WO2024088190A1 - 一种抑制lpa基因表达的rna抑制剂及其应用 - Google Patents

一种抑制lpa基因表达的rna抑制剂及其应用 Download PDF

Info

Publication number
WO2024088190A1
WO2024088190A1 PCT/CN2023/125861 CN2023125861W WO2024088190A1 WO 2024088190 A1 WO2024088190 A1 WO 2024088190A1 CN 2023125861 W CN2023125861 W CN 2023125861W WO 2024088190 A1 WO2024088190 A1 WO 2024088190A1
Authority
WO
WIPO (PCT)
Prior art keywords
mvip
nucleotides
antisense strand
rna inhibitor
sense strand
Prior art date
Application number
PCT/CN2023/125861
Other languages
English (en)
French (fr)
Inventor
崔坤元
卢雪琴
Original Assignee
厦门甘宝利生物医药有限公司
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 厦门甘宝利生物医药有限公司 filed Critical 厦门甘宝利生物医药有限公司
Publication of WO2024088190A1 publication Critical patent/WO2024088190A1/zh

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
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing

Definitions

  • the present invention belongs to the field of biomedicine, and in particular relates to an RNA inhibitor for inhibiting LPA gene expression and an application thereof.
  • RNAi RNA interference
  • dsRNA double-stranded RNA
  • RNA inhibitor process a nuclease called “Dicer” cuts or “dices” the long-chain dsRNA into small fragments of 21 to 25 nucleotides. These small fragments, called small interfering RNA (siRNA), have their antisense strands loaded onto the Argonaute protein (AGO2).
  • AGO2 loading occurs in the RISC-loading complex, a ternary complex consisting of the Argonaute protein, Dicer, and the dsRNA binding protein (TRBP for short).
  • the sense strand Passenger strand
  • Passenger strand is cleaved and expelled by AGO2.
  • AGO2 then uses the antisense strand to bind to mRNAs containing fully complementary sequences and then catalyzes the cleavage of these mRNAs, causing the mRNA split to lose its role as a translation template, thereby preventing the synthesis of related proteins. After cleavage, the cleaved mRNA is released, and the RISC-loading complex loaded with the antisense strand is recycled for another round of cleavage.
  • RNAi technology treats diseases at the mRNA level, which is more efficient than chemical small molecule drugs and biological macromolecule drugs at the protein level.
  • RNAi technology we can design the sense and antisense chain sequences of siRNA with high specificity and good inhibitory effect according to specific gene sequences, synthesize these single-stranded sequences through solid phase, and then pair the sense and antisense chains into siRNA according to the base pairing principle in a specific annealing buffer, and finally deliver them to the corresponding target in the body through a carrier system to degrade the target mRNA and destroy the function of the target mRNA as a translation template, thereby preventing the synthesis of related proteins.
  • siRNA is unstable in blood and tissues and is easily degraded by nucleases.
  • the sense strand and/or antisense strand of siRNA can be modified, but these chemical modifications only provide limited protection from nuclease degradation and may ultimately affect the activity of siRNA. Therefore, a corresponding delivery system is also needed to ensure that siRNA passes through the cell membrane safely and efficiently. Since siRNA has a large molecular weight, carries a large amount of negative charge, and has high water solubility, it cannot pass through the cell membrane smoothly and reach the cell.
  • liposomes The basic structure of liposomes is composed of a hydrophilic core and a phospholipid bilayer. It has a phospholipid bilayer similar to a biological membrane and has high biocompatibility. Therefore, liposomes have become the most popular and widely used siRNA carrier.
  • Liposome-mediated siRNA delivery mainly encapsulates siRNA into liposomes to protect siRNA from degradation by nucleases, improve the efficiency of siRNA passing through cell membrane barriers, and thus promote cell absorption.
  • anionic liposomes, pH-sensitive liposomes, immunoliposomes Although some progress has been made in fusogenic liposomes and cationic lipids, liposomes themselves are prone to induce inflammatory responses.
  • a variety of antihistamines and hormones such as cilitizine and dexamethasone must be used to reduce possible acute inflammatory responses. Therefore, they are not suitable for all treatment areas in actual clinical applications, especially in the treatment of some chronic diseases.
  • the cumulative toxicity that may occur due to long-term use is a potential safety hazard. Therefore, a safer and more effective carrier system is needed to deliver siRNA.
  • the asialoglycoprotein receptor (ASGPR) in the liver is a receptor specifically expressed by hepatocytes and is a highly efficient endocytic receptor. Since the secondary end of various glycoproteins exposed after enzyme or acid hydrolysis of sialic acid in the body under physiological conditions is a galactose residue, the sugar that ASGPR specifically binds is galactosyl, so it is also called galactose-specific receptor. Monosaccharide and polysaccharide molecules such as galactose, galactosamine, and N-acetylgalactosamine have high affinity for ASGPR.
  • ASGPR The main physiological function of ASGPR is to mediate the clearance of asialoglycoproteins, lipoproteins and other substances in the blood, and it is closely related to the occurrence and development of liver diseases such as viral hepatitis, cirrhosis, and liver cancer.
  • liver diseases such as viral hepatitis, cirrhosis, and liver cancer.
  • the discovery of this characteristic of ASGPR plays an important role in the diagnosis and treatment of hepatic diseases (Ashwell G, Harford J, Carbohydrate specific Receptors of the Liver, Ann Rev Biochem 1982 51:531-554).
  • the therapeutic drugs for liver-derived diseases containing galactose or galactosamine and their derivatives in the structure can specifically bind to ASGPR, thereby having active liver targeting and requiring no other carrier system for delivery.
  • LPA is the name of the gene encoding apolipoprotein (a) (apo(a)), which is mainly expressed in the liver, and its expression is limited to humans and non-primates.
  • the hydrophilic apolipoprotein component apolipoprotein (a) is attached to apo(B)-100 through a disulfide bond and is combined with the lipid core to form lipoprotein (a) (Lp(a)) particles.
  • Lp(a) particles are a special type of cholesterol-rich macromolecular lipoprotein, with the surface coated by cholesterol and phospholipids, and embedded with apolipoprotein (a) and apo(B)-100 as described above.
  • Lp(a) can enter and deposit on the blood vessel wall, which has the effect of promoting atherosclerosis.
  • Lp(a) is structurally homologous to plasminogen (PLG) and can compete with plasminogen for binding to fibrin sites, thereby inhibiting fibrinogen hydrolysis and promoting thrombosis. Therefore, LP(a) is closely related to atherosclerosis and thrombosis. Studies have shown that the level of Lp(a) in the blood is an independent risk factor for cardiovascular disease, stroke and atherosclerotic stenosis.
  • High Lp(a) levels are mainly related to genetics and will not change significantly with diet, exercise and other lifestyle changes. Lp(a) levels in humans greater than 300 mg/L are considered high. High Lp(a) levels often indicate a significantly increased risk of atherosclerosis and thrombosis. Testing Lp(a) is of great significance for early identification of the risk of atherosclerosis. There are approximately 330 million people suffering from cardiovascular diseases in China, but the general public's awareness, treatment and control rates of dyslipidemia are generally at a low level, and the awareness of Lp(a) risks is even lower. Most hospitals do not include this item in their routine blood lipid tests, and there are currently no targeted therapeutic drugs in clinical practice at home and abroad. Therefore, there is an urgent need in this field for a drug that effectively inhibits LPA gene expression.
  • the present invention provides an RNA inhibitor for inhibiting LPA gene expression or a pharmaceutically acceptable salt thereof.
  • the RNA inhibitor of the present invention is formed by base pairing of a sense strand and an antisense strand with a chain length of 15-30, preferably 19-23, and at least 85% of the bases between the sense strand and the antisense strand are complementary; the -OH at the 2' position of the nucleotide sugar of some or all of the sense strands and/or the antisense strands can be substituted, wherein the substituent group is fluorine or methoxy, and the phosphate bonds between three adjacent nucleotides at at least one of the ends of the sense strand and/or the antisense strand can be thiolated.
  • the RNA inhibitor or a pharmaceutically acceptable salt thereof of the present invention wherein the antisense strand forms a complementary region with a target sequence
  • the target sequence is a plurality of regions at different positions of LPA mRNA, the plurality of regions having at least 15 identical consecutive nucleotides
  • the target sequence is selected from any one of nucleotide regions between 312-332, 654-674, 996-1016, 1338-1358, 1680-1700, 2022-2042 and 2364-2384 in LPA mRNA (NM_005577.4).
  • the starting positions of these regions may vary depending on the version number of LPA mRNA, such as the nucleotide region between 654-674 in LPA mRNA NM_005577.4.
  • the RNA inhibitor of the present invention or a pharmaceutically acceptable salt thereof wherein the antisense strand forms a complementary region with a target sequence, the target sequence is a plurality of regions at different positions of LPA mRNA, the plurality of regions have at least 15 identical consecutive nucleotides, and the target sequence is selected from any one of the nucleotide regions between 493-512, 1861-1880, and 2203-2222 in LPA mRNA (NM_005577.4).
  • the starting positions of these regions may vary depending on the version number of LPA mRNA, such as the nucleotide region between 1861-1880 in LPA mRNA NM_005577.4.
  • the antisense strand of the RNA inhibitor or a pharmaceutically acceptable salt thereof of the present invention is selected from the following sequences:
  • g guanylate
  • a adenylate
  • u uridylate
  • c cytidine
  • the sense strand of the RNA inhibitor or a pharmaceutically acceptable salt thereof is selected from the following sequences:
  • g guanylate
  • a adenylate
  • u uridylate
  • c cytidine
  • the sense strand of the RNA inhibitor of the present invention or a pharmaceutically acceptable salt thereof is SEQ ID NO.11 or a sequence having at least 15 consecutive nucleotides identical thereto, or a sequence differing therefrom by one, two, or three nucleotides; and the antisense strand is SEQ ID NO.25 or a sequence having at least 15 consecutive nucleotides identical thereto, or a sequence differing therefrom by one, two, or three nucleotides:
  • Antisense strand 5'ucguauaacaauaaggagcug 3' SEQ ID NO.25;
  • the sense strand is SEQ ID NO.8 or a sequence having at least 15 consecutive nucleotides identical thereto, or a sequence differing therefrom by one, two or three nucleotides
  • the antisense strand is SEQ ID NO.21 or a sequence having at least 15 consecutive nucleotides identical thereto, or a sequence differing therefrom by one, two or three nucleotides:
  • Antisense strand 5'auaacucuguccauuaccaug 3' SEQ ID NO.21;
  • g guanylate
  • a adenylate
  • u uridylate
  • c cytidine
  • the RNA inhibitor of the present invention or a pharmaceutically acceptable salt thereof wherein the sense strand is SEQ ID NO.270 or a sequence that differs therefrom by one, two or three nucleotides, and the antisense strand is SEQ ID NO.278 or a sequence that differs therefrom by one, two or three nucleotides:
  • Antisense strand 5'UsfCsGfUAfUAACAAfUAfAGfGAfGCsfUsG 3' SEQ ID NO.278;
  • the sense strand is SEQ ID NO.239 or a sequence that differs therefrom by one, two or three nucleotides
  • the antisense strand is SEQ ID NO.344 or a sequence that differs therefrom by one, two or three nucleotides:
  • Antisense strand 5'AsfUsAfACdTCfUGUCCAfUUfACCAsUsG 3' SEQ ID NO.344;
  • G 2'-O-methylguanylate
  • A 2'-O-methyladenylate
  • U 2'-O-methyluridylate
  • C 2'-O-methylcytidine
  • Gs 2'-O-methyl-3'-thioguanylate
  • As 2'-O-methyl-3'-thioadenylate
  • Us 2'-O-methyl-3'-thiouridylate
  • Cs 2'-O-methyl-3'-thiocytidine
  • fG 2'-fluoroguanylic acid
  • fA 2'-fluoroadenylic acid
  • fU 2'-fluorouridylic acid
  • fC 2'-fluorocytidylic acid
  • fGs 2'-fluoro-3'-thioguanylic acid
  • fAs 2'-fluoro-3'-thioadenylic acid
  • fUs 2'-fluoro-3'-thiouridylic acid
  • the sense strand or antisense strand of the RNA inhibitors described herein can accommodate no more than 3 mismatched nucleotides, for example, within 5, 4, 3, or 2 nucleotides of the 5' end and/or 3' end.
  • RNA inhibitor or its pharmaceutically acceptable salt described in the present invention further contains The structure of the RNA inhibitor is shown in Formula Ia, Ib or Ic:
  • the vector structure includes 5'MVIP (5'MultiValent Import Platform) and 3'MVIP (3'MultiValent Import Platform);
  • the 5'MVIP is composed of a transfer point R 1 , a connecting chain D, a linker B, a side chain L and a liver-targeting specific ligand X, which is connected to the 5' end of the sense chain or the 5' end of the antisense chain through the transfer point R 1. Its structure is shown in general formula I:
  • the 3'MVIP is composed of a transfer point R 2 , a connecting chain D, a linker B, a side chain L and a liver-targeting specific ligand X, which is connected to the 3' end of the sense chain or the 3' end of the antisense chain through the transfer point R 2 , and its structure is shown in general formula II:
  • the transition point R1 is a heterocyclic or carbocyclic structure containing N, S or O as shown below:
  • R 1 is -NH(CH 2 ) x CH 2 O-, wherein x is any integer from 3 to 12, preferably any integer from 4 to 6;
  • the transition point R2 is a heterocyclic or carbocyclic structure containing N, S or O as shown below:
  • transition point R 2 is -NH(CH 2 ) x1 CH(OH)(CH 2 ) x2 CH 2 O-, wherein x1 is any integer from 1 to 4, and x2 is any integer from 0 to 4;
  • the liver-targeting specific ligand X is selected from a structure used to enhance the uptake of RNA inhibitors by hepatocytes, and is the same or different within each of 5'MVIP and 3'MVIP or between 5'MVIP and 3'MVIP, and is selected from monosaccharides and their derivatives, preferably N-acetylgalactosamine and its derivatives, and more preferably selected from the following structures:
  • W is selected from one or two of -OH, -NHCOOH and -NHCO(CH 2 ) q CH 3 , wherein q is an integer of 0-4;
  • the branched chain L is the same or different within each of the 5'MVIP and the 3'MVIP or between the 5'MVIP and the 3'MVIP, and is selected from one or more of the following structures:
  • r1 is any integer from 1 to 12
  • r2 is any integer from 0 to 20
  • Z is H, an alkyl group or an amide group, wherein the alkyl group is, for example, a C 1 -C 5 alkyl group;
  • the linker B is the same or different within each of the 5'MVIP and the 3'MVIP or between the 5'MVIP and the 3'MVIP, and is selected from the following structures:
  • A1 and A2 are each independently C, O, S, -NH-, carbonyl, amide, phosphoryl or thiophosphoryl, and r is any integer from 0 to 4;
  • the connecting chain D is the same or different within each of the 5'MVIP and the 3'MVIP or between the 5'MVIP and the 3'MVIP, and is selected from the following structures:
  • the 5’MVIP is selected from any one of 5’MVIP01 to 5’MVIP22 in Table 11.
  • the 3’MVIP is selected from any one of 3’MVIP01 to 3’MVIP27 in Table 12.
  • the RNA inhibitor of the present invention or a pharmaceutically acceptable salt thereof wherein the 5'MVIP is 5'MVIP01 or 5'MVIP09 as shown below, and the 3'MVIP is 3'MVIP01, 3'MVIP09 or 3'MVIP17:
  • the RNA inhibitor of the present invention or a pharmaceutically acceptable salt thereof wherein the combination of the sense chain 5’MVIP and the antisense chain 3’MVIP is 5’MVIP01/3’MVIP01, 5’MVIP01/3’MVIP17 or 5’MVIP09/3’MVIP09, or the combination of the sense chain 5’MVIP and the antisense chain 3’MVIP is 5’MVIP01/3'MVIP09 or 5’MVIP09/3’MVIP01.
  • the present invention also provides the use of the above-mentioned RNA inhibitor or a pharmaceutically acceptable salt thereof in the preparation of a medicament for treating and/or preventing diseases associated with elevated LP(a) levels, wherein the diseases associated with elevated LP(a) levels include hepatovascular diseases, including inflammatory diseases, cardiovascular and cerebrovascular diseases, and metabolic diseases, wherein the cardiovascular and cerebrovascular diseases include hyperLP(a)emia, hyperlipidemia, stroke, atherosclerosis, thrombosis, coronary heart disease, and aortic valve stenosis.
  • diseases associated with elevated LP(a) levels include hepatovascular diseases, including inflammatory diseases, cardiovascular and cerebrovascular diseases, and metabolic diseases, wherein the cardiovascular and cerebrovascular diseases include hyperLP(a)emia, hyperlipidemia, stroke, atherosclerosis, thrombosis, coronary heart disease, and aortic valve stenosis.
  • the present invention provides a pharmaceutical composition, which comprises the above-mentioned RNA inhibitor for inhibiting LPA gene expression or a pharmaceutically acceptable salt thereof and optional pharmaceutically acceptable excipients, wherein the pharmaceutically acceptable excipients may be pharmaceutically acceptable excipients, carriers and/or diluents, and the dosage form of the pharmaceutical composition is an oral agent, an intravenous injection, or a subcutaneous or intramuscular injection, preferably a subcutaneous injection.
  • the present invention also provides a method for treating and/or preventing diseases, disorders or syndromes associated with elevated levels of LP(a), the method comprising administering to a subject or patient in need thereof a therapeutically effective amount of an RNA inhibitor or a pharmaceutically acceptable salt thereof that inhibits LPA gene expression, or a pharmaceutical composition comprising the RNA inhibitor or a pharmaceutically acceptable salt thereof and optional pharmaceutically acceptable excipients, wherein the therapeutically effective amount is 1.0 mg/kg to 10 mg/kg of the above-mentioned RNA inhibitor or a pharmaceutically acceptable salt thereof.
  • the mode of administration (administration mode) to the subject or patient includes oral, intravenous, subcutaneous or intramuscular injection, rectal or intraperitoneal application.
  • FIG1 is a graph showing the inhibitory effect of the RNA inhibitors in Table 3 on the LPA mRNA level in Huh7 cells at different concentrations obtained in Example 2 of the present application;
  • FIG2 is a high-resolution mass spectrum of ERCd-01-c2 synthesized in 3.1.15 in Example 3 of the present application;
  • FIG3 is a high-resolution mass spectrum of 3'MVIP17-c1 synthesized in 3.1.2.6 of Example 3 of the present application;
  • FIG4 is a high-resolution mass spectrum of 5'MVIP09-ERCd-PFP-c2 synthesized in 3.2.1.2 of Example 3 of the present application;
  • FIG5 is a graph showing the inhibitory effect of the RNA inhibitor in Table 4 on the LPA mRNA level in Huh7 cells at different concentrations obtained in Example 5 of the present application;
  • FIG6 is a graph showing the inhibitory effect of the RNA inhibitor in Table 5 on the LPA mRNA level in Huh7 cells at different concentrations obtained in Example 6 of the present application;
  • FIG7 is a graph showing the inhibitory effect of the RNA inhibitor in Table 6 on the LPA mRNA level in Huh7 cells at different concentrations obtained in Example 7 of the present application;
  • FIG8 is a graph showing the inhibitory effect of the RNA inhibitor in Table 7 on the LPA mRNA level in Huh7 cells at different concentrations obtained in Example 8 of the present application;
  • FIG9 is a graph showing the inhibitory effect of the RNA inhibitor in Table 8 on the LPA mRNA level in Huh7 cells at different concentrations obtained in Example 9 of the present application;
  • FIG10 is a graph showing the effect of the RNA inhibitor in Example 10 of the present application on reducing the LDL-c level in the plasma of cynomolgus monkeys;
  • FIG. 11 is a graph showing the effect of the RNA inhibitor in Example 10 of the present application on reducing Lp(a) levels in cynomolgus monkey plasma.
  • LPA includes human LPA and cynomolgus monkey LPA, where the human LPA mRNA sequence can be found in, for example, GenBank NM_005577.4.
  • target sequence refers to a continuous portion of the nucleotide sequence of the mRNA molecule formed during the transcription of the LPA gene, including mRNA that is a product of RNA processing of the primary transcription product.
  • the target portion of the sequence will be at least long enough to serve as a substrate for RNA inhibitor-directed degradation at or near the portion of the nucleotide sequence of the mRNA molecule formed during the transcription of the LPA gene.
  • the length of the "target sequence” is generally about 15-30 nucleotides.
  • region refers to the starting position to the ending position of the target sequence in the LPA mRNA included in GenBank.
  • region 312-332 refers to the nucleotide from the 312th position to the 332nd position in the LPA mRNA (NM_005577.4).
  • RNA inhibitor generally refers to an agent comprising RNA as defined in the present terminology, and which can mediate targeted cleavage of RNA transcripts through the RNA-induced silencing complex (RISC) pathway.
  • RISC RNA-induced silencing complex
  • the RNA inhibitor can be a single-stranded siRNA (ssRNA inhibitor) introduced into a cell or organism to inhibit a target mRNA (i.e., the LPA gene).
  • the single-stranded RNA inhibitor binds to the RISC endonuclease Argonaute 2, which then cleaves the target mRNA.
  • Single-stranded siRNAs are generally 15 to 30 nucleotides in length and are chemically modified.
  • the "RNA inhibitor” used in the present application is a double-stranded RNA, and is referred to as a "double-stranded RNA inhibitor", “double-stranded RNA (dsRNA, DS) molecule", “dsRNA agent” or “dsRNA” in the present invention.
  • dsRNA refers to a complex of ribonucleic acid molecules, which has a duplex structure comprising two antiparallel and substantially complementary nucleic acid strands, referred to as having "Sense” and “antisense” orientation relative to the target mRNA.
  • double-stranded RNA dsRNA triggers degradation of the target mRNA through a post-transcriptional gene silencing mechanism (referred to herein as RNA inhibition or RNA interference).
  • the duplex structure can be any length that triggers specific degradation of LPA mRNA through the RISC pathway, and can be in the range of about 15 to 36 base pairs in length, for example, about 15-30 base pairs in length, for example, about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or 36 base pairs in length.
  • the RNA inhibitor of the present application is a dsRNA of 15-30 nucleotides that interacts with the target sequence to guide the cleavage of LPA mRNA.
  • nucleotides in the sense strand and antisense strand of a dsRNA molecule are ribonucleotides, but as described in detail in the present invention, one or more non-ribonucleotides, such as deoxyribonucleotides or modified nucleotides, may also be included.
  • the RNA inhibitors involved in this specification may include chemically modified ribonucleotides, which may have modified nucleotides in multiple regions.
  • modified nucleotides used in the present invention means independently having a modified sugar moiety, a modified internucleotide connection or a modified nucleobase, or a nucleotide in any combination thereof.
  • modified nucleotides encompasses replacement, addition or removal of, for example, functional groups or atoms of internucleotide connections, sugar moieties or nucleobases. Modifications applicable to the RNA inhibitors of the present application include all types of modifications disclosed in the present invention or known in the art.
  • nucleotide sequence generally refers to a series or a certain order of nucleotides, whether modified or unmodified, using standard nucleotide nomenclature and the symbol table of modified nucleotides described in this application as a series of letters.
  • the nucleotide sequence described in this application is a polymer composed of phosphodiester bonds (or its related structural variants or synthetic analogs), including naturally occurring nucleotide polymers, but it should be understood that the scope of the term also includes various analogs, including but not limited to: peptide nucleic acids (PNA), aminophosphoroesters, thiophosphates, methylphosphonates and 2'-O-methyl ribonucleic acids, etc. There are usually about 15-30 nucleotides, but the term can also refer to molecules of any length.
  • the nucleotide sequence comprises one or more unmodified ribonucleosides (RNA) and/or unmodified deoxyribonucleosides (DNA) and/or one or more modified nucleotides.
  • modified nucleotide sequence generally means a series or a certain order of nucleotides comprising at least one modification and/or at least one modified internucleotide linkage.
  • modified nucleotide generally means a nucleotide comprising at least one chemical modification compared to a naturally occurring RNA or DNA nucleotide.
  • 2'-deoxy-thymidylate 2'-O-methyl modified nucleotides, 2'-fluoro modified nucleotides, 2'-deoxy-modified nucleotides, locked nucleotides, abasic nucleotides, 2'-amino-modified nucleotides, 2'-O-long chain alkyl-modified nucleotides (such as hexadecyl), morpholino nucleotides, phosphoramidate nucleotides, non-natural nucleobase nucleotides, 5'-phosphorothioate nucleotides, and nucleotides attached with a cholesterol derivative or a dodecanoic acid didecylamide group.
  • Modified nucleotides contain modified sugar groups and/or modified nucleobases.
  • nucleobase or “base” generally refers to a heterocyclic pyrimidine or purine compound, which is a component of all nucleic acids and includes adenine, guanine, cytosine, thymine and uracil. Nucleotides can include modified nucleotides or nucleotide mimetics, abasics or surrogate replacement parts.
  • unmodified nucleobase or “naturally occurring nucleobase” generally refers to the naturally occurring heterocyclic nucleobases of RNA or DNA: the purine bases adenine and guanine; and the pyrimidine bases thymine, cytosine and uracil.
  • Modified nucleobase generally refers to any nucleobase that is not a naturally occurring nucleobase.
  • sugar generally refers to a naturally occurring sugar or a modified sugar of a nucleotide.
  • naturally occurring sugar generally refers to a ribofuranosyl as found in naturally occurring RNA or a deoxyribofuranosyl as found in naturally occurring DNA.
  • Modified sugar refers to a substituted sugar or sugar surrogate, for example, a fluoro or methoxy substitution at the 2' position of the sugar.
  • internucleotide linkage generally means a covalent linkage between adjacent nucleotides in a nucleotide sequence.
  • Naturalally occurring internucleotide linkage means a 3' to 5' phosphodiester linkage.
  • Modified internucleotide linkage means any internucleotide linkage other than a naturally occurring internucleotide linkage.
  • AS antisense strand
  • RNA inhibitor e.g., dsRNA
  • region of complementarity generally refers to a region on the antisense strand that is substantially complementary to a sequence defined in the present application (e.g., a target sequence).
  • the term "sense strand” generally refers to a strand of an RNA inhibitor (e.g., dsRNA) that Includes a region that is substantially complementary to the region of the "antisense strand” (AS).
  • the "sense” strand is sometimes referred to as the “sense” strand, the “passenger” strand or the "anti-guide” strand.
  • the antisense strand targets the desired mRNA, while the sense strand may target different targets or be degraded. Therefore, if the antisense strand is incorporated into RISC, the correct target is targeted.
  • the incorporation of the sense strand can result in off-target effects. These off-target effects can be limited by using modifications or using 5' end caps on the sense strand.
  • the term “complementary” refers to the ability of two nucleotide sequences to hybridize under certain conditions, form base pair hydrogen bonds, and form a duplex or double helix structure.
  • the antisense strand of an RNA inhibitor hybridizes with the sense strand of an RNA inhibitor or LPA mRNA to form Watson-Crick base pairs or non-Watson-Crick base pairs, and includes natural or modified nucleotides or nucleotide mimetics.
  • “Complementary” does not necessarily have nucleobase complementarity on every nucleoside. On the contrary, some mismatches can be tolerated.
  • mismatch refers to when the complementary region is not completely complementary to the target sequence, and the mismatch can be in the internal or terminal region of the molecule. Usually, the most tolerated mismatch is in the terminal region, for example, within 5, 4, 3 or 2 nucleotides of the 5' end and/or 3' end, and no more than 3 mismatches.
  • the term "ligand” generally refers to any compound or molecule that can covalently or otherwise chemically bind to a biologically active substance (such as dsRNA).
  • the ligand can interact directly or indirectly with another compound such as a receptor, and the receptor that interacts with the ligand can be present on the cell surface, or alternatively can be an intracellular and/or intercellular receptor, and the interaction of the ligand with the receptor can result in a biochemical reaction, or can be simply a physical interaction or binding.
  • the term "pharmaceutically acceptable” generally refers to one or more non-toxic substances that do not inhibit the effectiveness of the biological activity of the active ingredient.
  • Such preparations may generally contain salts, excipients, buffers, preservatives, compatible carriers and optional other therapeutic agents.
  • Such pharmaceutically acceptable preparations may also generally contain compatible solid or liquid fillers, diluents or encapsulation materials suitable for administration to people.
  • salts should be pharmaceutically acceptable salts, but non-pharmaceutically acceptable salts can be conveniently used to prepare pharmaceutically acceptable salts, and they cannot be excluded from the scope of this application.
  • Such pharmacological and pharmaceutically acceptable salts include, but are not limited to, salts prepared from the following acids: hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, maleic acid, acetic acid, salicylic acid, citric acid, boric acid, formic acid, malonic acid, succinic acid, etc.
  • Pharmaceutically acceptable salts may also be prepared into alkali metal salts or alkaline earth metal salts, such as sodium salts, potassium salts or calcium salts.
  • lipid nanoparticle generally refers to a vesicle comprising a lipid layer encapsulating a pharmacologically active molecule (e.g., dsRNA).
  • LNP is described in, for example, Chinese Patent No. CN103189057B, the entire contents of which are incorporated herein by reference.
  • the present invention provides an RNA inhibitor for inhibiting LPA gene expression or a pharmaceutically acceptable salt thereof.
  • the target sequence of the RNA inhibitor or a pharmaceutically acceptable salt thereof comprises multiple regions of LPA mRNA NM_005577.4 (SEQ ID NO.1):
  • g guanylate
  • a adenylate
  • t thymidylate
  • c cytidine
  • the target sequence of the RNA inhibitor described in the present invention or a pharmaceutically acceptable salt thereof includes the nucleotide region with double underline and single underline in SEQ ID NO.1.
  • the target sequence of the RNA inhibitor described in the present invention or a pharmaceutically acceptable salt thereof is a nucleotide region between 301-2401 in SEQ ID NO.1 and has a sequence of at least 15 consecutive nucleotides identical thereto.
  • the target sequence of the RNA inhibitor described in the present invention is at least 15 consecutive nucleotides in these regions.
  • the target sequence of the RNA inhibitor described in the present invention is at least 30 consecutive nucleotides extending before and after these regions.
  • the target sequence of the RNA inhibitor described in the present invention is a target sequence that differs from SEQ ID NO.2 by one, two or three nucleotides.
  • the target sequence of the RNA inhibitor described in the present invention is a target sequence having at least 15 consecutive nucleotides identical to SEQ ID NO.2.
  • the target sequence of the RNA inhibitor described in the present invention is a target sequence that differs from SEQ ID NO.3 by one, two or three nucleotides.
  • the target sequence of the RNA inhibitor described in the present invention is a target sequence having at least 15 consecutive nucleotides identical to SEQ ID NO.3.
  • the target sequence of the RNA inhibitor described in the present invention can be a sequence of 15-30 nucleotides in any other region other than those described above in SEQ ID NO.1.
  • the RNA inhibitors described herein include double-stranded ribonucleic acid (dsRNA) molecules for inhibiting LPA gene expression in cells, such as cells of a subject (e.g., a mammal, such as a human susceptible to LPA-related disorders such as high LP(a) levels).
  • the antisense strand of the dsRNA includes a complementary region that is substantially complementary to the target sequence described above and is usually completely complementary.
  • the positive strand includes a region that is complementary to the antisense strand so that when combined under appropriate conditions, the two strands can hybridize and form a duplex structure.
  • the length of the duplex structure is 15 to 30 base pairs.
  • the length of the complementary region to the target sequence is 15 to 30 nucleotides.
  • the RNA inhibitor of the present invention is formed by base pairing of a sense strand and an antisense strand with a chain length of 15-30, wherein the chain length is preferably 19-23.
  • RNA inhibitor of the present invention there is at least 85% base complementarity between the sense strand and the antisense strand in the RNA inhibitor of the present invention
  • the sense strand of the RNA inhibitor of the present invention is selected from the sequences in Table 1, as shown in Table 1 below:
  • g guanylate
  • a adenylate
  • u uridylate
  • c cytidine
  • the antisense strand of the RNA inhibitor of the present invention is selected from the sequences in Table 2, as shown in Table 2 below:
  • g guanylate
  • a adenylate
  • u uridylate
  • c cytidine
  • t thymidylate
  • the base pairs of the sense strand in Table 1 and the corresponding antisense strand in Table 2 are complementary to form dsRNA, which may be partially complementary or completely complementary.
  • the partial complementarity may be at least 85% base pairing.
  • RNA inhibitor of the present invention is selected from the following Table 3:
  • the RNA inhibitor can be added to the cell line for sequence screening by cell transfection or liposome-nucleic acid nanoparticles, which are well known to those skilled in the art.
  • Patents US9233971B2, US9080186B2, CN102985548B and CN103189057B on lipid compounds and methods for preparing liposome-nucleic acid nanoparticles are fully introduced into this specification.
  • amphoteric lipids in the lipid compound are preferably macrocyclic lipid compounds D1C1, T1C1, T1C6, T4C4, B2C1, B2C6, B2C7 and M10C1.
  • dsRNAs having a duplex structure of about 20 to 23 base pairs, for example, 21 base pairs have been found to be particularly effective in inducing RNA inhibition (Elbashir et al., EMBO 2001, 20: 6877-6888). However, others have found that shorter or longer RNA duplex structures are also effective (Chu and Rana (2007) RNA 14: 1714-1719; Kim et al. (2005) Nat Biotech 23: 222-226). It is reasonable to expect that a duplex having a few nucleotides minus or added at one or both ends of a sequence in Tables 1, 2 and 3 may be similarly effective compared to the dsRNA.
  • inhibitory dsRNAs having a sequence of at least 15, 16, 17, 18, 19, 20, 21 or more consecutive nucleotides derived from a sequence in Tables 1, 2 and 3 and differing by no more than about 5, 10, 15, 20, 25 or 30% in their ability to inhibit LPA gene expression from a dsRNA comprising the entire sequence are included within the scope of the present application.
  • the dsRNA described in the present application may further include one or more single-stranded nucleotide overhangs, for example, 1, 2, 3 or 4 nucleotides.
  • the nucleotide overhangs may contain nucleotide/nucleoside analogs or a combination thereof, including deoxynucleotides.
  • the overhang may be on the sense strand, the antisense strand or a combination thereof.
  • the nucleotides of the overhang may be present at the 5' end, the 3' end or both ends of the antisense strand or the sense strand of the dsRNA.
  • the overhang may be formed by one strand being longer than the other, or by two strands of the same length being staggered.
  • the overhang is on the antisense strand and may form a mismatch or complementarity with LPA mRNA or may be another sequence.
  • the overhang is located at the 3' end of the sense strand, or alternatively, at the 3' end of the antisense strand.
  • the dsRNA may also have a blunt end, which means that there are no unpaired nucleotides at that end of the dsRNA, i.e., no nucleotide overhangs.
  • the blunt end may be located at the 5' end of the antisense strand and the 3' end of the sense strand, or vice versa, or a double-ended blunt end, which is a double-stranded dsRNA over its entire length, i.e., there are no nucleotide overhangs at either end of the molecule.
  • the sense strand or antisense strand of the dsRNA has a nucleotide overhang at the 3' end, the overhang contains 1, 2, 3 or 4 nucleotides, and the 5' end is blunt.
  • an overhang is present at the 3' end of both the sense strand and the antisense strand, and the overhang contains 1, 2, 3, or 4 nucleotides.
  • the dsRNA is a double-ended blunt-ended dsRNA of 19, 21, or 23 nucleotides in length, which is double-stranded throughout its entire length, ie, there are no nucleotide overhangs at either end of the molecule.
  • the dsRNA is 21 nucleotides in length, and both the sense and antisense strands have 2 nucleotide overhangs at the 3' end.
  • the sense strand and antisense strand of the RNA inhibitor may be modified without affecting its activity or even enhancing its activity, wherein the nucleotides may have a modifying group, and the entire strand or a portion thereof may be modified.
  • one or more nucleotides on the sense strand and/or antisense strand are modified to form modified nucleotides.
  • the sense strand and antisense strand of the RNA inhibitor (e.g., dsRNA) described herein are unmodified.
  • the sense strand and antisense strand of the RNA inhibitor described herein are chemically modified or coupled as known in the art and described herein to enhance stability or other favorable properties.
  • all nucleotides or substantially all nucleotides of the RNA inhibitor described herein may be modified, i.e., the strand of the RNA inhibitor has no more than 5, 4, 3, 2 or 1 unmodified nucleotides.
  • the sense strand and antisense strand of the RNA inhibitor as described in the present application can be synthesized and/or modified by methods known in the art, such as those described in "Current protocols in nucleic acid chemistry", Beaucage, S.L. et al. (eds.), John Wiley & Sons, Inc., New York, NY, USA, which is incorporated by reference into the present invention.
  • Modifications include, for example, terminal modifications, for example, 5' terminal modifications (phosphorylation, coupling, reverse connection) or 3' terminal modifications (coupling, DNA nucleotides, reverse connection, etc.); base modifications, for example, the use of stabilized bases, destabilized bases or the removal of bases (basic nucleotides) or coupled bases; sugar modifications (e.g., 2'-position or 4'-position) or sugar substitutions; or backbone modifications, including modifications or replacements of phosphodiester linkages.
  • the sense strand and antisense strand of the RNA inhibitor do not need to be uniformly modified, and one or more modifications can be incorporated into their individual nucleotides.
  • the modified nucleotide is selected from the group consisting of: deoxyribonucleotides, nucleotide mimetics, abasic nucleoside Nucleotides, 2'-modified nucleotides, 3' to 3' linked (inverted) nucleotides, nucleotides containing unnatural bases, bridged nucleotides, peptide nucleic acids (PNAs), unlocked nucleobase analogs, locked nucleotides, 3'-O-methoxy (2' internucleoside linkage) nucleotides, 2'-F-arabino nucleotides, 5'-Me/2'-fluoro nucleotides, morpholino nucleotides, vinylphosphonate deoxyribonucleotides, vinylphosphonate-containing nucleotides, and cyclopropylphosphonate-containing nucleotides.
  • PNAs peptide nucleic acids
  • the 2'-modified nucleotides include: 2'-O-methyl nucleotides, 2'-deoxy-2'-fluoro nucleotides, 2'-deoxy nucleotides, 2'-methoxyethyl nucleotides, 2'-amino nucleotides and/or 2'-alkyl nucleotides.
  • the 2' position of the nucleotide sugar group at at least two or more even-numbered positions starting from the 5' end of the antisense strand is fluorine.
  • all 2' positions of the nucleotide sugars at the even-numbered positions starting from the 5' end of the antisense strand are fluorine.
  • At least one of the 2' positions of the nucleotide sugars at positions 2, 4, 6, 8, 12, and 14 starting from the 5' end of the antisense strand is fluorine.
  • the 2' positions of the nucleotide sugars at positions 2, 4, 6, 8, 12, and 14 starting from the 5' end of the antisense strand are all fluorine.
  • At least one of the 2' positions of the remaining nucleotide sugar groups is a methoxy group.
  • At least one of the 2' positions of the remaining nucleotide sugar groups is a methoxy group.
  • At least one of the 2' positions of the remaining nucleotide sugar groups is a methoxy group.
  • At least two or more nucleotide sugar groups at odd-numbered positions starting from the 5' end of the sense strand have fluorine at the 2' position.
  • the 2' positions of the nucleotide sugars at the odd-numbered positions starting from the 5' end of the sense strand are all fluorine.
  • At least one of the 2' positions of the 5th, 7th, 8th, and 9th nucleotide sugars starting from the 5' end of the sense strand is fluorine.
  • the 2' positions of the 5th, 7th, 8th, and 9th nucleotide sugars starting from the 5' end of the sense strand are all fluorine.
  • At least one of the 2' positions of the remaining nucleotide sugar groups is a methoxy group.
  • At least one of the 2' positions of the sugar moieties at positions 7, 9, 10, and 11 starting from the 5' end of the sense strand is fluorine.
  • the 2' positions of the sugar moieties at positions 7, 9, 10, and 11 starting from the 5' end of the sense strand are all fluorine.
  • At least one of the 2' positions of the remaining nucleotide sugar groups is a methoxy group.
  • At least one of the 2' positions of the nucleotide sugars at positions 3, 5, 7, 9, 10, 11, 13, and 15 starting from the 5' end of the sense strand is fluorine.
  • the 2' positions of the nucleotide sugars at positions 3, 5, 7, 9, 10, 11, 13, and 15 starting from the 5' end of the sense strand are all fluorine.
  • At least one of the 2' positions of the remaining nucleotide sugar groups is a methoxy group.
  • At least one of the 2' positions of the remaining nucleotide sugar groups is a methoxy group.
  • the -OH at the 2' position of some or all of the nucleotide sugar groups of the sense strand and/or antisense strand can be substituted, wherein the substituent group is fluorine or methoxy, preferably the 2' position of the nucleotide sugar groups at positions 9, 10, and 11 from the 5' end of the sense strand is fluorine, and the 2' position of the nucleotide sugar groups at positions 2, 4, 6, 12, 14, 16, 18, and 20 from the 5' end of the antisense strand is fluorine, and the 2' positions of the remaining nucleotide sugar groups are all methoxy, or preferably the 2' position of the nucleotides at positions 5, 7, 8, and 9 from the 5' end of the sense strand is fluorine, and the 2' position of the nucleotide sugar groups at positions 2, 4, 8, 14, and 16 from the 5' end of the antisense strand is fluorine, and the 2' positions of the remaining nucleotide sugar groups are all methoxy.
  • At least two consecutive phosphorothioate bonds exist between three consecutive nucleotides at at least one end of the sense strand end and/or the antisense strand end.
  • the 2' position of the nucleotide sugar group at positions 9, 10, and 11 from the 5' end of the sense strand is fluorine
  • the 2' position of the nucleotide sugar group at positions 2, 4, 6, 12, 14, 16, 18, and 20 from the 5' end of the antisense strand is fluorine
  • the 2' position of the remaining nucleotide sugar groups are all methoxy, and there are at least two consecutive phosphorothioate bonds between three consecutive nucleotides at the 5' and 3' ends of the sense and antisense strands.
  • the 2' position of some nucleotides of the sense strand is fluorine or methoxy, and at least three phosphate bonds between adjacent nucleotides at the end of the antisense strand can be thiolated.
  • the 2' position of the 5th, 7th, 8th, 9th or the 3rd, 5th, 7th, 8th, 9th, 11th, 13th, 15th nucleotides starting from the 5' end of the sense strand is fluorine, and the 2' position of the remaining nucleotides is methoxy, and at least three phosphate bonds between adjacent nucleotides at the end of the antisense strand can be thiolated.
  • the 2' position of some nucleotides of the sense strand is fluorine or methoxy, and at least three phosphate bonds between adjacent nucleotides at the end of the antisense strand can be thiolated.
  • the 2' position of the 9th, 10th, 11th or the 3rd, 5th, 7th, 8th, 9th, 11th, 13th, 15th and/or 17th nucleotides starting from the 5' end of the sense strand is fluorine, and the 2' position of the remaining nucleotides is methoxy, and at least three phosphate bonds between adjacent nucleotides at the end of the antisense strand can be thiolated.
  • the sense strand and antisense strand in the RNA inhibitor of the present invention are selected from the following Table 4:
  • G 2'-O-methylguanylate
  • A 2'-O-methyladenylate
  • U 2'-O-methyluridylate
  • C 2'-O-methylcytidine
  • Gs 2'-O-methyl-3'-thioguanylate
  • As 2'-O-methyl-3'-thioadenylate
  • Us 2'-O-methyl-3'-thiouridylate
  • Cs 2'-O-methyl-3'-thiocytidine
  • fG 2'-fluoroguanylate
  • fA 2'-fluoroadenylate
  • fU 2'-fluorouridylate
  • fC 2'-fluorocytidine
  • fGs 2'-fluoro-3'-thioguanylate
  • fAs 2'-fluoro-3'-thioadenylate
  • fUs 2'-fluoro-3'-thiouridylate
  • fCs 2'-fluoro-3'
  • G 2'-O-methylguanylate
  • A 2'-O-methyladenylate
  • U 2'-O-methyluridylate
  • C 2'-O-methylcytidine
  • Gs 2'-O-methyl-3'-thioguanylate
  • As 2'-O-methyl-3'-thioadenylate
  • Us 2'-O-methyl-3'-thiouridylate
  • Cs 2'-O-methyl-3'-thiocytidine
  • fG 2'-fluoroguanylate
  • fA 2'-fluoroadenylate
  • fU 2'-fluorouridylate
  • fC 2'-fluorocytidine
  • fGs 2'-fluoro-3'-thioguanylate
  • fAs 2'-fluoro-3'-thioadenylate
  • fUs 2'-fluoro-3'-thiouridylate
  • fCs 2'-fluoro-2'
  • the sense strand and antisense strand in the RNA inhibitor of the present invention are selected from the following Table 6:
  • G 2'-O-methylguanylate
  • A 2'-O-methyladenylate
  • U 2'-O-methyluridylate
  • C 2'-O-methylcytidine
  • Gs 2'-O-methyl-3'-thioguanylate
  • As 2'-O-methyl-3'-thioadenylate
  • Us 2'-O-methyl-3'-thiouridylate
  • Cs 2'-O-methyl-3'-thiocytidine
  • fG 2'-fluoroguanylate
  • fA 2'-fluoroadenylate
  • fU 2'-fluorouridylate
  • fC 2'-fluorocytidine
  • fGs 2'-fluoro-3'-thioguanylate
  • fAs 2'-fluoro-3'-thioadenylate
  • fUs 2'-fluoro-3'-thiouridylate
  • fCs 2'-fluoro-2'
  • the sense strand and antisense strand in the RNA inhibitor of the present invention are selected from the following Table 7:
  • G 2'-O-methylguanylate
  • A 2'-O-methyladenylate
  • U 2'-O-methyluridylate
  • C 2'-O-methylcytidine
  • Gs 2'-O-methyl-3'-thioguanylate
  • As 2'-O-methyl-3'-thioadenylate
  • Us 2'-O-methyl-3'-thiouridylate
  • Cs 2'-O-methyl-3'-thiocytidine
  • fG 2'-fluoroguanylate
  • fA 2'-fluoroadenylate
  • fU 2'-fluorouridylate
  • fC 2'-fluorocytidine
  • fGs 2'-fluoro-3'-thioguanylate
  • fAs 2'-fluoro-3'-thioadenylate
  • fUs 2'-fluoro-3'-thiouridylate
  • fCs 2'-fluoro-2'
  • the sense strand and antisense strand in the RNA inhibitor of the present invention are selected from the following Table 8:
  • G 2'-O-methylguanylate
  • A 2'-O-methyladenylate
  • U 2'-O-methyluridylate
  • C 2'-O-methylcytidine
  • Gs 2'-O-methyl-3'-thioguanylate
  • As 2'-O-methyl-3'-thioadenylate
  • Us 2'-O-methyl-3'-thiouridylate
  • Cs 2'-O-methyl-3'-thiocytidine
  • fG 2'-fluoroguanylate
  • fA 2'-fluoroadenylate
  • fU 2'-fluorouridylate
  • fC 2'-fluorocytidine
  • fGs 2'-fluoro-3'-thioguanylate
  • fAs 2'-fluoro-3'-thioadenylate
  • fUs 2'-fluoro-3'-thiouridylate
  • fCs 2'-fluoro-2'
  • the sense strand or antisense strand of the RNA inhibitor described in the present invention has a sequence of at least 15 consecutive nucleotides identical to the sense strand or antisense strand in Tables 4 to 8, or a sequence that differs by one, two or three nucleotides.
  • the distribution, targeting or stability of the RNA inhibitor is altered by introducing a ligand for a target tissue receptor into the vector.
  • a specific ligand can provide enhanced affinity for a selected target (e.g., a molecule, a cell or cell type, a compartment (e.g., a cell or organ compartment, a body tissue, an organ or region)) compared to a species in which the ligand is not present.
  • Ligands can include naturally occurring substances such as proteins (e.g., human serum albumin (HSA), low-density lipoprotein (LDL), or globulins); carbohydrates (e.g., dextran, pullulan, chitin, chitosan, inulin, cyclodextrin, N-acetylglucosamine, N-acetylgalactosamine, or hyaluronic acid); or lipids.
  • Ligands can also be recombinant or synthetic molecules, such as synthetic polymers, For example, synthetic polyamino acids.
  • the ligand can also include a targeting group, such as a cell or tissue targeting agent that is combined with a specified cell type such as a kidney cell, such as a lectin, a glycoprotein, a lipid or a protein, such as an antibody.
  • a targeting group such as a cell or tissue targeting agent that is combined with a specified cell type such as a kidney cell, such as a lectin, a glycoprotein, a lipid or a protein, such as an antibody.
  • the targeting group can be thyrotropin, melanocyte stimulating hormone, a lectin, a glycoprotein, a surfactant protein A, a mucin carbohydrate, a multivalent lactose, a multivalent galactose, N-acetyl-galactosamine, N-acetyl-glucosamine multivalent mannose, a multivalent fucose, a glycosylated polyamino acid, a multivalent galactose, transferrin, a bisphosphonate, polyglutamic acid, polyaspartic acid, a lipid, cholesterol, a steroid, bile acid, folic acid, vitamin B12, vitamin A, biotin, or an RGD peptide or RGD peptide mimetic.
  • the ligand is a multivalent galactose, such as N-acetyl-galactosamine.
  • the sense strand and antisense strand contained in the RNA inhibitor of the present invention can be conveniently and routinely prepared by the well-known technique of solid phase synthesis. Any other method known in the art for such synthesis, such as liquid phase synthesis or fermentation, can be used additionally or alternatively.
  • the sense strand and antisense strand contained in the RNA inhibitor of the present application can be synthesized by an automatic synthesizer using a phosphoramidite method derived from a carrier-nucleoside phosphoramidite monomer.
  • the ligand described in the present invention is coupled to the 5' end and/or 3' end of the antisense chain, and/or the 5' end and/or 3' end of the sense chain through a carrier structure.
  • the carrier structure can be coupled to the 5' end and/or the 3' end of the sense strand; or the carrier structure can be coupled to the 5' end of the antisense strand and the carrier structure can be coupled to the 3' end of the sense strand; or the carrier structure can be coupled to the 3' end of the antisense strand and the ligand can be coupled to the 5' end of the sense strand.
  • the carrier structure includes 5'MVIP and 3'MVIP, wherein the 5'MVIP is coupled to the 5' end of the sense strand and/or the antisense strand, and the 3'MVIP is coupled to the 3' end of the antisense strand and/or the sense strand, the structure of the 5'MVIP is as shown in Formula I, and the structure of the 3'MVIP is as shown in Formula II.
  • X is a liver-targeting specific ligand
  • L is a branched chain
  • R 1 and R 2 are transfer points
  • connection between R 1 or R 2 and the sense strand or antisense strand is through phosphate or modified phosphate, and R 1 or R 2 is preferably connected to the sense strand or antisense strand through phosphate or phosphorothioate.
  • m or n may be 0, i.e., there is no 3'MVIP or 5'MVIP.
  • the structure of the 3'MVIP may be:
  • the structure of the 3'MVIP may be:
  • the structure of the 3'MVIP may be:
  • the structure of the 3'MVIP may be:
  • the structure of the 3'MVIP may be:
  • the n refers to the sum of n placed in the 5'MVIP at the 5' end of the sense strand and the antisense strand of the RNA inhibitor
  • the m refers to the sum of m placed in the 3'MVIP at the 3' end of the sense strand and the antisense strand of the RNA inhibitor.
  • the R1 and R2 structures contain -NH-, -S- and/or -O-, and R1 and R2 are connected to the connecting chain D and the 5' end and 3' end of the sense chain and/or antisense chain respectively through the -NH-, -S- or -O- in the structure, and R1 and R2 are the same or different.
  • R1 and R2 are optionally straight carbon chains, or straight carbon chains with amide, carboxyl or alkyl branches, or cyclic structures, wherein the cyclic structure includes a saturated or unsaturated aliphatic carbocyclic group, or a five-membered or six-membered heterocyclic group or aromatic hydrocarbon group containing sulfur, oxygen or nitrogen atoms.
  • the R 1 and/or R 2 is -E 1 (CH 2 ) x CH 2 E 2 -, wherein x is any integer from 3 to 12, and the groups E 1 and E 2 can be -NH-, -S- or -O-, respectively.
  • R1 and/or R2 is -E1 ( CH2 ) x1CH (OH)( CH2 ) x2E2- , wherein x1 or x2 is each independently any integer from 3 to 10, and E1 and E2 can be -NH-, -S- or -O- , respectively.
  • the R 1 is a heterocyclic or carbocyclic structure containing N, S or O as shown below:
  • the transition point R 1 is -NH(CH 2 ) x CH 2 O-, wherein x is any integer from 3 to 12, preferably any integer from 4 to 6, and can be introduced by the following two phosphoramidite monomers.
  • One -O- or -S- in the iR 1 structure is used for the synthesis of the R 1 phosphoramidite monomer, which is connected to the 5' end of the sense chain or antisense chain of the RNA inhibitor by solid phase synthesis.
  • the -NH-, -S- or -O- in the structure is used to connect with the connecting chain D in the 5'MVIP, thereby introducing the liver-targeting specific ligand X at the 5' end of the sense chain or antisense chain in the RNA inhibitor.
  • the exemplary structure of the monomer introduced into the 5' end of the sense chain or antisense chain of the RNA inhibitor is as follows:
  • One -NH-, -S- or -O- in the R1 structure is first connected to the connecting chain D, and the other -NH-, -S- or -O- is used to form an ester with the phosphoramidite in the synthesis of the 5'MVIP phosphoramidite monomer.
  • the structure of the sense chain or antisense chain 5'MVIP phosphoramidite monomer is shown below:
  • the 5'MVIP phosphoramidite monomer of the sense strand or antisense strand preferably has the following structure:
  • the linker B part in the above-mentioned monomer is branched 1 to 4 times to obtain the corresponding monomer compound.
  • the liver-targeting specific ligand X is introduced into the 5' end of the sense chain or the antisense chain through solid phase synthesis.
  • the transition point R1 is -NH( CH2 ) xCH2O- , wherein x can be any integer from 3 to 12, preferably any integer from 4 to 6.
  • the 5'MVIP phosphoramidite monomer structure is selected from the following structures:
  • the transition point R2 is a heterocyclic or carbocyclic structure containing N, S or O as shown below:
  • the transition point R2 is -NH( CH2 ) x1CH (OH)( CH2 ) x2CH2O- , wherein x1 is any integer from 1 to 4, and x2 is any integer from 0 to 4.
  • the transfer point R2 described in the present application is formed by esterification or amide formation between succinic anhydride and -NH-, -S- or -O- in the R2 structure, and at the same time, coupling with -NH- in the blank Solid Support to form a 3'MVIP solid spport, and then introducing 3'MVIP into the 3' end of the sense chain or antisense chain through the phosphoramidite solid phase synthesis method.
  • the heterocyclic ring in the transition point R2 structure is a pyrrole ring or a piperidine ring, which is connected to the connecting chain D of 3'MVIP through the nitrogen heteroatom in the ring.
  • the exemplary structure of the introduced 3'MVIP solid spport is as follows:
  • the transition point R2 is -B4 ( CH2 ) x1CH (OH)( CH2 ) x2CH2B5- , wherein x1 is any integer from 1 to 4, x2 is any integer from 0 to 4, B4 and B5 are -NH- , -S- or -O-, respectively, and the exemplary structure of the introduced 3'MVIP solid spport is as follows:
  • R 2 is -NHCH 2 CH(OH)CH 2 O-, and the exemplary structure of the introduced 3'MVIP solid spport is as follows:
  • the 3'MVIP solid support structure is as follows:
  • the liver-targeting specific ligand X is selected from structures used to enhance the uptake of RNA inhibitors by hepatocytes, and can be lipids, steroids, vitamins, sugars, proteins, peptides, polyamines, and peptide mimetic structures.
  • the liver-targeting specific ligand X introduced into the ends of the sense strand or antisense strand of the RNA inhibitor can be the same, They may also be different, for example, in terms of properties, some may enhance liver targeting, some may be regulatory structures of the RNA inhibitor in vivo pharmacokinetics, and some may be structures with in vivo dissolution activity.
  • the liver-targeting specific ligand X is selected from one or more monosaccharides and their derivatives in the following structures.
  • the monosaccharide is selected from one or more of the following structures: mannose, galactose, D-arabinose, glucose, fructose, xylose, glucosamine, ribose.
  • the monosaccharide derivative is selected from mannose derivatives, galactose derivatives, glucose derivatives, ribose derivatives and other derivatives.
  • the liver-targeting specific ligand X is selected from galactose, galactosamine, N-acetylgalactosamine and derivatives thereof, and its general structural formula is as follows:
  • W1 is hydrogen or a hydroxyl protecting group, which may be the same or different; W is -OH, -NHCOOH or -NHCO( CH2 ) qCH3 , wherein q is an integer of 0-4; W2 is -NH-, O, S or C.
  • the liver-targeting specific ligand X is N-acetylgalactosamine and its derivatives.
  • liver-targeting specific ligand X is selected from the following structures:
  • W is selected from one or two of -OH, -NHCOOH or -NHCO(CH 2 ) q CH 3 , wherein q is an integer of 0-4.
  • the liver-targeting specific ligand X in the same 5'MVIP or 3'MVIP structure may be the same or different.
  • X between 5'MVIP and 3'MVIP may be the same or different.
  • the branched chain L further has a hydroxyethyl group or a carboxylic acid side chain.
  • the branched chain L is a C 7 -C 18 carbon chain containing an amide group or a six-membered aliphatic carbocyclic group.
  • the side chain L is selected from one or more of the following structures:
  • r1 is any integer of 1-12
  • r2 is any integer of 0-20
  • Z is H, an alkyl group or an amide group, and the alkyl group is, for example, a C 1 -C 5 alkyl group.
  • the structure of the connector B is related to the number of Xs that can be introduced.
  • the connector B contains -NH-, C, O, S, amide, phosphoryl, thiophosphoryl, and when n or m is 1, it is a straight carbon chain. When n or m is 2, 3 or 4, the number of forks is 2, 3 or 4, respectively.
  • the linker B is selected from the following structures:
  • A1 and A2 are each independently C, O, S, -NH-, carbonyl, amide, phosphoryl or thiophosphoryl, and r is an integer of 0-4.
  • the linker B is selected from the following structures:
  • r is any integer from 0 to 4.
  • the linker B is selected from the following structures:
  • the linker B is selected from the following structures:
  • the connecting chain D further has a side chain of a hydroxymethyl group, a methyl tert-butyl group, a methylphenol group, or a C 5 -C 6 aliphatic ring group.
  • the connecting chain D is a C 3 -C 10 carbon chain containing two C ⁇ O groups, a six-membered aliphatic carbocyclic group, or a phenyl group.
  • the connecting chain D is a C 3 -C 10 carbon chain containing two C ⁇ O groups.
  • the connecting chain D is selected from the following structures:
  • the connecting chain D is selected from the following structures:
  • the connecting chain D is selected from the following structures:
  • the (XL) n -BD- in the 5'MVIP structure and the (XL) m -BD- in the 3'MVIP structure are selected from one or more of the following structures:
  • the X, L, B and D are the same or different within each of the 5'MVIP and the 3'MVIP or between the 5'MVIP and the 3'MVIP.
  • the (XL) n -BD- in the 5'MVIP structure is selected from the structures shown in Table 9:
  • 5’MVIP may not exist, in which case m may be any integer from 2 to 4.
  • the (XL) m -BD- in the 3'MVIP structure is selected from the structures shown in Table 10:
  • the combination of (XL) n -BD- and R 1 in the carrier structure 5′MVIP is as shown in Table 11.
  • 3'MVIP may not exist, in which case n may be any integer from 2 to 4.
  • the combination of (XL) m -BD- and R 2 in the carrier structure 3'MVIP is as shown in Table 12.
  • the 5’MVIP is selected from any one or more of 5’MVIP01 to 5’MVIP22 in Table 11.
  • the 3’MVIP is selected from any one or more of 3’MVIP01 to 3’MVIP27 in Table 12.
  • the sense strand in the RNA inhibitor may be a sequence selected from the following Table 13:
  • the sense strand of the RNA inhibitor described herein has a sequence of at least 15 consecutive nucleotides identical to the sense strand in Table 13, or a sequence that differs from the sense strand in Table 13 by one, two, or three nucleotides.
  • the antisense strand in the RNA inhibitor can be selected from the sequences in Table 14 below:
  • the antisense strand of the RNA inhibitor described herein has a sequence of at least 15 consecutive nucleotides identical to the antisense strand in Table 14, or a sequence that differs from the antisense strand in Table 14 by one, two, or three nucleotides.
  • RNA inhibitor described in the present application is selected from the sequences in Table 15:
  • the sense strand and antisense strand of the RNA inhibitor described in the present invention have a sequence of at least 15 consecutive nucleotides identical to the sense strand and antisense strand in Table 15, or differ from each sequence in Table 15 by one, two or three nucleotides.
  • the antisense strand of the RNA inhibitor of the present invention is:
  • UsfCsGfUAfUAACAAfUAfAGfGAfGCsfUsG (SEQ ID NO: 278), the 5' end and/or 3' end of which is connected to 5'MVIP and/or 3'MVIP of different structures, and the antisense strand of the connection vector structure is selected from the following Table 16:
  • the antisense strand of the RNA inhibitor described in the present invention has a sequence of at least 15 consecutive nucleotides identical to the antisense strand in Table 16, or a sequence that differs from the antisense strand in Table 16 by one, two, or three nucleotides.
  • the antisense strand of the RNA inhibitor of the present invention can be obtained by coupling the antisense strands in Tables 5 to 8 with 5'MVIP and/or 3'MVIP.
  • the antisense strand of the RNA inhibitor described in the present invention has a sequence of at least 15 consecutive nucleotides identical to the antisense strand in Tables 5 to 8, or a sequence that differs from the antisense strand in Tables 5 to 8 by one, two or three nucleotides coupled to 5'MVIP and/or 3'MVIP.
  • the sense strand of the RNA inhibitor of the present invention is: CsAsGCUCCUfUfAfUUGUUAUACsGsA (SEQ ID NO: 270), the 5' end and/or 3' end of which is connected to a 5'MVIP and/or 3'MVIP of different structures, and the sense strand of the connection vector structure is selected from the following Table 17:
  • the sense strand of the RNA inhibitor described in the present invention has a sequence of at least 15 consecutive nucleotides identical to the sense strand in Table 17, or a sequence that differs from the sense strand in Table 17 by one, two or three nucleotides.
  • the sense strand of the RNA inhibitor of the present invention can be obtained by coupling the sense strands in Tables 5 to 8 with 5'MVIP and/or 3'MVIP.
  • the sense strand of the RNA inhibitor described in the present invention has a sequence of at least 15 consecutive nucleotides identical to the sense strand in Tables 5 to 8, or is a sequence that differs from the sense strand in Tables 5 to 8 by one, two or three nucleotides coupled to 5'MVIP and/or 3'MVIP.
  • the RNA inhibitors described herein are formed by random pairing of the antisense strands in Table 16 or sequences differing from these antisense strands by one, two, or three nucleotides and the sense strands in Table 17 or sequences differing from these sense strands by one, two, or three nucleotides.
  • the sense strand and/or antisense strand of the RNA inhibitor is identical to the sense strand and/or antisense strand in Table 19 for at least 15 consecutive nucleotides, or differs by one, two, or three nucleotides.
  • Patent CN113171371B examines in detail the effects of different X, L, B, D, R1 and R2 in the 5'MVIP and/or 3'MVIP structures on the activity of RNA inhibitors, and the entire text of the patent is incorporated into the present invention.
  • N-acetylgalactosamine and its derivatives are preferably used as liver-targeting specific ligands:
  • the length of L has a great influence on the effect of RNA inhibitors.
  • the L chain should be neither too short nor too long.
  • linker B when X, L, D and R1 / R2 are consistent with those in the combination 5'MVIP09/3'MVIP09, A1 and A2 in the general formula of linker B are each independently C, O, S, -NH-, carbonyl, amide, phosphoryl or thiophosphoryl, r is any integer from 0 to 4, and when linker B is the same or different between 5'MVIP and 3'MVIP, the resulting RNA inhibitor activity is not much different.
  • n+m in the RNA inhibitor described in the present invention is 2, 3, 4, 5 and 6 respectively.
  • the positions for coupling of 5'MVIP and/or 3'MVIP include the 5' end and/or 3' end of the antisense chain, the 5' end and/or 3' end of the sense chain, the 5' end of the antisense chain and the 3' end of the sense chain, and the 5' end of the sense chain and the 3' end of the antisense chain.
  • n+m in the RNA inhibitor of the present invention is 2, 3, 4, 5 and 6, respectively.
  • the positions of 5'MVIP and/or 3'MVIP coupling include the 5' end and/or 3' end of the antisense strand in Table 5-Table 8, the 5' end and/or 3' end of the sense strand in Table 5-Table 8, the 5' end of the antisense strand and the 3' end of the sense strand in Table 5-Table 8, and the 5' end of the sense strand and the 3' end of the antisense strand in Table 5-Table 8.
  • the obtained 5'MVIP and 3'MVIP combinations are shown in Table 18:
  • the RNA inhibitor is selected from Table 19:
  • the sense strand and/or antisense strand of the RNA inhibitor has a sequence of at least 15 consecutive nucleotides identical to the sense strand and/or antisense strand in Table 19, or a sequence that differs from the sense strand and/or antisense strand in Table 19 by one, two, or three nucleotides.
  • the RNA inhibitor described herein or a pharmaceutically acceptable salt thereof is preferably prepared or synthesized in the form of a sodium salt, a triethylamine salt or other pharmaceutically acceptable salts.
  • the RNA inhibitor or its pharmaceutically acceptable salt described in the present application is more preferably its sodium salt or triethylamine salt.
  • the present application also provides a pharmaceutical composition comprising the above RNA inhibitor or a pharmaceutically acceptable salt thereof.
  • the present invention provides a pharmaceutical composition comprising the above-mentioned RNA inhibitor or a pharmaceutically acceptable salt thereof and an optional pharmaceutically acceptable excipient.
  • the pharmaceutical composition comprising the above-mentioned RNA inhibitor provided by the present invention can be used to prevent and/or treat related disorders, for example, hypertension.
  • Such pharmaceutical compositions are formulated according to the mode of delivery.
  • An example method is to formulate a composition for systemic administration by parenteral delivery, for example, subcutaneous (SC), intramuscular (IM) or intravenous (IV) delivery.
  • the pharmaceutical composition provided herein can be administered at a dose sufficient to inhibit LPA gene expression.
  • a pharmaceutically acceptable "excipient” or “excipient” is a pharmaceutically acceptable solvent, suspending agent, or any other pharmaceutically inert vehicle for delivering one or more nucleic acids to an animal.
  • Excipients can be liquid or solid and are selected taking into account the planned mode of administration to provide the desired volume, consistency, etc. when combined with the nucleic acid and other components in a given pharmaceutical composition.
  • the RNA inhibitors described herein can be delivered in a manner that targets specific tissues (e.g., hepatocytes).
  • the pharmaceutical composition of the present invention further comprises a delivery vehicle (such as nanoparticles, dendrimers, polymers, liposomes or cationic delivery systems).
  • a delivery vehicle such as nanoparticles, dendrimers, polymers, liposomes or cationic delivery systems.
  • the delivery vehicle described herein comprises a liposome.
  • the delivery vehicle described herein includes nanolipids that can form liposome-nucleic acid nanoparticles with nucleic acid molecules.
  • the delivery vehicle described herein comprises the amphiphilic lipid compound M10C1.
  • compositions provided by the present invention include, but are not limited to, solutions, emulsions, and formulations containing liposomes. These compositions can be produced from a variety of components, including, but not limited to, preformed liquids, self-emulsifying solids, and self-emulsifying semisolids.
  • the formulations include those that target the liver.
  • the pharmaceutical formulations of the present application that can be conveniently presented in unit dosage form can be prepared according to conventional techniques known to the pharmaceutical industry. Such techniques include the step of combining the active ingredient with a pharmaceutically acceptable adjuvant or excipient.
  • the present application provides a method for reducing LPA mRNA or protein expression in cells or tissues, which comprises contacting the cells or tissues with an effective amount of the aforementioned RNA inhibitor that inhibits LPA gene expression or a pharmaceutically acceptable salt thereof, and/or the aforementioned pharmaceutical composition.
  • Cells suitable for treatment using the method of the present application may be any cell expressing the LPA gene, for example, liver cells, brain cells, gallbladder cells, heart cells or kidney cells, but preferably liver cells.
  • Cells suitable for use in the method of the present application may be mammalian cells, and when contacted with cells expressing the LPA gene, the RNA inhibitor inhibits the expression of the LPA gene (e.g., human, primate, non-primate or rat LPA gene) by at least about 50%, for example, as determined by PCR or branched DNA (bDNA)-based methods, or by protein-based methods such as immunofluorescence analysis, Western blotting or flow cytometry.
  • LPA gene e.g., human, primate, non-primate or rat LPA gene
  • the tissue is liver tissue.
  • the cells and tissues are ex vivo.
  • the cells and tissues are in a subject.
  • the term “inhibit” may be used interchangeably with “reduce,” “reduced,” “silenced,” “down-regulated,” “suppressed,” and other similar terms, and includes any level of inhibition.
  • the expression of the LPA gene may be evaluated based on the level or change in level of any variable associated with the expression of the LPA gene, for example, the level of LPA mRNA. This level may be analyzed in a single cell or in a population of cells (including, for example, a sample from a subject).
  • the control level may be any type of control level used in the art, for example, a baseline level before administration or a level measured from a similar subject, cell, or sample that has not been treated or has been treated with a control (such as, for example, a buffer-only control or a no-active-agent control).
  • Inhibition of LPA gene expression can be achieved by treating a subject in which the LPA gene is transcribed and treated (e.g., by contacting one or more cells with the RNA inhibitor of the present application, or by administering the RNA inhibitor of the present application to a subject in which the cells are present).
  • the method can be performed by a first cell or cell population (such cells can be present in a sample derived from a subject) in which LPA gene expression is inhibited, as compared to a second cell or cell population that is substantially the same as the first cell or cell population but has not been treated in this way (control cells that have not been treated with an RNA inhibitor or have not been treated with an RNA inhibitor targeting a gene of interest).
  • the method is performed by evaluating the mRNA level in a cell line that highly expresses LPA using an appropriate concentration of siRNA, and expressing the mRNA level in the treated cells as a percentage of the mRNA level in the non-treated control cells.
  • inhibition of LPA gene expression can be assessed by a decrease in a parameter functionally associated with LPA gene expression, e.g., LP(a) levels in the blood or serum of a subject.
  • LPA gene inhibition can be measured in any cell expressing LPA (endogenous or exogenous from an expression construct) and by any assay known in the art.
  • Inhibition of LPA expression can be manifested by a decrease in the level of LP(a) expressed by a cell or cell population or a sample from a subject (eg, protein levels in a blood sample derived from a subject).
  • Control cells, cell groups or subject samples that can be used to evaluate LPA gene inhibition include cells, cell groups or subject samples that have not been contacted with the RNA inhibitor of the present application.
  • control cells, cell groups or subject samples can be derived from a single subject (e.g., a human or animal subject) or an appropriately matched population control before treatment with an RNA inhibitor.
  • the level of LPA mRNA expressed by a cell or cell population can be measured by any method known in the art for evaluating mRNA expression. For example, qRT-PCR, evaluates the reduction of gene expression. The reduction of protein production can be evaluated by any method known in the art, for example, ELISA.
  • a puncture liver biopsy sample is used as a tissue material to monitor the reduction of LPA gene expression.
  • a blood sample is used as a subject sample to monitor the reduction of LP(a) expression.
  • the present application provides the use of the aforementioned RNA inhibitor for inhibiting LPA gene expression or a pharmaceutically acceptable salt thereof, or the aforementioned pharmaceutical composition in the preparation of a drug for preventing and/or treating a disease or condition or reducing the risk of a disease or condition.
  • the diseases or conditions described herein include diseases or conditions associated with the LPA gene, such as cardiovascular disease.
  • the disease or condition described in the present invention is selected from: hyperLP(a)emia, hyperlipidemia, stroke, atherosclerosis, thrombosis, coronary heart disease, aortic valve stenosis, vascular disease, myocardial infarction, angina pectoris, kidney disease, renal failure, obesity, glucose intolerance, type 2 diabetes (non-insulin-dependent diabetes mellitus) and metabolic syndrome.
  • the present application provides a method for preventing and/or treating a disease or condition, comprising administering to a subject in need thereof an effective amount of the aforementioned RNA inhibitor for inhibiting LPA gene expression or a pharmaceutically acceptable salt thereof, and/or the aforementioned pharmaceutical composition.
  • the in vivo method of the present application may include administering to a subject a pharmaceutical composition comprising an RNA inhibitor, wherein the RNA inhibitor comprises a nucleotide sequence complementary to at least a portion of the LPA mRNA of a mammal to which the RNA inhibitor is administered.
  • the pharmaceutical composition of the present invention may be administered in any manner known in the art, including (but not limited to): oral, intraperitoneal or parenteral routes, including intracranial (e.g., intraventricular, intraparenchymal and intrathecal), intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), nasal, rectal and topical (including buccal and sublingual) administration.
  • the pharmaceutical composition is administered by intravenous infusion or injection.
  • the pharmaceutical composition is administered by subcutaneous injection.
  • the composition is administered by intramuscular injection.
  • RNA inhibitors provided herein can also be administered as "free RNA inhibitors". Free RNA inhibitors are administered in the absence of a pharmaceutical composition. Naked RNA inhibitors can be in a suitable buffer.
  • the buffer may contain acetate, citrate, prolamin, carbonate or phosphate, or any combination thereof.
  • the buffer is phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • RNA inhibitors provided herein can be administered as a pharmaceutical composition, such as a liposomal formulation.
  • RNA inhibitor described herein is in the range of about 0.001 to about 200.0 mg per kilogram of subject body weight per day, typically in the range of about 1 to 50 mg per kilogram of body weight per day.
  • a suitable dose of the RNA inhibitor described herein is in the range of about 0.1 mg/kg to about 5.0 mg/kg, for example, in the range of about 0.3 mg/kg to about 3.0 mg/kg.
  • the method comprises administering the pharmaceutical composition of the present invention to reduce target LPA gene expression, such as about 1, 2, 3, 4, 5, 6, 1-6, 1-3 or 3-6 months per dose.
  • the pharmaceutical composition is administered once every 3-6 months.
  • the treatment is administered less frequently.
  • Repeated dosage regimens may include regularly administering a therapeutic amount of the RNA inhibitor, such as once a month to once a year.
  • the RNA inhibitor is administered about once a month to about once every three months, or about once every three months to about once every six months.
  • the RNA inhibitor can be administered less frequently for treatment.
  • the duration of treatment can be determined based on the severity of the disease.
  • a single dose of the pharmaceutical composition can be long-acting, so that the dosage is applied at intervals of no more than 1, 2, 3 or 4 months.
  • a single dose of the pharmaceutical composition described herein is applied approximately once a month.
  • a single dose of the pharmaceutical composition described herein is applied quarterly (i.e., approximately every 3 months).
  • a single dose of the pharmaceutical composition described herein is applied 2 times per year (i.e., approximately once every 6 months).
  • preventing and/or treating a subject as needed may include a single treatment or a series of treatments.
  • the method further comprises determining the level of LP(a) in a sample from the subject.
  • the method further comprises determining the level of LP(a) in a blood sample, a serum sample, or a urine sample from the subject.
  • the method further comprises administering to the subject an additional therapeutic agent for treating hyperLP(a)emia, hyperlipidemia, stroke, atherosclerosis, thrombosis, coronary heart disease, and aortic stenosis.
  • the additional therapeutic agent can be selected from: statins, such as atorvastatin, rosuvastatin, etc.; cholesterol absorption inhibitors such as ezetimibe; PCSK9 inhibitors; ANGPTL3 inhibitors; APOC3 inhibitors and AGT inhibitors.
  • statins such as atorvastatin, rosuvastatin, etc.
  • cholesterol absorption inhibitors such as ezetimibe
  • PCSK9 inhibitors PCSK9 inhibitors
  • ANGPTL3 inhibitors ANGPTL3 inhibitors
  • APOC3 inhibitors and AGT inhibitors APOC3 inhibitors and AGT inhibitors.
  • the present application provides a cell comprising the aforementioned RNA inhibitor for inhibiting LPA gene expression or a pharmaceutically acceptable salt thereof.
  • the present application provides a drug kit comprising the aforementioned RNA inhibitor for inhibiting LPA gene expression or a pharmaceutically acceptable salt thereof, or the aforementioned pharmaceutical composition.
  • DMSO dimethyl sulfoxide
  • the Chinese name of DMF is N,N-dimethylformamide
  • HOBt 1-hydroxybenzotriazole
  • HBTU O-benzotriazole-tetramethyluronium hexafluorophosphate
  • DIPEA N,N-diisopropylethylamine
  • the Chinese name of DCM is dichloromethane
  • the Chinese name of DMAP is 4-dimethylaminopyridine
  • DMT-CL 4,4'-dimethoxytriphenylmethane
  • the Chinese name of MEOH is methanol
  • the Chinese name of TBTU is O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate;
  • solid phase carrier such as macroporous aminomethyl resin (Resin).
  • the sense strand and antisense strand of the uncoupled carrier structure are synthesized by a standard solid phase phosphoramidite method using a multi-channel solid phase synthesizer, and then the sense strand is complementary annealed with the corresponding antisense strand to prepare the corresponding RNA inhibitor.
  • the basic steps of the solid phase phosphoramidite method include:
  • Oxidation oxidizing the obtained nucleoside phosphite to a more stable nucleoside phosphate (i.e., oxidizing trivalent phosphorus to pentavalent phosphorus);
  • Blocking Block the 5’-OH of the failed nucleotide sequence in the previous step to prevent it from further participating in the reaction; repeat the above steps until the last phosphoramidite monomer is connected; then use methylamine aqueous solution and ammonia water to cleave the ester bond between Solid Support and the starting monomer, and remove the protecting groups on each base and phosphate on the resulting nucleotide sequence; separate and purify by HPLC, filter and sterilize, and lyophilize to obtain the corresponding sense chain or antisense chain.
  • RNA inhibitors of this example are selected from Table 3 and prepared by the method described in Example 1. Plasmid DNA (LPA_PSICHECK(TM)-2 plasmid) was transferred into Huh7 cells using Fugene HD. The transfected cells were inoculated into a 96-well plate at a density of 10,000 cells per well, and the culture medium in each well was 100 ⁇ L. The cells were placed in a 5% CO 2 , 37°C incubator and cultured overnight. Then, the RNA inhibitor was prepared with PBS to prepare the RNA inhibitor sample solution of the corresponding concentration of nanolipid encapsulated RNA inhibitor.
  • RNAiMAX/Opti-MEM was added to each well at the corresponding position, and the RNA inhibitor sample solution diluted with the corresponding concentration was added to the well, mixed and incubated, and the incubated mixed solution was mixed evenly with DMEM containing 10% FBS.
  • the culture medium in each well was aspirated, and then a new culture medium containing the sample was added. After adding, it was placed in a 5% CO 2 , 37°C incubator for culture. The final concentration of the sample test was 5nM, 0.5nM, and 0.05nM.
  • RNA inhibitors in Table 3 exhibited different degrees of inhibitory effects on the LPA mRNA level in Huh7 cells at different concentrations, and were significantly dose-dependent. At a low concentration of 0.05nM, except for Kylo-11-DS-03, Kylo-11-DS-17, Kylo-11-DS-20, Kylo-11-DS-24 and Kylo-11-DS-27 with inhibition rates lower than 40%, the remaining inhibitors all had inhibition rates higher than 40%.
  • the solid support of 3'MVIP is used as the starting monomer of solid phase synthesis.
  • the 5' end of the sense strand or antisense strand of the RNA inhibitor of the present application is coupled with the carrier structure 5'MVIP, the 5'MVIP phosphoramidite monomer is used as the last monomer of solid phase synthesis.
  • the linker B part in the general formula is branched 1 to 4 times to obtain the corresponding Solid Support of 3’MVIP.
  • the linker B part in the general formula is branched 1 to 4 times to obtain the corresponding 5'MVIP phosphoramidite monomer.
  • ERCd-01-c1 (3.24 g, 2.6 mmol) was dissolved in methanol (60 mL), and 10% palladium carbon (0.3 g) and acetic acid (2.0 mL) were added. Then hydrogenation was added under normal pressure and the reaction was allowed to proceed overnight. The reaction solution was filtered through diatomaceous earth, and the filtrate was evaporated to dryness under reduced pressure to obtain 2.9 g of an oily substance ERCd-01-c2, the high-resolution mass spectrum of which is shown in Figure 2.
  • the synthesis steps refer to the synthesis of 3'MVIP09-c1 in 3.1.1.6., and the high-resolution mass spectrum of the synthesized 3'MVIP17-c1 is shown in Figure 3.
  • the synthesis steps refer to the synthesis of 3.1.1.6.3’MVIP09-c1.
  • the synthesis steps refer to the synthesis of 3.1.1.7.3'MVIP09-c2.
  • the synthesis steps refer to 3.1.1.8.3’MVIP09’s Solid Support synthesis.
  • Phosphoramidite monomer of 5'MVIP01 Weigh YICd-01-c2 (1.12 g, 2.0 mmol), and refer to 3.2.1.1. to 3.2.1.5 for the remaining operations.
  • the instrument After the synthesis method is set up, the instrument is ready to work. Click Run to start the synthesis. Observe and record the area of each detritylation peak online. During the synthesis process, add the deprotection reagent according to the actual amount used.
  • argon is purged into the synthesis column for ⁇ 2h, and the synthesis column is unloaded according to the operating procedures.
  • the solid phase carrier in the synthesis column is transferred to the reaction bottle, methylamine aqueous solution and ammonia water are added, and the reaction bottle is placed in a shaker at 35°C for 2-3 hours.
  • the solution is filtered into a round-bottom flask, and the residual solid phase is washed with a 50% ethanol aqueous solution, filtered again and combined with the previous filtrate, the round-bottom flask is connected to a rotary evaporator, the water temperature is set to 50°C and evaporated until no distillation, ethanol is added to the round-bottom flask, mixed, and evaporated again until no distillation, and the operation is repeated until white powder appears at the bottom of the bottle.
  • the obtained white powder is prepared into a solution, purified using a reverse chromatography column, and sampled to detect OD260 and purity.
  • the purified antisense chain solution is divided into a syringe bottle and freeze-dried for standby use, and the product is sealed and stored in a -20°C refrigerator.
  • the synthesis of the sense strand (5'MVIP09 coupling) of the coupling carrier is the same as the antisense strand, where the Solid Support loaded on the column is the Universal carrier.
  • Add DIPEA to the intermediate to make a solution add 5'MVIP phosphoramidite monomer, mix well, and place the reaction bottle in a shaker at 35°C for 2-3 hours.
  • RNA inhibitors in Table 14 Take out the double-stranded solution after centrifugation, remove the supernatant, add ultrapure water to completely dissolve the solid, take samples to test OD260 and purity, and obtain the RNA inhibitors in Table 14.
  • the purified finished solution is divided into vials for freeze-drying for use, and the product is sealed and stored in a -20°C refrigerator.
  • RNA inhibitors described in the present invention are all applicable to this rule, that is, when the 3' end of the sense strand or antisense strand of the RNA inhibitor is coupled with the carrier structure 3'MVIP, the solid support of 3'MVIP is used as the starting monomer for solid phase synthesis; when the 5' end of the sense strand or antisense strand of the RNA inhibitor is coupled with the carrier structure 5'MVIP, the 5'MVIP phosphoramidite monomer is used as the last monomer for solid phase synthesis.
  • those skilled in the art can easily synthesize the synthesis of the remaining RNA inhibitors involved in the present invention.
  • RNA inhibitor of this example is selected from Table 4, and the 2' position of the sugar group of different nucleotides in the sense strand and the antisense strand has a methoxy or fluorine modification.
  • RNA inhibitor was prepared by the method described in Example 4. Referring to the experimental method of Example 2, the inhibitory effect of the RNA inhibitor on LPA mRNA in Huh7 cells at concentrations of 5 nM and 0.5 nM was investigated. The obtained test results are shown in Table 21 and Figure 5.
  • RNA inhibitor Kylo-11-DS53 had a significant inhibitory effect on the LPA mRNA level of Huh7 cells at different concentrations. Its characteristics are that the 2' position of the sugar group of the 5th, 7th, 8th and 9th nucleotides starting from the 5' end of the sense chain are all fluorine, and the rest are methoxy, and the phosphate bonds between the three consecutive nucleotides at the 5' and 3' ends are thio.
  • the 2' position of the sugar group of the 2nd, 4th, 8th, 14th and 16th nucleotides starting from the 5' end of the antisense chain are all fluorine, and the rest are methoxy, and the phosphate bonds between the three consecutive nucleotides at the 5' and 3' ends are thio.
  • RNA inhibitor of this embodiment is selected from Table 5, and the 2' position of different nucleotide sugar groups in the sense chain and the antisense chain has a methoxy or fluorine modification.
  • RNA inhibitor was prepared by the method described in Example 4. Referring to the test method of Example 2, the inhibitory effect of the RNA inhibitor on the LPAmRNA of Huh7 cells at concentrations of 5 nM and 0.5 nM was investigated. The test results are shown in Table 22 and Figure 6.
  • RNA inhibitors Kylo-11-DS65, Kylo-11-DS71, Kylo-11-DS72, and Kylo-11-DS74 had significant inhibitory effects on the LPA mRNA level of Huh7 cells at different concentrations.
  • Kylo-11-DS65 is characterized by fluorine at the 2' position of the sugar group of the 5th, 7th, 8th, and 9th nucleotides starting from the 5' end of the sense chain, and the rest are methoxy groups, and the phosphate bonds between the three consecutive nucleotides at the 5' and 3' ends have thio groups, and the 2' position of the sugar group of the 2nd, 4th, 8th, 14th, and 16th nucleotides starting from the 5' end of the antisense chain are all fluorine, and the rest are methoxy groups, and the phosphate bonds between the three consecutive nucleotides at the 5' and 3' ends have thio groups.
  • Kylo-11-DS71 and Kylo-11-DS72 are characterized in that the 2' positions of the sugar groups of the 3rd, 5th, 7th, 8th, 9th, 10th, 11th, 13th, and 15th nucleotides starting from the 5' end of the sense chain are all fluorine, the rest are methoxy, and the phosphate bonds between the three consecutive nucleotides at the 5' and 3' ends are thiolated, and the 2' positions of the sugar groups of the 2nd, 4th, 6th, 8th, 14th, and 16th nucleotides starting from the 5' end of the antisense chain are all fluorine, the rest are methoxy, and the phosphate bonds between the three consecutive nucleotides at the 5' and 3' ends are thiolated.
  • Kylo-11-DS74 The characteristic of Kylo-11-DS74 is that the 2' positions of the sugar groups of the 9th, 10th and 11th nucleotides starting from the 5' end of the sense chain are all fluorine, the rest are methoxy, and the phosphate bonds between the three consecutive nucleotides at the 5' and 3' ends are thiolated.
  • the 2' positions of the sugar groups of the 2nd, 4th, 6th, 8th, 14th, 16th, 18th and 20th nucleotides starting from the 5' end of the antisense chain are all fluorine, the rest are methoxy, and the phosphate bonds between the three consecutive nucleotides at the 5' and 3' ends are thiolated.
  • the RNA inhibitor of this example is selected from the RNA inhibitors in Table 6.
  • the candidate RNA inhibitors Kylo-11-DS81 to Kylo-11-DS105 have methoxy or fluorine modifications at the 2' position of different nucleotide sugars in the sense strand and antisense strand.
  • RNA inhibitor was prepared by the method described in Example 4. Referring to the experimental method of Example 2, the inhibitory effect of the RNA inhibitor on the LPAmRNA of Huh7 cells at concentrations of 5 nM and 0.5 nM was investigated. The obtained test results are shown in Table 23 and Figure 7.
  • RNA inhibitors Kylo-11-DS87, Kylo-11-DS88, Kylo-11-DS91, Kylo-11-DS92, Kylo-11-DS97, Kylo-11-DS98, Kylo-11-DS101, Kylo-11-DS102 and Kylo-11-DS104 had significant inhibitory effects on the LPA mRNA level of Huh7 cells at different concentrations.
  • Kylo-11-DS87 is characterized in that the 2' positions of the 7th, 8th, 9th and 10th nucleotide sugar groups starting from the 5' end of the sense chain are all fluorine, and the 2' positions of the 2nd, 4th, 6th, 8th and 14th nucleotide sugar groups starting from the 5' end of the antisense chain are all fluorine;
  • Kylo-11-DS88 is characterized in that the 2' positions of the 5th, 8th, 9th and 10th nucleotide sugar groups starting from the 5' end of the sense chain are all fluorine, and the 2' positions of the 2nd, 15th and 17th nucleotide sugar groups starting from the 5' end of the antisense chain are all fluorine;
  • Kylo-11-DS91 is characterized in that the 2' positions of the 8th, 9th and 10th nucleotide sugar groups starting from the 5' end of the sense chain are all fluorine, and the 2' positions of the 2nd, 4
  • Kylo-11-DS97 is characterized in that the 2' positions of the nucleotide sugar groups at the 5' end of the sense strand and the 2' positions of the nucleotide sugar groups at the 7' end, 12' end and 14' end of the antisense strand are all fluorine
  • Kylo-11-DS98 is characterized in that the 2' positions of the nucleotide sugar groups at the 7' end, 9' end and 10' end of the sense strand and the 2' positions of the nucleotide sugar groups at the 2' end of the antisense strand are all fluorine
  • Kylo-11-DS101 is characterized in that the 2' positions of the nucleotide sugar groups at the 7' end, 9' end and 10' end of the sense strand and the 2' positions of the nucleotide sugar groups at the 2' end of the antisense strand are all fluorine
  • Kylo-11-DS101 is characterized in that the 2' positions of the nucleotide sugar groups at
  • the 2' position of the nucleotide sugars is fluorine, and the 2' position of the nucleotide sugars at the 2nd, 6th, 8th, 10th, 14th, and 16th positions starting from the 5' end of the antisense chain is fluorine;
  • Kylo-11-DS104 is characterized in that the 2' position of the nucleotide sugars at the 9th, 10th, and 11th positions starting from the 5' end of the sense chain is fluorine, and the 2' position of the nucleotide sugars at the 14th and 16th positions starting from the 5' end of the antisense chain is fluorine.
  • RNA inhibitors have the common characteristics that, except for the fluorine 2' position of the nucleotide sugars at the above positions, the 2' position of the remaining nucleotide sugars is methoxy, and the phosphate bonds between the three consecutive nucleotides at the 5' and 3' ends are thiolated.
  • Kylo-11-DS102 has the best inhibitory effect on the LPA mRNA level of Huh7 cells, with an inhibition rate of 72.99% at a concentration of 0.5nM.
  • RNA inhibitor of this embodiment is selected from Table 7, and the 2' position of different nucleotide sugar groups in the sense chain and the antisense chain has a methoxy or fluorine modification.
  • RNA inhibitor was prepared by the method described in Example 4. Referring to the experimental method of Example 2, the inhibitory effect of the RNA inhibitor on LPA mRNA of Huh7 cells at concentrations of 5 nM and 0.5 nM was investigated. The obtained test results are shown in Table 24 and Figure 8.
  • RNA inhibitors Kylo-11-DS106 ⁇ Kylo-11-DS110, Kylo-11-DS112 ⁇ Kylo-11-DS117 and Kylo-11-DS119 had significant inhibitory effects on LPA mRNA in Huh7 cells at different concentrations.
  • Kylo-11-DS106 ⁇ Kylo-11-DS110, Kylo-11-DS112 ⁇ Kylo-11-DS114 and Kylo-11-DS116 have at least 15 consecutive nucleotides in common with each other, and the 2' position of the sugar group of the 3rd, 5th, 7th, 8th, 9th, 10th, 11th, 13th and 15th nucleotides starting from the 5' end of the sense chain are all fluorine, and the remaining 2' positions are methoxyl groups, and the phosphate ester bonds between the three consecutive nucleotides at the 5' and 3' ends have thiols, and the 2' positions of the sugar groups of the 2nd, 4th, 6th, 8th, 14th and 16th nucleotides starting from the 5' end of the antisense chain are all fluorine, and the remaining methoxyl groups, and the phosphate ester bonds between the three consecutive nucleotides at the 5' and 3' ends have thio
  • the unmodified sequences of the sense strand and antisense strand of Kylo-11-DS116 and Kylo-11-DS117 are the same, both are SEQ ID NO.8 and SEQ ID NO.21, respectively, and the modification characteristics of the two are different, the 2' position of the sugar group of the 5th, 7th, 8th and 9th nucleotides from the 5' end of the sense strand of the latter are all fluorine, and the 2' position of the sugar group of the 2nd, 6th, 8th, 10th, 14th and 16th nucleotides from the 5' end of the antisense strand are all fluorine.
  • the unmodified sequences of the sense chain and antisense chain of the RNA inhibitor Kylo-11-DS119 are SEQ ID NO.11 and SEQ ID NO.25, respectively.
  • the modification characteristics are that the 2' position of the sugar group of the 9th, 10th and 11th nucleotides starting from the 5' end of the sense chain are all fluorine, and the 2' position of the sugar group of the 2nd, 4th, 6th, 12th, 14th, 16th, 18th and 20th nucleotides starting from the 5' end of the antisense chain are all fluorine.
  • Both the sense chain and the antisense chain contain 21 nucleotides, and both ends are blunt ends.
  • the RNA inhibitor of this example is selected from Table 8, and the 2' position of different nucleotide sugars of the sense chain and the antisense chain has a methoxy or fluorine modification.
  • the RNA inhibitor was prepared by the method described in Example 4. Referring to the test method of Example 2, the inhibitory effect of the RNA inhibitor on the LPA mRNA of Huh7 cells at concentrations of 5nM and 0.5nM was examined. The test results are shown in Table 25 and Figure 9.
  • RNA inhibitors Kylo-11-DS-124, Kylo-11-DS-126, Kylo-11-DS-130 and Kylo-11-DS-131 had significant inhibitory effects on LPA mRNA in Huh7 cells at different concentrations.
  • the common characteristics of these RNA inhibitors are that the 2' position of the sugar group of the 9th, 10th and 11th nucleotides starting from the 5' end of the sense chain are all fluorine, the rest are methoxy, and the phosphate bond between the three consecutive nucleotides at the 5' and 3' ends has a thio group.
  • the 2' position of the sugar group of the 20 nucleotides is fluorine.
  • the sense and antisense chains of Kylo-11-DS-126 and Kylo-11-DS-131 both contain 21 nucleotides, and both have two nucleotide protrusions at the 3' end.
  • the difference is that the 6th nucleotide starting from the 5' end of the antisense chain of the former is fA, and the same position of the latter is dT.
  • the activities of the two are similar.
  • the sense chains of Kylo-11-DS-124 and Kylo-11-DS-130 contain 19 nucleotides, and the antisense chains contain 21 nucleotides. There are two nucleotide protrusions at the 3' end of the antisense chain. The 3' end of the sense chain and the 5' end of the antisense chain are blunt ends. The activities of the two are similar.
  • RNA inhibitors Kylo-11-DS146, Kylo-11-DS 148 and Kylo-11-DS163 could reduce the LDL-c level in the plasma of crab-eating macaques and significantly and continuously reduce the Lp(a) level in the plasma.
  • the Lp(a) level in the plasma of the individual could be reduced by up to 95.25% compared with before administration. By day 77, the average reduction rate could still be maintained at 89.98%.
  • CN202210241706.1 (hereinafter referred to as D1) is a Chinese invention patent application filed by the applicant "Xiamen Ganbaoli Biopharmaceutical Co., Ltd.” on March 11, 2022. Its invention name is LPA inhibitors and their uses, and its publication number is CN114703184A. The applicant performed the following experimental operations on different duplexes:
  • Huh7 cells were washed with PBS, digested with 0.05% trypsin, and then gently perfused with DMEM containing 10% FBS until the cells were single cells and counted. Plasmid DNA was transferred into Huh7 cells using Fugene HD. The transfected cells were seeded into 96-well plates at a density of 10,000 cells per well, with 100 ⁇ L of culture medium per well. The cells were cultured in a 5% CO 2 , 37°C incubator overnight.
  • the final concentrations of the compound test are 5nM, 0.5nM and 0.05nM.
  • the instructions for the dual luciferase detection system are as follows: remove the cells from the incubator, discard the supernatant, add 75 ⁇ L of fresh culture medium and 75 ⁇ L of detection reagent, shake for 10 minutes in the dark, and after the cells are fully lysed, transfer 100 ⁇ L of the sample to an opaque white plate to detect the luminescent signal of fireflies; add 50 ⁇ L of Detection reagent, shake in the dark for 10 minutes, and then detect the Renilla luciferase signal. Calculate the ratio of the main reporter gene to the internal reference reporter gene signal in each well, and calculate the inhibition rate % based on the ratio.
  • the two target sequences for the original sequences claimed to be protected in the present application contain a region of LPA mRNA NM_005577.4 (SEQ ID NO.1), which is different from that in D1 and is expanded by 2 positions forward or 1 position backward compared with the region in D1.
  • the inhibition rates of Kylo-11-DS13 and Kylo-11-DS11 at concentrations of 5nM, 0.5nM and 0.05nM are respectively higher than those of Ky-11-DS12 and Ky-11-DS08 in D1.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Obesity (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Neurosurgery (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Neurology (AREA)
  • Vascular Medicine (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Urology & Nephrology (AREA)
  • Epidemiology (AREA)

Abstract

提供一种抑制LPA基因表达的RNA抑制剂或其药学上可接受的盐,该RNA抑制剂由链长为15-30的正义链和反义链通过碱基配对形成,链长优选为19-23,正义链和反义链之间至少有85%的碱基互补,正义链和/或反义链的部分或全部核苷酸糖基2'位的-OH可被氟或甲氧基取代,且正义链和/或反义链的末端中至少有一个末端的三个相邻核苷酸之间的磷酸酯键可以被硫代。RNA抑制剂的结构中还可含有载体结构5'MVIP和3'MVIP。本申请提供的RNA抑制剂干扰LPA mRNA的翻译模板功能,持续高效地抑制LPA基因表达,可用于治疗和/或预防与LP(a)的水平升高相关的疾病。

Description

一种抑制LPA基因表达的RNA抑制剂及其应用
相关申请的交叉引用
本申请要求于2022年10月24日向中国国家知识产权局提交的申请号为202211304230.8、发明名称为“一种抑制LPA基因表达的RNA抑制剂及其应用”的中国专利申请的优先权,其全部内容通过引用方式并入在本申请中。
技术领域
本发明属于生物医药领域,具体涉及一种抑制LPA基因表达的RNA抑制剂及其应用。
背景技术
RNAi
RNAi(RNA干扰)于1998年,由安德鲁·法厄(Andrew Z.Fire)等在秀丽隐杆线虫中进行反义RNA抑制实验时发现,并将这一过程称为RNA干扰。这一发现被《Science》杂志评为2001年的十大科学进展之一,并名列2002年十大科学进展之首。自此以后,以RNA干扰为作用机理的siRNA作为潜在的基因治疗药物得到人们广泛的关注,2006年,安德鲁·法厄与克雷格·梅洛(Craig C.Mello)由于在RNA干扰机制研究中的贡献获得诺贝尔生理或医学奖。RNAi是在许多生物中,包括动物、植物和真菌,都可由双链RNA(dsRNA)触发的,在RNA抑制剂过程中,一种称为“Dicer”的核酸内切酶将长链dsRNA切割或“切丁”成21~25个核苷酸长的小片段。这些小片段,被称为小干扰RNA(siRNA),其中的反义链(Guide strand)被加载到Argonaute蛋白(AGO2)上。AGO2加载发生在RISC-loading复合物中,这是一个三元复合物,由Argonaute蛋白、Dicer和dsRNA结合蛋白(简称为TRBP)组成。在装载过程中,正义链(Passenger strand)链被AGO2裂解并排出。然后,AGO2使用反义链与包含完全互补序列的mRNA结合,然后催化这些mRNA的切割,致使mRNA分裂丧失翻译模板的作用,进而阻止相关蛋白质的合成。切割后,被切割的mRNA被释放,加载着反义链的RISC-loading复合物被循环用于另一轮的切割。
据统计,在人体内的疾病相关蛋白中,大约超过80%的蛋白质不能被目前常规的小分子药物以及生物大分子制剂所靶向,属于不可成药蛋白。旨在通过基因的表达、沉默等功能治疗疾病的基因治疗被业界认为是继化学小分子药物、生物大分子药物之后的第三代治疗药物,这种疗法在基因水平上实现对疾病的治疗,不受不可成药蛋白的制约。作为基因治疗中RNAi技术最主流的类型,RNAi技术是从mRNA的水平对疾病进行治疗,相比化学小分子药物及生物大分子药物在蛋白质水平的治疗具有更高的效率。利用RNAi技术,可以根据特定基因序列,设计出特异性高、抑制效果好的siRNA的正义链和反义链序列,通过固相合成这些单链序列,然后正义链与反义链在特定的退火缓冲液中按照碱基配对原则配对成siRNA,最后通过载体系统输送到体内相应靶点,降解目标mRNA,破坏目标mRNA作为翻译模板的功能,从而阻止相关蛋白的合成。
siRNA的递送系统
siRNA在血液和组织中不稳定,容易被核酸酶降解,为了提高siRNA的稳定性,可以通过对siRNA的正义链和/或反义链修饰,但这些化学修饰只提供有限的免受核酸酶降解的保护作用并且可能最终影响siRNA的活性。因此,还需要相应的传递系统来保障siRNA安全高效的穿过细胞膜。由于siRNA分子质量较大,且带有大量负电荷,而且具有高水溶解性,所以自身无法顺利穿越细胞膜到达细胞内。
脂质体基本结构是由亲水核和磷脂双分子层构成,具备类似生物膜的磷脂双分子层,拥有很高的生物相容性,所以脂质体一度成为最受欢迎、应用最广泛的siRNA载体。脂质体介导的siRNA递送主要将siRNA包裹到脂质体内,保护siRNA不被核酸酶降解,提高siRNA的通过细胞膜障碍的效率,从而促进细胞的吸收。例如阴离子脂质体、pH敏感性脂质体、免疫脂质体、 膜融合脂质体(fusogenic liposome)和阳离子脂质等等,尽管取得了一定的进展,但脂质体本身容易引发炎症反应,给药前必须使用多种抗组胺和激素类如西利替嗪和地塞米松类等药物,以减少可能发生的急性炎症反应,因此在实际临床应用中并不适合所有治疗领域,尤其一些慢性疾病治疗领域,长期使用可能产生的积蓄毒性是潜在的安全隐患,因此需要一种更安全有效的载体系统来递送siRNA。
肝脏中去唾液酸糖蛋白受体(ASGPR),是肝细胞特异性表达的受体,是一种高效的内吞型受体。由于体内生理情况下各种糖蛋白在酶或酸水解唾液酸后,暴露出的次末端是半乳糖残基,所以ASGPR特异性结合的糖为半乳糖基,故又称半乳糖特异性受体。半乳糖、半乳糖胺、N-乙酰半乳糖胺等单糖和多糖分子都对ASGPR有高亲和性。ASGPR主要生理功能是介导血液中去唾液酸糖蛋白、脂蛋白等物质的清除,且与病毒性肝炎、肝硬化、肝癌等肝脏疾病的发生发展有着密切联系。ASGPR这一特性的发现,对肝源性疾病的诊断及治疗起着重要作用(Ashwell G、Harford J,Carbohydrate specific Receptors of the Liver,Ann Rev Biochem 1982 51:531-554)。结构中含有半乳糖或半乳糖胺及其衍生物的肝源性疾病治疗药物可以特异性地与ASGPR亲和,从而具有主动肝靶向性,不需要其它的载体系统来输送。
LPA、apo(a)和Lp(a)
LPA是编码载脂蛋白(a)(apo(a))的基因的名称,主要表达在肝脏,其表达仅限于人类和非灵长类动物。亲水性载脂蛋白组分载脂蛋白(a)通过二硫键附接至apo(B)-100,与脂质核心一起组合成脂蛋白(a)(Lp(a))颗粒。Lp(a)颗粒是一种富含胆固醇的特殊大分子脂蛋白,表面由胆固醇及磷脂包裹,嵌有前面所述的载脂蛋白(a)和apo(B)-100。Lp(a)可以进入并沉积在血管壁上,有促进动脉粥样硬化的作用。Lp(a)与纤溶酶原(PLG)结构同源,可以与纤维酶原竞争结合纤维蛋白位点,从而抑制纤维蛋白原水解作用,促进血栓形成。因此,LP(a)与动脉粥样硬化和血栓形成有着密切的相关性。研究显示血液中Lp(a)水平是心血管疾病、卒中和动脉粥样硬化性狭窄的独立风险因子。
Lp(a)水平偏高主要跟遗传有关,不会随着饮食、运动和其他生活方式改变而显著改变。人类体内的Lp(a)水平大于300毫克每升属于Lp(a)偏高。Lp(a)水平偏高常常预示患动脉粥样硬化和血栓形成的风险明显增加,检测Lp(a)对于早期识别动脉粥样硬化的风险,具有比较重要的意义。在中国患心血管疾病人数大约有3.3.亿,但普通大众对血脂异常的知晓率、治疗率和控制率总体处于较低的水平,对Lp(a)风险的认识更低,绝大多数医院常规的血脂检查也没有纳入这一项,目前国内外临床上也没有针对性的治疗药物。因此,本领域中急需一种有效抑制LPA基因表达的药物。
发明内容
一方面,本发明提供了一种抑制LPA基因表达的RNA抑制剂或其药学上可接受的盐。
在一些实施方式中,本发明所述的RNA抑制剂由链长为15-30的正义链和反义链通过碱基配对形成,所述链长优选为19-23,并且所述正义链和反义链之间至少有85%的碱基互补;所述正义链和/或反义链的部分或全部核苷酸糖基2'位的-OH可以被取代,其中,所述取代基团为氟或甲氧基,且所述正义链和/或反义链的末端中至少有一个末端的三个相邻核苷酸之间的磷酸酯键可以被硫代。
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐,其中,所述反义链通过与靶序列形成互补性区域,所述靶序列为LPA mRNA不同位置处的多个区域,所述多个区域具有相同的至少15个连续的核苷酸。
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐,其中,所述反义链与靶序列形成互补性区域,所述靶序列为LPA mRNA不同位置处的多个区域,所述多个区域具有相同的至少15个连续的核苷酸,所述靶序列选自LPA mRNA(NM_005577.4)中312-332、654-674、996-1016、1338-1358、1680-1700、2022-2042和2364-2384之间的任意一个核苷酸区域。这些区域的起始位置可能会因LPA mRNA的版本号不同而有所不同,例如在LPA mRNA NM_005577.4中654-674之间的核苷酸区域。
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐,其中,所述反义链与靶序列形成互补性区域,所述靶序列为LPA mRNA不同位置处的多个区域,所述多个区域具有相同的至少15个连续的核苷酸,所述靶序列选自LPA mRNA(NM_005577.4)中493-512、1861-1880和2203-2222之间的任意一个核苷酸区域。这些区域的起始位置可能会因LPA mRNA的版本号不同而有所不同,例如在LPA mRNA NM_005577.4中1861-1880之间的核苷酸区域。
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐的反义链选自以下序列:
5'ucguauaacaauaaggagcug 3'  SEQ ID NO.25
5'auaacucuguccauuaccaug 3'  SEQ ID NO.21
或与上述反义链具有相同的至少15个连续核苷酸的序列,或与上述反义链相差一个、两个或三个核苷酸的序列,
其中,g=鸟苷酸,a=腺苷酸,u=尿苷酸,c=胞苷酸。
在一些实施方式中,所述RNA抑制剂或其药学上可接受的盐的正义链选自以下序列:
5'cagcuccuuauuguuauacga 3'  SEQ ID NO.11
5'ugguaauggacagaguuauca 3'  SEQ ID NO.8
或与上述正义链连续至少15个核苷酸相同,或与上述正义链相差一个、两个或三个核苷酸的序列,
其中,g=鸟苷酸,a=腺苷酸,u=尿苷酸,c=胞苷酸。
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐的正义链为SEQ ID NO.11或与其具有相同的至少15个连续核苷酸的序列,或与其相差一个、两个或三个核苷酸的序列;且所述反义链为SEQ ID NO.25或与其具有相同的至少15个连续核苷酸的序列,或与其相差一个、两个或三个核苷酸的序列:
正义链:5'cagcuccuuauuguuauacga 3'  SEQ ID NO.11
反义链:5'ucguauaacaauaaggagcug 3'  SEQ ID NO.25;
或者,所述正义链为SEQ ID NO.8或与其具有相同的至少15个连续核苷酸的序列,或与其相差一个、两个或三个核苷酸的序列;且所述反义链为SEQ ID NO.21或与其具有相同的至少15个连续核苷酸的序列,或与其相差一个、两个或三个核苷酸的序列:
正义链:5'ugguaauggacagaguuauca 3'  SEQ ID NO.8
反义链:5'auaacucuguccauuaccaug 3'  SEQ ID NO.21;
其中,g=鸟苷酸,a=腺苷酸,u=尿苷酸,c=胞苷酸。
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐,其中,所述正义链为SEQ ID NO.270或与其相差一个、两个或三个核苷酸的序列,且所述反义链为SEQ ID NO.278或与其相差一个、两个或三个核苷酸的序列:
正义链:5'CsAsGCUCCUfUfAfUUGUUAUACsGsA 3'  SEQ ID NO.270
反义链:5'UsfCsGfUAfUAACAAfUAfAGfGAfGCsfUsG 3'  SEQ ID NO.278;
或者,所述正义链为SEQ ID NO.239或与其相差一个、两个或三个核苷酸的序列,且所述反义链为SEQ ID NO.344或与其相差一个、两个或三个核苷酸的序列:
正义链:5'UsGsGUfAAfUfGfGACAGAGUUAUsCsA 3'  SEQ ID NO.239
反义链:5'AsfUsAfACdTCfUGUCCAfUUfACCAsUsG 3'  SEQ ID NO.344;
其中,G=2'-O-甲基鸟苷酸,A=2'-O-甲基腺苷酸,U=2'-O-甲基尿苷酸,C=2'-O-甲基胞苷酸;Gs=2'-O-甲基-3'-硫代鸟苷酸,As=2'-O-甲基-3'-硫代腺苷酸,Us=2'-O-甲基-3'-硫代尿苷酸,Cs=2'-O-甲基-3'-硫代胞苷酸;fG=2'-氟鸟苷酸,fA=2'-氟腺苷酸,fU=2'-氟尿苷酸,fC=2'-氟胞苷酸;fGs=2'-氟-3'-硫代鸟苷酸,fAs=2'-氟-3'-硫代腺苷酸,fUs=2'-氟-3'-硫代尿苷酸,fCs=2'-氟-3'-硫代胞苷酸,dT=2'‐脱氧‐胸苷酸。
在一些实施方式中,本发明所述的RNA抑制剂的正义链或反义链中可容纳不多于3个错配的核苷酸,例如,在5'端和/或3'端的5、4、3或2个核苷酸内。
在上述技术方式中,优选地,本发明所述的RNA抑制剂或其药学上可接受的盐还含有载 体结构5'MVIP和3'MVIP,所述RNA抑制剂的结构如式Ia、Ib或Ic所示:
其中,
所述载体结构包括5'MVIP(5'MultiValent Import Platform)和3'MVIP(3'MultiValent Import Platform);
所述5'MVIP由转接点R1、连接链D、接头B、支链L和肝靶向特异性配体X组成,其通过转接点R1与正义链5'端或反义链5'端连接,其结构如通式I所示:
(X-L)n-B-D-R1-
I
所述3'MVIP由转接点R2、连接链D、接头B、支链L和肝靶向特异性配体X组成,其通过转接点R2与正义链3'端或反义链3'端连接,其结构如通式II所示:
(X-L)m-B-D-R2-
II
其中,
n和m各自独立地为0-4的任意整数,各自独立地优选为1-3的整数,且n+m=2-6的整数,优选n+m=2、3或4,更优选为4;
所述转接点R1为如下所示的含有N、S或O的杂环或碳环结构:
或者,所述R1为-NH(CH2)xCH2O-,其中x为3-12的任意整数,优选为4-6的任意整数;
所述转接点R2为如下所示的含有N、S或O的杂环或碳环结构:
或者,所述转接点R2为-NH(CH2)x1CH(OH)(CH2)x2CH2O-,其中x1为1-4的任意整数,x2为0-4的任意整数;
所述肝靶向特异性配体X选自用于增强肝细胞对RNA抑制剂的摄取的结构,在5’MVIP与3’MVIP各自的内部或5’MVIP与3’MVIP之间相同或不同,其选自单糖及其衍生物,优选为N-乙酰半乳糖胺及其衍生物,更优选地选自以下结构:
其中,W选自-OH、-NHCOOH和-NHCO(CH2)qCH3中的一种或两种,其中q为0-4的整数;
所述支链L在5’MVIP与3’MVIP各自的内部或5’MVIP与3’MVIP之间相同或不同,其选自如下结构中的一种或多种:
其中,r1是1-12的任意整数,r2为0-20的任意整数,Z为H、烷基或酰胺基,所述烷基如C1-C5烷基;
所述接头B在5’MVIP与3’MVIP各自的内部或5’MVIP与3’MVIP之间相同或不同,其选自以下结构:
其中,A1和A2各自独立地是C、O、S、-NH-、羰基、酰胺基、磷酰基或硫代磷酰基,r为0-4的任意整数;
所述连接链D在5’MVIP与3’MVIP各自的内部或5’MVIP与3’MVIP之间相同或不同,其选自以下结构:


其中,每个p各自独立地为1-20的任意整数;s为2-13的任意整数;Z1和Z2为相同或者不同的取代基团,如C3-C10烷基。
在一些实施方式中,所述5’MVIP选自表11中5’MVIP01至5’MVIP22中的任一个。
在一些实施方式中,所述3’MVIP选自表12中3’MVIP01至3’MVIP27中的任一个。
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐,其中,所述5’MVIP为如下所示的5’MVIP01或5’MVIP09,所述3’MVIP为如下所示的3’MVIP01、3’MVIP09或 3’MVIP17:
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐,其中,所述正义链5’MVIP和反义链3’MVIP的组合为5’MVIP01/3’MVIP01、5’MVIP01/3’MVIP17或5’MVIP09/3’MVIP09,或者所述正义链5’MVIP和正义链3’MVIP的组合为5’MVIP01/3'MVIP09或5’MVIP09/3'MVIP01。
另一方面,本发明还提供了上述RNA抑制剂或其药学上可接受的盐在制备用于治疗和/或预防与LP(a)水平升高相关的疾病的药物中的应用,所述与LP(a)的水平升高相关的疾病包括肝源性疾病,其包括包括炎性疾病、心脑血管疾病和代谢疾病,其中,所述心脑血管疾病包括高LP(a)血症、高脂血症、中风、动脉粥样硬化、血栓形成、冠心病和主动脉瓣狭窄。
再一方面,本发明又提供了一种药物组合物,该药物组合物包括上述抑制LPA基因表达的RNA抑制剂或其药学上可接受的盐和任选的药学上可接受的辅料,其中,所述药学上可接受的辅料可以为药学上可接受的赋形剂、载体和/或稀释剂,该药物组合物的剂型为口服剂、静脉注射剂或者皮下或肌内注射剂,优选为皮下注射剂。
又一方面,本发明还提供了一种治疗和/或预防与LP(a)的水平升高相关的疾病、病症或综合征的方法,所述方法包括向有此需要的受试者或患者施用治疗有效量的包含抑制LPA基因表达的RNA抑制剂或其药学上可接受的盐或者包括该RNA抑制剂其药学上可接受的盐和任选的药学上可接受的辅料的药物组合物,其中,所述治疗有效量为1.0mg/kg~10mg/kg的上述RNA抑制剂或其药学上可接受的盐。其中,向受试者或患者施用方式(给药方式)包括口服、静脉注射、皮下或肌内注射、经直肠或腹膜内施加。本领域技术人员能够从下文的详细描述中容易地洞察到本申请的其它方面和优势。下文的详细描述中仅显示和描述了本申请的示例性实施方式。如本领域技术人员将认识到的,本申请的内容使得本领域技术人员能够对所公开的具体实施方式进行改动而不脱离本申请所涉及发明的精神和范围。相应地,本申请的附图和说明书中的描述仅仅是示例性的,而非为限制性的。
附图说明
本申请所涉及的发明的具体特征如所附权利要求书所显示。通过参考下文中详细描述的示例性实施方式和附图能够更好地理解本申请所涉及发明的特点和优势。对附图简要说明如下:
图1为本申请实施例2制得的表3中RNA抑制剂在不同浓度下对Huh7细胞中LPA mRNA水平的抑制效果图;
图2为本申请实施例3中3.1.15合成的ERCd-01-c2的高分辨质谱图;
图3为本申请实施例3中3.1.2.6合成的3'MVIP17-c1的高分辨质谱图;
图4为本申请实施例3中3.2.1.2合成的5'MVIP09-ERCd-PFP-c2的高分辨率质谱图;
图5为本申请实施例5制得的表4中RNA抑制剂在不同浓度下对Huh7细胞中LPA mRNA水平的抑制效果图;
图6为本申请实施例6制得的表5中RNA抑制剂在不同浓度下对Huh7细胞中LPA mRNA水平的抑制效果图;
图7为本申请实施例7制得的表6中RNA抑制剂在不同浓度下对Huh7细胞中LPA mRNA水平的抑制效果图;
图8为本申请实施例8制得的表7中RNA抑制剂在不同浓度下对Huh7细胞中LPA mRNA水平的抑制效果图;
图9为本申请实施例9制得的表8中RNA抑制剂在不同浓度下对Huh7细胞中LPA mRNA水平的抑制效果图;
图10为本申请实施例10中RNA抑制剂对食蟹猴血浆中的LDL-c水平的降低效果图;
图11为本申请实施例10中RNA抑制剂对食蟹猴血浆中的Lp(a)水平的降低效果图。
具体实施方式
以下由特定的具体实施例说明本发明的实施方式,熟悉此技术的人士可由本说明书所公开的内容容易地了解本申请发明的其他优点及效果。
术语定义
在本申请中,术语“LPA”包括人类LPA和食蟹猴LPA,其中人类LPA mRNA序列可见于例如,GenBank NM_005577.4。
在本申请中,“靶序列”是指在LPA基因转录过程中形成的mRNA分子的核苷酸序列的一个连续部分,包括作为原代转录产物的RNA加工的产物的mRNA。在一些实施方式中,序列的靶部分将是至少足够长的,以用作在LPA基因转录过程中形成的mRNA分子的核苷酸序列的所述部分处或附近用于RNA抑制剂引导的降解的底物。“靶序列”的长度通常是约15‐30个核苷酸。
在本申请中,术语“区域”是指在GenBank所收载的LPA mRNA中靶序列开始位置到结束位置。例如,所述“区域”312-332是指在LPA mRNA(NM_005577.4)的从第312位上的核苷酸到第332位上的核苷酸。这些“区域”会由于GenBank收载的LPA mRNA序列的更新,位置会有所不同,但区域涵盖的核苷酸个数和种类不会改变。
在本申请中,术语“RNA抑制剂”,通常指包含如本发明术语所定义的RNA的药剂,且其可通过RNA诱导沉默复合物(RISC)途径介导RNA转录物的靶向切割。经由称为RNA抑制的过程指导mRNA的序列特异性降解,调控(例如,抑制)LPA基因在细胞(例如,受试者如哺乳动物受试者中的细胞)中的表达。
在一些实施方式中,RNA抑制剂可为引入细胞或生物体中来抑制靶mRNA(即LPA基因)的单链siRNA(ssRNA抑制剂)。单链RNA抑制剂结合RISC内切核酸酶Argonaute 2,其然后切割靶mRNA。单链siRNA的长度一般为15至30个核苷酸且经化学修饰。
在一些实施方式中,本申请所使用的“RNA抑制剂”是双链RNA,且本发明中称为“双链RNA抑制剂”、“双链RNA(dsRNA,DS)分子”、“dsRNA剂”或“dsRNA”。术语“dsRNA”指核糖核酸分子的复合物,其具有包含两个反向平行且基本上互补的核酸链的双链体结构,称为具有 相对于靶mRNA的“正义”和“反义”取向。在本申请的一些实施方式中,双链RNA(dsRNA)通过转录后基因沉默机制(本发明中称为RNA抑制或RNA干扰)触发靶mRNA的降解。
双链体结构可为触发LPA mRNA通过RISC途径的特异性降解的任何长度,且可在约15至36碱基对的长度范围内,例如,约15-30碱基对的长度,例如,约15、16、17、18、19、20、21、22、23、24、25、26、27、28、29、30或36碱基对的长度。在某些实施方式中,本申请的RNA抑制剂为15-30个核苷酸的dsRNA,其与靶序列相互作用以指导LPA mRNA的切割。
通常,dsRNA分子的正义链和反义链大多数核苷酸为核糖核苷酸,但如本发明详细说明的,也可包括一个或多个非核糖核苷酸,例如,脱氧核糖核苷酸或修饰的核苷酸。此外,本说明书所涉及的RNA抑制剂可包括具有化学修饰的核糖核苷酸,可在多个区域上有修饰的核苷酸。本发明所用术语“修饰的核苷酸”意指独立地具有修饰的糖部分、修饰的核苷酸间连接或修饰的核碱基,或其任何组合的核苷酸。因此,术语“修饰的核苷酸”涵盖对核苷酸间连接、糖部分或核碱基的例如官能团或原子的置换、添加或移除。适用于本申请的RNA抑制剂的修饰包括本发明所公开或本领域已知的所有类型的修饰。
在本申请中,术语“核苷酸序列”通常是指一连串或一定顺序的核苷酸,无论是经修饰还是未修饰,使用标准核苷酸命名和本申请所述的经修饰的核苷酸的符号表用一连串字母描述。本申请所述的核苷酸序列是通过磷酸二酯键(或其相关的结构变体或合成类似物)连接组成的聚合物,包括天然产生的核苷酸聚合物,但应理解,该术语的范围也包括各种类似物,包括但不限于:肽核酸(PNA)、氨基磷酸酯、硫代磷酸酯、甲基膦酸酯和2'-O-甲基核糖核酸等。通常约有15-30个核苷酸,但该术语也可指任何长度的分子。
在一些实施方式中,核苷酸序列包含一个或多个未修饰的核糖核苷(RNA)和/或未修饰的脱氧核糖核苷(DNA)和/或一个或多个修饰核苷酸。术语“修饰核苷酸序列”通常意指包含至少一个修饰和/或至少一个修饰的核苷酸间键联的一连串或一定顺序的核苷酸。
在本申请中,术语“修饰的核苷酸”通常意指与天然存在的RNA或DNA核苷酸相比包含至少一个化学修饰。例如,2'‐脱氧‐胸苷酸2'‐O‐甲基修饰的核苷酸、2'‐氟代修饰的核苷酸、2'‐脱氧‐修饰的核苷酸、锁定核苷酸、脱碱基核苷酸、2'‐氨基‐修饰的核苷酸、2'‐O‐长链烷基‐修饰的核苷酸(如十六烷基)、吗啉代核苷酸、氨基磷酸酯核苷酸、非天然核碱基核苷酸、5'‐硫代磷酸酯核苷酸以及连接有胆固醇基衍生物或十二烷酸二癸酰胺基团的核苷酸。
修饰的核苷酸包含修饰的糖基和/或修饰的核碱基。
在本申请中,术语“核碱基”或“碱基”通常意指杂环嘧啶或嘌呤化合物,它是所有核酸的组分且包括腺嘌呤、鸟嘌呤、胞嘧啶、胸腺嘧啶和尿嘧啶。核苷酸可包括经修饰的核苷酸或核苷酸模拟物、无碱基(Abasics)或替代物替代部分。术语“未修饰的核碱基”或“天然存在的核碱基”通常意指RNA或DNA的天然存在的杂环核碱基:嘌呤碱基腺嘌呤和鸟嘌呤;以及嘧啶碱基胸腺嘧啶、胞嘧啶和尿嘧啶。“修饰的核碱基”通常意指并非天然存在的核碱基的任何核碱基。
在本申请中,术语“糖基”通常意指核苷酸的天然存在的糖基或修饰的糖基。术语“天然存在的糖基”通常意指如在天然存在的RNA中发现的呋喃核糖基或如在天然存在的DNA中发现的脱氧呋喃核糖基。“修饰的糖基”意指取代的糖基或糖替代物,例如在糖基2'位上有氟代或甲氧基取代。
在本申请中,术语“核苷酸间键联”通常意指核苷酸序列中相邻核苷酸之间的共价键联。“天然存在的核苷酸间键联”意指3'至5'磷酸二酯键联。“修饰的核苷酸间键联”意指除了天然存在的核苷酸间键联之外的任何核苷酸间键联。
在本申请中,术语“反义链”(AS)通常是指RNA抑制剂(例如dsRNA)的包括与靶序列实质上互补的区域的链。在本发明中使用时,术语“互补性区域”通常指反义链上与本申请定义的序列(例如靶序列)实质上互补的区域。
在本申请中,术语“正义链”(S)通常是指RNA抑制剂(例如dsRNA)的这样一条链,所述链 包括有与“反义链”(AS)的区域基本互补的区域。“正义”链有时被称为“有义”链,“过客”链或“反引导”链。借助正义链的序列,反义链靶向所希望的mRNA,同时正义链可能靶向不同靶标或被降解。因此,如果反义链被掺入RISC中,则正确的靶标被靶向。正义链的掺入可以导致脱靶效应。这些脱靶效应可以通过在正义链上使用修饰或使用5'端帽加以限制。
在本申请中,术语“互补”是指两条核苷酸序列在一定条件下杂交,形成碱基对氢键,并形成双链体或双螺旋结构的能力。如RNA抑制剂反义链与RNA抑制剂正义链或LPA mRNA杂交形成沃森-克里克碱基对(Watson-Crick base pairs)或非沃森-克里克碱基对,并且包括天然或经修饰的核苷酸或核苷酸模拟物。“互补”不必每个核苷上均具有核碱基互补性。相反,可以容忍一些错配。
在本申请中,术语“错配”,当互补性区域与靶序列不完全互补时,错配可以在分子的内部或末端区域。通常,最被容许的错配在末端区域,例如,在5'端和/或3'端的5、4、3或2个核苷酸内,且不多于3个错配。
在本申请中,术语“配体”通常是指能够共价地或以其它化学方式与生物活性物质(如dsRNA)结合的任何化合物或分子。在一些实施方式中,配体能够与另一种化合物例如受体直接或间接地相互作用,与配体相互作用的受体可以存在于细胞表面上,或可替代地可以是细胞内和/或细胞间受体,配体与受体的相互作用可以导致生化反应,或可以仅仅是物理相互作用或结合。
在本申请中,术语“药学上可接受的”通常是指不抑制活性成分生物学活性的有效性的一种或多种无毒物质。这类制剂通常可含有盐、赋形剂、缓冲剂、防腐剂、相容性载体和任选的其它治疗剂。这类药学上可接受的制剂通常也可包含适合给予人的相容性固体或液体填料、稀释剂或包囊材料。用于医药时,盐应该是药学上可接受的盐,但可方便地使用非药学上可接受的盐来制备药学上可接受的盐,不能将它们排除在本申请范围以外。这类药理学和药学上可接受的盐包括但不限于由以下酸制备的盐:氢氯酸、氢溴酸、硫酸、硝酸、磷酸、马来酸、乙酸、水杨酸、柠檬酸、硼酸、甲酸、丙二酸、琥珀酸等。药学上可接受的盐也可制备成碱金属盐或碱土金属盐,如钠盐、钾盐或钙盐。
在本申请中,术语“脂质纳米颗粒”或“LNP”通常指包含包封药理活性分子(如,dsRNA)的脂质层的囊泡。LNP描述于例如,中国专利号CN103189057B中,其完整内容以引用方式并入本发明中。
发明详述
一方面,本发明提供了一种抑制LPA基因表达的RNA抑制剂或其药学上可接受的盐。
在一些实施方式中,RNA抑制剂或其药学上可接受的盐,其作用的靶序列包含LPA mRNA NM_005577.4(SEQ ID NO.1)的多个区域:
SEQ ID NO.1:



其中,g=鸟苷酸,a=腺苷酸,t=胸苷酸,c=胞苷酸。
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐,其作用的靶序列包含SEQ ID NO.1中的下划双实线和下划单实线的核苷酸区域。
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐,其作用的靶序列为包含SEQ ID NO.1中的301-2401之间的核苷酸区域且与其具有相同的至少15个连续核苷酸的序列。
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐,其中,所述反义链与靶序列形成互补性区域,所述靶序列为SEQ ID NO.1中312-332、654-674、996-1016、1338-1358、1680-1700、2022-2042和2364-2384之间的任意一个核苷酸区域。
在一些实施方式中,本发明所述的RNA抑制剂或其药学上可接受的盐,其中,所述反义链与靶序列形成互补性区域,所述靶序列为SEQ ID NO.1中493-512、1861-1880和2203-2222之间的任意一个核苷酸区域。
在一些实施方式中,本发明所述的RNA抑制剂的靶序列为这些区域处的至少连续15个核苷酸。
在一些实施方式中,本发明所述的RNA抑制剂的靶序列为这些区域处前后延展的至少连续30个核苷酸。
在一些实施方式中,本发明所述的RNA抑制剂的靶序列为:cagctcc ttattgttat acga(SEQ ID NO.2),其中,g=鸟苷酸,a=腺苷酸,t=胸苷酸,c=胞苷酸。
在一些实施方式中,本发明所述的RNA抑制剂的靶序列为:tggtaatggacagagttat(SEQ ID NO.3),其中,g=鸟苷酸,a=腺苷酸,t=胸苷酸,c=胞苷酸。
在一些实施方式中,本发明所述的RNA抑制剂的靶序列为与SEQ ID NO.2相差一个、两个或三个核苷酸的靶序列。
在一些实施方式中,本发明所述的RNA抑制剂的靶序列为与SEQ ID NO.2具有相同的至少15个连续核苷酸的靶序列。
在一些实施方式中,本发明所述的RNA抑制剂的靶序列为与SEQ ID NO.3相差一个、两个或三个核苷酸的靶序列。
在一些实施方式中,本发明所述的RNA抑制剂的靶序列为与SEQ ID NO.3具有相同的至少15个连续核苷酸的靶序列。
在一些实施方式中,本发明所述的RNA抑制剂的靶序列可以是SEQ ID NO.1中的前面所述的那些区域外的其它任意区域上的15-30个核苷酸的序列。
在一些实施方式中,本发明所述的RNA抑制剂包括用于抑制在细胞中,如受试者(例如,哺乳动物,如易于发生LPA相关障碍例如高LP(a)水平的人)的细胞中LPA基因表达的双链核糖核酸(dsRNA)分子。dsRNA的反义链包括与前面所述的靶序列基本上互补且通常完全互补的互补区。正义链包括与反义链互补的区域,使得在合适条件下组合时,两条链可杂交并形成双链体结构。通常,双链体结构的长度为15至30个碱基对。类似地,与靶序列的互补区的长度为15至30个核苷酸。
在一些实施方式中,本发明所述的RNA抑制剂由链长为15-30的正义链和反义链通过碱基配对形成,其中链长优选为19-23。
在一些实施方式中,本发明所述的RNA抑制剂中正义链和反义链之间至少有85%的碱基互补;
在一些实施方式中,本发明所述的RNA抑制剂的正义链选自表1中的序列,如下表1所示:
表1



其中,g=鸟苷酸,a=腺苷酸,u=尿苷酸,c=胞苷酸。
在一些实施方式中,本发明所述的RNA抑制剂的反义链选自表2中的序列,如下表2所示:
表2


其中,g=鸟苷酸,a=腺苷酸,u=尿苷酸,c=胞苷酸,t=胸苷酸。
在一些实施方式中,表1中的正义链和表2中对应的反义链的碱基对互补形成dsRNA,可以部分互补,也可以完全互补。所述部分互补,可以是至少85%的碱基配对。
在一些实施方式中,本发明所述的RNA抑制剂选自下表3:
表3 RNA抑制剂
在一些实施方式中,所述RNA抑制剂可以通过本领域技术人员所熟知的细胞转染方式或脂质体-核酸纳米颗粒的方式加药到细胞系中进行序列筛选。专利US9233971B2,US9080186B2,CN102985548B和CN103189057B有关脂质化合物及脂质体-核酸纳米颗粒制备的方法全文引入本说明书。
在一些实施方式中,其中所述的脂质化合物中的两性脂质优选大环脂类化合物D1C1、T1C1、T1C6、T4C4、B2C1、B2C6、B2C7和M10C1。
本领域技术人员公知,具有约20至23碱基对,例如,21碱基对的双链体结构的dsRNA已经被认为能特别有效地诱导RNA抑制(Elbashir等人,EMBO 2001,20:6877-6888)。然而,其他人已发现更短或更长的RNA双链体结构也有效(Chu和Rana(2007)RNA14:1714-1719;Kim等人(2005)Nat Biotech 23:222-226)。可以合理地预期,由表1、2和3中一个序列在一端或两端减去或增加几个核苷酸的双链体与所述dsRNA相比可以类似地有效。因此,具有源自表1、2和3中一个序列的至少15、16、17、18、19、20、21或更多个连续核苷酸的序列且在抑制LPA基因表达的能力方面与包含全序列的dsRNA的差异不超过约5、10、15、20、25或30%的抑制的dsRNA均包括在本申请范围内。
本申请所述dsRNA可进一步包括一个或多个单链核苷酸突出端,例如,1、2、3或4个核苷酸。核苷酸突出端可包含核苷酸/核苷类似物或其组合,包括脱氧核苷酸。该突出端可在正义链、反义链或其组合上。此外,突出端的核苷酸可存在于dsRNA的反义链或正义链的5’端、3’端或两端。该突出端可由一条链长于另一条链形成,或由相同长度的两条链交错形成。当该突出端在反义链且可与LPA mRNA形成错配或互补或可为另一个序列。例如,突出端位于正义链的3’端,或可选地,在反义链的3’端。
所述dsRNA还可以具有平端,平端意指在dsRNA该端处不存在不成对的核苷酸,即无核苷酸突出端。平端可位于反义链的5’端和正义链的3’端,反之亦然,或双端平端体,其整个长度上为双链的dsRNA,即,在分子的任一端处没有核苷酸突出端。
在一些实施方式中,所述dsRNA的正义链或反义链在3’端具有核苷酸突出端,突出端含有1、2、3或4个核苷酸,而5’端是平端。
在一些实施方式中,突出端同时存在于正义链和反义链的3’端,突出端含有1、2、3或4个核苷酸。
在一些实施方式中,所述dsRNA是19、21或23个核苷酸长度的双端平端体,其整个长度上为双链的dsRNA,即,在分子的任一端处没有核苷酸突出端。
在一些实施方式中,所述dsRNA具有21个核苷酸长度,且正义链和反义链在3’端均具有2个核苷酸的突出端。
为了增强本申请所述的RNA抑制剂在体内的稳定性,在不影响其活性甚至增强其活性的情况下,可以对上述RNA抑制剂的正义链和反义链进行修饰,其中的核苷酸可以有修饰基团,可以整条链或者部分修饰。在一些实施方式中,所述正义链和/或反义链上的一个或多个核苷酸被修饰以形成修饰的核苷酸。
在一些实施方式中,本申请所述的RNA抑制剂(例如,dsRNA)的正义链和反义链未修饰。在其他实施方式中,本申请所述的RNA抑制剂的正义链和反义链经本领域已知及本发明所述的化学修饰或偶联以加强稳定性或其他有利特性。本申请的其他实施方式中,本申请所述的RNA抑制剂的所有核苷酸或基本上所有核苷酸可以被修饰,即RNA抑制剂的链存在不超过5、4、3、2或1个未修饰的核苷酸。
如本申请所述的RNA抑制剂的正义链和反义链可采用本领域上公知的方法合成和/或修饰,如那些描述于“Current protocols in nucleic acid chemistry”,Beaucage,S.L.等人(编辑),JohnWiley&Sons,Inc.,New York,NY,USA中的,其以引用方式并入本发明中。修饰包括例如,末端修饰,例如,5’端修饰(磷酸化、偶联、反向连接)或3’端修饰(偶联、DNA核苷酸、反向连接等);碱基修饰,例如,使用稳定化碱基、失稳碱基或移除碱基(无碱基核苷酸)或偶联碱基;糖修饰(例如,2’-位或4’-位)或糖的替代;或主链修饰,包括磷酸二酯连接的修饰或替换。在本申请提供的RNA抑制剂中,所述RNA抑制剂的正义链和反义链均不需要均匀修饰,可在其单个核苷酸中掺入一种或一种以上的修饰。
在一些实施方式中,所述修饰的核苷酸选自:脱氧核糖核苷酸、核苷酸模拟物、脱碱基核 苷酸、2’-修饰核苷酸、3’至3’连接(倒置)核苷酸、含非天然碱基的核苷酸、桥接核苷酸、肽核酸(PNA)、解锁的核碱基类似物、锁定核苷酸、3’-O-甲氧基(2’核苷间连接)核苷酸、2’-F-阿拉伯糖核苷酸、5’-Me/2’-氟代核苷酸、吗啉代核苷酸、乙烯基膦酸酯脱氧核糖核苷酸、含乙烯基膦酸酯的核苷酸和含环丙基膦酸酯的核苷酸。
在一些实施方式中,所述2’-修饰核苷酸包括:2’-O-甲基核苷酸、2’-脱氧-2’-氟核苷酸、2’-脱氧核苷酸、2’-甲氧基乙基核苷酸、2’-氨基核苷酸和/或2’-烷基核苷酸。
在一些实施方式中,其中所述反义链5’端开始的至少两个或两个以上偶数位上的核苷酸糖基2’位是氟。
在一些实施方式中,所述反义链5’端开始的偶数位上的核苷酸糖基2’位全部是氟。
在一些实施方式中,所述反义链5’端开始的第2、4、6、8、12、14位核苷酸糖基2’位中的至少一个是氟。例如,其中所述反义链5’端开始的第2、4、6、8、12、14位核苷酸糖基2’位均是氟。
在一些实施方式中,所述反义链除了5’端开始的第2、6、8、10、14、16位核苷酸之外,其余的核苷酸糖基2’位中的至少一个是甲氧基。
在一些实施方式中,所述反义链除了5’端开始的第2、4、6、8、14、16位核苷酸之外,其余的核苷酸糖基2’位中的至少一个是甲氧基。
在一些实施方式中,其中所述反义链除了5’端开始的第2、4、6、8、14、16、18、20位核苷酸之外,其余的核苷酸糖基2’位中的至少一个是甲氧基。
在一些实施方式中,所述正义链5’端开始的奇数位上至少有两个或两个以上核苷酸糖基2’位是氟。
在一些实施方式中,所述正义链5’端开始的奇数位上的核苷酸糖基2’位全部是氟。
在一些实施方式中,所述正义链5’端开始的第5、7、8、9位核苷酸糖基2’位中的至少一个是氟。例如,所述正义链5’端开始的第5、7、8、9位核苷酸糖基2’位均是氟。
在一些实施方式中,所述正义链除了5’端开始的第5、7、8、9位核苷酸之外,其余的核苷酸糖基2’位中的至少一个是甲氧基。
在一些实施方式中,所述正义链5’端开始的第7、9、10、11位核苷酸糖基2’位中的至少一个是氟。例如,所述正义链5’端开始的第7、9、10、11位核苷酸糖基2’位均是氟。
在一些实施方式中,所述正义链除了5’端开始的第7、9、10、11位核苷酸之外,其余的核苷酸糖基2’位中的至少一个是甲氧基。
在一些实施方式中,所述正义链5’端开始的第3、5、7、9、10、11、13、15位核苷酸糖基2’位中的至少一个是氟。例如,所述正义链5’端开始的第3、5、7、9、10、11、13、15位核苷酸糖基2’位均是氟。
在一些实施方式中,所述正义链除了5’端开始的第3、5、7、9、10、11、13、15位核苷酸之外,其余的核苷酸糖基2’位中的至少一个是甲氧基。
在一些实施方式中,所述正义链除了5’端开始的第7、8、9、10位核苷酸之外,其余的核苷酸糖基2’位中的至少一个是甲氧基。
例如,所述正义链和/或反义链的部分或全部核苷酸糖基2’位的-OH可以被取代,其中,所述取代基团为氟或甲氧基,优选从正义链5’端开始的第9、10、11位的核苷酸糖基2’位是氟且从反义链的5’端开始的第2、4、6、12、14、16、18、20位核苷酸2’位是氟,其余的核苷酸糖基2’位均是甲氧基,或优选从正义链5’端开始的第5、7、8、9位的核苷酸2’位是氟且从反义链的5’端开始的第2、4、8、14、16位核苷酸糖基2’位是氟,其余的核苷酸糖基2’位均是甲氧基。
在一些实施方式中,所述正义链和/或反义链的核苷酸之间存在至少两个连续的硫代磷酸酯键。
在一些实施方式中,所述正义链末端和/或反义链末端至少有一端的3个连续的核苷酸之间存在至少两个连续的硫代磷酸酯键。
例如,所述正义链和反义链的5’端和3’端的3个连续的核苷酸之间存在至少两个连续的硫代磷酸酯键。
又例如,从正义链5’端开始的第9、10、11位的核苷酸糖基2’位是氟且从反义链的5’端开始的第2、4、6、12、14、16、18、20位核苷酸糖基2’位是氟,其余的核苷酸糖基2’位均是甲氧基,并且所述正义链和反义链的5’端和3’端的3个连续的核苷酸之间存在至少两个连续的硫代磷酸酯键。
在一些实施方式中,所述正义链的部分核苷酸2’位是氟或甲氧基,且所述反义链的末端至少有3个相邻核苷酸之间的磷酸酯键可以被硫代。从正义链5’端开始的第5、7、8、9位或者第3、5、7、8、9、11、13、15的核苷酸2’位是氟,其余的核苷酸2’位是甲氧基,且所述反义链的末端至少有3个相邻核苷酸之间的磷酸酯键可以被硫代。
在一些实施方式中,所述正义链的部分核苷酸2’位是氟或甲氧基,且所述反义链的末端至少有3个相邻核苷酸之间的磷酸酯键可以被硫代。从正义链5’端开始的第9、10、11位或者第3、5、7、8、9、11、13、15和/或17的核苷酸2’位是氟,其余的核苷酸2’位是甲氧基,且所述反义链的末端至少有3个相邻核苷酸之间的磷酸酯键可以被硫代。
在一些实施方式中,本发明所述的RNA抑制剂中的正义链与反义链选自下表4:
表4序列被修饰的RNA抑制剂

其中,G=2'-O-甲基鸟苷酸,A=2'-O-甲基腺苷酸,U=2'-O-甲基尿苷酸,C=2'-O-甲基胞苷酸;Gs=2'-O-甲基-3’-硫代鸟苷酸,As=2'-O-甲基-3'-硫代腺苷酸,Us=2'-O-甲基-3'-硫代尿苷酸,Cs=2'-O-甲基-3'-硫代胞苷酸;fG=2'-氟鸟苷酸,fA=2'-氟腺苷酸,fU=2'-氟尿苷酸,fC=2'-氟胞苷酸;fGs=2'-氟-3'-硫代鸟苷酸,fAs=2'-氟-3'-硫代腺苷酸,fUs=2'-氟-3'-硫代尿苷酸,fCs=2'-氟 -3'-硫代胞苷酸,T=2'-脱氧-胸苷酸,dT=2'-脱氧‐胸苷酸。在一些实施方式中,本发明所述的RNA抑制剂中的正义链与反义链选自下表5:
表5序列被修饰的RNA抑制剂

其中,G=2'-O-甲基鸟苷酸,A=2'-O-甲基腺苷酸,U=2'-O-甲基尿苷酸,C=2'-O-甲基胞苷酸;Gs=2'-O-甲基-3’-硫代鸟苷酸,As=2'-O-甲基-3'-硫代腺苷酸,Us=2'-O-甲基-3'-硫代尿苷酸,Cs=2'-O-甲基-3'-硫代胞苷酸;fG=2'-氟鸟苷酸,fA=2'-氟腺苷酸,fU=2'-氟尿苷酸,fC=2'-氟胞苷酸;fGs=2'-氟-3'-硫代鸟苷酸,fAs=2'-氟-3'-硫代腺苷酸,fUs=2'-氟-3'-硫代尿苷酸,fCs=2'-氟-3'-硫代胞苷酸,dT=2'-脱氧‐胸苷酸。
在一些实施方式中,本发明所述的RNA抑制剂中的正义链与反义链选自下表6:
表6序列被修饰的RNA抑制剂

其中,G=2'-O-甲基鸟苷酸,A=2'-O-甲基腺苷酸,U=2'-O-甲基尿苷酸,C=2'-O-甲基胞苷酸;Gs=2'-O-甲基-3’-硫代鸟苷酸,As=2'-O-甲基-3'-硫代腺苷酸,Us=2'-O-甲基-3'-硫代尿苷酸,Cs=2'-O-甲基-3'-硫代胞苷酸;fG=2'-氟鸟苷酸,fA=2'-氟腺苷酸,fU=2'-氟尿苷酸,fC=2'-氟胞苷酸;fGs=2'-氟-3'-硫代鸟苷酸,fAs=2'-氟-3'-硫代腺苷酸,fUs=2'-氟-3'-硫代尿苷酸,fCs=2'-氟-3'-硫代胞苷酸,T=2'-脱氧-胸苷酸,dT=2'-脱氧‐胸苷酸。
在一些实施方式中,本发明所述的RNA抑制剂中的正义链与反义链选自下表7:
表7序列被修饰的RNA抑制剂

其中,G=2'-O-甲基鸟苷酸,A=2'-O-甲基腺苷酸,U=2'-O-甲基尿苷酸,C=2'-O-甲基胞苷酸;Gs=2'-O-甲基-3’-硫代鸟苷酸,As=2'-O-甲基-3'-硫代腺苷酸,Us=2'-O-甲基-3'-硫代尿苷酸,Cs=2'-O-甲基-3'-硫代胞苷酸;fG=2'-氟鸟苷酸,fA=2'-氟腺苷酸,fU=2'-氟尿苷酸,fC=2'-氟胞苷酸;fGs=2'-氟-3'-硫代鸟苷酸,fAs=2'-氟-3'-硫代腺苷酸,fUs=2'-氟-3'-硫代尿苷酸,fCs=2'-氟-3'-硫代胞苷酸,dT=2'-脱氧‐胸苷酸。
在一些实施方式中,本发明所述的RNA抑制剂中的正义链与反义链选自下表8:
表8序列被修饰的RNA抑制剂

其中,G=2'-O-甲基鸟苷酸,A=2'-O-甲基腺苷酸,U=2'-O-甲基尿苷酸,C=2'-O-甲基胞苷酸;Gs=2'-O-甲基-3’-硫代鸟苷酸,As=2'-O-甲基-3'-硫代腺苷酸,Us=2'-O-甲基-3'-硫代尿苷酸,Cs=2'-O-甲基-3'-硫代胞苷酸;fG=2'-氟鸟苷酸,fA=2'-氟腺苷酸,fU=2'-氟尿苷酸,fC=2'-氟胞苷酸;fGs=2'-氟-3'-硫代鸟苷酸,fAs=2'-氟-3'-硫代腺苷酸,fUs=2'-氟-3'-硫代尿苷酸,fCs=2'-氟-3'-硫代胞苷酸,dT=2'-脱氧‐胸苷酸。
在一些实施方式中,本发明所述的RNA抑制剂的正义链或反义链为与表4至表8中正义链或反义链具有相同的至少15个连续核苷酸的序列,或相差一个、两个或三个核苷酸的序列。
在一些实施方式中,通过在载体中引入靶组织受体的配体,以改变RNA抑制剂的分布、靶向或稳定性。例如,与不存在配体的物种相比,专属性的配体可以提供针对所选靶(例如分子、细胞或细胞类型、区室(例如细胞或器官区室、身体组织、器官或区域))的增强的亲和力。
配体可以包括天然存在的物质,如蛋白质(例如人血清白蛋白(HSA)、低密度脂蛋白(LDL)或球蛋白);碳水化合物(例如葡聚糖、茁霉多糖、壳多糖、壳聚糖、菊糖、环糊精、N-乙酰葡糖胺、N-乙酰半乳糖胺或透明质酸);或脂质。配体也可以是重组或合成分子,如合成聚合物, 例如合成的聚氨基酸。
配体也可以包括靶向基团,例如与指定的细胞类型如肾细胞结合的细胞或组织靶向剂,例如凝集素、糖蛋白、脂质或蛋白质,例如抗体。靶向基团可以是促甲状腺激素、促黑素、凝集素、糖蛋白、表面活性蛋白质A、黏蛋白碳水化合物、多价乳糖、多价半乳糖、N-乙酰基-半乳糖胺、N-乙酰基-葡糖胺多价甘露糖、多价岩藻糖、糖基化聚氨基酸、多价半乳糖、转铁蛋白、双膦酸盐、聚谷氨酸、聚天冬氨酸、脂质、胆固醇、类固醇、胆酸、叶酸、维生素B12、维生素A、生物素、或RGD肽或RGD肽模拟物。在一些实施方式中,该配体为多价半乳糖,例如,N-乙酰基-半乳糖胺。
本发明所述的RNA抑制剂所包含的正义链和反义链可以通过固相合成的公知技术方便且常规地制备。可另外地或替代地使用本领域中已知的用于这类合成的任何其他方法,如液相合成或发酵。
在一些实施方式中,除了可商购以及常规使用的标准核苷亚磷酰胺单体以及非标准核苷亚磷酰胺单体之外,本申请的RNA抑制剂所包含的正义链和反义链可以通过自动合成仪使用衍生自载体-核苷亚磷酰胺单体的亚磷酰胺法合成。
在一些实施方式中,本发明所述配体通过载体结构偶联于反义链的5’端和/或3’端,和/或正义链的5’端和/或3’端。
例如,所述载体结构可以偶联于正义链的5’端和/或3’端;或所述载体结构可以偶联于反义链的5’端且所述载体结构偶联于正义链的3’端;或所述载体结构可以偶联于反义链的3’端,且所述配体偶联于正义链的5’端。
在一些实施方式中,所述载体结构包括5’MVIP和3’MVIP,其中,所述5’MVIP偶联在所述正义链和/或反义链5’端,所述3’MVIP偶联在所述反义链和/或正义链3’端,所述5’MVIP的结构如式I所示,所述3’MVIP结构如式II所示,
(X-L)n-B-D-R1-,
I
(X-L)m-B-D-R2-,
II
其中,
X为肝靶向特异性配体;
L为支链;
B为接头;
D为连接链;
R1和R2为转接点;
所述5’MVIP通过转接点R1与正义链5’端或反义链5’端连接,所述3’MVIP通过转接点R2与正义链3’端或反义链3’端连接,n和m各自独立地为0-4的任意整数,且n+m=2-6的整数,优选n+m=2、3或4,更优选为4。
在一些实施方式中,所述R1或R2与所述正义链或反义链的连接通过磷酸酯或经修饰的磷酸酯,R1或R2优选地通过磷酸酯或硫代磷酸酯与所述正义链或反义链相连接。
在一些实施方式中,m或n可以为0,即不存在3’MVIP或5’MVIP。
在一些实施方式中,当n=0(即不存在5’MVIP)时,所述3’MVIP的结构可以为:
在一些实施方式中,当n=1时,所述3’MVIP的结构可以为:
在一些实施方式中,当n=2时,所述3’MVIP的结构可以为:
在一些实施方式中,当n=3时,所述3’MVIP的结构可以为:
在一些实施方式中,当n=4时,所述3’MVIP的结构可以为:
在一些实施方式中,所述的n是指同时放在所述RNA抑制剂的正义链和反义链5’端5'MVIP中n之和,所述的m是指同时放在所述RNA抑制剂的正义链和反义链3’端3'MVIP中m之和。
在一些实施方式中,所述R1和R2结构中带有-NH-、-S-和/或-O-,R1和R2通过结构中-NH-、-S-或-O-分别与连接链D以及正义链和/或反义链5’端和3’端相连,R1和R2相同或不相同。
在一些实施方式中,所述R1和R2是任选直碳链,或带有酰胺基、羧基或烷基类支链的直碳链或者环状结构,所述环状结构包括饱和或不饱和的脂肪族碳环基,或者含有硫、氧或氮原子的五元或六元杂环基或芳香烃基。
在一些实施方式中,所述R1和/或R2为-E1(CH2)xCH2E2-,其中x为3-12的任意整数,基团E1和E2可以分别为-NH-、-S-或-O-。
在一些实施方式中,所述R1和/或R2为-E1(CH2)x1CH(OH)(CH2)x2E2-,其中x1或x2各自独立地为3-10的任意整数,E1和E2可以分别为-NH-、-S-或-O-。
在一些实施方式中,所述R1为如下所示的含有N、S或O的杂环或碳环结构:
在一些实施方式中,所述转接点R1为-NH(CH2)xCH2O-,其中x为3-12的任意整数,优选为4-6的任意整数,可以通过以下两种亚磷酰胺单体的方式引入。
i.R1结构中的一个-O-或-S-用于R1亚磷酰胺单体的合成,通过固相合成的方法接入RNA抑制剂正义链或反义链的5’端。结构中-NH-、-S-或-O-用于与5'MVIP中的连接链D连接,从而在RNA抑制剂中的正义链或反义链的5’端引入肝靶向特异性配体X。引入到RNA抑制剂正义链或反义链5’端的单体示例性结构如下:
在一些实施方式中,优选以下结构:
ii.R1结构中的一个-NH-、-S-或-O-先与连接链D连接,另外一个-NH-、-S-或-O-用于5'MVIP亚磷酰胺单体的合成中与亚磷酰胺成酯,正义链或反义链5’MVIP亚磷酰胺单体结构示例如下:
在一些实施方式中,正义链或反义链5’MVIP亚磷酰胺单体优选以下结构:
当通式中n为1-4时,上述的单体中接头B部分分别支化1至4次,以获得对应的单体化合物,借助上述的单体化合物,肝靶向特异性配体X通过固相合成被引入到正义链或反义链5’端。
在一些实施方式中,所述转接点R1为-NH(CH2)xCH2O-,其中x可以是3-12的任意整数,优选为4-6的任意整数。
在一些实施方式中,5'MVIP亚磷酰胺单体结构选自如下结构中:


在一些实施方式中,所述转接点R2为如下所示的含有N、S或O的杂环或碳环结构:
在一些实施方式中,所述转接点R2为-NH(CH2)x1CH(OH)(CH2)x2CH2O-,其中x1为1-4的任意整数,x2为0-4的任意整数。
本申请所述的转接点R2是通过丁二酸酐与R2结构中-NH-、-S-或-O-成酯或酰胺的同时,又与空白Solid Support中-NH-进行偶联,形成3'MVIP solid spport,再通过亚磷酰胺固相合成法,将3’MVIP引入到正义链或反义链的3’端。
在一些实施方式中,所述转接点R2结构中的杂环为吡咯环或哌啶环,其通过环中的氮杂原子与3'MVIP的连接链D连接,引入3'MVIP solid spport示例性结构如下:
当通式中m为1-4时,上述的单体中接头B部分分别支化1至4次,以获得对应的Solid Support。
在一些实施方式中,所述转接点R2为-B4(CH2)x1CH(OH)(CH2)x2CH2B5-,其中x1为1-4的任意整数,x2为0-4的任意整数,B4和B5分别为-NH-、-S-或-O-,引入3’MVIP solid spport示例性结构如下:
当通式中m为1-4时,上述的单体中接头B部分分别支化1至4次,以获得对应的Solid Support。
在一些实施方式中,R2为-NHCH2CH(OH)CH2O-,引入3’MVIP solid spport示例性结构如下:
当通式中m为1-4时,上述的单体中接头B部分分别支化1至4次,以获得对应的Solid Support。
在一些实施方式中,3’MVIP solid support结构如下:


在一些实施方式中,所述肝靶向特异性配体X选自用于增强肝细胞对RNA抑制剂的摄取的结构,可以是脂质、类固醇、维生素、糖、蛋白质、肽、多胺及肽模拟结构。在本申请提供的RNA抑制剂中,引入所述RNA抑制剂正义链或反义链末端的肝靶向特异性配体X可以相同, 也可以不同,例如在特性上,有些可以是增强肝靶向性,有些可以是所述RNA抑制剂在体内药代动力学的调节结构,有些可以是具有体内溶解活性的结构。在一些实施方式中,所述肝靶向特异性配体X选自以下结构中的一种或多种单糖及其衍生物。
在一些实施方式中,所述单糖选自以下结构中的一种或多种:甘露糖、半乳糖、D-阿拉伯糖、葡萄糖、果糖、木糖、葡糖胺、核糖。所述单糖衍生物选自甘露糖衍生物、半乳糖衍生物、葡萄糖衍生物、核糖衍生物以及其他衍生物。
在一些实施方式中,所述肝靶向特异性配体X选自半乳糖、半乳糖胺、N-乙酰半乳糖胺及其衍生物,其结构通式如下:
其中,W1为氢或羟基保护基,可以相同也可以不同;W为-OH、-NHCOOH或-NHCO(CH2)qCH3,其中q为0-4的整数;W2为-NH-、O、S或C。
在一些实施方式中,所述肝靶向特异性配体X为N-乙酰半乳糖胺及其衍生物。
在一些实施方式中,所述肝靶向特异性配体X选自以下结构:
其中,W选自-OH、-NHCOOH或-NHCO(CH2)qCH3中的一种或两种,其中q为0-4的整数。
在一些实施方式中,所述肝靶向特异性配体X在同一个5'MVIP或3'MVIP结构中可以相同,也可以不同。
在一些实施方式中,5'MVIP与3'MVIP彼此之间的X可以相同,也可以不同。
在一些实施方式中,所述支链L是含有-NH-、-C(=O)-、-O-、-S-、酰胺基、磷酰基、硫代磷酰基、C4-C10脂肪族碳环基、苯基或者这些基团的组合的C4-C18碳链。
在一些实施方式中,所述支链L还带有羟乙基或羧酸类的侧链。
在一些实施方式中,所述支链L为含有酰胺基或六元脂肪族碳环基的C7-C18碳链。
在一些实施方式中,所述支链L选自如下结构中的一种或多种:
其中,r1是1-12的任意整数,r2为0-20的任意整数,Z为H、烷基或酰胺基,所述烷基如C1-C5烷基。
在一些实施方式中,所述接头B的结构与能引入的X的数量有关,所述接头B中含-NH-、C、O、S、酰胺基、磷酰基、硫代磷酰基,当n或m为1时,其为一条直碳链,当n或m为2、3或4时,其分叉的次数分别为2、3或4。
在一些实施方式中,所述接头B选自以下结构:
其中,A1和A2各自独立地是C、O、S、-NH-、羰基、酰胺基、磷酰基或硫代磷酰基,r为0-4的整数。
在一些实施方式中,所述接头B选自以下结构:

其中,r为0-4的任意整数。
在一些实施方式中,所述接头B选自以下结构:


在一些实施方式中,所述接头B选自以下结构:
在一些实施方式中,所述连接链D是含有-NH-、C=O、O、S、酰胺基、磷酰基、硫代磷酰基、芳香烃基、C4-C10脂肪族碳环基、含1-3个氮的五元或六元杂环基或者这些基团的组合的C3-C18碳链。
在一些实施方式中,所述连接链D还带有羟甲基、甲基叔丁基、甲基苯酚基、C5-C6脂肪环基的侧链。
在一些实施方式中,所述连接链D为含有两个C=O、六元脂肪族碳环基或苯基的C3-C10碳链。
在一些实施方式中,所述连接链D为含两个C=O的C3-C10碳链。
在一些实施方式中,所述连接链D选自以下结构:

其中,每个p各自独立地为1-20的任意整数;s为2-13的整数;Z1和Z2为相同或者不同的取代基团,如C3-C10烷基。
在一些实施方式中,所述连接链D选自以下结构:


在一些实施方式中,所述连接链D选自以下结构:
在一些实施方式中,所述5’MVIP结构中的(X-L)n-B-D-和3’MVIP结构中的(X-L)m-B-D-选自以下结构中的一种或多种:











在一些实施方式中,所述X、L、B及D在5’MVIP与3’MVIP各自的内部或5’MVIP与3’MVIP之间相同或不同。
在一些实施方式中,所述5’MVIP结构中的(X-L)n-B-D-选自表9所示的结构:
表9 5’MVIP的(X-L)n-B-D-结构



在一些实施方式中,5’MVIP也可以不存在,这时候m可以为2-4的任意整数。
在一些实施方式中,所述3’MVIP结构中的(X-L)m-B-D-选自表10中所示的结构:
表10 3’MVIP的(X-L)m-B-D-结构



在一些实施方式中,所述载体结构5’MVIP中的(X-L)n-B-D-与R1的组合如表11所示。
表11 5’MVIP中(X-L)n-B-D-与R1的组合

在一些实施方式中,3’MVIP可以不存在,这时候n可以是2-4的任意整数。
在一些实施方式中,所述载体结构3’MVIP中的(X-L)m-B-D-与R2组合如表12所示。
表12 3’MVIP的(X-L)m-B-D-与R2组合


在一些实施方式中,所述5’MVIP选自表11中5’MVIP01至5’MVIP22中的任一个或多个。
在一些实施方式中,所述3’MVIP选自表12中3’MVIP01至3’MVIP27中的任一个或多个。
在一些实施方式中,表11中的5’MVIP与表12中的3’MVIP任一个存在组合的可能性,其中n+m=2、3、4、5或6。
在一些实施方式中,所述RNA抑制剂中的正义链可以为选自下列表13中的序列:
表13与5’MVIP09偶联的正义链

在一些实施方式中,本申请所述的RNA抑制剂的正义链为与表13中正义链具有相同的至少15个连续核苷酸的序列,或为与表13中正义链相差一个、两个或三个核苷酸的序列。
在一些实施方式中,所述RNA抑制剂中的反义链可以选自下列表14中的序列:
表14与3’MVIP09偶联的反义链
在一些实施方式中,本申请所述的RNA抑制剂的反义链为与表14中反义链具有相同的至少15个连续核苷酸的序列,或为与表14中反义链相差一个、两个或三个核苷酸的序列。
在一些体内试验实施方式中,本申请所述的RNA抑制剂选自表15中的序列:
表15含5’MVIP09/3’MVIP09组合的RNA抑制剂
在一些实施方式中,本发明所述的RNA抑制剂的正义链和反义链为与表15中正义链和反义链具有相同的至少15个连续核苷酸的序列,或为与表15中各序列相差一个、两个或三个核苷酸。
在一些实施方式中,本发明所述RNA抑制剂的反义链为:
UsfCsGfUAfUAACAAfUAfAGfGAfGCsfUsG(SEQ ID NO:278),其5’端和/或3'端与不同结构的5'MVIP和/或3'MVIP连接,所述连接载体结构的反义链选自下表16:
表16 5'MVIP和/或3'MVIP偶联的反义链

在一些实施方式中,本发明所述的RNA抑制剂的反义链为与表16中反义链具有相同的至少15个连续核苷酸的序列,或为与表16中反义链的相差一个、两个或三个核苷酸的序列。
在一些实施方式中,本发明所述的RNA抑制剂的反义链可以是表5-表8中的反义链与5'MVIP和/或3'MVIP偶联得到。
在一些实施方式中,本发明所述的RNA抑制剂的反义链是与表5-表8中的反义链具有相同的至少15个连续核苷酸的序列,或与表5-表8中反义链相差一个、两个或三个核苷酸的序列与5'MVIP和/或3'MVIP偶联得到的。
在一些实施方式中,本发明所述RNA抑制剂的正义链为:CsAsGCUCCUfUfAfUUGUUAUACsGsA(SEQ ID NO:270),其5’端和/或3'端与不同结构的5'MVIP和/或3'MVIP连接,所述连接载体结构的正义链选自下表17:
表17 5'MVIP和/或3'MVIP偶联的正义链

在一些实施方式中,本发明所述的RNA抑制剂的正义链为与表17中正义链具有相同的至少15个连续核苷酸的序列,或为与表17中正义链相差一个、两个或三个核苷酸的序列。
在一些实施方式中,本发明所述的RNA抑制剂的正义链可以是表5-表8中的正义链与5'MVIP和/或3'MVIP偶联得到的。
在一些实施方式中,本发明所述的RNA抑制剂的正义链是与表5-表8中正义链具有相同的至少15个连续核苷酸的序列,或为与表5-表8中正义链相差一个、两个或三个核苷酸的序列与5'MVIP和/或3'MVIP偶联得到的。
在一些实施方式中,本申请所述的RNA抑制剂是由表16的反义链或与这些反义链相差一个、两个或三个核苷酸的序列和表17中的正义链或与这些正义链相差一个、两个或三个核苷酸的序列随机配对形成的。
在一些实施方式中,所述的RNA抑制剂的正义链和/或反义链与表19中正义链和/或反义链连续至少15个核苷酸相同,或与相差一个、两个或三个核苷酸。
专利CN113171371B中详细考察了5'MVIP和/或3'MVIP结构中X、L、B、D、R1和R2不同对RNA抑制剂活性效果影响,该专利全文引入本发明书。
当X分别为半乳糖、半乳糖胺、N-乙酰半乳糖胺及其衍生物时,在本发明提供的RNA抑制剂中,优选N-乙酰半乳糖胺及其衍生物作为肝靶向特异性配体:
L的长短对RNA抑制剂的作用效果影响较大,L链不能太短也不能太长;当含有-NH-、C=O、O、S、酰胺基、磷酰基、硫代磷酰基、脂肪族碳环基如环己烷或者这些基团的组合时,或者在同一个5'MVIP、3'MVIP结构中或5'MVIP与3'MVIP彼此L结构不同时,在碳链长为C7-C18这个范围,所得RNA抑制剂的活性相差不大。

除接头B结构改变外,而X、L、D及R1/R2与组合5’MVIP09/3’MVIP09中的一致时,接头B中通式中的A1和A2各自独立地是C、O、S、-NH-、羰基、酰胺基、磷酰基或硫代磷酰基,r为0-4的任意整数,并且接头B在5’MVIP与3’MVIP之间相同或不同时,所得RNA抑制剂活性相差不大。






在MVIP结构及RNA抑制剂相同的情况下,不同的连接链D会对RNA抑制剂活性有影响,其中D1、D2、D4的效果接近且优于D3。

不同的转接点R1会对RNA抑制剂活性有影响,其中R1-1作为转接点的所得的RNA抑制剂活性最好。
不同的转接点R2会对RNA抑制剂活性有影响,其中R2-1作为转接点时RNA抑制剂最佳。

在一些实施方式中,本发明所述的RNA抑制剂中的n+m分别为2、3、4、5和6。5’MVIP和/或3’MVIP偶联的位置包括反义链的5’端和/或3’端、正义链的5’端和/或3’端、反义链的5’端和正义链的3’端、正义链的5’端和反义链的3’端。
在一些实施方式中,本发明所述的RNA抑制剂中的n+m分别为2、3、4、5和6。5’MVIP和/或3’MVIP偶联的位置包括表5-表8中反义链的5’端和/或3’端、表5-表8中正义链的5’端和/或3’端、表5-表8中反义链的5’端和正义链的3’端、表5-表8中正义链的5’端和反义链的3’端,所获得的5’MVIP和3’MVIP组合如表18中所示:
表18 5’MVIP和3’MVIP组合列表

在一些实施方式中,n和m各自独立地为0-4的任意整数,各自独立地优选为1-3的整数,且n+m=2-6的整数,优选n+m=2、3或4,更优选为4。
在一些实施方式中,所述的RNA抑制剂选自表19:
表19 RNA抑制剂
在一些实施方式中,所述的RNA抑制剂的正义链和/或反义链为与表19中正义链和/或反义链具有相同的至少15个连续核苷酸的序列,或与表19中正义链和/或反义链相差一个、两个或三个核苷酸的序列。
在一些实施方式中,本申请所述的RNA抑制剂或其药学上可接受的盐优选以钠盐和三乙胺盐或其它可药用盐的形式制备或合成。
在一些实施方式中,本申请所述的RNA抑制剂或其药学上可接受的盐更优选为其钠盐或三乙胺盐。
另一方面,本申请还提供了一种包含上述RNA抑制剂或其药学上可接受的盐的药物组合物。
在一些实施方式中,本发明提供一种包含上述RNA抑制剂或其药学上可接受的盐和任选的药学上可接受的辅料的药物组合物。本发明提供的包含上述RNA抑制剂的药物组合物可用于预防和/或治疗相关障碍,例如,高血压。这类药物组合物依据递送模式配制。一个实例方式为配制用于以肠胃外递送全身性施用的组合物,例如,皮下(SC)、肌内(IM)或静脉内(IV)递送。本申请提供的药物组合物可以足以抑制LPA基因表达的剂量施用。
药学上可接受的“辅料”或“赋形剂”是用于递送一种或多种核酸至动物的药学上可接受的溶剂、悬浮剂或任何其他药学上惰性的媒介物。赋形剂可为液体或固体,并考虑计划的施用方式进行选择,以在与核酸及给定药物组合物中的其他组分组合时提供所需的体积、稠度等。本发明所述的RNA抑制剂可以靶向特定组织(例如,肝细胞)的方式递送。
在一些实施方式中,本发明所述的药物组合物,其还包含递送媒介物(如纳米颗粒、树状聚合物、聚合物、脂质体或阳离子递送系统),
在一些实施方式中,本发明所述的递送媒介物包括脂质体。
在一些实施方式中,本发明所述的递送媒介物包括纳米脂质,其能够与核酸分子形成脂质体-核酸纳米颗粒。
在一些实施方式中,本发明所述的递送媒介物包括两性脂质化合物M10C1。
本发明提供的药物组合物包括(但不限于)溶液、乳液和包含脂质体的制剂。这些组合物可由多种组分产生,包括(但不限于)预形成液体、自乳化固体和自乳化半固体。所述制剂包括靶向肝脏的那些。可以单位剂型方便地存在的本申请药物制剂可依据制药业公知的常规技术制备。这类技术包括将活性成分与药学上可接受的辅料或赋形剂结合的步骤。
用途
另一方面,本申请提供一种减少细胞或组织中LPA mRNA或蛋白质表达的方法,其包括使细胞或组织与有效量的前述的抑制LPA基因表达的RNA抑制剂或其药学上可接受的盐,和/或前述的药物组合物接触。
适合使用本申请方法处理的细胞可为任何表达LPA基因的细胞,例如,肝脏细胞、脑细胞、胆囊细胞、心脏细胞或肾脏细胞,但优选为肝脏细胞。适合用于本申请方法的细胞可为哺乳动物细胞,当与表达LPA基因的细胞接触时,RNA抑制剂抑制LPA基因(例如,人类、灵长类、非灵长类或大鼠LPA基因)的表达至少约50%,例如可通过PCR或基于分支DNA(bDNA)的方法,或由基于蛋白质的方法,如免疫荧光分析法,蛋白质印迹法或流式细胞分析技术测定的。
在一些实施方式中,所述组织是肝脏组织。
在一些实施方式中,所述细胞和组织是离体的。
在一些实施方式中,所述细胞和组织在受试者体内。
本发明所用术语“抑制”可与“减少”、“降低”、“沉默”、“下调”、“压制”及其他类似术语交换使用,且包括任何抑制水平。LPA基因的表达可依据与LPA基因表达相关的任何变量的水平或水平变化来评价,例如,LPA mRNA水平。这一水平可在单个细胞中或细胞群中(包括例如,源自受试者的样品)中分析。对照水平可为本领域上采用的任何类型的对照水平,例如,给药前基线水平或从未处理或接受对照(如例如,仅缓冲剂对照或无活性剂对照)处理的类似受试者、细胞或样本测得的水平。
LPA基因表达的抑制可通过其中LPA基因被转录且已处理(例如,通过一个或多个细胞与本申请的RNA抑制剂接触,或通过施用本申请的RNA抑制剂于其中存在该细胞的受试者)使 得抑制LPA基因表达的第一细胞或细胞群(这类细胞可例如存在于源自受试者的样品中)表达的mRNA量与基本上与该第一细胞或细胞群相同但未如此处理的第二细胞或细胞群(未用RNA抑制剂处理或未用靶向目的基因的RNA抑制剂处理的对照细胞)相比的降低来表现。
在优选的实施方式中,通过实施使用siRNA合适的浓度在高表达LPA的细胞系中评价,并将被干预细胞中的mRNA水平表示为非干预对照细胞中mRNA水平的百分比。
在其他实施方式中,LPA基因表达的抑制可通过功能上与LPA基因表达相关的参数的降低来评价,例如,受试者血液或血清中的LP(a)水平。LPA基因抑制可在任何表达LPA的细胞(内源性或来自表达构建体的外源性)中且通过本领域已知的任何分析法测定。
LPA表达的抑制可由细胞或细胞群或受试者样品表达的LP(a)水平(例如,源自受试者的血液样品中的蛋白质水平)的降低来表现。
可用于评价LPA基因抑制的对照组细胞、细胞群或受试者样品包括未与本申请RNA抑制剂接触的细胞、细胞群或受试者样品。例如,对照细胞、细胞群或受试者样品可源自于用RNA抑制剂治疗前的单个受试者(例如,人类或动物受试者)或适当匹配的群体对照。
细胞或细胞群表达的LPA mRNA水平可采用本领域已知用于评价mRNA表达的任何方法测定。例如,qRT-PCR,评价基因表达的降低。可通过本领域中已知的任何方法,例如,ELISA,评价蛋白质产生的降低。在一些实施方式中,穿刺肝脏活检样品用作监测LPA基因表达降低的组织材料。其他实施方式中,血液样品用作监测LP(a)表达降低的受试者样品。
另一方面,本申请提供的前述抑制LPA基因表达的RNA抑制剂或其药学上可接受的盐,或前述的药物组合物在制备药物中的用途,所述药物用于预防和/或治疗疾病或病症或者降低疾病或病症的风险。
在一些实施方式中,本发明所述的疾病或病症包括与LPA基因相关的疾病或病症,如心血管疾病。
在一些实施方式中,本发明所述的疾病或病症选自:高LP(a)血症、高脂血症、中风、动脉粥样硬化、血栓形成、冠心病、主动脉瓣狭窄、血管病变、心肌梗塞、心绞痛、肾疾病、肾衰竭、肥胖、葡萄糖耐受不良、2型糖尿病(非胰岛素依赖型糖尿病)和代谢综合征。
另一方面,本申请提供一种预防和/或治疗疾病或病症的方法,所述方法包括向有此需要的受试者施用有效量的前述抑制LPA基因表达的RNA抑制剂或其药学上可接受的盐,和/或前述的药物组合物。
本申请的体内方法可包括对受试者施用包含RNA抑制剂的药物组合物,其中,该RNA抑制剂包括与接受施用RNA抑制剂的哺乳动物的LPA mRNA的至少一部分互补的核苷酸序列。本发明所述的药物组合物可采用本领域已知的任何方式施用,包括(但不限于):经口、腹膜内或肠胃外途径,包括颅内(例如,脑室内、脑实质内和鞘内)、静脉内、肌内、皮下、透皮、气道(气雾剂)、经鼻、直肠和局部(包括颊内及舌下)施用。在一些实施方式中,所述药物组合物通过静脉内输注或注射施用。在一些实施方式中,所述药物组合物通过皮下注射施用。在一些实施方式中,该组合物通过肌内注射施用。
本申请提供的RNA抑制剂还可作为“游离RNA抑制剂”施用。游离RNA抑制剂是在没有药物组合物的存在下施用。裸RNA抑制剂可在合适缓冲液中。所述缓冲液可包含乙酸盐、柠檬酸盐、醇溶谷蛋白、碳酸盐或磷酸盐,或其任何组合。在一个实施方式中,所述缓冲液为磷酸盐缓冲盐水(PBS)。可以调整包含所述RNA抑制剂的缓冲液的pH和渗透压,以便适合施用于受试者。
或者,本申请提供的RNA抑制剂可作为药物组合物施用,如脂质体制剂。
本申请提供的药物组合物可以足以抑制LPA基因表达的剂量施用。通常,本申请所述的RNA抑制剂的合适剂量在每天每千克受试者体重约0.001至约200.0mg的范围内,通常在每天每千克体重约1至50mg的范围内。通常,本申请所述的RNA抑制剂的合适剂量在约0.1mg/kg至约5.0mg/kg的范围内,例如约0.3mg/kg至约3.0mg/kg的范围内。
在一个实施方式中,该方法包括施用本发明所述的药物组合物,使得降低靶LPA基因表达,如每剂约1、2、3、4、5、6、1-6、1-3或3-6个月。在一些实施方式中,该药物组合物每3-6个月施用一次。
在一些实施方式中,在初始治疗方式后,以较少的频率施用治疗。重复剂量方式可包括规律地施用治疗量的所述RNA抑制剂,如每月一次至一年一次。在一些实施方式中,所述RNA抑制剂约每月一次至约每三个月一次施用,或约每三个月一次至约每六个月一次施用。
在初始治疗方式后,可以较低的频率施用所述RNA抑制剂来治疗。可依据疾病严重性确定治疗持续时间。
在其他实施方式中,单一剂量的药物组合物可以为长效的,使得剂量以不超过1、2、3或4个月的间隔施用。在本申请的一些实施方式中,本申请所述的药物组合物的单一剂量约每月施用一次。在本申请的其他实施方式中,本申请所述的药物组合物的单一剂量按季(即约每3个月)施用。在本申请的其他实施方式中,本申请所述的药物组合物的单一剂量每年施用2次(即约每6个月一次)。
本领域技术人员应理解,一些因素可影响有效治疗受试者所需的剂量和施用时间,包括(但不限于):受试者中存在的突变、之前的治疗、受试者的一般健康或年龄和存在的其他疾病。此外,按照需要以预防和/或治疗受试者可包括单次治疗或一系列治疗。
在一些实施方式中,该方法进一步包括测定来自所述受试者的样品中的LP(a)水平。
例如,该方法进一步包括测定来自所述受试者血液样品、血清样品或尿液样品中的LP(a)水平。
在一些实施方式中,该方法进一步包括对所述受试者施用用于治疗高LP(a)血症、高脂血症、中风、动脉粥样硬化、血栓形成、冠心病和主动脉瓣狭窄的另外的治疗剂。
例如,所述另外的治疗剂可以选自:他汀类药物,如阿托伐他汀、瑞舒伐他汀等;胆固醇吸收抑制剂如依折麦布;PCSK9抑制剂;ANGPTL3抑制剂;APOC3抑制剂和AGT抑制剂。
又一方面,本申请提供一种细胞,其包含前述的抑制LPA基因表达的RNA抑制剂或其药学上可接受的盐。
另一方面,本申请提供一种药盒,其包含前述的抑制LPA基因表达的RNA抑制剂或其药学上可接受的盐,或者前述的药物组合物。
不欲被任何理论所限,下文中的实施例仅仅是为了阐释本申请提供的RNA抑制剂、制备方法和用途等,而不用于限制本申请发明的范围。
实施例
说明:
DMSO的中文名称为二甲基亚砜;
DMF的中文名称为N,N-二甲基甲酰胺;
HOBt的中文名称为1-羟基苯并三氮唑;
HBTU的中文名称为O-苯并三氮唑-四甲基脲六氟磷酸酯;
DIPEA(DIEA)的中文名称为N,N-二异丙基乙胺;
DCM的中文名称为二氯甲烷;
DMAP的中文名称为4-二甲氨基吡啶;
DMT-CL的中文名称为4,4'-二甲氧基三苯基氯甲烷;
MEOH的中文名称为甲醇;
TBTU的中文名称为O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸;
的名称为固相载体,如大孔氨甲基树脂(Resin)。
实施例1 RNA抑制剂的合成
未偶联载体结构的正义链和反义链是利用标准的固相亚磷酰胺法、使用多通道固相合成仪合成的,然后所述正义链与对应的反义链互补退火制备得到相应的RNA抑制剂。
固相亚磷酰胺法基本步骤包括:
1)脱保护:脱掉起始单体Solid Support羟基保护基(DMTr);
2)偶联:加上第一个亚磷酰胺单体,通过3’至5’方向发生偶联反应;
3)氧化:将所得的核苷亚磷酸酯氧化成更稳定的核苷磷酸酯(即三价磷氧化成五价磷);
4)封闭:将前步骤失败核苷酸序列5’-OH加冒封死,使其不再进一步参与反应;重复上述步骤,直至最后一个亚磷酰胺单体的接入;然后用甲胺水溶液和氨水裂解Solid Support与起始单体之间酯键,并将所得核苷酸序列上的各个碱基与磷酸上的保护基脱掉;经HPLC分离纯化后,过滤除菌,冻干得到相应的正义链或反义链。
RNA抑制剂的合成工艺描述:
将正义链和反义链冻干粉分别复溶、等摩尔混合,加入注射用水适量,投入适量的TRIS缓冲溶液。轻晃约1~2min,使溶液混合均匀。将水浴锅升温至92℃~95℃。将上述反应液置水浴锅中加热3min~5min,轻晃使溶液受热均匀。自然冷却至室温。得到无色或微黄色透明液体,取样送检,测浓度。
实施例2 RNA抑制剂体外抑制LPA基因表达试验1
本实施例的RNA抑制剂选自表3,由实施例1所述方法制备得到。应用Fugene HD将质粒DNA(LPA_PSICHECK(TM)-2质粒)转入Huh7细胞。将转染后的细胞以每孔10,000个细胞的密度接种到96孔板中,每孔培养液为100μL。细胞置于5%CO2、37℃孵箱中培养过夜。然后将RNA抑制剂用PBS配制相应浓度的纳米脂质包裹的RNA抑制剂样品溶液。向对应位置每孔加入RNAiMAX/Opti-MEM,并取相应浓度稀释好的RNA抑制剂样品溶液加入孔中,混匀并孵育,取孵育后的混合液与含10%FBS的DMEM混合均匀。吸去每孔中的培养基,然后加入新的含样品的培养基,加好后置于5%CO2、37℃培养箱中培养,样品测试终浓度为5nM,0.5nM,0.05nM。
将细胞从培养箱中取出,吸弃上清,加入新鲜培养基和检测试剂,避光震荡,待细胞充分裂解后,将样品转移至不透光的白板中检测萤火虫的发光信号;向各孔中加入 检测试剂,避光震荡,检测海肾萤光素酶信号。计算各孔主报告基因与内参报告基因信号的比值。所得试验结果见下表20和图1。
表20 RNA抑制剂对Huh7细胞中LPA mRNA的抑制效果
试验结果显示,表3中RNA抑制剂在不同浓度下对Huh7细胞中LPA mRNA水平呈现出不同程度的抑制效果,且有显著的剂量依赖性,在低浓度0.05nM剂量下,除Kylo-11-DS-03、Kylo-11-DS-17、Kylo-11-DS-20、Kylo-11-DS-24和Kylo-11-DS-27的抑制率低于40%,其余的抑制剂都具有高于40%的抑制率。
实施例3载体结构的合成
当本申请的RNA抑制剂的正义链或反义链的3’端偶联有载体结构3'MVIP时,3'MVIP的solid support作为固相合成的起始单体。当本申请的RNA抑制剂的正义链或反义链的5’端偶联有载体结构5'MVIP时,5’MVIP亚磷酰胺单体作为固相合成的最后一个单体。
3’MVIP的solid spport通式如下:
m为1-4时,通式中接头B部分分别支化1至4次,以获得对应的3’MVIP的Solid Support。
5’MVIP亚磷酰胺单体通式如下:
n为1-4时,通式中接头B部分分别支化1至4次,以获得对应的5’MVIP亚磷酰胺单体。
以下仅示例性列举出几种3’MVIP Solid Support和5’MVIP亚磷酰胺单体化学合成过程。参照实施例所述的方法,本领域技术人员可以容易的合成本发明所涉及的其余的3’MVIP Solid Support和5’MVIP亚磷酰胺单体。合成工艺过程描述如下:
3.1 3’MVIP的Solid Support的合成
3.1.1 3'MVIP09的Solid Support的合成
3'MVIP09的Solid Support
合成过程描述:
3.1.1.1 ERC-01-c1的合成
称取2-氨基-1,3-丙二醇(5.0g,54.9mmol),加入DMSO 50mL、氢氧化钠溶液(1g/mL)5mL,降温到0℃,滴加丙烯酸叔丁酯(20mL,137.8mol),2小时滴加完,室温反应48h,加石油醚(100mL),饱和食盐水洗2次,有机层干燥。过层析柱(洗脱液:乙酸乙酯:石油醚=25%-75%),上柱加0.05%的三乙胺,得无色油状物6.2g。
3.1.1.2 ERC-01-c2的合成
称取ERC-01-c1(6.2g,17.9mmol),加二氯甲烷50mL、碳酸钠溶液(25%)23mL,室温滴加氯甲酸苄酯(8.2mL,57.4mmol),2小时滴加完,室温反应过夜,饱和食盐水洗涤3次,无水硫酸钠干燥,蒸干溶剂,过层析柱(乙酸乙酯:石油醚=5%-30%)得油状物4.0g。
3.1.1.3 ERC-01-c3的合成
取ERC-01-c2(4.0g,8.3mmol),加甲酸12mL,室温反应过夜,减压蒸干溶剂,得产品2.8g。
3.1.1.4 ERCd-01-c1的合成
将化合物ERC-01-c3(1.11g,3.0mmol)和dlSANC-c4(3.6g,8.04mmol)加到DMF(60mL)中,然后加入HOBt(2.24g)和HBTU(3.36g),然后缓慢加入DIEA(4.16mL)。反应液室温下搅拌反应3小时。然后加入水,水层用二氯甲烷萃取(2x10mL)。合并有机层,然后依次用饱和碳酸氢钠(80mL)、水(2x60mL)、饱和食盐水(60mL)洗。用无水硫酸钠干燥,减压蒸干,用硅胶柱层析纯化(洗脱液:3-15%MeOH in DCM)。得淡黄色固3.24g。
3.1.1.5 ERCd-01-c2的合成
ERCd-01-c1(3.24g,2.6mmol)用甲醇(60mL)溶解,加入10%钯碳(0.3g)、乙酸(2.0mL)。然后常压下加氢,反应过夜。反应液用硅藻土过滤,滤液减压蒸干,得油状物ERCd-01-c2 2.9g,其高分辨质谱图见图2。
3.1.1.6 3’MVIP09-c1的合成
向反应瓶内依次加入SANCd-01-c0(0.824g,1.5mmol)和ERCd-01-c2(1.09g,1.0mmol),再加入10mL的DCM,搅拌溶解,再依次加入TBTU(0.963g)和DIPEA(0.517g),反应过夜,加水,用DCM萃取,有机相再用饱和食盐水洗涤,干燥、过滤、浓缩,最后过硅胶柱进行纯化,得产品1.3g。
3.1.1.7 3’MVIP09-c2的合成
向反应瓶内依次加入3’MVIP09-c1(1.62g,1μmol)和10mL的DCM,室温搅拌溶解,再依次加入DMAP(0.366g)和丁二酸酐(0.2g,3μmol),室温搅拌反应,TLC分析,反应合格浓缩掉DCM,加水,用DCM萃取,有机相再用饱和食盐水洗涤,有机相经无水硫酸钠干燥、过滤、浓缩,最后过硅胶柱进行纯化,得到产品为1.55g。
3.1.1.8 3’MVIP09的Solid Support合成
向反应瓶内依次加入3’MVIP09-c2(0.86g,0.5μmol)和10mL DMF,溶解,再依次加入HBTU(0.19g)、DIPEA(0.194g)和大孔氨甲基树脂(2.0g),摇床24h,过滤,树脂用10%甲醇/DCM洗涤,再用25%醋酸/吡啶进行封端,取代度150μmol/g。
3.1.2 3'MVIP17的Solid Support的合成
3'MVIP17 Solid Support
3.1.2.1 SANC-01-c1的合成
合成步骤参照3.1.1.1.ERC-01-c1的合成。
3.1.2.2 SANC-01-c2的合成
合成步骤参照3.1.1.2.ERC-01-c2的合成。
3.1.2.3 SANC-01-c3的合成
合成步骤参照3.1.1.3.ERC-01-c3的合成。
3.1.2.4 SANCd-01-c1的合成
合成步骤参照3.1.1.4.ERCd-01-c1的合成。
3.1.2.5 SANCd-01-c2的合成
合成步骤参照3.1.1.5.ERCd-01-c2的合成。
3.1.2.6 3’MVIP17-c1的合成
合成步骤参照3.1.1.6.3’MVIP09-c1的合成,合成所得3’MVIP17-c1的高分辨质谱图见图3。
3.1.2.7 3’MVIP17-c2的合成
合成步骤参照3.1.1.7 3’MVIP09-c2的合成。
3.1.2.8 3’MVIP17的Solid Support合成
合成步骤参照3.1.1.8 3’MVIP09的Solid Support合成。
3.1.3 3'MVIP01的Solid Support的合成:
3'MVIP01 Solid Support
合成过程描述:
3.1.3.1 3’MVIP01-c1的合成
合成步骤参照3.1.1.6.3’MVIP09-c1的合成。
3.1.3.2 3’MVIP01-c2的合成
合成步骤参照3.1.1.7.3’MVIP09-c2的合成。
3.1.3.3 3’MVIP01的Solid Support合成
合成步骤参照3.1.1.8.3’MVIP09的Solid Support合成。
3.2 5’MVIP亚磷酰胺单体的合成
3.2.1 5’MVIP09亚磷酰胺单体的合成:
5'MVIP09亚磷酰胺单体
3.2.1.1 5’MVIP09-ERCd-PFP-c1的合成
称量ERCd-01-c2(2.18g,2.0mmol)溶于DMF(50mL),加戊二酸单苄酯(0.53g,2.4mmol)、DIPEA(0.78g)与TBTU(0.84g),室温搅拌过夜,加水淬灭(50mL),DCM(30mL*3)萃取,10%柠檬酸(50mL*3)、饱和碳酸氢钠50mL和吡啶100mL洗涤,无水硫酸钠干燥,过滤,旋蒸,过柱纯化得产品5’MVIP09-ERCd-PFP-c1(2.15g)。
3.2.1.2 5’MVIP09-ERCd-PFP-c2的合成
称量5’MVIP09-ERCd-PFP-c1(2.15g,1.66mmol)和10%钯碳(0.21g),加甲醇(50mL),室温搅拌加氢过夜,反应结束后硅藻土过滤钯碳,旋蒸得5’MVIP09-ERCd-PFP-c2粗品(1.9g),其高分辨率质谱图如图4所示。
3.2.1.3 5’MVIP09-ERCd-PFP的合成
称量5’MVIP09-ERCd-PFP-c2粗品(1.9g,1.58mmol)溶于DCM(60mL),加DIPEA(1.33g),冷却,加三氟乙酸五氟苯酚酯(2.21g,7.9mmol),室温搅拌反应2h后旋蒸,再溶于DCM(60mL),饱和碳酸氢钠(30mL*3)、10%柠檬酸(30mL*1)、饱和食盐水(50mL*1)洗涤,无水硫酸钠干燥,过滤,旋蒸得5’MVIP09-ERCd-PFP粗品(2.35g),抽干后无纯化直接用于下一步反应。
3.2.1.4 5’MVIP09亚磷酰胺单体-c1的合成
5’MVIP09-ERCd-PFP粗品(2.35g,1.58mmol)溶于DCM(60mL),加DIPEA(0.82g,6.32mmol)、6-氨基-1-己醇(0.37g,3.16mmol),室温搅拌反应过夜。加10%柠檬酸(30mL),DCM(30mL*3)萃取,饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤、旋蒸,过柱纯化得产品5’MVIP09单体-c1(1.73g)。
3.2.1.5 5’MVIP09亚磷酰胺单体
称量5’MVIP09亚磷酰胺单体-c1(1.3g、1.0mmol)溶于乙腈(30mL),加入二异丙胺三氮唑(0.22g),冰浴下滴加双-(二异丙基氨基)(2-氰基乙氧基)膦(0.36g,1.2mmol),室温反应4h,HPLC中控,反应合格后,浓缩过柱纯化得到产品5’MVIP09单体(1.2g)。
3.2.2 5'MVIP01亚磷酰胺单体的合成:
5'MVIP01亚磷酰胺单体
5'MVIP01的亚磷酰胺单体称量YICd-01-c2(1.12g,2.0mmol),剩余操作参照3.2.1.1.~3.2.1.5。
实施例4带载体偶联的RNA抑制剂的合成
偶联载体的反义链(3’MVIP 09偶联)合成描述:用氩气吹扫试剂瓶至少2min。向试剂瓶中依次加入亚磷酰胺单体或和乙腈,拧紧瓶盖后,震荡直至目测固体全溶。然后加入3A分子筛,静置8h以上待用。用氩气吹扫试剂瓶至少2min。向试剂瓶中依次加入氢化黄原素和干燥的吡啶,拧紧瓶盖后,震荡直至目测固体全溶,暂存待用。确认在室温20~30℃环境条件下进行以下操作:称取3’MVIP Solid Support,加入到试剂瓶中,再加入乙腈,震荡混合均匀,转移至合成柱内,并用乙腈将试剂瓶中残余的部分淋洗转移至合成柱内。淋洗完毕后加乙腈充满合成柱,记录使用乙腈的用量。按仪器操作安装固定合成柱。
将上述配制的单体溶液、CAP A、CAP B、氧化剂、硫代试剂、活化剂、脱帽剂以及乙腈,连接至AKTA PILOT100对应的管路,确保管路插入试剂瓶底。
合成方法设置完毕后,仪器各项工作准备就绪,点击运行,开始合成。在线观察记录每个detritylation峰面积。在合成过程中根据脱保护试剂实际使用量,进行补加操作。
合成结束后,氩气吹扫合成柱≥2h,按操作规程卸载合成柱。向合成柱内固相载体转移至反应瓶,加入甲胺水溶液和氨水,将反应瓶放入摇床中,35℃,2-3小时。将溶液过滤至圆底烧瓶中,再使用50%乙醇水溶液洗涤残留固相,再次过滤与之前滤液合并,将圆底烧瓶连接旋转蒸发仪,设置水温50℃蒸至无馏出,向圆底烧瓶内加入乙醇,混匀,再次蒸至无馏出,重复操作至瓶底出现白色粉末。将得到的白色粉末配制成溶液,使用反向层析柱进行纯化,取样检测OD260、纯度。将纯化的反义链溶液分装于西林瓶中冻干备用,并将产品密封储存于-20℃冰箱中。
偶联载体的正义链(5’MVIP09偶联)的合成操作同反义链,其中装柱的Solid Support为Universal载体。所得中间体加DIPEA配制成溶液,加入5’MVIP亚磷酰胺单体,混匀,将反应瓶放入摇床中,35℃,2-3小时。
RNA抑制剂的合成退火工艺描述:
取所得的正义链和反义链,1:1等摩尔混合在反应瓶中,水浴95℃5分钟后,关闭水浴锅电源,使其自然降温至40℃以下。向双链溶液中加入3M醋酸钠水溶液,混合均匀后,再加入适量体积的无水乙醇,混合均匀,将反应液放入-20℃冰箱内45min。冷冻高速离心机设置4℃预制冷,温度达到后,放入双链溶液,启动离心机。取出离心后的双链溶液,去除上清液,加入超纯水使固体完全溶解,取样检测OD260、纯度,可得到表14中的RNA抑制剂,将纯化的成品溶液分装于西林瓶中冻干备用,并将产品密封储存于-20℃冰箱中。
以上仅示例性列举出带有5'MVIP09/3'MVIP09偶联的RNA抑制剂的合成,本发明中所述的但未列举出来的RNA抑制剂都适用于这个规则,即当RNA抑制剂的正义链或反义链的3'端偶联有载体结构3'MVIP时,3'MVIP的solid support作为固相合成的起始单体;当RNA抑制剂的正义链或反义链的5'端偶联有载体结构5'MVIP时,5'MVIP亚磷酰胺单体作为固相合成的最后一个单体。参照本实施例所述的方法,本领域技术人员可以容易的合成本发明所涉及的其余的RNA抑制剂的合成。
实施例5 RNA抑制剂体外抑制LPA基因表达试验2
本实施例的RNA抑制剂选自表4,正义链和反义链的不同核苷酸糖基2'位具有甲氧基或氟代修饰。
由实施例4所述的方法制备RNA抑制剂。参照实施例2的试验方式,考察RNA抑制剂在浓度5nM和0.5nM下对Huh7细胞中的LPA mRNA抑制效果。所得试验结果见表21和图5。
表21 RNA抑制剂对Huh7细胞中LPA mRNA的抑制效果
试验结果显示,RNA抑制剂Kylo-11-DS53在不同浓度下,对Huh7细胞的LPA mRNA水平抑制效果显著,其特征是正义链5’端开始的第5、7、8、9位核苷酸糖基2’位均是氟,其余为甲氧基,5’端和3’端连续三个核苷酸之间磷酸酯键有硫代,反义链5’端开始的第2、4、8、14、16位核苷酸糖基2’位均是氟,其余为甲氧基,5’端和3’端连续三个核苷酸之间的磷酸酯键有硫代。
实施例6 RNA抑制剂体外抑制LPA基因表达试验3
本实施例的RNA抑制剂选自表5,正义链和反义链的不同核苷酸糖基2’位具有甲氧基或氟代修饰。
由实施例4所述的方法制备RNA抑制剂。参照实施例2的试验方式,考察RNA抑制剂在浓度5nM和0.5nM下对Huh7细胞的LPAmRNA抑制效果。所得试验结果见表22和图6。
表22 RNA抑制剂对Huh7细胞中LPA mRNA的抑制效果
试验结果显示,RNA抑制剂Kylo-11-DS65、Kylo-11-DS71、Kylo-11-DS72、和Kylo-11-DS74在不同浓度下,对Huh7细胞的LPA mRNA水平抑制效果显著。Kylo-11-DS65其特征是正义链5’端开始的第5、7、8、9位核苷酸糖基2’位均是氟,其余为甲氧基,5’端和3’端连续三个核苷酸之间磷酸酯键有硫代,反义链5’端开始的第2、4、8、14、16位核苷酸糖基2’位均是氟,其余为甲氧基,5’端和3’端连续三个核苷酸之间的磷酸酯键有硫代。
Kylo-11-DS71和Kylo-11-DS72其特征是正义链5’端开始的第3、5、7、8、9、10、11、13、15位核苷酸糖基2’位均是氟,其余为甲氧基,5’端和3’端连续三个核苷酸之间磷酸酯键有硫代,反义链5’端开始的第2、4、6、8、14、16位核苷酸糖基2’位均是氟,其余为甲氧基,5’端和3’端连续三个核苷酸之间的磷酸酯键有硫代。Kylo-11-DS74的特征是是正义链5’端开始的第9、10、11位核苷酸糖基2’位均是氟,其余为甲氧基,5’端和3’端连续三个核苷酸之间磷酸酯键有硫代,反义链5’端开始的第2、4、6、8、14、16、18、20位核苷酸糖基2’位均是氟,其余为甲氧基,5’端和3’端连续三个核苷酸之间的磷酸酯键有硫代。
实施例7 RNA抑制剂体外抑制LPA基因表达试验4
本实施例的RNA抑制剂选自表6中的RNA抑制剂。候选的RNA抑制剂Kylo-11-DS81~Kylo-11-DS105的正义链和反义链的不同核苷酸糖基2’位具有甲氧基或氟代修饰。
由实施例4所述的方法制备RNA抑制剂。参照实施例2的实验方式,考察RNA抑制剂在浓度5nM和0.5nM下对Huh7细胞的LPAmRNA抑制效果。所得试验结果见表23和图7。
表23 RNA抑制剂对Huh7细胞中LPA mRNA的抑制效果
试验结果显示,RNA抑制剂Kylo-11-DS87、Kylo-11-DS88、Kylo-11-DS91、Kylo-11-DS92、Kylo-11-DS97、Kylo-11-DS98、Kylo-11-DS101、Kylo-11-DS102和Kylo-11-DS104在不同浓度下对Huh7细胞的LPA mRNA水平抑制效果显著。Kylo-11-DS87其特征是正义链5’端开始的第7、8、9、10位核苷酸糖基2’位均是氟,反义链5’端开始的第2、4、6、8、14位核苷酸糖基2’位均是氟;Kylo-11-DS88其特征是正义链5’端开始的第5、8、9、10位核苷酸糖基2’位均是氟,反义链5’端开始的第2、15、17位核苷酸糖基2’位均是氟;Kylo-11-DS91其特征是正义链5’端开始的第8、9、10位核苷酸糖基2’位均是氟,反义链5’端开始的第2、4、12、14位核苷酸糖基2’位均是氟;Kylo-11-DS92其特征是正义链5’端开始的第7、8、9、10位核苷酸糖基2’位均是氟,反义链5’端开始的第2、5、14、16位核苷酸糖基2’位均是氟;Kylo-11-DS97其特征是正义链5’端开始的第5、7、8、9位核苷酸糖基2’位均是氟,反义链5’端开始的第7、12、14位核苷酸糖基2’位均是氟;Kylo-11-DS98其特征是正义链5’端开始的第7、9、10、11位核苷酸糖基2’位均是氟,反义链5’端开始的第2、14位核苷酸糖基2’位均是氟;Kylo-11-DS101其特征是正义链5’端开始的第7、9、10、11位核苷酸糖基2’位均是氟,反义链5’端开始的第2、14位核苷酸糖基2’位均是氟;Kylo-11-DS102其特征是正义链5’端开始的第5、7、8、9位核 苷酸糖基2’位均是氟,反义链5’端开始的第2、6、8、10、14、16位核苷酸糖基2’位均是氟;Kylo-11-DS104其特征是正义链5’端开始的第9、10、11位核苷酸糖基2’位均是氟,反义链5’端开始的第14、16位核苷酸糖基2’位均是氟。这些RNA抑制剂共同特征是,除上述位置的核苷酸糖基2’位氟代外,其余的核苷酸糖基2’位为甲氧基,5’端和3’端连续三个核苷酸之间的磷酸酯键有硫代。其中Kylo-11-DS102对Huh7细胞的LPA mRNA水平的抑制效果最佳,在0.5nM浓度下达72.99%的抑制率。
实施例8 RNA抑制剂体外抑制LPA基因表达试验5
本实施例的RNA抑制剂选自表7,正义链和反义链的不同核苷酸糖基2’位具有甲氧基或氟代修饰。
由实施例4所述的方法制备RNA抑制剂。参照实施例2的试验方式,考察RNA抑制剂在浓度5nM和0.5nM下对Huh7细胞的LPA mRNA抑制效果。所得试验结果见表24和图8。
表24 RNA抑制剂对Huh7细胞中LPA mRNA的抑制效果
试验结果显示,RNA抑制剂Kylo-11-DS106~Kylo-11-DS110、Kylo-11-DS112~Kylo-11-DS117和Kylo-11-DS119在不同浓度下对Huh7细胞的LPA mRNA抑制效果显著。其中,Kylo-11-DS106~Kylo-11-DS110、Kylo-11-DS112~Kylo-11-DS114和Kylo-11-DS116彼此之间具有相同的至少15个连续核苷酸,且正义链5’端开始的第3、5、7、8、9、10、11、13、15位核苷酸糖基2’位均是氟,其余的2’位为甲氧基,5’端和3’端连续三个核苷酸之间的磷酸酯键有硫代,反义链5’端开始的第2、4、6、8、14、16位核苷酸糖基2’位均是氟,其余为甲氧基,5’端和3’端连续三个核苷酸之间的磷酸酯键有硫代,正义链和反义链都包含有21个核苷酸(除 Kylo-11-DS109),在正义链和反义链的3’端各有两个核苷酸的突出端;Kylo-11-DS106与Kylo-11-DS115仅相差一个核苷酸,从反义链5’端开始的第6位核苷酸,前者为fU,后者为dT,两者活性相近,都具有较高的抑制活性。Kylo-11-DS116和Kylo-11-DS117的正义链和反义链的未修饰序列一样,都分别为SEQ ID NO.8和SEQ ID NO.21,两者的修饰特征不一样,后者的正义链5’端开始的第5、7、8、9位核苷酸糖基2’位均是氟,反义链5’端开始的第2、6、8、10、14、16位核苷酸糖基2’位均是氟。RNA抑制剂Kylo-11-DS119的正义链和反义链的未修饰序列分别为SEQ ID NO.11和SEQ ID NO.25,其修饰特征是正义链5’端开始的第9、10、11位核苷酸糖基2’位均是氟,反义链5’端开始的第2、4、6、12、14、16、18、20位核苷酸糖基2’位均是氟,正义链与反义链都含有21个核苷酸,两端都为平端。
实施例9 RNA抑制剂体外抑制LPA基因表达试验6
本实施例的RNA抑制剂选自表8,正义链和反义链的不同核苷酸糖基2’位具有甲氧基或氟代修饰。由实施例4所述的方法制备RNA抑制剂。参照实施例2的试验方式,考察RNA抑制剂在浓度5nM和0.5nM下对Huh7细胞的LPA mRNA抑制效果。所得试验结果见表25和图9。
表25 RNA抑制剂对Huh7细胞中LPA mRNA的抑制效果
试验结果显示,RNA抑制剂Kylo-11-DS-124、Kylo-11-DS-126、Kylo-11-DS-130和Kylo-11-DS-131在不同浓度下对Huh7细胞的LPA mRNA抑制效果显著。这些RNA抑制剂共同特征是正义链5’端开始的第9、10、11位核苷酸糖基2’位均是氟,其余为甲氧基,5’端和3’端连续三个核苷酸之间的磷酸酯键有硫代。反义链5’端开始的第2、4、6、12、14、16、18、 20位核苷酸糖基2’位均是氟。其中,Kylo-11-DS-126和Kylo-11-DS-131正义链与反义链都含有21个核苷酸,3’端都有两个核苷酸的突出端,所不同的是前者的反义链5’端开始的第6位核苷酸为fA,后者同样位置为dT,两者呈现的活性相近。Kylo-11-DS-124和Kylo-11-DS-130正义链含有19个核苷酸,反义链含有21个核苷酸,反义链3’端有两个核苷酸突出端,正义链3’端与反义链5’端为平端,两者呈现的活性相近。
实施例10应用食蟹猴评估RNA抑制剂的体内活性
取适龄的食蟹猴做实验评估RNA抑制剂Kylo-11-DS148、Kylo-11-DS146和Kylo-11-DS163。在Day0分别通过皮下注射给药6mg/kg。在给药后每周采血,测LDL-c和Lp(a)水平。RNA抑制剂干预后,LDL-c和Lp(a)水平结果见表26-27及图10-11。
表26 RNA抑制剂对食蟹猴血浆的LDL-c水平的降低效果
表27 RNA抑制剂对食蟹猴血浆的Lp(a)水平的降低效果
试验结果显示,RNA抑制剂Kylo-11-DS146、Kylo-11-DS 148和Kylo-11-DS163可以使食蟹猴血浆中的LDL-c水平有所降低,可以显著持续的降低血浆中Lp(a)水平,其中Kylo-11-DS146干预后,与给药前相比,个体最高可使血浆中Lp(a)水平降低95.25%,至day77,仍可维持89.98%的平均降低率。
实施例11不同双链体的体外抑制效果对比
CN202210241706.1(以下简称D1)是本申请人“厦门甘宝利生物医药有限公司”在2022年3月11日提交的中国发明专利申请,其发明名称为LPA抑制剂及其用途,其公开号为CN114703184A。本申请人对不同的双链体进行如下试验操作:
取Huh7细胞,先用PBS洗涤后,用0.05%胰蛋白酶进行消化,用含10%FBS的DMEM将细胞轻轻吹打至单个细胞后计数。应用Fugene HD将质粒DNA转入Huh7细胞。将转染后的细胞以每孔10,000个细胞的密度接种到96孔板中,每孔培养液为100μL。细胞置于5%CO2、37℃孵箱中培养过夜。
配制RNAiMAX转染试剂,以RNAiMAX:Opti-MEM=1.5:48.5比例按需配置适量体积于15mL离心管,涡旋15秒,混匀,室温孵育15分钟。向对应位置每孔加入60μL RNAiMAX/Opti-MEM,并取相应浓度60μL稀释好的化合物加入孔中,混匀并孵育15分钟。吸去每孔中的培养基,然后加入新的含化合物的培养基,此时每孔含有培养基120μL,加好后置于5%CO2、37℃培养箱箱中培养,化合物测试终浓度为5nM、0.5nM和0.05nM。
实验步骤参照双萤光素酶检测系统说明书,简述方法如下:将细胞从培养箱中取出,吸弃上清,加入75μL新鲜培养基和75μL检测试剂,避光震荡10分钟,待细胞充分裂解后,将100μL样品转移至不透光的白板中检测萤火虫的发光信号;向各孔中加入50μL检测试剂,避光震荡10分钟后检测海肾萤光素酶信号。计算各孔主报告基因与内参报告基因信号的比值,并通过该比值计算抑制率%。
试验结果如下:
由上述试验结果所示,本申请要求获得保护的2对原始序列作用的靶序列包含LPA mRNA NM_005577.4(SEQ ID NO.1)的区域与D1中的不同,比D1中的区域向前扩大了2位或向后扩大了1位,本申请中Kylo-11-DS13和Kylo-11-DS11在5nM、0.5nM和0.05nM浓度的抑制率分别高于D1中的Ky-11-DS12和Ky-11-DS08。

Claims (13)

  1. 一种抑制LPA基因表达的RNA抑制剂或其药学上可接受的盐,其中,
    所述RNA抑制剂由链长为15-30的正义链和反义链通过碱基配对形成,所述链长优选为19-23,并且所述正义链和反义链之间至少有85%的碱基互补;
    所述正义链和/或反义链的部分或全部核苷酸糖基2'位的-OH可以被取代,其中,所述取代基团为氟或甲氧基;
    且所述正义链和/或反义链的末端中至少有一个末端的三个相邻核苷酸之间的磷酸酯键可以被硫代。
  2. 根据权利要求1所述的RNA抑制剂或其药学上可接受的盐,其中,所述反义链与靶序列形成互补性区域,所述靶序列为LPA mRNA不同位置处的多个区域,所述多个区域具有相同的至少15个连续的核苷酸,所述靶序列选自LPA mRNA(NM_005577.4)中312-332、654-674、996-1016、1338-1358、1680-1700、2022-2042和2364-2384之间的任意一个核苷酸区域。
  3. 根据权利要求1所述的RNA抑制剂或其药学上可接受的盐,,其中,所述反义链与靶序列形成互补性区域,所述靶序列为LPA mRNA不同位置处的多个区域,所述多个区域具有相同的至少15个连续的核苷酸,所述靶序列选自LPA mRNA(NM_005577.4)中493-512、1861-1880和2203-2222之间的任意一个核苷酸区域。
  4. 根据权利要求1-3中任一项所述的RNA抑制剂或其药学上可接受的盐,其中,所述反义链选自以下序列:
    5'ucguauaacaauaaggagcug 3'  SEQ ID NO.25
    5'auaacucuguccauuaccaug 3'  SEQ ID NO.21
    或与上述反义链具有相同的至少15个连续核苷酸的序列,或与上述反义链相差一个、两个或三个核苷酸的序列,
    其中,g=鸟苷酸,a=腺苷酸,u=尿苷酸,c=胞苷酸。
  5. 根据权利要求1-3中任一项所述的RNA抑制剂或其药学上可接受的盐,其中,所述正义链选自以下序列:
    5'cagcuccuuauuguuauacga 3'  SEQ ID NO.11
    5'ugguaauggacagaguuauca 3'  SEQ ID NO.8
    或与上述正义链具有相同的至少15个连续核苷酸的序列,或与上述正义链相差一个、两个或三个核苷酸的序列,
    其中,g=鸟苷酸,a=腺苷酸,u=尿苷酸,c=胞苷酸。
  6. 根据权利要求1-5中任一项所述的RNA抑制剂或其药学上可接受的盐,其中,所述正义链为SEQ ID NO.11或与其具有相同的至少15个连续核苷酸的序列,或与其相差一个、两个或三个核苷酸的序列;且所述反义链为SEQ ID NO.25或与其具有相同的至少15个连续核苷酸的序列,或与其相差一个、两个或三个核苷酸的序列:
    正义链:5'cagcuccuuauuguuauacga 3'  SEQ ID NO.11
    反义链:5'ucguauaacaauaaggagcug 3'  SEQ ID NO.25;
    或者,所述正义链为SEQ ID NO.8或与其具有相同的至少15个连续核苷酸的序列,或与其相差一个、两个或三个核苷酸的序列;且所述反义链为SEQ ID NO.21或与其具有相同的至少15个连续核苷酸的序列,或与其相差一个、两个或三个核苷酸的序列:
    正义链:5'ugguaauggacagaguuauca 3'  SEQ ID NO.8
    反义链:5'auaacucuguccauuaccaug 3'  SEQ ID NO.21;
    其中,g=鸟苷酸,a=腺苷酸,u=尿苷酸,c=胞苷酸。
  7. 根据权利要求6所述的RNA抑制剂或其药学上可接受的盐,其中,所述正义链为SEQ ID NO.270或与其相差一个、两个或三个核苷酸的序列,且所述反义链为SEQ ID NO.278或与其相差一个、两个或三个核苷酸的序列:
    正义链:5'CsAsGCUCCUfUfAfUUGUUAUACsGsA 3'  SEQ ID NO.270
    反义链:5'UsfCsGfUAfUAACAAfUAfAGfGAfGCsfUsG 3'  SEQ ID NO.278;
    或者,所述正义链为SEQ ID NO.239或与其相差一个、两个或三个核苷酸的序列,且所述反义链为SEQ ID NO.344或与其相差一个、两个或三个核苷酸的序列:
    正义链:5'UsGsGUfAAfUfGfGACAGAGUUAUsCsA 3'  SEQ ID NO.239
    反义链:5'AsfUsAfACdTCfUGUCCAfUUfACCAsUsG 3'  SEQ ID NO.344;
    其中,G=2'-O-甲基鸟苷酸,A=2'-O-甲基腺苷酸,U=2'-O-甲基尿苷酸,C=2'-O-甲基胞苷酸;Gs=2'-O-甲基-3'-硫代鸟苷酸,As=2'-O-甲基-3'-硫代腺苷酸,Us=2'-O-甲基-3'-硫代尿苷酸,Cs=2'-O-甲基-3'-硫代胞苷酸;fG=2'-氟鸟苷酸,fA=2'-氟腺苷酸,fU=2'-氟尿苷酸,fC=2'-氟胞苷酸;fGs=2'-氟-3'-硫代鸟苷酸,fAs=2'-氟-3'-硫代腺苷酸,fUs=2'-氟-3'-硫代尿苷酸,fCs=2'-氟-3'-硫代胞苷酸,dT=2'-脱氧‐胸苷酸。
  8. 根据权利要求1-7中任一项所述的RNA抑制剂或其药学上可接受的盐,其中,所述RNA抑制剂还含有载体结构5'MVIP和3'MVIP,所述RNA抑制剂的结构如式Ia、Ib或Ic所示:

    其中,
    所述5'MVIP由转接点R1、连接链D、接头B、支链L和肝靶向特异性配体X组成,其通过转接点R1与正义链5'端或反义链5'端连接,其结构如通式I所示:
    (X-L)n-B-D-R1-
    I
    所述3'MVIP由转接点R2、连接链D、接头B、支链L和肝靶向特异性配体X组成,其通过转接点R2与正义链3'端或反义链3'端连接,其结构如通式II所示:
    (X-L)m-B-D-R2-
    II
    其中,
    n和m各自独立地为0-4的任意整数,各自独立地优选为1-3的整数,且n+m=2-6的整数,优选n+m=2、3或4,更优选为4;
    所述转接点R1为如下所示的含有N、S或O的杂环或碳环结构:
    或者,所述R1为-NH(CH2)xCH2O-,其中x为3-12的任意整数,优选为4-6的任意整数;
    所述转接点R2为如下所示的含有N、S或O的杂环或碳环结构:
    或者,所述转接点R2为-NH(CH2)x1CH(OH)(CH2)x2CH2O-,其中x1为1-4的任意整数,x2为0-4的任意整数;
    所述肝靶向特异性配体X在5'MVIP与3'MVIP各自的内部或5'MVIP与3'MVIP之间相同或不同,其选自单糖及其衍生物,优选为为N-乙酰半乳糖胺及其衍生物,更优选地选自以下结构:
    其中,W选自-OH、-NHCOOH和-NHCO(CH2)qCH3中的一种或两种,其中q为0-4的整数;
    所述支链L在5'MVIP与3'MVIP各自的内部或5'MVIP与3'MVIP之间相同或不同,其选自如下结构中的一种或多种:

    其中,r1是1-12的任意整数,r2为0-20的任意整数,Z为H、烷基或酰胺基,所述烷基如C1-C5烷基;
    所述接头B在5'MVIP与3'MVIP各自的内部或5'MVIP与3'MVIP之间相同或不同,其选自以下结构:
    其中,A1和A2各自独立地是C、O、S、-NH-、羰基、酰胺基、磷酰基或硫代磷酰基,r为0-4的任意整数;
    所述连接链D在5'MVIP与3'MVIP各自的内部或5'MVIP与3'MVIP之间相同或不同,其选自以下结构:

    其中,每个p各自独立地为1-20的任意整数;s为2-13的任意整数;Z1和Z2为相同或者不同的取代基团。
  9. 根据权利要求8所述的RNA抑制剂或其药学上可接受的盐,其中,所述5'MVIP为如下所示的5'MVIP01或5'MVIP09,所述3'MVIP为如下所示的3'MVIP01、3'MVIP09或3'MVIP17:
  10. 根据权利要求9所述的RNA抑制剂或其药学上可接受的盐,其中,所述正义链5'MVIP和反义链3'MVIP的组合为5'MVIP01/3'MVIP01、5'MVIP01/3'MVIP17或5'MVIP09/3'MVIP09,或者所述正义链5'MVIP和正义链3'MVIP的组合为5'MVIP01/3'MVIP09或5'MVIP09/3'MVIP01。
  11. 根据权利要求10所述的RNA抑制剂或其药学上可接受的盐,其中,所述RNA抑制剂选自Kylo-11-DS146、Kylo-11-DS148和Kylo-11-DS163。
  12. 如权利要求1-11中任一项所述的RNA抑制剂剂或其药学上可接受的盐在制备用于治疗和/或预防与LP(a)的水平升高相关的疾病的药物中的应用,其中,所述疾病包括但不限于炎性疾病、心脑血管疾病和代谢疾病,其中,所述心脑血管疾病包括高LP(a)血症、高脂血症、中风、动脉粥样硬化、血栓形成、冠心病和主动脉瓣狭窄。
  13. 一种药物组合物,该药物组合物包括权利要求1-11中任一项所述的RNA抑制剂或其药学上可接受的盐和药学上可接受的辅料,其剂型为口服剂、静脉注射剂或者皮下或肌内注射剂,优选为皮下注射剂。
PCT/CN2023/125861 2022-10-24 2023-10-23 一种抑制lpa基因表达的rna抑制剂及其应用 WO2024088190A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211304230.8 2022-10-24
CN202211304230.8A CN115851723B (zh) 2022-10-24 2022-10-24 一种抑制lpa基因表达的rna抑制剂及其应用

Publications (1)

Publication Number Publication Date
WO2024088190A1 true WO2024088190A1 (zh) 2024-05-02

Family

ID=85661740

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/125861 WO2024088190A1 (zh) 2022-10-24 2023-10-23 一种抑制lpa基因表达的rna抑制剂及其应用

Country Status (2)

Country Link
CN (1) CN115851723B (zh)
WO (1) WO2024088190A1 (zh)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114703184A (zh) * 2022-03-11 2022-07-05 厦门甘宝利生物医药有限公司 Lpa抑制剂及其用途
CN115851723B (zh) * 2022-10-24 2023-10-03 厦门甘宝利生物医药有限公司 一种抑制lpa基因表达的rna抑制剂及其应用

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108368506A (zh) * 2015-10-01 2018-08-03 箭头药业股份有限公司 用于抑制lpa的基因表达的组合物和方法
CN111465694A (zh) * 2017-11-13 2020-07-28 赛伦斯治疗有限责任公司 用于抑制细胞中lpa的表达的核酸
CN113227372A (zh) * 2018-11-13 2021-08-06 赛伦斯治疗有限责任公司 用于抑制细胞中lpa的表达的核酸
WO2022032288A1 (en) * 2020-08-05 2022-02-10 Dicerna Pharmaceuticals, Inc. Compositions and methods for inhibiting lpa expression
WO2022098841A1 (en) * 2020-11-05 2022-05-12 Amgen Inc. METHODS FOR TREATING ATHEROSCLEROTIC CARDIOVASCULAR DISEASE WITH LPA-TARGETED RNAi CONSTRUCTS
WO2022121959A1 (zh) * 2020-12-09 2022-06-16 纳肽得有限公司 siRNA分子及其在治疗冠状动脉疾病中的应用
CN114703184A (zh) * 2022-03-11 2022-07-05 厦门甘宝利生物医药有限公司 Lpa抑制剂及其用途
CN115851723A (zh) * 2022-10-24 2023-03-28 厦门甘宝利生物医药有限公司 一种抑制lpa基因表达的rna抑制剂及其应用

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108368506A (zh) * 2015-10-01 2018-08-03 箭头药业股份有限公司 用于抑制lpa的基因表达的组合物和方法
CN111465694A (zh) * 2017-11-13 2020-07-28 赛伦斯治疗有限责任公司 用于抑制细胞中lpa的表达的核酸
CN113227372A (zh) * 2018-11-13 2021-08-06 赛伦斯治疗有限责任公司 用于抑制细胞中lpa的表达的核酸
WO2022032288A1 (en) * 2020-08-05 2022-02-10 Dicerna Pharmaceuticals, Inc. Compositions and methods for inhibiting lpa expression
WO2022098841A1 (en) * 2020-11-05 2022-05-12 Amgen Inc. METHODS FOR TREATING ATHEROSCLEROTIC CARDIOVASCULAR DISEASE WITH LPA-TARGETED RNAi CONSTRUCTS
WO2022121959A1 (zh) * 2020-12-09 2022-06-16 纳肽得有限公司 siRNA分子及其在治疗冠状动脉疾病中的应用
CN114703184A (zh) * 2022-03-11 2022-07-05 厦门甘宝利生物医药有限公司 Lpa抑制剂及其用途
CN115851723A (zh) * 2022-10-24 2023-03-28 厦门甘宝利生物医药有限公司 一种抑制lpa基因表达的rna抑制剂及其应用

Also Published As

Publication number Publication date
CN115851723A (zh) 2023-03-28
CN115851723B (zh) 2023-10-03

Similar Documents

Publication Publication Date Title
WO2024088190A1 (zh) 一种抑制lpa基因表达的rna抑制剂及其应用
US20220056454A1 (en) RNAi Agents for Inhibiting Expression of 17beta-HSD Type 13 (HSD17B13), Compositions Thereof, and Methods of Use
CN111107853A (zh) 用于抑制载脂蛋白C-III (APOC3)的表达的RNAi试剂和组合物
JP7383308B2 (ja) 新規化合物及びその適用
WO2023169548A1 (zh) Lpa抑制剂及其用途
KR20080066987A (ko) siRNA에 대한 전달 운반체로서의 펩티드-다이서 기질RNA 접합체들
WO2023066236A1 (zh) Agt抑制剂及其用途
CN113234725B (zh) 一种抑制pcsk9基因表达的rna抑制剂及其应用
JP2022511866A (ja) 化学修飾されたRNAiコンストラクト及びその使用
US20220389430A1 (en) Chemical modifications of small interfering rna with minimal fluorine content
JP7476422B2 (ja) Lpa発現を阻害するための組成物及び方法
CN114716518A (zh) 一种能够抑制pcsk9表达的分子构造及药物组合物
CN111212909A (zh) 用于抑制去唾液酸糖蛋白受体1的表达的RNAi试剂和组合物
WO2023134705A1 (zh) 抑制angptl3表达的rna干扰剂及其用途
WO2024027809A1 (zh) 一种抑制apoc3基因表达的rna抑制剂及其应用
CN117858949A (zh) 用于抑制粘蛋白5AC(MUC5AC)的表达的RNAi试剂、其组合物及其使用方法
JP2024520522A (ja) ムチン5AC(MUC5AC)の発現を阻害するためのRNAi剤、その組成物、及び使用方法
TW202345873A (zh) 調節scap活性之組合物及方法
JP2024514880A (ja) Pnpla3発現を調節するための組成物及び方法
TW202405174A (zh) 用於抑制超氧化物歧化酶1(SOD1)表現之RNAi藥劑、其組合物及使用方法
WO2023102469A2 (en) Compositions and methods for modulating apoc3 expression