CN111377984B - Compounds and conjugates, methods of making and uses thereof - Google Patents

Compounds and conjugates, methods of making and uses thereof Download PDF

Info

Publication number
CN111377984B
CN111377984B CN201811637314.7A CN201811637314A CN111377984B CN 111377984 B CN111377984 B CN 111377984B CN 201811637314 A CN201811637314 A CN 201811637314A CN 111377984 B CN111377984 B CN 111377984B
Authority
CN
China
Prior art keywords
nucleotide
group
conjugate
sirna
formula
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.)
Active
Application number
CN201811637314.7A
Other languages
Chinese (zh)
Other versions
CN111377984A (en
Inventor
张鸿雁
杨志伟
曹力强
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Suzhou Ruibo Biotechnology Co ltd
Original Assignee
Suzhou Ruibo Biotechnology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Suzhou Ruibo Biotechnology Co ltd filed Critical Suzhou Ruibo Biotechnology Co ltd
Priority to CN201811637314.7A priority Critical patent/CN111377984B/en
Publication of CN111377984A publication Critical patent/CN111377984A/en
Application granted granted Critical
Publication of CN111377984B publication Critical patent/CN111377984B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/02Acyclic radicals, not substituted by cyclic structures
    • C07H15/04Acyclic radicals, not substituted by cyclic structures attached to an oxygen atom of the saccharide radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal 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
    • A61K47/51Medicinal 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
    • A61K47/54Medicinal 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal 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
    • A61K47/51Medicinal 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
    • A61K47/54Medicinal 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
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal 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
    • A61K47/51Medicinal 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
    • A61K47/54Medicinal 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
    • A61K47/545Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal 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
    • A61K47/51Medicinal 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
    • A61K47/54Medicinal 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
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H1/00Processes for the preparation of sugar derivatives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

A compound capable of forming a conjugate with an oligonucleotide, the compound having a structure as shown in formula (101). The present disclosure also provides corresponding oligonucleotide conjugates. The oligonucleotide conjugate disclosed by the disclosure can specifically target liver cells, so that the in-vivo delivery problem of an oligonucleotide drug is effectively solved, the toxicity is low, and the delivered oligonucleotide has high stability.

