CN116615472A - Polymer conjugated lipid compounds and lipid nanoparticle compositions - Google Patents

Polymer conjugated lipid compounds and lipid nanoparticle compositions Download PDF

Info

Publication number
CN116615472A
CN116615472A CN202280007596.0A CN202280007596A CN116615472A CN 116615472 A CN116615472 A CN 116615472A CN 202280007596 A CN202280007596 A CN 202280007596A CN 116615472 A CN116615472 A CN 116615472A
Authority
CN
China
Prior art keywords
independently
compound
alkyl
moiety
integer
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.)
Pending
Application number
CN202280007596.0A
Other languages
Chinese (zh)
Inventor
英博
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 Aibo Biotechnology Co ltd
Original Assignee
Suzhou Aibo 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 Aibo Biotechnology Co ltd filed Critical Suzhou Aibo Biotechnology Co ltd
Publication of CN116615472A publication Critical patent/CN116615472A/en
Pending legal-status Critical Current

Links

Classifications

    • 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
    • A61K47/56Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/58Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. poly[meth]acrylate, polyacrylamide, polystyrene, polyvinylpyrrolidone, polyvinylalcohol or polystyrene sulfonic acid resin
    • A61K47/585Ion exchange resins, e.g. polystyrene sulfonic acid resin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • A61K48/0025Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid
    • A61K48/0041Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid the non-active part being polymeric
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08FMACROMOLECULAR COMPOUNDS OBTAINED BY REACTIONS ONLY INVOLVING CARBON-TO-CARBON UNSATURATED BONDS
    • C08F120/00Homopolymers of compounds having one or more unsaturated aliphatic radicals, each having only one carbon-to-carbon double bond, and only one being terminated by only one carboxyl radical or a salt, anhydride, ester, amide, imide or nitrile thereof
    • C08F120/02Monocarboxylic acids having less than ten carbon atoms; Derivatives thereof
    • C08F120/10Esters
    • C08F120/34Esters containing nitrogen, e.g. N,N-dimethylaminoethyl (meth)acrylate
    • C08F120/36Esters containing nitrogen, e.g. N,N-dimethylaminoethyl (meth)acrylate containing oxygen in addition to the carboxy oxygen, e.g. 2-N-morpholinoethyl (meth)acrylate or 2-isocyanatoethyl (meth)acrylate
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08FMACROMOLECULAR COMPOUNDS OBTAINED BY REACTIONS ONLY INVOLVING CARBON-TO-CARBON UNSATURATED BONDS
    • C08F120/00Homopolymers of compounds having one or more unsaturated aliphatic radicals, each having only one carbon-to-carbon double bond, and only one being terminated by only one carboxyl radical or a salt, anhydride, ester, amide, imide or nitrile thereof
    • C08F120/02Monocarboxylic acids having less than ten carbon atoms; Derivatives thereof
    • C08F120/10Esters
    • C08F120/38Esters containing sulfur
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08FMACROMOLECULAR COMPOUNDS OBTAINED BY REACTIONS ONLY INVOLVING CARBON-TO-CARBON UNSATURATED BONDS
    • C08F130/00Homopolymers of compounds having one or more unsaturated aliphatic radicals, each having only one carbon-to-carbon double bond, and containing phosphorus, selenium, tellurium or a metal
    • C08F130/02Homopolymers of compounds having one or more unsaturated aliphatic radicals, each having only one carbon-to-carbon double bond, and containing phosphorus, selenium, tellurium or a metal containing phosphorus
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08FMACROMOLECULAR COMPOUNDS OBTAINED BY REACTIONS ONLY INVOLVING CARBON-TO-CARBON UNSATURATED BONDS
    • C08F220/00Copolymers of compounds having one or more unsaturated aliphatic radicals, each having only one carbon-to-carbon double bond, and only one being terminated by only one carboxyl radical or a salt, anhydride ester, amide, imide or nitrile thereof
    • C08F220/02Monocarboxylic acids having less than ten carbon atoms; Derivatives thereof
    • C08F220/10Esters
    • C08F220/34Esters containing nitrogen, e.g. N,N-dimethylaminoethyl (meth)acrylate
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08FMACROMOLECULAR COMPOUNDS OBTAINED BY REACTIONS ONLY INVOLVING CARBON-TO-CARBON UNSATURATED BONDS
    • C08F2438/00Living radical polymerisation
    • C08F2438/01Atom Transfer Radical Polymerization [ATRP] or reverse ATRP

Abstract

This document relates to polymer conjugated lipid compounds and lipid nanoparticle compositions. In particular, to polymer conjugated lipid compounds that can be used in combination with other lipid components (e.g., cationic lipids, neutral lipids, and cholesterol) to form lipid nanoparticles to deliver therapeutic agents (e.g., nucleic acid molecules) for therapeutic or prophylactic purposes (including vaccination). Also provided herein are lipid nanoparticle compositions comprising the polymer conjugated lipid compounds.

Description

Polymer conjugated lipid compounds and lipid nanoparticle compositions
1. Cross-reference to related applications
The present application claims priority from chinese patent application No. 202110051372.7 filed on 1 month 14 of 2021 and U.S. provisional application No. 63/140,685 filed on 22 of 2021, 1, the entire contents of which are incorporated herein by reference.
2. Sequence listing
This specification is presented with a Computer Readable Form (CRF) copy of the sequence listing. CRF is named 14639-005-228_seqlisting_st25.txt, created at 2022, 1/3 and of size 736 bytes, and is incorporated herein by reference in its entirety.
3. Technical field
The present invention relates generally to a polymer conjugated lipid compound that can be used in combination with other lipid components, such as cationic lipids, neutral lipids, and cholesterol, to form lipid nanoparticles for intracellular and extracellular use in delivering therapeutic agents, such as nucleic acid molecules including nucleic acid mimics of Lock (LNA), peptide Nucleic Acid (PNA), and morpholino oligonucleotides, for therapeutic or prophylactic purposes, including vaccination.
4. Background art
Therapeutic nucleic acids have the potential to radically alter vaccination, gene therapy, protein replacement therapy and other genetic disease therapies. Since the first clinical study of therapeutic nucleic acids, beginning in the 2000 s, significant progress has been made through the design of nucleic acid molecules and improvements in their delivery methods. However, nucleic acid therapeutics still face several challenges, including low cell permeability and high sensitivity to degradation by certain nucleic acid molecules (including RNA). Thus, there is a need to develop new nucleic acid molecules and related methods and compositions to facilitate their extracellular or intracellular delivery for therapeutic and/or prophylactic purposes.
One such method involves the use of a lipid nanoparticle composition. The lipid nanoparticle may be formed from lipids formulated with other lipid components, such as neutral lipids, cholesterol, polymer conjugated lipids. One of the most commonly used polymer conjugated lipids is a pegylated lipid. Several carboxylate-based zwitterionic polymer conjugated lipids have also been reported. See, e.g., murou et al, journal of Colloid and Interface Science 2013,390,47-53; li et al, theranostics 2015,5 (5), 583-596; and Cao et al, langmuir 2012,28,11625-11632. There remains a need for systematic studies of zwitterionic polymer conjugated lipids, including, for example, molecular weight effects, structural effects, and end groups. There is also a need for alternative or improved polymer conjugated lipids and lipid nanoparticles for delivery of therapeutic nucleic acids.
5. Summary of the invention
In one embodiment, provided herein are polymer conjugated lipid compounds, including pharmaceutically acceptable salts or stereoisomers thereof, that can be used alone or in combination with other lipid components such as cationic lipids, neutral lipids, charged lipids, steroids (including, for example, all sterols) and/or their analogs, and/or polymers to form lipid nanoparticles for the delivery of therapeutic agents (e.g., nucleic acid molecules including nucleic acid mimics such as Locked Nucleic Acids (LNAs), peptide Nucleic Acids (PNAs), and morpholino cyclic oligonucleotides). In some cases, the lipid nanoparticle is used to deliver nucleic acids, such as antisense and/or messenger RNAs. It also provides methods of using such lipid nanoparticles to treat various diseases or conditions, such as diseases or conditions caused by infectious entities and/or protein deficiencies.
In one embodiment, the polymer conjugated lipid compounds provided herein are derived from monomers comprising a zwitterionic moiety.
In one embodiment, provided herein are compounds represented by formula (I) or (II):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein L, X, X 1 、X 2 、R 3 、R 4 、Y 1 、G 4 、G 5 、Z 1 、Z 2 "ran", n and T are as defined herein or elsewhere.
In one embodiment, the present invention provides nanoparticle compositions comprising a compound provided herein and a therapeutic or prophylactic agent. In one embodiment, the therapeutic or prophylactic agent comprises at least one mRNA encoding an antigen or fragment or epitope thereof.
Additional features of the present disclosure will become apparent to those skilled in the art upon consideration of the following detailed description of specific embodiments.
6. Description of the drawings
Figure 1 shows the effect of selected polymer compounds on hEPO expression levels in animal studies. Figure 2 shows the effect of selected polymer compounds on hEPO expression levels in animal studies. Figure 3 shows Cryo-EM imaging of lipid nanoparticles comprising selected polymer compounds.
7. Detailed description of the invention
7.1 general technique
The techniques and methods described or referenced in this disclosure include conventional methods commonly understood or commonly used by those skilled in the art, such as Molecular Cloning: A Laboratory Manual (3 rd edition 2001); current Protocols in Molecular Biology (Ausubel et al, edit 2003) and the like.
7.2 terminology
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. For the purposes of explaining the present specification, the following description of terms will be used, and terms used in the singular form will also include the plural and vice versa, as appropriate. The disclosures of all patents and other publications cited herein are incorporated by reference in their entirety. To the extent that any description of a term herein conflicts with any document incorporated by reference, the description and illustration of the following term shall govern.
Unless otherwise indicated herein, the term "lipid" refers to a group of organic compounds that include, but are not limited to, esters of fatty acids and are characterized by generally poor solubility in water, but are soluble in many non-polar organics. Although lipids generally have poor solubility in water, certain classes of lipids (e.g., lipids modified with polar groups such as DMG-PEG 2000) have limited water solubility and can be dissolved in water under certain conditions. Known types of lipids include biomolecules such as fatty acids, waxes, sterols, fat-soluble vitamins, monoglycerides, diglycerides, triglycerides and phospholipids. Lipids can generally be divided into at least three classes: (1) "simple lipids", including fats and oils and waxes; (2) "Compound lipids" including phospholipids and glycolipids (such as DMPE-PEG 2000); and (3) "derived lipids", such as steroids and the like. Furthermore, as used herein, lipids also include lipid compounds. The term "lipid compound" also simply referred to as "lipid" refers to a lipid compound such as an amphiphilic compound having a physical property of a lipid.
The term "lipid nanoparticle" or "LNP" refers to particles having a nanometer scale (nm) (e.g., 1nm to 1,000 nm) that comprise one or more types of lipid molecules. The LNPs provided herein can further comprise at least one non-lipid payload molecule (e.g., one or more nucleic acid molecules). In some embodiments, the LNP comprises a non-lipid payload molecule partially or fully encapsulated inside a lipid shell. In particular, in some embodiments, wherein the payload is a negatively charged molecule (e.g., mRNA encoding a viral protein), and the lipid component of the LNP comprises at least one cationic lipid. It is contemplated that cationic lipids can interact with negatively charged payload molecules and facilitate payload incorporation and/or encapsulation into the LNP during LNP formation. As provided herein, other lipids that may form part of the LNP include, but are not limited to, neutral lipids and charged lipids, such as steroids, polymer conjugated lipids, and various zwitterionic lipids. In certain embodiments, LNPs according to the invention comprise one or more polymer conjugated lipids of formula (I) or (II) (and subformulae thereof) described herein.
The term "cationic lipid" refers to a lipid that is positively charged at any pH or hydrogen ion activity of the environment in which it is located, or that is capable of being positively charged in response to the pH or hydrogen ion activity of the environment in which it is located (e.g., the environment in which it is intended to be used). Thus, the term "cation" encompasses the scope of "permanent cations" and "cationizable". In certain embodiments, the positive charge in the cationic lipid results from the presence of a quaternary nitrogen atom. In certain embodiments, the cationic lipid comprises a zwitterionic lipid that is positively charged in the environment in which it is intended to be administered (e.g., at physiological pH).
The term "polymer conjugated lipid" or "polymer conjugated lipid" refers to a molecule comprising both a lipid moiety and a polymer moiety. An example of a polymer conjugated lipid is a pegylated lipid (PEG-lipid), wherein the polymer moiety comprises polyethylene glycol. In certain embodiments, LNPs in accordance with the present disclosure comprise one or more polymer conjugated lipids of formula (I) or (II) (and subformulae thereof) as described herein.
The term "neutral lipid" encompasses any lipid molecule that exists in an uncharged form or a neutral zwitterionic form at a selected pH. In some embodiments, the selected useful pH value or range corresponds to the pH condition of the environment in which the lipid is intended to be used, e.g., physiological pH. As non-limiting examples, neutral lipids that may be used in conjunction with the disclosure herein include, but are not limited to, phosphatidylcholine, such as 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1, 2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1, 2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1, 2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), phosphatidylethanolamine such as 1, 2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 2- (((2, 3-bis (oleoyloxy) propyl)) dimethylammonium phosphate) ethyl hydrogen (DOCP), sphingomyelin (SM), ceramides, steroids such as sterols, and derivatives thereof. Neutral lipids may be synthetic or derived (isolated or modified) from natural sources or compounds.
The term "charged lipid" encompasses any lipid molecule that exists in a positively or negatively charged form at a selected pH or range. In some embodiments, the selected pH value or range corresponds to the pH condition of the intended use environment of the lipid, e.g., physiological pH. As non-limiting examples, charged lipids that may be used in conjunction with the disclosure herein include, but are not limited to, phosphatidylserine, phosphatidic acid, phosphatidylglycerol, phosphatidylinositol, sterol hemisuccinate, dialkyltrimethylammonium-propane (e.g., DOTAP, DOTMA), dialkyldimethylaminopropane, ethylphosphocholine, dimethylaminoethane carbamoylsterol (e.g., DC-Chol), 1, 2-dioleoyl-sn-glycerol-3-phosphate-L-serine sodium salt (DOPS-Na), 1, 2-dioleoyl-sn-glycerol-3-phosphate- (1' -rac-glycerol) sodium salt (DOPG-Na), and 1, 2-dioleoyl-sn-glycerol-3-phosphate sodium salt (DOPA-Na). Charged lipids provided herein may be synthetic or derived (isolated or modified) from natural sources or compounds.
As used herein, unless otherwise indicated, the term "alkyl" refers to a straight or branched hydrocarbon chain group consisting of only saturated carbon and hydrogen atoms. In one embodiment, the alkyl group has, for example, 1 to 24 carbon atoms (C 1 -C 24 Alkyl), 4 to 20 carbon atoms (C 4 -C 20 Alkyl), 6 to 16 carbon atoms (C 6 -C 16 Alkyl), six to nine carbon atoms (C 6 -C 9 Alkyl), one to fifteen carbon atoms (C 1 -C 15 Alkyl), one to twelve carbon atoms (C 1 -C 12 Alkyl), one to eight carbon atoms (C 1 -C 8 Alkyl) or one to six carbon atoms (C 1 -C 6 Alkyl) and is attached to the remainder of the molecule by a single bond. Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl, 1-methylethyl (isopropyl), n-butyl, n-pentyl, 1-dimethylethyl (t-butyl), 3-methylhexyl, 2-methylhexyl, and the like. Unless otherwise indicated, alkyl groups are optionally substituted.
As used herein, unless otherwise indicated, the term "alkenyl" refers to a straight or branched hydrocarbon chain group consisting of only carbon and hydrogen atoms, which contains one or more carbon-carbon double bonds. As understood by one of ordinary skill in the art, the term "alkenyl" also includes groups having "cis" and "trans" configurations, or "E" and "Z" configurations. In one embodiment, the alkenyl group has, for example, 2 to 24 carbon atoms (C 2 -C 24 Alkenyl), 4 to 20 carbon atoms (C 4 -C 20 Alkenyl), 6 to 16 carbon atoms (C 6 -C 16 Alkenyl), six to nine carbon atoms (C 6 -C 9 Alkenyl), two to fifteen carbon atoms (C 2 -C 15 Alkenyl), two to twelve carbon atoms (C 2 -C 12 Alkenyl), two to eight carbon atoms (C 2 -C 8 Alkenyl) or 2 to 6 carbon atoms (C 2 -C 6 Alkenyl) and is attached to the remainder of the molecule by a single bond. Examples of alkenyl groups include, but are not limited to, vinyl, prop-1-enyl, but-1-enyl, pent-1, 4-dienyl, and the like. Unless otherwise indicated, alkenyl groups are optionally substituted.
As used herein, unless otherwise indicated, the term "alkynyl" refers to a straight or branched hydrocarbon chain group consisting of only carbon and hydrogen atoms, which contains one or more carbon-carbon triple bonds. In one embodiment, the alkynyl group has, for example, 2 to 24 carbon atoms (C 2 -C 24 Alkynyl), 4 to 20 carbon atoms (C 4 -C 20 Alkynyl), 6 to 16 carbon atoms (C 6 -C 16 Alkynyl), six to nine carbon atoms (C 6 -C 9 Alkynyl), two to fifteen carbon atoms (C 2 -C 15 Alkynyl), two to twelve carbon atoms (C 2 -C 12 Alkynyl), two to eight carbon atoms (C 2 -C 8 Alkynyl) or two to six carbon atoms (C 2 -C 6 Alkynyl) and is attached to the remainder of the molecule by a single bond. Examples of alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, and the like. Unless otherwise indicated, alkynyl groups are optionally substituted.
As used herein, unless otherwise indicated, the term "alkylene" or "alkylene chain" refers to a straight or branched divalent hydrocarbon chain that connects the remainder of the molecule to a group consisting of only saturated carbons and hydrogens. In one embodiment, the alkylene group has, for example, 1 to 24 carbon atoms (C 1 -C 24 Alkylene), 1 to 15 carbon atoms (C 1 -C 15 Alkylene), 1 to 12 carbon atoms (C 1 -C 12 Alkylene), 1 to 8 carbon atoms (C 1 -C 8 Alkylene), 1 to 6 carbon atoms (C 1 -C 6 Alkylene), 2 to 4 carbon atoms (C 2 -C 4 Alkylene), 1 to 2 carbon atoms (C 1 -C 2 An alkylene group). Examples of alkylene groups include, but are not limited to, methylene, ethylene, propylene, n-butene, and the like. The alkylene chain is linked to the rest of the molecule by a single bond and to the radical group by a single bond. The alkylene chain may be attached to the remainder of the molecule and to the radical group by one or any two carbons in the chain. Unless otherwise indicated, the alkylene chain is optionally substituted.
As used herein, unless otherwise indicated, the term "alkenylene" refers to a straight or branched divalent hydrocarbon chain that links the remainder of the molecule to a radical consisting of only carbon and hydrogen, the radical comprising one or more carbon-carbon double bonds. In one embodiment, the alkenylene group has, for example, 2 to 24 carbon atoms (C 2 -C 24 Alkenylene), 2 to 15 carbon atoms (C 2 -C 15 Alkenylene), 2 to 12 carbon atoms (C 2 -C 12 Alkenylene), 2 to 8 carbon atoms (C 2 -C 8 Alkenylene), 2 to 6 carbon atoms (C 2 -C 6 Alkenylene) or 2 to 4 carbon atoms (C 2 -C 4 Alkenylene). Examples of alkenylene groups include, but are not limited to, ethenylene, propenylene, n-butenyl, and the like. Alkenylene is attached to the remainder of the molecule by a single or double bond and to a free radical group by a single or double bond. The alkenylene group may be attached to the remainder of the molecule and to the radical group through one or any two carbons in the chain. Unless otherwise indicated, alkenylene is optionally substituted.
As used herein, unless otherwise indicated, the term "cycloalkyl" refers to a non-aromatic monocyclic or polycyclic hydrocarbon group consisting of only carbon and hydrogen atoms and being saturated. Cycloalkyl groups may include fused or bridged ring systems. In one embodiment, cycloalkyl groups have, for example, 3 to 15 ring carbon atoms (C 3 -C 15 Cycloalkyl), 3 to 10 ring carbon atoms (C 3 -C 10 Cycloalkyl) or 3 to 8 ring carbon atoms (C 3 -C 8 Cycloalkyl). Cycloalkyl groups are linked to the rest of the molecule by single bonds. Examples of monocyclic cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Examples of polycyclic cycloalkyl groups include, but are not limited to, adamantyl, norbornyl, decahydroalkyl, 7-dimethyl-bicyclo [2.2.1]Heptyl, and the like. Unless otherwise indicated, cycloalkyl groups are optionally substituted.
As described herein, unless otherwise indicated, the term "cycloalkylene" is a divalent cycloalkyl group. Unless otherwise indicated, cycloalkylene groups are optionally substituted.
As used herein, unless otherwise indicated, the term "cycloalkenyl" refers to a non-aromatic monocyclic or polycyclic hydrocarbon group consisting of only carbon and hydrogen atoms and including one or more carbon-carbon double bonds. Cycloalkenyl groups may include fused or bridged ring systems. In one embodiment, cycloalkenyl groups have, for example, 3 to 15 ring carbon atoms (C 3 -C 15 Cycloalkenyl), 3 to 10 ring carbon atoms (C 3 -C 10 Cycloalkenyl) or 3 to 8 ring carbon atoms (C 3 -C 8 Cycloalkenyl group). The cycloalkenyl group is linked to the rest of the molecule by a single bond. Monocyclic cycloalkenyl groupsExamples include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, and the like. Unless otherwise indicated, cycloalkenyl groups are optionally substituted.
As described herein, the term "cycloalkenyl" is a divalent cycloalkenyl group unless otherwise indicated. Unless otherwise indicated, cycloalkenyl groups are optionally substituted.
As described herein, unless otherwise indicated, the term "heterocyclyl" refers to a monocyclic or polycyclic moiety comprising one or more (e.g., one or two, one to three, or one to four) non-aromatic groups independently selected from nitrogen, oxygen, phosphorus, and sulfur heteroatoms. The heterocyclyl may be attached to the main structure at any heteroatom or carbon atom. The heterocyclyl may be a monocyclic, bicyclic, tricyclic, tetracyclic or other polycyclic ring system, wherein the polycyclic ring system may be a fused, bridged or spiro ring system. Heterocyclic polycyclic ring systems may include one or more heteroatoms in one or more rings. The heterocyclyl groups may be saturated or partially unsaturated. Saturated heterocycloalkyl groups may be referred to as "heterocycloalkyl groups". If the heterocyclyl contains at least one double bond, then the partially unsaturated heterocycloalkyl group may be referred to as "heterocycloalkenyl"; if the heterocyclyl contains at least one triple bond, it may be referred to as "heterocyclylalkynyl". In one embodiment, the heterocyclyl has, for example, 3 to 18 ring atoms (3 to 18 membered heterocyclyl), 4 to 18 ring atoms (4 to 18 membered heterocyclyl), 5 to 18 ring atoms (5 to 18 membered heterocyclyl), 4 to 8 ring atoms (4 to 8 membered heterocyclyl) or 5 to 8 ring atoms (5 to 8 membered heterocyclyl). In this context, whenever a numerical range such as "3 to 18" is present, it refers to each integer within the given range. For example, a "3-to 18-membered heterocyclic group" means that the heterocyclic group may be composed of 3 ring atoms, 4 ring atoms, 5 ring atoms, 6 ring atoms, 7 ring atoms, 8 ring atoms, 9 ring atoms, 10 ring atoms, up to 18 ring atoms, and the like. Examples of heterocyclyl groups include, but are not limited to, imidazolyl, imidazolidinyl, oxazolyl, oxazolidinyl, thiazolyl, thiazolidinyl, pyrazolidinyl, pyrazolyl, isoxazolidinyl, isothiazolidinyl, isothiazolyl, morpholinyl, pyrrolyl, pyrrolidinyl, furyl, tetrahydrofuryl, thienyl, pyridyl, piperidyl, quinolinyl, and isoquinolinyl. Unless otherwise indicated, heterocyclyl groups are optionally substituted.
As described herein, unless otherwise indicated, the term "heterocyclyl" is a divalent heterocyclyl. Unless otherwise indicated, the heterocyclylene group is optionally substituted.
As described herein, unless otherwise indicated, the term "aryl" refers to a monocyclic aromatic group and/or a polycyclic monovalent aromatic group comprising at least one aromatic hydrocarbon ring. In certain embodiments, aryl groups have 6 to 18 ring carbon atoms (C 6 -C 18 Aryl), 6 to 14 ring carbon atoms (C 6 -C 14 Aryl) or 6 to 10 ring carbon atoms (C 6 -C 10 Aryl). Examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, azulenyl (azulenyl), anthracenyl, phenanthryl, pyrenyl (pyrenyl), biphenyl, and terphenyl. The term "aryl" also refers to bicyclic, tricyclic, or other polycyclic hydrocarbon rings in which at least one ring is aromatic, and the other rings may be saturated, partially unsaturated, or aromatic, such as dihydronaphthyl, indenyl, indanyl, or tetrahydronaphthyl (tetrahydronaphthyl). Unless otherwise indicated, aryl groups are optionally substituted.
As used herein, the term "arylene" is a divalent aryl group unless otherwise indicated. Unless otherwise indicated, arylene groups are optionally substituted.
As used herein, unless otherwise indicated, the term "heteroaryl" refers to a monocyclic aromatic group and/or polycyclic aromatic group containing at least one aromatic ring, wherein at least one aromatic ring contains one or more heteroatoms selected independently from one to three or one to four of O, S and N. The heteroatom in the heteroaryl group may be attached to the main structure at any carbon atom. In certain embodiments, heteroaryl groups have 5 to 20, 5 to 15, or 5 to 10 ring atoms. The term "heteroaryl" also refers to bicyclic, tricyclic, or other polycyclic rings in which at least one ring is aromatic, and the other rings may be saturated, partially unsaturated, or aromatic, in which at least one aromatic ring contains one or more monocyclic heteroaryl examples including, but not limited to, pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, and triazinyl. Examples of bicyclic heteroaryl groups include, but are not limited to, indolyl, benzothiazolyl, benzoxazolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuranyl, isobenzofuranyl, chromonyl, coumarin, cinnolinyl, quinoxalinyl, indazolyl, purinyl, pyrrolopyridinyl, furanpyridinyl, thienopyridinyl, dihydroisoindolyl, and tetrahydroquinolinyl. Examples of tricyclic heteroaryl groups include, but are not limited to, carbazolyl, benzindolyl, phenanthrolinyl, acridinyl, phenanthridinyl, and xanthenyl. Unless otherwise indicated, heteroaryl groups are optionally substituted.
As described herein, unless otherwise indicated, the term "heteroarylene" is a divalent heteroaryl group. Unless otherwise indicated, heteroarylene is optionally substituted.
Where groups described herein are "substituted," they may be substituted with any suitable substituent or substituents. Illustrative examples of substituents include, but are not limited to, those set forth in the exemplary compounds and embodiments provided herein, and: halogen atoms such as F, cl, br or I; cyano group; oxo (=o); hydroxyl (-OH); an alkyl group; alkenyl alkynyl cycloalkylaryl- (c=o) OR'; -O (c=o) R'; -C (=o) R'; s (O) x R’;-S-SR’;-C(=O)SR’;-SC(=O)R’;-NR’R’;-NR’C(=O)R’;-C(=O)NR’R’;-NR’C(=O)NR’R’;-OC(=O)NR’R’;-NR’C(=O)OR’;-NR’S(O) x N R’R’;-NR’S(O) x R'; and-S (O) x NR ' R ', wherein R ' is independently H, C at each occurrence 1 -C 15 Alkyl or cycloalkyl, and x is 0, 1 or 2. In some embodiments, the substituent is C 1 -C 12 An alkyl group. In other embodiments, the substituent is cycloalkyl. In other embodiments, the substituent is a halogen group, such as fluoro.In other embodiments, the substituent is an oxo group. In other embodiments, the substituent is hydroxy. In other embodiments, the substituent is an alkoxy (-OR'). In other embodiments, the substituent is a carboxyl group. In other embodiments, the substituent is an amino group (-NR 'R').
As used herein, unless otherwise indicated, the term "optional" or "optionally" (e.g., optionally substituted) means that the subsequently described event may or may not occur, and that the description includes instances where the event or event occurs as well as instances where the event or event does not occur. For example, "optionally substituted alkyl" means that the alkyl group may or may not be substituted, and is described to include substituted alkyl groups and unsubstituted alkyl groups.
"prodrug" refers to a compound that can be converted to a biologically active compound under physiological conditions or by solvolysis. Thus, the term "prodrug" refers to a metabolic precursor of a pharmaceutically acceptable bioactive compound. Prodrugs can be inactive when administered to a subject in need thereof, but are converted in vivo to the biologically active compounds of the invention. Prodrugs are typically rapidly converted in vivo to the parent bioactive compounds of the present invention, for example, by hydrolysis in the blood. Prodrug compounds generally provide solubility, histocompatibility or delayed release advantages in mammalian organisms (see Bundgard, h., design of Prodrugs (1985), pages 7-9, 21-24 (Elsevier, amsterdam)). Discussion of prodrugs is provided in Higuchi, t et al, a.c. s. Symposium Series, volume 14, and Bioreversible Carriers in Drug Design, edward b.roche edit, american Pharmaceutical Association and Pergamon Press,1987.
In some embodiments, the term "prodrug" is also meant to include any carrier that is covalently bonded, and when such a prodrug is administered to a mammalian subject, they release the active compound of the invention in vivo. Prodrugs of the compounds of the present invention may be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either by routine manipulation or in vivo, to the parent compound of the present invention. Prodrugs include the following compounds of the invention: wherein the hydroxyl, amino or sulfhydryl group is bonded to any group that cleaves to form a free hydroxyl, free amino or free sulfhydryl group, respectively, when the prodrug of the compounds of the invention is administered to a mammalian subject.
Examples of "prodrugs" include, but are not limited to, acetate, formate, benzoate derivatives, and the like of amide derivatives of alcohol or amine functional groups in the compounds provided herein.
As used herein, unless otherwise indicated, the term "pharmaceutically acceptable salt" includes both acid addition salts and base addition salts.
Examples of "pharmaceutically acceptable acid addition salts" include, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, as well as organic acids such as, but not limited to, acetic acid, 2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclic amic acid, dodecyl sulfuric acid, ethane-1, 2-disulfonic acid, ethane sulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactonic acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxoglutaric acid, glycerophosphate, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-1, 5-dicarboxylic acid, naphthalene-2-sulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, p-toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like.
Examples of "pharmaceutically acceptable base addition salts" include, but are not limited to, salts prepared by addition of an inorganic or organic base to a free acid compound. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Preferably, the inorganic salts are ammonium, sodium, potassium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of the following primary, secondary and tertiary amines, substituted amines (including naturally occurring substituted amines), cyclic amines, and basic ion exchange resins: such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, dealcoholization, 2-dimethylaminoethanol, 2-diethylaminoethanol, lysine, arginine, histidine, caffeine, procaine, hydrazinaniline, choline, betaine, benazepine (bennethamine), benzathine (benzathine), ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, purine, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like. Preferably, the organic base is isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
The compounds provided herein may contain one or more asymmetric centers and thus may produce enantiomers, diastereomers, and other stereoisomeric forms, which may be defined as (R) -or (S) -or as (D) -or (L) -for amino acids, depending on the absolute stereochemistry. Unless otherwise indicated, the compounds provided herein are intended to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R) -and (S) -or (D) -and (L) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, e.g., chromatography and fractional crystallization. Conventional techniques for preparing/separating individual enantiomers include chiral synthesis from suitable optically pure precursors or resolution of racemates (or racemates of salts or derivatives) using, for example, chiral High Pressure Liquid Chromatography (HPLC). When a compound described herein contains an olefinic double bond or other geometric asymmetric center, unless specified otherwise, the compound is meant to include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.
As used herein, unless otherwise indicated, the term "isomer" refers to different compounds having the same formula. "stereoisomers" are isomers in which only the atoms differ in their arrangement in space. "atropisomers" are stereoisomers in which the rotation of an atom about a single bond is hindered. "enantiomers" are a pair of stereoisomers that are mirror images that do not overlap each other. Any ratio of a pair of enantiomers can be referred to as a "racemic" mixture. "diastereomers" are stereoisomers that have at least two asymmetric atoms, but are not mirror images of each other.
"stereoisomers" may also include E and Z isomers or mixtures thereof, and cis and trans isomers or mixtures thereof. In certain embodiments, the compounds described herein are isolated as the E or Z isomers. In other embodiments, the compounds described herein are mixtures of the E and Z isomers.
"tautomer" refers to the isomeric forms of the compounds that are in equilibrium with each other. The concentration of the isomeric forms will vary depending upon the environment in which the compound is located and may depend upon whether the compound is a solid or in a state present in an organic or aqueous solution.
The compounds described herein may contain an atomic isotope of an unnatural moiety on one or more atoms. For example, the compounds may be radiolabeled with a radioisotope, such as tritium 3 # 3 H) Iodine-125% 125 I) Sulfur 35% 35 S) or carbon 14% 14 C) Or may be deuterium 2 H) 13% of carbon 13 C) Or nitrogen 15% 15 N) isotopically enriched. As used herein, an "isotope" is an isotopically enriched compound. The term "isotopically enriched" refers to an atom having an isotopic composition different from the natural isotopic composition of the atom. "isotopically enriched" may also refer to a compound containing at least one atom whose isotopic composition differs from the natural isotopic composition of the atom. The term "isotopic composition" refers to the amount of each isotope present for a given atom. Radiolabeled and isotopically enriched compounds are useful as therapeutic agents, for example, cancer therapeutic agents, research reagents (e.g., binding assay reagents), and diagnostic agents (e.g., in vivo imaging agents). All isotopic variations of the compounds described herein, whether radioactive or not, are intended to be encompassed within the scope of the embodiments provided herein. In some embodiments, the method comprises Isotopes of the compounds described herein are provided, for example, isotopes that are deuterium-enriched, carbon-13 and/or nitrogen 15. As used herein, "deuterated" refers to where at least one hydrogen (H) is deuterated (in D or 2 H represents) substituted compounds, i.e., compounds enriched in deuterium at least at one position.
It should be noted that if there is a difference between the structure described herein and the name of the structure, the structure described should have a greater weight.
As used herein, unless otherwise indicated, the term "pharmaceutically acceptable carrier, diluent or excipient" includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent or emulsifying agent that has been approved by the U.S. food and drug administration for use in humans or domestic animals.
The term "composition" is intended to encompass a product comprising the specified ingredients (e.g., mRNA molecules), optionally in the specified amounts.
The terms "polynucleotide" or "nucleic acid" are used interchangeably herein to refer to a polymer of nucleotides of any length, including, for example, DNA and RNA. The nucleotide may be a deoxyribonucleotide, a ribonucleotide, a modified nucleotide or base and/or analogue thereof, or may be any substrate that is incorporated into the polymer by a DNA polymerase or RNA polymerase or by a synthetic reaction. Polynucleotides may comprise modified nucleotides, such as methylated nucleotides and analogs thereof. The nucleic acid may be in single-stranded or double-stranded form. As described herein and unless otherwise indicated, "nucleic acid" also includes nucleic acid mimics, such as Locked Nucleic Acids (LNAs), peptide Nucleic Acids (PNAs) and morpholino loop oligonucleotides. As used herein, "oligonucleotide" refers to a short synthetic polynucleotide, typically but not necessarily less than about 200 nucleotides in length. The terms "oligonucleotide" and "polynucleotide" are not mutually exclusive. The above description of polynucleotides applies equally and entirely to oligonucleotides. Unless otherwise indicated, the left end of any single stranded polynucleotide sequence disclosed herein is the 5' end; the left-hand orientation of the double-stranded polynucleotide sequence is referred to as the 5' orientation. The direction of addition of nascent RNA transcripts from 5 'to 3' is referred to as the transcription direction; the region of the sequence on the DNA strand having the same sequence as the RNA transcript that is located at the 5 'to 5' end of the RNA transcript is referred to as the "upstream sequence"; the region of the DNA strand having the sequence 3 'to 3' end of the same sequence as the RNA transcript is referred to as the "downstream sequence".
"isolated nucleic acid" refers to a nucleic acid, which may be, for example, RNA, DNA or a mixture of nucleic acids, which are substantially naturally separated from other genomic DNA sequences and proteins or complexes (e.g., ribosomes and polymerases), including native sequences. An "isolated" nucleic acid molecule is a nucleic acid molecule that is separated from other nucleic acid molecules in natural sources. Furthermore, an "isolated" nucleic acid molecule (e.g., an mRNA molecule) may be substantially free of other cellular material or culture medium when produced by recombinant techniques, or may be substantially free of chemical precursors or other chemicals when chemically synthesized. In a specific embodiment, one or more nucleic acid molecules encoding the antigens described herein are isolated or purified. The term includes nucleic acid sequences that have been removed from their naturally occurring environment, and includes recombinant or cloned DNA or RNA isolates as well as chemically synthesized analogs or analogs biosynthesized by heterologous systems. Substantially pure molecules may include isolated forms of the molecule.
The term "encoding nucleic acid" or grammatical equivalents thereof includes: (a) In a native state or by methods well known to those skilled in the art, can be transcribed to produce a nucleic acid molecule capable of translation into mRNA for the peptide and/or polypeptide, and (b) the mRNA molecule itself. The antisense strand is the complement of a nucleic acid molecule and the coding sequence can be deduced therefrom. The term "coding region" refers to that portion of a coding nucleic acid sequence that can be translated into a peptide or polypeptide. The term "untranslated region" or "UTR" refers to a portion of a coding nucleic acid that is not translated into a peptide or polypeptide. Depending on the orientation of the UTR relative to the coding region of the nucleic acid molecule, if the UTR is located at the 5 'end of the coding region, the UTR is referred to as a 5' -UTR; if located 3 'of the coding region, this UTR is referred to as the 3' -UTR.
As used herein, the term "mRNA" refers to a messenger RNA molecule comprising one or more Open Reading Frames (ORFs) that can be translated by a cell or organism to produce one or more peptide or protein products. The region comprising one or more ORFs is referred to as the coding region of an mRNA molecule. In certain embodiments, the mRNA molecule further comprises one or more untranslated regions (UTRs).
In certain embodiments, the mRNA is a monocistronic mRNA comprising only one ORF. In certain embodiments, the monocistronic mRNA encodes a peptide or protein comprising at least one epitope of a selected antigen (e.g., a pathogenic antigen or a tumor-associated antigen). In other embodiments, the mRNA is a polycistronic mRNA comprising two or more ORFs. In certain embodiments, the polycistronic mRNA encodes two or more peptides or proteins that are the same or different from each other. In certain embodiments, each peptide or protein encoded by the polycistronic mRNA comprises at least one epitope of the selected antigen. In certain embodiments, the different peptides or proteins encoded by the polycistronic mRNA each comprise at least one epitope of a different antigen. In any of the embodiments described herein, the at least one epitope may be at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 epitopes of one antigen.
The term "nucleobase" encompasses purines and pyrimidines, including the natural compounds adenine, thymine, guanine, cytosine, uracil, inosine and natural or synthetic analogs or derivatives thereof.
As used herein, the term "functional nucleotide analog" refers to a modified form of a canonical nucleotide A, G, C, U or T that (a) retains the base pairing properties of the corresponding canonical nucleotide, and (b) comprises at least one chemical modification of (i) a nucleobase, (ii) a glycosyl, (iii) a phosphate group, or (iv) any combination of (i) through (iii) of the corresponding natural nucleotide. As described herein, base pairs encompass not only standard watson-crick a-T, A-U or C-G base pairs, but also base pairs formed between a canonical nucleotide and a functional nucleotide analog or between a pair of functional nucleotide analogs, wherein the arrangement of the hydrogen bond donor and hydrogen bond acceptor allows hydrogen bonding between a modified nucleobase and a standard nucleobase or between two complementary modified nucleobase structures. For example, functional analogs of guanosine (G) retain the ability to base pair with cytosine (C) or functional analogs of cytosine. An example of such non-canonical base pairing is base pairing between the modified nucleotide inosine and adenine, cytosine or uracil. As described herein, functional nucleotide analogs can be naturally occurring or non-naturally occurring. Thus, a nucleic acid molecule comprising a functional nucleotide analog may have at least one modified nucleobase, sugar group, or internucleoside linkage. Exemplary chemical modifications to nucleobases, glycosyls, or internucleoside linkages of nucleic acid molecules are provided herein.
As used herein, the terms "translation enhancing element," "TEE," and "translation enhancer" refer to a region in a nucleic acid molecule that functions to facilitate translation of a coding sequence of a nucleic acid into a protein or peptide product, such as by cap-dependent or non-cap-dependent translation. TEE is typically located in the UTR region of a nucleic acid molecule (e.g., mRNA) and is capable of enhancing the level of translation of coding sequences located upstream or downstream. For example, a TEE in the 5' -UTR of a nucleic acid molecule may be located between the promoter and the start codon of the nucleic acid molecule. Various TEE sequences are known in the art (Wellensiek et al Genome-wide profiling of human cap-independent tr anslation-enhancing elements, nature Methods, month 8 of 2013; 10 (8): 747-750; chappell et al PNAS, 6/29/101 (26) 9590-9594). Certain TEEs are known to be conserved across species (P nek et al Nucleic Acids Res earch, volume 41, 16, 2013, 9, 1, pages 7625-7634).
As used herein, the term "stem-loop sequence" refers to a single-stranded polynucleotide sequence having at least two regions that are complementary or substantially complementary to each other when read in opposite directions to form at least one double helix and a non-complementary loop, the resulting loop structure being referred to as a stem-loop structure, hairpin or hairpin loop, which is also a secondary structure present in many RNA molecules.
As used herein, the term "peptide" refers to a polymer containing 2 to 50 amino acid residues linked by one or more covalent peptide bonds. The term applies to naturally occurring amino acid polymers, where one or more amino acid residues is a non-naturally occurring amino acid (e.g., an amino acid analog or a non-natural amino acid).
The terms "polypeptide" and "protein" are used interchangeably herein to refer to a polymer of more than fifty amino acid residues linked by covalent peptide bonds. That is, the description for polypeptides applies equally to the description for proteins and vice versa. The term applies to naturally occurring amino acid polymers, where one or more amino acid residues is a non-naturally occurring amino acid (e.g., an amino acid analog). As used herein, the term encompasses amino acid chains of any length, including full-length proteins (e.g., antigens).
The term "antigen" refers to a substance that is capable of being recognized by the immune system of a subject (including the adaptive immune system) and is capable of generating an immune response (including an antigen-specific immune response) upon contact with an antigen in a subject. In certain embodiments, the antigen is a protein (e.g., a tumor-associated antigen (TAA)) associated with a diseased cell (e.g., a pathogen or neoplastic cell-infected cell).
In the context of peptides or polypeptides, the term "fragment" refers to a peptide or polypeptide comprising less than the full-length amino acid sequence. Such fragments may result from N-terminal truncations, C-terminal truncations and/or deletions of residues within the amino acid sequence. Fragments may be produced by alternative RNA splicing or in vivo proteases. In certain embodiments, a fragment refers to a polypeptide comprising at least 5 consecutive amino acid residues, at least 10 consecutive amino acid residues, at least 15 consecutive amino acid residues, at least 20 consecutive amino acid residues, at least 25 consecutive amino acid sequences, at least 30 consecutive amino acid residues, at least 40 consecutive amino acid residues, at least 50 consecutive amino acid residues, at least 60 consecutive amino acid residues, at least 70 consecutive amino acid residues, at least 80 consecutive amino acid residues, at least 90 consecutive amino acid residues, at least 100 consecutive amino acid residues, at least 125 consecutive amino acid residues, at least 150 consecutive amino acid residues, at least 175 consecutive amino acid residues, at least 200 consecutive amino acid residues, at least 250, at least 300, at least 350, at least 400, at least 450, at least 500, at least 550, at least 600, at least 650, at least 700, at least 750, at least 800, at least 850, at least 900 or at least 950 consecutive amino acid residue sequences. In a specific embodiment, a fragment of a polypeptide retains at least 1, at least 2, at least 3 or more functions of the polypeptide.
An "epitope" is a site on the surface of an antigen molecule to which a specific antibody molecule binds, e.g., a localized region on the surface of an antigen that is capable of binding to one or more antigen binding regions of an antibody, has antigen or immunogenic activity in an animal, e.g., a mammal (e.g., a human), and is capable of eliciting an immune response. An epitope with immunogenic activity is a portion of a polypeptide that elicits an antibody response in an animal. Epitopes having antigenic activity are part of an antibody-bound polypeptide as determined by any method known in the art, including, for example, by an immunoassay. The epitope need not be immunogenic. Epitopes are generally composed of a collection of chemically active surface groups of a molecule, such as amino acids or sugar side chains, and generally have specific three-dimensional structural features, as well as specific charge features. The antibody epitope may be a linear epitope or a conformational epitope. Linear epitopes are formed by the continuous sequence of amino acids in a protein. Conformational epitopes are formed by discrete amino acids in the protein sequence, but bind together when the protein is folded into its three-dimensional structure. An induced epitope is formed when the three-dimensional structure of a protein is in an altered configuration, such as after activation or binding of another protein or ligand. In certain embodiments, the epitope is a three-dimensional surface feature of the polypeptide. In other embodiments, the epitope is a linear characteristic of the polypeptide. Typically, an antigen has several or many different epitopes and can react with many different antibodies.
As used herein, the term "genetic vaccine" refers to a therapeutic or prophylactic composition comprising at least one nucleic acid molecule encoding an antigen associated with a target disease (e.g., an infectious disease or neoplastic disease). The production of peptides or proteins is encoded by administering a vaccine (vaccination) to a subject, thereby eliciting an immune response in the subject against the target disease. In certain embodiments, the immune response includes an adaptive immune response, such as the production of antibodies to the encoded antigen, and/or immune cells capable of activating and proliferating diseased cells for specific elimination of the expressed antigen. In certain embodiments, the immune response further comprises an innate immune response. According to the present invention, the vaccine may be administered to the subject either before or after the onset of clinical symptoms of the disease of interest. In some embodiments, vaccination of healthy or asymptomatic subjects renders the vaccinated subjects immune or less susceptible to the disease process of interest. In some embodiments, vaccination of a subject with symptoms of a disease may improve the disease condition of the vaccinated subject or treat the disease.
The terms "innate immune response" and "innate immunity" are well known in the art and refer to the non-specific defense mechanisms that the human immune system initiates upon recognition of pathogen-associated molecules, which are involved in different forms of cellular activity, including cytokine production and cell death of various pathways. As described herein, the innate immune response includes, but is not limited to, increased production of inflammatory cytokines (e.g., type I interferon or IL-10 production), activation of the nfkb pathway, increased proliferation, maturation, differentiation and/or survival of immune cells, and in some cases, induced apoptosis. Activation of innate immunity can be detected using methods known in the art, for example by measuring activation of (NF) - κb.
The terms "adaptive immune response" and "adaptive immunity" are well known in the art and refer to antigen-specific defense mechanisms, including humoral and cell-mediated responses, initiated by the immune system of the human body upon recognition of a particular antigen. An adaptive immune response, as described herein, includes a cellular response triggered and/or enhanced by a vaccine composition (e.g., a genetic composition as described herein). In some embodiments, the vaccine composition comprises an antigen that is a target of an antigen-specific adaptive immune response. In other embodiments, the vaccine composition, upon administration, allows for the production of an antigen in the subject that is a target of an antigen-specific adaptive immune response. Activation of the adaptive immune response may be detected using methods known in the art, such as by monitoring the production of antigen-specific antibodies or monitoring antigen-specific cell-mediated cytotoxicity levels.
The term "antibody" is intended to include a polypeptide product secreted by effector b cells that consists of two identical pairs of polypeptide chains, wherein each pair of polypeptide chains has a heavy chain (about 50-70 kDa) and a light chain (about 25 kDa), the N-terminal portion of each chain comprising a variable region of about 100 to about 130 or more amino acids, the C-terminal portion of each chain comprising a constant region capable of binding to a particular molecular antigen, and immunoglobulins are not merely antibodies. See, for example, antibody Engineering (Borrebaeck, editions, 2d edition 1995) and Kuby, immunology (3 d edition 1997). In certain embodiments, a particular molecular antigen comprises a polypeptide, fragment or epitope thereof, which can bind to an antibody described herein. Antibodies also include, but are not limited to, synthetic antibodies, antibodies produced by recombinant means, camelized antibodies, intracellular antibodies (intracelluar antibodies), anti-Id antibodies and functional fragments of these antibodies, which refer to functional polypeptide fragments isolated from the heavy or light chain of the aforementioned antibodies that retain a portion of their full binding activity. Some non-limiting examples of functional fragments include single chain antibodies (scFv) (including monospecific, bispecific, and the like), fab fragments, F (ab ') fragments, F (ab) 2 fragments, F (ab') 2 fragments, disulfide stabilized antibodies (dsFv), fd fragments, fv fragments, diabodies, triabodies, tetrabodies, and minibodies. In particular, antibodies described herein include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, such as may be antigen binding domains or molecules that contain an antigen binding site (e.g., one or more CDRs of an antibody). Such antibody fragments may be those described in Harlow and Lane, antibodies: A Laboratory Manual (1989); mol. Biology and Biotechnology: A Comprehensive Desk Reference (Myers editions, 1995); huston et al, 1993,Cell Biophysics22:189-224; pluckthun and Skerra,1989, meth. Enzymol.178:497-515; and Day, advanced Immunochemistry (2 d edition 1990). Antibodies provided by the invention may be of any type (e.g., igG, igE, igM, igD and IgA types, etc.) or any subclass (e.g., igG1, igG2, igG3, igG4, igA1 and IgA2 types, etc.) of immunoglobulin molecules.
The term "administration" refers to the act of delivering an in vitro substance (e.g., a lipid nanoparticle composition as described herein) to a patient, for example, via mucosal, intramuscular/subcutaneous injection, intravenous injection, or in other physical means known in the art. When used to treat a disease, disorder, condition, or symptom thereof, administration of the substance is typically performed after the onset of the disease, disorder, condition, or symptom thereof. When used to prevent a disease, disorder, condition or symptom, administration of the substance is typically performed prior to onset of the disease, disorder, condition or symptom.
By "chronic" administration is meant administration in a continuous mode (e.g., for a period of time such as days, weeks, months or years) as opposed to an acute mode of administration, to maintain the initial therapeutic effect (activity) over an extended period of time. The "intermittent" administration is not continuous but periodic, without interrupting the treatment.
The term "targeted delivery" or verb form of "target" refers to the process of facilitating delivery of an agent (e.g., a therapeutic payload molecule in a lipid nanoparticle composition described herein) to a particular organ, tissue, cell, and/or intracellular compartment (referred to as a target site) such that the target site is delivered more than any other organ, tissue, cell, or intracellular compartment (referred to as a non-target site). Targeted delivery may be detected by methods known in the art, for example, by comparing the concentration of the agent delivered in the target cell population to the concentration of the agent delivered in the non-target cell population following systemic administration. In certain embodiments, targeted delivery results in at least a 2-fold higher concentration at the target location as compared to the non-target location.
An "effective amount" is generally an amount sufficient to reduce the severity and/or frequency of symptoms, eliminate symptoms and/or underlying etiology, prevent the occurrence of symptoms and/or their etiology, and/or ameliorate or remedy damage. Diseases caused by or associated with a disease, disorder or condition include infection, neoplasia, and the like. In some embodiments, the effective amount is a therapeutically effective amount or a prophylactically effective amount.
As used herein, the term "therapeutically effective amount" refers to an amount of an agent (e.g., a vaccine composition) sufficient to reduce and/or ameliorate the severity and/or duration of a symptom associated with a given disease, disorder, or condition (e.g., an infectious disease caused by a viral infection, or a neoplastic disease of a cancer, etc.). The "therapeutically effective amount" of a substance/molecule/agent of the present disclosure (lipid nanoparticle compositions as described herein) can vary depending on factors such as the disease state, age, sex, and weight of the individual, and the ability of the substance/molecule/agent to elicit a desired response in the individual, and the like. Therapeutically effective amounts include amounts in which any toxic or deleterious effects of the substance/molecule/agent are offset by the beneficial effects of the treatment. In certain embodiments, the term "therapeutically effective amount" refers to an amount of a lipid nanoparticle composition or therapeutic or prophylactic agent (e.g., therapeutic mRNA) contained therein that is effective to "treat" a disease, disorder, or condition in a subject or mammal.
A "prophylactically effective amount" is an amount of a pharmaceutical composition that, when administered to a subject, will have the intended prophylactic effect, e.g., preventing, delaying or reducing the likelihood of the onset (or recurrence) of a disease, disorder, and related symptoms, such as an infectious disease caused by a viral infection or a neoplastic disease such as cancer. Condition or related symptoms. Typically, but not necessarily, the prophylactically effective amount may be less than the therapeutically effective amount because the prophylactic dose is administered to the subject prior to or at an earlier stage of the disease, disorder, or condition. The complete therapeutic or prophylactic effect does not necessarily occur by administration of one dose, but may occur only after administration of a series of doses. Thus, a therapeutically or prophylactically effective amount can be administered in one or more administrations.
The term "preventing" refers to reducing the likelihood of suffering from a disease, disorder, condition, or related symptom (e.g., infectious disease, e.g., infection by a virus or neoplastic disease, e.g., cancer).
The term "managing" refers to the beneficial effect a subject obtains from treatment (e.g., prophylactic or therapeutic agent) that does not result in a cure of the disease. In certain embodiments, one or more therapies (e.g., prophylactic or therapeutic agents, such as lipid nanoparticle compositions described herein) are administered to a subject to "manage" one or more symptoms of an infectious or neoplastic disease, thereby preventing progression or worsening of the disease.
The term "prophylactic agent" refers to any agent that can completely or partially inhibit the development, recurrence, onset, or spread of a disease and/or symptoms associated therewith in a subject.
The term "therapeutic agent" refers to any drug that can be used to treat, prevent, or ameliorate a disease, disorder, or condition, including one or more symptoms of a disease, disorder, or condition and related symptoms.
The term "therapy" refers to any regimen, method and/or agent that can be used to prevent, manage, treat and/or ameliorate a disease, disorder or condition. In certain embodiments, the term "therapy" refers to biological therapies, supportive therapies, and/or other therapies that can be used to prevent, control, treat, and/or ameliorate a known disease, disorder, or condition, as known to those skilled in the art, such as medical personnel.
"prophylactically effective serum titer" is the serum titer of an antibody in a subject (e.g., a human) that completely or partially inhibits the development, recurrence, onset, or spread of a disease, disorder, or condition and symptoms associated therewith.
In certain embodiments, a "therapeutically effective serum titer" is a serum titer of antibodies in a subject (e.g., a human) that reduces the severity, duration, and/or symptoms associated with a disease, disorder, or condition.
The term "serum titer" refers to the average serum titer in a subject from multiple samples (e.g., at multiple time points) or in a population of subjects of at least 10, at least 20, at least 40 subjects, up to about 100, 1000, or more.
The term "side effects" encompasses undesired and/or adverse effects of a therapy (e.g., a prophylactic or therapeutic agent). The detrimental effects are not necessarily detrimental. Adverse effects of treatment (e.g., prophylactic or therapeutic agents) can be detrimental, uncomfortable, or risky. Examples of side effects include diarrhea, cough, gastroenteritis, wheezing, nausea, vomiting, anorexia, abdominal cramps, fever, pain, weight loss, dehydration, alopecia, dyspnea, insomnia, dizziness, mucositis, nerve and muscle effects, fatigue, dry mouth, loss of appetite, rash or swelling at the site of administration, symptoms like influenza such as fever, coldness, tiredness, digestive tract problems and allergic reactions, etc. Other undesirable effects experienced by patients are known in the art, and related disclosures are made in the Physics' Desk Reference (68 th edition 2014).
The terms "subject" and "patient" may be used interchangeably. As described herein, in certain embodiments, the subject is a mammal, e.g., a non-primate (e.g., cow, pig, horse, cat, dog, rat, etc.) or a primate (e.g., monkey and human). In particular embodiments, the subject is a human. In one embodiment, the subject is a mammal (e.g., a human) having an infectious disease or neoplastic disease. In another embodiment, the subject is a mammal (e.g., a human) at risk of developing an infectious or neoplastic disease.
The term "detectable probe" refers to a composition that provides a detectable signal. The term includes, but is not limited to, any fluorophore, chromophore, radiolabel, enzyme, antibody or antibody fragment, etc. that provides a detectable signal by its activity.
The term "detectable agent" refers to a substance that can be used to determine the presence of a desired molecule in a sample or subject, such as an antigen encoded by an mRNA molecule as described herein. The detectable agent may be a substance that can be visualized or a substance that can be determined and/or measured (e.g., by quantification).
"substantially all" means at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or about 100%.
As used herein, unless otherwise indicated, the term "about" or "approximately" refers to an acceptable error for a particular value determined by one of ordinary skill in the art, which depends in part on the manner in which the value is measured or determined. In certain embodiments, the term "about" or "approximately" means within 1, 2, 3, or 4 standard deviations. In certain embodiments, the term "about" or "approximately" refers to within 20%,15%,10%,9%,8%,7%,6%,5%,4%,3%,2%,1%,0.5%,0.05% or less of a given value or range.
As used herein, the singular terms "a," "an," and "the" include the plural forms thereof unless the context clearly dictates otherwise.
As used herein and unless otherwise indicated, the term "target moiety" or "targeting group" refers to a moiety or group that is capable of specifically binding to a molecule on the surface of a target cell, such as a cell within a target tissue of a subject.
All publications, patent applications, accession numbers, and other references cited in this specification are herein incorporated by reference in their entirety and each individual publication or patent application is specifically and individually indicated to be incorporated by reference. The publications discussed herein are publications that are disclosed prior to the filing date of the present application. Nothing herein is to be construed as an admission that the application is not entitled to antedate such publication by virtue of prior application. Further, the release date provided herein may be different from the actual release date, which may require independent confirmation.
The present application has been described with respect to a number of embodiments. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the application. Accordingly, the description in the experimental section and examples is intended to illustrate and not limit the scope of the application as described in the claims.
7.3 Polymer conjugated lipid Compounds
In one embodiment, provided herein are compounds of formula (I):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
l is a lipid;
x is a linker;
each R 3 Independently H or C 1 -C 6 An alkyl group;
each Y 1 Independently is a bond, O, S or NR a
Each G 4 Independently is a bond or C 1 -C 12 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -S-or-NR a -optionally substituted;
each G 5 Independently is a bond or C 1 -C 12 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -S-or-NR a -optionally substituted;
each R a H, C independently 1 -C 12 Alkyl or C 2 -C 12 Alkenyl groups;
Z 1 and Z 2 One of which is a positively charged moiety and Z 1 And Z 2 The other of which is a negatively charged moiety;
n is an integer from 2 to 100;
t is hydrogen, halogen, alkyl, alkenyl, -OR ', -SR', -COOR ', -OCOR', -NR 'R', -N + (R”) 3 、-P + (R”) 3 -S-C (=s) -S-R ", -S-C (=s) -O-R", -S-C (=s) -NR "R", -S-C (=s) -aryl, cyano, azido, aryl, heteroaryl, or a targeting group, wherein R "in each occurrence is independently hydrogen or alkyl; and
wherein each alkyl, alkenyl, alkylene, aryl, and heteroaryl is independently optionally substituted; and is also provided with
Provided that the compound is not:
in one embodiment, when Z 1 Or Z is 2 Where the negatively charged moiety of (2) is a carboxylate (-COO-) T is not bromine.
In one embodiment, the compound is a compound represented by formula (II):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
l is a lipid;
x is a linker;
each R 3 Independently H or C 1 -C 6 An alkyl group;
each R 4 Independently H or C 1 -C 6 An alkyl group;
each X is 1 Independently a bond or-C (O) -Y 1 -;
Each X is 2 Independently a bond or-C (O) -Y 2 -;
Each Y 1 Independently is a bond, O, S or NR a
Each Y 2 Independently is a bond, O, S or NR a
Each G 4 Independently is a bond or C 1 -C 12 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -S-or-NR a -optionally substituted;
each G 5 Independently is a bond or C 1 -C 12 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -S-or-NR a -optionally substituted;
each R a H, C independently 1 -C 12 Alkyl or C 2 -C 12 Alkenyl groups;
Z 1 and Z 2 One of which is a positively charged moiety and Z 1 And Z 2 The other of which is a negatively charged moiety;
n is an even integer from 2 to 100;
"ran" means eachUnit and each->The units appear in any order within { };
t is hydrogen, halogen, alkyl, alkenyl, -OR ', -SR', -COOR ', -OCOR', -NR 'R', -N + (R”) 3 、-P + (R”) 3 -S-C (=s) -S-R ", -S-C (=s) -O-R", -S-C (=s) -NR "R", -S-C (=s) -aryl, cyano, azido, aryl, heteroaryl, or a targeting group, wherein R "in each occurrence is independently hydrogen or alkyl; and
wherein each alkyl, alkenyl, alkylene, aryl, and heteroaryl is independently optionally substituted.
In formula (II), there are equal numbers of positively charged moieties and negatively charged moieties, so that the compound is generally neutral. Positively charged moieties (e.g., when Z 1 When positively chargedUnit) and negatively charged moiety (e.g., when Z 2 When negatively charged->The cells) may occur in any order within { }. For example, the charged moiety within { } may be +, -, +, - …; can be +, - …; can be +, - …; can be-, +, -, …
In one embodiment, X 1 Is a key. In one embodiment, X 1 is-C (O) -Y 1 -。
In one embodiment, X 2 Is a key. In one embodiment, X 2 is-C (O) -Y 2 -。
In one embodiment, Y 1 Is O. In one embodiment, Y 1 S. In one embodiment, Y 1 Is NR (NR) a . In one embodiment, Y 1 Is NH. In one embodiment, Y 1 Is a key.
In one embodiment, Y 2 Is O. In one embodiment, Y 2 S. In one embodiment, Y 2 Is NR (NR) a . In one embodiment, Y 2 Is NH. In one embodiment, Y 2 Is a key.
In one embodiment, G 4 Is a key. In one embodiment, G 4 Is C 1 -C 6 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -S-or-NR a -optionally substituted. In one embodiment, G 4 Is C 1 -C 3 An alkylene group. In one embodiment, G 4 Is C 1 An alkylene group. In one embodiment, G 4 Is C 2 An alkylene group. In one embodiment, G 4 Is C 3 An alkylene group.
In one embodiment, G 5 Is a key. In one embodiment, G 5 Is C 1 -C 6 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -S-or-NR a -optionally substituted. In one embodiment, G 5 Is C 1 -C 3 An alkylene group. In one embodiment, G 5 Is C 1 An alkylene group. In one embodiment, G 5 Is C 2 An alkylene group. In one embodiment, G 5 Is C 3 An alkylene group.
In one embodiment, Z 1 Is a positively charged moiety and Z 2 Is a negatively charged moiety. In one embodiment, Z 2 Is a positively charged moiety and Z 1 Is a negatively charged moiety. As used herein and unless otherwise indicated, "charged moiety" means charged at any pH or hydrogen ion activity of its environment, or capable of being charged to itThe pH of the environment (e.g., the environment in which it is intended to be used) or the portion of the hydrogen ion activity that is charged in response.
In one embodiment, Z 1 Or Z is 2 Is a quaternary amine cation.
In one embodiment, Z 1 Or Z is 2 Is a carboxylate, sulfonate or phosphate anion.
In one embodiment, Z 1 Or Z is 2 Is carboxylate, and corresponding X 1 And G 4 Or corresponding X 2 And G 5 Are all keys. In one embodiment, theThe units are->In one embodiment, said ∈ ->The units are->
In one embodiment, the compound is a compound represented by formula (I-A):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
In one embodiment, the compound is a compound represented by formula (I-B):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
In one embodiment, the compound is a compound represented by formula (I-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment, the compound is a compound represented by formula (II-A):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment, the compound is a compound represented by formula (II-B):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment, the compound is a compound represented by formula (II-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
each R p Independently C 1 -C 6 Alkyl or-O- (C) 1 -C 6 An alkyl group); and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment, L is a compound comprising one or more C 6 -C 24 Hydrocarbon chains (e.g. one or more C' s 6 -C 24 Alkyl or C 6 -C 24 Alkenyl) lipids. In one embodiment, L is a compound comprising two C 8 -C 24 Lipids of hydrocarbon chains. In one embodiment, the two C' s 8 -C 24 The hydrocarbon chain being selected from two C 8 -C 22 Hydrocarbon chain, two C 8 -C 20 Hydrocarbon chain, two C 8 -C 19 Hydrocarbon chain, two C 8 -C 18 Hydrocarbon chain, two C 8 -C 17 Hydrocarbon chain, two C 10 -C 24 Hydrocarbon chain, two C 10 -C 22 Hydrocarbon chain, two C 10 -C 20 Hydrocarbon chain, two C 10 -C 19 Hydrocarbon chain, two C 10 -C 18 Hydrocarbon chain, two C 10 -C 17 Hydrocarbon chain, two C 12 -C 24 Hydrocarbon chain, two C 12 -C 22 Hydrocarbon chain, two C 12 -C 20 Hydrocarbon chain, two C 12 -C 19 Hydrocarbon chain, two C 12 -C 18 Hydrocarbon chain, two C 12 -C 17 Hydrocarbon chain, two C 13 -C 24 Hydrocarbon chain, two C 13 -C 22 Hydrocarbon chain, two C 13 -C 20 Hydrocarbon chain, two C 13 -C 19 Hydrocarbon chain, two C 13 -C 18 Hydrocarbon chain or two C' s 13 -C 17 A hydrocarbon chain. In one embodiment, the lipid is glycerophospholipid. In one embodiment, suitable glycerophospholipids include 1, 2-dilauroyl-sn-glycero-3-phosphoethanolamine, 1, 2-ditridecyl-sn-glycero-3-phosphoethanolamine, 1, 2-dimyristoyl-sn-glycero-3-phosphoethanolamine (DMPE), 1, 2-ditridecyl-sn-glycero-3-phosphoethanolamine1, 2-dipalmitoyl-sn-glycero-3-phosphate ethanolamine (DPPE), 1, 2-bipearoyl-sn-glycero-3-phosphate ethanolamine, 1, 2-distearoyl-sn-glycero-3-phosphate ethanolamine (DSPE), 1, 2-bisnonadecanoyl-sn-glycero-3-phosphate ethanolamine, 1, 2-diacetyleneyl-sn-glycero-3-phosphate ethanolamine, 1, 2-dimyristoyl oleoyl-sn-glycero-3-phosphate ethanolamine, 1, 2-dipalmitoyl-sn-glycero-3-phosphate ethanolamine, 1, 2-diterpene acyl (distienoyl) -sn-glycero-3-phosphate ethanolamine, 1, 2-dioleoyl (dielaiyl) -sn-glycero-3-phosphate ethanolamine, 1, 2-octacosenoyl (dioleoyl) -sn-glycero-3-phosphate ethanolamine, 1, 2-dioleoyl-glycero-3-phosphate ethanolamine, 1, 2-di- α -linolenoyl (sn-glycero-3-phosphoethanolamine and 1, 2-di-arachidonoyl-sn-glycero-3-phosphoethanolamine. In one embodiment, the lipid is a glyceride. In one embodiment, suitable glycerides include tetracosacylglycerol (DMG), distearoyl glycerol (DSG), dipalmitoyl glycerol (DPG), dilauryl glycerol, ditridoyl glycerol, ditpentadecanoyl glycerol, bisheptadecanoyl glycerol, bisnonadecanoyl glycerol, ditolyl glycerol, dimyristoyl glycerol, dipalmitoyl glycerol, distalinyl glycerol, dioleoyl glycerol, ditolyl (dieleayl) glycerol, octacosoyl (divalinyl) glycerol, dillenoyl (dilineleayl) glycerol, di-alpha-linolenoyl (linolenoyl) glycerol, and ditolyl glycerol.
In one embodiment, L is a sterol lipid. In one embodiment, the lipid is selected from cholesterol, cholesteryl hemisuccinate, stigmasterol indoside (sitoindoside) I, stigmasterol indoside (sitoindoside) II, glucosyl stigmasterol, 16:0 stigmasterol glucose, 18:1 stigmasterol glucose, glucosyl sitosterol B, cholesterol sulfate, DHEA sulfate, FF-MAS, campesterol, zymosterol, dihydrositosterol, sitosterol, stigmasterol, dioscin, 7-dehydrocholesterol, lanosterol-95, dihydrolanosterol, 14-desmethyl (desmethyl) -lanosterol, zymosterol, 24-dehydrocholesterol, 7-alkenecholanol, and pregnenolone. In one embodiment, the lipid is cholesterol.
In one embodiment, L is sphingolipid. In one embodiment, the sphingolipid is selected from the group consisting of N-octanoic acid-sphingosine, sphinganine-1-phosphate (d17:0), sphingosine-1-phosphate (d17:1), sphinganine-1-phosphate (d18:0), sphingosine-1-phosphate (d18:1), sphinganine-1-phosphate (d20:0), sphingosine-1-phosphate (d20:1), 1-deoxysphinganine, sphinganine (d17:0), sphinganine (d18:0), sha Fenge, sphinganine (d20:0), sphingosine (d14:1), sphingosine (d17:1), sphingosine (d18:1), sphingosine (d20:1), 1-deoxysphingosine, 4E, 8Z-sphingosine, 4E, 11Z-sphingosine and 4E, 14Z-dienol. In one embodiment, the sphingolipid is N-octanoic acid-sphingosine.
In one embodiment, L is a lipid comprising the formula, and in one embodiment, L is a lipid comprising the formula:
wherein:
G 1 、G 2 and G 3 Are each independently a bond, C 1 -C 12 Alkylene or C 2 -C 12 Alkenylene;
L 1 is-OC (=O) R 1 、-C(=O)OR 1 、-OC(=O)OR 1 、-C(=O)R 1 、-OR 1 、-S(O) x R 1 、-S-SR 1 、-C(=O)SR 1 、-SC(=O)R 1 、-NR a C(=O)R 1 、-C(=O)NR b R c 、-NR a C(=O)NR b R c 、-OC(=O)NR b R c 、-NR a C(=O)OR 1 、-SC(=S)R 1 、-C(=S)SR 1 、-C(=S)R 1 、-CH(OH)R 1 、-P(=O)(OR b )(OR c )、-(C 6 -C 10 Arylene) -R 1 (6-to 10-membered heteroarylene) -R 1 Or R is 1
L 2 is-OC (=O) R 2 、-C(=O)OR 2 、-OC(=O)OR 2 、-C(=O)R 2 、-OR 2 、-S(O) x R 2 、-S-SR 2 、-C(=O)SR 2 、-SC(=O)R 2 、-NR d C(=O)R 2 、-C(=O)NR e R f 、-NR d C(=O)NR e R f 、-OC(=O)NR e R f 、-NR d C(=O)OR 2 、-SC(=S)R 2 、-C(=S)SR 2 、-C(=S)R 2 、-CH(OH)R 2 、-P(=O)(OR e )(OR f ) - (arylene) -R 2 (6-to 10-membered heteroarylene) -R 2 Or R is 2
L 3 is-OC (=O) -, -C (=O) O-; -OC (=o O-, O- -C (=o) -, -O-, -S (O) x -、-S-S-、-C(=O)S-、-SC(=O)-、-NR a C(=O)-、-C(=O)NR b -、-NR a C(=O)NR b -、-OC(=O)NR b -、-NR a C(=O)O-、-SC(=S)-、-C(=S)S-、-C(=S)-、-CH(OH)-、-P(=O)(OR b )O-、-(C 6 -C 10 Arylene) -or- (6-to 10-membered heteroarylene) -;
R 1 and R is 2 Each independently is C 6 -C 24 Alkyl or C 6 -C 24 Alkenyl groups;
R a 、R b 、R d and R is e H, C each independently of the other 1 -C 12 Alkyl or C 2 -C 12 Alkenyl groups;
R c and R is f Each independently is C 1 -C 12 Alkyl or C 2 -C 12 Alkenyl groups;
x is 0, 1 or 2, and
wherein each alkyl, alkenyl, alkylene, alkenylene, arylene, and heteroarylene is independently optionally substituted.
In one embodiment, the compound is a compound of formula (I-D):
or a pharmaceutically acceptable salt or stereoisomer thereof.
In one embodiment, the compound is a compound of formula (II-D):
or a pharmaceutically acceptable salt or stereoisomer thereof.
In one embodiment, L is a lipid comprising the formula, and in one embodiment, L is a lipid comprising the formula:
In one embodiment, L is a lipid comprising the formula, and in one embodiment, L is a lipid comprising the formula:
in one embodiment, the compound is a compound of formula (I-E):
or a pharmaceutically acceptable salt or stereoisomer thereof.
In one embodiment, the compound is a compound of formula (II-E):
or a pharmaceutically acceptable salt or stereoisomer thereof.
In one embodiment, G 1 Is a key. In one embodiment, G 1 Is C 1 An alkylene group. In one embodiment, G 1 Is C 2 -C 12 An alkylene group. In one embodiment, G 1 Is C 4 -C 8 An alkylene group. In one embodiment, G 1 Is C 5 -C 7 An alkylene group. In one embodiment, G 1 Is C 5 An alkylene group. In one embodiment, G 1 Is C 7 An alkylene group. In one embodiment, G 1 Is C 2 -C 12 Alkenylene radicals. In one embodiment, G 1 Is C 4 -C 8 Alkenylene radicals. In one embodiment, G 1 Is C 5 -C 7 Alkenylene radicals. In one embodiment, G 1 Is C 5 Alkenylene radicals. In one embodiment, G 1 Is C 7 Alkenylene radicals.
In one embodiment, G 2 Is a key. In one embodiment, G 2 Is C 1 An alkylene group. In one embodiment, G 2 Is C 2 -C 12 An alkylene group. In one embodiment, G 2 Is C 4 -C 8 An alkylene group. In one embodiment, G 2 Is C 5 -C 7 An alkylene group. In one embodiment, G 2 Is C 5 An alkylene group. In one embodiment, G 2 Is C 7 An alkylene group. In one embodiment, G 2 Is C 2 -C 12 Alkenylene radicals. In one embodiment, G 2 Is C 4 -C 8 Alkenylene radicals. In one embodimentIn (G) 2 Is C 5 -C 7 Alkenylene radicals. In one embodiment, G 2 Is C 5 Alkenylene radicals. In one embodiment, G 2 Is C 7 Alkenylene radicals.
In one embodiment, G 1 And G 2 Each independently is a bond or C 2 -C 12 Alkylene (e.g., C 4 -C 8 Alkylene groups, e.g. C 5- C 7 Alkylene groups, e.g. C 5 Alkylene or C 7 An alkylene group). In one embodiment, G 1 And G 2 Are all keys. In one embodiment, G 1 And G 2 One being a bond and the other being C 2 -C 12 Alkylene (e.g., C 4 -C 8 Alkylene groups, e.g. C 5 -C 7 Alkylene groups, e.g. C 5 Alkylene or C 7 An alkylene group). In one embodiment, G 1 And G 2 Each independently is C 2 -C 12 Alkylene (e.g., C 4 -C 8 Alkylene groups, e.g. C 5 -C 7 Alkylene groups, e.g. C 5 Alkylene or C 7 An alkylene group). In one embodiment, G 1 And G 2 Are each independently a bond, C 5 Alkylene or C 7 An alkylene group. In one embodiment, G 1 And G 2 Each independently is a bond or C 1 An alkylene group. In one embodiment, G 1 And G 2 One being a bond and the other being C 1 An alkylene group. In one embodiment, G 1 And G 2 Are all C 1 An alkylene group.
In one embodiment, G 3 Is a key. In one embodiment, G 3 Is C 1 An alkylene group. In one embodiment, G 3 Is C 2 -C 12 An alkylene group. In one embodiment, G 3 Is C 4 -C 8 An alkylene group. In one embodiment, G 3 Is C 5 -C 7 An alkylene group. In one embodiment, G 3 Is C 5 An alkylene group. In a real worldIn embodiments, G 3 Is C 7 An alkylene group. In one embodiment, G 3 Is C 2 -C 12 Alkenylene radicals. In one embodiment, G 3 Is C 4 -C 8 Alkenylene radicals. In one embodiment, G 3 Is C 5 -C 7 Alkenylene radicals. In one embodiment, G 3 Is C 5 Alkenylene radicals. In one embodiment, G 3 Is C 7 Alkenylene radicals.
In one embodiment, L 1 Is R 1
In one embodiment, L 1 is-OC (=O) R 1 、-C(=O)OR 1 、-OC(=O)OR 1 、-C(=O)R 1 、-OR 1 、-S(O) x R 1 、-S-SR 1 、-C(=O)SR 1 、-SC(=O)R 1 、-NR a C(=O)R 1 、-C(=O)NR b R c 、-NR a C(=O)NR b R c 、-OC(=O)NR b R c 、-NR a C(=O)OR 1 、-SC(=S)R 1 、-C(=S)SR 1 、-C(=S)R 1 、-CH(OH)R 1 OR-P (=O) (OR b )(OR c ). In one embodiment, L 1 is-OC (=O) R 1 、-C(=O)OR 1 、-C(=O)SR 1 、-SC(=O)R 1 、-NR a C(=O)R 1 or-C (=O) NR b R c . In one embodiment, L 1 is-OC (=O) R 1 、-C(=O)OR 1 、-NR a C(=O)R 1 or-C (=O) NR b R c . In one embodiment, L 1 is-OC (=O) R 1 . In one embodiment, L 1 is-C (=O) OR 1 . In one embodiment, L 1 is-NR a C(=O)R 1 . In one embodiment, L 1 is-C (=O) NR b R c
In one embodiment, L 2 Is R 2
In one embodiment, L 2 is-OC (=O) R 2 、-C(=O)OR 2 、-OC(=O)OR 2 、-C(=O)R 2 、-OR 2 、-S(O) x R 2 、-S-SR 2 、-C(=O)SR 2 、-SC(=O)R 2 、-NR d C(=O)R 2 、-C(=O)NR e R f 、-NR d C(=O)NR e R f 、-OC(=O)NR e R f 、-NR d C(=O)OR 2 、-SC(=S)R 2 、-C(=S)SR 2 、-C(=S)R 2 、-CH(OH)R 2 OR-P (=O) (OR e )(OR f ). In one embodiment, L 2 is-OC (=O) R 2 、-C(=O)OR 2 、-C(=O)SR 2 、-SC(=O)R 2 、-NR d C(=O)R 2 or-C (=O) NR e R f . In one embodiment, L 2 is-OC (=O) R 2 、-C(=O)OR 2 、-NR d C(=O)R 2 or-C (=O) NR e R f . In one embodiment, L 2 is-OC (=O) R 2 . In one embodiment, L 2 is-C (=O) OR 2 . In one embodiment, L 2 is-NR d C(=O)R 2 . In one embodiment, L 2 is-C (=O) NR e R f
In one embodiment, G 1 Is a bond and L 1 Is R 1 . In one embodiment, G 1 Is a bond and L 1 is-OC (=O) R 1 . In one embodiment, G 1 Is C 1 Alkylene and L 1 is-OC (=O) R 1
In one embodiment, G 2 Is a bond and L 2 Is R 2 . In one embodiment, G 2 Is a bond and L 2 is-OC (=O) R 2 . In one embodiment, G 2 Is C 1 Alkylene and L 2 is-OC (=O) R 2
In one embodiment, L 3 is-O-. In one embodiment, L 3 is-OC (=O)) -. In one embodiment, L 3 is-C (=O) O-. As described herein and unless otherwise indicated, at L 3 The connection point on the left side is connected to G 3 And is positioned at L 3 The connection point on the right side is X. For example, when L 3 In the case of-OC (=O) -it means G 3 -OC(=O)-X。
In one embodiment, L is a compound of formula (I)The lipids shown. In one embodiment, L is of the formula +.>The lipids shown or stereoisomers thereof.
In one embodiment, X is C 1 -C 12 An alkylene group, wherein:
one or more-CH 2 -independently by-O-, -NR a -、-OC(=O)-、-C(=O)O-、-OC(=O)O-、-C(=O)-、-S(O) x -、-S-S-、-C(=O)S-、-SC(=O)-、-NR a C(=O)-、-C(=O)NR b -、-NR a C(=O)NR b -、-SC(=S)-、-C(=S)S-、-C(=S)-、-P(=O)(OR b )-O-、-O-P(=O)(OR b )-O-、-(C 6 -C 10 Arylene) -or- (6-to 10-membered heteroarylene) -optionally substituted;
x is 0, 1 or 2;
R a and R is b H, C each independently of the other 1 -C 12 Alkyl or C 2 -C 12 Alkenyl groups; and
and each of alkyl, alkenyl, alkylene, arylene, and heteroarylene is optionally substituted.
In one embodiment, X is C 1 -C 12 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -NR a -、-OC(=O)-、-C(=O)O-、-NR a C (=o) -or-C (=o) NR b -optionally substituted. In one embodiment, X is C 1 -C 8 Alkylene groups, one or more ofCH 2 -independently by-O-, -NR a -、-OC(=O)-、-C(=O)O-、-NR a C (=o) -or-C (=o) NR b -optionally substituted.
In one embodiment, X is-C (=o) -C 1 -C 11 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -NR a -、-OC(=O)-、-C(=O)O-、-NR a C (=o) -or-C (=o) NR b -optionally substituted. In one embodiment, X is-C (=o) -C 1 -C 7 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -NR a -、-OC(=O)-、-C(=O)O-、-NR a C (=o) -or-C (=o) NR b -optionally substituted.
In one embodiment, X is-P (=o) (OR b )-O-C 1 -C 11 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -NR a -、-OC(=O)-、-C(=O)O-、-NR a C (=o) -or-C (=o) NR b -optionally substituted. In one embodiment, X is-P (=o) (OR b )-O-C 1 -C 7 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -NR a -、-OC(=O)-、-C(=O)O-、-NR a C (=o) -or-C (=o) NR b -optionally substituted.
In one embodiment, the alkylene of X is optionally substituted with one or more C 1 -C 6 Alkyl (e.g., methyl) or cyano substitution.
In one embodiment, X has one of the following structures:
in one embodiment, the polymer conjugated lipid compounds provided herein can be prepared by Controlled Radical Polymerization (CRP). In one embodiment, the CRP is Atom Transfer Radical Polymerization (ATRP). In one embodiment, the CRP is a reversible addition fragmentation chain transfer (RAFT) polymerization. In one embodiment, T is a polymer chain-end group or modification of a chain-end group produced by CRP. One common chain-end group from ATRP is-Br. One common chain-end group from RAFT polymerization is thiocarbonylthio. For a review of polymer chain-end modifications, see, for example, lunn et al, journal of Polymer Science, part A: polymer Chemistry 2017,55,2903-2914, the entire contents of which are incorporated herein by reference.
In one embodiment of the present invention, in one embodiment, T is hydrogen, halogen, alkyl, alkenyl, -OR ', -SR', -COOR ', -OCOR', -NR 'R', -N + (R”) 3 、-P + (R”) 3 -S-C (=s) -S-R ", -S-C (=s) -O-R", -S-C (=s) -NR "R", -S-C (=s) -aryl, cyano, azido, aryl, heteroaryl, or a targeting group, wherein each occurrence of R "is independently hydrogen or alkyl, and wherein each alkyl, alkenyl, aryl, and heteroaryl is independently optionally substituted.
In one embodiment, T is hydrogen. In one embodiment, T is bromo. In one embodiment, T is allyl. In one embodiment, T is-R ". In one embodiment, T is-S-alkyl, wherein alkyl is optionally substituted with one or more hydroxy, carboxy, or alkoxy groups. In one embodiment, T is-S-CH 2 -CH 2 -OH. In one embodiment, T is-S-CH 2 -CH 2 -COOH. In one embodiment, T is-S-CH 2 -CH 2 OMe. In one embodiment, T is-NH-alkyl, wherein alkyl is optionally substituted with one or more hydroxy, carboxy, or alkoxy groups. In one embodiment, T is-NH-CH 2 -CH 2 -OH. In one embodiment, T is-NH-CH 2 -CH 2 -COOH. In one embodiment, T is-NH-CH 2 -CH 2 -OMe。
In one embodiment, T is an azido (-N) 3 ). In one embodiment, T is optionally substituted 1,2, 3-triazole.
In one embodiment, T is-S-C (=s) -S-Et. In one embodiment, T is-S-C (=s) -S-C 12 H 25 . In one embodiment, T is-S-C (=s) -phenyl. In one embodiment, T is-S-C (=s) -N (Me) (phenyl).
In one embodiment, the targeting group specifically recognizes a molecule on the surface of a target cell, such as, for example, a cell surface receptor. Particularly suitable targeting groups include antibodies, antibody-like molecules, polypeptides, proteins (e.g., insulin-like growth factor II (IGF-II)), peptides (e.g., integrin-binding peptides, such as RGD-containing peptides), and small molecules such as, for example, sugars (e.g., lactose, galactose, N-acetylgalactosamine (GalNAc), mannose-6-phosphate (M6P)) or vitamins (e.g., folic acid). In one embodiment, the targeting group is a small molecule targeting group (e.g., folic acid). In one embodiment, the targeting group is a peptide, linker or monoclonal antibody fragment. In one embodiment, the targeting group comprises one or more N-acetylgalactosamine (GalNAc) residues.
In one embodiment, the compound is a compound represented by formula (III):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
each R 5 H, C independently 1 -C 6 Alkyl or cyano.
In one embodiment, the compound is a compound of formula (IV):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
each R 5 H, C independently 1 -C 6 Alkyl or cyano.
In one embodiment, the compound is a compound of formula (III-A):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
In one embodiment, the compound is a compound of formula (III-B):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
In one embodiment, the compound is a compound of formula (III-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment, the compound is Sup>A compound of formulSup>A (IV-Sup>A):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment, the compound is a compound of formula (IV-B):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment, the compound is a compound of formula (IV-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
each R p Independently C 1 -C 6 Alkyl or-O- (C) 1 -C 6 An alkyl group); and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment, the compound is a compound of formula (V):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
each R 5 H, C independently 1 -C 6 Alkyl or cyano.
In one embodiment, the compound is a compound of formula (VI):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
each R 5 H, C independently 1 -C 6 Alkyl or cyano.
In one embodiment, the compound is Sup>A compound of formulSup>A (V-Sup>A):
Or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
In one embodiment, the compound is a compound of formula (V-B):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
In one embodiment, the compound is a compound of formula (V-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment, the compound is a compound of formula (VI-A):
Or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment, the compound is a compound of formula (VI-B):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment, the compound is a compound of formula (VI-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
Each R p Independently C 1 -C 6 Alkyl or-O- (C) 1 -C 6 An alkyl group); and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
In one embodiment of formulSup>A (I-A), (I-B), (I-C), (II-A), (II-B), (II-C), (III-A), (III-B), (III-C), (IV-A), (IV-B), (IV-C), (V-A), (V-B), (V-C), (VI-A), (VI-B) or (VI-C), s is 1. In one embodiment, s is 2. In one embodiment, s is 3. In one embodiment, s is 4. In one embodiment, s is 5. In one embodiment, s is 6.
In one embodiment of formulSup>A (I-A), (I-B), (I-C), (II-B), (II-C), (III-A), (III-B), (III-C), (IV-B), (IV-C), (V-A), (V-B), (V-C), (VI-B) or (VI-C), t is 1. In one embodiment, t is 2. In one embodiment, t is 3. In one embodiment, t is 4. In one embodiment, t is 5. In one embodiment, t is 6.
In one embodiment of formulSup>A (I-A), (I-B), (I-C), (II-B), (II-C), (III-A), (III-B), (III-C), (IV-B), (IV-C), (V-A), (V-B), (V-C), (VI-B) or (VI-C), s is an integer from 1 to 3 and t is an integer from 1 to 3. In one embodiment, s is 1 and t is 1. In one embodiment, s is 1 and t is 2. In one embodiment, s is 1 and t is 3. In one embodiment, s is 2 and t is 1. In one embodiment, s is 2 and t is 2. In one embodiment, s is 2 and t is 3. In one embodiment, s is 3 and t is 1. In one embodiment, s is 3 and t is 2. In one embodiment, s is 3 and t is 3.
In one embodiment of formulSup>A (I-A), (III-A) or (V-A), s is 2 and t is 1.
In one embodiment of formula (I-B), (III-B) or (V-B), s is 2 and t is 3.
In one embodiment of formula (I-C), (III-C) or (V-C), s is 2 and t is 2.
In one embodiment of formula (II-B), (IV-B) or (VI-B), s is 2 and t is 1.
In one embodiment of formula (II-C), (IV-C) or (VI-C), s is 2 and t is 2.
In one embodiment, each R o Independently C 1 -C 6 An alkyl group. In one embodiment, each R o Independently C 1 -C 3 An alkyl group. In one embodiment, all R o Is methyl. In one embodiment, all R o Is ethyl. In one embodiment, two R' s o Together with the nitrogen to which they are attached form a ring moiety. In one embodiment, 3R o Together with the nitrogen to which they are attached form a bicyclic moiety.
In one embodiment, R p Is C 1 -C 6 An alkyl group. In one embodiment, R p Is C 1 -C 3 An alkyl group. In one embodiment, R p Is methyl. In one embodiment, R p Is ethyl.
In one embodiment, R p is-O- (C) 1 -C 6 Alkyl). In one embodiment, R p is-O- (C) 1 -C 3 Alkyl). In one embodiment, R p Is methoxy. In one embodiment, R p Is ethoxy.
In one embodiment, R 1 And R is 2 Each independently is a straight chain C 6 -C 24 Alkyl, branched C 6 -C 24 Alkyl or straight-chain C 6 -C 24 Alkenyl groups.
In one embodiment, R 1 And R is 2 Each independently is a straight chain C 6 -C 18 Alkyl, -R 7 -CH(R 8 )(R 9 ) Or C 6 -C 18 Alkenyl, wherein R is 7 Is C 0 -C 5 Alkylene group, and R 8 And R is 9 Independently C 2 -C 10 An alkyl group.
In one embodiment, R 1 And R is 2 Each independently is a straight chain C 7 -C 15 Alkyl or-R 7 -CH(R 8 )(R 9 ) Wherein R is 7 Is C 0 -C 1 Alkylene group, and R 8 And R is 9 Independently C 4 -C 8 An alkyl group.
In one embodiment, R 1 And R is 2 Each independently is a straight chain C 6 -C 24 An alkyl group. In one embodiment, R 1 And R is 2 Each independently is a straight chain C 7 -C 15 An alkyl group. In one embodiment, R 1 And R is 2 Each independently is a straight chain C 7 An alkyl group. In one embodiment, R 1 And R is 2 Each independently is a straight chain C 9 An alkyl group. In one embodiment, R 1 And R is 2 Each independently is a straight chain C 11 An alkyl group. In one embodiment, R 1 And R is 2 Each independently is a straight chain C 13 An alkyl group. In one placeIn one embodiment, R 1 And R is 2 Each independently is a straight chain C 15 An alkyl group.
In one embodiment, R 1 And R is 2 Each independently is a branched chain C 6 -C 24 Alkyl or branched C 6 -C 24 Alkenyl groups.
In one embodiment, R 1 And R is 2 Each independently is-R 7 -CH(R 8 )(R 9 ) Wherein R is 7 Is C 1 -C 5 Alkylene group, and R 8 And R is 9 Independently C 2 -C 10 Alkyl or C 2 -C 10 Alkenyl groups.
In one embodiment, R 1 And R is 2 Each independently is a straight chain C 6 -C 24 Alkenyl groups. In one embodiment, R 1 And R is 2 Each independently is a straight chain C 6 -C 18 Alkenyl groups. In one embodiment, R 1 And R is 2 Each independently is a straight chain C 17 Alkenyl groups.
In one embodiment, R 1 Or R is 2 Or both independently have one of the following structures:
in one embodiment, R a And R is d Each independently H.
In one embodiment, R b 、R c 、R e And R is f Each independently is n-hexyl or n-octyl.
In one embodiment, R 3 H. In one embodiment, R 3 Is C 1 -C 6 An alkyl group. In one embodiment, R 3 Is C 1 -C 4 An alkyl group. In one embodiment, R 3 Is methyl. In one embodiment, R 3 Is ethyl. In one embodiment, R 3 Is n-propyl. In one embodiment, R 3 Is isopropyl. In one embodiment, R 3 Is n-butyl. In one embodiment, R 3 Is n-amyl. In one embodiment, R 3 Is n-hexyl.
In one embodiment, n is an integer from 2 to 100. In one embodiment, n is an integer from 2 to 50. In one embodiment, n is an integer from 5 to 20. In one embodiment, n is 5. In one embodiment, n is 10. In one embodiment, n is 15. In one embodiment, n is 20.
In one embodiment, the compound is a compound of table 1 or a pharmaceutically acceptable salt, prodrug, or stereoisomer thereof.
Table 1.
/>
/>
In one embodiment, the compound is a compound of table 2, or a pharmaceutically acceptable salt, prodrug, or stereoisomer thereof.
Table 2.
/>
It is to be understood that any embodiment of the compounds provided herein as described above, as well as any particular substituent and/or variable of the compounds provided herein as described above, may be independently combined with other embodiments and/or substituents and/or various variables of the compounds to form an embodiment not specifically set forth. In addition, where a list of substituents and/or variables is listed for any particular group or variable, it is to be understood that each individual substituent and/or variable may be deleted from a particular embodiment and/or claim and that the remaining list of substituents and/or variables is to be considered within the scope of embodiments provided herein.
It is to be understood that in this specification, it is only permissible if combinations of substituents and/or variables of the described formulae have such that the compounds are stable.
7.4 nanoparticle compositions
In one aspect, described herein are nanoparticle compositions comprising the lipid compounds described herein. In certain embodiments, the nanoparticle composition comprises a compound according to formula (I) or (II) (and sub-formulae thereof) described herein.
In one embodiment, provided herein are compositions comprising a compound provided herein (e.g., a compound according to formula (I) or (II) (and sub-formulae thereof)) and a therapeutic or prophylactic agent.
In one embodiment, the composition further comprises one or more cationic lipids. In one embodiment, the molar ratio of cationic lipid to compound is from about 100:1 to about 20:1.
In one embodiment, the composition further comprises one or more neutral lipids. In one embodiment, the one or more neutral lipids is DSPC. In one embodiment, the molar ratio of cationic lipid to neutral lipid is from about 2:1 to about 8:1.
In one embodiment, the composition further comprises a steroid. In one embodiment, the steroid is cholesterol. In one embodiment, the molar ratio of cationic lipid to steroid is in the range of about 5:1 to about 1:1.
In one embodiment, the therapeutic or prophylactic agent comprises at least one mRNA encoding an antigen or fragment or epitope thereof. In one embodiment, the mRNA is a monocistronic mRNA. In one embodiment, the mRNA is a polycistronic mRNA. In one embodiment, the antigen is a pathogenic antigen. In one embodiment, the antigen is a tumor-associated antigen. In one embodiment, the mRNA comprises one or more functional nucleotide analogs. In one embodiment, the functional nucleotide analog is one or more selected from the group consisting of pseudouridine, 1-methyl-pseudouridine, and 5-methylcytosine.
In one embodiment, the composition is a nanoparticle. In one embodiment, provided herein are lipid nanoparticles comprising a compound provided herein (e.g., a compound according to formula (I) or (II) (and sub-formulae thereof)) or a composition provided herein. In one embodiment, provided herein are pharmaceutical compositions comprising a compound provided herein (e.g., a compound according to formula (I) or (II) (and sub-formulae thereof)), a composition provided herein, or a lipid nanoparticle provided herein, and a pharmaceutically acceptable excipient or diluent.
In some embodiments, nanoparticle compositions provided herein have a maximum dimension of 1 μm or less (e.g., 1 μm, 900nm, 800nm, 700nm, 600nm, 500nm, 400nm, 300nm, 200nm, 175nm, 150nm, 125nm, 100nm, 75nm, 50nm or less) when measured by Dynamic Light Scattering (DLS), transmission electron microscopy, scanning electron microscopy, or other methods. In an embodiment, at least one dimension of the lipid nanoparticle provided herein has a size in the range of about 40 to about 200 nm. In one embodiment, at least one dimension is in the range of about 40 to about 100 nm.
Nanoparticle compositions that can be used in connection with the present invention include Lipid Nanoparticles (LNP), nanolipoprotein particles, liposomes, lipid vesicles, lipid complexes, and the like. In some embodiments, the nanoparticle composition comprises one or more lipid bilayer vesicles. In some embodiments, the nanoparticle composition comprises two or more concentric bilayers separated by an aqueous compartment. Lipid bilayers can be functionalized and/or crosslinked to each other. The lipid bilayer may include one or more ligands, proteins, or channels.
The characteristics of the nanoparticle composition may depend on its components. For example, nanoparticle compositions comprising cholesterol as a structural lipid may have different properties than nanoparticle compositions comprising a different structural lipid. Similarly, the characteristics of a nanoparticle composition may depend on the absolute or relative amounts of its components. For example, nanoparticle compositions comprising a higher mole fraction of phospholipids may have different properties than nanoparticle compositions comprising a lower mole fraction of phospholipids. The characteristics may also vary depending on the method and conditions of preparation of the nanoparticle composition.
Nanoparticle compositions can be characterized by a variety of methods. For example, a microscope (transmission electron microscope or scanning electron microscope, etc.) can be used to detect morphology and size distribution of the nanoparticle composition. Dynamic light scattering or potentiometry (e.g., potentiometry) can be used to measure zeta potential. Dynamic light scattering can also be used to determine particle size. Instruments such as Zetasizer Nano ZS (Malvem Instruments Ltd, malvem, and Worcestershire, UK) can also be used to measure various characteristics of nanoparticle compositions, such as particle size, polydispersity index, and zeta potential.
Dh (size): the average size of the nanoparticle composition may be between 10s nm and 100s nm. For example, the average size may be about 40nm to about 150nm, e.g., about 40nm,45nm,50nm,55nm,60nm,65nm,70nm,75nm,80nm,85nm,90nm,95nm,100nm,105nm,110nm,115nm,120nm,125nm,130nm,135nm,140nm,145nm, or 150nm. In some embodiments, the nanoparticle composition can have an average size of about 50nm to about 100nm, about 50nm to about 90nm, about 50nm to about 80nm, about 50nm to about 70nm, about 50nm to about 60nm, about 60nm to about 100nm, about 60nm to about 90nm, about 60nm to about 80nm, about 60nm to about 70nm, about 70nm to about 70nm 100nm, about 70nm to about 90nm, about 70nm to about 80nm, about 80nm to about 100nm, about 80nm to about 90nm, or about 90nm to about 100nm. In certain embodiments, the nanoparticle composition can have an average size of about 70nm to about 100nm. In some embodiments, the average size may be about 80nm. In other embodiments, the average size may be about 100nm.
PDI: the composition of the nanoparticles may be relatively uniform. The polydispersity index may be used to indicate the uniformity of the nanoparticle composition, e.g., the particle size distribution of the nanoparticle composition. A small (e.g., less than 0.3) polydispersity index generally indicates a narrow particle size distribution. The nanoparticle composition can have a polydispersity index of about 0 to about 0.25, such as 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17,0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, or 0.25. In some embodiments, the nanoparticle composition can have a polydispersity index of about 0.10 to about 0.20.
Encapsulation efficiency: the encapsulation efficiency of the therapeutic and/or prophylactic agent means the ratio of the amount of therapeutic and/or prophylactic agent that is encapsulated or combined with the nanoparticle composition after preparation relative to the amount originally provided. High encapsulation efficiency (e.g., near 100%) is desirable. Encapsulation efficiency can be measured by comparing the amount of therapeutic and/or prophylactic agent comprising the nanoparticle composition before decomposition of the nanoparticle composition with one or more organic solvents or detergents, and after decomposition in solution. Fluorescence can be used to measure the amount of free therapeutic and/or prophylactic agent (e.g., RNA) in a solution. For nanoparticle compositions described herein, the encapsulation efficiency of the therapeutic and/or prophylactic agent can be at least 50%, such as 50%,55%,60%,65%,70%,75%,80%,85%,90%,91%,92%,93%,94%,95%,96%,97%,98%,99% or 100%. In some embodiments, the packaging efficiency may be at least 80%. In certain embodiments, the packaging efficiency may be at least 90%.
Apparent pKa: the zeta potential of the nanoparticle composition can be used to indicate the electromotive force of the composition. For example, the zeta potential may describe the surface charge of the nanoparticle composition. For nanoparticle compositions, it is generally desirable to have a relatively low positive or negative charge, as higher charge materials may interact poorly with cells, tissues, and other elements of the human body. In some embodiments, the zeta potential of the nanoparticle composition may be from about-10 mV to about +20mV, from about-10 mV to about +15mV, from about-10 mV to about +10mV, about-10 mV. To about +5mV, about-10 mV to about 0mV, about-10 mV to about-5 mV, about-5 mV to about +20mV, about-5 mV to about +15mV, about-5 mV to about +10mV, about-5 mV to about +5mV, about-5 mV to about 0mV, about 0mV to about +20mV, about 0mV to about +15mV, about 0mV to about +10mV, about 0mV to about +5mV, about +5mV to about +20mV, about +5mV to about +15mV or about +5mV to about +10mV.
In another embodiment, self-replicating RNA can be formulated in liposomes. As a non-limiting example, self-replicating RNA can be formulated in liposomes as described in international publication No. WO20120067378, which is incorporated herein by reference in its entirety. In one aspect, the liposome may comprise a lipid that facilitates delivery of pKa values to the mRNA. In another aspect, liposomes can have a substantially neutral surface charge at physiological pH and thus can be effectively used for immunization (see, e.g., liposomes described in international publication No. WO20120067378, which is incorporated herein by reference in its entirety).
In some embodiments, the nanoparticle composition comprises a polymer-conjugated lipid component comprising at least one polymer-conjugated lipid, such as a compound according to formula (I) or (II) (and sub-formulae thereof) described herein. For example, in some embodiments, nanoparticle compositions can include a polymer conjugated lipid component that includes one of the compounds provided herein. The nanoparticle composition may also include one or more other lipid or non-lipid components as described below.
7.4.1 cationic/ionizable lipid
As described herein, in some embodiments, nanoparticle compositions provided herein comprise one or more charged or ionizable lipids in addition to the polymer conjugated lipid according to formula (I) or (II) (and sub-formulae thereof). It is contemplated that certain charged or zwitterionic lipid components of the nanoparticle composition are similar to the lipid components in the cell membrane, and thus may improve cellular uptake of the nanoparticle. Exemplary charged or ionizable lipids that may form part of the nanoparticle compositions of the present invention include, but are not limited to, 3- (didodecylamino) -N1, N1, 4-trilauryl-1-piperazineethylamine (KL 10), N1- [2- (didodecylamino) ethyl ] -N1, N4, N4-trilauryl-1, 4-piperazinediethylamine (KL 22), 14, 25-ditridecyl-15,18,21,24-tetraaza-trioctadecyl (KL 25), 1, 2-dioleoyloxy-N, N-dimethylaminopropane (DLinDMA), 2-dioleyl-4-dimethylaminomethyl- [1,3] -dioxolane (DLin-K-DMA), thirty-heptadecyl-6,9,28,31-tetraen-19-ester (DLin-MC 3-DMA), 2-dioleoyl-4- (2-dimethylaminoethyl) - [1,3] -dioxolane (DLin-KC-2, 2-dioleoyl-N-2-dioleyloxy) -2- [ (dioleyl-4-dimethylaminomethyl ] -2- (. Beta. -2-dioleyl) -2- [ (2-dioleyl-N-3-DMA), n-dimethyl-3 [ (9Z, 12Z) -octadec-9, 12-dien-1-yloxy ] propan-1-amine (octyl-CLinDMA), (2R) -2- ({ 8- [ (3Z-, 12Z) -cholest-5-en-3-yloxy ] octyl } oxy) -N, N-dimethyl-3- [ [ (9Z, 12Z) -octadec-9, 12-dien-1-yloxy ] propan-1-amine (octyl-CLinDMA (2R)), (2S) -2- ({ 8- [ (3Z-, 12Z) -cholest-5-en-3-yloxy ] octyl } oxy) -N, N-dimethyl-3- [ ((9Z-, 12Z) -octadec-9, 12-dien-1-yloxy ] propan-1-amine (octyl-CLinDMA (2S)), (12Z, 15Z) -N-dimethyl-2-nonyldien-12, 15-dien-1-amine, additional exemplary charged or ionizable lipids that may form part of the nanoparticle compositions of the present invention, such as lipid 5, include Sabnis et al, "A Novel Am ino Lipid Series for mRNA Delivery: improved Endosomal Escape and Sustained Pharmacology and Safety in Non-human matrices", volume Molecular Therapy, volume 26, phase 6, 2018, described in (1 s,2 r) -2-octylcyclopropyl } heptadecan-8-amine.
In some embodiments, suitable cationic lipids include N- [1- (2, 3-dioleyloxy) propyl]-N, N-trimethylammonium chloride (DOTMA); n- [1- (2, 3-dioleoyloxy) propyl]-N, N-trimethylammonium chloride (DOTAP); 1, 2-dioleoyl-sn-glycero-3-ethylcholine phosphate (DOEPC); 1, 2-dilauryl-sn-glycero-3-ethylcholine phosphate (DLEPC); 1, 2-dimyristoyl-sn-glycero-3-ethylcholine phosphate (DMEPC); 1, 2-dimyristoyl-sn-glycero-3-ethylcholine phosphate (14:1); n1- [2- ((1S) -1- [ (3-aminopropyl) amino)]-4- [ bis (3)-amino-propyl) amino group]Butyl carboxamide) ethyl]-3, 4-bis [ oleyloxy ]]-benzamide (MVL 5); dioctadecylamino-glycyl spermine (DOGS); 3b- [ N- (N ', N' -dimethylaminoethyl) carbamoyl]Cholesterol (DC-Chol); dioctadecyl Dimethyl Ammonium Bromide (DDAB); SAINT-2, N-methyl-4- (dioleenyl) picoline; 1, 2-dimyristoxypropyl-3-dimethylhydroxyethyl ammonium bromide (dmrii); 1, 2-dioleoyl-3-dimethyl-hydroxyethyl ammonium bromide (dorrie); 1, 2-dioleoyloxypropyl-3-dimethylhydroxyethyl ammonium chloride (DORI); di-alkylated amino acids (DILA) 2 ) (e.g., C18:1-norArg-C16); dioleyldimethyl Ammonium Chloride (DODAC); 1-palmitoyl-2-oleoyl-sn-glycero-3-ethylcholine phosphate (poe pc); 1, 2-dimyristoyl-sn-glycero-3-ethylcholine phosphate (MOEPC); (R) -5- (dimethylamino) pentane-1, 2-diyl dioleate hydrochloride (DODAPEN-Cl); (R) -5-guanidinopentane-1, 2-diyldioleate hydrochloride (DOPen-G); and (R) -N, N, N-trimethyl-4, 5-bis (oleoyloxy) penta-1-ammonium chloride (DOTAPen). Cationic lipids having head groups charged at physiological pH, such as primary amines (e.g., DODAG N ', N' -dioctadecyl-N-4, 8-diaza-10-aminodecanoyl glycine amide) and guanidinium head groups (e.g., bis-guanidinium-spermidine-cholesterol (BGSC), bis-guanidyl amine-cholesterol (BGTC), PONA, and (R) -5-guanidinium pentane-1, 2-diyldioleate hydrochloride (DOPen-G)) are also suitable. Another suitable cationic lipid is (R) -5- (dimethylamino) pentane-1, 2-diyldioleate hydrochloride (DODAPEN-Cl). In certain embodiments, the cationic lipid is in a particular enantiomer or racemic form, and includes various salt forms (e.g., chloride or sulfate) of the cationic lipid as above. For example, in some embodiments, the cationic lipid is N- [1- (2, 3-dioleoyloxy) propyl ]-N, N, N-trimethylammonium chloride (DOTAP-Cl) or N- [1- (2, 3-dioleoyloxy) propyl group]-N, N-trimethylammonium sulfate (DOTAP-sulfate). In some embodiments, the cationic lipid is an ionizable cationic lipid, such as, for example, dioctadecyl Dimethyl Ammonium Bromide (DDAB); 1, 2-dioleyloxy-3-dimethylaminopropane (DLinDMA); 2, 2-Dilinoleyl-4- (2-dimethylaminoethyl)Radical) - [1,3 ]]-dioxolane (DLin-KC 2-DMA); hepta-triacontan-6,9,28,31-tetraen-19-yl 4- (dimethylamino) butyrate (DLin-MC 3-DMA); 1, 2-dioleoyloxy-3-dimethylaminopropane (DODAP); 1, 2-dioleyloxy-3-dimethylaminopropane (DODMA); and morpholino cholesterol (Mo-CHOL). In certain embodiments, the lipid nanoparticle comprises a combination of two or more cationic lipids (e.g., two or more cationic lipids as above).
Additionally, in some embodiments, the charged or ionizable lipid that may form part of the present nanoparticle compositions is a lipid that includes a cyclic amine group. Additional cationic lipids suitable for use in the formulations and methods disclosed herein include those described in WO2015199952, WO2016176330 and WO2015011633, the entire contents of which are incorporated herein by reference in their entirety.
7.4.2 Polymer conjugated lipids
In some embodiments, the lipid component of the nanoparticle composition may include one or more additional polymer-conjugated lipids (polymer-conjugated lipids), such as pegylated lipids (PEG lipids), in addition to the polymer-conjugated lipids of formula (I) or (II) (and sub-formulae thereof) described herein. It is expected that the polymer conjugated lipid component in the nanoparticle composition may improve colloidal stability and/or reduce protein absorption by the nanoparticle. Exemplary polymer conjugated lipids that can be used in connection with the present disclosure include, but are not limited to, PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramide, PEG-modified dialkylamine, PEG-modified diacylglycerol, PEG-modified dialkylglycerol, and mixtures thereof. For example, the PEG lipid may be PEG-c-DOMG, PEG-DMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, PEG-DSPE, ceramide-PEG 2000 or Chol-PEG2000.
In one embodiment, the additional polymer conjugated lipid is a pegylated lipid. Some embodiments include pegylated diacylglycerols (PEG-DAG), such as 1- (monomethoxy-polyethylene glycol) -2, 3-dimyristoylglycerol (PEG-DMG), pegylated phosphatidylethanolamine (PEG-PE), PEG succinic diacylglycerols (PEG-S-DAG), such as 4-O- (2 ',3' -di (tetradecanoyloxy) propyl-1-O- (omega-methoxy (polyethoxy) ethyl) succinate (PEG-S-DMG), pegylated ceramides (PEG-cer), or PEG dialkoxypropyl carbamates, such as omega-methoxy (polyethoxy) ethyl-N- (2, 3-di (tetradecyloxy) propyl) carbamate or 2, 3-di (tetracdecyloxy) propyl-N- (omega-methoxy) (polyethoxy) ethyl) carbamate.
In one embodiment, the polymer conjugated lipid is present at a molar concentration of 1.0 to 2.5%. In one embodiment, the polymer conjugated lipid is present at a molar concentration of about 1.7%. In one embodiment, the polymer conjugated lipid is present at a molar concentration of about 1.5%.
In one embodiment, the molar ratio of cationic lipid to polymer conjugated lipid is from about 35:1 to about 25:1. In one embodiment, the molar ratio of cationic lipid to polymer conjugated lipid is from about 100:1 to about 20:1.
In one embodiment, the pegylated lipid has the formula:
or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein:
R 12 and R is 13 Each independently is a linear or branched, saturated or unsaturated alkyl chain containing from 10 to 30 carbon atoms, wherein the alkyl chain is optionally interrupted by one or more ester linkages; and
w has an average value between 30 and 60.
In one embodiment, R 12 And R is 13 Each independently is a straight saturated alkyl chain containing from 12 to 16 carbon atoms. In other embodiments, w averages in the range of 42 to 55, e.g., w averages 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, or 55. In a particular embodiment, the average w is about 49.
In one embodiment, the pegylated lipid has the formula:
wherein w has an average value of about 49.
7.4.3 structural lipids
In some embodiments, the lipid component of the nanoparticle composition may include one or more structural lipids. It is contemplated that the structural lipid may stabilize the amphiphilic structure of the nanoparticle, such as, but not limited to, the lipid bilayer structure of the nanoparticle. Exemplary structural lipids that can be used in connection with the present disclosure include, but are not limited to, cholesterol, non-sterols, sitosterols, ergosterols, campesterols, stigmasterols, brassicasterol, lycorine, ursolic acid, alpha-tocopherol, and mixtures thereof. In certain embodiments, the structural lipid is cholesterol. In some embodiments, the structural lipids include cholesterol and corticosteroids (e.g., prednisolone, dexamethasone, prednisone, and hydrocortisone) or combinations thereof.
In one embodiment, the lipid nanoparticle provided herein comprises a steroid or steroid analog. In one embodiment, the steroid or steroid analog is cholesterol. In one embodiment, the steroid is present in a molar concentration range of 39-49%,40-46%,40-44%,40-42%,42-44%, or 44-46%. In one embodiment, the steroid is present at a molar concentration of 40, 41, 42, 43, 44, 45, or 46%.
In one embodiment, the molar ratio of cationic lipid to steroid is 1.0:0.9 to 1.0:1.2, or 1.0:1.0 to 1.0:1.2. in one embodiment, the molar ratio of cationic lipid to cholesterol is from about 5:1 to 1:1. In one embodiment, the steroid is present at a molar concentration of 32-40% of the steroid.
7.4.4 phospholipids
In some embodiments, the lipid component of the nanoparticle composition may include one or more phospholipids, such as one or more (poly) unsaturated lipids. It is contemplated that phospholipids may be assembled into one or more lipid bilayer structures. Exemplary phospholipids that may form part of the present nanoparticle compositions include, but are not limited to, 1, 2-distearoyl-sn-glycero-3-phosphorylcholine (DSPC), 1, 2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPC), 1, 2-dioleoyl-sn-glycero-3-phosphorylcholine (DLPC), 1, 2-dimyristoyl-sn-glycero-phosphorylcholine (DMPC), 1, 2-dioleoyl-sn-glycero-3-phosphorylcholine (DOPC), 1, 2-dipalmitoyl-sn-glycero-3-phosphorylcholine (DPPC), 1, 2-di-undecoyl-sn-glycero-phosphorylcholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphorylcholine (POPC), 1, 2-dioleoyl-sn-glycero-3-phosphorylcholine (18:0 diner PC), 1, 2-dioleoyl-sn-glycero-3-phosphorylcholine (dpp), 1, 2-dioleoyl-sn-glycero-3-phosphorylcholine (DPPC), 1, 2-dioleoyl-sn-glycero-3-phosphorylcholine (dapc), 1, 2-dioleoyl-glycero-2-phosphorylcholine (dicarpc), 2-oleoyl-2-phosphorylcholine (16) and 16-dioleoyl-glycero-3-phosphorylcholine (npc) 1, 2-docosahexaenoic acid-sn-glycerol-3-phosphorylcholine, 1, 2-biphytoyl-sn-glycerol-3-phosphoethanolamine (ME 16.0 PE), 1, 2-distearoyl-sn-glycerol-3-phosphoethanolamine, 1, 2-dioleoyl-1-sn-glycerol-3-phosphoethanolamine, 1, 2-dioleoyl-sn-glycerol-3-phosphoethanolamine, 1, 2-diacetarachidonoyl-sn-glycerol-3-phosphoethanolamine, 1, 2-docosahexaenoic acid-sn-glycerol-3-phosphoethanolamine, 1, 2-dioleoyl-sn-glycerol-3-phospho-rac- (1-glycerol) sodium salt (DOPG), and sphingomyelin. In certain embodiments, the nanoparticle composition comprises DSPC. In certain embodiments, the nanoparticle composition comprises DOPE. In some embodiments, the nanoparticle composition includes DSPC and DOPE.
Additional exemplary neutral lipids include dipalmitoyl phosphatidylglycerol (DPP G), palmitoyl Oleoyl Phosphatidylethanolamine (POPE), and dioleoyl phosphatidylethanolamine 4- (N-maleimidomethyl) -cyclohexane-1 carboxylate (DOPE-mal), dipalmitoyl phosphatidylethanolamine (DPPE), distearoyl phosphatidylethanolamine (DSPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, 1-stearoyl-2-oleoyl phosphatidylethanolamine (SOPE), and 1, 2-dipentamyl-sn-glycero-3-phosphate ethanolamine (transDOP E). In one embodiment, the neutral lipid is 1, 2-distearoyl-sn-glycero-3 phosphorylcholine (DSPC). In one embodiment, the neutral lipid is selected from DSPC, DPPC, DM PC, DOPC, POPC, DOPE and SM.
In one embodiment, the neutral lipid is Phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylserine (PS), phosphatidic Acid (PA) or Phosphatidylglycerol (PG).
Additional phospholipids that may form part of the nanoparticle compositions of the present invention also include those described in WO2017/112865, the entire contents of which are incorporated herein by reference.
7.4.5 therapeutic payload
Nanoparticle compositions according to the present disclosure may further comprise one or more therapeutic and/or prophylactic agents. These therapeutic and/or prophylactic agents are sometimes referred to herein as "therapeutic payloads" or "payloads". In some embodiments, the therapeutic payload may be administered in vivo or in vitro using the nanoparticle as a delivery vehicle.
In some embodiments, nanoparticle compositions comprise small molecule compounds (e.g., small molecule drugs) as therapeutic payloads, for example, antineoplastic agents (e.g., vincristine, doxorubicin, mitoxantrone, camptothecin, cisplatin, bleomycin, cyclophosphamide, methotrexate, and streptozotocin), antineoplastic agents (e.g., actinomycin D, vincristine, vinblastine, cytarabine, anthracyclines, alkylating agents, platinum compounds, antimetabolites, and nucleoside analogs such as methotrexate, purine and pyrimidine analogs), antiinfectives, local anesthetics (e.g., dibucaine and chlorpromazine), beta-adrenergic blockers (such as propranolol, timolol, and labetalol), antihypertensives (such as clonidine and hydralazine), antidepressants (such as imipramine, amitriptyline, and doxepin), anticonvulsants (such as phenytoin sodium), antihistamines (such as diphenhydramine, chlorphenamine, and promethazine), antibiotics/antiseptics (gentamicin, ciprofloxacin, and cefoxitin, etc.), antifungals (such as miconazole, terconazole, econazole, isoconazole, ding Kang, clotrimazole, itraconazole, nystatin, naftifine, and amphotericin B), antiparasitics, hormones, hormone antagonists, immunomodulators, neurotransmitter antagonists, anti-glaucoma agents, anesthetics, and imaging agents.
In some embodiments, the therapeutic payload comprises a cytotoxin, a radioactive ion, a chemotherapeutic agent, a vaccine, a compound that elicits an immune response, and/or another therapeutic and/or prophylactic agent. Cytotoxins or cytotoxic agents include any substance that may be deleterious to the cell. Examples include, but are not limited to, paclitaxel, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, teniposide, vincristine, vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxyanthraquinone, ketansetron, 1-nortestosterone, aspergillus oryzae, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, puromycin, maytansinoids, maytansinol, rapamycin (CC-1065), and analogs or homologs thereof. Radioions include, but are not limited to, iodine (e.g., iodine 125 or iodine 131), strontium 89, phosphorus, palladium, cesium, iridium, phosphate, cobalt, yttrium 90, samarium 153, and praseodymium.
In other embodiments, the therapeutic payloads of the present nanoparticle compositions may include, but are not limited to, therapeutic and/or prophylactic agents such as antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil, dacarbazine), alkylating agents (e.g., mechlorethamine, thiabendazole, rapamycin (CC-1065), melphalan, carmustine (BSNU), lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cisplatin (II) (DDP) cisplatin), anthracyclines (e.g., daunomycin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, light godomycin (AMC)) and antimitotics (e.g., vinblastine, vincristine, paclitaxel, and alkaloids).
In some embodiments, the nanoparticle composition comprises biomolecules, such as peptides and polypeptides, as a therapeutic payload. The biomolecules forming part of the nanoparticle composition may be of natural or synthetic origin. For example, in some embodiments, the therapeutic payload of the nanoparticle composition may include, but is not limited to, gentamicin, amikacin, insulin, erythropoietin (EPO), granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), factor VIR, luteinizing Hormone Releasing Hormone (LHRH) analogs, interferons, heparin, hepatitis b surface antigens, typhoid vaccines, cholera vaccines, and peptides and polypeptides.
7.4.5.1 nucleic acids
In some embodiments, the nanoparticle composition comprises one or more nucleic acid molecules (e.g., DNA or RNA molecules) as a therapeutic payload. Exemplary forms of nucleic acid molecules that may be included as therapeutic payloads in the present nanoparticle compositions include, but are not limited to, deoxyribonucleic acid (DNA), ribonucleic acid (RNA) including messenger mRNA (mRNA), and hybrid forms thereof, RNAi-inducing agents, RNAi agents, siRNA, shRNA, miRN a, antisense RNA, ribozymes, catalytic DNA, triple helix-inducing RNA, aptamers, vectors, and the like. In certain embodiments, the therapeutic payload comprises RNA. RNA molecules that may be included in the nanoparticle compositions of the present invention as therapeutic payloads include, but are not limited to: short isoforms, agonists (agomir), antagonists (antagomir), antisense molecules, ribozymes, small interfering RNAs (siRNA), asymmetric interfering RNAs (aiRNA), small microR NA (miRNA)), cleavage substrate RNAs (Dicer-substrate RNAs) (dsRNA), small hairpin RNAs (shRNA), transfer RNAs (tRNA), messenger RNAs (mRNA) and other forms of RNA molecules known in the art. In a specific embodiment, the RNA is mRNA.
In other embodiments, the nanoparticle composition comprises an siRNA molecule as a therapeutic payload. In particular, in some embodiments, the siRNA molecules are capable of selectively interfering with and down-regulating expression of a gene of interest. In some embodiments, when a nanoparticle composition comprising an siRNA is administered to a subject of administration, the siRNA payload selectively silences a gene associated with a particular disease, disorder, or condition. In some embodiments, the siRNA molecule comprises a sequence complementary to an mRNA sequence encoding a protein product of interest. In some embodiments, the siRNA molecule is an immunomodulatory siRNA.
In some embodiments, the nanoparticle composition comprises an shRNA molecule or vector encoding an shRNA molecule as a therapeutic payload. In particular, in some embodiments, the therapeutic payload produces shRNA within the target cell after administration to the target cell. Constructs and mechanisms related to shRNA are known in the art.
In some embodiments, the nanoparticle composition comprises an mRNA molecule as a therapeutic payload. In particular, in some embodiments, the mRNA molecule encodes a polypeptide of interest, including any naturally or non-naturally occurring or modified polypeptide. The polypeptide encoded by the mRNA may be of any size and may have any secondary structure or activity. In some embodiments, the polypeptide encoded by the mRNA payload may have a therapeutic effect when expressed in a cell.
In some embodiments, the nucleic acid molecules of the present disclosure comprise mRNA molecules. In particular embodiments, the nucleic acid molecule comprises at least one coding region (e.g., an Open Reading Frame (ORF)) encoding a peptide or polypeptide of interest. In some embodiments, the nucleic acid molecule further comprises at least one untranslated region (UTR). In certain embodiments, the untranslated region (UTR) is located upstream (5 'to) the coding region, referred to herein as the 5' -UTR. In certain embodiments, the untranslated region (UTR) is located downstream (3 'end) of the coding region, referred to herein as the 3' -UTR. In particular embodiments, the nucleic acid molecule comprises both a 5'-UTR and a 3' -UTR. In some embodiments, the 5'-UTR comprises a 5' -cap structure. In some embodiments, the nucleic acid molecule comprises a Kozak sequence (e.g., in the 5' -UTR). In some embodiments, the nucleic acid molecule comprises a poly-a region (e.g., in the 3' -UTR). In some embodiments, the nucleic acid molecule comprises a polyadenylation signal (e.g., in the 3' -UTR). In some embodiments, the nucleic acid molecule comprises a conserved region (as in the 3' -UTR). In some embodiments, the nucleic acid molecule comprises a secondary structure. In some embodiments, the secondary structure is a stem loop. In some embodiments, the nucleic acid molecule comprises a stem loop sequence (e.g., in the 5'-UTR and/or 3' -UTR). In some embodiments, the nucleic acid molecule comprises one or more intron regions capable of being excised during splicing. In a specific embodiment, the nucleic acid molecule comprises one or more regions selected from the group consisting of 5' -UTR and coding region. In a specific embodiment, the nucleic acid molecule comprises one or more regions selected from the group consisting of coding regions and 3' -UTRs. In a specific embodiment, the nucleic acid molecule comprises one or more regions selected from the group consisting of 5'-UTR, coding region and 3' -UTR.
Coding region
In some embodiments, the nucleic acid molecules of the present disclosure comprise at least one coding region. In some embodiments, the coding region is an Open Reading Frame (ORF) encoding a single peptide or protein. In some embodiments, the coding region comprises at least two ORFs, each ORF encoding a peptide or protein. In embodiments where the coding region comprises more than one ORF, the peptides and/or proteins encoded by the ORFs may be the same or different from each other. In some embodiments, the multiple ORFs in the coding region are separated by non-coding sequences. In a particular embodiment, the non-coding sequence separating the two ORFs comprises an Internal Ribosome Entry Site (IRES).
It is contemplated that an Internal Ribosome Entry Site (IRES) can serve as the sole ribosome binding site, or as one of the multiple ribosome binding sites of an mRNA. mRNA molecules containing more than one functional ribosome binding site can encode several peptides or polypeptides (e.g., polycistronic mRNA) that are translated independently from the ribosome. Thus, in some embodiments, a nucleic acid molecule (e.g., mRNA) of the present disclosure comprises one or more Internal Ribosome Entry Sites (IRES). Examples of IRES sequences that may be used in connection with the present disclosure include, but are not limited to, sequences from polyomaviruses (such as FMDV), pest viruses (CFFV), polioviruses (PV), encephalomyocarditis viruses (ECMV), hand-foot-mouth viruses (FMDV), hepatitis C Viruses (HCV), classical Swine Fever Viruses (CSFV), murine Leukemia Viruses (MLV), simian Immunodeficiency Viruses (SIV), or paralytic viruses (CrPV).
In various embodiments, the nucleic acid molecules of the invention encode at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 peptides or proteins. The peptides and proteins encoded by the nucleic acid molecules may be the same or different. In some embodiments, the nucleic acid molecules of the present disclosure encode dipeptides (e.g., carnosine and anserine). In some embodiments, the nucleic acid molecule encodes a tripeptide. In some embodiments, the nucleic acid molecule encodes a tetrapeptide. In some embodiments, the nucleic acid molecule encodes a pentapeptide. In some embodiments, the nucleic acid molecule encodes a hexapeptide. In some embodiments, the nucleic acid molecule encodes a heptapeptide. In some embodiments, the nucleic acid molecule encodes an octapeptide. In some embodiments, the nucleic acid molecule encodes a nonapeptide. In some embodiments, the nucleic acid molecule encodes a decapeptide. In some embodiments, the nucleic acid molecule encodes a peptide or polypeptide having at least about 15 amino acids. In some embodiments, the nucleic acid molecule encodes a peptide or polypeptide having at least about 50 amino acids. In some embodiments, the nucleic acid molecule encodes a peptide or polypeptide having at least about 100 amino acids. In some embodiments, the nucleic acid molecule encodes a peptide or polypeptide having at least about 150 amino acids. In some embodiments, the nucleic acid molecule encodes a peptide or polypeptide having at least about 300 amino acids. In some embodiments, the nucleic acid molecule encodes a peptide or polypeptide having at least about 500 amino acids. In some embodiments, the nucleic acid molecule encodes a peptide or polypeptide having at least about 1000 amino acids.
In some embodiments, the nucleic acid molecules of the present disclosure are at least about 30 nucleotides (nt) in length. In some embodiments, the nucleic acid molecule is at least about 35nt in length. In some embodiments, the nucleic acid molecule is at least about 40nt in length. In some embodiments, the nucleic acid molecule is at least about 45nt in length. In some embodiments, the nucleic acid molecule is at least about 50nt in length. In some embodiments, the nucleic acid molecule is at least about 55nt in length. In some embodiments, the nucleic acid molecule is at least about 60nt in length. In some embodiments, the nucleic acid molecule is at least about 65nt in length. In some embodiments, the nucleic acid molecule is at least about 70nt in length. In some embodiments, the nucleic acid molecule is at least about 75nt in length. In some embodiments, the nucleic acid molecule is at least about 80nt in length. In some embodiments, the nucleic acid molecule is at least about 85nt in length. In some embodiments, the nucleic acid molecule is at least about 90nt in length. In some embodiments, the nucleic acid molecule is at least about 95nt in length. In some embodiments, the nucleic acid molecule is at least about 100nt in length. In some embodiments, the nucleic acid molecule is at least about 120nt in length. In some embodiments, the nucleic acid molecule is at least about 140nt in length. In some embodiments, the nucleic acid molecule is at least about 160nt in length. In some embodiments, the nucleic acid molecule is at least about 180nt in length. In some embodiments, the nucleic acid molecule is at least about 200nt in length. In some embodiments, the nucleic acid molecule is at least about 250nt in length. In some embodiments, the nucleic acid molecule is at least about 300nt in length. In some embodiments, the nucleic acid molecule is at least about 400nt in length. In some embodiments, the nucleic acid molecule is at least about 500nt in length. In some embodiments, the nucleic acid molecule is at least about 600nt in length. In some embodiments, the nucleic acid molecule is at least about 700nt in length. In some embodiments, the nucleic acid molecule is at least about 800nt in length. In some embodiments, the nucleic acid molecule is at least about 900nt in length. In some embodiments, the nucleic acid molecule is at least about 1000nt in length. In some embodiments, the nucleic acid molecule is at least about 1100nt in length. In some embodiments, the nucleic acid molecule is at least about 1200nt in length. In some embodiments, the nucleic acid molecule is at least about 1300nt in length. In some embodiments, the nucleic acid molecule is at least about 1400nt in length. In some embodiments, the nucleic acid molecule is at least about 1500nt in length. In some embodiments, the nucleic acid molecule is at least about 1600nt in length. In some embodiments, the nucleic acid molecule is at least about 1700nt in length. In some embodiments, the nucleic acid molecule is at least about 1800nt in length. In some embodiments, the nucleic acid molecule is at least about 1900nt in length. In some embodiments, the nucleic acid molecule is at least about 2000nt in length. In some embodiments, the nucleic acid molecule is at least about 2500nt in length. In some embodiments, the nucleic acid molecule is at least about 3000nt in length. In some embodiments, the nucleic acid molecule is at least about 3500nt in length. In some embodiments, the nucleic acid molecule is at least about 4000nt in length. In some embodiments, the nucleic acid molecule is at least about 4500nt in length. In some embodiments, the nucleic acid molecule is at least about 5000nt in length.
In certain embodiments, the therapeutic payload comprises a vaccine composition (e.g., a genetic vaccine) as described herein. In some embodiments, the therapeutic payload comprises a compound capable of eliciting immunity against one or more target conditions or diseases. In some embodiments, the symptom of interest is associated with a pathogen, such as coronavirus (e.g., 2019-nCoV), influenza, measles, human Papilloma Virus (HPV), rabies, meningitis, pertussis, tetanus, plague, hepatitis, and tuberculosis, or an infection caused by the same. In some embodiments, the therapeutic payload comprises a nucleic acid sequence (e.g., mRNA) encoding a pathogen protein or an antigenic fragment or epitope thereof that is characteristic of the pathogen. The vaccine, upon inoculation into a subject, expresses the encoded pathogen protein (or antigenic fragment or epitope thereof) thereby eliciting immunity against the pathogen in the subject.
In some embodiments, the target disorder is associated with or caused by neoplastic growth of a cell, such as cancer. In some embodiments, the therapeutic payload comprises a nucleic acid sequence (e.g., mRNA) encoding a tumor-associated antigen (TAA) characteristic of cancer or an antigenic fragment or epitope thereof. The vaccine, upon administration to a vaccinated subject, expresses the encoded TAA (or an antigenic fragment or epitope thereof) thereby eliciting immunity in the subject against tumor cells that express the TAA.
5' -cap structure
It is expected that the 5' -cap structure of the polynucleotide is involved in nuclear export and enhances polynucleotide stability and binds to mRNA Cap Binding Protein (CBP) responsible for polynucleotide stability in cells. The translational capacity is obtained by the binding of CBP to poly-a binding protein to form mature circular mRNA. The 5 '-cap structure further assists in the removal of the 5' intron during mRNA splicing. Thus, in some embodiments, the nucleic acid molecules of the present disclosure comprise a 5' -cap.
The nucleic acid molecule may be capped at the 5 'end by the endogenous transcription machinery of the cell, thereby creating a 5' -ppp-5 '-triphosphate bond between the guanine cap terminal residue and the 5' transcribed sense nucleotide of the polynucleotide. This 5' -guanylate cap is then methylated to produce an N7-methyl-guanylate residue. The ribose of the nucleotide transcribed from the 5 '-end and/or the pre-end of the polynucleotide may also optionally be 2' -O-methylated. 5' -uncapping by hydrolysis and cleavage of guanylate cap structures can target nucleic acid molecules, such as mRNA molecules, for degradation.
In some embodiments, the nucleic acid molecules of the present disclosure comprise one or more alterations in the native 5' -cap structure resulting from endogenous processes. Modification of the 5' -cap can increase the stability of the polynucleotide, increase the half-life of the polynucleotide, and can increase the translation efficiency of the polynucleotide.
Exemplary alterations to the native 5' -cap structure include the creation of a non-hydrolyzable cap structure, thereby preventing uncapping and increasing the half-life of the polynucleotide. In some embodiments, since hydrolysis of the cap structure requires cleavage of the 5'-ppp-5' phosphodiester linkage, modified nucleotides may be used during the capping reaction in some embodiments. For example, in some embodiments, vaccinia capping enzyme from New England Biolabs can be used with an α -thioguanosine nucleotide to produce a phosphorothioate linkage in 5' -ppp-5' according to the manufacturer's instructions. Other modified guanosine nucleotides, such as alpha-methylphosphonate and selenophosphate nucleotides, may also be used.
Other exemplary alterations of the natural 5' -cap structure also include modifications at the 2' -and/or 3' -positions of the blocked Guanosine Triphosphate (GTP), substitution of sugar epoxy (oxygen participating in the carbocycle) for a methylene moiety (CH 2), modification of the triphosphate bridge moiety of the cap structure or modification of the nucleobase (G) moiety.
Other exemplary alterations of the native 5 '-cap structure include, but are not limited to, 2' -O-methylation of the 5 '-end of the polynucleotide and/or the 2' -hydroxyl of the 5 '-end nucleic acid at ribose, a number of different 5' -cap structures of the polynucleotide (e.g., mRNA molecules) can be generated. Additional exemplary 5' -cap structures that may be used in connection with the present disclosure also include those described in international patent publication nos. WO2008127688, WO 2008016473 and WO 2011015347, the entire contents of which are incorporated herein by reference.
In various embodiments, the 5' -cap may comprise a cap analogue. Cap analogs, also referred to herein as synthetic cap analogs, chemical caps, chemical cap analogs, or structural or functional cap analogs, are chemically different from the native (i.e., endogenous, wild-type or physiological) 5' -cap structure while retaining the function of the cap. Cap analogs can be chemically (i.e., non-enzymatically) or enzymatically synthesized and/or linked to polynucleotides.
For example, an inverted cap analogue (ARCA) cap comprises two guanosine groups linked by a 5'-5' -triphosphate group, wherein one guanosine group comprises an N7-methyl group and a 3 '-O-methyl group (i.e., N7,3' -O-dimethyl-guanosine-5 '-triphosphate-5' -guanosine, m7G-3'mppp-G, which may equivalently be referred to as 3' O-Me-m7G (5 ') ppp (5') G). The other unchanged guanosine 3'-O atom is attached to the 5' -terminal nucleotide of the end-capped polynucleotide (e.g., mRNA). N7-and 3' -O-methylated guanosine provide the terminal portion of the end-capped polynucleotide (e.g., mRNA). Another exemplary cap structure is a mCAP, which is similar to ARCA, but has a 2 '-O-methyl group on guanosine (i.e., N7,2' -O-dimethyl-guanosine-5 '-triphosphate-5' -guanosine, m) 7 Gm-ppp-G)。
In some embodiments, the cap analog can be a dinucleotide cap analog. As non-limiting examples, dinucleotide cap analogs can be modified with a boronate phosphate group or a phosphoselenate group at different phosphate positions, such as in U.S. patent No.: 8,519,110, the entire contents of which are incorporated herein by reference.
In some embodiments, the cap analog can be an N7- (4-chlorophenoxyethyl) -substituted dinucleotide cap analog known in the art and/or described herein. Non-limiting examples of N7- (4-chlorophenoxyethyl) -substituted dinucleotide cap analogs include N7- (4-chlorophenoxyethyl) -G (5 ') ppp (5 ') G and N7- (4-chlorophenoxyethyl) -m3' -OG (5 ') ppp (5 ') G cap analogs (see, e.g., kore et al Bioorganic & Medicinal Chemistry 201321:4570-4574, methods of synthesizing cap analogs, the disclosure of which is incorporated herein by reference). In other embodiments, cap analogs useful in the nucleic acid molecules of the present disclosure are 4-chloro/bromophenoxyethyl analogs.
In various embodiments, the cap analogue may comprise a guanosine analogue. Useful guanosine analogs include, but are not limited to, inosine, N1-methyl-guanosine, 2' -fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido.
It is expected that up to 20% of transcripts remain uncapped, although cap analogues allow for simultaneous capping of polynucleotides in an in vitro transcription reaction. This is a structural difference from the cap analogue of the natural 5' -cap structure of polynucleotides produced from the endogenous transcription machinery of the cell, possibly resulting in reduced translation capacity and reduced cell stability.
Thus, in some embodiments, the nucleic acid molecules of the present disclosure may also be capped post-transcriptionally using enzymes to produce a more realistic 5' -cap structure. As used herein, the phrase "more realistic" refers to features that closely reflect or mimic endogenous or wild-type features, either structurally or functionally. That is, a "more authentic" characteristic represents a better endogenous, wild-type, natural, or physiological cellular function and/or structure, or a natural type, nature, or physiological characteristic that performs better than a corresponding endogenous, wild-type, or analog thereof. Non-limiting examples of more realistic 5 '-cap structures for use in conjunction with the nucleic acid molecules of the present disclosure are those with enhanced binding of cap binding proteins, increased half-life, reduced sensitivity to 5'. Reduced 5' -uncapping of the beta-endonuclease compared to synthetic 5' -cap structures known in the art (or to wild-type, natural or physiological 5' -cap structures). For example, in some embodiments, the recombinant vaccinia virus capping enzyme and the recombinant 2 '-O-methyltransferase may generate canonical 5' -5 '-triphosphates between the 5' -terminal nucleotide and guanosine cap nucleotide of a polynucleotide. The cap guanine contains N7-methylation, while the 5 '-terminal nucleotide of the polynucleotide contains a 2' -O-methyl group. This structure is called Cap1 structure. The cap results in higher translational capacity, cell stability, and reduced activation of the cell pro-inflammatory cytokines compared to, for example, other 5' cap analog structures known in the art. Other exemplary cap structures include 7mG (5 ') ppp (5 ') N, pN2p (cap 0), 7mG (5 ') ppp (5 ') NlmpNp (cap 1), 7mG (5 ') -ppp (5 ') NlmpN2mp (cap 2) and m (7) Gpppm (3) (6,6,2 ') Apm (2 ') Cpm (2) (3, 2 ') Up (cap 4).
It is contemplated that the nucleic acid molecules of the present disclosure may be capped post-transcriptionally, and that, because the process is more efficient, nearly 100% of the nucleic acid molecules may be capped.
Untranslated region (UTR)
In some embodiments, the nucleic acid molecules of the present disclosure comprise one or more untranslated regions (UTRs). In some embodiments, the UTR is located upstream of the coding region of the nucleic acid molecule, termed the 5' -UTR. In some embodiments, the UTR is located downstream of the coding region of the nucleic acid molecule, referred to as the 3' -UTR. The sequence of the UTR may be homologous or heterologous to the sequence of the coding region in the nucleic acid molecule. The nucleic acid molecule may comprise a plurality of UTRs, which may be of identical or different sequences and/or genetic origin. In accordance with the present disclosure, any portion of the UTR in a nucleic acid molecule (including none) may be codon optimized, and may independently comprise one or more different structural or chemical modifications, prior to and/or after codon optimization.
In some embodiments, a nucleic acid molecule (e.g., mRNA) of the present disclosure comprises UTR and coding regions that are homologous to each other. In other embodiments, the nucleic acid molecules (e.g., mRNA) of the present disclosure comprise UTR and coding regions that are heterologous with respect to each other. In some embodiments, to detect the activity of a UTR sequence, a nucleic acid molecule comprising a UTR and a detectable probe coding sequence may be administered in vitro (e.g., a cell or tissue culture) or in vivo (e.g., to a subject). And the effect of the UTR sequence (e.g., modulation of expression levels, cellular localization of the encoded product, or half-life of the encoded product) can be detected using methods known in the art.
In some embodiments, the UTR of a nucleic acid molecule (e.g., mRNA) of the present disclosure comprises at least one Translational Enhancer Element (TEE) that functions to increase the yield of a polypeptide or protein produced from the nucleic acid molecule. In some embodiments, the TEE is located in the 5' -UTR of the nucleic acid molecule. In other embodiments, the TEE is located at the 3' -UTR of the nucleic acid molecule. In other embodiments, at least two TEEs are located in the 5'-UTR and 3' -UTR, respectively, of a nucleic acid molecule. In some embodiments, a nucleic acid molecule (e.g., mRNA) of the present disclosure may comprise one or more copies of a TEE sequence or comprise more than one different TEE sequence. In some embodiments, different TEE sequences in a nucleic acid molecule may be homologous or heterologous to each other.
Various TEE sequences are known in the art that may be used in connection with the present disclosure. For example, in some embodiments, the TEE may be an Internal Ribosome Entry Site (IRES), HCV-IRES, or IRES element. Chappell et al Proc. Natl. Acad. Sci. USA 101:9590-9594,2004; zhou et al Proc. Natl. Acad. Sci.102:6273-6278,2005. Additional Internal Ribosome Entry Sites (IRES) that can be used in connection with the present disclosure include, but are not limited to, those described in U.S. patent No. 7,468,275, U.S. patent publication No. 2007/0048776 and U.S. patent publication No. 2011/0123410 and international patent publication No. WO2007/025008, and international patent publication No. WO2001/055369, which are incorporated herein by reference in their entireties. In some embodiments, the TEE may be that described in Wellensiek et al Genome-wide profiling of human cap-independent translation-enhancing elements, nature Methods, month 8 of 2013; 10 (8) 747-750, the contents of which are incorporated herein by reference in their entirety, complement those described in tables 1 and 2.
Additional exemplary TEEs that may be used in connection with the present disclosure include, but are not limited to, TEE sequences disclosed in U.S. patent No. 6,310,197, U.S. patent No. 6,849,405, U.S. patent No. 7,456,273, U.S. patent No. 7,183,395, U.S. patent publication No. 2009/0226470, U.S. patent publication No. 2013/0177581, U.S. patent publication No. 2007/0048776, U.S. patent publication No. 2011/012340, U.S. patent publication No. 2009/0093049, international patent publication No. WO2009/075886, international patent publication No. WO2012/009644 and international patent publication No. WO 1999/02595, international patent publication No. WO2007/025008, international patent publication No. WO2001/055371, european patent No. 2610341, european patent No. 2610340, the entire contents of which are incorporated herein by reference.
In various embodiments, a nucleic acid molecule (e.g., mRNA) of the present disclosure comprises at least one UTR comprising at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, or greater than the case of 60 TEE sequences. In some embodiments, the TEE sequence in the UTR of the nucleic acid molecule is a copy of the same TEE sequence. In other embodiments, at least two TEE sequences in the UTR of the nucleic acid molecule have different sequences. In some embodiments, a plurality of different TEE sequences are arranged in one or more repeating patterns in the UTR region of the nucleic acid molecule. For illustration purposes only, the repeating pattern may be, for example, ABABAB, ABABBAABBAABB, ABCABCABC, etc., wherein in these exemplary patterns each capital letter (a, B, or C) represents a different TEE sequence. In some embodiments, at least two TEE sequences are contiguous with each other (i.e., without a spacer sequence therebetween) in the UTR of the nucleic acid molecule. In other embodiments, at least two TEE sequences are separated by a spacer sequence. In some embodiments, UTRs may comprise a TEE sequence-spacer sequence module that is repeated at least once, at least twice, at least 3 times, at least 4 times, at least 5 times, at least 6 times, at least 7 times, at least 8 times, at least 9 times, or more than 9 times. In any of the embodiments described in this paragraph, the UTR may be the 5'-UTR, the 3' -UTR, or both the 5'-UTR and the 3' -UTR of the nucleic acid molecule.
In some embodiments, the UTR of a nucleic acid molecule (e.g., mRNA) of the present disclosure comprises at least one translational inhibiting element that functions to reduce the amount of polypeptide or protein produced from the nucleic acid molecule. In some embodiments, the UTR of the nucleic acid molecule comprises one or more miR sequences or fragments thereof (e.g., miR seed sequences) that are recognized by one or more micrornas. In some embodiments, the UTR of the nucleic acid molecule comprises one or more stem loop structures that down-regulate the translational activity of the nucleic acid molecule. Other mechanisms for inhibiting the translational activity associated with nucleic acid molecules are known in the art. In any of the embodiments described in this paragraph, the UTR may be the 5'-UTR, the 3' -UTR, or both the 5'-UTR and the 3' -UTR of the nucleic acid molecule.
Polyadenylation (poly-A) region
During natural RNA processing, long-chain adenosine nucleotide (poly-a) regions are typically added to messenger RNA (mRNA) molecules to increase the stability of the molecules. Immediately after transcription, the 3 '-end of the transcript is cleaved to release the 3' -hydroxyl group. Then, the poly-A polymerase adds an adenosine nucleotide chain to the RNA. This process is called polyadenylation and adds a poly-A region of 100 to 250 residues in length. It is contemplated that the poly-A region may confer a variety of advantages to the nucleic acid molecules of the invention.
Thus, in some embodiments, a nucleic acid molecule (e.g., mRNA) of the present disclosure comprises a polyadenylation signal. In some embodiments, a nucleic acid molecule (e.g., mRNA) of the present disclosure comprises one or more polyadenylation (poly-a) regions. In some embodiments, the poly-a region consists entirely of adenine nucleotides or functional analogs thereof. In some embodiments, the nucleic acid molecule comprises at least one poly-a region at its 3' end. In some embodiments, the nucleic acid molecule comprises at least one poly-a region at its 5' end. In some embodiments, the nucleic acid molecule comprises at least one poly-a region at its 5 'end and at least one poly-a region at its 3' end.
In accordance with the present disclosure, the poly-a regions may have varying lengths in different embodiments. In particular, in some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 30 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 35 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 40 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 45 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 50 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 55 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 60 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 65 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 70 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 75 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 80 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 85 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 90 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 95 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 100 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 110 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 120 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 130 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 140 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 150 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 160 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 170 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 180 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 190 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 200 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 225 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 250 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 275 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 300 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 350 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 400 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 450 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 500 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 600 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 700 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 800 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 900 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 1000 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 1100 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 1200 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 1300 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 1400 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 1500 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 1600 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 1700 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 1800 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 1900 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 2000 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 2250 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 2500 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 2750 nucleotides in length. In some embodiments, the poly-a region of a nucleic acid molecule of the present disclosure is at least 3000 nucleotides in length.
In some embodiments, the length of the poly-a region in a nucleic acid molecule can be selected based on the total length of the nucleic acid molecule or portion thereof (e.g., the length of the coding region or the length of the open reading frame). For example, in some embodiments, the poly-a region comprises about 5%,10%,15%,20%,25%,30%,35%,40%,45%,50%,55%,60%,65%,70%,75%,80%,85%,90%,95% or more of the total length of the nucleic acid molecule comprising the poly-a region.
It is contemplated that certain RNA binding proteins may bind to the poly-A region located at the 3' end of the mRNA molecule. These poly-a binding proteins (PABPs) can modulate mRNA expression, for example, interact with translation initiation mechanisms in cells and/or protect the 3' -poly-a tail from degradation. Thus, in some embodiments, a nucleic acid molecule (e.g., mRNA) of the present disclosure comprises at least one binding site for a poly-a binding protein (PABP). In other embodiments, the nucleic acid molecule is conjugated or complexed to the PABP prior to loading into a delivery vehicle (e.g., a lipid nanoparticle).
In some embodiments, a nucleic acid molecule (e.g., mRNA) of the present disclosure comprises a poly-A-G tetramer. G tetramers are cyclic hydrogen bond arrays of four guanosine nucleotides, which can be formed from G-rich sequences in DNA and RNA. In this embodiment, the G tetramer is attached at the end of the poly-A region. The stability of the resulting polynucleotide (e.g., mRNA), protein production, and other parameters, including half-life at various time points, can be determined. Studies have shown that the polyA-G tetramer structure produces a protein yield at least equal to 75% of the protein yield produced by the 120 nucleotide poly-a region alone.
In some embodiments, a nucleic acid molecule (e.g., mRNA) of the present disclosure may include a poly-a region, and may be stabilized by the addition of a 3' stabilizing region. In some embodiments, 3' stabilizing regions useful for stabilizing nucleic acid molecules (e.g., mRNA), including poly-a or poly-a-G tetramer structures, are described in international patent publication No. WO2013/103659, which is incorporated herein by reference in its entirety.
In other embodiments, the 3' stability region that can be used in conjunction with the nucleic acid molecules of the present disclosure includes chain terminating nucleosides such as, but not limited to, 3' -deoxyadenosine (cordic-sine), 3' -deoxyuridine, 3' -deoxycytosine, 3' -deoxyguanosine, 3' -deoxythymine, 2',3' -dideoxynucleoside, 2',3' -dideoxyadenosine, 2',3' -dideoxyuridine, 2',3' -dideoxycytosine, 2',3' -dideoxyguanosine, 2',3' -dideoxythymine, 2' -deoxynucleoside or O-methyl nucleoside, 3' -deoxynucleoside, 2',3' -dideoxynucleoside 3' -O-methyl nucleoside, 3' -O-ethyl nucleoside, 3' -arabinoside, other substituted nucleosides described herein or known in the art.
Two-stage structure
The stem loop structure can direct RNA folding, preserve the structural stability of the nucleic acid molecule (e.g., mRNA), provide recognition sites for RNA binding proteins, and serve as substrates for enzymatic reactions. For example, the integration of miR sequences and/or TEE sequences alters the shape of the stem-loop region, which may increase and/or decrease translation (Kedde et al A Pumilio-induced RNA structure switch in p-3'UTR controls miR-221and miR-222accessibility.Nat Cell Biol; 10. 2010; 12 (10): 1014-20, the contents of which are incorporated herein by reference in their entirety).
Thus, in some embodiments, a nucleic acid molecule (e.g., mRNA) or a portion thereof described herein may take the form of a stem loop structure, such as, but not limited to, a histone stem loop. In some embodiments, the stem-loop structure is formed from a stem-loop sequence of about 25 or about 26 nucleotides in length, which may be, but is not limited to, those described in international patent publication No. WO2013/103659, which is incorporated herein by reference in its entirety. Other examples of stem-loop sequences include those described in international patent publication No. WO2012/019780 and international patent publication No. WO201502667, the contents of which are incorporated herein by reference. In some embodiments, the stem-loop sequence comprises a TEE as described herein. In some embodiments, the stem-loop sequence comprises a miR sequence as described herein. In particular embodiments, the stem loop sequence may comprise a miR-122 seed sequence. In a particular embodiment, the nucleic acid molecule comprises the stem-loop sequence CAAAG GCTCTTTTCAGAGCCACCA (SEQ ID NO: 1). In other embodiments, the nucleic acid molecule comprises a stem-loop sequence CAAAGGCUCUUUUCAGAGCCACCA (SEQ ID NO: 2).
In some embodiments, a nucleic acid molecule (e.g., mRNA) of the present disclosure comprises a stem-loop sequence upstream (5' to) the coding region of the nucleic acid molecule. In some embodiments, the stem-loop sequence is located within the 5' -UTR of the nucleic acid molecule. In some embodiments, a nucleic acid molecule (e.g., mRNA) of the present disclosure comprises a stem-loop sequence downstream (3' to) the coding region of the nucleic acid molecule. In some embodiments, the stem-loop sequence is located within the 3' -UTR of the nucleic acid molecule. In some cases, the nucleic acid molecule may comprise more than one stem loop sequence. In some embodiments, the nucleic acid molecule comprises at least one stem-loop sequence in the 5'-UTR and at least one stem-loop sequence in the 3' -UTR.
In some embodiments, the nucleic acid molecule comprising a stem-loop structure further comprises a stabilizing region. In some embodiments, the stabilizing region comprises at least one chain terminating nucleoside that acts to slow degradation and thus increases the half-life of the nucleic acid molecule. Exemplary chain terminating nucleosides that can be used in connection with the present disclosure include, but are not limited to, 3 '-deoxyadenosine (cordic), 3' -deoxyuridine, 3 '-deoxycytosine, 3' -deoxyguanosine, 3 '-deoxythymine, 2',3 '-dideoxynucleoside, 2',3 '-dideoxyadenosine, 2',3 '-dideoxyuridine, 2',3 '-dideoxyguanosine, 2',3 '-dideoxythymine, 2' -deoxynucleoside or O-methyl nucleoside, 3 '-deoxynucleoside, 2',3 '-dideoxynucleoside 3' -O-methyl nucleoside, 3 '-O-ethyl nucleoside, 3' -arabinoside, other substitute nucleosides described herein or known in the art. In other embodiments, the stem-loop structure may be stabilized by altering the 3' region of the polynucleotide, which may prevent and/or inhibit the addition of oligo (U) (international patent publication No. WO2013/103659, which is incorporated herein by reference in its entirety).
In some embodiments, the nucleic acid molecules of the present disclosure comprise at least one stem-loop sequence and a poly-a region or polyadenylation signal. Non-limiting examples of polynucleotide sequences comprising at least one stem-loop sequence and a poly-a region or polyadenylation signal, including those described in international patent publication No. WO2013/120497, international patent publication No. WO2013/120629, international patent publication No. WO2013/120500, international patent No. WO2013/120627, international patent No. WO2013/120498, international patent publication No. WO2013/120626, international patent publication No. WO2013/120499, and international patent publication No. WO2013/120628, the entire contents of which are incorporated herein by reference.
In some embodiments, a nucleic acid molecule comprising a stem-loop sequence and a poly-a region or polyadenylation signal may encode a pathogen antigen or fragment thereof, as described in international patent publication No. WO2013/120499 and international patent publication No. WO2013/120628, the contents of which are incorporated herein by reference in their entirety.
In some embodiments, a nucleic acid molecule comprising a stem-loop sequence and a poly-a region or polyadenylation signal may encode a therapeutic protein, as described in international patent publication nos. WO2013/120497 and WO2013/120629, the contents of which are incorporated herein by reference in their entirety.
In some embodiments, a nucleic acid molecule comprising a stem-loop sequence and a poly-a region or polyadenylation signal may encode a tumor antigen or fragment thereof, as described in international patent publication No. WO2013/120500 and international patent publication No. WO2013/120627, the contents of which are incorporated herein by reference in their entirety.
In some embodiments, a nucleic acid molecule comprising a stem-loop sequence and a poly-a region or polyadenylation signal may encode an allergenic antigen or an autoimmune autoantigen, as described in international patent publication nos. WO2013/120498 and WO2013/120626, the contents of which are incorporated herein by reference in their entirety.
Functional nucleotide analogues
In some embodiments, a nucleic acid molecule comprising a stem-loop sequence and a poly-a region or polyadenylation signal may encode an allergenic antigen or an autoimmune autoantigen, as described in international patent publication nos. WO2013/120498 and WO2013/120626, the contents of which are incorporated herein by reference in their entirety.
Thus, in some embodiments, the payload nucleic acid molecule comprises at least one functional nucleotide analog described herein. In some embodiments, the functional nucleotide analog comprises at least one chemical modification to a nucleobase, a sugar group, and/or a phosphate group. Thus, a payload nucleic acid molecule comprising at least one functional nucleotide analogue contains at least one chemical modification to a nucleobase, a sugar group and/or a nucleoside linkage. Exemplary chemical modifications to nucleobases, glycosyls, or nucleoside linkages of nucleic acid molecules are provided herein.
As described herein, all nucleotides in the payload nucleic acid molecule can be in the range of 0% to 100% functional nucleotide analogs as described herein. For example, in various embodiments, from about 1% to about 20%, from about 1% to about 25%, from about 1% to about 50%, from about 1% to about 60%, from about 1% to about 70%, from about 1% to about 80%, from about 1% to about 90%, from about 1% to about 95%, from about 10% to about 20%, from about 10% to about 25%, from about 10% to about 50%, from about 10% to about 60%, from about 10% to about 70%, from about 10% to about 80%, from about 10% to about 90%, from about 10% to about 95%, from about 10% to about 100%, from about 20% to about 25%, from about 20% to about 50%, from about 20% to about 60%, from about 20% to about 70%, from about 20% to about 90%, from about 20% to about 95%, from about 20% to about 100%, from about 50% to about 70%, from about 50% to about 80%, from about 50% to about 90%, from about 50% to about 95%, from about 50% to about 100%, from about 70% to about 80%, from about 80% to about 95%, or from about 80% to about 95% to about 100% of the nucleoside is similar. In any of these embodiments, the functional nucleotide analog may be present at any position of the nucleic acid molecule, including the 5 '-terminus, the 3' -terminus, and/or one or more internal positions. In some embodiments, a single nucleic acid molecule may comprise different sugar modifications, different nucleobase modifications, and/or different types of nucleoside linkages (e.g., backbone structures).
As described herein, a functional nucleotide analog described herein can be found in 0% to 100% of all nucleotides of a type (e.g., all purine-containing nucleotides of a type, or all pyrimidine-containing nucleotides of a type, or "as one" in all a, G, C, T, or U ranging from 0% to 100% payload nucleic acid molecules). For example, in various embodiments, from about 1% to about 20%, from about 1% to about 25%, from about 1% to about 50%, from about 1% to about 60%, from about 1% to about 70%, from about 1% to about 80%, from about 1% to about 90%, from about 1% to about 95%, from about 10% to about 20%, from about 10% to about 25%, from about 10% to about 50%, from about 10% to about 60%, from about 10% to about 70%, from about 10% to about 80%, from about 10% to about 90%, from about 10% to about 95%, from about 10% to about 100%, from about 20% to about 25%, from about 20% to about 50%, from about 20% to about 60%, from about 20% to about 70%, about 20% to about 80%, about 20% to about 90%, about 20% to about 95%, about 20% to about 100%, about 50% to about 60%, about 50% to about 70%, about 50% to about 80%, about 50% to about 90%, about 50% to about 95%, about 50% to about 100%, about 70% to about 80%, about 70% to about 90%, about 70% to about 95%, about 70% to about 100%, about 80% to about 90%, about 80% to about 95%, about 80% to about 100%, about 90% to about 95%, about 90% to about 100%, or about 95% to about 100% are functional nucleotide analogs described herein. In any of these embodiments, the functional nucleotide analog may be present at any position of the nucleic acid molecule, including the 5 '-terminus, the 3' -terminus, and/or one or more internal positions. In some embodiments, a single nucleic acid molecule may comprise different sugar modifications, different nucleobase modifications, and/or different types of nucleoside linkages (e.g., backbone structures).
Modification of bases
In some embodiments, the functional nucleotide analog comprises a non-standard nucleobase. In some embodiments, standard nucleobases (e.g., adenine, guanine, uracil, thymine, and cytosine) in a nucleotide may be modified or substituted to provide one or more functional analogs of the nucleotide. Exemplary modifications of nucleobases include, but are not limited to, one or more substitutions or modifications including, but not limited to, alkyl, aryl, halogen, oxo, hydroxy, alkoxy, and/or thio substitutions; one or more condensed rings or ring-opening, oxidation and/or reduction.
In some embodiments, the nonstandard nucleobase is a modified uracil. Exemplary nucleobases and nucleosides with modified uracils include pseudouridine (ψ), pyridin-4-one ribonucleoside, 5-azauracil, 6-azauracil, 2-thio-5-azauracil, 2-thiouracil(s) 2 U), 4-thio-uracil(s) 4 U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxy-uracil (ho) 5 U), 5-aminoallyl-uracil, 5-halo-uracil (e.g., 5-iodo-uracil or 5-bromouracil), 3-methyluracil (m) 3 U), 5-methoxyuracil (mo) 5 U), uracil 5-oxyacetic acid (cmo) 5 U), uracil 5-oxoacetic acid methyl ester (mcmo) 5 U), 5-carboxymethyl-uracil (cm) 5 U), 1-carboxymethyl-pseudouridine, 5-carboxyhydroxymethyl-uracil (chm) 5 U), 5-carboxyhydroxymethyl-uracil methyl ester (mchm) 5 U), 5-methoxycarbonyl methyl uracil (mcm) 5 U), 5-methoxycarbonylmethyl-2-thiouracil (mcm) 5 s 2 U), 5-aminomethyl-2-thiouracil (nm) 5 s 2 U), 5-methylaminomethyl-2-uracil (mn) 5 U), 5-methylaminomethyl-2-thiouracil (mn) 5 s 2 U), 5-methylaminomethyl-2-selenouracil (mn) 5 se 2 U), 5-carbamoyl methyluracil (ncm U), 5-carboxymethyl aminomethyluracil (cmnm) 5 U), 5-carboxymethylaminomethyl-2-thiouracil (cmnm) 5 s 2 U), 5-propynyluracil, 1-propynyl-pseudouracil, 5-taurine methyl uracil (τm) 5 U), 1-taurine methyl-pseudo-uridine, 5-taurine methyl-2-thiouracil (τm) 5 s 2 U), 1-Niu Huangji methyl-4-thio-pseudouridine, 5-methyl-uracil (m) 5 U, i.e. having the nucleobase deoxythymine), 1-methyl-pseudouridine (m 1 Psi), 1-ethyl-pseudouridine (Et) 1 Psi), 5-methyl-2-thiouracil (m) 5 s 2 U), 1-methyl-4-thio-Gu Duli (m) 1 s 4 Psi), 4-thio-1-methyl-Gu Duli, 3-methyl-Gu Duli (m) 3 ψ), 2-thio-1-methyl-Du Duli, 1-methyl-1-deaza-pseudo-uridine, 2-thio-1-methyl-1-deaza-pseudo-uridine, dihydro-uracil (D), dihydro-pseudo-uridine, 5, 6-dihydro-uracil, 5-methyl-dihydro-uracil (m) 5 D) 2-thio-dihydro-uracil, 2-thio-dihydro-pseudouridine, 2-methoxy-uracil, 2-methoxy-4-thiouracil, 4-methoxy-pseudouridine, 4-methoxy-2-thio-pseudouridine, N1-methyl-pseudouridine, 3- (3-amino-3-carboxypropyl) uracil (acp) 3 U), 1-methyl-3- (3-amino-3-carboxypropyl) pseudouridine (acp) 3 Psi), 5- (isopentenyl aminomethyl) uracil (m) 5 U), 5- (isopentenyl) aminomethyl) -2-thiouracil (m) 5 s 2 U), 5,2' -O-dimethyluridine (m) 5 Um), 2-thio-2' -O-methyluridine(s) 2 Um), 5-methoxycarbonylmethyl-2' -O-methyluridine (mcm) 5 Um), 5-carbamoylmethyl-2' -O-methyluridine (ncm) 5 Um), 5-carboxymethylaminomethyl-2' -O-methyluridine (cmnm) 5 Um), 3,2' -O-dimethyluridine (m) 3 Um) and 5- (isopentenylaminomethyl) -2' -O-methyl-uridine (mm) 5 Um), 1-thio-uracil, deoxythymidine, 5- (2-carbomethoxyvinyl) -uracil, 5- (carbamoyl hydroxymethyl) -uracil, 5-carbamoylmethyl-2-thiouracil, 5-carboxymethyl-2-thiouracil, 5-cyanomethyl-uracil, 5-methoxy-2-thiouracil and 5-3- (1-E-acrylamido) uracil.
In some embodiments, the nonstandard nucleobase is a modified cytosine. Exemplary nucleobases and nucleosides having modified cytosines include 5-azacytosine, 6-azacytosine, pseudoisocytosine, 3-methylcytosine (m 3C), N4-acetylcytosine (ac 4C), 5-formylcytosine (f 5C), N4-methyl-cytosine (m 4C), 5-methyl-cytosine (m 5C), 5-halo-cytosine (e.g., 5-iodo-cytosine), 5-hydroxymethyl-cytosine (hm 5C), 1-methyl-pseudoisocytosine, pyrrolo-cytosine, pyrrolo-pseudoisocytosine, 2-thiocytosine (s 2C), 2-thio-5-methylcytosine nucleoside, 4-thio-pseudoisocytosine, 4-thio-1-methyl-azadeaza-pseudoisocytosine, 1-methyl-1-deaza-pseudoisocytosine, buzeine (buzeine), 5-methyl-5-thiozeine (buzeine), 2-thiozeine (s 2C), 2-thio-5-thiocytosine, 4-thio-1-azazeine (C), 5-thiozeine (2-thiozeine), 5-thiozeine (C) and the like, 2-methoxy-5-methylcytidine, 4-methoxy-pseudoisocytosine, 4-methoxy-1-methyl-pseudoisocytosine, lysine (k 2C), 5,2' -O-dimethylcytidine (m 5 Cm), N4-acetyl-2 ' -O-methylcytidine (ac 4 Cm), N4,2' -O-dimethylcytidine (m 4 Cm), 5-formyl-2 ' -O-methylcytidine (fSCm), N4,2' -O-trimethylcytidine (m 42 Cm), 1-thiocytosine, 5-hydroxy-cytosine, 5- (3-azidopropyl) -cytosine, and 5- (2-azidoethyl) -cytosine.
In some embodiments, the nonstandard nucleobase is a modified adenine. Exemplary nucleobases and nucleosides having substituted adenine include 2-aminopurine, 2, 6-diaminopurine, 2-amino-6-halopurine (e.g., 2-amino-6-chloropurine), 6-halopurine (e.g., 6-chloropurine), 2-amino-6-methylpurine, 8-azidoadenine, 7-deazaadenine, 7-deaza-8-azaadenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2, 6-diaminopurine, 7-deaza-8-aza-2, 6-diaminopurine, 1-methyladenine (m 1A), 2-methyladenine (m 2A), N6-methyladenine (m 6A), 2-methylthio-N6-methyladenine (ms 2m 6A), N6-isopentenyl adenine (i 6A), 2-methylthio-N6-isopentenyl adenine (ms 2i 6A), N6- (hydroxy-isopentenyl) adenine (m 6A), 2-hydroxy-5-adenine (m 6A), 2-methyladenine (m 6A), 2-methylsulfanyl-N6-methyladenine (m 6A), N6-methyl adenine (m 6A), N6-isopentenyl adenine (m 6A), N6-methyl adenine (m 6-methyl) N6-methyl-N6-Su Anjia carbamoyl-adenine (m 6t 6A), 2-methylsulfanyl-N6-Su Anjia carbamoyl-adenine (ms 2g 6A), N6-dimethyl-adenine (m 62A), N6-hydroxy-N-pentylcarbamoyl-adenine (hn 6A), 2-methylsulfanyl-N6-hydroxy-N-pentylcarbamoyl-adenine (ms 2hn 6A), N6-acetyladenine (ac 6A), 7-methyladenine, 2-methylsulfanyl-adenine, 2-methoxyadenine, N6,2' -O-dimethyladenine (m 6 Am), N6,2' -O-trimethyladenine (m 62 Am), 1,2' -O-dimethyladenine (m 1 Am), 2-amino-N6-methylpurine, 1-thioadenine, 8-azaadenine, N6- (19-amino-pentaoxadodecane) -adenine, 2, 8-dimethyl adenine, N6-O-methyladenine and N6-hydroxy-methyladenine
In some embodiments, the nonstandard nucleobase is a modified guanine. Exemplary nucleobases and nucleosides with modified guanines include inosine (I), 1-methyl inosine (m 1I), inosine (imG), methyl inosine (mimG), 4-desmethylinosine (imG-14), iso-tyrosine (imG 2), huai Dinggan (wybutosine) (yW), peroxo tyrosine (o 2 yW), hydroxy tyrosine (OHyW), under-modified hydroxy tyrosine (OHyW), 7-deazaguanine, quinine (Q), epoxy quinine (oQ), galactosyl quinine (galQ), mannosyquinine, 7-cyano-7-deazaguanine (preQO), 7-aminomethyl-7-deazaguanine (preQ 1), archaea (G+), 7-deazaguanine (6-thioguanine, 6-thio-7-deaza-guanine (OHyW), 6-thio-7-deaza-8-deaza-methyl guanine (G7-m), N-methyl guanine (N, N-2-methyl guanine (G1), N-methyl guanine (N2G 2, N-methyl guanine (2G 2), n2, 7-dimethylguanine (m 2,2,7G), 8-oxoguanine, 7-methyl-8-oxoguanine, 1-methyl-6-thioguanine, N2-dimethyl-6-thioguanine, N2-methyl-2 '-O-methyl-guanine (m 2 Gm), N2-dimethyl-2' -O-methylguanosine (m 22 Gm), 1-methyl-2 '-O-methylguanosine (m 1 Gm), N2, 7-dimethyl-2' -O-methylguanosine (m 2,7 Gm), 2 '-O-methylguanosine (Im), 1,2' -O-dimethylinosine (mIm), 1-thioguanosine and O-6-methylguanosine.
In some embodiments, the nonstandard nucleobases of the functional nucleotide analogs can independently be purines, pyrimidines, purines, or pyrimidine analogs. For example, in some embodiments, the non-canonical nucleobase can be a modified adenine, cytosine, guanine, uracil, or hypoxanthine. In other embodiments, non-canonical nucleobases can also include naturally occurring and synthetic derivatives of bases, including pyrazolo [3,4-d ] pyrimidine, 5-methylcytosine (5-me-C), 5-hydroxymethylcytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-propynyluracil and cytosine, 6-azouracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo (e.g., 8-bromo), 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxy and other 8-substituted adenine and guanine, 5-halo, particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracil and cytosine, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, deazaguanine, 7-deazaguanine, 3-deazaguanine, deazaadenosine, 7-deazaadenosine, 3-deazaadenosine, pyrazolo [3,4-d ] pyrimidine, imidazo [1,5-a ]1,3,5 triazinone, 9-deazapurine, imidazo [4,5-d ] pyrazine, thiazolo [4,5-d ] pyrimidine, pyrazin-2-one, 1,2, 4-triazine, pyridazine, or 1,3, 5-triazine.
Modification of sugar
In some embodiments, the functional nucleotide analog comprises a non-standard sugar group. In various embodiments, the non-standard sugar group may be a 5-carbon or 6-carbon sugar (e.g., pentose, ribose, arabinose, xylose, glucose, galactose, or deoxy derivatives thereof) having one or more substituents that may be halogen, hydroxy, thiol, alkyl, alkoxy, alkenyloxy, alkynyloxy, cycloalkyl, aminoalkoxy, alkoxyalkoxy, hydroxyalkoxy, amino, azido groups, aryl, aminoalkyl, aminoalkenyl, aminoalkylynyl, and the like.
Typically, RNA molecules contain a ribose group, which is a five-membered ring with oxygen. Exemplary non-limiting alternative nucleotides include substitution of oxygen in ribose (e.g., with S, se, or alkylene groups such as methylene or ethylene); addition of double bonds (e.g., substitution of ribose with cyclopentenyl or cyclohexenyl); a ribose ring (e.g., a four-membered ring that forms a cyclobutane or oxetane ring); expansion of ribose (e.g., forming 6 or 7 membered rings with additional carbon or heteroatoms, such as anhydrohexitols, arabitol, mannitol, cyclohexyl, cyclohexenyl, and morpholino (also having phosphoramidate backbones)); polycyclic forms (e.g., tricyclic and "unlocked" forms), such as ethylene Glycol Nucleic Acid (GNA) (e.g., R-GNA or S-GNA, wherein ribose is replaced with an ethylene glycol unit attached to a phosphodiester linkage), threose nucleic acid (TNA, wherein ribose is replaced with an α -L-threofuranosyl- (3 '→2') moiety) and peptide nucleic acid (PNA, wherein 2-amino-ethyl-glycine linkages replace ribose and phosphodiester backbones).
In some embodiments, the sugar group comprises one or more carbons having a stereochemical configuration opposite to the corresponding carbon in ribose. Thus, a nucleic acid molecule may comprise a nucleotide containing, for example, arabinose or L-ribose as sugar. In some embodiments, the nucleic acid molecule comprises at least one nucleoside, wherein the sugar is L-ribose, 2 '-O-methyl ribose, 2' -fluoro ribose, arabinose, hexitol, LNA, or PNA.
Modification of nucleoside bonds
In some embodiments, a payload nucleic acid molecule of the present disclosure may comprise one or more modified nucleoside linkages (e.g., phosphate backbone). The phosphate group of the backbone may be altered by substitution of one or more oxygen atoms with different substituents.
In some embodiments, the functional nucleotide analog may include another nucleoside linkage substituted for an unaltered phosphate moiety. Examples of alternative phosphate groups include, but are not limited to, phosphorothioates, selenophosphate, phosphoroborates, phosphonates, phosphoramidates, phosphorodiamidates, alkyl or aryl phosphonates and phosphotriesters. Both non-linking oxygens of the dithiophosphate are replaced by sulfur. The altered phosphate linkages can also be attached by replacing oxygen with nitrogen (bridged phosphoramidate), sulfur (bridged phosphorothioate) and carbon (bridged methylphosphonate).
Alternative nucleosides and nucleotides include a borane moiety (BH 3 ) Sulfur (thio), methyl, ethyl and/or methoxy instead of one or more non-bridging oxygens. As a non-limiting example, two non-bridged oxygens at the same position (e.g., the α, β or γ position) may be substituted with sulfur (thio) and methoxy. RNA and DNA stability (e.g., for exonucleases and endonucleases) is enhanced by substitution of one or more oxygen atoms at the position of the phosphate moiety (e.g., alpha-phosphorothioate) with an unnatural phosphorothioate backbone linkage. Phosphorothioate DNA and RNA have enhanced nuclease resistance and therefore have a longer half-life in the cellular environment.
Other nucleoside linkages used in accordance with the present disclosure include nucleoside linkages that do not include a phosphorus atom.
Other examples of nucleic acid molecules (e.g., mRNA), compositions, formulations, and/or methods related thereto that may be used in conjunction with the present disclosure further include those described in WO2002/098443, WO 2003/051401, WO2008/052770, WO 200927230, WO2006122828, WO2008/083949, WO2010088927, WO2010/037539, WO2004/004743, WO2005/016376, WO2006/024518, WO2007/095976, WO2008/014979, WO2008/077592, WO2009/030481, WO2009/095226, WO 201201069586, WO 201026641, WO 2012012012019780, WO 2013326, WO2012089338, WO 20120120120113, WO 2016811, WO 201211311350, WO 2013111, WO2013113736, WO 20131698, WO 20131699, WO 201313, WO 20131626, WO 2012012012110629, WO 20120120120131626, WO 2012012012012117, WO 2012010629, WO2015024667, WO2015/024665, WO2015/024666, WO2015/024664, WO 20151101415, WO 20151101414, WO 201514667, WO 201515538, WO 20151101416, the contents of each of which are incorporated herein in their entirety.
7.5 dosage forms
According to the present disclosure, nanoparticle compositions described herein can comprise at least one lipid component and one or more other components, such as therapeutic and/or prophylactic agents. Nanoparticle compositions can be designed for one or more specific applications or targets. The elements of the nanoparticle composition can be selected based on the particular application or goal and/or based on the efficacy, toxicity, cost, ease of use, availability, or other characteristics of one or more elements. Similarly, a particular formulation of the nanoparticle composition may be selected for a particular application or target based on the efficacy and toxicity of a particular combination of elements.
The lipid component of the nanoparticle composition can include, for example, polymer conjugated lipids, cationic lipids, phospholipids (such as unsaturated lipids, e.g., DOPE or DSPC), and structural lipids according to formula (I) or (II) (and sub-formulae thereof) described herein. The elements of the lipid component may be provided in specific parts.
In one embodiment, provided herein are nanoparticle compositions comprising a polymer conjugated lipid compound provided herein, a therapeutic agent, and one or more excipients. In one embodiment, the polymer-conjugated lipid compound comprises a compound according to formula (I) or (II) (and sub-formulae thereof) as described herein and optionally one or more other polymer-conjugated lipid compounds. In one embodiment, the one or more excipients are selected from cationic lipids, neutral lipids and steroids. In one embodiment, the therapeutic agent is encapsulated within or associated with a lipid nanoparticle.
In one embodiment, provided herein is a nanoparticle composition (lipid nanoparticle) comprising:
i) 40 to 50 mole percent of a cationic lipid;
ii) neutral lipids;
iii) A steroid;
iv) a polymer conjugated lipid; and
v) a therapeutic agent.
As used herein, "mole percent" refers to the mole percent of a component relative to the total moles of all lipid components in the LNP (i.e., the total moles of cationic lipid, neutral lipid, steroid, and polymer conjugated lipid).
In one embodiment, the lipid nanoparticle comprises 41 to 49 mole percent, 41 to 48 mole percent, 42 to 48 mole percent, 43 to 48 mole percent, 44 to 48 mole percent, 45 to 48 mole percent, the content of cationic lipid is 46-48 mole percent, or 47.2-47.8 mole percent. In one embodiment, the lipid nanoparticle comprises about 47.0, 47.1, 47.2, 47.3, 47.4, 47.5, 47.6, 47.7, 47.8, 47.9, or 48.0 mole percent of the cationic lipid.
In one embodiment, the neutral lipid is present at a concentration of 5 to 15 mole percent, 7 to 13 mole percent, or 9 to 11 mole percent. In one embodiment, the neutral lipid is present at a concentration of about 9.5, 10, or 10.5 mole percent. In one embodiment, the molar ratio of cationic lipid to neutral lipid is about 4.1:1.0 to about 4.9:1.0, about 4.5:1.0 to about 4.8:1.0, or about 4.7:1.0 to 4.8:1.0.
In one embodiment, the steroid is present in a concentration in the range of 39 to 49 mole percent, 40 to 46 mole percent, 40 to 44 mole percent, 40 to 42 mole percent, 42 to 44 mole percent, or 44 to 46 mole percent. In one embodiment, the steroid is present at a concentration of 40, 41, 42, 43, 44, 45 or 46 mole percent. In one embodiment, the molar ratio of cationic lipid to steroid is 1.0:0.9 to 1.0:1.2, or 1.0:1.0 to 1.0:1.2. in one embodiment, the steroid is cholesterol.
In one embodiment, the ratio of therapeutic agent to lipid in the LNP (i.e., N/P, N representing the moles of cationic lipid, P representing the moles of phosphate present as part of the nucleic acid backbone) is 2:1 to 2. 30:1, for example 3:1 to 22:1. in one embodiment, N/P is 6:1 to 20:1 or 2:1 to 12:1. An exemplary N/P range includes about 3:1. about 6:1, about 12:1 and about 22:1.
in one embodiment, provided herein is a lipid nanoparticle comprising:
i) A cationic lipid having an effective pKa greater than 6.0;
ii) 5 to 15 mole percent of neutral lipid;
iv) 30 to 45 mole percent of a steroid;
v) a polymer conjugated lipid; and
vi) a therapeutic agent or a pharmaceutically acceptable salt or prodrug thereof,
wherein the mole percent is determined based on the total moles of lipids present in the lipid nanoparticle.
In one embodiment, the cationic lipid may be any of a variety of lipids that carry a net positive charge at a selected pH (e.g., physiological pH). Exemplary cationic lipids are described below. In one embodiment, the cationic lipid has a pKa greater than 6.25. In one embodiment, the cationic lipid has a pKa greater than 6.5. In one embodiment, the cationic lipid has a pKa of greater than 6.1, greater than 6.2, greater than 6.3, greater than 6.35, greater than 6.4, greater than 6.45, greater than 6.55, greater than 6.6, greater than 6.65, or greater than 6.7.
In one embodiment, the lipid nanoparticle comprises 40 to 45 mole percent of the cationic lipid. In one embodiment, the lipid nanoparticle comprises 45 to 50 mole percent of the cationic lipid.
In one embodiment, the molar ratio of cationic lipid to neutral lipid is from about 2:1 to about 8:1. In one embodiment, the neutral lipids comprise 5 to 10 mole percent of the lipids in the lipid nanoparticle.
Exemplary anionic lipids include, but are not limited to, phosphatidylglycerol, dioleoyl phosphatidylglycerol (DOPG), dipalmitoyl phosphatidylglycerol (DPPG), or 1, 2-distearoyl-sn-glycerol-3-phosphate- (1' -rac-glycerol) (DSPG).
In one embodiment, the lipid nanoparticle contains 1 to 10 mole% anionic lipid. In one embodiment, the lipid nanoparticle contains 1 to 5 mole% anionic lipid. In one embodiment, the lipid nanoparticle contains 1 to 9 mole%, 1 to 8 mole%, 1 to 7 mole%, or 1 to 6 mole% of the anionic lipid. In one embodiment, the molar ratio of anionic lipid to neutral lipid is 1:1 to 1:10.
In one embodiment, the steroid cholesterol. In one embodiment, the molar ratio of cationic lipid to cholesterol is from about 5:1 to 1:1. In one embodiment, the lipid nanoparticle contains 32 to 40 mole% of a steroid.
In one embodiment, the sum of the mole percent of neutral lipid and the mole percent of anionic lipid is 5 to 15 mole percent. In one embodiment, wherein the sum of the mole percent of neutral lipids and the mole percent of anionic lipids is 7 to 12 mole percent.
In one embodiment, the molar ratio of anionic lipid to neutral lipid is 1:1 to 1:10. In one embodiment, the sum of the mole percentages of neutral lipids and steroids is from 35 to 45 mole percent.
In one embodiment, the lipid nanoparticle contains 1.0 to 2.5 mole percent of polymer conjugated lipid. In one embodiment, the polymer conjugated lipid is present at a concentration of about 1.5 mole percent.
In one embodiment, the neutral lipid is present at a concentration of 5 to 15 mole percent, 7 to 13 mole percent, or 9 to 11 mole percent. In one embodiment, the neutral lipid is present at a concentration of about 9.5, 10, or 10.5 mole percent. In one embodiment, the molar ratio of cationic lipid to neutral lipid is about 4.1:1.0 to about 4.9:1.0, about 4.5:1.0 to about 4.8:1.0, or about 4.7:1.0 to 4.8:1.0.
in one embodiment, the steroid is cholesterol. In some embodiments, the steroid is present in a concentration ranging from 39 to 49 mole percent, 40 to 46 mole percent, 40 to 44 mole percent, 40 to 42 mole percent, 42 to 44 mole percent, or 44 to 46 mole percent. In one embodiment, the steroid is present at a concentration of 40, 41, 42, 43, 44, 45 or 46 mole percent. In certain embodiments, the molar ratio of cationic lipid to steroid is 1.0:0.9 to 1.0:1.2, or 1.0:1.0 to 1.0:1.2.
In one embodiment, the molar ratio of cationic lipid to steroid is from 5:1 to 1:1.
In one embodiment, the lipid nanoparticle contains 1.0 to 2.5 mole percent of polymer conjugated lipid. In one embodiment, the polymer conjugated lipid is present at a concentration of about 1.5 mole percent.
In one embodiment, the molar ratio of cationic lipid to polymer conjugated lipid is from about 100:1 to about 20:1. In one embodiment, the molar ratio of cationic lipid to polymer conjugated lipid is from about 35:1 to about 25:1.
In one embodiment, the lipid nanoparticle has an average diameter of 50nm to 100nm, or 60nm to 85nm.
In one embodiment, the composition comprises a cationic lipid, DSPC, cholesterol, and polymer conjugated lipid and mRNA. In one embodiment, the cationic lipid, DSPC, cholesterol, and polymer conjugated lipid provided herein are in a molar ratio of about 50:10:38.5:1.5.
Nanoparticle compositions can be designed for one or more specific applications or targets. For example, nanoparticle compositions can be designed to deliver therapeutic and/or prophylactic agents (e.g., RNA) to specific cells, tissues, organs, or systems thereof, etc., within a mammalian body. The physicochemical properties of the nanoparticle composition can be altered to increase selectivity for a particular bodily target. For example, the particle size may be adjusted based on the fenestration size (fenestration size) of the different organs. The therapeutic and/or prophylactic agents included in the nanoparticle composition may also be selected based on the desired delivery target(s). For example, therapeutic and/or prophylactic agents can be selected for a particular indication, condition, disease or disorder and/or delivered to a particular cell, tissue, organ or system, etc. (e.g., local or specific delivery). In certain embodiments, the nanoparticle composition may comprise an mRNA encoding a polypeptide of interest that is capable of being translated in a cell. Such compositions may be specifically designed for delivery to a particular organ. In certain embodiments, the compositions may be designed for specific delivery to the liver of a mammal.
The amount of therapeutic and/or prophylactic agent in the nanoparticle composition can depend on the size, composition, intended target and/or other properties of the nanoparticle composition, as well as the nature of the therapeutic and/or prophylactic agent. For example, the amount of RNA that can be used in the nanoparticle composition can depend on the size, sequence, and other characteristics of the RNA. The relative amounts of therapeutic and/or prophylactic agents and other elements (e.g., lipids) in the nanoparticle composition can also be adjusted. In some embodiments, the weight/weight ratio of lipid component to therapeutic and/or prophylactic agent in the nanoparticle composition can be about 5:1 to about 60:1, for example 5: 1. 6: 1. 7: 1. 8: 1. 9: 1. 10: 1. 11: 1. 12: 1. 13: 1. 14: 1. 15: 1. 16: 1. 17: 1. 18: 1. 19: 1. 20: 1. 25: 1. 30:1. 35: 1. 40: 1. 45: 1. 50:1 and 60:1. the weight/weight ratio of lipid component to therapeutic and/or prophylactic agent may be from about 10:1 to about 40:1. In certain embodiments, the weight to weight ratio is about 20:1. The amount of therapeutic and/or prophylactic agent in the nanoparticle composition can be measured by absorption spectroscopy (e.g., ultraviolet-visible spectroscopy).
In some embodiments, the nanoparticle composition comprises one or more RNAs, and the one or more RNAs, lipids, and amounts thereof may be selected to provide a particular N: p ratio. N of the composition: the P ratio refers to the molar ratio of nitrogen atoms in one or more lipids to the number of phosphate groups in the RNA. In some embodiments, a lower N is selected: p ratio. One or more RNAs, lipids, and amounts thereof may be selected such that N: the P ratio is about 2:1 to about 30:1, for example 2: 1. 3: 1. 4: 1. 5:1,6: 1. 7: 1. 8: 1. 9: 1. 10: 1. 12: 1. 14: 1. 16: 1. 18: 1. 20: 1. 22: 1. 24: 1. 26: 1. 28:1 or 30:1. in certain embodiments, the N:P ratio may be from about 2:1 to about 8:1. In other embodiments, the N to P ratio is from about 5:1 to about 8:1. For example, N: the P ratio may be about 5.0:1, about 5.5:1, about 5.67:1, about 6.0:1, about 6.5:1 or about 7.0:1. for example, the N:P ratio may be about 5.67:1.
The physical properties of the nanoparticle composition may depend on its components. For example, nanoparticle compositions comprising cholesterol as a structural lipid may have different properties compared to nanoparticle compositions comprising different structural lipids. Similarly, the characteristics of a nanoparticle composition may depend on the absolute or relative amounts of its components. For example, nanoparticle compositions comprising a higher mole fraction of phospholipids have different characteristics than nanoparticle compositions comprising a lower mole fraction of phospholipids. The characteristics may also vary depending on the method and conditions of preparation of the nanoparticle composition.
Nanoparticle compositions can be characterized by a variety of methods. For example, a microscope (e.g., a transmission electron microscope or a scanning electron microscope) can be used to examine the morphology and size distribution of the nanoparticle composition. Dynamic light scattering or potentiometry (e.g., potentiometry) can be used to measure zeta potential. Dynamic light scattering can also be used to determine particle size. Zetasizer Nano ZS (Malvem Instruments Ltd, malvem, worcestershire, UK) can also be used to measure various characteristics of nanoparticle compositions, such as particle size, polydispersity index, and Zeta potential.
In various embodiments, the nanoparticle composition can have an average size between 10 and 100nm. For example, the average size may be about 40nm to about 150nm, such as about 40nm,45nm,50nm,55nm,60nm,65nm,70nm,75nm,80nm,85nm,90nm,95nm,100nm,105nm,110nm,115nm,120nm,125nm,130nm,135nm,140nm,145nm, or 150nm. In some embodiments, the nanoparticle composition can have an average size of about 50nm to about 100nm, about 50nm to about 90nm, about 50nm to about 80nm, about 50nm to about 70nm, about 50nm to about 60nm, about 60nm to about 100nm, about 60nm to about 90nm, about 60nm to about 80nm, about 60nm to about 70nm, about 70nm to about 70nm 100nm, about 70nm to about 90nm, about 70nm to about 80nm, about 80nm to about 100nm, about 80nm to about 90nm, or about 90nm to about 100nm. In certain embodiments, the nanoparticle composition can have an average size of about 70nm to about 100nm. In some embodiments, the average size may be about 80nm. In other embodiments, the average size may be about 100nm.
The nanoparticle composition may be relatively uniform. The polydispersity index may be used to indicate the uniformity of the nanoparticle composition, e.g., the particle size distribution of the nanoparticle composition. A small (e.g., less than 0.3) polydispersity index generally indicates a narrow particle size distribution. The nanoparticle composition can have a polydispersity index of about 0 to about 0.25, such as 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, or 0.25. In some embodiments, the nanoparticle composition can have a polydispersity index of about 0.10 to about 0.20.
The zeta potential of a nanoparticle composition can be used to indicate the electromotive force of the composition. For example, the zeta potential can characterize the surface charge of the nanoparticle composition. Nanoparticle compositions having relatively low positive or negative charges are generally desirable because more highly charged materials can interact poorly with cells, tissues and other elements of the human body. In some embodiments, the zeta potential of the nanoparticle composition can be from about-10 to about +20mV, from about-10 to about +15mV, from about-10 to about +10mV, from about-10 to about +5mV, from about-10 to about 0mV, from about-10 to about-5 mV, from about-5 to about +20mV, from about-5 to about +15mV, from about-5 to about +10mV, from about-5 to about +5mV, from about-5 to about 0mV, from about 0 to about +20mV, from about 0 to about +15mV, from about 0 to about +10mV, from about 0 to about +5mV, from about +5 to about +20mV, from about +5 to about +15mV, or from about +5 to about +10mV.
Encapsulation efficiency of therapeutic and/or prophylactic agents describes the amount of therapeutic and/or prophylactic agent that is encapsulated or associated with the nanoparticle composition after preparation relative to the initial amount provided. High encapsulation efficiency (e.g., near 100%) is desirable. Encapsulation efficiency may be measured, for example, by comparing the amount of therapeutic and/or prophylactic agent before decomposing the nanoparticle composition with one or more organic solvents or detergents and after treatment in a solution comprising the nanoparticle composition. Fluorescence can be used to measure the amount of free therapeutic and/or prophylactic agent (e.g., RNA) in a solution. For nanoparticle compositions described herein, the encapsulation efficiency of the therapeutic and/or prophylactic agent can be at least 50%, e.g., 50%,55%,60%,65%,70%,75%,80%,85%,90%,91%,92%,93%,94%,95%,96%,97%,98%,99%, or 100%. In some embodiments, the packaging efficiency may be at least 80%. In certain embodiments, the packaging efficiency may be at least 90%.
The nanoparticle composition may optionally comprise one or more coatings. For example, the nanoparticle composition can be formulated as a capsule, film, or tablet with a coating. The capsules, films or tablets of the compositions described herein may have any useful size, tensile strength, hardness or density.
7.6 pharmaceutical compositions
Nanoparticle compositions according to the present disclosure may be formulated as part or all of a pharmaceutical composition. The pharmaceutical composition may include one or more nanoparticle compositions. For example, the pharmaceutical composition may include one or more nanoparticle compositions, and one or more different therapeutic and/or prophylactic agents. The pharmaceutical composition may further comprise one or more pharmaceutically acceptable excipients or auxiliary ingredients, such as those described herein. General guidelines for the formulation and production of pharmaceutical compositions and formulations are described, for example, in Remington's The Science and Practice of Pharmacy, 21 st edition, a.r. gennaro; related descriptions are provided in Lippincott, williams & Wilkins, baltimore, md.,2006, etc. Conventional excipients and adjunct ingredients can be used in any pharmaceutical composition unless they are incompatible with one or more components of the nanoparticle composition. If the excipient or adjunct ingredient is incompatible with the components of the nanoparticle composition, its combination can result in a poor biological or deleterious effect.
In some embodiments, one or more excipients or adjunct ingredients can comprise greater than 50% of the total mass or volume of the pharmaceutical composition including the nanoparticle composition. For example, typically one or more excipients or adjunct ingredients can comprise 50%,60%,70%,80%,90% or more of the pharmaceutical composition. In some embodiments, the pharmaceutically acceptable excipient is at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% pure. In some embodiments, the excipient is approved for human and veterinary use. In some embodiments, the excipient is approved by the U.S. food and drug administration. In some embodiments, the excipient is pharmaceutical grade. In some embodiments, the excipient meets the standards of the United States Pharmacopeia (USP), the European Pharmacopeia (EP), the british pharmacopeia, and/or the international pharmacopeia.
The relative amounts of one or more nanoparticle compositions, one or more pharmaceutically acceptable excipients, and/or any other ingredients in the pharmaceutical compositions according to the present disclosure are adjusted to vary depending on their characteristics, size, etc. associated conditions, and further depending on the subject and route of administration of the composition. For example, the pharmaceutical composition may comprise from 0.1% to 100% (wt/wt) of one or more nanoparticle compositions.
In certain embodiments, the nanoparticle compositions and/or pharmaceutical compositions of the present disclosure are refrigerated or frozen for storage and transport. For example, at a temperature of 4 ℃ or less, between about-150 ℃ and 0 ℃ or at a temperature of about-80 ℃ to about-20 ℃, such as at a temperature of about-5 ℃, -10 ℃, -15 ℃, -20 ℃, -25 ℃, -30 ℃, -40 ℃, -50 ℃, -60 ℃, -70 ℃, -80 ℃, -90 ℃, -130 ℃ or-150 ℃). The pharmaceutical compositions comprising the compounds of formula (I) and their sub-formulae in solution form are refrigerated for storage or transport under conditions such as about-20 ℃, -30 ℃, -40 ℃, -50 ℃, -60 ℃, -70 ℃ or-80 ℃. In some embodiments, the present disclosure also relates to methods of improving the stability of nanoparticle compositions and/or pharmaceutical compositions comprising compounds of formula (I) or (II) (and sub-formulae thereof). By storing the nanoparticle composition and/or the pharmaceutical composition at a temperature of 4 ℃ or less, such as between about-150 ℃ and about 0 ℃ or between about-80 ℃ and about-20 ℃, such as between about-5 ℃, about-10 ℃, about-15 ℃, about-20 ℃, about-25 ℃, about-30 ℃, about-40 ℃, about-50 ℃, about-60 ℃, about-70 ℃, about-80 ℃, about-90 ℃, about-130 ℃ or about-150 ℃. The nanoparticle compositions and/or pharmaceutical compositions disclosed herein are stable at a temperature of 4 ℃ or less (e.g., between about 4 ℃ and-20 ℃) for about at least 1 week, at least 2 weeks, at least 3 weeks, at least 4 weeks, at least 5 weeks, at least 6 weeks, at least one month, at least 2 months, at least 4 months, at least 6 months, at least 8 months, at least 10 months, at least 12 months, at least 14 months, at least 16 months, at least 18 months, at least 20 months, at least 22 months, or at least 24 months. In one embodiment, the formulation is stable at about 4 ℃ for at least 4 weeks. In certain embodiments, the pharmaceutical compositions of the present disclosure comprise a nanoparticle composition as disclosed herein and a pharmaceutically acceptable carrier selected from one or more of Tris, acetate (e.g., acetic acid), citrate (e.g., sodium citrate), saline, PBS, and sucrose. In certain embodiments, the pharmaceutical compositions of the present disclosure have a pH of between about 7 and 8 (e.g., 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0, or between 7.5 and 8 or 7 and 7.8). The pharmaceutical compositions of the present disclosure comprise the nanoparticle compositions disclosed herein, tris, saline, and sucrose, and have a pH of about 7.5-8, which is suitable for storage or transport at about-20 ℃. For example, the pharmaceutical compositions of the present disclosure comprise the nanoparticle compositions disclosed herein and PBS and have a pH of about 7-7.8, suitable for storage or transport at a temperature of about 4 ℃ or less. In the context of the present disclosure, "stable" and "stability" refer to the resistance of a nanoparticle composition or pharmaceutical composition disclosed herein to chemical or physical changes (e.g., degradation, particle size change, aggregation change) under given manufacturing, transportation, storage, and/or use conditions (e.g., applied stress (shear force, freeze/thaw stress, etc.).
The nanoparticle composition and/or pharmaceutical composition comprising one or more nanoparticle compositions can be administered to any patient or subject, including can provide a beneficial therapeutic effect by delivering therapeutic and/or prophylactic agents to a patient or subject specific cell, tissue, organ, or system thereof, such as the renal system. Although the description herein of nanoparticle compositions and pharmaceutical compositions comprising nanoparticle compositions is primarily directed to compositions suitable for administration to humans, it will be understood by those skilled in the art that such compositions are generally suitable for administration to any other mammal. Modification of compositions suitable for administration to humans in order to render the compositions suitable for administration to a variety of animals is well known and can be designed and/or carried out by ordinary skill veterinarian pharmacologists simply through ordinary experimentation. Subjects to whom the compositions are contemplated include, but are not limited to, humans, other primates, and other mammals, including commercially relevant mammals such as cows, pigs, horses, sheep, cats, dogs, mice, and rats.
Pharmaceutical compositions comprising one or more nanoparticle compositions may be prepared by any method known in the pharmacological arts or later developed. Typically, such preparation involves combining the active ingredient with excipients and/or one or more other auxiliary ingredients, and if necessary, shaping and/or packaging the product separately into the desired single or mixed form of multiple dosage units.
Pharmaceutical compositions according to the present disclosure may be prepared, packaged and/or sold in bulk as single unit doses and/or as a plurality of single unit doses. A "unit dose" is a discrete amount of a pharmaceutical composition comprising a predetermined amount of an active ingredient (e.g., a nanoparticle composition). The amount of active ingredient is typically equal to the dose of active ingredient to be administered to the subject and/or a convenient fraction of the dose, e.g., half or one third of the dose.
The pharmaceutical compositions may be prepared in a variety of forms suitable for a variety of routes and methods of administration. For example, pharmaceutical compositions may be prepared in liquid dosage forms (such as emulsions, microemulsions, nanoemulsions, solutions, suspensions, syrups and elixirs), injectable dosage forms, solid dosage forms (such as capsules, tablets, pills, powders and granules), dosage forms for topical and/or transdermal administration (such as ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants and patches), suspensions, powders and other forms.
Liquid dosage forms for oral and parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, nanoemulsions, solutions, suspensions, syrups and/or elixirs. In addition to the active ingredient, the liquid dosage form may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. In addition to inert diluents, the oral compositions can include other therapeutic and/or prophylactic agents, such as other agents including wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents. In certain embodiments for parenteral administration, the composition is admixed with a solubilizing agent such as cremophor (TM), an alcohol, an oil, a modified oil, a glycol, a polysorbate, a cyclodextrin polymer, and/or combinations thereof.
Injectable formulations, such as sterile injectable aqueous or oleaginous suspensions, may be formulated according to the known art using suitable dispersing, wetting and/or suspending agents. The sterile injectable preparation may be a sterile injectable solution, suspension and/or emulsion in a non-toxic parenterally acceptable diluent and/or solvent, for example, as a solution in 1, 3-butanediol. Acceptable vehicles and solvents that may be used include water, ringer's solution and isotonic sodium chloride solution. Sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. Fatty acids such as oleic acid find use in the preparation of injectables.
The injectable formulation may be filtered through a bacterial-retaining filter and/or sterilized by incorporating sterilizing agents in the form of sterile solid compositions which are dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
Disclosed herein are methods of delivering a therapeutic and/or prophylactic agent to a mammalian cell or organ, producing a polypeptide of interest in the mammalian cell, and treating a disease or disorder in a mammal comprising contacting the mammalian cell with a nanoparticle composition comprising the therapeutic and/or prophylactic agent.
8. Examples
The embodiments in this section are provided by way of example only and not by way of limitation.
8.1 example 1: preparation of Compound 1.
Step 0: preparation of monomer 1
To a solution of ethyl 2- (dimethylamino) methacrylate (7.8 g,50.0mmol,1.0 eq) in ACN (75.0 ml) was added tert-butyl 2-bromoacetate (14.6 g,75.0mmol,1.5 eq) at RT. The mixture was stirred for 16 hours. LCMS showed the reaction was complete and after evaporation of the mixture under reduced pressure, monomer 1 was obtained as a white solid (14.0 g, crude).
Step 1: preparation of Compounds 1-2
EDCI (6.5 g,34.2mmol,2.5 eq), DMAP (0.344 g,2.74mmol,0.2 eq) and DIEA (8.8 g,68.5mmol,5.0 eq) were added to a solution of 1-1 (2.5 g,13.7mmol,1.0 eq) and myristic acid (7.8 g,34.2mmol,2.5 eq) in DCM (30.0 ml) at RT. The mixture was stirred at 50℃for 16 hours. TLC showed the reaction was complete and the mixture was evaporated under reduced pressure and purified by FCC (PE/ea=1/0-20/1) to give compound 1-2 as a white solid (6.9 g,83% yield).
Step 2: preparation of Compounds 1-3
Pd/C (7.5 g,0.1 eq) was added to a solution of 1-2 (6.9 g,11.5mmol,1.0 eq) in EA (75.0 ml) at RT. The mixture was heated to 50deg.C at H 2 (5 MPa) for 16 hours. TLC showed the reaction was complete, and after filtering the mixture and evaporating under reduced pressure, compounds 1-3 (5.3 g, crude product) were obtained as white solids.
Step 3: preparation of Compounds 1-5
Et is added to a solution of 1-3 (2.56 g,5.0mmol,1.0 eq) and 1-4 (1.47 g,6.5mmol,1.5 eq) in DCM (20.0 ml) at RT 3 N (1.01 g,10.0mmol,2.0 eq). The mixture was stirred at RT for 16 h. TLC showed the reaction was complete and the mixture was evaporated under reduced pressure and purified by FCC (PE/ea=1/0-100/1) to give compounds 1-5 as white solids (3.0 g,90% yield).
Step 4: preparation of Compounds 1-6
To a solution of compounds 1-5 (165.0 mg,0.25mmol,1.0 eq) and monomer 1 (0.44 g,1.25mmol,5.0 eq) in DMF (1.0 ml) and MeOH (1.0 ml) at RT were added CuBr (36.0 mg,0.25mmol,1.0 eq) and 1,1,4,7,10,10-Hexamethyltriethylenetetramine (HMTETA) (57.5 mg,0.25mmol,1.0 eq). The mixture is heated to 50 DEG CStirred for 3 hours, then 2-mercaptoethanol (195.0 mg,2.5mmol,10.0 eq) and DIEA (0.32 g,2.5mmol,10.0 eq) were added. The mixture was stirred at RT for 16 h. Addition of Et 2 O, after filtration, gives compounds 1-6 (0.8 g, crude product) as a white solid.
Step 5: preparation of Compound 1
To a solution of compounds 1-6 (0.8 g,0.4mmol,1.0 eq) in toluene (5.0 ml) was added TFA (2.0 ml) at RT. The mixture was stirred at 100℃for 16 hours. Addition of Et 2 O, after filtration, gives the crude product, which is purified by dialysis bag and lyophilized to give compound 1 as a white solid (160.0 mg,20% yield).
1 H NMR(400MHz,D 2 O)δ:0.82-1.26(m,56H),1.53-1.91(m,25H),3.24-3.32(m,50H),3.76-3.78(m,4H),4.04-4.13(m,10H),4.37-4.41(m,5H)。
The following compounds were prepared in a similar manner to compound 1 using different equivalents of monomer 1.
/>
8.2 example 2: preparation of Compound 4.
Step 1: preparation of Compound 4
To a solution of compounds 1-5 (165.0 mg,0.25mmol,1.0 eq) and commercially available monomer 3 (370.0 mg,1.25mmol,5.0 eq) in DMF (1.0 ml) and MeOH (1.0 ml) was added CuBr (36.0 mg,0.25mmol,1.0 eq) and HMTETA (57.5 mg,0.25mmol,1.0 eq) at RT. The mixture was stirred at 50℃for 3 hours, then 2-mercaptoethanol (195.0 mg,2.5mmol,10.0 eq) and DIEA (0.32 g,2.5mmol,10.0 eq) were added. The mixture was stirred at RTMix for 16 hours. Addition of Et 2 O, after filtration, gives the crude product, which is purified by dialysis bag and lyophilized to give compound 4 as a white solid (200 mg,37% yield).
1 H NMR(400MHz,D 2 O)δ:0.82-1.22(m,60H),1.49-1.70(m,5H),1.96(s,10H),3.18(s,46H),3.63(s,14H),4.03-4.24(m,35H)。
The following compounds were prepared in a similar manner to compound 4 using different equivalents of monomer 3.
8.3 example 3: preparation of Compound 7.
Step 0: preparation of monomer 2
At N 2 To a solution of ethyl 2- (dimethylamino) methacrylate (5.0 g,31.8mmol,1.0 eq) in ACN (20 ml) was added a solution of 1, 2-oxathiolane 2, 2-dioxide (3.9 g,31.8mmol,1.0 eq) in ACN (4 ml) at room temperature. The reaction mixture was stirred at room temperature for 16 hours, filtered and washed with ACN. After drying the solid in vacuo, monomer 2 was obtained as a white solid (8.2 g,92% yield).
1 H NMR(400MHz,D 2 O)δ:1.88(s,3H),2.13-2.21(m,2H),2.90-2.93(m,2H),3.16(s,6H),3.51-3.55(m,2H),3.75-3.76(m,2H),4.58(s,2H),5.72(s,1H),6.10(s,1H)。
Step 1: preparation of Compound 7
To a solution of compounds 1-5 (165.0 mg,0.25mmol,1.0 eq) and monomer 2 (0.35 g,1.25mmol,5.0 eq) in DMF (1.0 ml) and MeOH (1.0 ml) at RT was added CuBr (36.0 mg,0.25mmol,1.0 eq) and HMTETA (57.5 mg,0.25mmol,1.0 eq). The mixture was stirred at 50℃for 3 hours, then 2-mercaptoethanol (195.0 mg,2.5mmol,10.0 eq) and DIEA (0.32 g,2.5mmol,10.0 eq) were added. The mixture was stirred at RT for 16 h. Addition of Et 2 O, after filtration, gives the crude product, which is purified by dialysis bag and lyophilized to give compound 7 as a white solid (12.0 mg,2.4% yield).
1 H NMR(400MHz,D 2 O)δ:0.774-1.29(m,53H),1.83-1.97(m,10H),2.20(s,30H),2.92(s,28H),3.17(s,78H),3.51(s,29H),3.69(s,20H),4.37-4.42(m,20H)。
The following compounds were prepared in a similar manner to compound 7 using different equivalents of monomer 2.
8.4 example 4: preparation of Compound 10.
Step 1: preparation of Compound 10-2
To a solution of stearyl alcohol (10.0 g,37.0mmol,1.0 eq) and 1-4 (12.0 g,55.0mmol,1.5 eq) in DCM (150.0 ml) at RT was added Et 3 N (7.5 g,74.0mmol,2.0 eq). The mixture was stirred at RT for 16 h. TLC showed the reaction was complete and the mixture was evaporated under reduced pressure and purified by FCC (PE/ea=1/0-100/1) to give compound 10-2 (12.0 g,77% yield) as a colourless oil.
Step 2: preparation of Compound 10-3
To a solution of 10-2 (105.0 mg,0.25mmol,1.0 eq) and monomer 1 (0.44 g,1.25mmol,5.0 eq) in DMF (1.0 ml) and MeOH (1.0 ml) at RT were added CuBr (36.0 mg,0.25mmol,1.0 eq) and HMTETA (57.5 mg,0.25mmol,1.0 eq). The mixture was stirred at 50℃for 3 hours, then 2-mercaptoethanol (195.0 mg,2.5mmol,10.0 eq) and DIEA (0.32 g,2.5mmol,10.0 eq) were added. The mixture was stirred at RT for 16 h. Addition of Et 2 O, after filtration, gives 10-3 (0.5 g, crude product) as a white solid.
Step 3: preparation of Compound 10
To a solution of 10-3 (0.5 g,0.3mmol,1.0 eq) in toluene (5.0 ml) at RT was added TFA (2.0 ml). The mixture was stirred at 100℃for 16 hours. Addition of Et 2 O, after filtration, gives the crude product, which is purified by dialysis bag and lyophilized to give compound 10 as a white solid (240.0 mg,44% yield).
1 H NMR(400MHz,D 2 O)δ:0.73-1.23(m,50H),1.43-1.63(m,6H),1.73-2.06(m,10H),3.24-3.28(m,30H),3.75(s,2H),3.95-4.12(m,20H),4.29-4.44(m,12H)。
The following compounds were prepared in a similar manner to compound 10 using different equivalents of monomer 1.
8.5 example 5: preparation of Compound 13.
Step 1: preparation of Compound 13
To a solution of compound 10-2 (105.0 mg,0.25mmol,1.0 eq) and monomer 3 (368.0 mg,1.25mmol,5.0 eq) in DMF (1.0 ml) and MeOH (1.0 ml) at RT was added CuBr (36.0 mg,0.25mmol,1.0 eq) and HMTETA (57.5 mg,0.25mmol,1.0 eq). The mixture was stirred at 50℃for 3 hours, then mercaptoethanol (195.0 mg,2.5mmol,10.0 eq) and DIEA (0.32 g,2.5mmol,10.0 eq) were added. The mixture was stirred at RT for 16 h. Addition of Et 2 O, after filtration, gives the crude product, which is purified by dialysis bag and lyophilized to give compound 13 as a white solid (220 mg,45% yield).
1 H NMR(400MHz,D 2 O)δ:0.72-1.23(m,41H),1.54-2.00(m,25H),3.17(s,44H),3.62(s,15H),4.02(s,16H),4.16-4.23(m,15H)。
The following compounds were prepared in a similar manner to compound 13 using different equivalents of monomer 3.
8.6 example 6: preparation of Compound 16.
Step 1: preparation of Compound 16
To a solution of 10-2 (105.0 mg,0.25mmol,1.0 eq) and monomer 2 (0.2 g,0.75mmol,5.0 eq) in DMF (1.0 ml) and MeOH (1.0 ml) at RT were added CuBr (36.0 mg,0.25mmol,1.0 eq) and HMTETA (57.5 mg,0.25mmol,1.0 eq). The mixture was stirred at 50℃for 3 hours, then mercaptoethanol (195.0 mg,2.5mmol,10.0 eq) and DIEA (0.32 g,2.5mmol,10.0 eq) were added. The mixture was stirred at RT for 16 h. Addition of Et 2 O, filtered to give the crude product, which was purified by dialysis bag and lyophilized to give compound 16 as a white solid (32.0 mg,2.5% yield).
1 H NMR(400MHz,D 2 O)δ:0.78-1.19(m,35H),1.56-1.79(m,6H),1.93(s,10H),2.22(s,15H),2.92(s,12H),3.17(s,30H),3.54(s,14H),3.75(s,13H),4.45(s,17H)。
The following compounds were prepared in a similar manner to compound 16 using different equivalents of monomer 2.
8.7 example 7: preparation of Compound 19.
At RT, 10-2 (105.0 mg,0.25mmol,1.0 eq) monomers5 (215 mg,2.5mmol,10.0 eq) and monomer 4 (393 mg,2.5mmol,10.0 eq) to a solution of CuBr (36.0 mg,0.25mmol,1.0 eq) and HMTETA (57.5 mg,0.25mmol,1.0 eq) in DMF (1.0 ml) and MeOH (1.0 ml). The mixture was stirred at 50℃for 3 hours, then mercaptoethanol (195.0 mg,2.5mmol,10.0 eq) and DIEA (0.32 g,2.5mmol,10.0 eq) were added. The mixture was stirred at RT for 16 h. Addition of Et 2 O, after filtration, gives the crude product, which is purified by dialysis bag and lyophilized to give compound 19 (139.0 mg,19.0% yield) as a white solid.
1 H NMR(400MHz,D 2 O)δ:0.89(t,3H),1.10-1.31(m,94H),1.55-1.89(b,42H),3.16(s,60H),3.54(t,20H),4.2-4.45(m,22H)。
8.8 example 8: preparation and characterization of lipid nanoparticles
Briefly, cationic lipids, DSPC, cholesterol, and polymer conjugated lipids provided herein were conjugated to a lipid as described in 50:10:38.5:1.5 in ethanol and mRNA was diluted in 10 to 50mM citrate buffer, ph=4. Using a microfluidic device, the ethanolic lipid solution was mixed with the mRNA aqueous solution at a flow rate of 9-30mL/min at 1:3 in a total lipid to mRNA weight ratio of about 10:1 to 30:1 preparation of LNP. And ethanol was removed using dialysis with PBS instead of ethanol. Finally, the lipid nanoparticles were filtered through a 0.2 μm sterile filter.
The size of the liposome nanoparticles was determined by dynamic light scattering using Malvern Zetasizer Nano ZS (Malvern UK) of 173 ° backscatter detection mode. The encapsulation efficiency of lipid nanoparticles was determined using the Quant-it Ribogreen RNA quantification kit (Thermo Fisher Scientific, UK) according to the manufacturer's instructions.
As reported in the literature, the apparent pKa of LNP formulations correlates with the efficiency of LNP delivery with nucleic acids in vivo. The apparent pKa of each formulation was determined using a fluorometry based on 2- (p-tolyl) -6-naphthalenesulfonic acid (TNS). LNP formulations comprising cationic lipid/DSPC/cholesterol/DMG-PEG (50/10/38.5/1.5 mol%) were prepared as described above. TNS was prepared as 300uM distilled water stock solution. The LNP formulation was diluted to 0.1mg/mL of total lipid in 3mL of a buffer solution containing 50mM sodium citrate, 50mM sodium phosphate, 50mM sodium borate and 30mM sodium chloride, wherein the pH was 3 to 9. TNS solution was added to a final concentration of 0.1mg/ml, and after vortexing, fluorescence intensity was measured at room temperature in a Molecular Devices Spectramax iD spectrometer using excitation wavelengths of 325nm and 435 nm. The fluorescence data were subjected to an S-shaped best fit analysis and the pKa was measured when the pH reached half the maximum fluorescence intensity.
8.9 example 9
Lipid nanoparticles comprising 50% cationic lipid C1, 38.5% cholesterol, 10% dspc and 1.5% polymeric compound encapsulating human erythropoietin (hEPO) mRNA were administered systemically to 6-8 week old female ICR mice (xiguer-Bikai, shanghai) by tail vein injection at a dose of 0.5mg/kg and blood was collected 6 hours after administration. Except for the above groups, lipid nanoparticles comprising DMG-PEG2000 encapsulating hEPO mRNA were similarly administered at the same dose to age and sex comparison groups of mice as positive controls. The results are shown in table 3 and fig. 1.
Table 3.
Polymer Cationic lipids Z-avg.(nm) PDI EE%
6 C1 88.41 0.075 93.16%
12 C1 100.4 0.11 91.80%
DMG-PEG2000 C1 82.79 0.056 90.73%
8.10 example 10
Lipid nanoparticles comprising 50% cationic lipid (one of: lipid 5, MC3, C1 and C2), 38.5% cholesterol, 10% DSPC and 1.5% polymeric compound encapsulating human erythropoietin (hEPO) mRNA were systemically administered to 6-8 week old female ICR mice (xipeer-Bikai, shangai) at a dose of 0.5mg/kg by tail vein injection, and mouse blood was collected 6 hours after administration. Except for the above groups, lipid nanoparticles comprising DMG-PEG2000 encapsulating hEPO mRNA were similarly administered at the same dose to age and sex comparison groups of mice as positive controls. The results are shown in table 4 and fig. 2.
Table 4.
8.11 example 11
Lipid nanoparticles comprising 50% cationic lipid C1, 38.5% cholesterol, 10% dspc, and 1.5% polymeric compound encapsulating firefly luciferase (FLuc) mRNA were prepared. Mu.l of each LNP sample was deposited on a glow discharge porous carbon grid (Quantifoil R1.2/1.3) and vitrified using a Vitrobot Mark IV (ThermoFisher Scientific) instrument. Cyro-EM imaging was performed on a Talos F200C equipped with a Ceta 4kX 4k camera operating at 200kV acceleration voltage. The results are shown in FIG. 3.
The structure of the cationic lipids used in the examples is shown below:
/>

Claims (43)

1. a compound of formula (I):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
l is a lipid;
x is a linker;
each R 3 Independently H or C 1 -C 6 An alkyl group;
each Y 1 Independently is a bond, O, S or NR a
Each G 4 Independently is a bond or C 1 -C 12 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -S-or-NR a -optionally substituted;
each G 5 Independently is a bond or C 1 -C 12 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -S-or-NR a -optionally substituted;
each R a H, C independently 1 -C 12 Alkyl orC 2 -C 12 Alkenyl groups;
Z 1 and Z 2 One of which is a positively charged moiety and Z 1 And Z 2 The other of which is a negatively charged moiety;
n is an integer from 2 to 100;
t is hydrogen, halogen, alkyl, alkenyl, -OR ', -SR', -COOR ', -OCOR', -NR 'R', -N + (R”) 3 、-P + (R”) 3 -S-C (=s) -S-R ", -S-C (=s) -O-R", -S-C (=s) -NR "R", -S-C (=s) -aryl, cyano, azido, aryl, heteroaryl, or a targeting group, wherein R "in each occurrence is independently hydrogen or alkyl; and
wherein each alkyl, alkenyl, alkylene, aryl, and heteroaryl is independently optionally substituted; and is also provided with
Provided that the compound is not:
2. the compound of claim 1, provided that when Z 1 Or Z is 2 Is carboxylate (-COO) ) When T is not bromine.
3. A compound of formula (II):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
l is a lipid;
x is a linker;
each R 3 Independently H or C 1 -C 6 An alkyl group;
each R 4 Independently H or C 1 -C 6 An alkyl group;
each X is 1 Independently a bond or-C (O) -Y 1 -;
Each X is 2 Independently a bond or-C (O) -Y 2 -;
Each Y 1 Independently is a bond, O, S or NR a
Each Y 2 Independently is a bond, O, S or NR a
Each G 4 Independently is a bond or C 1 -C 12 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -S-or-NR a -optionally substituted;
each G 5 Independently is a bond or C 1 -C 12 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -S-or-NR a -optionally substituted;
each R a H, C independently 1 -C 12 Alkyl or C 2 -C 12 Alkenyl groups;
Z 1 and Z 2 One of which is a positively charged moiety and Z 1 And Z 2 The other of which is a negatively charged moiety;
n is an even integer from 2 to 100;
"ran" means eachUnit and each->The units appear in any order within { };
t is hydrogen, halogen, alkyl, alkenyl, -OR ', -SR', -COOR ', -OCOR', -NR 'R', -N + (R”) 3 、-P + (R”) 3 -S-C (=s) -S-R ", -S-C (=s) -O-R", -S-C (=s) -NR "R", -S-C (=s) -aryl, cyano, azido, aryl, heteroaryl, or a targeting group, wherein R "in each occurrence is independently hydrogen or alkyl; and
wherein each alkyl, alkenyl, alkylene, aryl, and heteroaryl is independently optionally substituted.
4. A compound according to any one of claims 1 to 3 wherein Y 1 Is O and Y 2 Is O; g 4 Is C 1 -C 3 An alkylene group; g 5 Is C 1 -C 3 An alkylene group; z is Z 1 Or Z is 2 Is a quaternary amine cation; and/or Z 1 Or Z is 2 Is a carboxylate, sulfonate or phosphate anion.
5. The compound of claim 1, which is a compound of formula (I-a):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
6. The compound of claim 5, wherein s is 2 and t is 1.
7. The compound of claim 1, which is a compound of formula (I-B):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
8. The compound of claim 7, wherein s is 2 and t is 3.
9. The compound of claim 1, which is a compound of formula (I-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
10. The compound of claim 9, wherein s is 2 and t is 2.
11. A compound according to claim 3, which is a compound of formula (II-a), (II-B) or (II-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety;
each R p Independently C 1 -C 6 Alkyl or-O- (C) 1 -C 6 An alkyl group); and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
12. The compound of claim 11, wherein in formula (II-a), s is 2; or in the formula (II-B) or (II-C), s is 2 and t is 2.
13. The compound of any one of claims 1 to 12, wherein
L is C or C 6 -C 24 Alkyl or C 6 -C 24 Alkenyl, sterol or sphingolipid lipids; or (b)
L is a lipid of the formula:
wherein:
G 1 、G 2 and G 3 Each independently is a bond, C 1 -C 12 Alkylene or C 2 -C 12 Alkenylene;
L 1 is-OC (=O) R 1 、-C(=O)OR 1 、-OC(=O)OR 1 、-C(=O)R 1 、-OR 1
-S(O) x R 1 、-S-SR 1 、-C(=O)SR 1 、-SC(=O)R 1 、-NR a C(=O)R 1 、-C(=O)NR b R c 、-NR a C(=O)NR b R c 、-OC(=O)NR b R c 、-NR a C(=O)OR 1 、-SC(=S)R 1 、-C(=S)SR 1 、-C(=S)R 1 、-CH(OH)R 1 、-P(=O)(OR b )(OR c )、-(C 6 -C 10 Arylene) -R 1 (6-to 10-membered heteroarylene) -R 1 Or R is 1
L 2 is-OC (=O) R 2 、-C(=O)OR 2 、-OC(=O)OR 2 、-C(=O)R 2 、-OR 2
-S(O) x R 2 、-S-SR 2 、-C(=O)SR 2 、-SC(=O)R 2 、-NR d C(=O)R 2 、-C(=O)NR e R f 、-NR d C(=O)NR e R f 、-OC(=O)NR e R f 、-NR d C(=O)OR 2 、-SC(=S)R 2 、-C(=S)SR 2 、-C(=S)R 2 、-CH(OH)R 2 、-P(=O)(OR e )(OR f )、-(C 6 -C 10 Arylene) -R 2 (6-to 10-membered heteroarylene) -R 2 Or R is 2
L 3 is-OC (=O) -, -C (=O) O-; -OC (=o O-, O- -C (=o) -, -O-, -S (O) x -、
-S-S-、-C(=O)S-、-SC(=O)-、-NR a C(=O)-、-C(=O)NR b -、-NR a C(=O)NR b -、-OC(=O)NR b -、-NR a C(=O)O-、-SC(=S)-、-C(=S)S-、-C(=S)-、-CH(OH)-、-P(=O)(OR b )O-、-(C 6 -C 10 Arylene) -or- (6-to 10-membered heteroarylene) -;
R 1 and R is 2 Each independently is C 6 -C 24 Alkyl or C 6 -C 24 Alkenyl groups;
R a 、R b 、R d and R is e Each independently of the otherThe ground is H, C 1 -C 12 Alkyl or C 2 -C 12 Alkenyl groups;
R c and R is f Each independently is C 1 -C 12 Alkyl or C 2 -C 12 Alkenyl groups;
x is 0, 1 or 2, and
wherein each alkyl, alkenyl, alkylene, alkenylene, arylene, and heteroarylene is independently optionally substituted.
14. The compound of claim 13, which is a compound of formula (I-D), (II-D), (I-E), or (II-E):
or a pharmaceutically acceptable salt or stereoisomer thereof.
15. The compound of claim 13 or 14, wherein G 1 And G 2 Each independently is a bond or C 1 An alkylene group; l (L) 1 is-OC (=O) R 1 、-C(=O)OR 1 、-C(=O)NR b R c Or R is 1 The method comprises the steps of carrying out a first treatment on the surface of the And/or L 2 is-OC (=O) R 2 、-C(=O)OR 2 、-C(=O)NR e R f Or R is 2
16. The compound of any one of claims 1 to 15, wherein X is C 1 -C 12 An alkylene group, wherein:
one or more-CH 2 -independently by-O-, -NR a -、-OC(=O)-、-C(=O)O-、-OC(=O)O-、-C(=O)-、-S(O) x -、-S-S-、-C(=O)S-、-SC(=O)-、-NR a C(=O)-、-C(=O)NR b -、-NR a C(=O)NR b -、-SC(=S)-、-C(=S)S-、-C(=S)-、-P(=O)(OR b )-O-、-O-P(=O)(OR b )-O-、-(C 6 -C 10 Arylene) -or- (6-to 10-membered heteroaryleneGroup) -optionally substituted;
x is 0, 1 or 2;
R a and R is b Each independently H, C 1 -C 12 Alkyl or C 2 -C 12 Alkenyl groups; and
and each of alkyl, alkenyl, alkylene, arylene, and heteroarylene is optionally substituted.
17. The compound of claim 16, wherein X is-C (=o) -C 1 -C 11 Alkylene OR-P (=o) (OR b )-O-C 1 -C 11 Alkylene groups, wherein one or more-CH 2 -independently by-O-, -NR a -、-OC(=O)-、-C(=O)O-、-NR a C (=o) -or-C (=o) NR b -optionally substituted.
18. The compound of claim 16 or 17, wherein the alkylene of X is optionally substituted with one or more C 1 -C 6 Alkyl or cyano substitution.
19. The compound of claim 13, which is a compound of formula (III) or (IV):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
each R 5 H, C independently 1 -C 6 Alkyl or cyano.
20. The compound of claim 19, which is a compound of formula (III-a):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
21. The compound of claim 20, wherein s is 2 and t is 1.
22. The compound of claim 19, which is a compound of formula (III-B):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
23. The compound of claim 22, wherein s is 2 and t is 3.
24. The compound of claim 19, which is a compound of formula (III-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
25. The compound of claim 24, wherein s is 2 and t is 2.
26. The compound of claim 19, which is Sup>A compound of formulSup>A (IV-Sup>A), (IV-B), or (IV-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
each R p Independently C 1 -C 6 Alkyl or-O- (C) 1 -C 6 An alkyl group); and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
27. The compound of claim 26, wherein in formulSup>A (IV-Sup>A), s is 2; or in formula (IV-B) or (IV-C), s is 2 and t is 2.
28. The compound of claim 13, which is a compound of formula (V) or (VI):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
each R 5 H, C independently 1 -C 6 Alkyl or cyano.
29. The compound of claim 28, which is Sup>A compound of formulSup>A (V-Sup>A):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
30. The compound of claim 29, wherein s is 2 and t is 1.
31. The compound of claim 28, which is a compound of formula (V-B):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; and
wherein each alkyl and ring moiety is independently optionally substituted.
32. The compound of claim 31, wherein s is 2 and t is 3.
33. The compound of claim 28, which is a compound of formula (V-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
34. The compound of claim 33, wherein s is 2 and t is 2.
35. The compound of claim 28, which is a compound of formula (VI-a), (VI-B), or (VI-C):
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:
s is an integer of 1 to 6;
t is an integer from 1 to 6;
each R o Independently C 1 -C 6 An alkyl group; or two R o Forms a ring moiety with the nitrogen to which they are attached; or three R o Together with the nitrogen to which they are attached, form a bicyclic moiety; and
each R p Independently C 1 -C 6 Alkyl or-O- (C) 1 -C 6 An alkyl group); and
wherein each alkyl, cyclic moiety and bicyclic moiety is independently optionally substituted.
36. The compound of claim 35, wherein in formula (VI-a), s is 2; or in the formula (VI-B) or (VI-C), s is 2 and t is 2.
37. The compound of any one of claims 13 to 36, wherein R 1 And R is 2 Each independently is a straight chain C 6 -C 24 Alkyl, branched C 6 -C 24 Alkyl or straight-chain C 6 -C 24 Alkenyl groups; r is R a And R is d Each independently is H; and/or R b 、R c 、R e And R is f Each independently is n-hexyl or n-octyl.
38. The compound of claim 37, wherein R 1 And R is 2 Each independently is a straight chain C 6 -C 18 Alkyl, -R 7 -CH(R 8 )(R 9 ) Or C 6 -C 18 Alkenyl, wherein R is 7 Is C 0 -C 5 Alkylene group, and R 8 And R is 9 Independently C 2 -C 10 An alkyl group.
39. The compound of any one of claims 1 to 38, wherein R 3 Is methyl; t is-S-CH 2 -CH 2 -OH; and/or n is an integer from 5 to 20.
40. A compound of table 1 or table 2, or a pharmaceutically acceptable salt or stereoisomer thereof.
41. A composition comprising a compound of any one of claims 1 to 40 and a therapeutic or prophylactic agent.
42. A lipid nanoparticle comprising a compound according to any one of claims 1 to 40 or a composition according to claim 41.
43. A pharmaceutical composition comprising a compound according to any one of claims 1 to 40, a composition according to claim 41 or a lipid nanoparticle according to claim 42 and a pharmaceutically acceptable excipient or diluent.
CN202280007596.0A 2021-01-14 2022-01-12 Polymer conjugated lipid compounds and lipid nanoparticle compositions Pending CN116615472A (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CN2021100513727 2021-01-14
CN202110051372 2021-01-14
US202163140685P 2021-01-22 2021-01-22
US63/140685 2021-01-22
PCT/CN2022/071492 WO2022152141A2 (en) 2021-01-14 2022-01-12 Polymer conjugated lipid compounds and lipid nanoparticle compositions

Publications (1)

Publication Number Publication Date
CN116615472A true CN116615472A (en) 2023-08-18

Family

ID=81327156

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280007596.0A Pending CN116615472A (en) 2021-01-14 2022-01-12 Polymer conjugated lipid compounds and lipid nanoparticle compositions

Country Status (2)

Country Link
CN (1) CN116615472A (en)
WO (1) WO2022152141A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117257965A (en) * 2023-11-21 2023-12-22 深圳瑞吉生物科技有限公司 Nucleic acid delivery carrier composition and application thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024046448A1 (en) * 2022-09-02 2024-03-07 Suzhou Abogen Biosciences Co., Ltd. Lyophilized formulations and liquid formulations of lipid nanoparticles

Family Cites Families (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2309535A1 (en) 1997-11-12 1999-05-20 Brigham And Women's Hospital, Inc. The translation enhancer element of the human amyloid precursor protein gene
US7468275B2 (en) 2000-01-28 2008-12-23 The Scripps Research Institute Synthetic internal ribosome entry sites and methods of identifying same
BR0107943A (en) 2000-01-28 2003-01-28 Scripps Research Inst Synthetic Internal Ribosome Entry Sites and Methods of Identifying Them
EP1800697B1 (en) 2001-06-05 2010-04-14 CureVac GmbH Stabilised mRNA with increased G/C-content for use in gene therapy
DE10162480A1 (en) 2001-12-19 2003-08-07 Ingmar Hoerr The application of mRNA for use as a therapeutic agent against tumor diseases
DE10229872A1 (en) 2002-07-03 2004-01-29 Curevac Gmbh Immune stimulation through chemically modified RNA
DE10335833A1 (en) 2003-08-05 2005-03-03 Curevac Gmbh Transfection of blood cells with mRNA for immune stimulation and gene therapy
DE102004042546A1 (en) 2004-09-02 2006-03-09 Curevac Gmbh Combination therapy for immune stimulation
DE102005023170A1 (en) 2005-05-19 2006-11-23 Curevac Gmbh Optimized formulation for mRNA
JP5225087B2 (en) 2005-08-24 2013-07-03 ザ スクリプス リサーチ インスティチュート Translation enhancer element-dependent vector system
DE102006007433A1 (en) 2006-02-17 2007-08-23 Curevac Gmbh Immunostimulant adjuvant useful in vaccines against cancer or infectious diseases comprises a lipid-modified nucleic acid
JP2009544754A (en) 2006-07-28 2009-12-17 アプライド バイオシステムズ, エルエルシー Dinucleotide MRNA cap analog
EP2046954A2 (en) 2006-07-31 2009-04-15 Curevac GmbH NUCLEIC ACID OF FORMULA (I): GIXmGn, OR (II): CIXmCn, IN PARTICULAR AS AN IMMUNE-STIMULATING AGENT/ADJUVANT
DE102006051516A1 (en) 2006-10-31 2008-05-08 Curevac Gmbh (Base) modified RNA to increase the expression of a protein
DE102006061015A1 (en) 2006-12-22 2008-06-26 Curevac Gmbh Process for the purification of RNA on a preparative scale by HPLC
DE102007001370A1 (en) 2007-01-09 2008-07-10 Curevac Gmbh RNA-encoded antibodies
WO2008127688A1 (en) 2007-04-13 2008-10-23 Hart Communication Foundation Synchronizing timeslots in a wireless communication protocol
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
WO2009075886A1 (en) 2007-12-11 2009-06-18 The Scripps Research Institute Compositions and methods related to mrna translational enhancer elements
EP2176408B9 (en) 2008-01-31 2015-11-11 Curevac GmbH NUCLEIC ACIDS COMPRISING FORMULA (NuGiXmGnNv)a AND DERIVATIVES THEREOF AS AN IMMUNOSTIMULATING AGENTS /ADJUVANTS
WO2009127230A1 (en) 2008-04-16 2009-10-22 Curevac Gmbh MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION
PL215513B1 (en) 2008-06-06 2013-12-31 Univ Warszawski New borane phosphate analogs of dinucleotides, their application, RNA particle, method of obtaining RNA and method of obtaining peptides or protein
WO2010037408A1 (en) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
WO2010088927A1 (en) 2009-02-09 2010-08-12 Curevac Gmbh Use of pei for the improvement of endosomal release and expression of transfected nucleic acids, complexed with cationic or polycationic compounds
EP2281579A1 (en) 2009-08-05 2011-02-09 BioNTech AG Vaccine composition comprising 5'-Cap modified RNA
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
CN102906127A (en) * 2009-11-06 2013-01-30 华盛顿大学商业中心 Self-assembled particles from zwitterionic polymers and related methods
WO2011069529A1 (en) 2009-12-09 2011-06-16 Curevac Gmbh Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
EP2387999A1 (en) 2010-05-21 2011-11-23 CureVac GmbH Histidine-containing solution for transfection and/or injection of nucleic acids and uses thereof
US20130230884A1 (en) 2010-07-16 2013-09-05 John Chaput Methods to Identify Synthetic and Natural RNA Elements that Enhance Protein Translation
ES2558106T3 (en) 2010-07-30 2016-02-02 Curevac Ag Formation of nucleic acid complexes with disulfide-cross-linked cationic components for transfection and immunostimulation
WO2012019630A1 (en) 2010-08-13 2012-02-16 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
KR101648986B1 (en) 2010-11-17 2016-08-30 두산인프라코어 주식회사 Sliding bearing having improved lubrication property
WO2012089225A1 (en) 2010-12-29 2012-07-05 Curevac Gmbh Combination of vaccination and inhibition of mhc class i restricted antigen presentation
WO2012116715A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in newborns and infants
WO2012113413A1 (en) 2011-02-21 2012-08-30 Curevac Gmbh Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
WO2012116714A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in elderly patients
WO2013103659A1 (en) 2012-01-04 2013-07-11 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Stabilizing rna by incorporating chain-terminating nucleosides at the 3'-terminus
WO2013113325A1 (en) 2012-01-31 2013-08-08 Curevac Gmbh Negatively charged nucleic acid comprising complexes for immunostimulation
WO2013113326A1 (en) 2012-01-31 2013-08-08 Curevac Gmbh Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen
EP2623121A1 (en) 2012-01-31 2013-08-07 Bayer Innovation GmbH Pharmaceutical composition comprising a polymeric carrier cargo complex and an antigen
WO2013120499A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly (a) sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen
WO2013120500A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded tumour antigen
WO2013120497A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
WO2013120498A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded allergenic antigen or an autoimmune self-antigen
ES2654205T3 (en) 2012-03-27 2018-02-12 Curevac Ag Artificial nucleic acid molecules for enhanced protein or peptide expression
US9890391B2 (en) 2012-03-27 2018-02-13 Curevac Ag RNA vector with an open reading frame, an albumin 3′-UTR, and a histone stem loop
CA2866945C (en) 2012-03-27 2021-05-04 Curevac Gmbh Artificial nucleic acid molecules comprising a 5'top utr
EP2854857B1 (en) 2012-05-25 2018-11-28 CureVac AG Reversible immobilization and/or controlled release of nucleic acid containing nanoparticles by (biodegradable) polymer coatings
SG11201506052PA (en) 2013-02-22 2015-09-29 Curevac Gmbh Combination of vaccination and inhibition of the pd-1 pathway
WO2015002667A1 (en) 2013-07-01 2015-01-08 Myq, Inc. A location regulated point-of-sale system and enhancements
EP3033325B1 (en) 2013-07-23 2019-12-04 Arbutus Biopharma Corporation Compositions and methods for delivering messenger rna
SG10201801431TA (en) 2013-08-21 2018-04-27 Curevac Ag Respiratory syncytial virus (rsv) vaccine
WO2015024669A1 (en) 2013-08-21 2015-02-26 Curevac Gmbh Combination vaccine
CN105530952A (en) 2013-08-21 2016-04-27 库瑞瓦格股份公司 Composition and vaccine for treating lung cancer
CA2915712A1 (en) 2013-08-21 2015-02-26 Margit SCHNEE Rabies vaccine
BR112016000889A2 (en) 2013-08-21 2017-12-12 Curevac Ag composition and vaccine for prostate cancer treatment
ES2702466T3 (en) 2013-08-21 2019-03-01 Curevac Ag Procedure to increase the expression of proteins encoded by RNA
ES2806575T3 (en) 2013-11-01 2021-02-18 Curevac Ag Modified RNA with decreased immunostimulatory properties
SG10201805660WA (en) 2013-12-30 2018-08-30 Curevac Ag Methods for rna analysis
JP6584414B2 (en) 2013-12-30 2019-10-02 キュアバック アーゲー Artificial nucleic acid molecule
RU2717986C2 (en) 2013-12-30 2020-03-27 Куревак Аг Artificial molecules of nucleic acid
CA3179824A1 (en) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
AU2016253972B2 (en) 2015-04-27 2020-01-02 Acuitas Therapeutics Inc. Nucleoside-modified RNA for inducing an adaptive immune response
RS63051B1 (en) 2015-12-22 2022-04-29 Modernatx Inc Compounds and compositions for intracellular delivery of agents
IL243285A0 (en) * 2015-12-22 2016-04-21 Yeda Res & Dev Lipid analogs and liposomes comprising same
WO2018226854A2 (en) * 2017-06-06 2018-12-13 Wayne State University Antifouling polymer coatings and reverse coating method

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117257965A (en) * 2023-11-21 2023-12-22 深圳瑞吉生物科技有限公司 Nucleic acid delivery carrier composition and application thereof
CN117257965B (en) * 2023-11-21 2024-02-23 深圳瑞吉生物科技有限公司 Nucleic acid delivery carrier composition and application thereof

Also Published As

Publication number Publication date
WO2022152141A3 (en) 2022-08-11
WO2022152141A2 (en) 2022-07-21

Similar Documents

Publication Publication Date Title
US11472766B2 (en) Lipid nanoparticle composition
CN114391008A (en) Lipid compounds and lipid nanoparticle compositions
CN116615472A (en) Polymer conjugated lipid compounds and lipid nanoparticle compositions
US20230348362A1 (en) Lipid compounds and lipid nanoparticle compositions
WO2023056917A1 (en) Lipid compounds and lipid nanoparticle compositions
WO2023056914A1 (en) Lipid compounds and lipid nanoparticle compositions
TW202330457A (en) Lipid compounds and lipid nanoparticle compositions
CN116472275A (en) Lipid compounds and lipid nanoparticle compositions
WO2024022263A1 (en) Lipid compound and lipid nanoparticle composition
US11964052B2 (en) Lipid compounds and lipid nanoparticle compositions
CN116332830A (en) Lipid compounds and lipid nanoparticle compositions
CN114206463A (en) Lipid compounds and lipid nanoparticle compositions
CN117069785A (en) Lipid compounds and lipid nanoparticle compositions
AU2022358824A1 (en) Lipid compounds and lipid nanoparticle compositions
JP2024517529A (en) Lipid Compounds and Lipid Nanoparticle Compositions
CA3234156A1 (en) Lipid compounds and lipid nanoparticle compositions
CN116064598A (en) Nucleic acid vaccine for coronavirus

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination