WO2023246935A1 - Target-specific nucleic acid molecule having quinoline modification - Google Patents

Target-specific nucleic acid molecule having quinoline modification Download PDF

Info

Publication number
WO2023246935A1
WO2023246935A1 PCT/CN2023/102119 CN2023102119W WO2023246935A1 WO 2023246935 A1 WO2023246935 A1 WO 2023246935A1 CN 2023102119 W CN2023102119 W CN 2023102119W WO 2023246935 A1 WO2023246935 A1 WO 2023246935A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
modified
acid molecule
target
specific nucleic
Prior art date
Application number
PCT/CN2023/102119
Other languages
French (fr)
Chinese (zh)
Inventor
张戈
何伊欣
阿木古布
Original Assignee
安沛治疗有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 安沛治疗有限公司 filed Critical 安沛治疗有限公司
Publication of WO2023246935A1 publication Critical patent/WO2023246935A1/en

Links

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
    • A61K31/7115Nucleic acids or oligonucleotides having modified bases, i.e. other than adenine, guanine, cytosine, uracil or thymine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • 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
    • C12Q1/6811Selection methods for production or design of target specific oligonucleotides or binding molecules

Definitions

  • the invention relates to the field of biomedicine. Specifically, it relates to a target-specific nucleic acid molecule comprising a quinoline modification that can increase the binding affinity of the nucleic acid molecule to a target.
  • Nucleic acid aptamers are single-stranded DNA or RNA that can form a complementary structure with the target. They have specificity and affinity comparable to monoclonal antibody drugs. They are also non-immunogenic, easy to produce, low cost, and thermally stable. Good to wait for the advantage.
  • Osteosclerostin is a negative regulator of bone formation and is an ideal target for the development of drugs that promote bone formation and treat osteoporosis and other related diseases.
  • osteosclerostin also plays an important regulatory role in the occurrence and development of diseases such as osteogenesis imperfecta, hypophosphatemic rickets, and triple-negative breast cancer, and is therefore an important target for the treatment of these diseases.
  • Sclerostin monoclonal antibody Romosozumab can promote bone formation.
  • Drug regulatory agencies in the United States, Europe and Asia have approved osteosclerostin monoclonal antibody for marketing. The indication is postmenopausal patients with osteoporosis who are at high risk of fracture.
  • Sclerostin protein has three structural regions, namely loop1, loop2 and loop3. Among them, osteosclerostin monoclonal antibody mainly acts on loop2. Studies have found that loop2 is involved in both bone formation inhibition and cardiovascular protection, while osteosclerostin loop3 is only involved in bone formation inhibition but not cardiovascular protection. Therefore, the nucleic acid aptamer-APC001, which specifically targets osteosclerostin loop3, was screened through SELEX technology, and the aptamer was verified to have the ability to promote bone anabolism in the treatment of osteoporosis models and osteogenesis imperfecta models. Therapeutic potential without increasing cardiovascular disease risk. This therapeutic aptamer received Osteogenesis Imperfecta orphan drug certification from the U.S. FDA in 2019 (FDA, DRU-2019-6966).
  • Embodiment 1 A modified target-specific nucleic acid molecule comprising one or more quinoline-modified nucleotides, wherein the modified target-specific nucleic acid molecule is compared to a corresponding control nucleic acid molecule. Target binding affinity is increased.
  • Embodiment 2 The modified target-specific nucleic acid molecule of embodiment 1, wherein said nucleic acid molecule is selected from the group consisting of an aptamer (aptamer), an antisense nucleic acid (antisense RNA), dsRNA (eg, siRNA, shRNA).
  • aptamer an aptamer
  • antisense RNA an antisense nucleic acid
  • dsRNA eg, siRNA, shRNA
  • Embodiment 3 The modified target-specific nucleic acid molecule of embodiment 1 or 2, wherein the modified target-specific nucleic acid molecule has a binding affinity to a target that is increased by at least about 10% compared to a corresponding control nucleic acid molecule. At least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100% or more; or all
  • the binding affinity of the modified target-specific nucleic acid molecule to the target is at least about 1.5 times, at least about 2 times, at least about 3 times, at least about 4 times, at least about 5 times, at least about the affinity of the unmodified corresponding nucleic acid molecule.
  • Embodiment 4 The modified target-specific nucleic acid molecule of any one of embodiments 1-3, wherein the modified target-specific nucleic acid molecule has an affinity for the target of less than 100 nM, preferably less than 50 nM, preferably less than 40 nM, preferably less than 30 nM, preferably less than 20 nM, preferably less than 10 nM, preferably less than 5 nM or less KD (dissociation constant).
  • Embodiment 5 The modified target-specific nucleic acid molecule of any one of embodiments 1-4, wherein the quinoline modification is quinolin-2-yl substitution, quinolin-3-yl substitution, quinolin-4 - base substitution, quinolin-5-yl substitution, quinolin-6-yl substitution, quinolin-7-yl substitution or quinolin-8-yl substitution, preferably, the quinoline modification is quinolin-5- base substitution.
  • Embodiment 6 The modified target-specific nucleic acid molecule of any one of embodiments 1-5, wherein the quinoline-modified nucleotide comprises a substituent group R selected from ag:
  • Embodiment 7 The modified target-specific nucleic acid molecule of any one of embodiments 1-6, wherein the quinoline-modified nucleotide comprises the base moiety of Formula I:
  • Embodiment 8 The modified target-specific nucleic acid molecule of any one of embodiments 1-7, wherein the quinoline-modified nucleotide comprises the following structure of Formula II:
  • Embodiment 9 The modified target-specific nucleic acid molecule of any one of embodiments 1-8, wherein one or more naturally occurring nucleotides in the target-specific nucleic acid molecule are modified with the quinoline Nucleotide substitutions.
  • Embodiment 10 The modified target-specific nucleic acid molecule of any one of embodiments 1-9, wherein the target-specific nucleic acid molecule may further comprise one or more modifications that extend its half-life in vivo.
  • Embodiment 11 The modified target-specific nucleic acid molecule of any one of embodiments 1-10, wherein the modified target-specific nucleic acid molecule is a modified osteosclerostin-specific aptamer.
  • Embodiment 12 The modified target-specific nucleic acid molecule of embodiment 11, wherein the modified osteosclerostin-specific aptamer is derived from the osteosclerostin-specific aptamer of SEQ ID NO: 2.
  • Embodiment 13 The modified target-specific nucleic acid molecule of embodiment 12, wherein the modified osteosclerostin-specific aptamer is at one or more positions selected from the group consisting of corresponding to SEQ ID NO:2
  • the nucleotides are replaced by the quinoline modified nucleotides: position 1, position 2, position 3, position 4, position 5, position 6, position 8, position 11, position 13 , 15th, 29th, 30th, 31st, 32nd, 33rd, 34th, 35th, 36th, 37th, 38th, 39th and 39th 40 people.
  • Embodiment 14 The modified target-specific nucleic acid molecule of embodiment 12 or 13, wherein the modified osteosclerostin-specific nucleic acid molecule is at position 6, position 8, position corresponding to SEQ ID NO: 2 One or more nucleotides at position 11, 13, 32 and/or 34 are substituted with the quinoline modified nucleotide.
  • Embodiment 15 The modified target-specific nucleic acid molecule of any one of embodiments 12-14, wherein the modified osteosclerostin-specific nucleic acid molecule comprises a nucleotide selected from the group consisting of SEQ ID NOs: 3-23 Sequence, where x is the quinoline modified nucleotide.
  • Embodiment 16 The modified target-specific nucleic acid molecule of any one of embodiments 12-15, wherein the modified osteosclerostin-specific nucleic acid molecule comprises a nucleic acid molecule selected from the group consisting of SEQ ID NO: 8, 10, 11, 16 and the nucleotide sequence of 18, wherein x is the quinoline-modified nucleotide.
  • Embodiment 17 The modified target-specific nucleic acid molecule of any one of embodiments 12-16, wherein the quinoline-modified nucleotide comprises the structure of Formula II:
  • Embodiment 18 A composition comprising the modified target-specific nucleic acid molecule of any one of embodiments 1-17, and optionally a pharmaceutically or physiologically acceptable carrier.
  • Embodiment 19 A method of increasing the binding affinity of a target-specific nucleic acid molecule to a target, the method comprising replacing one or more of the target-specific nucleic acid molecules with one or more quinoline-modified nucleotides. Naturally occurring nucleotides.
  • Embodiment 20 The method of Embodiment 19, wherein the quinoline modification is quinolin-2-yl substitution, quinolin-3-yl substitution, quinolin-4-yl substitution, quinolin-5-yl substitution, quinolin-5-yl substitution, quinolin-6-yl substitution, quinolin-7-yl substitution or quinolin-8-yl substitution.
  • the quinoline modification is quinolin-5-yl substitution.
  • Embodiment 21 The method of embodiment 19 or 20, wherein the quinoline modified nucleotide comprises a substituent group R selected from the following ag:
  • Embodiment 22 The method of any one of embodiments 19-21, wherein the quinoline modified nucleotide comprises the base moiety of Formula I:
  • Embodiment 23 The method of any one of embodiments 19-22, wherein the quinoline modified nucleotide comprises the following structure of Formula II:
  • Embodiment 24 The method of any one of embodiments 19-23, said one or more quinoline modified nucleotide substitutions being introduced during chemical synthesis of said target-specific nucleic acid molecule.
  • Embodiment 25 The method of embodiment 24, wherein a modified phosphoramidite monomer selected from the group consisting of 2a-2g is added during chemical synthesis of the target-specific nucleic acid molecule:
  • Embodiment 26 A modified phosphoramidite monomer selected from the following 2a-2g, which is used to increase the binding affinity of a target-specific nucleic acid molecule to a target:
  • Figure 1 Measurement results of the binding ability of osteosclerostin aptamers modified with different subtypes of quinoline at key sites and targets.
  • the highlighted parts in the sequence are key sites.
  • the horizontal axis in the heat map represents the key sites modified with quinoline, and the vertical axis represents different subtypes of quinoline.
  • the binding ability was tested using the ELONA (Enzyme Ligated OligoNucleotide Assay) method.
  • Figure 2 Results of measuring the affinity of quinoline-modified aptamers for targets using Biolayer interferometry (BLI). a) Binding and dissociation curves of interaction between each aptamer and target. b) The equilibrium dissociation constant KD value calculated by the final fitting.
  • the invention provides a modified target-specific nucleic acid molecule comprising one or more quinoline-modified nucleotides, wherein the modified target-specific nucleic acid molecule has a higher The binding affinity of the nucleic acid molecule to the target is increased.
  • the corresponding control nucleic acid molecule refers to a target-specific nucleic acid molecule that does not comprise the one or more quinoline-modified nucleotides.
  • a "target-specific nucleic acid molecule” refers to a nucleic acid molecule that is capable of specifically binding to a particular target.
  • the targets include but are not limited to proteins, nucleic acids, small molecule compounds, etc.
  • the nucleic acid molecules include but are not limited to aptamers, antisense nucleic acids (antisense RNA), dsRNA (such as siRNA, shRNA), preferably aptamers.
  • the modified target-specific nucleic acid molecule has an increased binding affinity to the target by at least about 10%, at least about 20%, at least about 30%, at least about 40% compared to a corresponding control nucleic acid molecule. , at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100% or more. In some embodiments, the modified target-specific nucleic acid molecule binds the target with an affinity that is at least about 1.5 times, at least about 2 times, at least about 3 times, at least about 4 times greater than the affinity of the corresponding unmodified nucleic acid molecule.
  • binding affinity can be measured, for example, by enzyme-linked oligonucleotide assay (ELONA) or by biofilm interferometry (BLI).
  • the modified target-specific nucleic acid molecule has a KD for the target of less than 100 nM, preferably less than 50 nM, preferably less than 40 nM, preferably less than 30 nM, preferably less than 20 nM, preferably less than 10 nM, preferably less than 5 nM or less. (dissociation constant).
  • the KD is measured, for example, by enzyme-linked oligonucleotide assay (ELONA) or by biofilm interferometry (BLI).
  • the quinoline modification is quinolin-2-yl substitution, quinolin-3-yl substitution, quinolin-4-yl substitution, quinolin-5-yl substitution, quinolin-6-yl substitution Substituted, quinolin-7-yl substituted or quinolin-8-yl substituted.
  • the quinoline modification is a quinolin-5-yl substitution.
  • the substitution position numbers on quinoline are as follows:
  • the quinoline modified nucleotide comprises a substituent group R selected from the following ag:
  • the quinoline modified nucleotide contains the substituent group R in the base portion thereof.
  • the quinoline modified nucleotide is derived from uridine, such as deoxyuridine.
  • the quinoline modified nucleotide includes the base moiety of Formula I:
  • the quinoline modified nucleotides comprise the structure of Formula II:
  • one or more nucleotides (natural nucleotides) in the target-specific nucleic acid molecule are replaced with the quinoline-modified nucleotides.
  • the target-specific nucleic acid molecule may also comprise one or more modifications that extend its half-life in vivo.
  • the modifications that extend its half-life in vivo include, for example, 3' and 5' modifications, such as 3' and 5' capping.
  • the aptamer is capped at the 3' end with inverted deoxythymidine, i.e., 3' inverted deoxythymidine (3'idT) modification.
  • the modification to extend its half-life in vivo may also include replacing one or more naturally occurring nucleotides with modified nucleotides.
  • the modified nucleotides for extending half-life include, but are not limited to, 2'-fluoro, 2'-methoxyethyl, 2'-methoxy and/or 2'propenoxy modified nucleosides Acid (that is, the hydroxyl group at the 2' position of ribose is replaced by fluorine, methoxyethyl, methoxy or propyleneoxy, etc.).
  • the modified nucleotides for extending half-life may also include C-5 modified pyrimidines.
  • C-5 modified pyrimidine refers to a pyrimidine with a modification at the C-5 position.
  • C-5 modified pyrimidines can enhance the nuclease resistance of oligonucleotides and are known in the art. For example, see International Patent Application WO 2011/130195 and its cited documents.
  • Modifications for extending half-life also include modifications between nucleotides, such as nucleotides with uncharged bonds (e.g., methylphosphonate, phosphotriester, phosphate amine ester, carbamate, etc.) Inter-modifications and inter-nucleotide modifications with charged bonds (e.g. phosphorothioates, phosphorodithioates, etc.), inter-nucleotide modifications with intercalating agents (e.g.
  • Internucleotide modifications containing chelating agents such as metals, radioactive metals, boron, oxidizing metals, etc.
  • internucleotide modifications containing alkylating agents such as alpha anomeric nucleic acids, etc.
  • the modification for extending half-life may also include pegylation modification (PEG modification).
  • PEG modification pegylation modification
  • the invention also provides a composition, such as a pharmaceutical composition, comprising a modified target-specific nucleic acid molecule of the invention.
  • a composition such as a pharmaceutical composition, comprising a modified target-specific nucleic acid molecule of the invention.
  • the composition further includes a pharmaceutically or physiologically acceptable carrier or excipient.
  • the composition may, for example, be used to detect the target, or to diagnose and/or treat a disease associated with the target.
  • the invention provides a method for increasing the binding affinity of a target-specific nucleic acid molecule to a target, the method comprising replacing the nucleotides in the target-specific nucleic acid molecule with one or more quinoline-modified nucleotides.
  • the quinoline modified nucleotide is as defined above.
  • the nucleotide substitutions are introduced during chemical synthesis (eg, solid phase) of the target-specific nucleic acid molecule.
  • a modified phosphoramidite monomer selected from 2a-2g below is added during chemical synthesis of the target-specific nucleic acid molecule:
  • the present invention also provides modified phosphoramidite monomers selected from the following 2a-2g, which are used to improve the binding affinity of target-specific nucleic acid molecules to targets:
  • the target-specific nucleic acid molecule is an osteosclerostin-specific nucleic acid molecule. Accordingly, the present invention also provides modified osteosclerostin-specific nucleic acid molecules comprising one or more quinoline-modified nucleotides as described above.
  • the target is sclerostin, such as human sclerostin.
  • sclerostin such as human sclerostin.
  • An exemplary human osteosclerostin contains the amino acid sequence:
  • the target-specific nucleic acid molecule is an aptamer that specifically binds osteosclerostin (modified osteosclerostin-specific aptamer).
  • the aptamer (unmodified) that specifically binds osteosclerostin comprises the nucleotide sequence:
  • the modified osteosclerostin-specific aptamer is a nucleoside modified by the quinoline at one or more positions of a nucleotide corresponding to SEQ ID NO: 2 selected from: Acid substitution: 1st, 2nd, 3rd, 4th, 5th, 6th, 8th, 11th, 13th, 15th, 29th, 30th , 31st, 32nd, 33rd, 34th, 35th, 36th, 37th, 38th, 39th and 40th.
  • the modified osteosclerostin-specific nucleic acid molecule is at position 6, position 8, position 11, position 13, position 32 and/or corresponding to SEQ ID NO:2
  • the nucleotide at position 34 is substituted with the quinoline modified nucleotide.
  • the modified osteosclerostin-specific nucleic acid molecule comprises a nucleotide sequence selected from:
  • x is the quinoline modified nucleotide
  • the modified osteosclerostin-specific nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 8, wherein x is the quinoline modified nucleotide. In some embodiments, the modified osteosclerostin-specific nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 10, wherein x is the quinoline modified nucleotide. In some embodiments, the modified osteosclerostin-specific nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 11, wherein x is the quinoline modified nucleotide.
  • the modified osteosclerostin-specific nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 16, wherein x is the quinoline modified nucleotide. In some embodiments, the modified osteosclerostin-specific nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 18, wherein x is the quinoline modified nucleotide.
  • the quinoline modified nucleotides comprise the structure of Formula II:
  • the binding affinity of the modified osteosclerostin-specific nucleic acid molecule of the present invention to osteosclerosis is the affinity of the unmodified osteosclerostin-specific nucleic acid molecule shown in SEQ ID NO: 2 At least about 1.5 times, at least about 2 times, at least about 3 times, at least about 4 times, at least about 5 times, at least about 6 times, at least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, At least about 15 times, at least about 16 times or more.
  • the binding affinity can be measured, for example, by enzyme-linked oligonucleotide assay (ELONA) or by biofilm interferometry (BLI).
  • the modified osteosclerostin-specific nucleic acid molecule has a KD (dissociation constant) for osteosclerosis of less than 10 nM or even less than 5 nM.
  • the KD is measured, for example, by enzyme-linked oligonucleotide assay (ELONA) or by biofilm interferometry (BLI).
  • the present invention also provides a composition comprising the modified osteosclerostin-specific nucleic acid molecule of the present invention, and optionally a pharmaceutically or physiologically acceptable carrier.
  • the composition is used, for example, for detecting osteosclerostin, or for diagnosing and/or treating osteosclerostin-related diseases.
  • the invention also provides a method of treating a disease by a modified osteosclerostin-specific nucleic acid molecule of the invention, the method comprising administering to a subject in need thereof a therapeutically effective amount of a modified osteosclerostin of the invention Specific nucleic acid molecules.
  • the disease is, for example, an osteosclerostin-related disease, such as an osteosclerostin-mediated disease.
  • osteosclerostin-related disease includes conditions in which bone mineral density (BMD) is abnormally and/or pathologically low relative to healthy subjects.
  • Diseases characterized by low BMD and/or bone fragility include, but are not limited to: primary and secondary osteoporosis, osteopenia, osteomalacia, osteogenesis imperfecta (OI), avascular necrosis (bone necrosis), fractures and implant healing (dental implants and hip implants), bone loss due to other conditions (eg, associated with HIV infection, cancer, and arthritis).
  • Other "osteosclerostin-related diseases” include, but are not limited to: hypophosphatemic rickets, rheumatoid arthritis, osteoarthritis, arthritis, and osteolytic lesions.
  • osteosclerosis-related cancers such as myeloma (eg, multiple myeloma with osteolytic lesions), breast cancer (eg, triple-negative breast cancer), colon cancer, Melanoma, hepatocellular carcinoma, epithelial cancer, esophageal cancer, brain cancer, lung cancer, prostate cancer or pancreatic cancer, and any metastases thereto.
  • “Osteosclerin-related diseases” may also include renal and cardiovascular diseases caused by at least expression of osteosclerostin in the kidneys and in the cardiovascular system.
  • Such conditions include, but are not limited to, renal diseases such as: glomerular diseases (e.g., acute and chronic glomerulonephritis, rapidly progressive glomerulonephritis, nephrotic syndrome, focal proliferative glomerulonephritis, and Glomerular damage associated with systemic diseases such as systemic lupus erythematosus, Goodpasture's syndrome, multiple myeloma, diabetes, polycystic kidney disease, neoplasia, sickle cell disease, and chronic inflammation), renal tubules Diseases (e.g., acute tubular necrosis and acute renal failure, polycystic kidney disease, medullary sponge kidney disease, medullary cystic disease, nephrogenic diabetes, and tubular acidosis), tubulointerstitial disease (e.g
  • the osteosclerostin-related diseases also include, but are not limited to, cardiovascular diseases such as: ischemic heart disease (eg, angina pectoris, myocardial infarction, and chronic ischemic heart disease), hypertensive heart disease, cor pulmonale, heart disease, Valvular disease (eg, rheumatic fever and rheumatic heart disease, endocarditis, mitral valve prolapse, and aortic stenosis), congenital heart disease (eg, occlusive lesions of valves and blood vessels, atrial or ventricular septal defects , and persistent ductus arteriosus), or cardiomyopathies (eg, myocarditis, congestive cardiomyopathy, and hypertrophic cardiomyopathy).
  • cardiovascular diseases such as: ischemic heart disease (eg, angina pectoris, myocardial infarction, and chronic ischemic heart disease), hypertensive heart disease, cor pulmonale, heart disease, Valvular disease (eg,
  • treating means that the subject's symptoms are partially or completely alleviated, or remain unchanged following treatment. Treatment therefore includes prevention, treatment and/or cure. Prevention means preventing underlying disease and/or preventing symptoms from worsening or disease progression.
  • a “therapeutically effective amount” or “therapeutically effective dose” refers to an amount of a substance, compound, material or composition containing a compound that is at least sufficient to produce a therapeutic effect upon administration to a subject. Thus, it is an amount necessary to prevent, cure, ameliorate, block or partially block the symptoms of a disease or condition.
  • therapeutic effect means an effect resulting from treatment of a subject that alters, generally ameliorates, or ameliorates the symptoms of a disease or disease condition, or cures a disease or disease condition.
  • Dosage regimens utilizing the nucleic acid molecules are selected based on a variety of factors, including, for example, the type, species, age, weight, gender, and medical condition of the patient; the severity of the condition to be treated; the route of administration; the The patient's kidney and liver function; and the specific nucleic acid molecule or salt thereof used.
  • the ordinarily skilled physician can readily determine and prescribe the effective amount of the composition required to prevent, combat, or inhibit the progression of the disorder.
  • Example 1 synthetic routes of compounds 1a to 1g and 2a to 2g
  • the NMR data are as follows:
  • Synthetic route of 2a ⁇ 2g Under nitrogen protection and ice water bath conditions, add tetrazole to 50mL 3.3mmol 1a ⁇ 1g (1a is 2.2g, 1b-1g is 2.4g) anhydrous dichloromethane solution (0.69g, 9.8mmol) and the phosphorus reagent 2-cyanoethyl-N,N,N',N'-tetraisopropylphosphoramidite (2.9g, 9.8mmol). The solution was then moved to room temperature and stirred for 2 hours, and the solution was concentrated in vacuo.
  • Example 2 Modification of nucleic acid aptamer APC001 by quinoline to increase its target binding affinity
  • nucleic acid aptamer APC001 which specifically targets osteosclerostin, was screened through protein-SELEX technology.
  • the specific sequence is: CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC.
  • 22 key sites for aptamer-target binding were screened (respectively C1, G2, G3, G4, G5, T6, T8, G11, T13, G15, T29, T30 , G31, G32, C33, A34, G35, C36, T37, G38, C39, C40).
  • the present invention uses chemical modification technology to modify different subtypes of quinolines at these key sites.
  • different quinolines are coupled to nucleosides to obtain compounds 1a-1g, and then reacted with phosphorus reagents to construct different quinoline isoforms modified phosphoramidite monomers 2a-2g.
  • DNA solid-phase synthesis technology is used to obtain compounds in different Aptamers with quinoline modification at key sites.
  • the phosphoramidite monomers 2a to 2g were inserted into the 22 key sites of the osteosclerostin aptamer through standard DNA solid-phase synthesis technology.
  • the sequence and insertion position are shown in Table 1 below:
  • ELONA B&W buffer PBS, 1mM MgCl2, 0.05% Tween 20;
  • ELONA assay blocking buffer PBS,0.1% Tween 20and 1%BSA;
  • ELONA assay washing buffer PBS, 1mM MgCl2, 0.1% Tween 20 and 0.1% BSA;
  • ELONA assay Strep-HRP binding buffer Strep-HRP 1:10000 diluted into ELONA assay washing buffer
  • the results are shown in Figure 1.
  • the obtained quinoline-modified osteosclerostin nucleic acid aptamer generally has improved target-binding ability compared with the natural APC001.
  • the improvement effect is most significant (nearly 16 times).
  • Biolayer interferometry determines the affinity between quinoline-modified aptamers and targets
  • Biolayer interferometry was used to determine the affinity of the six sequences with the highest binding ability in Figure 1, namely APC001-d6, APC001-d8, APC001-d11, APC001-d13, APC001-d32, and APC001-d34.
  • the instrument used to analyze the interaction between quinoline-modified aptamers and osteosclerostin was Octet 96/96e system (ForteBio), and all operations were performed at room temperature.
  • the program running procedure is as follows:
  • Baseline 1 ⁇ PBS buffer (10mM phosphate buffer, 2.7mM KCl and 137mM NaCl, pH 7.4), 120s, 1000rpm;

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Plant Pathology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to the field of biomedicines. Specifically, the present invention relates to a target-specific nucleic acid molecule having a quinoline modification, the quinoline modification being capable of improving the binding affinity between the nucleic acid molecule and a target.

Description

包含喹啉修饰的靶特异性核酸分子Target-specific nucleic acid molecules comprising quinoline modifications 技术领域Technical field
本发明涉及生物医药领域。具体而言,涉及一种包含喹啉修饰的靶特异性核酸分子,所述喹啉修饰能够提高所述核酸分子与靶的结合亲和力。The invention relates to the field of biomedicine. Specifically, it relates to a target-specific nucleic acid molecule comprising a quinoline modification that can increase the binding affinity of the nucleic acid molecule to a target.
发明背景Background of the invention
核酸适配子(Aptamer)是单链DNA或RNA,可与靶标形成互补结构,具有与单克隆抗体药物媲美的特异性与亲和力,同时还具有无免疫原性、易于生产、成本低、热稳定好等优势。Nucleic acid aptamers (Aptamers) are single-stranded DNA or RNA that can form a complementary structure with the target. They have specificity and affinity comparable to monoclonal antibody drugs. They are also non-immunogenic, easy to produce, low cost, and thermally stable. Good to wait for the advantage.
骨硬化素是骨形成中的负调节因子,是开发促进骨形成从而治疗骨质疏松等相关疾病的药物的理想靶点。除此之外,研究发现,骨硬化素在成骨不全、低磷佝偻病和三阴性乳腺癌等病症的发生、发展过程中也发挥重要调控作用,所以也是这些疾病治疗的重要靶点。骨硬化素单抗Romosozumab能够促进骨形成,美国、欧洲和亚洲地区的药品监管机构已经批准骨硬化素单抗上市,适应症是绝经后骨质疏松症具有骨折高风险的患者。由于骨硬化素单抗临床存在严重的心血管风险,美国药品监管机构FDA要求患者必须与医生共同评估成骨治疗的益处与心血管风险之后,才能使用,而且只能允许使用一年;欧洲药品监管机构EMA进一步严格限制,临床使用骨硬化素单抗的骨质疏松骨折高风险患者既往不能有心血管疾病史。Osteosclerostin is a negative regulator of bone formation and is an ideal target for the development of drugs that promote bone formation and treat osteoporosis and other related diseases. In addition, studies have found that osteosclerostin also plays an important regulatory role in the occurrence and development of diseases such as osteogenesis imperfecta, hypophosphatemic rickets, and triple-negative breast cancer, and is therefore an important target for the treatment of these diseases. Sclerostin monoclonal antibody Romosozumab can promote bone formation. Drug regulatory agencies in the United States, Europe and Asia have approved osteosclerostin monoclonal antibody for marketing. The indication is postmenopausal patients with osteoporosis who are at high risk of fracture. Due to the serious clinical cardiovascular risks of osteosclerostin monoclonal antibody, the US drug regulatory agency FDA requires patients to jointly evaluate the benefits and cardiovascular risks of osteogenic treatment with their doctors before using it, and it can only be used for one year; European drugs The regulatory agency EMA has further strictly restricted the clinical use of sclerostin monoclonal antibody in patients with high risk of osteoporotic fractures who must not have a history of cardiovascular disease.
骨硬化素蛋白有三个结构区,分别是loop1、loop2和loop3。其中,骨硬化素单抗主要作用于loop2。研究发现loop2既参与骨形成抑制,也参与心血管保护,而骨硬化素loop3只参与骨形成抑制但不参与心血管保护的。因此通过SELEX技术筛选得到特异性靶向骨硬化素loop3的核酸适配子—APC001,并在骨质疏松症模型和成骨不全症模型的治疗中验证了该适配子具有促进骨合成代谢的治疗潜能且不增加心血管疾病风险。该治疗性适配子于2019年获得美国FDA成骨不全孤儿药认证(FDA,DRU-2019-6966)。Sclerostin protein has three structural regions, namely loop1, loop2 and loop3. Among them, osteosclerostin monoclonal antibody mainly acts on loop2. Studies have found that loop2 is involved in both bone formation inhibition and cardiovascular protection, while osteosclerostin loop3 is only involved in bone formation inhibition but not cardiovascular protection. Therefore, the nucleic acid aptamer-APC001, which specifically targets osteosclerostin loop3, was screened through SELEX technology, and the aptamer was verified to have the ability to promote bone anabolism in the treatment of osteoporosis models and osteogenesis imperfecta models. Therapeutic potential without increasing cardiovascular disease risk. This therapeutic aptamer received Osteogenesis Imperfecta orphan drug certification from the U.S. FDA in 2019 (FDA, DRU-2019-6966).
提高适配子对靶标的亲和力有助于改善适配子药物药效偏低的成药性瓶颈。目前,适配子APC001对骨硬化素的亲和力(KD=39.9nM)比骨硬化素单抗(KD=0.12nM)低。因此,为进一步提高该适配子抑制骨硬化素的能力(甚至超过单抗),有必要采取有效措施提高其对靶标骨硬化素的亲和力。Improving the affinity of aptamers for targets can help improve the druggability bottleneck of low efficacy of aptamer drugs. Currently, the affinity of aptamer APC001 for osteosclerostin (KD=39.9nM) is lower than that of osteosclerostin monoclonal antibody (KD=0.12nM). Therefore, in order to further improve the aptamer's ability to inhibit osteosclerostin (even beyond that of monoclonal antibodies), it is necessary to take effective measures to increase its affinity for the target osteosclerostin.
已有研究表明,在适配子上进行疏水性修饰可以提高适配子与靶标的亲和力。发展新的适配子修饰方法,拓展适配子的化学多样性,对于适配子进一步的临床应用有非常重要意义。Studies have shown that hydrophobic modifications on aptamers can improve the affinity of aptamers to targets. Developing new aptamer modification methods and expanding the chemical diversity of aptamers are of great significance for the further clinical application of aptamers.
发明简述 Brief description of the invention
本发明至少提供以下实施方案:The present invention at least provides the following embodiments:
实施方案1.一种经修饰的靶特异性核酸分子,其包含一或多个喹啉修饰的核苷酸,其中与相应的对照核酸分子相比,所述经修饰的靶特异性核酸分子与靶的结合亲和力提高。Embodiment 1. A modified target-specific nucleic acid molecule comprising one or more quinoline-modified nucleotides, wherein the modified target-specific nucleic acid molecule is compared to a corresponding control nucleic acid molecule. Target binding affinity is increased.
实施方案2.实施方案1的经修饰的靶特异性核酸分子,其中所述核酸分子选自适配子(aptamer)、反义核酸(反义RNA)、dsRNA(如siRNA、shRNA)。Embodiment 2. The modified target-specific nucleic acid molecule of embodiment 1, wherein said nucleic acid molecule is selected from the group consisting of an aptamer (aptamer), an antisense nucleic acid (antisense RNA), dsRNA (eg, siRNA, shRNA).
实施方案3.实施方案1或2的经修饰的靶特异性核酸分子,其中与相应的对照核酸分子相比,所述经修饰的靶特异性核酸分子与靶的结合亲和力提高至少约10%、至少约20%、至少约30%、至少约40%、至少约50%、至少约60%、至少约70%、至少约80%、至少约90%、至少约100%或更多;或者所述经修饰的靶特异性核酸分子与靶的结合亲和力是未修饰的相应核酸分子的亲和力的至少约1.5倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约15倍、至少约16倍、至少约20倍、至少约25倍、至少约30倍、至少约40倍、至少约50倍、至少约75倍、至少约100倍或更多倍。Embodiment 3. The modified target-specific nucleic acid molecule of embodiment 1 or 2, wherein the modified target-specific nucleic acid molecule has a binding affinity to a target that is increased by at least about 10% compared to a corresponding control nucleic acid molecule. At least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100% or more; or all The binding affinity of the modified target-specific nucleic acid molecule to the target is at least about 1.5 times, at least about 2 times, at least about 3 times, at least about 4 times, at least about 5 times, at least about the affinity of the unmodified corresponding nucleic acid molecule. About 6 times, at least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, at least about 15 times, at least about 16 times, at least about 20 times, at least about 25 times, at least about 30 times, at least About 40 times, at least about 50 times, at least about 75 times, at least about 100 times or more.
实施方案4.实施方案1-3中任一项的经修饰的靶特异性核酸分子,其中所述经修饰的靶特异性核酸分子对靶具有小于100nM,优选小于50nM,优选小于40nM,优选小于30nM,优选小于20nM,优选小于10nM,优选小于5nM或更小的KD(解离常数)。Embodiment 4. The modified target-specific nucleic acid molecule of any one of embodiments 1-3, wherein the modified target-specific nucleic acid molecule has an affinity for the target of less than 100 nM, preferably less than 50 nM, preferably less than 40 nM, preferably less than 30 nM, preferably less than 20 nM, preferably less than 10 nM, preferably less than 5 nM or less KD (dissociation constant).
实施方案5.实施方案1-4中任一项的经修饰的靶特异性核酸分子,其中所述喹啉修饰是喹啉-2-基取代、喹啉-3-基取代、喹啉-4-基取代、喹啉-5-基取代、喹啉-6-基取代、喹啉-7-基取代或喹啉-8-基取代,优选地,所述喹啉修饰是喹啉-5-基取代。Embodiment 5. The modified target-specific nucleic acid molecule of any one of embodiments 1-4, wherein the quinoline modification is quinolin-2-yl substitution, quinolin-3-yl substitution, quinolin-4 - base substitution, quinolin-5-yl substitution, quinolin-6-yl substitution, quinolin-7-yl substitution or quinolin-8-yl substitution, preferably, the quinoline modification is quinolin-5- base substitution.
实施方案6.实施方案1-5中任一项的经修饰的靶特异性核酸分子,其中所述喹啉修饰的核苷酸包含取代基团R,所述R选自以下a-g:
Embodiment 6. The modified target-specific nucleic acid molecule of any one of embodiments 1-5, wherein the quinoline-modified nucleotide comprises a substituent group R selected from ag:
优选地, Preferably,
实施方案7.实施方案1-6中任一项的经修饰的靶特异性核酸分子,其中所述喹啉修饰的核苷酸包括以下式I的碱基部分:
Embodiment 7. The modified target-specific nucleic acid molecule of any one of embodiments 1-6, wherein the quinoline-modified nucleotide comprises the base moiety of Formula I:
其中
in
优选地, Preferably,
实施方案8.实施方案1-7中任一项的经修饰的靶特异性核酸分子,其中所述喹啉修饰的核苷酸包括以下式II的结构:
Embodiment 8. The modified target-specific nucleic acid molecule of any one of embodiments 1-7, wherein the quinoline-modified nucleotide comprises the following structure of Formula II:
其中
in
优选地, Preferably,
实施方案9.实施方案1-8中任一项的经修饰的靶特异性核酸分子,其中所述靶特异性核酸分子中的一或多个天然存在的核苷酸被所述喹啉修饰的核苷酸取代。Embodiment 9. The modified target-specific nucleic acid molecule of any one of embodiments 1-8, wherein one or more naturally occurring nucleotides in the target-specific nucleic acid molecule are modified with the quinoline Nucleotide substitutions.
实施方案10.实施方案1-9中任一项的经修饰的靶特异性核酸分子,其中所述靶特异性核酸分子还可以包含一或多种延长其体内半衰期修饰。Embodiment 10. The modified target-specific nucleic acid molecule of any one of embodiments 1-9, wherein the target-specific nucleic acid molecule may further comprise one or more modifications that extend its half-life in vivo.
实施方案11.实施方案1-10中任一项的经修饰的靶特异性核酸分子,其中所述经修饰的靶特异性核酸分子是经修饰的骨硬化素特异性适配子。Embodiment 11. The modified target-specific nucleic acid molecule of any one of embodiments 1-10, wherein the modified target-specific nucleic acid molecule is a modified osteosclerostin-specific aptamer.
实施方案12.实施方案11的经修饰的靶特异性核酸分子,其中经修饰的骨硬化素特异性适配子衍生自SEQ ID NO:2的骨硬化素特异性适配子。Embodiment 12. The modified target-specific nucleic acid molecule of embodiment 11, wherein the modified osteosclerostin-specific aptamer is derived from the osteosclerostin-specific aptamer of SEQ ID NO: 2.
实施方案13.实施方案12的经修饰的靶特异性核酸分子,其中所述经修饰的骨硬化素特异性适配子在相应于SEQ ID NO:2的选自以下的一或多个位置的核苷酸被所述喹啉修饰的核苷酸取代:第1位、第2位、第3位、第4位、第5位、第6位、第8位、第11位、第13位、第15位、第29位、第30位、第31位、第32位、第33位、第34位、第35位、第36位、第37位、第38位、第39位和第40位。 Embodiment 13. The modified target-specific nucleic acid molecule of embodiment 12, wherein the modified osteosclerostin-specific aptamer is at one or more positions selected from the group consisting of corresponding to SEQ ID NO:2 The nucleotides are replaced by the quinoline modified nucleotides: position 1, position 2, position 3, position 4, position 5, position 6, position 8, position 11, position 13 , 15th, 29th, 30th, 31st, 32nd, 33rd, 34th, 35th, 36th, 37th, 38th, 39th and 39th 40 people.
实施方案14.实施方案12或13的经修饰的靶特异性核酸分子,其中所述经修饰的骨硬化素特异性核酸分子在相应于SEQ ID NO:2的第6位、第8位、第11位、第13位、第32位和/或第34位的一或多个核苷酸被所述喹啉修饰的核苷酸取代。Embodiment 14. The modified target-specific nucleic acid molecule of embodiment 12 or 13, wherein the modified osteosclerostin-specific nucleic acid molecule is at position 6, position 8, position corresponding to SEQ ID NO: 2 One or more nucleotides at position 11, 13, 32 and/or 34 are substituted with the quinoline modified nucleotide.
实施方案15.实施方案12-14中任一项的经修饰的靶特异性核酸分子,其中所述经修饰的骨硬化素特异性核酸分子包含选自SEQ ID NO:3-23的核苷酸序列,其中x为所述喹啉修饰的核苷酸。Embodiment 15. The modified target-specific nucleic acid molecule of any one of embodiments 12-14, wherein the modified osteosclerostin-specific nucleic acid molecule comprises a nucleotide selected from the group consisting of SEQ ID NOs: 3-23 Sequence, where x is the quinoline modified nucleotide.
实施方案16.实施方案12-15中任一项的经修饰的靶特异性核酸分子,其中所述经修饰的骨硬化素特异性核酸分子包含选自SEQ ID NO:8、10、11、16和18的核苷酸序列,其中x为所述喹啉修饰的核苷酸。Embodiment 16. The modified target-specific nucleic acid molecule of any one of embodiments 12-15, wherein the modified osteosclerostin-specific nucleic acid molecule comprises a nucleic acid molecule selected from the group consisting of SEQ ID NO: 8, 10, 11, 16 and the nucleotide sequence of 18, wherein x is the quinoline-modified nucleotide.
实施方案17.实施方案12-16中任一项的经修饰的靶特异性核酸分子,其中所述喹啉修饰的核苷酸包含下式II的结构:
Embodiment 17. The modified target-specific nucleic acid molecule of any one of embodiments 12-16, wherein the quinoline-modified nucleotide comprises the structure of Formula II:
其中, in,
实施方案18.一种组合物,其包含实施方案1-17中任一项的经修饰的靶特异性核酸分子,以及任选的药学上或生理上可接受的载体。Embodiment 18. A composition comprising the modified target-specific nucleic acid molecule of any one of embodiments 1-17, and optionally a pharmaceutically or physiologically acceptable carrier.
实施方案19.一种提高靶特异性核酸分子与靶的结合亲和力的方法,所述方法包括用一或多个喹啉修饰的核苷酸取代所述靶特异性核酸分子中的一个或多个天然存在的核苷酸。Embodiment 19. A method of increasing the binding affinity of a target-specific nucleic acid molecule to a target, the method comprising replacing one or more of the target-specific nucleic acid molecules with one or more quinoline-modified nucleotides. Naturally occurring nucleotides.
实施方案20.实施方案19的方法,其中所述喹啉修饰是喹啉-2-基取代、喹啉-3-基取代、喹啉-4-基取代、喹啉-5-基取代、喹啉-6-基取代、喹啉-7-基取代或喹啉-8-基取代,优选地,所述喹啉修饰是喹啉-5-基取代。Embodiment 20. The method of Embodiment 19, wherein the quinoline modification is quinolin-2-yl substitution, quinolin-3-yl substitution, quinolin-4-yl substitution, quinolin-5-yl substitution, quinolin-5-yl substitution, quinolin-6-yl substitution, quinolin-7-yl substitution or quinolin-8-yl substitution. Preferably, the quinoline modification is quinolin-5-yl substitution.
实施方案21.实施方案19或20的方法,其中所述喹啉修饰的核苷酸包含取代基团R,所述R选自以下a-g:
Embodiment 21. The method of embodiment 19 or 20, wherein the quinoline modified nucleotide comprises a substituent group R selected from the following ag:
优选地, Preferably,
实施方案22.实施方案19-21中任一项的方法,其中所述喹啉修饰的核苷酸包括以下式I的碱基部分:
Embodiment 22. The method of any one of embodiments 19-21, wherein the quinoline modified nucleotide comprises the base moiety of Formula I:
其中
in
优选地, Preferably,
实施方案23.实施方案19-22中任一项的方法,其中所述喹啉修饰的核苷酸包括以下式II的结构:
Embodiment 23. The method of any one of embodiments 19-22, wherein the quinoline modified nucleotide comprises the following structure of Formula II:
其中
in
优选地, Preferably,
实施方案24.实施方案19-23中任一项的方法,所述一或多个喹啉修饰的核苷酸取代在化学合成所述靶特异性核酸分子期间引入。Embodiment 24. The method of any one of embodiments 19-23, said one or more quinoline modified nucleotide substitutions being introduced during chemical synthesis of said target-specific nucleic acid molecule.
实施方案25.实施方案24的方法,其中在化学合成所述靶特异性核酸分子期间加入选自以下2a-2g的经修饰的亚磷酰胺单体:
Embodiment 25. The method of embodiment 24, wherein a modified phosphoramidite monomer selected from the group consisting of 2a-2g is added during chemical synthesis of the target-specific nucleic acid molecule:
实施方案26.一种选自以下2a-2g的经修饰的亚磷酰胺单体,其用于提高靶特异性核酸分子与靶的结合亲和力:
Embodiment 26. A modified phosphoramidite monomer selected from the following 2a-2g, which is used to increase the binding affinity of a target-specific nucleic acid molecule to a target:
附图简述Brief description of the drawings
图1、在关键位点分别修饰不同亚型喹啉修饰的骨硬化素适配子与靶标的结合能力测定结果。序列中高亮部分为关键位点。热图中横轴为用以喹啉修饰的关键位点,纵轴为不同亚型的喹啉。结合能力考察采用ELONA(Enzyme Ligated OligoNucleotide Assay)方法。Figure 1. Measurement results of the binding ability of osteosclerostin aptamers modified with different subtypes of quinoline at key sites and targets. The highlighted parts in the sequence are key sites. The horizontal axis in the heat map represents the key sites modified with quinoline, and the vertical axis represents different subtypes of quinoline. The binding ability was tested using the ELONA (Enzyme Ligated OligoNucleotide Assay) method.
图2、生物膜干涉法(Biolayer interferometry,BLI)测定喹啉修饰适配子对靶标亲和力的结果。a)各适配子与靶标相互作用结合和解离曲线图。b)最终拟合计算得到的平衡解离常数KD值。Figure 2. Results of measuring the affinity of quinoline-modified aptamers for targets using Biolayer interferometry (BLI). a) Binding and dissociation curves of interaction between each aptamer and target. b) The equilibrium dissociation constant KD value calculated by the final fitting.
发明详述Detailed description of the invention
除非另有指示或定义,否则所有所用术语均具有本领域中的通常含义,该含义将为本领域技术人员所了解。参考例如标准手册,如Sambrook et al.,“Molecular Cloning:A  Laboratory Manual”;Lewin,“Genes VIII”;及Roitt et al.,“Immunology”(第8版),以及本文中引用的一般现有技术;此外,除非另有说明,否则未具体详述的所有方法、步骤、技术及操作均可以且已经以本身已知的方式进行,该方式将为本领域技术人员所了解。亦参考例如标准手册、上述一般现有技术及其中引用的其他参考文献。Unless otherwise indicated or defined, all terms used have their ordinary meanings in the art, which meanings will be understood by those skilled in the art. Reference, for example, standard manuals such as Sambrook et al., "Molecular Cloning: A Laboratory Manual"; Lewin, "Genes VIII"; and Roitt et al., "Immunology" (8th ed.), as well as general prior art references herein; in addition, unless otherwise indicated, all references not specifically recited Methods, steps, techniques and operations can and have been performed in a manner known per se, which manner will be understood by a person skilled in the art. Reference is also made to, for example, standard manuals, the general prior art mentioned above and other references cited therein.
I.结合亲和力提高的经修饰的靶特异性核酸分子I. Modified target-specific nucleic acid molecules with increased binding affinity
在一方面,本发明提供一种经修饰的靶特异性核酸分子,其包含一或多个喹啉修饰的核苷酸,其中与相应的对照核酸分子相比,所述经修饰的靶特异性核酸分子与靶的结合亲和力提高。在一些实施方案中,所述相应的对照核酸分子是指不包含所述一或多个喹啉修饰的核苷酸的靶特异性核酸分子。In one aspect, the invention provides a modified target-specific nucleic acid molecule comprising one or more quinoline-modified nucleotides, wherein the modified target-specific nucleic acid molecule has a higher The binding affinity of the nucleic acid molecule to the target is increased. In some embodiments, the corresponding control nucleic acid molecule refers to a target-specific nucleic acid molecule that does not comprise the one or more quinoline-modified nucleotides.
如本文所用,“靶特异性核酸分子”指的是能够与特定的靶特异性结合的核酸分子。所述靶包含但不限于蛋白质、核酸、小分子化合物等。所述核酸分子包括但不限于适配子(aptamer)、反义核酸(反义RNA)、dsRNA(如siRNA、shRNA),优选是适配子。As used herein, a "target-specific nucleic acid molecule" refers to a nucleic acid molecule that is capable of specifically binding to a particular target. The targets include but are not limited to proteins, nucleic acids, small molecule compounds, etc. The nucleic acid molecules include but are not limited to aptamers, antisense nucleic acids (antisense RNA), dsRNA (such as siRNA, shRNA), preferably aptamers.
在一些实施方案中,与相应的对照核酸分子相比,所述经修饰的靶特异性核酸分子与靶的结合亲和力提高至少约10%、至少约20%、至少约30%、至少约40%、至少约50%、至少约60%、至少约70%、至少约80%、至少约90%、至少约100%或更多。在一些实施方案中,所述经修饰的靶特异性核酸分子与靶的结合亲和力是未修饰的相应核酸分子的亲和力的至少约1.5倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约15倍、至少约16倍、至少约20倍、至少约25倍、至少约30倍、至少约40倍、至少约50倍、至少约75倍、至少约100倍或更多倍。所述结合亲和力例如可以通过酶联寡核苷酸测定法(ELONA)测定或通过生物膜干涉法(Biolayer interferometry,BLI)测定。In some embodiments, the modified target-specific nucleic acid molecule has an increased binding affinity to the target by at least about 10%, at least about 20%, at least about 30%, at least about 40% compared to a corresponding control nucleic acid molecule. , at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100% or more. In some embodiments, the modified target-specific nucleic acid molecule binds the target with an affinity that is at least about 1.5 times, at least about 2 times, at least about 3 times, at least about 4 times greater than the affinity of the corresponding unmodified nucleic acid molecule. , at least about 5 times, at least about 6 times, at least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, at least about 15 times, at least about 16 times, at least about 20 times, at least about 25 times , at least about 30 times, at least about 40 times, at least about 50 times, at least about 75 times, at least about 100 times or more. The binding affinity can be measured, for example, by enzyme-linked oligonucleotide assay (ELONA) or by biofilm interferometry (BLI).
在一些实施方案中,所述经修饰的靶特异性核酸分子对靶具有小于100nM,优选小于50nM,优选小于40nM,优选小于30nM,优选小于20nM,优选小于10nM,优选小于5nM或更小的KD(解离常数)。所述KD例如通过酶联寡核苷酸测定法(ELONA)测定或通过生物膜干涉法(Biolayer interferometry,BLI)测定。In some embodiments, the modified target-specific nucleic acid molecule has a KD for the target of less than 100 nM, preferably less than 50 nM, preferably less than 40 nM, preferably less than 30 nM, preferably less than 20 nM, preferably less than 10 nM, preferably less than 5 nM or less. (dissociation constant). The KD is measured, for example, by enzyme-linked oligonucleotide assay (ELONA) or by biofilm interferometry (BLI).
在一些实施方案中,所述喹啉修饰是喹啉-2-基取代、喹啉-3-基取代、喹啉-4-基取代、喹啉-5-基取代、喹啉-6-基取代、喹啉-7-基取代或喹啉-8-基取代。在一些优选实施方案中,所述喹啉修饰是喹啉-5-基取代。喹啉上的取代位置编号如下式所示:
In some embodiments, the quinoline modification is quinolin-2-yl substitution, quinolin-3-yl substitution, quinolin-4-yl substitution, quinolin-5-yl substitution, quinolin-6-yl substitution Substituted, quinolin-7-yl substituted or quinolin-8-yl substituted. In some preferred embodiments, the quinoline modification is a quinolin-5-yl substitution. The substitution position numbers on quinoline are as follows:
在一些实施方案中,所述喹啉修饰的核苷酸包含取代基团R,所述R选自以下a-g:
In some embodiments, the quinoline modified nucleotide comprises a substituent group R selected from the following ag:
在一些优选实施方案中, In some preferred embodiments,
在一些实施方案中,所述喹啉修饰的核苷酸在其碱基部分包含所述取代基团R。在一些实施方案中,所述喹啉修饰的核苷酸衍生自尿嘧啶核苷,例如脱氧尿嘧啶核苷。In some embodiments, the quinoline modified nucleotide contains the substituent group R in the base portion thereof. In some embodiments, the quinoline modified nucleotide is derived from uridine, such as deoxyuridine.
在一些实施方案中,所述喹啉修饰的核苷酸包括以下式I的碱基部分:
In some embodiments, the quinoline modified nucleotide includes the base moiety of Formula I:
其中
in
在一些优选实施方案中, In some preferred embodiments,
在一些实施方案中,所述喹啉修饰的核苷酸包括以下式II的结构:
In some embodiments, the quinoline modified nucleotides comprise the structure of Formula II:
其中
in
在一些优选实施方案中, In some preferred embodiments,
在一些实施方案中,所述靶特异性核酸分子中的一或多个核苷酸(天然核苷酸)被所述喹啉修饰的核苷酸取代。In some embodiments, one or more nucleotides (natural nucleotides) in the target-specific nucleic acid molecule are replaced with the quinoline-modified nucleotides.
在一些实施方案中,所述靶特异性核酸分子还可以包含一或多种延长其体内半衰期修饰。In some embodiments, the target-specific nucleic acid molecule may also comprise one or more modifications that extend its half-life in vivo.
所述延长其体内半衰期修饰包括例如3’和5’的修饰,如3’和5’加帽。在一些实施方案中,所述适体在3’末端用反向脱氧胸苷加帽,即3’反向脱氧胸苷(3’idT)修饰。The modifications that extend its half-life in vivo include, for example, 3' and 5' modifications, such as 3' and 5' capping. In some embodiments, the aptamer is capped at the 3' end with inverted deoxythymidine, i.e., 3' inverted deoxythymidine (3'idT) modification.
所述延长其体内半衰期的修饰还可以包括用经修饰的核苷酸取代一或多个天然存在的核苷酸。例如,所述用于延长半衰期的经修饰的核苷酸包括但不限于2’-氟、2’-甲氧乙基、2’-甲氧基和/或2’丙烯氧基修饰的核苷酸(即核糖的2’位置羟基被氟、甲氧乙基、甲氧基或丙烯氧基等取代)。所述用于延长半衰期的经修饰的核苷酸还可以包括C-5修饰的嘧啶。术语“C-5修饰的嘧啶”是指C-5位上有修饰的嘧啶。C-5修饰的嘧啶能够增强寡核苷酸的核酸酶抗性,且是本领域已知的,例如可以参见国际专利申请WO 2011/130195及其引用的文献。The modification to extend its half-life in vivo may also include replacing one or more naturally occurring nucleotides with modified nucleotides. For example, the modified nucleotides for extending half-life include, but are not limited to, 2'-fluoro, 2'-methoxyethyl, 2'-methoxy and/or 2'propenoxy modified nucleosides Acid (that is, the hydroxyl group at the 2' position of ribose is replaced by fluorine, methoxyethyl, methoxy or propyleneoxy, etc.). The modified nucleotides for extending half-life may also include C-5 modified pyrimidines. The term "C-5 modified pyrimidine" refers to a pyrimidine with a modification at the C-5 position. C-5 modified pyrimidines can enhance the nuclease resistance of oligonucleotides and are known in the art. For example, see International Patent Application WO 2011/130195 and its cited documents.
所述用于延长半衰期的修饰还包括核苷酸间的修饰,例如具有不带电荷的键(例如甲基膦酸酯、磷酸三酯、磷酸胺酯、氨基甲酸酯等)的核苷酸间修饰和具有带电荷的键(例如硫代磷酸酯、二硫代磷酸酯等)的核苷酸间修饰,有嵌入剂(例如吖啶、补骨脂素等)的核苷酸间修饰,含有螯合剂(例如金属、放射性金属、硼、氧化性金属等)的核苷酸间修饰,含有烷化剂的核苷酸间修饰和有修饰的键(例如阿尔法异头核酸等)的核苷酸间修饰。Modifications for extending half-life also include modifications between nucleotides, such as nucleotides with uncharged bonds (e.g., methylphosphonate, phosphotriester, phosphate amine ester, carbamate, etc.) Inter-modifications and inter-nucleotide modifications with charged bonds (e.g. phosphorothioates, phosphorodithioates, etc.), inter-nucleotide modifications with intercalating agents (e.g. acridine, psoralen, etc.), Internucleotide modifications containing chelating agents (such as metals, radioactive metals, boron, oxidizing metals, etc.), internucleotide modifications containing alkylating agents, and nucleosides with modified bonds (such as alpha anomeric nucleic acids, etc.) Inter-acid modification.
所述用于延长半衰期的修饰还可以包括聚乙二醇化修饰(PEG修饰)。The modification for extending half-life may also include pegylation modification (PEG modification).
在另一方面,本发明还提供在一种组合物如药物组合物,其包括本发明的经修饰的靶特异性核酸分子。在一些实施方案中,所述组合物还包含药学上或生理上可接受的载体或赋形剂。所述组合物可以例如用于检测所述靶,或用于诊断和/或治疗所述靶相关的疾病。In another aspect, the invention also provides a composition, such as a pharmaceutical composition, comprising a modified target-specific nucleic acid molecule of the invention. In some embodiments, the composition further includes a pharmaceutically or physiologically acceptable carrier or excipient. The composition may, for example, be used to detect the target, or to diagnose and/or treat a disease associated with the target.
在另一方面,本发明提供一种提高靶特异性核酸分子与靶的结合亲和力的方法,所述方法包括用一或多个喹啉修饰的核苷酸取代所述靶特异性核酸分子中的一个或多个天然存在的核苷酸。所述喹啉修饰的核苷酸如上文所定义。In another aspect, the invention provides a method for increasing the binding affinity of a target-specific nucleic acid molecule to a target, the method comprising replacing the nucleotides in the target-specific nucleic acid molecule with one or more quinoline-modified nucleotides. One or more naturally occurring nucleotides. The quinoline modified nucleotide is as defined above.
在一些实施方案中,所述核苷酸取代在化学合成(如固相)所述靶特异性核酸分子期间引入。In some embodiments, the nucleotide substitutions are introduced during chemical synthesis (eg, solid phase) of the target-specific nucleic acid molecule.
在一些实施方案中,在化学合成所述靶特异性核酸分子期间加入选自以下2a-2g的经修饰的亚磷酰胺单体:
In some embodiments, a modified phosphoramidite monomer selected from 2a-2g below is added during chemical synthesis of the target-specific nucleic acid molecule:
在另一方面,本发明还提供选自以下2a-2g的经修饰的亚磷酰胺单体,其用于提高靶特异性核酸分子与靶的结合亲和力:
In another aspect, the present invention also provides modified phosphoramidite monomers selected from the following 2a-2g, which are used to improve the binding affinity of target-specific nucleic acid molecules to targets:
II.结合亲和力提高的经修饰的骨硬化素特异性核酸分子II. Modified osteosclerostin-specific nucleic acid molecules with increased binding affinity
在本发明的一些具体实施方案中,所述靶特异性核酸分子是骨硬化素特异性核酸分子。因此,本发明也提供经修饰的骨硬化素特异性核酸分子,其包含一或多个上文所述喹啉修饰的核苷酸。In some embodiments of the invention, the target-specific nucleic acid molecule is an osteosclerostin-specific nucleic acid molecule. Accordingly, the present invention also provides modified osteosclerostin-specific nucleic acid molecules comprising one or more quinoline-modified nucleotides as described above.
在一些实施方案中,所述靶是骨硬化素(sclerostin)如人骨硬化素。示例性人骨硬化素包含氨基酸序列:
In some embodiments, the target is sclerostin, such as human sclerostin. An exemplary human osteosclerostin contains the amino acid sequence:
在一些实施方案中,所述靶特异性核酸分子是与骨硬化素特异性结合的适配子(经修饰的骨硬化素特异性适配子)。在一些实施方案中,所述与骨硬化素特异性结合的适配子(未修饰的)包含核苷酸序列:In some embodiments, the target-specific nucleic acid molecule is an aptamer that specifically binds osteosclerostin (modified osteosclerostin-specific aptamer). In some embodiments, the aptamer (unmodified) that specifically binds osteosclerostin comprises the nucleotide sequence:
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID  NO:2)。5'-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3'(SEQ ID NO:2).
在一些实施方案中,所述经修饰的骨硬化素特异性适配子在相应于SEQ ID NO:2的选自以下的一或多个位置的核苷酸被所述喹啉修饰的核苷酸取代:第1位、第2位、第3位、第4位、第5位、第6位、第8位、第11位、第13位、第15位、第29位、第30位、第31位、第32位、第33位、第34位、第35位、第36位、第37位、第38位、第39位和第40位。In some embodiments, the modified osteosclerostin-specific aptamer is a nucleoside modified by the quinoline at one or more positions of a nucleotide corresponding to SEQ ID NO: 2 selected from: Acid substitution: 1st, 2nd, 3rd, 4th, 5th, 6th, 8th, 11th, 13th, 15th, 29th, 30th , 31st, 32nd, 33rd, 34th, 35th, 36th, 37th, 38th, 39th and 40th.
在一些优选实施方案中,所述经修饰的骨硬化素特异性核酸分子在相应于SEQ ID NO:2的第6位、第8位、第11位、第13位、第32位和/或第34位的核苷酸被所述喹啉修饰的核苷酸取代。In some preferred embodiments, the modified osteosclerostin-specific nucleic acid molecule is at position 6, position 8, position 11, position 13, position 32 and/or corresponding to SEQ ID NO:2 The nucleotide at position 34 is substituted with the quinoline modified nucleotide.
在一些实施方案中,所述经修饰的骨硬化素特异性核酸分子包含选自以下的核苷酸序列:In some embodiments, the modified osteosclerostin-specific nucleic acid molecule comprises a nucleotide sequence selected from:
5’-xGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:3),5’-xGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:3),
5’-CxGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:4),5’-CxGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:4),
5’-CGxGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:5),5’-CGxGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:5),
5’-CGGxGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:6),5’-CGGxGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:6),
5’-CGGGxTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:7),5’-CGGGxTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:7),
5’-CGGGGxGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:8),5’-CGGGGxGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:8),
5’-CGGGGTGxGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:9),5’-CGGGGTGxGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:9),
5’-CGGGGTGTGGxTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:10),5’-CGGGGTGTGGxTTCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:10),
5’-CGGGGTGTGGGTxCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:11),5’-CGGGGTGTGGGTxCGTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:11),
5’-CGGGGTGTGGGTTCxTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:12),5’-CGGGGTGTGGGTTCxTCGTTAGCTTGATTTGGCAGCTGCC-3’(SEQ ID NO:12),
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATxTGGCAGCTGCC-3’(SEQ ID NO:13),5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATxTGGCAGCTGCC-3’(SEQ ID NO:13),
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTxGGCAGCTGCC-3’(SEQ ID NO:14),5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTxGGCAGCTGCC-3’(SEQ ID NO:14),
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTxGCAGCTGCC-3’(SEQ ID  NO:15),5'-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTxGCAGCTGCC-3'(SEQ ID NO:15),
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGxCAGCTGCC-3’(SEQ ID NO:16),5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGxCAGCTGCC-3’(SEQ ID NO:16),
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGxAGCTGCC-3’(SEQ ID NO:17),5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGxAGCTGCC-3’(SEQ ID NO:17),
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCxGCTGCC-3’(SEQ ID NO:18),5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCxGCTGCC-3’(SEQ ID NO:18),
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAxCTGCC-3’(SEQ ID NO:19),5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAxCTGCC-3’(SEQ ID NO:19),
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGxTGCC-3’(SEQ ID NO:20),5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGxTGCC-3’(SEQ ID NO:20),
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCxGCC-3’(SEQ ID NO:21),5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCxGCC-3’(SEQ ID NO:21),
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTxCC-3’(SEQ ID NO:22),5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTxCC-3’(SEQ ID NO:22),
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGxC-3’(SEQ ID NO:23),和5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGxC-3’ (SEQ ID NO: 23), and
5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCx-3’(SEQ ID NO:24),5’-CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCx-3’(SEQ ID NO:24),
其中x为所述喹啉修饰的核苷酸。Where x is the quinoline modified nucleotide.
在一些实施方案中,所述经修饰的骨硬化素特异性核酸分子包含SEQ ID NO:8的核苷酸序列,其中x为所述喹啉修饰的核苷酸。在一些实施方案中,所述经修饰的骨硬化素特异性核酸分子包含SEQ ID NO:10的核苷酸序列,其中x为所述喹啉修饰的核苷酸。在一些实施方案中,所述经修饰的骨硬化素特异性核酸分子包含SEQ ID NO:11的核苷酸序列,其中x为所述喹啉修饰的核苷酸。在一些实施方案中,所述经修饰的骨硬化素特异性核酸分子包含SEQ ID NO:16的核苷酸序列,其中x为所述喹啉修饰的核苷酸。在一些实施方案中,所述经修饰的骨硬化素特异性核酸分子包含SEQ ID NO:18的核苷酸序列,其中x为所述喹啉修饰的核苷酸。In some embodiments, the modified osteosclerostin-specific nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 8, wherein x is the quinoline modified nucleotide. In some embodiments, the modified osteosclerostin-specific nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 10, wherein x is the quinoline modified nucleotide. In some embodiments, the modified osteosclerostin-specific nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 11, wherein x is the quinoline modified nucleotide. In some embodiments, the modified osteosclerostin-specific nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 16, wherein x is the quinoline modified nucleotide. In some embodiments, the modified osteosclerostin-specific nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 18, wherein x is the quinoline modified nucleotide.
在一些优选实施方案中,所述喹啉修饰的核苷酸包含下式II的结构:
In some preferred embodiments, the quinoline modified nucleotides comprise the structure of Formula II:
其中, in,
在一些实施方案,本发明的所述经修饰的骨硬化素特异性核酸分子对骨硬化素的结合亲和力是SEQ ID NO:2所示的未经修饰的骨硬化素特异性核酸分子的亲和力的至少约1.5倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约15倍、至少约16倍或更多倍。所述结合亲和力例如可以通过酶联寡核苷酸测定法(ELONA)测定或通过生物膜干涉法(Biolayer interferometry,BLI)测定。In some embodiments, the binding affinity of the modified osteosclerostin-specific nucleic acid molecule of the present invention to osteosclerosis is the affinity of the unmodified osteosclerostin-specific nucleic acid molecule shown in SEQ ID NO: 2 At least about 1.5 times, at least about 2 times, at least about 3 times, at least about 4 times, at least about 5 times, at least about 6 times, at least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, At least about 15 times, at least about 16 times or more. The binding affinity can be measured, for example, by enzyme-linked oligonucleotide assay (ELONA) or by biofilm interferometry (BLI).
在一些实施方案中,所述经修饰的骨硬化素特异性核酸分子对骨硬化素具有小于10nM或甚至小于5nM的KD(解离常数)。所述KD例如通过酶联寡核苷酸测定法(ELONA)测定或通过生物膜干涉法(Biolayer interferometry,BLI)测定。In some embodiments, the modified osteosclerostin-specific nucleic acid molecule has a KD (dissociation constant) for osteosclerosis of less than 10 nM or even less than 5 nM. The KD is measured, for example, by enzyme-linked oligonucleotide assay (ELONA) or by biofilm interferometry (BLI).
在另一方面,本发明还提供一种组合物,其包含本发明的经修饰的骨硬化素特异性核酸分子,以及任选的药学上或生理上可接受的载体。所述组合物例如用于检测骨硬化素,或用于诊断和/或治疗骨硬化素相关的疾病。In another aspect, the present invention also provides a composition comprising the modified osteosclerostin-specific nucleic acid molecule of the present invention, and optionally a pharmaceutically or physiologically acceptable carrier. The composition is used, for example, for detecting osteosclerostin, or for diagnosing and/or treating osteosclerostin-related diseases.
在另一方面,本发明还提供通过本发明的经修饰的骨硬化素特异性核酸分子治疗疾病的方法,该方法包括给有需要的对象施用治疗有效量的本发明的经修饰的骨硬化素特异性核酸分子。In another aspect, the invention also provides a method of treating a disease by a modified osteosclerostin-specific nucleic acid molecule of the invention, the method comprising administering to a subject in need thereof a therapeutically effective amount of a modified osteosclerostin of the invention Specific nucleic acid molecules.
所述疾病例如是骨硬化素相关疾病,例如骨硬化素介导的疾病。The disease is, for example, an osteosclerostin-related disease, such as an osteosclerostin-mediated disease.
如本文所使用,“骨硬化素相关疾病”包括其中骨矿质密度(BMD)相对于健康对象不正常和/或病理上低的病症。由低BMD和/或骨易碎性表征的疾病包括但不限于:原发性和继发性骨质疏松症、骨质减少、骨软化、成骨不全(OI)、缺血性坏死(骨坏死)、骨折和植入物愈合(牙种植体和髋植入物)、由于其它病症的骨丧失(例如,与HIV感染、癌症和关节炎相关)。其它“骨硬化素相关疾病”包括但不限于:低磷佝偻病、类风湿性关节炎、骨关节炎、关节炎,以及溶骨性病变。As used herein, "osteosclerostin-related disease" includes conditions in which bone mineral density (BMD) is abnormally and/or pathologically low relative to healthy subjects. Diseases characterized by low BMD and/or bone fragility include, but are not limited to: primary and secondary osteoporosis, osteopenia, osteomalacia, osteogenesis imperfecta (OI), avascular necrosis (bone necrosis), fractures and implant healing (dental implants and hip implants), bone loss due to other conditions (eg, associated with HIV infection, cancer, and arthritis). Other "osteosclerostin-related diseases" include, but are not limited to: hypophosphatemic rickets, rheumatoid arthritis, osteoarthritis, arthritis, and osteolytic lesions.
如本文所使用,“骨硬化素相关疾病”包括骨硬化素相关癌症,例如骨髓瘤(例如,伴随溶骨性病变的多发性骨髓瘤)、乳腺癌(例如三阴性乳腺癌)、结肠癌、黑色素瘤、肝细胞癌、上皮癌、食道癌、脑癌、肺癌、前列腺癌或胰癌,及其任何转移瘤。As used herein, "osteosclerostin-related disease" includes osteosclerosis-related cancers, such as myeloma (eg, multiple myeloma with osteolytic lesions), breast cancer (eg, triple-negative breast cancer), colon cancer, Melanoma, hepatocellular carcinoma, epithelial cancer, esophageal cancer, brain cancer, lung cancer, prostate cancer or pancreatic cancer, and any metastases thereto.
“骨硬化素相关疾病”还可包括至少由骨硬化素在肾脏和在心血管中表达而引起的肾病及心血管疾病。所述病症包括但不限于诸如以下的肾病:肾小球疾病(例如,急性和慢性肾小球性肾炎,急进性肾小球性肾炎,肾病综合征,局灶增生性肾小球肾炎,与诸如系统性红斑狼疮、古德帕斯彻氏综合征、多发性骨髓瘤、糖尿病、多囊肾病、瘤形成、镰刀形红细胞病、以及慢性炎症等全身疾病相关的肾小球损害)、肾小管疾病(例如,急性肾小管坏死和急性肾衰竭、多囊肾病、髓质海绵肾、髓质囊性病、肾原性糖尿病,以及肾小管性酸中毒)、小管间质性疾病(例如,肾盂肾炎、药和毒素诱导的小管间质性肾炎、高钙性肾病,以及低钾血症性肾病)、急性和急进性肾衰竭、慢性肾衰竭、肾结 石、痛风、血管疾病(例如,高血压和肾硬化、微血管病性溶血性贫血、动脉粥样硬化栓塞肾病、扩散皮层的坏死以及肾梗塞),或肿瘤(例如,肾细胞癌和肾胚细胞瘤)。"Osteosclerin-related diseases" may also include renal and cardiovascular diseases caused by at least expression of osteosclerostin in the kidneys and in the cardiovascular system. Such conditions include, but are not limited to, renal diseases such as: glomerular diseases (e.g., acute and chronic glomerulonephritis, rapidly progressive glomerulonephritis, nephrotic syndrome, focal proliferative glomerulonephritis, and Glomerular damage associated with systemic diseases such as systemic lupus erythematosus, Goodpasture's syndrome, multiple myeloma, diabetes, polycystic kidney disease, neoplasia, sickle cell disease, and chronic inflammation), renal tubules Diseases (e.g., acute tubular necrosis and acute renal failure, polycystic kidney disease, medullary sponge kidney disease, medullary cystic disease, nephrogenic diabetes, and tubular acidosis), tubulointerstitial disease (e.g., pyelonephritis , drug- and toxin-induced tubulointerstitial nephritis, hypercalcaemic nephropathy, and hypokalemic nephropathy), acute and rapidly progressive renal failure, chronic renal failure, renal tuberculosis stones, gout, vascular disease (eg, hypertension and nephrosclerosis, microangiopathic hemolytic anemia, atheroembolic nephropathy, diffuse cortical necrosis, and renal infarction), or tumors (eg, renal cell carcinoma and nephroblastoma) tumor).
所述骨硬化素相关疾病还包括但不限于诸如以下的心血管疾病:缺血性心脏病(例如,心绞痛、心肌梗死,以及慢性缺血性心脏病)、高血压心脏病、肺心病、心脏瓣膜病(例如,风湿热和风湿性心脏病、心内膜炎、二尖瓣脱垂以及主动脉瓣狭窄)、先天性心脏病(例如,瓣和血管堵塞性损害、房或室中隔缺损、以及久存性动脉导管),或心肌病(例如,心肌炎、充血型心肌病,以及肥厚型心肌病)。The osteosclerostin-related diseases also include, but are not limited to, cardiovascular diseases such as: ischemic heart disease (eg, angina pectoris, myocardial infarction, and chronic ischemic heart disease), hypertensive heart disease, cor pulmonale, heart disease, Valvular disease (eg, rheumatic fever and rheumatic heart disease, endocarditis, mitral valve prolapse, and aortic stenosis), congenital heart disease (eg, occlusive lesions of valves and blood vessels, atrial or ventricular septal defects , and persistent ductus arteriosus), or cardiomyopathies (eg, myocarditis, congestive cardiomyopathy, and hypertrophic cardiomyopathy).
如本文所用,“治疗”患有疾病的对象表示所述对象的症状部分或全部缓解,或者在治疗后保持不变。因此,治疗包括预防、治疗和/或治愈。预防指防止潜在疾病和/或防止症状恶化或疾病发展。As used herein, "treating" a subject suffering from a disease means that the subject's symptoms are partially or completely alleviated, or remain unchanged following treatment. Treatment therefore includes prevention, treatment and/or cure. Prevention means preventing underlying disease and/or preventing symptoms from worsening or disease progression.
如本文所用,“治疗有效量”或“治疗有效剂量”指施用于对象之后至少足以产生疗效的物质、化合物、材料或包含化合物的组合物的量。因此,其为防止、治愈、改善、阻滞或部分阻滞疾病或病症的症状所必需的量。如本文所用,“疗效”表示由对象的治疗所导致的效果,其改变、通常改良或改善疾病或疾病状况的症状,或者治愈疾病或疾病状况。As used herein, a "therapeutically effective amount" or "therapeutically effective dose" refers to an amount of a substance, compound, material or composition containing a compound that is at least sufficient to produce a therapeutic effect upon administration to a subject. Thus, it is an amount necessary to prevent, cure, ameliorate, block or partially block the symptoms of a disease or condition. As used herein, "therapeutic effect" means an effect resulting from treatment of a subject that alters, generally ameliorates, or ameliorates the symptoms of a disease or disease condition, or cures a disease or disease condition.
按照多种因素选择利用所述核酸分子的剂量方案,所述因素包括例如,所述患者的类型、种类、年龄、体重、性别和医疗病症;所要治疗的病症的严重性;施用途径;所述患者的肾与肝的功能;和所使用的特定的核酸分子或其盐。普通熟练的医生可以容易地确定并指定预防、对抗或抑制所述病症进展所需要的组合物的有效量。Dosage regimens utilizing the nucleic acid molecules are selected based on a variety of factors, including, for example, the type, species, age, weight, gender, and medical condition of the patient; the severity of the condition to be treated; the route of administration; the The patient's kidney and liver function; and the specific nucleic acid molecule or salt thereof used. The ordinarily skilled physician can readily determine and prescribe the effective amount of the composition required to prevent, combat, or inhibit the progression of the disorder.
实施例Example
实施例1、化合物1a~1g和2a~2g的合成路线Example 1, synthetic routes of compounds 1a to 1g and 2a to 2g
1)不同喹啉亚型修饰核苷(1a~1g)结构如下:
1) The structures of different quinoline subtype modified nucleosides (1a~1g) are as follows:
2)化合物1a~1g与磷试剂反应后得到不同喹啉亚型修饰亚磷酰胺单体(2a~2g),可作为固相合成原料,2a~2g的结构如下:
2) Compounds 1a to 1g react with phosphorus reagents to obtain different quinoline subtype modified phosphoramidite monomers (2a to 2g), which can be used as raw materials for solid phase synthesis. The structures of 2a to 2g are as follows:
3)1a~1g和2a~2g的合成路线:
3) Synthetic routes of 1a~1g and 2a~2g:
实验部分:Experimental part:
1a~1g的合成:向100mL高压釜中加入5'-DMT-5-碘脱氧尿嘧啶(2.0g,3.0mmol)、氨基甲基喹啉(0.95g,6.0mmol)和四(三苯基膦)钯(35mg,Pb 10%)。随后加入20mL无水THF以溶解上述反应物,加入Et3N(2.1mL,15mmol)。盖上釜盖,打开一氧化碳钢瓶减压阀。将剩余空间用一氧化碳置换3次,然后在0.30-0.40MPa压力下用一氧化碳填充。将反应釜置于60-70℃油浴中,搅拌3-4天。在此过程中,如果釜内压力降至0.30MPa以下,则需要补充一氧化碳气体以保持压力在0.30-0.40MPa之间。将反应釜置于冰水中20分钟,然后在通风橱中释放压力。打开釜,将反应液倒入100ml冰水中,用50ml乙酸乙酯萃取3次。用无水Na2SO4干燥有机相,并真空浓缩。接下来,使用中性二氧化硅纯化最终产品,洗脱液为正己烷/乙酸乙酯=1/2。浓缩蒸发至干后,得到粘性固体产物(收率:1a:66%,1b:26%,1c:65%,1d:72%,1e:60%,1f:68%,1g:53%)。Synthesis of 1a ~ 1g: Add 5'-DMT-5-iododeoxyuracil (2.0g, 3.0mmol), aminomethylquinoline (0.95g, 6.0mmol) and tetrakis(triphenylphosphine) to a 100mL autoclave )Palladium (35mg, Pb 10%). Then 20 mL of anhydrous THF was added to dissolve the above reactants, and Et3N (2.1 mL, 15 mmol) was added. Cover the kettle lid and open the pressure reducing valve of the carbon monoxide cylinder. Replace the remaining space with carbon monoxide three times, and then fill it with carbon monoxide under a pressure of 0.30-0.40MPa. Place the reaction kettle in an oil bath at 60-70°C and stir for 3-4 days. During this process, if the pressure in the kettle drops below 0.30MPa, carbon monoxide gas needs to be added to maintain the pressure between 0.30-0.40MPa. Place the kettle in ice water for 20 minutes and then release the pressure in a fume hood. Open the kettle, pour the reaction solution into 100 ml of ice water, and extract three times with 50 ml of ethyl acetate. The organic phase was dried over anhydrous Na2SO4 and concentrated in vacuo. Next, the final product is purified using neutral silica, and the eluent is n-hexane/ethyl acetate = 1/2. After concentration and evaporation to dryness, a viscous solid product was obtained (yield: 1a: 66%, 1b: 26%, 1c: 65%, 1d: 72%, 1e: 60%, 1f: 68%, 1g: 53%).
核磁数据如下:The NMR data are as follows:
1a:1H NMR(400MHz,DMSO)δ11.98(s,1H),9.18(t,J=5.8Hz,1H),8.89(d,J=2.8Hz,1H),8.58(d,J=8.5Hz,1H),8.54(s,1H),7.96(d,J=8.4Hz,1H),7.73–7.64(m,1H),7.56(d,J=7.0Hz,1H),7.49– 7.34(m,3H),7.23(tt,J=14.4,7.4Hz,7H),6.87(d,J=8.7Hz,4H),6.08(t,J=6.3Hz,1H),5.35(d,J=4.3Hz,1H),5.07–4.89(m,2H),4.11(s,1H),3.97(d,J=4.0Hz,1H),3.68(d,J=3.7Hz,6H),3.20(d,J=15.7Hz,2H),2.39–2.14(m,2H).13C NMR(101MHz,DMSO)δ163.7,161.9,158.5,150.7,149.9,148.5,146.2,145.3,136.0,135.8,132.4,130.3,130.2,129.5,129.1,128.3,128.1,127.1,126.4,121.8,113.7,105.4,86.7,86.5,86.3,70.9,64.1,55.4,39.93.1a: 1 H NMR (400MHz, DMSO) δ11.98 (s, 1H), 9.18 (t, J = 5.8Hz, 1H), 8.89 (d, J = 2.8Hz, 1H), 8.58 (d, J = 8.5 Hz,1H),8.54(s,1H),7.96(d,J=8.4Hz,1H),7.73–7.64(m,1H),7.56(d,J=7.0Hz,1H),7.49– 7.34(m,3H),7.23(tt,J=14.4,7.4Hz,7H),6.87(d,J=8.7Hz,4H),6.08(t,J=6.3Hz,1H),5.35(d,J =4.3Hz,1H),5.07–4.89(m,2H),4.11(s,1H),3.97(d,J=4.0Hz,1H),3.68(d,J=3.7Hz,6H),3.20(d ,J=15.7Hz,2H),2.39–2.14(m,2H). 13 C NMR(101MHz,DMSO)δ163.7,161.9,158.5,150.7,149.9,148.5,146.2,145.3,136.0,135.8,132.4,130.3, 130.2,129.5,129.1,128.3,128.1,127.1,126.4,121.8,113.7,105.4,86.7,86.5,86.3,70.9,64.1,55.4,39.93.
1b:1H NMR(400MHz,DMSO)δ12.02(s,1H),9.27(t,J=6.0Hz,1H),8.86(dd,J=4.1,1.6Hz,1H),8.54(s,1H),8.21(d,J=7.8Hz,1H),7.97(d,J=8.7Hz,1H),7.81(s,1H),7.70(dd,J=8.7,1.7Hz,1H),7.48(dd,J=8.3,4.2Hz,1H),7.38(d,J=7.5Hz,2H),7.27(dd,J=8.7,4.3Hz,6H),7.13(t,J=7.3Hz,1H),6.87(dd,J=8.8,1.8Hz,4H),6.11(t,J=6.4Hz,1H),5.36(d,J=4.1Hz,1H),4.83–4.59(m,2H),4.11(s,1H),4.01–3.92(m,1H),3.68(s,6H),3.22(s,2H),2.37–2.18(m,2H).13C NMR(101MHz,DMSO)δ163.7,162.2,158.5,150.7,149.9,147.5,146.2,145.4,138.1,136.2,136.0,135.8,130.3,130.2,129.9,129.5,128.3,128.2,128.1,127.1,126.0,122.1,113.7,105.6,86.6,86.5,86.3,70.9,64.1,55.4,42.6.1c:1H NMR(400MHz,DMSO)δ12.01(s,1H),9.29(t,J=6.1Hz,1H),8.88(dd,J=4.2,1.7Hz,1H),8.52(s,1H),8.33(d,J=8.2Hz,1H),7.92(d,J=8.7Hz,2H),7.55(dd,J=8.4,1.5Hz,1H),7.49(dd,J=8.3,4.2Hz,1H),7.37(d,J=7.5Hz,2H),7.33–7.21(m,6H),7.15(t,J=7.3Hz,1H),6.87(d,J=8.9Hz,4H),6.10(t,J=6.4Hz,1H),5.36(d,J=4.6Hz,1H),4.81–4.64(m,2H),4.12(dt,J=8.9,4.4Hz,1H),3.95(dd,J=8.8,4.4Hz,1H),3.69(d,J=3.4Hz,6H),3.21(t,J=7.9Hz,2H),2.37–2.16(m,2H).13C NMR(101MHz,DMSO)δ163.7,162.2,158.5,151.2,149.9,146.2,145.3,141.4,139.6,136.2,136.0,135.8,130.3,130.2,128.6,128.3,128.1,127.3,127.1,126.8,121.6,113.7,105.6,86.5,86.4,86.2,70.8,64.1,55.4,42.7.1b: 1 H NMR (400MHz, DMSO) δ12.02(s,1H),9.27(t,J=6.0Hz,1H),8.86(dd,J=4.1,1.6Hz,1H),8.54(s,1H ),8.21(d,J=7.8Hz,1H),7.97(d,J=8.7Hz,1H),7.81(s,1H),7.70(dd,J=8.7,1.7Hz,1H),7.48(dd ,J=8.3,4.2Hz,1H),7.38(d,J=7.5Hz,2H),7.27(dd,J=8.7,4.3Hz,6H),7.13(t,J=7.3Hz,1H),6.87 (dd,J=8.8,1.8Hz,4H),6.11(t,J=6.4Hz,1H),5.36(d,J=4.1Hz,1H),4.83–4.59(m,2H),4.11(s, 1H),4.01–3.92(m,1H),3.68(s,6H),3.22(s,2H),2.37–2.18(m,2H). 13 C NMR(101MHz,DMSO)δ163.7,162.2,158.5,150.7 ,149.9,147.5,146.2,145.4,138.1,136.2,136.0,135.8,130.3,130.2,129.9,129.5,128.3,128.2,128.1,127.1,126.0,122.1,113.7,105.6,86 .6,86.5,86.3,70.9,64.1 ,55.4,42.6.1c: 1 H NMR(400MHz,DMSO)δ12.01(s,1H),9.29(t,J=6.1Hz,1H),8.88(dd,J=4.2,1.7Hz,1H), 8.52(s,1H),8.33(d,J=8.2Hz,1H),7.92(d,J=8.7Hz,2H),7.55(dd,J=8.4,1.5Hz,1H),7.49(dd,J =8.3,4.2Hz,1H),7.37(d,J=7.5Hz,2H),7.33–7.21(m,6H),7.15(t,J=7.3Hz,1H),6.87(d,J=8.9Hz ,4H),6.10(t,J=6.4Hz,1H),5.36(d,J=4.6Hz,1H),4.81–4.64(m,2H),4.12(dt,J=8.9,4.4Hz,1H) 13 C NMR (101MHz, DMSO) δ163.7,162.2,158.5,151.2,149.9,146.2,145.3,141.4,139.6,136.2,136.0,135.8,130.3,130.2,128.6,128.3,128.1,127.3,127. 1,126.8,121.6,113.7, 105.6,86.5,86.4,86.2,70.8,64.1,55.4,42.7.
1d:1H NMR(400MHz,DMSO)δ12.04(s,1H),9.29(t,J=5.9Hz,1H),8.77(d,J=4.4Hz,1H),8.54(s,1H),8.19(d,J=8.3Hz,1H),8.06(d,J=8.4Hz,1H),7.77(t,J=7.6Hz,1H),7.61(t,J=7.5Hz,1H),7.37(d,J=7.7Hz,2H),7.32(d,J=4.3Hz,1H),7.26(dd,J=11.1,6.7Hz,6H),7.16(t,J=7.2Hz,1H),6.85(d,J=8.5Hz,4H),6.10(t,J=6.3Hz,1H),5.36(d,J=4.4Hz,1H),5.09–4.94(m,2H),4.11(d,J=4.3Hz,1H),3.99–3.93(m,1H),3.65(d,J=4.7Hz,6H),3.20(d,J=4.0Hz,2H),2.38–2.20(m,2H).13C NMR(101MHz,DMSO)δ163.7,162.3,158.5,150.8,149.9,148.1,146.3,145.3,144.9,135.9,135.8,130.3,130.2,130.1,129.8,128.3,128.1,127.1,126.4,124.0,119.4,113.7,105.4,86.7,86.5,86.3,70.9,64.1,55.4,39.7.1d: 1 H NMR (400MHz, DMSO) δ12.04 (s, 1H), 9.29 (t, J = 5.9Hz, 1H), 8.77 (d, J = 4.4Hz, 1H), 8.54 (s, 1H), 8.19(d,J=8.3Hz,1H),8.06(d,J=8.4Hz,1H),7.77(t,J=7.6Hz,1H),7.61(t,J=7.5Hz,1H),7.37( d,J=7.7Hz,2H),7.32(d,J=4.3Hz,1H),7.26(dd,J=11.1,6.7Hz,6H),7.16(t,J=7.2Hz,1H),6.85( d,J=8.5Hz,4H),6.10(t,J=6.3Hz,1H),5.36(d,J=4.4Hz,1H),5.09–4.94(m,2H),4.11(d,J=4.3 13 C NMR (101MHz, DMSO) δ163.7,162.3,158.5,150.8,149.9,148.1,146.3,145.3,144.9,135.9,135.8,130.3,130.2,130.1,129.8,128.3,128.1,127.1,126.4, 124.0,119.4,113.7,105.4 ,86.7,86.5,86.3,70.9,64.1,55.4,39.7.
1e:1H NMR(400MHz,DMSO)δ11.99(s,1H),9.28(t,J=6.1Hz,1H),8.89(t,J=3.1Hz,1H),8.50(s,1H),8.17(s,1H),8.01(d,J=8.4Hz,1H),7.85(d,J=8.1Hz,1H),7.75–7.69(m,1H),7.60–7.54(m,1H),7.36(d,J=7.5Hz,2H),7.28–7.22(m,6H),7.12(t,J=7.3Hz,1H),6.86(dd,J=8.9,1.8Hz,4H),6.08(t,J=6.4Hz,1H),5.37(d,J=4.5Hz,1H),4.78–4.63(m,2H),4.18–4.05(m,1H),3.95(dd,J=8.5,4.3Hz,1H),3.69(s,6H),3.18(t,J=4.7Hz,2H),2.33–2.17(m,2H).13C NMR(101MHz,DMSO)δ163.6,162.3,158.5,151.4,149.9,147.2,146.2,145.3,135.9,135.8,134.1,133.0,130.2,129.6,129.1,128.3,128.1,127.9,127.2,127.0,113.7,105.6,86.6,86.4,86.2,70.9,64.1,55.4,40.5.1e: 1 H NMR (400MHz, DMSO) δ11.99 (s, 1H), 9.28 (t, J = 6.1Hz, 1H), 8.89 (t, J = 3.1Hz, 1H), 8.50 (s, 1H), 8.17(s,1H),8.01(d,J=8.4Hz,1H),7.85(d,J=8.1Hz,1H),7.75–7.69(m,1H),7.60–7.54(m,1H),7.36 (d,J=7.5Hz,2H),7.28–7.22(m,6H),7.12(t,J=7.3Hz,1H),6.86(dd,J=8.9,1.8Hz,4H),6.08(t, J=6.4Hz,1H),5.37(d,J=4.5Hz,1H),4.78–4.63(m,2H),4.18–4.05(m,1H),3.95(dd,J=8.5,4.3Hz,1H ), 3.69 (s, 6H), 3.18 (t, J = 4.7Hz, 2H), 2.33–2.17 (m, 2H). 13 C NMR (101MHz, DMSO) δ 163.6, 162.3, 158.5, 151.4, 149.9, 147.2, 146.2,145.3,135.9,135.8,134.1,133.0,130.2,129.6,129.1,128.3,128.1,127.9,127.2,127.0,113.7,105.6,86.6,86.4,86.2,70.9,64.1,55 .4,40.5.
1f:1H NMR(400MHz,DMSO)δ12.01(s,1H),9.65(t,J=5.4Hz,1H),8.52(s,1H),8.30(d,J=8.5Hz,1H),7.97(t,J=7.8Hz,2H),7.81–7.72(m,1H),7.59(dd,J=11.5,4.5Hz,1H),7.48(d,J=8.5Hz,1H),7.37(d,J=7.4Hz,2H),7.31–7.23(m,6H),7.15(t,J=7.3Hz,1H),6.87(d,J=8.9Hz,4H),6.11(t,J=6.4Hz,1H),5.36(d,J=4.6Hz,1H),4.79(d,J=5.4Hz,2H),4.13(dt,J=8.9,4.3Hz,1H),3.96(q,J=4.3Hz,1H),3.69(d,J=2.1Hz,6H),3.20(d,J=4.3Hz,2H),2.36–2.19(m,2H).13C NMR(101MHz, DMSO)δ163.6,162.1,158.6,158.5,150.0,146.1,145.3,137.1,136.0,135.8,130.3,130.2,128.9,128.4,128.3,128.1,127.4,127.1,126.7,120.2,113.7,105.7,86.5,86.4,86.2,70.8,64.1,55.4,45.3.1f: 1 H NMR (400MHz, DMSO) δ12.01 (s, 1H), 9.65 (t, J = 5.4Hz, 1H), 8.52 (s, 1H), 8.30 (d, J = 8.5Hz, 1H), 7.97(t,J=7.8Hz,2H),7.81–7.72(m,1H),7.59(dd,J=11.5,4.5Hz,1H),7.48(d,J=8.5Hz,1H),7.37(d ,J=7.4Hz,2H),7.31–7.23(m,6H),7.15(t,J=7.3Hz,1H),6.87(d,J=8.9Hz,4H),6.11(t,J=6.4Hz ,1H),5.36(d,J=4.6Hz,1H),4.79(d,J=5.4Hz,2H),4.13(dt,J=8.9,4.3Hz,1H),3.96(q,J=4.3Hz ,1H),3.69(d,J=2.1Hz,6H),3.20(d,J=4.3Hz,2H),2.36–2.19(m,2H). 13 C NMR(101MHz, DMSO)δ163.6,162.1,158.6,158.5,150.0,146.1,145.3,137.1,136.0,135.8,130.3,130.2,128.9,128.4,128.3,128.1,127.4,127.1,126.7,120.2, 113.7,105.7,86.5,86.4, 86.2,70.8,64.1,55.4,45.3.
1g:1H NMR(400MHz,DMSO)δ11.93(s,1H),9.42(t,J=6.1Hz,1H),8.98(dd,J=4.2,1.7Hz,1H),8.48(s,1H),8.39(dt,J=8.6,4.3Hz,1H),7.90(d,J=8.1Hz,1H),7.64(d,J=7.0Hz,1H),7.59(dd,J=8.3,4.2Hz,1H),7.56–7.50(m,1H),7.36(d,J=7.5Hz,2H),7.30–7.22(m,6H),7.17(t,J=7.2Hz,1H),6.87(d,J=8.8Hz,4H),6.09(t,J=6.4Hz,1H),5.34(d,J=4.6Hz,1H),5.08(d,J=6.0Hz,2H),4.17–4.08(m,1H),3.94(q,J=4.4Hz,1H),3.68(d,J=3.7Hz,6H),3.19(d,J=4.4Hz,2H),2.35–2.11(m,2H).13C NMR(101MHz,DMSO)δ163.6,161.9,158.5,150.3,149.9,146.2,146.0,145.4,136.9,135.9,135.8,130.3,130.1,128.4,128.3,128.1,127.8,127.1,126.7,122.0,113.7,105.8,86.4,86.3,86.2,70.8,64.1,55.4.1g: 1 H NMR (400MHz, DMSO) δ11.93 (s, 1H), 9.42 (t, J = 6.1Hz, 1H), 8.98 (dd, J = 4.2, 1.7Hz, 1H), 8.48 (s, 1H ),8.39(dt,J=8.6,4.3Hz,1H),7.90(d,J=8.1Hz,1H),7.64(d,J=7.0Hz,1H),7.59(dd,J=8.3,4.2Hz ,1H),7.56–7.50(m,1H),7.36(d,J=7.5Hz,2H),7.30–7.22(m,6H),7.17(t,J=7.2Hz,1H),6.87(d, J=8.8Hz,4H),6.09(t,J=6.4Hz,1H),5.34(d,J=4.6Hz,1H),5.08(d,J=6.0Hz,2H),4.17–4.08(m, 1H),3.94(q,J=4.4Hz,1H),3.68(d,J=3.7Hz,6H),3.19(d,J=4.4Hz,2H),2.35–2.11(m,2H). 13 C NMR (101MHz, DMSO) δ163.6,161.9,158.5,150.3,149.9,146.2,146.0,145.4,136.9,135.9,135.8,130.3,130.1,128.4,128.3,128.1,127.8,127.1,126. 7,122.0,113.7,105.8, 86.4,86.3,86.2,70.8,64.1,55.4.
2a~2g的合成路线:在氮气保护下且冰水浴的条件下,向50mL 3.3mmol 1a~1g(1a为2.2g,1b-1g为2.4g)的无水二氯甲烷溶液中加入四氮唑(0.69g,9.8mmol)和磷试剂2-氰乙基-N,N,N',N'-四异丙基亚磷酰胺(2.9g,9.8mmol)。然后将溶液移至室温搅拌2小时,将溶液真空浓缩。然后通过快速色谱法(正己烷/乙酸乙酯=1/1)纯化残余物,得到所需产物2a-2g,为粘稠和淡黄色固体(收率:2a:70%,2b:72%,2c:48%,2d:70%,2e:58%,2f:79%,2g:52%)。Synthetic route of 2a~2g: Under nitrogen protection and ice water bath conditions, add tetrazole to 50mL 3.3mmol 1a~1g (1a is 2.2g, 1b-1g is 2.4g) anhydrous dichloromethane solution (0.69g, 9.8mmol) and the phosphorus reagent 2-cyanoethyl-N,N,N',N'-tetraisopropylphosphoramidite (2.9g, 9.8mmol). The solution was then moved to room temperature and stirred for 2 hours, and the solution was concentrated in vacuo. The residue was then purified by flash chromatography (n-hexane/ethyl acetate = 1/1) to obtain the desired product 2a-2g as a viscous and light yellow solid (yield: 2a: 70%, 2b: 72%, 2c:48%, 2d:70%, 2e:58%, 2f:79%, 2g:52%).
2a:1H NMR(400MHz,DMSO)δ11.93(s,1H),9.18(d,J=5.8Hz,1H),8.89(s,1H),8.63–8.52(m,2H),7.95(d,J=8.4Hz,1H),7.73–7.63(m,1H),7.56(d,J=7.0Hz,1H),7.47–7.35(m,3H),7.30–7.23(m,6H),7.18(dd,J=7.6,5.9Hz,1H),6.86(d,J=7.9Hz,4H),6.13–6.04(m,1H),5.05–4.91(m,2H),4.40–4.29(m,1H),4.10(dd,J=15.9,4.1Hz,1H),3.70–3.67(m,6H),3.63–3.37(m,4H),3.23(d,J=3.8Hz,2H),2.75(t,J=5.9Hz,1H),2.64(t,J=5.4Hz,1H),2.47–2.35(m,2H),1.10(dd,J=15.1,7.7Hz,9H),0.96(d,J=6.7Hz,3H).13C NMR(101MHz,DMSO)δ163.7,161.9,158.5,150.7,149.8,148.5,146.7,146.5,145.2,136.0,135.9,135.8,135.7,132.4,130.2,129.5,129.2,128.3,128.1,127.1,126.4,121.8,119.4,119.2,113.7,105.5,87.1,86.3,85.6,73.2,63.7,59.0,58.8,55.4,43.1,43.0,24.7,20.2.31P NMR(162MHz,DMSO)δ147.6,147.3.2a: 1 H NMR (400MHz, DMSO) δ11.93 (s, 1H), 9.18 (d, J = 5.8Hz, 1H), 8.89 (s, 1H), 8.63–8.52 (m, 2H), 7.95 (d ,J=8.4Hz,1H),7.73–7.63(m,1H),7.56(d,J=7.0Hz,1H),7.47–7.35(m,3H),7.30–7.23(m,6H),7.18( dd,J=7.6,5.9Hz,1H),6.86(d,J=7.9Hz,4H),6.13–6.04(m,1H),5.05–4.91(m,2H),4.40–4.29(m,1H) ,4.10(dd,J=15.9,4.1Hz,1H),3.70–3.67(m,6H),3.63–3.37(m,4H),3.23(d,J=3.8Hz,2H),2.75(t,J =5.9Hz,1H),2.64(t,J=5.4Hz,1H),2.47–2.35(m,2H),1.10(dd,J=15.1,7.7Hz,9H),0.96(d,J=6.7Hz ,3H). 13 C NMR (101MHz, DMSO) δ163.7, 161.9, 158.5, 150.7, 149.8, 148.5, 146.7, 146.5, 145.2, 136.0, 135.9, 135.8, 135.7, 132.4, 130.2, 129.5, 129.2, 128.3, 128.1,127.1,126.4,121.8,119.4,119.2,113.7,105.5,87.1,86.3,85.6,73.2,63.7,59.0,58.8,55.4,43.1,43.0,24.7,20.2. 31 P NMR (162MHz, DMSO) δ147. 6,147.3.
2b:1H NMR(400MHz,DMSO)δ11.93(s,1H),9.27(s,1H),8.85(d,J=2.9Hz,1H),8.57(d,J=7.0Hz,1H),8.21(t,J=8.2Hz,1H),7.96(d,J=8.6Hz,1H),7.80(s,1H),7.70(d,J=8.6Hz,1H),7.47(dd,J=7.4,3.2Hz,1H),7.37(d,J=7.3Hz,2H),7.25(t,J=6.3Hz,6H),7.16–7.11(m,1H),6.86(d,J=7.7Hz,4H),6.15–6.04(m,1H),4.70(s,2H),4.34(s,1H),4.15–4.05(m,1H),3.68(s,6H),3.61–3.46(m,4H),3.24(s,2H),2.75(t,J=5.8Hz,1H),2.64(d,J=6.1Hz,1H),2.47–2.38(m,2H),1.10(dd,J=14.7,7.5Hz,9H),0.96(d,J=6.7Hz,3H).13C NMR(101MHz,DMSO)δ163.7,162.2,158.5,150.7,149.8,147.5,146.6,146.4,145.2,138.1,136.2,135.9,135.8,135.7,130.2,130.1,129.8,129.5,128.3,128.2,128.1,127.1,126.0,122.1,119.4,119.2,113.7,105.6,87.0,86.4,85.6,73.4,63.7,59.0,58.7,55.4,43.1,42.6,24.7,20.2.31P NMR(162MHz,DMSO)δ147.7,147.4.2b: 1 H NMR (400MHz, DMSO) δ11.93 (s, 1H), 9.27 (s, 1H), 8.85 (d, J = 2.9Hz, 1H), 8.57 (d, J = 7.0Hz, 1H), 8.21(t,J=8.2Hz,1H),7.96(d,J=8.6Hz,1H),7.80(s,1H),7.70(d,J=8.6Hz,1H),7.47(dd,J=7.4 ,3.2Hz,1H),7.37(d,J=7.3Hz,2H),7.25(t,J=6.3Hz,6H),7.16–7.11(m,1H),6.86(d,J=7.7Hz,4H ),6.15–6.04(m,1H),4.70(s,2H),4.34(s,1H),4.15–4.05(m,1H),3.68(s,6H),3.61–3.46(m,4H), 3.24(s,2H),2.75(t,J=5.8Hz,1H),2.64(d,J=6.1Hz,1H),2.47–2.38(m,2H),1.10(dd,J=14.7,7.5Hz ,9H),0.96(d,J=6.7Hz,3H). 13 C NMR(101MHz,DMSO)δ163.7,162.2,158.5,150.7,149.8,147.5,146.6,146.4,145.2,138.1,136.2,135.9,135.8, 135.7,130.2,130.1,129.8,129.5,128.3,128.2,128.1,127.1,126.0,122.1,119.4,119.2,113.7,105.6,87.0,86.4,85.6,73.4,63.7,59.0,58. 7,55.4,43.1,42.6, 24.7,20.2. 31 P NMR (162MHz, DMSO) δ147.7,147.4.
2c:1H NMR(400MHz,DMSO)δ11.92(s,1H),9.29(t,J=5.5Hz,1H),8.88(d,J=2.7Hz,1H),8.55(d,J=5.6Hz,1H),8.33(d,J=8.2Hz,1H),7.92(d,J=9.9Hz,2H),7.55(d,J=8.4Hz,1H),7.49(dd,J=8.2,4.2Hz,1H),7.41–7.33(m,2H),7.26(s,6H),7.15(t,J=7.2Hz,1H),6.86(dd,J=8.6,3.0Hz,4H), 6.16–6.04(m,1H),4.73(d,J=5.5Hz,2H),4.36(d,J=4.5Hz,1H),4.09(dd,J=9.2,4.2Hz,1H),3.69(d,J=1.4Hz,6H),3.63–3.39(m,4H),3.26(dd,J=12.2,7.2Hz,2H),2.75(t,J=5.8Hz,1H),2.64(t,J=5.5Hz,1H),2.42(dd,J=12.8,5.9Hz,2H),1.10(dd,J=14.3,7.4Hz,9H),0.96(d,J=6.7Hz,3H).13C NMR(101MHz,DMSO)δ163.7,162.2,158.5,151.2,149.9,148.3,146.6,146.5,145.2,141.4,136.2,135.9,135.8,135.7,130.2,130.1,128.6,128.3,128.1,127.3,127.1,126.8,121.6,119.4,119.2,113.7,105.6,87.0,86.3,85.5,73.5,63.8,59.0,58.8,55.4,43.1,42.7,24..7,20.2.31P NMR(162MHz,DMSO)δ147.7,147.4.2c: 1 H NMR (400MHz, DMSO) δ11.92 (s, 1H), 9.29 (t, J = 5.5Hz, 1H), 8.88 (d, J = 2.7Hz, 1H), 8.55 (d, J = 5.6 Hz,1H),8.33(d,J=8.2Hz,1H),7.92(d,J=9.9Hz,2H),7.55(d,J=8.4Hz,1H),7.49(dd,J=8.2,4.2 Hz,1H),7.41–7.33(m,2H),7.26(s,6H),7.15(t,J=7.2Hz,1H),6.86(dd,J=8.6,3.0Hz,4H), 6.16–6.04(m,1H),4.73(d,J=5.5Hz,2H),4.36(d,J=4.5Hz,1H),4.09(dd,J=9.2,4.2Hz,1H),3.69(d ,J=1.4Hz,6H),3.63–3.39(m,4H),3.26(dd,J=12.2,7.2Hz,2H),2.75(t,J=5.8Hz,1H),2.64(t,J= 5.5Hz, 1H), 2.42 (dd, J=12.8, 5.9Hz, 2H), 1.10 (dd, J=14.3, 7.4Hz, 9H), 0.96 (d, J=6.7Hz, 3H). 13 C NMR ( 101MHz, DMSO) δ163.7,162.2,158.5,151.2,149.9,148.3,146.6,146.5,145.2,141.4,136.2,135.9,135.8,135.7,130.2,130.1,128.6,128.3,128.1, 127.3,127.1,126.8,121.6, 119.4,119.2,113.7,105.6,87.0,86.3,85.5,73.5,63.8,59.0,58.8,55.4,43.1,42.7,24..7,20.2. 31 P NMR (162MHz, DMSO) δ147.7,147.4.
2d:1H NMR(400MHz,DMSO)δ11.94(s,1H),9.28(d,J=4.3Hz,1H),8.77(t,J=4.7Hz,1H),8.57(d,J=6.1Hz,1H),8.19(d,J=8.4Hz,1H),8.06(d,J=8.4Hz,1H),7.77(dd,J=8.2,7.1Hz,1H),7.65–7.56(m,1H),7.40–7.35(m,2H),7.33(t,J=4.2Hz,1H),7.26(dt,J=9.0,5.0Hz,6H),7.17(dd,J=7.7,5.9Hz,1H),6.85(dd,J=8.9,2.5Hz,4H),6.15–6.06(m,1H),5.02(s,2H),4.41–4.30(m,1H),4.14–4.07(m,1H),3.67–3.63(m,6H),3.61–3.46(m,4H),3.23(d,J=4.4Hz,2H),2.75(t,J=5.9Hz,1H),2.64(t,J=6.1Hz,1H),2.43(dd,J=13.1,7.6Hz,2H),1.13–1.07(m,9H),0.96(d,J=6.7Hz,3H).13C NMR(101MHz,DMSO)δ163.7,162.3,158.5,150.8,149.9,148.1,146.7,146.6,145.2,144.9,135.9,135.8,135.7,130.2,130.1,129.8,128.3,128.1,127.1,126.4,124.0,119.4,119.2,113.7,105.5,87.1,86.4,85.6,73.4,63.8,59.0,58.8,55.4,43.1,43.0,24.7,20.2.31P NMR(162MHz,DMSO)δ147.6,147.3.2d: 1 H NMR (400MHz, DMSO) δ11.94 (s, 1H), 9.28 (d, J = 4.3Hz, 1H), 8.77 (t, J = 4.7Hz, 1H), 8.57 (d, J = 6.1 Hz,1H),8.19(d,J=8.4Hz,1H),8.06(d,J=8.4Hz,1H),7.77(dd,J=8.2,7.1Hz,1H),7.65–7.56(m,1H ),7.40–7.35(m,2H),7.33(t,J=4.2Hz,1H),7.26(dt,J=9.0,5.0Hz,6H),7.17(dd,J=7.7,5.9Hz,1H) ,6.85(dd,J=8.9,2.5Hz,4H),6.15–6.06(m,1H),5.02(s,2H),4.41–4.30(m,1H),4.14–4.07(m,1H),3.67 –3.63(m,6H),3.61–3.46(m,4H),3.23(d,J=4.4Hz,2H),2.75(t,J=5.9Hz,1H),2.64(t,J=6.1Hz, 1H), 2.43 (dd, J=13.1, 7.6Hz, 2H), 1.13–1.07 (m, 9H), 0.96 (d, J=6.7Hz, 3H). 13 C NMR (101MHz, DMSO) δ 163.7, 162.3, 158.5,150.8,149.9,148.1,146.7,146.6,145.2,144.9,135.9,135.8,135.7,130.2,130.1,129.8,128.3,128.1,127.1,126.4,124.0,119.4,119 .2,113.7,105.5,87.1,86.4, 85.6,73.4,63.8,59.0,58.8,55.4,43.1,43.0,24.7,20.2. 31 P NMR (162MHz, DMSO) δ147.6,147.3.
2e:1H NMR(400MHz,DMSO)δ11.93(s,1H),9.30(t,J=5.6Hz,1H),8.89(s,1H),8.54(d,J=6.9Hz,1H),8.17(s,1H),8.01(d,J=8.4Hz,1H),7.85(t,J=7.7Hz,1H),7.72(t,J=7.6Hz,1H),7.57(t,J=7.3Hz,1H),7.40–7.33(m,2H),7.25(td,J=6.4,2.9Hz,6H),7.13(t,J=7.2Hz,1H),6.91–6.81(m,4H),6.13–6.02(m,1H),4.70(d,J=5.6Hz,2H),4.41–4.30(m,1H),4.14–4.07(m,1H),3.69(s,6H),3.62–3.46(m,4H),3.22(d,J=4.5Hz,2H),2.75(t,J=5.9Hz,1H),2.64(dd,J=6.1,4.7Hz,1H),2.48–2.35(m,2H),1.10(dd,J=15.3,7.7Hz,9H),0.96(d,J=6.7Hz,3H).13C NMR(101MHz,DMSO)δ163.6,162.3,158.5,151.4,149.9,147.2,146.6,146.4,145.2,135.9,135.8,135.7,134.1,133.0,130.2,129.6,129.1,128.3,128.1,127.9,127.2,119.4,119.2,113.7,105.6,87.0,86.3,85.6,73.4,63.7,58.8,55.4,43.1,43.0,24.7,20.2.31P NMR(162MHz,DMSO)δ147.6,147.3.2e: 1 H NMR (400MHz, DMSO) δ11.93 (s, 1H), 9.30 (t, J = 5.6Hz, 1H), 8.89 (s, 1H), 8.54 (d, J = 6.9Hz, 1H), 8.17(s,1H),8.01(d,J=8.4Hz,1H),7.85(t,J=7.7Hz,1H),7.72(t,J=7.6Hz,1H),7.57(t,J=7.3 Hz,1H),7.40–7.33(m,2H),7.25(td,J=6.4,2.9Hz,6H),7.13(t,J=7.2Hz,1H),6.91–6.81(m,4H),6.13 –6.02(m,1H),4.70(d,J=5.6Hz,2H),4.41–4.30(m,1H),4.14–4.07(m,1H),3.69(s,6H),3.62–3.46(m ,4H),3.22(d,J=4.5Hz,2H),2.75(t,J=5.9Hz,1H),2.64(dd,J=6.1,4.7Hz,1H),2.48–2.35(m,2H) ,1.10(dd,J=15.3,7.7Hz,9H),0.96(d,J=6.7Hz,3H). 13 C NMR(101MHz,DMSO)δ163.6,162.3,158.5,151.4,149.9,147.2,146.6,146.4 ,145.2,135.9,135.8,135.7,134.1,133.0,130.2,129.6,129.1,128.3,128.1,127.9,127.2,119.4,119.2,113.7,105.6,87.0,86.3,85.6,73.4, 63.7,58.8,55.4,43.1 ,43.0,24.7,20.2. 31 P NMR (162MHz, DMSO) δ147.6,147.3.
2f:1H NMR(400MHz,DMSO)δ12.01(s,1H),9.66(t,J=5.1Hz,1H),8.56(d,J=6.0Hz,1H),8.31(dd,J=8.5,2.8Hz,1H),8.00–7.93(m,2H),7.76(dd,J=11.3,4.2Hz,1H),7.58(t,J=7.5Hz,1H),7.48(dd,J=8.5,2.5Hz,1H),7.37(dd,J=7.5,3.1Hz,2H),7.29–7.23(m,6H),7.15(t,J=7.2Hz,1H),6.86(dd,J=8.9,3.0Hz,4H),6.16–6.05(m,1H),4.83–4.76(m,2H),4.38(dd,J=10.1,4.8Hz,1H),4.14–4.07(m,1H),3.69(d,J=2.4Hz,6H),3.63–3.46(m,4H),3.29–3.18(m,2H),2.76(t,J=5.9Hz,1H),2.68–2.61(m,1H),2.48–2.37(m,2H),1.13–1.07(m,9H),0.96(d,J=6.7Hz,3H).13C NMR(101MHz,DMSO)δ163.6,162.1,158.6,149.9,147.3,146.5,146.3,145.2,137.1,135.9,135.8,135.7,130.2,130.1,128.9,128.3,128.1,127.4,127.1,126.7,120.2,119.4,119.2,113.7,105.8,86.9,86.4,85.5,73.1,63.5,59.0,55.4,45.3,43.1,24.8,20.2.31P NMR(162MHz,DMSO)δ147.6,147.3.2f: 1 H NMR (400MHz, DMSO) δ12.01 (s, 1H), 9.66 (t, J = 5.1Hz, 1H), 8.56 (d, J = 6.0Hz, 1H), 8.31 (dd, J = 8.5 ,2.8Hz,1H),8.00–7.93(m,2H),7.76(dd,J=11.3,4.2Hz,1H),7.58(t,J=7.5Hz,1H),7.48(dd,J=8.5, 2.5Hz,1H),7.37(dd,J=7.5,3.1Hz,2H),7.29–7.23(m,6H),7.15(t,J=7.2Hz,1H),6.86(dd,J=8.9,3.0 Hz,4H),6.16–6.05(m,1H),4.83–4.76(m,2H),4.38(dd,J=10.1,4.8Hz,1H),4.14–4.07(m,1H),3.69(d, J=2.4Hz,6H),3.63–3.46(m,4H),3.29–3.18(m,2H),2.76(t,J=5.9Hz,1H),2.68–2.61(m,1H),2.48–2.37 (m,2H),1.13–1.07(m,9H),0.96(d,J=6.7Hz,3H). 13 C NMR(101MHz,DMSO)δ163.6,162.1,158.6,149.9,147.3,146.5,146.3,145.2 ,137.1,135.9,135.8,135.7,130.2,130.1,128.9,128.3,128.1,127.4,127.1,126.7,120.2,119.4,119.2,113.7,105.8,86.9,86.4,85.5,73.1, 63.5,59.0,55.4,45.3 ,43.1,24.8,20.2. 31 P NMR (162MHz, DMSO) δ147.6,147.3.
2g:1H NMR(400MHz,DMSO)δ11.93(s,1H),9.42(t,J=5.7Hz,1H),8.98(d,J=2.7Hz,1H),8.51(d,J=5.7Hz,1H),8.40(d,J=7.0Hz,1H),7.91(d,J=8.1Hz,1H),7.68–7.56(m,2H),7.53(td,J=7.7,2.8Hz,1H),7.40–7.32(m,2H),7.31–7.21(m,6H),7.20–7.13(m,1H),6.86(dd,J=8.8,3.5Hz,4H),6.14–6.03(m,1H),5.08(d,J=5.8Hz,2H),4.41–4.29(m,1H),4.13–4.05(m,1H),3.71–3.66(m,6H),3.62–3.38(m,4H),3.24(dd,J=25.5,5.9Hz,2H),2.75(t,J=5.9Hz,1H),2.66–2.61(m,1H),2.46–2.33(m,2H),1.09(dd,J=16.0,7.1Hz,9H),0.95(d,J=6.7Hz,3H).13C NMR(101MHz,DMSO) δ163.6,161.1,158.5,150.3,149.9,146.4,146.2,145.2,136.9,135.9,135.8,135.7,130.2,130.1,128.4,128.3,128.1,127.8,127.1,126.7,122.0,119.4,119.2,113.7,105.9,86.9,86.4,85.5,72.9,63.6,59.0,55.4,43.1,43.0,24.7,20.2.31P NMR(162MHz,DMSO)δ147.7,147.3.2g: 1 H NMR (400MHz, DMSO) δ11.93 (s, 1H), 9.42 (t, J = 5.7Hz, 1H), 8.98 (d, J = 2.7Hz, 1H), 8.51 (d, J = 5.7 Hz,1H),8.40(d,J=7.0Hz,1H),7.91(d,J=8.1Hz,1H),7.68–7.56(m,2H),7.53(td,J=7.7,2.8Hz,1H ),7.40–7.32(m,2H),7.31–7.21(m,6H),7.20–7.13(m,1H),6.86(dd,J=8.8,3.5Hz,4H),6.14–6.03(m,1H ),5.08(d,J=5.8Hz,2H),4.41–4.29(m,1H),4.13–4.05(m,1H),3.71–3.66(m,6H),3.62–3.38(m,4H), 3.24(dd,J=25.5,5.9Hz,2H),2.75(t,J=5.9Hz,1H),2.66–2.61(m,1H),2.46–2.33(m,2H),1.09(dd,J= 16.0,7.1Hz,9H),0.95(d,J=6.7Hz,3H). 13 C NMR(101MHz,DMSO) δ163.6,161.1,158.5,150.3,149.9,146.4,146.2,145.2,136.9,135.9,135.8,135.7,130.2,130.1,128.4,128.3,128.1,127.8,127.1,126.7,12 2.0,119.4,119.2,113.7,105.9, 86.9,86.4,85.5,72.9,63.6,59.0,55.4,43.1,43.0,24.7,20.2. 31 P NMR (162MHz, DMSO) δ147.7,147.3.
实施例2、通过喹啉修饰核酸适配体APC001增加其靶结合亲和力Example 2. Modification of nucleic acid aptamer APC001 by quinoline to increase its target binding affinity
前期工作通过protein-SELEX技术筛选得到特异性靶向骨硬化素的核酸适配子APC001,具体序列为:CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC。通过对其进行化学生物学研究,筛查得到22个适配子-靶标结合的关键位点(分别是C1、G2、G3、G4、G5、T6、T8、G11、T13、G15、T29、T30、G31、G32、C33、A34、G35、C36、T37、G38、C39、C40)。In previous work, the nucleic acid aptamer APC001, which specifically targets osteosclerostin, was screened through protein-SELEX technology. The specific sequence is: CGGGGTGTGGGTTCGTCGTTAGCTTGATTTGGCAGCTGCC. Through chemical biology research, 22 key sites for aptamer-target binding were screened (respectively C1, G2, G3, G4, G5, T6, T8, G11, T13, G15, T29, T30 , G31, G32, C33, A34, G35, C36, T37, G38, C39, C40).
本发明用化学修饰技术,在这些关键位点上分别修饰不同亚型的喹啉。首先将不同的喹啉偶联到核苷上获得化合物1a-1g,然后再跟磷试剂反应构建不同喹啉亚型修饰亚磷酰胺单体2a-2g,最后通过DNA固相合成技术获得在不同关键位点进行喹啉修饰的适配子。The present invention uses chemical modification technology to modify different subtypes of quinolines at these key sites. First, different quinolines are coupled to nucleosides to obtain compounds 1a-1g, and then reacted with phosphorus reagents to construct different quinoline isoforms modified phosphoramidite monomers 2a-2g. Finally, DNA solid-phase synthesis technology is used to obtain compounds in different Aptamers with quinoline modification at key sites.
具体而言通过标准的DNA固相合成技术,将亚磷酰胺单体2a~2g分别插入骨硬化素适配子的22个关键位点中。序列和插入位置如下表1所示:Specifically, the phosphoramidite monomers 2a to 2g were inserted into the 22 key sites of the osteosclerostin aptamer through standard DNA solid-phase synthesis technology. The sequence and insertion position are shown in Table 1 below:
表1.喹啉修饰适配子序列(x=a、b、c、d、e、f、g)

Table 1. Quinoline modified adapter sequences (x=a, b, c, d, e, f, g)

ELONA方法比较各喹啉修饰适配子与靶标骨硬化素的结合能力ELONA method to compare the binding ability of each quinoline-modified aptamer to the target osteosclerostin
预先制备以下缓冲液:Prepare the following buffer in advance:
ELONA B&W buffer:PBS,1mM MgCl2,0.05%Tween 20;ELONA B&W buffer: PBS, 1mM MgCl2, 0.05% Tween 20;
ELONA assay blocking buffer:PBS,0.1%Tween 20and 1%BSA;ELONA assay blocking buffer:PBS,0.1% Tween 20and 1%BSA;
ELONA assay washing buffer:PBS,1mM MgCl2,0.1%Tween 20and 0.1%BSA;ELONA assay washing buffer: PBS, 1mM MgCl2, 0.1% Tween 20 and 0.1% BSA;
ELONA assay Strep-HRP binding buffer:Strep-HRP 1:10000稀释到ELONA assay washing buffer中;ELONA assay Strep-HRP binding buffer: Strep-HRP 1:10000 diluted into ELONA assay washing buffer;
ELONA assay reaction stopping buffer:2M H2SO4。ELONA assay reaction stopping buffer:2M H2SO4.
往含100μl SELEX B&W buffer中的96孔中加入160ng目标蛋白4℃下孵育过夜。然后将96孔板放在室温下用Blocking buffer封闭1小时,然后用SELEX B&W buffer洗涤4次。将末端生物素化的适配子以1μM溶于100μL SELEX B&W buffer中添加到每个孔中,室温下孵育45分钟,同时持续轻轻晃动。接着,用SELEX B&W buffer洗涤4次以去除非特异性和非常弱的结合。每孔加入100μl Strap-HRP(1:10000稀释),孵育30分钟,用washing buffer洗涤4次。每孔加入50μl TMB,孵育20分钟。加入50μl2M H2SO4终止反应。用酶标仪测量450nm处的吸光度。Add 160ng of target protein to 96 wells containing 100μl SELEX B&W buffer and incubate overnight at 4°C. The 96-well plate was then blocked with Blocking buffer for 1 hour at room temperature, and then washed 4 times with SELEX B&W buffer. Add terminally biotinylated aptamers at 1 μM in 100 μL SELEX B&W buffer to each well and incubate at room temperature for 45 minutes while continuing to shake gently. Next, wash 4 times with SELEX B&W buffer to remove non-specific and very weak binding. Add 100 μl Strap-HRP (1:10000 dilution) to each well, incubate for 30 minutes, and wash 4 times with washing buffer. Add 50 μl TMB to each well and incubate for 20 minutes. The reaction was stopped by adding 50 μl 2M H2SO4 . Measure the absorbance at 450 nm with a microplate reader.
结果如图1所示。所获得的喹啉修饰的骨硬化素核酸适配子,相对天然的APC001对靶标的结合能力普遍提高。其中,其中在适配子6位和11位进行5喹啉(d)修饰后(即适配子APC001-d6和APC001-d11),提高效果最为显著(近16倍)。The results are shown in Figure 1. The obtained quinoline-modified osteosclerostin nucleic acid aptamer generally has improved target-binding ability compared with the natural APC001. Among them, after modification of 5-quinoline (d) at positions 6 and 11 of the aptamer (i.e. aptamers APC001-d6 and APC001-d11), the improvement effect is most significant (nearly 16 times).
生物膜干涉法(Biolayer interferometry,BLI)测定喹啉修饰适配子与靶标的亲和力Biolayer interferometry (BLI) determines the affinity between quinoline-modified aptamers and targets
采用生物膜干涉法(Biolayer interferometry,BLI)测定图1中结合能力最高的6条序列即APC001-d6、APC001-d8、APC001-d11、APC001-d13、APC001-d32、APC001-d34的亲和力。Biolayer interferometry (BLI) was used to determine the affinity of the six sequences with the highest binding ability in Figure 1, namely APC001-d6, APC001-d8, APC001-d11, APC001-d13, APC001-d32, and APC001-d34.
分析喹啉修饰适配子与骨硬化素的相互作用所用仪器为Octet 96/96e system(ForteBio),所有操作在常温下进行。程序运行程序如下:The instrument used to analyze the interaction between quinoline-modified aptamers and osteosclerostin was Octet 96/96e system (ForteBio), and all operations were performed at room temperature. The program running procedure is as follows:
Baseline:1×PBS buffer(10mM phosphate buffer,2.7mM KCl and 137mM NaCl,pH 7.4),120s,1000rpm;Baseline: 1×PBS buffer (10mM phosphate buffer, 2.7mM KCl and 137mM NaCl, pH 7.4), 120s, 1000rpm;
Loading:100nM 5-biotinated aptamer in 1×PBST buffer(1×PBS+0.02%Tween20,pH  7.4),600s,500rpm;Loading:100nM 5-biotinated aptamer in 1×PBST buffer(1×PBS+0.02%Tween20, pH 7.4),600s,500rpm;
Baseline 2:1×PBST buffer,300s,500rpm;Baseline 2:1×PBST buffer,300s,500rpm;
Association:sclerostin(20nM,16nM,12nM,8nM,4nM,0nM)in 1×PBST buffer,600s,500rpm;Association: sclerostin (20nM, 16nM, 12nM, 8nM, 4nM, 0nM) in 1×PBST buffer, 600s, 500rpm;
Dissociation:1×PBST buffer,600s,500rpm;Dissociation:1×PBST buffer,600s,500rpm;
Regenration:5M NaCl,30s,500rpm;Regenration:5M NaCl,30s,500rpm;
Neutralization:1×PBS buffer,60s,500rpm。Neutralization: 1×PBS buffer, 60s, 500rpm.
最终,平衡解离常数KD值用分析软件ForteBio Data analysis 11.0以1:1结合模式拟合计算得到。Finally, the equilibrium dissociation constant KD value was calculated using the analysis software ForteBio Data analysis 11.0 with a 1:1 binding mode fitting.
结果如图2所示。本发明中的喹啉结构大大提高了适配子与靶标的亲和力。 The results are shown in Figure 2. The quinoline structure in the present invention greatly improves the affinity between the aptamer and the target.

Claims (26)

  1. 一种经修饰的靶特异性核酸分子,其包含一或多个喹啉修饰的核苷酸,其中与相应的对照核酸分子相比,所述经修饰的靶特异性核酸分子与靶的结合亲和力提高。A modified target-specific nucleic acid molecule comprising one or more quinoline-modified nucleotides, wherein the modified target-specific nucleic acid molecule has a binding affinity to a target compared to a corresponding control nucleic acid molecule improve.
  2. 权利要求1的经修饰的靶特异性核酸分子,其中所述核酸分子选自适配子(aptamer)、反义核酸(反义RNA)、dsRNA(如siRNA、shRNA)。The modified target-specific nucleic acid molecule of claim 1, wherein the nucleic acid molecule is selected from the group consisting of aptamers, antisense nucleic acids (antisense RNA), dsRNA (such as siRNA, shRNA).
  3. 权利要求1或2的经修饰的靶特异性核酸分子,其中与相应的对照核酸分子相比,所述经修饰的靶特异性核酸分子与靶的结合亲和力提高至少约10%、至少约20%、至少约30%、至少约40%、至少约50%、至少约60%、至少约70%、至少约80%、至少约90%、至少约100%或更多;或者所述经修饰的靶特异性核酸分子与靶的结合亲和力是未修饰的相应核酸分子的亲和力的至少约1.5倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约15倍、至少约16倍、至少约20倍、至少约25倍、至少约30倍、至少约40倍、至少约50倍、至少约75倍、至少约100倍或更多倍。The modified target-specific nucleic acid molecule of claim 1 or 2, wherein the modified target-specific nucleic acid molecule has a binding affinity to a target that is increased by at least about 10%, at least about 20% compared to a corresponding control nucleic acid molecule. , at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100% or more; or the modified The target-specific nucleic acid molecule has a binding affinity to the target that is at least about 1.5 times, at least about 2 times, at least about 3 times, at least about 4 times, at least about 5 times, at least about 6 times, the affinity of the unmodified corresponding nucleic acid molecule. At least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, at least about 15 times, at least about 16 times, at least about 20 times, at least about 25 times, at least about 30 times, at least about 40 times, At least about 50 times, at least about 75 times, at least about 100 times or more.
  4. 权利要求1-3中任一项的经修饰的靶特异性核酸分子,其中所述经修饰的靶特异性核酸分子对靶具有小于100nM,优选小于50nM,优选小于40nM,优选小于30nM,优选小于20nM,优选小于10nM,优选小于5nM或更小的KD(解离常数)。The modified target-specific nucleic acid molecule of any one of claims 1-3, wherein the modified target-specific nucleic acid molecule has an effect on the target of less than 100 nM, preferably less than 50 nM, preferably less than 40 nM, preferably less than 30 nM, preferably less than 20 nM, preferably less than 10 nM, preferably less than 5 nM or less KD (dissociation constant).
  5. 权利要求1-4中任一项的经修饰的靶特异性核酸分子,其中所述喹啉修饰是喹啉-2-基取代、喹啉-3-基取代、喹啉-4-基取代、喹啉-5-基取代、喹啉-6-基取代、喹啉-7-基取代或喹啉-8-基取代,优选地,所述喹啉修饰是喹啉-5-基取代。The modified target-specific nucleic acid molecule of any one of claims 1-4, wherein the quinoline modification is quinolin-2-yl substitution, quinolin-3-yl substitution, quinolin-4-yl substitution, Quinolin-5-yl substitution, quinolin-6-yl substitution, quinolin-7-yl substitution or quinolin-8-yl substitution. Preferably, the quinoline modification is quinolin-5-yl substitution.
  6. 权利要求1-5中任一项的经修饰的靶特异性核酸分子,其中所述喹啉修饰的核苷酸包含取代基团R,所述R选自以下a-g:
    The modified target-specific nucleic acid molecule of any one of claims 1-5, wherein the quinoline-modified nucleotide comprises a substituent group R selected from the following ag:
    优选地, Preferably,
  7. 权利要求1-6中任一项的经修饰的靶特异性核酸分子,其中所述喹啉修饰的核苷酸包括以下式I的碱基部分: The modified target-specific nucleic acid molecule of any one of claims 1-6, wherein the quinoline-modified nucleotide comprises the base moiety of Formula I:
    其中
    in
    优选地, Preferably,
  8. 权利要求1-7中任一项的经修饰的靶特异性核酸分子,其中所述喹啉修饰的核苷酸包括以下式II的结构:
    The modified target-specific nucleic acid molecule of any one of claims 1-7, wherein the quinoline-modified nucleotide comprises the following structure of Formula II:
    其中
    in
    优选地, Preferably,
  9. 权利要求1-8中任一项的经修饰的靶特异性核酸分子,其中所述靶特异性核酸分子中的一或多个天然存在的核苷酸被所述喹啉修饰的核苷酸取代。The modified target-specific nucleic acid molecule of any one of claims 1-8, wherein one or more naturally occurring nucleotides in the target-specific nucleic acid molecule are replaced by the quinoline-modified nucleotides .
  10. 权利要求1-9中任一项的经修饰的靶特异性核酸分子,其中所述靶特异性核酸分子还可以包含一或多种延长其体内半衰期修饰。The modified target-specific nucleic acid molecule of any one of claims 1-9, wherein the target-specific nucleic acid molecule may further comprise one or more modifications that extend its half-life in vivo.
  11. 权利要求1-10中任一项的经修饰的靶特异性核酸分子,其中所述经修饰的靶特异性核酸分子是经修饰的骨硬化素特异性适配子。The modified target-specific nucleic acid molecule of any one of claims 1-10, wherein the modified target-specific nucleic acid molecule is a modified osteosclerostin-specific aptamer.
  12. 权利要求11的经修饰的靶特异性核酸分子,其中经修饰的骨硬化素特异性适配子衍生自SEQ ID NO:2的骨硬化素特异性适配子。The modified target-specific nucleic acid molecule of claim 11, wherein the modified osteosclerostin-specific aptamer is derived from the osteosclerostin-specific aptamer of SEQ ID NO: 2.
  13. 权利要求12的经修饰的靶特异性核酸分子,其中所述经修饰的骨硬化素特异性适配子在相应于SEQ ID NO:2的选自以下的一或多个位置的核苷酸被所述喹啉修饰的核苷酸取代:第1位、第2位、第3位、第4位、第5位、第6位、第8位、第11位、第13位、第15位、第29位、第30位、第31位、第32位、第33位、第34位、第35位、第36位、第37位、第38位、第39位和第40位。 The modified target-specific nucleic acid molecule of claim 12, wherein the modified osteosclerostin-specific aptamer is substituted at a nucleotide corresponding to one or more positions selected from SEQ ID NO: 2 The quinoline modified nucleotide substitutions: 1st, 2nd, 3rd, 4th, 5th, 6th, 8th, 11th, 13th, 15th , 29th, 30th, 31st, 32nd, 33rd, 34th, 35th, 36th, 37th, 38th, 39th and 40th.
  14. 权利要求12或13的经修饰的靶特异性核酸分子,其中所述经修饰的骨硬化素特异性核酸分子在相应于SEQ ID NO:2的第6位、第8位、第11位、第13位、第32位和/或第34位的一或多个核苷酸被所述喹啉修饰的核苷酸取代。The modified target-specific nucleic acid molecule of claim 12 or 13, wherein the modified osteosclerostin-specific nucleic acid molecule is at position 6, position 8, position 11, and position corresponding to SEQ ID NO: 2 One or more nucleotides at position 13, 32 and/or 34 are substituted by the quinoline modified nucleotide.
  15. 权利要求12-14中任一项的经修饰的靶特异性核酸分子,其中所述经修饰的骨硬化素特异性核酸分子包含选自SEQ ID NO:3-24的核苷酸序列,其中x为所述喹啉修饰的核苷酸。The modified target-specific nucleic acid molecule of any one of claims 12-14, wherein the modified osteosclerostin-specific nucleic acid molecule comprises a nucleotide sequence selected from SEQ ID NOs: 3-24, wherein x It is the quinoline modified nucleotide.
  16. 权利要求12-15中任一项的经修饰的靶特异性核酸分子,其中所述经修饰的骨硬化素特异性核酸分子包含选自SEQ ID NO:8、10、11、16和18的核苷酸序列,其中x为所述喹啉修饰的核苷酸。The modified target-specific nucleic acid molecule of any one of claims 12-15, wherein the modified osteosclerostin-specific nucleic acid molecule comprises a core selected from the group consisting of SEQ ID NOs: 8, 10, 11, 16 and 18 A nucleotide sequence, where x is the quinoline-modified nucleotide.
  17. 权利要求12-16中任一项的经修饰的靶特异性核酸分子,其中所述喹啉修饰的核苷酸包含下式II的结构:
    The modified target-specific nucleic acid molecule of any one of claims 12-16, wherein the quinoline-modified nucleotide comprises the structure of Formula II:
    其中, in,
  18. 一种组合物,其包含权利要求1-17中任一项的经修饰的靶特异性核酸分子,以及任选的药学上或生理上可接受的载体。A composition comprising the modified target-specific nucleic acid molecule of any one of claims 1-17, and optionally a pharmaceutically or physiologically acceptable carrier.
  19. 一种提高靶特异性核酸分子与靶的结合亲和力的方法,所述方法包括用一或多个喹啉修饰的核苷酸取代所述靶特异性核酸分子中的一个或多个天然存在的核苷酸。A method of increasing the binding affinity of a target-specific nucleic acid molecule to a target, the method comprising replacing one or more naturally occurring nuclei in the target-specific nucleic acid molecule with one or more quinoline-modified nucleotides glycosides.
  20. 权利要求19的方法,其中所述喹啉修饰是喹啉-2-基取代、喹啉-3-基取代、喹啉-4-基取代、喹啉-5-基取代、喹啉-6-基取代、喹啉-7-基取代或喹啉-8-基取代,优选地,所述喹啉修饰是喹啉-5-基取代。The method of claim 19, wherein the quinoline modification is quinolin-2-yl substitution, quinolin-3-yl substitution, quinolin-4-yl substitution, quinolin-5-yl substitution, quinolin-6-yl substitution base substitution, quinolin-7-yl substitution or quinolin-8-yl substitution. Preferably, the quinoline modification is quinolin-5-yl substitution.
  21. 权利要求19或20的方法,其中所述喹啉修饰的核苷酸包含取代基团R,所述R选自以下a-g:
    The method of claim 19 or 20, wherein the quinoline modified nucleotide comprises a substituent group R selected from the following ag:
    优选地, Preferably,
  22. 权利要求19-21中任一项的方法,其中所述喹啉修饰的核苷酸包括以下式I的碱基部分:
    The method of any one of claims 19-21, wherein the quinoline modified nucleotide comprises the base moiety of Formula I:
    其中
    in
    优选地, Preferably,
  23. 权利要求19-22中任一项的方法,其中所述喹啉修饰的核苷酸包括以下式II的结构:
    The method of any one of claims 19-22, wherein the quinoline modified nucleotide comprises the following structure of Formula II:
    其中
    in
    优选地, Preferably,
  24. 权利要求19-23中任一项的方法,所述一或多个喹啉修饰的核苷酸取代在化学合成所述靶特异性核酸分子期间引入。The method of any one of claims 19-23, said one or more quinoline modified nucleotide substitutions being introduced during chemical synthesis of said target-specific nucleic acid molecule.
  25. 权利要求24的方法,其中在化学合成所述靶特异性核酸分子期间加入选自以下2a-2g的经修饰的亚磷酰胺单体:
    The method of claim 24, wherein modified phosphoramidite monomers selected from the following 2a-2g are added during chemical synthesis of the target-specific nucleic acid molecules:
  26. 一种选自以下2a-2g的经修饰的亚磷酰胺单体,其用于提高靶特异性核酸分子与靶的结合亲和力:
    A modified phosphoramidite monomer selected from the following 2a-2g, which is used to increase the binding affinity of target-specific nucleic acid molecules to the target:
PCT/CN2023/102119 2022-06-23 2023-06-25 Target-specific nucleic acid molecule having quinoline modification WO2023246935A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNPCT/CN2022/100727 2022-06-23
CN2022100727 2022-06-23

Publications (1)

Publication Number Publication Date
WO2023246935A1 true WO2023246935A1 (en) 2023-12-28

Family

ID=89379205

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/102119 WO2023246935A1 (en) 2022-06-23 2023-06-25 Target-specific nucleic acid molecule having quinoline modification

Country Status (1)

Country Link
WO (1) WO2023246935A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103154014A (en) * 2010-04-28 2013-06-12 Isis制药公司 Modified nucleosides and oligomeric compounds prepared therefrom
CN103492569A (en) * 2010-11-05 2014-01-01 米拉根医疗公司 Base modified oligonucleotides
CN104053777A (en) * 2011-11-18 2014-09-17 塔古西库斯生物株式会社 Nucleic acid fragment binding to target protein
CN104975020A (en) * 2008-02-11 2015-10-14 阿克赛医药公司 Modified Rnai Polynucleotides And Uses Thereof
CN107353317A (en) * 2010-05-28 2017-11-17 萨勒普塔医疗公司 The oligonucleotide analogs of key and/or end group between subunit with modification
CN110997917A (en) * 2017-12-01 2020-04-10 苏州瑞博生物技术有限公司 Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
CN113025617A (en) * 2021-04-30 2021-06-25 福建农林大学 Nucleic acid aptamer for specifically recognizing quinclorac and application thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104975020A (en) * 2008-02-11 2015-10-14 阿克赛医药公司 Modified Rnai Polynucleotides And Uses Thereof
CN103154014A (en) * 2010-04-28 2013-06-12 Isis制药公司 Modified nucleosides and oligomeric compounds prepared therefrom
CN107353317A (en) * 2010-05-28 2017-11-17 萨勒普塔医疗公司 The oligonucleotide analogs of key and/or end group between subunit with modification
CN103492569A (en) * 2010-11-05 2014-01-01 米拉根医疗公司 Base modified oligonucleotides
CN104053777A (en) * 2011-11-18 2014-09-17 塔古西库斯生物株式会社 Nucleic acid fragment binding to target protein
CN110997917A (en) * 2017-12-01 2020-04-10 苏州瑞博生物技术有限公司 Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
CN113025617A (en) * 2021-04-30 2021-06-25 福建农林大学 Nucleic acid aptamer for specifically recognizing quinclorac and application thereof

Similar Documents

Publication Publication Date Title
Pal et al. Molecular imaging of EGFR kinase activity in tumors with 124 I-labeled small molecular tracer and positron emission tomography
TWI686391B (en) New compounds
CN104507944B (en) Pteridines as FGFR inhibitors
CN104507931B (en) Noval chemical compound
US20200338201A1 (en) Conjugates and preparation and use thereof
RU2688435C2 (en) Kit for synthesis reaction mixture synthesis 3′-o-propargyl-modified nucleic acid
CN107286077B (en) Selective C-KIT kinase inhibitor
CN104011052B (en) Compound
JP5603933B2 (en) Nucleic acid aptamer capable of specifically binding to pancreatic cancer cells or tissues and use thereof
JP6898919B2 (en) New compound
Roy et al. Interaction of individual structural domains of hnRNP LL with the BCL2 promoter i-motif DNA
EP3719125A1 (en) Nucleic acid, composition and conjugate containing same, and preparation method and use
SA517380651B1 (en) Inhibitors of Lysine Specific Demethylase-1
KR20090090365A (en) Pyridine or pyrimidine derivative having excellent cell growth inhibition effect and excellent anti-tumor effect on cell strain having amplification of hgfr gene
CN104725362A (en) Pyrazolyl quinazoline kinase inhibitors
US10155946B2 (en) Particle-nucleic acid conjugates and therapeutic uses related thereto
US9096856B2 (en) Nucleotides and aptamers containing boronic acid groups having biased binding to glycosylated proteins, and uses thereof
WO2014007438A1 (en) Aptamer for periostin and anti-cancer composition including same
WO2023246935A1 (en) Target-specific nucleic acid molecule having quinoline modification
KR20220059472A (en) RNA-targeting ligands, compositions thereof, and methods of making and using same
WO2014071824A1 (en) 4-quinazolinamine heterocyclic compound and use thereof
CN104177342B (en) The indoles and naphthalene ketone derivant and its medical usage of heterocyclic radical substitution
JP6896711B2 (en) Biheteroaryl substitutions 1,4-benzodiazepines and their use for the treatment of cancer
Ding et al. Synthesis, and biological evaluation of EGFR/HER2-NAMPT conjugates for tumor treatment
CN107446012B (en) Tumor fluorescent imaging agent, preparation and application thereof

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: 23826580

Country of ref document: EP

Kind code of ref document: A1