Description

Compounds and conjugates, methods of making and uses thereof
Technical Field
The present application relates to compounds and conjugates for drug delivery, and methods for their preparation and use.
Background
Delivery systems are one of the key technologies in the development of small nucleic acid drugs, and the most widely studied class of delivery systems for small nucleic acid delivery systems worldwide is currently targeted conjugated delivery technology.
Disclosure of Invention
In one embodiment, the present disclosure provides a conjugate molecule having a structure represented by formula (101):
wherein:
n 1 is an integer selected from 1-2;
each n 2 Independently an integer selected from 1-2;
m 1 is an integer selected from 1-6;
R 1 is a group capable of binding to an active agent via a covalent bond; the method comprises the steps of carrying out a first treatment on the surface of the
Each R 2 Each independently selected from H, C 1 -C 10 Alkyl, C 1 -C 10 Haloalkyl or C 1 -C 10 An alkoxy group;
each L 1 Is a linear alkylene group of 1 to 70 carbon atoms in length, wherein one or more carbon atoms are optionally replaced by one or more selected from the group consisting of: c (O), NH, O, S, CH = N, S (O) 2 、C 2 -C 10 Alkenylene, C 2 -C 10 Alkynylene, C 6 -C 10 Arylene group, C 3 -C 18 Heterocyclylene and C 5 -C 10 Heteroarylene; and wherein L 1 Optionally substituted with any one or more of the group consisting of: c (C) 1 -C 10 Alkyl, C 6 -C 10 Aryl, C 5 -C 10 Heteroaryl, C 1 -C 10 Haloalkyl, -OC 1 -C 10 Alkyl, -OC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-OH, -OC 1 -C 10 Haloalkyl, -SC 1 -C 10 Alkyl, -SC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-SH, -SC 1 -C 10 Haloalkyl, halogen substituent, -OH, -SH, -NH 2 、-C 1 -C 10 alkyl-NH 2 、-N(C 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -NH (C) 1 -C 10 Alkyl), cyano, nitro, -CO 2 H、-C(O)O(C 1 -C 10 Alkyl), -CON (C) 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -CONH (C) 1 -C 10 Alkyl), -CONH 2 ,-NHC(O)(C 1 -C 10 Alkyl), -NHC (O) (phenyl), -N (C) 1 -C 10 Alkyl) C (O) (C 1 -C 10 Alkyl), -N (C) 1 -C 10 Alkyl) C (O) (phenyl), -C (O) C 1 -C 10 Alkyl, -C (O) C 1 -C 10 Alkylphenyl, -C (O) C 1 -C 10 Haloalkyl, -OC (O) C 1 -C 10 Alkyl, -SO 2 (C 1 -C 10 Alkyl), -SO 2 (phenyl) -SO 2 (C 1 -C 10 HaloalkanesRadical) -SO 2 NH 2 、-SO 2 NH(C 1 -C 10 Alkyl), -SO 2 NH (phenyl) -NHSO 2 (C 1 -C 10 Alkyl), -NHSO 2 (phenyl) and-NHSO 2 (C 1 -C 10 A haloalkyl group);
each S 1 Independently M 1 Wherein any active hydroxyl groups, if any, are protected by hydroxyl protecting groups;
each M 1 Independently selected from ligands capable of binding to cell surface receptors.
In some embodiments, each L 1 A linked combination of one or more independently selected from the groups of formulae A1-a 26:
wherein each j1 is independently an integer from 1 to 20; each j2 is independently an integer from 1 to 20;
each R' is independently C1-C10 alkyl;
each Ra is independently selected from one of the groups of formulae a27-a 45:
each Rb is independently C1-C10 alkyl;
represents the site where the groups are linked by covalent bonds;
in one embodiment, the present disclosure provides a conjugate having a structure represented by formula (201):
wherein:
n 1 is an integer selected from 1-2;
each n 2 Independently selected from integers from 1-2;
m 1 is an integer selected from 1-6;
each R 2 Each independently H, C 1 -C 10 Alkyl, C 1 -C 10 Haloalkyl or C 1 -C 10 An alkoxy group;
R 6 is an active drug;
R 5 is a linear alkylene group of 1 to 20 carbon atoms in length, wherein one or more carbon atoms are optionally replaced by one or more selected from the group consisting of: c (O), NH, O, S, CH = N, S (O) 2 、C 2 -C 10 Alkenylene, C 2 -C 10 Alkynylene, C 6 -C 10 Arylene group, C 3 -C 18 Heterocyclylene and C 5 -C 10 Heteroarylene; and wherein R is 5 Optionally having substituents of any one or more of the group consisting of: c (C) 1 -C 10 Alkyl, C 6 -C 10 Aryl, C 5 -C 10 Heteroaryl, C 1 -C 10 Haloalkyl, -OC 1 -C 10 Alkyl, -OC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-OH, -OC 1 -C 10 Haloalkyl, -SC 1 -C 10 Alkyl, -SC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-SH, -SC 1 -C 10 Haloalkyl, halogen substituent, -OH, -SH, -NH 2 、-C 1 -C 10 alkyl-NH 2 、-N(C 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -NH (C) 1 -C 10 Alkyl), cyano, nitro, -CO 2 H、-C(O)O(C 1 -C 10 Alkyl group),-CON(C 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -CONH (C) 1 -C 10 Alkyl), -CONH 2 ,-NHC(O)(C 1 -C 10 Alkyl), -NHC (O) (phenyl), -N (C) 1 -C 10 Alkyl) C (O) (C 1 -C 10 Alkyl), -N (C) 1 -C 10 Alkyl) C (O) (phenyl), -C (O) C 1 -C 10 Alkyl, -C (O) C 1 -C 10 Alkylphenyl, -C (O) C 1 -C 10 Haloalkyl, -OC (O) C 1 -C 10 Alkyl, -SO 2 (C 1 -C 10 Alkyl), -SO 2 (phenyl) -SO 2 (C 1 -C 10 Haloalkyl) -SO 2 NH 2 、-SO 2 NH(C 1 -C 10 Alkyl), -SO 2 NH (phenyl) -NHSO 2 (C 1 -C 10 Alkyl), -NHSO 2 (phenyl) and-NHSO 2 (C 1 -C 10 A haloalkyl group);
each L 1 Is a linear alkylene group of 1 to 70 carbon atoms in length, wherein one or more carbon atoms are optionally replaced by one or more selected from the group consisting of: c (O), NH, O, S, CH = N, S (O) 2 、C 2 -C 10 Alkenylene, C 2 -C 10 Alkynylene, C 6 -C 10 Arylene group, C 3 -C 18 Heterocyclylene and C 5 -C 10 Heteroarylene; and wherein L 1 Optionally substituted with any one or more of the group consisting of: c (C) 1 -C 10 Alkyl, C 6 -C 10 Aryl, C 5 -C 10 Heteroaryl, C 1 -C 10 Haloalkyl, -OC 1 -C 10 Alkyl, -OC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-OH, -OC 1 -C 10 Haloalkyl, -SC 1 -C 10 Alkyl, -SC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-SH, -SC 1 -C 10 Haloalkyl, halogen substituent, -OH, -SH, -NH 2 、-C 1 -C 10 alkyl-NH 2 、-N(C 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -NH (C) 1 -C 10 Alkyl), cyano, nitro, -CO 2 H、-C(O)O(C 1 -C 10 Alkyl), -CON (C) 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -CONH (C) 1 -C 10 Alkyl), -CONH 2 ,-NHC(O)(C 1 -C 10 Alkyl), -NHC (O) (phenyl), -N (C) 1 -C 10 Alkyl) C (O) (C 1 -C 10 Alkyl), -N (C) 1 -C 10 Alkyl) C (O) (phenyl), -C (O) C 1 -C 10 Alkyl, -C (O) C 1 -C 10 Alkylphenyl, -C (O) C 1 -C 10 Haloalkyl, -OC (O) C 1 -C 10 Alkyl, -SO 2 (C 1 -C 10 Alkyl), -SO 2 (phenyl) -SO 2 (C 1 -C 10 Haloalkyl) -SO 2 NH 2 、-SO 2 NH(C 1 -C 10 Alkyl), -SO 2 NH (phenyl) -NHSO 2 (C 1 -C 10 Alkyl), -NHSO 2 (phenyl) and-NHSO 2 (C 1 -C 10 A haloalkyl group);
each M 1 Selected from ligands capable of binding to cell surface receptors.
In some embodiments, each L 1 A linked combination of one or more independently selected from the groups of formulae A1-a 26:
wherein each j1 is independently an integer from 1 to 20; each j2 is independently an integer from 1 to 20;
Each R' is independently C1-C10 alkyl;
each Ra is independently selected from one of the groups of formulae a27-a 45:
each Rb is independently C1-C10 alkyl;
represents the site where the groups are linked by covalent bonds;
in some embodiments, there is provided the use of a compound of the present disclosure in the manufacture of a medicament for the treatment and/or prevention of a pathological condition or disease caused by expression of a particular gene.
In some embodiments, the present disclosure provides a method of treating a pathological condition or disease caused by expression of a particular gene in hepatocytes, comprising providing to a subject an effective dose of a conjugate of the present disclosure.
In some embodiments, the present disclosure provides a method of inhibiting expression of a particular gene in a hepatocyte, the method comprising contacting with a conjugate of the present disclosure.
In some embodiments, the present disclosure provides a kit comprising a conjugate of the present disclosure.
Additional features and advantages of the present disclosure will be set forth in the detailed description which follows.
Incorporated by reference
All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
Detailed Description
The following describes specific embodiments of the present disclosure in detail. It should be understood that the detailed description and specific examples, while indicating and illustrating the disclosure, are not intended to limit the disclosure.
Definition of the definition
In the above and below, capital C, G, U, A indicates the base composition of nucleotides unless otherwise specified; the lower case letter m indicates that the adjacent nucleotide to the left of the letter m is a 2' -methoxy modified nucleotide; the lower case letter f indicates that the adjacent nucleotide to the left of the letter f is a 2' -fluoro modified nucleotide; the lower case letter s indicates that phosphorothioate linkages are between two nucleotides adjacent to the letter s; p1 represents that one nucleotide adjacent to the right of P1 is a nucleotide modified with 5 '-phosphate or a 5' -phosphate analogue, particularly a nucleotide modified with vinyl phosphate (denoted as VP in the following examples), a nucleotide modified with 5 '-phosphate (denoted as P in the following examples), or a nucleotide modified with 5' -phosphorothioate (denoted as Ps in the following examples).
In the present context, the terms "complementary" or "reverse complementary" are used interchangeably and have the meaning well known to those skilled in the art, i.e., in a double stranded nucleic acid molecule, the bases of one strand pair with the bases on the other strand in a complementary manner. In DNA, the purine base adenine (a) is always paired with the pyrimidine base thymine (T) (or uracil (U) in RNA); the purine base guanine (G) is always paired with the pyrimidine base cytosine (C). Each base pair includes a purine and a pyrimidine. When adenine on one strand always pairs with thymine (or uracil) on the other strand, and guanine always pairs with cytosine, the two strands are considered complementary to each other, and the sequence of the strand may be deduced from the sequence of its complementary strand. Accordingly, "mismatch" means in the art that bases at corresponding positions do not exist in complementary pairs in a double-stranded nucleic acid.
In the above and in the following, essentially reverse complement, unless otherwise indicated, means that there are no more than 3 base mismatches between the two nucleotide sequences involved; substantially complete reverse complement means that there is no more than 1 base mismatch between the two nucleotide sequences; complete complementarity refers to the absence of a base mismatch between two nucleotide sequences. In the above and below, the nucleotide difference between one nucleotide sequence and another nucleotide sequence means that the base type of the nucleotide at the same position is changed as compared with the former, for example, when one nucleotide base is A in the latter, when the corresponding nucleotide base at the same position in the former is U, C, G or T, it is determined that the nucleotide difference exists between the two nucleotide sequences at the position. In some embodiments, a nucleotide difference is also considered to occur at an original position when the nucleotide is replaced with an abasic nucleotide or nucleotide analog.
In the above and in the following, particularly when describing the preparation method of the conjugate molecule of the present disclosure or the preparation method of the siRNA conjugate, the nucleoside monomer (nucleoside monomer) means, unless otherwise specified, that the "unmodified or modified RNA phosphoramidite (unmodified or modified RNA phosphoramidite)" is used for so-called solid phase phosphoramidite synthesis, respectively, which is a well-known method in the art for synthesizing RNA, depending on the RNA sequence to be prepared. RNA phosphoramidite is also referred to herein as nucleoside phosphoramidite (nucleoside phosphoramidites). Nucleoside monomers used in the present disclosure are commercially available unless otherwise indicated.
As used herein, a short dash ("-") that is not between two letters or between two symbols is used to indicate the position of a substituent attachment point. For example: -C 1 -C 10 alkyl-NH 2 Through C 1 -C 10 Alkyl groups.
As used herein, "optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "Optionally substitutedThe (optinally substituted) alkyl group "of (a) includes" alkyl group "and" substituted alkyl group "defined below. Those skilled in the art will appreciate that forIn any group comprising one or more substituents, these groups are not intended to introduce any substitution or pattern of substitution that is sterically impractical, synthetically infeasible, and/or inherently unstable.
As used herein, "alkyl" refers to straight and branched chains having the indicated number of carbon atoms, typically 1 to 20 carbon atoms, for example 1 to 10 carbon atoms, such as 1 to 8 or 1 to 6 carbon atoms. For example, C 1 -C 6 The alkyl groups comprise straight and branched alkyl groups of 1 to 6 carbon atoms. When naming alkyl residues having a specific number of carbons, it is intended to cover all branched and straight chain forms having that number of carbons; thus, for example, "butyl" is meant to include n-butyl, sec-butyl, isobutyl, and tert-butyl; "propyl" includes n-propyl and isopropyl. Alkylene is a subset of alkyl groups, referring to residues that are identical to alkyl groups, but have two points of attachment.
As used herein, "alkenyl" refers to an unsaturated branched or straight chain alkyl group having at least one carbon-carbon double bond obtained by removing one hydrogen molecule from an adjacent carbon atom of the parent alkyl group. The group may be in the cis or trans configuration of the double bond. Typical alkenyl groups include, but are not limited to: vinyl; propenyl, such as prop-1-en-1-yl, prop-1-en-2-yl, prop-2-en-1-yl (i.e., allyl), prop-2-en-2-yl; butenyl, such as but-1-en-1-yl, but-1-en-2-yl, 2-methylpropan-1-en-1-yl, but-2-en-2-yl, but-1, 3-dien-1-yl, but-1, 3-dien-2-yl, and the like. In certain embodiments, alkenyl groups have 2 to 20 carbon atoms, and in other embodiments 2 to 10, 2 to 8, or 2 to 6 carbon atoms. Alkenylene is a subset of alkenyl groups and refers to residues that are identical to alkenyl groups but have two points of attachment.
As used herein, "alkynyl" refers to an unsaturated branched or straight chain alkyl group having at least one carbon-carbon triple bond obtained by removing two hydrogen molecules from adjacent carbon atoms of the parent alkyl group. Typical alkynyl groups include, but are not limited to: ethynyl; propynyl, such as prop-1-yn-1-yl, prop-2-yn-1-yl; butynyl, such as but-1-yn-1-yl, but-1-yn-3-yl, but-3-yn-1-yl, and the like. In certain embodiments, alkynyl groups have 2 to 20 carbon atoms, while in other embodiments, 2 to 10, 2 to 8, or 2 to 6 carbon atoms. Alkynylene is a subset of alkynyl groups and refers to residues that are identical to alkynyl groups but have two points of attachment.
As used herein, "alkoxy" refers to an alkyl group of the specified number of carbon atoms attached through an oxygen bridge, e.g., methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, pentyloxy, 2-pentyloxy, isopentyloxy, neopentyloxy, hexyloxy, 2-hexyloxy, 3-methylpentyloxy, and the like. Alkoxy groups typically have 1 to 10, 1 to 8, 1 to 6, or 1 to 4 carbon atoms attached through an oxygen bridge.
As used herein, "aryl" refers to a group derived from an aromatic mono-or polycyclic hydrocarbon ring system formed by removal of a hydrogen atom from a ring carbon atom. The aromatic mono-or polycyclic hydrocarbon ring system contains only hydrogen and carbon of 6 to 18 carbon atoms, wherein at least one ring of the ring system is fully unsaturated, i.e. it comprises a cyclic, delocalized (4n+2) pi-electron system according to Huckel theory. Aryl groups include, but are not limited to, phenyl, fluorenyl, and naphthyl groups. Arylene is a subset of aryl groups and refers to residues that are identical to aryl groups but have two points of attachment.
As used herein, "cycloalkyl" refers to a non-aromatic carbocyclic ring, typically having 3 to 7 cyclic carbon atoms. The ring may be saturated or have one or more carbon-carbon double bonds. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl and cyclohexenyl, as well as bridged and caged ring groups such as norbornane (norbornane).
As used herein, "halogen substituent" or "halo" refers to fluoro, chloro, bromo, and iodo, and the term "halogen" includes fluoro, chloro, bromo, and iodo.
As used herein, "haloalkyl" refers to an alkyl group as defined above wherein a specified number of carbon atoms are replaced with one or more up to the maximum allowable number of halogen atoms. Examples of haloalkyl include, but are not limited to, trifluoromethyl, difluoromethyl, 2-fluoroethyl and pentafluoroethyl.
"heterocyclyl" means a stable 3-to 18-membered non-aromatic ring radical containing 2-12 carbon atoms and 1-6 heteroatoms selected from nitrogen, oxygen and sulfur. Unless otherwise indicated in the specification, heterocyclyl is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. The heteroatoms in the heterocyclic radical may optionally be oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heterocyclyl groups are partially saturated or fully saturated. The heterocyclyl may be attached to the remainder of the molecule through any atom of the ring. Examples of such heterocyclyl groups include, but are not limited to: dioxanyl, thienyl [1,3] dithioyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxapiperazinyl, 2-oxapiperidinyl, 2-oxapyrimidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuranyl, trithioyl, tetrahydropyranyl, thiomorpholinyl, 1-oxathiomorpholinyl, and 1, 1-dioxathiomorpholinyl.
"heteroaryl" refers to groups derived from 3-to 18-membered aromatic ring radicals containing 2 to 17 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur. As used herein, heteroaryl groups may be monocyclic, bicyclic, tricyclic or tetracyclic systems, wherein at least one ring of the ring system is fully unsaturated, i.e. comprises a cyclic delocalized (4n+2) pi-electron system according to the huckel theory. Heteroaryl groups include fused or bridged ring systems. The heteroatoms in the heteroaryl group are optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. Heteroaryl groups are attached to the remainder of the molecule through any atom in the ring. Examples of heteroaryl groups include, but are not limited to: azetidinyl, acridinyl, benzimidazolyl, benzindolyl, 1, 3-benzodioxazolyl, benzofuranyl, benzoxazolyl, benzo [ d ] thiazolyl, benzothiadiazolyl, benzo [ b ] [1,4] dioxazolyl, benzo [ b ] [1,4] oxazolyl, 1, 4-benzodioxazolyl, benzonaphthofuranyl, benzodiazolyl, benzodioxaphenyl, benzopyranyl, benzopyronyl, benzofuranyl, benzothienyl, benzothiophene [3,2-d ] pyrimidinyl, benzotriazolyl, benzo [4,6] imidazo [1,2-a ] pyridinyl, carbazolyl, cinnolinyl, cyclopentyl [ d ] pyrimidinyl, 6, 7-dihydro-5H-cyclopentyl [4,5] thiophene [2,3-d ] pyrimidinyl, 5, 6-dihydrobenzo [ H ] quinazolinyl 5, 6-dihydrobenzo [ H ] Xin Nuolin yl, 6, 7-dihydro-5H-benzo [6,7] cyclohepta [1,2-c ] pyridazinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, furan [3,2-c ] pyridinyl, 5,6,7,8,9, 10-hexahydrocyclohepta [ d ] pyrimidinyl, 5,6,7,8,9, 10-hexahydrocyclooctanoic acid [ d ] pyridazinyl, 5,6,7,8,9, 10-hexahydrocyclooctanoic acid [ d ] pyridinyl, isothiazolyl, indazolyl, imidazolyl, indolyl, isoindolyl, indolizinyl, isooxazolyl, 5, 8-methyl-5, 6,7, 8-tetrahydroquinazolinyl, naphthyridine, 1, 6-naphthyridonyl, oxadiazolyl, 2-oxazinyl, oxazolyl, 6a, 10-dihydrobenzoquinazolinyl, 10-a, 10H-benzoquinazolinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthaloyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyrazolo [3,4-d ] pyrimidinyl, pyridinyl, pyrido [3,2-d ] pyrimidinyl, pyrido [3,4-d ] pyrimidinyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, 5,6,7, 8-tetrahydroquinazolinyl, 5,6,7, 8-tetrahydrobenzo [4,5] thiophen [2,3-d ] pyrimidinyl, 6,7,8,9 tetrahydro-5H-cyclohepta [4,5] thiophen [2,3-d ] pyrimidinyl, 5,6,7, 8-tetrahydropyrido [4,5-c ] pyridazinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, thiophen [2,3-d ] pyrimidinyl, 3,2-d ] thiophenyl, 2,3-d ] thienyl and naphthyridinyl.
Various hydroxyl protecting groups may be used in the present disclosure. In general, the protecting group renders the chemical functionality insensitive to the particular reaction conditions and may be appended and removed from that functionality in the molecule without substantially damaging the remainder of the molecule. Representative hydroxyl protecting groups are disclosed in Beaucage et al, tetrahedron 1992,48,2223-2311, and Greeneand Wuts, protective Groups in Organic Synthesis, chapter 2,2d ed,John Wiley&Sons,New York,1991, which are incorporated herein by reference in their entirety. In some embodiments, the protecting group is stable under alkaline conditions, but can be removed under acidic conditions. In some embodiments, non-exclusive examples of hydroxyl protecting groups that may be used herein include Dimethoxytrityl (DMT), monomethoxytrityl, 9-phenylxanthin-9-yl (Pixyl), and 9- (p-methoxyphenyl) xanthin-9-yl (Mox). In some embodiments, non-exclusive examples of hydroxyl protecting groups that may be used herein include Tr (trityl), MMTr (4-methoxytrityl), DMTr (4, 4 '-dimethoxytrityl), and TMTr (4, 4',4 "-trimethoxytrityl).
The term "subject" as used herein refers to any animal, such as a mammal or a pouched animal. Subjects of the present disclosure include, but are not limited to, humans, non-human primates (e.g., rhesus monkeys or other types of macaques), mice, pigs, horses, donkeys, cattle, sheep, rats, and any variety of poultry.
As used herein, "treatment," "treatment" alleviation, or "improvement" may be used interchangeably herein. These terms refer to methods of achieving a beneficial or desired result, including but not limited to therapeutic benefit. By "therapeutic benefit" is meant eradication or amelioration of the underlying disorder being treated. In addition, therapeutic benefit is obtained by eradicating or ameliorating one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, although the subject may still be afflicted with the underlying disorder.
As used herein, "prevent" and "prevent" are used interchangeably. These terms refer to methods of achieving a beneficial or desired result, including but not limited to prophylactic benefit. To obtain a "prophylactic benefit," the conjugate or composition may be administered to a subject at risk of suffering from a particular disease, or to a subject reporting one or more pathological symptoms of the disease, even though a diagnosis of the disease may not have been made.
Conjugation molecules
In one aspect, a conjugate molecule for delivering an active agent or active drug is disclosed. In some embodiments, the conjugate molecules of the present disclosure facilitate tissue-specific targeting. In some embodiments, the conjugate molecules of the present disclosure bind to a cell surface receptor. For this purpose, any cell surface receptor or biomarker or portion thereof is considered suitable. In some embodiments, the conjugate molecules of the present disclosure specifically bind to specific receptors of a particular tissue, thereby achieving tissue-specific targeting. In some embodiments, the conjugate molecules of the present disclosure are directed specifically to hepatocyte surface receptors, such that the patent is directed to liver tissue. In some embodiments, the conjugate molecules of the present disclosure are directed specifically to cell surface receptors specific to hepatocytes. In some embodiments, the conjugate molecules of the present disclosure are directed specifically to the asialoglycoprotein receptor (asialoglycoprotein receptors, ASGPR) of the liver surface.
As used herein, "active agent" and "active drug" are used interchangeably, and refer to a molecule capable of being delivered by a conjugated molecule of the present disclosure. In some embodiments, the active agent is an agent capable of being delivered to hepatocytes. Such reagents are well known to those skilled in the art and include, but are not limited to, functional nucleotides, such as functional oligonucleotides, particularly those disclosed herein.
In some embodiments, the present disclosure provides a conjugate molecule having a structure represented by formula (101):
wherein:
n 1 is an integer selected from 1-2; each n 2 Independently an integer selected from 1-2;
m 1 is an integer selected from 1-6;
R 1 is a group capable of binding to an active agent via a covalent bond;
each L 1 Is a linear alkylene group of 1 to 70 carbon atoms in length, wherein one or more carbon atoms are optionally replaced by one or more selected from the group consisting of: c (O), NH, O, S, CH = N, S (O) 2 、C 2 -C 10 Alkenylene, C 2 -C 10 Alkynylene, C 6 -C 10 Arylene group, C 3 -C 18 Heterocyclylene and C 5 -C 10 Heteroarylene; and wherein L 1 Optionally substituted with any one or more of the group consisting of: c (C) 1 -C 10 Alkyl, C 6 -C 10 Aryl, C 5 -C 10 Heteroaryl, C 1 -C 10 Haloalkyl, -OC 1 -C 10 Alkyl, -OC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-OH, -OC 1 -C 10 Haloalkyl, -SC 1 -C 10 Alkyl, -SC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-SH, -SC 1 -C 10 Haloalkyl, halogen substituent, -OH, -SH, -NH 2 、-C 1 -C 10 alkyl-NH 2 、-N(C 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -NH (C) 1 -C 10 Alkyl), cyano, nitro, -CO 2 H、-C(O)O(C 1 -C 10 Alkyl), -CON (C) 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -CONH (C) 1 -C 10 Alkyl), -CONH 2 ,-NHC(O)(C 1 -C 10 Alkyl), -NHC (O) (phenyl), -N (C) 1 -C 10 Alkyl) C (O) (C 1 -C 10 Alkyl), -N (C) 1 -C 10 Alkyl) C (O) (phenyl), -C (O) C 1 -C 10 Alkyl, -C (O) C 1 -C 10 Alkylphenyl, -C (O) C 1 -C 10 Haloalkyl, -OC (O) C 1 -C 10 Alkyl, -SO 2 (C 1 -C 10 Alkyl), -SO 2 (phenyl) a radical,-SO 2 (C 1 -C 10 Haloalkyl) -SO 2 NH 2 、-SO 2 NH(C 1 -C 10 Alkyl), -SO 2 NH (phenyl) -NHSO 2 (C 1 -C 10 Alkyl), -NHSO 2 (phenyl) and-NHSO 2 (C 1 -C 10 A haloalkyl group);
each S 1 Independently M 1 Wherein any active hydroxyl groups, if any, are protected by hydroxyl protecting groups;
each M 1 Independently selected from ligands capable of binding to cell surface receptors.
In some embodiments, m 1 May be an integer of 1 to 6, thereby ensuring S in the conjugate molecule 1 The number of groups is at least 2; in one embodiment, m 1 Is an integer selected from 2-6 such that M in the oligonucleotide conjugate formed from the conjugate molecule 1 The number of ligands is at least 3, so that M 1 The ligand binds more readily to hepatic surface asialoglycoprotein receptors, thereby facilitating entry of the conjugate into cells by endocytosis. Experiments show that when M 1 When the number of the ligands is more than 3, M 1 The increased ease of ligand binding to hepatic surface asialoglycoprotein receptor is not significant and thus, in some embodiments, m is a combination of ease of synthesis, structural/process costs and delivery efficiency 1 2.
In some embodiments, n 1 Is an integer selected from 1-2, each n 2 Independently an integer selected from 1-2, can be such that in an oligonucleotide conjugate formed from the conjugate molecule, a plurality of M 1 Spatial position between ligands is suitable for M 1 Binding of ligand to hepatic surface asialoglycoprotein receptor in order to make the conjugate molecule provided by the present disclosure simpler, easier to synthesize and/or lower cost, n in one embodiment according to the present disclosure 1 =n 2 =2。
As one of ordinary skill in the art will appreciate, when each R 2 Each independently selected from H, C 1 -C 10 Alkyl, C 1 -C 10 Haloalkyl or C 1 -C 10 The alkoxy groups, without altering the nature of the conjugate molecules provided by the present disclosure, may achieve the objects of the present disclosure. In some embodiments, each R 2 Independently selected from H, methyl or ethyl. In some embodiments, each R 2 All are H.
R 1 Is a group capable of being delivered by the conjugate molecules of the present disclosure and bound to an active drug (also referred to as an active agent). In some embodiments, R 1 To be able to bind to a group of an oligonucleotide, the oligonucleotide will be delivered by a conjugate molecule of the present disclosure. In some embodiments, R 1 Is a group capable of binding to an oligonucleotide by a covalent bond. In some embodiments, R 1 Is a group capable of binding to an oligonucleotide through a phosphodiester bond. In some embodiments, R 1 Is selected to achieve ligation to N on a nitrogen-containing backbone and to provide a suitable reaction site for synthesis of the oligonucleotide conjugate. In the context of the present disclosure, "nitrogen-containing backbone" means that R is attached 2 A chain structure in which carbon atoms of (a) and N are connected to each other. In some embodiments, R 1 May be a group capable of being attached to an N atom on a nitrogen-containing backbone in an appropriate manner. In some embodiments, R 1 The group contains a linking site for linking to the N on the nitrogen-containing backbone and any functional group that may be reacted to conjugate to the oligonucleotide via a phosphodiester linkage.
In some embodiments, R 1 And a 2 nd functional group, wherein the 2 nd functional group can form a covalent bond with a hydroxyl group or an amino group, or is a solid phase carrier capable of being connected through a covalent bond with a hydroxyl group or an amino group; in yet another embodiment, the 1 st functional group is a phosphoramidite, hydroxyl, or protected hydroxyl, in some embodiments, the 2 nd functional group is a phosphoramidite, carboxyl, or carboxylate; in some embodiments, the carboxylate is a carboxylate with a metal cation, an ammonium carboxylate salt, a tertiary amine carboxylate salt, or a quaternary ammonium carboxylate salt; in some embodiments, the carboxylate is triethylamine carboxylate or N, N-diiso Propyl ethylamine carboxylate. In some embodiments, the solid support that can be linked by a covalent bond formed with a hydroxyl group or an amino group is a solid support that is linked by a phosphate bond, a carboxylate bond, and/or an amide bond. In some embodiments, the solid support is a resin.
In some embodiments, the 1 st functional group contains a hydroxyl group, -OR k Or a group represented by the formula (C3); and/or the 2 nd functional group contains a structure represented by formula (C1), (C2), (C3), (C1 ') or (C3'):
wherein q is 1 Is an integer of 1-4, X is O or NH, M + Is cationic, SPS represents a solid support,indicating the site of covalent attachment of the group.
In some embodiments, the 1 st functional group contains a phosphoramidite functionality, as shown in formula (C3), that can be coupled to a hydroxyl group at any position on a nucleotide, such as the 2 'hydroxyl or the 3' hydroxyl, and oxidized to form a phosphodiester linkage, conjugating the conjugated molecule to an oligonucleotide. At this time, the conjugate molecule of the present disclosure is capable of being conjugated to a nucleotide even if the 2 nd functional group is not present. At this point, the conjugate molecules of the present disclosure are adapted to react with hydroxyl groups on terminal nucleotides in the nucleotide sequence and form phosphodiester linkages during subsequent oxidation, thereby conjugating the conjugate molecules of the present disclosure to oligonucleotides.
In some embodiments, the 1 st functional group contains a protected hydroxyl group. In some embodiments, the 2 nd functional group contains a group that is reactive with the solid support to provide a conjugated molecule containing the solid support. In some embodiments, the 2 nd functional group contains a carboxylic acid functional group, a carboxylate functional group, or a phosphoramidite functional group, as shown in formula (C1), (C2), or (C3), which can undergo an esterification reaction or an amidation reaction with a hydroxyl or amino group on a solid support, such as a resin, to form a conjugate molecule containing a solid support linked via a carboxylic acid ester linkage or a solid support linked via an amide linkage. The phosphoramidite functionality can be coupled to a common solid support, such as a hydroxyl group on a resin, and oxidized to form a solid support linked via a phosphodiester linkage. At this time, according to an aspect of the present invention, there is provided a method of preparing the conjugate of the present disclosure using such a conjugate molecule. In some embodiments, the method comprises first linking the conjugate molecule to a solid support by condensation or coupling reaction, and then adding nucleoside monomers according to a solid phase phosphoramidite synthesis method, thereby obtaining a conjugate of the present disclosure that conjugates the conjugate molecule of the present disclosure to an oligonucleotide. In some embodiments, during solid phase phosphoramidite synthesis, deprotection of the 1 st functional group occurs followed by coupling with a phosphoramidite group on a nucleotide under coupling reaction conditions.
In some embodiments, R 1 Containing a 1 st functional group and a 2 nd functional group, the 1 st functional group containing a hydroxyl group or a protected hydroxyl group; the 2 nd functional group contains a carboxylic ester bond, an amide bond, or a phosphodiester bond, or a solid support linked by a carboxylic ester bond, an amide bond, or a phosphodiester bond. In some embodiments, the 2 nd functional group is a group as shown in formula (C1 ') or (C3'). In some embodiments, when the 2 nd functional group comprises a solid support, a conjugate molecule comprising the solid support facilitates the preparation of the conjugates of the present disclosure. Thus, in one aspect of the invention, there is provided a method of preparing a conjugate of the present disclosure using the conjugate molecule. In some embodiments, the method comprises reacting a conjugate molecule comprising a solid support with a nucleoside monomer according to a phosphoramidite solid phase synthesis method, thereby conjugating the conjugate molecule of the present disclosure to an oligonucleotide. In some embodiments, the conjugate molecule comprising a solid support may be bound to a solid support via a conjugate moietyThe reaction of the molecule with the solid support is internally obtained, and the conjugated molecule is reacted with a carboxyl group, a carboxylate or a phosphoramidite. In some embodiments, the conjugate molecule may be commercially available.
In some embodiments, the carboxylate functionality may be represented by-COO-M + Wherein M is + Is a cation, e.g. selected from metal cations, ammonium cations NH 4 + One of the organic ammonium cations. In some embodiments, the metal ion is selected from one of the alkali metal ions, such as K + Or Na (or) + . In some embodiments, the organic ammonium ion is an ammonium cation formed from a tertiary amine or a quaternary ammonium cation, such as an ammonium ion formed from triethylamine or an ammonium ion formed from N, N-diisopropylethylamine, for reasons of improving solubility and facilitating the reaction. In some embodiments, the carboxylate is triethylamine carboxylate or N, N-diisopropylethylamine carboxylate.
In some embodiments of the disclosure, R 1 Has a structure represented by the formula (B9), (B10), (B9 '), (B10'), (B11), (B12), (B11 ') or (B12'):
wherein q 1 Is an integer of 1 to 4, q 2 Is an integer of 1-10, X is O or NH, M + Is a cation, R k Is a hydroxyl protecting group, SPS represents a solid support,indicating the site of covalent attachment of the group. In some embodiments, q 1 1 or 2. In some embodiments, q 2 Is an integer of 1 to 5. In some embodiments, R 1 Comprises a structure represented by the formula (B9) or (B10). In some embodiments, R 1 Comprises a structure represented by the formula (B11) or (B12). In some embodiments, R k Is Tr (trityl), MMTr (4-methoxytrityl), DMTr (4, 4 '-dimethoxytrityl), TMTr (4, 4'4 "-trimethoxybenzyl). In some embodiments, R k May be DMTr. L (L) 1 Is a linear alkylene group of 1 to 70 carbon atoms in length, wherein one or more carbon atoms are optionally replaced by one or more selected from the group consisting of: c (O), NH, O, S, CH = N, S (O) 2 、C 2 -C 10 Alkenylene, C 2 -C 10 Alkynylene, C 6 -C 10 Arylene group, C 3 -C 18 Heterocyclylene and C 5 -C 10 Heteroarylene; and wherein L 1 Optionally having substituents of any one or more of the group consisting of: c (C) 1 -C 10 Alkyl, C 6 -C 10 Aryl, C 5 -C 10 Heteroaryl, C 1 -C 10 Haloalkyl, -OC 1 -C 10 Alkyl, -OC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-OH, -OC 1 -C 10 Haloalkyl, -SC 1 -C 10 Alkyl, -SC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-SH, -SC 1 -C 10 Haloalkyl, halogen substituent, -OH, -SH, -NH 2 、-C 1 -C 10 alkyl-NH 2 、-N(C 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -NH (C) 1 -C 10 Alkyl), cyano, nitro, -CO 2 H、-C(O)O(C 1 -C 10 Alkyl), -CON (C) 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -CONH (C) 1 -C 10 Alkyl), -CONH 2 ,-NHC(O)(C 1 -C 10 Alkyl), -NHC (O) (phenyl), -N (C) 1 -C 10 Alkyl) C (O) (C 1 -C 10 Alkyl), -N (C) 1 -C 10 Alkyl) C (O) (phenyl), -C (O) C 1 -C 10 Alkyl, -C (O) C 1 -C 10 Alkylphenyl, -C (O) C 1 -C 10 Haloalkyl, -OC (O) C 1 -C 10 Alkyl, -SO 2 (C 1 -C 10 Alkyl), -SO 2 (phenyl)、-SO 2 (C 1 -C 10 Haloalkyl) -SO 2 NH 2 、-SO 2 NH(C 1 -C 10 Alkyl), -SO 2 NH (phenyl) -NHSO 2 (C 1 -C 10 Alkyl), -NHSO 2 (phenyl) and-NHSO 2 (C 1 -C 10 Haloalkyl). The skilled artisan will appreciate that although L is for convenience 1 Is defined as a linear alkyl group, but it may not be a linear group or be named differently, such as an amine or alkenyl group resulting from the substitution and/or displacement described above. For the purposes of this disclosure, L 1 Is the number of atoms in the chain connecting the two attachment points. For this purpose, the ring (e.g., heterocyclylene or heteroarylene) resulting from substitution of the carbon atom of the straight chain alkyl group is counted as one atom.
In some embodiments, L 1 Is used for M 1 The ligand is linked to N on a nitrogen-containing backbone, thereby providing liver targeting function to the oligonucleotide conjugates of the present disclosure. In some embodiments, L 1 A linked combination of one or more selected from the groups of formulae A1-a 26. In some embodiments, L 1 A linked combination of one or more selected from A1, A4, A5, A6, A8, a10, a11 and a 13. In some embodiments, L 1 A combination of linkages selected from at least 2 of A1, A4, A8, a10 and a 11. In some embodiments, L 1 A combination of linkages selected from at least 2 of A1, A8, a 10.
In some embodiments, L 1 May be 3-25, 3-20, 4-15 or 5-12 atoms in length. In some embodiments, L 1 Is 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 55, or 60 atoms in length. In accordance with some embodiments of the present disclosure, j1 is an integer from 2 to 10, in some embodiments, j1 is an integer from 3 to 5. In some embodiments, j2 is an integer from 2 to 10, and in some embodiments, j2 is an integer from 3 to 5. R 'is a C1-C4 alkyl group, and in some embodiments, R' is one of methyl, ethyl, and isopropyl. Ra is A27, A28, A29, A30And one of a31, in some embodiments Ra is a27 or a28.Rb is a C1-C5 alkyl group, and in some embodiments Rb is one of methyl, ethyl, isopropyl, and butyl. In some embodiments, j1, j2, R', ra, rb are each selected in formulas A1-A26 to achieve M 1 The ligand being linked to N on a nitrogen-containing skeleton and allowing M 1 The spatial position between ligands is more suitable for M 1 The ligand binds to hepatic surface asialoglycoprotein receptors.
Each M 1 Independently selected from ligands capable of binding to cell surface receptors. In some embodiments, at least one M 1 Is a ligand capable of binding to liver surface receptors. In some embodiments, at least one M 1 Is a ligand capable of binding to a mammalian cell surface receptor. In some embodiments, at least one M 1 Is a ligand capable of binding to human hepatocyte surface receptor. In some embodiments, at least one M 1 Is a ligand capable of binding to hepatic surface asialoglycoprotein receptor (ASGPR).
In some embodiments, M 1 May be any ligand having an affinity for the asialoglycoprotein receptor (ASGPR) on the surface of mammalian hepatocytes, the class of which is well known to those skilled in the art. In some embodiments, at least one M 1 Is a saccharide. In some embodiments, each M 1 Is sugar. In some embodiments, at least one M 1 Is monosaccharide, disaccharide, trisaccharide or polysaccharide. In some embodiments, each M 1 Is monosaccharide, disaccharide, trisaccharide or polysaccharide. In some embodiments, at least one M 1 Is a modified sugar. In some embodiments, each M 1 Is a modified sugar. In some embodiments, each M 1 Independently selected from the group consisting of polysaccharides, modified polysaccharides, monosaccharides, or monosaccharide derivatives. In some embodiments, each or at least one M 1 Possibly independently selected from the group consisting of: glucose and its derivatives, mannans and its derivatives, galactose and its derivatives, xylose and its derivatives, ribose and its derivatives, fucose and its derivativesLactose and maltose and derivatives thereof, arabinose and derivatives thereof, fructose and derivatives thereof, and sialic acid.
In some embodiments, each or at least one M 1 May be independently selected from D-mannopyranose, L-mannopyranose, D-arabinose, D-xylofuranose, L-xylofuranose, D-glucose, L-glucose, D-galactose, L-galactose, alpha-D-glucofuranose, beta 0-D-mannopyranose, beta 1-D-mannopyranose, beta 2-D-glucopyranose, beta 3-D-glucopyranose, alpha-D-glucofuranose, beta-D-glucofuranose, alpha-D-fructofuranose, alpha-D-galactopyranose, beta-D-galactopyranose, alpha-D-galactofuranose, beta-galactofuranose, glucosamine, sialic acid, galactosamine, N-acetylgalactosamine, N-trifluoroacetyl galactosamine, N-propionyl galactosamine, N-N-butyryl galactosamine, N-isobutyryl galactosamine, 2-amino-3-O- [ (R) -1-carboxyethyl group ]-2-deoxy- β -D-glucopyranose, 2-deoxy-2-methylamino-L-glucopyranose, 4, 6-dideoxy-4-carboxamido-2, 3-di-O-methyl-D-glucopyranose, 2-deoxy-2-sulphonamino-D-glucopyranose, N-glycolyl- α -neuraminic acid, 5-thio- β -D-glucopyranose, 2,3, 4-tri-O-acetyl-1-thio-6-O-trityl- α -D-glucopyranoside methyl ester, 4-thio- β -D-galactopyranose, 3,4,6, 7-tetra-O-acetyl-2-deoxy-1, 5-dithio- α -D-glucoheptopyranoside ethyl ester, 2, 5-anhydro-D-psicose, ribose, D-4-thioribose, L-ribose, L-4-thioribose. In some embodiments, each M 1 Are all N-acetylgalactosamine (GalNAc). In some embodiments, ligand selection may be found in, for example, the disclosure of CN105378082a, the entire disclosure of which is incorporated herein by reference.
CN105378082a discloses a compound comprising a modified oligonucleotide and a conjugate group comprising at least one phosphorus or neutral linking group and 1 or more ligands, each ligand is selected from the group consisting of polysaccharide, modified polysaccharide, mannose, galactose, mannose derivative, galactose derivative, D-mannopyranose, L-mannopyranose, D-arabinose, D-xylofuranose, L-xylofuranose, D-glucose, L-glucose, D-galactose, L-galactose, alpha-D-mannfuranose, beta-D-mannopyranose, alpha-D-mannopyranose, beta-D-mannopyranose, alpha-D-glucopyranose, beta-D-glucopyranose, alpha-D-glucopyranose, beta-D-glucofuranose, alpha-D-fructofuranose, alpha-D-fructopyranose, alpha-D-galactopyranose, beta-D-galactopyranose, alpha-D-galactofuranose, beta-D-galactofuranose, glucosamine, sialic acid, alpha-D-galactosamine, N-acetyl galactosamine, 2-amino-3-O- [ (R) -1-carboxyethyl ] -2-deoxy-beta-D-glucopyranose, 2-D-glucopyranose, 2-methyl-L-galactopyranose, 4, 6-dideoxy-4-carboxamide-2, 3-di-O-methyl-D-mannopyranose, 2-deoxy-2-sulphonamino-D-glucopyranose, N-glycolyl-alpha-neuraminic acid, 5-thio-beta-D-glucopyranose, 2,3, 4-tri-O-acetyl-1-thio-6-O-trityl-alpha-D-glucopyranoside methyl ester, 4-thio-beta-D-galactopyranose, 3,4,6, 7-tetra-O-acetyl-2-deoxy-1, 5-dithio-alpha-D-glucopyranoside ethyl ester, 2, 5-anhydro-D-psilonitrile, ribose, D-4-thioribose, L-ribose or L-4-thioribose. The compounds are said to reduce the amount or activity of nucleic acid transcripts in cells.
WO2016077321A1 discloses numerous sirnas specifically targeting HBV genes and methods of delivering them and enhances their serum stability by modifying the nucleotides of the sirnas. The document also discloses siRNA conjugates, and further specifically discloses several siRNA conjugates having specific structures.
WO2016168286A1 discloses numerous sirnas specifically targeting the ANGPTL3 gene and methods of delivering them, and by modifying the nucleotides of the sirnas, the serum stability thereof is enhanced. The document also discloses siRNA conjugates.
N-acetylgalactosamine (GalNAc), a ligand that binds to hepatic surface asialoglycoprotein receptors. The asialoglycoprotein receptor (asialoglycoprotein receptor, ASGPR) is an endocytic receptor for hepatocyte-specific expression. In recent years, N-acetylgalactosamine (GalNAc), a high affinity ligand of ASGPR, was used as a ligandThe targeting molecule has better effect in the aspect of liver targeting delivery of nucleic acid medicaments. For example, siRNA based on GalNAc conjugation technology was first reported to exert interfering activity in mice by alnilla corporation (Alnylam pharmaceuticals, inc.) (Nair et al, j.am. Chem. Soc.,2014, 136, 16958-16961). The article reports that three clusters of GalNAc conjugated sirnas exhibit good delivery activity in both in vivo and in vitro experiments. Single dose ED by in vivo experiments in subcutaneously administered mice 50 The single injection dosage is less than 1ml at 1 mg/kg. In long-term dosing experiments, once weekly subcutaneous injections, stable interfering activities of up to 9 months were obtained.
In some embodiments, S 1 Independently M 1 . In some embodiments, S 1 Independently M 1 A group formed by substitution of at least one active hydroxyl group with a hydroxyl protecting group. In some embodiments, S 1 Independently M 1 A group in which all of the hydroxyl groups are substituted with a hydroxyl protecting group. In some embodiments, any hydroxy protecting group known to those skilled in the art may be used to protect M 1 Active hydroxyl groups on the polymer. In some embodiments, the protected hydroxy group is represented by the formula YCOO-wherein each Y is independently selected from C 1 -C 10 Alkyl, C 1 -C 10 Aryl, substituted C 1 -C 10 Alkyl or substituted C 1 -C 10 Aryl groups. In some embodiments, substituted C 1 -C 10 Alkyl is selected from the group consisting of one or more halogen substituents and/or one or more C 1 -C 6 C of alkyl-substituted radical 1 -C 10 An alkyl group. In some embodiments, substituted C 1 -C 10 Aryl is selected from the group consisting of one or more halogen substituents and/or one or more C 1 -C 6 C of alkyl-substituted radical 1 -C 10 Aryl groups. In some embodiments, each Y is independently selected from the group consisting of: methyl, trifluoromethyl, difluoromethyl, monofluoromethyl, trichloromethyl, dichloromethyl, monochloromethyl, ethyl, n-propyl, isopropyl, phenyl, halophenyl and C1-C6-alkyl And (3) a phenyl group.
In some embodiments, each S 1 Each independently one of the groups of formulae A46 to A54:
in some embodiments, S 1 Is of formula A49 or A50.
In some embodiments, each Y is independently selected from one of methyl, trifluoromethyl, difluoromethyl, monofluoromethyl, trichloromethyl, dichloromethyl, monochloromethyl, ethyl, n-propyl, isopropyl, phenyl, halophenyl, and alkylphenyl; for the purpose of simplifying the conjugate molecules of the present disclosure, in some embodiments, Y is methyl.
In some embodiments, the conjugate molecules of the present disclosure have a structure represented by formula (301), (302), (303), (304), or (305):
/>
in the above formulae (301) - (305), R k Is a hydroxyl protecting group, M + Selected from one of metal cations, ammonium cations, tertiary amine cations or quaternary ammonium cations. In some embodiments, M + Is that
In some embodiments, the conjugate molecules of the present disclosure may have a structure represented by formula (501), (502), (503), (504), or (505):
/>
/>
in the above formulae (501) to (505), wherein X is O or NH, R k Being a hydroxyl protecting group, SPS represents a solid support.
According to some embodiments of the present disclosure, the conjugate molecule of the present disclosure has a structure represented by formula (601), (602), (603), (604), or (605):
/>
In the above formulae (601) to (605), DMTr represents a 4,4' -dimethoxytrityl group, structureRepresents the salt of the corresponding carboxylic acid with triethylamine.
In some specific embodiments, the conjugate molecules of the present disclosure may have a structure represented by formula (701), (702), (703), (704), or (705):
/>
