of the sEV group were both significantly greater than those of the Apt-sEV group (p ⁇ 0.0001).
  • the diagnostic validity of the aptamer is that it not only detects CRC cell-derived sEVs at a concentration ranging from 2.0 ⁇ 10 4 - 1.0 ⁇ 10 6 particles/ ⁇ L, and achieves a detection limit of 1.0 ⁇ 10 3 particles/ ⁇ L, but also detects sEVs in healthy serum, normal This was demonstrated by distinguishing between normal sEVs and non-target sEVs derived from cells and other cancer cells.
  • SW620 sEV biomarkers were closely investigated by systematic analyzes including cell survival, cell uptake, wound healing, transwell cell migration/invasion, and tube formation assays.
  • the biomarkers targeted by CCE-10F promote cell migration, invasion, and angiogenesis through sEV-mediated intercellular communication and may be responsible for additional tumor metastasis processes such as EMT. Therefore, it is expected that the DNA aptamer according to the present invention can be used for diagnosis and treatment in the field of colon cancer liquid biopsy.
  • SW620 human colon cancer
  • LS 174T human colon cancer
  • HT29 human colon cancer
  • CCD-18Co human normal colon
  • cells were replaced with conditioned medium supplemented with 5% (v/v) sEVs-depleted FBS (Gibco; Thermo Fisher Scientific) and 1% (v/v) penicillin-streptomycin and incubated for 48 h. Cultured in a 5% CO 2 incubator under humidified conditions at 37°C. The conditioned medium containing the secreted sEVs was recovered, and the sEVs were concentrated, separated, and purified for further use.
  • sEVs-depleted FBS Gibco; Thermo Fisher Scientific
  • penicillin-streptomycin penicillin-streptomycin
  • the conditioned medium was first subjected to a series of centrifugations at 300 xg for 5 min, 2,000 xg for 20 min, and 10,000 xg for 30 min to remove cells, cell debris, microvesicles, and apoptotic bodies, respectively. Afterwards, the supernatant was sequentially syringe filtered using filters with pore sizes of 0.45 and 0.22 ⁇ m (Sartorius, G ⁇ ttingen, Germany) to exclude extracellular vesicles larger than 200 nm in size. For concentration and purification, a tangential flow filtration system with a 300K membrane (Pall Corporation, New York, NY, USA) was used until the medium was concentrated to the desired volume.
  • sEVs were isolated from human serum (HS) (Sigma-Aldrich, St Louis, MO, USA) in the same manner as described above, except that they were diluted in 1 ⁇ PBS before serial centrifugation.
  • HS human serum
  • SW620 and HS-derived small extracellular vesicles Characterization of SW620 and HS-derived small extracellular vesicles (sEVs)
  • the integrity and sEV markers of sEVs isolated from SW620, LS174T, HT29 (positive target) cells and healthy human serum (HS, counter target) were assessed using NTA, cryo-TEM, and Western blot analysis.
  • NTA Nanoparticle Tracking Analysis
  • MONO Zetaview (PMX-130, Particle Metrix, Bavaria, Germany) was used in light scatter mode to determine the concentration and size of sEV (small extracellular vesicle; hereinafter sEV) samples.
  • sEV small extracellular vesicle
  • the instrument Prior to NTA analysis of samples, the instrument was calibrated using 100 nm polystyrene standard beads (diluted 1:250,000 in deionized water). sEV samples were diluted to 4 - 6 x 10 7 particles/mL using 1x PBS before applying to the instrument for measurement.
  • detailed parameters for NTA analysis were consistently set to minimum brightness of 30, sensitivity of 80, shutter of 100, and cell temperature of 23°C. Data from this experiment was analyzed using ZetaView analysis. Deionized water and PBS used in the analysis were filtered with a 0.1 ⁇ m syringe filter membrane for optimal measurement.
  • Figures 14 (A) to 14 (D) show the characteristics of sEVs analyzed according to this example.
  • Figure 14 (A) is the NTA result for SW620 sEV (insert image: cryo-TEM image)
  • Figure 14 (B) is the NTA result for LS 174T sEV (insert image: cryo-TEM image)
  • Figure 14 (C) NTA results for HT29 sEV (inset image: cryo-TEM image).
  • Figure 14 (D) shows the NTA results for HS sEV (insert image: cryo-TEM image). In each cryo-TEM image, scale bar: 100 nm.
  • Equal volumes of SW620, LS 174T, HT29 cells and HS-derived sEVs were concentrated using a qEV enrichment kit (Izon Science) according to the manufacturer's instructions. Each sample was supplemented with 5x SDS-PAGE loading buffer (Biosesang, Seongnam, Republic of Korea), heated at 95°C for 10 min, and incubated at 300 V for 10 min for 20 min using the Mini-Protean tetra system (Bio-Rad Laboratories). % TGX stain-free protein gel (Bio-Rad Laboratories, Hercules, CA, USA).
  • SW620 sEV, LS 174T sEV, HT29 sEV, and HS sEV were about 100 nm to 150 nm, which was consistent with the value obtained using cryo-TEM. Additionally, SW620, LS 174T, HT29 and HS-derived sEVs all showed circular morphology and were within a size range consistent with previous literature, confirming that sEVs were effectively isolated using the SEC method.
  • an important step in TEV-SELEX is to efficiently separate the bound and unbound aptamer pools.
  • immunoplates as a facile targeted sEVs immobilization platform that can not only maintain the integrity of biologically active sEVs but also facilitate the selection of high-quality aptamers with diagnostic value.
  • oligonucleotides used were synthesized by Integrated DNA Technologies (IDT; Coralville, IA, USA). The types of DNA included in the DNA library and the types of forward and reverse primers used are shown in Table 5 below.
  • a random library was designed with a central random region of 40 nucleotides (N40) between the primer regions at both ends used in the PCR amplification step.
  • sEVs derived from three different CRC cells SW620, LS 174T, HT29
  • HS sEVs derived from three different CRC cells (SW620, LS 174T, HT29) and HS were used as positive and counter targets, respectively, and TEV-SELEX was performed by a known method with some modifications.
  • ssDNA single-stranded DNA
  • Negative selection was performed first to remove ssDNA likely to bind to BSA and immune plates. 1 g/L of BSA solution was inoculated into the immune plate and incubated at 37°C for 2 hours. After washing three times with washing buffer (WB; DPBS with 5 mM MgCl 2 ) on a shaker (500 rpm) for 30 seconds each, the prepared ssDNA library was added and incubated for 1 hour. Afterwards, the supernatant containing unbound ssDNA was recovered and concentrated using Nucleospin Gel and PCR clean-up (Macherey-Nagel, Duren, Germany) according to the manufacturer's instructions.
  • WB washing buffer
  • DPBS DPBS with 5 mM MgCl 2
  • the recovered ssDNA was then amplified using asymmetric PCR (Asy-PCR) with different ratios of primer concentrations. Briefly, mixture buffer for Asy-PCR reactions containing 0.2 mM dNTPs, ssDNA template recovered from each selection, high-fidelity nPfu-forte DNA polymerase, and a forward-to-reverse primer ratio of 20:1 in 1x nPfu forte (Enzynomics , Daejeon, Republic of Korea).
  • the thermal cycling protocol was set up as follows: initial denaturation at 95°C for 3 min, followed by 20 cycles of 10 s at 95°C (denaturation), 1 min at 68°C (annealing and elongation), and a final extension of 5 min at 68°C. .
  • the Asy-PCR product was separated on a 2.5% agarose gel, and the ssDNA band was extracted and purified using Nucleospin gel and PCR clean-up.
  • each positive target (sEV, 20 ⁇ g/mL) was incubated in an immune plate at 37°C for 2 h and then washed (three times) by WB. Then, ssDNA from negative selection was added to the wells, then incubated at 37°C for 1 hour and washed (three times) with WB to remove unbound ssDNA. Next, samples from each well were suspended in BB and heated to 95°C for 10 min to recover bound ssDNA from sEVs and then processed as previously described.
  • Negative selection in the TEV-SELEX procedure was performed to remove non-target single-stranded DNA (ssDNA) that non-specifically binds to the buffer and/or components of the immune plate. Unbound ssDNA from negative selection was enriched for further selection containing three E-SELEX loops, where each loop (SW620 sEV, LS 174T sEV, HT29 sEV, HS sEV; 1 st -8 th loop in that order) ) included three consecutive positive screenings and one counterscreening.
  • TEV-SELEX Another important consideration for the success of TEV-SELEX is monitoring the TEV-SELEX procedure to determine the number of loops performed.
  • qPCR can study DNA pools of diverse sequences and provides important information about the homogeneity of ssDNA recovered through TEV-SELEX in terms of ssDNA pool diversity. The number of loops was selected by analyzing ssDNA obtained from each loop (1 st -8 th ) by qPCR.
  • the initial library of ssDNA is highly heterogeneous in sequence due to a random region of 40 nucleotides. However, heterogeneity will be reduced because an iterative process of selecting a pool of aptamers that specifically bind to the target will be continuously performed. This expected behavior was demonstrated through changes in amplification and melting curves ( Figure 16).
  • Figure 16 (A) amplification of the random library (red line; Lib) shows an initial increase and reaches maximum fluorescence intensity, but while the primer region is completely hybridized, the random region in the center partially achieves non-complementary hybridization, making it unstable. A rapid decrease in fluorescence occurred due to the formation of heteroduplex.
  • amplification curves were analyzed quantitatively by calculating the ratio of the fluorescence intensity in the last cycle ( 20th ) to the maximum fluorescence intensity in the final qPCR cycle.
  • a ratio higher than 0.95 was considered a plateau of amplification and was sufficient to ensure high homogeneity in ssDNA libraries.
  • the random ssDNA library (Lib) shows a low ratio, reflecting significant heterogeneity, but the ratio increases as the TEV-SELEX loop is continuously repeated. Only the 8 th -loop (light pink line) showed a fluorescence ratio greater than 0.95, proving that the 8 th -loop reached a plateau in amplification due to the increase in ssDNA homogeneity.
  • the ssDNA obtained in the 8th cycle of Example 2 was analyzed by NGS, and the top 10 candidate aptamers were selected based on frequency (%) for further evaluation.
  • NGS Next-generation sequencing
  • the concentrated ssDNA pool from the final loop ( 8th ) of TEV-SELEX was symmetrically PCR amplified with primers to generate dsDNA, resolved on an agarose gel (2.5%), and using Nucleospin Gel and PCR clean-up. Purified (Macherey-Nagel). Finally, the purified dsDNA was processed and the sequence of the candidate aptamer was analyzed through NGS (next-generation sequencing), which was performed by a sequencing company (Clinomics, Ulsan, Republic of Korea).
  • dsDNA was fragmented with Frag enzyme (MGI, Shenzhen, China) for paired-end (PE) 150 sequencing according to the manufacturer's instructions (MGI FS DNA library preparation set).
  • the fragmented DNA was further selected between 300 and 500 bp by DNA clean beads (MGI), and then recovered to generate a blunt-end and modified to have a single adenosine residue at the 3' end.
  • An adapter sequence with a 5' single thymine residue was ligated to both ends of the DNA fragment.
  • the ligation product was then amplified for 7 cycles and subjected to the following single-strand circularization process:
  • PCR product was heat denatured with a special reverse complementary molecule, a single-stranded circular DNA library was created using DNA ligase, and the remaining linear DNA was digested with exonuclease.
  • the DNA library was finally sequenced using DNBSEQ-T7 (MGI) with a PE read length of 150 bp and evaluated by FastQC (v0.11.8) to assess the overall sequencing quality of the MGI sequencing platform.
  • the forward primer of SEQ ID NO: 2 was included in the 5' part of the aptamer sequence, and the reverse primer of SEQ ID NO: 3 was included in the 3' part of the ssDNA library [Table 7 ], the forward and reverse primer portions are not indicated.
  • T stands for Toggle.
  • candidate aptamers modified with 5'-biotin groups were prepared and then tested against SW620, LS 174T, HT29 (positive target) and HS (counter target) sEVs. Binding affinity was assessed using the ELONA method.
  • Enzyme-linked oligonucleotide assay (ELONA)
  • ELONA was performed using a known method with some modifications. Maxi-binding immunoplate (SPL) wells were first coated with each sEV (1 Blocking was performed with a 3% BSA solution in PBS). After washing three times with PBST, the immune plate was incubated with prepared biotin-conjugated aptamer (IDT) in BB at 37°C for 1 hour and then washed three times with 0.1% PBST. The immune plate was further incubated with streptavidin-peroxidase polymer (Sigma-Aldrich; 1:1,000) diluted in 0.1% PBST for 1 hour at 37°C and then washed (three times) with 0.1% PBST.
  • IDT biotin-conjugated aptamer
  • 1x TMB substrate solution (Invitrogen, Carlsbad, CA, USA) was added to each well and incubated for 6 min in the dark and at room temperature. The reaction was terminated by adding 0.5 M sulfuric acid, and the absorbance was immediately measured at 450 nm using a Spectramax iD5 multi-mode microplate reader (Molecular Devices, San Jose, CA, USA). The normalized value ⁇ A was calculated based on the following equation 3.
  • a t is the absorbance of the sample containing both sEV and aptamer
  • a b is the absorbance of the blank. The concentration of aptamer varies depending on the analysis method, and all experiments were repeated three times.
  • the 10 candidate aptamers with the highest frequency (%) showed different affinities for SW620, LS 174T, HT29, and HS sEVs.
  • T6 affinity was highest for SW620, LS 174T, and HT29 sEVs, and affinity was lowest for HS sEVs.
  • these results confirmed the high binding affinity and specificity of T6 for SW620, LS 174T, and HT29 sEVs; Therefore, T6 was selected for optimization after SELEX.
  • T6 aptamers were included in the ssDNA library for PCR amplification during the TEV-SELEX process. Because shorter length aptamers are synthesized at lower cost and are more suitable for the development of aptamer-based biosensors, we performed post-SELEX optimization of the T6 aptamer by truncating the forward or reverse primer regions. T6 aptamers lacking the forward, reverse, or both regions were designated T6F, T6R, and T6FR, respectively, and the derived sequences of T6 with the additional regions removed were designated T6RA T6RB, T6RB1, T6RB2, and T6RB3, respectively (see Table 8). . The T6 aptamer is represented by SEQ ID NO: 23 in the above-mentioned [Table 7].
  • the binding affinities of the cleaved T6 aptamers (T6F, T6R, and T6FR) to SW620, LS 174T, HT29 sEVs (positive target), and HS sEVs (counter target) are comparable to those of the uncleaved T6 aptamer. was similar to the binding affinity ((A) of Figure 18). However, due to the high binding affinity of T6FR for HS sEVs (countertarget), the specificity was low, so we proceeded in the direction of not selecting.
  • the present inventors cut other regions of T6R (FIG. 19 (A) and (C)) to produce T6RA, T6RB, T6RB1, T6RB2 and T6RB3, and then each positive target ( After analyzing the binding affinity to SW620, LS174T, HT29) and the counter target (HS), the ⁇ A value for each cleaved aptamer was compared and evaluated to select the most appropriate aptamer for this study as T6RB. . As shown in Figure 19 (B), the value of the ⁇ A counter target in T6RB did not change, but the value of the ⁇ A positive target increased, showing the highest binding affinity and specificity.
  • T6RB The binding affinity of T6RB was quantitatively evaluated by estimating K d . Specifically, T6RB was exposed to a certain number of SW620, LS 174T, HT29 and HS sEVs at various concentrations (0-100 nM) and then analyzed using the ELONA method described above in Example 5.
  • Figure 20 (A) shows the dissociation constant ( K d ) curve of T6RB.
  • the absorbance signal increased as the concentration of T6RB increased, showing K d values of 3.848 nM, 5.904 nM, and 5.234 nM, respectively, while HS sEV
  • K d values in the low nanomolar range suggested high binding affinity of T6RB for SW620, LS 174T, and HT29 sEVs. Accordingly, T6RB was determined to be a suitable candidate aptamer for diagnostic applications.
  • T6RB at a concentration of 10 ⁇ M was denatured at 95°C for 5 minutes in a buffer containing 10mM Tris/HCl (pH 7.4) and/or 100mM KCl, and then slowly cooled to 25°C to form secondary structures.
  • CD spectra were recorded on a J-810 spectropolarimeter (Jasco, Tokyo, Japan) in the spectral range of 210–300 nm with a scan speed of 50 nm/min and a bandwidth of 1 nm. All measurements were performed in triplicate and averaged, and the background CD spectrum for the corresponding buffer was measured and subtracted from the experimental spectrum.
  • Spectral analysis was performed using Spectra Manager (Jasco), and DNA secondary structure was analyzed using the conformation index r (Equation 4 below).
  • CD 265 and CD 290 are the CD molar ellipticities [ ⁇ ] at 265 nm and 290 nm, respectively.
  • r ⁇ 0.5, 0 ⁇ r ⁇ 0.5, and r ⁇ 0 mainly correspond to parallel, hybrid, and anti-parallel topologies, respectively.
  • Figure 20 (B) shows the circular dichroism (CD) spectrum of T6RB in the absence/presence of potassium ions (K+).
  • the circular dichroism (CD) spectrum of T6RB shows positive peaks in the 210 nm and 265 nm regions, and as a result of calculation based on Equation 2 above, r > 0.5, T6RB It was confirmed that when a G-quadruplex secondary structure is formed, a parallel structure is formed.
  • T6RB at a concentration of 500 nM was denatured at 95°C for 5 min in a buffer containing 10 mM Tris/HCl (pH 7.4) with or without 100 mM KCl, then slowly cooled to 37°C and incubated for an additional 0.5 h. This allowed the secondary structure to be formed. Then, NMM (Cayman Chemical, Ann Arbor, MI, USA) was added to the mixture at a concentration of 5 ⁇ M and incubated for an additional 0.5 h to allow NMM to be inserted into the T6RB structure. Fluorescence intensity (FI) was measured at an excitation wavelength of 399 nm and an emission wavelength of 610 nm on a Spectramax iD5 multimode microplate reader (Molecular Devices).
  • FI Fluorescence intensity
  • lysates of SW620 sEVs were prepared using RIPA lysis buffer supplemented with Halt protease inhibitor cocktail (Thermo Fisher Scientific) at a final concentration of 1X. Briefly, sEVs were mixed with lysis buffer supplemented with protease inhibitors, incubated on ice for 30 min, and then incubated in a water bath sonicator (SH-2140D, SAE HAN ULTRASONIC, Seoul, Republic of) with 5 cycles of 30 s on/off at 40 kHz. Korea) was treated with ultrasonic waves.
  • H-2140D SAE HAN ULTRASONIC, Seoul, Republic of
  • SW620 sEVs were treated with proK (20 ⁇ g/mL, Enzynomics) for 1 h at 37°C with gentle vortexing every 20 min.
  • proK activity was inhibited by treatment with phenylmethylsulfonyl fluoride (PMSF, 5 mM) for 10 min at room temperature.
  • PMSF phenylmethylsulfonyl fluoride
  • trypsin-EDTA trypsin-ethylenediamine tetraacetic acid
  • Figure 20 (D) shows the results of the biomarker topology analysis (in that order, untreated/proteinase K (proK) treated/trypsin treated/RIPA treated sEVs). That is, Figure 20(D) shows that T6RB exhibits negligible binding affinity to proK and trypsin-treated sEVs. Meanwhile, T6RB showed comparable binding affinity to RIPA-lysed sEVs compared to intact sEVs without any treatment ( Figure 20(D)). These results confirmed that the biomarker targeted by T6RB was identified as a protein and was topologically located in the outer membrane of sEV.
  • proK proteinase K
  • TSA with biotin-tyramide (b-tyramide)/SA-PP system was used as an additional step in ELONA to improve the sensitivity of the assay.
  • Experimental conditions optimized for TSA were explored using HT29 sEV (2 ⁇ 10 7 particles/mL) coated on an immune plate, and the concentrations of T6RB, b-tyramide, H 2 O 2 and TSA reaction time were optimized.
  • the first incubation of SA-HRP (1:1000 dilution in PBST) was performed in ELONA under optimized conditions [15 ⁇ g/mL b-tyramide (APExBIO Technology LLC, Texas, USA) in 0.1 M borate (pH 8.5) and 0.001 ⁇ g/mL b-tyramide (APExBIO Technology LLC, Texas, USA). % H 2 O 2 (Sigma-Aldrich)] at 37°C for 25 minutes, followed by a washing step. Finally, a second culture of SA-HRP (1:1000 dilution in PBST) was treated at 37°C for 1 hour followed by a washing step. TMB reactions and absorbance value normalization were handled in the same way as ELONA described above and all experiments were performed in triplicate.
  • T6RB/b-tyramide/H 2 O 2 concentration and reaction time were optimized in TSA, and the results were 160 nM T6RB and 15 ⁇ g/mL, respectively. Results were obtained for b-tyramide, 0.001% H 2 O 2 and a reaction time of 25 minutes ((AD) of FIG. 21).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Veterinary Medicine (AREA)
  • Analytical Chemistry (AREA)
  • Plant Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Pathology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Hospice & Palliative Care (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Oncology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present invention relates to a DNA aptamer for the diagnosis of colorectal cancer and, specifically, to a composition and a kit for the diagnosis of colorectal cancer, and a colorectal cancer-specific drug delivery composition, each comprising the aptamer consisting of the nucleotide sequences of SEQ ID NOS: 4 to 35. Exhibiting high affinity and specificity for colorectal cancer cell-derived small extracellular vesicles (sEV), the aptamer of the present invention can be used for the specific and accurate diagnosis of colorectal cancer. With the strong binding with specific protein biomarkers present on the sEV membrane, the DNA aptamer suppresses the metastatic potential of cancer through sEV and is expected to find applications in the treatment of colorectal cancer.

Description

대장암 진단 또는 치료용 DNA 압타머DNA aptamer for colon cancer diagnosis or treatment

본 발명은 대장암 진단 및 치료용 DNA 압타머에 관한 기술로, 압타머를 활용하여 높은 친화성 및 특이성으로 대장암 진단이 가능하고, 대장암 유래 sEV와 특이적으로 결합하여 sEV 매개 암 전이성을 억제할 수 있는 잠재력을 지님으로 치료 가능성을 확인한 기술이다. 본 발명은 또한, 높은 이질성 특징을 지니는 대장암을 진단할 수 있는 DNA 압타머에 관한 기술로, 압타머를 활용하여 높은 친화성 및 특이성으로 대장암 진단 가능성을 확인한 기술을 제공한다.The present invention is a technology related to a DNA aptamer for colon cancer diagnosis and treatment. Using the aptamer, colon cancer can be diagnosed with high affinity and specificity, and it specifically binds to colon cancer-derived sEV to prevent sEV-mediated cancer metastasis. It is a technology that has been confirmed to have therapeutic potential as it has the potential to suppress it. The present invention also provides a technology related to a DNA aptamer that can diagnose colon cancer with high heterogeneity characteristics, and a technology that confirms the possibility of colon cancer diagnosis with high affinity and specificity using an aptamer.

본 발명은 과학기술정보통신부의 개인기초연구(과기정통부) [과제고유번호: 1711157003, 과제번호: 2020R1C1C1012275, 연구과제명: 엑소좀 SELEX 기술(E-SELEX)과 이를 이용한 암 진단 바이오]의 일환으로 수행한 연구로부터 도출된 것이다.This invention is as part of the individual basic research of the Ministry of Science and ICT (Ministry of Science and ICT) [Project identification number: 1711157003, task number: 2020R1C1C1012275, research project name: Exosome SELEX technology (E-SELEX) and cancer diagnosis bio using the same] It is derived from research conducted.

전 세계 건강 관리에 대한 위협인 대장암(Colorectal cancer, 이하 'CRC')은 전 세계적으로 세 번째로 많이 진단되는 암이자 두 번째로 많은 암 사망 원인이다. 2020년에 대장암은 전 세계 암 진단의 10%, 암 사망률의 9.4%를 차지했다. 대장암의 종양성 진행은 유전적 돌연변이, 후성 유전적 변형 및 환경적 요인을 포함한 요인의 조합에 의해 시작되며 궁극적으로 다양한 장기, 특히 간에서 재발 및 전이를 촉진한다. 대장암은 특히 높은 종양 이질성 특징을 지니므로 진단 및 치료가 어려운 암종으로 알려져 있다. 대장암 예방 및 통제의 큰 발전에도 불구하고 특정 증상과 정확한 바이오마커의 부족으로 인해 대장암의 검출 및 생존율의 예측이 만족스럽지 않은 상황이다. 따라서 대장암의 효과적인 진단을 위해 대장암 특이적 바이오마커를 탐색하는 것이 매우 중요하다.Colorectal cancer (CRC), a threat to global health care, is the third most diagnosed cancer and the second most common cause of cancer death worldwide. In 2020, colorectal cancer accounted for 10% of cancer diagnoses and 9.4% of cancer deaths worldwide. The neoplastic progression of colorectal cancer is initiated by a combination of factors, including genetic mutations, epigenetic modifications, and environmental factors, ultimately promoting recurrence and metastasis in various organs, especially the liver. Colon cancer is known to be a carcinoma that is particularly difficult to diagnose and treat because it is characterized by high tumor heterogeneity. Despite great progress in the prevention and control of colorectal cancer, detection of colorectal cancer and prediction of survival rates are unsatisfactory due to the lack of specific symptoms and accurate biomarkers. Therefore, it is very important to explore colon cancer-specific biomarkers for effective diagnosis of colon cancer.

한편, 엑소좀이라고도 알려진 작은 세포외소포체 (small extracellular vesicles, sEVs)는 모든 세포 유형에서 세포외로 분비되는 나노 크기의 지질막으로 둘러싸인 소포체(직경 50-200 nm)이다. sEVs은 혈액, 소변, 땀, 타액과 같은 다양한 체액에서 안정적이다. sEVs은 DNA, RNA 및 단백질과 같은 활성 생체 분자를 운반하여 기원 세포와 수용 세포 사이의 세포 간 신호전달을 국부적이고 체계적으로 매개한다. 특히, 종양 세포에서 유래한 sEVs은 암 발달, 진행, 침습 및 전이의 조절자 역할을 할 수 있다. 점점 더 많은 연구에서 CRC 유래 sEVs의 잠재적 응용이 입증되었다. 예를 들어, sEVs 마이크로RNA(miRNA), 긴 비암호화 RNA(lncRNA), 메신저 RNA(mRNA) 및 단백질의 발현 패턴은 CRC에서 상향 조절되며, 이는 CRC의 진단 가능성을 높여준다. 또한 전이, 화학내성 및 재발을 포함하여 CRC 예후에 대한 후보 sEVs 바이오마커를 확인하였다. 인체 체액을 순환하는 sEVs은 모세포의 미니 버전으로 간주되기 때문에 비침습적 대장암 진단이 가능하다. 따라서 sEVs을 진단 및 치료 응용을 위한 유망한 도구로 사용하는 것에 대한 연구 관심이 증가하고 있다. 특히, 대장암 유래 sEVs과 기원을 정확히 알기 어려운 정상인 혈청 유래 sEVs을 구별하는 것은 매우 중요하다.Meanwhile, small extracellular vesicles (sEVs), also known as exosomes, are nano-sized lipid membrane-enclosed endoplasmic reticulum (50-200 nm in diameter) that are secreted extracellularly by all cell types. sEVs are stable in various body fluids such as blood, urine, sweat, and saliva. sEVs transport active biomolecules such as DNA, RNA, and proteins to locally and systematically mediate intercellular signaling between cells of origin and recipient cells. In particular, sEVs derived from tumor cells can act as regulators of cancer development, progression, invasion, and metastasis. More and more studies have demonstrated the potential applications of CRC-derived sEVs. For example, the expression patterns of sEVs microRNAs (miRNAs), long non-coding RNAs (lncRNAs), messenger RNAs (mRNAs) and proteins are upregulated in CRC, which increases the diagnostic potential of CRC. Additionally, candidate sEVs biomarkers for CRC prognosis, including metastasis, chemoresistance, and recurrence, were identified. Since sEVs circulating in human body fluids are considered mini versions of parent cells, non-invasive diagnosis of colorectal cancer is possible. Therefore, there is increasing research interest in using sEVs as a promising tool for diagnostic and therapeutic applications. In particular, it is very important to distinguish between colon cancer-derived sEVs and normal human serum-derived sEVs, whose origin is difficult to accurately determine.

또한, 특정 표적에 결합할 수 있는 단일 가닥 DNA 또는 RNA인 압타머는 높은 안정성, 작은 크기, 합성 용이성 및 변형, 낮은 면역원성, 높은 결합 친화성 및 특이성 등의 탁월한 장점으로 인해 진단 및 치료제 개발과 바이오마커 발굴에 있어 큰 주목을 받고 있다. 압타머는 일반적으로 SELEX(Systematic Evolution of Ligands by EXponential Enrichment)에 의한 리간드의 체계적인 진화라고 하는 반복적인 시험관 내 선택 과정을 통해 스크리닝된다. SELEX의 주요 장점은 표적에 대한 사전 지식 없이도 세포, 조직 및 생체 내 모델과 같은 자연 형태를 유지하는 모든 생물학적 개체에 대해 압타머를 얻을 수 있다는 것이다.In addition, aptamers, which are single-stranded DNA or RNA that can bind to a specific target, are widely used in diagnostic and therapeutic development and biotechnology due to their outstanding advantages such as high stability, small size, ease of synthesis and modification, low immunogenicity, and high binding affinity and specificity. It is receiving great attention when it comes to marker discovery. Aptamers are typically screened through an iterative in vitro selection process called Systematic Evolution of Ligands by EXponential Enrichment (SELEX). The main advantage of SELEX is that aptamers can be obtained for any biological entity that maintains its natural form, such as cells, tissues, and in vivo models, without prior knowledge of the target.

이에, 본 발명자들은 sEVs를 통해 대장암을 진단할 수 있는 안정한 DNA 압타머를 찾고, sEVs 매개 암 전이성을 억제함으로 대장암을 치료할 수 있는 잠재력을 밝혔으며, 대장암을 진단할 수 있는 다양한 특성을 지닌 대장암 sEVs에 대한 결합성이 높은 DNA 압타머를 찾고, 이를 이용하여 높은 민감도의 바이오센서 개발한 결과 본 발명을 완성하였다. Accordingly, the present inventors found a stable DNA aptamer that can diagnose colon cancer through sEVs, revealed its potential to treat colon cancer by inhibiting sEVs-mediated cancer metastasis, and demonstrated various properties that can be used to diagnose colon cancer. The present invention was completed by finding a DNA aptamer with high binding to colon cancer sEVs and using it to develop a highly sensitive biosensor.

본 발명의 목적은 서열번호 4 내지 17의 염기서열로 이루어진 군에서 선택되는 어느 하나 이상의 대장암 진단용 DNA 압타머를 제공하는 것이다. The purpose of the present invention is to provide at least one DNA aptamer for colon cancer diagnosis selected from the group consisting of base sequences of SEQ ID NOs: 4 to 17.

본 발명의 또다른 목적은 상기 압타머를 포함하는 대장암 진단용 조성물, 키트 및 대장암 특이적 약물 전달 조성물을 제공하는 것이다. Another object of the present invention is to provide a composition for diagnosing colon cancer, a kit, and a colon cancer-specific drug delivery composition containing the aptamer.

본 발명의 또다른 목적은 상기 압타머의 제조 방법, 압타머를 이용한 대장암 진단을 위한 정보를 제공하는 방법 및 대장암 sEVs 특이적 표면 바이오마커의 검출방법을 제공하는 것이다. Another object of the present invention is to provide a method for producing the aptamer, a method for providing information for colon cancer diagnosis using the aptamer, and a method for detecting surface biomarkers specific to colon cancer sEVs.

본 발명의 또다른 목적은 서열번호 18 내지 35의 염기서열로 이루어진 군에서 선택되는 어느 하나 이상의 대장암 진단용 DNA 압타머를 제공하는 것이다. Another object of the present invention is to provide at least one DNA aptamer for colon cancer diagnosis selected from the group consisting of base sequences of SEQ ID NOs: 18 to 35.

본 발명의 또다른 목적은 상기 압타머를 포함하는 대장암 진단용 조성물, 키트 및 대장암 특이적 약물 전달 조성물을 제공하는 것이다. Another object of the present invention is to provide a composition for diagnosing colon cancer, a kit, and a colon cancer-specific drug delivery composition containing the aptamer.

본 발명의 또다른 목적은 상기 압타머의 제조 방법, 압타머를 이용한 대장암 진단을 위한 정보를 제공하는 방법 및 대장암 sEVs 특이적 표면 바이오마커의 검출방법을 제공하는 것이다. Another object of the present invention is to provide a method for producing the aptamer, a method for providing information for colon cancer diagnosis using the aptamer, and a method for detecting surface biomarkers specific to colon cancer sEVs.

그러나, 본 발명이 이루고자 하는 기술적 과제는 이상에서 언급한 과제에 제한되지 않으며, 언급되지 않은 또 다른 과제들은 아래의 기재로부터 당업자에게 명확하게 이해될 수 있을 것이다.However, the technical problem to be achieved by the present invention is not limited to the problems mentioned above, and other problems not mentioned will be clearly understood by those skilled in the art from the description below.

본 발명자들은 대장암 세포에 특이적인 고품질 압타머를 발굴하기 위해 대장암 sEV SELEX; E-SELEX를 구축했다. 각 루프가 4개의 연속적인 양성 선별과 하나의 대항 선별로 구성된 총 3개의 E-SELEX 루프로 진행 후, 상위 10개의 압타머 후보를 선택하고 효소 결합 올리고뉴클레오티드 어세이(Enzyme-linked oligonucleotide assay, ELONA) 방법을 사용하여 결합 친화도를 평가했다. 마지막으로 post-SELEX 최적화를 통해 최상의 압타머를 얻었으며, 이는 3.41 nM의 K d 값과 1.0 x 103 입자/μL의 검출한계값이 도출되어 혈청 또는 혈장 농도 범위 내에서 대장암 세포 유래 sEV를 검출하기에 충분한 감도를 갖는 것을 확인하였다. 또한 비표적 sEV(예: 건강한 혈청 유래 sEV 및 정상 세포 유래 sEV, 다른 암세포 유래 sEV)과 높은 특이성을 보이며 효과적으로 대장암 세포 유래 sEV을 구별했다. 선정된 최적의 압타머 (서열번호 4 내지 17)를 대장암 세포 유래 sEV의 바이오마커 예측을 위해 추가로 조사하였으며 sEV 매개 세포간 신호전달을 조절함으로써 세포 이동 및 증식을 억제하는 것을 입증하였다. The present inventors used colon cancer sEV SELEX; E-SELEX was built. After proceeding with a total of three E-SELEX loops, where each loop consists of four consecutive positive selections and one counter selection, the top 10 aptamer candidates are selected and subjected to enzyme-linked oligonucleotide assay (ELONA). ) method was used to evaluate binding affinity. Finally, the best aptamer was obtained through post-SELEX optimization, which resulted in a K d value of 3.41 nM and a detection limit of 1.0 It was confirmed that it had sufficient sensitivity for detection. In addition, it showed high specificity from non-target sEVs (e.g., sEVs derived from healthy serum and sEVs from normal cells, and sEVs derived from other cancer cells) and effectively distinguished sEVs derived from colon cancer cells. The selected optimal aptamers (SEQ ID NOs: 4 to 17) were further investigated to predict biomarkers of sEVs derived from colon cancer cells, and were demonstrated to inhibit cell migration and proliferation by regulating sEV-mediated intercellular signaling.

또한, 본 발명자들은 세 종류의 대장암 세포(SW620, LS 174T, HT29)에 특이적인 고품질 압타머를 발굴하기 위해 대장암 Colorectal Cancer (CRC) Toggle sEV-SELEX (이하, TEV-SELEX)를 구축했다. 각 루프를 세 번의 연속적인 양성 선별과 한 번의 대항 선별로 구성하여 총 여덟 번의 루프를 진행한 TEV-SELEX를 구축하였고, NGS 결과 기반으로 빈도수 (%) 상위 10 개의 압타머 후보를 선택하고 효소 결합 올리고뉴클레오티드 어세이(Enzyme-linked oligonucleotide assay, ELONA) 방법을 사용하여 결합 친화도를 평가했다. 마지막으로 post-SELEX 최적화를 통해 최상의 압타머를 얻었으며, 이는 SW620, LS 174T, HT29 세포에 대해 각 3.848 nM, 5.904 nM, 5.234 nM의 K d 값과 각 3.6 x 102 입자/μL, 3.5 x 103 입자/μL, 8.4 x 102 입자/μL의 검출한계값이 도출되어 혈청 또는 혈장 농도 범위 내에서 대장암 세포 유래 sEVs을 검출하기에 충분한 감도를 갖는 것을 확인하였다. 또한 비표적 sEVs(예: 건강한 혈청 유래 sEVs 및 정상 결장 세포 유래 sEVs)에 대해 높은 특이성을 보이며, 효과적으로 이질성이 높은 대장암 세포 유래 sEVs을 구별하였다. In addition, the present inventors constructed Colorectal Cancer (CRC) Toggle sEV-SELEX (hereinafter referred to as TEV-SELEX) to discover high-quality aptamers specific to three types of colon cancer cells (SW620, LS 174T, HT29). . TEV-SELEX was constructed with a total of eight loops, with each loop consisting of three consecutive positive selections and one counter selection. Based on the NGS results, the top 10 aptamer candidates by frequency (%) were selected and enzyme-linked. Binding affinity was evaluated using an oligonucleotide assay (Enzyme-linked oligonucleotide assay, ELONA) method. Finally, the best aptamer was obtained through post-SELEX optimization, with K d values of 3.848 nM, 5.904 nM, and 5.234 nM for SW620, LS 174T, and HT29 cells, respectively, and 3.6 × 10 2 particles/μL, 3.5 × Detection limits of 10 3 particles/μL and 8.4 In addition, it showed high specificity for non-target sEVs (e.g., sEVs derived from healthy serum and sEVs derived from normal colon cells) and effectively distinguished sEVs derived from colon cancer cells with high heterogeneity.

따라서, 본 발명은 서열번호 4 내지 17의 염기서열로 이루어진 군에서 선택되는 어느 하나 이상의 대장암 진단 또는 치료용 DNA 압타머, 서열번호 18 내지 35의 염기서열로 이루어진 군에서 선택되는 어느 하나 이상의 대장암 진단용 DNA 압타머를 제공한다. Therefore, the present invention provides a DNA aptamer for diagnosing or treating colon cancer selected from the group consisting of the base sequences of SEQ ID NOs: 4 to 17, and one or more colon cancers selected from the group consisting of the base sequences of SEQ ID NOs: 18 to 35 Provides DNA aptamers for cancer diagnosis.

본 발명은 서열번호 4 내지 17의 염기서열로 이루어진 군에서 선택되는 적어도 어느 하나의 서열을 포함하는 DNA 압타머의 대장암 치료 또는 진단 용도, 서열번호 18 내지 35의 염기서열로 이루어진 군에서 선택되는 적어도 어느 하나의 서열을 포함하는 DNA 압타머의 대장암 진단 용도를 제공한다. The present invention relates to the treatment or diagnosis of colon cancer of a DNA aptamer containing at least one sequence selected from the group consisting of the base sequences of SEQ ID NOs: 4 to 17, and the use of a DNA aptamer selected from the group consisting of the base sequences of SEQ ID NOs: 18 to 35. A DNA aptamer containing at least one sequence is provided for use in colon cancer diagnosis.

본 발명에 있어서 대장암은 "결장직장암", "Colorectal cancer", "CRC"와 동일한 의미로 사용된다. In the present invention, colon cancer is used with the same meaning as “colorectal cancer,” “Colorectal cancer,” and “CRC.”

본 발명에 있어서, "DNA 압타머"는 높은 친화도와 특이성으로 대상 표적에 결합하는 특성을 가지며, 각기 독특한 3차원 구조를 가지는 단일가닥 올리고뉴클레오티드를 의미한다. 반복된 생체 외 선별 및 농축 과정을 통해 DNA 압타머 라이브러리로부터 특정 대상 표적에 특이적으로 결합하는 DNA 분자, 즉 DNA 압타머의 선별이 가능하다.In the present invention, “DNA aptamer” refers to a single-stranded oligonucleotide that has the characteristic of binding to a target with high affinity and specificity and each has a unique three-dimensional structure. Through repeated in vitro selection and enrichment processes, it is possible to select DNA molecules that specifically bind to a specific target, that is, DNA aptamers, from a DNA aptamer library.

본 발명에 있어서, "진단"은 병리 상태의 존재 또는 특징을 확인하는 것을 의미한다. 본 발명에서의 진단은 DNA 압타머를 이용하여 대장암의 존재 또는 발생 여부를 확인하는 것이다.In the present invention, “diagnosis” means confirming the presence or characteristics of a pathological condition. Diagnosis in the present invention is to confirm the presence or occurrence of colon cancer using a DNA aptamer.

본 발명에 있어서, "치료"란, 본 발명의 DNA 압타머를 이용하여 이미 유발된 대장암의 증세가 호전되거나 진행을 억제하는 등의 이롭게 되는 모든 행위를 의미한다.In the present invention, “treatment” means any beneficial action, such as improving the symptoms or suppressing the progression of colon cancer that has already been caused by using the DNA aptamer of the present invention.

본 개시의 일 측면에 있어서, 서열번호 4 내지 35의 염기 서열과 90% 이상의 서열 상동성을 가지는 DNA 압타머를 포함할 수 있으며, "90% 이상의 서열 상동성을 가지는 염기 서열"이란 일 내지 수개의 뉴클레오티드가 추가, 결실 또는 치환되어 90% 이상 100% 미만의 서열에 공통성이 있는 것으로 유사한 암 특이적 결합능을 보이는 염기 서열을 의미한다.In one aspect of the present disclosure, it may include a DNA aptamer having 90% or more sequence homology to the nucleotide sequences of SEQ ID NOs. 4 to 35, and the term “base sequence having 90% or more sequence homology” ranges from 1 to 10. It refers to a nucleotide sequence that shows similar cancer-specific binding ability by adding, deleting, or substituting 90% or more but less than 100% of the sequences with nucleotides added, deleted, or substituted.

본 발명에 있어서, 상기 압타머는 대장암 세포 유래 sEVs에 특이적으로 결합할 수 있다. 본 발명의 일실시예에서, 양성 표적 및 대항 표적으로부터 sEVs를 분리하였으며, 본 발명의 DNA 압타머를 이용한 경우, 정상 결장세포, 인간 유방암 세포, 인간 교모세포종으로부터 유래한 sEV에는 결합특이성을 갖지 않으나, 대장암 세포 유래 sEV에만 특이적으로 결합하는 것을 확인하였다(도 8). In the present invention, the aptamer can specifically bind to sEVs derived from colon cancer cells. In one embodiment of the present invention, sEVs were isolated from positive and counter targets, and when the DNA aptamer of the present invention was used, it did not have binding specificity to sEVs derived from normal colon cells, human breast cancer cells, or human glioblastoma. , it was confirmed that it specifically bound only to sEV derived from colon cancer cells (Figure 8).

본 발명의 상기 압타머는 정방향 프라이머 및 역방향 프라이머 서열로 이루어진 군에서 선택되는 하나 이상을 추가로 포함할 수 있다.The aptamer of the present invention may further include one or more selected from the group consisting of forward primer and reverse primer sequences.

본 발명에서 "프라이머"는 짧은 자유 3' 말단 수산화기(free 3` hydroxyl group)을 가지는 핵산 서열로 상보적인 주형(template)과 염기쌍(base pair)을 형 성할 수 있고 주형의 복사를 위한 시작지점으로 기능을 하는 짧은 핵산 서열을 의 미한다. 본 발명에서는 전술한 DNA 압타머의 정방향 및 역방향 프라이머를 이용하여 PCR 증폭을 실시하여 대장암을 진단할 수 있다. PCR 조건, 정방향 및 역방향 프라이머의 길이는 당업계에 공지된 것을 기초로 변형할 수 있다. 예시적으로, 상기 정방향 프라이머의 서열은 서열번호 2의 염기서열로 이루어질 수 있으며, 역방향 프라이머의 서열은 서열번호 3의 염기서열로 이루어질 수 있으나, 이에 제한되는 것은 아니다. In the present invention, a "primer" is a nucleic acid sequence with a short free 3' terminal hydroxyl group that can form a base pair with a complementary template and serves as a starting point for copying the template. It refers to a short nucleic acid sequence that performs a function. In the present invention, colon cancer can be diagnosed by performing PCR amplification using the forward and reverse primers of the DNA aptamer described above. PCR conditions and lengths of forward and reverse primers can be modified based on those known in the art. Illustratively, the sequence of the forward primer may consist of the base sequence of SEQ ID NO: 2, and the sequence of the reverse primer may consist of the base sequence of SEQ ID NO: 3, but are not limited thereto.

구체적인 일실시예에서 프라이머 영역은 E-SELEX 또는 TEV-SELEX 과정 동안 상기 압타머 서열의 5' 부분에는 정방향 프라이머가, 3' 부분에는 역방향 프라이머 영역을 ssDNA 라이브러리에 포함시켜, PCR 증폭에 사용하였다. PCR 증폭을 위해 5' 부분 및 3' 부분에 프라이머 영역이 결합될 수 있음은 본 발명의 기술 분야에서 통상의 기술자에게 자명하다. In a specific example, the primer region was used for PCR amplification by including a forward primer in the 5' portion of the aptamer sequence and a reverse primer region in the 3' portion of the aptamer sequence in an ssDNA library during the E-SELEX or TEV-SELEX process. It is obvious to those skilled in the art that primer regions can be combined at the 5' and 3' portions for PCR amplification.

일 양태에 있어서, 상기 DNA 압타머는 서열번호 13의 염기서열로 이루어질 수 있다. 구체적인 일실시예에서 SELEX 이후 빈도수 (%) 상위 10개의 후보 압타머를 선별하였으며, 결합 친화도 및 특이성을 갖는 압타머를 선택하였다. 그 결과, 도 6에 나타난 바와 같이, CCE-10가 sEV에 대한 친화도가 가장 높았다. In one aspect, the DNA aptamer may consist of the base sequence of SEQ ID NO: 13. In a specific example, the top 10 candidate aptamers by frequency (%) after SELEX were selected, and aptamers with binding affinity and specificity were selected. As a result, as shown in Figure 6, CCE-10 had the highest affinity for sEV.

그 이후, 정방향 또는 역방향 프라이머 영역의 유무를 조절하여 CCE-10 압타머 서열의 최적화를 수행하였다. 따라서, [표 4]에 나타난 바와 같이, CCE-10, CCE-10F, CCE-10R, CCE-10FR을 이용하였다. 따라서, 예시적으로 상기 서열번호 13의 염기서열로 이루어진 DNA 압타머는, 정방향 프라이머 염기서열 및 역방향 프라이머 염기서열 중 어느 하나 이상이 추가적으로 연결될 수 있다. 그 결과, 정방향 프라이머를 제거한 서열벌호 15의 염기서열로 이루어진, DNA 압타머가 가장 높은 친화력을 나타냈다. 따라서, 상기 DNA 압타머는 서열벌호 15의 염기서열로 이루어진 것일 수 있다. 또한, 상기 DNA 압타머의 결합 친화도를 평가해 보았을 때 결합 해리상수로는 3 내지 4 nM일 수 있으며, 바람직하게는 3.2 내지 3.6일 수 있다. Afterwards, optimization of the CCE-10 aptamer sequence was performed by controlling the presence or absence of the forward or reverse primer region. Therefore, as shown in [Table 4], CCE-10, CCE-10F, CCE-10R, and CCE-10FR were used. Therefore, for example, the DNA aptamer consisting of the base sequence of SEQ ID NO: 13 may be additionally linked to one or more of the forward primer base sequence and the reverse primer base sequence. As a result, the DNA aptamer consisting of the base sequence of sequence number 15 with the forward primer removed showed the highest affinity. Therefore, the DNA aptamer may be composed of the base sequence of sequence number 15. Additionally, when evaluating the binding affinity of the DNA aptamer, the binding dissociation constant may be 3 to 4 nM, preferably 3.2 to 3.6.

본 발명의 다른 일 양태에 있어서, 상기 DNA 압타머는 서열번호 32의 염기서열로 이루어질 수 있다. 구체적인 일실시예에서 SELEX 이후 빈도수(%) 상위 10 개의 후보 압타머를 선별하였으며, 높은 결합 친화도 및 특이성을 갖는 압타머를 선택하였다. 그 결과, 도 17에 나타난 바와 같이, T6가 sEVs에 대한 친화도가 가장 높았다. 그 이후, T6 압타머 서열의 최적화를 수행하였다. 따라서, [표 8]에 나타난 바와 같이, T6F T6R T6FR T6RA T6RB T6RB1 T6RB2 T6RB3 서열을 이용하였다. In another aspect of the present invention, the DNA aptamer may consist of the base sequence of SEQ ID NO: 32. In a specific example, the top 10 candidate aptamers by frequency (%) after SELEX were selected, and aptamers with high binding affinity and specificity were selected. As a result, as shown in Figure 17, T6 had the highest affinity for sEVs. Afterwards, optimization of the T6 aptamer sequence was performed. Therefore, as shown in [Table 8], the T6F T6R T6FR T6RA T6RB T6RB1 T6RB2 T6RB3 sequence was used.

본 발명은 또한, 상기의 압타머를 포함하는 대장암 진단 또는 치료용 조성물을 제공한다. The present invention also provides a composition for diagnosing or treating colon cancer containing the aptamer above.

본 발명에 있어서, 상기 조성물은 생리학적으로 허용 가능한 부형제, 담체, 또는 첨가제를 더 포함할 수 있으며, 이에는 전분, 젤라틴화 전분, 미결정셀룰로오스, 유당, 포비돈, 콜로이달실리콘디옥사이드, 인산수소칼슘, 락토스, 만니톨, 엿, 아라비아고무, 전호화전분, 옥수수전분, 분말셀룰로오스, 히드록시프로필셀룰로오스, 오파드라이, 전분글리콜산나트륨, 카르나우바납, 합성규산알루미늄, 스테아린산, 스테아린산마그네슘, 스테아린산알루미늄, 스테아린산칼슘, 백당, 덱스트로스, 소르비톨 및 탈크 등이 사용될 수 있으나, 이에 제한되지 않는다. In the present invention, the composition may further include physiologically acceptable excipients, carriers, or additives, including starch, gelatinized starch, microcrystalline cellulose, lactose, povidone, colloidal silicon dioxide, calcium hydrogen phosphate, Lactose, mannitol, taffy, gum arabic, pregelatinized starch, corn starch, powdered cellulose, hydroxypropyl cellulose, Opadry, sodium starch glycolate, carnauba lead, synthetic aluminum silicate, stearic acid, magnesium stearate, aluminum stearate, calcium stearate. , white sugar, dextrose, sorbitol, and talc may be used, but are not limited thereto.

본 발명의 다른 실시예에서는 상기 DNA 압타머를 포함하는 조성물을 이용하는 경우, 대장암 세포주 이외 정상 혈청이나 정상 결장 세포주 또는 다른 인간 암 세포주에 대해서는 거의 결합하지 않아 (다양한) 대장암 세포주를 특이적으로 검출함을 확인하였다.In another embodiment of the present invention, when using a composition containing the DNA aptamer, it hardly binds to normal serum, normal colon cell lines, or other human cancer cell lines other than colon cancer cell lines, so it can be used to specifically target (various) colon cancer cell lines. Detection was confirmed.

본 발명은 또한 상기 조성물을 포함하는, 대장암 진단 또는 치료용 키트를 제공한다.The present invention also provides a kit for diagnosing or treating colon cancer, comprising the composition.

일실시예로, 대장암 유래 sEV에 특이적으로 결합하는 압타머를 함유하는 대장암 진단용 키트(kit)의 형태로 제공될 수 있다. 이 대장암 진단용 키트는 필요에 따라 완충용액 및 검출의 수행과 분석을 위한 용기들을 포함할 수 있는데, 병, 통(tub), 작은 봉지(sachet), 봉투(envelope), 튜브, 앰플(ampoule) 등과 같 은 형태를 취할 수 있으며 이들은 부분적으로 또는 전체적으로 플라스틱, 유리, 종이, 호일, 왁스 등으로부터 형성될 수 있다. 용기는, 처음에는 용기의 일부이거나 또는 기계적, 접착성, 또는 기타 수단에 의해 용기에 부착될 수 있는, 완전히 또는 부분적으로 분리가 가능한 마개를 장착할 수 있다. 용기는 또한 주사바늘에 의해 내용물에 접근할 수 있는, 스토퍼가 장착될 수 있다. 상기 키트는 외부 패키지를 포함할 수 있으며, 외부 패키지는 구성 요소들의 사용에 관한 사용설명서를 포함할 수 있다.In one embodiment, it may be provided in the form of a colon cancer diagnosis kit containing an aptamer that specifically binds to colon cancer-derived sEV. This colon cancer diagnostic kit may include a buffer solution and containers for detection and analysis as needed, such as bottles, tubs, sachets, envelopes, tubes, and ampoules. They may be formed in part or entirely from plastic, glass, paper, foil, wax, etc. The container may be equipped with a completely or partially removable closure that may initially be part of the container or may be attached to the container by mechanical, adhesive, or other means. The container may also be equipped with a stopper, allowing access to the contents by means of a needle. The kit may include an external package, and the external package may include instructions for use of the components.

상기 키트는 진단용 센서, RT-PCR 키트, 경쟁적 RT-PCR 키트, 실시간 RT-PCR 키트, DNA 칩 키트 및 단백질 칩 키트일 수 있다. The kit may be a diagnostic sensor, RT-PCR kit, competitive RT-PCR kit, real-time RT-PCR kit, DNA chip kit, and protein chip kit.

본 발명의 키트에는 상기 DNA 압타머를 인식하는 프라이머, 프로브 뿐만 아니라 분석 방법에 적합한 하나 이상의 다른 구성 성분을 포함하는 조성물, 용액 또는 장치가 포함될 수 있다.The kit of the present invention may include a composition, solution, or device containing not only primers and probes that recognize the DNA aptamer, but also one or more other components suitable for the analysis method.

예시적으로, 상기 대장암 유래 sEV에 특이적으로 결합하는 DNA 압타머는 비드, 입자, 딥스틱(dipstick), 섬유, 필터, 막 및 유리 슬라이드와 같은 통상적인 지지체 및 실란 또는 실리케이트 지지체 등의 고체 지지체에 고정되어 검출센서로 제공됨으로써, 대장암 진단에 이용될 수 있는바, 본 발명은, 상기 대장암 유래 sEV 에 특이적으로 결합하는 DNA 압타머가 고정되어 있는 대장암 진단용 센서일 수 있다. Illustratively, the DNA aptamer that specifically binds to the colon cancer-derived sEV may be used on conventional supports such as beads, particles, dipsticks, fibers, filters, membranes, and glass slides, and solid supports such as silane or silicate supports. By being fixed to and provided as a detection sensor, it can be used for colon cancer diagnosis. The present invention may be a sensor for colon cancer diagnosis in which a DNA aptamer that specifically binds to the colon cancer-derived sEV is fixed.

상기 고체 지지체는 적어도 하나의 실질적으로 단단한 표면을 포함하는데, 그 표면 위에 상기 DNA 압타머들이 움직일 수 없게 고정될 수 있다. 이때, 상기 DNA 압타머는 모든 통상적인 화학적 커플링 방법에 의하여 고정화 될 수 있다. 예컨대, 상기 DNA 압타머의 말단에 바이오틴을 결합시켜 복합체를 형성하고, 상기 칩 등의 기판 표면에 스트렙타비딘 혹은 아비딘을 고정화시켜, 상기 바이오틴과 기판 표면에 고정화된 스트렙타비딘 혹은 아비딘의 상호작용에 의하여 상기 DNA 압타머를 기판 표면에 고정화시길 수 있다.The solid support includes at least one substantially rigid surface, on which the DNA aptamers can be immovably fixed. At this time, the DNA aptamer can be immobilized by any conventional chemical coupling method. For example, biotin is bound to the end of the DNA aptamer to form a complex, and streptavidin or avidin is immobilized on the surface of a substrate such as the chip, so that the interaction between the biotin and streptavidin or avidin immobilized on the substrate surface The DNA aptamer can be immobilized on the surface of the substrate.

또한, 상기 키트에는 대장암 sEV에 결합된 DNA 압타머를 검출하기 위한 신호 생성 방식으로 다양한 방식이 적용될 수 있다. 예시적으로, colorimetry, fluorescence, 및 electrochemistry 등일 수 있으며, 이에 제한되지 않고, 각각의 신호생성에 적합한 labeling 방법을 통해서 구현이 가능한 방법이라면 제한없이 사용가능하다. In addition, various methods can be applied to the kit as a signal generation method to detect DNA aptamers bound to colon cancer sEVs. Examples include colorimetry, fluorescence, and electrochemistry, but are not limited thereto, and any method that can be implemented through a labeling method suitable for each signal generation can be used without limitation.

한편, 본 개시의 일 측면에 있어서, DNA 압타머는 전술한 그 자체, 조성물, 또는 키트를 통해 관련 기술 분야의 통상의 기술자에 의해 이해된 바와 같이 선택된 투여 경로에 따라 다양한 형태로 대상체에 투여될 수 있다. 예를 들어, 국소, 경장 또는 비경구 적용에 의해 투여될 수 있다. 국소 적용은 표피, 흡입, 관장제, 점안제, 점이제 및 신체 내의 점막을 통한 적용을 포함하나, 이에 제한되지 않는다. 경장 적용은 경구 투여, 직장 투여, 질 투여 및 위 영양 공급 튜브 등을 포함할 수 있다. 비경구 투여는 정맥내, 동맥내, 피막내, 안와내, 심장내, 피내, 경기관, 각피하, 관절내, 피막 하, 지주막하, 척수내, 경막외, 흉골내, 복강내, 피하, 근육내, 경상피, 비강, 폐내, 척수강내, 직장 및 국소 투여 방식을 포함할 수 있다. Meanwhile, in one aspect of the present disclosure, the DNA aptamer can be administered to the subject in various forms according to the selected administration route as understood by those skilled in the art through the above-described itself, composition, or kit. there is. For example, it may be administered by topical, enteral or parenteral application. Topical applications include, but are not limited to, epidermis, inhalation, enemas, eye drops, ear drops, and application through mucous membranes within the body. Enteral applications may include oral administration, rectal administration, vaginal administration, and gastric feeding tubes. Parenteral administration is intravenous, intraarterial, intracapsular, intraorbital, intracardiac, intradermal, transtracheal, subcutaneous, intraarticular, subcapsular, subarachnoid, intrathecal, epidural, intrasternal, intraperitoneal, subcutaneous, Methods of administration may include intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal, and topical.

아울러, 본 개시의 일 측면에 있어서, DNA 압타머는 전술한 그 자체, 조성물, 또는 키트는 투여 경로 등에 따라 적절한 형태로 제제화될 수 있다. 제제화 할 경우에는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제 등을 사용하여 조제될 수 있으나, 이에 제한되지 않는다.In addition, in one aspect of the present disclosure, the DNA aptamer itself, the composition, or the kit described above may be formulated in an appropriate form depending on the route of administration, etc. When formulated, it may be prepared using diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrants, and surfactants, but is not limited thereto.

본 발명은 또한, 상기 DNA 압타머 제조 방법을 제공한다.The present invention also provides a method for producing the DNA aptamer.

구체적으로, 상기 방법은Specifically, the method is

a) 랜덤 라이브러리를 생성하는 단계;a) generating a random library;

b) E-SELEX 또는 TEV-SELEX를 수행하는 단계; b) performing E-SELEX or TEV-SELEX;

c) NGS 기법을 통해 DNA 압타머 서열 분석하는 단계; 및c) analyzing the DNA aptamer sequence through NGS technology; and

d) DNA 압타머 특성화하는 단계;를 포함할 수 있다. d) characterizing the DNA aptamer;

또한, DNA 압타머 제조 방법에 있어서도 상기 압타머는 대장암 세포 유래 sEV에 특이적으로 결합할 수 있으나, 이는 DNA 압타머의 기재에서 전술하였는 바 중복을 피하기 위해 기재를 생략한다. In addition, in the method of producing a DNA aptamer, the aptamer can specifically bind to sEV derived from colon cancer cells, but this description is omitted to avoid duplication as described above in the description of the DNA aptamer.

본 발명은 또한, 상기 DNA 압타머를 이용한 대장암 진단 또는 치료를 위한 정보를 제공하는 방법을 제공한다. The present invention also provides a method of providing information for diagnosis or treatment of colon cancer using the DNA aptamer.

본 발명은 상기 압타머를 이용한 대장암 sEV 특이적 표면 바이오마커의 검출방법을 제공한다. The present invention provides a method for detecting colorectal cancer sEV-specific surface biomarkers using the aptamer.

이때, 상기 방법은 상기 DNA 압타머와 대장 조직, 대장 세포, 혈액, 혈청, 혈장, 타액, 객담 및 뇨 등의 시료와 접촉시키는 단계를 포함할 수 있다. 상기 시료는 포유류, 바람직하게는 인체에서 분리된 것으로, 최소 침습으로 확보가능한 시료 또는 분비체액, in vitro 세포배양액 성분 시료 등일 수 있으나, 바람직하게는 대장암 세포 유래 sEV일 수 있으며, 대장암 마커를 포함하고 있을 수 있는 시료이면, 상기에 한정되지 않음은 자명하다. At this time, the method may include contacting the DNA aptamer with samples such as colon tissue, colon cells, blood, serum, plasma, saliva, sputum, and urine. The sample is isolated from a mammal, preferably from the human body, and may be a sample that can be obtained with minimal invasiveness, secreted body fluid, or in vitro cell culture component sample, but preferably may be sEV derived from colon cancer cells, and may be a colon cancer marker. It is obvious that the sample is not limited to the above as long as it is a sample that may be included.

구체적으로, 상기 DNA 압타머를 시료와 접촉시키는 경우, 시료 중 존재하는 대장암 세포 유래 sEV 내 존재하는 마커와 상기 DNA 압타머간 특이적인 결합이 일어나게 된다. 따라서, 형광 등으로 DNA 압타머를 표지하여 결합시킨 다음 신호의 발생 존부를 확인함으로써, 대장암을 검출할 수 있게 된다.Specifically, when the DNA aptamer is brought into contact with a sample, specific binding occurs between the marker present in sEVs derived from colon cancer cells present in the sample and the DNA aptamer. Therefore, it is possible to detect colon cancer by labeling the DNA aptamer with fluorescence, etc., binding it, and then checking whether a signal is generated.

시료 중 상기 DNA 압타머와 결합한 물질의 분석을 통하여, 대장암 바이오마커를 분리하여 낼 수 있는바, 본 발명은 상기 DNA 압타머를 이용한 대장암 세포 특이적 표면 바이오마커의 검출에 이용될 수 있다. Colon cancer biomarkers can be separated through analysis of substances bound to the DNA aptamer in the sample, and the present invention can be used to detect colon cancer cell-specific surface biomarkers using the DNA aptamer. .

본 발명은 또한, 상기 압타머를 포함하는 대장암 특이적 약물전달 조성물을 제공한다. 대장암 세포 유래 sEV에 압타머가 특이적으로 결합하여 sEV을 매개로 하여, 암의 진행을 저해하여 대장암의 치료에 이용할 수 있음은, 본 발명이 속하는 기술분야에서 공지된 사실이므로, 본 발명에 따른 압타머가 대장암 세포 유래 sEV에 특이적으로 결합하여 대장암 진행 기작을 저해할 것인 바, 이를 함유하는 조성물이 대장암 치료를 위한 조성물로 제공할 수 있음은 본 발명이 속하는 기술분야에서 당업자에게 자명하다. The present invention also provides a colon cancer-specific drug delivery composition containing the aptamer. It is a known fact in the art that aptamers can be used to treat colon cancer by specifically binding to sEVs derived from colon cancer cells and inhibiting the progression of cancer through sEVs. Therefore, the present invention Since the aptamer will specifically bind to sEV derived from colon cancer cells and inhibit the mechanism of colon cancer progression, it is known to those skilled in the art that a composition containing this can be provided as a composition for treating colon cancer. It is self-explanatory.

또한, 압타머를 리포좀이나 나노입자의 표면에 부착함으로써, 리포좀이나 나노입자 내부에 탑재된 항암제, 톡신, 암성장 저해 유전자, siRNA (small interfering RNA) 등을 대장암 세포 유래 sEV에 선택적으로 전달할 수 있다. 또한, 본 발명에 따른 압타머에 공지된 대장암 특이적 약물, 암세포 사멸을 유도하는 톡신 및 항암제, 또는 Herpes simplex virusthymidine kinase(HSV-TK), 사이토신 데아미네이즈(cytosine deaminase, CD) 등과 같은 공지된 자살 유전자, 또는 대장암 성장 및 전이에 중요한 역할을 하는 유전자의 발현을 저해하는 siRNA(small interfering RNA)를 직접 부착하여 대장암 세포 유래 sEV로 전달할 수 있는바, 본 발명은 상기 DNA 압타머를 함유하는 대장암 특이적 약물전달 조성물의 형태로 제공될 수 있다.In addition, by attaching aptamers to the surface of liposomes or nanoparticles, anticancer drugs, toxins, cancer growth inhibitory genes, siRNA (small interfering RNA), etc. loaded inside liposomes or nanoparticles can be selectively delivered to sEVs derived from colon cancer cells. there is. In addition, the aptamer according to the present invention includes known colon cancer-specific drugs, toxins and anticancer agents that induce cancer cell death, or Herpes simplex virusthymidine kinase (HSV-TK), cytosine deaminase (CD), etc. siRNA (small interfering RNA) that inhibits the expression of known suicide genes or genes that play an important role in colon cancer growth and metastasis can be directly attached and delivered to sEVs derived from colon cancer cells. The present invention provides the DNA aptamer It may be provided in the form of a colon cancer-specific drug delivery composition containing.

본 발명에 따르면, 대장암 세포 유래 세포에만 특이적으로 결합하는 압타머를 이용하여 대장암의 진단 및 치료에 이용할 수 있다.According to the present invention, an aptamer that specifically binds only to colon cancer cell-derived cells can be used for diagnosis and treatment of colon cancer.

도 1은 분리된 sEV의 특성화를 나타낸다. 도 1의 (A)는 SW620 sEV에 대한 NTA 결과(삽입: cryo-TEM 이미지), 도 1의 (B)는 HS(인간혈청; Human serum) sEV에 대한 NTA 결과. cryo-TEM 이미지의 스케일 바: 100 nm. 도 1의 (C)는 CD63, CD9, Hsp90α 및 칼넥신 발현의 웨스턴 블랏 분석 결과이다.Figure 1 shows characterization of isolated sEVs. Figure 1 (A) shows the NTA results for SW620 sEV (insert: cryo-TEM image), and Figure 1 (B) shows the NTA results for HS (human serum) sEV. Scale bar of cryo-TEM image: 100 nm. Figure 1 (C) shows the results of Western blot analysis of CD63, CD9, Hsp90α, and calnexin expression.

도 2는 각각 (A) E-SELEX, (B) E-SELEX의 워크플로우를 나타낸다.Figure 2 shows the workflow of (A) E-SELEX and (B) E-SELEX, respectively.

도 3은 대칭 및 비대칭 PCR 결과를 비교하기 위한 아가로스(2.5%) 겔 이미지를 나타낸다. M: 25/100 bp DNA 크기 마커; 1 및 2: 대칭 PCR에서 w/o 및 w/ssDNA 템플릿; 레인 3 & 4: 비대칭 PCR에서 w/o 및 w/ssDNA 템플릿; 레인 5: ssDNA 템플릿(비교군)Figure 3 shows an agarose (2.5%) gel image for comparing symmetric and asymmetric PCR results. M: 25/100 bp DNA size marker; 1 and 2: w/o and w/ssDNA templates in symmetric PCR; Lanes 3 & 4: w/o and w/ssDNA templates from asymmetric PCR; Lane 5: ssDNA template (comparison)

도 4는 qPCR을 사용한 E-SELEX 모니터링결과를 나타낸다. (A) 증폭 곡선. (B) 용융 곡선 분석.Figure 4 shows the E-SELEX monitoring results using qPCR. (A) Amplification curve. (B) Melting curve analysis.

도 5는 균질성 정도를 추정하기 위해 Fmax 최대 형광 강도에 대한 F20th 20 번째 주기 형광 강도의 비율 (F20th/Fmax)을 나타낸다.Figure 5 shows the ratio of F20th 20th cycle fluorescence intensity to Fmax maximum fluorescence intensity ( F20th / Fmax ) to estimate the degree of homogeneity.

도 6은 ELONA 방법을 사용한 압타머 평가결과를 나타낸다. 도 6의 (A)는 상위 10개 후보 압타머의 결합 친화도, 도 6의 (B)는 프라이머 영역이 있거나 없는 CCE-10의 결합 친화도를 나타낸다. 결합 친화도 분석에 사용된 압타머의 농도는 250 nM이었다.Figure 6 shows the results of aptamer evaluation using the ELONA method. Figure 6 (A) shows the binding affinity of the top 10 candidate aptamers, and Figure 6 (B) shows the binding affinity of CCE-10 with and without the primer region. The concentration of aptamer used in the binding affinity analysis was 250 nM.

도 7은 CCE-10F(A) 및 CCE-10FR(B)의 해리 상수(K d) 곡선을 나타낸다.Figure 7 shows the dissociation constant ( K d ) curves of CCE-10F (A) and CCE-10FR (B).

도 8은 최적화된 압타머 CCE-10F의 평가결과를 나타낸다. 도 8의 (A) 압타머의 예측된 2차 구조의 2차원 및 3차원 형태, 도 8의 (B) 임의의 ssDNA 라이브러리에 있는 것과 결합 친화도 비교, 도 8의 (C) 감도, 도 8의 (D) 특이성 평가 결과이며, 평가에 사용된 압타머의 농도는 10 nM이었다. 도 8의 (E)는 인간 혈청에서 압타머(Apt) 및 sEV 결합 압타머(Apt-Exosomes)의 안정성 분석결과이며, 각 시점에서 압타머의 밴드 세기(좌)와 각각의 안정성(우)을 보여주는 대표적인 겔 이미지이다.Figure 8 shows the evaluation results of the optimized aptamer CCE-10F. Figure 8 (A) Two-dimensional and three-dimensional shapes of the predicted secondary structure of the aptamer, Figure 8 (B) Binding affinity comparison with that in any ssDNA library, Figure 8 (C) Sensitivity, Figure 8 (D) This is the specificity evaluation result, and the concentration of the aptamer used in the evaluation was 10 nM. Figure 8 (E) shows the stability analysis results of aptamer (Apt) and sEV-bound aptamer (Apt-Exosomes) in human serum, showing the band intensity (left) and each stability (right) of the aptamer at each time point. This is a representative gel image shown.

도 9는 바이오마커 토폴로지 분석결과이다. I: 온전한 sEV, II: 프로테이나제 K(proK) 처리, III: 트립신 처리 sEV, IV: RIPA 처리 sEV.Figure 9 shows the results of biomarker topology analysis. I: intact sEV, II: proteinase K (proK) treated, III: trypsin treated sEV, IV: RIPA treated sEV.

도 10은 세포독성 분석 결과이다. 도 10의 (A) LS 174T, 도 10의 (B) HUVEC 에서의 분석 결과이다. Figure 10 shows the results of cytotoxicity analysis. This is the analysis result for LS 174T in Figure 10 (A) and HUVEC in Figure 10 (B).

도 11은 LS 174T에 의한 sEV 흡수 분석결과이다. 도 11은 각각 (A) 공초점 현미경 이미지, (B) 유세포 분석 결과; 대표적인 히스토그램(왼쪽) 및 형광 정량화(오른쪽)를 나타내며, 현미경 이미지의 스케일 바: 10 μm이다.Figure 11 shows the results of sEV absorption analysis by LS 174T. Figure 11 shows (A) confocal microscopy image, (B) flow cytometry result; Representative histograms (left) and fluorescence quantification (right) are shown, scale bar of microscopy images: 10 μm.

도 12는 sEV 및 Apt-sEV이 LS 174T의 이동 및 침입에 미치는 영향을 확인한 결과이다. 도 12에서 각각 (A) 0h 및 24h에서 상처 치유 분석의 대표적인 이미지(축척 막대: 200 μm), (B) 갭 폐쇄의 정량적 분석, (C) 48h에서 트랜스웰 마이그레이션 분석의 대표적인 이미지(스케일 막대: 100 μm), (D) 이동 영역의 정량적 분석, (E) 48시간에서 트랜스웰 침습 분석의 대표적인 이미지(스케일 막대: 100 μm), (F) 침입 지역의 정량적 분석 결과이다.Figure 12 shows the results confirming the effect of sEV and Apt-sEV on the migration and invasion of LS 174T. In Figure 12, (A) representative images of wound healing assay at 0 h and 24 h (scale bar: 200 μm), (B) quantitative analysis of gap closure, and (C) representative image of transwell migration assay at 48 h (scale bar: 100 μm), (D) quantitative analysis of migration area, (E) representative image of transwell invasion analysis at 48 hours (scale bar: 100 μm), (F) quantitative analysis of invasion area.

도 13은 HUVEC 혈관신생에 대한 sEV 및 Apt-sEV의 효과를 확인한 결과이다. 도 13에서 각각 (A) 4h에서 튜브 형성 분석의 대표적인 이미지(축척 막대: 100 μm), (B-C) 접합의 상대 수(B) 및 상대 총 분기 길이(C)의 정량적 분석 결과를 나타낸다.Figure 13 shows the results confirming the effect of sEV and Apt-sEV on HUVEC angiogenesis. Figure 13 shows (A) representative images of the tube formation assay at 4 h (scale bar: 100 μm), (B-C) quantitative analysis results of the relative number of junctions (B) and relative total branch length (C), respectively.

도 14는 분리된 sEV의 특성화를 나타낸다. 도 14의 (A)는 SW620 sEV에 대한 NTA 결과(삽입: cryo-TEM 이미지), 도 14의 (B)는 LS 174T sEV에 대한 NTA 결과(삽입: cryo-TEM 이미지), 도 14의 (C)는 HT29 sEV에 대한 NTA 결과(삽입: cryo-TEM 이미지), 도 14의 (D)는 HS sEV에 대한 NTA 결과(삽입: cryo-TEM 이미지)를 나타낸다. 도 14의 (E)는 CD63, Hsp90α 및 칼넥신 발현에 대한 웨스턴 블롯 분석결과이며, 각각의 cryo-TEM 이미지의 스케일 바: 100 nm이다.Figure 14 shows characterization of isolated sEVs. Figure 14 (A) shows NTA results for SW620 sEV (insert: cryo-TEM image), Figure 14 (B) shows NTA results for LS 174T sEV (insert: cryo-TEM image), Figure 14 (C) ) shows the NTA results for HT29 sEVs (insert: cryo-TEM image), and (D) in Figure 14 shows the NTA results for HS sEVs (insert: cryo-TEM image). Figure 14 (E) shows the results of Western blot analysis for the expression of CD63, Hsp90α, and calnexin, and the scale bar of each cryo-TEM image is 100 nm.

도 15는 본 발명의 일 실시예에 따라 수행된 Colorectal Cancer Toggle sEV-SELEX (CRC TEV-SELEX)의 워크플로우를 나타낸다.Figure 15 shows the workflow of Colorectal Cancer Toggle sEV-SELEX (CRC TEV-SELEX) performed according to an embodiment of the present invention.

도 16은 TEV-SELEX 모니터링 결과를 나타낸다. 도 16의 (A)는 증폭 곡선, 도 3의 (B) 무작위 라이브러리(Lib) 및 1~8번째 루프로부터 ssDNA의 균질성 정도를 추정하기 위한 Fmax 최대 형광 강도에 대한 F20th 20 번째 주기 형광 강도의 비율(F20th/Fmax)을 나타내며, 도 16의 (C)는 용융 분석 곡선이다. 도 16에서 모든 막대는 평균 ± SD(n=3)를 나타낸다.Figure 16 shows TEV-SELEX monitoring results. Figure 16 (A) is an amplification curve, Figure 3 (B) is the ratio of F20th 20th cycle fluorescence intensity to Fmax maximum fluorescence intensity for estimating the degree of homogeneity of ssDNA from random library (Lib) and 1st to 8th loops. (F 20th /F max ), and (C) in Figure 16 is the melt analysis curve. All bars in Figure 16 represent mean ± SD (n=3).

도 17는 본 발명에 따라 선정된 상위 10개 후보 압타머(T1 내지 T10)의 결합 친화도 분석 결과이다.Figure 17 shows the binding affinity analysis results of the top 10 candidate aptamers (T1 to T10) selected according to the present invention.

도 18은 본 발명에 따른 압타머 후보군의 결합 친화도 분석 결과이다. 도 18의 (A)는 T6 및 그 파생서열(T6F, T6R 및 T6FR)의 결합 친화도를, 도 18의 (B) 내지 (D)는 대항 표적(HS) 대비 양성 표적(SW620, LS 174T, HT29)에 대한 T6 및 그 파생서열의 비율과 함께 결합 친화도를 나타낸다.Figure 18 shows the results of binding affinity analysis of aptamer candidates according to the present invention. Figure 18 (A) shows the binding affinity of T6 and its derivative sequences (T6F, T6R, and T6FR), and Figure 18 (B) to (D) shows the binding affinity of the positive target (SW620, LS 174T, The binding affinity is shown along with the ratio of T6 and its derived sequences to HT29).

도 19는 본 발명에 따른 압타머와 그 파생서열들의 구조 모식도 및 결합 친화도를 나타낸다. 도 19의 (A) 및 (B)는 T6R 및 이의 잘린 버전인 파생서열(T6RA, T6RB)의 구조 및 결합 친화도를 나타내며, 도 19의 (C) 및 (D)는 T6RB 및 이의 잘린 버전인 파생서열(T6RB1, T6RB2, T6RB3)의 구조 및 결합 친화도를 나타낸다.Figure 19 shows the structural schematic diagram and binding affinity of the aptamer and its derived sequences according to the present invention. Figures 19 (A) and (B) show the structure and binding affinity of T6R and its truncated versions of derived sequences (T6RA, T6RB), and Figures 19 (C) and (D) show T6RB and its truncated versions. The structure and binding affinity of the derived sequences (T6RB1, T6RB2, T6RB3) are shown.

도 20은 최적의 압타머인 T6RB의 평가 결과를 나타낸다. 도 20의 (A) T6RB 해리 상수(K d) 곡선 및 그 값을, 도 20의 (B)는 칼륨 이온(K+)의 부재/존재에서 T6RB의 원형 이색성(CD) 스펙트럼을, 도 20의 (C)는 K+의 부재/존재에서 T6RB에 결합된 NMM의 형광 강도(FI)를, 도 20의 (D)는 바이오마커 토폴로지 분석결과이다.Figure 20 shows the evaluation results of T6RB, the optimal aptamer. Figure 20 (A) shows the T6RB dissociation constant ( K d ) curve and its value, Figure 20 (B) shows the circular dichroism (CD) spectrum of T6RB in the absence/presence of potassium ions (K + ), Figure 20 (C) shows the fluorescence intensity (FI) of NMM bound to T6RB in the absence/presence of K + , and (D) in Figure 20 shows the results of the biomarker topology analysis.

도 21은 TSA 반응을 위한 매개변수 최적화 결과이다. 도 21의 (A)는 압타머 농도, 도 21의 (B) 비오틴-티라미드(b-티라미드) 농도, 도 21의 (C)는 H2O2 농도, 도 21의 (D)는 TSA 반응 시간에 따른 최적화 결과를 나타낸다.Figure 21 shows parameter optimization results for TSA reaction. Figure 21 (A) is aptamer concentration, Figure 21 (B) biotin-tyramide (b-tyramide) concentration, Figure 21 (C) is H 2 O 2 concentration, Figure 21 (D) is TSA. Shows optimization results according to reaction time.

도 22는 T6RB 압타머의 평가결과를 나타낸다. 도 22에서는 ELONA와 TSA를 탑재한 ELONA를 비교하여 분석하였다.Figure 22 shows the evaluation results of T6RB aptamer. In Figure 22, ELONA and ELONA equipped with TSA are compared and analyzed.

도 23은 최적화된 조건에서 TSA에 의한 CRC sEV 검출을 확인한 결과이다. 도 23의 (A)는 SW620 sEV의 감도를, 도 23의 (B)는 LS 174T sEV의 감도를, 도 23의 (C)는 HT29 sEV의 감도를, 도 23의 (D)는 CRC, HS 및 정상 결장에서 유래된 sEV에 대한 T6RB의 특이성을 나타낸다.Figure 23 shows the results confirming CRC sEV detection by TSA under optimized conditions. Figure 23 (A) shows the sensitivity of SW620 sEV, Figure 23 (B) shows the sensitivity of LS 174T sEV, Figure 23 (C) shows the sensitivity of HT29 sEV, and Figure 23 (D) shows CRC, HS and specificity of T6RB for sEVs derived from normal colon.

이하, 실시예를 통하여 본 발명의 구성 및 효과를 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 예시하기 위한 것일 뿐, 본 발명의 범위가 이들 실시예에 의해 한정되는 것은 아니다.Hereinafter, the configuration and effects of the present invention will be described in more detail through examples. These examples are only for illustrating the present invention, and the scope of the present invention is not limited by these examples.

[실시예 1][Example 1]

[준비예][Preparation example]

세포 배양cell culture

SW620(인간 대장암), LS 174T(인간 대장암), CCD-18Co(인간 정상 결장), SKBR3(인간 유방암), U-87 MG(인간 교모세포종) 세포주는 한국 세포주 은행(KCLB)으로부터 획득했다. SW620, LS 174T 및 CCD-18Co 세포는 DMEM(Welgene, 한국 경산)에서 배양한 반면 SKBR3 및 U-87 MG 세포는 RPMI 1640 및 MEM에서 각각 배양하였다. 모든 세포주는 37℃, 5% CO2의 습한 조건의 10%(v/v) FBS(Gibco; Thermo Fisher Scientific, Waltham, MA, USA) 및 1%(v/v) 페니실린-스트렙토마이신(Welgene)이 보충된 각각의 배지에서 배양되었다. SW620 (human colon cancer), LS 174T (human colon cancer), CCD-18Co (human normal colon), SKBR3 (human breast cancer), and U-87 MG (human glioblastoma) cell lines were obtained from the Korea Cell Line Bank (KCLB). . SW620, LS 174T, and CCD-18Co cells were cultured in DMEM (Welgene, Gyeongsan, Korea), while SKBR3 and U-87 MG cells were cultured in RPMI 1640 and MEM, respectively. All cell lines were incubated with 10% (v/v) FBS (Gibco; Thermo Fisher Scientific, Waltham, MA, USA) and 1% (v/v) penicillin-streptomycin (Welgene) in humidified conditions at 37°C with 5% CO2. Cultured in each supplemented medium.

sEV의 분리Isolation of sEVs

약 80% confluence 상태에서, 세포를 5%(v/v) sEV가 고갈된 FBS(Gibco; Thermo Fisher Scientific) 및 1%(v/v) 페니실린-스트렙토마이신이 보충된 조건 배지로 교체하고 48시간 동안 37℃에서 가습된 조건의 5% CO2 인큐베이터에서 배양하였다. 분비된 sEV를 함유하는 조절 배지를 회수하여 sEV를 농축, 분리 및 정제하여 추가 사용을 하였다. 컨디셔닝된 배지는 먼저 5분 동안 300 x g, 20분 동안 2,000 x g 및 30분 동안 10,000 x g에서 일련의 원심분리를 수행하여 세포, 세포 파편, 미세소포체 및 세포자멸체를 각각 제거했다. 그 후, 0.45 및 0.22 μm의 기공 크기를 갖는 필터(Sartorius, Gφttingen, Germany)를 사용하여 상청액을 연속적으로 시린지 여과하여 200 nm보다 큰 크기의 세포외 소포체를 배제하였다. 농축 및 정제를 위해 배지가 원하는 부피로 농축될 때까지 300K 멤브레인(Pall Corporation, New York, NY, USA)이 있는 접선 흐름 여과 시스템(tangential flow filtration system)을 사용했다. 다음으로, 농축된 배지는 qEV 자동화 분획 수집기(Izon Science, Christchurch, NZ)에서 qEV10/35 nm 컬럼을 사용하여 크기 배제 크로마토그래피를 수행하여 sEV를 높은 수율 및 순도로 분리했다. 이어서, SEC의 sEV를 포함하는 분획을 풀링하고 3K 멤브레인이 있는 Macrosep(Pall Corporation)에서 5,000 x g에서 20분 동안 다시 원심분리하여 sEV를 추가로 농축한 다음, -80℃에서 PBS에 사용할 때까지 보관했다. 모든 원심분리 단계는 sEV 안정성을 보장하기 위해 4℃에서 수행하였다. 일련의 원심분리 전에 1X PBS로 희석한 것을 제외하고는 위에서 설명한 것과 동일한 방식으로 인간 혈청(HS)(Sigma-Aldrich, St Louis, MO, USA)으로부터 sEV를 분리하였다.At approximately 80% confluence, cells were replaced with conditioned medium supplemented with 5% (v/v) sEV-depleted FBS (Gibco; Thermo Fisher Scientific) and 1% (v/v) penicillin-streptomycin and incubated for 48 h. Cultured in a 5% CO 2 incubator under humidified conditions at 37°C. The conditioned medium containing the secreted sEVs was recovered, and the sEVs were concentrated, isolated, and purified for further use. The conditioned medium was first subjected to a series of centrifugations at 300 xg for 5 min, 2,000 xg for 20 min, and 10,000 xg for 30 min to remove cells, cell debris, microvesicles, and apoptotic bodies, respectively. Afterwards, the supernatant was sequentially syringe filtered using filters with pore sizes of 0.45 and 0.22 μm (Sartorius, Gϕttingen, Germany) to exclude extracellular vesicles larger than 200 nm in size. For concentration and purification, a tangential flow filtration system with a 300K membrane (Pall Corporation, New York, NY, USA) was used until the medium was concentrated to the desired volume. Next, the concentrated medium was subjected to size exclusion chromatography using a qEV10/35 nm column in a qEV automated fraction collector (Izon Science, Christchurch, NZ) to isolate sEVs in high yield and purity. Fractions containing sEVs from SEC were then pooled and centrifuged again at 5,000 x g for 20 min in a Macrosep with 3K membrane (Pall Corporation) to further concentrate sEVs and then stored in PBS at -80°C until use. did. All centrifugation steps were performed at 4°C to ensure sEV stability. sEVs were isolated from human serum (HS) (Sigma-Aldrich, St Louis, MO, USA) in the same manner as described above, except that they were diluted in 1× PBS before serial centrifugation.

실시예 1-1.Example 1-1.

SW620 및 HS 유래 sEV의 특성화Characterization of SW620 and HS-derived sEVs

SW620(양성 표적) 세포 및 건강한 인간 혈청(HS, 반대 표적)에서 분리된 sEV의 무결성 및 sEV 마커는 NTA, cryo-TEM 및 웨스턴 블롯 분석을 사용하여 평가하였다. The integrity of sEVs and sEV markers isolated from SW620 (positive target) cells and healthy human serum (HS, counter target) were assessed using NTA, cryo-TEM, and Western blot analysis.

나노입자 추적 분석(NTA)Nanoparticle Tracking Analysis (NTA)

NanoSight NS300 기기(Malvern Panalytical, Malvern, UK)를 사용하여 분리된 sEV의 농도 및 크기 분포를 분석했다. 레이저는 532nm로 설정되었으며 sCMOS 카메라를 사용하여 초당 25프레임으로 각 샘플에 대해 3개의 60초 비디오를 기록했다. 모든 비디오 녹화에 대해 카메라 수준은 잘 조정된 카메라 초점과 함께 14로 설정되었으며 감지 임계값은 5로 설정하였다. 입자의 브라운 운동은 NanoSight 소프트웨어(Malvern Panalytical)를 사용하여 평가하였다. NTA 설정은 샘플 간에 일관되게 유지되었고 샘플은 최적의 측정을 위해 PBS로 희석하였다.The concentration and size distribution of isolated sEVs were analyzed using a NanoSight NS300 instrument (Malvern Panalytical, Malvern, UK). The laser was set at 532 nm and three 60-second videos were recorded for each sample at 25 frames per second using an sCMOS camera. For all video recordings, the camera level was set to 14 with well-adjusted camera focus and the detection threshold was set to 5. Brownian motion of particles was evaluated using NanoSight software (Malvern Panalytical). NTA settings were kept consistent between samples and samples were diluted in PBS for optimal measurements.

저온 투과 전자 현미경(Cryo-TEM)Cryo-transmission electron microscopy (Cryo-TEM)

Cryo-TEM은 TEM 이미지에서 SW620 세포 및 HS 유래 sEV의 크기를 결정하는 데 사용하였다. 샘플 적용 전에 그리드를 글로우 방전하여 친수성으로 만들었다. 각 sEV 샘플(3 μL)을 구멍이 있는 탄소 코팅 구리 그리드(200 메쉬; Quantifoil, Großlφbichau, Germany)에 놓고 Thermo Scientific Vitrobot(Thermo Fisher Scientific)을 사용하여 액체 에탄에 넣고 온도를 유지하여 유리화했다. 액체 질소. 유리화 후 샘플을 액체 질소에 보관했다. 이미징 전에 저장된 샘플을 액체 질소를 사용하여 약 -180℃에서 온도를 지속적으로 유지하는 cryoholder로 옮겼다. Cryo-TEM 이미지는 CMOS 카메라와 결합된 JEM-2100PLUS 전자 현미경(JEOL, Tokyo, Japan)을 사용하여 25kV에서 얻었다.Cryo-TEM was used to determine the size of SW620 cells and HS-derived sEVs in TEM images. Before sample application, the grid was glow discharged to make it hydrophilic. Each sEV sample (3 μL) was placed on a perforated carbon-coated copper grid (200 mesh; Quantifoil, Großlϕbichau, Germany) and vitrified using a Thermo Scientific Vitrobot (Thermo Fisher Scientific) by placing it in liquid ethane and maintaining the temperature. liquid nitrogen. After vitrification, the samples were stored in liquid nitrogen. Before imaging, the stored samples were transferred to a cryoholder where the temperature was continuously maintained at approximately -180°C using liquid nitrogen. Cryo-TEM images were obtained at 25 kV using a JEM-2100PLUS electron microscope (JEOL, Tokyo, Japan) coupled with a CMOS camera.

웨스턴 블로팅Western blotting

제조업체의 지침에 따라 30K 필터(Merck Millipore, Burlington, MA, USA)가 있는 Amicon Ultra-0.5 장치를 사용하여 동일한 부피의 SW620 세포 및 HS 유래 sEV를 농축했다. 각 샘플에 5x SDS-PAGE loading buffer (Biosesang, Seongnam, Republic of Korea)를 보충하고, 95℃에서 10분간 가열하고, Mini-Protean tetra 시스템(Bio-Rad Laboratories)을 사용하여 300V에서 20분 동안 10% TGX stain-free protein gel (Bio-Rad Laboratories, Hercules, CA, USA)에 용해하였다. 분해된 단백질을 제조업체의 지침에 따라 Trans-Blot Turbo Transfer System(Bio-Rad Laboratories)에서 0.2μm PVDF 멤브레인(Bio-Rad Laboratories)으로 옮겼다. 멤브레인을 0.1% TBST(0.1% Tween-20이 포함된 1x TBS)로 용해한 5% BSA TBST 용액으로 블랑킹하고, 1차 항체[마우스 항-CD63(Santa Cruz, Dallas, TX, USA), 마우스 항-CD9, 토끼 항- Hsp90α 및 토끼 항 칼넥신(ABclonal, Woburn, MA, USA), 1:1,000] Can Get Signal 용액 1(Toyobo, New York, NY, USA)와 함께 4℃에서 밤새 인큐베이션한 다음 0.1% TBST로 3회 세척하였다. 그런 다음 멤브레인을 Can Get Signal 용액 2(Toyobo)에 희석된 해당 HRP-접합 2차 항체[HRP 염소 항-토끼 IgG(ABclonal), 1:10,000 및 HRP 염소 항-마우스 IgG(Biolegend, San Diego, CA, USA), 1:5000]와 함께 실온에서 1시간 동안 인큐베이션하고, 0.1% TBST로 4회 세척했다. 멤브레인의 단백질은 ChemiDoc Imaging System(Bio-Rad Laboratories)에서 Clarity Western ECL 기질(Bio-Rad Laboratories)을 사용하여 시각화하였다.Equal volumes of SW620 cells and HS-derived sEVs were concentrated using an Amicon Ultra-0.5 device with a 30K filter (Merck Millipore, Burlington, MA, USA) according to the manufacturer's instructions. Each sample was supplemented with 5x SDS-PAGE loading buffer (Biosesang, Seongnam, Republic of Korea), heated at 95°C for 10 min, and incubated at 300 V for 10 min for 20 min using the Mini-Protean tetra system (Bio-Rad Laboratories). % TGX stain-free protein gel (Bio-Rad Laboratories, Hercules, CA, USA). Digested proteins were transferred to 0.2 μm PVDF membranes (Bio-Rad Laboratories) in the Trans-Blot Turbo Transfer System (Bio-Rad Laboratories) according to the manufacturer's instructions. Membranes were blanked with 5% BSA TBST solution dissolved in 0.1% TBST (1x TBS with 0.1% Tween-20) and incubated with primary antibody [mouse anti-CD63 (Santa Cruz, Dallas, TX, USA), mouse anti-CD63 (Santa Cruz, Dallas, TX, USA). -CD9, rabbit anti-Hsp90α and rabbit anti-calnexin (ABclonal, Woburn, MA, USA), 1:1,000] followed by incubation with Can Get Signal solution 1 (Toyobo, New York, NY, USA) overnight at 4°C. Washed three times with 0.1% TBST. The membrane was then incubated with the corresponding HRP-conjugated secondary antibodies [HRP goat anti-rabbit IgG (ABclonal), 1:10,000 and HRP goat anti-mouse IgG (Biolegend, San Diego, CA) diluted in Can Get Signal solution 2 (Toyobo). , USA), 1:5000] for 1 hour at room temperature and washed four times with 0.1% TBST. Proteins on the membrane were visualized using Clarity Western ECL substrate (Bio-Rad Laboratories) on a ChemiDoc Imaging System (Bio-Rad Laboratories).

그 결과, 도 1의 (A) 및 1의 (B)에 표시된 바와 같이 SW620 및 HS sEV의 크기는 각각 115 및 129 nm이며, 이는 cryo-TEM을 사용하여 얻은 값과 잘 일치하였다. 또한 SW620 및 HS 유래 sEV는 모두 원형 형태를 보였고 이전 문헌과 일치하는 크기 범위 내에 존재하여 sEV가 SEC 방법을 사용하여 효과적으로 분리되었음을 확인했다. 또한, 도 1의 (C)의 웨스턴 블롯 이미지는 분리된 sEV가 잘 알려진 sEV 바이오마커인 CD63, CD9, Hsp90α에 대해 양성이고, 소포체(ER) 막에 대한 마커인 칼넥신에 대해 음성임을 확인하였다. 이러한 모든 분석을 통해 분리된 sEV이 E-SELEX에 적합한 표적임을 확인했다.As a result, as shown in Figures 1(A) and 1(B), the sizes of SW620 and HS sEV were 115 and 129 nm, respectively, which matched well with the values obtained using cryo-TEM. Additionally, both SW620- and HS-derived sEVs showed circular morphology and were within a size range consistent with previous literature, confirming that sEVs were effectively isolated using the SEC method. In addition, the Western blot image in Figure 1 (C) confirmed that the isolated sEVs were positive for well-known sEV biomarkers CD63, CD9, and Hsp90α, and negative for calnexin, a marker for the endoplasmic reticulum (ER) membrane. . All these analyzes confirmed that isolated sEV was a suitable target for E-SELEX.

실시예 1-2.Example 1-2.

SW620 유래 sEV에 대한 압타머의 시험관 내 선택In vitro selection of aptamers against SW620-derived sEVs

도 2에서 볼 수 있듯이 E-SELEX의 중요한 단계는 결합한 압타머 풀과 결합하지 않은 압타머 풀을 효율적으로 분리하는 것이다. 이를 위해 생물학적으로 활성인 sEV의 무결성을 유지할 수 있을 뿐만 아니라 진단 가치가 있는 고품질 압타머의 선택을 용이하게 할 수 있는 손쉬운 표적 sEV 고정화 플랫폼으로 면역 플레이트를 사용했다.As can be seen in Figure 2, an important step in E-SELEX is to efficiently separate the bound and unbound aptamer pools. To this end, we used immunoplates as a facile targeted sEV immobilization platform that can not only maintain the integrity of biologically active sEVs but also facilitate the selection of high-quality aptamers with diagnostic value.

sEV-SELEX(E-SELEX)sEV-SELEX (E-SELEX)

사용된 모든 올리고뉴클레오티드는 Integrated DNA Technologies(IDT; Coralville, IA, USA)에 의해 합성되었다. DNA 라이브러리에 포함된 DNA 형태와 사용되는 정방향 및 역방향 프라이머의 형태는 하기 표 1과 같다.All oligonucleotides used were synthesized by Integrated DNA Technologies (IDT; Coralville, IA, USA). The types of DNA included in the DNA library and the types of forward and reverse primers used are shown in Table 1 below.

Sequence (5'→3')Sequence (5'→3') 서열번호sequence number Random libraryRandom library ATCCAGAGTGACGCAGCA-N40-CTGGCTCGAACAAGCTTGCATCCAGAGTGACGCAGCA-N 40 -CTGGCTCGAACAAGCTTGC 1One Forward primerForward primer ATCCAGAGTGACGCAGCAATCCAGAGTGACGCAGCA 22 Reverse primerReverse primer GCAAGCTTGTTCGAGCCAGGCAAGCTTGTTCGAGCCAG 33

PCR 증폭 단계에 사용된 양 말단 프라이머 영역 사이에 40개의 뉴클레오티드(N40)의 중앙 랜덤 영역으로 랜덤 라이브러리를 설계하였다. SW620 세포 유래 sEV 및 정상 인간 혈청(HS) 유래 sEV을 각각 양성 및 대항 표적으로 사용하였고, E-SELEX 과정 전반에 걸쳐 맥시 결합 면역 플레이트를 사용하여 선별을 수행하였다. 표적 sEV과 함께 인큐베이션하기 전에, 결합 완충액(BB, 1 g/L BSA, 5 mM MgCl2 및 0.5 mg/mL 연어 정자 ssDNA가 포함된 DPBS)에 용해된 초기 단일 가닥 DNA(ssDNA) 라이브러리(100 nM)를 95℃에서 5분동안 변성하였고, 10분 동안 얼음에서 급속 냉각하였다. A random library was designed with a central random region of 40 nucleotides (N40) between the primer regions at both ends used in the PCR amplification step. SW620 cell-derived sEVs and normal human serum (HS)-derived sEVs were used as positive and counter targets, respectively, and selection was performed using maxi-binding immunoplates throughout the E-SELEX process. Prior to incubation with target sEVs, a nascent single-stranded DNA (ssDNA) library (100 nM ) was denatured at 95°C for 5 minutes and rapidly cooled on ice for 10 minutes.

음성 선별은 BSA 및 면역 플레이트에 결합할 가능성이 있는 ssDNA를 제거하기 위해 먼저 수행하였다. 1g/L의 BSA 용액을 면역 플레이트에 접종하고 37℃에서 2시간 동안 인큐베이션했다. 세척 완충액(WB; DPBS with 5 mM MgCl2)으로 각각 30초 동안 진탕기(500 rpm)에서 3회 세척한 후, 준비된 ssDNA 라이브러리를 첨가하고 1시간 동안 배양하였다. 그 후, 결합되지 않은 ssDNA를 포함하는 상층액을 회수하고 제조업체의 지침에 따라 Nucleospin Gel 및 PCR clean-up(Macherey-Nagel, Duren, Germany)을 사용하여 농축했다. Negative selection was performed first to remove ssDNA likely to bind to BSA and immune plates. 1 g/L of BSA solution was inoculated into the immune plate and incubated at 37°C for 2 hours. After washing three times with washing buffer (WB; DPBS with 5 mM MgCl 2 ) on a shaker (500 rpm) for 30 seconds each, the prepared ssDNA library was added and incubated for 1 hour. Afterwards, the supernatant containing unbound ssDNA was recovered and concentrated using Nucleospin Gel and PCR clean-up (Macherey-Nagel, Duren, Germany) according to the manufacturer's instructions.

그런 다음 회수된 ssDNA를 프라이머 농도의 비율이 다른 대칭 PCR(Asy-PCR)을 사용하여 증폭했다. 간단히 말해서, 0.2 mM dNTP, 각 선택에서 회수된 ssDNA 주형, 충실도가 높은 nPfu-forte DNA 중합효소 및 1x nPfu forte에서 20:1의 정방향-역방향 프라이머 비율을 포함하는 Asy-PCR 반응용 혼합물 버퍼(Enzynomics, 대전, 대한민국)를 준비하였다. The recovered ssDNA was then amplified using symmetric PCR (Asy-PCR) with different ratios of primer concentrations. Briefly, mixture buffer for Asy-PCR reactions containing 0.2 mM dNTPs, ssDNA template recovered from each selection, high-fidelity nPfu-forte DNA polymerase, and a forward-to-reverse primer ratio of 20:1 in 1x nPfu forte (Enzynomics , Daejeon, Republic of Korea).

열 순환 프로토콜은 다음과 같이 설정하였다: 95℃에서 3분 동안 초기 변성, 이어서 95℃에서 10초(변성), 68℃에서 1분(어닐링 및 신장), 최종 연장의 20주기 68℃에서 5분. 다음으로, Asy-PCR 산물을 2.5% agarose gel에서 분리하고 Nucleospin gel과 PCR clean-up을 이용하여 ssDNA band를 추출 및 정제하였다. 양성 선별을 위해 SW620 sEV(20 μg/mL))을 면역 플레이트에서 37℃에서 2시간 동안 배양한 다음 WB로 세척(3회)했다. 이어서, 음성 선별으로부터 얻은 ssDNA를 웰에 첨가한 다음, 37℃에서 1시간 동안 배양하고 결합되지 않은 ssDNA를 제거하기 위해 WB로 세척(3회)하였다. 다음으로, 각 웰의 샘플을 BB에 현탁시키고 10분 동안 95℃로 가열하여 sEV에서 결합된 ssDNA를 회수한 다음 이전에 설명한 대로 처리했다. ssDNA 풀과 함께 HS 유래 sEV의 인큐베이션에 의존하는 대항 선별도 음성 선별과 동일한 방식으로 수행하여 정상 sEV에 대한 ssDNA 결합을 제거했다. SW620 sEV에 대해서만 표적으로 하는 압타머 풀의 최대 결합 능력을 보장하기 위해 [표 2]에 설명된 대로 연속 E-SELEX 루프에서 엄격한 조건으로 진행하였다. The thermal cycling protocol was set up as follows: initial denaturation at 95°C for 3 min, followed by 20 cycles of 10 s at 95°C (denaturation), 1 min at 68°C (annealing and elongation), and a final extension of 5 min at 68°C. . Next, the Asy-PCR product was separated on a 2.5% agarose gel, and the ssDNA band was extracted and purified using Nucleospin gel and PCR clean-up. For positive selection, SW620 sEV (20 μg/mL)) were incubated in immunoplates at 37°C for 2 h and then washed (three times) by WB. Then, ssDNA from negative selection was added to the wells, then incubated at 37°C for 1 hour and washed (three times) with WB to remove unbound ssDNA. Next, samples from each well were suspended in BB and heated to 95°C for 10 min to recover bound ssDNA from sEVs and then processed as previously described. Counter selection, which relies on incubation of HS-derived sEVs with ssDNA pools, was also performed in the same manner as negative selection to eliminate ssDNA binding to normal sEVs. To ensure maximum binding capacity of the aptamer pool targeting only SW620 sEV, stringent conditions were performed in continuous E-SELEX loops as described in [Table 2].

양성 선별positive screening 음성 선별voice screening 배양시간Incubation time 1 h (A)*
40 min (B)
20 min (C)
1 h (A) *
40min (B)
20min (C)
1 h (A)
1.5 h (B)
2 h (C)
1 h (A)
1.5 h (B)
2 h (C)
세척
(30 s, 500 rpm)
wash
(30 s, 500 rpm)
3 times (A)
6 times (B)
10 times (C)
3 times (A)
6 times (B)
10 times (C)
No Washing requiredNo washing required
Competitor
(Salmon sperm DNA)
Competitor
(Salmon sperm DNA)
0.5 mg/ml (A)
1.0 mg/ml (B)
1.5 mg/ml (C)
0.5 mg/ml (A)
1.0 mg/ml (B)
1.5 mg/ml (C)

*: A-C는 E-SELEX루프 사이클에서의 각 단계를 의미한다. * : AC refers to each step in the E-SELEX loop cycle.

E-SELEX 절차(도 2)에서 음성 선별은 버퍼 및/또는 면역 플레이트 자체의 구성 요소에 비특이적으로 결합하는 표적 외 단일 가닥 DNA(ssDNA)를 제거하기 위해 수행하였다. 음성 선별에서 얻은 결합되지 않은 ssDNA는 3개의 E-SELEX 루프를 포함하는 추가 선택을 위해 강화되었으며, 여기서 각 루프(A-, B- 및 C-루프로 표시됨)는 4개의 연속적인 양성 선별 및 1번의 대항 선별을 포함했다. 높은 전이 가능성을 고려하여 SW620 세포 유래 sEV을 양성 선별 대상으로 선택하였고, HS 유래 sEV은 잠재적인 생체 내 적용을 위한 임상적 환경을 설정하기 위해 대항 선별 대상으로 결정하였다. 루프는 총 15개의 개별 선택 단계로 세 번 반복되었으며, 그 동안 압타머의 결합 친화도 및 특이성을 개선하기 위해 엄격한 조건을 점진적으로 증가시켜 수행하였다(표 2).Negative selection in the E-SELEX procedure (Figure 2) was performed to remove non-target single-stranded DNA (ssDNA) that binds nonspecifically to components of the buffer and/or the immune plate itself. Unbound ssDNA obtained from negative selection was enriched for further selection containing three E-SELEX loops, where each loop (denoted A-, B-, and C-loop) was used for four consecutive positive selections and one It included one counter-screening. Considering the high metastatic potential, SW620 cell-derived sEVs were selected as a positive selection target, and HS-derived sEVs were selected as a counter-selection target to establish a clinical environment for potential in vivo application. The loop was repeated three times for a total of 15 individual selection steps, during which the stringency conditions were gradually increased to improve the binding affinity and specificity of the aptamer (Table 2).

또한, 자기 분리, 효소 분해 또는 추가 NaOH 처리를 위한 비오틴 또는 인산염 그룹 접합을 필요로 하지 않는 Asy-PCR을 사용하여 ssDNA 증폭과 생성을 한 단계로 수행하여 E-SELEX의 전체 프로세스를 단순화하였다. 최적화된 조건에서 20:1의 정방향-역방향 프라이머 비율을 사용한 Asy-PCR을 겔 전기영동 분석을 통해 프라이머 농도가 균일한 대칭 PCR과 비교했다. 도 3의 결과에서 dsDNA 밴드와 구별되는 ssDNA 밴드에 의해 입증된 바와 같이, Asy-PCR의 경우에는 ssDNA가 생성되는 것을 확인하였고, ssDNA 생성과정으로 Asy-PCR의 적합성을 재증명하였다. Additionally, the overall process of E-SELEX was simplified by performing ssDNA amplification and generation in one step using Asy-PCR, which does not require magnetic separation, enzymatic digestion, or biotin or phosphate group conjugation for additional NaOH treatment. Asy-PCR using a forward-to-reverse primer ratio of 20:1 under optimized conditions was compared with symmetric PCR with uniform primer concentration through gel electrophoresis analysis. As evidenced by the ssDNA band distinct from the dsDNA band in the results of Figure 3, it was confirmed that ssDNA was generated in the case of Asy-PCR, and the suitability of Asy-PCR was re-proven through the ssDNA generation process.

E-SELEX의 성공을 위한 또 다른 중요한 고려 사항은 E-SELEX 절차를 모니터링하여 수행되는 루프 수를 결정하는 것이다. qPCR은 다양한 서열의 DNA 풀에 대해 연구할 수 있고, ssDNA pool 다양성 측면에서 E-SELEX를 통해 회수한 ssDNA의 균질성에 대한 중요한 정보를 제공한다. 각 루프(A-C)에서 얻는 ssDNA를 qPCR로 분석하여 루프 수를 선정하였다 Another important consideration for the success of E-SELEX is monitoring the E-SELEX procedure to determine the number of loops performed. qPCR can study DNA pools of diverse sequences and provides important information about the homogeneity of ssDNA recovered through E-SELEX in terms of ssDNA pool diversity. The number of loops was selected by analyzing ssDNA obtained from each loop (A-C) by qPCR.

정량적 중합효소 연쇄 반응(qPCR)Quantitative polymerase chain reaction (qPCR)

qPCR을 사용하여 E-SELEX 동안 선택 효율성을 평가했다. 각 선택에서 회수된 ssDNA 주형과 1X TOPreal qPCR 프리믹스(Enzynomics)에서 1:1의 정방향-역방향 프라이머 비율을 포함하는 혼합물에서 PCR을 수행했다. 열 순환 프로토콜은 다음과 같이 설정하였다: 95℃에서 15분 동안 초기 변성, 이어서 95℃에서 10초(변성), 60℃에서 15초(어닐링), 72℃에서 15초의 20주기 (연장). 그 다음에는 0.5℃의 증분 속도로 55℃에서 95℃로 온도를 점진적으로 증가시키면서 용융 온도(Tm) 분석이 이어졌다.Selection efficiency was assessed during E-SELEX using qPCR. PCR was performed on a mixture containing the ssDNA template recovered from each selection and a forward-reverse primer ratio of 1:1 in 1X TOPreal qPCR premix (Enzynomics). The thermal cycling protocol was set up as follows: initial denaturation at 95°C for 15 min, followed by 20 cycles of 10 s at 95°C (denaturation), 15 s at 60°C (annealing), and 15 s at 72°C (extension). This was followed by melting temperature (Tm) analysis while gradually increasing the temperature from 55°C to 95°C at an increment rate of 0.5°C.

ssDNA의 초기 라이브러리는 40개 뉴클레오티드의 무작위 영역으로 인해 서열이 매우 이질적이다. 그러나 표적에 특이적으로 결합하는 압타머 풀을 지속적으로 반복적인 선택 과정이 수행될 것이기 때문에 이질성은 감소될 것이다. 이러한 예상되는 동작은 증폭 및 용융 곡선의 변화를 통해 입증하였다(도 4). 도 4A에서 랜덤 라이브러리(적색선; Lib)의 증폭은 초기 증가를 보여 최대 형광 강도에 도달하지만, 프라이머 영역은 완전히 혼성화되는 반면, 중앙의 무작위 영역은 부분적으로 비상보적 혼성화를 이루게 되어 불안정한 헤테로듀플렉스의 형성으로 인해 급격한 형광 감소가 나타났다. 이러한 결과는 도 4의 (B)에서 69℃의 낮은 용융 온도(Tm)는 헤테로듀플렉스의 불안정성과 잘 일치하였음을 나타낸다. 대조적으로, E-SELEX의 반복적인 루프는 높은 결합 친화도로 결합하는 특정 ssDNA 풀을 풍부하게 하여 균질성을 증가시켰다. 따라서 C-loop(파란색 선) 후 얻은 ssDNA는 qPCR 동안 극한 안정기를 갖는 정상 지수 곡선을 나타냈고 Tm 피크는 낮은 온도에서 Tm 피크가 80.5℃의 더 높은 온도로 완전히 이동하였다. 이는 높은 안정성을 가지는 호모듀플렉스의 비율이 크게 증가했음을 의미한다.The initial library of ssDNA is highly heterogeneous in sequence due to a random region of 40 nucleotides. However, heterogeneity will be reduced because an iterative process of selecting a pool of aptamers that specifically bind to the target will be continuously performed. This expected behavior was demonstrated through changes in amplification and melting curves (Figure 4). In Figure 4A, amplification of the random library (red line; Lib) shows an initial increase and reaches maximum fluorescence intensity, but while the primer region hybridizes completely, the central random region partially undergoes noncomplementary hybridization, resulting in the formation of an unstable heteroduplex. This resulted in a rapid decrease in fluorescence. These results indicate that the low melting temperature (Tm) of 69°C in Figure 4 (B) is consistent with the instability of the heteroduplex. In contrast, the repetitive loops of E-SELEX increased homogeneity by enriching specific ssDNA pools that bind with high binding affinity. Therefore, the ssDNA obtained after C-loop (blue line) showed a normal exponential curve with an extreme plateau during qPCR, and the Tm peak was completely shifted from the lower temperature to the higher temperature of 80.5°C. This means that the proportion of homoduplexes with high stability has increased significantly.

또한, 균질성의 정도를 추정하기 위해 최종 qPCR 사이클에서 최대 형광 강도에 대한 마지막 사이클 (20th)의 형광 강도의 비율을 계산하여 증폭 곡선을 정량적으로 분석했다. 이 연구에서는 0.95보다 높은 비율을 증폭의 안정기로 간주하였고, ssDNA 라이브러리에서 높은 균질성을 보장하기에 충분했다. 도 5에서 무작위 ssDNA 라이브러리(Lib)는 가장 낮은 비율을 보여 상당한 이질성을 반영하지만 E-SELEX 루프가 지속적으로 반복될수록 비율이 증가한다. C-loop 단독의 비율은 0.95보다 큰 값을 보여, ssDNA 균질성의 증가로 인해 C-loop가 증폭에서 안정기에 도달했음을 증명했다. Additionally, to estimate the degree of homogeneity, amplification curves were analyzed quantitatively by calculating the ratio of the fluorescence intensity in the last cycle ( 20th ) to the maximum fluorescence intensity in the final qPCR cycle. In this study, a ratio higher than 0.95 was considered a plateau of amplification and was sufficient to ensure high homogeneity in ssDNA libraries. In Figure 5, the random ssDNA library (Lib) shows the lowest ratio, reflecting significant heterogeneity, but the ratio increases as the E-SELEX loop is continuously repeated. The ratio of C-loop alone showed a value greater than 0.95, proving that C-loop reached a plateau in amplification due to the increase in ssDNA homogeneity.

qPCR 모니터링의 이러한 모든 결과는 C-루프가 ssDNA 풀의 최대 농축을 달성하는 데 최적임을 보여주었다. 따라서 C 루프에서 얻은 ssDNA를 NGS 분석에 적용했다.All these results from qPCR monitoring showed that the C-loop was optimal to achieve maximum enrichment of the ssDNA pool. Therefore, ssDNA obtained from the C loop was subjected to NGS analysis.

실시예 1-3.Example 1-3.

후보 압타머 평가Candidate aptamer evaluation

C-loop에서 얻은 ssDNA를 NGS로 분석하고 빈도수(%)를 기준으로 상위 10개 후보 압타머를 선택하여 추가 평가를 수행했다. The ssDNA obtained from the C-loop was analyzed by NGS, and the top 10 candidate aptamers were selected based on frequency (%) for further evaluation.

차세대 시퀀싱(NGS)Next-generation sequencing (NGS)

3개의 E-SELEX 루프 후, 농축된 ssDNA 풀을 프라이머로 대칭적으로 PCR 증폭하여 dsDNA를 생성하고, 아가로스 겔(2.5%)에서 분해하고, Nucleospin Gel 및 PCR clean-up을 사용하여 정제했다. 마지막으로 정제된 dsDNA를 처리하여 후보 압타머의 서열을 NGS(next-generation sequencing)를 통해 분석하였으며, 이는 시퀀싱 업체(Clinomics, Ulsan, Republic of Korea)에서 수행하였다.After three E-SELEX loops, the concentrated ssDNA pool was symmetrically PCR amplified with primers to generate dsDNA, resolved on an agarose gel (2.5%), and purified using Nucleospin Gel and PCR clean-up. Finally, the purified dsDNA was processed and the sequence of the candidate aptamer was analyzed through NGS (next-generation sequencing), which was performed by a sequencing company (Clinomics, Ulsan, Republic of Korea).

상위 10개 후보 압타머의 서열은 하기 [표 3]과 같다. The sequences of the top 10 candidate aptamers are shown in [Table 3] below.

Sequence (5'→3')*# Sequence (5'→3') *# ReadRead Frequency (%)Frequency (%) 서열번호sequence number CCE-1CCE-1 CCCCGAAGCCTCCTATTCTTAAATTTCCCCATCTTTCTCACCCCGAAGCCTCCTATTCTTAAATTTCCCCATCTTTCTCA 345188345188 2.182.18 44 CCE-2CCE-2 GGCGGAGTAGGGATAAACGGCAGGAAGAGGTGACAGGATGGGCGGAGTAGGGATAAACGGCAGGAAGAGGTGACAGGATG 285719285719 1.811.81 55 CCE-3CCE-3 GTGAGAAAGATGGGGAAATTTAAGAATAGGAGGCTTCGGGGTGAGAAAGATGGGGGAAATTTAAGAATAGGAGGCTTCGGG 283314283314 1.791.79 66 CCE-4CCE-4 CCACCGCCGTGAACTAACGTTGGTATTCTCCTCTCCCTTACCACCGCCGTGAACTAACGTTGGTATTCTCCTCTCCCTTA 225555225555 1.431.43 77 CCE-5CCE-5 CCACCGGCCGACATAGTCATATCTGACCTCTACACTCTCACCACCGGCCGACATAGTCATATCTGACCTCTACACTCTCA 200686200686 1.341.34 88 CCE-6CCE-6 CCACATCCCCACAACACCGTCACATCCCCTCATTTCACTGCCACATCCCCACAACACCGTCACATCCCCTCATTTCACTG 211848211848 1.271.27 99 CCE-7CCE-7 GACAAAAGAGTTCATGGAAGAAAGGAGAGGAGCCTAGGGTGACAAAAGAGTTCATGGAAGAAAGGAGAGGAGCCTAGGGT 199772199772 1.261.26 1010 CCE-8CCE-8 CCACCCTATCACTTCTTCATTAAACCGTACCCTCCCTACGCCACCCTATCACTTCTTCATTAAACCGTACCCTCCCTACG 185809185809 1.171.17 1111 CCE-9CCE-9 CCATCCTGTCACCTCTTCCTGCCGTTTATCCCTACTCCGCCCATCCTGTCACCTCTTCCTGCCGTTTATCCCTACTCCGC 182099182099 1.151.15 1212 CCE-10CCE-10 GGCCTAAGAAGGGACAGGGTGACAATCAGGGGGAGAGGGGGGCCTAAGAAGGGACAGGGTGACAATCAGGGGGGAGAGGGG 181328181328 1.151.15 1313

프라이머 영역은 E-SELEX 과정 동안 PCR 증폭시키기 위하여 상기 압타머 서열의 5' 부분에는 서열번호 2의 정방향 프라이머가, 3' 부분에는 서열번호 3의 역방향 프라이머 영역이 ssDNA 라이브러리에 포함되었으나, [표 3]에서 정방향과 역방향 프라이머 부분은 표시되지 않았다. CCE는 colorectal cancer exosome aptamer의 약자이다. 높은 결합 친화도 및 특이성을 갖는 압타머를 선택하기 위해, 5'-비오틴 그룹으로 변형된 후보 압타머를 제조한 후, SW620(양성표적) 및 HS(대항표적) sEV에 대한 결합 친화도는 ELONA 방법을 사용하여 평가하였다 In order to PCR amplify the primer region during the E-SELEX process, the forward primer of SEQ ID NO: 2 was included in the 5' part of the aptamer sequence, and the reverse primer of SEQ ID NO: 3 was included in the 3' part of the ssDNA library [Table 3 ], the forward and reverse primer portions are not indicated. CCE stands for colorectal cancer exosome aptamer. To select aptamers with high binding affinity and specificity, candidate aptamers modified with 5'-biotin groups were prepared, and then the binding affinities for SW620 (positive target) and HS (countertarget) sEV were measured using ELONA. It was evaluated using the method

효소 결합 올리고뉴클레오티드 분석(ELONA)Enzyme-linked oligonucleotide assay (ELONA)

맥시 결합 면역 플레이트(SPL) 웰을 37℃에서 2시간 동안 sEV(1 x 108 입자)으로 먼저 코팅한 다음, 0.1% PBST(0.1% Tween-20이 포함된 1x PBS)로 세 번 세척했다. 그 후, 면역 플레이트를 0.1% PBST에 3% BSA로 37℃에서 2시간 동안 블락킹 후, 0.1% PBST로 3회 세척하였다. 그런 다음 면역 플레이트를 준비된 비오틴 접합 압타머(IDT)와 함께 BB에서 37℃에서 1시간 동안 인큐베이션한 후 0.1% PBST로 3회 세척했다. 면역 플레이트를 0.1% PBST에 희석된 streptavidin-peroxidase polymer(Sigma-Aldrich; 1:1,000)와 함께 37℃에서 1시간 동안 추가로 인큐베이션한 다음, 0.1% PBST로 세척(3회)하였다. 다음으로, 1x TMB 기질 용액(Invitrogen, Carlsbad, CA, USA)을 각 웰에 첨가하고, 암실 및 실온에서 6분 동안 인큐베이션하였다. 0.5 M 황산을 첨가하여 반응을 종료하고, Spectramax iD5 multi-mode microplate reader(Molecular Devices, San Jose, CA, USA)를 이용하여 450 nm에서 흡광도를 즉시 측정하였다. 정규화 값 ΔA는 다음 방정식을 기반으로 계산하였다. Maxi-binding immunoplate (SPL) wells were first coated with sEVs (1 x 10 8 particles) for 2 hours at 37°C and then washed three times with 0.1% PBST (1x PBS with 0.1% Tween-20). Afterwards, the immune plate was blocked with 3% BSA in 0.1% PBST at 37°C for 2 hours and then washed three times with 0.1% PBST. The immune plate was then incubated with the prepared biotin-conjugated aptamer (IDT) in BB at 37°C for 1 h and then washed three times with 0.1% PBST. The immune plate was further incubated with streptavidin-peroxidase polymer (Sigma-Aldrich; 1:1,000) diluted in 0.1% PBST for 1 hour at 37°C and then washed (three times) with 0.1% PBST. Next, 1x TMB substrate solution (Invitrogen, Carlsbad, CA, USA) was added to each well and incubated for 6 min in the dark and at room temperature. The reaction was terminated by adding 0.5 M sulfuric acid, and the absorbance was immediately measured at 450 nm using a Spectramax iD5 multi-mode microplate reader (Molecular Devices, San Jose, CA, USA). The normalized value ΔA was calculated based on the following equation.

<식 1><Equation 1>

ΔA = At - AcΔA = At - Ac

여기서 At는 sEV과 압타머를 모두 포함하는 샘플의 흡광도이고, Ac는 sEV만 포함하는 대조군의 흡광도이다. 압타머의 농도는 분석법에 따라 달라지며, 모든 실험은 3회 반복하였다.Here, At is the absorbance of the sample containing both sEV and aptamer, and Ac is the absorbance of the control containing only sEV. The concentration of aptamer varies depending on the analysis method, and all experiments were repeated three times.

도 6에 도시된 바와 같이, 상위 빈도수(%) 10개 후보 압타머는 SW620 및 HS sEV에 대해 서로 다른 친화성을 보여주었다. 흥미롭게도, 빈도수(%)가 가장 낮은 CCE-10은 SW620 sEV에 대한 친화도가 가장 높았고 HS sEV에 대한 친화도가 가장 낮았다. 이것은 더 높은 빈도(%)를 가진 압타머가 더 낮은 빈도(%)를 가진 압타머보다 더 높은 결합 친화도를 가질 것이라는 초기 가정과 달랐다. 또한 ΔAHS에 대한 ΔASW620의 비율을 추정할 때 CCE-10도 가장 높은 값을 보였다. 전반적으로, 이러한 결과는 SW620 sEV에 대한 CCE-10의 높은 결합 친화도 및 특이성을 확인한 것이며; 따라서 CCE-10을 SELEX 후 최적화를 위해 선택하였다.As shown in Figure 6, the top 10 candidate aptamers with high frequency (%) showed different affinities for SW620 and HS sEV. Interestingly, CCE-10, which had the lowest frequency (%), had the highest affinity for SW620 sEVs and the lowest affinity for HS sEVs. This differed from the initial assumption that aptamers with higher frequency (%) would have higher binding affinity than aptamers with lower frequency (%). Additionally, when estimating the ratio of ΔA SW620 to ΔA HS , CCE-10 also showed the highest value. Overall, these results confirmed the high binding affinity and specificity of CCE-10 for SW620 sEV; Therefore, CCE-10 was selected for optimization after SELEX.

실시예 1-4Example 1-4

CCE-10 압타머의 SELEX 이후 최적화Post-SELEX optimization of CCE-10 aptamer

E-SELEX 과정 동안 PCR 증폭을 위해 프라이머 영역을 ssDNA 라이브러리에 포함하였다. 더 짧은 길이의 압타머는 더 저렴한 비용으로 합성되고 압타머 기반 바이오센서 개발에 더 적합하기 때문에 정방향 또는 역방향 프라이머 영역을 절단하여 CCE-10 압타머의 사후 SELEX 최적화를 수행했다. 정방향, 역방향 또는 두 영역이 모두 없는 CCE-10 압타머를 각각 CCE-10F, CCE-10R 및 CCE-10FR로 지정하였다. CCE-10 압타머는 전술한 [표 3]에서 서열번호 13으로 표시하였으나, 하기 [표 4]에서는 정방향과 역방향 프라이머 부분을 포함하여 CCE 10'으로 명명하고, 서열번호 14로 기재하였다. Primer regions were included in the ssDNA library for PCR amplification during the E-SELEX process. Because shorter length aptamers are synthesized at lower cost and are more suitable for aptamer-based biosensor development, we performed post-SELEX optimization of the CCE-10 aptamer by truncating the forward or reverse primer regions. CCE-10 aptamers lacking the forward, reverse, or both regions were designated CCE-10F, CCE-10R, and CCE-10FR, respectively. The CCE-10 aptamer was indicated by SEQ ID NO: 13 in the above-mentioned Table 3, but in [Table 4] below, it was named CCE 10' including the forward and reverse primer portions, and was listed as SEQ ID NO: 14.

Sequence (5'→3')Sequence (5'→3') 서열번호sequence number Random libraryRandom library ATCCAGAGTGACGCAGCA-N40-CTGGCTCGAACAAGCTTGCATCCAGAGTGACGCAGCA-N 40 -CTGGCTCGAACAAGCTTGC 1One Forward primerForward primer ATCCAGAGTGACGCAGCAATCCAGAGTGACGCAGCA 22 Reverse primerReverse primer GCAAGCTTGTTCGAGCCAGGCAAGCTTGTTCGAGCCAG 33 CCE-10CCE-10 ATCCAGAGTGACGCAGCAGGCCTAAGAAGGGACAGGGTGACAATCAGGGGGAGAGGGGCTGGCTCGAACAAGCTTGCATCCAGAGTGACGCAGCAGGCCTAAGAAGGGACAGGGTGACAATCAGGGGGAGAGGGGCTGGCTCGAACAAGCTTGC 1414 CCE-10FCCE-10F GGCCTAAGAAGGGACAGGGTGACAATCAGGGGGAGAGGGGCTGGCTCGAACAAGCTTGCGGCCTAAGAAGGGACAGGGTGACAATCAGGGGGAGAGGGGCTGGCTCGAACAAGCTTGC 1515 CCE-10RCCE-10R ATCCAGAGTGACGCAGCAGGCCTAAGAAGGGACAGGGTGACAATCAGGGGGAGAGGGGATCCAGAGTGACGCAGCAGGCCTAAGAAGGGACAGGGTGACAATCAGGGGGGAGAGGGG 1616 CCE-10FRCCE-10FR GGCCTAAGAAGGGACAGGGTGACAATCAGGGGGAGAGGGGGGCCTAAGAAGGGACAGGGTGACAATCAGGGGGGAGAGGGG 1717

도 6의 (B)에서 볼 수 있듯이, 절단된 CCE-10 압타머(CCE-10F, CCE-10R 및 CCE-10FR)와 SW620 sEV(양성 표적)의 결합 친화도는 절단되지 않은 CCE-10' 압타머의 결합 친화도와 비슷했다. 그러나, HS sEV(반대 표적)에 대한 결합 친화성은 현저하게 달랐다. 구체적으로, 정방향 프라이머 영역이 없는 압타머(CCE-10F 및 CCE-10FR)는 HS sEV에 대한 결합 친화도가 추가로 감소한 반면, 역방향 프라이머 영역이 없는 압타머(CCE-10R)는 결합 친화도가 크게 증가하는 것으로 나타났다. 이러한 결과로부터 본 연구에서 평가된 정방향 프라이머 영역이 HS sEV에 대한 압타머의 결합에 참여하여 표적외 결합의 확률을 증가시킬 수 있다고 판단하였고, CCE-10에서 정방향 프라이머 영역을 제거하는 것이 암 유래 sEV을 비표적 sEV과 구별하는 데 도움이 될 것이라고 결론지었다. CCE-10F와 CCE-10FR은 SW620 및 HS sEV에 필적할만한 결합 친화력을 나타내므로 각 절단된 압타머에 대한 ΔASW620 대 ΔAHS의 비율을 평가하여 이 연구에 가장 적합한 압타머를 가장 적합한 압타머로 선정했다. 도 6B에 도시된 바와 같이, CCE-10F의 비율이 가장 높아 결합 친화도 및 특이성 측면에서 그 우수성을 입증하였다.As shown in Figure 6 (B), the binding affinity of cleaved CCE-10 aptamers (CCE-10F, CCE-10R, and CCE-10FR) and SW620 sEV (positive target) is similar to that of uncleaved CCE-10'. The binding affinity was similar to that of the aptamer. However, the binding affinity to HS sEV (opposite target) was significantly different. Specifically, aptamers without the forward primer region (CCE-10F and CCE-10FR) had further reduced binding affinity for HS sEVs, whereas aptamers without the reverse primer region (CCE-10R) had a reduced binding affinity. appeared to increase significantly. From these results, it was determined that the forward primer region evaluated in this study could participate in the binding of aptamers to HS sEVs and increase the probability of off-target binding, and that removing the forward primer region from CCE-10 could reduce the likelihood of cancer-derived sEVs It was concluded that it would be helpful in distinguishing sEVs from non-targeted sEVs. Since CCE-10F and CCE-10FR exhibit comparable binding affinities to SW620 and HS sEV, the ratio of ΔA SW620 to ΔA HS for each truncated aptamer was evaluated to select the most suitable aptamer for this study. did. As shown in Figure 6B, the ratio of CCE-10F was the highest, demonstrating its superiority in terms of binding affinity and specificity.

실시예 1-5.Examples 1-5.

해리 상수, Kd의 결정Determination of the dissociation constant, Kd

CCE-10F의 결합 친화도는 Kd를 추정하여 정량적으로 평가하였다. 구체적으로, 다양한 농도(0-100 nM)에서 CCE-10F를 일정한 수의 SW620 및 HS sEV에 노출시킨 다음 실시예3에서 전술한 ELONA 방법을 사용하여 분석했다. The binding affinity of CCE-10F was quantitatively evaluated by estimating Kd. Specifically, CCE-10F was exposed to a certain number of SW620 and HS sEVs at various concentrations (0-100 nM) and then analyzed using the ELONA method described above in Example 3.

결합 친화도 분석Binding affinity analysis

선택된 압타머 CCE-10F의 결합 친화도를 평가하기 위해, 비오틴(0-100 nM)으로 표지된 선택된 압타머의 농도를 변화시키고 위에서 설명한 ELONA 방법을 사용하여 평형 해리 상수(K d)를 결정했다. 정규화된 ΔA 값은 압타머 농도의 함수로 표시되었으며 K d는 비선형 회귀 방정식 Y = (Bmax Х X)/(K d + X)를 사용하여 결정하였다. 여기서 Y는 ΔA, Bmax는 최대 ΔA, K d는 해리 상수, X는 압타머 농도이다.To assess the binding affinity of the selected aptamer CCE-10F, we varied the concentration of the selected aptamer labeled with biotin (0–100 nM) and determined the equilibrium dissociation constant ( K d ) using the ELONA method described above. . Normalized ΔA values were expressed as a function of aptamer concentration and K d was determined using the nonlinear regression equation Y = (Bmax Х X)/( K d + X). where Y is ΔA, Bmax is the maximum ΔA, K d is the dissociation constant, and X is the aptamer concentration.

그 결과, 도 7은 CCE-10F(A) 및 CCE-10FR(B)의 해리 상수(K d) 곡선을 나타낸다. 도 7의 (A)에서 보는 바와 같이 SW620 sEV의 경우 CCE-10F의 농도가 증가함에 따라 흡광도 신호가 증가하여 3.41 nM의 K d 값을 나타내는 반면, HS sEV의 경우, 흡광도 신호가 변하지 않으므로 K d 값을 결정할 수 없었다. 낮은 나노몰 범위의 K d 값은 SW620 sEV에 대한 CCE-10F의 높은 결합 친화도를 시사했다. 또한, SW620 및 HS sEV에 대한 CCE-10FR의 K d 값도 CCE-10F에서와 동일한 방식으로 측정한 결과, SW620 sEV에 대해서는 6.34 nM의 K d을 나타내었고, HS sEV에 대해서는 K d 값이 검출되지 않았다(도 7의 (B)). CCE-10F의 친화력은 CCE-10FR의 친화력보다 1.9배 높았지만 CCE-10F와 CCD-10FR 모두 진단 및 치료 응용 분야에 적합한 후보 압타머로 판단하였다. 추가 실험을 위해 SW620 sEV에 대한 결합 친화도가 더 높은 CCE-10F를 선택했다.As a result, Figure 7 shows the dissociation constant ( K d ) curves of CCE-10F (A) and CCE-10FR (B). As shown in Figure 7 (A), for SW620 sEVs, the absorbance signal increases as the concentration of CCE-10F increases, showing a K d value of 3.41 nM, whereas for HS sEVs, the absorbance signal does not change, resulting in K d The value could not be determined. K d values in the low nanomolar range suggested high binding affinity of CCE-10F for SW620 sEV. In addition, the K d value of CCE-10FR for SW620 and HS sEV was measured in the same manner as for CCE-10F. As a result, a K d of 6.34 nM was detected for SW620 sEV, and a K d value was detected for HS sEV. It did not work ((B) in Figure 7). Although the affinity of CCE-10F was 1.9 times higher than that of CCE-10FR, both CCE-10F and CCD-10FR were judged to be suitable candidate aptamers for diagnostic and therapeutic applications. CCE-10F, which has a higher binding affinity to SW620 sEV, was selected for further experiments.

실시예 1-6Example 1-6

최고의 압타머 평가(CCE-10F)Best Aptamer Evaluation (CCE-10F)

OligoAnalyzer Tool (IDT)와 Mol* 3D Viewer (RCSB PDB)를 사용하여 각각 CCE-10F 2차 구조의 2차원 및 3차원 구조를 예측분석하였다. The two-dimensional and three-dimensional structures of CCE-10F secondary structure were predicted and analyzed using OligoAnalyzer Tool (IDT) and Mol * 3D Viewer (RCSB PDB), respectively.

도 8의 (A)는 CCE-10F가 37℃에서 최소 깁스 자유 에너지가 -2.39kcal/mol인 두 개의 서로 다른 줄기 루프 모티프를 포함한다는 것을 보여준다. 본 발명자들은 2개의 스템-루프 모티프의 존재가 CCE-10F에 SW620 sEV에 대한 높은 결합 친화도 및 특이성을 부여할 것이라고 가정했다. 다음으로, CCE-10F의 친화도를 랜덤 라이브러리(Lib) 및 정방향 프라이머 영역이 없는 랜덤 라이브러리(Lib-F)의 친화도와 비교하였다. 도 8의 (B)에 도시된 바와 같이, CCE-10F는 SW620 sEV에 대한 가장 높은 결합 친화도를 가지며, 이는 랜덤 라이브러리(Lib 및 Lib-F)보다 상당히 더 높다. Figure 8 (A) shows that CCE-10F contains two different stem-loop motifs with a minimum Gibbs free energy of -2.39 kcal/mol at 37°C. We hypothesized that the presence of two stem-loop motifs would endow CCE-10F with high binding affinity and specificity for SW620 sEV. Next, the affinity of CCE-10F was compared with that of a random library (Lib) and a random library without the forward primer region (Lib-F). As shown in Figure 8 (B), CCE-10F has the highest binding affinity to SW620 sEV, which is significantly higher than the random libraries (Lib and Lib-F).

또한, SW620 sEV에 대한 CCE-10F의 검출 가능성을 ELONA 방법을 사용하여 조사하였다. 도 8의 (C)에 표시된 결과는 흡광도 신호(ΔA)가 SW620 sEV의 농도(2.0 Х 104 - 1.0 Х 106 입자/μL)에 비례하고 선형 회귀함수가 Y = 0.01986X + 0.08825 (R2 = 0.9903)가 다음과 같이 획득되었음을 보여준다. 또한, SW620 sEV에 대한 검출한계 (LOD, limit of detection)을 계산한 결과, 1.0 x 103 입자/μL로 도출되었다. 혈청 또는 혈장 내 sEV의 농도가 105 입자/μL 범위로 존재하는 것을 고려하면, CRC 세포 유래 sEV에 대한 CCE-10F의 진단 능력을 강력하게 검증하기 정밀함을 시사한다. Additionally, the detectability of CCE-10F against SW620 sEV was investigated using the ELONA method. The results shown in (C) of Figure 8 show that the absorbance signal (ΔA) is proportional to the concentration of SW620 sEV (2.0 Х 10 4 - 1.0 Х 10 6 particles/μL) and the linear regression function is Y = 0.01986X + 0.08825 (R 2 = 0.9903) is obtained as follows. Additionally, as a result of calculating the limit of detection (LOD) for SW620 sEV, it was derived to be 1.0 x 10 3 particles/μL. Considering that the concentration of sEV in serum or plasma is in the range of 10 5 particles/μL, this suggests that it is accurate to strongly verify the diagnostic ability of CCE-10F for CRC cell-derived sEV.

마지막으로, 정상 결장 및 기타 암 세포주에서 유래한 sEV와 SW620 및 HS 유래 sEV에 대한 CCE-10F의 결합 특이성을 평가하였다. 도 8의 (D)는 CCD-18Co 세포(정상 결장)-, SKBR3 세포(인간 유방암) 및 U-87 MG 세포(인간 교모세포종)유래 sEV에 대해서는 결합성을 보이지 않지만, SW620 세포 유래 sEV에 대한 CCE-10F의 높은 결합 특이성을 갖음을 나타낸다. CCE-10F의 높은 특이성은 CCD-18Co 세포, SKBR3 세포 및 U-87 MG 세포 유래 sEV이 대항 선별의 대상으로 포함되지 않았음에도 불구하고, CCD-18Co 세포, SKBR3 세포 및 U-87 MG 세포 유래 sEV에 대한 이 압타머의 친화도가 낮으며, HS 유래 sEV에 대한 결합 친화도와 비슷하다는 것을 확인했다. Finally, we evaluated the binding specificity of CCE-10F to sEVs derived from normal colon and other cancer cell lines and to sEVs derived from SW620 and HS. Figure 8 (D) does not show binding to sEVs derived from CCD-18Co cells (normal colon), SKBR3 cells (human breast cancer), and U-87 MG cells (human glioblastoma), but binds to sEVs derived from SW620 cells. This indicates that CCE-10F has high binding specificity. The high specificity of CCE-10F allows it to detect sEVs derived from CCD-18Co cells, SKBR3 cells, and U-87 MG cells, even though sEVs derived from CCD-18Co cells, SKBR3 cells, and U-87 MG cells were not included as targets of counterscreening. It was confirmed that the affinity of this aptamer for is low and is similar to the binding affinity for HS-derived sEV.

인간 혈청 내 압타머 안정성 분석Aptamer stability analysis in human serum

마지막으로, HS(80%)에서 압타머(Apt) 단독 및/또는 Apt-sEV의 안정성은 압타머의 잠재적인 생체내 사용을 확인하기 위해 추가로 평가하였다.Finally, the stability of aptamer (Apt) alone and/or Apt-sEV in HS (80%) was further evaluated to confirm the potential in vivo use of the aptamer.

혈청 내 압타머 안정성을 조사하기 위해 압타머(Apt) 단독 또는 압타머-SW620 sEV을 80% 인간 혈청(Sigma-Aldrich)에서 0-48시간 동안 배양했다. 각 시간 간격으로 회수된 시료는 혈청 또는 sEV 유래 단백질을 제거하기 위해 먼저 37℃에서 1시간 동안 proK(200 μg/mL)를 처리하여 -80℃에서 사용할 때까지 보관하였다. 다음으로, 각 샘플을 Novex TBE-urea 샘플 버퍼(Invitrogen)와 혼합하고, 95℃에서 10분 동안 가열하고, 요소 변성 폴리아크릴아미드 겔(15%)에 용해시켰다. 젤을 GreenStar 핵산 염색 용액(Bioneer, 대전, 대한민국)으로 염색하고 ChemiDoc Imaging System(Bio-Rad Laboratories)을 사용하여 이미지화했다. 밴드 세기는 Image Lab 소프트웨어(Bio-Rad Laboratories)를 사용하여 분석하였다. 0시간에 압타머 또는 압타머-SW620 sEV의 밴드 세기는 100%로 간주되었다.To investigate aptamer stability in serum, aptamer (Apt) alone or Aptamer-SW620 sEV were cultured in 80% human serum (Sigma-Aldrich) for 0–48 h. Samples recovered at each time interval were first treated with proK (200 μg/mL) at 37°C for 1 hour to remove serum or sEV-derived proteins and stored at -80°C until use. Next, each sample was mixed with Novex TBE-urea sample buffer (Invitrogen), heated at 95°C for 10 min, and dissolved in a urea-modified polyacrylamide gel (15%). Gels were stained with GreenStar Nucleic Acid Staining Solution (Bioneer, Daejeon, South Korea) and imaged using the ChemiDoc Imaging System (Bio-Rad Laboratories). Band intensity was analyzed using Image Lab software (Bio-Rad Laboratories). The band intensity of aptamer or aptamer-SW620 sEV at time 0 was considered 100%.

그 결과, 도 8의 (E)는 0에서 48시간까지 다양한 시간 프레임에서 배양된 압타머의 젤 이미지와 안정성(%)을 보여준다. 압타머 단독과 Apt-sEV은 각각 33%와 44%의 안정성을 유지하면서 최대 24시간 동안 안정적이었다. 흥미롭게도, 최대 3시간 동안 유의미한 차이가 관찰되지 않았으며, 그 후 Apt-sEV는 압타머 단독보다 더 높은 안정성을 보여주었다. 이는 압타머가 sEV에 결합하여 혈청의 뉴클레아제에 대해 더 많은 저항성을 제공하기 때문일 수 있다. 혈청 안정성 분석에 사용된 압타머는 변형이 없는 것이므로 포스포티오에이트 결합 및 2' fluoro-base와 같은 작용기로 변형하면 뉴클레아제에 대한 압타머의 저항성이 증가하고 결합 능력을 손상시키지 않으면서 안정성이 확보될 것으로 예상된다.As a result, Figure 8 (E) shows gel images and stability (%) of aptamers cultured in various time frames from 0 to 48 hours. Aptamer alone and Apt-sEV were stable for up to 24 hours, maintaining stability of 33% and 44%, respectively. Interestingly, no significant differences were observed for up to 3 hours, after which Apt-sEV showed higher stability than the aptamer alone. This may be because the aptamer binds to sEVs, providing more resistance to nucleases in serum. Since the aptamer used in the serum stability analysis is unmodified, modification with functional groups such as phosphothioate bond and 2' fluoro-base increases the resistance of the aptamer to nucleases and improves stability without compromising binding ability. It is expected to be secured.

이러한 흥미로운 결과는 형광 편광(FP) 및 표면 강화 라만 분광법(SERS)과 같은 다양한 신호 전달 방법과 함께 CRC 세포 유래 sEV을 특이적으로 검출하기 위한 다양한 압타머 기반 바이오센서의 개발을 위한 길을 열 수 있음을 확인하였다.These interesting results could pave the way for the development of various aptamer-based biosensors for specific detection of CRC cell-derived sEVs together with various signaling methods such as fluorescence polarization (FP) and surface-enhanced Raman spectroscopy (SERS). It was confirmed that it exists.

실시예 1-7.Example 1-7.

SW620 sEV에 대한 압타머 표적 바이오마커의 위상(topology) 분석Topology analysis of aptamer target biomarkers for SW620 sEV

CCE-10F에 의해 표적화된 SW620 sEV에 대한 바이오마커 위치를 조사하기 위해, sEV을 프로테이나제 K(proK), 트립신 및 RIPA 용해에 적용하였다.To investigate the biomarker location on SW620 sEVs targeted by CCE-10F, sEVs were subjected to proteinase K (proK), trypsin and RIPA lysis.

sEV 용해를 위해 1X Halt 프로테아제 억제제 칵테일(Thermo Fisher Scientific)이 보충된 RIPA 용해 완충액을 사용하여 SW620 유래 sEV의 용해물을 제조했다. sEV을 프로테아제 억제제가 보충된 용해 완충액과 혼합하고, 얼음 위에서 20분 동안 인큐베이션하고 40kHz에서 30초 온/오프의 5주기로 수조 초음파 처리기(SH-2140D, SAE HAN ULTRASONIC, Seoul, Republic of Korea)에서 초음파 처리하였다. 프로테이나제 K(proK) 처리를 위해 SW620 세포 유래 sEV을 20분마다 부드러운 볼텍싱으로 37℃에서 1시간 동안 20 μg/mL proK(엔지노믹스)로 처리했다. 프로테이나제 활성은 실온에서 10분 동안 5 mM 페닐메틸설포닐 플루오라이드(PMSF)의 첨가에 의해 억제되었다. 트립신 처리를 위해 0.25% 트립신-EDTA(Welgene)를 SW620 세포 유래 sEV에 첨가하고 37℃에서 30분 동안 인큐베이션했다. 트립신 활성은 10% FBS를 함유하는 동일한 부피의 PBS를 첨가함으로써 종결되었다. 각 처리의 최종 조성물은 사용 전까지 -80℃에 보관하였다.For sEV lysis, lysates of SW620-derived sEVs were prepared using RIPA lysis buffer supplemented with 1X Halt protease inhibitor cocktail (Thermo Fisher Scientific). sEVs were mixed with lysis buffer supplemented with protease inhibitors, incubated on ice for 20 min and sonicated in a water bath sonicator (SH-2140D, SAE HAN ULTRASONIC, Seoul, Republic of Korea) with 5 cycles of 30 s on/off at 40 kHz. Processed. For proteinase K (proK) treatment, SW620 cell-derived sEVs were treated with 20 μg/mL proK (Engenomics) for 1 h at 37°C with gentle vortexing every 20 min. Proteinase activity was inhibited by addition of 5 mM phenylmethylsulfonyl fluoride (PMSF) for 10 min at room temperature. For trypsinization, 0.25% trypsin-EDTA (Welgene) was added to SW620 cell-derived sEVs and incubated at 37°C for 30 min. Trypsin activity was terminated by adding an equal volume of PBS containing 10% FBS. The final composition of each treatment was stored at -80°C until use.

proK와 트립신의 분해효소 활성은 소낭 외막단백질(extravesicular membrane protein)에만 작용하며, 소낭 내 단백질(intravesicular protein)은 영향을 받지 않아 그대로 유지되기 때문에 바이오마커의 위치를 구별할 수 있다. RIPA 용해 완충액을 사용하면 sEV의 루멘에서 소포 내 단백질과 지질 이중층에서 막 단백질을 방출하여 원형태로 노출시킨다. The decomposing enzyme activities of proK and trypsin only act on the extravesicular membrane protein of the vesicle, and the intravesicular protein remains unaffected, making it possible to distinguish the location of the biomarker. Using RIPA lysis buffer releases proteins within vesicles from the lumen of sEVs and membrane proteins from the lipid bilayer, exposing them in their original form.

그 결과, 도 9는 바이오마커 토폴로지 분석결과를 나타낸다. I: 온전한 sEV, II: proteinase K(proK) 처리, III: 트립신 처리, IV: RIPA 처리 sEV 결과이다. 즉, 도 9(I-III)는 CCE-10F가 proK 및 트립신 처리된 sEV에 대해 무시할 수 있는 결합 친화도를 나타냄을 나타낸다. 한편, CCE-10F는 온전한 sEV과 비교하여 RIPA 용해 sEV에 대해 필적할만한 결합 친화도를 나타냈다(도 9, I 및 IV). 이러한 결과로, CCE-10F가 표적으로 하는 바이오마커가 단백질로 식별되고 SW620 sEV의 외막에 위상적으로 위치함을 확인하였다. As a result, Figure 9 shows the results of biomarker topology analysis. I: intact sEV, II: proteinase K (proK) treated, III: trypsin treated, IV: RIPA treated sEV results. That is, Figure 9 (I-III) shows that CCE-10F exhibits negligible binding affinity to proK and trypsin-treated sEV. Meanwhile, CCE-10F showed comparable binding affinity to RIPA-lysed sEVs compared to intact sEVs (Figure 9, I and IV). These results confirmed that the biomarker targeted by CCE-10F was identified as a protein and was topologically located in the outer membrane of SW620 sEV.

실시예 1-8.Examples 1-8.

sEV 및 Apt-sEV의 세포 흡수Cellular uptake of sEV and Apt-sEV

LS 174T 세포의 생존력은 SW620 세포 유래 sEV, CCE-10F 압타머-sEV 복합체(Apt-sEV) 및 CCE-10F 압타머 단독(Apt)으로 처리한 후 먼저 평가되었다.The viability of LS 174T cells was first assessed after treatment with SW620 cell-derived sEVs, CCE-10F aptamer-sEV complex (Apt-sEV), and CCE-10F aptamer alone (Apt).

세포독성 분석Cytotoxicity assay

제조업체의 지침에 따라 CCK-8(Dojindo, Rockville, MD, USA)을 사용하여 SW620 및 압타머-SW620 sEV의 존재하에 LS 174T 세포의 생존력을 테스트했다. 세포(2 x 104 cells/well)를 96웰 플레이트에 접종했다. 밤새 배양한 후, 세포를 PBS, SW620 및 압타머-SW620 sEV으로 37℃에서 24시간 동안 처리하였다. 이 분석에 사용된 sEV 및 압타머의 농도는 각각 1 x 1010 입자/mL 및 200 nM이었다. 다음으로, CCK-8 시약을 각 웰에 첨가하고 37℃에서 추가로 2시간 동안 인큐베이션하였다. 마지막으로 마이크로플레이트 리더를 이용하여 450 nm에서 시료의 흡광도를 측정하였으며, PBS로 처리한 시료는 100% 세포생존율로 간주하였다. The viability of LS 174T cells was tested in the presence of SW620 and aptamer-SW620 sEVs using CCK-8 (Dojindo, Rockville, MD, USA) according to the manufacturer's instructions. Cells (2 x 10 4 cells/well) were inoculated into a 96-well plate. After overnight culture, cells were treated with PBS, SW620, and Aptamer-SW620 sEV for 24 hours at 37°C. The concentrations of sEV and aptamer used in this analysis were 1 x 10 10 particles/mL and 200 nM, respectively. Next, CCK-8 reagent was added to each well and incubated for an additional 2 hours at 37°C. Finally, the absorbance of the sample was measured at 450 nm using a microplate reader, and the sample treated with PBS was considered to have 100% cell viability.

그 결과, 도 10의 (A)에서 볼 수 있듯이 sEV(주황색), Apt-sEV(녹색) 및 Apt(파란색)는 LS 174T 세포에 대해 세포 독성을 나타내지 않았다. 통계 분석으로 또한 모든 경우의 세포 생존율이 대조군의 세포 생존율과 크게 다르지 않음을 확인했다(one-way ANOVA, p > 0.05). 따라서, 세포 생존력 분석에 사용된 실험 조건은 추가 실험을 위해 채택되었다.As a result, as shown in Figure 10 (A), sEV (orange), Apt-sEV (green), and Apt (blue) did not show cytotoxicity to LS 174T cells. Statistical analysis also confirmed that the cell survival rate in all cases was not significantly different from that of the control group (one-way ANOVA, p > 0.05). Therefore, the experimental conditions used for cell viability analysis were adopted for further experiments.

세포 흡수 분석Cellular uptake assay

LS 174T 세포에 의한 sEV 흡수에 대한 CCE-10F의 효과는, SW620 sEV에 대한 CCE-10F 압타머의 특이적 결합이 sEV에 의해 유도된 다운스트림 세포 반응을 억제하는지 여부를 조사하기 전에 공초점 현미경 이미지 및 유세포 분석을 통해 평가하였다.The effect of CCE-10F on sEV uptake by LS 174T cells was examined by confocal microscopy before examining whether specific binding of the CCE-10F aptamer to SW620 sEVs inhibits downstream cellular responses induced by sEVs. Evaluated through image and flow cytometry.

sEV(1 x 1010 입자)을 100 μM CFSE(Abcam, Cambridge, UK)로 표지하고 어둠 속에서 2시간 동안 400 rpm으로 진탕기에서 37℃에서 배양했다. 과잉 CFSE를 제거하기 위해 qEV 자동 분획 수집기에서 qEV10 / 35 nm 컬럼을 사용하는 크기 배제 크로마토그래피(SEC)를 제조업체의 지침에 따라 수행했다. 분획을 모아 사용할 때까지 -80℃에서 보관했다. LS 174T 세포(2 x 105 cells/well)를 24웰 플레이트에 접종하고 37℃에서 밤새 배양했다. 다음으로, 세포를 새로운 배지로 교체하고 SW620 및 압타머-SW620 sEV으로 37℃에서 24시간 동안 처리하였다. 이 분석에 사용된 sEV과 압타머의 농도는 각각 1 x 109 입자와 200 nM이었다. 공초점 현미경 분석을 위해 세포를 DPBS로 세척 후, 4% 파라포름알데히드 (PFA)로 세포 고정 후, 핵염색을 위해 DAPI를 처리하였다. 공초점 이미지는 LSM 900 공초점 현미경(Carl Zeiss, Thornwood, NY, USA)에서 수집하고 Zen 소프트웨어(Carl Zeiss)로 분석했다. 유세포 분석을 위해 트립신 처리로 세포를 분리하고 DPBS로 세척하고 2% FBS가 첨가된 DPBS에 재현탁하고 CyFlow Cube 6 유세포 분석기(Sysmex Corporation, Kobe, Japan)를 사용하여 분석하였다. 3 × 104 이벤트를 기록한 후 FlowJo 소프트웨어(BD Biosciences, San Jose, CA, USA)를 사용하여 결과를 분석했다.sEVs (1 × 10 particles) were labeled with 100 μM CFSE (Abcam, Cambridge, UK) and incubated at 37°C on a shaker at 400 rpm for 2 h in the dark. To remove excess CFSE, size exclusion chromatography (SEC) using a qEV10/35 nm column on a qEV automated fraction collector was performed according to the manufacturer's instructions. Fractions were collected and stored at -80°C until use. LS 174T cells (2 x 10 5 cells/well) were seeded in a 24-well plate and cultured at 37°C overnight. Next, the cells were replaced with fresh medium and treated with SW620 and Aptamer-SW620 sEV at 37°C for 24 hours. The concentrations of sEV and aptamer used in this analysis were 1 x 10 9 particles and 200 nM, respectively. For confocal microscopy analysis, cells were washed with DPBS, fixed with 4% paraformaldehyde (PFA), and treated with DAPI for nuclear staining. Confocal images were collected on an LSM 900 confocal microscope (Carl Zeiss, Thornwood, NY, USA) and analyzed with Zen software (Carl Zeiss). For flow cytometry, cells were separated by trypsinization, washed with DPBS, resuspended in DPBS supplemented with 2% FBS, and analyzed using a CyFlow Cube 6 flow cytometer (Sysmex Corporation, Kobe, Japan). After recording 3 × 104 events, the results were analyzed using FlowJo software (BD Biosciences, San Jose, CA, USA).

그 결과, 도 11의 (A)의 공초점 이미지는 녹색 형광 신호에 의해 입증된 바와 같이 CFSE로 표지된 sEV 및 Apt-sEV 모두가 LS 174T 세포에 의해 흡수되었음을 보여준다. sEV 및 Apt-sEV의 흡수도 유세포분석에 의해 정량적으로 평가하였다. 도 11B(왼쪽)에서 볼 수 있듯이 sEV(주황색)과 Apt-sEV(녹색)을 처리한 LS 174T 세포에서 대조군(빨간색)과 비교할 때 CFSE 세기의 현저한 변화가 관찰되었다. 또한 도 11의 (B)(오른쪽)에서 CFSE의 평균 형광 강도(MFI)가 sEV 및 Apt-sEV에서 유의하게 증가하여 LS 174T 세포에 의한 sEV의 효과적인 흡수를 입증하였다. 흥미롭게도 Apt-sEV의 흡수는 sEV 단독의 흡수와 다르지 않았다(ns; p > 0.05). 이러한 결과는 CCE-10F와 sEV의 잠재적 바이오마커의 분자 상호작용이 세포 흡수에 유의미한 영향을 미치지 않는다는 것을 강력하게 증명하며, 이는 잠재적 바이오마커의 생물학적 기능이 sEV의 세포 내재화와 관련이 없음을 시사한다.As a result, the confocal image in Figure 11 (A) shows that both sEV and Apt-sEV labeled with CFSE were taken up by LS 174T cells, as evidenced by the green fluorescence signal. Uptake of sEV and Apt-sEV was also quantitatively assessed by flow cytometry. As shown in Figure 11B (left), significant changes in CFSE intensity were observed in LS 174T cells treated with sEV (orange) and Apt-sEV (green) compared to the control (red). Additionally, in Figure 11 (B) (right), the mean fluorescence intensity (MFI) of CFSE was significantly increased in sEV and Apt-sEV, demonstrating effective uptake of sEV by LS 174T cells. Interestingly, uptake of Apt-sEV did not differ from that of sEV alone (ns; p > 0.05). These results strongly demonstrate that the molecular interaction of CCE-10F with potential biomarkers of sEVs does not significantly affect cellular uptake, suggesting that the biological function of potential biomarkers is not related to cellular internalization of sEVs. .

실시예 1-9.Example 1-9.

sEV 및 Apt-sEV의 상처 치유Wound healing in sEV and Apt-sEV

SW620 CRC 세포주는 원래 전이성 림프절에서 유래한 것으로 간 전이 능력을 나타낸다. 따라서 SW620 세포에서 파생된 sEV은 또한 전이 가능성을 가질 수 있으며, 이는 세포에서 높은 침입 및 이동 능력의 발달을 유도한다. CCE-10F에 의해 특별히 표적화된 SW620 sEV에 대한 잠재적인 바이오마커를 탐색하기 위해, 본 발명자들은 모델 시스템으로 전이 가능성이 낮은 CRC 세포주인 LS 174T를 선택하고, CCE-10F가 SW620 세포 유래 sEV의 전이 가능성에 영향을 미치는지 조사하였다. The SW620 CRC cell line is originally derived from metastatic lymph nodes and exhibits the ability to metastasize to the liver. Therefore, sEVs derived from SW620 cells may also have metastatic potential, which leads to the development of high invasion and migration abilities in the cells. To explore potential biomarkers for SW620 sEVs specifically targeted by CCE-10F, we selected LS 174T, a CRC cell line with a low metastatic potential, as a model system and demonstrated that CCE-10F inhibits the metastasis of SW620 cell-derived sEVs. We investigated whether it affected the possibility.

LS 174T 세포의 이동 및 증식 능력에 대한 sEV 및 압타머 결합 sEV의 효과를 평가하기 위해 상처 치유 분석을 수행했다. sEV은 특히 종양 주변의 미세 환경에 세포 정보를 전달할 수 있기 때문에 전이 가능성이 높은 SW620 세포에서 파생된 sEV이 sEV 매개 신호 전달 경로를 통해 세포 증식, 이동 및 궁극적으로 전이를 유도한다고 가정했다. Wound healing assays were performed to evaluate the effect of sEVs and aptamer-conjugated sEVs on the migration and proliferation abilities of LS 174T cells. Because sEVs can specifically convey cellular information to the microenvironment surrounding the tumor, we hypothesized that sEVs derived from SW620 cells, which have a high metastatic potential, induce cell proliferation, migration, and ultimately metastasis through the sEV-mediated signaling pathway.

LS 174T 세포(7 Х 105 세포/웰)를 24웰 플레이트에 접종하고 표준 배양 조건에서 밤새 인큐베이션했다. 다음으로, 멸균 200 μL 피펫 팁을 사용하여 웰의 중앙을 가로질러 세포 단층을 긁어 상처를 유도했다. 이어서, 세포를 DPBS로 부드럽게 2회 세척하고 1% FBS가 보충된 조건화된 DMEM으로 교체하였다. 세포에 SW620 또는 압타머-SW620 sEV을 처리한 후 Optinity 도립현미경(MDM Instruments, Seoul, Republic of Korea)을 사용하여 0시간 및 24시간에 상처 간극 이미지를 촬영하였다. 상처 갭 영역은 ImageJ 소프트웨어(NIH, Bethesda, MD, USA)를 사용하여 분석하였다. 상처 치유(%)는 다음 식 2를 사용하여 결정하였다.LS 174T cells (7 Х 10 5 cells/well) were seeded in 24-well plates and incubated overnight under standard culture conditions. Next, a sterile 200 μL pipette tip was used to induce wounding by scraping the cell monolayer across the center of the well. Cells were then gently washed twice with DPBS and replaced with conditioned DMEM supplemented with 1% FBS. After treating cells with SW620 or aptamer-SW620 sEV, wound gap images were taken at 0 and 24 hours using an Optinity inverted microscope (MDM Instruments, Seoul, Republic of Korea). Wound gap area was analyzed using ImageJ software (NIH, Bethesda, MD, USA). Wound healing (%) was determined using Equation 2 below.

<식 2><Equation 2>

Y = 100 - (A f /A i Х 100)Y = 100 - ( A f /A i Х 100)

여기서 Y는 상처 치유(%), A f는 24시간에서의 상처 간격 면적, A i는 0시간에서의 상처 간격 면적을 의미한다. 이 분석에 사용된 SW620 sEV 및 압타머의 농도는 각각 2 x 1010 입자/ml 및 200 nM이었다.Here, Y is wound healing (%), A f is the wound gap area at 24 hours, and A i is the wound gap area at 0 hours. The concentrations of SW620 sEV and aptamer used in this analysis were 2 x 10 10 particles/ml and 200 nM, respectively.

그 결과, 도 12의 (A)는 초기(0시간; 상부 패널) 및 최종(24시간; 하부 패널) 인큐베이션 시간에서 대조군 세포 및 sEV 또는 Apt-sEV으로 처리된 세포의 상처 갭 이미지를 예시한다. 도 12의 (B)에서 정량적으로 분석했을 때, 32%의 갭 폐쇄가 sEV 단독 처리된 LS 174T에서 유도되었음을 알 수 있었다. 이러한 결과는 SW620 sEV이 LS 174T 이동 및 증식을 유발할 수 있으며 전이 가능성이 낮은 LS 174T도 이동 능력을 획득할 수 있음을 보여준다. 반면 Apt-sEV을 처리한 LS 174T의 갭 폐쇄는 18%에 불과하여 sEV 단독 처리군에 비해 유의하게 감소하였다(p < 0.005). Apt-sEV에 의한 이동 및 증식의 억제는 Apt와 sEV 간의 상호작용이 단백질 기반 바이오마커의 구조를 차단하여 궁극적으로 세포 반응의 다운스트림을 방지하는 것으로 추론할 수 있다.As a result, Figure 12 (A) illustrates wound gap images of control cells and cells treated with sEV or Apt-sEV at initial (0 h; upper panel) and final (24 h; lower panel) incubation times. When quantitatively analyzed in Figure 12 (B), it was found that 32% of gap closure was induced in LS 174T treated with sEV alone. These results show that SW620 sEV can induce LS 174T migration and proliferation, and that LS 174T, which has a low metastatic potential, can also acquire migration ability. On the other hand, the gap closure of LS 174T treated with Apt-sEV was only 18%, which was significantly reduced compared to the group treated with sEV alone (p < 0.005). The inhibition of migration and proliferation by Apt-sEVs can be inferred that the interaction between Apt and sEVs blocks the structure of protein-based biomarkers, ultimately preventing downstream cellular responses.

상처 치유가 세포 흡수 분석에서 나타난 바와 같이 sEV 및 Apt-sEV의 흡수율에 의해 영향을 받지 않았다는 점은 주목할 만하다(도 11). 이러한 결과는 CCE-10F 압타머가 원래 LS 174T 세포의 이동 및 증식 능력을 촉진하는 SW620 sEV의 단백질에 특이적으로 결합하여 sEV 매개 세포 신호 경로를 억제할 수 있음을 나타낸다.It is noteworthy that wound healing was not affected by the uptake rate of sEV and Apt-sEV as shown in the cellular uptake assay (Figure 11). These results indicate that CCE-10F aptamer can specifically bind to proteins of SW620 sEVs that originally promote the migration and proliferation ability of LS 174T cells and inhibit sEV-mediated cell signaling pathways.

실시예 1-10.Examples 1-10.

sEV 및 Apt-sEV에 의한 트랜스웰 세포 이동 및 침입Transwell cell migration and invasion by sEV and Apt-sEV

sEV 매개 종양 진행에 대한 Apt의 가능한 억제 효과를 추가로 평가하기 위해, sEV 및 Apt-sEV으로 처리된 LS 174T 세포에서 트랜스웰 이동 분석(마트리겔 없음) 및 트랜스웰 침입 분석(마트리겔 포함)을 수행했다. To further evaluate the possible inhibitory effect of Apt on sEV-mediated tumor progression, transwell migration assay (without Matrigel) and transwell invasion assay (with Matrigel) were performed in LS 174T cells treated with sEV and Apt-sEV. performed.

트랜스웰 세포 이동 및 침입 분석은 직경 6.5 mm(Corning Inc., Corning, NY, USA)의 8 μm 폴리에틸렌 테레프탈레이트(PET) 멤브레인이 있는 삽입물을 사용하여 24웰 플레이트에서 수행되었다. 침입 분석을 위해 PET 멤브레인을 세포 파종 전에 마트리겔(Corning Inc.)로 미리 코팅했다. 무혈청 DMEM에 재현탁된 LS 174T 세포를 인서트의 상부 챔버(5 x 104 cells/insert)에 접종하고 PBS, SW620 또는 압타머-SW620 sEV으로 처리했다. 1% FBS가 보충된 DMEM을 화학유인물질로 하부 챔버에 첨가하였다. 37℃에서 48시간 배양 후, 세포를 DPBS로 부드럽게 두 번 세척하고, 삽입물의 아래쪽 표면에 있는 세포를 4% PFA로 고정하고, 차가운 90% 메탄올/PBS 투과화하고, 25% 메탄올에 용해된 1% 크리스탈 바이올렛으로 염색했다. 잔여 크리스탈 바이올렛을 제거한 후, 인서트 상부 표면의 세포를 젖은 면봉으로 제거하고, 인서트를 완전히 공기 건조시킨 후 Optinity 도립 현미경(MDM Instruments)을 사용하여 사진을 촬영하였다. ImageJ 소프트웨어(NIH)를 사용하여 이동 및 침입 영역을 분석했다. 이 분석에 사용된 SW620 sEV 및 압타머의 농도는 각각 2 x 109 입자/mL 및 200 nM이었다.Transwell cell migration and invasion assays were performed in 24-well plates using inserts with an 8 μm polyethylene terephthalate (PET) membrane with a diameter of 6.5 mm (Corning Inc., Corning, NY, USA). For invasion assays, PET membranes were precoated with Matrigel (Corning Inc.) prior to cell seeding. LS 174T cells resuspended in serum-free DMEM were seeded in the upper chamber of the insert (5 x 10 4 cells/insert) and treated with PBS, SW620, or aptamer-SW620 sEV. DMEM supplemented with 1% FBS was added to the lower chamber as a chemoattractant. After 48 h of incubation at 37°C, cells were gently washed twice with DPBS, cells on the lower surface of the insert were fixed with 4% PFA, permeabilized with cold 90% methanol/PBS, and 1 dissolved in 25% methanol. % Dyed with crystal violet. After removing residual crystal violet, cells on the upper surface of the insert were removed with a wet cotton swab, the insert was completely air-dried, and photographs were taken using an Optinity inverted microscope (MDM Instruments). Migration and invasion areas were analyzed using ImageJ software (NIH). The concentrations of SW620 sEV and aptamer used in this analysis were 2 x 10 9 particles/mL and 200 nM, respectively.

그 결과, 도 12의 (C) 및 12의 (D)는 Apt-sEV이 sEV 처리군에 비해 처리되었을 때 LS 174T 세포의 수직 이동이 유의하게 억제되었음을 나타낸다(p < 0.0001). Apt-sEV의 동일한 억제 효과는 트랜스웰 침입 분석에서도 관찰되었으며(도 11E 및 11F) sEV과 Apt-sEV 처리군 사이의 침윤 면적(%)은 유의한 차이를 나타냈다(p < 0.0001). 이전 문헌의 다양한 바이오마커는 CRC 세포주의 이동 및 침습에 관여하는 것으로 확인되었으며, 그 중 일부는 혈관신생, 상피-중간엽 전이(EMT) 및 궁극적으로 종양 전이에도 관여한다. 또한 CRC 세포 유래 sEV의 바이오마커가 CRC 간 전이를 촉진하는 것으로 확인되었다. As a result, Figures 12 (C) and 12 (D) show that the vertical migration of LS 174T cells was significantly inhibited when Apt-sEV was treated compared to the sEV treatment group (p < 0.0001). The same inhibitory effect of Apt-sEV was also observed in the transwell invasion assay (Figures 11E and 11F), and the invasion area (%) between sEV and Apt-sEV treatment groups showed a significant difference (p < 0.0001). Various biomarkers from previous literature have been identified to be involved in migration and invasion of CRC cell lines, some of which are also involved in angiogenesis, epithelial-mesenchymal transition (EMT) and ultimately tumor metastasis. Additionally, biomarkers of CRC cell-derived sEVs were confirmed to promote CRC liver metastasis.

본 발명자들은 SW620 sEV에서 CCE-10F에 의해 표적화된 바이오마커가 세포 이동 및 침입 촉진에 관여할 것이며 sEV에 대한 CCE-10F의 특이적 결합에 의해 강화된 억제 효과에서 입증된 바와 같이 CRC에 대한 유망한 치료 표적이 될 수 있음을 확인하였다. We believe that the biomarkers targeted by CCE-10F in SW620 sEVs will be involved in promoting cell migration and invasion and are promising against CRC, as evidenced by the enhanced inhibitory effect by the specific binding of CCE-10F to sEVs. It was confirmed that it could be a therapeutic target.

실시예 1-11.Example 1-11.

sEV과 Apt-sEV의 튜브 형성Tube formation of sEV and Apt-sEV

본 발명자들는 또한 HUVEC 세포의 혈관 형성에 대한 CCE-10F의 항혈관신생 효과를 평가했다. We also evaluated the anti-angiogenic effect of CCE-10F on angiogenesis of HUVEC cells.

HUVEC 세포의 생존력은 초기에 SW620 sEV, CCE-10F 압타머-sEV 복합체(Apt-sEV) 및 CCE-10F 압타머 단독(Apt)으로 처리한 후 평가되었다. 실시예 8에서와 동일한 방법으로 세포 독성을 분석한 결과, 도 10의 (B)는 Apt-sEV(녹색)과 Apt(파란색)가 대조군과 비교하여 HUVEC 세포에 대해 세포 독성을 나타내지 않음을 확인하였다. 또한, 통계 분석으로 Apt-sEV 및 Apt 그룹의 세포 생존율이 대조군의 세포 생존율과 유의하게 다르지 않음을 확인했다(one-way ANOVA, p > 0.05). 이에 반해 sEV을 처리한 HUVEC 세포는 대조군에 비해 세포 생존율이 유의하게 증가하는 것으로 나타났다(p < 0.0001). 이는 SW620 sEV이 sEV 매개 신호전달 경로를 통해 HUVEC의 증식 능력을 촉진함을 시사한다. The viability of HUVEC cells was initially assessed after treatment with SW620 sEV, CCE-10F aptamer-sEV complex (Apt-sEV), and CCE-10F aptamer alone (Apt). As a result of analyzing cytotoxicity in the same manner as in Example 8, Figure 10 (B) confirmed that Apt-sEV (green) and Apt (blue) did not show cytotoxicity to HUVEC cells compared to the control group. . In addition, statistical analysis confirmed that the cell survival rates of Apt-sEV and Apt groups were not significantly different from those of the control group (one-way ANOVA, p > 0.05). In contrast, HUVEC cells treated with sEV showed a significant increase in cell survival rate compared to the control group (p < 0.0001). This suggests that SW620 sEV promotes the proliferation ability of HUVEC through the sEV-mediated signaling pathway.

튜브 형성 분석은 96웰 플레이트(SPL Life Sciences)에서 수행되었다. 세포 시딩 전에 96-웰 플레이트를 1% FBS가 포함된 CEFOgro-HUVEC 배지에 현탁된 마트리겔(Corning Inc.) HUVEC로 미리 코팅하고 마트리겔-코팅된 플레이트(1 x 104 cells/well)에 시딩하고 PBS, SW620 또는 압타머-SW620 sEV으로 처리했다. 37℃에서 4시간 배양 후, Optinity 도립 현미경(MDM Instruments)을 사용하여 세포를 사진 촬영하고 ImageJ 소프트웨어로 분석했다. PBS로 처리한 시료에 대해 sEV 및 Apt-sEV을 처리한 시료의 상대적 접합 수 및 총 가지 길이로 정의하였다. 이 분석에 사용된 SW620 sEV 및 압타머의 농도는 각각 1 x 109 입자/mL 및 200 nM이었다.Tube formation assays were performed in 96-well plates (SPL Life Sciences). Before cell seeding, 96-well plates were pre-coated with Matrigel (Corning Inc.) HUVECs suspended in CEFOgro-HUVEC medium containing 1% FBS and seeded on Matrigel-coated plates (1 x 10 cells/well). and treated with PBS, SW620, or Aptamer-SW620 sEV. After 4 hours of incubation at 37°C, cells were photographed using an Optinity inverted microscope (MDM Instruments) and analyzed with ImageJ software. It was defined as the relative number of junctions and total branch length of samples treated with sEV and Apt-sEV for samples treated with PBS. The concentrations of SW620 sEV and aptamer used in this analysis were 1 x 10 9 particles/mL and 200 nM, respectively.

그 결과, 도 13의 (A)는 튜브 형성의 대표적인 이미지를 나타내며, 이는 sEV 처리된 세포만이 튜브 형성을 발달시킨 반면, Apt-sEV 처리된 세포는 대조군에 필적하는 튜브 형성에 효과적이지 않다는 것을 명확하게 보여주었다. 도 13의 (B) 및 13의 (C)에서 명백한 바와 같이, sEV 그룹의 상대적 접합 수와 총 분지 길이는 모두 Apt-sEV 그룹보다 유의하게 컸다(p < 0.0001). As a result, Figure 13 (A) shows a representative image of tube formation, showing that only sEV treated cells developed tube formation, whereas Apt-sEV treated cells were not effective in forming tubes comparable to the control group. clearly shown. As evident in Figures 13 (B) and 13 (C), the relative junction number and total branch length of the sEV group were both significantly greater than those of the Apt-sEV group (p < 0.0001).

종합하면, 세포 이동 및 침입 분석에서 관찰된 결과(도 12)와 일치하여, 이러한 결과는 CCE-10F가 SW620 sEV에 대한 잠재적 바이오마커에 대한 특이적 결합에 의해 항혈관신생 효과를 발휘할 수 있음을 종합적으로 입증하여 결과적으로 다운스트림 세포 반응을 억제함을 확인하였다. Taken together, consistent with the results observed in cell migration and invasion assays (Figure 12), these results demonstrate that CCE-10F can exert anti-angiogenic effects by specific binding to potential biomarkers for SW620 sEVs. It was comprehensively verified and ultimately confirmed to inhibit downstream cell responses.

결론적으로, 설계한 E-SELEX 방법을 사용하여 CRC 유래 sEV을 표적으로 하는 상위 10개의 추정 압타머를 선별하고 사후 분석을 통해 가장 높은 결합 친화도(K d = 3.41 nM)와 특이성을 나타내는 최고의 압타머 CCE-10F를 선택하였다. 압타머의 진단적 타당성은 2.0 Х 104 - 1.0 Х 106 입자/μL 범위의 농도에서 CRC 세포 유래 sEV을 검출하고, 1.0 Х 103 입자/μL의 검출한계를 달성했을 뿐만 아니라 건강한 혈청, 정상 세포 및 기타 암세포에서 유래한 정상 sEV 및 비표적 sEV을 구별함으로써 입증하였다. 또한, SW620 sEV 바이오마커의 생물학적 활성은 세포 생존, 세포 흡수, 상처 치유, 트랜스웰 세포 이동/침습 및 튜브 형성 분석을 포함한 체계적인 분석으로 면밀히 조사하였다. 본 발명자들은 CCE-10F가 표적으로 하는 바이오마커가 sEV 매개 세포간 통신을 통해 세포 이동, 침입 및 혈관신생을 촉진하고 EMT와 같은 추가 종양 전이 과정을 담당할 수 있음을 확인하였다. 따라서 본 발명에 의한 DNA 압타머는 대장암 액체 생검 분야의 진단 및 치료용으로 이용할 수 있을 것으로 기대한다.In conclusion, the designed E-SELEX method was used to screen the top 10 putative aptamers targeting CRC-derived sEVs, and post-hoc analysis revealed that the best aptamer exhibited the highest binding affinity ( K d = 3.41 nM) and specificity. Tamer CCE-10F was selected. The diagnostic validity of the aptamer is that it not only detects CRC cell-derived sEVs at a concentration ranging from 2.0 Х 10 4 - 1.0 Х 10 6 particles/μL, and achieves a detection limit of 1.0 Х 10 3 particles/μL, but also detects sEVs in healthy serum, normal This was demonstrated by distinguishing between normal sEVs and non-target sEVs derived from cells and other cancer cells. Additionally, the biological activity of SW620 sEV biomarkers was closely investigated by systematic analyzes including cell survival, cell uptake, wound healing, transwell cell migration/invasion, and tube formation assays. We confirmed that the biomarkers targeted by CCE-10F promote cell migration, invasion, and angiogenesis through sEV-mediated intercellular communication and may be responsible for additional tumor metastasis processes such as EMT. Therefore, it is expected that the DNA aptamer according to the present invention can be used for diagnosis and treatment in the field of colon cancer liquid biopsy.

통계 분석statistical analysis

모든 통계 분석은 GraphPad Prism 8 소프트웨어(GraphPad Software, San Diego, CA, USA)를 사용하여 수행하였다. 데이터는 평균 ± 표준 편차(SD)로 표시된다. 두 샘플 간, 두 개 이상의 샘플 사이의 통계적 차이는 스튜던트 t-검정과 one-way ANOVA(일원배치 분산분석)을 사용하여 평가하였다. p < 0.05, 0.01, 0.001 및 0.0001은 유의한 것으로 간주되어 각각 *, **, *** 및 ****로 표시되는 반면, p > 0.05는 유의하지 않음(ns)을 나타낸다. 모든 실험은 삼중으로 수행하였다.All statistical analyzes were performed using GraphPad Prism 8 software (GraphPad Software, San Diego, CA, USA). Data are expressed as mean ± standard deviation (SD). Statistical differences between two samples and between two or more samples were evaluated using Student's t-test and one-way ANOVA (one-way analysis of variance). p < 0.05, 0.01, 0.001 and 0.0001 are considered significant and are indicated as *, **, *** and ****, respectively, while p > 0.05 indicates not significant (ns). All experiments were performed in triplicate.

[실시예 2][Example 2]

[준비예][Preparation example]

세포 배양cell culture

SW620(인간 대장암), LS 174T(인간 대장암), HT29 (인간 대장암), CCD-18Co(인간 정상 결장) 세포주는 한국 세포주 은행(KCLB)으로부터 획득했다. SW620, LS 174T, HT29 및 CCD-18Co 세포는 DMEM(Welgene, 한국 경산)에서 배양하였다. 모든 세포주는 37℃ 5% CO2의 습한 조건의 10%(v/v) FBS(Gibco; Thermo Fisher Scientific, Waltham, MA, USA) 및 1%(v/v) 페니실린-스트렙토마이신(Welgene)이 보충된 각각의 배지에서 배양되었다. SW620 (human colon cancer), LS 174T (human colon cancer), HT29 (human colon cancer), and CCD-18Co (human normal colon) cell lines were obtained from the Korea Cell Line Bank (KCLB). SW620, LS 174T, HT29, and CCD-18Co cells were cultured in DMEM (Welgene, Gyeongsan, Korea). All cell lines were incubated with 10% (v/v) FBS (Gibco; Thermo Fisher Scientific, Waltham, MA, USA) and 1% (v/v) penicillin-streptomycin (Welgene) in humidified conditions at 37°C and 5% CO2. Cultured in each supplemented medium.

sEV (small extracellular vesicle)의 분리Isolation of small extracellular vesicles (sEV)

약 80% confluence 상태에서, 세포를 5%(v/v) sEVs이 고갈된 FBS(Gibco; Thermo Fisher Scientific) 및 1%(v/v) 페니실린-스트렙토마이신이 보충된 조건 배지로 교체하고 48시간 동안 37℃에서 가습된 조건의 5% CO2 인큐베이터에서 배양하였다. 분비된 sEVs을 함유하는 조절 배지를 회수하여 sEVs을 농축, 분리 및 정제하여 추가 사용을 하였다. 컨디셔닝된 배지는 먼저 5분 동안 300 x g, 20분 동안 2,000 x g 및 30분 동안 10,000 x g에서 일련의 원심분리를 수행하여 세포, 세포 파편, 미세소포체 및 세포자멸체를 각각 제거했다. 그 후, 0.45 및 0.22 μm의 기공 크기를 갖는 필터(Sartorius, Gφttingen, Germany)를 사용하여 상청액을 연속적으로 시린지 여과하여 200 nm보다 큰 크기의 세포외 소포체를 배제하였다. 농축 및 정제를 위해 배지가 원하는 부피로 농축될 때까지 300K 멤브레인(Pall Corporation, New York, NY, USA)이 있는 접선 흐름 여과 시스템(tangential flow filtration system)을 사용했다. 다음으로, 농축된 배지는 qEV 자동화 분획 수집기(Izon Science, Christchurch, NZ)에서 qEV10/35 nm 컬럼을 사용하여 크기 배제 크로마토그래피를 수행하여 sEVs을 높은 수율 및 순도로 분리했다. 이어서, SEC의 sEVs을 포함하는 분획을 풀링하고 3K 멤브레인이 있는 Macrosep(Pall Corporation)에서 5,000 x g에서 20분 동안 다시 원심분리하여 sEVs을 추가로 농축한 다음, -80℃에서 PBS에 사용할 때까지 보관했다. 모든 원심분리 단계는 sEVs 안정성을 보장하기 위해 4℃에서 수행하였다. 일련의 원심분리 전에 1X PBS로 희석한 것을 제외하고는 위에서 설명한 것과 동일한 방식으로 인간 혈청(HS)(Sigma-Aldrich, St Louis, MO, USA)으로부터 sEVs을 분리하였다.At approximately 80% confluence, cells were replaced with conditioned medium supplemented with 5% (v/v) sEVs-depleted FBS (Gibco; Thermo Fisher Scientific) and 1% (v/v) penicillin-streptomycin and incubated for 48 h. Cultured in a 5% CO 2 incubator under humidified conditions at 37°C. The conditioned medium containing the secreted sEVs was recovered, and the sEVs were concentrated, separated, and purified for further use. The conditioned medium was first subjected to a series of centrifugations at 300 xg for 5 min, 2,000 xg for 20 min, and 10,000 xg for 30 min to remove cells, cell debris, microvesicles, and apoptotic bodies, respectively. Afterwards, the supernatant was sequentially syringe filtered using filters with pore sizes of 0.45 and 0.22 μm (Sartorius, Gϕttingen, Germany) to exclude extracellular vesicles larger than 200 nm in size. For concentration and purification, a tangential flow filtration system with a 300K membrane (Pall Corporation, New York, NY, USA) was used until the medium was concentrated to the desired volume. Next, the concentrated medium was subjected to size exclusion chromatography using a qEV10/35 nm column in a qEV automated fraction collector (Izon Science, Christchurch, NZ) to isolate sEVs in high yield and purity. Fractions containing sEVs from SEC were then pooled and centrifuged again for 20 min at 5,000 did. All centrifugation steps were performed at 4°C to ensure sEVs stability. sEVs were isolated from human serum (HS) (Sigma-Aldrich, St Louis, MO, USA) in the same manner as described above, except that they were diluted in 1× PBS before serial centrifugation.

실시예 2-1.Example 2-1.

SW620 및 HS 유래 sEV (small extracellular vesicle; 이하 sEV)의 특성화Characterization of SW620 and HS-derived small extracellular vesicles (sEVs)

SW620, LS174T, HT29(양성 표적) 세포 및 건강한 인간 혈청(HS, 대항 표적)에서 분리된 sEV의 무결성 및 sEV 마커는 NTA, cryo-TEM 및 웨스턴 블롯 분석을 사용하여 평가하였다. The integrity and sEV markers of sEVs isolated from SW620, LS174T, HT29 (positive target) cells and healthy human serum (HS, counter target) were assessed using NTA, cryo-TEM, and Western blot analysis.

나노입자 추적 분석(NTA)Nanoparticle Tracking Analysis (NTA)

본 실시예에서는 MONO Zetaview(PMX-130, Particle Metrix, Bavaria, Germany)를 광 산란(light scatter) 모드에서 사용하여 sEV (small extracellular vesicle; 이하 sEV) 샘플의 농도와 크기를 결정했다. 샘플에 대한 NTA 분석에 앞서, 기기는 100 nm 폴리스티렌 표준 비드(탈이온수에서 1:250,000 희석)를 사용하여 보정하였다. sEV 샘플은 측정을 위해 기기에 적용하기 전에 1x PBS를 사용하여 4 - 6 x 107 입자/mL로 희석하였다. 모든 샘플에 대해 NTA 분석을 위한 세부 매개변수는 최소 밝기 30, 감도 80, 셔터 100, 셀 온도 23℃로 일관되게 설정하였다. 본 실험의 데이터는 ZetaView 분석을 사용하여 분석되었다. 분석에 사용된 탈이온수와 PBS는 최적의 측정을 위해 0.1 μm 주사기 필터 멤브레인으로 필터하였다.In this example, MONO Zetaview (PMX-130, Particle Metrix, Bavaria, Germany) was used in light scatter mode to determine the concentration and size of sEV (small extracellular vesicle; hereinafter sEV) samples. Prior to NTA analysis of samples, the instrument was calibrated using 100 nm polystyrene standard beads (diluted 1:250,000 in deionized water). sEV samples were diluted to 4 - 6 x 10 7 particles/mL using 1x PBS before applying to the instrument for measurement. For all samples, detailed parameters for NTA analysis were consistently set to minimum brightness of 30, sensitivity of 80, shutter of 100, and cell temperature of 23°C. Data from this experiment was analyzed using ZetaView analysis. Deionized water and PBS used in the analysis were filtered with a 0.1 μm syringe filter membrane for optimal measurement.

저온 투과 전자 현미경(Cryo-TEM)Cryo-transmission electron microscopy (Cryo-TEM)

sEV 샘플의 크기와 형태를 분석하기 위해 샘플을 Cryo-TEM 장비에 적용했다. 탄소 코팅 구리 그리드 (200 메쉬; Quantifoil, Großlφbichau, Germany)를 먼저 글로우 방전(glow-discharge)하여 친수성으로 만들고, 각 sEV 샘플(3 μL)을 적용했다. 이후, Thermo Scientific Vitrobot(Thermo Fisher Scientific)을 사용하여 액체 에탄에 넣고 액체 질소로 유지한 다음, 액체 질소를 사용하여 온도를 약 -180℃로 지속적으로 유지하는 냉동 홀더(cryoholder)로 옮겨 유리화했다. Cryo-TEM 이미지는 CMOS 카메라와 결합된 JEM-2100PLUS 전자 현미경(JEOL, Tokyo, Japan)을 사용하여 25kV에서 획득하였다.To analyze the size and morphology of sEV samples, the samples were subjected to Cryo-TEM equipment. Carbon-coated copper grids (200 mesh; Quantifoil, Großlϕbichau, Germany) were first made hydrophilic by glow-discharge, and then each sEV sample (3 μL) was applied. Then, they were placed in liquid ethane using a Thermo Scientific Vitrobot (Thermo Fisher Scientific), maintained in liquid nitrogen, and then transferred to a cryoholder where the temperature was continuously maintained at approximately -180°C using liquid nitrogen for vitrification. Cryo-TEM images were acquired at 25 kV using a JEM-2100PLUS electron microscope (JEOL, Tokyo, Japan) coupled with a CMOS camera.

도 14의 (A) 내지 도 14의 (D)는 본 실시예에 따라 분석된 sEV의 특성을 나타낸다. 도 14의 (A)는 SW620 sEV에 대한 NTA 결과 (삽입 이미지: cryo-TEM 이미지), 도 14의 (B)는 LS 174T sEV에 대한 NTA 결과(삽입 이미지: cryo-TEM 이미지), 도 14의 (C)는 HT29 sEV에 대한 NTA 결과(삽입 이미지: cryo-TEM 이미지). 도 14의 (D)는 HS sEV에 대한 NTA 결과(삽입 이미지: cryo-TEM 이미지)를 나타낸다. 각 cryo-TEM 이미지에서, 스케일 바: 100 nm이다. Figures 14 (A) to 14 (D) show the characteristics of sEVs analyzed according to this example. Figure 14 (A) is the NTA result for SW620 sEV (insert image: cryo-TEM image), Figure 14 (B) is the NTA result for LS 174T sEV (insert image: cryo-TEM image), Figure 14 (C) NTA results for HT29 sEV (inset image: cryo-TEM image). Figure 14 (D) shows the NTA results for HS sEV (insert image: cryo-TEM image). In each cryo-TEM image, scale bar: 100 nm.

웨스턴 블로팅Western blotting

제조업체의 지침에 따라 동일한 부피의 SW620, LS 174T, HT29 세포 및 HS 유래 sEVs을 qEV 농축 키트 (Izon Science)을 이용하여 농축했다. 각 샘플에 5x SDS-PAGE loading buffer (Biosesang, Seongnam, Republic of Korea)를 보충하고, 95℃에서 10분간 가열하고, Mini-Protean tetra 시스템(Bio-Rad Laboratories)을 사용하여 300V에서 20분 동안 10% TGX stain-free protein gel (Bio-Rad Laboratories, Hercules, CA, USA)에 용해하였다. 분해된 단백질을 제조업체의 지침에 따라 Trans-Blot Turbo Transfer System(Bio-Rad Laboratories)에서 0.2 μm PVDF 멤브레인(Bio-Rad Laboratories)으로 옮겼다. 멤브레인을 0.1% TBST(0.1% Tween-20이 포함된 1x TBS)로 용해한 5% BSA TBST 용액으로 블락킹하고, 1차 항체[마우스 항-CD63(Santa Cruz, Dallas, TX, USA), 토끼 항- Hsp90α 및 토끼 항 칼넥신(ABclonal, Woburn, MA, USA), 1:1,000] Can Get Signal 용액 1(Toyobo, New York, NY, USA)와 함께 4℃에서 밤새 인큐베이션한 다음 0.1% TBST로 3회 세척하였다. 그런 다음 멤브레인을 Can Get Signal 용액 2(Toyobo)에 희석된 해당 HRP-접합 2차 항체[HRP 염소 항-토끼 IgG(ABclonal), 1:10,000 및 HRP 염소 항-마우스 IgG(Biolegend, San Diego, CA, USA), 1:5000]와 함께 실온에서 1시간 동안 인큐베이션하고, 0.1% TBST로 4회 세척했다. 멤브레인의 단백질은 ChemiDoc Imaging System(Bio-Rad Laboratories)에서 Clarity Western ECL 기질(Bio-Rad Laboratories)을 사용하여 시각화하였다.Equal volumes of SW620, LS 174T, HT29 cells and HS-derived sEVs were concentrated using a qEV enrichment kit (Izon Science) according to the manufacturer's instructions. Each sample was supplemented with 5x SDS-PAGE loading buffer (Biosesang, Seongnam, Republic of Korea), heated at 95°C for 10 min, and incubated at 300 V for 10 min for 20 min using the Mini-Protean tetra system (Bio-Rad Laboratories). % TGX stain-free protein gel (Bio-Rad Laboratories, Hercules, CA, USA). Digested proteins were transferred to 0.2 μm PVDF membranes (Bio-Rad Laboratories) in the Trans-Blot Turbo Transfer System (Bio-Rad Laboratories) according to the manufacturer's instructions. Membranes were blocked with 5% BSA TBST solution dissolved in 0.1% TBST (1x TBS with 0.1% Tween-20) and incubated with primary antibodies [mouse anti-CD63 (Santa Cruz, Dallas, TX, USA), rabbit anti-CD63 (Santa Cruz, Dallas, TX, USA). - Hsp90α and rabbit anti-calnexin (ABclonal, Woburn, MA, USA), 1:1,000] incubated overnight at 4°C with Can Get Signal solution 1 (Toyobo, New York, NY, USA), followed by 3 with 0.1% TBST. Washed twice. The membrane was then incubated with the corresponding HRP-conjugated secondary antibodies [HRP goat anti-rabbit IgG (ABclonal), 1:10,000 and HRP goat anti-mouse IgG (Biolegend, San Diego, CA) diluted in Can Get Signal solution 2 (Toyobo). , USA), 1:5000] for 1 hour at room temperature and washed four times with 0.1% TBST. Proteins on the membrane were visualized using Clarity Western ECL substrate (Bio-Rad Laboratories) on a ChemiDoc Imaging System (Bio-Rad Laboratories).

그 결과, 도 14의 (A) 내지 (D)에 표시된 바와 같이 SW620 sEV, LS 174T sEV, HT29 sEV, HS sEV는 약 100nm 내지 150nm이며, 이는 cryo-TEM을 사용하여 얻은 값과 일치하였다. 또한 SW620, LS 174T, HT29 및 HS 유래 sEV 은 모두 원형 형태를 보였고 이전 문헌과 일치하는 크기 범위 내에 존재하여 sEV 이 SEC 방법을 사용하여 효과적으로 분리되었음을 확인했다. 또한, 도 14의 (E)에 표시된 웨스턴 블롯 이미지는 분리된 sEVs이 잘 알려진 sEVs 바이오마커인 CD63 및 Hsp90α에 대해 양성이고, 소포체(ER) 막에 대한 마커인 칼넥신(calnexin)에 대해 음성임을 확인하였다. 이러한 모든 분석을 통해 분리된 sEVs이 E-SELEX에 적합한 표적임을 확인했다.As a result, as shown in (A) to (D) of Figure 14, SW620 sEV, LS 174T sEV, HT29 sEV, and HS sEV were about 100 nm to 150 nm, which was consistent with the value obtained using cryo-TEM. Additionally, SW620, LS 174T, HT29 and HS-derived sEVs all showed circular morphology and were within a size range consistent with previous literature, confirming that sEVs were effectively isolated using the SEC method. In addition, the Western blot image shown in Figure 14 (E) shows that the isolated sEVs were positive for CD63 and Hsp90α, well-known sEVs biomarkers, and negative for calnexin, a marker for the endoplasmic reticulum (ER) membrane. Confirmed. All these analyzes confirmed that isolated sEVs were suitable targets for E-SELEX.

실시예 2-2.Example 2-2.

SW620 유래 sEVs에 대한 압타머의 시험관 내 선택In vitro selection of aptamers against SW620-derived sEVs

도 15에서 볼 수 있듯이 TEV-SELEX의 중요한 단계는 결합한 압타머 풀과 결합하지 않은 압타머 풀을 효율적으로 분리하는 것이다. 이를 위해 생물학적으로 활성인 sEVs의 무결성을 유지할 수 있을 뿐만 아니라 진단 가치가 있는 고품질 압타머의 선택을 용이하게 할 수 있는 손쉬운 표적 sEVs 고정화 플랫폼으로 면역 플레이트를 사용했다.As can be seen in Figure 15, an important step in TEV-SELEX is to efficiently separate the bound and unbound aptamer pools. To this end, we used immunoplates as a facile targeted sEVs immobilization platform that can not only maintain the integrity of biologically active sEVs but also facilitate the selection of high-quality aptamers with diagnostic value.

Toggle sEV-SELEX(TEV-SELEX)Toggle sEV-SELEX(TEV-SELEX)

사용된 모든 올리고뉴클레오티드는 Integrated DNA Technologies(IDT; Coralville, IA, USA)에 의해 합성되었다. DNA 라이브러리에 포함된 DNA 형태와 사용되는 정방향 및 역방향 프라이머의 형태는 하기 표 5와 같다.All oligonucleotides used were synthesized by Integrated DNA Technologies (IDT; Coralville, IA, USA). The types of DNA included in the DNA library and the types of forward and reverse primers used are shown in Table 5 below.

Sequence (5'→3')Sequence (5'→3') 서열번호sequence number Random libraryRandom library ATCCAGAGTGACGCAGCA-N40-CTGGCTCGAACAAGCTTGCATCCAGAGTGACGCAGCA-N 40 -CTGGCTCGAACAAGCTTGC 1One Forward primerForward primer ATCCAGAGTGACGCAGCAATCCAGAGTGACGCAGCA 22 Reverse primerReverse primer GCAAGCTTGTTCGAGCCAGGCAAGCTTGTTCGAGCCAG 33

PCR 증폭 단계에 사용된 양 말단 프라이머 영역 사이에 40개의 뉴클레오티드(N40)의 중앙 랜덤 영역으로 랜덤 라이브러리를 설계하였다. A random library was designed with a central random region of 40 nucleotides (N40) between the primer regions at both ends used in the PCR amplification step.

세 종류의 다른 CRC 세포(SW620, LS 174T, HT29) 및 HS에서 유래된 sEV는 각각 양성 및 대항 표적으로 사용하였고, TEV-SELEX는 약간의 수정을 거쳐 공지된 방법으로 수행되었다.sEVs derived from three different CRC cells (SW620, LS 174T, HT29) and HS were used as positive and counter targets, respectively, and TEV-SELEX was performed by a known method with some modifications.

TEV-SELEX 과정 전반에 걸쳐 BSA에 대한 ssDNA결합 또는 맥시-결합(Maxi-binding) 면역 플레이트 자체를 제거하는 음성 선별이 초기에 수행되었다 (SPL Life Sciences, Pocheon, Republic of Korea). 표적 sEVs과 함께 인큐베이션하기 전에, 결합 완충액(BB, 1 g/L BSA, 5 mM MgCl2 및 0.5 mg/mL 연어 정자 ssDNA가 포함된 DPBS)에 용해된 초기 단일 가닥 DNA(ssDNA) 라이브러리(100 nM)를 95℃에서 5분동안 변성하였고, 10분 동안 얼음에서 급속 냉각하였다.Throughout the TEV-SELEX process, negative selection was performed initially to remove ssDNA binding to BSA or the Maxi-binding immunoplate itself (SPL Life Sciences, Pocheon, Republic of Korea). Prior to incubation with target sEVs, a nascent single-stranded DNA (ssDNA) library (100 nM ) was denatured at 95°C for 5 minutes and rapidly cooled on ice for 10 minutes.

음성 선별은 BSA 및 면역 플레이트에 결합할 가능성이 있는 ssDNA를 제거하기 위해 먼저 수행하였다. 1 g/L의 BSA 용액을 면역 플레이트에 접종하고 37℃에서 2시간 동안 인큐베이션했다. 세척 완충액(WB; DPBS with 5 mM MgCl2)으로 각각 30초 동안 진탕기(500 rpm)에서 3회 세척한 후, 준비된 ssDNA 라이브러리를 첨가하고 1시간 동안 배양하였다. 그 후, 결합되지 않은 ssDNA를 포함하는 상층액을 회수하고 제조업체의 지침에 따라 Nucleospin Gel 및 PCR clean-up(Macherey-Nagel, Duren, Germany)을 사용하여 농축했다. Negative selection was performed first to remove ssDNA likely to bind to BSA and immune plates. 1 g/L of BSA solution was inoculated into the immune plate and incubated at 37°C for 2 hours. After washing three times with washing buffer (WB; DPBS with 5 mM MgCl 2 ) on a shaker (500 rpm) for 30 seconds each, the prepared ssDNA library was added and incubated for 1 hour. Afterwards, the supernatant containing unbound ssDNA was recovered and concentrated using Nucleospin Gel and PCR clean-up (Macherey-Nagel, Duren, Germany) according to the manufacturer's instructions.

그런 다음 회수된 ssDNA를 프라이머 농도의 비율이 다른 비대칭 PCR(Asy-PCR)을 사용하여 증폭했다. 간단히 말해서, 0.2 mM dNTP, 각 선택에서 회수된 ssDNA 주형, 충실도가 높은 nPfu-forte DNA 중합효소 및 1x nPfu forte에서 20:1의 정방향-역방향 프라이머 비율을 포함하는 Asy-PCR 반응용 혼합물 버퍼(Enzynomics, 대전, 대한민국)를 준비하였다. The recovered ssDNA was then amplified using asymmetric PCR (Asy-PCR) with different ratios of primer concentrations. Briefly, mixture buffer for Asy-PCR reactions containing 0.2 mM dNTPs, ssDNA template recovered from each selection, high-fidelity nPfu-forte DNA polymerase, and a forward-to-reverse primer ratio of 20:1 in 1x nPfu forte (Enzynomics , Daejeon, Republic of Korea).

열 순환 프로토콜은 다음과 같이 설정하였다: 95℃에서 3분 동안 초기 변성, 이어서 95℃에서 10초(변성), 68℃에서 1분(어닐링 및 신장), 최종 연장의 20주기 68℃에서 5분. 다음으로, Asy-PCR 산물을 2.5% agarose gel에서 분리하고 Nucleospin gel과 PCR clean-up을 이용하여 ssDNA band를 추출 및 정제하였다. The thermal cycling protocol was set up as follows: initial denaturation at 95°C for 3 min, followed by 20 cycles of 10 s at 95°C (denaturation), 1 min at 68°C (annealing and elongation), and a final extension of 5 min at 68°C. . Next, the Asy-PCR product was separated on a 2.5% agarose gel, and the ssDNA band was extracted and purified using Nucleospin gel and PCR clean-up.

양성 선별을 위해 각 양성 표적(sEV, 20 μg/mL)을 면역 플레이트에서 37℃에서 2시간 동안 배양한 다음 WB로 세척(3회)했다. 이어서, 음성 선별로부터 얻은 ssDNA를 웰에 첨가한 다음, 37℃에서 1시간 동안 배양하고 결합되지 않은 ssDNA를 제거하기 위해 WB로 세척(3회)하였다. 다음으로, 각 웰의 샘플을 BB에 현탁시키고 10분 동안 95℃로 가열하여 sEV에서 결합된 ssDNA를 회수한 다음 이전에 설명한 대로 처리했다. For positive selection, each positive target (sEV, 20 μg/mL) was incubated in an immune plate at 37°C for 2 h and then washed (three times) by WB. Then, ssDNA from negative selection was added to the wells, then incubated at 37°C for 1 hour and washed (three times) with WB to remove unbound ssDNA. Next, samples from each well were suspended in BB and heated to 95°C for 10 min to recover bound ssDNA from sEVs and then processed as previously described.

ssDNA 풀과 함께 HS 유래 sEV의 인큐베이션에 의존하는 대항 선별도 음성 선별과 동일한 방식으로 수행하여 정상 혈청 sEV에 대한 ssDNA 결합을 제거했다. 양성 표적 sEV에 대해서만 높은 결합 친화력과 특이성을 갖는 ssDNA 풀을 얻기 위해, 각 TEV-SELEX 루프(하기 표 6)에서 엄격한 조건으로 진행하였다. Counter selection, relying on incubation of HS-derived sEVs with ssDNA pools, was also performed in the same manner as negative selection to eliminate ssDNA binding to normal serum sEVs. To obtain a ssDNA pool with high binding affinity and specificity only for positive target sEVs, stringent conditions were applied in each TEV-SELEX loop (Table 6 below).

TEV-SELEX에 대한 엄격한 조건Strict conditions for TEV-SELEX 양성 선별positive screening 음성 선별voice screening 배양시간Incubation time 1 h (1st-2nd)*
45 min (3rd-4th)
30 min (5-6th)
20 min (7-8th)
1 h (1 st -2 nd ) *
45min ( 3rd -4th )
30 minutes (5-6 days )
20min ( 7-8th )
1 h (1st-2nd)
1.25 h (3rd-4th)
1.5 h (5-6th)
2 h (7-8th)
1 h (1 st -2 nd )
1.25 h ( 3rd -4th )
1.5 h (5-6 th )
2 h (7-8 th )
세척
(30 s, 500 rpm)
wash
(30 s, 500 rpm)
3 Х (1st-2nd)
5 Х (3rd-4th)
7 Х (5-6th)
9 Х (7-8th)
3 Х (1 st -2 nd )
5 Х (3 rd -4 th )
7 Х (5-6 th )
9 Х (7-8 th )
No Washing requiredNo washing required
Competitor
(Salmon sperm DNA)
Competitor
(Salmon sperm DNA)
0.5 mg/ml (2nd-8th)0.5 mg/ml ( 2nd -8th )

*: () 내 숫자는 TEV-SELEX에서 루프 순서를 의미한다. * : () My numbers refer to the loop order in TEV-SELEX.

TEV-SELEX 절차(도 15)에서 음성 선별은 버퍼 및/또는 면역 플레이트의 구성 요소에 비특이적으로 결합하는 표적 외 단일 가닥 DNA(ssDNA)를 제거하기 위해 수행하였다. 음성 선별에서 얻은 결합되지 않은 ssDNA는 3개의 E-SELEX 루프를 포함하는 추가 선택을 위해 강화되었으며, 여기서 각 루프(SW620 sEV, LS 174T sEV, HT29 sEV, HS sEV; 순서대로 1st-8th 루프)는 세 번의 연속적인 양성 선별 및 한 번의 대항 선별을 포함했다. 이질성이 높은 대장암 특성을 고려하여 SW620, LS 174T, HT29 세 종류의 CRC 세포 유래 sEVs을 양성 표적으로 선택하였고, HS 유래 sEVs은 잠재적인 생체 내 적용을 위한 임상적 환경을 설정하기 위해 대항 표적으로 결정하였다. 루프는 총 8 회 반복되었으며, 그 동안 압타머의 결합 친화도 및 특이성을 개선하기 위해 엄격한 조건을 점진적으로 증가시켜 수행하였다(표 2). 또한, 자기 분리, 효소 분해 또는 추가 NaOH 처리를 위한 비오틴 또는 인산염 그룹 접합을 필요로 하지 않는 Asy-PCR을 사용하여 ssDNA 증폭과 생성을 한 단계로 수행하여 TEV-SELEX의 전체 프로세스를 단순화하였다. Negative selection in the TEV-SELEX procedure (Figure 15) was performed to remove non-target single-stranded DNA (ssDNA) that non-specifically binds to the buffer and/or components of the immune plate. Unbound ssDNA from negative selection was enriched for further selection containing three E-SELEX loops, where each loop (SW620 sEV, LS 174T sEV, HT29 sEV, HS sEV; 1 st -8 th loop in that order) ) included three consecutive positive screenings and one counterscreening. Considering the highly heterogeneous characteristics of colorectal cancer, three types of CRC cell-derived sEVs, SW620, LS 174T, and HT29, were selected as positive targets, and HS-derived sEVs were selected as countertargets to establish a clinical environment for potential in vivo application. decided. The loop was repeated a total of eight times, during which the stringency conditions were gradually increased to improve the binding affinity and specificity of the aptamer (Table 2). Additionally, the overall process of TEV-SELEX was simplified by performing ssDNA amplification and generation in one step using Asy-PCR, which does not require magnetic separation, enzymatic digestion, or biotin or phosphate group conjugation for additional NaOH treatment.

TEV-SELEX의 성공을 위한 또 다른 중요한 고려 사항은 TEV-SELEX 절차를 모니터링하여 수행되는 루프 수를 결정하는 것이다. qPCR은 다양한 서열의 DNA 풀에 대해 연구할 수 있고, ssDNA pool 다양성 측면에서 TEV-SELEX를 통해 회수한 ssDNA의 균질성에 대한 중요한 정보를 제공한다. 각 루프(1st-8th)에서 얻는 ssDNA를 qPCR로 분석하여 루프 수를 선정하였다.Another important consideration for the success of TEV-SELEX is monitoring the TEV-SELEX procedure to determine the number of loops performed. qPCR can study DNA pools of diverse sequences and provides important information about the homogeneity of ssDNA recovered through TEV-SELEX in terms of ssDNA pool diversity. The number of loops was selected by analyzing ssDNA obtained from each loop (1 st -8 th ) by qPCR.

정량적 중합효소 연쇄 반응(qPCR)Quantitative polymerase chain reaction (qPCR)

qPCR을 사용하여 TEV-SELEX 동안 선택 효율성을 평가했다. TOPreal qPCR 프리믹스(Enzynomics)에서 최종농도 1X의 qPCR완충액을 각 선택에서 회수된 ssDNA 주형과 혼합하였다. qPCR은 다음과 같은 열순환 매개변수를 사용하여 CFX Connect Real-Time PCR 감지 시스템(Bio-Rad Laboratories)에서 수행되었다: 95℃에서 15분 동안 초기 변성(활성화), 이어서 95℃에서 10초(변성), 60℃에서 15초(어닐링), 72℃에서 15초의 20주기(연장). 그 다음에는 0.5℃의 증분 속도로 55℃에서 95℃로 온도를 점진적으로 증가시키면서 용융 온도(Tm) 분석이 이어졌다. qPCR 증폭 곡선과 Tm은 CFX Maestro Software(Bio-Rad Laboratories)로 분석되었다.Selection efficiency was assessed during TEV-SELEX using qPCR. In TOPreal qPCR premix (Enzynomics), qPCR buffer at a final concentration of 1X was mixed with the ssDNA template recovered from each selection. qPCR was performed on a CFX Connect Real-Time PCR Detection System (Bio-Rad Laboratories) using the following thermocycling parameters: initial denaturation (activation) at 95°C for 15 min, followed by 10 s at 95°C (denaturation). ), 20 cycles of 15 s at 60°C (annealing) and 15 s at 72°C (extension). This was followed by melting temperature (Tm) analysis while gradually increasing the temperature from 55°C to 95°C at an increment rate of 0.5°C. qPCR amplification curves and Tm were analyzed with CFX Maestro Software (Bio-Rad Laboratories).

ssDNA의 초기 라이브러리는 40개 뉴클레오티드의 무작위 영역으로 인해 서열이 매우 이질적이다. 그러나 표적에 특이적으로 결합하는 압타머 풀을 지속적으로 반복적인 선택 과정이 수행될 것이기 때문에 이질성은 감소될 것이다. 이러한 예상되는 동작은 증폭 및 용융 곡선의 변화를 통해 입증하였다(도 16). 도 16의 (A)에서 랜덤 라이브러리(적색선; Lib)의 증폭은 초기 증가를 보여 최대 형광 강도에 도달하지만, 프라이머 영역은 완전히 혼성화되는 반면, 중앙의 무작위 영역은 부분적으로 비상보적 혼성화를 이루게 되어 불안정한 헤테로듀플렉스의 형성으로 인해 급격한 형광 감소가 나타났다. 이러한 결과는 도 16의 (C)에서 68.5℃의 낮은 용융 온도(Tm)는 헤테로듀플렉스의 불안정성과 잘 일치하였음을 나타낸다. 대조적으로, TEV-SELEX의 반복적인 루프는 높은 결합 친화도로 결합하는 특정 ssDNA 풀을 풍부하게 하여 균질성을 증가시켰다. 따라서 8th-loop(연한 핑크선) 후 얻은 ssDNA는 qPCR 동안 극한 안정기를 갖는 정상 지수 곡선을 나타냈고 Tm 피크는 낮은 온도에서 Tm 피크가 81.5℃의 더 높은 온도로 완전히 이동하였다. 이는 높은 안정성을 가지는 호모듀플렉스의 비율이 크게 증가했음을 의미한다.The initial library of ssDNA is highly heterogeneous in sequence due to a random region of 40 nucleotides. However, heterogeneity will be reduced because an iterative process of selecting a pool of aptamers that specifically bind to the target will be continuously performed. This expected behavior was demonstrated through changes in amplification and melting curves (Figure 16). In Figure 16 (A), amplification of the random library (red line; Lib) shows an initial increase and reaches maximum fluorescence intensity, but while the primer region is completely hybridized, the random region in the center partially achieves non-complementary hybridization, making it unstable. A rapid decrease in fluorescence occurred due to the formation of heteroduplex. These results indicate that the low melting temperature (Tm) of 68.5°C in Figure 16 (C) is consistent with the instability of the heteroduplex. In contrast, the repetitive loops of TEV-SELEX increased homogeneity by enriching specific ssDNA pools that bind with high binding affinity. Therefore, the ssDNA obtained after 8 th -loop (light pink line) showed a normal exponential curve with an extreme plateau during qPCR, and the Tm peak completely shifted from the lower temperature to the higher temperature of 81.5°C. This means that the proportion of homoduplexes with high stability has increased significantly.

또한, 균질성의 정도를 추정하기 위해 최종 qPCR 사이클에서 최대 형광 강도에 대한 마지막 사이클 (20th)의 형광 강도의 비율을 계산하여 증폭 곡선을 정량적으로 분석했다. 이 연구에서는 0.95보다 높은 비율을 증폭의 안정기로 간주하였고, ssDNA 라이브러리에서 높은 균질성을 보장하기에 충분했다. 도 16의 (B)에서 무작위 ssDNA 라이브러리(Lib)는 낮은 비율을 보여 상당한 이질성을 반영하지만 TEV-SELEX 루프가 지속적으로 반복될수록 비율이 증가한다. 8th-loop(연한 핑크선)만이 형광 비율이 0.95보다 큰 값을 보여, ssDNA 균질성의 증가로 인해 8th-loop가 증폭에서 안정기에 도달했음을 증명했다. Additionally, to estimate the degree of homogeneity, amplification curves were analyzed quantitatively by calculating the ratio of the fluorescence intensity in the last cycle ( 20th ) to the maximum fluorescence intensity in the final qPCR cycle. In this study, a ratio higher than 0.95 was considered a plateau of amplification and was sufficient to ensure high homogeneity in ssDNA libraries. In Figure 16 (B), the random ssDNA library (Lib) shows a low ratio, reflecting significant heterogeneity, but the ratio increases as the TEV-SELEX loop is continuously repeated. Only the 8 th -loop (light pink line) showed a fluorescence ratio greater than 0.95, proving that the 8 th -loop reached a plateau in amplification due to the increase in ssDNA homogeneity.

qPCR 모니터링의 이러한 모든 결과는 8th-loop이 ssDNA 풀의 최대 농축을 달성하는 데 최적임을 보여주었다. 따라서 8th-loop에서 얻은 ssDNA를 NGS 분석에 적용했다.All these results from qPCR monitoring showed that 8 th -loops were optimal to achieve maximum enrichment of the ssDNA pool. Therefore, ssDNA obtained from the 8 th -loop was subjected to NGS analysis.

실시예 2-3.Example 2-3.

후보 압타머 평가Candidate aptamer evaluation

상기 실시예 2의 8번째 사이클에서 얻은 ssDNA를 NGS로 분석하고 빈도수(%)를 기준으로 상위 10개 후보 압타머를 선택하여 추가 평가를 수행했다. The ssDNA obtained in the 8th cycle of Example 2 was analyzed by NGS, and the top 10 candidate aptamers were selected based on frequency (%) for further evaluation.

차세대 시퀀싱(NGS)Next-generation sequencing (NGS)

TEV-SELEX의 최종 루프(8th)에서 농축된 ssDNA 풀을 프라이머로 대칭적으로 PCR 증폭하여 dsDNA를 생성하고, 아가로스 겔(2.5%)에서 분해하고, Nucleospin Gel 및 PCR clean-up을 사용하여 정제했다(Macherey-Nagel). 마지막으로 정제된 dsDNA를 처리하여 후보 압타머의 서열을 NGS(next-generation sequencing)를 통해 분석하였으며, 이는 시퀀싱 업체(Clinomics, Ulsan, Republic of Korea)에서 수행하였다.The concentrated ssDNA pool from the final loop ( 8th ) of TEV-SELEX was symmetrically PCR amplified with primers to generate dsDNA, resolved on an agarose gel (2.5%), and using Nucleospin Gel and PCR clean-up. Purified (Macherey-Nagel). Finally, the purified dsDNA was processed and the sequence of the candidate aptamer was analyzed through NGS (next-generation sequencing), which was performed by a sequencing company (Clinomics, Ulsan, Republic of Korea).

정제된 dsDNA는 제조업체의 지침(MGI FS DNA 라이브러리 준비 세트)에 따라 페어드-엔드(PE) 150 시퀀싱을 위해 Frag 효소(MGI, Shenzhen, China)로 단편화되었다. 단편화된 DNA는 DNA 클린 비드(MGI)에 의해 300~500 bp 사이로 추가로 선택되었으며, 그런 다음 복구되어 blunt-end를 생성하고 3'말단에서 단일 아데노신 잔기를 갖도록 변형되었다. 5'단일 티민 잔기를 갖는 어댑터 서열은 DNA 단편의 양쪽 말단에 결찰시켰다. 그런 다음, 결찰 생성물을 7주기 동안 증폭시키고 다음과 같은 단일 가닥 순환화 과정을 거쳤다:Purified dsDNA was fragmented with Frag enzyme (MGI, Shenzhen, China) for paired-end (PE) 150 sequencing according to the manufacturer's instructions (MGI FS DNA library preparation set). The fragmented DNA was further selected between 300 and 500 bp by DNA clean beads (MGI), and then recovered to generate a blunt-end and modified to have a single adenosine residue at the 3' end. An adapter sequence with a 5' single thymine residue was ligated to both ends of the DNA fragment. The ligation product was then amplified for 7 cycles and subjected to the following single-strand circularization process:

간략하게 설명하면, PCR 산물을 역상보적인 특수 분자와 함께 열변성시키고, DNA 리가제를 이용하여 단일 가닥 원형 DNA 라이브러리를 제작하고, 나머지 선형 DNA를 엑소뉴클레아제로 분해하였다. DNA 라이브러리는 PE 판독 길이가 150 bp인 DNBSEQ-T7(MGI)을 사용하여 최종적으로 시퀀싱되었으며 FastQC(v0.11.8)에 의해 평가되어 MGI 시퀀싱 플랫폼의 전체 시퀀싱 품질을 평가했다.Briefly, the PCR product was heat denatured with a special reverse complementary molecule, a single-stranded circular DNA library was created using DNA ligase, and the remaining linear DNA was digested with exonuclease. The DNA library was finally sequenced using DNBSEQ-T7 (MGI) with a PE read length of 150 bp and evaluated by FastQC (v0.11.8) to assess the overall sequencing quality of the MGI sequencing platform.

본 실시예에서 상위 10개 후보 압타머의 서열은 하기 [표 7]과 같다. The sequences of the top 10 candidate aptamers in this example are shown in Table 7 below.

Sequence (5'→3')*#Sequence (5'→3')*# ReadRead Frequency (%)Frequency (%) 서열번호sequence number T1T1 ATCCAGAGTGACGCAGCACCTAGAACCCGCACTAACACTCACCACGACTAACACACACCTGGCTCGAACAAGCTTGC ATCCAGAGTGACGCAGCACCTAGAACCCGCACTAACACTCACCACGACTAACACACACCTGGCTCGAACAAGCTTGC 357817357817 3.233.23 1818 T2T2 ATCCAGAGTGACGCAGCAACACTGAAGGGAAGGGAGAGAGGGAGTGTGGAGGGTAAAACTGGCTCGAACAAGCTTGC ATCCAGAGTGACGCAGCAACACTGAAGGGAAGGGAGAGAGGAGTGTGGAGGGTAAAACTGGCTCGAACAAGCTTGC 197439197439 1.781.78 1919 T3T3 ATCCAGAGTGACGCAGCACCCCCAATCCGCCTATGCTATCTGGCCTCCATCTCTCTGTCTGGCTCGAACAAGCTTGC ATCCAGAGTGACGCAGCACCCCCAATCCGCCTATGCTATCTGGCCTCCATCTCTCTGTCTGGCTCGAACAAGCTTGC 188125188125 1.71.7 2020 T4T4 ATCCAGAGTGACGCAGCAACACAGACAAGGCGGTAGAGGAGAGGAGAGGAACTGGCCACTGGCTCGAACAAGCTTGC ATCCAGAGTGACGCAGCAACACAGACAAGGCGGTAGAGGAGAGGAGAGGAACTGGCCACTGGCTCGAACAAGCTTGC 178936178936 1.611.61 2121 T5T5 ATCCAGAGTGACGCAGCAGTGGCCAGTTCCTCTCCTCTCCTCTACCGCCTTGTCTGTGCTGGCTCGAACAAGCTTGC ATCCAGAGTGACGCAGCAGTGGCCAGTTCTCTCTCCTCTCCTCTCTACCGCCTTGTCTGTGCTGGCTCGAACAAGCTTGC 170334170334 1.541.54 2222 T6T6 ATCCAGAGTGACGCAGCAGGGACAAAGGACACAGGTGGGGGGTGTTGGGATCGGGGGTGCTGGCTCGAACAAGCTTGC ATCCAGAGTGACGCAGCAGGGACAAAGGACACAGGTGGGGGGTGTTGGGATCGGGGGTGCTGGCTCGAACAAGCTTGC 159087159087 1.441.44 2323 T7T7 ATCCAGAGTGACGCAGCAGCCTCGCCTCTACTAGATCATACCTCCCTTCCCCTCCGCTCTGGCTCGAACAAGCTTGC ATCCAGAGTGACGCAGCAGCCTCGCCTCTACTAGATCATACCTCCCTTCCCCTCCGCTCTGGCTCGAACAAGCTTGC 147763147763 1.331.33 2424 T8T8 ATCCAGAGTGACGCAGCATGCCACGCCTTTATTTTACGTCCTCTCCCACCCTCTCCTCTCTGGCTCGAACAAGCTTGC ATCCAGAGTGACGCAGCATGCCACGCCTTTATTTTACGTCCTCTCCCACCCTCTCCTCTCTGGCTCGAACAAGCTTGC 146723146723 1.321.32 2525 T9T9 ATCCAGAGTGACGCAGCAGACTAACGGTGCAAAAGTGTGGCAAGAGGGAGAGAGGGGGTCTGGCTCGAACAAGCTTGC ATCCAGAGTGACGCAGCAGACTAACGGTGCAAAAGTGTGGCAAGAGGGAGAGAGGGGGTCTGGCTCGAACAAGCTTGC 145779145779 1.321.32 2626 T10T10 ATCCAGAGTGACGCAGCACACCCCCTCTCTCCCTCTTGCCACACTTTTGCACCGTTAGTCTGGCTCGAACAAGCTTGC ATCCAGAGTGACGCAGCACACCCCCTCTCTCCCTCTTGCCACACTTTTGCACCGTTAGTCTGGCTCGAACAAGCTTGC 136694136694 1.231.23 2727

프라이머 영역은 TEV-SELEX 과정 동안 PCR 증폭시키기 위하여 상기 압타머 서열의 5' 부분에는 서열번호 2의 정방향 프라이머가, 3' 부분에는 서열번호 3의 역방향 프라이머 영역이 ssDNA 라이브러리에 포함되었으나, [표 7]에서 정방향과 역방향 프라이머 부분은 표시되지 않았다. 상기 표 7에서 T는 Toggle의 약자이다In order to PCR amplify the primer region during the TEV-SELEX process, the forward primer of SEQ ID NO: 2 was included in the 5' part of the aptamer sequence, and the reverse primer of SEQ ID NO: 3 was included in the 3' part of the ssDNA library [Table 7 ], the forward and reverse primer portions are not indicated. In Table 7 above, T stands for Toggle.

높은 결합 친화도 및 특이성을 갖는 압타머를 선택하기 위해, 5'-비오틴 그룹으로 변형된 후보 압타머를 제조한 후, SW620, LS 174T, HT29 (양성 표적) 및 HS(대항 표적) sEVs에 대한 결합 친화도는 ELONA 방법을 사용하여 평가하였다. To select aptamers with high binding affinity and specificity, candidate aptamers modified with 5'-biotin groups were prepared and then tested against SW620, LS 174T, HT29 (positive target) and HS (counter target) sEVs. Binding affinity was assessed using the ELONA method.

효소 결합 올리고뉴클레오티드 분석(ELONA)Enzyme-linked oligonucleotide assay (ELONA)

ELONA는 약간의 수정을 거쳐 공지된 방법으로 수행되었다. 맥시 결합 면역 플레이트(SPL) 웰을 37℃에서 2시간 동안 각각의 sEV(1 x 109 입자/mL)으로 먼저 코팅한 다음, 37℃에서 1시간 동안 PBST(0.1% Tween-20이 포함된 1x PBS)에서 3% BSA 용액으로 블락킹하였다. PBST로 3회 세척한 후, 면역 플레이트를 준비된 비오틴 접합 압타머(IDT)와 함께 BB에서 37℃에서 1시간 동안 인큐베이션한 후 0.1% PBST로 3회 세척했다. 면역 플레이트를 0.1% PBST에 희석된 streptavidin-peroxidase polymer(Sigma-Aldrich; 1:1,000)와 함께 37℃에서 1시간 동안 추가로 인큐베이션한 다음, 0.1% PBST로 세척(3회)하였다. 다음으로, 1x TMB 기질 용액(Invitrogen, Carlsbad, CA, USA)을 각 웰에 첨가하고, 암실 및 실온에서 6분 동안 인큐베이션하였다. 0.5 M 황산을 첨가하여 반응을 종료하고, Spectramax iD5 multi-mode microplate reader(Molecular Devices, San Jose, CA, USA)를 이용하여 450 nm에서 흡광도를 즉시 측정하였다. 정규화 값 ΔA는 다음 식 3을 기반으로 계산하였다. ELONA was performed using a known method with some modifications. Maxi-binding immunoplate (SPL) wells were first coated with each sEV (1 Blocking was performed with a 3% BSA solution in PBS). After washing three times with PBST, the immune plate was incubated with prepared biotin-conjugated aptamer (IDT) in BB at 37°C for 1 hour and then washed three times with 0.1% PBST. The immune plate was further incubated with streptavidin-peroxidase polymer (Sigma-Aldrich; 1:1,000) diluted in 0.1% PBST for 1 hour at 37°C and then washed (three times) with 0.1% PBST. Next, 1x TMB substrate solution (Invitrogen, Carlsbad, CA, USA) was added to each well and incubated for 6 min in the dark and at room temperature. The reaction was terminated by adding 0.5 M sulfuric acid, and the absorbance was immediately measured at 450 nm using a Spectramax iD5 multi-mode microplate reader (Molecular Devices, San Jose, CA, USA). The normalized value ΔA was calculated based on the following equation 3.

<식 3><Equation 3>

ΔA = At - Ab ΔA = A t - A b

여기서 At는 sEV와 압타머를 모두 포함하는 샘플의 흡광도이고, Ab는 공백(blank)의 흡광도이다. 압타머의 농도는 분석법에 따라 달라지며, 모든 실험은 3회 반복하였다.Here, A t is the absorbance of the sample containing both sEV and aptamer, and A b is the absorbance of the blank. The concentration of aptamer varies depending on the analysis method, and all experiments were repeated three times.

도 17에 도시된 바와 같이, 상위 빈도수(%) 10개 후보 압타머는 SW620, LS 174T, HT29 및 HS sEVs에 대해 서로 다른 친화성을 보여주었다. SW620, LS 174T, HT29 sEVs에 대한 T6 친화도가 가장 높았고 HS sEVs에 대한 친화도가 낮았다. 전반적으로, 이러한 결과는 SW620, LS 174T, HT29 sEVs에 대한 T6의 높은 결합 친화도 및 특이성을 확인한 것이며; 따라서 T6을 SELEX 후 최적화를 위해 선택하였다.As shown in Figure 17, the 10 candidate aptamers with the highest frequency (%) showed different affinities for SW620, LS 174T, HT29, and HS sEVs. T6 affinity was highest for SW620, LS 174T, and HT29 sEVs, and affinity was lowest for HS sEVs. Overall, these results confirmed the high binding affinity and specificity of T6 for SW620, LS 174T, and HT29 sEVs; Therefore, T6 was selected for optimization after SELEX.

실시예 2-4.Example 2-4.

T6 압타머의 post-SELEX 최적화Post-SELEX optimization of the T6 aptamer

TEV-SELEX 과정 동안 PCR 증폭을 위해 프라이머 영역을 ssDNA 라이브러리에 포함하였다. 더 짧은 길이의 압타머는 더 저렴한 비용으로 합성되고 압타머 기반 바이오센서 개발에 더 적합하기 때문에 정방향 또는 역방향 프라이머 영역을 절단하여 T6 압타머의 post-SELEX 최적화를 수행했다. 정방향, 역방향 또는 두 영역이 모두 없는 T6 압타머를 각각 T6F, T6R, T6FR로 명명하였으며, 추가 영역에 대해 제거한 T6의 파생서열들은 각각 T6RA T6RB, T6RB1, T6RB2 및 T6RB3로 지정하였다 (표 8 참조). T6 압타머는 전술한 [표 7]에서 서열번호 23으로 표시된다. Primer regions were included in the ssDNA library for PCR amplification during the TEV-SELEX process. Because shorter length aptamers are synthesized at lower cost and are more suitable for the development of aptamer-based biosensors, we performed post-SELEX optimization of the T6 aptamer by truncating the forward or reverse primer regions. T6 aptamers lacking the forward, reverse, or both regions were designated T6F, T6R, and T6FR, respectively, and the derived sequences of T6 with the additional regions removed were designated T6RA T6RB, T6RB1, T6RB2, and T6RB3, respectively (see Table 8). . The T6 aptamer is represented by SEQ ID NO: 23 in the above-mentioned [Table 7].

Sequence (5'→3')Sequence (5'→3') 서열번호sequence number Random libraryRandom library ATCCAGAGTGACGCAGCA-N40-CTGGCTCGAACAAGCTTGCATCCAGAGTGACGCAGCA-N 40 -CTGGCTCGAACAAGCTTGC 1One Forward primerForward primer ATCCAGAGTGACGCAGCAATCCAGAGTGACGCAGCA 22 Reverse primerReverse primer GCAAGCTTGTTCGAGCCAGGCAAGCTTGTTCGAGCCAG 33 T6FT6F GGGACAAAGGACACAGGTGGGGGGTGTTGGGATCGGGGGTGCTGGCTCGAACAAGCTTGCGGGACAAAGGACACAGGTGGGGGGTGTTGGGATCGGGGGTGCTGGCTCGAACAAGCTTGC 2828 T6RT6R ATCCAGAGTGACGCAGCAGGGACAAAGGACACAGGTGGGGGGTGTTGGGATCGGGGGTGATCCAGAGTGACGCAGCAGGGACAAAGGACACAGGTGGGGGGTGTTGGGATCGGGGGTG 2929 T6FRT6FR GGGACAAAGGACACAGGTGGGGGGTGTTGGGATCGGGGGTGGGGACAAAGGACACAGGTGGGGGGTGTTGGGATCGGGGTG 3030 T6RAT6RA ATCCAGAGTGACGCAGCAGGGACAAAGGATCCAGAGTGACGCAGCAGGGACAAAGG 3131 T6RBT6RB CAAAGGACACAGGTGGGGGGTGTTGGGATCGGGGGTGCAAAGGACACAGGTGGGGGGGTGTTGGGATCGGGGTGTG 3232 T6RB1T6RB1 ACACAGGTGGGGGGTGTTGGGATCGGGGGTGACACAGGTGGGGGGTGTTGGGATCGGGGGTG 3333 T6RB2T6RB2 CAAAGGACACAGGTGGGGGGTGTCAAAGGACACAGGTGGGGGGTGT 3434 T6RB3T6RB3 ACACAGGTGGGGGGTGTACACAGGTGGGGGGTGT 3535

도 18에서 볼 수 있듯이, 절단된 T6 압타머(T6F, T6R 및 T6FR)와 SW620, LS 174T, HT29 sEVs (양성 표적) 및 HS sEV(대항 표적)에 대한 결합 친화도는 절단되지 않은 T6 압타머의 결합 친화도와 비슷했다 (도 18의 (A)). 단, HS sEVs(대항 표적)에 대한 T6FR의 결합 친화도가 높음으로 특이성이 낮아 선택하지 않는 방향으로 진행하였다. 보다 구체적으로, T6, T6F 및 T6R에 대한 결합 친화도를 정략적으로 비교하기 위해 ΔA대항표적에 대한 ΔA양성표적의 비율을 분석하였고, 그 결과 T6R이 가장 높은 값을 보였다 (도 18의 (B-D)). 전반적으로, 이러한 결과는 SW620, LS 174T, HT29 sEVs에 대한 T6R의 높은 결합 친화도 및 특이성을 확인한 것이며; 따라서 T6R을 최종 선택 후 보다 구체적인 서열 최적화 과정을 진행했다. As can be seen in Figure 18, the binding affinities of the cleaved T6 aptamers (T6F, T6R, and T6FR) to SW620, LS 174T, HT29 sEVs (positive target), and HS sEVs (counter target) are comparable to those of the uncleaved T6 aptamer. was similar to the binding affinity ((A) of Figure 18). However, due to the high binding affinity of T6FR for HS sEVs (countertarget), the specificity was low, so we proceeded in the direction of not selecting. More specifically, to quantitatively compare the binding affinities for T6, T6F and T6R, the ratio of ΔA positive target to ΔA counter target was analyzed, and as a result, T6R showed the highest value ((BD) in Figure 18) ). Overall, these results confirmed the high binding affinity and specificity of T6R for SW620, LS 174T, and HT29 sEVs; Therefore, after final selection of T6R, a more detailed sequence optimization process was performed.

이후, 본 발명자들은 T6R의 다른 영역들을 절단하여(도 19의 (A) 및 (C)), T6RA, T6RB, T6RB1, T6RB2 및 T6RB3를 제작한 후, 상기 실시예에 기재된 바와 같이 각 양성표적(SW620, LS174T, HT29) 및 대항표적(HS)에 대한 결합 친화도를 분석 후 각 절단된 압타머에 대한 ΔA 값을 비교평가하여 이 연구에 가장 적합한 압타머를 가장 적합한 압타머를 T6RB로 선정했다. 도 19의 (B)에 도시된 바와 같이, T6RB에서 ΔA대항표적의 값은 변함이 없지만 ΔA양성표적에 대한 값이 증가하여 가장 높은 결합 친화도 및 특이성을 보였다. 또한, 도 19의 (C)에 도시된 바와 같이, T6RB의 추가적인 서열 최적화는 오히려 ΔA양성표적에 대한 값이 낮았고, 이는 곧 결합 친화도를 감소시키는 서열로 판단되어 최종적으로 T6RB를 최적의 서열로 선정하였다. Afterwards, the present inventors cut other regions of T6R (FIG. 19 (A) and (C)) to produce T6RA, T6RB, T6RB1, T6RB2 and T6RB3, and then each positive target ( After analyzing the binding affinity to SW620, LS174T, HT29) and the counter target (HS), the ΔA value for each cleaved aptamer was compared and evaluated to select the most appropriate aptamer for this study as T6RB. . As shown in Figure 19 (B), the value of the ΔA counter target in T6RB did not change, but the value of the ΔA positive target increased, showing the highest binding affinity and specificity. In addition, as shown in Figure 19 (C), the additional sequence optimization of T6RB resulted in a lower value for the ΔA positive target , which was judged to be a sequence that reduces binding affinity, and T6RB was finally selected as the optimal sequence. selected.

실시예 2-5.Example 2-5.

해리 상수, Kd의 결정Determination of the dissociation constant, Kd

T6RB의 결합 친화도는 K d를 추정하여 정량적으로 평가하였다. 구체적으로, 다양한 농도(0-100 nM)에서 T6RB를 일정한 수의 SW620, LS 174T, HT29 및 HS sEVs에 노출시킨 다음 실시예 5에서 전술한 ELONA 방법을 사용하여 분석했다. The binding affinity of T6RB was quantitatively evaluated by estimating K d . Specifically, T6RB was exposed to a certain number of SW620, LS 174T, HT29 and HS sEVs at various concentrations (0-100 nM) and then analyzed using the ELONA method described above in Example 5.

결합 친화도 분석Binding affinity analysis

선택된 압타머 T6RB의 결합 친화도를 평가하기 위해, 비오틴으로 표지된 선택된 압타머의 농도(0-100 nM)를 변화시키고 위에서 설명한 ELONA 방법을 사용하여 평형 해리 상수(K d)를 결정했다. 정규화된 ΔA 값은 압타머 농도의 함수로 표시되었으며 K d는 비선형 회귀 방정식 Y = (Bmax Х X)/(K d + X)를 사용하여 결정하였다. 여기서 Y는 ΔA, Bmax는 최대 ΔA, K d는 해리 상수, X는 압타머 농도이다.To assess the binding affinity of selected aptamers T6RB, the concentration (0–100 nM) of selected aptamers labeled with biotin was varied and the equilibrium dissociation constant ( K d ) was determined using the ELONA method described above. Normalized ΔA values were expressed as a function of aptamer concentration and K d was determined using the nonlinear regression equation Y = (Bmax Х X)/( K d + X). where Y is ΔA, Bmax is the maximum ΔA, K d is the dissociation constant, and X is the aptamer concentration.

그 결과, 도 20의 (A)는 T6RB의 해리 상수(K d) 곡선을 나타낸다. 도 20의 (A)에서 보는 바와 같이 SW620, LS 174T, HT29 sEV의 경우 T6RB의 농도가 증가함에 따라 흡광도 신호가 증가하여 각각 3.848 nM, 5.904 nM, 5.234 nM의 K d 값을 나타내는 반면, HS sEV의 경우, 흡광도 신호가 변하지 않으므로 K d 값을 결정할 수 없었다. 낮은 나노몰 범위의 K d 값은 SW620, LS 174T, HT29 sEV에 대한 T6RB의 높은 결합 친화도를 시사했다. 이에, T6RB를 진단 응용 분야에 적합한 후보 압타머로 판단하였다. As a result, Figure 20 (A) shows the dissociation constant ( K d ) curve of T6RB. As shown in Figure 20 (A), in the case of SW620, LS 174T, and HT29 sEV, the absorbance signal increased as the concentration of T6RB increased, showing K d values of 3.848 nM, 5.904 nM, and 5.234 nM, respectively, while HS sEV In the case of , the absorbance signal did not change, so the K d value could not be determined. K d values in the low nanomolar range suggested high binding affinity of T6RB for SW620, LS 174T, and HT29 sEVs. Accordingly, T6RB was determined to be a suitable candidate aptamer for diagnostic applications.

원형 이색성(Circular dichroism, CD) 측정Circular dichroism (CD) measurement

10 mM Tris/HCl(pH 7.4) 및/또는 100 mM KCl이 포함된 완충액에 10 μM 농도의 T6RB를 95℃에서 5분간 변성시킨 후 25℃까지 천천히 냉각시켜 2차 구조를 형성시켰다. CD 스펙트럼은 50 nm/min의 스캔 속도와 1 nm의 대역폭으로 210~300 nm의 스펙트럼 범위에서 J-810 분광편광계(Jasco, Tokyo, Japan)에 기록되었다. 모든 측정은 3회 수행하여 평균을 냈으며, 해당 완충액에 대한 배경 CD 스펙트럼을 측정하여 실험 스펙트럼에서 뺐다. 스펙트럼 분석은 Spectra Manager(Jasco)를 사용하여 수행되었으며, 형태 지수 r(하기 식 4)을 사용하여 DNA 이차구조를 분석하였다.T6RB at a concentration of 10 μM was denatured at 95°C for 5 minutes in a buffer containing 10mM Tris/HCl (pH 7.4) and/or 100mM KCl, and then slowly cooled to 25°C to form secondary structures. CD spectra were recorded on a J-810 spectropolarimeter (Jasco, Tokyo, Japan) in the spectral range of 210–300 nm with a scan speed of 50 nm/min and a bandwidth of 1 nm. All measurements were performed in triplicate and averaged, and the background CD spectrum for the corresponding buffer was measured and subtracted from the experimental spectrum. Spectral analysis was performed using Spectra Manager (Jasco), and DNA secondary structure was analyzed using the conformation index r (Equation 4 below).

<식 4><Equation 4>

Figure PCTKR2023017433-appb-img-000001
Figure PCTKR2023017433-appb-img-000001

여기서 CD265와 CD290은 각각 265 nm와 290 nm에서의 CD 몰 타원율[θ]이다. r ≥ 0.5, 0 ≤ r < 0.5, r < 0은 각각 주로 병렬, 하이브리드 및 역병렬 토폴로지에 해당한다.Here, CD 265 and CD 290 are the CD molar ellipticities [θ] at 265 nm and 290 nm, respectively. r ≥ 0.5, 0 ≤ r < 0.5, and r < 0 mainly correspond to parallel, hybrid, and anti-parallel topologies, respectively.

도 20의 (B)는 칼륨 이온(K+)의 부재/존재 시 T6RB의 원형 이색성(CD) 스펙트럼을 나타낸다. 칼륨 이온(K+) 존재시, T6RB의 원형 이색성 (CD) 스펙트럼을 살펴보면, 210 nm와 265 nm 영역에서 양성 피크를 나타내고, 상기 식 2를 기반으로 계산한 결과 r > 0.5이상인 것으로 나타나, T6RB는 G-quadruplex 이차 구조를 이루면 그 중 병렬구조를 형성하는 것을 확인하였다. Figure 20 (B) shows the circular dichroism (CD) spectrum of T6RB in the absence/presence of potassium ions (K+). In the presence of potassium ions (K+), the circular dichroism (CD) spectrum of T6RB shows positive peaks in the 210 nm and 265 nm regions, and as a result of calculation based on Equation 2 above, r > 0.5, T6RB It was confirmed that when a G-quadruplex secondary structure is formed, a parallel structure is formed.

N-메틸 메소포르피린 IX(NMM)의 형광 측정Fluorescence measurement of N-methyl mesoporphyrin IX (NMM)

100 mM KCl이 첨가되거나 첨가되지 않은 10 mM Tris/HCl(pH 7.4)을 함유한 완충액에서 500 nM 농도의 T6RB를 95℃에서 5분 동안 변성시킨 후 천천히 37℃로 냉각하고 0.5시간 동안 추가로 배양하여, 2차 구조가 형성될 수 있도록 하였다. 그런 다음, NMM(Cayman Chemical, Ann Arbor, MI, USA)을 5 μM 농도로 혼합물에 첨가하고 NMM이 T6RB 구조에 삽입될 수 있도록 추가로 0.5시간 동안 배양했다. 형광 강도(FI)는 Spectramax iD5 다중 모드 마이크로플레이트 판독기(Molecular Devices)에서 399 nm의 여기 파장과 610 nm의 방출 파장에서 측정되었다.T6RB at a concentration of 500 nM was denatured at 95°C for 5 min in a buffer containing 10 mM Tris/HCl (pH 7.4) with or without 100 mM KCl, then slowly cooled to 37°C and incubated for an additional 0.5 h. This allowed the secondary structure to be formed. Then, NMM (Cayman Chemical, Ann Arbor, MI, USA) was added to the mixture at a concentration of 5 μM and incubated for an additional 0.5 h to allow NMM to be inserted into the T6RB structure. Fluorescence intensity (FI) was measured at an excitation wavelength of 399 nm and an emission wavelength of 610 nm on a Spectramax iD5 multimode microplate reader (Molecular Devices).

도 20의 (C)에서 알 수 있듯이, 원형 이색성 스펙트럼 결과와 동일하게, 칼륨 이온 (K+)의 존재 시 T6RB에 결합된 NMM의 형광 강도가 더 강하게 나타나는 것을 확인할 수 있었고, 이는 곧 T6RB가 G-quadruplex 이차 구조를 형성함을 재확인하였다.As can be seen in (C) of Figure 20, consistent with the circular dichroism spectrum results, it was confirmed that the fluorescence intensity of NMM bound to T6RB appears stronger in the presence of potassium ions (K + ), which means that T6RB It was confirmed again that a G-quadruplex secondary structure was formed.

T6RB sEV에 대한 압타머 표적 바이오마커의 위상(topology) 분석 (sEV 표면 단백질의 sEV 용해 및 효소적 단백질 분해)Topology analysis of aptamer target biomarkers for T6RB sEV (sEV lysis and enzymatic proteolysis of sEV surface proteins)

sEV 용해를 위해 최종 농도가 1X인 Halt 프로테아제 억제제 칵테일(Thermo Fisher Scientific)이 보충된 RIPA 용해 완충액을 사용하여 SW620 sEV의 용해물을 준비했다. 간단히 말하면, sEV를 프로테아제 억제제가 보충된 용해 완충액과 혼합하고 얼음 위에서 30분 동안 배양한 다음, 40kHz에서 30초 온/오프의 5주기로 수조 초음파 처리기(SH-2140D, SAE HAN ULTRASONIC, Seoul, Republic of Korea)에서 초음파 처리하였다. 프로테이나제 K(proK) 처리를 위해 SW620 sEV를 20분마다 부드럽게 볼텍싱하면서 37℃에서 1시간 동안 proK(20 μg/mL, Enzynomics)로 처리했다. proK 활성은 실온에서 페닐메틸설포닐 플루오라이드(PMSF, 5 mM)로 10분간 처리하여 억제되었다. 트립신 처리를 위해 트립신-에틸렌디아민 테트라아세트산(트립신-EDTA, 0.25%)을 SW620 sEV와 함께 37℃에서 30분 동안 배양하고 1x PBS에 동량의 10% FBS를 첨가하여 활성을 중단했다. 각 처리의 최종 조성물은 사용 전까지 -80℃에 보관하였다.For sEV lysis, lysates of SW620 sEVs were prepared using RIPA lysis buffer supplemented with Halt protease inhibitor cocktail (Thermo Fisher Scientific) at a final concentration of 1X. Briefly, sEVs were mixed with lysis buffer supplemented with protease inhibitors, incubated on ice for 30 min, and then incubated in a water bath sonicator (SH-2140D, SAE HAN ULTRASONIC, Seoul, Republic of) with 5 cycles of 30 s on/off at 40 kHz. Korea) was treated with ultrasonic waves. For proteinase K (proK) treatment, SW620 sEVs were treated with proK (20 μg/mL, Enzynomics) for 1 h at 37°C with gentle vortexing every 20 min. proK activity was inhibited by treatment with phenylmethylsulfonyl fluoride (PMSF, 5 mM) for 10 min at room temperature. For trypsinization, trypsin-ethylenediamine tetraacetic acid (trypsin-EDTA, 0.25%) was incubated with SW620 sEVs at 37°C for 30 min, and activity was stopped by adding an equal amount of 10% FBS in 1x PBS. The final composition of each treatment was stored at -80°C until use.

그 결과, 도 20의 (D)는 바이오마커 토폴로지 분석결과를 나타낸다 (순서대로, 미처리/proteinase K(proK) 처리/트립신 처리/RIPA 처리 sEVs). 즉, 도 20의 (D)는 T6RB가 proK 및 트립신 처리된 sEVs에 대해 무시할 수 있는 결합 친화도를 나타냄을 나타낸다. 한편, T6RB는 아무 처리하지 않은 (미처리) 온전한 sEV와 비교하여 RIPA 용해 sEV에 대해 필적할만한 결합 친화도를 나타냈다(도 20의 (D)). 이러한 결과로, T6RB가 표적으로 하는 바이오마커가 단백질로 식별되고 sEV의 외막에 위상적으로 위치함을 확인하였다. As a result, Figure 20 (D) shows the results of the biomarker topology analysis (in that order, untreated/proteinase K (proK) treated/trypsin treated/RIPA treated sEVs). That is, Figure 20(D) shows that T6RB exhibits negligible binding affinity to proK and trypsin-treated sEVs. Meanwhile, T6RB showed comparable binding affinity to RIPA-lysed sEVs compared to intact sEVs without any treatment (Figure 20(D)). These results confirmed that the biomarker targeted by T6RB was identified as a protein and was topologically located in the outer membrane of sEV.

티라마이드 신호 증폭(TSA) 기능을 탑재한 ELONAELONA with Tyramide Signal Amplification (TSA) function

비오틴-티라미드(b-티라미드)/SA-PP 시스템을 갖춘 TSA는 ELONA의 추가 단계로 사용되어 분석의 감도를 향상시켰다. 면역 플레이트에 코팅된 HT29 sEV(2×107 입자/mL)를 이용하여 TSA에 최적화된 실험조건을 탐색하였고, T6RB, b-tyramide, H2O2의 농도와 TSA의 반응시간을 최적화하였다. SA-HRP(PBST에 1:1000 희석)의 첫 번째 배양이 ELONA에서 수행되면 최적화된 조건[0.1 M 붕산염(pH 8.5) 중 15μg/mL b-티라미드(APExBIO Technology LLC, Texas, USA) 및 0.001% H2O2(Sigma-Aldrich)]을 37℃에서 25분간 수행한 후 세척 단계를 거쳤다. 마지막으로 SA-HRP(PBST에 1:1000 희석)의 두 번째 배양을 37℃에서 1시간 동안 처리한 후 세척 단계를 거쳤다. TMB 반응 및 흡광도 값 정규화는 위에서 설명한 ELONA와 동일한 방식으로 처리되었으며 모든 실험은 3회 수행되었다.TSA with biotin-tyramide (b-tyramide)/SA-PP system was used as an additional step in ELONA to improve the sensitivity of the assay. Experimental conditions optimized for TSA were explored using HT29 sEV (2×10 7 particles/mL) coated on an immune plate, and the concentrations of T6RB, b-tyramide, H 2 O 2 and TSA reaction time were optimized. The first incubation of SA-HRP (1:1000 dilution in PBST) was performed in ELONA under optimized conditions [15 μg/mL b-tyramide (APExBIO Technology LLC, Texas, USA) in 0.1 M borate (pH 8.5) and 0.001 μg/mL b-tyramide (APExBIO Technology LLC, Texas, USA). % H 2 O 2 (Sigma-Aldrich)] at 37°C for 25 minutes, followed by a washing step. Finally, a second culture of SA-HRP (1:1000 dilution in PBST) was treated at 37°C for 1 hour followed by a washing step. TMB reactions and absorbance value normalization were handled in the same way as ELONA described above and all experiments were performed in triplicate.

TSA가 탑재된 ELONA 방식의 최적 조건을 찾기 위해 TSA에서 주요 변수들 (T6RB/b-티라미드/H2O2 농도 및 반응시간)을 최적화를 진행하였고 그 결과 각각 160 nM T6RB, 15 μg/mL b-티라미드, 0.001% H2O2 및 25분의 반응시간의 결과가 도출되었다 (도 21의 (A-D)). In order to find the optimal conditions for the ELONA method equipped with TSA, key variables (T6RB/b-tyramide/H 2 O 2 concentration and reaction time) were optimized in TSA, and the results were 160 nM T6RB and 15 μg/mL, respectively. Results were obtained for b-tyramide, 0.001% H 2 O 2 and a reaction time of 25 minutes ((AD) of FIG. 21).

도 22에서 TSA 최적화 조건으로 일반 ELONA와 TSA 탑재 ELONA를 비교 분석한 결과, 낮은 농도의 sEV 상에서 일반 ELONA는 양성 표적 (SW620, LS 174T, HT29 sEVs)에 대해 검출할 수 없는 ΔA이 도출되었다. 대조적으로, TSA를 탑재한 ELONA의 경우, sEV가 없는 control 군에서는 ΔA을 유의미한 값을 나타내지 않았으며, 양성 표적 (SW620, LS 174T, HT29 sEVs)에 대해서는 훨씬 큰 ΔA을 나타냈다. 이는 일반 ELONA에 비해 TSA에 의한 신호 증폭으로 인해 최종 ΔA 값을 크게 상승한 것으로 해석할 수 있고, 최종적으로 민감도 증가효과를 가져올 것으로 예상하였다. In Figure 22, as a result of comparative analysis of regular ELONA and TSA-equipped ELONA under TSA optimization conditions, the ΔA of regular ELONA was undetectable for positive targets (SW620, LS 174T, HT29 sEVs) on low concentrations of sEVs. In contrast, in the case of ELONA loaded with TSA, the sEV-free control group did not show a significant ΔA value and showed a much larger ΔA for positive targets (SW620, LS 174T, HT29 sEVs). This can be interpreted as a significant increase in the final ΔA value due to signal amplification by TSA compared to regular ELONA, and is expected to ultimately lead to increased sensitivity.

실시예 2-6.Example 2-6.

최고의 압타머 평가(T6RB)Best aptamer evaluation (T6RB)

최종적으로 SW620, LS 174T 및 HT29 sEVs에 대한 검출 가능성을 고결합성/특이성을 지닌 T6RB와 TSA-탑재 ELONA 방법을 활용하여 분석하였다. 도 23의 (A)에 표시된 결과는 흡광도 신호(ΔA)가 SW620 sEV의 농도(2.0 Х 104 - 1.0 Х 106 입자/μL)에 비례하고 선형 회귀함수가 Y = 0.02782X + 0.03430 (R2 = 0.9871)가 다음과 같이 획득되었음을 보여준다. 또한, 10의 (B)에서 흡광도 신호(ΔA)가 LS 174T sEV의 농도에 비례하고 선형 회귀함수가 Y = 0.01075X + 0.03711 (R2 = 0.9850)가 다음과 같이 획득되었음을 보여주며, 도 23의 (C)에서는 흡광도 신호(ΔA)가 HT29 sEV의 농도에 비례하고 선형 회귀함수가 Y = 0.03014X + 0.2759 (R2 = 0.9861)가 획득되었음을 나타낸다. 또한, 각 sEV에 대한 검출한계 (LOD, limit of detection)을 계산한 결과, 3.6 x 102 입자/μL (SW620), 3.5 x 103 입자/μL (LS 174T), 8.4 x 102 입자/μL (HT29)로 도출되었다. 이는 혈청 또는 혈장 내 sEV의 농도가 105 입자/μL 범위로 존재하는 것을 고려하면, CRC 유래 sEV에 대한 T6RB의 진단 능력을 강력하게 검증하기 정밀함을 시사한다. Finally, the detection potential for SW620, LS 174T, and HT29 sEVs was analyzed using the T6RB and TSA-equipped ELONA methods with high binding/specificity. The results shown in (A) of Figure 23 show that the absorbance signal (ΔA) is proportional to the concentration of SW620 sEV (2.0 Х 10 4 - 1.0 Х 10 6 particles/μL) and the linear regression function is Y = 0.02782X + 0.03430 (R 2 = 0.9871) is obtained as follows. In addition, (B) in Fig. 10 shows that the absorbance signal (ΔA) is proportional to the concentration of LS 174T sEV and the linear regression function Y = 0.01075X + 0.03711 (R 2 = 0.9850) was obtained as follows, in Figure 23 (C) shows that the absorbance signal (ΔA) is proportional to the concentration of HT29 sEV and the linear regression function Y = 0.03014X + 0.2759 (R 2 = 0.9861) was obtained. In addition, the limit of detection (LOD) for each sEV was calculated as follows: 3.6 x 10 2 particles/μL (SW620), 3.5 x 10 3 particles/μL (LS 174T), 8.4 x 10 2 particles/μL It was derived as (HT29). This suggests that it is accurate to strongly verify the diagnostic ability of T6RB for CRC-derived sEV, considering that the concentration of sEV in serum or plasma is in the range of 10 5 particles/μL.

결론적으로, 설계한 TEV-SELEX 방법을 사용하여 다양한 CRC 세포 유래 sEV를 채택하여 이질성이 높은 CRC를 보다 넑게 표적할 수 있도록 설계하였고, 상위 10 개의 추정 압타머를 선별하고 분석을 통해 가장 높은 결합 친화도(K d = 3.848(SW620), 5.904(LS 174T), 5.234(HT29) nM)와 특이성을 나타내는 최고의 압타머 T6RB을 선택하였다. 압타머의 진단적 타당성은 3.6 x 102 입자/μL (SW620), 3.5 x 103 입자/μL (LS 174T), 8.4 x 102 입자/μL (HT29)의 검출한계값을 지니고 CRC 세포 유래 sEVs를 검출하고 건강한 혈청 및 정상 세포 sEVs인 비표적 sEVs를 구별함으로써 입증하였다. 또한, T6RB는 sEV의 외막에 존재하는 단백질을 표적하는 것을 확인하였고, 추후 연구를 통해 바이오마커 동정을 하여 새로운 바이오마커 구축에도 이용할 수 있을 것으로 예상된다.In conclusion, using the designed TEV-SELEX method, various CRC cell-derived sEVs were adopted to enable broader targeting of CRC with high heterogeneity, and the top 10 putative aptamers were selected and analyzed to determine the highest binding affinity. The best aptamer T6RB was selected, showing specificity ( K d = 3.848 (SW620), 5.904 (LS 174T), 5.234 (HT29) nM). The diagnostic validity of the aptamer is that it has detection limits of 3.6 was demonstrated by detecting and distinguishing between non-target sEVs, which are healthy serum and normal cell sEVs. In addition, T6RB was confirmed to target proteins present in the outer membrane of sEV, and it is expected that it can be used to construct new biomarkers by identifying biomarkers through future research.

Claims (28)

서열번호 4 내지 17의 염기서열로 이루어진 군에서 선택되는 어느 하나 이상의 대장암 진단 또는 치료용 DNA 압타머. At least one DNA aptamer for diagnosis or treatment of colon cancer selected from the group consisting of base sequences of SEQ ID NOs: 4 to 17. 제1항에 있어서, According to paragraph 1, 상기 압타머는 대장암 세포 유래의 sEV에 특이적으로 결합하는, DNA 압타머. The aptamer is a DNA aptamer that specifically binds to sEV derived from colon cancer cells. 제1항에 있어서, According to paragraph 1, 상기 압타머는 정방향 프라이머 및 역방향 프라이머 서열로 이루어진 군에서 선택되는 하나 이상을 추가로 포함하는, DNA 압타머.The aptamer is a DNA aptamer, further comprising at least one selected from the group consisting of a forward primer and a reverse primer sequence. 제3항에 있어서, According to clause 3, 상기 정방향 프라이머의 서열은 서열번호 2의 염기서열로 이루어진, DNA 압타머.The sequence of the forward primer is a DNA aptamer consisting of the base sequence of SEQ ID NO: 2. 제3항에 있어서, According to clause 3, 상기 역방향 프라이머의 서열은 서열번호 3의 염기서열로 이루어진, DNA 압타머.The sequence of the reverse primer is a DNA aptamer consisting of the base sequence of SEQ ID NO: 3. 제1항에 있어서, According to paragraph 1, 상기 DNA 압타머는 서열번호 13의 염기서열로 이루어진, DNA 압타머.The DNA aptamer is a DNA aptamer consisting of the base sequence of SEQ ID NO: 13. 제6항에 있어서, According to clause 6, 상기 서열번호 13의 염기서열로 이루어진 DNA 압타머는, 정방향 프라이머 염기서열 및 역방향 프라이머 염기서열 중 어느 하나 이상이 추가적으로 연결된 DNA 압타머.The DNA aptamer consisting of the base sequence of SEQ ID NO: 13 is a DNA aptamer in which at least one of the forward primer base sequence and the reverse primer base sequence is additionally linked. 제7항에 있어서, In clause 7, 상기 DNA 압타머는 서열벌호 15의 염기서열로 이루어진, DNA 압타머. The DNA aptamer is a DNA aptamer consisting of the base sequence of sequence number 15. 제8항에 있어서, According to clause 8, 상기 DNA 압타머는 결합 해리상수로 3 내지 4 nM를 갖는, DNA 압타머.The DNA aptamer is a DNA aptamer having a binding dissociation constant of 3 to 4 nM. 서열번호 18 내지 35의 염기서열로 이루어진 군에서 선택되는 어느 하나 이상의 대장암 진단용 DNA 압타머. At least one DNA aptamer for colon cancer diagnosis selected from the group consisting of base sequences of SEQ ID NOs: 18 to 35. 제10항에 있어서, According to clause 10, 상기 압타머는 다양한 대장암 세포 유래 sEVs에 특이적으로 결합하는, DNA 압타머.The aptamer is a DNA aptamer that specifically binds to sEVs derived from various colon cancer cells. 제10항에 있어서, According to clause 10, 상기 압타머는 상기 압타머는 정방향 프라이머 및 역방향 프라이머 서열로 이루어진 군에서 선택되는 하나 이상을 추가로 포함하는, DNA 압타머.The aptamer is a DNA aptamer, wherein the aptamer further includes one or more selected from the group consisting of a forward primer and a reverse primer sequence. 제12항에 있어서, According to clause 12, 상기 정방향 프라이머의 서열은 서열번호 2의 염기서열로 이루어진 것인, DNA 압타머.The sequence of the forward primer is a DNA aptamer consisting of the base sequence of SEQ ID NO: 2. 제12항에 있어서, According to clause 12, 상기 역방향 프라이머의 서열은 서열번호 3의 염기서열로 이루어진, DNA 압타머.The sequence of the reverse primer is a DNA aptamer consisting of the base sequence of SEQ ID NO: 3. 제10항에 있어서, According to clause 10, 상기 DNA 압타머는 서열번호 32의 염기서열로 이루어진 것인, DNA 압타머.The DNA aptamer is a DNA aptamer consisting of the base sequence of SEQ ID NO: 32. 제10항에 있어서, According to clause 10, 상기 DNA 압타머는 결합 해리상수로 3 내지 6 nM를 갖는, DNA 압타머.The DNA aptamer is a DNA aptamer having a binding dissociation constant of 3 to 6 nM. 제1항 내지 제9항 중 어느 한 항의 압타머를 포함하는 대장암 진단 또는 치료용 조성물. A composition for diagnosing or treating colon cancer comprising the aptamer of any one of claims 1 to 9. 제10항의 조성물을 포함하는, 대장암 진단 또는 치료용 키트. A kit for diagnosing or treating colon cancer, comprising the composition of claim 10. 제11항에 있어서, According to clause 11, 상기 키트는 진단용 센서, RT-PCR 키트, 경쟁적 RT-PCR 키트, 실시간 RT-PCR 키트, DNA 칩 키트 및 단백질 칩으로 이루어진 군에서 선택되는 어느 하나인, 키트.The kit is any one selected from the group consisting of a diagnostic sensor, RT-PCR kit, competitive RT-PCR kit, real-time RT-PCR kit, DNA chip kit, and protein chip. 제10항 내지 제16항 중 어느 한 항의 압타머를 포함하는 대장암 진단용 조성물. A composition for diagnosing colon cancer comprising the aptamer of any one of claims 10 to 16. 제20항의 조성물을 포함하는, 대장암 진단용 키트. A kit for diagnosing colon cancer, comprising the composition of claim 20. 제20항에 있어서, According to clause 20, 상기 키트는 진단용 센서, RT-PCR 키트, 경쟁적 RT-PCR 키트, 실시간 RT-PCR 키트, DNA 칩 키트 및 단백질 칩으로 이루어진 군에서 선택되는 어느 하나인, 키트.The kit is any one selected from the group consisting of a diagnostic sensor, RT-PCR kit, competitive RT-PCR kit, real-time RT-PCR kit, DNA chip kit, and protein chip. 제1항 내지 제16항 중 어느 한 항의 DNA 압타머 제조 방법. A method for producing a DNA aptamer according to any one of claims 1 to 16. 제23항에 있어서, 상기 방법은The method of claim 23, wherein the method a) 랜덤 라이브러리를 생성하는 단계;a) generating a random library; b) E-SELEX 또는 TEV-SELEX를 수행하는 단계; b) performing E-SELEX or TEV-SELEX; c) NGS 기법을 통해 DNA 압타머 서열 분석하는 단계; 및c) analyzing the DNA aptamer sequence through NGS technology; and d) DNA 압타머 특성화하는 단계를 포함하는, 방법. d) a method comprising characterizing the DNA aptamer. 제23항에 있어서,According to clause 23, 상기 압타머는 대장암 세포 유래 sEV에 특이적으로 결합하는, 방법. The method wherein the aptamer specifically binds to sEV derived from colon cancer cells. 제1항 내지 제16항 중 어느 한 항의 DNA 압타머를 이용한 대장암 진단 또는 치료를 위한 정보를 제공하는 방법. A method of providing information for diagnosis or treatment of colon cancer using the DNA aptamer of any one of claims 1 to 16. 제1항 내지 제16항 중 어느 한 항의 압타머를 이용한 대장암 sEVs 특이적 표면 바이오마커의 검출방법.A method for detecting colorectal cancer sEVs-specific surface biomarkers using the aptamer of any one of claims 1 to 16. 제1항 내지 제16항 중 어느 한 항의 압타머를 포함하는 대장암 특이적 약물전달 조성물. A colon cancer-specific drug delivery composition comprising the aptamer of any one of claims 1 to 16.
PCT/KR2023/017433 2022-11-02 2023-11-02 Dna aptamer for diagnosis or treatment of colorectal cancer Ceased WO2024096642A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR1020220144790A KR20240066514A (en) 2022-11-02 2022-11-02 DNA Aptamer for colorectal cancer diagnostics and therapeutics
KR10-2022-0144790 2022-11-02
KR10-2023-0150082 2023-11-02
KR1020230150082A KR20250064751A (en) 2023-11-02 2023-11-02 Dna aptamer for colorectal cancer diagnostics

Publications (1)

Publication Number Publication Date
WO2024096642A1 true WO2024096642A1 (en) 2024-05-10

Family

ID=90930935

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/017433 Ceased WO2024096642A1 (en) 2022-11-02 2023-11-02 Dna aptamer for diagnosis or treatment of colorectal cancer

Country Status (1)

Country Link
WO (1) WO2024096642A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN118957063A (en) * 2024-07-22 2024-11-15 宁夏医科大学总医院 A colorectal cancer LncRNA diagnostic marker, diagnostic kit and application

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109682973A (en) * 2019-01-02 2019-04-26 中国科学院化学研究所 Lesion detection approach and kit based on aptamer
KR20190134683A (en) * 2017-03-30 2019-12-04 어그매니티 나노 엘티디 Methods and Compositions for Selection of Functional Oligonucleotides
CN112501173A (en) * 2020-11-09 2021-03-16 苏州吉玛基因股份有限公司 GPC1DNA aptamer and application thereof
KR20210112875A (en) * 2020-03-06 2021-09-15 연세대학교 원주산학협력단 Exosomes with drug trapped and aptamer attached to the surface and methods of thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190134683A (en) * 2017-03-30 2019-12-04 어그매니티 나노 엘티디 Methods and Compositions for Selection of Functional Oligonucleotides
CN109682973A (en) * 2019-01-02 2019-04-26 中国科学院化学研究所 Lesion detection approach and kit based on aptamer
KR20210112875A (en) * 2020-03-06 2021-09-15 연세대학교 원주산학협력단 Exosomes with drug trapped and aptamer attached to the surface and methods of thereof
CN112501173A (en) * 2020-11-09 2021-03-16 苏州吉玛基因股份有限公司 GPC1DNA aptamer and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHINNAPPAN, R. et al. An integrated lab-on-a-chip platform for pre-concentration and detection of colorectal cancer exosomes using anti-CD63 aptamer as a recognition element. Biosensors and Bioelectronics. published online : 29 October 2022, vol. 220, thesis no. : 114856 (pp. 1-8). *
HUI WANG: "DNase I enzyme-aided fluorescence signal amplification based on graphene oxide-DNA aptamer interactions for colorectal cancer exosome detection", TALANTA, ELSEVIER, AMSTERDAM, NL, vol. 184, 1 July 2018 (2018-07-01), NL , pages 219 - 226, XP093168967, ISSN: 0039-9140, DOI: 10.1016/j.talanta.2018.02.083 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN118957063A (en) * 2024-07-22 2024-11-15 宁夏医科大学总医院 A colorectal cancer LncRNA diagnostic marker, diagnostic kit and application

Similar Documents

Publication Publication Date Title
WO2010140834A9 (en) Nucleic acid aptamer specifically binding to pancreatic cancer cells or tissues and use thereof
WO2016018087A1 (en) Novel biomarker for predicting sensitivity to egfr-targeting agent, and use thereof
WO2021086107A1 (en) Method for determining reactivity to parp inhibitor
WO2024096642A1 (en) Dna aptamer for diagnosis or treatment of colorectal cancer
WO2015023146A1 (en) Method for detecting precancerous lesions
WO2016036172A1 (en) Biomarker for predicting sensitivity to protein kinase inhibitor and use thereof
WO2015002511A1 (en) Improved nanoparticle type oligonucleotide structure having high efficiency and method for preparing same
WO2010090452A2 (en) Small interference rna complex with increased intracellular transmission capacity
Cha et al. Development of a Novel DNA Aptamer Targeting Colorectal Cancer Cell‐Derived Small Extracellular Vesicles as a Potential Diagnostic and Therapeutic Agent
WO2021006570A1 (en) Aptamer selection method and immunity analysis method using aptamer
WO2021225423A1 (en) Novel nucleic acid ligand, and method for identifyng same
WO2024158242A1 (en) Compound for inhibiting kras g12d mutation, and composition for preventing or treating cancer disease, comprising same as active ingredient
WO2018110980A1 (en) Pharmaceutical composition for preventing or treating hepatitis b
WO2015130115A1 (en) Novel antibody specific for tspan8 and uses thereof
WO2015108328A1 (en) Novel ntrk1 fusion gene as colorectal cancer marker and use thereof
WO2017082655A1 (en) Methods for determining resistance to anticancer therapy and composition used therefor
WO2024177416A1 (en) Pharmaceutical composition for treating egfr targeted therapy-resistant cancer
WO2015002512A1 (en) Dengue virus-specific sirna, double helix oligo-rna structure comprising sirna, and composition for suppressing proliferation of dengue virus comprising rna structure
WO2011118994A9 (en) Nlk as a marker for diagnosis of liver cancer and as a therapeutic agent thereof
WO2023167544A1 (en) Biomarker comprising fam167a for diagnosis of resistance against bcr-abl-independent tyrosine kinase inhibitor, and composition targeting fam167a for preventing or treating chronic myeloid leukemia
WO2022065968A1 (en) Pharmaceutical composition comprising evodiamine as active ingredient for prevention or treatment of non-small cell lung cancer
WO2017188707A1 (en) Dicer substrate rna nanostructures with enhanced gene silencing effect and preparation method thereof
WO2022169342A2 (en) Pharmaceutical composition for inhibiting cancer metastasis
WO2018088826A2 (en) Composition for diagnosing colorectal cancer metastasis or predicting prognosis and use thereof
WO2025249887A1 (en) Composition for increasing stability or expression of exogenous rna in cells, comprising inhibitor of trim25 expression or activity

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: 23886336

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE