WO2019177061A1 - 核酸複合体 - Google Patents
核酸複合体 Download PDFInfo
- Publication number
- WO2019177061A1 WO2019177061A1 PCT/JP2019/010392 JP2019010392W WO2019177061A1 WO 2019177061 A1 WO2019177061 A1 WO 2019177061A1 JP 2019010392 W JP2019010392 W JP 2019010392W WO 2019177061 A1 WO2019177061 A1 WO 2019177061A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- nucleic acid
- acid strand
- acid complex
- strand
- group
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/54—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
- A61K47/549—Sugars, nucleosides, nucleotides or nucleic acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/711—Natural deoxyribonucleic acids, i.e. containing only 2'-deoxyriboses attached to adenine, guanine, cytosine or thymine and having 3'-5' phosphodiester links
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/713—Double-stranded nucleic acids or oligonucleotides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/54—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
- A61K47/543—Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
- A61K47/544—Phospholipids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-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
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/11—Antisense
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/11—Antisense
- C12N2310/113—Antisense targeting other non-coding nucleic acids, e.g. antagomirs
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/30—Chemical structure
- C12N2310/34—Spatial arrangement of the modifications
- C12N2310/341—Gapmers, i.e. of the type ===---===
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/50—Physical structure
- C12N2310/53—Physical structure partially self-complementary or closed
Definitions
- the present invention relates to a nucleic acid complex or a salt thereof capable of producing an antisense effect in the nervous system, particularly the central nervous system, and a composition containing the same.
- nucleic acid pharmaceuticals In recent years, oligonucleotides have attracted interest in the ongoing development of pharmaceuticals called nucleic acid pharmaceuticals, and in particular, nucleic acid pharmaceuticals using antisense methods in view of high target gene selectivity and low toxicity. Development is actively underway.
- the antisense method uses a partial sequence of mRNA or miRNA transcribed from a target gene as a target sense strand and a complementary oligonucleotide (antisense oligonucleotide: often referred to as "ASO (AntiSense Oligonucleotide)" in this specification)
- ASO AntiSense Oligonucleotide
- Patent Document 1 comprises a first oligomer compound and a second oligomer compound containing a conjugated group such as cholesterol, and regulates the amount and activity of target nucleic acid in extrahepatic tissue or extrahepatic cells, or in hepatic tissue or hepatocytes.
- a conjugated group such as cholesterol
- the brain has a mechanism called the blood-brain barrier (hereinafter referred to as “BBB”) that selects and restricts substances that are transferred to the brain via blood. . While this BBB mechanism protects the brain from harmful substances, it also serves as a barrier to drug delivery to the brain. Therefore, there is a need for a method of delivering a nucleic acid agent such as ASO to the central nervous system including the brain.
- BBB blood-brain barrier
- the present invention relates to a nucleic acid agent that is efficiently delivered to the nervous system, particularly the central nervous system where drug delivery is inhibited by the BBB mechanism, and that provides an antisense effect to a target transcript at the delivery site, and a composition comprising the same It is an issue to provide.
- the present inventors have found that a nucleic acid complex obtained by annealing ASO and a complementary strand of ASO bound with phosphatidylethanolamine or an analog thereof is a central nervous system. It has been found that it is delivered efficiently and has a high antisense effect. Based on these findings, the present inventors have completed the present invention. That is, the present invention includes the following.
- nucleic acid complex comprising a first nucleic acid strand and a second nucleic acid strand or a salt thereof (in the present specification, these may be collectively abbreviated as “the nucleic acid complex of the present invention” in some cases)
- the first nucleic acid strand includes a base sequence capable of hybridizing to at least a part of the target transcript, and has an antisense effect on the target transcript, and the second nucleic acid strand is the second nucleic acid strand.
- the nucleic acid complex or salt thereof comprising a base sequence complementary to one nucleic acid strand and bound to phosphatidylethanolamine or an analogue thereof, wherein the first nucleic acid strand is annealed to the second nucleic acid strand .
- R 1 and R 2 each independently represents a substituted or unsubstituted C 5 to C 32 alkyl group, or a substituted or unsubstituted C 5 to C 32 alkenyl group.
- n 0 or 1
- (6) The nucleic acid complex according to (5), wherein phosphatidylethanolamine or an analog thereof is bonded to the 5 ′ end of the second nucleic acid strand via the linker.
- (7) The nucleic acid complex or a salt thereof according to (5) or (6), wherein the linker-bound phosphatidylethanolamine or an analog thereof is represented by the general formula III. (Wherein 5 'oligo 5 oligonucleotide' to a terminal.
- R 1 and R 2, and R 1 and R 2 according to the general formula I, and n is the general It is synonymous with n described in Formula II.
- (8) The nucleic acid complex or a salt thereof according to any one of (1) to (7), wherein the first nucleic acid strand includes at least 4 consecutive deoxyribonucleosides.
- nucleic acid strand comprises at least 4 consecutive ribonucleosides complementary to at least 4 consecutive deoxyribonucleosides in the first nucleic acid strand Or a salt thereof.
- the nucleic acid complex or salt thereof according to any one of (1) to (13), wherein the second nucleic acid strand does not contain a natural ribonucleoside.
- nucleic acid complex according to any one of (1) to (14), wherein the nucleic acid part of the second nucleic acid strand is composed of deoxyribonucleoside and / or sugar-modified nucleoside linked by a modified or unmodified internucleoside bond Body or salt thereof.
- a composition for regulating the expression or editing of a target transcript in the central nervous system comprising the nucleic acid complex according to any one of (1) to (15) or a salt thereof.
- the composition according to (16) which is used for treatment of central nervous system diseases.
- a composition for delivering a central nervous system drug comprising the nucleic acid complex according to any one of (1) to (15) or a salt thereof.
- the central nervous system is the frontal lobe, temporal lobe, hippocampus, parahippocampal, parietal lobe, occipital lobe, striatum, pallidum, forehead, thalamus, hypothalamic nucleus, midbrain, substantia nigra, bridge
- composition according to any one of (16) to (20), which is for intravenous administration or subcutaneous administration (22) The composition according to any one of (16) to (21), which comprises 5 mg / kg or more of the nucleic acid complex or a salt thereof in one dose.
- composition according to any one of (16) to (22), wherein the nucleic acid complex or a salt thereof passes through the BBB This specification includes the disclosure of Japanese Patent Application No. 2018-047294, which is the basis of the priority of the present application.
- nucleic acid agent that is efficiently delivered to the central nervous system and provides an antisense effect at the delivery location, and a composition containing the same.
- FIG. 1 is a schematic diagram showing the basic structure of the nucleic acid complex of the present invention. This figure shows two modes depending on the binding position of phosphatidylethanolamine in the second nucleic acid strand.
- Figure 1a shows a nucleic acid complex with phosphatidylethanolamine bound to the 5 'end of the second nucleic acid strand
- Figure 1b shows a nucleic acid complex with phosphatidylethanolamine bound to the 3' end of the second nucleic acid strand. Showing the body.
- phosphatidylethanolamine may be bound to both ends of the 5 ′ end and 3 ′ end of the second nucleic acid strand.
- FIG. 1 is a schematic diagram showing the basic structure of the nucleic acid complex of the present invention. This figure shows two modes depending on the binding position of phosphatidylethanolamine in the second nucleic acid strand.
- Figure 1a shows a nucleic acid complex with phosphatidylethanolamine bound to the 5 '
- FIG. 2 is a diagram showing an example of a general mechanism of the antisense method.
- “X” indicates a place to be suppressed or inhibited in the process from gene expression to translation.
- the diagram in the broken line is a schematic diagram in which the heteroduplex portion is recognized by RNase H and the target gene mRNA is degraded.
- FIG. 3 shows the structures of various cross-linked nucleic acids.
- FIG. 4 shows the structure of various natural or non-natural nucleotides.
- the first aspect of the present invention is a nucleic acid complex, more preferably a blood brain barrier-passing nucleic acid complex.
- the nucleic acid complex includes a first nucleic acid strand and a second nucleic acid strand.
- the second nucleic acid strand is a nucleotide strand containing a base sequence complementary to the first nucleic acid strand.
- the first nucleic acid strand is annealed to the second nucleic acid strand.
- the second nucleic acid strand is linked to phosphatidylethanolamine or an analog thereof.
- FIG. 1a shows a nucleic acid complex in which phosphatidylethanolamine is bound to the 5 ′ end of the second nucleic acid strand.
- FIG. 1b shows a nucleic acid complex in which phosphatidylethanolamine is bound to the 3 ′ end of the second nucleic acid strand.
- phosphatidylethanolamine or an analog thereof may be bound to both ends of the 5 ′ end and 3 ′ end of the second nucleic acid strand.
- phosphatidylethanolamine or an analog thereof may be bound to the nucleotide inside the second nucleic acid strand.
- the first nucleic acid strand is a nucleotide strand comprising a base sequence capable of hybridizing to at least a part of the target transcript. In certain embodiments, the first nucleic acid strand is a transcript of the target gene or a nucleotide strand that has an antisense effect on the target transcript.
- target transcript refers to any RNA that can be a target of the nucleic acid complex of the present invention and is synthesized by a DNA-dependent RNA polymerase. Generally, it corresponds to the transcript of the target gene. Specifically, mRNA (including mature mRNA, mRNA precursor, mRNA not subjected to base modification, etc.) transcribed from the target gene, non-coding RNA (non-coding RNA, ncRNA) such as miRNA can be included.
- the “target gene” is not particularly limited, and examples thereof include a gene derived from an organism into which the nucleic acid complex of the present invention is introduced, for example, a gene whose expression increases in various diseases.
- the target transcript is mRNA transcribed from genomic DNA encoding the target gene, and further includes mRNA not subjected to base modification, unprocessed mRNA precursor, and the like.
- the “target transcript” may include not only mRNA but also non-coding RNA (non-coding RNA, ncRNA) such as miRNA. More generally, a “transcript” can be any RNA synthesized by a DNA-dependent RNA polymerase.
- the “target transcript” is, for example, scavenger receptor B1 (scavenger receptor B1: scavenger receptor B1: often referred to herein as “SR-B1 mRNA”) or metastasis-related Non-coding RNA of lung adenocarcinoma transcript 1 (metastasis associated lung adenocarcinoma transcript 1: often referred to as “Malat1” in this specification)
- SR-B1 mRNA metastasis-related Non-coding RNA of lung adenocarcinoma transcript 1
- Malat1 metastasis-related Non-coding RNA of lung adenocarcinoma transcript 1
- the nucleotide sequence of mouse Malat1 noncoding RNA is shown in SEQ ID NO: 3. No.
- SEQ ID NO: 4 shows the base sequence of human Malat1 non-coding RNA
- SEQ ID NO: 5 shows the base sequence of mouse SR-B1 mRNA
- SEQ ID NO: 6 shows the base sequence of human SR-B1 mRNA
- SEQ ID NO: 7 Shows the base sequence of mouse DMPK mRNA
- the base sequence of human DMPK mRNA is shown in SEQ ID NO: 8.
- the base sequence of mRNA is replaced with the base sequence of DNA. .
- Nucleotide sequence information for these genes and transcripts for example available from known databases such as NCBI (National Center for Biotechnology Information) database.
- the term “antisense oligonucleotide (ASO)” or “antisense nucleic acid” includes at least a part of a target transcript, for example, a complementary base sequence that can hybridize to any target region. It refers to a single-stranded oligonucleotide capable of suppressing and controlling the expression of the target gene transcript or the level of the target transcript depending on the effect.
- the first nucleic acid strand functions as ASO, and its target region is 3′UTR, 5′UTR, exon, intron, coding region, translation initiation region, translation termination region, or any other
- the nucleic acid region may be included.
- the target region of the target transcript can be at least 8 bases long, eg, 10-35 bases long, 12-25 bases long, 13-20 bases long, 14-19 bases long, or 15-18 bases long .
- Antisense effect refers to an effect of regulating expression or editing brought about by a target transcript by hybridization of ASO with the target transcript (eg, RNA sense strand).
- Regulatory the expression or editing of a target transcript refers to the expression of a target gene or the expression level of a target transcript (in this specification, “the expression level of a target transcript” is often referred to as “the level of a target transcript”). Suppression or reduction, translational inhibition, splicing function modification effect (for example, exon skipping, etc.), or degradation of transcripts. For example, as shown in FIG.
- ASO in the inhibition of translation, when an oligonucleotide (eg, RNA) is introduced into a cell as ASO, ASO binds to mRNA or the like, which is a transcription product of the target gene, and partially doubles. A chain is formed. This partial duplex serves as a cover to prevent translation by the ribosome, thereby inhibiting the expression of the protein encoded by the target gene at the translational level (marked outside the broken line in FIG. 2).
- a partial DNA-RNA heteroduplex is formed.
- the mRNA of the target gene is degraded, and the expression of the protein encoded by the target gene is inhibited at the expression level (in the broken line in FIG. 2). This is referred to as the “RNase H-dependent pathway”.
- the antisense effect can be brought about by targeting introns of the mRNA precursor. Antisense effects may also be brought about by targeting the miRNA, in which case the function of the miRNA is inhibited and the expression of the gene for which the miRNA normally controls expression may be increased. In one embodiment, modulation of target transcript expression may be a reduction in the amount of target transcript.
- nucleic acid or “nucleic acid molecule” means a nucleoside or nucleotide for a monomer, an oligonucleotide for an oligomer, and a polynucleotide for a polymer.
- Nucleoside generally refers to a molecule composed of a combination of a base and a sugar. Although the sugar moiety of the nucleoside is not limited, it is usually composed of a pentofuranosyl sugar, and specific examples thereof include ribose and deoxyribose.
- the base part (nucleobase) of a nucleoside is usually a heterocyclic base part. Although not limited, adenine, cytosine, guanine, thymine, or uracil and other modified nucleobases (modified bases) can be mentioned.
- Nucleotide refers to a molecule in which a phosphate group is covalently bonded to the sugar moiety of the nucleoside.
- a phosphate group is usually linked to the hydroxyl group at the 2′-position, 3′-position or 5′-position of the sugar.
- Oligonucleotide refers to a linear oligomer formed by linking several to several tens of hydroxyl groups and phosphate groups of sugar moieties by covalent bonds between adjacent nucleotides.
- Polynucleotide refers to a linear polymer formed by linking several tens or more, preferably several hundred or more, of nucleotides more than oligonucleotides by the covalent bond.
- phosphate groups are generally considered to form internucleoside linkages.
- nucleic acid strand or simply “strand” means an oligonucleotide or a polynucleotide.
- the nucleic acid chain can be produced as a full-length chain or a partial chain by, for example, a chemical synthesis method using an automatic synthesizer, or by an enzymatic process using a polymerase, ligase, or restriction reaction.
- the nucleic acid strand can include natural nucleotides and / or non-natural nucleotides.
- naturally nucleoside refers to a nucleoside that exists in nature. Examples thereof include ribonucleosides composed of ribose and a base such as adenine, cytosine, guanine, or uracil, and deoxyribonucleosides composed of deoxyribose and a base such as adenine, cytosine, guanine, or thymine.
- ribonucleoside found in RNA and the deoxyribonucleoside found in DNA are sometimes referred to as “DNA nucleoside” and “RNA nucleoside”, respectively.
- a “natural nucleotide” refers to a molecule that exists in nature and has a phosphate group covalently bonded to the sugar moiety of the natural nucleoside.
- examples thereof include ribonucleotides known as RNA constituent units in which phosphate groups are bound to ribonucleosides, and deoxyribonucleotides known as DNA constituent units in which phosphate groups are bound to deoxyribonucleosides.
- unnatural nucleoside refers to any nucleoside other than natural nucleosides.
- modified nucleosides and nucleoside mimetics are included.
- modified nucleoside means a nucleoside having a modified sugar moiety and / or a modified nucleobase.
- Nucleic acid strands containing non-naturally occurring oligonucleotides often have desirable properties such as enhanced cellular uptake, enhanced affinity for nucleic acid targets, increased stability in the presence of nucleases, or increased inhibitory activity. , More preferable than the natural type.
- mimic refers to a functional group that replaces a sugar, a nucleobase, and / or an internucleoside bond. In general, mimetics are used in place of sugars or sugar-internucleoside linkage combinations, and nucleobases are maintained for hybridization to a selected target.
- a “nucleoside mimetic” is used to substitute a sugar at one or more positions of an oligomeric compound, to substitute a sugar and a base, or to substitute a bond between monomer subunits constituting the oligomeric compound. Contains the structure used for.
- “Oligomer compound” means a polymer of linked monomer subunits capable of at least hybridizing to a region of a nucleic acid molecule.
- Nucleoside mimetics include, for example, morpholino, cyclohexenyl, cyclohexyl, tetrahydropyranyl, bicyclic or tricyclic sugar mimics, eg, nucleoside mimics having non-furanose sugar units.
- bicyclic nucleoside refers to a modified nucleoside containing a bicyclic sugar moiety.
- Nucleic acids containing a bicyclic sugar moiety are commonly referred to as bridged nucleic acids (BNA).
- BNA bridged nucleic acids
- a nucleoside that includes a bicyclic sugar moiety may be referred to as a “bridged nucleoside”.
- FIG. 3 illustrates a part of the cross-linked nucleic acid.
- Bicyclic sugars may be sugars in which the 2 ′ carbon atom and the 4 ′ carbon atom are bridged by two or more atoms. Examples of bicyclic sugars are known to those skilled in the art.
- One subgroup of nucleic acids containing bicyclic sugars (BNA) is 4 ′-(CH 2 ) p —O-2 ′, 4 ′-(CH 2 ) p —CH 2 -2 ′, 4 ′-( CH 2 ) p -S-2 ', 4'-(CH 2 ) p -O CH 2 O-2 ', 4'-(CH 2 ) n -N (R 3 ) -O- (CH 2 ) m- 2 ′
- R 3 represents a hydrogen atom, an alkyl group, an alkenyl group, a cyclo Represents an alkyl group, ary
- a hydroxyl group protecting group for nucleic acid synthesis alkyl group, alkenyl group, cycloalkyl group, aryl group, aralkyl group, acyl group, sulfonyl group Group, silyl group, phosphate group, phosphate group protected by a protecting group for nucleic acid synthesis, or P (R 4 ) R 5 [wherein R 4 and R 5 may be the same as each other May be different, hydroxyl group, hydroxyl group protected by protecting group for nucleic acid synthesis, mercapto group, mercapto group protected by protecting group for nucleic acid synthesis, amino group, 1-5 Have carbon atoms Represents an alkoxy group, an alkylthio group having 1 to 5 carbon atoms, a cyanoalkoxy group having 1 to 6 carbon atoms, or an amino group substituted with an alkyl group having 1 to 5 carbon atoms] It may be
- Non-limiting examples of such BNA include methyleneoxy (4′-CH 2 -O-2 ′) BNA (LNA (Locked Nucleic Acid®, also known as 2 ′, 4′-BNA).
- LNA Locked Nucleic Acid®
- ⁇ -L-methyleneoxy (4′-CH 2 -O-2 ′) BNA or ⁇ -D-methyleneoxy (4′-CH 2 -O-2 ′) BNA ethyleneoxy (4 '-(CH 2 ) 2 -O-2') BNA (also known as ENA), ⁇ -D-thio (4'-CH 2 -S-2 ') BNA, aminooxy (4'-CH 2 -ON (R 3 ) -2 ') BNA, oxyamino (4'-CH 2 -N (R 3 ) -O-2') BNA (also known as 2 ', 4'-BNA NC ) , 2 ', 4'-BNA coc , 3'- amino -2', 4'-
- non-natural nucleotide refers to any nucleotide other than natural nucleotides, including modified nucleotides and nucleotide mimetics.
- modified nucleotide means a nucleotide having any one or more of a modified sugar moiety, a modified internucleoside linkage, and a modified nucleobase.
- nucleic acid strands comprising non-naturally occurring oligonucleotides often include, for example, enhanced cellular uptake, enhanced affinity for nucleic acid targets, increased stability in the presence of nucleases, or increased inhibitory activity, etc. It has desirable characteristics. Therefore, it is preferred over natural nucleotides.
- modified internucleoside linkage refers to an internucleoside linkage having a substitution or any change from a naturally occurring internucleoside linkage (ie, phosphodiester linkage). Modified internucleoside linkages include phosphorus-containing internucleoside linkages that contain a phosphorus atom and non-phosphorus-containing internucleoside linkages that do not contain a phosphorus atom.
- Typical phosphorus-containing internucleoside linkages include phosphodiester linkages, phosphorothioate linkages, phosphorodithioate linkages, phosphotriester linkages, alkyl phosphonate linkages, alkylthiophosphonate linkages, boranophosphate linkages, and phosphoramidate linkages.
- the phosphorothioate bond is an internucleoside bond in which the non-bridging oxygen atom of the phosphodiester bond is replaced with a sulfur atom. Methods for preparing phosphorus-containing and non-phosphorus-containing bonds are well known.
- the modified internucleoside linkage is preferably a linkage that has higher nuclease resistance than naturally occurring internucleoside linkages.
- modified nucleobase or “modified base” means any nucleobase other than adenine, cytosine, guanine, thymine, or uracil.
- modified nucleobases include 5-methylcytosine, 5-fluorocytosine, 5-bromocytosine, 5-iodocytosine, N4-methylcytosine, N6-methyladenine, 8-bromoadenine, N2-methylguanine, or 8 -Bromoguanine includes, but is not limited to A preferred modified nucleobase is 5-methylcytosine.
- Unmodified nucleobase or “unmodified base” is synonymous with natural nucleobase, and is a purine base, adenine (A) and guanine (G), and a pyrimidine base, thymine (T), cytosine (C ) And uracil (U).
- a “modified sugar” has a substitution and / or any change from a natural sugar moiety (ie, a sugar moiety found in DNA (2′-H) or RNA (2′-OH)).
- a nucleic acid strand may include one or more modified nucleosides that optionally include a modified sugar.
- Sugar-modified nucleosides may confer enhanced nuclease stability, increased binding affinity, or some other beneficial biological property to the nucleic acid strand.
- the nucleoside may contain a chemically modified ribofuranose ring moiety.
- Examples of chemically modified ribofuranose rings include, but are not limited to, bicyclic nucleic acids (bridged nucleic acids, BNAs) by the addition of substituents (including 5 ′ and 2 ′ substituents) and the formation of bridges on non-geminal ring atoms. formation of), S ribosyl ring oxygen atom, N (R), or C (R1) (R2) ( R, R1 and R2 are each, independently, H, to C 1 -C 12 alkyl and or a protective group And combinations thereof, and combinations thereof.
- BNAs bicyclic nucleic acids
- substituents including 5 ′ and 2 ′ substituents
- R1 and R2 are each, independently, H, to C 1 -C 12 alkyl and or a protective group And combinations thereof, and combinations thereof.
- nucleosides having modified sugar moieties include, but are not limited to, 5′-vinyl, 5′-methyl (R or S), 4′-S, 2′-F (2 ′ -Fluoro groups), 2′-OCH 3 (2′-OMe group or 2′-O-methyl group), and nucleosides containing 2′-O (CH 2 ) 2 OCH 3 substituents.
- “2′-modified sugar” means a furanosyl sugar modified at the 2 ′ position.
- nucleobase moiety (natural, modified, or combinations thereof) may be maintained for hybridization with an appropriate nucleic acid target.
- modification can be performed such that nucleotides in the same strand can be independently subjected to different modifications.
- the same nucleotide has a modified internucleoside linkage (e.g., a phosphorothioate linkage) and is further modified with a modified sugar (e.g., 2'-O-methyl modified sugar or bicyclic Sugar).
- the same nucleotide also has a modified nucleobase (eg, 5-methylcytosine) and can further have a modified sugar (eg, a 2′-O-methyl modified sugar or a bicyclic sugar).
- the number, type, and position of non-natural nucleotides in a nucleic acid chain can affect the antisense effect and the like provided by the nucleic acid complex of the present invention.
- the selection of modification may vary depending on the sequence of the target gene, etc., but those skilled in the art will be able to explain the literature related to the antisense method (for example, WO 2007/143315, WO 2008/043753, and WO 2008/049085).
- the preferred embodiment can be determined by reference.
- the antisense effect of the nucleic acid complex of the present invention after modification is measured, the measurement value thus obtained is significantly compared with the measurement value of the nucleic acid complex of the present invention before modification. If not low (for example, if the measurement obtained after modification is 70% or more, 80% or more or 90% or more of the measurement of the nucleic acid complex of the present invention before modification), evaluate the relevant modification. Can do.
- the term “complementary” refers to a so-called Watson-Crick base pair (natural base pair) or non-Watson-Crick base pair (Hoogsteen-type base) via hydrogen bonding. It means a relationship that can form a peer).
- the first nucleic acid strand does not necessarily have to be completely complementary to at least a part of a target transcript (for example, a transcript of a target gene), and has a base sequence of at least 70%, preferably at least 70%. It is acceptable if it has 80%, even more preferably at least 90% complementarity (eg, 95%, 96%, 97%, 98%, or 99% or more).
- the complementary region in the second nucleic acid strand does not necessarily have to be completely complementary to at least a part of the first nucleic acid strand, and has a base sequence of at least 70%, preferably at least 80%, More preferably, at least 90% (eg, 95%, 96%, 97%, 98%, or 99% or more) complementarity is acceptable.
- alkyl means a linear or branched, acyclic saturated aliphatic hydrocarbon.
- linear or branched alkyl having 1 to 32 carbon atoms includes methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, 1-ethylpropyl , Hexyl, isohexyl, 1,1-dimethylbutyl, 2,2-dimethylbutyl, 3,3-dimethylbutyl, 2-ethylbutyl, octyl, decyl, dodecyl, tridecyl, tetradecyl, 2,6,10-trimethylundecyl, Pentadecyl, 3,7,11-trimethyldodecyl, hexadecyl, heptadecy
- alkenyl means linear or branched alkenyl containing at least one double bond. Alkenyl includes both cis and trans isomers.
- straight chain or branched alkenyl having 2 to 32 carbon atoms includes ethenyl, 1-propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 3-methyl-2-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 4-methyl-3-pentenyl, 1-hexenyl, 3-hexenyl, 5-hexenyl, 8-heptadecenyl, (E ) -8-heptadecenyl, (Z) -8-heptadecenyl, heptadesa-8,11-dienyl, (8Z, 11Z) -heptadesa-8,11-dienyl, (8
- blood-brain barrier is a mechanism that selects and restricts substances that migrate to the brain, as described above, and plays a role in protecting the brain from harmful substances. .
- central nervous system is a tissue that consists of the brain and spinal cord and constitutes the nervous system together with the peripheral nervous system.
- the brain includes the cerebrum (cerebral cortex, cerebral white matter, basal ganglia), diencephalon (thalamus, hypothalamic nucleus), cerebellum (cerebellar cortex, cerebellar nucleus) and brain stem (midbrain, substantia nigra, pons, medulla oblongata).
- the spinal cord includes the cervical spinal cord, thoracic spinal cord, lumbar spinal cord, sacral spinal cord and tail spinal cord.
- the central nervous system in the present specification may be any of these areas, but the cerebral cortex (frontal lobe, temporal lobe, parietal lobe, occipital lobe), cerebellum, striatum, pallidum, palsy, The hippocampus, parahippocampal, brain stem, cervical spinal cord, thoracic spinal cord or lumbar spinal cord are preferred.
- cerebral cortex frontal lobe, temporal lobe, parietal lobe, occipital lobe
- cerebellum cerebellum
- striatum pallidum
- palsy palsy
- the hippocampus, parahippocampal, brain stem, cervical spinal cord, thoracic spinal cord or lumbar spinal cord are preferred.
- the “salt thereof” is a salt of the nucleic acid complex of the present invention, and is a physiologically and pharmaceutically acceptable salt of the nucleic acid complex of the present invention, that is, a desired salt of the nucleic acid complex.
- salts include alkali metal salts such as sodium salt, potassium salt and lithium salt; alkaline earth metal salts such as calcium salt and magnesium salt; aluminum salt, iron salt, zinc salt, copper salt, Metal salts such as nickel salts and cobalt salts; inorganic salts such as ammonium salts; t-octylamine salts, dibenzylamine salts, morpholine salts, glucosamine salts, phenylglycine alkyl ester salts, ethylenediamine salts, guanidine salts, diethylamine salts, Triethylamine salt, dicyclohexylamine salt, N, N'-dibenzylethylenediamine salt, chloroprocaine salt, procaine salt, diethanolamine salt, N-benzyl-phenethylamine salt, piperazine salt, tetramethylammonium salt, tris (hydroxymethyl) aminomethane salt , Diora Amine salts such as thiol
- the nucleic acid complexes of the invention include any pharmaceutically acceptable salt of the nucleic acid complex, an ester of the nucleic acid complex, or a salt of the ester.
- suitable pharmaceutically acceptable salts include, but are not limited to, sodium, potassium and meglumine salts.
- the first nucleic acid strand is a single-stranded oligonucleotide strand that contains a base sequence capable of hybridizing to at least a part of the target transcript and provides an antisense effect to the target transcript.
- the second nucleic acid strand is a single-stranded oligonucleotide strand containing a base sequence complementary to the first nucleic acid strand.
- the second nucleic acid strand is bound to phosphatidylethanolamine or an analog thereof.
- the second nucleic acid strand is annealed to the first nucleic acid strand through complementary base pair hydrogen bonding.
- the base length of the first and second nucleic acid strands is usually at least 8 bases long, at least 9 bases long, at least 10 bases long, at least 11 bases long, at least 12 bases long, at least 13 bases long, at least 14 bases long Or at least 15 bases in length, but not particularly limited.
- the base lengths of the first and second nucleic acid strands are 35 bases or less, 30 bases or less, 25 bases or less, 24 bases or less, 23 bases or less, 22 bases or less, 21 bases or less 20 base length or less, 19 base length or less, 18 base length or less, 17 base length or less, or 16 base length or less.
- the first nucleic acid strand and the second nucleic acid strand may be about 100 bases in length, or 10 to 35 bases, 12 to 25 bases, 13 to 20 bases, 14 to 19 bases, or 15 It may be up to 18 bases long.
- the first nucleic acid strand and the second nucleic acid strand may be the same length or different lengths (for example, either one is 1 to 3 bases shorter or longer).
- the double-stranded structure formed by the first nucleic acid strand and the second nucleic acid strand may contain a bulge.
- the choice of length can be determined by the balance between the strength of the antisense effect and the specificity of the nucleic acid strand for the target, among other factors such as cost, synthesis yield, and the like.
- the internucleoside linkage in the first nucleic acid strand and the second nucleic acid strand may be a naturally occurring internucleoside linkage and / or a modified internucleoside linkage.
- at least one, at least two, or at least three internucleoside linkages from the end (5 ′ end, 3 ′ end or both ends) of the first nucleic acid strand and / or the second nucleic acid strand are between the modified nucleosides.
- a bond is preferred.
- two internucleoside bonds from the end of the nucleic acid chain mean an internucleoside bond that is closest to the end of the nucleic acid chain and an internucleoside bond that is adjacent to and opposite to the end.
- Modified internucleoside linkages in the terminal region of the nucleic acid strand are preferred because they can suppress or inhibit undesired degradation of the nucleic acid strand.
- all the internucleoside linkages of the first nucleic acid strand and / or the second nucleic acid strand may be modified internucleoside linkages.
- the modified internucleoside linkage may be a phosphorothioate linkage.
- the at least one (for example, 3) internucleoside bond from the 3 ′ end of the second nucleic acid strand may be a modified internucleoside bond such as a phosphorothioate bond having high RNase resistance. It is preferable to include a modified internucleoside bond such as phosphorothioate modification at the 3 ′ end of the second nucleic acid strand because the gene suppression activity of the double-stranded nucleic acid complex is improved.
- the internucleoside linkage of 2 to 6 bases at the phosphatidylethanolamine unbound end may be a modified internucleoside linkage (eg, phosphorothioate linkage).
- the at least one (for example, 3) nucleoside from the 3 ′ end of the second nucleic acid strand may be, for example, a modified nucleoside such as 2′F-RNA or 2′-OMe having high RNase resistance. Including a modified nucleoside such as 2′F-RNA or 2′-OMe at the 3 ′ end of the second nucleic acid strand is preferable because the gene suppression activity of the double-stranded nucleic acid complex is enhanced.
- nucleosides at the phosphatidylethanolamine-unbound end may be modified nucleosides such as 2′F-RNA having high RNase resistance, for example.
- the nucleoside in the first nucleic acid strand and the second nucleic acid strand may be a natural nucleoside (deoxyribonucleoside, ribonucleoside, or both) and / or a non-natural nucleoside.
- the base sequence of the first nucleic acid strand is complementary to at least a part of the base sequence of the target transcript, it can hybridize (or anneal) to the target transcript.
- Complementarity of base sequences can be determined by using a BLAST program or the like.
- a person skilled in the art can easily determine the conditions (temperature, salt concentration, etc.) under which two strands can hybridize in consideration of the degree of complementarity between strands.
- those skilled in the art can easily design an antisense nucleic acid complementary to the target transcript based on, for example, information on the base sequence of the target gene.
- Hybridization conditions may be various stringent conditions such as low stringency conditions and high stringency conditions.
- Low stringent conditions may be relatively low temperature and high salt conditions, eg, 30 ° C., 2 ⁇ SSC, 0.1% SDS.
- High stringency conditions may be relatively high temperature and low salt conditions, eg, 65 ° C., 0.1 ⁇ SSC, 0.1% SDS.
- 1 ⁇ SSC contains 150 mM sodium chloride and 15 mM sodium citrate.
- the first nucleic acid strand can comprise at least 4, at least 5, at least 6, or at least 7 consecutive nucleosides that are recognized by RNase H when hybridized to the target transcript. Usually, it may be a region containing a continuous nucleoside of 4 to 20 bases, 5 to 16 bases, or 6 to 12 bases.
- a nucleoside recognized by RNase H for example, natural deoxyribonucleoside can be used. Modified deoxyribonucleosides and suitable nucleosides containing other bases are well known in the art. It is also known that a nucleoside having a hydroxy group at the 2 ′ position, such as a ribonucleoside, is inappropriate as the nucleoside. The suitability of a nucleoside can be readily determined for use in this region containing “at least 4 consecutive nucleosides”. In one embodiment, the first nucleic acid strand can comprise at least 4 consecutive deoxyribonucleosides.
- the full length of the first nucleic acid strand is not composed only of natural ribonucleosides. It is preferred that the natural ribonucleoside of the first nucleic acid strand is not more than half of the full length or does not contain it.
- the second nucleic acid strand may comprise at least 4 consecutive ribonucleosides that are complementary to at least 4 consecutive nucleosides (eg, deoxyribonucleosides) in the first nucleic acid strand. .
- the second nucleic acid strand forms a partial DNA-RNA heteroduplex with the first nucleic acid strand, and is recognized and cleaved by RNaseH.
- At least four consecutive ribonucleosides in the second nucleic acid strand are preferably linked by naturally occurring internucleoside linkages, ie phosphodiester linkages.
- all nucleosides may be composed of ribonucleosides and / or modified nucleosides. All nucleosides of the second nucleic acid strand may be composed of deoxyribonucleosides and / or modified nucleosides, and may not include ribonucleosides.
- the first nucleic acid strand and / or the second nucleic acid strand constituting the nucleic acid complex of the present invention may be a gapmer.
- gapmer refers to a single-stranded nucleic acid consisting of a central region (DNA gap region) and a 5 ′ wing region and a 3 ′ wing region arranged on both sides of the 5 ′ end and 3 ′ end.
- the central region in the gapmer contains at least 4 consecutive deoxyribonucleosides, and the 5 ′ wing region and 3 ′ wing region contain unnatural nucleosides.
- the gapmer is specifically referred to as a “BNA / DNA gapmer”.
- the number of bridged nucleosides contained in the 5 ′ wing region and the 3 ′ wing region may be 2 or 3.
- the bridged nucleosides contained in the 5 ′ wing region and the 3 ′ wing region may exist continuously or discontinuously in the 5 ′ wing region and the 3 ′ wing region.
- the bridged nucleoside can further comprise a modified nucleobase (eg, 5-methylcytosine).
- the gapmer When the bridged nucleoside is an LNA nucleoside, the gapmer is referred to as “LNA / DNA gapmer”.
- the gapmer When the non-natural nucleoside constituting the 5 ′ wing region and the 3 ′ wing region comprises or consists of a peptide nucleic acid, the gapmer is specifically referred to as a “peptide nucleic acid gapmer”.
- the gapmer When the non-natural nucleoside constituting the 5 ′ wing region and the 3 ′ wing region comprises or consists of a peptide nucleic acid, the gapmer is particularly referred to as a “morpholino nucleic acid gapmer”.
- the base lengths of the 5 ′ wing region and the 3 ′ wing region may be independently at least 2 bases, for example, 2 to 10 bases, 2 to 7 bases, or 3 to 5 bases.
- the 5 ′ wing region and the 3 ′ wing region may contain at least one non-natural nucleoside, and may further contain a natural nucleoside.
- the first nucleic acid strand and / or the second nucleic acid strand constituting the gapmer is a bridged nucleoside having a length of 2 to 7 bases or 3 to 5 bases, 4 to 15 bases or 8 to 12 bases in order from the 5 ′ end. It may be composed of a long ribonucleoside or deoxyribonucleoside and a bridged nucleoside having a length of 2 to 7 bases or 3 to 5 bases.
- the first nucleic acid strand and / or the second nucleic acid strand constituting the nucleic acid complex of the present invention may be a mixmer.
- “mixmer” refers to a nucleic acid chain that includes alternating natural nucleosides and non-natural nucleosides of periodic or random segment length, and that does not include four or more consecutive deoxyribonucleosides and ribonucleosides. .
- the mixmer in which the non-natural nucleoside is a bridged nucleoside and the natural nucleoside is a deoxyribonucleoside is particularly referred to as a “BNA / DNA mixmer”.
- the mixmer in which the non-natural nucleoside is a peptide nucleic acid and the natural nucleoside is a deoxyribonucleoside is particularly referred to as a “peptide nucleic acid / DNA mixmer”.
- the mixmer in which the non-natural nucleoside is a morpholino nucleic acid and the natural nucleoside is a deoxyribonucleoside is particularly referred to as a “morpholino nucleic acid / DNA mixmer”.
- a mixmer is not limited to containing only two nucleosides.
- a mixmer can contain any number of types of nucleosides, whether or not they are natural or modified nucleosides or nucleoside mimetics.
- the bridged nucleoside may further comprise a modified nucleobase (eg, 5-methylcytosine).
- the modified nucleoside may be at least 1, at least 2, at least 3, or at least 4 nucleosides from the end (5 ′ end, 3 ′ end, or both ends) of the second nucleic acid strand.
- the modified nucleoside may contain a modified sugar and / or a modified nucleobase.
- the modified sugar may be a 2′-modified sugar (eg, a sugar containing a 2′-O-methyl group).
- the modified nucleobase can also be 5-methylcytosine.
- the second nucleic acid strand is, in order from the 5 ′ end, a modified nucleoside having a length of 2 to 7 bases or 3 to 5 bases (for example, a modified nucleoside containing a 2′-modified sugar), 4 to 15 bases or 8 to 12 bases From long (optionally linked by modified internucleoside linkages) ribonucleosides or deoxyribonucleosides and modified nucleosides of 2-7 bases or 3-5 bases in length (eg, modified nucleosides containing 2′-modified sugars) It may be configured.
- the first nucleic acid strand may be a gapmer.
- the first nucleic acid strand and the second nucleic acid strand may include a nucleoside mimic or nucleotide mimic in whole or in part.
- Nucleotide mimetics may be peptide nucleic acids and / or morpholino nucleic acids.
- the first nucleic acid strand may include at least one modified nucleoside.
- the modified nucleoside may contain a 2′-modified sugar.
- the 2′-modified sugar may be a sugar containing a 2′-O-methyl group.
- the first nucleic acid strand and the second nucleic acid strand may contain any combination of the above-described modified internucleoside bond and the modified nucleoside.
- the second nucleic acid strand is bound to phosphatidylethanolamine (PhosphatidylEthanolamine: often referred to herein as “PE”) or an analog thereof.
- PhosphatidylEthanolamine often referred to herein as “PE”
- Phosphatidylethanolamine is a neutral phospholipid having a structure in which ethanolamine is ester-bonded to the phosphate group of phosphatidic acid.
- the phosphatidic acid is a glycerophospholipid in which various carboxyl groups (fatty acids) are ester-bonded to the hydroxyl groups at the 1-position and 2-position of glycerol and phosphoric acid is ester-bonded to the hydroxyl group at the 3-position.
- PE also called cephalin, is an important component of biological cell membranes and functions in cell membrane division, fusion, maintenance, and membrane protein stabilization.
- PE group A group derived from PE or an analog thereof bound to the second nucleic acid strand (sometimes abbreviated as “PE group” in the present specification) is represented by the following general formula I.
- R 1 and R 2 each independently represent an alkyl group (examples of C 5 ⁇ C 32, preferably C 15 ⁇ 19 that are not been substituted or unsubstituted, pentadecyl, heptadecyl, 2, 6, 10, 14-tetramethylpentadecyl), more preferably a C 17 alkyl group (eg, heptadecyl) or a substituted or unsubstituted C 5 -C 32, preferably C 17 alkenyl group (eg, heptadecenyl (eg, Represents 8-heptadecenyl (eg, (E) -8-heptadecenyl, (Z) -8-heptadecenyl), heptadec-8,11-dienyl (eg, (8E, 11E) -heptadesa-8,11-dienyl)) .
- alkyl group examples of C 5 ⁇ C 32
- a group derived from PE or an analog thereof that binds to the second nucleic acid strand is represented by the following general formulas XV-XXII.
- analog refers to a compound having a similar structure and properties having the same or similar basic skeleton. Analogs include, for example, biosynthetic intermediates, metabolites, and the like. A person skilled in the art can determine whether a compound is an analog of another compound. PE or an analog thereof can be produced by a method known per se by those skilled in the art.
- the second nucleic acid strand contains one or more PE or its analogs.
- PE or its analog is linked to the 5 ′ end, 3 ′ end or the nucleotide inside the second nucleic acid strand of the second nucleic acid strand.
- it is the 5 ′ end or the 3 ′ end.
- the second nucleic acid strand includes two or more PE and / or analogs thereof, they may be linked to a plurality of positions of the second nucleic acid strand and / or one position of the second nucleic acid strand. May be connected as a group. It is preferable that one is connected to each of the 5 ′ end and the 3 ′ end of the second nucleic acid strand.
- the bond between the second nucleic acid strand and PE or an analog thereof may be a direct bond or an indirect bond.
- Direct binding refers to the direct binding of two molecules.
- Indirect binding refers to the binding of two molecules to be bound via another substance.
- the binding between the second nucleic acid strand and PE and / or its analogs is a phosphate ester bond to the 5 ′ end, 3 ′ end of the second nucleic acid strand, or a nucleotide inside the second nucleic acid strand.
- they may be bonded via a phosphorothioate bond.
- the second nucleic acid strand is bound to the 5 ′ end via a phosphate ester bond or a phosphorothioate bond.
- the second nucleic acid strand is bound to the 5 ′ end via a phosphate ester bond.
- the second nucleic acid strand and PE or an analog thereof may be bound via a linking group (often referred to as “linker” in the present specification).
- the linker may be bound to the 5 ′ end, 3 ′ end of the second nucleic acid strand, or the nucleotide inside the second nucleic acid strand via a phosphate ester bond or a phosphorothioate bond.
- the linker is bound to the 5 ′ end of the second nucleic acid strand via a phosphate ester bond or a phosphorothioate bond.
- the linker is bound to the 5 ′ end of the second nucleic acid strand via a phosphate ester bond.
- linker examples include a linker represented by the following general formula II.
- n 0 or 1.
- cleavable linker refers to a linking group that can be cleaved under physiological conditions, eg, in a cell or animal body (eg, the human body).
- the cleavable linker may be selectively cleaved by endogenous enzymes such as nuclease and peptidase, acidic conditions, reducing environment, and the like.
- Examples of such include, for example, amide bonds, ester bonds, phosphate ester bonds, ester bonds of one or both of phosphodiester bonds, carbamate bonds, and disulfide bonds, and nucleotide linkers such as natural DNA linkers.
- Non-cleavable linker refers to a linking group that is not cleaved under physiological conditions. Examples of such a non-cleavable linker include a phosphorothioate bond and a linker composed of a modified or unmodified deoxyribonucleoside linked by a phosphorothioate bond, or a modified or unmodified ribonucleoside.
- the PE group bound to the linker is represented by the following general formula IV.
- R 1 and R 2 are the same as R 1 and R 2 in the general formula I, and n is the same meaning as n in the general formula II.
- the bond between the second nucleic acid strand and the group represented by the general formula IV is bound to the 5 ′ end, 3 ′ end of the second nucleic acid strand, or a nucleotide inside the second nucleic acid strand via a phosphate ester bond or a phosphorothioate bond. You may do it.
- the group represented by the general formula IV is bonded to the 5 ′ end of the second nucleic acid strand via a phosphate ester bond or a phosphorothioate bond.
- the group represented by the general formula IV is bonded to the 5 ′ end of the second nucleic acid strand via a phosphate bond.
- the PE group bound to the linker is represented by the following general formula III.
- 5 ′ oligo indicates the 5 ′ end of the oligonucleotide to be bound.
- R 1 and R 2 have the same meanings as R 1 and R 2 in the general formula I, and n is the same meaning as n in the general formula II.
- the chain length of the cleavable linker or non-cleavable linker is not particularly limited in the case of nucleic acids such as DNA or oligonucleotides, but is usually 1 to 20 bases, 1 to 10 bases or 1 to 6 bases long. Good.
- nucleic acid complex of the present invention when the nucleic acid complex of the present invention contains an optical isomer, a stereoisomer, a positional isomer, or a rotational isomer, these are also contained as the nucleic acid complex of the present invention.
- Each can be obtained as a single product by a synthesis method and a separation method known per se.
- the optical isomer resolved from the compound is also included in the nucleic acid complex of the present invention.
- the nucleic acid complex of the invention comprises a prodrug and a pharmaceutically acceptable salt of the prodrug.
- the prodrug of the nucleic acid complex of the present invention and the pharmaceutically acceptable salt of the prodrug is a compound that is converted into the nucleic acid complex of the present invention by a reaction with an enzyme, gastric acid or the like under physiological conditions in vivo, that is, the enzyme
- it refers to a compound that changes to the nucleic acid complex of the present invention by oxidation, reduction, hydrolysis, etc., or a compound that changes to the nucleic acid complex of the present invention by hydrolysis due to gastric acid or the like.
- a prodrug of a nucleic acid complex of the invention comprises one or more PEs or analogs thereof bound to a first nucleic acid strand or a second nucleic acid strand.
- the nucleic acid complex of the present invention binds PE or its analog to the end of the second nucleic acid strand of the double-stranded nucleic acid complex, the double-stranded nucleic acid complex passes through the blood-brain barrier (BBB), It has not been known in the art that the delivery efficiency to the central nervous system such as the brain is increased. The present invention is based on this unexpected finding.
- BBB blood-brain barrier
- the antisense effect on the target transcription product of the first nucleic acid strand can be measured by a method known in the art.
- a method known in the art such as Northern blotting, quantitative PCR, or Western blotting.
- the expression level of the target gene in the cell or the level of the target transcript is reduced by the antisense effect as described above. What is necessary is just to verify using a well-known technique.
- Measurement of the antisense effect in the central nervous system of the nucleic acid complex of the present invention and determination of passage through the blood brain barrier can also be measured by methods known in the art.
- the nucleic acid complex of the present invention is administered to a subject (eg, a mouse), and the expression of the target gene in the central nervous system after several days to several months (for example, 2 to 7 days or 1 month later)
- the determination can be made by measuring whether the amount or level of the target transcript is suppressed.
- the criterion for the determination is that the expression level of the target gene or the measured value of the target transcript is at least 5%, at least 10%, at least 15%, at least 20% compared to the measured value of the negative control (for example, vehicle administration).
- the nucleic acid complex of the present invention has crossed the blood-brain barrier and brought an antisense effect to the central nervous system.
- the determination of passage through the blood brain barrier is carried out by administering the nucleic acid complex of the present invention to a subject (eg, a mouse), and after several days to several months (for example, 2 to 7 days or 1 month later), the central nervous system This can be determined by measuring the abundance (concentration) of the nucleic acid complex of the present invention.
- each nucleic acid molecule is designed based on the base sequence information of the target transcript (for example, the base sequence of the target gene).
- the base sequence information of the target transcript for example, the base sequence of the target gene.
- GE Healthcare, Thermo ⁇ Fisher Scientific, Beckman Coulter, etc. Can be produced by synthesizing a nucleic acid using a commercially available automatic nucleic acid synthesizer, and then purifying the obtained oligonucleotide using a reverse phase column or the like.
- the nucleic acid complex of the present invention to which a functional moiety is bound can be produced by performing the above synthesis, purification, and annealing using a nucleic acid species to which a functional moiety has been bound in advance. it can.
- the second nucleic acid strand can be produced by performing the above synthesis and purification using a nucleic acid species to which PE or an analog thereof has been previously bound.
- PE or an analog thereof can be bound to the second nucleic acid strand produced by carrying out the synthesis and purification described above by a method known per se.
- Methods for linking functional moieties to nucleic acids are well known in the art. Nucleic acids produced in this way are mixed in a suitable buffer solution and denatured at about 90 ° C. to 98 ° C. for several minutes (eg 5 minutes), after which the nucleic acids are annealed at about 30 ° C. to 70 ° C. for about 1 to 8 hours.
- one of the nucleic acid complexes of the present invention can be produced.
- the nucleic acid chain can be ordered and obtained from various manufacturers (for example, Gene Design Co., Ltd.) by designating the base sequence and modification site and type. You can also
- the annealing step can be performed by standing at room temperature (about 10 ° C. to about 35 ° C.) for about 5 to 60 minutes.
- the first nucleic acid strand and the second nucleic acid strand are each dissolved in a buffer solution (eg, phosphate buffered saline) or water at about 70 ° C. to 98 ° C., and the obtained two solutions are mixed. In some embodiments, hold at 70 ° C. to 98 ° C. for several minutes (eg, 5 minutes) and then hold the mixture at about 30 ° C. to 70 ° C. (or 30 ° C. to 50 ° C.) for about 1 to 8 hours.
- the nucleic acid complex of the present invention may be prepared.
- Each of the first nucleic acid strand and the second nucleic acid strand can also be dissolved in a buffer (eg, phosphate buffered saline) or water at room temperature (about 10 ° C. to about 35 ° C.).
- the annealing conditions (time and temperature) in producing the nucleic acid complex of the present invention are not limited to the above conditions. Also suitable conditions for promoting nucleic acid strand annealing are well known in the art.
- Embodiment 1 A nucleic acid complex comprising a first nucleic acid strand and a second nucleic acid strand or a salt thereof,
- the first nucleic acid strand includes a base sequence capable of hybridizing to at least a part of the target transcript, and has an antisense effect on the target transcript;
- the second nucleic acid strand includes a base sequence complementary to the first nucleic acid strand, and is bound to phosphatidylethanolamine or an analog thereof;
- Embodiment 2 A nucleic acid complex comprising a first nucleic acid strand and a second nucleic acid strand or a salt thereof,
- the first nucleic acid strand includes (1) a base sequence having a length of 13 to 20 bases, (2) can hybridize to at least part of the target transcript; (3) comprises at least 4 consecutive deoxyribonucleosides; and (4) has an antisense effect on the target transcript,
- the second nucleic acid strand comprises (1) at least four consecutive ribonucleosides complementary to at least four consecutive deoxyribonucleosides in the first nucleic acid strand; and (2) It is bound to phosphatidylethanolamine or an analog thereof represented by the general formula I (preferably, the general formulas XV to XXII), The nucleic acid complex or a salt thereof, wherein the first nucleic acid strand is annealed to the second nucleic acid strand.
- Embodiment 3 A nucleic acid complex comprising a first nucleic acid strand and a second nucleic acid strand or a salt thereof,
- the first nucleic acid strand comprises (1) a nucleoside and optionally a non-natural nucleoside, and the total number of the nucleoside and the optionally included non-natural nucleoside in the nucleic acid strand is 13 to 20, (2) can hybridize to at least part of the target transcript; (3) comprising at least four consecutive deoxyribonucleosides recognized by RNaseH; (4) an unnatural nucleoside that is one or more sugar-modified nucleotides located 5 ′ and / or 3 ′ of the four consecutive nucleosides, and (5) has an antisense effect on the target transcript
- the second nucleic acid strand comprises (1) at least 4 consecutive ribonucleosides complementary to at least 4 consecutive deoxyribonucleosides in the first nucleic acid strand; (2) comprising one or more unnatural nucle
- Embodiment 4 A nucleic acid complex comprising a first nucleic acid strand and a second nucleic acid strand or a salt thereof,
- the first nucleic acid strand comprises (1) a nucleoside and optionally a non-natural nucleoside, and the total number of the nucleoside and the optionally included non-natural nucleoside in the nucleic acid strand is 13 to 20, (2) can hybridize to at least part of the target transcript; (3) comprising at least four consecutive deoxyribonucleosides recognized by RNaseH; (4) an unnatural nucleoside that is one or more sugar-modified nucleotides located 5 ′ and / or 3 ′ of the four consecutive nucleosides, and (5) has an antisense effect on the target transcript
- the second nucleic acid strand comprises (1) at least 4 consecutive ribonucleosides complementary to at least 4 consecutive deoxyribonucleosides in the first nucleic acid strand; (2) comprising one or more unnatural nucle
- the nucleic acid complex of the present invention can inhibit the effect of the target miRNA in the central nervous system of the subject.
- the first nucleic acid strand includes a base sequence capable of hybridizing to at least a part of the target miRNA, and has an antisense effect on the target miRNA
- the second nucleic acid includes a nucleic acid complex that includes a base sequence complementary to the first nucleic acid strand and is bonded to PE or an analog thereof and anneals to the first nucleic acid strand and the second nucleic acid strand. It is done.
- the nucleic acid complex of the present invention can regulate the expression or editing of target RNA in the central nervous system of a subject.
- the first nucleic acid strand includes a base sequence capable of hybridizing to at least a part of the target RNA, and has an antisense effect on the target RNA
- the second nucleic acid strand Includes a nucleic acid complex comprising a base sequence complementary to the first nucleic acid strand and bound to PE or an analog thereof, wherein the first nucleic acid strand and the second nucleic acid strand are annealed to each other.
- “regulation of target RNA expression” includes, for example, up-regulation and down-regulation of the expression level.
- “Target RNA editing control” includes splicing control by RNA editing, such as exon skipping and exon inclusion.
- the target RNA may be viral or bacterial RNA, or toxic RNA (Toxic RNA).
- the nucleic acid complex of the present invention can inhibit translation of a target mRNA in the central nervous system of a subject.
- the first nucleic acid strand includes a base sequence capable of hybridizing to at least a part of the target mRNA, and has an antisense effect on the target mRNA
- the second nucleic acid strand Includes a nucleic acid complex comprising a base sequence complementary to the first nucleic acid strand and bound to PE or an analog thereof, wherein the first nucleic acid strand and the second nucleic acid strand are annealed with each other. .
- a steric block is generated and translation of the mRNA is inhibited.
- the second aspect of the present invention is a composition.
- the composition of the present invention includes the nucleic acid complex of the present invention of the first aspect as an active ingredient and / or as a drug delivery molecule.
- the nucleic acid complex of the present invention of the first aspect can pass through the BBB and regulate the expression level of the target transcript by the antisense effect (for example, decrease the expression level) in the central nervous system. Therefore, the composition of the present invention may be a composition for delivering the nucleic acid complex of the present invention and treating the subject by being administered to the subject, or may be a pharmaceutical composition.
- the present invention relates to a treatment method for treating each central nervous system disease by administering a composition containing the nucleic acid complex of the present invention.
- the composition can be formulated by a method known per se.
- the composition can be used in capsules, tablets, pills, liquids, powders, granules, fine granules, film coatings, pellets, troches, sublinguals, peptizers, buccals, pastes , Syrup, suspension, elixir, emulsion, coating, ointment, plaster, cataplasm, transdermal agent, lotion, inhalant, aerosol, eye drop, injection and seat It can be used orally or parenterally in the form of an agent.
- a pharmaceutically acceptable carrier or solvent or a carrier or solvent acceptable as food and beverage can be appropriately incorporated.
- a carrier or solvent specifically, sterilized water, physiological saline, vegetable oil, base, emulsifier, suspending agent, surfactant, pH adjusting agent, stabilizer, flavoring agent, flavoring agent, Excipients, vehicles, preservatives, binders, diluents, tonicity agents, sedatives, bulking agents, disintegrating agents, buffering agents, coating agents, lubricants, colorants, sweeteners, thickeners, taste-masking Agents, solubilizers, and other additives.
- Dosage form / dosage As used herein, there is no particular limitation on the preferred dosage form of the composition. For example, it may be oral administration or parenteral administration. Specific examples of parenteral administration include intravenous administration, intraarterial administration, intraperitoneal administration, subcutaneous administration, intradermal administration, tracheal / bronchial administration, rectal administration, intramuscular administration, and administration by blood transfusion. It can also be administered by intramuscular injection, intravenous infusion, or implantable continuous subcutaneous administration. Subcutaneous administration is preferred because self-injection by the patient himself is possible.
- the amount of the nucleic acid complex of the present invention contained in one dose of the composition is, for example, 0.001 mg / kg or more, 0.005 mg / kg or more, 0.01 mg / kg or more, 00.25 mg / kg or more, 0.5 mg / kg or more, 1 mg / kg or more, 2.5 mg / kg or more, 5 mg / kg or more, 10 mg / kg or more, 20 mg / kg or more, 30 mg / kg or more, 40 mg / kg or more, 50 mg / kg or more, 75 mg / kg or more, 100 mg / kg or more, 150 mg / kg or more, 200 mg / kg or more, 300 mg / kg or more, 400 mg / kg or more, or 500 mg / kg or more It can be.
- any amount within the range of 0.001 mg / kg to 500 mg / kg (eg, 0.001 mg / kg, 0.01 mg / kg, 0.1 mg / kg, 1 mg / kg, 5 mg / kg, 10 mg / kg, 50 mg / kg kg, 100 mg / kg, or 200 mg / kg) can be appropriately selected.
- subject refers to a subject to which the composition of the present invention is applied.
- the subject includes organs, tissues, and cells in addition to individuals.
- the composition of the present invention can be applied to any animal including humans.
- subjects other than humans for example, various livestock, poultry, pets, laboratory animals, and the like can be applied.
- the subject may be an individual who needs to reduce the expression level of the target transcript in the central nervous system, or an individual who needs to treat a central nervous system disease.
- composition of the present invention can reduce the expression level of the target transcript in the central nervous system by the BBB passing action and the antisense effect of the nucleic acid complex of the first aspect of the invention.
- the target disease is related to a central nervous system disease associated with an increase or decrease in gene expression, particularly an increase in expression of a target transcript or target gene.
- Diseases include, but are not limited to, brain tumors, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, Huntington's disease and the like.
- the delivery site of the composition of the present invention is not particularly limited, but it is more effective by being delivered to an appropriate site according to each disease. Results can be obtained.
- FDD frontotemporal dementia
- FTLD frontotemporal lobar degeneration
- SD semantic dementia
- PNFA progressive non-fluent aphasia
- Pick's disease Drug delivery to the frontal lobe, temporal lobe and / or substantia nigra can be effective.
- Parkinson's disease dementia drug delivery to the occipital lobe, substantia nigra and / or striatum may be effective. Also, in the treatment of Parkinson's disease, drug delivery to the substantia nigra and / or striatum may be effective.
- CBD corticobasal degeneration
- PSP progressive supranuclear palsy
- drug delivery to the frontal lobe, basal ganglia and / or substantia nigra can be effective.
- SCD spinocerebellar degeneration
- DPLA dentate nucleus red nucleus pallidal leucodegeneration
- SBMA bulbar spinal atrophy
- FFA Friedreich ataxia
- drug delivery to the brainstem and / or cerebellum can be effective.
- drug delivery to the striatum, frontal lobe, parietal lobe and / or basal ganglia can be effective.
- prion diseases including mad cow disease, GSS
- drug delivery to the cerebral cortex, cerebral white matter, basal ganglia and / or substantia nigra can be effective.
- cerebral white matter encephalopathy drug delivery to the cerebral white matter can be effective.
- encephalitis including viral, bacterial, fungal, tuberculosis
- meningitis including viral, bacterial, fungal, tuberculosis
- drug delivery to the entire brain can be effective.
- metabolic encephalopathy toxic encephalopathy, dystrophic encephalopathy
- drug delivery to the entire brain can be effective.
- cerebral white matter encephalopathy drug delivery to the cerebral white matter can be effective.
- cerebral infarction cerebral hemorrhage, subarachnoid hemorrhage, moyamoya disease, anoxic encephalopathy
- drug delivery to the entire brain can be effective.
- cerebral white matter encephalopathy drug delivery to the cerebral white matter can be effective.
- axon injury In the treatment of diffuse axon injury (Diffuse axonal injury), drug delivery to the cerebral white matter can be effective. In the treatment of head trauma, drug delivery to the entire brain can be effective. In the treatment of multiple sclerosis (MS), optic neuromyelitis (NMO), drug delivery to the cerebral white matter, cerebral cortex, optic nerve and / or spinal cord can be effective. In the treatment of myotonic dystrophy (DM1, DM2), drug delivery to skeletal muscle, myocardium, cerebral cortex and / or cerebral white matter can be effective. For the treatment of familial spastic paraplegia (HSP), drug delivery to the parietal lobe and / or spinal cord can be effective.
- MS multiple sclerosis
- NMO optic neuromyelitis
- DM1, DM2 myotonic dystrophy
- HSP familial spastic paraplegia
- the dosage or ingestion depends on the subject's age (including age and age), weight, symptoms and health status, type of composition (pharmaceutical, food and beverage) Etc.) and so on.
- the effective ingestion amount of the composition of the present invention to the subject is, for example, 0.00001 mg / kg / day to 10000 mg / kg / day, or 0.001 mg / kg / day to 100 mg / kg of the nucleic acid complex of the present invention to be included. / Day.
- the composition may be a single dose or multiple doses.
- One dose of the nucleic acid complex of the present invention is, for example, 0.001 mg / kg or more, 0.005 mg / kg or more, 0.01 mg / kg or more, 00.25 mg / kg or more, 0.5 mg / kg or more, 1 mg / kg kg or more, 2.5 mg / kg or more, 0.5 mg / kg or more, 1.0 mg / kg or more, 2.0 mg / kg or more, 3.0 mg / kg or more, 4.0 mg / kg or more, 5 mg / kg or more, 10 mg / kg or more, 20 mg / kg or more, 30 mg / kg or more, 40 mg / kg or more, 50 mg / kg or more, 75 mg / kg or more, 100 mg / kg or more, 150 mg / kg or more, 200 mg
- the nucleic acid complex of the present invention may be administered four times at a frequency of 0.01 to 10 mg / kg (for example, about 6.25 mg / kg) twice a week. Further, the nucleic acid complex of the present invention is administered at a dose of 0.05 to 30 mg / kg (for example, about 25 mg / kg) 2 to 4 times a week, 2 times a week, for example, twice a week. Also good.
- a dose for example, about 25 mg / kg 2 to 4 times a week, 2 times a week, for example, twice a week. Also good.
- the BBB passage amount and BNB passage amount by single administration of the nucleic acid complex of the present invention are limited (upper limit), but it is considered that the inhibitory effect works additively even in repeated administration. That is, at higher doses (for example, 25 mg / kg or higher) than the limits for passage through the BBB and BNB, the increase in efficacy is reduced by a single dose increase, but a certain interval between administrations (eg, more than half a day) should be avoided. Efficacy can be improved by repeated administration.
- the nucleic acid complex of the present invention has excellent solubility in water, the second liquid of the Japanese Pharmacopoeia dissolution test, or the second liquid of the Japanese Pharmacopoeia disintegration test, and has a pharmacokinetics (eg, blood drug half-life). , Brain transfer, metabolic stability, CYP inhibition), low toxicity (eg acute toxicity, chronic toxicity, genotoxicity, reproductive toxicity, cardiotoxicity, drug interaction, carcinogenicity, phototoxicity, etc.) Therefore, it also has excellent properties as a pharmaceutical product such as a superior side effect (eg, suppression of oversedation).
- a superior side effect eg, suppression of oversedation.
- the composition of the present invention utilizes the fact that the nucleic acid complex of the first aspect of the present invention contained as an active ingredient passes through the BBB and can be efficiently delivered to the central nervous system.
- the agent By binding to the nucleic acid strand and / or the second nucleic acid strand, the agent can be delivered to the nervous system, particularly the central nervous system.
- the drug delivered to the nervous system is not particularly limited, and includes peptides, proteins or nucleic acid drugs, or other organic compounds such as antitumor drugs, hormone drugs, antibiotics, antiviral drugs, anti-inflammatory drugs, and the like. .
- the drug is preferably a small molecule drug. “Small molecule drugs” are well understood in the art.
- Small molecule drugs typically refer to drugs having a molecular weight of less than 1,000 daltons.
- the drug may be a lipophilic drug.
- the nucleic acid drug include, but are not limited to, ASO, antagomiR, splice switching oligonucleotide, aptamer, single-stranded siRNA, microRNA, pre-microRNA and the like.
- the binding position and type of binding in the second nucleic acid strand of the drug is as described above for binding of phosphatidylethanolamine or its analog to the second nucleic acid strand.
- the composition of the present invention is delivered to the central nervous system with high efficiency as disclosed in the following examples, and can effectively modify or suppress the expression of a target gene or the level of a target transcript. Accordingly, there is provided a method for reducing the expression level of a target transcript in the central nervous system of a subject, comprising administering to the subject a composition comprising the nucleic acid complex of the present invention described above. .
- the method may be a method for treating a central nervous system disease in a subject.
- a method for delivering a drug to the central nervous system of a subject comprising administering to the subject a composition comprising the nucleic acid complex of the present invention described above.
- Root temperature in the following examples usually indicates about 10 ° C. to about 35 ° C.
- the ratio shown in the mixed solvent is a volume ratio unless otherwise specified. Unless otherwise indicated, “%” indicates “% by weight”.
- HPLC high performance liquid chromatography
- octadecyl-bonded silica gel was used.
- the ratio shown in the elution solvent indicates a volume ratio unless otherwise specified.
- -DIPEA N, N-diisopropylethylamine-NMP: N-methyl-2-pyrrolidone-DMAP: 4-dimethylaminopyridine-HATU: O- (7-azabenzotriazol-1-yl) -N, N, N ' , N'-Tetramethyluronium hexafluorophosphate ⁇
- PBS Phosphate buffered saline
- TEAA Triethylamine acetate
- THF Tetrahydrofuran
- oligonucleotides used in the following examples are summarized in Table 1.
- ASO Merat1
- HA-cRNA Merat1
- HA The structure of HA described in Table 1 is shown by the following formula V.
- the designation “5 ′ oligo” represents the 5 ′ end of the oligonucleotide.
- the designation “5 ′ oligo” represents the 5 ′ end of the oligonucleotide.
- the designation “5 ′ oligo” represents the 5 ′ end of the oligonucleotide.
- the designation “5 ′ oligo” represents the 5 ′ end of the oligonucleotide.
- the designation “5 ′ oligo” represents the 5 ′ end of the oligonucleotide.
- the designation “5 ′ oligo” represents the 5 ′ end of the oligonucleotide.
- the designation “5 ′ oligo” represents the 5 ′ end of the oligonucleotide.
- the designation “5 ′ oligo” represents the 5 ′ end of the oligonucleotide.
- the designation “5 ′ oligo” represents the 5 ′ end of the oligonucleotide.
- Step 1 Synthesis of DSPE-cRNA (Malat1) 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) at the 5 ′ end
- a cRNA (DSPE-cRNA (Malat1)) of a metastasis-associated lung adenocarcinoma transcript (Malat1) conjugated with a glutaryl group was synthesized by the following procedure.
- ASO (Malat1) and HA-cRNA (Malat1) were outsourced to Gene Design Co., Ltd.
- COATSOME FE-8080SU5 50 mM NMP solution (800 ⁇ L), distilled water (271 ⁇ L), NMP (7200 ⁇ L), ⁇ 10 PBS (1000 ⁇ L) in an aqueous solution (4000 nmol) of the RNA strand shown in Table 1 (HA-cRNA (Malat1)) , DIPEA (125 ⁇ L) in the order of Eppen, stirred and settled, and then reacted at 70 ° C. for 2 hours.
- the sample was subjected to solvent exchange using gel filtration chromatography (Sephadex G-25: manufactured by GE, moving bed: distilled water), and then freeze-dried.
- the residue is purified by preparative HPLC (column: Xbridge OST C18 2.5 ⁇ m, 10mmID ⁇ 50mm: Waters, mobile phase: TEAA / acetonitrile), and removed by ultrafiltration (Amicon ultra: Merck Millipore, distilled water). Salt treatment was performed. To the resulting solution (300 ⁇ L), 10 times the amount of 1M meglumine acetate was added, stirred well, and allowed to stand for 5 minutes for ion exchange. Thereafter, desalting was performed by ultrafiltration (Amicon ultrapore 3kDa: manufactured by Merck Millipore, distilled water). The final product was filtered through a 0.20 ⁇ m membrane filter and lyophilized to obtain 730 nmol using the title compound as a 5% glucose solution.
- Step 2 Synthesis of Double-Stranded Nucleic Acid Agent DSPE-HDO ASO (Malat1) shown in Table 1 is a 16mer single-stranded LNA / DNA gapmer targeting Malat1 non-coding RNA. Includes 3 and 3 LNA nucleosides at the 3 'end and 10 DNA nucleosides between them. This LNA / DNA gapmer has a base sequence complementary to 1316 to 1331 of mouse Malat1 non-coding RNA (GenBank accession number NR — 002847: SEQ ID NO: 3).
- both nucleic acid strands were mixed in equimolar amounts, and the solution was heated to 70 ° C. For 7 minutes and then slowly cooled to room temperature. As a result, both nucleic acid strands were annealed to prepare “DSPE-glutaryl-binding heteroduplex oligonucleotide (DSPE-conjugated heteroduplex oligonucleotide, DSPE-HDO)” which is a nucleic acid complex of the present invention.
- DSPE-glutaryl-binding heteroduplex oligonucleotide DSPE-conjugated heteroduplex oligonucleotide, DSPE-HDO
- Step 1 Synthesis of DOPE-cRNA (Malat1) 1,2-Dioleoyl-sn-glycero-3-phosphatidylethanolamine (DOPE) glutaryl group at the 5 'end
- DOPE-cRNA (Malat1) 1,2-Dioleoyl-sn-glycero-3-phosphatidylethanolamine (DOPE) glutaryl group at the 5 'end
- DOPE-cRNA (Malat1) 1,2-Dioleoyl-sn-glycero-3-phosphatidylethanolamine
- RNA strand (HA-cRNA (Malat1)) shown in Table 1
- DOPE- cRNA (Malat1) was synthesized to obtain 1050 nmol as a 5% glucose solution.
- Step 2 Synthesis of double-stranded nucleic acid agent DOPE-HDO ASO (Malat1) is the first nucleic acid strand, and DOPE-cRNA (Malat1) obtained in Step 1 of Example 2 is the second nucleic acid strand. Nucleic acid strands are mixed in equimolar amounts, and in the same manner as in Step 2 of Example 1, the nucleic acid complex of the present invention, “DOPE-conjugated heteroduplex oligonucleotide (DOPE-HDO) ) ”Was prepared.
- DOPE-HDO ASO is the first nucleic acid strand
- DOPE-cRNA (Malat1) obtained in Step 1 of Example 2 is the second nucleic acid strand.
- Nucleic acid strands are mixed in equimolar amounts, and in the same manner as in Step 2 of Example 1, the nucleic acid complex of the present invention, “DOPE-conjugated heteroduplex oligonucleotide (DOPE-HDO
- Malat1 cRNA (DPPE-cRNA) with 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine (DPPE) glutaryl group attached to the 5 'end (Malat1) was synthesized by the following procedure.
- RNA strand (HA-cRNA (Malat1)) shown in Table 1
- HA-cRNA a cRNA having a DPPE glutaryl group bonded to the 5 ′ end in the same procedure as in Step 1 of Example 1
- DPPE- cRNA (Malat1) was synthesized to obtain 891 nmol as a 5% glucose solution.
- Step 2 Synthesis of Double-Stranded Nucleic Acid Agent
- DPPE-HDO ASO (Malat1) is the first nucleic acid strand
- DPPE-cRNA (Malat1) obtained in Step 1 of Example 3 is the second nucleic acid strand.
- Nucleic acid strands were mixed in equimolar amounts, and “DPPE-conjugated heteroduplex oligonucleotide: DPPE-HDO”, which is the nucleic acid complex of the present invention, was prepared in the same manner as in Step 2 of Example 1. ) ”Was prepared.
- Step 1 Synthesis of POPE-cRNA (Malat1) 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine is attached to the 5 ′ end of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine.
- POPE A cRNA of Malat1 (POPE-cRNA (Malat1)) to which a glutaryl group was bound was synthesized by the following procedure.
- Step 2 Synthesis of Double-Stranded Nucleic Acid Agent
- POPE-HDO Double-Stranded Nucleic Acid Agent
- ASO Malat1
- POPE-cRNA Malat1
- Nucleic acid strands are mixed in equimolar amounts, and in the same manner as in Step 2 of Example 1, the POPE-conjugated heteroduplex oligonucleotide (POPE-HDO) is a nucleic acid complex of the present invention. ) ”Was prepared.
- Step 1 Synthesis of Glutaryl-cRNA (Malat1)
- An aqueous solution (5000 nmol) of the RNA strand (HA-cRNA (Malat1)) shown in Table 1 was added to a commercially available 50 mM NMP solution of glutaric anhydride (2000 ⁇ L), distilled water (415 ⁇ L). ), NMP (6000 ⁇ L), ⁇ 10 PBS (1250 ⁇ L), and commercially available DMAP 50 mM NMP solution (2000 ⁇ L) were mixed in an Eppen, stirred and settled, and reacted at 70 ° C. for 2 hours.
- the sample was subjected to solvent exchange using gel filtration chromatography (Sephadex G-25: manufactured by GE, moving bed: distilled water) and then freeze-dried to obtain 4542 nmol of the title compound.
- Step 2 Synthesis of 18: 1 (delta9-Trans) PE-cRNA (Malat1) 1,2-dielideoyl-sn-glycero-3-phosphoethanolamine (1,2-dielaidoyl-sn-glycero) -3-phosphoethanolamine: 18: 1 (delta9-Trans) PE)
- a malat1 cRNA (18: 1 (delta9-Trans) PE-cRNA (Malat1)) to which glutaryl group was bound was synthesized by the following procedure.
- the reaction solution was purified with an ODS column (column: Purif-Pack (registered trademark) -EX ODS-50 size 60, manufactured by Shoko Science, mobile phase: TEAA / acetonitrile), and ultrafiltration (Amicon ultra, manufactured by Merck Millipore) , Distilled water).
- ODS column column: Purif-Pack (registered trademark) -EX ODS-50 size 60, manufactured by Shoko Science, mobile phase: TEAA / acetonitrile
- ultrafiltration Analogenethoxys
- To the resulting solution 10 times the amount of 1M meglumine acetate was added, stirred well, and allowed to stand for 5 minutes for ion exchange. Thereafter, desalting was performed by ultrafiltration (Amicon ultra, manufactured by Merck Millipore, distilled water). The final product was filtered through a 0.20 ⁇ m membrane filter and freeze-dried to obtain 1071 nmol of the title compound.
- Step 3 Synthesis of double-stranded nucleic acid agent 18: 1 (delta9-Trans) PE-HDO
- ASO (Malat1) is the first nucleic acid strand, and 18: 1 (delta9-Trans obtained in Step 2 of Example 5)
- PE-cRNA (Malat1) as the second nucleic acid strand
- both nucleic acid strands are mixed in equimolar amounts, and in the same manner as in Step 2 of Example 1, the nucleic acid complex of the present invention “18: 1 (delta9 -Trans) PE glutaryl-linked heteroduplex oligonucleotide (18: 1 (delta9-Trans) PE-conjugated heteroduplex oligonucleotide: 18: 1 (delta9-Trans) PE-HDO) "was prepared.
- Step 1 Synthesis of 18: 0-18: 1 PE-cRNA (Malat1)
- 1-stearoyl-2-oleoyl -sn-glycero-3-phosphoethanolamine 18: 0-18: 1 PE
- Malat1 cRNA (18: 0-18: 1 PE-cRNA (Malat1)) to which glutaryl group was bound was synthesized by the following procedure.
- RNA strand (Glutaryl-cRNA (Malat1)) synthesized in the same manner as in Step 1 of Example 5, the procedure was the same as in Step 2 of Example 5, and 18: 0 was added to the 5 ′ end. 18: 0-18: 1 ⁇ ⁇ PE-cRNA (Malat1) was synthesized as cRNA bound with -18: 1 PE glutaryl group, and 721 nmol was obtained as a 5% glucose solution.
- Step 2 Synthesis of double-stranded nucleic acid agent 18: 0-18: 1 PE-HDO ASO (Malat1) was used as the first nucleic acid strand, and 18: 0-18: 1 obtained in Step 1 of Example 6 Using PE-cRNA (Malat1) as the second nucleic acid strand, both nucleic acid strands are mixed in an equimolar amount, and the nucleic acid complex of the present invention “18: 0-18: 1 PE glutaryl-linked heteroduplex oligonucleotide (18: 0-18: 1 PE-conjugated heteroduplex oligonucleotide: 18: 0-18: 1 PE-HDO) ”was prepared.
- Step 1 Synthesis of 18: 2 PE-cRNA (Malat1) 1,2-Dilinoleoyl-sn-glycero-3-phosphoethanolamine: 18: 2 PE)
- Malat1 cRNA (18: 2 PE-cRNA (Malat1)) bound with glutaryl group was synthesized by the following procedure.
- RNAPE-cRNA (Malat1) was synthesized as a cRNA to which a glutaryl group was bound, and 544 nmol was obtained as a 5% glucose solution.
- Step 2 Synthesis of double-stranded nucleic acid agent 18: 2 PE-HDO ASO (Malat1) was used as the first nucleic acid strand, and 18: 2 PE-cRNA (Malat1) obtained in Step 1 of Example 7 was used as the first nucleic acid strand.
- both nucleic acid strands are mixed in equimolar amounts, and in the same manner as in Step 2 of Example 1, the nucleic acid complex of the present invention, “18: 2 PE glutaryl-linked heteroduplex oligonucleotide ( 18: 2 PE-conjugated heteroduplex oligonucleotide: 18: 2 PE-HDO) ”.
- Step 1 Synthesis of DPyPE-cRNA (Malat1) 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE) glutaryl at the 5 ′ end
- DPyPE-cRNA (Malat1) 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine
- DPyPE glutaryl group was bound to the 5 ′ end in the same procedure as in Step 2 of Example 5.
- DPyPE-cRNA (Malat1) was synthesized as cRNA, and 961 nmol was obtained as a 5% glucose solution.
- Step 2 Synthesis of Double-Stranded Nucleic Acid Agent DPyPE-HDO Both ASO (Malat1) is the first nucleic acid strand, and DPyPE-cRNA (Malat1) obtained in Step 1 of Example 8 is the second nucleic acid strand. Nucleic acid strands were mixed in equimolar amounts, and the nucleic acid complex of the present invention, “DPyPE-glutaryl-linked heteroduplex oligonucleotide (DPyPE-HDO), was synthesized in the same manner as in Step 2 of Example 1. ) ”Was prepared.
- DPyPE-glutaryl-linked heteroduplex oligonucleotide DPyPE-HDO
- Example 9 In vivo experiment Seven-week-old male C57BL / 6J mice (Nippon Charles River) were used as experimental animals, and two mice per group were subjected to the experiment. In the experimental group, the solution containing the nucleic acid was administered once intravenously at a dose of 5 mL / kg from the tail vein of the mouse. In the comparative group, the solvent (5% glucose solution) used for preparing the nucleic acid solution was intravenously administered to mice in the same procedure as in the experimental group.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Genetics & Genomics (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Biochemistry (AREA)
- Wood Science & Technology (AREA)
- Biotechnology (AREA)
- General Engineering & Computer Science (AREA)
- Zoology (AREA)
- Epidemiology (AREA)
- Biophysics (AREA)
- Microbiology (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Neurology (AREA)
- Neurosurgery (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicinal Preparation (AREA)
Abstract
Description
近年、核酸医薬と呼ばれる医薬品の現在進行中の開発において、オリゴヌクレオチドが関心を集めており、また特に、標的遺伝子の高い選択性及び低毒性の点から考えて、アンチセンス法を利用する核酸医薬の開発が積極的に進められている。アンチセンス法とは、標的遺伝子より転写されたmRNAやmiRNAの部分配列を標的センス鎖として、それに相補的なオリゴヌクレオチド(アンチセンスオリゴヌクレオチド:本明細書ではしばしば「ASO(AntiSense Oligonucleotide)」と表記する)を細胞に導入することによって、標的遺伝子によってコードされたタンパク質の発現やmiRNAの活性を選択的に改変又は阻害することを含む方法である。
(2)前記ホスファチジルエタノールアミン又はその類縁体が一般式Iで表される、(1)に記載の核酸複合体又はその塩。
(3)前記R1及びR2が、それぞれ独立に、C15~C19のアルキル基、又はC17のアルケニル基である、(2)に記載の核酸複合体又はその塩。
(4)前記ホスファチジルエタノールアミン又はその類縁体が一般式XV~XXIIで表される、(1)~(3)のいずれかに記載の核酸複合体又はその塩。
(6)前記第2核酸鎖の5'末端に、前記リンカーを介してホスファチジルエタノールアミン又はその類縁体が結合している、(5)に記載の核酸複合体。
(7)前記リンカー結合された前記ホスファチジルエタノールアミン又はその類縁体が一般式IIIで表される、(5)又は(6)に記載の核酸複合体又はその塩。
(8)前記第1核酸鎖が少なくとも4個の連続するデオキシリボヌクレオシドを含む、(1)~(7)のいずれかに記載の核酸複合体又はその塩。
(9)前記第1核酸鎖がギャップマーである、(1)~(8)のいずれかに記載の核酸複合体又はその塩。
(10)前記ギャップマーがLNA/DNAギャップマーである、(9)に記載の核酸複合体又はその塩。
(11)前記第2核酸鎖が前記第1核酸鎖中の少なくとも4個の連続するデオキシリボヌクレオシドに相補的な、少なくとも4個の連続するリボヌクレオシドを含む、(8)~(10)のいずれかに記載の核酸複合体又はその塩。
(12)前記第1核酸鎖がミックスマーである、(1)~(7)のいずれかに記載の核酸複合体又はその塩。
(13)前記第1核酸鎖が13~20塩基長である、(1)~(12)のいずれかに記載の核酸複合体又はその塩。
(14)前記第2核酸鎖が天然リボヌクレオシドを含まない、(1)~(13)のいずれかに記載の核酸複合体又はその塩。
(15)前記第2核酸鎖の核酸部分が修飾若しくは非修飾のヌクレオシド間結合により連結されたデオキシリボヌクレオシド及び/又は糖修飾ヌクレオシドから成る、(1)~(14)のいずれかに記載の核酸複合体又はその塩。
(16)(1)~(15)のいずれかに記載の核酸複合体又はその塩を含む、中枢神経系において標的転写産物の発現又は編集を調節するための組成物。
(17)中枢神経系疾患治療用である、(16)に記載の組成物。
(18)(1)~(15)のいずれかに記載の核酸複合体又はその塩を含む、中枢神経系薬剤送達用組成物。
(19)前記中枢神経系が大脳皮質、大脳基底核、大脳白質、間脳、脳幹、小脳、及び脊髄からなる群から選択される、(16)~(18)のいずれかに記載の組成物。
(20)前記中枢神経系が前頭葉、側頭葉、海馬、海馬傍回、頭頂葉、後頭葉、線条体、淡蒼球、前障、視床、視床下核、中脳、黒質、橋、延髄、小脳皮質、小脳核、頸髄、胸髄及び腰髄からなる群から選択される、(16)~(18)のいずれかに記載の組成物。
(21)静脈内投与用又は皮下投与用である、(16)~(20)のいずれかに記載の組成物。
(22)1回の投与量中に5mg/kg以上の前記核酸複合体又はその塩を含む、(16)~(21)のいずれかに記載の組成物。
(23)前記核酸複合体又はその塩がBBBを通過する、(16)~(22)のいずれかに記載の組成物。
本明細書は本願の優先権の基礎となる日本国特許出願番号2018-047294号の開示内容を包含する。
1.核酸複合体
本発明の第1態様は、核酸複合体、より好ましくは血液脳関門通過型核酸複合体である。この核酸複合体は、第1核酸鎖と第2核酸鎖とを含む。第2核酸鎖は、第1核酸鎖に相補的な塩基配列を含むヌクレオチド鎖である。核酸複合体において、第1核酸鎖は、第2核酸鎖にアニールしている。一実施形態では、第2核酸鎖は、ホスファチジルエタノールアミン又はその類縁体と結合している。
本明細書において「標的転写産物」とは、本発明の核酸複合体の標的となり得、かつDNA依存性RNAポリメラーゼによって合成される任意のRNAをいう。一般的には標的遺伝子の転写産物が該当する。具体的には、標的遺伝子から転写されるmRNA(成熟mRNA、mRNA前駆体、塩基修飾を受けていないmRNA等を含む)、miRNA等のノンコーディングRNA(non-coding RNA、ncRNA)を含み得る。
第1核酸鎖は、標的転写産物の少なくとも一部にハイブリダイズすることが可能な塩基配列を含み、標的転写産物に対してアンチセンス効果をもたらす一本鎖オリゴヌクレオチド鎖である。
第1核酸鎖と第2核酸鎖とを含む核酸複合体又はその塩であって、
該第1核酸鎖は標的転写産物の少なくとも一部にハイブリダイズすることが可能な塩基配列を含み、かつ、標的転写産物に対してアンチセンス効果を有し、
該第2核酸鎖は該第1核酸鎖に相補的な塩基配列を含み、かつ、ホスファチジルエタノールアミン又はその類縁体と結合しており、
該第1核酸鎖は該第2核酸鎖にアニールしている前記核酸複合体又はその塩。
第1核酸鎖と第2核酸鎖とを含む核酸複合体又はその塩であって、
該第1核酸鎖は
(1)13~20塩基長の塩基配列を含み、
(2)標的転写産物の少なくと一部にハイブリダイズすることができ、
(3)少なくとも4個の連続するデオキシリボヌクレオシドを含み、かつ、
(4)標的転写産物に対してアンチセンス効果を有し、
該第2核酸鎖は
(1)前記第1核酸鎖中の少なくとも4個の連続するデオキシリボヌクレオシドに相補的な、少なくとも4個の連続するリボヌクレオシドを含み、かつ、
(2)一般式I(好ましくは、一般式XV~XXII)で表される、ホスファチジルエタノールアミン又はその類縁体と結合しており、
該第1核酸鎖は該第2核酸鎖にアニールしている前記核酸複合体又はその塩。
第1核酸鎖と第2核酸鎖とを含む核酸複合体又はその塩であって、
該第1核酸鎖は
(1)ヌクレオシドと任意に非天然ヌクレオシドとを含み、該核酸鎖における該ヌクレオシド及び任意に含まれる該非天然ヌクレオシドの総数は13~20であり、
(2)標的転写産物の少なくと一部にハイブリダイズすることができ、
(3)RNaseHによって認識される少なくとも4個の連続するデオキシリボヌクレオシドを含み、
(4)前記4つの連続したヌクレオシドの5’及び/又は3’側に位置する1又は複数の糖修飾ヌクレオチドである非天然ヌクレオシドを含み、かつ
(5)標的転写産物に対してアンチセンス効果を有し、
該第2核酸鎖は
(1)前記第1核酸鎖中の少なくとも4個の連続するデオキシリボヌクレオシドに相補的な、少なくとも4個の連続するリボヌクレオシドを含み、
(2)前記少なくとも4つの連続したリボヌクレオシドの5’側に位置する1以上の非天然ヌクレオシドを含み、
(3)前記少なくとも4つの連続したリボヌクレオシドの3’側に位置する1以上の非天然ヌクレオシドを含み、かつ
(4)一般式I(好ましくは、一般式XV~XXII)で表される、ホスファチジルエタノールアミン又はその類縁体と結合しており、
該第1核酸鎖は該第2核酸鎖にアニールしている前記核酸複合体又はその塩。
第1核酸鎖と第2核酸鎖とを含む核酸複合体又はその塩であって、
該第1核酸鎖は
(1)ヌクレオシドと任意に非天然ヌクレオシドとを含み、該核酸鎖における該ヌクレオシド及び任意に含まれる該非天然ヌクレオシドの総数は13~20であり、
(2)標的転写産物の少なくと一部にハイブリダイズすることができ、
(3)RNaseHによって認識される少なくとも4個の連続するデオキシリボヌクレオシドを含み、
(4)前記4つの連続したヌクレオシドの5’及び/又は3’側に位置する1又は複数の糖修飾ヌクレオチドである非天然ヌクレオシドを含み、かつ
(5)標的転写産物に対してアンチセンス効果を有し、
該第2核酸鎖は
(1)前記第1核酸鎖中の少なくとも4個の連続するデオキシリボヌクレオシドに相補的な、少なくとも4個の連続するリボヌクレオシドを含み、
(2)前記少なくとも4つの連続したリボヌクレオシドの5’側に位置する1以上の非天然ヌクレオシドを含み、
(3)前記少なくとも4つの連続したリボヌクレオシドの3’側に位置する1以上の非天然ヌクレオシドを含み、
(4)一般式I(好ましくは、一般式XV~XXII)で表される、ホスファチジルエタノールアミン又はその類縁体と結合しており、かつ
(5)前記第2核酸鎖が、一般式IIで表されるリンカーを介して、(好ましくは、前記第2核酸鎖の5'末端にて)ホスファチジルエタノールアミン又はその類縁体と結合している、
該第1核酸鎖は該第2核酸鎖にアニールしている前記核酸複合体又はその塩。
本発明の核酸複合体は、被験体の中枢神経系において標的miRNAの効果を阻害することができる。例えば、具体的には、第1核酸鎖は、標的miRNAの少なくとも一部にハイブリダイズすることが可能な塩基配列を含み、かつ、その標的miRNAに対してアンチセンス効果を有し、第2核酸鎖は、前記第1核酸鎖に相補的な塩基配列を含み、かつ、PE又はその類縁体と結合しており、第1核酸鎖と第2核酸鎖に互いにアニールしている核酸複合体が挙げられる。この核酸複合体によって標的miRNAの効果を阻害することにより、当該標的miRNAが通常ダウンレギュレートしている遺伝子の発現をアップレギュレートすることができる。
本発明の第2態様は組成物である。本発明の組成物は、前記第1態様の本発明の核酸複合体を有効成分として、及び/又は薬剤送達分子として包含する。前記第1態様の本発明の核酸複合体は、BBBを通過し、中枢神経系にて標的転写産物の発現量をアンチセンス効果によって調節する(例えば、発現量を減少させる)ことができる。したがって、本発明の組成物は、被験体に投与することで、本発明の核酸複合体を送達し、被験体を治療する組成物であってもよく、また医薬組成物であってもよい。
本明細書において、組成物は自体公知の方法により製剤化することができる。例えば、本組成物は、カプセル剤、錠剤、丸剤、液剤、散剤、顆粒剤、微粒剤、フィルムコーティング剤、ペレット剤、トローチ剤、舌下剤、解膠剤(peptizer)、バッカル剤、ペースト剤、シロップ剤、懸濁剤、エリキシル剤、乳剤、コーティング剤、軟膏、硬膏剤(plaster)、パップ剤(cataplasm)、経皮剤、ローション剤、吸入剤、エアロゾル剤、点眼剤、注射剤及び坐剤の形態で、経口的に又は非経口的に使用することができる。
本明細書において、組成物の好ましい投与形態には特定の限定はない。例えば、経口投与又は非経口投与であればよい。非経口投与の具体例として、静脈内投与、動脈内投与、腹腔内投与、皮下投与、皮内投与、気管/気管支投与、直腸投与、及び筋肉内投与、並びに輸血による投与が挙げられる。筋肉内注射投与、静脈内点滴投与、又は埋め込み型持続皮下投与で投与することもできる。皮下投与は、患者自身による自己注射が可能であるので好適である。また、静脈内投与の場合、組成物1回の投与量中に含まれる本発明の核酸複合体の量、すなわち本発明の核酸複合体の単回投与量は、例えば、0.001mg/kg以上、0.005mg/kg以上、0.01mg/kg以上、00.25mg/kg以上、0.5mg/kg以上、1mg/kg以上、2.5mg/kg以上、5mg/kg以上、10mg/kg以上、20mg/kg以上、30mg/kg以上、40mg/kg以上、50mg/kg以上、75mg/kg以上、100mg/kg以上、150mg/kg以上、200mg/kg以上、300mg/kg以上、400mg/kg以上、若しくは500mg/kg以上とすることができる。例えば、0.001mg/kg~500mg/kgの範囲に含まれる任意の量(例えば、0.001mg/kg、0.01mg/kg、0.1mg/kg、1mg/kg、5mg/kg、10mg/kg、50mg/kg、100mg/kg、若しくは200mg/kg)を適宜選択することができる。
本明細書で「被検体」とは、本発明の組成物を適用する対象をいう。被検体は、個体の他、器官、組織、及び細胞を含む。被検体が個体の場合、本発明の組成物は、ヒトを含むあらゆる動物に適用することができる。ヒト以外の対象としては、例えば、様々な家畜、家禽、ペット、実験動物等が適用対象となり得る。被験体は、中枢神経系で標的転写産物の発現量を減少させることが必要な個体や、中枢神経系疾患の治療が必要な個体であってもよい。
本発明の組成物は、有効成分として含有する第1態様の本発明の核酸複合体がBBBを通過し、中枢神経系に効率的に送達され得ることを利用して、特定の薬剤を第1核酸鎖及び/又は第2核酸鎖に結合させることによって、その薬剤を神経系、特に中枢神経系に送達することができる。神経系に送達される薬剤は、特に限定されないが、ペプチド、タンパク質又は核酸薬剤、あるいはその他の有機化合物等、例えば抗腫瘍薬、ホルモン薬、抗生物質、抗ウイルス剤、抗炎症薬等が挙げられる。薬剤は、好ましくは、小分子薬剤である。「小分子薬剤」とは、当該技術分野において十分に理解されている。小分子薬剤は、典型的には、1,000ダルトン未満の分子量を有する薬剤を指す。薬剤は、親油性薬剤であってもよい。核酸薬剤としては、特に限定されないが、ASO、アンタゴmiR、スプライススイッチングオリゴヌクレオチド、アプタマー、一本鎖siRNA、マイクロRNA、プレ-マイクロRNA等が挙げられる。薬剤の第2核酸鎖における結合位置及び結合の種類は、ホスファチジルエタノールアミン又はその類縁体と第2核酸鎖との結合について上に記載される通りである。
分取HPLC(高速液体クロマトグラフィー)において、C18と記載した場合は、オクタデシル結合シリカゲルを用いた。溶出溶媒において示した比は、特に断らない限り容量比を示す。
・DIPEA:N,N-ジイソプロピルエチルアミン
・NMP:N-メチル-2-ピロリドン
・DMAP:4-ジメチルアミノピリジン
・HATU:O-(7-アザベンゾトリアゾール-1-イル)-N,N,N',N'-テトラメチルウロニウム ヘキサフルオロリン酸塩
・PBS:リン酸緩衝生理食塩水
・TEAA:トリエチルアミンアセタート
・THF:テトラヒドロフラン
(工程1)DSPE-cRNA(Malat1)の合成
5'末端に、1,2-ジステアロイル-sn-グリセロ-3-ホスホエタノールアミン(1,2-Dioctadecanoyl-sn-glycero-3-phosphoethanolamine:DSPE)グルタリル基を結合させた転移関連肺腺癌転写産物(Malat1)のcRNA(DSPE-cRNA(Malat1))を以下の手順により合成した。なお、ASO(Malat1)及びHA-cRNA(Malat1)は、株式会社ジーンデザインに合成委託した。
表1で示すASO(Malat1)は、Malat1ノンコーディングRNAを標的とする16merの一本鎖LNA/DNAギャップマーであり、5'末端の3個及び3'末端の3個のLNAヌクレオシド、並びにそれらの間の10個のDNAヌクレオシドを含む。このLNA/DNAギャップマーは、マウスのMalat1ノンコーディングRNA(GenBankアクセッション番号NR_002847:配列番号3)の1316~1331に相補的な塩基配列を有する。
(工程1)DOPE-cRNA(Malat1)の合成
5'末端に1,2-ジオレオイル-sn-グリセロ-3-ホスホエタノールアミン(1,2-Dioleoyl-sn-glycero-3-phosphatidylethanolamine:DOPE)グルタリル基を結合させたMalat1のcRNA(DOPE-cRNA(Malat1))を以下の手順により合成した。
ASO(Malat1)を第1核酸鎖とし、また実施例2の工程1で得られたDOPE-cRNA(Malat1)を第2核酸鎖として、両核酸鎖を等モル量で混合し、実施例1の工程2と同様にして、本発明の核酸複合体である「DOPEグルタリル結合型ヘテロ二本鎖オリゴヌクレオチド(DOPE-conjugated heteroduplex oligonucleotide:DOPE-HDO)」を調製した。
(工程1)DPPE-cRNA(Malat1)の合成
ASO(Malat1)を第1核酸鎖とし、また実施例3の工程1で得られたDPPE-cRNA(Malat1)を第2核酸鎖として、両核酸鎖を等モル量で混合し、実施例1の工程2と同様にして、本発明の核酸複合体である「DPPEグルタリル結合型ヘテロ二本鎖オリゴヌクレオチド(DPPE-conjugated heteroduplex oligonucleotide:DPPE-HDO)」を調製した。
(工程1)POPE-cRNA(Malat1)の合成
5'末端に1-パルミトイル-2-オレオイル-sn-グリセロ-3-ホスホエタノールアミン(1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine:POPE)グルタリル基を結合させたMalat1のcRNA(POPE-cRNA(Malat1))を以下の手順により合成した。
ASO(Malat1)を第1核酸鎖とし、また実施例4の工程1で得られたPOPE-cRNA(Malat1)を第2核酸鎖として、両核酸鎖を等モル量で混合し、実施例1の工程2と同様にして、本発明の核酸複合体である「POPEグルタリル結合型ヘテロ二本鎖オリゴヌクレオチド(POPE-conjugated heteroduplex oligonucleotide:POPE-HDO)」を調製した。
(工程1)Glutaryl-cRNA(Malat1)の合成
表1に示すRNA鎖(HA-cRNA(Malat1))の水溶液(5000nmol)に市販のグルタル酸無水物の50mM NMP溶液(2000μL)、蒸留水(415μL)、NMP(6000μL)、×10PBS(1250μL)、市販のDMAP 50mM NMP溶液(2000μL)の順にエッペン中で混合し、撹拌、沈降した後、70℃で2時間反応させた。サンプルをゲルろ過クロマトグラフィー(Sephadex G-25:GE製、移動層:蒸留水)を用いて溶媒交換した後、凍結乾燥し標題化合物を4542nmol得た。
5'末端に、1,2-ジエライドイル-sn-グリセロ-3-ホスホエタノールアミン(1,2-dielaidoyl-sn-glycero-3-phosphoethanolamine:18:1 (delta9-Trans) PE)グルタリル基を結合させたMalat1のcRNA(18:1 (delta9-Trans) PE -cRNA(Malat1))を以下の手順により合成した。
ASO(Malat1)を第1核酸鎖とし、また実施例5工程2で得られた18:1 (delta9-Trans) PE-cRNA(Malat1)を第2核酸鎖として、両核酸鎖を等モル量で混合し、実施例1の工程2と同様にして、本発明の核酸複合体である「18:1 (delta9-Trans) PEグルタリル結合型ヘテロ二本鎖オリゴヌクレオチド(18:1 (delta9-Trans) PE -conjugated heteroduplex oligonucleotide:18:1 (delta9-Trans) PE -HDO)」を調製した。
(工程1)18:0-18:1 PE-cRNA(Malat1)の合成
5'末端に、1-ステアロイル-2-オレオイル-sn-グリセロ-3-ホスホエタノールアミン(1-stearoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine:18:0-18:1 PE)グルタリル基を結合させたMalat1のcRNA(18:0-18:1 PE-cRNA(Malat1))を以下の手順により合成した。
ASO(Malat1)を第1核酸鎖とし、また実施例6の工程1で得られた18:0-18:1 PE-cRNA(Malat1)を第2核酸鎖として、両核酸鎖を等モル量で混合し、実施例1の工程2と同様にして、本発明の核酸複合体である「18:0-18:1 PEグルタリル結合型ヘテロ二本鎖オリゴヌクレオチド(18:0-18:1 PE-conjugated heteroduplex oligonucleotide:18:0-18:1 PE-HDO)」を調製した。
(工程1)18:2 PE-cRNA(Malat1)の合成
5'末端に、1,2-ジリノレオイル-sn-グリセロ-3-ホスホエタノールアミン(1,2-dilinoleoyl-sn-glycero-3-phosphoethanolamine:18:2 PE)グルタリル基を結合させたMalat1のcRNA(18:2 PE-cRNA(Malat1))を以下の手順により合成した。
ASO(Malat1)を第1核酸鎖とし、また実施例7の工程1で得られた18:2 PE-cRNA(Malat1)を第2核酸鎖として、両核酸鎖を等モル量で混合し、実施例1の工程2と同様にして、本発明の核酸複合体である「18:2 PEグルタリル結合型ヘテロ二本鎖オリゴヌクレオチド(18:2 PE-conjugated heteroduplex oligonucleotide:18:2 PE-HDO)」を調製した。
(工程1)DPyPE-cRNA(Malat1)の合成
5'末端に、1,2-ジフィタノイル-sn-グリセロ-3-ホスホエタノールアミン(1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine:DPyPE)グルタリル基を結合させたMalat1のcRNA(DPyPE-cRNA(Malat1))を以下の手順により合成した。
ASO(Malat1)を第1核酸鎖とし、また実施例8の工程1で得られたDPyPE-cRNA(Malat1)を第2核酸鎖として、両核酸鎖を等モル量で混合し、実施例1の工程2と同様にして、本発明の核酸複合体である「DPyPEグルタリル結合型ヘテロ二本鎖オリゴヌクレオチド(DPyPE-conjugated heteroduplex oligonucleotide:DPyPE-HDO)」を調製した。
(A)in vivo実験
実験動物には、7週齢の雄性C57BL/6Jマウス(日本チャールズリバー)を使用し、一群あたり2匹を実験に供した。実験群では、核酸を含む溶液をマウスの尾静脈から5mL/kgの投与量で静脈内に単回投与した。また、比較対象群では、核酸溶液の調製に用いた溶媒(5%ブドウ糖液)を実験群と同様の手順でマウスに静脈内投与した。
核酸溶液を投与して72時間後に、ペントバルビタールを50mg/kgで前記マウスに腹腔内投与によって麻酔し、採血及び屠殺し、脳(大脳半球又は大脳皮質)を摘出した。脳組織からの全RNA抽出は、RNA抽出用試薬ISOGEN(ニッポン・ジーン社)を用いた。ISOGEN溶液中で摘出した脳組織を破砕した後、クロロホルムでRNA画分として分離した。その後に、核酸分離システムQuickGene RNA tissue kit SII(倉敷紡績社)で実施した。全RNAからのcDNA合成は、ReverTra Ace qPCR RT Kit(東洋紡社)を用い、THUNDERBIRD qPCR Mix(東洋紡社)による定量PCRを実施した。定量PCRには蛍光プローブ法を採用し、蛍光プローブにはマウスMalat1(Integrated DNA Technologies社)及びマウスGapdh(Thermo Fisher Scientific社)を用いた。定量PCRの遺伝子断片増幅反応条件は、上記THUNDERBIRD qPCR Mix(東洋紡社)のプロトコルに従った。マウスMalat1及びGapdh(内部標準遺伝子)の発現量を相対検量線によって算出し、相対発現レベルをMalat1/Gapdhとして計算した。一群あたり2匹のマウスの結果から、相対発現レベルの平均値を算出した。比較対象群の相対発現レベルの平均値を100%とした場合の実験群の相対発現レベルの平均値の割合を、相対的Malat1 ncRNA発現レベルとして算出した。
実施例9の結果を表2に示す。表中、投与量はASO(Malat1)の量を示す。
二本鎖番号1のDSPE-HDO及び二本鎖番号2のDOPE-HDOは、いずれも大脳皮質におけるMalat1ノンコーディングRNAの発現を抑制した。また、二本鎖番号3のDPPE-HDO、二本鎖番号4のPOPE-HDO、二本鎖番号5の18:1 (delta9-Trans) PE-HDO、二本鎖番号6の18:0-18:1 PE-HDO、二本鎖番号7の18:2 PE-HDOおよび二本鎖番号8のDPyPE-HDOは、いずれも大脳半球におけるMalat1ノンコーディングRNAの発現を抑制した。この結果から、ASOと、ホスファチジルエタノールアミン又はその類縁体が結合した相補鎖で構成される二本鎖の核酸複合体は、脳に送達され、そこでアンチセンス効果をもたらし得ることが立証された。
Claims (21)
- 第1核酸鎖と第2核酸鎖とを含む核酸複合体であって、
該第1核酸鎖は標的転写産物の少なくとも一部にハイブリダイズすることが可能な塩基配列を含み、かつ、標的転写産物に対してアンチセンス効果を有し、
該第2核酸鎖は該第1核酸鎖に相補的な塩基配列を含み、かつ、ホスファチジルエタノールアミン又はその類縁体と結合しており、
該第1核酸鎖は該第2核酸鎖にアニールしている前記核酸複合体。 - R1及びR2が、それぞれ独立に、C15~C19のアルキル基、又はC17のアルケニル基である、請求項2に記載の核酸複合体。
- 前記第2核酸鎖の5'末端に、前記リンカーを介してホスファチジルエタノールアミン又はその類縁体が結合している、請求項5に記載の核酸複合体。
(式中、nは0又は1である) - 前記第1核酸鎖が少なくとも4個の連続するデオキシリボヌクレオシドを含む、請求項1~6のいずれか一項に記載の核酸複合体。
- 前記第1核酸鎖がギャップマーである、請求項1~7のいずれか一項に記載の核酸複合体。
- 前記第2核酸鎖が前記第1核酸鎖中の少なくとも4個の連続するデオキシリボヌクレオシドに相補的な、少なくとも4個の連続するリボヌクレオシドを含む、請求項7又は8に記載の核酸複合体。
- 前記第1核酸鎖がミックスマーである、請求項1~6のいずれか一項に記載の核酸複合体。
- 前記第1核酸鎖が13~20塩基長である、請求項1~10のいずれか一項に記載の核酸複合体。
- 前記第2核酸鎖が天然リボヌクレオシドを含まない、請求項1~11のいずれか一項に記載の核酸複合体。
- 前記第2核酸鎖の核酸部分が修飾若しくは非修飾のヌクレオシド間結合により連結されたデオキシリボヌクレオシド及び/又は糖修飾ヌクレオシドから成る、請求項1~12のいずれか一項に記載の核酸複合体。
- 請求項1~13のいずれか一項に記載の核酸複合体を含む、中枢神経系において標的転写産物の発現又は編集を調節するための組成物。
- 中枢神経系疾患治療用である、請求項14に記載の組成物。
- 請求項1~13のいずれか一項に記載の核酸複合体を含む、中枢神経系薬剤送達用組成物。
- 前記中枢神経系が大脳皮質、大脳基底核、大脳白質、間脳、脳幹、小脳、及び脊髄からなる群から選択される、請求項14~16のいずれか一項に記載の組成物。
- 前記中枢神経系が前頭葉、側頭葉、海馬、海馬傍回、頭頂葉、後頭葉、線条体、淡蒼球、前障、視床、視床下核、中脳、黒質、橋、延髄、小脳皮質、小脳核、頸髄、胸髄及び腰髄からなる群から選択される、請求項14~16のいずれか一項に記載の組成物。
- 静脈内投与用又は皮下投与用である、請求項14~18のいずれか一項に記載の組成物。
- 1回の投与量中に5mg/kg以上の前記核酸複合体を含む、請求項14~19のいずれか一項に記載の組成物。
- 前記核酸複合体が血液脳関門(BBB)を通過する、請求項14~20のいずれか一項に記載の組成物。
Priority Applications (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US16/980,191 US11597928B2 (en) | 2018-03-14 | 2019-03-13 | Nucleic acid complex |
| EP19767189.4A EP3766972B1 (en) | 2018-03-14 | 2019-03-13 | Nucleic acid complex |
| JP2020506627A JP7394392B2 (ja) | 2018-03-14 | 2019-03-13 | 核酸複合体 |
| US18/163,506 US20230295628A1 (en) | 2018-03-14 | 2023-02-02 | Nucleic acid complex |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2018-047294 | 2018-03-14 | ||
| JP2018047294 | 2018-03-14 |
Related Child Applications (2)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| US16/980,191 A-371-Of-International US11597928B2 (en) | 2018-03-14 | 2019-03-13 | Nucleic acid complex |
| US18/163,506 Continuation US20230295628A1 (en) | 2018-03-14 | 2023-02-02 | Nucleic acid complex |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2019177061A1 true WO2019177061A1 (ja) | 2019-09-19 |
Family
ID=67907945
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2019/010392 Ceased WO2019177061A1 (ja) | 2018-03-14 | 2019-03-13 | 核酸複合体 |
Country Status (4)
| Country | Link |
|---|---|
| US (2) | US11597928B2 (ja) |
| EP (1) | EP3766972B1 (ja) |
| JP (1) | JP7394392B2 (ja) |
| WO (1) | WO2019177061A1 (ja) |
Cited By (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2022106695A1 (en) | 2020-11-23 | 2022-05-27 | Alpha Anomeric Sas | Nucleic acid duplexes |
| WO2024071099A1 (ja) | 2022-09-29 | 2024-04-04 | 国立大学法人東京医科歯科大学 | 5'-シクロプロピレン修飾を含む核酸分子 |
Families Citing this family (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| GB202215614D0 (en) | 2022-10-21 | 2022-12-07 | Proqr Therapeutics Ii Bv | Heteroduplex rna editing oligonucleotide complexes |
| US20240153803A1 (en) | 2022-11-07 | 2024-05-09 | Applied Materials, Inc. | Semiconductor process equipment |
| US12273051B2 (en) | 2022-12-14 | 2025-04-08 | Applied Materials, Inc. | Apparatus and method for contactless transportation of a carrier |
Citations (9)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2007143315A2 (en) | 2006-05-05 | 2007-12-13 | Isis Pharmaceutical, Inc. | Compounds and methods for modulating expression of pcsk9 |
| WO2008043753A2 (en) | 2006-10-09 | 2008-04-17 | Santaris Pharma A/S | Rna antagonist compounds for the modulation of pcsk9 |
| WO2008049085A1 (en) | 2006-10-18 | 2008-04-24 | Isis Pharmaceuticals, Inc. | Antisense compounds |
| WO2009123185A1 (ja) * | 2008-03-31 | 2009-10-08 | 独立行政法人産業技術総合研究所 | Rna干渉効果が高い2本鎖脂質修飾rna |
| WO2011008857A1 (en) * | 2009-07-14 | 2011-01-20 | Northeastern University | SiRNA PHOSPHOLIPID CONJUGATE |
| JP2016524588A (ja) * | 2013-05-30 | 2016-08-18 | 国立大学法人 東京医科歯科大学 | 治療用オリゴヌクレオチドを送達するための二本鎖剤 |
| JP2016526529A (ja) * | 2013-06-16 | 2016-09-05 | 国立大学法人 東京医科歯科大学 | エクソンスキッピング効果を有する二本鎖アンチセンス核酸 |
| WO2017053999A1 (en) | 2015-09-25 | 2017-03-30 | Ionis Pharmaceuticals, Inc. | Conjugated antisense compounds and their use |
| JP2018047294A (ja) | 2017-11-27 | 2018-03-29 | ファンプルワークス インコーポレイテッド | デバイスの領域情報に基づいてゲームを進行するサーバ及びデバイス |
Family Cites Families (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP3517610A4 (en) * | 2016-09-23 | 2020-05-27 | National University Corporation Tokyo Medical and Dental University | Blood-brain barrier permeable heteroduplex nucleic acid |
| JP7262816B2 (ja) * | 2018-03-22 | 2023-04-24 | 国立大学法人 東京医科歯科大学 | ヘテロ核酸のbbb通過脂質リガンド |
-
2019
- 2019-03-13 WO PCT/JP2019/010392 patent/WO2019177061A1/ja not_active Ceased
- 2019-03-13 EP EP19767189.4A patent/EP3766972B1/en active Active
- 2019-03-13 US US16/980,191 patent/US11597928B2/en active Active
- 2019-03-13 JP JP2020506627A patent/JP7394392B2/ja active Active
-
2023
- 2023-02-02 US US18/163,506 patent/US20230295628A1/en active Pending
Patent Citations (9)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2007143315A2 (en) | 2006-05-05 | 2007-12-13 | Isis Pharmaceutical, Inc. | Compounds and methods for modulating expression of pcsk9 |
| WO2008043753A2 (en) | 2006-10-09 | 2008-04-17 | Santaris Pharma A/S | Rna antagonist compounds for the modulation of pcsk9 |
| WO2008049085A1 (en) | 2006-10-18 | 2008-04-24 | Isis Pharmaceuticals, Inc. | Antisense compounds |
| WO2009123185A1 (ja) * | 2008-03-31 | 2009-10-08 | 独立行政法人産業技術総合研究所 | Rna干渉効果が高い2本鎖脂質修飾rna |
| WO2011008857A1 (en) * | 2009-07-14 | 2011-01-20 | Northeastern University | SiRNA PHOSPHOLIPID CONJUGATE |
| JP2016524588A (ja) * | 2013-05-30 | 2016-08-18 | 国立大学法人 東京医科歯科大学 | 治療用オリゴヌクレオチドを送達するための二本鎖剤 |
| JP2016526529A (ja) * | 2013-06-16 | 2016-09-05 | 国立大学法人 東京医科歯科大学 | エクソンスキッピング効果を有する二本鎖アンチセンス核酸 |
| WO2017053999A1 (en) | 2015-09-25 | 2017-03-30 | Ionis Pharmaceuticals, Inc. | Conjugated antisense compounds and their use |
| JP2018047294A (ja) | 2017-11-27 | 2018-03-29 | ファンプルワークス インコーポレイテッド | デバイスの領域情報に基づいてゲームを進行するサーバ及びデバイス |
Non-Patent Citations (2)
| Title |
|---|
| "GenBank", Database accession no. NR_002847 |
| See also references of EP3766972A4 |
Cited By (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2022106695A1 (en) | 2020-11-23 | 2022-05-27 | Alpha Anomeric Sas | Nucleic acid duplexes |
| WO2024071099A1 (ja) | 2022-09-29 | 2024-04-04 | 国立大学法人東京医科歯科大学 | 5'-シクロプロピレン修飾を含む核酸分子 |
Also Published As
| Publication number | Publication date |
|---|---|
| JP7394392B2 (ja) | 2023-12-08 |
| EP3766972B1 (en) | 2025-04-23 |
| JPWO2019177061A1 (ja) | 2021-03-11 |
| EP3766972A4 (en) | 2022-03-02 |
| US20210024929A1 (en) | 2021-01-28 |
| US20230295628A1 (en) | 2023-09-21 |
| US11597928B2 (en) | 2023-03-07 |
| EP3766972A1 (en) | 2021-01-20 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP7687682B2 (ja) | 筋疾患治療用医薬組成物 | |
| JP7554488B2 (ja) | 血液脳関門通過型ヘテロ2本鎖核酸 | |
| JP7669602B2 (ja) | ヘテロ核酸のbbb通過脂質リガンド | |
| JP7394392B2 (ja) | 核酸複合体 | |
| WO2018062510A1 (ja) | オーバーハングを有する二本鎖核酸複合体 | |
| WO2023022229A1 (ja) | モルホリノ核酸を含む修飾ヘテロ核酸 | |
| US12305169B2 (en) | BBB-crossing lipid ligand of hetero nucleic acid | |
| JP7737667B2 (ja) | 核酸複合体 | |
| WO2021187392A1 (ja) | モルホリノ核酸を含むヘテロ核酸 | |
| WO2024071099A1 (ja) | 5'-シクロプロピレン修飾を含む核酸分子 | |
| JP6934695B2 (ja) | 核酸医薬とその使用 | |
| WO2021187540A1 (ja) | Scn1a遺伝子の発現及び/又は機能調節剤 | |
| WO2024005156A1 (ja) | 核酸医薬の毒性軽減剤 | |
| WO2022250050A1 (ja) | scpBNA又はAmNAを含むヘテロ核酸 | |
| WO2024071362A1 (ja) | 神経系送達促進剤 | |
| WO2024106545A1 (ja) | 髄腔内投与のためのヘテロ核酸 | |
| WO2023042876A1 (ja) | 2'-修飾ヌクレオシドを含むヘテロ核酸 | |
| CN117412773A (zh) | 配体结合核酸复合物 |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 19767189 Country of ref document: EP Kind code of ref document: A1 |
|
| ENP | Entry into the national phase |
Ref document number: 2020506627 Country of ref document: JP Kind code of ref document: A |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| ENP | Entry into the national phase |
Ref document number: 2019767189 Country of ref document: EP Effective date: 20201014 |
|
| WWG | Wipo information: grant in national office |
Ref document number: 2019767189 Country of ref document: EP |