in the above formulae (701) to (705), SPS represents a solid phase carrier, and DMTr represents 4,4' -dimethoxytrityl.
Preparation of conjugate molecules of the present disclosure
Any reasonable synthetic route can be used by one of skill in the art to prepare the conjugate molecules of the present disclosure.
In some embodiments of the present disclosure, a method for preparing a conjugate molecule of formula (101) includes contacting a compound of formula (102) with a cyclic anhydride in an organic solvent under esterification reaction conditions and in the presence of a base and an ester-forming catalyst, ion-exchanging, and isolating to obtain a compound of formula (101):
wherein:
R 7 to provide R in formula (101) 1 Is a group of (2). In some embodiments, for example, R 7 Has a structure represented by formula (A61):
n 1 、n 2 、m 1 、R 2 、L 1 、S 1 the respective definitions and optional ranges are as previously described, R i To achieve N connection with nitrogen-containing skeleton, R k O is attached to and has attached to it an optional radical of free hydroxy, R k Is a hydroxyl protecting group. At this time, R is obtained 1 The compound contains a 1 st functional group and a 2 nd functional group as hydroxyl protecting groups, and the 2 nd functional group contains a compound of formula (101) having a structure shown as formula (C1) or (C2). In some embodiments, R 7 The structure shown as B7 or B8:
wherein q 2 And R is k The respective definitions are as described above.
The esterification reaction conditions include a reaction temperature of 0 to 100 ℃ and a reaction time of 8 to 48 hours, and in one embodiment, the esterification reaction conditions include a reaction temperature of 10 to 40 ℃ and a reaction time of 20 to 30 hours.
In some embodiments, the organic solvent is one or more of an epoxy solvent, an ether solvent, an haloalkane solvent, dimethyl sulfoxide, N-dimethylformamide, and N, N-diisopropylethylamine. In one embodiment, the epoxy solvent is dioxane and/or tetrahydrofuran, the ether solvent is diethyl ether and/or methyl tertiary butyl ether, and the haloalkane solvent is one or more of dichloromethane, chloroform and 1, 2-dichloroethane. In one embodiment, the organic solvent is methylene chloride. The amount of the organic solvent used is 3 to 50L/mol, and in one embodiment 5 to 20L/mol, relative to the compound represented by the formula (102).
In some embodiments, the cyclic anhydride is one of succinic anhydride, glutaric anhydride, adipic anhydride, or pimelic anhydride, in one embodiment succinic anhydride. The molar ratio of the cyclic anhydride to the compound of formula (102) is 1:1 to 10:1, and in one embodiment 2:1 to 5:1.
The ester-forming catalyst may be any catalyst that catalyzes the esterification reaction, for example, the catalyst may be 4-dimethylaminopyridine. The molar ratio of the catalyst to the compound of formula (102) is from 1:1 to 10:1, and in one embodiment from 2:1 to 5:1.
In some embodiments, the base may be any inorganic base, organic base, or combination thereof. The base may be, for example, a tertiary amine organic base in view of solubility and product stability. In one embodiment, the tertiary amine organic base is triethylamine or N, N-diisopropylethylamine. The molar ratio of the tertiary amine organic base to the compound of formula (102) is 1:1 to 20:1, and in one embodiment 3:1 to 10:1.
The ion exchange is the conversion of the compound of formula (101) to the desired carboxylic acid or carboxylate salt form, and methods of ion exchange are well known to those skilled in the art, Suitable ion exchange solutions and exchange conditions can be used to obtain the aforementioned cation M + Is not described in detail herein. In one embodiment, the ion exchange reaction is carried out using a triethylamine phosphate solution having a concentration of 0.2 to 0.8M, in one embodiment 0.4 to 0.6M, in an amount of 3 to 6L/mol, in one embodiment 4 to 5L/mol, relative to the compound of formula (102).
The compound of formula (101) may be isolated from the reaction mixture using any suitable isolation method. In some embodiments, the compound of formula (101) may be removed by evaporation followed by separation by chromatographic methods, e.g., separation may be performed using the following chromatographic conditions: (1) normal phase purification silica gel: 200-300 mesh silica gel packing, using dichloromethane containing 1 wt%o triethylamine, methanol=100:18-100:20 gradient elution; or (2) reverse phase purification: c18, C8 reversed phase packing, eluting with methanol: acetonitrile=0.1:1-1:0.1 gradient. In some embodiments, the solvent may be directly removed to provide a crude compound of formula (101), which may be directly used in subsequent reactions.
In some embodiments, the method for preparing a compound of formula (101) further comprises contacting the product obtained by the ion exchange reaction with a solid support containing an amino group or a hydroxyl group in an organic solvent in the presence of a condensing agent and a tertiary amine organic base under condensation reaction conditions. At this time, R is obtained 1 The compound contains a 1 st functional group and a 2 nd functional group, wherein the 1 st functional group contains a hydroxyl protecting group, and the 2 nd functional group contains a compound of formula (101) with a structure shown as a formula (C1').
The solid support is one of the supports used in solid phase synthesis of siRNA, some of which are well known to those skilled in the art. For example, the solid support may be selected from solid supports containing reactive hydroxyl or amino functional groups, in one embodiment amino resins or hydroxyl resins. In some embodiments, the amino or hydroxyl resin has the following parameters in one embodiment: particle size of 100-400 mesh, and surface amino or hydroxyl loading of 0.2-0.5mmol/g. The ratio of the compound represented by formula (101) to the solid carrier is 10 to 400. Mu. Mol of the compound per gram of the solid carrier (. Mu. Mol/g). In some embodiments, the compound of formula (101) is used in an amount of 50 to 200. Mu. Mol/g relative to the solid support.
The organic solvent may be any suitable solvent or mixed solvent known to those skilled in the art. In some embodiments, the organic solvent is one or more of acetonitrile, an epoxy-based solvent, an ether-based solvent, an haloalkane-based solvent, dimethyl sulfoxide, N-dimethylformamide, and N, N-diisopropylethylamine. In some embodiments, the epoxide-based solvent is dioxane and/or tetrahydrofuran, the ether-based solvent is diethyl ether and/or methyl tert-butyl ether, and the haloalkane-based solvent is one or more of dichloromethane, chloroform, and 1, 2-dichloroethane. In some embodiments, the organic solvent is acetonitrile. The organic solvent is used in an amount of 20 to 200L/mol, and in one embodiment 50 to 100L/mol, relative to the compound of formula (102).
In some embodiments, the condensing agent may be benzotriazol-1-yl-oxy-tripyrrolidinylphosphine hexafluorophosphate, 3-diethoxyphosphoryl-1, 2, 3-benzooxazol 4 (3H) -one, and/or O-benzotriazol-tetramethylurea hexafluorophosphate, in one embodiment, the condensing agent is O-benzotriazol-tetramethylurea hexafluorophosphate. The molar ratio of condensing agent to compound of formula (102) is 1:1 to 20:1, and in one embodiment 1:1 to 5:1.
In some embodiments, the tertiary amine organic base is triethylamine and/or N, N-diisopropylethylamine, in some embodiments N, N-diisopropylethylamine; the molar ratio of the tertiary amine organic base to the compound of formula (102) is 1:1 to 20:1, and in one embodiment 1:1 to 5:1.
In some embodiments, the method for preparing the compound of formula (101) may further include contacting the obtained condensation product with a capping reagent and an acylation catalyst in an organic solvent under capping reaction conditions, and separating to obtain the compound of formula (101). The capping reaction serves to remove any reactive functional groups that have not yet reacted to completion, to avoid the production of unwanted byproducts in subsequent reactions. The conditions under which the cap reacts include a reaction temperature of 0-50 ℃, in some embodiments 15-35 ℃, for a period of 1-10 hours, in some embodiments 3-6 hours. Capping reagents used in solid phase synthesis of siRNA can be used, and capping reagents used in solid phase synthesis of siRNA are well known to those skilled in the art.
In some embodiments, the capping reagent consists of capping reagent a (capA) and capping reagent B (capB), wherein capping reagent a is N-methylimidazole, in some embodiments provided in the form of a pyridine/acetonitrile mixed solution of N-methylimidazole, wherein the volume ratio of pyridine to acetonitrile is 1:10-1:1. In some embodiments 1:3 to 1:1. In some embodiments, the total volume of pyridine and acetonitrile and the volume of N-methylimidazole is 1:1 to 10:1, and in some embodiments 3:1 to 7:1. In some embodiments, the capping reagent B is acetic anhydride, in some embodiments, the capping reagent B is provided in the form of an acetonitrile solution of acetic anhydride, wherein the volumes of acetic anhydride and acetonitrile are 1:1-1:10, in other embodiments 1:2-1:6.
In some embodiments, the ratio of the volume of the pyridine/acetonitrile mixed solution of N-methylimidazole to the mass of the compound of formula (102) is 5ml/g to 50ml/g, and in some embodiments, 15ml/g to 30ml/g. The ratio of the volume of the acetonitrile solution of acetic anhydride to the mass of the compound of formula (102) is from 0.5ml/g to 10ml/g, and in some embodiments, from 1ml/g to 5ml/g.
In some embodiments, the capping reagent uses equimolar amounts of acetic anhydride and N-methylimidazole. In some embodiments, the organic solvent is one or more of acetonitrile, an epoxy-based solvent, an ether-based solvent, an haloalkane-based solvent, dimethyl sulfoxide, N-dimethylformamide, and N, N-diisopropylethylamine. In some embodiments, the organic solvent is acetonitrile. The organic solvent is used in an amount of 10 to 50L/mol, and in some embodiments 5 to 30L/mol, relative to the compound of formula (102).
In some embodiments, the acylation catalyst may be selected from any catalyst useful for ester-forming or amide-forming condensation, such as basic heterocyclic compounds. In some embodiments, the acylation catalyst is 4-dimethylaminopyridine. The mass ratio of the catalyst to the compound of formula (102) is from 0.001:1 to 1:1, in one embodiment from 0.01:1 to 0.1:1.
In some embodiments, the compound of formula (101) may be isolated from the reaction mixture using any suitable isolation method. In some embodiments, the compound of formula (101) may be obtained by washing thoroughly with an organic solvent selected from acetonitrile, dichloromethane, methanol, in some embodiments acetonitrile, and filtering to remove unreacted reactants, excess capping reagent, and other impurities.
In some embodiments, the preparation method of the conjugate molecule shown in the formula (101) comprises contacting the compound shown in the formula (102) with phosphoramidite under the condition of coupling reaction and in the presence of a coupling reagent, and separating to obtain the compound shown in the formula (101). At this time, R is obtained 1 The compound contains a 1 st functional group and a 2 nd functional group, wherein the 1 st functional group contains a hydroxyl protecting group, and the 2 nd functional group contains a compound of a formula (101) with a structure shown as a formula (C3).
In some embodiments, the coupling reaction conditions include a temperature of 0 to 50 ℃, e.g., 15 to 35 ℃, and a molar ratio of the compound of formula (102) to the phosphoramidite of 1:1 to 1:50, e.g., 1:5 to 1:15; the molar ratio of the compound of formula (102) to the coupling reagent is from 1:1 to 1:100, for example from 1:50 to 1:80; the reaction time is 200 to 3000 seconds, for example 500 to 1500 seconds. The phosphoramidite may be, for example, bis (diisopropylamino) (2-cyanoethoxy) phosphine, which is commercially available or synthetically obtained according to methods well known in the art. The coupling reagent is selected from one or more of 1H-tetrazole, 5-ethylthio 1H-tetrazole and 5-benzylthio 1H-tetrazole, for example, 5-ethylthio 1H-tetrazole. The coupling reaction may be carried out in an organic solvent selected from one or more of anhydrous acetonitrile, anhydrous DMF, anhydrous dichloromethane, for example, anhydrous acetonitrile. In some embodiments, the organic solvent is used in an amount of 3 to 50L/mol, for example, 5 to 20L/mol, relative to the compound of formula (102). By performing this coupling reaction, the hydroxyl group in the compound of formula (102) reacts with the phosphoramidite to form a phosphoramidite group. In some embodiments, the solvent may be directly removed to provide a crude compound of formula (101), which may be directly used in subsequent reactions.
In some embodiments, the method of preparing a compound of formula (101) further comprises the steps of: the isolated product is further contacted with a solid support containing hydroxyl groups under coupling reaction conditions in an organic solvent and in the presence of a coupling reagent. Then, the compound of formula (101) is isolated by capping reaction and oxidation reaction. At this time, R is obtained 1 The compound contains a 1 st functional group and a 2 nd functional group, wherein the 1 st functional group contains a hydroxyl protecting group, and the 2 nd functional group has a structure shown as a formula (C3') in the formula (101).
In some embodiments, the solid phase carrier is a solid phase carrier known in the art as useful for solid phase synthesis of nucleic acids, for example, a commercially available universal solid phase carrier after deprotection reactionHL UnyLinker TM 300Oligonucleotide Synthesis Support,Kinovate Life Sciences company, structure shown as formula B80):
deprotection reactions are well known to those skilled in the art. In some embodiments, the deprotection conditions include a temperature of 0-50 ℃, e.g., 15-35 ℃; the reaction time is 30 to 300 seconds, for example 50 to 150 seconds. The deprotecting reagent may be selected from one or more of trifluoroacetic acid, trichloroacetic acid, dichloroacetic acid, monochloroacetic acid, and in some embodiments, the deprotecting reagent is dichloroacetic acid. The molar ratio of deprotection reagent to-DMTr (4, 4' -dimethoxytrityl) protecting group on the stationary phase is 2:1-100:1, e.g., 3:1-50:1. By performing the deprotection, a free hydroxyl group having reactivity is obtained on the surface of the solid support, thereby facilitating the next coupling reaction.
The coupling reaction conditions and the coupling reagents are selected as described above. By carrying out this coupling reaction, the free hydroxyl groups formed in the deprotection reaction react with phosphoramidite groups to form phosphite linkages.
In some embodiments, the capping reaction conditions include a temperature of 0-50 ℃, e.g., 15-35 ℃, and a reaction time of 5-500 seconds, e.g., 10-100 seconds, with the capping reaction being performed in the presence of a capping reagent. The capping reagent is selected and used as described above.
The oxidation reaction conditions include a temperature of 0-50 ℃, e.g., 15-35 ℃, a reaction time of 1-100 seconds, e.g., 5-50 seconds, and the oxidizing agent, e.g., iodine (provided in some embodiments in the form of iodine water). In some embodiments, the molar ratio of oxidizing agent to phosphite groups is from 1:1 to 100:1, e.g., can be from 5:1 to 50:1. In some specific embodiments, the oxidation reaction is performed in a mixed solvent of tetrahydrofuran: water: pyridine=3:1:1 to 1:1:3.
In some embodiments, the compound of formula (102) may be obtained by the following preparation method: in an organic solvent, in the presence of an amide-forming reaction condensing agent and a tertiary amine organic base, contacting a compound shown in a formula (103) with a compound shown in a formula (104) under condensation reaction conditions, and separating to obtain a compound shown in a formula (102):
Wherein n is 1 、n 2 、m 1 、R 2 、R 7 、L 1 、S 1 The respective definitions and optional ranges are as previously described.
The compounds of formula (104) may be prepared using, for example, the compounds disclosed in j.am.chem.soc.2014,136,16958-16961, or the compounds of formula (104) may be prepared by a variety of methods by those skilled in the art, for example, certain compounds of formula (104) may be prepared by reference to the methods disclosed in example 1 of US8106022B2, the entire contents of which are incorporated herein by reference.
In some embodiments, the condensation reaction conditions include a reaction temperature of 0 to 100 ℃, a reaction time of 0.1 to 24 hours, and in one embodiment 10 to 40 ℃, a reaction time of 0.5 to 16 hours.
The molar ratio of the compound of formula (104) to the compound of formula (103) is from 2:1 to 10:1, and in one embodiment from 2.5:1 to 5:1.
In some embodiments, the organic solvent is one or more of acetonitrile, an epoxy-based solvent, an ether-based solvent, an haloalkane-based solvent, dimethyl sulfoxide, N-dimethylformamide, and N, N-diisopropylethylamine, the epoxy-based solvent in one embodiment is dioxane and/or tetrahydrofuran, the ether-based solvent in one embodiment is diethyl ether and/or methyl tert-butyl ether, the haloalkane-based solvent in one embodiment is one or more of dichloromethane, chloroform, and 1, 2-dichloroethane, and the organic solvent in one embodiment is acetonitrile. The organic solvent is used in an amount of 3 to 50L/mol, and in one embodiment 5 to 20L/mol, relative to the compound of formula (103).
In some embodiments, the amide forming reaction condensing agent is benzotriazol-1-yl-oxy-tripyrrolidinylphosphine hexafluorophosphate, 3-diethoxyphosphoryl-1, 2, 3-benzozol 4 (3H) -one (DEPBT), O-benzotriazol-tetramethylurea hexafluorophosphate, or 4- (4, 6-dimethoxytriazin-2-yl) -4-methylmorpholine hydrochloride, in one embodiment 4- (4, 6-dimethoxytriazin-2-yl) -4-methylmorpholine hydrochloride. The molar ratio of the amide-forming reaction condensing agent to the compound of formula (103) is from 2:1 to 10:1, in one embodiment from 2.5:1 to 5:1;
the tertiary amine organic base is N-methylmorpholine, triethylamine or N, N-diisopropylethylamine, in one embodiment N-methylmorpholine; the molar ratio of the tertiary amine organic base to the compound of formula (103) is from 3:1 to 20:1, and in one embodiment from 5:1 to 10:1.
Similar to the above, the compound of formula (102) may be isolated from the reaction mixture using any suitable isolation method. In some embodiments, the solvent may be removed by evaporation followed by chromatographic separation of the compound of formula (102), e.g., separation may be performed using the following chromatographic conditions: (1) normal phase purification silica gel: silica gel packing of 200-300 mesh, eluting with dichloromethane: methanol=100:5-100:7 gradient; and (2) reverse phase purification: c18, C8 reversed phase packing, eluting with methanol: acetonitrile=0.1:1-1:0.1 gradient. In some embodiments, the solvent may be directly removed to provide a crude compound of formula (102), which may be directly used in subsequent reactions.
In some embodiments, the compound of formula (103) is reacted with a sufficient amount of a compound of formula (104) in one step to form the desired compound of formula (102), in which case each S 1 -L 1 The parts are identical to each other. In some embodiments, the compound of formula (103) may be prepared by batchwise reacting a different compound of formula (104), i.e., L, as desired 1 And/or S 1 The different compounds of formula (104) react so that the resulting compound of formula (102) contains more than two S 1 And/or L 1 . For example, for 1eq of a compound of formula (103), it may be contacted with 2eq of a compound of formula (104) to attach a first S to the two terminal primary amine groups in the compound of formula (103) 1 -L 1 Part, then, it is continued with (m) 1 -1) eq of a compound of the second formula (104) to (m) 1 Definition and value ranges of (a) are as defined above), whereby (m) in the compound of formula (103) 1 -1) linking a second S to a secondary amine group 1 -L 1 Part(s).
In one embodiment, R 7 In this case, the compound represented by the formula (103) can be obtained by the following preparation method: contacting the compound shown in the formula (105) with the compound shown in the formula (A-1) or the compound shown in the formula (A-2) in an organic solvent under the condition of amide forming reaction and in the presence of an amide forming reaction condensing agent and a tertiary amine organic base, and separating to obtain the compound shown in the formula (103):
Wherein n is 1 、n 2 、m 1 、R 2 、R 7 、L 1 、S 1 The respective definitions and optional ranges are as previously described.
The amide forming reaction conditions are a reaction temperature of from 0 to 100 ℃ for a reaction time of from 1 to 48 hours, in some embodiments, the amide forming reaction conditions are a reaction temperature of from 10 to 40 ℃ for a reaction time of from 2 to 16 hours.
In some embodiments, the organic solvent is one or more of an alcohol solvent, an epoxy solvent, an ether solvent, an alkyl halide solvent, dimethyl sulfoxide, N-dimethylformamide, and N, N-diisopropylethylamine. The alcoholic solvent is one or more of methanol, ethanol, propanol in one embodiment, and ethanol in some embodiments. The epoxy-based solvent is dioxane and/or tetrahydrofuran in some embodiments. The ether solvent is diethyl ether and/or methyl tert-butyl ether in some embodiments. The haloalkane-based solvent is in some embodiments one or more of methylene chloride, chloroform, and 1, 2-dichloroethane. In some embodiments, the organic solvent is dichloromethane. The amount of organic solvent used is 3 to 50L/mol, and in one embodiment 3 to 20L/mol, relative to the compound of formula (105).
In some embodiments, the amide forming reaction condensing agent is benzotriazol-1-yl-oxy-tripyrrolidinylphosphine hexafluorophosphate, 3-diethoxyphosphoryl-1, 2, 3-benzooxazol 4 (3H) -one, 4- (4, 6-dimethoxytriazin-2-yl) -4-methylmorpholine hydrochloride, 2-ethoxy-1-ethoxycarbonyl-1, 2-dihydroquinoline (EEDQ), or O-benzotriazol-tetramethylurea hexafluorophosphate, in one embodiment 3-diethoxyphosphoryl-1, 2, 3-benzooxazol 4 (3H) -one. The molar ratio of the amide-forming reaction condensing agent to the compound of formula (105) is 1:1 to 10:1, and in one embodiment is 2.5:1 to 5:1.
In some embodiments, the tertiary amine organic base is triethylamine or N, N-diisopropylethylamine, in one embodiment N, N-diisopropylethylamine. The molar ratio of the tertiary amine organic base to the compound of formula (105) is from 3:1 to 20:1, and in one embodiment from 5:1 to 10:1.
In some embodiments, the compounds of formula (A-1) and formula (A-2) may be prepared by any suitable means. For example, when R k In the case of DMTr groups, the compounds of formula (A-1) can be prepared by reacting calcium glycerate with DMTrCl; similarly, 3-amino-1, 2-propanediol may be contacted with a cyclic anhydride, which may be a cyclic anhydride having 4 to 13 carbon atoms, in one embodiment 4 to 8 carbon atoms, followed by reaction with DMTrCl to produce the compound of formula (A-2). As will be readily appreciated by those skilled in the art, the cyclic anhydride is selected to correspond to q in the (A-2) compound 2 For example, when the cyclic anhydride is succinic anhydride, q 2 When the cyclic anhydride is glutaric anhydride, =1, q 2 =2, and so on.
In some variations, the compound of formula (103) may also be prepared by reacting the compound of formula (105) with the cyclic anhydride, 3-amino-1, 2-propanediol, and DMTrCl in that order. It will be readily appreciated by those skilled in the art that these modifications do not affect the structure and function of the compound of formula (103) and that these modifications are readily achievable by those skilled in the art based on the above-described methods.
Similarly as described above, the compound of formula (103) may be isolated from the reaction mixture using any suitable isolation method. In some embodiments, the compound of formula (103) may be removed by evaporation followed by separation by chromatographic methods, e.g., separation may be performed using the following chromatographic conditions: (1) normal phase purification silica gel: 200-300 mesh silica gel filler, petroleum ether, ethyl acetate, dichloromethane and N, N-dimethylformamide=1:1:0.5-1:1:1:0.6 gradient elution; and (2) reverse phase purification: c18, C8 reversed phase packing, eluting with methanol: acetonitrile=0.1:1-1:0.1 gradient. In some embodiments, the solvent may be directly removed to provide a crude compound of formula (103), which may be directly used in subsequent reactions.
In some embodiments, each R 2 Are all the same, and each n 2 And n 1 At the same time, two of the formulas (105)NH number 2 The groups are chemically equivalent. In some embodiments, the compound of formula (a-1) or (a-2) is reacted with an equimolar amount of the compound of formula (105), followed by isolation to obtain the compound of formula (103); in some embodiments, the compound of formula (A-1) or (A-2) is reacted with an excess of the compound of formula (105), followed by isolation to obtain the compound of formula (103).
The compounds of formula (105) are commercially available or are obtained by a person skilled in the art using known methods. For example, when m 1 =2、n 1 And each n 2 Are all 2, and each R 2 In the case of H, the compound of formula (105) is commercially available from the company alfa.
Oligonucleotide conjugates
In another aspect, the present disclosure provides an oligonucleotide conjugate having a structure as shown in formula (201):
wherein:
n 1 is an integer selected from 1-2;
each n 2 Independently selected from integers from 1-2;
m 1 is an integer selected from 1-6;
each R 2 Each independently is selected from H, C 1 -C 10 Alkyl, C 1 -C 10 Haloalkyl or C 1 -C 10 Alkoxy, in some embodiments, R 10 、R 11 、R 12 、R 13 、R 14 And R is 15 Each independently selected from one of H, methyl or ethyl;
R 6 is an active agent, in some embodiments, R 6 Containing a functional oligonucleotide;
R 5 is a linear alkylene group of 1 to 20 carbon atoms in length, wherein one or more carbon atoms are optionally replaced by one or more selected from the group consisting of: c (O), NH, O, S, CH = N, S (O) 2 、C 2 -C 10 Alkenylene, C 2 -C 10 Alkynylene, C 6 -C 10 Arylene group, C 3 -C 18 Heterocyclylene and C 5 -C 10 Heteroarylene; and wherein R is 5 Optionally having substituents of any one or more of the group consisting of: c (C) 1 -C 10 Alkyl, C 6 -C 10 Aryl, C 5 -C 10 Heteroaryl, C 1 -C 10 Haloalkyl, -OC 1 -C 10 Alkyl, -OC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-OH, -OC 1 -C 10 Haloalkyl, -SC 1 -C 10 Alkyl, -SC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-SH, -SC 1 -C 10 Haloalkyl, halogen substituent, -OH, -SH, -NH 2 、-C 1 -C 10 alkyl-NH 2 、-N(C 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -NH (C) 1 -C 10 Alkyl), cyano, nitro, -CO 2 H、-C(O)O(C 1 -C 10 Alkyl), -CON (C) 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -CONH (C) 1 -C 10 Alkyl), -CONH 2 ,-NHC(O)(C 1 -C 10 Alkyl), -NHC (O) (phenyl), -N (C) 1 -C 10 Alkyl) C (O) (C 1 -C 10 Alkyl), -N (C) 1 -C 10 Alkyl) C (O) (phenyl), -C (O) C 1 -C 10 Alkyl, -C (O) C 1 -C 10 Alkylphenyl, -C (O) C 1 -C 10 Haloalkyl, -OC (O) C 1 -C 10 Alkyl, -SO 2 (C 1 -C 10 Alkyl), -SO 2 (phenyl) -SO 2 (C 1 -C 10 Haloalkyl) -SO 2 NH 2 、-SO 2 NH(C 1 -C 10 Alkyl), -SO 2 NH (phenyl) -NHSO 2 (C 1 -C 10 Alkyl), -NHSO 2 (phenyl) and-NHSO 2 (C 1 -C 10 Halogenated compoundsAn alkyl group);
each L 1 Is a linear alkylene group of 1 to 70 carbon atoms in length, wherein one or more carbon atoms are optionally replaced by one or more selected from the group consisting of: c (O), NH, O, S, CH = N, S (O) 2 、C 2 -C 10 Alkenylene, C 2 -C 10 Alkynylene, C 6 -C 10 Arylene group, C 3 -C 18 Heterocyclylene and C 5 -C 10 Heteroarylene; and wherein L 1 Optionally having substituents of any one or more of the group consisting of: c (C) 1 -C 10 Alkyl, C 6 -C 10 Aryl, C 5 -C 10 Heteroaryl, C 1 -C 10 Haloalkyl, -OC 1 -C 10 Alkyl, -OC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-OH, -OC 1 -C 10 Haloalkyl, -SC 1 -C 10 Alkyl, -SC 1 -C 10 Alkylphenyl radicals C 1 -C 10 alkyl-SH, -SC 1 -C 10 Haloalkyl, halogen substituent, -OH, -SH, -NH 2 、-C 1 -C 10 alkyl-NH 2 、-N(C 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -NH (C) 1 -C 10 Alkyl), cyano, nitro, -CO 2 H、-C(O)O(C 1 -C 10 Alkyl), -CON (C) 1 -C 10 Alkyl) (C) 1 -C 10 Alkyl), -CONH (C) 1 -C 10 Alkyl), -CONH 2 ,-NHC(O)(C 1 -C 10 Alkyl), -NHC (O) (phenyl), -N (C) 1 -C 10 Alkyl) C (O) (C 1 -C 10 Alkyl), -N (C) 1 -C 10 Alkyl) C (O) (phenyl), -C (O) C 1 -C 10 Alkyl, -C (O) C 1 -C 10 Alkylphenyl, -C (O) C 1 -C 10 Haloalkyl, -OC (O) C 1 -C 10 Alkyl, -SO 2 (C 1 -C 10 Alkyl), -SO 2 (phenyl) -SO 2 (C 1 -C 10 HaloalkanesRadical) -SO 2 NH 2 、-SO 2 NH(C 1 -C 10 Alkyl), -SO 2 NH (phenyl) -NHSO 2 (C 1 -C 10 Alkyl), -NHSO 2 (phenyl) and-NHSO 2 (C 1 -C 10 A haloalkyl group); in some embodiments, L 1 A combination of linkages which may be selected from one or more of the groups of formulae A1 to A26, A1 to A26 being as defined above.
n 1 、n 2 、m 1 、R 2 、L 1 、M 1 The respective definitions and optional ranges are as previously described.
In some embodiments, R 5 Is R in the compound of formula (101) 1 The group is linked to the active drug via a group reaction to form a linking group. In some embodiments, R 5 Is R in the compound of formula (101) 1 The group is attached to the functional oligonucleotide by reaction to form a linking group. In some embodiments, R 5 The group contains a connecting site connected with N on a nitrogen-containing framework and R 6 A junction site for P-phase junction in (C). In some embodiments, R 5 Wherein the site linked to N on the nitrogen-containing skeleton forms an amide bond with N, and R 6 The P-linked site in (2) forms a phosphate bond with P. In some embodiments, R 5 Can be B5, B6, B5 'or B6':
wherein,represents the site of covalent bonding of the groups, q 2 The selection and value ranges of (a) are as described above.
In some embodiments, R 6 A group of the structure shown as a 59:
wherein E is 1 Is OH, SH or BH 2 In some embodiments, E 1 OH or SH; nu is an oligonucleotide.
In the context of the present disclosure, unless otherwise indicated, a "conjugate" group or molecule refers to a group or molecule capable of forming a covalent bond with a corresponding ligand, and the conjugate group or molecule and its ligand all have a specific function. Accordingly, "conjugate" refers to a compound formed by covalent linkage between the chemical moieties. Further, "oligonucleotide conjugate" refers to a compound formed by covalently attaching one or more conjugate moieties having a specific function to an oligonucleotide. In the context of the present disclosure, a "conjugate molecule" may be understood as a specific compound that can be conjugated to an oligonucleotide by reaction, ultimately forming an oligonucleotide conjugate of the present disclosure. In some embodiments, the oligonucleotide is an siRNA, in which case the conjugate of the present disclosure is an siRNA conjugate.
In some embodiments, the conjugates of the present disclosure have a structure represented by formula (401), (402), (403), (404), or (405):
/>
in some embodiments, the oligonucleotides in the oligonucleotide conjugates of the present disclosure are functional oligonucleotides. Functional oligonucleotides refer to such oligonucleotides: the oligonucleotides are capable of up-regulating or down-regulating the expression of a target gene or causing alternative splicing of mRNA by creating stable and specific hybridization with the target sequence using principles such as RNA activation (RNAa), RNA interference (RNAi), antisense nucleic acid technology, exon skipping (exon skip) technology, etc. In some aspects, a functional oligonucleotide may also be a nucleic acid structure that produces stable and specific binding to a target protein. Furthermore, it will be readily appreciated by those skilled in the art that polynucleotides (e.g., mRNA itself or fragments thereof) are equally suitable for conjugation with the conjugate molecules provided by the present disclosure to form conjugates for targeted delivery, such as liver targeted delivery, to modulate expression of proteins transcribed from mRNA. Thus, in this context, the concept of "functional oligonucleotide" may also encompass mRNA or fragments thereof.
In some embodiments, the functional oligonucleotide is capable of interacting with the target sequence, thereby affecting the normal function of the target sequence molecule, such as causing mRNA cleavage or translational repression or exon skipping to initiate alternative splicing of the mRNA, etc. In some embodiments, the functional oligonucleotide may be substantially complementary to a base of the target sequence. In some embodiments, the functional oligonucleotide may be 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more base complementary to the target sequence, or fully complementary to the target sequence. In some embodiments, the functional oligonucleotide may contain 1, 2, or 3 bases that are not complementary to the target sequence. In some embodiments, the functional oligonucleotides include deoxyribonucleotides or ribonucleotides, as well as nucleotides with modifications. In some embodiments, the functional oligonucleotide may be single-stranded DNA, RNA, or DNA-RNA chimeras (chimers), or double-stranded DNA, RNA, or DNA-RNA hybrids.
Thus, in some embodiments, a functional oligonucleotide suitable for inclusion in an oligonucleotide conjugate of the present disclosure may be one of a small interfering RNA (siRNA), a microRNA (microRNA), an anti-microRNA (anti-micror), a microRNA antagonist (anti-microRNA), a microRNA mimetic (microRNA), a decoy oligonucleotide (decoy), an immunostimulatory substance (immune stimulatory), a G-quadrupole (G-quadraplex), an alternative splice (splice), a single-stranded RNA (ssRNA), an antisense nucleic acid (antisense), a nucleic acid aptamer (Nucleic Acid Aptamer), a small activating RNA (small activating RNA, saRNA), a stem-loop RNA (stem-loop RNA), or DNA. WO2015/006740A2 discloses a conjugate of a different ligand conjugated to an oligonucleotide, wherein the ligand is linked to the oligonucleotide by a linker (linker) selected from one of small interfering RNAs (siRNA), micrornas (micrornas), anti-micrornas (anti-micrornas), microRNA antagonists (anti-micrornas), microRNA mimics (microRNA mimics), decoy oligonucleotides (decoy), immunostimulants (immune stimulatory), G-quadrupoles (G-quales), alternative spliceosomes (splice), single-stranded RNAs (ssrnas), antisense nucleic acids (anti-sense), aptamers (stem-loop RNAs) or DNA. These conjugates exhibit good stability in the in vivo delivery of oligonucleotides. In a further embodiment, the functional oligonucleotides suitable for inclusion in the oligonucleotide conjugates of the present disclosure may be the oligonucleotides disclosed in WO2009082607A2, WO2009073809A2 or WO2015006740A2, the entire contents of which are incorporated herein by reference.
The oligonucleotide conjugates of the present disclosure can modulate abnormal expression of a particular gene in a particular cell, such as a hepatocyte, by increasing the liver targeted delivery efficiency of an active agent, such as a functional oligonucleotide, thereby enhancing the interaction between the functional oligonucleotide and the targeting sequence in the cell. In some embodiments, the specific gene may be an endogenous gene expressed in the liver, or may be a pathogen gene propagated in the liver. The genes abnormally expressed in hepatocytes may be, for example, apoB, apoC, ANGPTL3, PCSK9, SCD1, FVII, p53, HBV, HCV, etc. In some embodiments, the gene that is abnormally expressed in hepatocytes is an HBV gene, an ANGPTL3 gene, or an APOC3 gene. In the context of the present disclosure, HBV gene refers to a gene whose sequence is shown as Genbank accession number nc_ 003977.1; ANGPTL3 gene refers to an mRNA sequence such as that shown in Genbank accession No. nm_ 014495.3; the APOC3 gene refers to a gene whose mRNA sequence is shown in Genbank accession No. nm_ 000040.1.
In some embodiments, a "target sequence" is a target mRNA. In the context of the present disclosure, "target mRNA" refers to mRNA corresponding to a gene that is abnormally expressed in hepatocytes, either as mRNA corresponding to an overexpressed gene or as mRNA corresponding to an underexpressed gene. Since most diseases result from overexpression of mRNA, in the present disclosure, target mRNA refers in particular to mRNA corresponding to the overexpressed gene. In some embodiments of the disclosure, the target mRNA may be mRNA corresponding to the genes of ApoB, apoC, ANGPTL, PCSK9, SCD1, FVII, p53, HBV, HCV, etc., corresponding to the genes expressed abnormally as described above. In some embodiments, the target mRNA may be mRNA transcribed from the corresponding HBV gene, or mRNA corresponding to the ANGPTL3 gene, or mRNA corresponding to the APOC3 gene.
P in formula A59 may be attached to any possible position in the oligonucleotide sequence, for example, to any one nucleotide of the oligonucleotide. In some embodiments, the functional oligonucleotide in the oligonucleotide conjugates of the present disclosure is a single stranded oligonucleotide (e.g., a single stranded RNA or an aptamer), at which point P in formula a59 may be attached to the end of the single stranded oligonucleotide, which refers to the first 4 nucleotides of the single stranded oligonucleotide from one end. In some embodiments, P in formula a59 is attached to the end of the single stranded oligonucleotide.
In some embodiments, the functional oligonucleotides in the oligonucleotide conjugates of the present disclosure are double-stranded oligonucleotides (e.g., siRNA, microRNA or DNA) comprising a sense strand and an antisense strand. In some embodiments, P in formula a59 is attached to the end of the sense strand or the antisense strand in the double-stranded oligonucleotide, the end referring to the first 4 nucleotides from one end in the sense strand or the antisense strand, in one embodiment, P in formula a59 is attached to the end of the sense strand or the antisense strand; in a further embodiment, P in formula a59 is attached to the 3' end of the sense strand. In the case where P in formula A59 is attached to the above-described position of the sense strand of a double-stranded oligonucleotide, the oligonucleotide conjugates provided by the present disclosure, upon entry into a cell, upon unwinding, can release the separate double-stranded oligonucleotide antisense strand to block the process of translation of the protein by the target mRNA, inhibiting expression of the particular gene.
P in formula A59 may be attached to any possible position on the nucleotide in the oligonucleotide sequence, for example, the 5' position of the nucleotide, the 2' position of the nucleotide, the 3' position of the nucleotide or the base of the nucleotide. In some embodiments, P in formula a59 may be attached to the 2', 3', or 5' position of a nucleotide in the oligonucleotide sequence by formation of a phosphodiester bond. In some embodiments, P in formula A59 is attached to an oxygen atom formed after dehydrogenation of the 3' -hydroxyl group of the 3' -terminal nucleotide of the sense strand in the double-stranded oligonucleotide sequence (in which case P in A59 can also be considered as P in the phosphate group contained in the siRNA), or P in formula A59 is attached to the nucleotide by substitution of hydrogen in the 2' -hydroxyl group of one nucleotide in the sense strand in the double-stranded oligonucleotide sequence, or P in formula A59 is attached to the nucleotide by substitution of hydrogen in the 5' -hydroxyl group of the 5' -terminal nucleotide of the sense strand in the double-stranded oligonucleotide sequence.
Without wishing to be limited, in the following embodiments and examples, the case where the functional oligonucleotides in the oligonucleotide conjugates of the present disclosure are small interfering RNAs (sirnas) is described in detail. At this time, the oligonucleotide conjugate of the present disclosure is an siRNA conjugate. In the context of this document, for convenience of description, the siRNA conjugates in these embodiments are also referred to as the siRNA conjugates of the present disclosure. This does not represent that the oligonucleotides in the oligonucleotide conjugates of the present disclosure may only be siRNA, but rather that the oligonucleotides may even be alternative drugs of the present disclosure or well known to those skilled in the art. Based on the detailed description of the siRNA conjugates, it is contemplated that other active agents or functional oligonucleotides will have similar effects when conjugated to the conjugate molecules provided by the present disclosure.
It is well known to those skilled in the art that siRNA contains a nucleotide group as a basic structural unit, the nucleotide group containing a phosphate group, a ribose group, and a base. Typically active, i.e., functional, siRNAs are about 12-40 nucleotides in length, in some embodiments about 15-30 nucleotides in length, each of which can independently be a modified or unmodified nucleotide, and at least one of which is a modified nucleotide for increased stability.
The inventors of the present disclosure found that the sirnas described in the following embodiments have higher activity and/or stability, and thus can be the object of the invention of the sirnas in the present disclosure.
In some embodiments, each nucleotide in an siRNA conjugate of the present disclosure (hereinafter also referred to as an siRNA of the present disclosure) is independently a modified or unmodified nucleotide, the siRNA comprising a sense strand and an antisense strand, wherein the sense strand comprises nucleotide sequence 1 and the antisense strand comprises nucleotide sequence 2, the nucleotide sequence 1 and the nucleotide sequence 2 are each 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 nucleotides in length, and are at least partially reverse-complementary to form a complementary duplex region, at least a portion of the nucleotide sequence 2 is complementary to a first stretch of nucleotide sequence that is a stretch of nucleotide sequence in a target mRNA.
In some embodiments, the siRNA of the present disclosure refers to an siRNA capable of inhibiting at least 50% hepatitis b virus gene expression, at least 50% angiopoietin-like protein 3 gene expression, or at least 50% apolipoprotein C3 gene expression at a concentration of 3 mg/kg. In some embodiments, the siRNA of the present disclosure is capable of inhibiting at least 55%, 60%, 65%, 70%, 75% or 80% hbv gene, ANGPTL3 gene or APOC3 gene expression at a concentration of 3 mg/kg.
In some embodiments, the nucleotide sequence 1 is equal in length to the first stretch of nucleotide sequences and does not differ by more than 3 nucleotides; the nucleotide sequence 2 is equal to the nucleotide sequence B in length and does not differ by more than 3 nucleotides; the nucleotide sequence B is a nucleotide sequence which is completely reverse complementary to the first nucleotide sequence. Without wishing to be limited, these specific nucleotide differences do not significantly reduce the target gene inhibition capacity of the siRNA conjugates, and these siRNA conjugates comprising the specific nucleotide differences are also within the scope of the present disclosure.
In some embodiments, the nucleotide sequence 1 and the nucleotide sequence 2 are substantially reverse complementary, substantially fully reverse complementary, or fully reverse complementary.
In some embodiments, the nucleotide sequence 1 differs from the first stretch of nucleotides by no more than 1 nucleotide, and/or the nucleotide sequence 2 differs from the nucleotide sequence B by no more than 1 nucleotide. In some embodiments, the nucleotide difference between the nucleotide sequence 2 and the nucleotide sequence B comprises a difference in the Z ' position of the first nucleotide on the nucleotide sequence 2 in a 5' end to 3' end direction. In some embodiments, the last nucleotide Z on the nucleotide sequence 1 is a nucleotide complementary to Z ' in a 5' end to 3' end orientation.
In some embodiments, the sense strand further comprises nucleotide sequence 3, the antisense strand further comprises nucleotide sequence 4, the nucleotide sequence 3 and the nucleotide sequence 4 are equal in length and each is 1-4 nucleotides, the nucleotide sequence 3 is linked at the 5 'end of the nucleotide sequence 1, and the nucleotide sequence 4 is linked at the 3' end of the nucleotide sequence 2, the nucleotide sequence 4 is complementary to a second nucleotide sequence that is adjacent to the first nucleotide sequence and is identical in length to the nucleotide sequence 4 in the target mRNA. In some embodiments, the nucleotide sequence 3 and the nucleotide sequence 4 are substantially fully reverse complementary or fully reverse complementary. Thus, the sense and antisense strands may be 19-23 nucleotides in length.
In some embodiments, the siRNA of the present disclosure further comprises a nucleotide sequence 5, the nucleotide sequence 5 being 1 to 3 nucleotides in length, attached to the 3 'end of the antisense strand, thereby constituting a 3' overhang of the antisense strand; in some embodiments, the nucleotide sequence 5 is 1 or 2 nucleotides in length. Thus, in some embodiments, the ratio of the lengths of the sense strand and the antisense strand of the siRNAs of the present disclosure can be 19/20, 19/21, 20/22, 21/23, 22/24, 23/24, or 23/25.
In one embodiment, the nucleotide sequence 5 is 2 nucleotides in length and in the 5 'to 3' end direction, the nucleotide sequence 5 is a contiguous 2 deoxythymidine nucleotides, a contiguous 2 uracil nucleotides, or is complementary to a third nucleotide sequence that is adjacent to the first nucleotide sequence or adjacent to the second nucleotide sequence in the target mRNA and is equal in length to the nucleotide sequence 5. In one embodiment, the ratio of the length of the sense strand to the length of the antisense strand of the siRNA of the present disclosure is 19/21 or 21/23, at which time the siRNA of the present disclosure has better hepatocyte mRNA silencing activity.
In some embodiments, the nucleotides in the siRNA of the present disclosure are each independently modified or unmodified nucleotides. In some embodiments, the siRNA of the present disclosure does not contain modified nucleotide groups; in some embodiments, the siRNA of the present disclosure contains modified nucleotide groups.
Currently, there are a variety of ways available in the art for modifying siRNA, including backbone modifications (also known as internucleotide linkage modifications, such as phosphate group modifications), ribose group modifications, and base modifications, among others (see, e.g., watts, J.K., G.F.Deleavey and M.J.damha, chemically modified siRNA: tools and applications. Drug discovery Today,2008.13 (19-20): p.842-55, the entire contents of which are incorporated herein by reference).
In the context of the present disclosure, the term "modified nucleotide" is used to refer to a nucleotide or nucleotide analogue in which the ribosyl group of the nucleotide is modified, such as where the 2' -hydroxyl group is replaced with another group, or where the base on the nucleotide is a modified base.
In some embodiments of the disclosure, at least one nucleotide in the sense strand or the antisense strand is a modified nucleotide, and/or at least one phosphate group is a phosphate group having a modification group. In other words, at least a portion of the phosphate groups and/or ribose groups in at least one single-stranded phosphate-sugar backbone in the sense strand and the antisense strand are phosphate groups and/or ribose groups (or modified phosphate groups and/or modified ribose groups) having a modifying group. In some embodiments of the disclosure, all nucleotides in the sense strand and/or the antisense strand are modified nucleotides.
In some embodiments, each nucleotide in the sense strand and the antisense strand is independently a fluoro-modified nucleotide or a non-fluoro-modified nucleotide.
The fluoro-modified nucleotide refers to a nucleotide in which the hydroxyl group at the 2' -position of the ribosyl group of the nucleotide is substituted with fluorine, and has a structure represented by the following formula (807).
Non-fluoro modified nucleotide refers to a nucleotide or nucleotide analogue formed by substituting the hydroxyl group at the 2' -position of the ribosyl of the nucleotide with a non-fluoro group. In some embodiments, each non-fluoro modified nucleotide is independently selected from one of the nucleotides or nucleotide analogs formed by substitution of the hydroxyl group at the 2' position of the ribosyl of the nucleotide with a non-fluoro group.
Nucleotides in which the hydroxyl group at the 2 '-position of the ribosyl group is substituted with a non-fluorine group are well known to those skilled in the art and may be selected from one of 2' -alkoxy-modified nucleotides, 2 '-substituted alkoxy-modified nucleotides, 2' -alkyl-modified nucleotides, 2 '-substituted alkyl-modified nucleotides, 2' -amino-modified nucleotides, 2 '-substituted amino-modified nucleotides, 2' -deoxynucleotides.
In some embodiments, the 2 '-alkoxy-modified nucleotide is a methoxy-modified nucleotide (2' -OMe), as shown in formula (808). The 2' -substituted alkoxy-modified nucleotide may be, for example, a 2' -O-methoxyethyl-modified nucleotide (2 ' -MOE), as shown in formula (809). 2 '-amino modified nucleotide (2' -NH) 2 ) As shown in equation (810). The 2' -Deoxynucleotide (DNA) is represented by formula (811).
Nucleotide analogs refer to groups that are capable of replacing nucleotides in a nucleic acid, but that differ in structure from adenine ribonucleotide, guanine ribonucleotide, cytosine ribonucleotide, uracil ribonucleotide, or thymine. In some embodiments, the nucleotide analog may be, for example, an isonucleotide, a bridged nucleic acid (bridged nucleic acid, abbreviated BNA) nucleotide, or an acyclic nucleotide.
BNA nucleotides refer to constrained or inaccessible nucleotides. BNA may contain a five-, six-, or seven-membered ring bridging structure with "fixed" C3' -endo-saccharides tucked. The bridge is typically incorporated at the 2'-, 4' -position of the ribose ring to provide a 2',4' -BNA nucleotide, such as LNA, ENA, cret BNA, etc., where LNA is shown as formula (812), ENA is shown as formula (813), cret BNA is shown as formula (814).
Acyclic nucleotides are a class of nucleotides in which the sugar ring of the nucleotide is opened, such as Unlocking Nucleic Acid (UNA) nucleotides or Glycerol Nucleic Acid (GNA) nucleotides, where UNA is represented by formula (815) and GNA is represented by formula (816).
Wherein R is selected from H, OH or alkoxy (O-alkyl).
An isopucleotide refers to a compound in which the position of a base on the ribose ring is changed in a nucleotide, for example, a compound in which a base is shifted from the 1' -position to the 2' -position or the 3' -position of the ribose ring, as shown in formula (817) or (818).
Wherein Base represents a Base, such as A, U, G, C or T; r is selected from H, OH, F or a non-fluorine group as described above.
In some embodiments, the nucleotide analog is selected from one of an iso-nucleotide, LNA, ENA, cET, UNA, and GNA. In some embodiments, each non-fluoro modified nucleotide is a methoxy modified nucleotide, which refers to a nucleotide formed by substitution of the 2' -hydroxy group of the ribosyl group with a methoxy group.
In the above and in the following, the meaning of "fluoro modified nucleotide", "2 '-fluoro modified nucleotide", "nucleotide in which the 2' -hydroxyl group of the ribose group is replaced with fluorine" and "2 '-fluoro ribose group" are the same, and refer to a compound having a structure as shown in formula (807) formed by replacing the 2' -hydroxyl group of the nucleotide with fluorine; "methoxy modified nucleotide", "2 '-methoxy modified nucleotide", "nucleotide in which the 2' -hydroxy group of the ribose group is replaced by methoxy" and "2 '-methoxyribosyl" are the same meaning, and each refers to a nucleotide in which the 2' -hydroxy group of the ribose group is replaced by methoxy to form a structure shown in formula (808).
In some embodiments, the siRNA of the present disclosure is an siRNA with modifications of: the nucleotides at positions 7, 8 and 9 of the nucleotide sequence 1 in the sense strand of the siRNA are-fluoro modified nucleotides according to the direction from the 5 'end to the 3' end, and the nucleotides at the rest positions in the sense strand are methoxy modified nucleotides; in the antisense strand, the nucleotides at the 2 nd, 6 th, 14 th and 16 th positions of the nucleotide sequence 2 are fluoro modified nucleotides, and the nucleotides at the rest positions in the antisense strand are methoxy modified nucleotides; in some embodiments, the siRNA of the present disclosure is an siRNA with modifications of: or according to the direction from the 5 'end to the 3' end, the nucleotides at the 5 th, 7 th, 8 th and 9 th positions of the nucleotide sequence 1 in the sense strand of the siRNA are fluoro modified nucleotides, and the nucleotides at the rest positions in the sense strand are methoxy modified nucleotides; in the antisense strand, the nucleotides at positions 2, 6, 8, 9, 14 and 16 of the nucleotide sequence 2 are fluoro-modified nucleotides, and the nucleotides at the rest positions in the antisense strand are methoxy-modified nucleotides; in some embodiments, the siRNA of the present disclosure is an siRNA with modifications of: the nucleotides at positions 7, 8 and 9 of the nucleotide sequence 1 in the sense strand of the siRNA are fluoro-modified nucleotides, the nucleotides at the remaining positions in the sense strand are methoxy-modified nucleotides, and the nucleotides at positions 2, 6, 14 and 16 of the nucleotide sequence 2 in the antisense strand of the siRNA are fluoro-modified nucleotides, in the 5 'to 3' direction.
In some embodiments of the siRNA described in the present disclosure, the nucleotide contains a phosphate group modification. In the context of the present disclosure, the phosphate group modification is in one embodiment a phosphorothioate modification as shown in formula (801) below, i.e., substitution of one sulfur atom for a non-bridging oxygen atom in the phosphodiester linkage, thereby replacing the phosphodiester linkage with a phosphorothioate linkage. The modification can stabilize the structure of siRNA, and maintain high specificity and high affinity of base pairing.
According to some embodiments of the present disclosure, the siRNA wherein the phosphorothioate linkage is present at least one of the group consisting of: between the first and second nucleotides at either end of the sense strand or the antisense strand; between the second and third nucleotides at either end of the sense strand or the antisense strand; or any combination of the above. In some embodiments, phosphorothioate linkages are present at all of the above positions except the 5' end of the sense strand. In some embodiments, phosphorothioate linkages are present at all of the above positions except the 3' end of the sense strand. In some embodiments, the phosphorothioate linkage is present in at least one of the following positions:
A linkage between nucleotide 1 and nucleotide 2 of the 5' terminal end of the sense strand;
a linkage between nucleotide 2 and nucleotide 3 of the 5' terminal end of the sense strand;
a linkage between nucleotide 1 and nucleotide 2 of the 3' -terminal end of the sense strand;
a linkage between nucleotide 2 and nucleotide 3 of the 3' -terminal end of the sense strand;
a linkage between nucleotide 1 and nucleotide 2 of the 5' terminal end of the antisense strand;
a linkage between nucleotide 2 and nucleotide 3 of the 5' terminal end of the antisense strand;
a linkage between nucleotide 1 and nucleotide 2 of the 3' -terminal end of the antisense strand; and
the 3' -terminal end of the antisense strand is linked between nucleotide 2 and nucleotide 3.
According to some embodiments of the present disclosure, the 5' -terminal nucleotide of the antisense strand sequence of the siRNA molecule is a 5' -phosphonucleotide or a 5' -phosphoanalog modified nucleotide.
In some embodiments, the 5' -phosphate nucleotide may have a structure represented by formula (802):
meanwhile, the kinds of commonly used 5' -phosphate analogue-modified nucleotides are well known to those skilled in the art, for example, anastasia Khvorova and Jonathan K.Watts, the chemical evolution of oligonucleotide therapies of clinical units.Nature Biotechnology,2017,35 (3): 4 nucleotides as shown in the following formulas (803) to (806) are disclosed in 238-48:
Wherein R represents a group selected from the group consisting of H, OH, F, and methoxy;
base represents a Base selected from A, U, C, G or T.
In one embodiment, the 5 '-phosphonucleotide or 5' -phosphoanalogue modified nucleotide is a vinyl phosphate (E-vinylphosphate, E-VP) -containing nucleotide represented by formula (803), a 5 '-phospho-modified nucleotide represented by formula (802), or a 5' -phosphorothioate-modified nucleotide represented by formula (805).
The inventors of the present disclosure unexpectedly found that the siRNA conjugates of the present disclosure, while having significantly improved serum stability, also exhibit target mRNA silencing activity that is not significantly reduced, as well as excellent gene expression inhibition effects. According to one embodiment of the present disclosure, the oligonucleotide conjugates of the present disclosure are siRNA conjugates comprising, for example, the siRNAs shown in tables 1A-4E:
TABLE 1A
TABLE 1B
TABLE 1C
TABLE 1D
TABLE 1E
TABLE 2A
TABLE 2B
TABLE 2C
TABLE 2D
/>
TABLE 2E
/>
TABLE 3A
TABLE 3B
TABLE 3C
TABLE 3D
TABLE 3E
TABLE 4A
TABLE 4B
TABLE 4C
TABLE 4D
TABLE 4E
* S: a sense strand; AS: antisense strand
Wherein capital C, G, U, A indicates the base composition of the nucleotide; the lower case letter m indicates that the adjacent nucleotide to the left of the letter m is a 2' -methoxy modified nucleotide; the lower case letter f indicates that the adjacent nucleotide to the left of the letter f is a 2' -fluoro modified nucleotide; the lower case letter s indicates that the linkage between two nucleotides adjacent to the letter s is a phosphorothioate linkage; p1 represents that the adjacent nucleotide to the right of P1 is a 5' -phosphonucleotide or a 5' -phosphoanalog modified nucleotide, in one embodiment a vinyl phosphate modified nucleotide (denoted as VP in the examples below), a 5' -phospho modified nucleotide (denoted as P in the examples below), or a phosphorothioate modified nucleotide (denoted as Ps in the examples below).
It will be apparent to those skilled in the art that the methods of preparing nucleoside monomers having corresponding modifications and methods of introducing modified nucleotide groups into siRNA described in the present disclosure can be incorporated by using nucleoside monomers having corresponding modifications as well as methods of introducing modified nucleotide groups into siRNA are well known to those skilled in the art. All modified nucleoside monomers are commercially available or can be prepared using known methods.
Preparation of oligonucleotide conjugates
Any reasonable synthetic route can be used to prepare the oligonucleotide conjugates of the present disclosure.
For example, the oligonucleotide conjugates of the present disclosure may be prepared by a method comprising sequentially ligating nucleoside monomers in a 3 'to 5' direction under conditions of phosphoramidite solid phase synthesis, respectively, according to the nucleotide species and sequence of the oligonucleotides, the ligating of each nucleoside monomer comprising a deprotection, coupling, capping, oxidation, or sulfidation four-step reaction; in some embodiments, the method further comprises contacting the compound of formula (101) with a nucleoside monomer or a nucleotide sequence attached to a solid support in the presence of a coupling reagent and coupling reaction conditions, such that the compound of formula (101) is attached to the nucleotide sequence via a coupling reaction.
In some embodiments, the method further comprises a step of removing the protecting group and cleaving from the solid support, a separation and purification step, and optionally an annealing step.
In some embodiments, the oligonucleotide is a double-stranded oligonucleotide, and the method of making comprises the steps of: contacting a compound shown in a formula (101) with a first nucleoside monomer at the 3' -end of a sense strand or an antisense strand under coupling reaction conditions and in the presence of a coupling reagent, connecting the compound shown in the formula (101) with a first nucleotide in a sequence, and sequentially connecting the nucleoside monomers in a 3' -to 5' -direction to synthesize the sense strand or the antisense strand of the double-stranded oligonucleotide; wherein the (101) compound is R 2 The compound of formula (101) having a structure represented by formula (C1 ') or (C3'), wherein the compound of formula (101) is deprotected before the compound is connected to the first nucleoside monomer; the connection of each nucleoside monomer comprises four steps of deprotection, coupling, capping, oxidation or vulcanization reaction; obtaining a sense strand or an antisense strand of the nucleic acid to which the conjugate molecule is attached; sequentially connecting nucleoside monomers in a 3 'to 5' direction to synthesize the other strand of the double-stranded oligonucleotide, wherein the connection of each nucleoside monomer comprises four steps of deprotection, coupling, capping, oxidation or vulcanization; removing protecting group, cutting with solid phase carrier, separating and purifying to obtain sense strand and antisense strand of nucleic acid Chain, annealing.
In some embodiments, the oligonucleotide is a double-stranded oligonucleotide, and the method of making comprises the steps of: sequentially connecting nucleoside monomers according to the nucleotide types and sequences of a sense strand or an antisense strand in the double-stranded oligonucleotide and the direction from 3 'to 5' to synthesize the sense strand and the antisense strand, wherein the connection of each nucleoside monomer comprises four steps of deprotection, coupling, capping, oxidation or sulfuration reaction to obtain the sense strand connected to a solid carrier and the antisense strand connected to the solid carrier; contacting a compound represented by formula (101) with a sense strand attached to a solid support or an antisense strand attached to a solid support in the presence of a coupling reagent under coupling reaction conditions to thereby attach the compound of formula (101) to the sense strand or the antisense strand, wherein the compound of formula (101) is R 1 A compound of formula (101) containing phosphoramidite as the 1 st functional group; removing protecting groups, cutting with a solid phase carrier, separating and purifying to obtain a sense strand or an antisense strand of the oligonucleotide, and annealing, wherein the sense strand or the antisense strand of the oligonucleotide is connected with a conjugated molecule.
In a specific embodiment, P in formula a59 is attached to the 3' end of the sense strand in the siRNA, and the method of preparing the siRNA conjugate of the disclosure comprises:
(1) Removing the hydroxyl protecting group R from the solid support-attached compound of formula (101) (hereinafter, also referred to as a solid support-attached conjugate molecule) k The method comprises the steps of carrying out a first treatment on the surface of the Contacting the conjugate molecule connected with the solid phase carrier with a nucleoside monomer under the condition of coupling reaction and in the presence of a coupling reagent to obtain the nucleoside monomer connected with the solid phase carrier through the conjugate molecule;
(2) Synthesizing the sense strand of the siRNA by a phosphoramidite solid phase synthesis method according to the 3'-5' direction starting from the nucleoside monomer attached to the solid phase carrier through the conjugate molecule;
(3) Synthesizing antisense strand of siRNA through phosphoramidite solid phase synthesis method;
(4) The sense and antisense strands of the siRNA were isolated and annealed to obtain the siRNA conjugates of the present disclosure.
Wherein in step (1), the connecting solids are removedProtecting group R in conjugate molecule of phase carrier k Comprising contacting a compound of formula (101) with a deprotection reagent under deprotection conditions. Deprotection conditions include a temperature of 0 to 50 ℃, in some embodiments 15 to 35 ℃, a reaction time of 30 to 300 seconds, in some embodiments 50 to 150 seconds, and the deprotection reagent may be selected from one or more of trifluoroacetic acid, trichloroacetic acid, dichloroacetic acid, monochloroacetic acid, in some embodiments dichloroacetic acid. The molar ratio of deprotecting reagent to compound of formula (101) is from 10:1 to 1000:1, in some embodiments from 50:1 to 500:1.
The coupling reaction conditions and coupling reagents may use any conditions and reagents capable of effecting the coupling reactions described above. In some embodiments, the same conditions and reagents are used as the coupling reaction in the solid phase synthesis method employed.
In some embodiments, the conditions of the coupling reaction include a reaction temperature of 0 to 50 ℃, in some embodiments 15 to 35 ℃. The molar ratio of compound of formula (101) to nucleoside monomer is from 1:1 to 1:50, in some embodiments from 1:2 to 1:5; the molar ratio of the compound of formula (101) to the coupling reagent is from 1:1 to 1:50, in some embodiments from 1:3 to 1:10, and the reaction time is from 200 to 3000 seconds, in some embodiments from 500 to 1500 seconds. The coupling reagent is selected from one or more of 1H-tetrazole, 5-ethylthio 1H-tetrazole, 5-benzylthio 1H-tetrazole, and in some embodiments 5-ethylthio 1H-tetrazole. The coupling reaction may be carried out in an organic solvent selected from one or more of anhydrous acetonitrile, anhydrous DMF, anhydrous dichloromethane, in some embodiments, anhydrous acetonitrile. The organic solvent is used in an amount of 3 to 50L/mol, and in some embodiments 5 to 20L/mol, relative to the compound of formula (101).
In step (2), the sense strand S of the siRNA conjugate is synthesized in the 3'-5' direction by the method of solid phase synthesis of phosphoramidite nucleic acid, starting with the nucleoside monomer attached to the solid support via the conjugate molecule prepared in the above step. At this point, the conjugate molecule is attached to the 3' end of the resulting sense strand.
Other conditions for the solid phase synthesis described in steps (2) and (3) include deprotection conditions for nucleoside monomers, types and amounts of deprotection reagents, coupling reaction conditions, types and amounts of coupling reagents, conditions for capping reactions, types and amounts of capping reagents, oxidation reaction conditions, types and amounts of oxidizing reagents, sulfidation reaction conditions, sulfidation reagents and amounts using various reagents, amounts and conditions conventionally used in the art.
For example, in some embodiments, the solid phase synthesis described in steps (2) and (3) may use the following conditions:
the nucleoside monomer deprotection conditions include a temperature of from 0 to 50 ℃, in some embodiments from 15 to 35 ℃, for a reaction time of from 30 to 300 seconds, in some embodiments from 50 to 150 seconds, and the deprotection reagent may be selected from one or more of trifluoroacetic acid, trichloroacetic acid, dichloroacetic acid, monochloroacetic acid, and dichloroacetic acid in some embodiments. The molar ratio of deprotection reagent to 4,4' -dimethoxytrityl protecting group on the solid support is from 2:1 to 100:1, and in some embodiments from 3:1 to 50:1.
Coupling reaction conditions include a temperature of 0 to 50 ℃, in some embodiments 15 to 35 ℃, and a molar ratio of nucleic acid sequence to nucleoside monomer attached to the solid support of 1:1 to 1:50, in some embodiments 1:5 to 1:15; the molar ratio of nucleic acid sequence to coupling reagent attached to the solid support is 1:1 to 1:100, in some embodiments 1:50 to 1:80, and the reaction time and coupling reagent selection is the same as described above.
The capping reaction conditions include a temperature of 0-50 ℃, in some embodiments 15-35 ℃, a reaction time of 5-500 seconds, in some embodiments 10-100 seconds, and the capping reagent is selected as described above. The molar ratio of the total amount of capping reagent to the nucleic acid sequence attached to the solid support is from 1:100 to 100:1, in some embodiments from 1:10 to 10:1. Where equimolar amounts of acetic anhydride to N-methylimidazole are used for the capping reagent, the molar ratio of acetic anhydride, N-methylimidazole, and nucleic acid sequences attached to the solid support is 1:1:10 to 10:10:1, in some embodiments 1:1:2 to 2:2:1.
The oxidation reaction conditions include a temperature of 0-50 ℃, in some embodiments 15-35 ℃, a reaction time of 1-100 seconds, in some embodiments 5-50 seconds, and an oxidizing agent, in some embodiments iodine (in further embodiments provided in the form of iodine water). The molar ratio of oxidizing reagent to nucleic acid sequence attached to the solid support during the coupling step is 1:1 to 100:1, in some embodiments 5:1 to 50:1. In some embodiments, the oxidation reaction is performed in a mixed solvent of tetrahydrofuran: water: pyridine=3:1:1 to 1:1:3. The sulfiding reaction conditions include a temperature of 0-50 ℃, in some embodiments 15-35 ℃, a reaction time of 50-2000 seconds, in some embodiments 100-1000 seconds, and a sulfiding agent of hydrogenation Huang Yuansu in some embodiments. The molar ratio of sulfiding reagent to nucleic acid sequence attached to the solid support during the coupling step is from 10:1 to 1000:1, in some embodiments from 10:1 to 500:1. In some embodiments, the sulfidation reaction is performed in a mixed solvent of acetonitrile: pyridine=1:3-3:1.
In accordance with the methods provided by the present disclosure, after ligating all nucleoside monomers, the methods further comprise isolating the sense strand and the antisense strand of the siRNA prior to annealing. Methods of isolation are well known to those skilled in the art and generally involve cleavage of the synthesized nucleotide sequence from the solid support, removal of protecting groups on the base, phosphate and ligand, purification and desalting.
The nucleotide sequence obtained by synthesis is cut off from the solid phase carrier, and the protecting groups on the base, the phosphate group and the ligand are removed according to the conventional cutting and deprotection method in siRNA synthesis. For example, the obtained nucleotide sequence linked to the solid phase carrier is contacted with concentrated ammonia water; in the deprotection process, the protecting group YCOO of the A46-A54 group is converted to a hydroxyl group, S 1 Conversion of the group to the corresponding M 1 A group, thereby producing a conjugate represented by formula (201). Wherein, the concentrated ammonia water is 25-30 wt% ammonia water, and the dosage of the concentrated ammonia water is 0.2 ml/mu mol-0.8 ml/mu mol compared with the target siRNA sequence.
In the presence of at least one 2'-TBDMS protection on the synthesized nucleotide sequence, the method further comprises contacting the solid support-removed nucleotide sequence with triethylamine trihydrofluoride to remove the 2' -TBDMS protection. At this time, the corresponding nucleoside having a free 2' -hydroxy group in the resulting target siRNA sequence. The amount of pure triethylamine-tricofluoride salt is 0.4 ml/. Mu.mol-1.0 ml/. Mu.mol compared with the target siRNA sequence. This resulted in the siRNA conjugates of the present disclosure.
Methods of purification and desalination are well known to those skilled in the art. For example, purification of nucleic acids can be accomplished by gradient elution with NaBr or NaCl using preparative ion chromatography purification columns; after the product is collected and combined, the desalination can be performed by adopting a reversed phase chromatographic purification column.
The purity and molecular weight of the nucleic acid sequence can be detected at any time during the synthesis process, so that the quality of the synthesis can be better controlled, and the detection method is well known to those skilled in the art. For example, the purity of the nucleic acid can be detected by ion exchange chromatography and the molecular weight can be determined by liquid chromatography.
Methods of annealing are also well known to those skilled in the art. For example, the synthesized sense strand (S strand) and antisense strand (AS strand) may simply be mixed in equimolar ratio in water for injection and heated to 70-95℃and then cooled at room temperature to form a double-stranded structure through hydrogen bonding. This resulted in the siRNA conjugates of the present disclosure.
After obtaining the conjugates of the present disclosure, in some embodiments, the synthesized siRNA conjugates can also be characterized by means of molecular weight detection, etc., using methods such as liquid chromatography, etc., to determine that the synthesized siRNA conjugates are targeted siRNA conjugates, and that the sequences of the synthesized sirnas correspond to the sequences of the sirnas to be synthesized, such as those listed in tables 1A-4E above.
Use of conjugates of the present disclosure
As shown in the present disclosure, the conjugates can deliver an active agent to cells for the treatment or prevention of a disease or condition that may require such delivery. Without wishing to be bound by any theory, we believe that the spatial arrangement of the conjugate molecules is particularly effective in targeting cell surface receptors, thereby bringing the supported active agent into contact with the cells. In some embodiments, such conjugates are oligonucleotide conjugates directed against hepatocytes.
In some embodiments, the oligonucleotide conjugates of the present disclosure have excellent liver targeting specificity, and thus are capable of efficiently delivering conjugated functional oligonucleotides to the liver, thereby effectively modulating the expression of specific genes within hepatocytes. Thus, the oligonucleotide conjugate of the present disclosure has a wide application prospect.
According to some embodiments of the present disclosure, the present disclosure provides the use of an oligonucleotide conjugate of the present disclosure in the manufacture of a medicament for the treatment and/or prevention of a pathological condition or disease caused by expression of a specific gene in a hepatocyte. The specific gene may be an endogenous gene expressed in the liver or a pathogen gene propagated in the liver. In some embodiments, the specific gene is selected from the group consisting of ApoB, apoC, ANGPTL, PCSK9, SCD1, FVII, p53, HBV, HCV, and the like. In some embodiments, the specific gene is selected from the group consisting of a hepatitis b virus gene, an angiopoietin-like protein 3 gene, or an apolipoprotein C3 gene. Accordingly, the disease is selected from the group consisting of chronic liver disease, hepatitis, liver fibrosis disease, liver hyperplasia disease and dyslipidemia. In some embodiments, the dyslipidemia is hypercholesterolemia, hypertriglyceridemia, or atherosclerosis. In some embodiments, the conjugates provided by the present disclosure may also be used to treat other liver diseases, including diseases characterized by unwanted cell proliferation, hematological diseases, metabolic diseases, and diseases characterized by inflammation. The proliferative disease of the liver may be a benign or malignant disease, such as cancer, hepatocellular carcinoma (HCC), liver metastasis or hepatoblastoma. The hematological or inflammatory disease of the liver may be a disease involving coagulation factors, complement mediated inflammation or fibrosis. Metabolic diseases of the liver include dyslipidemia and irregularities in glucose regulation. In some embodiments, liver disease is treated by administering one or more oligonucleotides having a high homology to the sequences of genes involved in liver disease.
According to another embodiment of the present disclosure, the present disclosure provides a method of inhibiting expression of a specific gene in a hepatocyte, the method comprising contacting the siRNA conjugate of the present disclosure with the hepatocyte.
By administering the oligonucleotide conjugates of the present disclosure to a patient in need thereof, the prevention and/or treatment of a pathological condition or disease caused by the expression of a specific gene in hepatocytes can be achieved by a mechanism that regulates gene expression. Thus, the oligonucleotide conjugates of the present disclosure may be used for the prevention and/or treatment of, or for the manufacture of a medicament for the prevention and/or treatment of, said pathological condition or disease.
The term "administration" as used herein refers to placement of a conjugate into a patient by a method or route that results in at least partial localization of the conjugate, such as an oligonucleotide conjugate, to a desired site to produce a desired effect. Routes of administration suitable for the methods of the present disclosure include, but are not limited to, topical and systemic administration. In general, topical administration results in more oligonucleotide conjugate being delivered to a particular site than the entire body of the patient; whereas systemic administration results in delivery of the oligonucleotide conjugate to substantially the entire body of the patient. It is contemplated that the present disclosure aims to provide means for preventing and/or treating pathological conditions or diseases caused by the expression of specific genes in hepatocytes, in some embodiments, modes of administration that are capable of delivering drugs to the liver.
The administration to the patient may be by any suitable route known in the art including, but not limited to: oral or parenteral routes include intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), pulmonary, nasal, rectal and topical (including buccal and sublingual). The frequency of administration may be 1 or more times daily, weekly, biweekly, monthly, or annually.
The dosages of oligonucleotide conjugates described in the present disclosure may be dosages conventional in the art, which dosages may be determined in accordance with various parameters, particularly the age, weight and sex of the patient. Toxicity and efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, for example, by determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose that causes 50% of the maximum intensity of response in dose response, and the dose that causes 50% of the subjects to develop a positive response in mass response). The range of doses for human use can be derived based on data obtained from cell culture assays and animal studies.
Upon administration of the conjugates described in the present disclosure, for example, for male or female, 6-12 week old, C57BL/6J or C3H/hencrl vr mice weighing 18-25g, based on the amount of oligonucleotide in the oligonucleotide conjugate: for oligonucleotide conjugates of functional oligonucleotides and conjugate molecules, the amount of oligonucleotide delivered by the conjugate may be from 0.001 to 100mg/kg body weight, in some embodiments from 0.01 to 50mg/kg body weight, in some embodiments from 0.05 to 20mg/kg body weight, and in one particular embodiment from 0.1 to 10mg/kg body weight. Reference is made to the above amounts when administering the oligonucleotide conjugates described in the present disclosure.
In addition, by introducing the oligonucleotide conjugate of the present disclosure into a hepatocyte in which a specific gene is abnormally expressed, the purpose of inhibiting the expression of the specific gene in the hepatocyte can also be achieved by a mechanism of gene expression regulation. In some embodiments, the hepatocyte is a hepatitis cell, in some embodiments a hepg2.2.15 cell. In some embodiments, the liver cells may be selected from liver cancer cell lines such as Hep3B, hepG2, huh7, etc., or isolated primary liver cells, in some embodiments Huh7 liver cancer cells.
The amount of functional oligonucleotide in the provided oligonucleotide conjugates to inhibit the expression of a particular gene in hepatocytes using the methods provided by the present disclosure is readily determinable by one of skill in the art based on the desired effect. For example, in some embodiments, the oligonucleotide conjugate is an siRNA conjugate, and the amount of siRNA in the provided siRNA conjugate is such that: it is sufficient to reduce expression of the target gene and results in an extracellular concentration of 1pM to 1. Mu.M, or 0.01nM to 100nM, or 0.05nM to 50nM, or to about 5 nM. The amount required to achieve this local concentration will vary depending on a variety of factors including the method of delivery, the site of delivery, the number of cell layers between the site of delivery and the target cell or tissue, whether the delivery is local or systemic, etc. The concentration at the delivery site may be significantly higher than the concentration at the surface of the target cell or tissue.
Advantageous effects
In some embodiments, the double-stranded oligonucleotides, compositions, or oligonucleotide conjugates provided by the present disclosure may have higher stability, lower toxicity, and/or higher activity in vivo. In some embodiments, the double-stranded oligonucleotide provided by the present disclosure is a saRNA. In some embodiments, the saRNA, saRNA compositions, or saRNA conjugates provided herein exhibit an increase in target gene expression of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95% in vivo. In some embodiments, the double stranded oligonucleotides provided by the present disclosure are siRNA. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit a target gene expression inhibition of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95% in vivo. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an HBV gene expression inhibition rate of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95% in vivo. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an inhibition of HBV gene expression in vivo of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95%. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an inhibition of HBV gene expression in vivo of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95% in animal models. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an HBV surface antigen expression inhibition rate of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95% in vivo. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an ANGPTL3 gene expression inhibition of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95% in vivo. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an inhibition of expression of an ANGPTL3 gene in vivo of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95%. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an inhibition of intrahepatic ANGPTL3 gene expression in an animal model of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95%. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an inhibition of intrahepatic ANGPTL3 gene expression in human subjects of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95% in vivo. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an APOC3 gene expression inhibition of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95% in vivo. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an in vivo inhibition of intrahepatic APOC3 gene expression of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95%. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an in vivo inhibition of intrahepatic APOC3 gene expression of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95% in animal models. In some embodiments, the siRNA, siRNA compositions, or siRNA conjugates provided by the present disclosure exhibit an intra-hepatic APOC3 gene expression inhibition rate of at least 20%,30%,40%,50%,60%,70%,80%,90%, or 95% in human subjects in vivo. In some embodiments, the double-stranded oligonucleotides, compositions, or oligonucleotide conjugates provided by the present disclosure do not exhibit significant off-target effects. The off-target effect may be, for example, inhibition of normal gene expression of non-target genes. It is believed that the off-target effect is not significant if the binding/inhibition of off-target gene expression is less than 50%, 40%, 30%, 20% or 10% compared to the effect at the target gene.
According to one embodiment of the present disclosure, when the oligonucleotide is an siRNA that inhibits the expression of hepatitis b virus (hepatitis B virus, HBV) genes, the siRNA conjugate provided by the present disclosure is capable of efficiently delivering siRNA to the liver and exhibits excellent HBV gene expression inhibiting properties: can inhibit 81.54% -83.8% of HBV gene expression in liver of hepatitis B model mice at a dose of 1mg/kg while having low off-target effect. Meanwhile, the siRNA conjugate can also effectively reduce the HBV surface antigen expression in a hepatitis B model mouse, and can reach 92.2 percent of HBV surface antigen expression inhibition rate and 89.2 percent of HBV DNA inhibition rate at a dose of 3 mg/kg. In particular, the specific siRNA conjugates formed by the specific modified siRNA and the specific conjugate molecule provided by the present disclosure can continuously exhibit excellent HBV expression inhibition for an experimental time up to 140 days while the dosage is low, compared to the conjugates formed by the conjugate molecules provided by the prior art.
According to one embodiment of the present disclosure, when the oligonucleotide is an siRNA that inhibits expression of hepatitis b virus gene, the siRNA conjugate provided by the present disclosure is capable of effectively delivering siRNA to the liver and exhibits excellent HBV gene expression inhibiting properties: can inhibit HBV gene expression in liver of hepatitis B model mice by more than 75% at a single administration dosage of 1mg/kg while having low off-target effect. Meanwhile, the siRNA conjugate can also effectively reduce HBV surface antigen expression in a hepatitis B model mouse, and can reach 95.2% HBV surface antigen expression inhibition rate and 91.6% HBV DNA inhibition rate at a dose of 3 mg/kg. In particular, the specific siRNA conjugates formed by the specific modified siRNA and the specific conjugate molecule provided by the present disclosure can continuously exhibit excellent HBV expression inhibition for an experimental time of up to 84 days while at a low administration dose, compared to conjugates formed by the conjugate molecules provided by the prior art.
According to one embodiment of the present disclosure, when the oligonucleotide is an siRNA that inhibits the expression of an angiopoietin-like protein 3 (ANGPTL 3) gene, the siRNA conjugates provided by the present disclosure are capable of efficiently delivering the siRNA to the liver and exhibit excellent properties of inhibiting the expression of the ANGPTL3 gene: inhibiting ANGPTL3 gene expression in the liver of a high-fat model mouse by at least 48.9% at a dose of 1 mg/kg; at a dose of 3mg/kg, the gene inhibition rate is as high as 80.8%. In particular, the specific modified siRNA conjugates provided by the present disclosure and specific conjugate molecules form specific siRNA conjugates that exhibit superior gene suppression rates compared to conjugates formed by conjugate molecules provided by the prior art; furthermore, the specific siRNA conjugates provided by the present disclosure are capable of continuously exhibiting excellent ANGPTL3 expression inhibition and hypolipidemic effects for an experimental period of up to 49 days with low dose, low dosing frequency.
According to one embodiment of the present disclosure, when the oligonucleotide is an siRNA that inhibits expression of an apolipoprotein C3 (ApoC 3) gene, the siRNA conjugate provided by the present disclosure is capable of efficiently delivering siRNA to the liver and exhibits excellent ApoC3 gene expression inhibiting properties: at least 68.2% of APOC3 gene expression was inhibited in the liver of the high fat model mice at a dose of 3 mg/kg. In particular, the specific modified siRNA conjugates provided by the present disclosure and specific conjugate molecules form specific siRNA conjugates that exhibit superior gene suppression rates compared to conjugates formed by conjugate molecules provided by the prior art; furthermore, the specific siRNA conjugates provided by the present disclosure are capable of continuously exhibiting excellent blood lipid inhibitory effect for an experimental period of up to 65 days with low dose, low frequency of administration.
In certain embodiments, the siRNA conjugates described in the present disclosure also exhibit low animal level toxicity and good safety, e.g., in some embodiments, no significant toxic response is observed for the conjugates of the present disclosure even when administered up to 100-fold of the onset concentration (3 mg/kg as the onset concentration) in C57BL/6J mice.
The above examples illustrate that the oligonucleotide conjugates provided by the present disclosure are capable of effectively delivering functional oligonucleotides to the liver and maintaining activity in vivo for a long period of time, thereby effectively treating and/or preventing pathological conditions and diseases caused by the expression of specific genes in hepatocytes.
Additional features and advantages of the present disclosure will be set forth in the detailed description which follows.
Kit for detecting a substance in a sample
In another aspect, provided herein are kits comprising conjugates as described above.
In some embodiments, the kits provided herein comprise a container comprising the conjugate. In some embodiments, the kits provided herein comprise a container of pharmaceutically acceptable excipients. In some embodiments, the kits provided herein further comprise pharmaceutically acceptable excipients, such as stabilizers or preservatives. In some embodiments, the kits provided herein comprise at least one additional therapeutic agent. In some embodiments, the kit comprises at least one additional therapeutic agent in a container other than the conjugates described in the present disclosure. In some embodiments, the kit may comprise instructions for mixing the conjugate with pharmaceutically acceptable excipients (for inclusion of excipients) or other ingredients.
In the kits of the present disclosure, the conjugate and optional pharmaceutically acceptable excipients may be provided in any form, such as liquid form, dry form, or lyophilized form. In some embodiments, the oligonucleotide conjugate and optional pharmaceutically acceptable excipients are substantially pure and/or sterile. In some embodiments, sterile water is provided in a kit of the present disclosure.
Examples
The present disclosure will be described in detail by way of examples. Unless otherwise specified, reagents and media used in the following examples are commercially available, and the procedures for nucleic acid electrophoresis, real-time PCR, and the like used are carried out by the method described in Molecular Cloning (Cold Spring Harbor LBboratory Press (1989)).
HEK239A cells were supplied from the nucleic acid technology laboratory of the university of beijing institute of molecular medicine and cultured with DMEM complete medium (Hyclone) containing 20% fetal bovine serum (FBS, hyclone), 0.2v% blueberry diabody (penicillin-streptomycin, gibco, invitrogen). The cells were incubated at 37℃in an incubator with 5% CO2/95% air.
Huh7 cells were purchased from ATCC and cultured in DMEM complete medium (Hyclone) containing 10% fetal bovine serum (FBS, hyclone), 1% non-essential amino acids (NEAA, corning) at 37℃in an incubator containing 5% CO2/95% air.
Unless otherwise stated, when cells were transfected with the siRNA conjugates synthesized in preparation examples 7-9 below, lipofectamine 2000 (Invitrogen) was used as the transfection reagent, and the specific procedure was referred to the manufacturer's instructions.
Unless otherwise indicated, the reagent ratios provided below are all calculated as volume ratios (v/v).
Unless otherwise stated, the animal models used were as follows:
c57BL/6J mice: purchased from beijing vernalia laboratory animal technology limited;
HBV transgenic mice C57BL/6J-Tg (Alb 1 HBV) 44Bri/J: purchased from the department of laboratory animal science, university of Beijing, medical department. Mice with S/CoV > 10 were selected prior to the experiment;
AAV-HBV transgenic mice: AAV-HBV models were prepared according to literature methods (Dong Xiaoyan et al, chin J Biotech 2010, may 25;26 (5): 679-686). rAAV8-1.3HBV, D (ayw) virus (available from Beijing Acanthopanax and molecular medicine research Co., ltd., 1X 10) 12 viral genome (v.g.)/mL, lot 2016123011) was diluted to 5X 10 with sterile PBS 11 v.g./mL, 200. Mu.L of diluted rAAV8-1.3HBV per mouse (i.e.1X 10 per mouse) 10 v.g). On day 28 post virus injection, all mice were bled through the orbit (about 100 μl) for collection of serum for detection of HBsAg and HBV DNA;
BALB/c mice: 6-8 weeks old, purchased from Beijing Vietnam laboratory animal technologies Co., ltd;
human APOC3 transgenic mice: b6; CBA-Tg (APOC 3) 3707Bres/J, available from Jackson Lab;
preparation example 1B-2 preparation of conjugate molecule (conjugate molecule 1)
In this preparation, a compound of conjugate molecule 1 (hereinafter, also referred to as B-2 conjugate molecule) was synthesized according to the following method:
(1-1) Synthesis of conjugated terminal segment GAL-5 (terminal molecule of B-2 conjugated molecule)
(1-1 a) Synthesis of GAL-2
100.0g of GAL-1 (N-acetyl-D-galactosamine hydrochloride, CAS number 1772-03-8, available from Ningbo paraglider Biochemical Co., ltd., 463.8 mmol) was dissolved in 1000ml of anhydrous pyridine, 540ml of acetic anhydride (available from Enox Co., ltd., 5565.6 mmol) was added to the solution in an ice water bath, and the reaction was stirred at room temperature for 1.5 hours. The reaction solution was poured into 10L of ice water, suction filtration was performed under reduced pressure, after the filter cake was washed with 2L of ice water, acetonitrile/toluene mixed solvent (volume ratio acetonitrile: toluene=1:1) was added until complete dissolution, and the solvent was evaporated to dryness, to obtain a white solid product GAL-2.0 g.
(1-1 b) Synthesis of GAL-3
GAL-2 (35.1 g,90.0 mmol) obtained in the step (1-1 a) was dissolved in 213ml of anhydrous 1, 2-dichloroethane, and 24.0g of TMSOTF (CAS number: 27607-77-8, available from Michael company, 108.0 mmol) was added under an ice-water bath and nitrogen protection, and reacted overnight at room temperature.
400ml of methylene chloride was added to the reaction solution to dilute it, the mixture was filtered through celite, then 1L of saturated aqueous sodium bicarbonate solution was added thereto and stirred uniformly, the organic phase was separated, the aqueous phase was extracted twice with dichloroethane, 300ml of each time, the organic phases were combined, washed with 300ml of saturated aqueous sodium bicarbonate solution and 300ml of saturated brine, the organic phase was separated, dried over anhydrous sodium sulfate, and the solvent was evaporated under reduced pressure to give GAL-3.9 g as a pale yellow viscous syrup-like product.
(1-1 c) Synthesis of GAL-4
GAL-3 (26.9 g,81.7 mmol) obtained in step (1-1 b) was dissolved in 136ml of anhydrous 1, 2-dichloroethane, and dried was addedMolecular sieve powder 30g, 9.0g of 5-hexen-1-ol (CAS number 821-41-0, available from Adamas-beta, 89.9 mmol) was added, stirred at room temperature for 30 minutes, 9.08g TMSOTF (40.9 mmol) was added under ice bath and nitrogen protection, and the reaction was stirred at room temperature overnight. Filtering to remove->Molecular sieve powder, adding 300ml of dichloroethane to the filtrate for dilution, filtering with diatomite, adding 500ml of saturated aqueous sodium bicarbonate solution for stirring and washing for 10 minutes, separating out an organic phase, extracting the aqueous phase once with 300ml of dichloroethane, combining the organic phases and washing with 300ml of saturated aqueous sodium bicarbonate solution and 300ml of saturated saline respectively, separating out the organic phase, drying with anhydrous sodium sulfate, evaporating the solvent under reduced pressure to obtain a yellow syrup-like product GAL-4.3 g, and directly carrying out the next oxidation reaction without purification.
(1-1 d) Synthesis of GAL-5
GAL-4 (14.9 g,34.7 mmol) obtained as described in step (1-1 c) was dissolved in a mixed solvent of 77ml of methylene chloride and 77ml of acetonitrile, 103ml of deionized water and 29.7g of sodium periodate (CAS No. 7790-28-5, available from Aba Ding Gongsi, 138.8 mmol) were added, respectively, and stirred in an ice-water bath for 10 minutes, and ruthenium trichloride (CAS No. 14898-67-0, available from Anagli, 238mg,1.145 mmol) was added to react overnight at room temperature. The reaction mixture was diluted with 300ml of water and stirred, saturated sodium bicarbonate was added to adjust the pH to about 7.5, the organic phase was separated and discarded, the aqueous phase was extracted three times with 200ml portions of dichloromethane and the organic phase was discarded. The aqueous phase was adjusted to pH 3 with citric acid solids, extracted three times with 200ml portions of methylene chloride, the organic phases combined, dried over anhydrous sodium sulfate and the solvent evaporated under reduced pressure to give GAL-5.5 g as a white foamy solid product. 1 H NMR(400MHz,DMSO)δ12.01(br,1H),7.83(d,J=9.2Hz,1H),5.21(d,J=3.2Hz,1H),4.96(dd,J=11.2,3.2Hz,1H),4.49(d,J=8.4Hz,1H),4.07–3.95(m,3H),3.92–3.85(m,1H),3.74–3.67(m,1H),3.48–3.39(m,1H),2.20(t,J=6.8Hz,2H),2.11(s,3H),2.00(s,3H),1.90(s,3H),1.77(s,3H),1.55–1.45(m,4H).
(1-2) Synthesis of A-1:
DMTrCl (4, 4' -dimethoxytrityl chloride, 38.12g,112.5 mm)ol) was dissolved in 450ml of anhydrous pyridine, calcium DL-glycerate hydrate (12.88 g,45.0 mmol) was added, the reaction was carried out at 45℃for 22h, the reaction solution was filtered, the cake was rinsed with 200ml of DCM, the filtrate was concentrated to dryness under reduced pressure, the residue was redissolved with 500ml of dichloromethane, 0.5M triethylamine phosphate (pH=7-8) was washed 2 times, 200ml each time, the aqueous phase was extracted 2 times with dichloromethane, 200ml each time, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure to solvent, 200-300 mesh normal phase silica gel column purified, elution was carried out with petroleum ether: ethyl acetate: dichloromethane: methanol=1:1:1:0.35-1:1:1:1:0.55 gradient, the product eluent was collected, the solvent was evaporated under reduced pressure, 500ml of dichloromethane was redissolved, 1 time was washed with 200ml of 0.5M triethylamine phosphate, the aqueous phase was extracted 2 times with dichloromethane, 200ml each time, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, the solvent was filtered, and vacuum oil was pumped to dryness overnight to obtain a white solid product A-20.7 g. 1 H NMR(400MHz,DMSO-d6)δ7.46(ddd,J=6.5,2.3,1.1Hz,1H),7.40–7.28(m,7H),6.89–6.81(m,4H),4.84(d,J=5.0Hz,1H),4.36–4.24(m,1H),4.29(s,6H),3.92(dd,J=12.4,7.0Hz,1H),3.67(dd,J=12.3,7.0Hz,1H),2.52(q,J=6.3Hz,6H),1.03(t,J=6.3Hz,9H).MS m/z:C24H23O6,[M-H]-, theory: 407.15, found: 406.92.
(1-3) Synthesis of A-2:
a-1 (5.100 g,10 mmol), benzotriazol-1-yl-oxy-tripyrrolidinylphosphine hexafluorophosphate (PyBOP, 10.410g,20 mmol), 1-hydroxybenzotriazole (HOBt, 2.700g,20 mmol), diisopropylethylamine (DIEA, 6.460g,50 mmol) obtained in step (1-2) were dissolved in 50ml of dichloromethane, and reacted at room temperature for 30 minutes with stirring, and the above reaction solution was poured into 50ml of dichloromethane solution in which triethylenetetramine (TETA, 11.700g,80 mmol) was dissolved and reacted at 25℃with stirring for 21 hours. Washing with 100ml saturated saline 1 times, extracting the aqueous phase with 100ml dichloromethane 2 times, mixing the organic phases, drying over anhydrous sodium sulfate, filtering, evaporating the solvent under reduced pressure, purifying with normal phase silica gel column (dichloromethane: methanol: ammonia=100:40:10-100):40:14 eluting the product), collecting the product eluent, and evaporating the solvent under reduced pressure to obtain 4.181g of the product. 1 H NMR(400MHz,DMSO-d 6 )δ7.97(t,J=4.4Hz,1H),7.57–7.49(m,4H),7.51–7.41(m,2H),7.35–7.24(m,3H),6.81–6.73(m,4H),5.86(d,J=7.0Hz,1H),5.08–4.98(m,2H),4.13(dt,J=6.9,6.3Hz,1H),3.92(dd,J=11.3,6.3Hz,1H),3.81(s,5H),3.67(dd,J=11.4,6.3Hz,1H),3.39(dtd,J=15.1,5.4,4.4Hz,1H),3.00(dtd,J=15.1,5.4,4.2Hz,1H),2.88–2.73(m,2H),2.72–2.57(m,3H),2.58–2.32(m,5H),1.33(p,J=4.2Hz,2H).MS m/z:C30H41N4O5,[M+H]++, theory: 537.68, found: 537.53.
the following B-2 conjugated molecules were synthesized by the following process route, using GAL-5 compounds obtained according to the above-described method:
(1-4) Synthesis of GAL-C6-1
GAL-5 (4.5 g,10.0 mmol), tert-butyl 6-aminocaproate hydrochloride (2.2 g,12.0 mmol), O-benzotriazol-tetramethylurea hexafluorophosphate (5.7 g,15.0 mmol) and diisopropylethylamine (3.9 g,30.0 mmol) were added to 40ml of N, N-dimethylformamide, and the reaction was stirred at room temperature for 4 hours. To the reaction solution, 100ml of a saturated aqueous sodium hydrogencarbonate solution was slowly added, extraction was performed 3 times with 100ml of ethyl acetate, the organic phases were combined, washed once with 100ml of a saturated brine, the organic phase was separated, dried over anhydrous sodium sulfate, the solvent was distilled off under reduced pressure and dried by pumping with an oil pump to obtain 10.5g of crude oil which was directly subjected to the next reaction.
(1-5) Synthesis of GAL-C6-2
The crude GAL-C6-1 (10.5 g,10 mmol) obtained in step (1-4) was dissolved in 60ml formic acid, and the reaction was stirred at room temperature for 16 hours. The reaction solution is dried by spin, and the target product is collected and concentrated by column chromatography (normal phase silica gel of 200-300 meshes, dichloromethane: methanol=100:18-100:20 gradient elution), thus obtaining 5.2g of target product. 1H NMR (400 MHz, DMSO-d 6) delta 7.87 (s, 0H), 7.46 (s, 0H), 6.05-5.94 (m, 0H), 5.17 (t, J=7.0 Hz, 0H), 4.54 (dd, J=12.4, 6.9Hz, 0H), 4.33 (q, J=7.0 Hz, 0H), 3.88 (t, J=7.0 Hz, 0H), 3.75-3.58 (m, 0H), 3.38 (td, J=12.3, 2.1Hz, 0H), 3.17-3.07 (m, 0H), 2.57-2.46 (m, 0H), 2.50-2.35 (m, 0H), 2.28 (ddd, J=12.3, 4.5,2.1Hz, 2.1H), 2.27-2.08 (m, 0H), 2.09-1.90 (m, 1.90), 1.1.1 Hz, 0H), 3.17-3.07 (m, 0H), 2.57-2.46 (m, 0H), 2.50-2.35 (m, 0H), 2.28 (m, 3.9-1.5 Hz, 1.1H): C25H39N2O12, [ M-H ] -, theory: 559.25, found: 559.32.
(1-6) Synthesis of B-1:
GAL-C6-2 (2.018 g,3.6 mmol), 3-diethoxyphosphoryl-1, 2, 3-benzozol 4 (3H) -one (DEPBT, 1.496g,5.0 mmol) and diisopropylethylamine (DIEA, 1.292g,10.0 mmol) obtained in step (1-5) were dissolved in 10ml of dichloromethane, and reacted at room temperature with stirring for 5 minutes, and then A-2 (0.537 g,1.0 mmol) was added thereto with stirring at 25℃for 24 hours. The reaction solution was washed 1 time with 20ml of saturated sodium bicarbonate, the aqueous phase was extracted 3 times with 20ml of methylene chloride, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the solvent was evaporated under reduced pressure, and the solvent was purified by column chromatography on normal phase silica gel (1% triethylamine to neutralize the acidity of the silica gel, petroleum ether: ethyl acetate: methylene chloride: methanol=1:1:0.2-0.25 eluting the product), and the solvent was evaporated under reduced pressure to give 1.79g of pure product. 1 H NMR(400MHz,DMSO-d 6 )δ8.01–7.87(m,2H),7.75(td,J=4.4,0.9Hz,1H),7.51–7.42(m,1H),7.35–7.25(m,1H),7.17–7.09(m,1H),6.81–6.73(m,1H),5.26(ddd,J=6.4,5.2,1.0Hz,1H),5.09(ddd,J=6.2,5.1,1.0Hz,1H),4.46(dd,J=6.3,0.9Hz,1H),4.34(ddd,J=12.1,2.3,1.0Hz,1H),4.14–4.03(m,2H),3.96(dddd,J=6.2,4.0,2.5,1.3Hz,1H),3.95–3.76(m,1H),3.81(s,2H),3.80–3.71(m,2H),3.72–3.57(m,1H),3.61–3.46(m,1H),3.47(dd,J=5.9,1.6Hz,1H),3.35(dt,J=11.8,6.0Hz,1H),3.11(dtd,J=14.3,6.2,4.4Hz,0H),2.99(tdd,J=6.2,4.3,1.6Hz,1H),2.87(dtd,J=14.2,6.2,4.3Hz,1H),2.39(dt,J=14.0,6.9Hz,1H),2.31–2.08(m,2H),2.09(dd,J=6.9,1.6Hz,1H),2.08–1.93(m,6H),1.97–1.88(m,3H),1.83–1.78(m,3H),1.73–1.08(m,10H).MS m/z:C105H155N10O38,[M+H]++, theory: 2165.43, found: 2165.62.
(1-7) Synthesis of B-2:
b-1 (2.727 g,1.26mmol, obtained by combining two batches) obtained according to the method described in steps (1-6), succinic anhydride (0.378 g,3.78 mmol) and 4-dimethylaminopyridine (DMAP, 0.4632 g,3.78 mmol)) Dissolved in 13ml of dichloromethane, DIEA (0.814 g,6.30 mmol) was added and the reaction stirred at 25℃for 24h.5ml of 0.5M triethylamine phosphate are used for washing the reaction solution, the aqueous phase is extracted with dichloromethane for 3 times, 5ml of each time, and the organic phases are combined and evaporated to dryness under reduced pressure to obtain a crude product. 60g of 200-300 mesh normal phase silica gel is used for column purification, 1% triethylamine is used for neutralizing the acidity of the silica gel, the column is balanced by methylene dichloride, the methylene dichloride containing 1 permillage triethylamine is eluted by methanol=100:18-20, and 2.719g of pure product is obtained by evaporating the solvent under reduced pressure. 1 H NMR(400MHz,DMSO-d 6 )δ8.01–7.87(m,2H),7.75(td,J=4.4,0.9Hz,1H),7.51–7.42(m,1H),7.34–7.25(m,1H),7.17–7.09(m,1H),6.81–6.73(m,1H),5.81(t,J=5.8Hz,1H),5.26(ddd,J=6.4,5.1,0.9Hz,1H),5.09(ddd,J=5.5,5.0,1.0Hz,1H),4.46(dd,J=6.3,0.9Hz,1H),4.34(ddd,J=12.2,2.3,1.0Hz,1H),4.14–4.04(m,2H),4.08–3.81(m,2H),3.85–3.71(m,5H),3.69–3.54(m,1H),3.59–3.46(m,1H),3.47(dd,J=5.9,1.6Hz,1H),3.35(dt,J=11.8,6.0Hz,0H),3.11(dtd,J=14.3,6.2,4.4Hz,1H),2.99(tdd,J=6.2,4.3,1.6Hz,1H),2.93–2.72(m,1H),2.71–2.47(m,3H),2.46–2.33(m,1H),2.31–2.05(m,3H),2.08–1.99(m,6H),2.03–1.88(m,4H),1.83–1.78(m,3H),1.71–1.56(m,1H),1.60–1.41(m,5H),1.46–1.38(m,1H),1.43–1.35(m,1H),1.40–1.32(m,1H),1.32(ddt,J=6.9,2.0,1.0Hz,1H),1.32–1.08(m,1H),1.03(t,J=7.2Hz,3H).MS m/z:C109H159N10O41,[M+H]++, theory: 2265.50, found: 2265.38. the structure of the resulting B-2 conjugated molecule is shown in formula (601).
Preparation of conjugate molecule 2D-6 (conjugate molecule 2)
In this preparation, the compound of conjugate molecule 2 (hereinafter, also referred to as D-6 conjugate molecule) was synthesized according to the following method:
(2-1) Synthesis of D-5:
GAL-5 (1.611 g,3.6 mmol), 3-diethoxyphosphoryl-1, 2, 3-benzole 4 (3H) -one (DEPBT, 1.496g,5.0 mmol) and diisopropylethylamine (DIEA, 1.292g,10.0 mmol) were dissolved in 10ml of dichloromethane, and reacted at room temperature with stirring for 5 minutes, followed by addition of A-2 (0.537 g,1.0 mmol) and stirred at 25℃for 24 hours. The reaction solution was washed 1 time with 20ml of saturated sodium bicarbonate, the aqueous phase was extracted 3 times with 20ml of dichloromethane each time, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the solvent was evaporated under reduced pressure, and the solvent was purified by column chromatography on normal phase silica gel (1% triethylamine neutralized silica gel acidity, petroleum ether: ethyl acetate: dichloromethane: methanol=1:1:1:0.2-1:1:1:0.25 eluted product), and the solvent was evaporated under reduced pressure to give 1.35g of pure product. 1 H NMR(400MHz,DMSO-d 6 )δ8.01–7.87(m,2H),7.51–7.41(m,1H),7.35–7.25(m,1H),7.17–7.09(m,1H),6.81–6.73(m,1H),5.86(d,J=7.0Hz,0H),5.26(ddd,J=6.4,5.2,1.0Hz,1H),5.09(ddd,J=5.5,5.0,1.0Hz,1H),4.74(dt,J=7.0,6.3Hz,0H),4.46(dd,J=6.3,0.9Hz,1H),4.34(ddd,J=12.2,2.3,1.0Hz,1H),4.14–4.03(m,2H),4.00–3.90(m,1H),3.94–3.76(m,1H),3.81(s,2H),3.80–3.71(m,2H),3.72–3.57(m,1H),3.61–3.46(m,1H),3.47(dd,J=5.9,1.6Hz,1H),3.35(dt,J=11.8,6.0Hz,0H),2.39(dt,J=14.0,6.9Hz,0H),2.31–2.09(m,1H),2.06–1.95(m,6H),1.99–1.90(m,3H),1.83–1.78(m,3H),1.68–1.22(m,4H).MS m/z:C87H122N7O35,[M+H]++, theory: 1825.95, found: 1825.77.
(2-2) Synthesis of D-6:
the resulting D-5 (2.299 g,1.26mmol, obtained by combining multiple batches) and succinic anhydride (0.378 g,3.78 mmol) obtained according to the method described in step (2-1) and 4-dimethylaminopyridine (DMAP, 0.463 g,3.78 mmol) were dissolved in 13ml dichloromethane and DIEA (0.814 g,6.30 mmol) was added thereto and reacted under stirring at 25℃for 24h. The reaction mixture was washed with 5ml of 0.5M triethylamine phosphate, the aqueous phase was extracted 3 times with 5ml of dichloromethane each time, and the organic phases were combined and evaporated to dryness under reduced pressure to give the crude product. Column purification Using 60g of 200-300 mesh normal phase silica gel column and neutralization with 1% triethylamineSilica gel is acidic, the column is equilibrated with dichloromethane, the product is eluted with dichloromethane containing 1% triethylamine in methanol=100:18-100:20, and the solvent is evaporated under reduced pressure to give pure product 1.836g. 1 H NMR(400MHz,DMSO-d 6 )δ8.01–7.87(m,2H),7.50–7.41(m,1H),7.35–7.25(m,1H),7.17–7.09(m,1H),6.81–6.73(m,1H),5.81(t,J=5.8Hz,0H),5.26(ddd,J=6.5,5.2,1.0Hz,1H),5.09(ddd,J=5.5,5.0,1.0Hz,1H),4.46(dd,J=6.3,0.9Hz,1H),4.34(ddd,J=12.2,2.3,1.0Hz,1H),4.14–4.07(m,1H),4.08(dq,J=2.4,1.7,1.2Hz,1H),4.08–3.94(m,1H),3.99–3.82(m,1H),3.86–3.71(m,5H),3.69–3.54(m,1H),3.59–3.46(m,1H),3.47(dd,J=5.9,1.6Hz,1H),3.35(dt,J=11.8,6.0Hz,1H),2.78(dt,J=14.6,8.5Hz,0H),2.71–2.47(m,3H),2.46–2.33(m,1H),2.31–2.09(m,1H),2.06–1.96(m,6H),2.00–1.90(m,3H),1.83–1.78(m,3H),1.68–1.22(m,4H),1.03(t,J=7.2Hz,3H).MS m/z:C91H126N7O38,[M+H]++, theory: 1926.02, found: 1926.15. the structure of the resulting D-6 conjugate molecule is shown in formula (602).
Preparation example 3 preparation of C-2 conjugate molecule (conjugate molecule 3)
In this preparation, a compound of conjugate molecule 3 (hereinafter, also referred to as a C-2 conjugate molecule) was synthesized according to the following method:
(3-1) Synthesis of GAL-C2-1
GAL-5 (4.5 g,10.0 mmol), glycine tert-butyl ester hydrochloride (2.0 g,12.0 mmol), O-benzotriazol-tetramethylurea hexafluorophosphate (5.7 g,15.0 mmol) and diisopropylethylamine (3.9 g,30.0 mmol) were added to 40ml of N, N-dimethylformamide, and the reaction was stirred at room temperature for 4 hours. To the reaction solution, 100ml of a saturated aqueous sodium hydrogencarbonate solution was added, extraction was performed 3 times with 100ml of ethyl acetate, the organic phases were combined, washed once with 100ml of a saturated brine, the organic phase was separated, dried over anhydrous sodium sulfate, the solvent was distilled off under reduced pressure and extracted with an oil pump to obtain 10.1g of crude oil which was directly subjected to the next reaction.
(3-2) Synthesis of GAL-C2-2
The crude GAL-C2-1 (10.1 g,10 mmol) was dissolved in 60ml formic acid and the reaction was stirred at room temperature for 16 hours. Spin-drying the reaction solution, purifying and collecting a target product by column chromatography (200-300 meshes of normal-phase silica gel and dichloromethane/methanol gradient elution), and concentrating to obtain 5.0g of the target product. 1H NMR (400 MHz, DMSO-d 6) delta 8.49 (s, 1H), 7.46 (s, 1H), 6.05-5.94 (m, 2H), 5.01-4.90 (m, 1H), 4.52-4.33 (m, 3H), 3.79-3.61 (m, 3H), 3.14 (td, J=12.2, 3.2Hz, 1H), 2.68 (td, J=12.2, 3.3Hz, 1H), 2.27 (td, J=12.6, 2.8Hz, 1H), 2.15 (td, J=12.6, 2.9Hz, 1H), 2.07-1.95 (m, 12H), 1.82 (qt, J=12.9, 2.8Hz, 1H), 1.54 (qt, J=12.6, 2.6Hz, 1H), 1.15-0.99 (m, 1.97/0.81). C21h31n2o12, [ m—h ] -, theory: 503.19, found: 503.26.
(3-3) Synthesis of C-1:
GAL-C2-2 (1.356 g,3.6 mmol), 3-diethoxyphosphoryl-1, 2, 3-benzozol 4 (3H) -one (DEPBT, 1.496g,5.0 mmol) and diisopropylethylamine (DIEA, 1.292g,10.0 mmol) were dissolved in 10ml of dichloromethane and reacted at room temperature with stirring for 5 minutes, and then A-2 (0.537 g,1.0 mmol) was added thereto and reacted at 25℃with stirring for 24 hours. The reaction solution was washed 1 time with 20ml of saturated sodium bicarbonate, the aqueous phase was extracted 3 times with 20ml of dichloromethane each time, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the solvent was evaporated under reduced pressure, and the solvent was purified by column chromatography on normal phase silica gel (1% triethylamine neutralized silica gel acidity, petroleum ether: ethyl acetate: dichloromethane: methanol=1:1:0.2-0.25 eluted product), and the solvent was evaporated under reduced pressure to give 1.74g of pure product. 1 H NMR(400MHz,DMSO-d6)δ8.14(td,J=5.8,1.0Hz,1H),8.01–7.87(m,1H),7.87–7.79(m,0H),7.51–7.42(m,1H),7.35–7.25(m,1H),7.17–7.09(m,1H),6.81–6.73(m,1H),5.86(d,J=7.0Hz,0H),5.26(ddd,J=6.4,5.2,1.0Hz,1H),5.09(ddd,J=5.6,5.0,1.0Hz,1H),4.74(dt,J=7.0,6.3Hz,0H),4.46(dd,J=6.3,0.9Hz,1H),4.34(ddd,J=12.2,2.3,1.0Hz,1H),4.14–4.03(m,2H),4.02–3.56(m,9H),3.60–3.46(m,1H),3.47(dd,J=5.9,1.6Hz,1H),3.35(dt,J=11.8,6.0Hz,1H),2.21(dt,J=15.1,6.9Hz,0H),2.14–2.05(m,1H),2.10–1.98(m,6H),2.01–1.90(m,3H),1.83–1.78(m,3H),1.68–1.22(m,4H).MS m/z:C93H131N10O38,[M+H]++, theory: 1997.10, found: 1997.32.
(3-4) Synthesis of C-2:
c-1 (2.515 g,1.26mmol, obtained by combining two batches) obtained according to the method described in step (3-3), succinic anhydride (0.378 g,3.78 mmol) and 4-dimethylaminopyridine (DMAP, 0.460 g,3.78 mmol) were dissolved in 13ml dichloromethane and DIEA (0.814 g,6.30 mmol) was added thereto and the reaction was stirred at 25℃for 24h.5ml of 0.5M triethylamine phosphate are used for washing the reaction solution, the aqueous phase is extracted with dichloromethane for 3 times, 5ml of each time, and the organic phases are combined and evaporated to dryness under reduced pressure to obtain a crude product. 60g of 200-300 mesh normal phase silica gel is used for column purification, 1% triethylamine is used for neutralizing the acidity of the silica gel, the column is balanced by methylene dichloride, the methylene dichloride containing 1 permillage triethylamine is eluted by methanol=100:18-20, and the solvent is evaporated under reduced pressure to obtain 2.469g of pure product. 1 H NMR(400MHz,DMSO-d 6 )δ8.14(td,J=5.8,1.0Hz,1H),8.01–7.87(m,1H),7.87–7.79(m,0H),7.51–7.41(m,1H),7.35–7.25(m,1H),7.17–7.09(m,1H),6.81–6.73(m,1H),5.81(t,J=5.8Hz,0H),5.26(ddd,J=6.5,5.2,1.0Hz,1H),5.09(ddd,J=5.5,5.1,1.0Hz,1H),4.46(dd,J=6.3,1.0Hz,1H),4.34(ddd,J=12.2,2.3,1.0Hz,1H),4.10(ddd,J=6.9,2.5,0.9Hz,1H),4.10–3.80(m,4H),3.84–3.68(m,5H),3.69–3.43(m,3H),3.35(dt,J=11.8,6.0Hz,0H),2.78(dt,J=14.6,8.5Hz,0H),2.71–2.47(m,3H),2.40(dt,J=14.9,8.6Hz,0H),2.21(dt,J=15.1,6.9Hz,1H),2.14–2.05(m,1H),2.10–1.99(m,6H),2.03–1.90(m,3H),1.83–1.78(m,3H),1.68–1.22(m,4H),1.03(t,J=7.2Hz,3H).MS m/z:C97H135N10O41,[M+H]++, theory: 2097.18, found: 2097.25. the structure of the resulting C-2 conjugate molecule is shown in formula (603).
Preparation example 4 preparation of P-2 conjugate molecule (conjugate molecule 4)
In this preparation, a compound of conjugate molecule 4 (hereinafter, also referred to as P-2 conjugate molecule) was synthesized according to the following method:
(4-1) Synthesis of GAL5-C4-1
GAL-5 (4.5 g,10.0 mmol), tert-butyl 4-amino acid hydrochloride (1.9 g,12.0 mmol), O-benzotriazol-tetramethylurea hexafluorophosphate (5.7 g,15.0 mmol), diisopropylethylamine (3.9 g,30.0 mmol) were added to 40ml of N, N-dimethylformamide, and the reaction was stirred at room temperature for 4 hours. To the reaction solution, 100ml of saturated aqueous sodium hydrogencarbonate solution was slowly added, extraction was performed 3 times with 100ml of ethyl acetate, the organic phases were combined, washed once with 100ml of saturated brine, the organic phase was separated, dried over anhydrous sodium sulfate, the solvent was distilled off under reduced pressure and extracted with an oil pump to obtain 10.3g of crude oil which was directly subjected to the next reaction.
(4-2) Synthesis of GAL5-C4-2
The crude GAL5-C4-1 (10.3 g,10 mmol) was dissolved in 60ml formic acid and the reaction was stirred at room temperature for 16 hours. The reaction solution is dried by spin, and the target product is collected and concentrated by column chromatography (normal phase silica gel of 200-300 meshes, dichloromethane: methanol=100:18-100:20 gradient elution), thus obtaining 5.1g of target product. 1 H NMR(400MHz,DMSO-d6)δ7.89(s,1H),7.46(s,1H),6.05–5.94(m,2H),5.25(t,J=7.0Hz,1H),4.53–4.35(m,2H),4.14(t,J=7.0Hz,1H),3.81(dd,J=12.1,6.8Hz,1H),3.46(td,J=12.1,3.3Hz,1H),3.30(td,J=12.4,3.0Hz,1H),3.01(td,J=12.1,2.8Hz,1H),2.75(td,J=12.4,3.0Hz,1H),2.45–2.08(m,6H),2.07–1.95(m,12H),1.81–1.49(m,3H),1.35–1.20(m,1H).MS m/z:C23H35N2O12,[M-H]-, theory: 531.22, found: 531.15.
(4-3) Synthesis of P-1:
GAL5-C4-2 (1.917 g,3.6 mmol), 3-diethoxyphosphoryl-1, 2, 3-benzozol 4 (3H) -one (DEPBT, 1.496g,5.0 mmol) and diisopropylethylamine (DIEA, 1.292g,10.0 mmol) were dissolved in 10ml of dichloromethane and reacted at room temperature with stirring for 5 minutes, and A-2 (0.537 g,1.0 mmol) was added thereto and reacted at 25℃with stirring for 24 hours. The reaction solution was washed 1 time with 20ml of saturated sodium bicarbonate, the aqueous phase was extracted 3 times with 20ml of methylene chloride each time, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was evaporated under reduced pressure, and the normal phase silica gel was purified (1% triethylamine neutralized silica gel acidity, petroleum ether: ethyl acetate: methylene chloride: methanol=1:1:1:0.2-0.25 eluted product) and the solvent was evaporated under reduced pressure to give 1.74g of pure product. 1 H NMR(400MHz,DMSO-d 6 )δ8.01–7.87(m,2H),7.75(td,J=4.4,0.9Hz,1H),7.51–7.41(m,1H),7.35–7.25(m,1H),7.17–7.09(m,1H),6.81–6.73(m,1H),5.86(d,J=7.0Hz,0H),5.26(ddd,J=6.4,5.2,1.0Hz,1H),5.09(ddd,J=5.6,5.0,1.0Hz,1H),4.74(dt,J=7.0,6.3Hz,0H),4.46(dd,J=6.3,0.9Hz,1H),4.34(ddd,J=12.2,2.3,1.0Hz,1H),4.14–4.03(m,2H),4.00–3.71(m,6H),3.72–3.57(m,1H),3.61–3.46(m,1H),3.47(dd,J=5.9,1.6Hz,1H),3.35(dt,J=11.8,6.0Hz,0H),3.14(dtd,J=14.2,6.3,4.4Hz,0H),3.02(tdd,J=6.2,4.4,1.6Hz,1H),2.90(dtd,J=14.3,6.3,4.4Hz,0H),2.50(dt,J=14.2,7.0Hz,1H),2.43–2.25(m,1H),2.28–2.05(m,2H),2.10–1.99(m,5H),2.01(s,1H),2.01–1.90(m,3H),1.83–1.78(m,3H),1.77–1.22(m,6H).MS m/z:C99H143N10O38,[M+H]++, theory: 2081.27, found: 2081.09.
(4-4) Synthesis of P-2:
p-1 (2.6271 g,1.26mmol, obtained by combining two batches) obtained according to the method described in step (4-3), succinic anhydride (0.378 g,3.78 mmol) and 4-dimethylaminopyridine (DMAP, 0.460 g,3.78 mmol) were dissolved in 13ml dichloromethane, DIEA (0.814 g,6.30 mmol) was added thereto, and the reaction was stirred at 25℃for 24 hours. The reaction mixture was washed with 5ml of 0.5M triethylamine phosphate, the aqueous phase was extracted 3 times with 5ml of dichloromethane each time, and the organic phases were combined and evaporated to dryness under reduced pressure to give the crude product. 60g of 200-300 mesh normal phase silica gel is used for column purification, 1% triethylamine is used for neutralizing the acidity of the silica gel, the column is balanced by methylene dichloride, the methylene dichloride containing 1%o triethylamine is eluted by methanol=100:18-100:20, and 2.654g of pure product is obtained by decompressing and evaporating the solvent. 1 H NMR(400MHz,DMSO-d 6 )δ8.01–7.87(m,2H),7.75(td,J=4.4,0.9Hz,1H),7.51–7.41(m,1H),7.34–7.25(m,1H),7.17–7.09(m,1H),6.81–6.73(m,1H),5.81(t,J=5.8Hz,0H),5.26(ddd,J=6.5,5.2,1.0Hz,1H),5.09(ddd,J=5.5,5.0,1.0Hz,1H),4.46(dd,J=6.3,1.0Hz,1H),4.34(ddd,J=12.2,2.3,1.0Hz,1H),4.14–4.04(m,2H),4.08–3.81(m,2H),3.85–3.71(m,5H),3.69–3.54(m,1H),3.59–3.46(m,1H),3.47(dd,J=5.9,1.6Hz,1H),3.35(dt,J=11.8,6.0Hz,1H),3.14(dtd,J=14.2,6.3,4.4Hz,0H),3.02(tdd,J=6.2,4.4,1.6Hz,1H),2.90(dtd,J=14.3,6.3,4.4Hz,0H),2.78(dt,J=14.6,8.5Hz,0H),2.71–2.44(m,3H),2.46–2.35(m,1H),2.40–2.19(m,1H),2.22–2.04(m,2H),2.06–1.96(m,6H),2.00–1.85(m,3H),1.83–1.78(m,3H),1.77–1.22(m,7H),1.03(t,J=7.2Hz,3H).MS m/z:C103H147N10O41,[M+H]+,Theory of: 2181.34, found: 2181.48. the structure of the resulting P-2 conjugate molecule is shown in formula (604).
Preparation example 5X-3 preparation of conjugate molecule (conjugate molecule 5)
In this preparation, a compound of conjugate molecule 5 (hereinafter, also referred to as X-3 conjugate molecule) was synthesized according to the following method:
(5-1) Synthesis of X-1:
a-1 (5.100 g,10 mmol), benzotriazol-1-yl-oxy-tripyrrolidinylphosphine hexafluorophosphate (PyBOP, 10.410g,20 mmol), 1-hydroxybenzotriazole (HOBt, 2.700g,20 mmol) and diisopropylethylamine (DIEA, 6.460g,50 mmol) were dissolved in 50ml of methylene chloride, and reacted at room temperature with stirring for 30 minutes, and the above reaction solution was poured into a 50ml of methylene chloride solution in which tetraethylenepentamine (15.145 g,80 mmol) was dissolved, and reacted at 25℃with stirring for 21 hours. Washing with 100ml of saturated saline 1 time, extracting the aqueous phase with dichloromethane 2 times each with 100ml of water, combining the organic phases, drying over anhydrous sodium sulfate, filtering, evaporating the solvent under reduced pressure, purifying the normal phase silica gel column (dichloromethane: methanol: ammonia water=100:40:10-100:40:14 eluting the product), collecting the product eluent, evaporating the solvent under reduced pressure to obtain 3.761g of the product. 1 H NMR(400MHz,DMSO-d 6 )δ7.97(t,J=4.4Hz,1H),7.60–7.52(m,4H),7.51–7.41(m,2H),7.35–7.24(m,3H),6.81–6.73(m,4H),5.86(d,J=7.0Hz,1H),5.11–5.00(m,2H),4.13(dt,J=6.9,6.2Hz,1H),3.92(dd,J=11.3,6.3Hz,1H),3.81(s,5H),3.67(dd,J=11.4,6.3Hz,1H),3.37(dtd,J=15.2,5.4,4.4Hz,1H),3.04(dtd,J=15.1,5.4,4.4Hz,1H),2.99–2.77(m,2H),2.71–2.35(m,12H),1.33(m,3H).MS m/z:C32H45N5O5,[M+H]++, theory: 579.74, found: 579.59.
(5-2) Synthesis of X-2:
GAL-5 (2.238 g,5.0 mmol), 3-diethoxyphosphoryl-1, 2, 3-benzole 4 (3H) -one (DEPBT, 1.795g,6.0 mmol) and diisopropylethylamine (DIEA, 1.550g,12.0 mmol) were dissolved in 10ml of dichloromethane and reacted at room temperature with stirring for 5 minutes,x-1 (0.580 g,1.0 mmol) was added thereto, and the reaction was stirred at 25℃for 24 hours. The reaction solution was washed 1 time with 20ml of saturated sodium bicarbonate, the aqueous phase was extracted 2 times with 20ml of dichloromethane each time, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the solvent was evaporated under reduced pressure, and the normal phase silica gel was purified (1% triethylamine neutralized silica gel acidity, petroleum ether: ethyl acetate: dichloromethane: methanol=1:1:0.2-0.25 eluted product), and the solvent was removed by evaporation under reduced pressure to give 1.73g of pure product. 1 H NMR(400MHz,DMSO-d 6 )δ8.01–7.87(m,2H),7.50–7.42(m,1H),7.35–7.25(m,1H),7.17–7.09(m,1H),6.81–6.73(m,1H),5.31–5.22(m,1H),5.13–5.05(m,1H),4.50–4.43(m,1H),4.39–4.30(m,1H),4.14–4.03(m,2H),4.00–3.81(m,2H),3.85–3.71(m,5H),3.72–3.43(m,3H),2.32–2.08(m,2H),2.06–1.90(m,10H),1.81(dd,J=4.1,1.7Hz,3H),1.68–1.22(m,5H).MS m/z:C108H154N9O45,[M+H]++, theory: 2298.44, found: 2298.31.
(5-3) Synthesis of X-3:
x-2 (2.895 g,1.26mmol, obtained by combining two batches) obtained according to the method described in step (5-2), succinic anhydride (0.378 g,3.78 mmol) and 4-dimethylaminopyridine (DMAP, 0.460 g,3.78 mmol) were mixed and dissolved in 13ml dichloromethane, and DIEA (0.814 g,6.30 mmol) was added thereto and reacted under stirring at 25℃for 24 hours. The reaction mixture was washed with 5ml of 0.5M triethylamine phosphate, the aqueous phase was extracted 3 times with 5ml of dichloromethane each time, and the organic phases were combined and evaporated to dryness under reduced pressure to give the crude product. 60g of 200-300 mesh normal phase silica gel is used for column purification, 1% triethylamine is used for neutralizing the acidity of the silica gel, the column is balanced by methylene dichloride, the methylene dichloride containing 1%o triethylamine is eluted by methanol=100:18-100:20, and 2.826g of pure product is obtained by decompressing and evaporating the solvent. 1 H NMR(400MHz,DMSO-d 6 )δ8.01–7.87(m,2H),7.50–7.42(m,1H),7.35–7.25(m,1H),7.17–7.09(m,1H),6.81–6.73(m,1H),5.31–5.22(m,1H),5.13–5.05(m,1H),4.50–4.43(m,1H),4.39–4.30(m,1H),4.14–4.05(m,2H),4.09–3.82(m,2H),3.86–3.71(m,5H),3.69–3.52(m,1H),3.56–3.43(m,2H),2.71–2.47(m,2H),2.46–2.33(m,1H),2.32–2.06(m,2H),2.06–1.90(m,10H),1.81(dd,J=4.1,1.7Hz,3H),1.68–1.22(m,5H),1.03(t,J=7.2Hz,2H).MS m/z:C112H158N9O48,[M+H]++, theory: 2398.51, found:2398.66. the structure of the resulting X-3 conjugate molecule is shown in formula (605).
Preparation example 6K-3 preparation of conjugate molecule (comparative conjugate molecule 1)
In this preparation, a compound of comparative conjugate molecule 1 (hereinafter, also referred to as K-3 conjugate molecule) was synthesized according to the following method.
(6-1) Synthesis of K-1:
a-1 (3.0 g,6.0 mmol), pyBOP (6.2 g,12.0 mmol), HOBt (1.6 g,2.0 mmol) and diisopropylethylamine (DIPEA, 3.9g,30.0 mmol) obtained as described in step (1-2) were added to 60ml of methylene chloride, and the reaction was stirred at room temperature for 10 minutes, followed by adding the above solution to K-0 (5.6 g,30.0 mmol) and reacting at room temperature for 1 hour and 50 minutes. The reaction solution was poured into 30ml of saturated sodium bicarbonate solution, the aqueous phase was extracted 3 times with 30ml of dichloromethane each time, the organic phases were combined and washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, and concentrated by filtration. Purifying with 200-300 mesh normal phase silica gel column, gradient eluting with dichloromethane/methanol/ammonia water (25wt%) =10:2:0.1-4:4:1, collecting product eluate, concentrating to remove solvent, and vacuum oil pump foaming and drying to obtain white solid product K-12.2g. 1 H NMR(400MHz,DMSO-d6)δ8.02(s,1H),7.43(d,J=7.8Hz,2H),7.34–7.17(m,7H),6.87(d,J=8.6Hz,4H),4.05(d,J=5.2Hz,1H),3.74(s,6H),3.20–3.01(m,5H),2.60–2.38(m,12H),1.60–1.39(m,8H),1.24(s,1H).MS m/z:C33H47N4O5,[M+H]++, theory: 579.35, found: 579.26.
(6-2) Synthesis of K-2:
GAL-5 (4813 mg,1.08 mmol), 3-diethoxyphosphoryl-1, 2, 3-benzooxazol 4 (3H) -one (356 mg,1.2 mmol), diisopropylethylamine (DIPEA, 310mg,2.4 mmol) obtained as described in (1-1) was added to 3ml of methylene chloride, and stirred at room temperature for 30 minutes, followed by K-1 (174 mg,0.3 mmol) and reacted at room temperature for 16 hours. The reaction solution was poured into 10ml of saturated sodium hydrogencarbonate solution, the aqueous phase was extracted 3 times with 10ml of methylene chloride each time, the organic phases were combined and washed with 10ml of saturated sodium chloride solution, dried over anhydrous sodium sulfate, and concentrated by filtration. Purifying with 200-300 mesh normal phase silica gel column, eluting with dichloromethane: methanol=20:1, collecting the product eluent, concentrating to remove solvent, and vacuum oil pump drying and foaming to obtain yellow solid product K-2 205mg.
(6-3) Synthesis of K-3:
to 1.1ml of methylene chloride were added K-2 (205 mg,0.11 mmol), succinic anhydride (22 mg,0.22 mmol), 4-dimethylaminopyridine (DMAP, 27mg,0.22 mmol) and diisopropylethylamine (DIPEA, 71mg,0.55 mmol), and the reaction was stirred at room temperature overnight. The reaction solution was washed 3 times with 0.5M triethylamine phosphate solution, 0.5ml each time, and each time the aqueous phase was back-extracted with 0.5ml of methylene chloride, dried over anhydrous sodium sulfate, concentrated to remove the solvent, and dried and foamed by vacuum oil pump to give 218mg of K-3 conjugate molecule (comparative conjugate molecule 1) as a pale yellow solid product.
Preparation example 7B preparation of 3-SiHBa1 conjugate (conjugate 6)
In this preparation, starting from the B-2 conjugate molecule (conjugate molecule 1), a B3-siHBa1 conjugate (hereinafter, also referred to as conjugate 6) was prepared according to the following method
(7-1) Synthesis of B-3 Compound:
in this step, the B-3 compound is prepared by attaching the B-2 conjugate molecule to a solid support.
B-2 conjugated molecule (0.455 g,0.22 mmol), O-benzotriazol-tetramethyluronium hexafluorophosphate (HBTU, 0.125g,0.33 mmol) and diisopropylethylamine (DIEA, 0.057g,0.44 mmol) obtained in step (1-7) were mixed and dissolved in 18ml of acetonitrile, stirred at room temperature for 5 minutes to obtain a homogeneous solution, aminomethyl resin (1.76 g,100-200 mesh, amino load 400. Mu. Mol/g, purchased from Nanking and Chengzhi) was added to the reaction solution, shaking reaction was started at 25℃for 150 revolutions per minute, the reaction was carried out for 18 hours, and then filtration was carried out, and the cake was rinsed with DCM for 2 times, acetonitrile for 3 times, 50ml of diethyl ether for 1 time, and dried with a vacuum oil pump for 2 hours to obtain a D-6-linked solid carrier, followed by capping according to the charge ratio shown in Table 5.
Table 5 cap reaction batch ratios
Raw materials Weight of (E) Specification of specification Lot number Manufacturing factories
Cap1 40ml —— —— Homemade
Cap2 4.5ml —— —— Homemade
DMAP 0.022g Analytical grade I1422139 Aladdin
Acetonitrile 4.5ml Spectral purity O15161001 Starfish-shaped food
Cap1 and Cap2 are Cap reagent solutions, cap1 is a pyridine/acetonitrile mixed solution of 20 volume percent of N-methylimidazole, and the volume ratio of pyridine to acetonitrile is 3:5; cap2 is a 20% acetic anhydride by volume acetonitrile solution;
cap1, cap2, 4-Dimethylaminopyridine (DMAP) and acetonitrile were added to the solid phase carrier connected with D-6, shaking reaction was started at 25℃for 5h at 200 rpm, the reaction solution was filtered, the filter cake was rinsed with acetonitrile 3 times, 50ml each time, suction filtered to dryness and dried with a vacuum oil pump overnight to give 2.127g of B-3 compound (i.e., D-6 conjugated molecule connected with solid phase carrier) with a loading of 106.59. Mu. Mol/g. The structure of the B-3 compound is shown as a formula (701).
(7-2) Synthesis of the sense strand of the B3-siHBa1 conjugate
In this step, the siRNA of the siRNA conjugate is the sequence numbered sibba 1:
siHBa1
sense strand: 5'-CCUUGAGGCAUACUUCAAA-3' (SEQ ID NO: 1),
antisense strand: 5'-UUUGAAGUAUGCCUCAAGGUU-3' (SEQ ID NO: 2);
by the method of phosphoramidite nucleic acid solid phase synthesis, the B-3 compound prepared by the above steps is used to initiate a cycle, and nucleoside monomers are linked one by one in the 3'-5' direction according to the above sequence order. Each nucleoside monomer connected comprises four steps of deprotection, coupling, capping and oxidation. The synthesis conditions were given as follows: the nucleoside monomer was provided as a 0.1M acetonitrile solution, the deprotection conditions were the same for each step, i.e., the temperature was 25 ℃, the reaction time was 70 seconds, the deprotection reagent was a dichloromethane solution of dichloroacetic acid (3% v/v), and the molar ratio of dichloroacetic acid to 4,4' -dimethoxytrityl protecting group on the solid support was 5:1.
The coupling reaction conditions of each step are the same, the temperature is 25 ℃, the mole ratio of the nucleic acid sequence connected on the solid carrier to the nucleoside monomer is 1:10, the mole ratio of the nucleic acid sequence connected on the solid carrier to the coupling reagent is 1:65, the reaction time is 600 seconds, and the coupling reagent is a 0.5M acetonitrile solution of 5-ethylthio-1H-tetrazole.
The capping conditions were the same for each step, including a temperature of 25℃and a reaction time of 15 seconds. The Cap reagent solution is a mixed solution of Cap1 and Cap2 with a molar ratio of 1:1, and the molar ratio of the Cap reagent to the nucleic acid sequence connected on the solid phase carrier is acetic anhydride to N-methylimidazole to the nucleic acid sequence connected on the solid phase carrier=1:1:1.
The oxidation reaction conditions are the same in each step, the temperature is 25 ℃, the reaction time is 15 seconds, and the oxidizing agent is iodine water with the concentration of 0.05M. The molar ratio of iodine to nucleic acid sequence attached to the solid support in the coupling step was 30:1. The reaction was carried out in a mixed solvent of tetrahydrofuran, water, pyridine=3:1:1.
The cleavage and deprotection conditions were as follows: the synthesized nucleotide sequence with the carrier attached was added to ammonia water at a concentration of 25wt% at an amount of 0.5 ml/. Mu.mol, reacted at 55℃for 16 hours, the liquid was removed, and concentrated to dryness in vacuo. After the ammonia treatment, the product was dissolved with 0.4 ml/. Mu.mol of N-methylpyrrolidone, followed by the addition of 0.3 ml/. Mu.mol of triethylamine and 0.6 ml/. Mu.mol of triethylamine-tricofluoride, relative to the amount of single-stranded nucleic acid, and the 2' -TBDMS protection on ribose was removed. Purifying and desalting: purification of nucleic acids was accomplished by gradient elution with NaCl using a preparative ion chromatography purification column (Source 15Q). Specifically, the method comprises the following steps: eluent A:20mM sodium phosphate (pH 8.1), solvent water/acetonitrile=9:1 (volume ratio); eluent B:1.5M sodium chloride, 20mM sodium phosphate (pH 8.1), solvent water/acetonitrile=9:1 (volume ratio); elution gradient: eluent a, eluent b=100:0-50:50 gradient elution. Collecting and combining product eluents, desalting by using a reversed phase chromatographic purification column, wherein specific conditions comprise desalting by using a sephadex column, eluting with deionized water by using sephadex G25 as a filler.
And (3) detection: detection was performed using ion exchange chromatography (IEX-HPLC) with a purity of 92.4%; molecular weight was analyzed by liquid chromatography-mass spectrometry (LC-MS), theoretical 7253.96, measured 7253.12.
Thus, in this step, a B-2 conjugate molecule is attached to the 3 '-end of the resulting sense strand, resulting in an siRNA sense strand S with a B-3 conjugate molecule conjugated to the 3' -end of the siRNA.
(7-3) Synthesis of antisense strand
In this step, a universal solid support (UnyLinker) TM loaded HL Solid Supports, kinovate Life Sciences company), the antisense strand AS of the B3-sibba 1 conjugate was synthesized. Deprotection, coupling, capping and oxidation reaction conditions in the solid phase synthesis method, deprotection and cleavage, and separation conditions are the same AS those of the synthesized sense strand, thus obtaining the siRNA antisense strand AS.
And (3) detection: purity was measured by ion exchange chromatography (IEX-HPLC), and as a result, purity was 93.2%; molecular weight was analyzed by liquid chromatography-mass spectrometry (LC-MS). Theoretical 6675.04, found 6674.50.
(7-4) Synthesis of B3-siHBa1 conjugate
S chain and AS chain are mixed in equimolar ratio, dissolved in water for injection and heated to 95 deg.c, cooled at room temperature to form double chain structure via hydrogen bond.
After the synthesis was completed, the conjugate was diluted to a concentration of 0.2mg/mL with ultra pure water (self-made with Milli-Q ultra pure water meter, resistivity 18.2MΩ. Cm (25 ℃)). Molecular weight measurements were performed using a liquid chromatography-mass spectrometer (LC-MS, liquid Chromatography-Mass Spectrometry, available from Waters, model: LCT Premier). As a result, theoretical value S:7253.96, AS:6675.04, found S:7253.24, as:6674.61, the actual measurement corresponds to the theoretical value, thereby determining that the synthesized conjugate is a double-stranded nucleic acid sequence with the D-6 conjugated molecule designed in the target. The structure of the B3-siHBa1 conjugate (conjugate 6) is shown in formula (401).
Preparation example 8 preparation of siRNA conjugates of conjugates 7-27, 156-157, 32-95, 158-159, 100-129, 160-161, 134-151 and 162-163
The subject siRNA conjugates were prepared using the same procedure as in preparation 7, except that: 1) The conjugated siRNAs had the sequences as shown in tables 6A-6D corresponding to conjugates 7-27, 156-157, 32-95, 158-159, 100-129, 160-161, 134-151 and 162-163; 2) When phosphorothioate linkage is between two nucleotides in the target sequence, the following sulfidation step is used to replace the oxidation step in the linkage of the latter one of the two nucleotides; the conditions for each step of sulfiding reaction were the same, including a temperature of 25 ℃, a reaction time of 300 seconds, and the sulfiding reagent was hydrogenation Huang Yuansu. The molar ratio of sulfiding reagent to nucleic acid sequence attached to the solid support in the coupling step was 120:1. The reaction is carried out in a mixed solvent of acetonitrile and pyridine=1:1; and 3) when all the nucleotides in the target sequence are modified hydroxyl groups at the 2 '-position, the step of removing 2' -TBDMS protection on ribose is not included in the cleavage and deprotection conditions. Thus, siRNA conjugates of conjugates 7-27, 156-157, 32-95, 158-159, 100-129, 160-161, 134-151 and 162-163 of the present disclosure were prepared and numbered according to tables 6A-6D, respectively. The molecular weight is detected by a liquid chromatograph-mass spectrometer, the detected molecular weight value of the conjugate is consistent with the theoretical value, and the structures of the conjugate are shown in a formula (401).
TABLE 6A siRNA conjugates
/>
TABLE 6B
/>
/>
/>
/>
TABLE 6C
/>
/>
TABLE 6D
/>
* S: a sense strand; AS: antisense strand
Note that: capital C, G, U, A indicates the base composition of the nucleotide; the lower case letter m indicates that the adjacent nucleotide to the left of the letter m is a 2' -methoxy modified nucleotide; the lower case letter f indicates that the adjacent nucleotide to the left of the letter f is a 2' -fluoro modified nucleotide; the lower case letter s indicates that the linkage between two nucleotides adjacent to the letter s is a phosphorothioate linkage; VP represents that the nucleotide to the right of the letter VP is a vinyl phosphate modified nucleotide; p represents that the nucleotide to the right of the letter P is a phosphate modified nucleotide; ps means that the nucleotide to the right of the letter Ps is a phosphorothioate modified nucleotide.
Wherein, the vinyl phosphate modified 2' -methoxy modified uracil nucleoside monomer (VP-Um) is synthesized according to the following method:
(8-1) Synthesis of VP-U-2
VP-U-2 molecules were synthesized according to the following procedure:
2 '-methoxy-modified uracil nucleotide (2' -OMe-U,51.30g,91.6 mmol), tert-butyldiphenylchlorosilane (TBDPSCl, 50.35g,183.2 mmol), imidazole (12.47 g,183.2 mmol) were dissolved in 450ml N, N-Dimethylformamide (DMF) and stirred at room temperature for 20h. DMF was distilled off, dissolved in 600ml of dichloromethane, washed with 300ml of saturated sodium bicarbonate, the aqueous phase was extracted 3 times with Dichloromethane (DCM), 300ml each time, the organic phases were combined, washed with 5% oxalic acid to pH <5, and the solvent was evaporated to dryness to give crude VP-U-1 which was used directly in the subsequent synthesis of VP-U-2.
After the VP-U-1 crude product is dissolved by using 100ml of dichloromethane, the mixture is stirred for 10 minutes by adding ice bath, 450ml of 2% p-toluenesulfonic acid solution (the solvent is a mixed solvent of methanol and dichloromethane with the volume ratio of 3:7) which is refrigerated in a refrigerator at the temperature of 4 ℃ in advance is added, and the reaction is carried out for 10 minutes. The reaction was quenched by addition of 200ml of saturated sodium bicarbonate and the organic phase was washed with saturated aqueous sodium bicarbonate to ph=8. The aqueous phases were combined, extracted 2 times with 200ml of dichloromethane each time, the organic phases were combined, washed once with 200ml of saturated brine and the solvent evaporated to dryness. Purifying by 200-300 mesh normal phase silica gel column, loading petroleum ether into column, gradient eluting with petroleum ether, ethyl acetate, dichloromethane, methanol=1:1:0.05-1:1:1:0.25, collecting product eluent, evaporating solvent under reduced pressure, and foaming and drying by vacuum oil pump to obtain pure VP-U-2 40.00g.1H NMR (400 mhz, dmso-d 6) delta 7.96 (d, j=7.8 hz, 1H), 7.64 (dtd, j=5.1, 4.0,2.2hz, 4H), 7.41-7.30 (m, 6H), 6.79 (d, j=4.7 hz, 1H), 5.73 (d, j=7.6 hz, 1H), 4.94 (t, j=7.0 hz, 1H), 4.12 (td, j=4.6, 3.9hz, 1H), 4.05 (dd, j=4.8, 4.0hz, 1H), 3.96 (t, j=4.7 hz, 1H), 3.68 (ddd, j=11.8, 7.0,4.6hz, 1H), 3.57-3.46 (m, 1H), 3.39 (s, 3H), 1.05 (s, 8 z)/m.ms: c26H33N2O6Si, [ m+h ] +, theory: 497.21, found: 497.45.
(8-2) Synthesis of VP-U-4:
VP-U-2 (19.84 g,40.0 mmol), dicyclohexylcarbodiimide (DCC, 16.48g,80.0 mmol), pyridine (4.20 g,53.2 mmol), trifluoroacetic acid (6.61 g,53.2 mmol) were mixed and dissolved in 200ml dimethyl sulfoxide (DMSO), and the reaction was stirred at room temperature for 20h. In addition, tetraethyl methylenediphosphate (21.44 g,74.4 mmol) was dissolved in 120ml THF, cooled in an ice bath, t-BuOK (11.36 g,101.2 mmol) was added at ice bath temperature, reacted for 10min at ice bath temperature, then cooled to room temperature for 0.5h, then added to the above reaction solution, and reacted for 1h at ice bath temperature and then cooled to room temperature for 18h. The reaction was quenched with water and the aqueous phase was extracted 3 times with 200ml of dichloromethane. The organic phases were combined, washed once with 200ml of saturated brine and the solvent was evaporated to dryness. Purifying with 200-300 mesh normal phase silica gel column, loading petroleum ether into column, gradient eluting with petroleum ether:ethyl acetate=1:1-1:4, collecting product eluent, evaporating solvent under reduced pressure, and vacuum oil pump foaming and drying to obtain pure VP-U-4 14.00g.1H NMR (400 MHz, DMSO-d 6) delta 7.96 (d, J=7.8 Hz, 1H), 7.64 (dtd, J=5.1, 4.0,2.2Hz, 4H), 7.41-7.30 (m, 6H), 6.82-6.71 (m, 2H), 5.90 (ddd, J=25.9, 15.0,1.0Hz, 1H), 5.73 (d, J=7.6 Hz, 1H), 4.36-4.21 (m, 3H), 4.18 (t, J=4.9 Hz, 1H), 4.05 (ddq, J=9.7, 8.5,6.9Hz, 2H), 3.87 (t, J=4.8 Hz, 1H), 3.39 (s, 3H), 1.32 (td, J=6.9, 0.7Hz, 6H), 1.05 (s, 8 z)/m MS/z: c31h42N2O8PSi, [ m+h ] +, theory: 629.24, found: 629.51.
(8-3) Synthesis of VP-U-5:
VP-U-4 (14.00 g,22.29 mmol) was dissolved in 100ml tetrahydrofuran, and triethylamine trihydrofluoric acid (17.96 g,111.45 mmol) was added thereto, followed by stirring at room temperature for 20 hours to complete the reaction. The solvent was evaporated directly to dryness, dissolved with dichloromethane and evaporated to dryness 2 times, using 50ml of dichloromethane each time, to give the crude product. Purifying with 200-300 mesh normal phase silica gel column, loading petroleum ether into column, gradient eluting with petroleum ether, ethyl acetate, dichloromethane, methanol=1:1:1:0.05-1:1:1:0.25, collecting product eluate, evaporating solvent under reduced pressure, and foaming and drying with vacuum oil pump to obtain pure VP-U-5 with total weight of 6.70g.1H NMR (400 mhz, dmso-d 6) delta 7.96 (d, j=7.8 hz, 1H), 6.77 (dd, j=15.0, 6.2hz, 1H), 5.99-5.82 (m, 2H), 5.73 (d, j=7.6 hz, 1H), 5.27 (d, j=5.1 hz, 1H), 5.10 (dd, j=5.3, 4.7hz, 1H), 4.29 (ddq, j=9.8, 8.6,7.0hz, 2H), 4.17 (ddd, j=6.2, 5.2,1.0hz, 1H), 4.12-3.98 (m, 3H), 3.39 (s, 2H), 1.32 (td, j=6.9, 0.6hz, 6H). MS m/z: c15h24n2o8p, [ m+h ] +, theory: 391.13, found: 391.38.
(8-4) Synthesis of VP-U-6:
VP-U-5 (399mg, 1.0 mmol), pyridine trifluoroacetate (0.232 g,1.2 mmol), N-methylimidazole (0.099 g,1.2 mmol) and bis (diisopropylamino) (2-cyanoethoxy) phosphine (0.452 g,1.5 mmol) were added to 10ml of anhydrous dichloromethane under argon atmosphere, and the mixture was stirred at room temperature for 5 hours. Evaporating the solvent to dryness, purifying by column chromatography (200-300 mesh normal phase silica gel, dichloromethane: acetonitrile (containing 0.5wt% triethylamine) =3:1-1:3 gradient elution), collecting product eluent, concentrating to remove the solvent to obtain the target product VP-U-6 of 508mg.31P NMR (161 MHz, DMSO-d 6) delta 150.34,150.29,17.07,15.50.MS m/z: c24h41N4O9P2, [ m+h ] +, theory: 591.23, found: 591.55. VP-U-6 is shown to be the target product VP-Um, and is used as a nucleoside monomer to participate in RNA chain synthesis.
The 5 '-phosphate modification was attached to the 5' end of the antisense strand using the following method:
the raw material is a phosphorylated structural monomer with the following CPR-I structure, which is provided by Suzhou Ji Ma, and the product number Cat#13-2601-XX:
after all nucleoside monomers of the antisense strand are connected, CPR-I monomers are connected to the 5' -end of the antisense strand through four steps of deprotection, coupling, capping and oxidation according to a phosphoramidite nucleic acid solid phase synthesis method. Cleavage and deprotection were then performed under the following conditions to obtain the antisense strand:
the synthesized nucleotide sequence with the carrier attached was added to ammonia water at a concentration of 25wt% at an amount of 0.5 ml/. Mu.mol, reacted at 55℃for 16 hours, the liquid was removed, and concentrated to dryness in vacuo. After the ammonia treatment, the product was dissolved with 0.4 ml/. Mu.mol of N-methylpyrrolidone, followed by the addition of 0.3 ml/. Mu.mol of triethylamine and 0.6 ml/. Mu.mol of triethylamine-tricofluoride, relative to the amount of single-stranded nucleic acid, and the 2' -TBDMS protection on ribose was removed. Purifying and desalting: purification of nucleic acids was accomplished by gradient elution with NaCl using a preparative ion chromatography purification column (Source 15Q). Specifically, the method comprises the following steps: eluent A:20mM sodium phosphate (pH 8.1), solvent water/acetonitrile=9:1 (volume ratio); eluent B:1.5M sodium chloride, 20mM sodium phosphate (pH 8.1), solvent water/acetonitrile=9:1 (volume ratio); elution gradient: eluent a, eluent b=100:0-50:50 gradient elution. Collecting and combining product eluents, desalting by using a reversed phase chromatographic purification column, wherein specific conditions comprise desalting by using a sephadex column, eluting with deionized water by using sephadex G25 as a filler.
The same procedure as described above was used for the case where the target product had 5' -phosphorothioate modification, except that the vulcanization reaction was carried out in place of the above oxidation reaction conditions at the time of ligation.
For the sense strand and antisense strand synthesized as described above, purity was detected using ion exchange chromatography (IEX-HPLC) and molecular weight was analyzed by liquid chromatography (LC-MS), confirming that the synthesized nucleic acid sequences were siRNA corresponding to each conjugate in tables 6A-6D and the conjugate of the comparative example.
Preparation example 9 Synthesis of siRNA conjugates of conjugates 28-31, 96-99, 130-133 and 152-155 and control conjugate 2-5
siRNA conjugates of conjugates 28-31, 96-99, 130-133 and 152-155 and control conjugates 2-5 were prepared by the same method as in preparation example 7, except that: 1) The conjugate molecules of the conjugates obtained in preparation examples 2-6 and the comparative conjugate described above were used in place of the B-2 conjugate molecule (e.g., when the B-2 conjugate molecule was replaced with the D-6 conjugate molecule of conjugate 2, the D7 conjugates of conjugates 28, 96, 130 and 152 were obtained, and when the D-6 conjugate molecule was replaced with the C-2 conjugate molecule of conjugate 3, the C3 conjugates of conjugates 29, 97, 131 and 153 were obtained, and so forth); 2) Conjugated siRNAs have the sequences shown in tables 6A-6D corresponding to conjugates 28-31, 96-99, 130-133 and 152-155, and comparative conjugates 2-5; 3) When phosphorothioate linkage is between two nucleotides in the target sequence, the oxidation step in the linkage of the latter one of the two nucleotides is replaced with the vulcanization step described in preparation example 8; and 4) when all nucleotides in the target sequence are modified hydroxyl groups at the 2 '-position, the step of removing 2' -TBDMS protection on ribose is not included in the cleavage and deprotection conditions. Thus, siRNA conjugates of conjugates 28-31, 96-99, 130-133 and 152-155 of the present disclosure and comparative conjugates 2-5 were prepared and numbered as shown in tables 6A-6D, respectively. Then, the molecular weight of the single strand and the double strand is detected by a liquid chromatograph/mass spectrometer, the actual measurement value accords with the theoretical value, the structure of each synthesized conjugate siRNA conjugate is confirmed to be shown as formulas (402), (403), (404) and (405), and the structure of each contrast conjugate is shown as formula (901):
Experimental example 1 this experiment illustrates animal level toxicity of the siRNA conjugates of the present disclosure.
300mg/kg (as siRNA) of the siRNA conjugates 21-31, 156-157, 83, 120 and 148, respectively, were administered subcutaneously to each of the C57BL/6J mice (purchased from Peking Vitre laboratory animal technologies Co., ltd.) in a single dose, and no death of the animals or clinical symptoms associated with adverse drug reactions were observed for 14 consecutive days, and no abnormalities were observed in the general anatomy. Thus, the above results indicate that the siRNA conjugates of the present disclosure have lower animal level toxicity.
In experimental examples 2 to 5 below, properties and effects of the siRNA conjugates of tables 6A to 6D were respectively verified according to siRNA target positions and sequence relatedness.
Experimental example 2 Effect of siRNA conjugates of Table 6A
Experimental example 2-1 this experiment demonstrates the inhibition efficiency of the siRNA conjugates of table 6A on HBV mRNA expression level in vitro (in vitro).
HepG2.2.15 cells were transfected in vitro with siRNA conjugates of conjugates 7-8, 13-15, 21-22, 24-31, 156-157 and control conjugate 2, at final siRNA concentrations of 50nM, 10nM, 1nM, respectively. At least 3 experiments were repeated for 3 duplicate wells per concentration. The expression level of HBV mRNA in the above-harvested cells was determined by real-time fluorescent quantitative PCR (real-time fluorescent qPCR) 48 hours after transfection, specifically: total RNA was extracted using RNeasy Mini Kit (QIAGEN, cat. No. 74106) according to the instructions thereof, and the extracted total RNA was reverse transcribed into cDNA, followed by measuring the inhibition efficiency of siRNA on HBV mRNA expression of HepG2.2.15 cells by the fluorescent quantitative PCR method.
In this fluorescent quantitative PCR method, HBV and GAPDH were detected using a primer for HBV and a primer for GAPDH, respectively, with the GAPDH gene as an internal gene, the primer sequences being shown in Table 7A:
TABLE 7A sequence of detection primers
In the fluorescent quantitative PCR method, the siRNA inhibitory activity was expressed as the remaining amount of HBV gene expression, calculated as follows:
HBV gene expression remaining = (copy number of HBV gene in test group/copy number of GAPDH in test group)/(copy number of HBV gene in mock group/copy number of GAPDH in mock group) ×100%,
the inhibition of mRNA by siRNA was then calculated according to the following formula:
mRNA inhibition ratio= (residual amount of 1-HBV gene expression) ×100%,
wherein each test group was hepg2.2.15 cells treated with the siRNA conjugates listed in table 6A, respectively, including conjugates 7-8, 13-15, 21-22, 24-31, 156-157 and control conjugate 2, and mock group was hepg2.2.15 cells not treated with any siRNA.
Table 8A below shows the results of the detection of HBV mRNA expression inhibitory activity of the siRNA conjugates listed in table 6A with the control conjugates in hepg2.2.15 cells.
TABLE 8A siRNA conjugated in vitro Activity test
/>
As can be seen from the results of table 8A, each siRNA conjugate in table 6A showed excellent HBV gene expression inhibitory activity at the cellular level.
Experimental example 2-2 this experiment demonstrates the stability of siRNA conjugates of Table 6A in human plasma in vitro
Conjugates 13-14 and 21-25 (0.9% NS solution for each siRNA concentration, 20. Mu.M, 12. Mu.l) were mixed with 108. Mu.L of 90% Human plasma (Human plasma, PBS dilution) respectively. Incubate at 37 ℃. 10 mu L of samples are taken out at 0, 8, 24 and 48 hours respectively, and immediately frozen in a refrigerator at-80 ℃ by liquid nitrogen. After sampling at each time point, 10. Mu.L of each sample was taken after 5-fold dilution with 1 XPBS (pH 7.4); meanwhile, an equimolar amount of siRNA conjugate (siRNA concentration 2. Mu.M, 2. Mu.l) was mixed with 8. Mu.l of 1 XPBS (pH 7.4) to prepare 10. Mu.l of a sample not treated with human plasma, which was designated as "untreated". A20 wt% non-denaturing polyacrylamide gel was prepared, and the sample was mixed with 4. Mu.L of loading buffer (20 mM EDTA,36 wt% glycerol, 0.06 wt% bromophenol blue) and loaded, followed by electrophoresis under 80mA constant current conditions for about 60 minutes. After electrophoresis was completed, the sample was stained with 1×sybr Gold dye (Invitrogen, cat.11494) for 15 minutes and then imaged, and the results are shown in table 9A.
Table 9A shows the results of semi-quantitative detection of stability of the test siRNA conjugates listed in table 6A versus control siRNA conjugates in human plasma in vitro. The results are expressed as the ratio of the longest fragment remaining after incubation of the test and control siRNA conjugates with human plasma to the longest fragment of untreated siRNA (ratio of the longest fragments, RL).
Table 9A plasma stability quantification results of siRNA conjugates
As can be seen from the results of table 9A, each siRNA conjugate has excellent stability in plasma.
Experimental examples 2-3 this experiment demonstrates the inhibition efficiency of siRNA conjugates of Table 6A on HBV Mra expression level in vivo (in vivo)
In this experimental example, the inhibition efficiency of the siRNA conjugates of examples 13 to 14, 21 to 22 and 156 to 157 and the control conjugate 2 on the HBV mRNA expression amount in HBV transgenic mice C57BL/6J-Tg (Alb 1 HBV) 44Bri/J was examined.
HBV transgenic mice C57BL/6J-Tg (Alb 1 HBV) 44Bri/J used in this experimental example were purchased from the university of Beijing, department of laboratory animal science.
First, C57BL/6J-Tg (Alb 1 HBV) 44Bri/J mice (all females) were numbered according to the siRNA conjugates in Table 3A for each group of 5 mice, and PBS control groups were added. All animals were dosed on a weight basis in a single dose (subcutaneous administration) at a dose of 1mg/kg and a volume of 5ml/kg. Animals were sacrificed on day 14 post-dose, livers were collected and saved with RNA later (Sigma Aldrich company); homogenizing liver tissue with tissue homogenizer, and extracting with Trizol according to standard procedure of total RNA extraction to obtain total RNA.
The expression level of HBV mRNA in liver tissue was detected using real-time fluorescent quantitative PCR, specifically: the extracted total RNA was reverse transcribed into cDNA using an ImProm-IITM reverse transcription kit (Promega company) according to the instructions thereof, and then the inhibition efficiency of siRNA on HBV mRNA expression in liver tissue was examined using a fluorescent quantitative PCR kit (Beijing kang is century Biotech Co.). In the fluorescent quantitative PCR method, the HBV and the beta-actin are detected by using a primer for HBV and a primer for beta-actin, respectively, with the beta-actin (beta-actin) gene as an internal reference gene.
The sequence of the detection primer is shown in Table 10A.
TABLE 10A sequence of detection primers
In the fluorescent quantitative PCR method, the siRNA inhibitory activity was expressed as the remaining amount of HBV gene expression, calculated as follows:
HBV gene expression residual amount = (copy number of HBV gene in test group/copy number of beta-actin in test group)/(copy number of HBV gene in control group/copy number of beta-actin in control group) ×100%,
mRNA inhibition was then calculated according to the following formula:
mRNA inhibition ratio= (residual amount of 1-HBV gene expression) ×100%,
wherein, the control group is a control group mouse to which PBS is applied in the experiment, and each test group is a dosing group mouse to which different siRNA conjugates are respectively applied. The results are shown in table 11A below.
Table 11A inhibition of HBV mRNA expression by siRNA conjugates in mouse liver
As can be seen from the above results, in one aspect, the conjugates of the various embodiments of the present disclosure all showed high HBV mRNA inhibition activity in mice; on the other hand, although the results of table 8A indicate that the conjugate of comparative conjugate 2 showed similar in vitro HBV gene inhibition activity as the conjugate of the present disclosure, it can be seen from the results of table 11A that the siRNA conjugates of examples 21 and 156-157 showed significantly higher inhibition rate of HBV gene mRNA of liver tissue of hepatitis b mouse in vivo experiments, compared to comparative conjugate 2 having different conjugate groups, in the case of identical nucleic acid sequence and identical base modification scheme. This also demonstrates that the siRNA conjugates of the present disclosure have good in vivo delivery efficiency.
Experimental examples 2-4 this experiment demonstrates the time-dependent test of the expression level of siRNA conjugate of Table 6A on serum HBsAg and the inhibition efficiency of HBV DNA in HBV transgenic mice
AAV-HBV models were prepared according to literature methods (Dong Xiaoyan et al, chin J Biotech 2010, may 25;26 (5): 679-686), rAAV8-1.3HBV, form D (ayw), purchased from Beijing acanthopanax and molecular medicine research, inc., 1X 10 12 viral genome (v.g.)/mL, lot 2016123011. Dilution with sterile PBS to 5X 10 before the experiment 11 g./mL. 200. Mu.L of each mouse, i.e., 1X 10 of each mouse, was injected 11 v.g. On day 28 post-viral injection, all mice were collected via the orbit (approximately 100 μl) for serum collection for detection of HBsAg and HBV DNA. After successful animal modeling, the siRNA conjugates of conjugates 13-14, 21-22, 24 and 156, respectively, were given as random groupings (5 per group) of serum HBsAg content, as well as PBS blank. All animals were dosed subcutaneously in a single dose of 3mg/kg and in a volume of 5ml/kg, calculated from body weight. Serum HBsAg levels were measured at various time points by bleeding from the orbital venous plexus of mice before and after dosing at days 7, 14, 21, 28, 56, 84, 112, 140.
The orbit blood is taken about 100 mu l each time, and the serum is not less than 20 mu l after centrifugation. Detecting the expression level of HBsAg in serum using HBsAg CLIA kit (amblycephala, CL 0310); DNA in serum was extracted by referring to QIAamp 96DNA Blood Kit instructions, and quantitative PCR was performed to detect the expression level of HBV DNA.
The HBsAg inhibition was calculated as follows:
HBsAg inhibition = (1-HBsAg content after dosing/HBsAg content before dosing) ×100%. Wherein the HBsAg content is expressed as the number of equivalents (UI) of HBsAg per milliliter (ml) of serum.
HBV DNA inhibition was calculated as follows:
HBV DNA inhibition = (1-HBV DNA content after administration/HBV DNA content before administration) ×100%.
Wherein HBV DNA content is expressed as how many copies of HBV DNA are contained per milliliter (ml) of serum.
The results are shown in Table 12A and Table 13A below.
Table 12A inhibition of HBsAg expression of siRNA conjugates in mouse serum
As can be seen from the results of table 12A, the PBS negative control group did not show any inhibition at different time points after administration; in contrast, the siRNA conjugates of each example exhibited excellent HBsAg inhibition effect on HBsAg at different time points after administration. In particular, conjugates 21 and 156 showed a high serum HBsAg inhibition rate for a long period of 140 days, indicating that they were able to stably and efficiently inhibit HBV gene expression for a long period of time.
TABLE 13 inhibition of HBV DNA expression of siRNA conjugates in mouse serum
As can be seen from the results of table 13A, the siRNA conjugates of the examples also showed high inhibition of HBV DNA expression, and maintained high inhibition rate for up to 84 days.
Experimental example 3 effect experiments of siRNA conjugates of table 6B
Experimental example 3-1 this experiment demonstrates the inhibition efficiency of the siRNA conjugates of table 6B on HBV mRNA expression level in vitro (in vitro).
HepG2.2.15 cells were transfected in vitro with siRNA conjugates of conjugates 53-56, 81-92, 158-159 and control conjugate 3, at final siRNA concentrations of 50nM, 10nM, 1nM, respectively. At least 3 experiments were repeated for 3 duplicate wells per concentration. The expression level of HBV mRNA in the above-harvested cells was determined by real-time fluorescent quantitative PCR (real-time fluorescent qPCR) 48 hours after transfection, specifically: total RNA was extracted using RNeasy Mini Kit (QIAGEN, cat. No. 74106) according to the instructions thereof, and the extracted total RNA was reverse transcribed into cDNA, followed by measuring the inhibition efficiency of siRNA on HBV mRNA expression of HepG2.2.15 cells by the fluorescent quantitative PCR method.
In this fluorescent quantitative PCR method, HBV and GAPDH were detected using a primer for HBV and a primer for GAPDH, respectively, with the GAPDH gene as an internal gene, the primer sequences being shown in Table 7B:
TABLE 7B sequence of detection primers
In the fluorescent quantitative PCR method, the siRNA inhibitory activity was expressed as the remaining amount of HBV gene expression, calculated as follows:
HBV gene expression remaining = (copy number of HBV gene in test group/copy number of GAPDH in test group)/(copy number of HBV gene in mock group/copy number of GAPDH in mock group) ×100%,
The inhibition of mRNA by siRNA was then calculated according to the following formula:
mRNA inhibition ratio= (residual amount of 1-HBV gene expression) ×100%,
wherein each test group was hepg2.2.15 cells treated with the siRNA conjugates listed in table 6B, respectively, including the siRNA conjugates of conjugates 53-56, 81-92, 158-159 and the control siRNA conjugate of control conjugate 3; mock groups were hepg2.2.15 cells not treated with any siRNA.
Table 8B below shows the results of the detection of HBV mRNA expression inhibitory activity of the test siRNA conjugates listed in table 6B with the control siRNA conjugates in hepg2.2.15 cells.
TABLE 8B siRNA conjugated in vitro Activity test
As can be seen from the results of table 8B, each siRNA conjugate in table 6B showed excellent HBV gene expression inhibitory activity at the cellular level.
Experimental example 3-2 this experiment demonstrates the stability of siRNA conjugates of Table 6B in human plasma in vitro
The siRNA conjugates of conjugates 53-58, 83-88 and control conjugate 3 (siRNA concentrations were 20. Mu.M, 12. Mu.l) were mixed with 108. Mu.L of 90% Human plasma (Human plasma, PBS dilution), respectively. Incubate at 37 ℃. 10 mu L of samples are taken out at 0, 8, 24 and 48 hours respectively, and immediately frozen in a refrigerator at-80 ℃ by liquid nitrogen. After sampling at each time point, 10. Mu.L of each sample was diluted 5 times with 1 XPBS (pH 7.4), 20 wt% of a non-denaturing polyacrylamide gel was prepared, and the sample was mixed with 4. Mu.L of a loading buffer (20 mM EDTA,36 wt% glycerol, 0.06 wt% bromophenol blue) and then loaded, and electrophoresis was performed under 80mA constant current for about 60 minutes. After electrophoresis was completed, the sample was stained with 1×sybr Gold dye (Invitrogen, cat.11494) for 15 minutes and then imaged, and the results are shown in table 9B.
Table 9B shows the results of semi-quantitative detection of stability of the siRNA conjugates listed in table 6B with the control conjugates in human plasma in vitro. The results are expressed as the Ratio (RL) of the longest fragment remaining after incubation of the siRNA conjugate with human plasma to the longest fragment of untreated siRNA.
Table 9B plasma stability quantification results of siRNA conjugates
As can be seen from the results of table 9B, each siRNA conjugate has excellent stability in plasma.
Experimental example 3-3 this experiment demonstrates the inhibition efficiency of siRNA conjugates of Table 6B on HBV mRNA expression in vivo (in vivo)
In this experimental example, the inhibition efficiency of the siRNA conjugates of conjugates 53 to 54, 57 to 58, 81 to 88 and control conjugate 3 on HBV mRNA expression level in HBV transgenic mice C57BL/6J-Tg (Alb 1 HBV) 44Bri/J was examined.
HBV transgenic mice C57BL/6J-Tg (Alb 1 HBV) 44Bri/J used in this experimental example were purchased from the university of Beijing, department of laboratory animal science.
First, C57BL/6J-Tg (Alb 1 HBV) 44Bri/J mice were randomly grouped (females each) with serum HbsAg content, 5 mice per group, numbered according to siRNA conjugates in Table 6B, and PBS control was added. All animals were dosed on a weight basis in a single dose (subcutaneous administration) at a dose of 1mg/kg and a volume of 5ml/kg. Animals were sacrificed on day 14 post-dose, livers were collected and saved with RNA later (Sigma Aldrich company); homogenizing liver tissue with tissue homogenizer, and extracting with Trizol according to standard procedure of total RNA extraction to obtain total RNA.
The expression level of HBV mRNA in liver tissue was detected using real-time fluorescent quantitative PCR, specifically: the extracted total RNA was reverse transcribed into cDNA using an ImProm-IITM reverse transcription kit (Promega company) according to the instructions thereof, and then the inhibition efficiency of siRNA on HBV mRNA expression in liver tissue was examined using a fluorescent quantitative PCR kit (Beijing kang is century Biotech Co.). In the fluorescent quantitative PCR method, the HBV and the beta-actin are detected by using a primer for HBV and a primer for beta-actin, respectively, with the beta-actin (beta-actin) gene as an internal reference gene.
The sequence of the detection primer is shown in Table 10B.
TABLE 10B sequence of detection primers
In the fluorescent quantitative PCR method, the siRNA inhibitory activity was expressed as the remaining amount of HBV gene expression, calculated as follows:
HBV gene expression residual amount = (copy number of HBV gene in test group/copy number of beta-actin in test group)/(copy number of HBV gene in control group/copy number of beta-actin in control group) ×100%,
mRNA inhibition was then calculated according to the following formula:
mRNA inhibition ratio= (residual amount of 1-HBV gene expression) ×100%,
wherein, the control group is a control group mouse to which PBS is applied in the experiment, and each test group is a dosing group mouse to which different siRNA conjugates are respectively applied. The results are shown in table 11B below.
Table 11B inhibition of HBV mRNA expression by siRNA conjugates in mouse liver
As can be seen from the above results, in one aspect, the conjugates of the various embodiments of the present disclosure all showed high HBV mRNA inhibition activity in mice; on the other hand, although the results of table 8B show that the comparative conjugate 3 showed similar in vitro HBV gene inhibition activity as the conjugate of the present disclosure, it is known from the results of table 11B that, in the case of the same nucleic acid sequence and the same base modification scheme, the conjugate 83 showed significantly higher inhibition rate of HBV gene mRNA of hepatitis B mouse liver tissue in vivo experiment as compared to the comparative conjugate 3 having different conjugate groups. This also demonstrates that the siRNA conjugates of the present disclosure have good in vivo delivery efficiency.
Experimental examples 3-4 this experiment demonstrates the time-dependent test of the expression level of siRNA of table 6B on serum HBsAg and the inhibition efficiency of HBV DNA in HBV transgenic mice
AAV-HBV models were prepared according to literature methods (Dong Xiaoyan et al, chin J Biotech 2010, may 25;26 (5): 679-686), rAAV8-1.3HBV, form D (ayw), purchased from Beijing acanthopanax and molecular medicine research, inc., 1X 10 12 viral genome(v.g.)/mLLot number 2016123011. Dilution with sterile PBS to 5X 10 before the experiment 11 g./mL. 200. Mu.L of each mouse, i.e., 1X 10 of each mouse, was injected 11 v.g. On day 28 post-viral injection, all mice were collected via the orbit (approximately 100 μl) for serum collection for detection of HBsAg and HBV DNA. After successful animal modeling, the siRNA conjugates of conjugates 87-88 and 158-159, respectively, were given as random groupings (5 per group) of serum HBsAg levels, as well as PBS blank. All animals were dosed subcutaneously in a single dose of 3mg/kg and in a volume of 5ml/kg, calculated from body weight. Serum HBsAg levels were measured at various time points by bleeding from the orbital venous plexus of mice before and after dosing on days 7, 14, 21, 28, 56, 84.
The orbit blood is taken about 100 mu l each time, and the serum is not less than 20 mu l after centrifugation. Detecting the expression level of HBsAg in serum using HBsAg CLIA kit (amblycephala, CL 0310); DNA in serum was extracted by referring to QIAamp 96DNA Blood Kit instructions, and quantitative PCR was performed to detect the expression level of HBV DNA.
The HBsAg inhibition was calculated as follows:
HBsAg inhibition = (1-HBsAg content after dosing/HBsAg content before dosing) ×100%. Wherein the HBsAg content is expressed as the number of equivalents (UI) of HBsAg per milliliter (ml) of serum.
HBV DNA inhibition was calculated as follows:
HBV DNA inhibition = (1-HBV DNA content after administration/HBV DNA content before administration) ×100%.
Wherein HBV DNA content is expressed as how many copies of HBV DNA are contained per milliliter (ml) of serum.
The results are shown in tables 12B and 13B below.
Table 12B inhibition of HBsAg expression of siRNA conjugates in mouse serum
As can be seen from the results in table 12B, the PBS negative control group did not show any inhibition at different time points after dosing; in contrast, the siRNA conjugates of each conjugate exhibited excellent HBsAg inhibition effect on HBsAg at different time points after administration.
TABLE 13 inhibition of HBV DNA expression of siRNA conjugates in mouse serum
As can be seen from table 13B, the siRNA conjugates of the respective examples also showed high-efficiency HBV DNA expression inhibition, similarly to the HBsAg inhibition effect, and the inhibition rate remained substantially stable for a period of up to 84 days.
Experimental example 4 Effect test of siRNA conjugates of Table 6C
Experimental example 4-1 this experiment demonstrates the inhibition efficiency of the siRNA conjugates of table 6C on ANGPTL3mRNA expression levels in vitro (in vitro).
Human hepatoma cell line Huh7 was transfected in vitro with siRNA conjugates of conjugates 100-107, 116-117, 124-133 and 160-161 and control conjugate 4 at final siRNA conjugate concentrations (based on siRNA) of 5nM, 0.5nM, 0.05nM, respectively, 3 replicates per concentration and at least 3 replicates were tested.
Specifically, huh7 cells were seeded in 24 well plates at a 4X 10 seeding density with DMEM complete medium containing 10% fetal bovine serum 5 Cells/well, 0.5mL of medium per well, were incubated overnight at 37 ℃.
Cell culture broth was pipetted off from the 24-well plate and 0.5mL Opti-MEM serum-free medium was added to each well. 1.5. Mu.L concentrations (based on siRNA) of 0.02. Mu.M, 0.2. Mu.M and 2. Mu.M siRNA conjugates, respectively, were diluted with 50. Mu.L of Opti-MEM serum-free medium; mu.L Lipofectamine TM 2000 (Invitrogen corporation) was diluted in 50. Mu.L of Opti-MEM serum-free medium, mixed and incubated at room temperature for 5 minutes; mixing diluted siRNA conjugates and diluted Lipofectamine TM 2000, gently mixed, and allowed to stand at room temperature for 20 minutes to allow the formation of transfection complexes. The final mixed solution described above was added at 100. Mu.L per well in 24-well plates seeded with Huh7 cells. The final concentration of siRNA conjugates was 0.05nM, 0.5nM, 5nM. Cells were cultured at 37℃for 4 hours and then to each1mL of DMEM complete medium containing 10% fetal bovine serum was added to the wells and the incubation continued overnight at 37 ℃.
The expression level of ANGPTL3mRNA in Huh7 cells transfected with each siRNA conjugate was separately detected by real-Time fluorescent quantitative PCR (Quantitative Real-Time PCR). The method comprises the following specific steps: after 24 hours of culturing the transfected cells, total RNA in the cells was extracted using RNAVzol (Vigorius, inc., cat. N002); mu.g of total RNA was taken and subjected to reverse transcription to obtain cDNA according to the method of using a reverse transcription kit (Promega Corp., cat. No. A3500). The amount of ANGPTL3mRNA expressed was detected by using a 2× Ultra SYBR Mixture (with ROX) (bejinkang, century biotechnology, inc., cat No. CW 0956) kit and using cDNA as a template according to the procedure of the specification. Among them, PCR primers for amplifying ANGPTL3 and beta-actin as an internal reference gene are shown in Table 7C.
TABLE 7C sequence of detection primers
The inhibition rate of the siRNA on ANGPTL3mRNA expression level was calculated as follows: inhibition ratio = [1- (expression amount of ANGPTL3mRNA in test group/expression amount of β -action mRNA in test group)/(expression amount of ANGPTL3mRNA in mock group/expression amount of β -action mRNA in mock group) ] ×100%. Wherein each test group was Huh7 cells treated with the siRNA conjugates listed in table 3E, respectively, including siRNA conjugates of conjugates 100-107, 116-117, 124-133 and 160-161 and control siRNA conjugate of control conjugate 4; mock groups were Huh7 cells not treated with any siRNA conjugate. The results of the detection of ANGPTL3mRNA expression inhibitory activity of the example siRNA conjugates listed in table 6C and the comparative example siRNA conjugates in Huh7 cells are shown in table 8C below.
TABLE 8C siRNA conjugated in vitro Activity test
/>
From the results of table 8C, it can be seen that each siRNA conjugate in table 6C showed excellent ANGPTL3mRNA expression inhibition activity at the cellular level at each concentration, the inhibition rate of the siRNA conjugate reached 50% or more at 5nM concentration, and some conjugates reached 70% or more inhibition rate.
Experimental example 4-2 this experiment demonstrates the stability of siRNA conjugates of Table 6C in human plasma in vitro
The siRNA conjugates of conjugates 124-125 and control conjugate 4 (20. Mu.M, 12. Mu.l based on siRNA concentration) were mixed with 108. Mu.L of 90% Human plasma (Human plasma, PBS dilution), respectively. Incubate at 37 ℃. 10 mu L of samples are taken out at 0, 8, 24 and 48 hours respectively, and immediately frozen in a refrigerator at-80 ℃ by liquid nitrogen. After sampling at each time point, 10. Mu.L of each sample was diluted 5 times with 1 XPBS (pH 7.4), 20 wt% of a non-denaturing polyacrylamide gel was prepared, and the sample was mixed with 4. Mu.L of a loading buffer (20 mM EDTA,36 wt% glycerol, 0.06 wt% bromophenol blue) and then loaded, and electrophoresis was performed under 80mA constant current for about 60 minutes. After electrophoresis was completed, the gel was stained with 1×sybr Gold dye (Invitrogen, cat.11494) for 15 minutes and then phase was formed, and the results are shown in table 9C.
Table 9C shows the results of semi-quantitative detection of stability of the siRNA conjugates listed in table 6C with the comparative siRNA conjugates in human plasma in vitro. The results are expressed as the Ratio (RL) of the longest fragment remaining after incubation of the siRNA conjugate with human plasma to the longest fragment of untreated siRNA and the comparative siRNA conjugate.
Table 9C plasma stability quantification results of siRNA conjugates
As can be seen from the results of table 9C, the siRNA conjugates of the present disclosure have excellent stability in plasma with little degradation within 48 hours.
Experimental example 4-3 this experiment demonstrates the inhibition efficiency of siRNA conjugates of Table 6C on the expression level of ANGPTL3mRNA in vivo (in vivo)
In this experimental example, the inhibition rate of ANGPTL3 expression levels in liver tissue by siRNA conjugates of conjugates 120, 123, 124, 160-161 and control conjugate 4 in normal BALB/c mice was examined.
Normal BALB/c mice (purchased from beijing villous laboratory animal technologies limited) 6-8 weeks old were randomly grouped, 6 (male and female halves) each, and each group was given siRNA conjugates of conjugates 120, 123, 124, 160-161 and control conjugate 4, respectively, along with PBS. All animals were dosed on a weight basis in a single dose by subcutaneous injection, with an siRNA conjugate dose (based on the amount of siRNA) of 3mg/kg and a dosing volume of 10mL/kg. Mice were sacrificed 14 days after dosing, livers were collected and saved with RNA later (Sigma Aldrich company); liver tissue was homogenized using a tissue homogenizer and total RNA was extracted using Trizol (Thermo Fisher Co.) according to standard procedures for total RNA extraction.
Detecting the expression level of ANGPTL3mRNA in liver tissue using real-time fluorescent quantitative PCR, specifically: using ImProm-II TM The total RNA extracted was reverse transcribed into cDNA using a reverse transcription kit (Promega) according to the instructions thereof, and the inhibition efficiency of siRNA on the expression of ANGPTL3mRNA in liver tissue was then examined using a fluorescent quantitative PCR kit (Beijing Ka, century Biotechnology Co., ltd.). In this fluorescent quantitative PCR method, the beta-actin (beta-actin) gene was used as a reference gene, and the ANGPTL3 and beta-actin were detected using a primer for ANGPTL3 and a primer for beta-actin, respectively.
The sequence of the detection primers is shown in Table 10C.
TABLE 10C sequence of detection primers
The inhibition rate of the siRNA on ANGPTL3mRNA expression level was calculated as follows: inhibition ratio = [1- (expression amount of ANGPTL3mRNA in test group/expression amount of β -action mRNA in test group)/(expression amount of ANGPTL3mRNA in control group/expression amount of β -action mRNA in control group) ]×100%. Wherein, the control group is a control group mouse to which PBS is applied in the experiment, and each test group is a dosing group mouse to which different siRNA conjugates are respectively applied. The results are shown in table 11C below.
TABLE 11 inhibition of ANGPTL3mRNA expression by siRNA conjugates in mouse liver
As can be seen from the results of table 11C, on the one hand, the siRNA conjugates of the conjugates 120, 123, 124, 160-161 of the present disclosure all showed extremely high inhibitory activity of ANGPTL3mRNA compared to PBS; on the other hand, in the case of the same nucleic acid sequence and the same modification scheme, the siRNA conjugates shown in conjugates 124 and 160-161 showed higher mRNA inhibition rate in vivo experiments compared to the comparative conjugate 4 having different conjugate groups, which also indicates that the siRNA conjugates of the present disclosure have good in vivo delivery efficiency.
Experimental example 4-4 this experiment demonstrates the inhibition efficiency of siRNA conjugates of Table 6C on the expression level of ANGPTL3mRNA in vivo (in vivo) and the effect on blood lipid
In this experimental example, the effect of the siRNA conjugates of example 120 (B3-sial 1M1 SP) and example 124 (B3-sial 1M3 SP) on the inhibition of the expression level of ANGPTL3 in liver tissue and on the total Cholesterol (CHO), triglyceride (TG) and low-density lipoprotein (LDL-c) levels in serum in vivo in ob/ob model mice was examined.
Female mice of 6-8 weeks old ob/ob (purchased from the company, kwangsi laboratory animal Co., ltd.) were randomly divided into 5 groups of 5 animals each, each group being as follows: (1) PBS control group; (2) conjugate 120.3 mg/kg group; (3) conjugate 124, 3mg/kg group; (4) conjugate 120 1mg/kg group; (5) conjugate 124 1mg/kg group. All animals were dosed on a single dose basis by subcutaneous injection, with a dosing volume of 10mL/kg, based on body weight.
Blood was collected from the orbit (about 100 μl) 2 days prior to dosing (noted as-2 days), and 7, 14, 21, 28, 35, 42, 49 days post dosing, respectively, for the detection of blood lipid levels.
Mice were sacrificed on day 49 and livers were collected and saved with RNA later (Sigma Aldrich company); liver tissue was homogenized using a tissue homogenizer and total RNA was extracted using Trizol (Thermo Fisher Co.) according to standard procedures for total RNA extraction.
The expression level of ANGPTL3mRNA in liver tissue was detected by real-time fluorescent quantitative PCR using the same method as in experimental example 4-3. The results are shown in table 12C below.
Table 12C inhibition of ANGPTL3mRNA expression in mouse liver by siRNA conjugates
Blood collected from the eyebox is centrifuged to obtain serum, the content of total Cholesterol (CHO), triglyceride (TG) and low density lipoprotein (LDL-c) in the serum is further detected by using a PM1P000/3 full-automatic serum biochemical analyzer (SABA, italy), the blood lipid result is standardized, and the inhibition rate of the blood lipid level is calculated according to the following equation: inhibition = (1-post-dose test group blood lipid content/pre-dose test group blood lipid content) ×100%. Blood lipid refers to total cholesterol, triglycerides or low density lipoproteins. The detection results are shown in tables 13C, 14C and 15C below.
TABLE 13 influence of siRNA conjugates on total cholesterol expression levels in mouse serum
TABLE 14 influence of siRNA conjugates on triglyceride expression levels in mouse serum
TABLE 15 influence of siRNA conjugates on the expression level of low density lipoproteins in mouse serum
As can be seen from the results of tables 13C, 14C and 15C above, the siRNA conjugates of conjugate 120 and conjugate 124 at different doses each significantly inhibited ANGPTL3 expression in mouse liver tissue, with a significant dose response; the siRNA conjugate (B3-sial 1M1 SP) of conjugate 120 had an inhibition rate of 48.9% on ANGPTL3 gene expression at a low dose of 1 mg/kg; the inhibition rate of the ANGPTL3 gene expression reaches 80.8% at a high dose of 3 mg/kg; at the same time, the content of CHO, TG and LDL-c in the serum of the mice was monitored, and the results showed that the content of CHO, TG and LDL-c in the serum of the mice treated with the siRNA conjugates of the conjugates 120 and 124 was significantly reduced, and still showed a higher blood lipid lowering effect at least at 49 days.
Experimental example 5 effect test of siRNA conjugates of table 6D
Experimental example 5-1 this experiment demonstrates the inhibition efficiency of the siRNA conjugates of table 6D on APOC3mRNA expression levels in vitro (in vitro).
Human hepatoma cell line Huh7 was transfected in vitro with siRNA conjugates of conjugates 134-135, 146-151, 162-163 and control conjugate 5, at final concentrations (based on siRNA) of 5nM, 0.5nM, 0.05nM, 3 replicates per concentration, at least 3 replicates.
Specifically, huh7 cells were seeded in 24 well plates at a 4X 10 seeding density with DMEM complete medium containing 10% fetal bovine serum 5 Cells/well, 0.5mL of medium per well, were incubated overnight at 37 ℃.
Cell culture broth was pipetted off from the 24-well plate and 0.5mL Opti-MEM serum-free medium was added to each well. 1.5. Mu.L concentrations (based on siRNA) of 0.02. Mu.M, 0.2. Mu.M and 2. Mu.M siRNA conjugates, respectively, were diluted with 50. Mu.L of Opti-MEM serum-free medium; mu.L Lipofectamine TM 2000 (Invitrogen corporation) was diluted in 50. Mu.L of Opti-MEM serum-free medium, mixed and incubated at room temperature for 5 minutes; mixing diluted siRNA conjugates and diluted Lipofectamine TM 2000, gently mix, and rest at room temperature for 20 minutes to allow transfectionFormation of a complex. The final mixed solution described above was added at 100. Mu.L per well in 24-well plates seeded with Huh7 cells. The final concentration of siRNA conjugates was 0.05nM, 0.5nM, 5nM. Cells were incubated at 37℃for 4 hours, and 1mL of DMEM complete medium containing 10% fetal bovine serum was added to each well, followed by incubation at 37℃overnight.
The expression level of APOC3mRNA in Huh7 cells transfected with each siRNA conjugate was detected by real-Time fluorescent quantitative PCR (Quantitative Real-Time PCR), respectively. The method comprises the following specific steps: after 24 hours of culturing the transfected cells, total RNA in the cells was extracted using RNAVzol (Vigorius, inc., cat. N002); mu.g of total RNA was taken and subjected to reverse transcription to obtain cDNA according to the method of using a reverse transcription kit (Promega Corp., cat. No. A3500). The detection of the expression level of APOC3mRNA was performed according to the procedure of the specification using a 2× Ultra SYBR Mixture (with ROX) (bekyokang, century biotechnology limited, cat No. CW 0956) kit and using cDNA as a template. Among them, PCR primers for amplifying APOC3 and beta-actin as an internal reference gene are shown in Table 7D.
TABLE 7D sequence of detection primers
The inhibition rate of the expression level of the APOC3mRNA by the siRNA is calculated according to the following equation: inhibition ratio = [1- (expression amount of ANGPTL3mRNA in test group/expression amount of β -action mRNA in test group)/(expression amount of APOC3mRNA in mock group/expression amount of β -action mRNA in mock group) ] ×100%. Wherein each test group was Huh7 cells treated with the siRNA conjugates listed in table 6D, respectively, including siRNA conjugates of conjugates 134-135, 146-151, 162-163 and control siRNA conjugate of control conjugate 5; mock groups were Huh7 cells not treated with any siRNA conjugate. The results of the detection of APOC3mRNA expression inhibitory activity of the example siRNA conjugates listed in table 6D versus the comparative siRNA conjugates in Huh7 cells are shown in table 8D below.
Table 8D siRNA conjugated in vitro activity test
As can be seen from the results of table 8D, each siRNA conjugate in table 6D showed excellent APOC3mRNA expression inhibitory activity at the cellular level at each concentration.
Experimental example 5-2 this experiment demonstrates the stability of siRNA conjugates of Table 6D in human plasma in vitro
The siRNA conjugates of conjugates 150-155, 162-163 and control conjugate 5 (20. Mu.M, 12. Mu.l based on siRNA concentration) were mixed with 108. Mu.L of 90% Human plasma (Human plasma, PBS dilution), respectively. Incubate at 37 ℃. 10 mu L of samples are taken out at 0, 8, 24 and 48 hours respectively, and immediately frozen in a refrigerator at-80 ℃ by liquid nitrogen. After sampling at each time point, 10. Mu.L of each sample was diluted 5 times with 1 XPBS (pH 7.4), 20 wt% of a non-denaturing polyacrylamide gel was prepared, and the sample was mixed with 4. Mu.L of a loading buffer (20 mM EDTA,36 wt% glycerol, 0.06 wt% bromophenol blue) and then loaded, and electrophoresis was performed under 80mA constant current for about 60 minutes. After electrophoresis was completed, the gel was stained with 1×sybr Gold dye (Invitrogen, cat.11494) for 15 minutes and then phase was formed, and the results are shown in table 9D.
Table 9D shows the results of semi-quantitative detection of the stability of the example siRNA conjugates listed in table 6D with the comparative example siRNA conjugates in human plasma in vitro. The results are expressed as the Ratio (RL) of the longest fragment remaining after incubation of the example siRNA conjugate and the comparative siRNA conjugate with human plasma to the longest fragment of untreated siRNA.
Table 9D plasma stability quantification results of siRNA conjugates
As can be seen from the results of table 9D, the siRNA conjugates of the present disclosure have excellent stability in plasma with little degradation within 48 hours.
Experimental example 5-3 this experiment demonstrates the inhibition efficiency of the siRNA conjugates of Table 6D on the expression level of APOC3mRNA in vivo (in vivo)
In this experimental example, the inhibition of the expression level of APOC3 in liver tissue by siRNA conjugates of conjugates 147, 148, 150, 162-163 and control conjugate 5 in human APOC3 transgenic mice (B6; CBA-Tg (APOC 3) 3707Bres/J, purchased from Jackson Lab) was examined.
6-8 week old human APOC3 transgenic mice were randomly grouped, 6 mice per group (male and female halves), and each group of mice was given siRNA conjugates of conjugates 147, 148, 150, 162-163 and control conjugate 5, respectively, along with PBS. All animals were dosed on a weight basis in a single dose by subcutaneous injection, with an siRNA conjugate dose (based on the amount of siRNA) of 3mg/kg and a dosing volume of 10mL/kg. Mice were sacrificed 28 days after dosing, livers were collected and saved with RNAlater (Sigma Aldrich company); liver tissue was homogenized using a tissue homogenizer and total RNA was extracted using Trizol (Thermo Fisher Co.) according to standard procedures for total RNA extraction.
Detecting the expression level of APOC3mRNA in liver tissue using real-time fluorescent quantitative PCR, specifically: using ImProm-II TM The total RNA extracted was reverse transcribed into cDNA using a reverse transcription kit (Promega) according to the instructions thereof, and the inhibition efficiency of siRNA on expression of APOC3mRNA in liver tissue was then examined using a fluorescent quantitative PCR kit (Beijing Ka, century Biotech Co., ltd.). In the fluorescent quantitative PCR method, the β -actin (β -actin) gene is used as an internal reference gene, and the APOC3 and β -actin are detected using a primer for APOC3 and a primer for β -actin, respectively.
The sequence of the detection primer is shown in Table 10D.
TABLE 10 sequence of detection primers
The inhibition rate of the expression level of the APOC3mRNA by the siRNA is calculated according to the following equation: inhibition ratio = [1- (expression amount of test set APOC3 mRNA/expression amount of test set β -action mRNA)/(expression amount of control set APOC3 mRNA/expression amount of control set β -action mRNA) ]x100%. Wherein, the control group is a control group mouse to which PBS is applied in the experiment, and each test group is a dosing group mouse to which different siRNA conjugates are respectively applied. The results are shown in table 11D below.
Table 11D inhibition of APOC3mRNA expression by siRNA conjugates in mouse liver
As can be seen from the results of table 11D, on the one hand, the conjugates 147, 148, 150 and 162-163 of the present disclosure all showed excellent inhibitory activity of APOC3mRNA compared to PBS; on the other hand, in the case of identical nucleic acid sequences and identical modification schemes, the siRNA conjugates shown by conjugates 150 and 162-163 showed higher mRNA inhibition rate in vivo experiments, as compared to the comparative conjugate 5, which was different in the conjugate group, which also indicates that the siRNA conjugates of the present disclosure have good in vivo delivery efficiency.
Experimental examples 5-4 this experiment demonstrates the effect of siRNA conjugates of conjugates 150 on blood lipid levels in vivo (in vivo)
In this experimental example, the effect of siRNA conjugate (B3-siAP 1M2 SP) of conjugate 150 on total Cholesterol (CHO) and Triglyceride (TG) levels in serum in human APOC3 transgenic mice (B6; CBA-Tg (APOC 3) 3707Bres/J, purchased from Jackson Lab) was examined.
The 6-8 week old human APOC3 transgenic mice were randomly divided into 3 groups of 6 mice (male and female halves) each, as follows: (1) PBS control group; (2) conjugate 150 3mg/kg group; (3) conjugate 150 1mg/kg group. All animals were dosed on a weight basis in a single dose by subcutaneous injection with a volume of 10mL/kg of siRNA conjugate.
Orbital blood collection (about 100 μl) was performed 1 day (noted as-1 day) before administration, and 7, 14, 21, 28, 35, 42, 49, 65 days after administration, and serum was obtained by centrifugation, and further the total Cholesterol (CHO) and Triglyceride (TG) contents in serum were measured using a PM1P000/3 fully automatic serum biochemistry meter (SABA, italy), and the blood lipid results were normalized, and the inhibition rate of blood lipid levels was calculated according to the following equation: inhibition = (1-post-dose test group blood lipid content/pre-dose test group blood lipid content) ×100%. Blood lipid refers to total cholesterol or triglycerides. The detection results are shown in table 12D below.
Table 12D effect of siRNA conjugates on total cholesterol and triglyceride expression levels in mouse serum
As can be seen from table 12D, the siRNA conjugate shown by conjugate 150 has a significant down-regulating effect on the total cholesterol and triglyceride content in the serum of mice, and still shows a high blood lipid lowering effect at least at 65 days.
The specific embodiments of the present disclosure have been described in detail above, but the present disclosure is not limited to the specific details of the above embodiments, and various simple modifications may be made to the technical solutions of the present disclosure within the scope of the technical concept of the present disclosure, and all the simple modifications belong to the protection scope of the present disclosure.
In addition, the specific features described in the foregoing embodiments may be combined in any suitable manner, and in order to avoid unnecessary repetition, the present disclosure does not further describe various possible combinations.
Moreover, any combination between the various embodiments of the present disclosure is possible as long as it does not depart from the spirit of the present disclosure, which should also be construed as the disclosure of the present disclosure.

Claims (19)

1. A compound having a structure represented by formula (101):
wherein:
n 1 and each n 2 Are all 2;
m 1 is selected from 2-an integer of 4;
R 1 has a structure represented by the formula (B9) or (B11):
wherein q 1 Is an integer of 1-4, X is O or NH, M + Is a cation, wherein the cation is one of alkali metal cation, ammonium cation, tertiary amine cation and quaternary ammonium cation, SPS represents a solid phase carrier, R k Is a hydroxyl protecting group, and is a hydroxyl protecting group,represents the site where the groups are linked by covalent bonds;
each R 2 Each independently selected from H, methyl or ethyl;
each L 1 Independently 4-15 atoms in length, and each L 1 Independently is a linked combination of at least 2 of A1, A8, a 10:
Wherein each j1 is independently an integer from 3 to 5;
represents the site where the groups are linked by covalent bonds;
each S 1 Independently M 1 Groups formed by substituting all hydroxyl groups with hydroxyl protecting groups;
each M 1 All are N-acetyl galactosamine.
2. The compound of claim 1, wherein each S 1 Is a group of formula A50:
each Y is methyl.
3. The compound according to claim 1, wherein the compound has a structure represented by formula (301), (302), (303), (304), (305), (501), (502), (503), (504) or (505):
wherein X is O or NH, R k One selected from trityl, 4-methoxytrityl, 4 '-dimethoxytrityl and 4,4' -trimethoxybenzyl, SPS is resin.
4. A conjugate having a structure represented by formula (201):
wherein:
n 1 and each n 2 Is 2;
m 1 is an integer selected from 2-4;
each R 2 Each independently is H, methyl or ethyl;
R 6 is a group of formula A59:
wherein E is 1 Is OH, SH or BH 2 Nu is a functional oligonucleotide;
R 5 and R is R 6 In (2) form a phosphate bond with the P atom, and R 5 Is a group of formula (B5):
wherein,represents sites linked by covalent bonds;
Each L 1 Independently 4-15 atoms in length, and each L 1 Independently is a linked combination of at least 2 of A1, A8, a 10:
wherein each j1 is independently an integer from 3 to 5;
represents the site where the groups are linked by covalent bonds;
each M 1 Is N-acetyl galactosamine;
the functional oligonucleotide is a double-stranded oligonucleotide comprising a sense strand and an antisense strand; the double-stranded oligonucleotide is an siRNA, each nucleotide in the siRNA is independently a modified or unmodified nucleotide, the sense strand comprises a nucleotide sequence 1, the antisense strand comprises a nucleotide sequence 2, the nucleotide sequence 1 and the nucleotide sequence 2 are each 19 nucleotides in length and are at least partially reverse-complementary to form a double-stranded region, the nucleotide sequence 2 is at least partially complementary to a first stretch of nucleotide sequence, the first stretch of nucleotide sequence is a stretch of nucleotide sequence in a target mRNA, the target mRNA refers to an mRNA corresponding to a gene that is abnormally expressed in a hepatocyte; the nucleotide sequence 1 is equal in length to the first stretch of nucleotide sequences and does not differ by more than 3 nucleotides; the nucleotide sequence 2 is equal to the nucleotide sequence B in length and does not differ by more than 3 nucleotides; the nucleotide sequence B is a nucleotide sequence which is equal to the first segment of nucleotide in length and is completely complementary in reverse direction.
5. The conjugate according to claim 4, wherein the conjugate has a structure represented by formula (401), (402), (403), (404) or (405):
where Nu is a functional oligonucleotide.
6. The conjugate of claim 4, wherein P in the formula a59 is linked to the 3 'position of the 3' terminal nucleotide of the sense strand in the oligonucleotide by formation of a phosphodiester bond.
7. The conjugate of claim 6, wherein the target mRNA is selected from the group consisting of mRNA of hepatitis b virus, mRNA expressed by an angiopoietin-like protein 3 gene, and mRNA expressed by an apolipoprotein C3 gene.
8. The conjugate of claim 6, wherein the nucleotide sequence 1 differs from the first stretch of nucleotide sequences by no more than 1 nucleotide, and/or the nucleotide sequence 2 differs from the nucleotide sequence B by no more than 1 nucleotide.
9. The conjugate of claim 6, wherein the siRNA further comprises a nucleotide sequence 5, the nucleotide sequence 5 being 2 nucleotides in length, attached to the 3 'end of the antisense strand, thereby constituting a 3' overhang of the antisense strand; and, according to the direction from the 5 '-end to the 3' -end, the nucleotide sequence 5 is a continuous 2-deoxythymidine nucleotide, a continuous 2-uracil nucleotide, or is complementary to a third nucleotide sequence which is adjacent to the first nucleotide sequence in the target mRNA and has the same length as the nucleotide sequence 5.
10. The conjugate of claim 9, wherein at least one nucleotide in the sense strand or the antisense strand is a modified nucleotide and/or at least one phosphate group is a phosphate group having a modification group;
the phosphate group having a modifying group has a phosphorothioate group having a structure represented by the formula (801):
11. the conjugate of claim 10, wherein the sense strand and the antisense strand each comprise a fluoro-modified nucleotide and a non-fluoro-modified nucleotide, the fluoro-modified nucleotides being located in nucleotide sequence 1 and nucleotide sequence 2, the nucleotides at positions 7, 8, 9 of the nucleotide sequence 1 being fluoro-modified nucleotides in the sense strand in a 5 'to 3' end direction, the nucleotides at the remaining positions in the sense strand being non-fluoro-modified nucleotides; in the antisense strand, the nucleotides at positions 2, 6, 14 and 16 of the nucleotide sequence 2 are fluoro-modified nucleotides, and the nucleotides at the rest positions in the sense strand are non-fluoro-modified nucleotides, wherein the non-fluoro-modified nucleotides are selected from one of 2' -alkoxy-modified nucleotides, 2' -substituted alkoxy-modified nucleotides and 2' -deoxynucleotides.
12. The conjugate of claim 11, wherein each non-fluoro modified nucleotide is a methoxy modified nucleotide, the methoxy modified nucleotide being a nucleotide in which the 2' -hydroxy group of the ribosyl group in the nucleotide is replaced with a methoxy group.
13. The conjugate of claim 10, wherein,
the phosphorothioate linkage is present at least one of:
the 5' terminal end of the sense strand is between nucleotide 1 and nucleotide 2;
the 5' terminal end of the sense strand is between nucleotide 2 and nucleotide 3;
the 3' -terminal end of the sense strand is between nucleotide 1 and nucleotide 2;
the 3' -terminal end of the sense strand is between nucleotide 2 and nucleotide 3;
the 5' terminal end of the antisense strand is between nucleotide 1 and nucleotide 2;
the 5' terminal end of the antisense strand is between nucleotide 2 and nucleotide 3;
the 3' -terminal end of the antisense strand is between nucleotide 1 and nucleotide 2; and
the 3' -terminal end of the antisense strand is between nucleotide 2 and nucleotide 3.
14. The conjugate of claim 10, wherein the 5' -terminal nucleotide of the antisense strand is a 5' -phosphonucleotide or a 5' -phosphoanalogue modified nucleotide, the 5' -phosphonucleotide or 5' -phosphoanalogue modified nucleotide being a nucleotide having a formula represented by one of the following formulas (802) - (806):
Wherein R represents a group selected from the group consisting of H, OH, F and methoxy, and Base represents a Base selected from A, U, C, G or T.
15. The conjugate of claim 14, wherein the 5 '-phosphonucleotide or 5' -phosphoanalogue modified nucleotide is a nucleotide represented by formula (802), formula (803), or formula (805).
16. Use of a conjugate according to any one of claims 4 to 15 in the manufacture of a medicament for the treatment of a pathological condition or disease caused by expression of a specific gene in hepatocytes; the specific gene is selected from hepatitis B virus gene, angiopoietin-like protein 3 gene or apolipoprotein C3 gene.
17. The use according to claim 16, wherein the specific gene is selected from the group consisting of hepatitis b virus genes and the disease is selected from the group consisting of chronic liver disease, hepatitis, liver fibrosis disease and liver hyperplasia disease.
18. The use according to claim 16, wherein the specific gene is selected from the group consisting of angiopoietin-like protein 3 gene or apolipoprotein C3 gene, the disease is dyslipidemia, and the dyslipidemia is hypercholesterolemia, hypertriglyceridemia or atherosclerosis.
19. A kit comprising the conjugate of any one of claims 4-15.
CN201811637314.7A 2018-12-29 2018-12-29 Compounds and conjugates, methods of making and uses thereof Active CN111377984B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811637314.7A CN111377984B (en) 2018-12-29 2018-12-29 Compounds and conjugates, methods of making and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811637314.7A CN111377984B (en) 2018-12-29 2018-12-29 Compounds and conjugates, methods of making and uses thereof

Publications (2)

Publication Number Publication Date
CN111377984A CN111377984A (en) 2020-07-07
CN111377984B true CN111377984B (en) 2024-03-05

Family

ID=71222443

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811637314.7A Active CN111377984B (en) 2018-12-29 2018-12-29 Compounds and conjugates, methods of making and uses thereof

Country Status (1)

Country Link
CN (1) CN111377984B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116333013A (en) * 2020-09-30 2023-06-27 纳肽得(青岛)生物医药有限公司 Targeting ligand
CN112142761B (en) * 2020-11-08 2022-03-22 江西师范大学 Synthesis method of tetrahydropyrano [3, 2-d ] oxazole ring compound

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015006740A2 (en) * 2013-07-11 2015-01-15 Alnylam Pharmaceuticals, Inc. Oligonucleotide-ligand conjugates and process for their preparation

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015006740A2 (en) * 2013-07-11 2015-01-15 Alnylam Pharmaceuticals, Inc. Oligonucleotide-ligand conjugates and process for their preparation

Also Published As

Publication number Publication date
CN111377984A (en) 2020-07-07

Similar Documents

Publication Publication Date Title
CN110997917B (en) Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
JP7436030B2 (en) Complexes and their preparation and use
CN110945130B (en) Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
CN110997919B (en) Double-stranded oligonucleotide, composition containing double-stranded oligonucleotide, conjugate, preparation method and application
CN111377985B (en) Compounds and conjugates, methods of making and uses thereof
CN110945132B (en) Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
WO2020135673A1 (en) Nucleic acid, composition and conjugate containing nucleic acid, preparation method therefor and use thereof
WO2019105435A1 (en) Nucleic acid, composition and conjugate containing nucleic acid, preparation method and use
CN111973618B (en) Nucleic acid, pharmaceutical composition and siRNA conjugate, and preparation method and application thereof
CN111973619B (en) Nucleic acid, pharmaceutical composition containing nucleic acid, siRNA conjugate, preparation method and application
CN113227376B (en) Nucleic acid, pharmaceutical composition and conjugate, and preparation method and application thereof
WO2020238766A1 (en) Nucleic acid, pharmaceutical composition, conjugate, preparation method, and use
CN113795280B (en) Nucleic acid, pharmaceutical composition and conjugate, and preparation method and application thereof
CN111973617A (en) Nucleic acid, pharmaceutical composition and conjugate, preparation method and application
WO2020238758A1 (en) Nucleic acid, pharmaceutical composition and conjugate, preparation method therefor and use thereof
CN111979237A (en) Nucleic acid, pharmaceutical composition containing nucleic acid, siRNA conjugate, preparation method and application
CN112390835A (en) Liver targeting compounds and conjugates
CN113330117A (en) Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
CN111377984B (en) Compounds and conjugates, methods of making and uses thereof
CN113891892B (en) Compounds and drug conjugates, methods of preparation and uses thereof
WO2020233651A1 (en) Nucleic acid, pharmaceutical composition, conjugate, preparation method, and use
CN112876534B (en) Liver targeting compounds and conjugates
CN113811613B (en) Nucleic acid, pharmaceutical composition and conjugate, and preparation method and application thereof
TW202409279A (en) Nucleic acids, pharmaceutical compositions and conjugates containing the same, as well as preparation methods, uses and kits

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
CB02 Change of applicant information

Address after: 215300, No. 168, Feng Feng Road, Yushan Town, Kunshan, Jiangsu, Suzhou

Applicant after: Suzhou Ruibo Biotechnology Co.,Ltd.

Address before: 215300, No. 168, Feng Feng Road, Yushan Town, Kunshan, Jiangsu, Suzhou

Applicant before: SUZHOU RIBO LIFE SCIENCE Co.,Ltd.

CB02 Change of applicant information
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant