RAPIDLY METABOLIZED LIPID COMPOUND

Information

  • Patent Application
  • 20250026713
  • Publication Number
    20250026713
  • Date Filed
    June 06, 2024
    8 months ago
  • Date Published
    January 23, 2025
    10 days ago
Abstract
Provided herein is a class of a rapidly metabolized lipid compound, and particularly relates to compounds represented by formula (I), or pharmaceutically acceptable salts, isotopic variants, tautomers, or stereoisomers thereof. Also provided is a nanoparticle pharmaceutical composition comprising said compound, and the application of said compound and its composition in the delivery of nucleic acids.
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

The present application claims the priority of the Chinese Patent Application No. 202310720951.5 filed on Jun. 16, 2023, Chinese Patent Application No. 202310723466.3 filed on Jun. 16, 2023, Chinese Patent Application No. 202410115561.X filed on Jan. 26, 2024, and Chinese Patent Application No. 202410437163.X filed on Apr. 11, 2024. The Chinese Patent Applications mentioned above are incorporated herein by reference as part of the disclosure of the present application.


TECHNICAL FIELD

The present disclosure relates to a novel class of ionizable cationic lipid compounds, or pharmaceutically acceptable salts, isotopic variants, tautomers, or stereoisomers thereof. The present disclosure also relates to lipid nanoparticles and pharmaceutical compositions comprising said compound, and the application of the lipid nanoparticles in the delivery of biologically active substances such as nucleic acids (e.g., mRNA, siRNA, ASO, and DNA).


BACKGROUND

Gene therapy refers to the introduction of exogenous genes into target cells to correct or compensate for genetic defects or abnormalities within the cell, so as to achieve the purpose of treatment. In the past few decades, research related to the treatment of clinical diseases through gene therapy has received more and more attention. Especially in recent years, siRNA-related drugs and mRNA vaccines have been approved by the FDA for clinical treatment, which has further promoted research and related investment in the field of gene therapy.


Nucleic acid substances are easily degraded by nucleases in organisms, and nucleic acid substances themselves are negatively charged, which makes it difficult for them to enter the cell through the cell membrane. As a nucleic acid delivery material, lipid nanoparticles (LNP) are one of the most important nucleic acid delivery systems with the advantages of easy to prepare, good biodegradability, no immunogenicity and good safety. The main components of LNP include cationic lipids, cholesterol, neutral lipids and polyethylene glycol-conjugated lipids. Among them, cationic lipid molecules are the core of LNP delivery system, and their molecular structure plays a decisive role in the delivery efficiency, targeting, and formulation stability, and the like, of the entire liposome nanoparticles.


Due to the different requirements of the delivery system for the delivery of different kinds of nucleic acid substances and the specific delivery of different targets, in order to meet the different needs of gene therapy, new lipid molecules need to be further developed.


SUMMARY

The present disclosure develops a new class of ionizable cationic lipid compounds that can be used to deliver various biologically active substances with high delivery efficiency.


The inventor's prior patent CN115850104A discloses that a class of cationic lipid compounds with a pair of geminal dialkyl in each tail has high delivery efficiency. Unexpectedly, after further research, it was found that when the geminal dialkyl structure was applied to cationic lipids containing central nitrogen atoms, the cationic lipids degraded slowly in the liver, making them suitable for applications with slow degradation. For some application scenarios that require accelerating metabolism in vivo, a new class of ionizable lipid compounds is developed.

    • the present disclosure provides a compound of formula (I) or a pharmaceutically acceptable salt, isotopic variant, tautomer or stereoisomer thereof:




embedded image




    • wherein each group is as defined herein.





In another aspect, the present disclosure provides a nanoparticle composition comprising lipid components, and optionally a load, wherein the lipid component comprises the compound of the present disclosure.


U.S. Ser. No. 11/246,933B1 discloses that incorporation of a biodegradable group into the tail chain of a lipid compound in a lipid nanoparticle results in faster metabolism and removal of the lipid from the body following delivery of the active agent to a target area. As a result, these lipids which contain the biodegradable groups have lower toxicity than similar lipids without the biodegradable groups. The tail chain of the cationic lipid compound of the present disclosure has biodegradable group(s), thereby has superior toxicity profile to similar lipids without biodegradable groups, such as DLin-MC3-DMA.


In another aspect, the present disclosure provides a pharmaceutical composition comprising a compound of the present disclosure or a nanoparticle composition of the present disclosure, and optionally pharmaceutically acceptable excipient(s), such as carrier(s), adjuvant(s) or vehicle(s).


In another aspect, the present disclosure provides use of the compound of the present disclosure, a nanoparticle composition of the present disclosure, or a pharmaceutical composition of the present disclosure in the manufacture of a medicament for treating, diagnosing, or preventing a disease. In one embodiment, the medicament for treating, diagnosing, or preventing a disease is a therapeutic or prophylactic mRNA vaccine.


In another aspect, the present disclosure provides use of the compound of the present disclosure, the nanoparticle composition of the present disclosure, or the pharmaceutical composition of the present disclosure in preparing a medicament for delivering a loading. In another aspect, the present disclosure provides the use of a compound of the present disclosure, a nanoparticle composition of the present disclosure, or a pharmaceutical composition of the present disclosure in the manufacture of a medicament for delivering a load.


In another aspect, the present disclosure provides a method of treating, diagnosing, or preventing a disease in a subject, comprising administering to the subject a compound of the present disclosure, a nanoparticle composition of the present disclosure, or a pharmaceutical composition of the present disclosure.


In another aspect, the present disclosure provides a compound of the present disclosure, a nanoparticle composition of the present disclosure, or a pharmaceutical composition of the present disclosure, for use in treating, diagnosing, and/or preventing a disease.


In another aspect, the present disclosure provides a method of delivering a load in a subject, comprising administering to the subject a compound of the present disclosure, a nanoparticle composition of the present disclosure, or a pharmaceutical composition of the present disclosure.


In another aspect, the present disclosure provides a compound of the present disclosure, a nanoparticle composition of the present disclosure, or a pharmaceutical composition of the present disclosure, for use in delivering a load.


In a specific embodiment, the load is selected from one or more of therapeutic agents, prophylactic agents, and diagnostic agents; alternatively, the therapeutic agent, prophylactic agent, or diagnostic agent is a nucleic acid.


In a more specific embodiment, the nucleic acid is selected from one or more of ASO, RNA and DNA.


In a more specific embodiment, the RNA is selected from one or more of small interfering RNA (siRNA), short hairpin RNA (shRNA), antisense RNA (aRNA), messenger RNA (mRNA), long non-coding RNA (lncRNA), microRNA (miRNA), small activating RNA (saRNA), multimeric coding nucleic acid (MCNA), polymeric coding nucleic acid (PCNA), guide RNA (gRNA), CRISPRRNA (crRNA), and a ribozyme, alternatively mRNA, and more alternatively modified mRNA.


Definitions
Chemical Definitions

Definitions of specific functional groups and chemical terms are described in more detail below.


When a range of values is listed, it is intended to encompass each value and sub-range within the range. For example, “C1-6 alkyl” includes C1, C2, C3, C4, C5, C6, C1-6, C1-5, C1-4, C1-3, C1-2, C2-6, C2-5, C2-4, C2-3, C3-6, C3-5, C3-4, C4-6, C4-5, and C5-6 alkyl.


“C1-20 alkyl” refers to a linear or branched saturated hydrocarbon group having 1 to 20 carbon atoms. In some embodiments, C4-20 alkyl, C6-14 alkyl, C7-12 alkyl, C8-12 alkyl, C9-12 alkyl, C4-10 alkyl, C7-11 alkyl, C8-11 alkyl, C9-11 alkyl, C10-11 alkyl, C6-10 alkyl, C7-10 alkyl, C8-10 alkyl, C9-10 alkyl, C10 alkyl, C8-9 alkyl, C4-9 alkyl, C6-9 alkyl, C7-9 alkyl, C9 alkyl, C10 alkyl, C11 alkyl, C2-8 alkyl, C4-8 alkyl, C5-8 alkyl, C6-8 alkyl, C7-8 alkyl, C8 alkyl, C6-7 alkyl, C7 alkyl, C4-6 alkyl, C1-20 alkyl, C1-14 alkyl, C2-14 alkyl, C1-13 alkyl, C1-12 alkyl, C1-10 alkyl, C1-9 alkyl, C1-8 alkyl, C1-7 alkyl, C2-7 alkyl, C1-6 alkyl, C2-6 alkyl, C1-5 alkyl, C5 alkyl, C1-4 alkyl, C2-4 alkyl, C1-3 alkyl, C2-3 alkyl, C1-2 alkyl, and Me are preferred. Examples of C1-6 alkyl include: methyl (C1), ethyl (C2), n-propyl (C3), isopropyl (C3), n-butyl (C4), tert-butyl (C4), sec-butyl (C4), isobutyl (C4), n-pentyl (C5), 3-pentyl (C5), pentyl (C5), neopentyl (C5), 3-methyl-2-butyl (C5), tert-pentyl (C5), and n-hexyl (C6). The term “C1-6 alkyl” also includes heteroalkyl, wherein one or more (e.g., 1, 2, 3, or 4) carbon atoms are substituted with heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, or phosphorus). An alkyl group may optionally be substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent. Conventional abbreviations of alkyl groups include: Me (—CH3), Et (—CH2CH3), iPr (—CH(CH3)2), nPr (—CH2CH2CH3), n-Bu (—CH2CH2CH2CH3), or i-Bu (—CH2CH(CH3)2). In some embodiments, alkyl is preferably linear alkyl.


“C2-13 alkenyl” refers to a linear or branched hydrocarbon group having 2 to 13 carbon atoms and at least one carbon-carbon double bond. “C4-20 alkenyl” refers to a linear or branched hydrocarbon group having 4 to 20 carbon atoms and at least one carbon-carbon double bond. In some embodiments, C4-14 alkenyl, C6-14 alkenyl, C7-12 alkenyl, C4-10 alkenyl, C2-10 alkenyl, C2-9 alkenyl, C2-6 alkenyl, and C2-4 alkenyl are preferred. Examples of C2-6 alkenyl include: ethenyl (C2), 1-propenyl (C3), 2-propenyl (C3), 1-butenyl (C4), 2-butenyl (C4), butadienyl (C4), pentenyl (C5), pentadienyl (C5), hexenyl (C6), etc. The term “C2-6 alkenyl” also includes heteroalkenyl where one or more (e.g., 1, 2, 3, or 4) carbon atoms are replaced with heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, or phosphorus). An alkenyl group may optionally be substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.


“C2-13 alkynyl” refers to a linear or branched hydrocarbon group having 2 to 13 carbon atoms, at least one carbon-carbon triple bond, and optionally one or more carbon-carbon double bonds. “C4-20 alkynyl” refers to a linear or branched hydrocarbon group having 4 to 20 carbon atoms, at least one carbon-carbon triple bond, and optionally one or more carbon-carbon double bonds. In some embodiments, C4-14 alkynyl, C6-14 alkynyl, C7-12 alkynyl, C4-10 alkynyl, C2-10 alkynyl, C2-9 alkynyl, C2-6 alkynyl, and C2-4 alkynyl are preferred. Examples of C2-6 alkynyl include, but are not limited to: ethynyl (C2), 1-propynyl (C3), 2-propynyl (C3), 1-butynyl (C4), 2-butynyl (C4), pentynyl (C5), hexynyl (C6), and the like. The term “C2-6 alkynyl” also includes heteroalkynyl where one or more (e.g., 1, 2, 3, or 4) carbon atoms are replaced with heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, or phosphorus). An alkynyl group may optionally be substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.


“C1-20 alkylene” refers to a bivalent group formed by removing another hydrogen of C1-20 alkyl, and can be substituted or unsubstituted. In some embodiments, C4-20 alkylene, C6-14 alkylene, C7-12 alkylene, C8-12 alkylene, C4-10 alkylene, C7-11 alkylene, C8-11 alkylene, C8-10 alkylene, C9-10 alkylene, C8-9 alkylene, C4-9 alkylene, C6-9 alkylene, C7-9 alkylene, C9 alkylene, C2-8 alkylene, C5-8 alkylene, C7-8 alkylene, C4-6 alkylene, C1-2 alkylene, C1-14 alkylene, C2-14 alkylene, C1-13 alkylene, C1-12 alkylene, C1-10 alkylene, C1-9 alkyl, C1-8 alkylene, C1-7 alkylene, C2-7 alkylene, C1-6 alkylene, C2-6 alkylene, C1-5 alkylene, C5 alkylene, C1-4 alkylene, C2-4 alkylene, C1-3 alkylene, C2-3 alkylene, C1-2 alkylene, and methylene are preferred. Unsubstituted alkylene groups include, but are not limited to: methylene (—CH2—), ethylene (—CH2CH2—), propylene (—CH2CH2CH2—), butylene (—CH2CH2CH2CH2—), pentylene (—CH2CH2CH2CH2CH2—), hexylene (—CH2CH2CH2CH2CH2CH2—), etc. Exemplary of substituted alkylene groups, such as those substituted with one or more alkyl (methyl) groups, include, but are not limited to: substituted methylene (—CH(CH3)—, and —C(CH3)2—), substituted ethylene (—CH(CH3)CH2—, —CH2CH(CH3)—, —C(CH3)2CH2—, —CH2C(CH3)2), substituted propylene (—CH(CH3)CH2CH2—, —CH2CH(CH3)CH2—, —CH2CH2CH(CH3)—, —C(CH3)2CH2CH2—, —CH2C(CH3)2CH2—, —CH2CH2C(CH3)2—), etc.


“C2-13 alkenylene” refers to a bivalent group formed by removing another hydrogen of C2-13 alkenyl, and can be substituted or unsubstituted. “C4-14 alkenylene” refers to a bivalent group formed by removing another hydrogen of C4-14 alkenyl, and can be substituted or unsubstituted. In some embodiments, C6-4 alkenylene, C4-10 alkenylene, C2-10 alkenylene, C2-9 alkenylene, C2-6 alkenylene, and C2-4 alkenylene are particularly preferred. Exemplary unsubstituted forms of the alkenylene include, but are not limited to: ethenylene (—CH═CH—) and propenylene (e.g., —CH═CHCH2— and —CH2—CH═CH—). Exemplary substituted alkenylene groups, for example, alkenylene substituted with one or more alkyl (methyl), include, but are not limited to: substituted ethenylene (—C(CH3)═CH— and —CH═C(CH3)—), substituted propenylene (—C(CH3)═CHCH2—, —CH═C(CH3)CH2—, —CH═CHCH(CH3)—, —CH═CHC(CH3)2—, —CH(CH3)—CH═CH—, —C(CH3)2—CH═CH—, —CH2—C(CH3)═CH—, —CH2—CH═C(CH3)—), etc.


“C2-13 alkynylene” refers to a bivalent group formed by removing another hydrogen of C2-13 alkynyl, and can be substituted or unsubstituted. “C4-14 alkynylene” refers to a bivalent group formed by removing another hydrogen of C4-14 alkynyl, and can be substituted or unsubstituted. In some embodiments, C6-14 alkynylene, C4-10 alkynylene, C2-10 alkynylene, C2-9 alkynylene, C2-6 alkynylene, and C2-4 alkynylene are particularly preferred. Exemplary alkynylene groups include, but are not limited to: ethynylene (—C≡C—), substituted or unsubstituted propynylene (—C≡CCH2—), etc.


“C0-6 alkylene” refers to a chemical bond and the “C1-6 alkylene” described above. “C0-4 alkylene” refers to a chemical bond and the “C1-4 alkylene” described above.


The term “variable A and variable B have a total length of x carbon atoms” means that the total number of carbon atoms of the main chain in the group represented by variable A and the number of carbon atoms of the main chain in the group represented by variable B is x.


The term “the substitution site of R1s on R2 is separated from M2 by x carbon atoms” means that the sum of the number of carbon atoms (including N atoms replaced by —NR′—) between the sites substituted by variable R1s on variable R2, and so on for other situations. For example:




embedded image


in compound 11b, the substitution site of R1s on R2 is separated from M2 by 3 carbon atoms.


“Halo” or “halogen” refers to fluorine (F), chlorine (Cl), bromine (Br), and iodine (I).


Thus, “C1-10 haloalkyl” refers to the above “C1-10 alkyl”, which is substituted with one or more halogen. In some embodiments, C1-8 haloalkyl, C1-6 haloalkyl, C1-4 haloalkyl, and C1-3 haloalkyl are particularly preferred, still alternatively C1-2 haloalkyl. Exemplary haloalkyl groups include, but are not limited to: —CF3, —CH2F, —CHF2, —CHFCH2F, —CH2CHF2, —CF2CF3, —CCl3, —CH2Cl, —CHCl2, 2,2,2-trifluoro-1,1-dimethyl-ethyl, etc. A haloalkyl group can be substituted at any accessible point of attachment with, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.


“C3-14 cycloalkyl” or “3- to 14-membered cycloalkyl” refers to a radical of a non-aromatic cyclic hydrocarbon group having 3 to 14 ring carbon atoms and zero heteroatoms, optionally containing 1, 2, or 3 double or triple bonds. In some embodiments, 3- to 10-membered cycloalkyl, 5- to 10-membered cycloalkyl, 3- to 8-membered cycloalkyl, 3- to 7-membered cycloalkyl, and 3- to 6-membered cycloalkyl are particularly preferred, still alternatively 5- to 7-membered cycloalkyl, 4- to 6-membered cycloalkyl, 3- to 5-membered cycloalkyl, 3- to 4-membered cycloalkyl, and 5- to 6-membered cycloalkyl, still alternatively 5-membered cycloalkyl, still alternatively 6-membered cycloalkyl, and still alternatively cyclopropyl. The cycloalkyl also includes a ring system in which the cycloalkyl ring described above is fused with one or more aryl or heteroaryl groups, wherein the point of attachment is on the cycloalkyl ring, and in such case, the number of carbon atoms continues to represent the number of carbon atoms in the cycloalkyl system. The cycloalkyl further comprises the cycloalkyl described above, in which the substituents on any non-adjacent carbon atoms are connected to form a bridged ring, together forming a polycyclic alkane sharing two or more carbon atoms. The cycloalkyl further comprises the cycloalkyl described above, in which the substituents on the same carbon atom are connected to form a ring, together forming a polycyclic alkane sharing one carbon atom. Exemplary cycloalkyl groups include, but are not limited to: cyclopropyl (C3), cyclopropenyl (C3), cyclobutyl (C4), cyclobutenyl (C4), cyclopentyl (C5), cyclopentenyl (C5), cyclohexyl (C6), cyclohexenyl (C6), cyclohexadienyl (C6), cycloheptyl (C7), cycloheptenyl (C7), cycloheptadienyl (C7), cycloheptatrienyl (C7), etc. A cycloalkyl can optionally be substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.


“C3-14 cycloalkylene” refers to a bivalent group formed by removing another hydrogen of C3-14 cycloalkyl and may be substituted or unsubstituted. In some embodiments, C3-10 cycloalkylene, C3-7 cycloalkylene, C3-6 cycloalkylene, C3-5 cycloalkylene, and C3-4 cycloalkylene are particularly preferred, even still alternatively cyclopropylene.


“3-14 membered heterocyclyl” or “3- to 14-membered heterocyclyl” refers to a saturated or unsaturated group of a 3- to 14-membered non-aromatic ring system having ring carbon atoms and 1 to 5 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon, optionally wherein 1, 2 or 3 double or triple bonds are contained. In a heterocyclyl containing one or more nitrogen atoms, the point of attachment can be a carbon atom or a nitrogen atom as long as the valence permits. In some embodiments, 3- to 10-membered heterocyclyl is preferred, which is a radical of 3- to 10-membered non-aromatic ring system having ring carbon atoms and 1 to 5 ring heteroatoms; in some embodiments, 5- to 10-membered heterocyclyl is preferred, which is a radical of 5- to 10-membered non-aromatic ring system having ring carbon atoms and 1 to 5 ring heteroatoms; in some embodiments, 3- to 8-membered heterocyclyl is preferred, which is a radical of 3- to 8-membered non-aromatic ring system having ring carbon atoms and 1 to 4 ring heteroatoms; in some embodiments, 3- to 7-membered heterocyclyl is preferred, which is a radical of 3- to 7-membered non-aromatic ring system having ring carbon atoms and 1 to 4 ring heteroatoms; 5- to 7-membered heterocyclyl is preferred, which is a radical of 5- to 7-membered non-aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms; 3- to 6-membered heterocyclyl is preferred, which is a radical of 3- to 6-membered non-aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms; 4- to 6-membered heterocyclyl is preferred, which is a radical of 4- to 6-membered non-aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms; 5- to 6-membered heterocyclyl is more preferred, which is a radical of 5- to 6-membered non-aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms; 5-membered heterocyclyl is preferred, which is a radical of 5-membered non-aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms; 6-membered heterocyclyl is preferred, which is a radical of 6-membered non-aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms. The heterocyclyl also includes a ring system wherein the heterocyclyl described above is fused with one or more cycloalkyl groups, wherein the point of attachment is on the heterocyclyl ring, or the heterocyclyl described above is fused with one or more aryl or heteroaryl groups, wherein the point of attachment is on the heterocyclyl ring; and in such cases, the number of ring members continues to represent the number of ring members in the heterocyclyl ring system. The heterocyclyl further comprises the heterocyclyl described above, in which the substituents on any non-adjacent carbon or nitrogen atoms are connected to form a bridge ring, together forming a polycyclic heteroalkane sharing two or more carbon or nitrogen atoms. The heterocyclyl further comprises the heterocyclyl described above, in which the substituents on the same carbon atom are connected to form a ring, together forming a polycyclic heteroalkane sharing one carbon atom. Exemplary 3-membered heterocyclyl groups containing one heteroatom include, but are not limited to: aziridinyl, oxiranyl, and thiorenyl. Exemplary 4-membered heterocyclyl groups containing one heteroatom include, but are not limited to: azetidinyl, oxetanyl, and thietanyl. Exemplary 5-membered heterocyclyl groups containing one heteroatom include, but are not limited to: tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl, and pyrrolyl-2,5-dione. Exemplary 5-membered heterocyclyl groups containing two heteroatoms include, but are not limited to: pyrazolidinyl, dioxolanyl, oxasulfuranyl, disulfuranyl, and oxazolidin-2-one. Exemplary 5-membered heterocyclyl groups containing three heteroatoms include, but are not limited to: triazolinyl, oxadiazolinyl, and thiadiazolinyl. Exemplary 6-membered heterocyclyl groups containing one heteroatom include, but are not limited to: piperidinyl, tetrahydropyranyl, dihydropyridinyl, and thianyl. Exemplary 6-membered heterocyclyl groups containing two heteroatoms include, but are not limited to: piperazinyl, morpholinyl, dithianyl, and dioxanyl. Exemplary 6-membered heterocyclyl groups containing three heteroatoms include, but are not limited to: triazinanyl. Exemplary 7-membered heterocyclyl groups containing one heteroatom include, but are not limited to: azepanyl, oxepanyl, and thiepanyl. Exemplary 5-membered heterocyclyl groups fused with a C6 aryl ring (also referred to as 5,6-bicyclic heterocyclyl herein) include, but are not limited to: indolinyl, isoindolinyl, dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolinonyl, and the like. Exemplary 6-membered heterocyclyl groups fused with a C6 aryl ring (also referred to as 6,6-bicyclic heterocyclyl herein) include, but are not limited to: tetrahydroquinolinyl, tetrahydroisoquinolinyl, etc. Heterocyclyl also includes the above heterocyclyl that shares one or two atoms with one cycloalkyl, heterocyclyl, aryl, or heteroaryl to form a bridged or spiro ring, where the shared atom may be a carbon or nitrogen atom as long as the valency permits. The heterocyclyl further includes the heterocyclyl described above, which optionally can be substituted with one or more substituents, e.g., with 1 to 5 substituents, 1 to 3 substituents or 1 substituent.


“C6-10 aryl” refers to a radical of monocyclic or polycyclic (e.g., bicyclic) 4n+2 aromatic ring system having 6-10 ring carbon atoms and zero heteroatoms (e.g., having 6 or 10 shared π electrons in a cyclic array). In some embodiments, an aryl group has six ring carbon atoms (“C6 aryl”; e.g., phenyl). In some embodiments, an aryl group has ten ring carbon atoms (“C10 aryl”; e.g., naphthyl such as 1-naphthyl and 2-naphthyl). The aryl group also includes a ring system in which the aryl ring described above is fused with one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on the aryl ring, in which case the number of carbon atoms continues to represent the number of carbon atoms in the aryl ring system. The aryl can be substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents or 1 substituent.


“5- to 14-membered heteroaryl” refers to a radical of 5- to 14-membered monocyclic or bicyclic 4n+2 aromatic ring system (e.g., having 6, 10 or 14 shared n electrons in a cyclic array) having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen and sulfur. In the heteroaryl group containing one or more nitrogen atoms, the point of attachment can be a carbon or nitrogen atom as long as the valence permits, heteroaryl bicyclic systems may include one or more heteroatoms in one or two rings, heteroaryl also includes ring systems wherein the heteroaryl ring described above is fused with one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on the heteroaryl ring. In such case, the number the carbon atoms continues to represent the number of carbon atoms in the heteroaryl ring system. In some embodiments, 5- to 10-membered heteroaryl groups are alternative, which are radicals of 5- to 10-membered monocyclic or bicyclic 4n+2 aromatic ring systems having ring carbon atoms and 1-4 ring heteroatoms. In other embodiments, 5- to 6-membered heteroaryl groups are yet alternative, which are radicals of 5- to 6-membered monocyclic or bicyclic 4n+2 aromatic ring systems having ring carbon atoms and 1-4 ring heteroatoms. Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to, pyrrolyl, furyl and thienyl. Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl. Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to, triazolyl, oxadiazolyl (such as, 1,2,4-oxadiazolyl), and thiadiazolyl. Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to, tetrazolyl. Exemplary 6-membered heteroaryl groups containing one heteroatom include, but are not limited to, pyridyl or pyridonyl. Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to, pyridazinyl, pyrimidinyl, and pyrazinyl. Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to, triazinyl and tetrazinyl, respectively. Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to, azepinyl, oxepinyl, and thiepinyl. Exemplary 5,6-bicyclic heteroaryl groups include, but are not limited to, indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothiophenyl, isobenzothiophenyl, benzofuranyl, benzoisofuranyl, benzimidazolyl, benzoxazolyl, benzoisoxazolyl, benzoxadiazolyl, benzothiazolyl, benzoisothiazolyl, benzothiadiazolyl, indolizinyl and purinyl. Exemplary 6,6-bicyclic heteroaryl groups include, but are not limited to, naphthyridinyl, pteridinyl, quinolyl, isoquinolyl, cinnolinyl, quinoxalinyl, phthalazinyl and quinazolinyl. The heteroaryl can be substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents or 1 substituent.


“Hydroxyalkyl” refers to an alkyl group that is substituted with one or more hydroxy groups.


“Alkoxy” refers to an oxyether form of a linear or branched-chain alkyl group, i.e., an —O-alkyl group. Similarly, “methoxy” refers to —O—CH3.


“Optionally substituted with . . . ” means, it can be substituted with the specified substituents or unsubstituted.


Bivalent groups formed by removing another hydrogen from the groups defined above such as alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl are collectively referred to as “ylene”. Ring-forming groups such as cycloalkyl, heterocyclyl, aryl, and heteroaryl are collectively referred to as “cyclic groups”.


Alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, etc, as defined herein are optionally substituted groups.


Exemplary substituents on carbon atoms include, but are not limited to: halogen, —CN, —NO2, —N3, —SO2H, —SO3H, —OH, —ORaa, —ON(Rbb)2, —N(Rbb)2, —N(Rbb)3+X, —N(ORcc)Rbb, —SH, —SRaa, —SSRcc, —C(═O)Raa, —CO2H, —CHO, —C(ORcc)2, —CO2Raa, —OC(═O)Raa, —OCO2Raa, —C(═O)N(Rbb)2, —OC(═O)N(Rbb)2, —NRbbC(═O)Raa, —NRbbCO2Raa, —NRbbC(═O)N(Rbb)2, —C(═NRbb)Raa, —C(═NRbb)ORaa, —OC(═NRbb)Raa, —OC(═NRbb)ORaa, —C(═NRbb)N(Rbb)2, —OC(═NRbb)N(Rbb)2, —NRbbC(═NRbb)N(Rbb)2, —C(═O)NRbbSO2Raa, —NRbbSO2Raa, —SO2N(Rbb)2, —SO2Raa, —SO2ORaa, —OSO2Raa, —S(═O)Raa, —OS(═O)Raa, —Si(Raa)3, —OSi(Raa)3, —C(═S)N(Rbb)2, —C(═O)SRaa, —C(═S)SRaa, —SC(═S)SRaa, —SC(═O)SRaa, —OC(═O)SRaa, —SC(═O)ORaa, —SC(═O)Raa, —P(═O)2Raa, —OP(═O)2Raa, —P(═O)(Raa)2, —OP(═O)(Raa)2, —OP(═O)(ORbb)2, —P(═O)2N(Rbb), —OP(═O)2N(Rbb)2, —P(═O)(NRbb)2, —OP(═O)(NRbb)2, —NRbbP(═O)(ORcc)2, —NRbbP(═O)(NRbb)2, —P(Rbb)2, —P(Rcc)3, —OP(Rcc)2, —OP(Rcc)3, —B(Raa)2, —B(ORcc)2, —BRaa(ORcc), alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein each of the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rdd groups;

    • or two geminal hydrogens on a carbon atom are replaced with the group ═O, ═S, ═NN(Rbb)2, =NNRbbC(═O)Raa, ═NNRbbC(═O)ORaa, =NNRbbS(═O)2Raa, =NRbb, or =NORcc;
    • each of the Raa is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or two Raa groups are joined to form a heterocyclyl or heteroaryl ring, wherein each of the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rdd groups;
    • each of the Rbb is independently selected from hydrogen, —OH, —ORaa, —N(Rcc)2, —CN, —C(═O)Raa, —C(═O)N(Rcc)2, —CO2Raa, —SO2Raa, —C(═NRcc)ORaa, —C(═NRcc)N(Rcc)2, —SO2N(Rcc)2, —SO2Rcc, —SO2ORcc, —SORaa, —C(═S)N(Rcc)2, —C(═O)SRcc, —C(═S)SRcc, —P(═O)2Raa, —P(═O)(Raa)2, —P(═O)2N(Rcc)2, —P(═O)(NRcc)2, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or two Rbb groups are combined to form a heterocyclyl or heteroaryl ring, wherein each of the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rdd groups;
    • each of the Rcc is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or two Rcc groups are joined to form a heterocyclyl or heteroaryl ring, wherein each of the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rdd groups;
    • each of the Rdd is independently selected from: halogen, —CN, —NO2, —N3, —SO2H, —SO3H, —OH, —ORee, —ON(Rff)2, —N(Rff)2, —N(Rff)3+X, —N(ORee)Rff, —SH, —SRee, —SSRee, —C(═O)Ree, —CO2H, —CO2Ree, —OC(═O)Ree, —OCO2Ree, —C(═O)N(Rff)2, —OC(═O)N(Rff)2, —NRffC(═O)Ret, —NRffCO2Ree, —NRffC(═O)N(Rff)2, —C(═NRff)ORee, —OC(═NRff)Ree, —OC(═NRff)ORee, —C(═NRff)N(Rff)2, —OC(═NRff)N(Rff)2, —NRffC(═NRff)N(Rff)2, —NRffSO2Ree, —SO2N(Rff)2, —SO2Ree, —SO2ORee, —OSO2Ree, —S(═O)Ree, —Si(Ree)3, —OSi(Ree)3, —C(═S)N(Rff)2, —C(═O)SRee, —C(═S)SRee, —SC(═S)SRee, —P(═O)2Ree, —P(═O)(Ree)2, —OP(═O)(Ree)2, —OP(═O)(ORee)2, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein each of the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rgg groups, or two geminal Rdd substituents may be combined to form ═O or ═S;
    • each of the Ree is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclyl, and heteroaryl, wherein each of the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rgg groups;
    • each of the Rff is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or two Rff groups are combined to form a heterocyclyl or heteroaryl ring, wherein each of the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rgg groups;
    • each of the Rgg is independently: halogen, —CN, —NO2, —N3, —SO2H, —SO2H, —OH, —OC1-6 alkyl, —ON(C1-6 alkyl)2, —N(C1-6 alkyl)2, —N(C1-6 alkyl)3+X, —NH(C1-6 alkyl)2+X, —NH2(C1-6 alkyl)+X, —NH3X, —N(OC1-6 alkyl)(C1-6 alkyl), —N(OH)(C1-6 alkyl), —NH(OH), —SH, —SC1-6 alkyl, —SS(C1-6 alkyl), —C(═O)(C1-6 alkyl), —CO2—H, —CO2(C1-6 alkyl), —OC(═O)(C1-6 alkyl), —OCO2(C1-6 alkyl), —C(═O)NH2, —C(═O)N(C1-6 alkyl)2, —OC(═O)NH(C1-6 alkyl), —NHC(═O)(C1-6 alkyl), —N(C1-6 alkyl)C(═O)(C1-6 alkyl), —NHCO2(C1-6 alkyl), —NHC(═O)N(C1-6 alkyl)2, —NHC(═O)NH(C1-6 alkyl), —NHC(═O)NH2, —C(═NH)O(C1-6 alkyl), —OC(═NH)(C1-6 alkyl), —OC(═NH)OC1-6 alkyl, —C(═NH)N(C1-6 alkyl)2, —C(═NH)NH(C1-6 alkyl), —C(═NH)NH2, —OC(═NH)N(C1-6 alkyl)2, —OC(NH)NH(C1-6 alkyl), —OC(NH)NH2, —NHC(NH)N(C1-6 alkyl)2, —NHC(═NH)NH2, —NHSO2(C1-6 alkyl), —SO2N(C1-6 alkyl)2, —SO2NH(C1-6 alkyl), —SO2NH2, —SO2C1-6 alkyl, —SO2OC1-6 alkyl, —OSO2C1-6 alkyl, —SOC1-6 alkyl, —SC(C1-6 alkyl)3, —OSi(C1-6 alkyl)3, —C(═S)N(C1-6 alkyl)2, C(═S)NH(C1-6 alkyl), C(═S)NH2, —C(═O)S(C1-6 alkyl), —C(═S)SC1-6 alkyl, —SC(═S)SC1-6 alkyl, —P(═O)2(C1-6 alkyl), —P(═O)(C1-6 alkyl)2, —OP(═O)(C1-6 alkyl)2, —OP(═O)(OC1-6 alkyl)2-, C1-6 alkyl, C1-6 haloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C7 cycloalkyl, C6-C10 aryl, 3-7 membered heterocyclyl, or 5-10 membered heteroaryl; or two geminal Rgg substituents may be combined to form ═O or ═S;
    • wherein, X is a counter ion.
    • exemplary substituents on nitrogen atoms include, but are not limited to: hydrogen, —OH, —ORaa—, —N(Rcc)2, —CN, —C(═O)Raa, —C(═O)N(Rcc)2, —CO2Raa, —SO2Raa, —C(═NRbb)Raa, —C(═NRcc)ORaa, —C(═NRcc)N(Rcc)2, —SO2N(Rcc)2, —SO2Rcc, —SO2ORcc, —SORaa, —C(═S)N(Rcc)2, —C(═O)SRcc, —C(═S)SRcc, P(═O)2Raa, —P(═O)(Raa)2, —P(═O)2N(Rcc)2, —P(═O)(NRcc)2, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or two RV groups attached to a nitrogen atom are combined to form a heterocyclyl or heteroaryl ring, wherein each of the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rdd groups, and wherein Raa, Rbb, Rcc, and Rdd are as described above.


“Nucleic acids” refers to single- or double-stranded deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) molecules and their heterozygous molecules. Examples of nucleic acid molecules include, but are not limited to, messenger RNA (mRNA), microRNA (miRNA), small interfering RNA (siRNA), self-amplified RNA (saRNA), and antisense oligonucleotides (ASO), etc. Nucleic acids may be further chemically modified, and the chemical modifier selected from one of, or a combination of: pseudouridine, N1-methyl-pseudouridine, 5-methoxyuridine, 5-methylcytosine. mRNA molecules contain protein coding regions and may further contain expression regulatory sequences. Typical expression regulatory sequences include, but are not limited to, 5′ cap, 5′ untranslated region (5′ UTR), 3′ untranslated region (3′ UTR), polyadenylate sequence (PolyA), miRNA binding sites.


“Cationic lipid” refers to a lipid molecule capable of being positively charged at physiological pH. In some embodiments, the cationic lipid is an amino lipid.


“Neutral lipid” refers to a lipid molecule that is not charged at specific pH, e.g., physiological pH. Examples of neutral lipids include, but are not limited to, 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2-dimyristoyl-sn-glycero-3-phosphoethanolamine (DMPE), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE), and 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine (DPPE).


“Structure lipids” refers to lipids that enhance the stability of nanoparticles by filling the gaps between lipids, commonly such as steroids. The steroid is a compound having a perhydrocyclopentanophenanthrene carbon framework. In an alternative embodiment, the steroid is selected from cholesterol, sitosterol, coprosterol, fucosterol, brassicasterol, ergosterol, tomatine, ursolic acid, α-tocopherol, stigmasterol, avenasterol, ergocalciferol and campesterol.


“Polymer lipids” refers to molecules containing a polymer moiety and a lipid moiety. In some embodiments, the polymer lipid is a polyethylene glycol (PEG) lipid. Other lipids that can reduce aggregation, such as products of compounds having uncharged, hydrophilic, space-barrier moieties coupled with lipid may also be used.


“Lipid nanoparticles” refers to particles containing lipid components of nanoscale size.


“Biodegradable groups” refers to functional groups that contain biodegradable bonds, such as esters, disulfide bonds and amides, etc. Biodegradation may affect the process of removing compounds from the body. The biodegradable groups of the present disclosure are oriented from the head to the tail in ionizable lipid molecules.


Other Definitions

The term “treating” as used herein relates to reversing, alleviating or inhibiting the progression or prevention of the disorders or conditions to which the term applies, or of one or more symptoms of such disorders or conditions. The noun “treatment” as used herein relates to the action of treating, which is a verb, and the latter is as just defined.


The term “pharmaceutically acceptable salt” as used herein refers to those carboxylate and amino acid addition salts of the compounds of the present disclosure, which are suitable for the contact with patients' tissues within a reliable medical judgment, and do not produce inappropriate toxicity, irritation, allergy, etc. They are commensurate with a reasonable benefit/risk ratio, and are effective for their intended use. The term includes, if possible, the zwitterionic form of the compounds of the disclosure.


The pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali metal and alkaline earth metal hydroxides or organic amines. Examples of the metals used as cations include sodium, potassium, magnesium, calcium, etc. Examples of suitable amines are N,N′-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine and procaine.


The base addition salt of the acidic compound can be prepared by contacting the free acid form with a sufficient amount of the required base to form a salt in a conventional manner. The free acid can be regenerated by contacting the salt form with an acid in a conventional manner and then isolating the free acid. The free acid forms are somewhat different from their respective salt forms in their physical properties, such as solubility in polar solvents. But for the purposes of the present disclosure, the salts are still equivalent to their respective free acids.


The salts can be prepared from the inorganic acids, which include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, nitrates, phosphates, monohydrogen phosphates, dihydrogen phosphates, metaphosphates, pyrophosphates, chlorides, bromides and iodides. Examples of the acids include hydrochloric acid, nitric acid, sulfuric acid, hydrobromic acid, hydroiodic acid, phosphoric acid, etc. The representative salts include hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthalate, methanesulfonate, glucoheptanate, lactobionate, lauryl sulfonate, isethionate, etc. The salts can also be prepared from the organic acids, which include aliphatic monocarboxylic and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxyalkanoic acids, alkanedioic acid, aromatic acids, aliphatic and aromatic sulfonic acids, etc. The representative salts include acetate, propionate, octanoate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, mandelate, benzoate, chlorobenzoate, methyl benzoate, dinitrobenzoate, naphthoate, besylate, tosylate, phenylacetate, citrate, lactate, maleate, tartrate, methanesulfonate, etc. The pharmaceutically acceptable salts can include cations based on alkali metals and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, etc., as well as non-toxic ammonium, quaternary ammonium, and amine cations including, but not limited to, ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, etc. Salts of amino acids are also included, such as arginine salts, gluconates, galacturonates, etc. (for example, see Berge S. M. et al., “Pharmaceutical Salts,” J. Pharm. Sci., 1977; 66: 1-19 for reference).


“Subjects” to which administration is contemplated include, but are not limited to, humans (e.g., males or females of any age group, e.g., paediatric subjects (e.g., infants, children, adolescents) or adult subjects (e.g., young adults, middle-aged adults or older adults) and/or non-human animals, such as mammals, e.g., primates (e.g., cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses, sheep, goats, rodents, cats and/or dogs. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human animal. The terms “human”, “patient” and “subject” can be used interchangeably herein.


“Disease”, “disorder”, and “condition” can be used interchangeably herein.


Unless otherwise indicated, the term “treatment” as used herein includes the effect on a subject who is suffering from a particular disease, disorder, or condition, which reduces the severity of the disease, disorder, or condition, or delays or slows the progression of the disease, disorder or condition (“therapeutic treatment”). The term also includes the effect that occurs before the subject begins to suffer from a specific disease, disorder or condition (“prophylactic treatment”).


Generally, the “effective amount” of a pharmaceutical composition refers to an amount sufficient to elicit a target biological response. As understood by those skilled in the art, the effective amount of the pharmaceutical composition of the disclosure can vary depending on the following factors, such as the desired biological endpoint, the pharmacokinetics of the pharmaceutical composition, the diseases being treated, the mode of administration, and the age, health status and symptoms of the subjects. The effective amount includes therapeutically effective amount and prophylactically effective amount.


Unless otherwise indicated, the “therapeutically effective amount” of the pharmaceutical composition as used herein is an amount sufficient to provide therapeutic benefits in the course of treating a disease, disorder or condition, or to delay or minimize one or more symptoms associated with the disease, disorder or condition. The therapeutically effective amount of a pharmaceutical composition refers to the amount of the therapeutic agent that, when used alone or in combination with other therapies, provides a therapeutic benefit in the treatment of a disease, disorder or condition. The term “therapeutically effective amount” can include an amount that improves the overall treatment, reduces or avoids the symptoms or causes of the disease or condition, or enhances the therapeutic effect of other therapeutic agents.


Unless otherwise indicated, the “prophylactically effective amount” of the pharmaceutical composition as used herein is an amount sufficient to prevent a disease, disorder or condition, or an amount sufficient to prevent one or more symptoms associated with a disease, disorder or condition, or an amount sufficient to prevent the recurrence of a disease, disorder or condition. The prophylactically effective amount of a pharmaceutical composition refers to the amount of a therapeutic agent that, when used alone or in combination with other agents, provides a prophylactic benefit in the prevention of a disease, disorder or condition. The term “prophylactically effective amount” can include an amount that improves the overall prevention, or an amount that enhances the prophylactic effect of other preventive agents.


“Combination” and related terms refer to the simultaneous or sequential administration of the pharmaceutical compositions of the present disclosure and other therapeutic agents. For example, the pharmaceutical compositions of the present disclosure can be administered simultaneously or sequentially in separate unit dosage with other therapeutic agents, or simultaneously in a single unit dosage with other therapeutic agents.







DETAILED DESCRIPTION

As used herein, “compounds of the present disclosure” refers to the following compounds, pharmaceutically acceptable salts, isotopic variants, tautomers or stereoisomers thereof.


In the present disclosure, compounds are named using standard nomenclature. For compounds having an asymmetric center, it should be understood, unless otherwise stated, that all optical isomers and mixtures thereof are included. Furthermore, unless otherwise specified, all isomer compounds and carbon-carbon double bonds included in the present disclosure may occur in the form of Z and E. Compounds which exist in different tautomeric forms, one of which is not limited to any particular tautomer, but is intended to cover all tautomeric forms.


In one embodiment, the present disclosure relates to a compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer or stereoisomer thereof:




embedded image




    • wherein

    • G1 and G2 are independently selected from a chemical bond, C1-13 linear alkylene, C2-13 linear alkenylene, and C2-13 linear alkynylene, each of which is optionally substituted with one or more RG1;





G1 and G2 have a total length of 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13 carbon atoms;

    • RG1 is independently H, C1-14 alkyl, -La-ORa, -La-SRa, or -LaNRaR′a;
    • G3 is C4-14 linear alkylene, C4-14 linear alkenylene, or C4-14 linear alkynylene, each of which is optionally substituted with one or more RG3;
    • RG3 is independently H, -La-ORa, -La-SRa, or -La-NRaR′a;
    • La is independently a chemical bond or C1-14 alkylene;
    • Ra and R′a are independently selected from H, C1-14 alkyl, C3-14 cycloalkyl, and 3- to 14-membered heterocyclyl;
    • G4 is a chemical bond, C1-6 alkylene, C2-6 alkenylene, or C2-6 alkynylene, each of which is optionally substituted with one or more RG4;
    • RG4 is independently H, C1-6 alkyl, -Lb-ORb, -Lb-SRh, or -L-NRbR′b;
    • Lb is independently a chemical bond or C1-6 alkylene;
    • Rb and R′b are independently selected from H, C1-6 alkyl, C3-10 cycloalkyl, and 3- to 10-membered heterocyclyl;
    • or, two RG4 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C3-14 cycloalkylene or 3- to 14-membered heterocyclylene, each of which is optionally substituted with one or more R4g;
    • R4g is independently H, halogen, cyano, C1-8 alkyl, C1-8 haloalkyl, -Le-ORe, -Le-SRe, or -Le-NReR′e;
    • Le is independently a chemical bond or C1-8 alkylene;
    • Re and R′e are independently selected from H, C1-8 alkyl, C3-4 cycloalkyl, and 3- to 14-membered heterocyclyl;
    • M1 and M2 are independently selected from —C(O)O—, —OC(O)—, —O—, —SC(O)O—, —OC(O)NR—, —NRC(O)NR—, —OC(O)S—, —OC(O)O—, —NRC(O)O—, —SC(O)—, —C(O)S—, —NR—, —C(O)NR—, —NRC(O)—, —NRC(O)S—, —SC(O)NR—, —C(O)—, —OC(S)—, —C(S)O—, —OC(S)NR—, —NRC(S)O—, —S—S—, and —S(O)0-2—;
    • Q is selected from a chemical bond, —C(O)O—, —O—, —SC(O)O—, —OC(O)NRf—, —NRfC(O)NRf—, —OC(O)S—, —OC(O)O—, —NRfC(O)O—, —OC(O)—, —SC(O)—, —C(O)S—, —NRf—, —C(O)NRf—, —NRfC(O)—, —NRfC(O)S—, —SC(O)NRf—, —C(O)—, —OC(S)—, —C(S)O—, —OC(S)NRf—, —NRfC(S)O—, —S—S—, —S(O)0-2—, phenylene, and pyridinylene, wherein the phenylene or the pyridinylene is optionally substituted with one or more R*;
    • R* is independently H, halogen, cyano, C1-10 alkyl, C1-10 haloalkyl, -Lf-ORf, -Lf-SRf, or -Lf-NRfR′f;
    • Lf is independently a chemical bond or C1-8 alkylene;
    • Rf and R′f are independently selected from H, C1-10 alkyl, C3-14 cycloalkyl, and 3- to 14-membered heterocyclyl;
    • R1 and R2 are independently selected from C4-20 alkyl, C4-20 alkenyl, and C4-20 alkynyl, each of which is optionally substituted with one or more R1s, and wherein one or more methylene units are optionally and independently replaced with —NR′—;
    • R1s is independently H, C1-20 alkyl, -Lc-ORc, -Lc-SRc, or -Lc-NRcR′c;
    • R and R′ are independently H or C1-20 alkyl;
    • Lc is independently a chemical bond or C1-20 alkylene;
    • Rc and R′c are independently selected from H, C1-20 alkyl, C3-14 cycloalkyl, and 3- to 14-membered heterocyclyl;
    • R3 is selected from CN, —ORg, —C(O)Rg, —OC(O)Rg, —NR″C(O)Rg, —NRgR′g, —NR″C(O)NRgR′g, —NR″C(O)Rg, —NR″S(O)2Rg, —OC(O)NRgR′g, —NR″C(O)ORg, —N(ORg)C(O)Rg, —N(ORg)S(O)2Rg, —N(ORg)C(O)ORg, —N(ORg)C(O)RgR′g, 3- to 14-membered heterocyclyl, and 5- to 14-membered heteroaryl;
    • R4 and R5 are independently H, C1-10 alkyl, C3-10 cycloalkyl, or 3- to 10-membered heterocyclyl;
    • R″ is independently H or C1-6 alkyl;
    • R4 and R5 are independently C1-8 alkyl, which is optionally substituted with one or more R4s;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-14 cycloalkylene or 3- to 14-membered heterocyclylene, each of which is optionally substituted with one or more R4s;
    • R4s is independently H, halogen, cyano, C1-8 alkyl, C1-8 haloalkyl, -Ld-ORd, -Ld-SRd, or -Ld-NRdR′d;
    • Ld is independently a chemical bond or C1-8 alkylene;
    • Rd and R′d are independently H, C1-8 alkyl, C3-14 cycloalkyl, or 3- to 14-membered heterocyclyl.


In another embodiment, the present disclosure relates to a compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, which has a structure of formula (II):




embedded image




    • wherein

    • a=1, 2, 3, 4, 5, or 6;

    • b=4, 5, 6, 7, 8, 9, or 10;

    • c=1, 2, 3, 4, 5, or 6;

    • d=0, 1, 2, 3, or 4;

    • c+d=3, 4, 5, 6, 7, 8, or 9;

    • the other groups are as defined herein.

    • G1 and G2





In one embodiment, G1 is a chemical bond; in another embodiment, G1 is C1-13 linear alkylene, alternatively C1-9 linear alkylene, alternatively C1-6 linear alkylene, and alternatively C2-6 linear alkylene; in another embodiment, G1 is C2-13 linear alkenylene, alternatively C2-9 linear alkenylene, and alternatively C2-6 linear alkenylene; in another embodiment, G1 is C2-13 linear alkynylene, alternatively C2-9 linear alkynylene, and alternatively C2-6 linear alkynylene; in another embodiment, G1 is optionally substituted with one or more RG1; in another embodiment, G1 is optionally substituted with 1, 2, 3, or 4 RG1; in another embodiment, G1 is unsubstituted.


In a more specific embodiment, G1 is a chemical bond, C1-9 linear alkylene, C2-9 linear alkenylene, or C2-9 linear alkynylene; in another more specific embodiment, G1 is C1-6 linear alkylene, C2-6 linear alkenylene, or C2-6 linear alkynylene; in another more specific embodiment, G1 is C1-6 linear alkylene; in another more specific embodiment, G1 is C2-6 linear alkylene.


In one embodiment, G2 is a chemical bond; in another embodiment, G2 is C1-13 linear alkylene, alternatively C1-9 linear alkylene, alternatively C1-6 linear alkylene, and alternatively C1-4 linear alkylene; in another embodiment, G2 is C2-13 linear alkenylene, alternatively C2-9 linear alkenylene, and alternatively C2-6 linear alkenylene; in another embodiment, G2 is C2-13 linear alkynylene, alternatively C2-9 linear alkynylene, and alternatively C2-6 linear alkynylene; in another embodiment, G2 is optionally substituted with one or more RG1; in another embodiment, G2 is optionally substituted with 1, 2, 3, or 4 RG1; in another embodiment, G2 is unsubstituted.


In a more specific embodiment, G2 is a chemical bond, C1-9 linear alkylene, C2-9 linear alkenylene, or C2-9 linear alkynylene; in another more specific embodiment, G2 is a chemical bond, C1-6 linear alkylene, C2-6 linear alkenylene, or C2-6 linear alkynylene; in another more specific embodiment, G2 is a chemical bond or C1-6 linear alkylene; in another more specific embodiment, G2 is a chemical bond or C3-4 linear alkylene.


In one embodiment, G1 and G2 have a total length of 3 carbon atoms; in another embodiment, G1 and G2 have a total length of 4 carbon atoms; in another embodiment, G1 and G2 have a total length of 5 carbon atoms; in another embodiment, G1 and G2 have a total length of 6 carbon atoms; in another embodiment, G1 and G2 have a total length of 7 carbon atoms; in another embodiment, G1 and G2 have a total length of 8 carbon atoms; in another embodiment, G1 and G2 have a total length of 9 carbon atoms; in another embodiment, G1 and G2 have a total length of 10 carbon atoms; in another embodiment, G1 and G2 have a total length of 11 carbon atoms; in another embodiment, G1 and G2 have a total length of 12 carbon atoms; in another embodiment, G1 and G2 have a total length of 13 carbon atoms.


In a more specific embodiment, G1 and G2 have a total length of 3, 4, 5, 6, 7, 8, or 9 carbon atoms; in another more specific embodiment, G1 and G2 have a total length of 4, 5, or 6 carbon atoms; in another more specific embodiment, G1 and G2 have a total length of 5 or 6 carbon atoms; in another more specific embodiment, G1 and G2 have a total length of 5, 6, or 7 carbon atoms; in another more specific embodiment, G1 and G2 have a total length of 6 or 7 carbon atoms.


RG1

In one embodiment, RG1 is H; in another embodiment, RG1 is C1-14 alkyl, alternatively C1-10 alkyl, and alternatively C1-6 alkyl; in another embodiment, RG1 is -La-ORa; in another embodiment, RG1 is -LaSRa; in another embodiment, RG1 is -La-NRaR′a.


In a more specific embodiment, RG1 is independently H or C1-10 alkyl; in another more specific embodiment, RG1 is independently H or C1-6 alkyl.


G3

In one embodiment, G3 is C4-14 linear alkylene, alternatively C4-10 linear alkylene, alternatively C4-9 linear alkylene, and alternatively C5-8 linear alkylene; in another embodiment, G3 is C4-14 linear alkenylene; in another embodiment, G3 is C4-14 linear alkynylene; in another embodiment, G3 is optionally substituted with one or more RG3; in another embodiment, G3 is optionally substituted with 1, 2, 3, or 4 RG3; in another embodiment, G3 is unsubstituted.


In a more specific embodiment, G3 is C4-10 linear alkylene, C4-10 linear alkenylene, or C4-10 linear alkynylene; in another more specific embodiment, G3 is C4-9 linear alkylene; in another more specific embodiment, G3 is C5-8 linear alkylene.


RG3

In one embodiment, RG3 is H; in another embodiment, RG3 is -La-ORa; in another embodiment, RG3 is -La-SRa; in another embodiment, RG3 is -La-NRaR′a.


La, Ra, and R′a


In one embodiment, La is a chemical bond; in another embodiment, La is C1-14 alkylene, alternatively C1-10 alkylene, and alternatively C1-6 alkylene.


In a more specific embodiment, La is independently a chemical bond or C1-6 alkylene; in another more specific embodiment, La is a chemical bond or C1-6 alkylene.


In one embodiment, Ra is H; in another embodiment, Ra is C1-14 alkyl, alternatively C1-10 alkyl, alternatively C1-6 alkyl; in another embodiment, Ra is C3-14 cycloalkyl, alternatively C3-10 cycloalkyl; in another embodiment, Ra is 3- to 14-membered heterocyclyl, alternatively 3- to 10-membered heterocyclyl.


In one embodiment, R′a is H; in another embodiment, R′a is C1-14 alkyl, alternatively C1-10 alkyl, and alternatively C1-6 alkyl; in another embodiment, R′a is C3-14 cycloalkyl, alternatively C3-10 cycloalkyl; in another embodiment, R′a is 3- to 14-membered heterocyclyl, alternatively 3- to 10-membered heterocyclyl.


In a more specific embodiment, Ra and R′a are independently H, C1-10 alkyl, C3-10 cycloalkyl, or 3- to 10-membered heterocyclyl; in another more specific embodiment, Ra and R′a are independently H or C1-6 alkyl.


G4

In one embodiment, G4 is a chemical bond; in another embodiment, G4 is C1-6 alkylene, alternatively C1-4 alkylene, alternatively C2-4 alkylene, and alternatively C2-3 alkylene; in another embodiment, G4 is C2-6 alkenylene; in another embodiment, G4 is C2-6 alkynylene; in another embodiment, G4 is optionally substituted with one or more RG4; in another embodiment, G4 is optionally substituted with 1, 2, 3, or 4 RG4; in another embodiment, G4 is unsubstituted.


In a more specific embodiment, G4 is C1-4alkylene, C2-4 alkenylene, or C2-4 alkynylene; in another more specific embodiment, G4 is C2-4 alkylene; in another more specific embodiment, G4 is C2-3 alkylene.


RG4

In one embodiment, RG4 is H; in another embodiment, Rea is CG4 alkyl, alternatively C1-4 alkyl; in another embodiment, RG4 is -Lb-ORb; in another embodiment, RG4 is -Lb-SRb; in another embodiment, RG4 is -Lb-NRbR′b.


In a more specific embodiment, RG4 is independently H or C1-6 alkyl.


In one embodiment, two RG4 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C3-14 cycloalkylene, alternatively C3-10 cycloalkylene, alternatively C3-7 cycloalkylene; in another embodiment, two RG4 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form 3- to 14-membered heterocyclylene, alternatively 3- to 10-membered heterocyclylene, alternatively 3- to 7-membered heterocyclylene; in another embodiment, a ring formed by two RG4 taken together with the carbon atom to which they are attached is optionally substituted with one or more R4g; in another embodiment, a ring formed by two RG4 taken together with the carbon atom to which they are attached is optionally substituted with 1, 2, or 3 R4g; in another embodiment, a ring formed by two RG4 taken together with the carbon atom to which they are attached is unsubstituted.


In a more specific embodiment, two RG4 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C3-10 cycloalkylene or 3- to 10-membered heterocyclylene; in another more specific embodiment, two RG4 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C3-7 cycloalkylene or 3- to 7-membered heterocyclylene.


Lb, Rb, and R′b


In one embodiment, Lb is a chemical bond; in another embodiment, Lb is C1-6 alkylene, alternatively C1-4 alkylene.


In a more specific embodiment, Lb is independently a chemical bond or C1-4 alkylene.


In one embodiment, Rb is H; in another embodiment, Rb is C1-6 alkyl, alternatively C1-4 alkyl; in another embodiment, Rb is C3-10 cycloalkyl, alternatively C3-7 cycloalkyl; in another embodiment, Rb is 3- to 10-membered heterocyclyl, alternatively 3- to 7-membered heterocyclyl.


In one embodiment, R′b is H; in another embodiment, R′b is C1-6 alkyl, alternatively C1-4 alkyl; in another embodiment, R′b is C3-10 cycloalkyl, alternatively C3-7 cycloalkyl; in another embodiment, R′b is 3- to 10-membered heterocyclyl, alternatively 3- to 7-membered heterocyclyl.


In a more specific embodiment, Rb and R′b are independently H, C1-6 alkyl, C3-7 cycloalkyl, or 3- to 7-membered heterocyclyl; in another more specific embodiment, Rb and R′b are independently H or C1-6 alkyl; in another more specific embodiment, Rb and R′b are independently H or C1-4 alkyl.


R4g

In one embodiment, R4g is H; in another embodiment, R4g is halogen; in another embodiment, R4g is cyano; in another embodiment, R4g is C1-8 alkyl, alternatively C1-6 alkyl; in another embodiment, R4g is C1-8 haloalkyl, alternatively C1-6 haloalkyl; in another embodiment, R4g is -Le-ORe; in another embodiment, R4g is -Le-SRe; in another embodiment, R4g is -Le-NReR′e.


In a more specific embodiment, R4g is independently H, halogen, cyano, C1-6 alkyl, or C1-6 haloalkyl.


Le, Re, and R′e


In one embodiment, Le is a chemical bond; in another embodiment, Le is C1-8 alkylene, alternatively C1-6 alkylene, and alternatively C1-4 alkylene.


In a more specific embodiment, Le is independently a chemical bond or C1-6 alkylene; in another more specific embodiment, La is a chemical bond or C1-4 alkylene.


In one embodiment, Re is H; in another embodiment, Re is C1-8 alkyl, alternatively C1-6 alkylene, and alternatively C1-4 alkylene; in another embodiment, Re is C3-14 cycloalkyl, alternatively C3-10 cycloalkyl; in another embodiment, Re is 3- to 14-membered heterocyclyl, alternatively 3- to 10-membered heterocyclyl.


In one embodiment, R′e is H; in another embodiment, R′e is C1-3 alkyl, alternatively C1-6 alkylene, and alternatively C1-4 alkylene; in another embodiment, R′e is C3-14 cycloalkyl, alternatively C3-10 cycloalkyl; in another embodiment, R′e is 3- to 14-membered heterocyclyl, alternatively 3- to 10-membered heterocyclyl.


In a more specific embodiment, Re and R′e are independently H, C1-6 alkyl, C3-10 cycloalkyl, or 3- to 10-membered heterocyclyl; in another more specific embodiment, Re and R′e are independently H or C1-4 alkyl.


M1 and M2


In one embodiment, M1 is —C(O)O—; in another embodiment, M1 is —OC(O)—; in another embodiment, M1 is —O—; in another embodiment, M1 is —SC(O)O—; in another embodiment, M1 is —OC(O)NR—; in another embodiment, M1 is —NRC(O)NR—; in another embodiment, M1 is —OC(O)S—; in another embodiment, M1 is —OC(O)O—; in another embodiment, M1 is —NRC(O)O—; in another embodiment, M1 is —SC(O)—; in another embodiment, M1 is —C(O)S—; in another embodiment, M1 is —NR—; in another embodiment, M1 is —C(O)NR—, for example, —C(O)NH—; in another embodiment, M1 is —NRC(O)—, for example, —NHC(O)—; in another embodiment, M1 is —NRC(O)S—; in another embodiment, M1 is —SC(O)NR—; in another embodiment, M1 is —C(O)—; in another embodiment, M1 is —OC(S)—; in another embodiment, M1 is —C(S)O—; in another embodiment, M1 is —OC(S)NR—; in another embodiment, M1 is —NRC(S)O—; in another embodiment, M1 is —S—S—; in another embodiment, M1 is —S(O)0-2—, such as —S—, —S(O)—, and —S(O)2—.


In one embodiment, M2 is —C(O)O—; in another embodiment, M2 is —OC(O)—; in another embodiment, M2 is —O—; in another embodiment, M2 is —SC(O)O—; in another embodiment, M2 is —OC(O)NR—; in another embodiment, M2 is —NRC(O)NR—; in another embodiment, M2 is —OC(O)S—; in another embodiment, M2 is —OC(O)O—; in another embodiment, M2 is —NRC(O)O—; in another embodiment, M2 is —SC(O)—; in another embodiment, M2 is —C(O)S—; in another embodiment, M2 is —NR—; in another embodiment, M2 is —C(O)NR—; in another embodiment, M2 is —NRC(O)—; in another embodiment, M2 is —NRC(O)S—; in another embodiment, M2 is —SC(O)NR—; in another embodiment, M2 is —C(O)—; in another embodiment, M2 is —OC(S)—; in another embodiment, M2 is —C(S)O—; in another embodiment, M2 is —OC(S)NR—; in another embodiment, M2 is —NRC(S)O—; in another embodiment, M2 is —S—S—; in another embodiment, M2 is —S(O)0-2—, such as —S—, —S(O)—, and —S(O)2—.


In a more specific embodiment, M1 and M2 are independently selected from —C(O)O—, —OC(O)—, —OC(O)O—, —SC(O)—, —C(O)S—, —C(O)NR—, and —NRC(O)—; in another more specific embodiment, M1 and M2 are independently selected from —C(O)O—, —OC(O)O—, —OC(O)—, —SC(O)—, and —C(O)S—; in another more specific embodiment, M1 and M2 are independently selected from —C(O)O—, —OC(O)—, —SC(O)—, and —C(O)S—; in another more specific embodiment, M1 and M2 are independently selected from —C(O)O— and —OC(O)—.


In a more specific embodiment, M1 and M2 are independently selected from —C(O)O— and —C(O)S—; in another more specific embodiment, M1 and M2 are —C(O)O—.


In a more specific embodiment, one of M1 and M2 is —C(O)O— or —C(O)S—, and the other is —OC(O)— or —SC(O)—; in another more specific embodiment, one of M1 and M2 is —C(O)O—, and the other is —OC(O)—.


In a more specific embodiment, M1 is —OC(O)— or —SC(O)—; in another more specific embodiment, M1 is —OC(O)—; in another more specific embodiment, M2 is —C(O)O— or —C(O)S—; in another more specific embodiment, M2 is —C(O)O—.


In a more specific embodiment, M1 and M2 are independently selected from —C(O)O—, —OC(O)—, —SC(O)—, —C(O)S—, —NHC(O)—, and —C(O)NH—; in another more specific embodiment, M1 and M2 are independently selected from —C(O)O—, —OC(O)—, —C(O)S—, and —C(O)NH—; in another more specific embodiment, M1 and M2 are independently selected from —C(O)O—, —OC(O)—, and —C(O)S—; in another more specific embodiment, one of M1 and M2 is —C(O)O— or —C(O)S—, alternatively —C(O)O—, and the other is —C(O)O—, —C(O)S—, —C(O)NH—, —OC(O)—, or —SC(O)—, alternatively —C(O)O—, —C(O)S—, —C(O)NH—, or —OC(O)—, alternatively —C(O)O—, —C(O)S—, —OC(O)—, or —SC(O)—, alternatively —C(O)O—, —C(O)S—, or —OC(O)—; in another more specific embodiment, one of M1 and M2 is —OC(O)O—, and the other is —C(O)O—, —OC(O)—, —SC(O)—, or —C(O)S—, alternatively —C(O)O— or —OC(O)—; in another more specific embodiment, M1 is —C(O)O—, —OC(O)—, —SC(O)—, or —C(O)S—, alternatively —C(O)O— or —OC(O)—, alternatively —C(O)O—, alternatively —OC(O)—, and M2 is —OC(O)O—; in another more specific embodiment, M1 is —OC(O)O—, and M2 is —OC(O)— or —C(O)O—, alternatively —OC(O)—; in another more specific embodiment, M1 and M2 are independently —C(O)O—, —C(O)S—, —OC(O)—, —SC(O)—, or —OC(O)O—; in another more specific embodiment, M1 and M2 are independently —C(O)O—, —OC(O)—, or —OC(O)O—; in another more specific embodiment, M1 and M2 are not simultaneously —OC(O)O—.


Q

In one embodiment, Q is a chemical bond; in another more specific embodiment, Q is —C(O)O—; in another more specific embodiment, Q is —O—; in another more specific embodiment, Q is —SC(O)O—; in another more specific embodiment, Q is —OC(O)NRf—; in another more specific embodiment, Q is —NRfC(O)NRf—; in another more specific embodiment, Q is —OC(O)S—; in another more specific embodiment, Q is —OC(O)O—; in another more specific embodiment, Q is —NRfC(O)O—; in another more specific embodiment, Q is —OC(O)—; in another more specific embodiment, Q is —SC(O)—; in another more specific embodiment, Q is —C(O)S—; in another more specific embodiment, Q is —NRf—; in another more specific embodiment, Q is —C(O)NRf—; in another more specific embodiment, Q is —NRfC(O)—; in another more specific embodiment, Q is —NRfC(O)S—; in another more specific embodiment, Q is —SC(O)NR—; in another more specific embodiment, Q is —C(O)—; in another more specific embodiment, Q is —OC(S)—; in another more specific embodiment, Q is —C(S)O—; in another more specific embodiment, Q is —OC(S)NRf—; in another more specific embodiment, Q is —NRfC(S)O—; in another more specific embodiment, Q is —S—S—; in another more specific embodiment, Q is —S(O)0-2—; in another more specific embodiment, Q is phenylene; in another more specific embodiment, Q is pyridinylene; in another more specific embodiment, when Q is phenylene, Q is optionally substituted with one or more R*, alternatively optionally substituted with 1, 2 or 3 R*; in another more specific embodiment, Q is unsubstituted phenylene; in another more specific embodiment, when Q is pyridinylene, Q is optionally substituted with one or more R*, alternatively optionally substituted with 1, 2 or 3 R*; in another more specific embodiment, Q is unsubstituted pyridinylene.


In another more specific embodiment, Q is a chemical bond, —OC(O)—, or —SC(O)—; in another more specific embodiment, Q is a chemical bond or —SC(O)—; in another more specific embodiment, Q is phenylene or pyridinylene, which is optionally substituted with 1, 2 or 3 R*.


R*

In one embodiment, R* is H; in another more specific embodiment, Q is halogen; in another more specific embodiment, R* is cyano; in another more specific embodiment, R* is C1-10 alkyl, alternatively C1-6 alkyl; in another more specific embodiment, R* is C1-10 haloalkyl, alternatively C1-6 haloalkyl; in another more specific embodiment, R* is -Lf-ORf; in another more specific embodiment, R* is -Lf-SRf; in another more specific embodiment, R* is -Lf-NRfR′f.


In a more specific embodiment, R* is independently H, halogen, cyano, C1-6 alkyl, or C1-6 haloalkyl.


Lf, Rf, and R′f


In one embodiment, Lf is a chemical bond; in another embodiment, Lf is C1-8 alkylene, alternatively C1-6 alkylene, alternatively C1-4 alkylene.


In a more specific embodiment, Lt is independently a chemical bond or C1-6 alkylene; in another more specific embodiment, Lf is independently a chemical bond or C1-4 alkylene.


In one embodiment, Rf is H; in another embodiment, Rf is C1-10 alkyl, alternatively C1-6 alkyl, and alternatively C1-4 alkyl; in another embodiment, Rf is C3-14 cycloalkyl, alternatively C3-10 cycloalkyl; in another embodiment, Rf is 3- to 14-membered heterocyclyl, alternatively 3- to 10-membered heterocyclyl.


In one embodiment, R′f is H; in another embodiment, R′f is C1-10 alkyl, alternatively C1-6 alkyl, alternatively C1-4 alkyl; in another embodiment, R′f is C3-14 cycloalkyl, alternatively C3-10 cycloalkyl; in another embodiment, R′f is 3- to 14-membered heterocyclyl, alternatively 3- to 10-membered heterocyclyl.


In a more specific embodiment, Rf and R′f are independently H, C1-6 alkyl, C3-10 cycloalkyl, or 3- to 10-membered heterocyclyl; in another more specific embodiment, Rf and R′f are independently H or C1-6 alkyl; in another more specific embodiment, Rf and R′f are independently H or C1-4 alkyl.


R1 and R2


In one embodiment, R1 is C4-20 alkyl, alternatively C6-14 alkyl, alternatively C7-12 alkyl, alternatively C8-12 alkyl, alternatively C8-11 alkyl, alternatively C9-11 alkyl, alternatively C8-10 alkyl, alternatively C9-10 alkyl, alternatively C8-9 alkyl, alternatively C9 alkyl, alternatively C8-12 linear alkyl, alternatively C7-11 linear alkyl, alternatively C8-11 linear alkyl, alternatively C9-11 linear alkyl, alternatively C10-11 linear alkyl, alternatively C8-10 linear alkyl, alternatively C9-10 linear alkyl, alternatively C8-9 linear alkyl, alternatively C11 linear alkyl, alternatively C10 linear alkyl, and alternatively C9 linear alkyl; in another embodiment, R1 is C4-20 alkenyl, alternatively C6-14 alkenyl, alternatively C7-12 alkenyl; alternatively C8-12 alkenyl; in another embodiment, R1 is C4-20 alkynyl, alternatively C6-14 alkynyl, alternatively C7-12 alkynyl, alternatively C8-12 alkynyl; in another embodiment, R1 is optionally substituted with one or more R1s; in another embodiment, R1 is optionally substituted with 1, 2, 3, or 4 R1s, alternatively optionally substituted with 1 R1s; in another embodiment, R1 is unsubstituted; in another embodiment, one or more methylene units in R1 are optionally and independently replaced with —NR′—, alternatively one methylene unit in R1 is optionally replaced with —NR′—; in another embodiment, R1 is optionally substituted with 1 C1-9 alkyl(alternatively C6-9 alkyl, alternatively C6-7 alkyl).


In one embodiment, R1 is —(CH2)5CH2; in another embodiment, R1 is —(CH2)6CH3; in another embodiment, R1 is —(CH2)7CH3; in another embodiment, R1 is —(CH2)8CH3; in another embodiment, R1 is —(CH2)9CH3; in another embodiment, R1 is —(CH2)10CH3; in another embodiment, R1 is —(CH2)11CH3; in another embodiment, R1 is —CH2—C≡C—(CH2)5CH3; in another embodiment, R1 is —CH2—C≡C—(CH2)6CH3; in another embodiment, R1 is —(CH2)2C≡C—(CH2)5CH3; in another embodiment, R1 is —(CH2)4—C≡C—(CH2)3CH3; in another embodiment, R1 is —CH2—CH═CH—(CH2)5CH3; in another embodiment, R1 is —CH2—CH═CH—(CH2)6CH3; in another embodiment, R1 is —(CH2)2—CH═CH—(CH2)5CH3; in another embodiment, R1 is —(CH2)4—CH═CH—(CH2)3CH3; in another embodiment, R1 is —(CH2)5—CH═CH—CH2CH3; in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


In one embodiment, R1 is —(CH2)2—C≡C—(CH2)4CH3; in another embodiment, R1 is —(CH2)3—C≡C—(CH2)3CH3; in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


in another embodiment, R1 is




embedded image


In one embodiment, R2 is C4-20 alkyl, alternatively C6-14 alkyl, alternatively C7-12 alkyl, alternatively C8-12 alkyl, alternatively C9-12 alkyl, alternatively C8-11 alkyl, alternatively C9-11 alkyl, alternatively C8-10 alkyl, alternatively C9-10 alkyl, alternatively C10 alkyl, alternatively C9 alkyl, alternatively C8-12 linear alkyl, alternatively C7-11 linear alkyl, alternatively C8-11 linear alkyl, alternatively C10-11 linear alkyl, alternatively C11 linear alkyl, alternatively C10 linear alkyl, alternatively C8-10 linear alkyl, alternatively C9-10 linear alkyl, and alternatively C9 linear alkyl; in another embodiment, R2 is C4-20 alkenyl, alternatively C6-14 alkenyl, alternatively C7-12 alkenyl, alternatively C8-12 alkenyl; in another embodiment, R2 is C4-20 alkynyl, alternatively C6-14 alkynyl, alternatively C7-12 alkynyl, alternatively C8-12 alkynyl; in another embodiment, R2 is optionally substituted with one or more R1s; in another embodiment, R2 is optionally substituted with 1, 2, 3, or 4 R1s, alternatively optionally substituted with 1 R1s, alternatively optionally substituted with 1 C1-3 alkyl, alternatively optionally substituted with 1 methyl; in another embodiment, R2 is unsubstituted; in another embodiment, one or more methylene units in R2 are optionally and independently replaced with —NR′—, alternatively one methylene unit in R2 is optionally replaced with —NR′—.


In one embodiment, only one of R1 and R2 is substituted; in another embodiment, R1 is substituted with R1s, and R2 is not substituted with R1s; in another embodiment, R2 is substituted with R1s, and R1 is not substituted with R1s;


In one embodiment, R2 is —(CH2)5CH3; in another embodiment, R2 is —(CH2)5CH3; in another embodiment, R2 is —(CH2)7CH3; in another embodiment, R2 is —(CH2)8CH3; in another embodiment, R2 is —(CH2)9CH3; in another embodiment, R2 is —(CH2)10CH3; in another embodiment, R2 is —(CH2)11CH3; in another embodiment, R2 is —CH2—C≡C—(CH2)5CH3; in another embodiment, R2 is —CH2—C≡C—(CH2)6CH3; in another embodiment, R2 is —(CH2)2—C≡C—(CH2)5CH3; in another embodiment, R2 is —(CH2)4—C≡C—(CH2)3CH3; in another embodiment, R2 is —CH—CH═CH—(CH2)5CH3; in another embodiment, R2 is —CH2—CH═CH—(CH2)6CH3; in another embodiment, R2 is —(CH2)2—CH═CH—(CH2)5CH3; in another embodiment, R2 is —(CH2)4—CH═CH—(CH2)3CH3; in another embodiment, R2 is —(CH2)5—CH═CH—CH2CH3; in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


In one embodiment, R2 is —(CH2)2—C≡C—(CH2)4CH3; in another embodiment, R2 is —(CH2)3—C≡C—(CH2)3CH3; in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


in another embodiment, R2 is




embedded image


In a more specific embodiment, R1 and R2 are independently selected from C6-4 alkyl, C6-14 alkenyl, and C6-14 alkynyl; in another more specific embodiment, R1 and R2 are independently selected from C7-12 alkyl, C7-12 alkenyl, and C7-12 alkynyl; in another more specific embodiment, R1 and R2 are independently selected from C3-12 alkyl, C8-12 alkenyl, and C8-12 alkynyl; in another more specific embodiment, R1 and R2 are independently selected from the following groups: —(CH2)5CH3, —(CH2)6CH3; —(CH2)7CH3, —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3; —(CH2)11CH3, —CH2C≡C—(CH2)5CH3; —CH2—C≡C—(CH2)6CH3, —(CH2)2—C≡C—(CH2)5CH3, —(CH2)4—C≡C—(CH2)3CH3, —CH2—CH═CH—(CH2)5CH3, —CH2—CH═CH—(CH2)6CH3, —(CH2)2—CH═CH—(CH2)5CH3, —(CH2)4—CH═CH—(CH2)3CH3, —(CH2)5—CH═CH—CH2CH3,




embedded image


embedded image


In a more specific embodiment, R1 and R2 are independently selected from C7-12 alkyl, alternatively C8-12 alkyl, alternatively C9-11 alkyl, alternatively C9-10 alkyl, which is optionally substituted with 1 R1s; and only one of R1 and R2 is substituted.


In a more specific embodiment, R1 and R2 are independently selected from: —(CH2)6CH3, —(CH2)7CH3, —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3,




embedded image


in another more specific embodiment, R1 and R2 are independently selected from: —(CH2)7CH3, —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3,




embedded image


in another more specific embodiment, R1 and R2 are independently selected from: —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3,




embedded image


In a more specific embodiment, R1 and R2 are independently selected from: —(CH2)6CH3; —(CH2)7CH3, —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3, —CH2—CH═CH—(CH2)5CH3, —CH2—C≡C—(CH2)5CH3, —(CH2)2—C≡C—(CH2)4CH3, —(CH2)3—C≡C—(CH2)3CH3,




embedded image


embedded image


in another more specific embodiment, R1 and R2 are independently selected from: —(CH2)7CH3, —(CH2)3CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3, —CH2—CH═CH—(CH2)5CH3, —CH2—C≡C—(CH2)5CH3, —(CH2)2—C≡C—(CH2)4CH3, —(CH2)3—C≡C—(CH2)3CH3,




embedded image


embedded image


in another more specific embodiment, R1 and R2 are independently selected from: —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3, —CH2—CH═CH—(CH2)5CH3, —CH2—C≡C—(CH2)5CH3, —(CH2)2—C≡C—(CH2)4CH3, —(CH2)3—C≡C—(CH2)3CH3,




embedded image


embedded image


In a more specific embodiment, R1 and R2 are independently selected from: —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3,




embedded image


in another more specific embodiment, R1 and R2 are independently selected from: —(CH2)7CH3, —(CH2)8CH3, —(CH2)9CH3,




embedded image


In a more specific embodiment, R1 is selected from:




embedded image


in another more specific embodiment, R1 is selected from:




embedded image


In a more specific embodiment, R1 is selected from:




embedded image


in another more specific embodiment, R1 is selected from:




embedded image


and in another more specific embodiment, R1 is selected from: —(CH2)8CH3, —(CH2)9CH3,




embedded image


in another more specific embodiment, R1 is selected from:




embedded image


in another more specific embodiment, R1 is selected from:




embedded image


In a more specific embodiment, R2 is selected from: —(CH2)9CH3, —(CH2)10CH3, and




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


In a more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


and




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


in another more specific embodiment, R2 is selected from:




embedded image


R1s

In one embodiment, R1s is H; in another embodiment, R1s is H; in another embodiment, R1s is C1-20 alkyl, alternatively C1-14 alkyl, alternatively C1-10 alkyl, alternatively C1-9 alkyl, alternatively C7-9 alkyl, alternatively C8-9 alkyl, alternatively C1-6 alkyl, and alternatively C1-4 alkyl, alternatively C7-11 alkyl, alternatively C4-10 alkyl, alternatively C6-10 alkyl, and alternatively C7-10 alkyl, alternatively C9-10 alkyl, alternatively C4-9 alkyl, alternatively C6-9 alkyl, alternatively C1-8 alkyl, alternatively C4-8 alkyl, alternatively C6-8 alkyl, alternatively C7-8 alkyl, alternatively C6-7 alkyl, alternatively C9 alkyl, alternatively C8 alkyl, alternatively C7 alkyl; in another embodiment, R1s is -Lc-ORc; in another embodiment, R1s is -Lc-SRc; in another embodiment, R1s is -Lc-NRcR′c.


In one embodiment, the substitution site of R1s on R1 is separated from M1 by 0 carbon atom; in another embodiment, the substitution site of R1s on R1 is separated from M1 by 1 carbon atom; in another embodiment, the substitution site of R1s on R1 is separated from M1 by 2 carbon atoms; in another embodiment, the substitution site of R1s on R1 is separated from M1 by 3 carbon atoms; in another embodiment, the substitution site of R1s on R1 is separated from M1 by 4 carbon atoms; in another embodiment, the substitution site of R1s on R1 is separated from M1 by 5 carbon atoms; in another embodiment, the substitution site of R1s on R1 is separated from M1 by 6 carbon atoms; in another embodiment, the substitution site of R1s on R1 is separated from M1 by 7 carbon atoms; in another embodiment, the substitution site of R1s on R1 is separated from M1 by 8 carbon atoms; in another embodiment, the substitution site of R1s on R1 is separated from M1 by 9 carbon atoms; in another embodiment, the substitution site of R1s on R1 is separated from M1 by 10 carbon atoms.


In one embodiment, the substitution site of R1s on R2 is separated from M2 by 0 carbon atom; in another embodiment, the substitution site of R1s on R2 is separated from M2 by 1 carbon atom; in another embodiment, the substitution site of R1s on R2 is separated from M2 by 2 carbon atoms; in another embodiment, the substitution site of R1s on R2 is separated from M2 by 3 carbon atoms; in another embodiment, the substitution site of R1s on R2 is separated from M2 by 4 carbon atoms; in another embodiment, the substitution site of R1s on R2 is separated from M2 by 5 carbon atoms; in another embodiment, the substitution site of R1s on R2 is separated from M2 by 6 carbon atoms; in another embodiment, the substitution site of R1s on R2 is separated from M2 by 7 carbon atoms; in another embodiment, the substitution site of R1s on R2 is separated from M2 by 8 carbon atoms; in another embodiment, the substitution site of R1s on R2 is separated from M2 by 9 carbon atoms; in another embodiment, the substitution site of R1s on R2 is separated from M2 by 10 carbon atoms.


In a more specific embodiment, R1s is independently H, C1-4 alkyl, -Lc-ORc, or -Lc-NRcR′c;


In another more specific embodiment, R1s is independently H or C1-14 alkyl; in another more specific embodiment, R1s is independently H or C1-10 alkyl; in another more specific embodiment, R1s is independently H or C1-9 alkyl; in another more specific embodiment, R1s is independently H or C1-6 alkyl; in another more specific embodiment, R1s is independently H or C1-4 alkyl.


In a more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 0-10 carbon atoms; in another more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 0-6 carbon atoms; in another more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 0-4 carbon atoms; in another more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 0-2 carbon atoms; in another more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 0 carbon atom.


In a more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 1-10 carbon atoms; in another more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 1-6 carbon atoms; in another more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 1-4 carbon atoms; in another more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 1-2 carbon atoms; in another more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 2-10 carbon atoms; in another more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 2-6 carbon atoms; in another more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 2-4 carbon atoms.


In another more specific embodiment, R1 is not substituted with Rs1, and the substitution site of R1s on R2 is separated from M2 by 0-10 carbon atoms, alternatively 1-10 carbon atoms, alternatively 1-6 carbon atoms, alternatively 1-4 carbon atoms, alternatively 1-2 carbon atoms; In another more specific embodiment, R1 is not substituted with R1s, and the substitution site of R1s on R2 is separated from M2 by 0-10 carbon atoms, alternatively 0-6 carbon atom, alternatively 0-4 carbon atoms, alternatively 0-2 carbon atoms; alternatively 0 carbon atom; In another more specific embodiment, R1 is not substituted with R1s, and the substitution site of R1s on R2 is separated from M2 by 2-10 carbon atoms, alternatively 2-6 carbon atom, alternatively 2-4 carbon atoms.


R and R′

In one embodiment, R is H; in another embodiment, R is C1-20 alkyl, alternatively C1-14 alkyl, alternatively C1-9 alkyl, and alternatively C1-6 alkyl.


In one embodiment, R′ is H; in another embodiment, R′ is C1-20 alkyl, alternatively C1-14 alkyl, alternatively C1-9 alkyl, and alternatively C1-6 alkyl.


In a more specific embodiment, R and R are each independently H or C10.20 alkyl; in another more specific embodiment, R and R′ are each independently H or C1-20 alkyl; in another more specific embodiment, R and R′ are each independently H or C1-9 alkyl; in another more specific embodiment, R and R′ are each independently H or C1-6 alkyl; in another more specific embodiment, R is H.


Lc, Rc, and R′c


In one embodiment, Lc is a chemical bond; in another embodiment, Lc is C1-20 alkylene, alternatively C1-14 alkylene, alternatively C1-10 alkylene, and alternatively C1-6 alkylene.


In a more specific embodiment, Lc is independently a chemical bond or C1-14 alkylene; in another more specific embodiment, Lc is independently a chemical bond or C1-10 alkylene; in another more specific embodiment, Lc is independently a chemical bond or C1-6 alkylene.


In one embodiment, Rc is H; in another embodiment, Rc is C1-20 alkyl, alternatively C1-14 alkyl, alternatively C1-10 alkyl, and alternatively C1-6 alkyl; in another embodiment, Rc is C3-14 cycloalkyl, alternatively C3-10 cycloalkyl; in another embodiment, Rc is 3- to 14-membered heterocyclyl, alternatively 3- to 10-membered heterocyclyl.


In one embodiment, R′c is H; in another embodiment, R′c is C1-20 alkyl, alternatively C1-14 alkyl, alternatively C1-10 alkyl, and alternatively C1-6 alkyl; in another embodiment, R′c is C3-14 cycloalkyl, alternatively C3-10 cycloalkyl; in another embodiment, R′c is 3- to 14-membered heterocyclyl, alternatively 3- to 10-membered heterocyclyl.


In a more specific embodiment, Rc and R′c are independently H or C1-14 alkyl; in another more specific embodiment, Rc and R′c are independently H or C1-10 alkyl; in another more specific embodiment, Rc and R′c are independently H or C1-6 alkyl.


R3

In one embodiment, R3 is CN; in another embodiment, R3 is —ORg, for example, —OH; in another embodiment, R3 is —C(O)Rg; in another embodiment, R3 is —OC(O)Rg; in another embodiment, R3 is —NR″C(O)Rg; in another embodiment, R3 is —NRgR′g, for example, —N(CH3)2; in another embodiment, R3 is —NR″C(O)NRgR′g; in another embodiment, Rg is —NR″C(O)Rg; in another embodiment, R3 is —NR″S(O)2Rg; in another embodiment, R3 is —OC(O)NRgR′g; in another embodiment, R3 is —NR″C(O)ORg; in another embodiment, R3 is —N(ORg)C(O)Rg; in another embodiment, R3 is —N(ORg)S(O)2Rg; in another embodiment, R3 is —N(ORg)C(O)ORg; in another embodiment, R3 is —N(ORg)C(O)RgR′g; in another embodiment, R3 is 3- to 14-membered heterocyclyl; in another embodiment, R3 is 5- to 14-membered heteroaryl.


In a more specific embodiment, R3 is CN, —ORg, or —NRgRg′; in another more specific embodiment, R3 is —ORg or —NRgR′g; in another more specific embodiment, R3 is —OH or —N(CH3)—;


In another more specific embodiment, R3 is —ORg; in another more specific embodiment, R3 is —OH.


Rg and R′g


In one embodiment, Rg is H; in another embodiment, Rg is C1-10 alkyl, alternatively C1-6 alkyl, alternatively C1-4 alkyl, for example, methyl; in another embodiment, Rg is C3-10 cycloalkyl, alternatively C3-7 cycloalkyl; in another embodiment, Rg is 3- to 10-membered heterocyclyl, alternatively 3- to 7-membered heterocyclyl.


In one embodiment, R′g is H; in another embodiment, R′g is C1-10 alkyl, alternatively C1-6 alkyl, and alternatively C1-4 alkyl; in another embodiment, R′g is C3-10 cycloalkyl, alternatively C3-7 cycloalkyl; in another embodiment, R′g is 3- to 10-membered heterocyclyl, alternatively 3- to 7-membered heterocyclyl.


In a more specific embodiment, Rg and R′g are independently H, C1-6 alkyl, C3-7 cycloalkyl, or 3- to 7-membered heterocyclyl; in another more specific embodiment, Rg and R′g are independently H or C1-6 alkyl; in another more specific embodiment, Rg and R′g are independently H or C1-4 alkyl; in another more specific embodiment, Rg and R′g are independently H or methyl.


R″

In one embodiment, R″ is H; in another embodiment, R″ is C1-6 alkyl, alternatively C1-4 alkyl.


R4 and R5


In one embodiment, R4 is C1-8 alkyl, alternatively C1-6 alkyl, alternatively C1-3 alkyl, and alternatively methyl; in another embodiment, R4 is optionally substituted with one or more R4s; in another embodiment, R4 is optionally substituted with 1, 2, or 3 R4s; in another embodiment, R4 is unsubstituted.


In one embodiment, R5 is C1-8 alkyl, alternatively C1-6 alkyl, alternatively C1-3 alkyl, and alternatively methyl; in another embodiment, R5 is optionally substituted with one or more R4s; in another embodiment, R5 is optionally substituted with 1, 2, or 3 R4s; in another embodiment, R5 is unsubstituted.


In one embodiment, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-14 cycloalkylene, alternatively C3-10 cycloalkylene, alternatively C3-6 cycloalkylene (e.g., cyclopropylene, cyclobutylene, cyclopentylene, and cyclohexylene), alternatively C3-5 cycloalkylene, alternatively C3-4 cycloalkylene, alternatively cyclopropylene, alternatively cyclopentylene; in another embodiment, R4 and R5 are taken together with the carbon atom to which they are attached to form 3- to 14-membered heterocyclylene, alternatively 3- to 10-membered heterocyclylene, alternatively 3- to 6-membered heterocyclylene; in another embodiment, a ring formed by R4, R5 taken together with the carbon atom to which they are attached is optionally substituted with one or more R4s; in another embodiment, a ring formed by R4, R5 taken together with the carbon atom to which they are attached is optionally substituted with 1, 2, or 3 R4s; in another embodiment, a ring formed by R4, R5 taken together with the carbon atom to which they are attached is unsubstituted.


In a more specific embodiment, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-10 cycloalkylene or 3- to 10-membered heterocyclylene; in another more specific embodiment, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-6 cycloalkylene or 3- to 6-membered heterocyclylene; in another more specific embodiment, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-6 cycloalkylene (e.g., cyclopropylene, cyclobutylene, cyclopentylene, or cyclohexylene); in another more specific embodiment, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-5 cycloalkylene; in another more specific embodiment, R4 and R5 are taken together with the carbon atom to which they are attached to form cyclopropylene or cyclopentylene; in another more specific embodiment, R4 and R5 are taken together with the carbon atom to which they are attached to form cyclopropylene; in another more specific embodiment, R4 and R5 are not taken together with the carbon atom to which they are attached to form a ring.


R4s

In one embodiment, R4s is H; in another embodiment, R4s is halogen; in another embodiment, R4s is cyano; in another embodiment, R4s is C1-8 alkyl, alternatively C1-6 alkyl, alternatively C1-3 alkyl; in another embodiment, R4s is C1-8 haloalkyl, alternatively C1-6 haloalkyl, and alternatively C1-3 haloalkyl; in another embodiment, R4s is -Ld-ORd; in another embodiment, R4s is -Ld-SRd; in another embodiment, R4s is -Ld-NRdR′d.


In a more specific embodiment, R4s is independently H, halogen, cyano, C1-6 alkyl, C1-6 haloalkyl, -Ld-ORd, -Ld-SRd, or -Ld-NRdR′d; in another more specific embodiment, R4s is independently H, halogen, cyano, C1-6 alkyl, or C1-6 haloalkyl; in another more specific embodiment, R4s is independently H, C1-3 alkyl, or C1-3 haloalkyl.


Ld, Rd, and R′d


In one embodiment, Ld is a chemical bond; in another embodiment, Ld is C1-3 alkylene, alternatively C1-6 alkylene, and alternatively C1-3 alkylene.


In a more specific embodiment, Ld is independently a chemical bond or C1-6 alkylene; in another more specific embodiment, Ld is independently a chemical bond or C1-3 alkylene.


In one embodiment, Rd is H; in another embodiment, Rd is C1-8 alkyl, alternatively C1-6 alkyl; in another embodiment, Rd is C3-14 cycloalkyl, alternatively C3-10 cycloalkyl; in another embodiment, Rd is 3- to 14-membered heterocyclyl, alternatively 3- to 10-membered heterocyclyl.


In one embodiment, R′d is H; in another embodiment, R′d is C1-8 alkyl, alternatively C1-6 alkyl; in another embodiment, R′d is C3-4 cycloalkyl, alternatively C3-10 cycloalkyl; in another embodiment, R′d is 3- to 14-membered heterocyclyl, alternatively 3- to 10-membered heterocyclyl.


In a more specific embodiment, Rd and R′d are independently H, C1-6 alkyl, C3-10 cycloalkyl, or 3- to 10-membered heterocyclyl; in another more specific embodiment, Rd and R′d are independently H or C1-6 alkyl.


a, b, c, and d


In one embodiment, a is 1; in another embodiment, a is 2; in another embodiment, a is 3; in another embodiment, a is 4; in another embodiment, a is 5; in another embodiment, a is 6.


In a more specific embodiment, a=2, 3, 4, 5, or 6; in another more specific embodiment, a=2, 3, or 4; in another more specific embodiment, a=2 or 4; in another more specific embodiment, a=2 or 3; in another more specific embodiment, a=3 or 4.


In one embodiment, b is 4; in another embodiment, b is 5; in another embodiment, b is 6; in another embodiment, b is 7; in another embodiment, b is 8; in another embodiment, b is 9; in another embodiment, b is 10.


In a more specific embodiment, b=5, 6, 7, or 8; in another more specific embodiment, b=5, 6, or 7; in another more specific embodiment, b=5 or 7; in another more specific embodiment, b=6 or 7.


In a more specific embodiment, b=4, 5, 6, 7, 8, 9, or 10; in another more specific embodiment, b=4, 5, 6, 7, 8, or 9; in another more specific embodiment, b=4, 5, 6, or 7; in another more specific embodiment, b=5 or 6; in another more specific embodiment, b=6, 7, or 8; in another more specific embodiment, b=5, 6, 7, or 8.


In one embodiment, c is 1; in another embodiment, c is 2; in another embodiment, c is 3; in another embodiment, c is 4; in another embodiment, c is 5; in another embodiment, c is 6.


In a more specific embodiment, c=2, 3, 4, 5, or 6; in another more specific embodiment, c=2, 3, or 4; in another more specific embodiment, c=3 or 4; in another more specific embodiment, c=5 or 6.


In a more specific embodiment, c=2, 4, 5, or 6; in another more specific embodiment, c=4, 5, or 6; in another more specific embodiment, c=4 or 6; in another more specific embodiment, c=2, 3, 4, or 5; in another more specific embodiment, c=2, 4, or 5; in another more specific embodiment, c=2 or 5.


In one embodiment, d is 0; in another embodiment, d is 1; in another embodiment, d is 2; in another embodiment, d is 3; in another embodiment, d is 4.


In a more specific embodiment, d=0, 1, 2, or 4; in another more specific embodiment, d=2, 3, or 4; in another more specific embodiment, d=0 or 1.


In a more specific embodiment, d=0, 1, 2, 3, or 4; in another more specific embodiment, d=0, 1, or 2; in another more specific embodiment, d=0 or 2; in another more specific embodiment, d is not 0; in another more specific embodiment, d=1, 2, 3, or 4; in another more specific embodiment, d=1 or 2; in another more specific embodiment, d=1, 2 or 4; in another more specific embodiment, d=1 or 4.


In one embodiment, c+d=3; in another embodiment, c+d=4; in another embodiment, c+d=5; in another embodiment, c+d=6; in another embodiment, c+d=7; in another embodiment, c+d=8; in another embodiment, c+d=9.


In a more specific embodiment, c+d=4, 5, or 6; in another more specific embodiment, c+d=5 or 6.


In a more specific embodiment, c+d=3, 4, 5, 6, 7, 8, or 9; in another more specific embodiment, c+d=4, 5, 6, or 7; in another more specific embodiment, c+d=4, 5, or 6; in another more specific embodiment, c+d=5, 6, or 7; in another more specific embodiment, c+d=6, 7, or 8; in another more specific embodiment, c+d=6 or 7.


Any of the above technical solutions in any specific embodiment or any combination thereof may be combined with any technical solution or any combination thereof in other specific embodiments. For example, any technical solutions of G1, or any combination thereof, may be combined with any technical solution of G2, RG1, G3, RG3, La, Ra, R′a, G4, RG4, Lb, Rb, R′b, R4g, Le, Re, R′e, M1, M2, Q, R*, Lf, Rf, R′f, R1, R2, R1s, R, R′, Lc, Rc, R′c, R3, Rg, R′g, R″, R4, R5, R4s, Ld, Rd, R′d, a, b, c, and d, etc. The present disclosure is intended to include all of these combinations of technical solutions and, for reasons of space, will not be listed.


In a more specific embodiment, the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof:




embedded image




    • wherein

    • G1 and G2 are independently selected from a chemical bond, C1-13 linear alkylene, C2-3 linear alkenylene, and C2-13 linear alkynylene, each of which is optionally substituted with one or more RG1;

    • G1 and G2 have a total length of 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13 carbon atoms;

    • RG1 is independently H, C1-14 alkyl, -La-ORa, -La-SRa, or -La-NRaR′a;

    • G3 is C4-14 linear alkylene, C4-14 linear alkenylene, or C4-14 linear alkynylene, each of which is optionally substituted with one or more RG3;

    • RG3 is independently H, -La-ORa, -La-SRa, or -La-NRaR′a;

    • La is independently a chemical bond or C1-14 alkylene;

    • Ra and R′a are independently selected from H, C1-14 alkyl, C3-14 cycloalkyl, and 3- to 14-membered heterocyclyl;

    • G4 is a chemical bond, C1-6 alkylene, C2-6 alkenylene, or C2-6 alkynylene, each of which is optionally substituted with one or more RG4;

    • RG4 is independently H, C1-6 alkyl, -Lb-ORb, -Lb-SRb, or -Lb-NRbR′b;

    • Lb is independently a chemical bond or C1-6 alkylene;





Rb and R′b are independently selected from H, C1-6 alkyl, C3-10 cycloalkyl, and 3- to 10-membered heterocyclyl;

    • or, two RG4 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C3-14 cycloalkylene or 3- to 14-membered heterocyclylene, each of which is optionally substituted with one or more R4g;
    • R4g is independently H, halogen, cyano, C1-8 alkyl, C1-8 haloalkyl, -Le-ORe, -Le-SRe, or -Le-NReR′e;
    • Le is independently a chemical bond or C1-8 alkylene;
    • Re and R′e are independently selected from H, C1-8 alkyl, C3-14 cycloalkyl, and 3- to 14-membered heterocyclyl;
    • M1, and M2 are independently selected from —C(O)O—, —OC(O)—, —O—, —SC(O)O—, —OC(O)NR—, —NRC(O)NR—, —OC(O)S—, —OC(O)O—, —NRC(O)O—, —SC(O)—, —C(O)S—, —NR—, —C(O)NR—, —NRC(O)—, —NRC(O)S—, —SC(O)NR—, —C(O)—, —OC(S)—, —C(S)O—, —OC(S)NR—, —NRC(S)O—, —S—S—, and —S(O)0-2;
    • Q is selected from a chemical bond, —C(O)O—, —O—, —SC(O)O—, —OC(O)NRf—, —NRfC(O)NRf—, —OC(O)S—, —OC(O)O—, —NRfC(O)O—, —OC(O)—, —SC(O)—, —C(O)S—, —NRf—, —C(O)NRf—, —NRfC(O)—, —NRfC(O)S—, —SC(O)NRf—, —C(O)—, —OC(S)—, —C(S)O—, —OC(S)NRf—, —NRfC(S)O—, —S—S—, —S(O)0-2, phenylene, and pyridinylene, wherein the phenylene or the pyridinylene is optionally substituted with one or more R*;
    • R* is independently H, halogen, cyano, C1-10 alkyl, C1-10 haloalkyl, -Lf-ORf, -Lf-SRf, or -Lf-NRfR′f;
    • Lf is independently a chemical bond or C1-8 alkylene;
    • R1 and R2 are independently selected from H, C1-10 alkyl, C3-14 cycloalkyl, and 3- to 14-membered heterocyclyl;
    • R1 and R2 are independently selected from C4-20 alkyl, C4-20 alkenyl, and C4-20 alkynyl, each of which is optionally substituted with one or more R1s, and wherein one or more methylene units are optionally and independently replaced with —NR′—;
    • R1s is independently H, C1-20 alkyl, -Lc-ORc, -Lc-SRc, or -Lc-NRcR′c;
    • R and R′ are independently H or C1-20 alkyl;


Lc is independently a chemical bond or C1-20 alkylene;

    • Rc and R′c are independently selected from H, C1-20 alkyl, C3-14 cycloalkyl, and 3- to 14-membered heterocyclyl;
    • R3 is selected from CN, —ORg, —C(O)Rg, —OC(O)Rg, —NR″C(O)Rg, —NRgR′g, —NR″C(O)NRgR′g, —NR″C(O)Rg, —NR″S(O)2Rg, —OC(O)NRgR′g, —NR″C(O)ORg, —N(ORg)C(O)Rg, —N(ORg)S(O)2Rg, —N(ORg)C(O)ORg, —N(ORg)C(O)RgR′g, 3- to 14-membered heterocyclyl, and 5- to 14-membered heteroaryl;
    • Rg and R′g are independently H, C1-10 alkyl, C3-10 cycloalkyl, or 3- to 10-membered heterocyclyl;
    • R″ is independently H or C1-6 alkyl;
    • R4 and R5 are independently C1-8 alkyl, which is optionally substituted with one or more R4s;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-14 cycloalkylene or 3- to 14-membered heterocyclylene, each of which is optionally substituted with one or more R4s;


R4s is independently H, halogen, cyano, C1-8 alkyl, C1-8 haloalkyl, -Ld-ORd, -Ld-SRd, or -Ld-NRdR′d;


Ld is independently a chemical bond or C1-8 alkylene;


Rd and R′d are independently H, C1-8 alkyl, C3-14 cycloalkyl, or 3- to 14-membered heterocyclyl.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein G1 and G2 are independently a chemical bond, C1-9 linear alkylene, C2-9 linear alkenylene, or C2-9 linear alkynylene.


In a more specific embodiment, G1 is C1-6 linear alkylene, C2-6 linear alkenylene, or C2-6 linear alkynylene, alternatively C1-6 linear alkylene, alternatively C2-6 linear alkylene.


In a more specific embodiment, G2 is a chemical bond, C1-6 linear alkylene, C2-6 linear alkenylene, or C2-6 linear alkynylene, alternatively a chemical bond or C1-6 linear alkylene, alternatively a chemical bond or C1-4 linear alkylene.


In a more specific embodiment, G1 and G2 have a total length of 3, 4, 5, 6, 7, 8, or 9 carbon atoms, alternatively a total length of 4, 5, or 6 carbon atoms, alternatively a total length of 5 or 6 carbon atoms; alternatively a total length of 5, 6 or 7 carbon atoms; alternatively a total length of 6 or 7 carbon atoms.

    • alternatively, G1 and G2 are optionally substituted with 1, 2, 3, or 4 RG1.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein RG1 is independently H or C1-10 alkyl, alternatively H or C1-6 alkyl.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein G3 is C4-10 linear alkylene, C4-10 linear alkenylene, or C4-10 linear alkynylene, alternatively C4-9 linear alkylene, alternatively C5-8 linear alkylene.


In a more specific embodiment, G3 is optionally substituted with 1, 2, 3, or 4 RG3.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein La is independently a chemical bond or C1-10 alkylene, alternatively a chemical bond or C1-6 alkylene.


In a more specific embodiment, Ra and R′a are independently H, C1-10 alkyl, C3-10 cycloalkyl, or 3- to 10-membered heterocyclyl, alternatively H or C1-6 alkyl.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein G4 is C1-4 alkylene, C2-4 alkenylene, or C2-4 alkynylene, alternatively C2-4 alkylene, alternatively C2-3 alkylene.


In a more specific embodiment, G4 is optionally substituted with 1, 2, 3, or 4 RG4.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein RG4 is independently H or C1-6 alkyl, alternatively C1-4 alkyl.


In a more specific embodiment, two RG4 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C3-10 cycloalkylene or 3- to 10-membered heterocyclylene, alternatively C3-7 cycloalkylene or 3- to 7-membered heterocyclylene, each of which is optionally substituted with 1, 2, or 3 R4g.


In a more specific embodiment, R4g is independently H, halogen, cyano, C1-6 alkyl, or C1-6 haloalkyl.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein Lb is independently a chemical bond or C1-4 alkylene.


In a more specific embodiment, Rb and R′b are independently H, C1-6 alkyl, C3-7 cycloalkyl, or 3- to 7-membered heterocyclyl, alternatively H or C1-6 alkyl, alternatively H or C1-4 alkyl.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein M1 and M2 are independently selected from —C(O)O—, —OC(O)—, —OC(O)O—, —SC(O)—, —C(O)S—, —C(O)NR—, and —NRC(O)—, alternatively —C(O)O—, —OC(O)O—, —OC(O)—, —SC(O)—, and —C(O)S—, alternatively —C(O)O—, —OC(O)—, —SC(O)—, and —C(O)S—, alternatively —C(O)O— and —OC(O)—.


In a more specific embodiment, M1 and M2 are independently selected from —C(O)O— and —C(O)S—, alternatively —C(O)O—.


In a more specific embodiment, one of M1 and M2 is —C(O)O— or —C(O)S—, alternatively —C(O)O—, and the other is —OC(O)— or —SC(O)—, alternatively —OC(O)—.


In a more specific embodiment, M1 is —OC(O)— or —SC(O)—, alternatively —OC(O)—; and M2 is —C(O)O— or —C(O)S—, alternatively —C(O)O—.


In a more specific embodiment, M1 is —C(O)O—, and M2 is —C(O)O— or —C(O)S—.


In a more specific embodiment, M1 and M2 are independently selected from —C(O)O—, —OC(O)—, —SC(O)—, —C(O)S—, —NHC(O)—, and —C(O)NH—.


In a more specific embodiment, M1 and M2 are independently selected from —C(O)O—, —OC(O)—, —C(O)S—, and —C(O)NH—, alternatively —C(O)O—, —OC(O)—, and —C(O)S—.


In a more specific embodiment, one of M1 and M2 is —C(O)O— or —C(O)S—, alternatively —C(O)O—, and the other is selected from —C(O)O—, —C(O)S—, —C(O)NH—, —OC(O)—, and —SC(O)—, alternatively —C(O)O—, —C(O)S—, —C(O)NH—, and —OC(O)—, alternatively —C(O)O—, —C(O)S—, —OC(O)—, and —SC(O)—, alternatively —C(O)O—, —C(O)S—, and —OC(O)—.


In a more specific embodiment, one of M1 and M2 is —OC(O)O—, and the other is selected from —C(O)O—, —OC(O)—, —SC(O)—, and —C(O)S—, alternatively —C(O)O— and —OC(O)—.


In a more specific embodiment, M1 is selected from —C(O)O—, —OC(O)—, —SC(O)—, and —C(O)S—, alternatively —C(O)O— and —OC(O)—, alternatively —C(O)O—, alternatively —OC(O)—, M2 is —OC(O)O—.


In a more specific embodiment, M1 is —OC(O)O—, M2 is —OC(O)— or —C(O)O—, alternatively —OC(O)—.


In a more specific embodiment, M1 and M2 are independently selected from —C(O)O—, —C(O)S—, —OC(O)—, —SC(O)—, and —OC(O)O—, alternatively —C(O)O—, —OC(O)—, and —OC(O)O—; alternatively M1 and M2 are not simultaneously —OC(O)O—.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein R1 and R2 are independently C6-14 alkyl, C6-14 alkenyl, or C6-14 alkynyl, alternatively C6-14 alkyl, alternatively C7-12 alkyl, alternatively C8-12 alkyl.


In a more specific embodiment, R1 and R2 are independently C7-12 alkyl, C7-12 alkenyl, or C7-12 alkynyl, alternatively C8-12 alkyl, C8-12 alkenyl, or C8-12 alkynyl.


In a more specific embodiment, R1 and R2 are optionally substituted with 1, 2, 3, or 4 R1s, alternatively optionally substituted with 1 R1s.


In a more specific embodiment, R1 and R2 are independently selected from the following groups: —(CH2)5CH3, —(CH2)6CH3, —(CH2)2CH3, —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3, —CH2—C≡C—(CH2)5CH3, —CH2—C≡C—(CH2)6CH3, —(CH2)2C≡C—(CH2)5CH3, —(CH2)4—C≡C—(CH2)3CH3, —CH2—CH≡CH—(CH2)5CH3, —CH2—CH═CH—(CH2)6CH3, —(CH2)2—CH═CH—(CH2)5CH3, —(CH2)4—CH═CH—(CH2)3CH3, —(CH2)5—CH═CH—CH2CH3,




embedded image


embedded image


embedded image


In a more specific embodiment, R1 and R2 are independently selected from the following groups: —(CH2)5CH3, —(CH2)6CH3, —(CH2)7CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3, —(CH2)11CH3, —CH2—C≡C—(CH2)5CH3, —CH2—C≡C—(CH2)6CH3, —(CH2)2C≡C—(CH2)5CH3, —(CH2)2—C≡C—(CH2)4CH3, —(CH2)3C≡C—(CH2)3CH3, —(CH2)4—C≡C—(CH2)3CH3, —CH2—CH═CH—(CH2)5CH3, —CH2—CH═CH—(CH2)6CH3—, —(CH2)2—CH═CH—(CH2)5CH3, —(CH2)4—CH═CH—(CH2)3CH3, —(CH2)5—CH═CH—CH2CH3,




embedded image


embedded image


embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein R1s is independently H, C1-4 alkyl, -LcORc, or -Lc-NRcR′c, alternatively H or C1-14 alkyl, alternatively H or C1-10 alkyl, alternatively H or C1-9 alkyl, alternatively H or C1-6 alkyl, alternatively H or C1-4 alky.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein R and R are each independently H or C1-20 alkyl, alternatively H or C1-14 alkyl, alternatively H or C1-9 alkyl, alternatively H or C1-6 alkyl; alternatively, R is H.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein Lc is independently a chemical bond or C1-14 alkylene, alternatively a chemical bond or C1-10 alkylene, alternatively a chemical bond or C1-6 alkylene.


In a more specific embodiment, Re and R′c are independently H or C1-14 alkyl, alternatively H or C1-10 alkyl, alternatively H or C1-6 alkyl.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein R3 is CN, —ORg, or —NRgR′g, alternatively —ORg or —NRgR′g, alternatively —ORg, alternatively —OH.


In a more specific embodiment, R3 is OH or —N(CH3)2.


In a more specific embodiment, Rg and R′g are independently H, C1-6 alkyl, C3-7 cycloalkyl, or 3- to 7-membered heterocyclyl, alternatively H or C1-6 alkyl, alternatively H or C1-4 alkyl, alternatively H or methyl.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein R4 and R5 are independently C1-6 alkyl, alternatively C1-3 alkyl, alternatively methyl.


In a more specific embodiment, R4 and R5 are optionally substituted with 1, 2, or 3 R4s.


In a more specific embodiment, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-10 cycloalkylene or 3- to 10-membered heterocyclylene, alternatively C3-6 cycloalkylene or 3- to 6-membered heterocyclylene, alternatively C3-6 cycloalkylene (e.g., cyclopropylene, cyclobutylene, cyclopentylene, or cyclohexylene), alternatively C3-5 cycloalkylene, alternatively cyclopropylene or cyclopentylene, alternatively cyclopropylene.


In a more specific embodiment, a ring formed by R4 and R5 with the carbon atom to which they are attached to is optionally substituted with 1, 2, or 3 R4s.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein R4s is independently H, halogen, cyano, C1-6 alkyl, C1-6 haloalkyl, -Ld-ORd, -Ld-SRd, or -Ld-NRdR′d, alternatively H, halogen, cyano, C1-6 alkyl, or C1-6 haloalkyl, alternatively H, C1-3 alkyl, or C1-3 haloalkyl.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein Ld is independently a chemical bond or C1-6 alkylene, alternatively a chemical bond or C1-3 alkylene.


In a more specific embodiment, Rd and R′d are independently H, C1-6 alkyl, C3-10 cycloalkyl, or 3- to 10-membered heterocyclyl, alternatively H or C1-6 alkyl.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein Le is independently a chemical bond or C1-6 alkylene, alternatively a chemical bond or C1-4 alkylene.


In a more specific embodiment, Re and R′e are independently H, C1-6 alkyl, C3-10 cycloalkyl, or 3- to 10-membered heterocyclyl, alternatively H or C1-6 alkyl, alternatively H or C1-4 alkyl.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein Q is a chemical bond, —OC(O)—, or —SC(O)—, alternatively a chemical bond or —SC(O)—.


In a more specific embodiment, when Q is phenylene or pyridinylene, Q is optionally substituted with 1, 2, or 3 R*.


In a more specific embodiment, R* is independently H, halogen, cyano, C1-3 alkyl, or C1-6 haloalkyl.


In a more specific embodiment, Lf is independently a chemical bond or C1-6 alkylene, alternatively a chemical bond or C1-4 alkylene.


In a more specific embodiment, Rf and R′f are independently H, C1-6 alkyl, C3-10 cycloalkyl, or 3- to 10-membered heterocyclyl, alternatively H or C1-6 alkyl, alternatively H or C1-4 alkyl.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, which has a structure of formula (II) or formula (III):




embedded image




    • wherein

    • a=1, 2, 3, 4, 5, or 6;

    • b=4, 5, 6, 7, 8, 9, or 10;

    • c=1, 2, 3, 4, 5, or 6;

    • d=0, 1, 2, 3, or 4;

    • c+d=3, 4, 5, 6, 7, 8, or 9;

    • the other groups are as defined herein.





In a more specific embodiment, the present disclosure provides the compound of formula (II) or formula (III), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,




embedded image




    • wherein, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 0-10 carbon atoms, alternatively 0-6 carbon atoms, alternatively 0-4 carbon atoms, alternatively 0-2 carbon atoms, alternatively 0 carbon atom.





In a more specific embodiment, the substitution site of R1s on R2 is separated from M2 by 1-10 carbon atoms, alternatively 1-6 carbon atoms, alternatively 1-4 carbon atoms, alternatively 1-2 carbon atoms; alternatively 2-10 carbon atoms, alternatively 2-6 carbon atoms, alternatively 2-4 carbon atoms.


In a more specific embodiment, R1 is not substituted with R1s, and the substitution site of R1s on R2 is separated from M2 by 0-10 carbon atoms, alternatively 1-10 carbon atoms, alternatively 1-6 carbon atoms, alternatively 1-4 carbon atoms, alternatively 1-2 carbon atoms; alternatively 2-10 carbon atoms, alternatively 2-6 carbon atoms, alternatively 2-4 carbon atoms.


In a more specific embodiment, R1 is substituted with R1s, and R2 is not substituted with R1s.


In a more specific embodiment, R4 and R5 are not taken together with the carbon atom to which they are attached to form a ring.


In a more specific embodiment, d is not 0.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,




embedded image




    • wherein

    • a=2, 3, 4, 5, or 6;

    • b=4, 5, 6, 7, 8, 9, or 10;

    • c=1, 2, 3, 4, 5, or 6;

    • d=0, 1, 2, 3, or 4;

    • c+d=3, 4, 5, 6, 7, 8, or 9; alternatively c+d=4, 5, or 6;

    • M1 and M2 are independently selected from —C(O)O—, —OC(O)—, —SC(O)—, and —C(O)S—; alternatively, M1 and M2 are independently selected from —C(O)O— and —C(O)S—; alternatively, one of M1 and M2 is —C(O)O— or —C(O)S—, and the other is —OC(O)— or —SC(O)—;

    • R1 and R2 are independently C6-14 alkyl, which is optionally substituted with 1, 2, 3, or 4 R1s;

    • R1s, is independently H, C1-14 alkyl, -Lc-ORc, or -Lc-NRcR′c, alternatively H or C1-14alkyl;

    • Lc is independently a chemical bond or C1-14 alkylene;

    • Rc and R′c are independently H or C1-14 alkyl;

    • R4 and R5 are independently C1-6 alkyl;

    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-6 cycloalkylene or 3- to 6-membered heterocyclylene.





In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4;
    • b=4, 5, 6, 7, 8, or 9; alternatively b=5, 6, 7, or 8; alternatively b=5, 6, or 7;
    • c=2, 3, 4, 5, or 6;
    • d=0, 1, 2, 3, or 4;
    • c+d=5 or 6;
    • M1 and M2 are independently —C(O)O— or —OC(O)—; alternatively, M1 and M2 are —C(O)O—;
    • alternatively, one of M1 and M2 is —C(O)O—, and the other is —OC(O)—;
    • R1 and R2 are independently C7-12 alkyl, alternatively C8-12 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently H or C1-10 alkyl, alternatively H or C1-9 alkyl;
    • R4 and R5 are independently C1-3 alkyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-6 cycloalkylene, alternatively C3-5 cycloalkylene.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4;
    • b=4, 5, 6, 7, 8, or 9; alternatively b=5, 6, 7, or 8; alternatively b=5 or 7;
    • c=2, 3, 4, 5, or 6;
    • d=0, 1, 2, 3, or 4; alternatively d=0, 1, 2, or 4;
    • c+d=5 or 6; alternatively c+d=6;
    • M1 and M2 are —C(O)O—;
    • or, one of M1 and M2 is —C(O)O—, and the other is —OC(O)—;
    • R1 and R2 are independently selected from: —(CH2)6CH3, —(CH2)7CH3, —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3,




embedded image


alternatively: —(CH2)7CH3, —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3,




embedded image


alternatively: —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3,




embedded image




    • R4 and R5 are methyl;

    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form cyclopropylene, cyclobutylene, cyclopentylene, or cyclohexylene, alternatively cyclopropylene or cyclopentylene, alternatively cyclopropylene.





In a more specific embodiment, only one of R1 and R2 is substituted.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2;
    • b=5;
    • c=2, 3, 4, 5, or 6; alternatively c=6;
    • d=0, 1, 2, 3, or 4; alternatively d=0;
    • c+d=5 or 6, alternatively 6;
    • M1 and M2 are —C(O)O—;
    • R1 is C7-12 alkyl, alternatively C3-12 alkyl, alternatively C8-9 alkyl, or alternatively C9 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently H or C1-10 alkyl, alternatively H or C1-9 alkyl, alternatively C5-8 alkyl, alternatively C6-8 alkyl, alternatively C7-8 alkyl;
    • R2 is C7-11 linear alkyl, alternatively C10-11 linear alkyl, alternatively C11 linear alkyl, which is optionally substituted with 1 C1-3 alkyl, alternatively optionally substituted with 1 methyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene.


In a more specific embodiment, R1 is




embedded image


alternatively




embedded image


In a more specific embodiment, R1 is




embedded image


alternatively




embedded image


In a more specific embodiment, R2 is —(CH2)9CH3, —(CH2)10CH3, or




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4; alternatively a=2;
    • b=7;
    • c=2, 3, 4, 5, or 6; alternatively c=2, 3, or 4; alternatively c=2;
    • d=0, 1, 2, 3, or 4; alternatively d=2, 3, or 4; alternatively d=4;
    • c+d=5 or 6, alternatively 6;
    • M1 and M2 are —C(O)O—;


R1 is C8-11 linear alkyl, alternatively C9-10 linear alkyl, alternatively C9 linear alkyl;

    • R2 is C7-12 alkyl, alternatively C8-12 alkyl, alternatively C9-10 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently H or C1-10 alkyl, alternatively H or C1-9 alkyl, alternatively C6-9 alkyl, alternatively C7-8 alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene.


In a more specific embodiment, R2 is




embedded image


alternatively




embedded image


alternatively




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2;
    • b=7;
    • c=3 or 4; alternatively c=4;
    • d=2;
    • M1 and M2 are —C(O)O—;
    • R1 is C7-12 alkyl, alternatively C8-12 alkyl, alternatively C8-10 alkyl, which is optionally substituted with 1 R1s;


R1s is independently H or C1-10 alkyl, alternatively H or C1-9 alkyl;


R2 is C7-11 linear alkyl, alternatively C9 linear alkyl;


R4 and R5 are independently C1-3 alkyl, alternatively methyl;


or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4; alternatively a=2 or 4;
    • b=6 or 7; alternatively b=7;
    • c=2, 3, 4, 5, or 6; alternatively c=5 or 6;
    • d=0, 1, 2, 3, or 4; alternatively d=0 or 1;
    • c+d=5 or 6, alternatively 6;
    • M1 is —OC(O)—; and M2 is —C(O)O—;
    • R1 is C7-11 linear alkyl, alternatively C9-10 linear alkyl;
    • R2 is C7-12 alkyl, alternatively C8-12 alkyl, alternatively C9-10 linear alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently C7-9 alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene.


In a more specific embodiment, when a=4, R4 and R5 are not taken together with the carbon atom to which they are attached to form a ring; alternatively, R1s is independently C8-9 alkyl.


In a more specific embodiment, R2 is




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,




embedded image




    • wherein

    • a=2, 3, 4, 5, or 6;

    • b=4, 5, 6, 7, 8, 9, or 10;

    • c=1, 2, 3, 4, 5, or 6;

    • d=0, 1, 2, 3, or 4;

    • c+d=3, 4, 5, 6, 7, 8, or 9; alternatively c+d=4, 5, 6, or 7;

    • M1 and M2 are independently selected from —C(O)O—, —OC(O)—, —SC(O)—, —C(O)S—, —NHC(O)—, and —C(O)NH—, alternatively —C(O)O—, —OC(O)—, —SC(O)—, and —C(O)S—; alternatively, one of M1 and M2 is —C(O)O— or —C(O)S—, and the other is —C(O)O—, —C(O)S—, —C(O)NH—, —OC(O)— or —SC(O)—, alternatively —C(O)O—, —C(O)S—, —OC(O)— or —SC(O)—;

    • R1 and R2 are independently C6-14 alkyl, C6-14 alkenyl, or C6-14 alkynyl, each of which is optionally substituted with 1, 2, 3, or 4 R1s;

    • R1s is independently H, C1-14 alkyl, -Lc-ORc, or -Lc-NRcR′c, alternatively H or C1-14alkyl;

    • Lc is independently a chemical bond or C1-14 alkylene;

    • Rc and R′c are independently H or C1-14 alkyl;

    • R4 and R5 are independently C1-6 alkyl;

    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-6 cycloalkylene or 3- to 6-membered heterocyclylene.





In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4;
    • b=4, 5, 6, 7, 8, or 9, alternatively b=5, 6, 7, or 8, alternatively b=5, 6, or 7;
    • c=2, 3, 4, 5, or 6;
    • d=0, 1, 2, 3, or 4;
    • c+d=5, 6, or 7;
    • M1 and M2 are independently —C(O)O—, —OC(O)—, —C(O)S— or —C(O)NH—, alternatively —C(O)O—, —OC(O)—, or —C(O)S—; alternatively, one of M1 and M2 is —C(O)O—, and the other is —C(O)O—, —C(O)S—, —C(O)NH—, or —OC(O)—, alternatively —C(O)O—, —C(O)S—, or —OC(O)—;
    • R1 and R2 are independently C7-12 alkyl, C7-12 alkenyl, or C7-12 alkynyl, alternatively C8-12 alkyl, C8-12 alkenyl, or C8-12 alkynyl, each of which is optionally substituted with 1 R1s;
    • R1s is independently H or C1-10 alkyl;
    • R4 and R5 are independently C1-3 alkyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-6 cycloalkylene, alternatively C3-5 cycloalkylene.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4;
    • b=4, 5, 6, 7, 8, or 9; alternatively b=5, 6, 7, or 8; alternatively b=5 or 7;
    • c=2, 3, 4, 5, or 6;
    • d=0, 1, 2, 3, or 4;
    • c+d=5, 6, or 7; alternatively c+d=6;
    • one of M1 and M2 is —C(O)O—, and the other is —C(O)O—, —C(O)S—, —C(O)NH—, or —OC(O)—, alternatively —C(O)O—, —C(O)S—, or —OC(O)—;
    • R1 and R2 are independently selected from: —(CH2)6CH3, —(CH2)7CH3, —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3, —CH2CH═CH—(CH2)5CH3, —CH2—C≡C—(CH2)5CH3, —(CH2)2—C≡C—(CH2)4CH3, —(CH2)3—C≡C—(CH2)3CH3,




embedded image


embedded image


alternatively: —(CH2)7CH3, —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3, —(CH2)11CH3, —CH2—CH═CH—(CH2)5CH3, —CH2—C≡C—(CH2)5CH3, —(CH2)2—C≡C—(CH2)4CH3, —(CH2)—C≡C—(CH2)3CH3,




embedded image


embedded image


alternatively: —(CH2)8CH3, —(CH2)9CH3, —(CH2)20CH3, —(CH2)11CH3, —CH2—CH═CH—(CH2)—CH3, —CH2—C≡C—(CH2)5CH3, —(CH)2—C≡C—(CH2)4CH3, —(CH2)3C≡C—(CH2)CH3,




embedded image


embedded image




    • R4 and R5 are methyl;

    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form cyclopropylene, cyclobutylene, cyclopentylene, or cyclohexylene, alternatively cyclopropylene or cyclopentylene, alternatively cyclopropylene;





In a more specific embodiment, only one of R1 and R2 is substituted.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2;
    • b=5;
    • c=5 or 6; alternatively c=6;
    • d=0 or 1; alternatively d=0;
    • c+d=5 or 6, alternatively 6;
    • M1 is —C(O)O—, and M2 is —C(O)O— or —C(O)S—; alternatively M1 and M2 are —C(O)O—;
    • R1 is C7-12 alkyl, alternatively C3-12 alkyl, alternatively C8-9 alkyl, or alternatively C9 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently H or C1-10 alkyl, alternatively H or C1-9 alkyl, alternatively C6-8 alkyl, alternatively C7-8 alkyl;
    • R2 is C7-11 linear alkyl, alternatively C10-11 linear alkyl, alternatively C11 linear alkyl, which is optionally substituted with 1 C1-3 alkyl, alternatively optionally substituted with 1 methyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene;


In a more specific embodiment, R1 is




embedded image


alternatively




embedded image


In a more specific embodiment, R2 is —(CH2)9CH3, —(CH2)10CH3, or




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4; alternatively a=2;
    • b=5, 6, or 7; alternatively b=7;
    • c=2, 3, 4, 5, or 6; alternatively c=2, 3, or 4; alternatively c=2;
    • d=0, 1, 2, 3, or 4; alternatively d=2, 3, or 4; alternatively d=4;
    • c+d=5 or 6, alternatively 6;
    • M1 and M2 are —C(O)O—;
    • R1 and R2 are independently C7-12 alkyl, alternatively C8-12 alkyl, alternatively C9-11 alkyl, alternatively C9-10 alkyl, which is optionally substituted with 1 R1s; and only one of R1 and R2 is substituted;
    • R1s is independently H or C1-10 alkyl, alternatively C6-10 alkyl, alternatively C7-9 alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene, alternatively not to form a ring;


In a more specific embodiment, R1 is C8-12 linear alkyl, alternatively C9-11 linear alkyl, alternatively C9-10 linear alkyl;


R2 is C7-12 alkyl, alternatively C8-12 alkyl, alternatively C9-11 alkyl, alternatively C9-10 alkyl, which is optionally substituted with 1 R1s; when R2 is C10 linear alkyl, R1s is C9-10 alkyl, alternatively C9 alkyl.


In a more specific embodiment, R2 is




embedded image


alternatively




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4;
    • b=6;
    • c=4, 5, or 6; alternatively c=5 or 6; alternatively c=5;
    • d=1 or 2; alternatively d=1;
    • c+d=6, 7, or 8; alternatively 6 or 7; alternatively 6;
    • M1 is —OC(O)—; M2 is —C(O)O—;
    • R1 is C7-12 linear alkyl, alternatively C8-11 linear alkyl, alternatively C9-11 linear alkyl;
    • R2 is C7-12 alkyl, alternatively C8-12 alkyl, alternatively C9-12 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently C7-11 alkyl, alternatively C7-10 alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene;
    • provided that, when c=4, a=2 or 3.


In a more specific embodiment, R2 is




embedded image


In a more specific embodiment, a=2 or 4; b=6; c=5 or 6, alternatively c=5; d=1;


R1 is C8-9 linear alkyl, alternatively C9 linear alkyl;

    • R2 is C9-10 alkyl, alternatively C10 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently C7-8 alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl.


In a more specific embodiment, a=2, 3 or 4, alternatively 2 or 4; b=6; c=5; d=2;

    • R1 is C9-10 linear alkyl, alternatively C9 linear alkyl;
    • R2 is C9-11 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently C7-9 alkyl,
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4; alternatively a=2 or 4;
    • b=7;
    • c=5 or 6; alternatively c=5;
    • d=0 or 1; alternatively d=1;
    • c+d=5 or 6, alternatively 6;
    • M1 is —OC(O)—; M2 is —C(O)O—;
    • R1 is C8-11 linear alkyl, alternatively C9-10 linear alkyl, which is optionally substituted with 1 C1-9 alkyl (alternatively C6-9 alkyl, alternatively C6-7 alkyl);
    • R2 is C7-12 alkyl, alternatively C8-12 alkyl, alternatively C9-10 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently C7-9 alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene.


In a more specific embodiment, R1 is —(CH2)8CH3, —(CH2)9CH3,




embedded image




    • R2 is







embedded image


In a more specific embodiment, when a=4,

    • R1 is C9-10 linear alkyl;
    • R2 is C9-10 alkyl, which is optionally substituted with 1 R1s;


R1s is C8-9 alkyl;


R4 and R5 are not taken together with the carbon atom to which they are attached to form a ring.


In a more specific embodiment, when a=2,

    • R1 is C10 linear alkyl;
    • alternatively R1s is C8-9 alkyl, alternatively C9 alkyl.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4; alternatively a=2 or 4;
    • b=7;
    • c=4;
    • d=2;
    • M1 is —OC(O)—; M2 is —C(O)O—;
    • R1 is C8-11 linear alkyl, alternatively C9-10 linear alkyl;
    • R2 is C8-11 alkyl, alternatively C9-10 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently C7-9 alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene, alternatively not to form a ring.


In a more specific embodiment, R2 is




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4; alternatively a=2;
    • b=8;
    • c=5;
    • d=1;
    • M1 is —OC(O)—; M2 is —C(O)O—;
    • R1 is C8-12 linear alkyl, alternatively C9-11 linear alkyl, alternatively C10-11 linear alkyl;
    • R2 is C8-11 alkyl, alternatively C9-10 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently C7-9 alkyl, alternatively C7-8 alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene, alternatively not to form a ring.


In a more specific embodiment, R2 is




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2;
    • b=6, 7, or 8, alternatively b=7;
    • c=5;
    • d=1;
    • M1 is —OC(O)—; M2 is —C(O)O—;


R1 is C8-11 linear alkyl, alternatively C9-10 linear alkyl, alternatively C9 linear alkyl;

    • R2 is C8-11 alkyl, alternatively C9-10 alkyl, alternatively C9 alkyl, which is optionally substituted with 1 R1s;


R1s is independently C7 alkyl;


R4 and R5 are independently C1-3 alkyl, alternatively methyl.


In a more specific embodiment, R2 is




embedded image


alternatively




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2 or 3, alternatively a=2;
    • b=7;
    • c=3;
    • d=3;
    • M1 is —OC(O)—; M2 is —C(O)O—;
    • R1 is C8-11 linear alkyl, alternatively C9-10 linear alkyl, alternatively C9 linear alkyl;
    • R2 is C8-11 alkyl, alternatively C9-10 alkyl, alternatively C9 alkyl, which is optionally substituted with 1 R1s;


R1s is independently C6-7 alkyl, alternatively C7 alkyl;


R4 and R5 are independently C1-3 alkyl, alternatively methyl.


In a more specific embodiment, R2 is




embedded image


alternatively




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,




embedded image




    • wherein

    • a=2, 3, 4, 5, or 6;

    • b=4, 5, 6, 7, 8, 9, or 10;

    • c=1, 2, 3, 4, 5, or 6;

    • d=0, 1, 2, 3, or 4;

    • c+d=3, 4, 5, 6, 7, 8, or 9, alternatively c+d=4, 5, or 6;

    • one of M1 and M2 is —OC(O)O—, and the other is —C(O)O—, —OC(O)—, —SC(O)—, or —C(O)S—;

    • R1 and R2 are independently C6-14 alkyl, which is optionally substituted with 1, 2, 3, or 4 R1s;

    • R1s is independently H, C1-14 alkyl, -Lc-ORc, or -Lc-NRcR′c; alternatively H or C1-14 alkyl;

    • Lc is independently a chemical bond or C1-14 alkylene;

    • Rc and R′c are independently H or C1-14 alkyl;





R4 and R5 are independently C1-6 alkyl;

    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-6 cycloalkylene or 3- to 6-membered heterocyclylene.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4;
    • b=4, 5, 6, or 7, alternatively b=5 or 6, alternatively b=6;
    • c=2, 3, 4, 5, or 6; alternatively c=2, 4, 5, or 6;
    • d=0, 1, 2, 3, or 4; alternatively d=0, 1, 2, or 4;
    • c+d=5 or 6, alternatively c+d=6;
    • one of M1 and M2 is —OC(O)O—, and the other is —C(O)O— or —OC(O)—;
    • R1 and R2 are independently C7-12 alkyl, alternatively C9-11 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently H or C1-10 alkyl; alternatively H or C4-10 alkyl;
    • R4 and R5 are independently C1-3 alkyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-6 cycloalkylene, alternatively C3-4 cycloalkylene.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3, or 4;
    • b=4, 5, 6, or 7, alternatively b=5 or 6, alternatively b=6;
    • c=2, 3, 4, 5, or 6; alternatively c=2, 4, 5, or 6;
    • d=0, 1, 2, 3, or 4; alternatively d=0, 1, 2, or 4;
    • c+d=5 or 6, alternatively c+d=6;
    • one of M1 and M2 is —OC(O)O—, and the other is —C(O)O— or —OC(O)—;
    • R1 and R2 are independently selected from —(CH2)8CH3, —(CH2)9CH3, —(CH2)10CH3,




embedded image




    • R4, and R5 are methyl;

    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form cyclopropylene.





In a more specific embodiment, only one of R1 and R2 is substituted;


In a more specific embodiment, M1 is —C(O)O— or —OC(O)—, alternatively —C(O)O—, and M2 is —OC(O)O—.


In a more specific embodiment, M1 is —OC(O)O—, and M2 is —OC(O)—.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2;
    • b=5 or 6, alternatively b=6;
    • c=4, 5, or 6; alternatively c=4 or 6; alternatively c=6;
    • d=0, 1, or 2; alternatively d=0 or 2;
    • c+d=6;
    • M1 is —C(O)O—, and M2 is —OC(O)O—;
    • R1 is C8-11 alkyl, alternatively C9-10 alkyl, alternatively C9 alkyl, which is optionally substituted with 1 R1s;
    • R1s is C7-9 alkyl, alternatively C7 alkyl;
    • R2 is C8-11 linear alkyl, alternatively C9-10 linear alkyl, alternatively C9 linear alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3 cycloalkylene, alternatively not to form a ring.


In a more specific embodiment, R1 is




embedded image


alternatively




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2;
    • b=6;
    • c=4, 5, or 6; alternatively c=5 or 6; alternatively c=6;
    • d=0, 1, or 2; alternatively d=0 or 1;
    • c+d=6;
    • M1 is —C(O)O—, and M2 is —OC(O)O—;


R1 is C8-10 linear alkyl, alternatively C9-10 linear alkyl, alternatively C9 linear alkyl;

    • R2 is C8-10 alkyl, alternatively C9-10 alkyl, alternatively C9 alkyl, which is optionally substituted with 1 R1s;
    • R1s is C6-7 alkyl, alternatively C7 alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively not to form a ring.


In a more specific embodiment, R2 is




embedded image


alternatively




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2;
    • b=5 or 6; alternatively b=6;
    • c=5;
    • d=1;
    • M1 is —OC(O)—, and M2 is —OC(O)O—;
    • R1 is C8-10 alkyl, alternatively C9-10 alkyl, alternatively C9 alkyl, which is optionally substituted with 1 R1s;
    • R1s is C6-8 alkyl, alternatively C6-7 alkyl;
    • R2 is C8-12 linear alkyl, alternatively C10-11 linear alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively not to form a ring.


In a more specific embodiment, R1 is




embedded image


alternatively




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • a=2, 3 or 4; alternatively a=2;
    • b=5, 6, 7, or 8; alternatively b=6 or 7; alternatively b=7;
    • c=4, 5, or 6; alternatively c=4;
    • d=0, 1, or 2; alternatively d=1;
    • c+d=5 or 6; alternatively c+d=5;
    • M1 is —OC(O)O—, M2 is —OC(O)—;
    • R1 is C8-12 alkyl, alternatively C8-10 alkyl, which is optionally substituted with 1 R1s;
    • R1s is C6-10 alkyl, alternatively C6-8 alkyl;
    • R2 is C8-12 linear alkyl, alternatively C8-10 linear alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively not to form a ring.


In a more specific embodiment, R1 is




embedded image


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein


In a more specific embodiment, the present disclosure provides the compound of formula (III), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,




embedded image




    • wherein

    • Q is —SC(O)— or —OC(O)—;

    • Rg and R′g are independently C1-6 alkyl;

    • a=2, 3, 4, 5, or 6; alternatively a=2, 3, or 4;

    • b=4, 5, 6, 7, 8, or 9; alternatively a=5, 6, or 7;

    • c=2, 3, 4, 5, or 6;

    • d=0, 1, 2, 3, or 4;

    • c+d=5 or 6;

    • M1 and M2 are independently selected from —C(O)O—, —C(O)S—, —OC(O)—, —SC(O)—, and —OC(O)O—;

    • R1 and R2 are independently C6-14 alkyl, which is optionally substituted with 1, 2, 3, or 4 R1s;

    • R1s is independently H, C1-14 alkyl, -Lc-ORc, or -Lc-NRcR′c; alternatively H or C1-14 alkyl;

    • Lc is independently a chemical bond or C1-14 alkylene;

    • Rc and R′c are independently H or C1-14a alkyl;

    • R4 and R5 are independently C1-6 alkyl,

    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-6 cycloalkylene or 3- to 6-membered heterocyclylene.





In a more specific embodiment, the present disclosure provides the compound of formula (III), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • Q is —SC(O)— or —OC(O)—;
    • Rg and R′g are independently C1-3 alkyl;
    • a=3 or 4;
    • b=6 or 7;
    • c=2, 3, 4, or 5; alternatively 2 or 5;
    • d=1, 2, 3, or 4; alternatively 1 or 4;
    • c+d=5 or 6;
    • M1 and M2 are independently selected from —C(O)O—, —OC(O)—, and —OC(O)O—; alternatively, M1 and M2 are not simultaneously —OC(O)O—;
    • R1 and R2 are independently C7-12 alkyl, alternatively C8-10 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently H or C1-10 alkyl; alternatively H or C1-9 alkyl;
    • R4 and R5 are independently C1-3 alkyl,
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-6 cycloalkylene.


In a more specific embodiment, the present disclosure provides the compound of formula (III), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • Q is —SC(O)—;
    • Rg and R′g are methyl;
    • a=3 or 4;
    • b=6 or 7;
    • c=2, 4, or 5; alternatively 2 or 5;
    • d=1, 2, or 4; alternatively 1 or 4;
    • c+d=6;
    • M1 and M2 are independently selected from —C(O)O—, —OC(O)—, and —OC(O)O—; alternatively, M1 and M2 are not simultaneously —OC(O)O—;
    • R1 and R2 are independently —(CH2)7CH3, —(CH2)8CH3, —(CH2)9CH3,




embedded image


R4 and R5 are methyl;


or, R4 and R5 are taken together with the carbon atom to which they are attached to form cyclopropylene, alternatively not to form a ring.


In a more specific embodiment, only one of R1 and R2 is substituted; alternatively, R1 is unsubstituted, and R2 is substituted.


In a more specific embodiment, the present disclosure provides the compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,




embedded image




    • wherein M1 and M2 are independently selected from —C(O)O— and —C(O)S—, alternatively —C(O)O—;

    • the other groups are as defined herein.





In a more specific embodiment, the present disclosure provides the compound of formula (II) or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,




embedded image




    • wherein one of M1 and M2 is —C(O)O— or —C(O)S—, alternatively —C(O)O—, and the other is —OC(O)— or —SC(O)—, alternatively —OC(O)—;

    • the other groups are as defined herein.





In a more specific embodiment, R1s is independently H or C1-6 alkyl, alternatively H or C1-4 alkyl.


In a more specific embodiment, M1 is —OC(O)— or —SC(O)—, alternatively —OC(O)—; and M2 is —C(O)O— or —C(O)S—, alternatively —C(O)O—.


In a more specific embodiment, R1s is independently C1-8 alkyl, alternatively C4-8 alkyl, alternatively C6-8 alkyl, alternatively C7-8 alkyl, alternatively C8 alkyl;


In a more specific embodiment, the present disclosure provides the compound of formula (II) or formula (III), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,




embedded image




    • wherein

    • Q is a chemical bond, —SC(O)— or —OC(O)—;

    • Rg and R′g are independently C1-6 alkyl;





a=2, 3, 4, 5, or 6;

    • b=6 or 7;
    • c=5 or 6;
    • d=0 or 1;
    • c+d=5 or 6;
    • M1 is —OC(O)— or —SC(O)—; and M2 is —C(O)O— or —C(O)S—;
    • R1 and R2 are independently C7-12 alkyl, alternatively C8-12 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently H or C1-10 alkyl;
    • R4 and R5 are independently C1-6 alkyl,
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-6 cycloalkylene.


In a more specific embodiment, only one of R1 and R2 is substituted; alternatively, R1 is unsubstituted, and R2 is substituted.


In a more specific embodiment, the present disclosure provides the compound of formula (II) or formula (III), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein

    • Q is —SC(O)— or —OC(O)—, alternatively —SC(O)—;
    • Rg and R′g are independently C1-8 alkyl, alternatively methyl;
    • a=2, 3, or 4; alternatively a=2 or 4; alternatively a=4;
    • b=6 or 7; alternatively b=7;
    • c=5 or 6; alternatively c=5;
    • d=0 or 1; alternatively d=1;
    • c+d=5 or 6; alternatively c+d=6;
    • M1 is —OC(O)—; M2 is —C(O)O—;
    • R1 is C7-11 linear alkyl, alternatively C9-10 linear alkyl; alternatively C9 linear alkyl;
    • R2 is C7-12 alkyl, alternatively C8-12 alkyl, alternatively C9-10 alkyl, alternatively C9 alkyl, which is optionally substituted with 1 R1s;
    • R1s is independently C1-9 alkyl, alternatively C1-8 alkyl, alternatively C4-9 alkyl, alternatively C4-8 alkyl;
    • R4 and R5 are independently C1-3 alkyl, alternatively methyl;
    • or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene.


In a more specific embodiment, the substitution site of R1s on R1 or R2 is separated from M1 or M2 by 0-10 carbon atoms, alternatively 0-6 carbon atoms, alternatively 0-4 carbon atoms, alternatively 0-2 carbon atoms, alternatively 0 carbon atom.


In a more specific embodiment, R1 is not substituted with R1s, and the substitution site of R2s, on R2 is separated from M2 by 0-10 carbon atoms, alternatively 1-10 carbon atoms, alternatively 1-6 carbon atoms, 1-4 carbon atoms, 1-2 carbon atoms; alternatively 2-10 carbon atoms, alternatively 2-6 carbon atoms, 2-4 carbon atoms.


In a more specific embodiment, R2 is




embedded image


alternatively




embedded image


alternatively




embedded image


alternatively




embedded image


In a more specific embodiment, R4 and R5 are not taken together with the carbon atom to which they are attached to form a ring.


In a more specific embodiment, the present disclosure provides the compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein the compound is selected from the compounds in Table (I).










TABLE I









embedded image


1a







embedded image


3a







embedded image


4a







embedded image


5a







embedded image


6a







embedded image


7a







embedded image


8a







embedded image


9a







embedded image


10a







embedded image


11a







embedded image


12a







embedded image


13a







embedded image


14a







embedded image


15a







embedded image


16a







embedded image


17a







embedded image


18a







embedded image


19a







embedded image


20a







embedded image


21a







embedded image


22a







embedded image


23a







embedded image


24a







embedded image


25a







embedded image


26a







embedded image


27a







embedded image


28a







embedded image


29a







embedded image


1b







embedded image


2b







embedded image


3b







embedded image


5b







embedded image


6b







embedded image


7b







embedded image


8b







embedded image


9b







embedded image


10b







embedded image


11b







embedded image


12b







embedded image


13b







embedded image


14b







embedded image


15b







embedded image


16b







embedded image


17b







embedded image


18b







embedded image


19b







embedded image


20b







embedded image


21b







embedded image


22b







embedded image


23b







embedded image


24b







embedded image


30a







embedded image


31a







embedded image


32a







embedded image


33a







embedded image


34a







embedded image


35a







embedded image


36a







embedded image


37a







embedded image


38a







embedded image


39a







embedded image


40a







embedded image


41a







embedded image


42a







embedded image


43a







embedded image


44a







embedded image


45a







embedded image


46a







embedded image


47a







embedded image


48a







embedded image


49a







embedded image


50a







embedded image


51a







embedded image


25b







embedded image


26b







embedded image


27b







embedded image


28b







embedded image


29b







embedded image


30b







embedded image


31b







embedded image


32b







embedded image


33b







embedded image


34b







embedded image


35b







embedded image


36b







embedded image


37b







embedded image


52a







embedded image


53a







embedded image


54a







embedded image


55a







embedded image


56a







embedded image


57a







embedded image


58a







embedded image


59a







embedded image


60a







embedded image


61a







embedded image


62a







embedded image


63a







embedded image


64a







embedded image


65a







embedded image


66a







embedded image


67a







embedded image


68a







embedded image


69a







embedded image


70a







embedded image


38b







embedded image


39b







embedded image


40b







embedded image


41b







embedded image


42b







embedded image


43b







embedded image


44b







embedded image


45b







embedded image


46b







embedded image


47b







embedded image


48b







embedded image


49b







embedded image


50b







embedded image


51b







embedded image


52b







embedded image


53b







embedded image


54b







embedded image


55b







embedded image


56b







embedded image


57b







embedded image


58b







embedded image


59b







embedded image


60b







embedded image


61b







embedded image


62b







embedded image


63b







embedded image


64b







embedded image


65b







embedded image


66b







embedded image


67b







embedded image


68b







embedded image


69b







embedded image


70b







embedded image


71b







embedded image


72b







embedded image


73b







embedded image


74b







embedded image


75b







embedded image


76b







embedded image


77b







embedded image


78b







embedded image


79b







embedded image


80b







embedded image


81b







embedded image


82b







embedded image


83b







embedded image


84b







embedded image


85b







embedded image


86b







embedded image


87b







embedded image


88b







embedded image


89b







embedded image


90b







embedded image


91b







embedded image


92b







embedded image


93b







embedded image


94b







embedded image


95b







embedded image


96b







embedded image


97b







embedded image


98b







embedded image


99b







embedded image


100b







embedded image


101b







embedded image


102b







embedded image


103b







embedded image


104b







embedded image


105b







embedded image


106b







embedded image


107b







embedded image


108b







embedded image


109b







embedded image


110b







embedded image


111b







embedded image


112b







embedded image


113b







embedded image


114b







embedded image


115b







embedded image


116b







embedded image


117b







embedded image


1c







embedded image


2c







embedded image


3c







embedded image


4c







embedded image


5c







embedded image


6c







embedded image


7c







embedded image


8c







embedded image


9c







embedded image


10c







embedded image


11c







embedded image


12c







embedded image


13c







embedded image


14c







embedded image


15c







embedded image


16c







embedded image


17c







embedded image


18c







embedded image


19c







embedded image


20c







embedded image


21c







embedded image


22c







embedded image


23c







embedded image


24c







embedded image


25c







embedded image


26c







embedded image


27c







embedded image


28c







embedded image


29c







embedded image


30c







embedded image


31c







embedded image


32c







embedded image


33c







embedded image


34c







embedded image


35c







embedded image


36c







embedded image


37c







embedded image


38c







embedded image


39c







embedded image


40c







embedded image


41c







embedded image


42c







embedded image


43c







embedded image


44c







embedded image


45c







embedded image


46c







embedded image


47c







embedded image


48c







embedded image


49c







embedded image


50c







embedded image


51c







embedded image


52c







embedded image


53c







embedded image


54c







embedded image


55c







embedded image


56c







embedded image


57c







embedded image


58c







embedded image


59c







embedded image


60c







embedded image


61c







embedded image


62c







embedded image


63c







embedded image


64c







embedded image


65c







embedded image


66c







embedded image


67c







embedded image


68c







embedded image


69c







embedded image


70c







embedded image


71c







embedded image


72c







embedded image


73c







embedded image


74c







embedded image


75c







embedded image


76c







embedded image


77c







embedded image


78c







embedded image


79c







embedded image


80c







embedded image


81c







embedded image


82c







embedded image


83c







embedded image


84c







embedded image


85c







embedded image


86c







embedded image


87c







embedded image


88c







embedded image


89c







embedded image


90c







embedded image


91c







embedded image


92c







embedded image


93c







embedded image


94c







embedded image


95c







embedded image


96c







embedded image


97c







embedded image


98c







embedded image


99c







embedded image


100c







embedded image


101c







embedded image


102c







embedded image


103c







embedded image


104c







embedded image


105c







embedded image


106c







embedded image


107c







embedded image


108c







embedded image


109c







embedded image


110c







embedded image


111c







embedded image


112c







embedded image


113c







embedded image


114c







embedded image


115c







embedded image


116c







embedded image


117c







embedded image


118c







embedded image


119c







embedded image


120c







embedded image


121c







embedded image


122c







embedded image


123c







embedded image


124c







embedded image


125c







embedded image


126c







embedded image


127c







embedded image


128c







embedded image


129c







embedded image


130c







embedded image


131c







embedded image


132c







embedded image


133c







embedded image


134c







embedded image


135c







embedded image


136c







embedded image


137c







embedded image


138c







embedded image


139c







embedded image


140c







embedded image


141c







embedded image


142c







embedded image


143c







embedded image


144c







embedded image


145c







embedded image


146c







embedded image


147c







embedded image


148c







embedded image


149c







embedded image


150c







embedded image


151c







embedded image


152c







embedded image


153c







embedded image


154c







embedded image


155c







embedded image


156c







embedded image


157c







embedded image


158c







embedded image


159c







embedded image


160c







embedded image


161c







embedded image


162c







embedded image


163c







embedded image


164c







embedded image


165c







embedded image


166c







embedded image


167c







embedded image


168c







embedded image


169c







embedded image


170c







embedded image


171c







embedded image


172c







embedded image


173c







embedded image


174c







embedded image


175c







embedded image


176c







embedded image


177c







embedded image


178c







embedded image


179c







embedded image


180c







embedded image


181c







embedded image


182c







embedded image


183c







embedded image


184c







embedded image


185c







embedded image


186c







embedded image


187c







embedded image


188c







embedded image


189c







embedded image


190c







embedded image


191c







embedded image


192c







embedded image


193c







embedded image


194c







embedded image


195c







embedded image


196c







embedded image


197c







embedded image


198c







embedded image


199c







embedded image


200c







embedded image


201c







embedded image


202c







embedded image


203c







embedded image


204c







embedded image


205c







embedded image


206c







embedded image


207c







embedded image


208c







embedded image


209c







embedded image


210c







embedded image


211c







embedded image


212c







embedded image


213c







embedded image


214c







embedded image


215c







embedded image


216c







embedded image


217c







embedded image


218c







embedded image


219c







embedded image


220c







embedded image


221c







embedded image


222c







embedded image


223c







embedded image


224c







embedded image


225c







embedded image


226c







embedded image


227c







embedded image


228c







embedded image


229c







embedded image


230c







embedded image


231c







embedded image


232c







embedded image


233c







embedded image


234c







embedded image


235c







embedded image


236c







embedded image


237c







embedded image


238c







embedded image


239c







embedded image


240c







embedded image


241c







embedded image


242c







embedded image


243c







embedded image


244c







embedded image


245c







embedded image


246c







embedded image


247c







embedded image


248c







embedded image


249c







embedded image


250c







embedded image


251c







embedded image


252c







embedded image


253c







embedded image


254c







embedded image


255c







embedded image


256c







embedded image


257c







embedded image


258c







embedded image


259c







embedded image


260c







embedded image


261c







embedded image


262c







embedded image


263c







embedded image


264c







embedded image


265c







embedded image


266c







embedded image


267c







embedded image


268c







embedded image


269c







embedded image


270c







embedded image


271c







embedded image


272c







embedded image


273c







embedded image


274c







embedded image


275c







embedded image


276c







embedded image


277c







embedded image


278c







embedded image


279c







embedded image


280c







embedded image


281c







embedded image


282c







embedded image


283c









The present disclosure further provides a method for preparing a compound of formula (II), comprising:

    • reacting a compound of formula (IIA) with a compound of formula (IIB) to give the compound of formula (II),




embedded image




    • wherein X is halogen, and the other variables are as defined herein.





The present disclosure further provides a method for preparing a compound of formula (II), comprising:

    • reacting a compound of formula (IIC) with a compound of formula (IID) to give the compound of formula (II),




embedded image




    • wherein X is halogen, and the other variables are as defined herein.





In a specific embodiment, the compound is selected from:




text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


In a more specific embodiment, the present disclosure provides a nanoparticle composition comprising a lipid component, and optionally a load, wherein the lipid component comprises the compound of the present disclosure.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the lipid component comprises the following components in molar percentage:

    • ionizable cationic lipids 20 mol %-85 mol %;
    • structural lipids 10 mol %-75 mol %;
    • neutral lipids 1.0 mol %-30 mol %;
    • polymeric lipids 0.25 mol %-10 mol %.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the lipid component comprises the following components in molar percentage:

    • any of the above-mentioned compound of the present disclosure 50 mol %;
    • neutral lipids 10 mol %;
    • structural lipids 38.5 mol %;
    • polymeric lipids 1.5 mol %.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the neutral lipids are selected from one or more of DSPC, DMPC, DOPC, DPPC, POPC, DOPE, DMPE, POPE and DPPE, alternatively DSPC and/or DOPE.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the structure lipids are selected from one or more of cholesterol, sitosterol, coprosterol, fucosterol, brassicasterol, ergosterol, tomatine, ursolic acid, α-tocopherol, stigmasterol, avenasterol, ergocalciferol and campesterol, alternatively cholesterol and/or β-sitosterol, still alternatively cholesterol.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the polymer lipids are polyethylene glycolated lipids.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the polyethylene glycolated lipids are selected from one or more of: PEG modified phosphatidylethanolamine, PEG modified phosphatidic acid, PEG modified ceramide, PEG modified dialkyl amine, PEG modified diacylglycerol, and PEG modified dialkylglycerol.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the polyethylene glycolated lipids contain a PEG moiety of about 1000 Da to about 20 kDa, alternatively a PEG moiety of about 1000 Da to about 5000 Da.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the polyethylene glycolated lipids are selected from one or more of DMPE-PEG1000, DPPE-PEG1000, DSPE-PEG1000, DOPE-PEG1000, DMG-PEG2000, Ceramide-PEG2000, DMPE-PEG2000, DPPE-PEG2000, DSPE-PEG2000, Azido-PEG2000, DSPE-PEG2000-Mannose, Ceramide-PEG5000, and DSPE-PEG5000, alternatively DMG-PEG2000.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the load is selected from one or more of therapeutic, prophylactic and diagnostic agents.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the therapeutic, prophylactic or diagnostic agent is a nucleic acid.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the nucleic acid is selected from one or more of ASO, RNA and DNA.


In a more specific embodiment, the present disclosure provides the nanoparticle composition described above, wherein, the RNA is selected from one or more of interfering RNA (RNAI), small interfering RNA (siRNA), short hairpin RNA (shRNA), antisense RNA (aRNA), messenger RNA (mRNA), modified messenger RNA (mmRNA), long non-coding RNA (lncRNA), microRNA (miRNA), small activating RNA (saRNA), multimeric coding nucleic acid (MCNA), polymeric coding nucleic acid (PCNA), guide RNA (gRNA), CRISPRRNA (crRNA), and nucleases, alternatively mRNA, still alternatively, modified mRNA.


The compounds of the present disclosure may include one or more asymmetric centers, and thus may exist in a variety of stereoisomeric forms, for example, enantiomers and/or diastereomers. For example, the compounds of the present disclosure may be in the form of an individual enantiomer, diastereomer or geometric isomer (e.g., cis- and trans-isomers), or may be in the form of a mixture of stereoisomers, including racemic mixture and a mixture enriched in one or more stereoisomers. The isomers can be separated from the mixture by the methods known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts; or alternative isomers can be prepared by asymmetric synthesis.


The compounds of the present disclosure may exist in tautomer forms. The tautomer is a functional group isomer resulting from the rapid shift of an atom between two positions in a molecule. The tautomer is a special functional group isomer, wherein a pair of tautomers can convert between each other, but usually exist in a relatively stable isomer as its main form. The most important examples are the enol and keto tautomers.


The present disclosure also comprises compounds that are labeled with isotopes (isotope variants), which are equivalent to those described in formula (IV), but one or more atoms are replaced with atoms having an atom mass or mass number that are different from that of atoms that are common in nature. Examples of isotopes which may be introduced into the compounds of the disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as 2H, 3H, 13C, 11C, 14C, 15N, 13O, 17O, 31P, 32P, 35S, 18F and 36Cl, respectively. Compounds of the present disclosure that comprise the above isotopes and/or other isotopes of other atoms, prodrugs thereof and pharmaceutically acceptable salts of said compounds or prodrugs all are within the scope of the present disclosure. Certain isotope-labeled compounds of the present disclosure, such as those incorporating radioactive isotopes (e.g., 3H and 14C), can be used for the measurement of the distribution of drug and/or substrate in tissue. Tritium, which is 3H and carbon-14, which is 14C isotope, are yet alternative, because they are easy to prepare and detect. Furthermore, replaced with heavier isotopes, such as deuterium, which is 2H, may provide therapeutic benefits due to the higher metabolic stability, such as prolonging the half-life in vivo or decreasing the dosage requirements, and thus may be alternative in some cases. Isotope-labeled compounds of formula (I) of the present disclosure and prodrugs thereof can be prepared generally by using readily available isotope-labeled reagents to replace non-isotope-labeled reagents in the following schemes and/or the procedures disclosed in the examples and preparation examples.


The present disclosure also provides a pharmaceutical formulation comprising a therapeutically effective amount of a compound of formula (I), or therapeutically acceptable salts thereof, and pharmaceutically acceptable carriers, diluents or excipients thereof. All of these forms belong to the present disclosure.


Pharmaceutical Compositions and Kits

In another aspect, the present disclosure provides a pharmaceutical composition comprising nanoparticle compositions of the present disclosure and pharmaceutically acceptable excipient(s), the nanoparticle composition comprises the compounds of the present disclosure.


A pharmaceutically acceptable excipient for use in the present disclosure refers to a non-toxic carrier, adjuvant or vehicle which does not destroy the pharmacological activity of the compound formulated together. Pharmaceutically acceptable carriers, adjuvants, or vehicles that may be used in the compositions of the present disclosure include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (e.g., human serum albumin), buffer substances (such as phosphate), glycine, sorbic acid, potassium sorbate, a mixture of partial glycerides of saturated plant fatty acids, water, salt or electrolyte (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based materials, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylate, wax, polyethylene-polyoxypropylene block polymers, polyethylene glycol and lanolin.


The present disclosure also includes kits (e.g., pharmaceutical packs). Kits provided may include a nanoparticle composition of the present disclosure and other therapeutic, or diagnostic, or prophylactic agents, and a first and a second containers (e.g., vials, ampoules, bottles, syringes, and/or dispersible packages or other materials) containing the nanoparticle composition of the present disclosure or other therapeutic, or diagnostic, or prophylactic agents. In some embodiments, kits provided can also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending the nanoparticle composition of the present disclosure and/or other therapeutic, or diagnostic, or prophylactic agent. In some embodiments, the nanoparticle composition of the present disclosure provided in the first container and the other therapeutic, or diagnostic, or prophylactic agents provided in the second container is combined to form a unit dosage form.


Administration

The pharmaceutical composition provided by the present disclosure can be administered by a variety of routes including, but not limited to, oral administration, parenteral administration, inhalation administration, topical administration, rectal administration, nasal administration, oral administration, vaginal administration, administration by implant or other means of administration. For example, parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intra-articular administration, intraarterial administration, intrasynovial administration, intrasternal administration, intracerebroventricular administration, intralesional administration, and intracranial injection or infusion techniques.


Generally, the pharmaceutical compositions provided herein are administered in an effective amount. The amount of the pharmaceutical composition actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated or prevented, the chosen route of administration, the actual pharmaceutical composition administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.


When used to prevent the disorder of the present disclosure, the pharmaceutical compositions provided herein will be administered to a subject at risk for developing the condition, typically on the advice and under the supervision of a physician, at the dosage levels described above. Subjects at risk for developing a particular condition generally include those that have a family history of the condition, or those who have been identified by genetic testing or screening to be particularly susceptible to developing the condition.


The pharmaceutical compositions provided herein can also be administered chronically (“chronic administration”). Chronic administration refers to administration of a compound or pharmaceutical composition thereof over an extended period of time, e.g., for example, over 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may be continued indefinitely, for example, for the rest of the subject's life. In certain embodiments, the chronic administration is intended to provide a constant level of the compound in the blood, e.g., within the therapeutic window over the extended period of time.


The pharmaceutical compositions of the present disclosure may be further delivered using a variety of dosing methods. For example, in certain embodiments, the pharmaceutical composition may be given as a bolus, e.g., in order to raise the concentration of the compound in the blood to an effective level. The placement of the bolus dose depends on the systemic levels of the active ingredient desired throughout the body, e.g., an intramuscular or subcutaneous bolus dose allows a slow release of the active ingredient, while a bolus delivered directly to the veins (e.g., through an IV drip) allows a much faster delivery which quickly raises the concentration of the active ingredient in the blood to an effective level. In other embodiments, the pharmaceutical composition may be administered as a continuous infusion, e.g., by IV drip, to provide maintenance of a steady-state concentration of the active ingredient in the subject's body. Furthermore, in still yet other embodiments, the pharmaceutical composition may be administered as first as a bolus dose, followed by continuous infusion.


Transdermal doses are generally selected to provide similar or lower blood levels than are achieved using injection doses, generally in an amount ranging from about 0.01 to about 20% by weight, alternatively from about 0.1 to about 20% by weight, alternatively from about 0.1 to about 10% by weight, and still alternatively from about 0.5 to about 15% by weight.


Injection dose levels range from about 0.1 mg/kg/hour to at least 10 mg/kg/hour, all for from about 1 to about 120 hours and especially 24 to 96 hours. A preloading bolus of from about 0.1 mg/kg to about 10 mg/kg or more may also be administered to achieve adequate steady state levels. The maximum total dose is not expected to exceed about 2 g/day for a 40 to 80 kg human patient.


Injectable compositions are typically based upon injectable sterile saline or phosphate-buffered saline or other injectable excipients known in the art. As before, the active compound in such compositions is typically a minor component, often being from about 0.05 to 10% by weight with the remainder being the injectable excipient and the like.


EXAMPLE

In order to make the technical solutions of the present disclosure clearer and more explicit, the present disclosure is further elaborated through the following examples. The following examples are used only to illustrate specific embodiments of the present disclosure so that a person skilled in the art can understand the present disclosure, but are not intended to limit the scope of protection of the present disclosure. The technical means or methods, etc. not specifically described in the specific embodiments of the present disclosure are conventional technical means or methods, etc. in the art. The materials, reagents, etc. used in examples are commercially available if not otherwise specified.












TABLE 1







Chemical




formula or



abbreviation
Full Name









THF
Tetrahydrofuran



ACN
Acetonitrile



DCM
Dichloromethane



MeOH
Methanol



DMF
N,N-dimethylformamide



DMSO
Dimethyl sulfoxide



MTBE
tert-Butyl methyl ether



CDCl3
Deuterated chloroform



TBAF
Tetrabutylammonium fluoride



TBAI
Tetrabutylammonium iodide



LDA
Lithium diisopropylamide



DMAP
4-Dimethylaminopyridine



(COCl)2
Oxalyl chloride



NaI
Sodium iodide



NaH
Sodium hydride



K2CO3
Potassium carbonate



EDCI
1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide



DIEA
N,N-diisopropylethylamine



LiAlH4
Lithium aluminum hydride



CMPE
Cyclopentyl methyl ether



DMPU
N,N-dimethylpropyleneurea



TMSCl
Trimethylchlorosilane



TMSOK
Potassium trimethylsilanolate



TEA
Triethylamine










Example 1: Synthesis of Compound 1a



embedded image


To a 500 mL three-neck round-bottom flask were added 1-nonanol (15 g, 104.0 mmol, 1.0 eq.), 8-bromooctanoic acid (25.5 g, 114.0 mmol, 1.1 eq.), DMAP (2.54 g, 20.8 mmol, 0.2 eq.), DIEA (40.3 g, 312.0 mmol, 3.0 eq.), and EDCI (25.9 g, 135 mmol, 1.3 eq.) at room temperature, and 250 mL of DCM was added. The reaction liquid was stirred at room temperature for 4 h, and the reaction was completed as monitored by TLC. The reaction liquid was poured into 200 mL of a saturated aqueous ammonium chloride solution, and the mixture was extracted with 3×100 mL of DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 21 g of a yellow oily compound 1a-2.


To a 250 mL three-neck round-bottom flask were added compound (1-2 (21 g, 60.1 mmol, 1.0 eq.) and 2-aminoethanol (110 g, 1.80 mom, 30.0 eq.) at room temperature, and the mixture was dissolved in 100 mL of methanol. The mixture was heated to 60° C., stirred for reaction for 18 h, and concentrated to remove the reaction solvent. Then a saturated aqueous ammonium chloride solution and ethyl acetate were added thereto, and the mixture was extracted after layer separation. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 14 g of a yellow oily compound 1a-3.


To a 250 mL three-neck round-bottom flask were added 9-heptadecanol (9.8 g, 38.2 mmol, 1.0 eq.) and triethylamine (15.5 g, 152.8 mmol, 4.0 eq.) at room temperature, and the mixture was dissolved in 100 mL of DCM. The reaction system was cooled in an ice bath, and then 2-methylpropionyl chloride (9.8 g, 91.7 mmol, 2.4 eq.) was slowly added. The mixture was warmed to room temperature and reacted overnight. After the reaction was completed as monitored by TLC, a saturated aqueous ammonium chloride solution was added to quench the reaction, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 8.7 g of a yellow oily compound 1a-5.


To a 250 mL three-neck round-bottom flask was added compound 1a-5 (8.7 g, 26.6 mmol, 1.0 eq.) at room temperature, and the mixture was dissolved in 60 mL of THF. The reaction system was cooled to −40° C., and then LDA (13.1 mL, 26.2 mmol, 0.98 eq.) was slowly added dropwise. The mixture was stirred for reaction for 1 h, and then 1,6-dibromohexane (9.03 g, 37.0 mmol, 1.39 eq.) and DMPU (0.48 g, 3.73 mmol, 0.14 eq.) were added to the reaction system. The mixture was warmed to room temperature and reacted overnight. After the reaction was completed as monitored by TLC, a saturated ammonium chloride ice water solution was added to quench the reaction, and the mixture was extracted with ethyl acetate. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 10.1 g of a yellow oily compound 1a-6.


To a 20 mL round-bottom flask were added compound 1a-6 (1.48 g, 3.04 mmol, 2.0 eq.), compound 1a-3 (500 mg, 1.52 mmol, 1.0 eq.), K2CO3 (628.2 mg, 4.55 mmol, 3.0 eq.), KI (302.3 mg, 1.82 mmol, 1.2 eq.), cyclopentyl methyl ether (7.5 ml), and acetonitrile (2.5 mL). The mixture was warmed to 80° C. and reacted. After the reaction was completed as monitored by TLC, a saturated ammonium chloride ice water solution was added to quench the reaction, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 96.98 mg of an oily compound 1a.



1H NMR (400 MHz, CDCl3) δ: 4.86-4.80 (m, 1H), 4.04 (t, J=7.2 Hz, 2H), 3.57 (m, 2H), 2.62 (t, J=7.2 Hz, 2H), 2.49 (t, J=7.2 Hz, 4H), 2.29 (t, J=7.2 Hz, 2H), 1.62 (m, 4H), 1.51-1.45 (m, 10H), 1.38-1.23 (m, 49H), 1.15 (s, 6H), 0.88 (t, J=7.2 Hz, 9H); ESI-MS m/z: 738.60 [M+H]+.


Example 2: Synthesis of Compound 2a



embedded image


To a 250 mL three-neck round-bottom flask were added 1-nonanol (8.8 g, 61.0 mmol, 1.0 eq.) and triethylamine (24.7 g, 244.0 mmol, 4.0 eq.) at room temperature, and the mixture was dissolved in 100 mL of DCM. The reaction system was cooled in an ice bath, and then 2-methylpropionyl chloride (15.6 g, 146.4 mmol, 2.4 eq.) was slowly added. The mixture was warmed to room temperature and reacted. After the reaction was completed as monitored by TLC, a saturated aqueous ammonium chloride solution was added to quench the reaction, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 10.8 g of a yellow oily compound 2a-2.


To a 250 mL three-neck round-bottom flask was added compound 2a-2 (4.8 g, 22.4 mmol, 1.0 eq.) at room temperature, and the mixture was dissolved in 50 mL of THF. The reaction system was cooled to −40° C., and then LDA (11.0 mL, 21.9 mmol, 0.98 eq.) was slowly added dropwise. The mixture was stirred for reaction for 1 h, and then 1,6-dibromohexane (7.59 g, 31.1 mmol, 1.39 eq.) and DMPU (0.57 g, 4.48 mmol, 0.2 eq.) were added to the reaction system. The mixture was warmed to room temperature and reacted overnight. After the reaction was completed as monitored by TLC, a saturated ammonium chloride ice water solution was added to quench the reaction, and the mixture was extracted with ethyl acetate. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 4.6 g of a yellow oily compound 2a-3.


To a 250 ml three-neck round-bottom flask were added compound 2a-3 (4.6 g, 12.2 mmol, 1.0 eq.) and 2-aminoethanol (14.9 g, 244.0 mmol, 20.0 eq.) at room temperature, and the mixture was dissolved in 50 mL of ethanol. The mixture was heated to 60° C., stirred for reaction for 18 h, and concentrated to remove the reaction solvent. Then a saturated aqueous ammonium chloride solution and ethyl acetate were added thereto, and the mixture was extracted after layer separation. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 2.2 g of a yellow oily compound 2a-4.


To a 20 mL round-bottom flask were added compound 2a-4 (500 mg, 1.4 mmol, 1.0 eq.), compound 1a-6 (1.37 g, 2.8 mmol, 2.0 eq.), K2CO3 (579 mg, 4.2 mmol, 3.0 eq.), KI (279 mg, 1.68 mmol, 1.2 eq.), cyclopentyl methyl ether (7.5 mL), and acetonitrile (2.5 mL). The mixture was warmed to 80° C. and reacted. After the reaction was completed as monitored by TLC, a saturated ammonium chloride ice water solution was added to quench the reaction, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 137.9 mg of an oily compound 2a.



1H NMR (400 MHz, CDCl3) δ: 4.83 (m, 1H), 4.04 (t, J=7.2 Hz, 2H), 3.55 (t, J=5.6 Hz, 2H), 2.60 (t, J=5.6 Hz, 2H), 2.47 (t, J=7.2 Hz, 4H), 1.62 (m, 2H), 1.52-1.40 (m, 12H), 1.36-1.20 (m, 48H), 1.15 (m, 12H), 0.88 (t, J=7.2 Hz, 9H); ESI-MS m/z: 766.50 [M+H]+.


Example 3: Synthesis of Compound 3a



embedded image


Compound 3a was prepared referring to the method of Example 1 to give 114.7 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.83 (m, 1H), 4.06 (t, J=7.2 Hz, 2H), 3.59 (m, 2H), 2.66 (m, 2H), 2.54 (m, 4H), 2.30 (t, J=7.2 Hz, 2H), 1.70-1.56 (m, 4H), 1.50 (m, 10H), 1.32-1.26 (m, 49H), 1.15 (s, 6H), 0.88 (t, J=7.2 Hz, 9H); ESI-MS m/z: 738.55 [M+H]+.


Example 4: Synthesis of Compound 4a



embedded image


Compound 4a was prepared referring to the method of Example 1 to give 112.1 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.83 (m, 1H), 4.05 (t, J=7.2 Hz, 2H), 3.80 (t, J=5.4 Hz, 2H), 2.51-2.73 (m, 6H), 2.29 (t, J=7.2 Hz, 2H), 1.65-1.44 (m, 16H), 1.26 (m, 49H), 1.15 (s, 6H), 0.93-0.82 (m, 9H); ESI-MS m/z: 752.70 [M+H]+.


Example 5: Synthesis of Compound 5a



embedded image


Compound 5a was prepared referring to the method of Example 1 to give 150.8 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.84 (m, 1H), 4.05 (t, J=7.2 Hz, 2H), 3.60 (m, 2H), 2.41-2.63 (m, 4H), 2.29 (t, J=7.2 Hz, 2H), 1.65-1.44 (m, 18H), 1.26-1.34 (m, 51H), 1.14 (s, 6H), 0.93-0.82 (m, 9H); ESI-MS m/z: 766.70 [M+H]+.


Example 6: Synthesis of Compound 6a



embedded image


tert-Butyl cyclopropanecarboxylate (15.0 g, 150.0 mmol, 1.0 eq.) was dissolved in 150 mL of anhydrous tetrahydrofuran, and the mixture was cooled to −60° C. LDA (150.0 mL, 300.0 mmol, 2.0 eq.) was added to the reaction liquid under nitrogen atmosphere. After the mixture was stirred at the same temperature for reaction for 1 h, 1-bromo-6-chlorohexane (44.9 g, 225.0 mmol, 1.5 eq.) was added, and the mixture was stirred at room temperature for reaction for another 3 h. After the reaction was completed as monitored by TLC, the reaction was quenched with a saturated aqueous ammonium chloride solution, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 19 g of a colorless oily compound 6a-1.


To a solution of compound 6a-1 (10.0 g, 38.3 mmol) in DCM (30.0 mL) was added 10.0 mL of trifluoroacetic acid, and the mixture was stirred at room temperature for 3 h. The reaction solvent was removed by pressurized rotary evaporation to give a crude product, which was purified by silica gel column chromatography to give 6.9 g of a colorless oily compound 6a-2.


Compound 6a-2 (500.0 mg, 2.45 mmol, 2.0 eq.) was dissolved in DCM (5.0 ml), and 2-3 drops of DMF and oxalyl chloride (311.1 mg, 2.45 mmol, 2.0 eq.) were added to the reaction system under an ice bath. After the mixture was stirred for 30 min, the solvent was removed by rotary evaporation under reduced pressure to give an acyl chloride intermediate. The resulting acyl chloride was dissolved in DCM (5.0 mL), then 9-heptadecanol (312.3 mg, 1.22 mmol, 1.0 eq.) and triethylamine (247.5 mg, 2.45 mmol, 2.0 eq.) were added, and the mixture was reacted at room temperature for 5 h. After the reaction was completed, the reaction liquid was poured into 50 mL of water, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 240 mg of an oily compound 6a-3.


To a solution of compound 6a-3 (200.0 mg, 0.45 mmol, 1.5 eq.) in DMF (3.0 mL) were added potassium carbonate (124.2 mg, 0.90 mmol, 3.0 eq.), sodium iodide (112.5 mg, 0.75 mmol, 2.5 eq.), and compound 1a-3 (99.0 mg, 0.30 mmol, 1.0 eq.). The reaction liquid was heated to 80° C. and stirred for reaction for 5 h. After the reaction was completed, the reaction system was cooled to room temperature. The reaction was quenched with a saturated aqueous sodium chloride solution, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by preparative liquid chromatography to give 75.2 mg of compound 6a. (Column model: XSelect C18 (19×150 mm, 5 μm), mobile phase A: water:acetonitrile=90:10 (10 mmol/L ammonium bicarbonate+0.5% aqueous ammonia), mobile phase B: isopropanol:acetonitrile=90:10; flow rate: 25 mL/min; gradient: the proportion of B was 75%-80% in 0-12 min, and the proportion of B was 90% after 12 min).



1H NMR (400 MHz, CDCl3) δ: 4.82 (m, 1H), 4.05 (t, J=7.2 Hz, 2H), 2.57 (m, 6H), 2.29 (t, J=7.2 Hz, 2H), 1.61 (m, 7H), 1.52 (m, 13H); 1.21-1.37 (m, 45H), 1.16 (q, J=4.4 Hz, 2H), 0.88 (t, J=7.2 Hz, 9H), 0.64 (q, J=4.4 Hz, 2H); ESI-MS m/z: 736.55 [M+H]+.


Example 7: Synthesis of Compound 7a



embedded image


Compound 7a was prepared referring to the method of Example 1 to give 102.4 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.08 (td, J=7.2 Hz, 4H), 3.58 (m, 2H), 2.57 (d, J=40.6 Hz, 6H), 2.31 (t, J=7.2 Hz, 2H), 1.73-1.40 (m, 14H), 1.20-1.39 (m, 45H), 1.17 (s, 6H), 0.96-0.85 (m, 9H); ESI-MS m/z: 710.55 [M+H]+.


Example 8: Synthesis of Compound 8a



embedded image


Compound 8a was prepared referring to the method of Example 1 to give 44.9 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.07-3.91 (m, 4H), 3.56 (m, 2H), 2.31-2.62 (m, 6H), 2.29 (t, J=7.2 Hz, 2H), 1.73-1.40 (m, 12H), 1.39-1.20 (m, 44H), 1.16 (s, 6H), 0.93-0.82 (m, 9H); ESI-MS m/z: 696.50 [M+H]+.


Example 9: Synthesis of Compound 9a



embedded image


In a 250 mL round-bottom flask, 5-bromo-1-pentanol (30 g, 180.0 mmol, 1.0 eq.) and imidazole (30.6 g, 449.0 mmol, 2.5 eq.) were dissolved in 120 mL of DMF. The reaction system was placed in an ice bath, and TBSCI (29.8 g, 198 mmol, 1.1 eq.) was added to the reaction system. The mixture was warmed to room temperature and reacted overnight. After the reaction was completed as monitored by TLC, a saturated aqueous sodium bicarbonate solution was added to quench the reaction, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 26 g of a light yellow oily compound 9a-1.


In a 250 mL three-neck round-bottom flask, Mg (1.70 g, 70.1 mmol, 1.6 eq.), b (0.01 g, 0.039 mmol), and 50 mL of anhydrous THF were added. The reaction system was warmed to 37° C., then compound 9a-1 (18.5 g, 65.7 mmol, 1.5 eq.) was slowly added within 30 min, and the mixture was reacted until magnesium powder almost disappeared completely to give a Grignard reagent. In a 250 mL three-neck round-bottom flask, CuBr (0.63 g, 4.38 mmol, 0.1 eq.), LiCl (0.37 g, 8.76 mmol, 0.2 eq.), and 50 mL of anhydrous THF were added under nitrogen atmosphere. After the mixture was stirred at 0° C. for 30 min, to the obtained copper-lithium reagent solution were added methyl 3,3-dimethacrylate (5 g, 43.8 mmol, 1.0 eq.) and TMSCl (7.14 g, 65.7 mmol, 1.5 eq.) dropwise. After the dropwise addition was completed, the mixture was stirred at 0° C. for another 30 min. The prepared Grignard reagent was slowly added dropwise to the reaction liquid, and after the dropwise addition was completed, the mixture was reacted for another 2 h. After the reaction was completed as monitored by TLC, the reaction was quenched with a saturated aqueous ammonium chloride solution, and the mixture was extracted with ethyl acetate. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 13 g of a light yellow oily compound 9a-2.


To a solution of compound 9a-2 (13 g, 41.1 mmol, 1.0 eq.) in THF was added TBAF (21.5 g, 82.1 mmol, 2.0 eq.) in batches under an ice bath, and the mixture was stirred at room temperature for 2 h. A saturated aqueous ammonium chloride solution was added to quench the reaction, and the mixture was extracted with ethyl acetate. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product of 9a-3, which was directly used in the next step without purification.


Compound 9a-3 (14 g, 69.2 mmol, 1.0 eq.) and PPh3 (21.8 g, 83.0 mmol, 1.2 eq.) were dissolved in 140 mL of dichloromethane, the reaction system was cooled to 0° C., and then CBr4 (25.3 g, 76.1 mmol, 1.1 eq.) was added in batches. The mixture was stirred at room temperature for reaction until the reaction was completed as monitored by TLC. The reaction was quenched with an ice saturated aqueous sodium chloride solution, and the mixture was extracted with ethyl acetate. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 7.6 g of a light yellow oily compound 9a-4.


To 60 mL of a solution of compound 9a-4 (6 g, 22.6 mmol, 1 eq.) in THF was added TMSOK (4.35 g, 33.9 mmol, 1.5 eq.). The mixture was stirred at room temperature for reaction until the reaction was completed as monitored by TLC, the pH of the reaction system was adjusted to 4.0 with 2.0 M hydrochloric acid, and the mixture was extracted with ethyl acetate. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 2.5 g of a light yellow oily compound 9a-5.


Compound 9a was prepared referring to the method of Example 1 to give 81.7 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.85 (m, 1H), 4.05 (t, J=7.2 Hz, 2H), 3.66 (s, 2H), 2.59 (m, 4H), 2.31 (t, J=7.2 Hz, 2H), 2.19 (s, 2H), 1.64-1.43 (m, 12H), 1.22-1.38 (m, 51H), 1.12 (s, 6H), 0.95-0.85 (m, 9H); ESI-MS m/z: 738.65 [M+H]+.


Example 10: Synthesis of Compound 10a



text missing or illegible when filed


In a 100 mL round-bottom flask, methyl 4-bromobutyrate (4 g, 22.1 mmol, 1.0 eq.) was dissolved in 40 mL of MTBE, the reaction system was placed in an ice bath, and methylmagnesium bromide (22.1 mL, 66.3 mmol, 3.0 eq.) was added to the reaction system. The mixture was warmed to room temperature and reacted until the reaction was completed as monitored by TLC, a saturated aqueous ammonium chloride solution was added to quench the reaction, and the mixture was extracted with EtOAc. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 3.5 g of compound 10a-2.


To a 100 mL round-bottom flask were added compound 10a-2 (3 g, 16.6 mmol, 1.0 eq.) triethylchlorosilane (7.49 g, 49.7 mmol, 3.0 eq.), zinc powder (3.25 g, 49.7 mmol, 3.0 eq.), Cp*TiCl3 (519 mg, 1.80 mmol, 0.1 eq.), and 5 A molecular sieves (2.4 g) under nitrogen atmosphere, and then 24 mL of anhydrous THF was added. The reaction system was heated to 50° C., and then ethyl acrylate (2.49 g, 24.9 mmol, 1.5 eq.) was added to the reaction system. The mixture was heated to 60° C. for reaction for another 12 h. The reaction liquid was cooled to room temperature, a saturated aqueous sodium chloride solution was added to quench the reaction, and the mixture was extracted with EtOAc. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 2.4 g of compound 10a-3.


In a 40 mL reaction flask, compound 10a-3 (1 g, 3.77 mmol, 1.0 eq.) was dissolved in a mixed solution of 10 mL ethanol and 2 ml water, and then lithium hydroxide (0.27 g, 11.3 mmol, 3.0 eq.) was added to the reaction system. The mixture was reacted at room temperature until the reaction was completed as monitored by TLC, the organic solvent was removed by rotary evaporation, and the pH of the solution was adjusted to about 4 with 2.0 M dilute hydrochloric acid. A saturated aqueous ammonium chloride solution was added to quench the reaction, and the mixture was extracted with EtOAc. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 800 mg of compound 10a-4.


Compound 10a was prepared referring to the method of Example 1 to give 93.4 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.92-4.72 (m, 1H), 4.05 (t, J=7.2 Hz, 2H), 3.59 (m, 2H), 2.64-2.51 (m, 6H), 2.33-2.18 (m, 4H), 1.55 (m, 14H), 1.34-1.24 (m, 45H), 0.83 (m, 15H); ESI-MS m/z: 724.55 [M+H]+.


Example 11: Synthesis of Compound 11a



text missing or illegible when filed


Compound 11a was prepared referring to the method of Example 1 to give 69.5 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.90-4.80 (m, 1H), 4.08 (t, J=7.2 Hz, 2H), 3.81 (t, J=5.2 Hz, 2H), 2.55 (m, 6H), 2.33 (t, J=7.2 Hz, 2H), 1.71-1.46 (m, 16H), 1.40-1.26 (m, 49H), 1.17 (s, 6H), 0.90 (t, J=7.2 Hz, 9H); ESI-MS m/z: 752.60 [M+H]+.


Example 12: Synthesis of Compound 12a



embedded image


Compound 12a was prepared referring to the method of Example 1 to give 178.6 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.88-4.77 (m, 1H), 4.05 (t, J=7.2 Hz, 2H), 3.60 (t, J=5.6 Hz, 2H), 2.57 (m, 6H), 2.30 (t, J=7.2 Hz, 2H), 1.71-1.46 (m, 12H), 1.43-1.15 (m, 55H), 1.11 (s, 6H), 0.89 (t, J=7.2 Hz, 9H); ESI-MS m/z: 766.70 [M+H]+.


Example 13: Synthesis of Compound 13a



embedded image


Compound 13a was prepared referring to the method of Example 1 to give 43.5 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.81 (p, J=6.4 Hz, 1H), 4.06 (t, J=6.8 Hz, 2H), 3.58 (m, 2H), 2.58-2.36 (m, 6H), 2.30 (t, J=7.6 Hz, 2H), 1.70-1.51 (m, 16H), 1.48-1.16 (m, 47H), 1.14 (t, J=4.0 Hz, 2H), 0.88 (t, J=7.2 Hz, 9H), 0.64 (t, J=4.0 Hz, 2H); ESI-MS m/z: 736.60 [M+H]+.


Example 14: Synthesis of Compound 14a



embedded image


Compound 14a was prepared referring to the method of Example 1 to give 56.3 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.08 (t, J=6.8 Hz, 2H), 4.01 (d, J=4.8 Hz, 2H), 3.97 (d, J=5.6 Hz, 2H), 3.17-3.06 (m, 5H), 2.36 (t, J=7.2 Hz, 2H), 1.87-1.63 (m, 16), 1.57-1.29 (m, 45H), 1.19 (s, 6H), 0.91 (t, J=7.2 Hz, 9H); ESI-MS m/z: 724.55 [M+H]+.


Example 15: Synthesis of Compound 15a



embedded image


Compound 15a was prepared referring to the method of Example 1 to give 120.3 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.09-4.03 (m, 4H), 3.56 (m, 2H), 2.62-2.49 (m, 6H), 2.30 (t, J=6.0 Hz, 2H), 1.66-1.55 (m, 14H), 1.52-1.26 (m, 45H), 1.15 (s, 6H), 0.88 (t, J=7.2 Hz, 9H); ESI-MS m/z: 710.55 [M+H]+.


Example 16: Synthesis of Compound 16a



embedded image


Compound 16a was prepared referring to the method of Example 1 to give 80.8 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.07-3.92 (m, 4H), 3.60 (m, 2H), 2.66-2.46 (m, 6H), 2.30 (t, J=7.6 Hz, 2H), 1.68-1.44 (m, 12H), 1.38-1.07 (m, 44H), 1.05 (s, 6H), 0.88 (t, J=7.2 Hz, 9H); ESI-MS m/z: 696.55 [M+H]+.


Example 17: Synthesis of Compound 17a



embedded image


Compound 17a was prepared referring to the method of Example 1 to give 125.7 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.86-4.80 (i, 1H), 4.05 (t, J=6.8 Hz, 2H), 3.55 (m, 2H), 2.60-2.47 (m, 6H), 2.29 (t, J=7.6 Hz, 2H), 1.66-1.48 (m, 14H), 1.43-1.26 (m, 49H), 1.15 (s, 6H), 0.88 (t, J=7.2 Hz, 9H); ESI-MS m/z: 738.65 [M+H]+.


Example 18: Synthesis of Compound 18a



text missing or illegible when filed


Compound 18a was prepared referring to the method of Example 1 to give 127.4 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.86-4.80 (m, 1H), 4.06 (t, J=6.8 Hz, 2H), 3.53 (t, J=4.8 Hz, 2H), 2.58 (t, J=4.8 Hz, 2H), 2.43 (t, J=4.8 Hz, 4H), 2.29 (t, J=7.6 Hz, 2H), 1.65-1.43 (m, 14H), 1.38-1.18 (m, 49H), 1.15 (s, 6H), 0.88 (t, J=7.2 Hz, 9H); ESI-MS m/z: 738.66 [M+H]+.


Example 19: Synthesis of Compound 19a



text missing or illegible when filed


Compound 19a was prepared referring to the method of Example 9 to give 79.7 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.89-4.82 (m, 1H), 4.06 (t, J=6.9 Hz, 2H), 3.67 (m, 2H), 2.70-2.34 (m, 4H), 2.31 (t, J=7.5 Hz, 2H), 2.17 (s, 2H), 1.70-1.49 (m, 12H), 1.38-1.18 (m, 49H), 0.99 (s, 6H), 0.88 (t, J=6.9 Hz, 9H); ESI-MS m/z: 738.65 [M+H]+.


Example 20: Synthesis of Compound 20a



text missing or illegible when filed


Compound 20a was prepared referring to the method of Example 10 to give 74.5 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.89-4.85 (m, 1H), 4.06 (t, J=7.2 Hz, 2H), 3.63 (m, 2H), 2.69-2.57 (m, 6H), 2.35-2.20 (m, 4H), 1.71-1.53 (m, 15H), 1.39-1.19 (m, 46H), 0.94-0.85 (m, 15H); ESI-MS m/z: 738.45 [M+H]+.


The compounds in Table 2 were synthesized using the methods of the above examples, or using similar methods of the corresponding intermediates.









TABLE 2









embedded image







Example 21: Compound 21a


[M + H]+: 752.65







embedded image







Example 22: Compound 22a


[M + H]+: 764.65







embedded image







Example 23: Compound 23a


[M + H]+: 696.65







embedded image







Example 24: Compound 24a


[M + H]+: 780.65







embedded image







Example 25: Compound 25a


[M + H]+: 738.60







embedded image







Example 26: Compound 26a


[M + H]+: 724.55







embedded image







Example 27: Compound 27a


[M + H]+: 710.50







embedded image







Example 28: Compound 28a


[M + H]+: 710.50







embedded image







Example 29: Compound 29a


[M + H]+: 808.70









Example 30: Synthesis of Compound 1b



text missing or illegible when filed


text missing or illegible when filed


9-Heptadecanol 1b-1 (200.0 mg, 0.78 mmol, 1.0 eq.) was dissolved in DCM (2.0 mL), and 8-bromooctanoic acid (226.2 mg, 1.01 mmol, 1.3 eq.), EDCl (179.4 mg, 0.94 mmol, 1.2 eq.), and DMAP (42.9 mg, 1.17 mmol, 1.5 eq.) were added to the solution. The reaction liquid was stirred at room temperature for 3 h, the reaction liquid was poured into 10 mL of a saturated aqueous sodium chloride solution, and the mixture was extracted with 3×20 mL of DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 300 mg of a yellow oily compound 1b-2.


Methyl isobutyrate (21 g, 205.0 mmol, 1.0 eq.) was dissolved in 200 mL of anhydrous THF, the mixture was cooled to 0° C., and LDA (205 mL, 410.0 mmol, 2.0 eq.) was added to the reaction liquid under nitrogen atmosphere. The reaction was heated to room temperature. After stirring for 30 min, 1,5-dibromopentane (47 g, 205.0 mmol, 1.0 eq.) was added. After the reaction was completed as monitored by TLC, a saturated aqueous ammonium chloride solution was added to quench the reaction, and the mixture was extracted with 3×300 mL of DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 30 g of a yellow oily compound 1b-4.


Compound 1b-4 (15.0 g, 40.0 mmol, 1.0 eq.) was dissolved in 30 mL of THF, the mixture was cooled to a ° C., and a borane-tetrahydrofuran solution (1 M, 100.0 mL) was added dropwise to the reaction system under nitrogen atmosphere. The mixture was heated to 75° C. and stirred for reaction for 3 h. After the reaction was completed, the reaction system was cooled to room temperature. The reaction was quenched with a saturated aqueous ammonium chloride solution, and the mixture was extracted with 3×300 mL of DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product of 1b-5 (13.6 g), which was directly used in the next step without purification.


Caprinoyl chloride (11.0 g, 58.0 mmol, 1.3 eq.) was added to a solution of compound 1b-5 (10.0 g, 44.0 mmol, 1.0 eq.) in DCM (100 ml), triethylamine (13.5 g, 134.0 mmol, 3.0 eq.) was added to the reaction system, and the mixture was reacted at room temperature for 3 h. The reaction liquid was poured into 100 mL of water, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 10 g of compound 1b-6.


Potassium carbonate (1.5 g, 11.1 mmol, 3.0 eq.) and compound 1b-6 (1.4 g, 3.7 mmol, 1.0 eq.) were added to a solution of ethanolamine (2.3 g, 37.2 mmol, 10.0 eq.) in acetonitrile (15.0 mL). The mixture was heated to 70° C. and stirred for reaction for 3 h. The reaction liquid was poured into 30 mL of water, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 1.4 g of compound 1b-7. 1H NMR (300 MHz, CDCl3) δ: 0.85-0.89 (m, 9H), 1.20-1.26 (m, 21H), 1.59-1.62 (m, 4H), 2.29-2.34 (m, 2H), 2.73-2.76 (m, 2H), 2.87-2.91 (m, 2H), 3.72-3.77 (m, 3H).


Compound 1b-2 (300.0 mg, 0.84 mmol, 1.0 eq.) and compound 1b-7 (503.4 mg, 1.09 mmol, 1.3 eq.) were dissolved in DMF (3.0 mL), then potassium carbonate (289.9 mg, 2.1 mmol, 2.5 eq.) and sodium iodide (314.4 mg, 2.1 mmol, 2.5 eq.) were added, and the mixture was heated to 70° C. and stirred for reaction for 2 h. The reaction liquid was poured into 20 ml of water, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give compound 1b (158.9 mg).



1H NMR (400 MHz, Methanol-d4) δ: 0.85-0.95 (m, 15H), 1.17.1.34 (m, 48H), 1.52-1.59 (m, 8H), 1.62.1.64 (m, 4H), 2.27-2.35 (m, 4H), 2.48-2.53 (m, 4H), 2.63 (t, J=6.3 Hz, 2H), 3.61 (t, J=6.3 Hz, 2H), 3.80 (s, 2H), 4.84.4.89 (m, 1H); ESI-MS m/z: 738.65 [M+H]°.


Example 31: Synthesis of Compound 2b



embedded image


Potassium carbonate (549.3 mg, 4.0 mmol, 3.0 eq.) and compound 1b-6 (500.0 mg, 1.33 mmol, 1.0 eq.) were added to a solution of 3-amino-1-propanol (1.0 g, 13.3 mmol, 10.0 eq.) in acetonitrile (10.0 mL). The mixture was heated to 70° C. and stirred for reaction for 3 h. The reaction liquid was poured into 30 mL of water, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 350 mg of a colorless oily compound 2b-1.


Compound 1b-2 (565 mg, 1.22 mmol, 1.3 eq.) and compound 2b-1 (350 mg, 0.94 mmol, 1.0 eq.) were dissolved in DMF (5.0 mL), then potassium carbonate (389 mg, 2.82 mmol, 3.0 eq.) and sodium iodide (353 mg, 2.35 mmol, 2.5 eq.) were added, and the mixture was heated to 70° C. and stirred for reaction for 5 h. The reaction liquid was poured into 20 mL of water, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give compound 2b (160 mg).



1H NMR (400 MHz, Methanol-d4) δ: 0.85-0.95 (m, 15H), 1.20-1.40 (m, 48H), 1.62-1.70 (m, 12H), 1.72-1.78 (m, 2H), 2.28-2.35 (m, 4H), 2.60-2.66 (m, 4H), 2.74-2.79 (m, 2H), 3.64 (t, J=6.0 Hz, 2H), 3.80 (s, 2H), 4.82-4.89 (m, 1H); ESI-MS m/z: 752.65 [M+H]+.


Example 32: Synthesis of Compound 3b



embedded image


Potassium carbonate (549 mg, 4.0 mmol, 3.0 eq.) and compound 1b-6 (500 mg, 1.33 mmol, 1.0 eq.) were added to a solution of 4-amino-1-butanol (1.2 g, 13.3 mmol, 10.0 eq.) in DMF (10.0 mL). The mixture was heated to 70° C. and stirred for reaction for 3 h. The reaction liquid was poured into 30 mL of water, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 300 mg of a colorless oily compound 3b-1.


Compound 1b-2 (462 mg, 1.0 mmol, 1.3 eq.) and compound 3b-1 (297 mg, 0.77 mmol, 1.0 eq.) were dissolved in DMF (5.0 mL), then potassium carbonate (269 mg, 1.9 mmol, 2.5 eq.) and sodium iodide (285 mg, 1.9 mmol, 2.5 eq.) were added, and the mixture was heated to 70° C. and stirred for reaction for 5 h. The reaction liquid was poured into 20 mL of water, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give compound 3b (228 mg).



1H NMR (400 MHz, CDCl3) δ: 0.80-0.90 (m, 15H), 1.25-1.40 (m, 48H), 1.49-1.66 (m, 8H), 1.74-1.81 (m, 8H), 2.24-2.37 (m, 4H), 2.47-2.58 (m, 6H), 3.56 (s, 2H), 3.77 (s, 2H), 4.81-4.88 (m, 1H); ESI-MS m/z: 766.85 [M+H]+.


Example 33: Synthesis of Compound 4b



text missing or illegible when filed


Methyl isobutyrate (21 g, 205 mmol, 1.0 eq.) was dissolved in 200 mL of anhydrous THF, the mixture was cooled to 0° C., and LDA (205 mL, 410 mmol, 2.0 eq.) was added to the reaction liquid under nitrogen atmosphere. The reaction was heated to room temperature. After stirring for 30 min, 1,6-dibromohexane (50 g, 205 mmol, 1.0 eq.) was added. After the reaction was completed as monitored by TLC, a saturated aqueous ammonium chloride solution was added to quench the reaction, and the mixture was extracted with 3×300 ml of DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 34 g of a yellow oily compound 4b-2. 1H NMR (400 MHz, CD3Cl) δ: 1.05-1.52 (m, 14H), 1.78-1.91 (m, 2H), 3.36-3.42 (m, 2H), 3.71 (s, 3H).


Compound 4b-2 (10.0 g, 37.6 mmol, 1.0 eq.) was added to a hydrobromic acid aqueous solution (50.0 ml). The reaction liquid was heated to 100° C. and stirred for reaction for 24 h. After the reaction was completed, the reaction system was cooled to room temperature. The reaction was quenched with a saturated aqueous sodium chloride solution, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 7.4 g of a yellow oily compound 4b-3. 1H NMR (400 MHz, CD3Cl) δ: 1.19 (s, 6H), 1.25-1.28 (m, 4H), 1.30-1.56 (m, 4H), 1.80-1.89 (m, 2H), 3.37.3.42 (m, 2H).


Compound 4b-3 (5.0 g, 19.9 mmol, 1.0 eq.) was dissolved in DCM (50.0 mL), and DMF (291.0 mg, 3.98 mmol, 0.2 eq.) and oxalyl chloride (5.1 g, 39.8 mmol, 2.0 eq.) were added to the reaction system under an ice bath. After the mixture was stirred for 30 min, the solvent was removed by rotary evaporation under reduced pressure to give an acyl chloride intermediate. The resulting acyl chloride was dissolved in DCM (50.0 ml), then 9-heptadecanol (4.08 g, 15.9 mmol, 0.8 eq.) and triethylamine (6.03 g, 59.7 mmol, 3.0 eq.) were added, and the mixture was reacted at room temperature overnight. After the reaction was completed, the reaction liquid was poured into 300 mL of water, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 4.3 g of an oily compound 4b-4.


To a solution of compound 4b-4 (492.9 mg, 1.0 mmol, 1.2 eq.) in DMF (3.0 ml) were added potassium carbonate (347.8 mg, 2.5 mmol, 3.0 eq.), sodium iodide (314.4 mg, 2.1 mmol, 2.5 eq.), and compound 1b-7 (300.0 mg, 0.8 mmol, 1.0 eq.). The reaction liquid was heated to 70° C. and stirred for reaction for 2 h. After the reaction was completed, the reaction system was cooled to room temperature. The reaction was quenched with a saturated aqueous sodium chloride solution, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by preparative liquid chromatography to give 106 mg of compound 4b. (Column model: XBridge Shield RP18 OBD Column 19×150 mm, 5 μm; mobile phase A: acetonitrile:water (10 mmol/L ammonium bicarbonate+0.05% aqueous ammonia), mobile phase B: isopropanol:acetonitrile; flow rate: 20 mL/min; gradient: the proportion of mobile phase B was gradually increased from 75% B to 95% B within 9 min).



1H NMR (400 MHz, Methanol-d4) δ: 0.85-0.95 (m, 15H), 1.15 (s, 6H), 1.20-1.40 (m, 48H), 1.47-1.54 (m, 10H), 1.60-1.64 (m, 2H), 2.31-2.34 (m, 2H), 2.47-2.51 (m, 4H), 2.61 (t, J=6.4 Hz, 2H), 3.61 (t, J=6.4 Hz, 2H), 3.80 (s, 2H), 4.81-4.88 (m, 1H); ESI-MS m/z: 766.70 [M+H]+.


Example 34: Synthesis of Compound 5b



embedded image


Compound 5b was prepared referring to the method of Example 30 to give 174 mg of an oily product.



1H NMR (400 MHz, Methanol-d4) δ: 0.85-0.95 (m, 15H), 1.20-1.40 (m, 43H), 1.50-1.60 (m, 4H), 1.61-1.70 (m, 7H), 2.28-2.33 (m, 4H), 2.48-2.64 (m, 5H), 3.51-3.63 (m, 2H), 3.80 (s, 2H), 4.04-4.08 (m, 2H); ESI-MS m/z: 696.60 [M+H]+.


Example 35: Synthesis of Compound 6b



embedded image


Compound 6b was prepared referring to the method of Example 30 to give 113 mg of an oily product.



1H NMR (400 MHz, Methanol-d4) δ: 0.85-0.95 (m, 9H), 1.25 (s, 6H), 1.35-1.55 (m, 48H), 1.58-1.65 (m, 14H), 2.27-2.32 (m, 2H), 2.46-2.51 (m, 4H), 2.58-2.62 (m, 2H), 3.58-3.64 (m, 2H), 4.04-4.08 (m, 2H), 4.83-4.87 (m, 1H); ESI-MS m/z: 738.65 [M+H]+.


Example 36: Synthesis of Compound 7b



embedded image


Compound 7b was prepared referring to the method of Example 30 to give 91 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 0.85-0.90 (m, 12H), 1.14 (s, 6H), 1.23-1.49 (m, 56H), 1.56-1.64 (m, 14H), 2.24-2.29 (m, 2H), 2.51-2.65 (m, 5H), 3.60 (s, 2H), 4.03-4.07 (m, 2H), 4.80-4.84 (m, 1H); ESI-MS m/z: 809.05 [M+H]+.


Example 37: Synthesis of Compound 8b



embedded image


Compound 8b was prepared referring to the method of Example 30 to give 124 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 0.85-0.95 (m, 6H), 1.27-1.49 (m, 42H), 1.52-1.63 (m, 12H), 2.26-2.31 (m, 2H), 2.52-2.57 (m, 4H), 2.65-2.68 (m, 2H), 3.47-3.64 (m, 2H), 4.01-4.07 (m, 4H); ESI-MS m/z: 626.65 [M+H]+.


Example 38: Synthesis of Compound 9b



embedded image


tert-Butyl cyclopropanecarboxylate (15.0 g, 150.0 mmol, 1.0 eq.) was dissolved in 150 mL of anhydrous tetrahydrofuran, and the mixture was cooled to −60° C. LDA (150.0 mL, 300.0 mmol, 2.0 eq.) was added to the reaction liquid under nitrogen atmosphere. After the mixture was stirred at the same temperature for reaction for 1 h, 1-bromo-6-chlorohexane (44.9 g, 225.0 mmol, 1.5 eq.) was added, and the mixture was stirred at room temperature for reaction for another 3 h. After the reaction was completed as monitored by TLC, the reaction was quenched with a saturated aqueous ammonium chloride solution, and the mixture was extracted with 3×300 mL of DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 19 g of a colorless oily compound 9b-1.


To a solution of compound 9b-1 (10.0 g, 38.3 mmol) in DCM (30.0 ml) was added 10.0 mL of trifluoroacetic acid, and the mixture was stirred at room temperature for 3 h. The reaction solvent was removed by pressurized rotary evaporation to give a crude product, which was purified by silica gel column chromatography to give 6.9 g of a colorless oily compound 9b-2.


Compound 9b-2 (500.0 mg, 2.45 mmol, 2.0 eq.) was dissolved in DCM (5.0 mL), and 2-3 drops of DMF and oxalyl chloride (311.1 mg, 2.45 mmol, 2.0 eq.) were added to the reaction system under an ice bath. After the mixture was stirred for 30 min, the solvent was removed by rotary evaporation under reduced pressure to give an acyl chloride intermediate. The resulting acyl chloride was dissolved in DCM (5.0 ml), then 9-heptadecanol (312.3 mg, 1.22 mmol, 1.0 eq.) and triethylamine (247.5 mg, 2.45 mmol, 2.0 eq.) were added, and the mixture was reacted at room temperature for 5 h. After the reaction was completed, the reaction liquid was poured into 50 mL of water, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 240 mg of an oily compound 9b-3.


To a solution of compound 9b-3 (200.0 mg, 0.45 mmol, 1.5 eq.) in DMF (3.0 ml) were added potassium carbonate (124.2 mg, 0.90 mmol, 3.0 eq.), sodium iodide (112.5 mg, 0.75 mmol, 2.5 eq.), and compound 6b-1 (99.0 mg, 0.30 mmol, 1.0 eq.). The reaction liquid was heated to 70° C. and stirred for reaction for 5 h. After the reaction was completed, the reaction system was cooled to room temperature. The reaction was quenched with a saturated aqueous sodium chloride solution, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by preparative liquid chromatography to give 85.4 mg of compound 9b. (Column model: XBridge Shield RP18 OBD Column 19×150 mm, 5 μm; mobile phase A: acetonitrile:water (10 mmol/L ammonium bicarbonate+0.05% aqueous ammonia), mobile phase B: isopropanol:acetonltrile; flow rate: 20 mL/min; gradient: 75% B to 95% B in 9 min).



1H NMR (400 MHz, CDCl3) δ: 0.60-0.70 (m, 2H), 0.85-0.95 (m, 9H), 1.13-1.31 (m, 48H), 1.46-1.48 (m, 12H), 1.57-1.63 (m, 4H), 2.26-2.31 (m, 2H), 2.42-2.47 (m, 4H), 2.56-2.59 (m, 2H), 3.50-3.54 (m, 2H), 4.03.4.07 (m, 2H), 4.80.4.84 (m, 1H); ESI-MS m/z: 736.65 [M+H]+.


Example 39: Synthesis of Compound 10b



embedded image


Compound 10b was prepared referring to the method of Example 30 to give 128 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 0.85-0.95 (m, 15H), 1.12-1.36 (m, 43H), 1.40-1.63 (m, 12H), 2.26-2.31 (m, 4H), 2.42-2.65 (m, 6H), 3.50-3.54 (m, 2H), 3.78 (s, 2H), 4.08 (t, J=7.2 Hz, 2H); ESI-MS m/z: 710.80 [M+H]+.


Example 40: Synthesis of Compound 11b



embedded image


Compound 11b was prepared referring to the method of Example 30 to give 92 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 0.83-0.94 (m, 15H), 1.18-1.42 (m, 43H), 1.48-1.67 (m, 10H), 2.28-2.35 (m, 4H), 2.46-2.52 (m, 4H), 2.61 (t, J=6.6 Hz, 2H), 3.61 (t, J=6.6 Hz, 2H), 3.80 (s, 2H), 4.05 (t, J=6.6 Hz, 2H); ESI-MS m/z: 696.60 [M+H]+.


Example 41: Synthesis of Compound 12b



embedded image


Compound 12b was prepared referring to the method of Example 30 to give 105 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 0.85-0.95 (m, 15H), 1.19-1.34 (m, 41H), 1.42-1.58 (m, 4H), 1.59-1.63 (m, 6H), 2.23 (q, J=7.6 Hz, 4H), 2.53 (q, J=7.6 Hz, 4H), 2.62 (q, J=7.6 Hz, 2H), 3.62 (t, J=6.4 Hz, 2H), 3.80 (s, 2H), 4.07 (t, J=6.4 Hz, 2H); ESI-MS m/z: 682.60 [M+H]+.


The compounds in Table 3 were synthesized using the methods of the above examples, or using similar methods of the corresponding intermediates.









TABLE 3









embedded image







Example 42: Compound 13b


[M + H]+: 724.80







embedded image







Example 43: Compound 14b


[M + H]+: 736.70







embedded image







Example 44: Compound 15b


[M + H]+: 764.70







embedded image







Example 45: Compound 16b


[M + H]+: 780.80







embedded image







Example 46: Compound 17b


[M + H]+: 808.80







embedded image







Example 47: Compound 18b


[M + H]+: 752.85







embedded image







Example 48: Compound 19b


[M + H]+: 738.65







embedded image







Example 49: Compound 20b


[M + H]+: 724.80







embedded image







Example 50: Compound 21b


[M + H]+: 778.75







embedded image







Example 51: Compound 22b


[M + H]+: 722.75







embedded image







Example 52: Compound 23b


[M + H]+: 708.70







embedded image







Example 53: Compound 24b


[M + H]+: 792.70









Example 54: Synthesis of Compound 30a



text missing or illegible when filed


In a 250 mL round-bottom flask, 9-heptadecanol (10.0 g, 39.0 mmol, 1.0 eq.) and pyridine (6.17 g, 78.0 mmol, 2.0 eq.) were dissolved in 100 mL of dichloromethane, and then the reaction system was cooled to 0° C. Isobutyryl chloride (10.39 g, 97.5 mmol, 2.5 eq.) was slowly added to the reaction liquid, and then the mixture was stirred at room temperature for 2 h. Water was added to quench the reaction at 0° C., and the mixture was extracted with DCM. The organic phases were combined, dried over anhydrous Na2SO4, and filtered to remove the drying agent, and the solvent was removed by rotary evaporation. The resulting crude product was purified by silica gel column chromatography to give a yellow oily compound 1a-5 (10.4 g).


Compound 1a-5 (10.0 g, 30.62 mmol, 1.0 eq.) was dissolved in anhydrous THF (100 ml), the reaction system was cooled to −40° C., and IDA (15.3 mL, 30.6 mmol, 1.0 eq.) was added to the reaction liquid under nitrogen atmosphere. After the mixture was stirred for reaction at -40° C. for 1 h, 1,6-dibromohexane (14.9 g, 61.2 mmol, 2.0 eq.) and DMPU (471 mg, 3.7 mmol, 0.12 eq.) were added at the same temperature. The reaction system was slowly warmed to room temperature and then reacted overnight. After the reaction was completed, the reaction liquid was added to a saturated NH4Cl solution, and the mixture was extracted with DCM. The organic phases were combined, dried over anhydrous Na2SO4, and filtered to remove the drying agent, and the solvent was removed by rotary evaporation to give a crude compound 1a-6, which was directly used in the next step without purification.


Compound 1a-6 (15 g, 30.6 mmol, 1.0 eq.) and ethanolamine (37.4 g, 612.0 mmol, 20.0 eq.) were dissolved in 80 mL of ethanol, and the reaction system was heated to 60° C. and reacted for 2 h. After the reaction was completed, the reaction system was cooled to room temperature, the ethanol solvent was removed by rotary evaporation, the crude product was dissolved in ethyl acetate, and then a saturated sodium chloride solution was added. After extraction, the organic phases were combined, dried over anhydrous Na2SO4, and filtered to remove the drying agent, and the solvent was removed by rotary evaporation. The resulting crude product was purified by silica gel column chromatography to give a yellow oily compound 30a-1 (12.4 g).


In a 50 mL reaction flask, 6-bromo-1-hexanol (1.5 g, 8.3 mmol, 1.0 eq.) and pyridine (1.31 g, 16.6 mmol, 2.0 eq.) were dissolved in 15 mL of DCM, n-nonyl chloroformate (1.88 g, 9.1 mmol, 1.1 eq.) was added dropwise within 15 min under an ice bath, and the mixture was left at room temperature overnight. A saturated aqueous ammonium chloride solution was added to quench the reaction, and the mixture was extracted with DCM. The organic phases were combined, dried over anhydrous Na2SO4, and filtered to remove the drying agent, and the solvent was removed by rotary evaporation. The resulting crude product was purified by silica gel column chromatography to give a yellow oily compound 30a-4 (2.5 g).


In an 8 mL sealed tube, compound 30a-4 (200 mg, 0.57 mmol, 1.0 eq.), compound 30a-1 (294.2 mg, 0.63 mmol, 1.1 eq.), KI (113.4 mg, 0.68 mmol, 1.2 eq.), K2CO3 (236.0 mg, 1.71 mmol, 3.0 eq.), and 5.0 mL of anhydrous acetonitrile were added. The mixture was heated to 80° C., stirred for reaction overnight, cooled to room temperature, and filtered. After the filter cake was washed with acetonitrile, the organic phases were combined, and the solvent was removed by rotary evaporation to give a reaction crude product. The crude product was purified by preparative liquid chromatography to give compound 30a (87.9 mg).



1H NMR (400 MHz, CDCl3) δ: 4.83 (m, 1H), 4.12 (t, J=6.6 Hz, 4H), 3.61 (m, 2H), 2.62 (m, 6H), 1.70-1.63 (m, 4H), 1.50 (m, 8H), 1.26 (m, 49H), 1.15 (s, 6H), 0.88 (t, J=6.6 Hz, 9H); ESI-MS m/z: 740.55 [M+H]+.


Example 55: Synthesis of Compound 31a



embedded image


Compound 31a was prepared referring to the method of Example 54 to give 80.7 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.83 (p, J=6.6 Hz, 1H), 4.12 (td, J=7.2, 2.4 Hz, 4H), 3.61 (m, 2H), 2.61 (m, 6H), 1.68 (m, 4H), 1.50 (m, 8H), 1.26 (m, 49H), 1.15 (s, 6H), 0.88 (t, J=6.6 Hz, 9H); ESI-MS m/z: 740.65 [M+H]4.


Example 56: Synthesis of Compound 32a



text missing or illegible when filed


Compound 32a was prepared referring to the method of Example 54 to give 93.9 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.85 (p, J=6.6 Hz, 1H), 4.12 (t, J=6.6 Hz, 4H), 3.79 (t, J=5.1 Hz, 2H), 2.65 (m, 2H), 2.42 (m, 4H), 1.77-1.64 (m, 6H), 1.51 (t, J=6.0 Hz, 10H), 1.28 (d, J=4.5 Hz, 47H), 1.17 (s, 6H), 0.88 (t, J=6.6 Hz, 9H); ESI-MS m/z: 754.60 [M+HJ].


Example 57: Synthesis of Compound 33a



embedded image


Compound 33a was prepared referring to the method of Example 54 to give 164.2 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.12 (t, J=6.8 Hz, 4H), 3.95 (d, J=5.6 Hz, 2H), 3.54 (t, J=5.2 Hz, 2H), 2.59 (t, J=5.2 Hz, 2H), 2.50-2.41 (m, 4H), 1.67 (m, 5H), 1.53-1.19 (m, 53H), 1.16 (s, 6H), 0.88 (t, J=6.8 Hz, 9H); ESI-MS m/z: 726.50 [M+H]+.


Example 58: Synthesis of Compound 34a



embedded image


Compound 34a was prepared referring to the method of Example 54 to give 150.1 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.12 (t, J=6.8 Hz, 4H), 4.08 (t, J=6.8 Hz, 2H), 3.56 (m, 2H), 2.61-2.48 (m, 6H), 1.66 (m, 5H), 1.61-1.26 (m, 51H), 1.15 (s, 6H), 0.88 (t, J=6.8 Hz, 9H); ESI-MS m/z: 712.60 [M+H]+.


Example 59: Synthesis of Compound 35a



embedded image


Compound 35a was prepared referring to the method of Example 54 to give 217.9 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.12 (t, J=6.8 Hz, 4H), 4.03 (t, J=6.8 Hz, 2H), 3.53 (t, J=5.2 Hz, 2H), 2.58 (t, J=5.2 Hz, 2H), 2.47-2.40 (m, 4H), 1.73-1.62 (m, 6H), 1.60-1.19 (m, 48H), 1.16 (s, 6H), 0.88 (t, J=6.8 Hz, 9H); ESI-MS m/z: 698.55 [M+H]+.


Example 60: Synthesis of Compound 36a



text missing or illegible when filed


Compound 36a was prepared referring to the method of Example 54 to give 169.7 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.83 (p, J=6.6 Hz, 1H), 4.16-4.10 (m, 4H), 3.53 (t, J=5.1 Hz, 2H), 2.58 (t, J=5.1 Hz, 2H), 2.52-2.42 (m, 4H), 1.67 (m, 4H), 1.59-1.18 (m, 57H), 1.15 (s, 6H), 0.89 (t, J=6.6 Hz, 9H); ESI-MS m/z: 740.60 [M+H]+.


Example 61: Synthesis of Compound 37a



text missing or illegible when filed


Compound 37a was prepared referring to the method of Example 54 to give 59.4 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.12 (q, J=6.8 Hz, 4H), 3.95 (d, J=5.2 Hz, 2H), 3.58 (m, 2H), 2.64-2.51 (m, 6H), 1.74-1.43 (m, 12H), 1.39-1.19 (m, 46H), 1.16 (s, 6H), 0.88 (t, J=6.8 Hz, 9H); ESI-MS m/z: 726.50 [M+H]4.


Example 62: Synthesis of Compound 38a



text missing or illegible when filed


Compound 38a was prepared referring to the method of Example 54 to give 44.8 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.16-4.04 (m, 6H), 3.62 (m, 2H), 2.69-2.58 (m, 6H), 1.74-1.41 (m, 12H), 1.39-1.18 (m, 44H), 1.15 (s, 6H), 0.92-0.87 (m, 9H); ESI-MS m/z: 712.55 [M+H]+.


Example 63: Synthesis of Compound 39a



embedded image


Compound 39a was prepared referring to the method of Example 54 to give 105.4 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.13-4.10 (m, 4H), 4.03 (t, J=6.8 Hz, 2H), 3.56 (m, 2H), 2.46 (m, 6H), 1.73-1.67 (m, 8H), 1.64-1.19 (m, 50H), 1.15 (s, 6H), 0.88 (m, 9H); ESI-MS m/z: 726.50 [M+H]+.


Example 64: Synthesis of Compound 40a



text missing or illegible when filed


In a 500 mL round-bottom flask, nonanoic acid (15.0 g, 94.8 mmol, 1.0 eq.) and THF (150 mL), were added at room temperature under nitrogen atmosphere. The system was cooled to 0° C., NaH (3.64 g, 151.7 mmol, 1.6 eq.) was sequentially added in batches, and LDA (85.3 mL, 170.6 mmol, 1.8 eq., 2 M in THF) was added dropwise. The mixture was stirred at room temperature for 1 h, and 1-iodoheptane (23.6 g, 104.3 mmol, 1.1 eq.) was added to the system at 0° C. The system was stirred at 80° C. overnight. The reaction was monitored by TLC, and a new spot of the product was found. The reaction was cooled to room temperature, ice water was added to quench the reaction, and the mixture was extracted 3 times with DCM. The organic phases were combined and washed once with a saturated NaCl solution, dried over Na2SO4, filtered, and concentrated. The crude product was purified by silica gel column chromatography to give yellow oily 2-heptyinonanoic acid (16.36 g).


In a 250 mL round-bottom flask, 2-heptylnonanoic acid (7.0 g, 27.3 mmol, 1.0 eq.) was dissolved in 70 mL of anhydrous THF at room temperature, the system was cooled to 0° C., and LiAlH4 (16.38 mL, 2.5 M in THF) was added dropwise within 30 min. After the dropwise addition was completed, the mixture was warmed to room temperature and stirred for 2 h. The reaction was monitored by TLC, and the conversion of the starting material was completed. The reaction liquid was poured into a saturated sodium chloride solution to quench the reaction, the mixture was extracted with ethyl acetate, and the organic phases were combined, dried over Na2SO4, filtered, and concentrated. The crude product was purified by silica gel column chromatography to give a light yellow oily product of 2-heptylnonanol (6.97 g).


In a 50 mL three-neck round-bottom flask, 2-heptylnonanol (400 mg, 1.65 mmol, 1.0 eq.), pyridine (261.0 mg, 3.30 mmol, 2.0 eq.), DMAP (40.3 mg, 0.33 mmol, 0.2 eq.), p-nitrophenyl chloroformate (399.0 mg, 1.98 mmol, 1.2 eq.), and DCM (4 mL) were added at room temperature. The reaction system was stirred at room temperature for 1 h. Thereafter, 6-bromo-1-hexanol (896.2 mg, 4.95 mmol, 3.0 eq.) and DIEA (639.7 mg, 4.95 mmol, 3.0 eq.) were added to the reaction system. The system was stirred at room temperature overnight. The mixture was diluted with DCM (4 ml) and washed once with a saturated sodium bicarbonate solution (6 ml). The organic phase was washed once with saturated NaCl (5 mL), dried over anhydrous Na2SO4, filtered, and concentrated. The crude product was purified by silica gel column chromatography to give a light yellow oily carbonate intermediate compound 40a-4 (325 mg).


In an 8 ml sealed tube, compound 40a-4 (250 mg, 0.56 mmol, 1.0 eq.) obtained in the previous step, compound 2a-4 (179.0 mg, 0.50 mmol, 0.9 eq.), potassium iodide (110.8 mg, 0.67 mmol, 1.2 eq.), potassium carbonate (230.6 mg, 1.68 mmol, 3.0 eq.), and acetonitrile (2.5 ml) were added at room temperature. The mixture was heated to 80° C. and stirred for reaction overnight. The reaction was monitored by LC-MS, and the reaction system was cooled to room temperature. The mixture was filtered, concentrated, and purified by Prep-HPLC to give a light yellow oily product of compound 40a (84.3 mg).



1H NMR (400 MHz, CDCl3) δ: 4.14 (t, J=6.8 Hz, 2H), 4.06 (dd, J=8.0, 6.0 Hz, 4H), 3.60 (m, 2H), 2.65-2.53 (m, 6H), 1.73-1.60 (m, 12H), 1.42-1.22 (m, 46H), 1.18 (s, 6H), 0.90 (t, J=6.8 Hz, 9H); ESI-MS m/z: 726.50 [M+H]+.


Example 65: Synthesis of Compound 41a



embedded image


Compound 41a was prepared referring to the method of Example 64 to give 113.3 mg of an oily product.



1H NMR (400 MHz, CDCl1) δ: 4.14 (td, J=9.2, 6.8 Hz, 4H), 4.06 (t, J=6.8 Hz, 2H), 3.57 (m, 2H), 2.62-2.49 (m, 6H), 1.71-1.63 (m, 6H), 1.57-1.22 (m, 50H), 1.17 (s, 6H), 0.90 (t, J=6.8 Hz, 9H); ESI-MS m/z: 712.55 [M+H]+.


Example 66: Synthesis of Compound 42a



embedded image


Compound 42a was prepared referring to the method of Example 64 to give 124.2 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.12 (td, J=6.6, 4.8 Hz, 4H), 4.04 (t, J=6.6 Hz, 2H), 3.55 (t, J=5.7 Hz, 2H), 2.60-2.47 (m, 6H), 1.72-1.62 (m, 6H), 1.59-1.19 (m, 48H), 1.15 (s, 6H), 0.93-0.83 (m, 9H); ESI-MS m/z: 698.55 [M+H]+.


Example 67: Synthesis of Compound 43a



embedded image


Compound 43a was prepared referring to the method of Example 64 to give 97.7 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.15 (td, J=6.8, 1.6 Hz, 4H), 4.06 (t, J=6.8 Hz, 2H), 3.64 (m, 2H), 2.68-2.56 (m, 6H), 1.76-1.60 (m, 8H), 1.54-1.22 (m, 44H), 1.18 (s, 6H), 0.96-0.86 (m, 9H); ESI-MS m/z: 684.45 [M+H]+.


Example 68: Synthesis of Compound 44a



text missing or illegible when filed


In a 20 mL round-bottom flask, compound 44a-1 (587.58 mg, 2.34 mmol, 1.2 eq.), 2-heptylnonanol (500 mg, 1.95 mmol, 1.0 eq.), EDCl (560.58 mg, 2.93 mmol, 1.5 eq.), and DMAP (47.63 mg, 0.39 mmol, 0.2 eq.) were dissolved in 5 ml. of dichloromethane, and the mixture was reacted at room temperature overnight. After the reaction was completed, a saturated aqueous sodium chloride solution was added to quench the reaction, the mixture was extracted with dichloromethane, and the organic phases were combined, dried over Na2SO4, filtered, and concentrated to give 1 g of crude 44a-2, which was directly used in the subsequent reaction.


Compound 44a was prepared referring to the method of Example 54 to give 165.3 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.12 (t, J=6.8 Hz, 4H), 3.96 (d, J=5.6 Hz, 2H), 3.59 (m, 2H), 2.53-2.42 (m, 6H), 2.26-2.20 (m, 2H), 1.70-1.63 (m, 7H), 1.56-1.19 (m, 49H), 0.92-0.83 (m, 15H); ESI-MS m/z: 726.50 [M+H]+.


Example 6: Synthesis of Compound 45a



text missing or illegible when filed


Compound 45a was prepared referring to the method of Example 68 to give 107.2 mg of an oily product.



1H NMR (300 M Hz, CDCl3) δ: 4.12 (t, J=6.6 Hz, 2H), 4.04 (t, J=6.6 Hz, 4H), 3.57 (m, 2H), 2.63-2.51 (i, 6H), 2.18 (s, 2H), 1.73-1.61 (m, 5H), 1.59-1.21 (m, 51H), 0.98 (s, 6H), 0.93-0.83 (in, 9H); ESI-MS mM/z: 726.55 [M+H]+.


Example 70: Synthesis of Compound 46a



text missing or illegible when filed


Compound 46a was prepared referring to the method of Example 68 to give 116.7 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.12 (t, J=6.6 Hz, 2H), 4.07-4.02 (m, 4H), 3.58 (m, 2H), 2.64-2.49 (m, 6H), 2.27-2.21 (m, 2H), 1.73-1.60 (m, 5H), 1.56-1.21 (m, 51H), 0.93-0.82 (m, 15H); ESI-MS m/z: 726.45 [M+H]+.


Example 71: Synthesis of Compound 47a



text missing or illegible when filed


Compound 47a was prepared referring to the method of Example 68 to give 24.2 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.12 (t, J=6.8 Hz, 2H), 4.07-4.02 (m, 4H), 3.73 (m, 2H), 2.81-2.63 (m, 6H), 2.31 (t, J=6.8 Hz, 2H), 1.76-1.59 (m, 10H), 1.58-1.22 (m, 46H), 0.92-0.87 (m, 15H); ESI-MS m/z: 726.55 [M+H]+.


Example 72: Synthesis of Compound 48a



text missing or illegible when filed


text missing or illegible when filed


Compound 48a was prepared referring to the method of Example 54 to give 258.1 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.85 (p, J=6.0 Hz, 1H), 4.12 (t, J=6.6 Hz, 4H), 3.59 (m, 2H), 2.65-2.52 (m, 6H), 1.67-1.62 (m, 5H), 1.49-1.20 (m, 60H), 1.15 (s, 6H), 0.88 (t, J=6.6 Hz, 9H); ESI-MS m/z: 782.72 [M+H]+.


Example 73: Synthesis of Compound 49a



text missing or illegible when filed


Compound 49a was prepared referring to the method of Example 54 to give 27.9 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.82 (p, J=6.0 Hz, 1H), 4.12 (t, J=6.6 Hz, 4H), 3.65 (m, 2H), 2.73-2.61 (m, 6H), 1.83-1.62 (m, 9H), 1.51-1.19 (m, 64H), 1.15 (s, 6H), 0.89 (t, J=6.6 Hz, 9H); ESI-MS m/z: 824.70 [M+H]+.


Example 74: Synthesis of Compound 50a



text missing or illegible when filed


Compound 50a was prepared referring to the method of Example 68 to give 75.9 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.12 (t, J=6.6 Hz, 2H), 4.03 (d, J=6.0 Hz, 2H), 3.96 (d, J=6.0 Hz, 2H), 3.61 (m, 2H), 2.67-2.55 (m, 6H), 2.32 (t, J=7.2 Hz, 2H), 1.72-1.54 (m, 10H), 1.52-1.20 (m, 57H), 0.89 (m, 18H); ESI-MS m/z: 824.65 [M+H]+.


Example 75: Synthesis of Compound 51a



embedded image


In a 250 mL reaction flask, methyl isobutyrate (3.5 g, 34.3 mmol, 1.0 eq.) and THF (40 mL) were added under nitrogen atmosphere, and the mixture was cooled to −40° C. LDA (17.2 mL, 34.3 mmol, 1.0 eq., 2 M in THF) was added dropwise within 10 min, and after the dropwise addition was completed, the mixture was reacted for another 1.5 h. Then 1,4-dibromobutane (14.80 g, 68.5 mmol, 2.0 eq.) and DMPU (0.88 g, 6.85 mmol, 0.2 eq.) were added dropwise within 5 min at the same temperature, and the mixture was slowly warmed to room temperature and reacted overnight. After the reaction was completed, a saturated aqueous ammonium chloride solution was added to quench the reaction, the mixture was extracted with ethyl acetate, the organic phases were combined, dried over Na2SO4, filtered, and concentrated, and the organic solvent was removed by rotary evaporation to give 7.5 g of a crude compound of methyl 2,2-dimethyl-6-bromohexanoate.


In a 250 mL round-bottom flask, methyl 2,2-dimethyl-6-bromohexanoate (7.5 g, crude) was dissolved in 75 ml of anhydrous THF. After the mixture was cooled to 0° C., LiAlH4 (10.12 mL, 25.3 mmol, 2.5 M in THF) was added dropwise. After the dropwise addition was completed, the mixture was reacted for another 30 min. After the reaction was completed, a saturated aqueous ammonium chloride solution was added to quench the reaction, the mixture was extracted with ethyl acetate, the organic phases were combined, dried over Na2SO4, filtered, and concentrated, and the organic solvent was removed by rotary evaporation to give 7.5 g of a crude product, which was purified by silica gel column chromatography to give 2.5 g of the compound 2,2-dimethyl-6-bromohexanol.


Compound 51a was prepared referring to the method of Example 64 to give 205.5 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.08-4.02 (m, 4H), 3.85 (s, 2H), 3.54 (t, J=5.1 Hz, 2H), 2.59 (t, J=5.1 Hz, 2H), 2.49-2.43 (m, 4H), 2.29 (t, J=7.5 Hz, 2H), 1.66-1.57 (m, 6H), 1.55-1.22 (m, 50H), 0.92-0.83 (m, 15H); ESI-MS m/z: 726.80 [M+H]+.


Example 76: Synthesis of Compound 25b



embedded image


Compound 25b was prepared referring to the method of Example 75 to give 152.1 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.08-4.02 (m, 4H), 3.85 (s, 2H), 3.58 (m, 2H), 2.64-2.51 (m, 6H), 2.29 (t, J=7.5 Hz, 2H), 1.79-1.59 (m, 6H), 1.47-1.24 (m, 50H), 0.95-0.85 (m, 15H); ESI-MS m/z: 726.50 [M+H]+.


Example 77: Synthesis of Compound 26b



text missing or illegible when filed


Methyl isobutyrate (21 g, 205.0 mmol, 1.0 eq.) was dissolved in 200 mL of anhydrous THF, the mixture was cooled to 0° C., and LDA (205 mL, 410.0 mmol, 2.0 eq.) was added to the reaction liquid under nitrogen atmosphere. The reaction was heated to room temperature. After stirring for 30 min, 1,5-dibromopentane (47 g, 205.0 mmol, 1.0 eq.) was added. After the reaction was completed as monitored by TLC, a saturated aqueous ammonium chloride solution was added to quench the reaction, and the mixture was extracted with DCM (3×300 ml). The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 30 g of a yellow oily compound methyl 2,2-dimethyl-7-bromoheptanoate.


Methyl 2-dimethyl-7-bromoheptanoate (15.0 g, 40.0 mmol, 1.0 eq.) was dissolved in 30 ml of THF, the mixture was cooled to 0° C., and a lithium aluminum hydride solution (2 M, 50.0 ml) was added dropwise to the reaction system under nitrogen atmosphere. The mixture was stirred for reaction for 3 h. After the reaction was completed, the reaction was quenched with a saturated aqueous ammonium chloride solution, and the mixture was extracted with DCM (3×300 ml). The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product of 2,2-dimethyl-7-bromoheptanol (13.6 g), which was directly used in the next step without purification.


2,2-Dimethyl-7-bromoheptanol (500.0 mg, 2.24 mmol, 1.0 eq.) was dissolved in DCM (10.0 ml), and 4-hexyldecanoic acid (746.2 mg, 2.91 mmol, 1.3 eq.), EDCl (515.7 mg, 2.69 mmol, 1.2 eq.), and DMAP (410.5 mg, 3.36 mmol, 1.5 eq.) were added to the solution. The reaction liquid was stirred at room temperature for 3 h, the reaction liquid was poured into 10 ml of a saturated aqueous sodium chloride solution, and the mixture was extracted with DCM. The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 858 mg of a yellow oily compound 26b-4.


Compound 26b was prepared referring to the method of Example 64 to give 99.0 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.86-0.91 (m, 15H), 1.21-1.40 (m, 50H), 1.58-1.71 (m, 6H), 2.32 (t, J=7.5 Hz, 2H), 2.48-2.53 (m, 4H), 2.62 (t, J=6.6 Hz, 2H), 3.61 (t, J=6.3 Hz, 2H), 3.80 (s, 2H), 4.10 (td, J=2.7, 6.3 Hz, 4H); ESI-MS m/z: 726.70 [M+H]+.


Example 78: Synthesis of Compound 27b



text missing or illegible when filed


Compound 27b was prepared referring to the method of Example 77 to give 70.0 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.85-0.95 (m, 15H), 1.25-1.55 (m, 48H), 1.59-1.83 (m, 10H), 2.25 (d, J=6.6 Hz, 2H), 2.48-2.53 (m, 4H), 2.62 (t, J=6.3 Hz, 2H), 3.61 (t, J=6.3 Hz, 2H), 3.79 (s, 2H), 4.11 (td, J=3.0, 6.6 Hz, 4H); ESI-MS m/z: 740.65 [M+H]+.


Example 79: Synthesis of Compound 28b



text missing or illegible when filed


Compound 28b was prepared referring to the method of Example 77 to give 97.8 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.85-0.95 (m, 15H), 1.21-1.55 (m, 48H), 1.60-1.84 (m, 10H), 2.25 (d, J=6.6 Hz, 2H), 2.47-2.53 (m, 4H), 2.61 (t, J=6.3 Hz, 2H), 3.61 (t, J=6.3 Hz, 2H), 3.79 (s, 2H), 4.11 (td, J=4.5, 6.3 Hz, 4H); ESI-MS m/z: 740.80 [M+H]+.


Example 80: Synthesis of Compound 29b



text missing or illegible when filed


Compound 29b was prepared referring to the method of Example 77 to give 107.7 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.85-0.95 (m, 15H), 1.21-1.55 (m, 53H), 1.60-1.84 (m, 5H), 2.25 (d, J=6.6 Hz, 2H), 2.50-2.55 (m, 4H), 2.64 (t, J=6.3 Hz, 2H), 3.60 (t, J=6.3 Hz, 2H), 3.79 (s, 2H), 4.11 (td, J=1.8, 6.6 Hz, 4H); ESI-MS m/z: 740.60 [M+H]+.


Example 81: Synthesis of Compound 30b



text missing or illegible when filed


Compound 30b was prepared referring to the method of Example 77 to give 82.4 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.87-0.92 (m, 15H), 1.21-1.53 (m, 49H), 1.55-1.83 (m, 13H), 2.25 (d, J=6.9 Hz, 2H), 2.46-2.52 (m, 6H), 3.55 (t, J=5.7 Hz, 2H), 3.79 (s, 2H), 4.11 (td, J=3.0, 6.6 Hz, 4H); ESI-MS m/z: 768.70 [M+H]+.


Example 82: Synthesis of Compound 31b



text missing or illegible when filed


Compound 31b was prepared referring to the method of Example 77 to give 107.7 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.86-0.92 (m, 15H), 1.21-1.53 (m, 49H), 1.55-1.83 (m, 13H), 2.25 (d, J=6.6 Hz, 2H), 2.47-2.54 (m, 6H), 3.55 (t, J=5.7 Hz, 2H), 3.79 (s, 2H), 4.10 (td, J=6.6, 12.3 Hz, 4H); ESI-MS m/z: 768.70 [M+H]+.


Example 83: Synthesis of Compound 32b



embedded image


Compound 32b was prepared referring to the method of Example 77 to give 84.3 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.88-0.93 (m, 15H), 1.22-1.53 (m, 49H), 1.56-1.83 (m, 13H), 2.25 (d, J=6.6 Hz, 2H), 2.49-2.54 (m, 6H), 3.55 (t, J=5.4 Hz, 2H), 3.79 (s, 2H), 4.10 (td, J=2.1, 6.6 Hz, 4H); ESI-MS m/z: 768.65 [M+H]+.


Example 84: Synthesis of Compound 33b



text missing or illegible when filed


Compound 33b was prepared referring to the method of Example 77 to give 74.9 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.88-0.92 (m, 15H), 1.22-1.53 (m, 51H), 1.48-1.69 (m, 9H), 2.33 (t, J=7.5 Hz, 2H), 2.50-2.55 (m, 4H), 2.63 (t, J=6.3 Hz, 2H), 3.62 (t, J=6.6 Hz, 2H), 3.80 (s, 2H), 4.02 (d, J=5.7 Hz, 2H), 4.11 (t, J=6.6 Hz, 2H); ESI-MS m/z: 754.60 [M+H]+.


Example 85: Synthesis of Compound 34b



text missing or illegible when filed


Compound 34b was prepared referring to the method of Example 77 to give 76.6 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.36-0.46 (m, 4H), 0.88-0.92 (m, 9H), 1.20-1.51 (m, 55H), 1.56-1.69 (m, 5H), 2.32 (t, J=7.5 Hz, 2H), 2.51-2.55 (m, 4H), 2.64 (t, J=6.3 Hz, 2H), 3.62 (t, J=6.3 Hz, 2H), 3.92 (s, 2H), 4.02 (d, J=5.7 Hz, 2H), 4.11 (t, J=6.6 Hz, 2H); ESI-MS m/z: 752.60 [M+H]+.


Example 86: Synthesis of Compound 35b



text missing or illegible when filed


Compound 35b was prepared referring to the method of Example 77 to give 100.0 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.88-0.92 (m, 15H), 1.24-1.72 (m, 64H), 2.33 (t, J=7.5 Hz, 2H), 2.45-2.51 (m, 6H), 3.55 (t, J=6.0 Hz, 2H), 3.80 (s, 2H), 4.02 (d, J=5.7 Hz, 2H), 4.11 (t, J=6.3 Hz, 2H); ESI-MS m/z: 782.65 [M+H]+.


Example 87: Synthesis of Compound 36b



embedded image


Compound 36b was prepared referring to the method of Example 77 to give 107.0 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.34-0.46 (m, 4H), 0.87-0.92 (m, 9H), 1.22-1.72 (m, 64H), 2.32 (t, J=7.2 Hz, 2H), 2.45-2.51 (m, 6H), 3.55 (t, J=5.7 Hz, 2H), 3.92 (s, 2H), 4.02 (d, J=5.7 Hz, 2H), 4.11 (t, J=6.3 Hz, 2H); ESI-MS m/z: 780.80 [M+H]+.


Example 88: Synthesis of Compound 37b



embedded image


Compound 37b was prepared referring to the method of Example 77 to give 71.6 mg of an oily product.



1H NMR (300 MHz, MeOH-d4) δ: 0.89-0.94 (m, 18H), 1.24-1.69 (m, 67H), 2.32 (t, J=7.5 Hz, 2H), 2.50-2.55 (m, 4H), 2.64 (t, J=6.3 Hz, 2H), 3.62 (t, J=6.3 Hz, 2H), 3.80 (s, 2H), 4.11 (td, J=6.3, 14.7 Hz, 4H); ESI-MS m/z: 824.70 [M+H]+.


Example 89: Synthesis of Compound 52a



embedded image


Compound 52a was prepared referring to the method of Example 1 to give 243.8 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.87-4.81 (m, 1H), 3.59 (m, 2H), 2.86 (t, J=7.2 Hz, 2H), 2.65-2.51 (m, 8H), 1.64-1.58 (m, 4H), 1.57-1.43 (m, 11H), 1.39-1.19 (m, 48H), 1.15 (s, 6H), 0.90 (t, J=7.2 Hz, 9H); ESI-MS m/z: 754.60 [M+H]+.


Example 90: Synthesis of Compound 53a



embedded image


Compound 53a was prepared referring to the method of Example 1 to give 262.7 mg of an oily product.


1H NMR (300 MHz, CDCl3) δ: 4.90-4.78 (m, 1H), 3.56 (t, J=5.1 Hz, 2H), 2.86 (t, J=7.2 Hz, 2H), 2.63-2.48 (m, 8H), 1.81-1.19 (m, 63H), 1.16 (s, 6H), 0.89 (t, J=7.2 Hz, 9H); ESI-MS m/z: 754.60 [M+H]+.


Example 91: Synthesis of Compound 54a



embedded image


Compound 54a was prepared referring to the method of Example 1 to give 287.7 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.89-4.79 (m, 1H), 4.66 (t, J=2.1 Hz, 2H), 3.53 (t, J=5.4 Hz, 2H), 2.58 (t, J=5.4 Hz, 2H), 2.45 (t, J=7.2 Hz, 4H), 2.34 (t, J=7.2 Hz, 2H), 2.24-2.20 (m, 2H), 1.79-1.17 (m, 57H), 1.15 (s, 6H), 0.88 (t, J=7.2 Hz, 9H); ESI-MS m/z: 734.60 [M+H]+.


Example 92: Synthesis of Compound 55a



embedded image


Compound 55a was prepared referring to the method of Example 1 to give 173.5 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.89-4.78 (m, 1H), 4.13 (t, J=7.2 Hz, 2H), 3.56 (m, 2H), 2.63 (m, 2H), 2.51-2.45 (m, 6H), 2.31 (t, J=7.2 Hz, 2H), 2.16-2.10 (m, 2H), 1.78-1.17 (m, 55H), 1.15 (s, 6H), 0.90 (m, 9H); ESI-MS m/z: 734.50 [M+H]+.


Example 93: Synthesis of Compound 56a



embedded image


Compound 56a was prepared referring to the method of Example 1 to give 112.2 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.87-4.79 (m, 1H), 4.16 (t, J=6.6 Hz, 2H), 3.60 (m, 2H), 2.67-2.53 (m, 6H), 2.32-2.11 (m, 6H), 2.32-2.11 (m, 6H), 1.85-1.76 (m, 2H), 1.66-1.18 (m, 53H), 1.14 (s, 6H), 0.88 (m, 9H); ESI-MS m/z: 734.55 [M+H]+.


Example 94: Synthesis of Compound 57a



embedded image


Compound 57a was prepared referring to the method of Example 1 to give 45.0 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 5.68-5.49 (m, 2H), 4.85-4.79 (m, 1H), 4.63 (d, J=6.8 Hz, 2H), 3.84 (m, 2H), 2.95-2.76 (m, 6H), 2.31 (t, J=7.2 Hz, 2H), 2.12-2.07 (m, 2H), 1.66-1.18 (m, 57H), 1.16 (s, 6H), 0.88 (m, 9H); ESI-MS m/z: 736.60 [M+H]+.


Example 95: Synthesis of Compound 58a



embedded image


In a 50 mL round-bottom flask, the reactants 1-nonanamine (286.5 mg, 2.0 mmol, 1.0 eq.), 8-bromooctanoic acid (535.5 mg, 2.4 mmol, 1.2 eq.), NMM (202.3 mg, 2.0 mmol, 1.0 eq.), and HATU (1.14 g, 3.0 mmol, 1.5 eq.) were dissolved in a mixed solution of DMF (2.0 ml) and DCM (10.0 mL), and the mixture was stirred at room temperature for reaction until the reaction of 1-nonanamine was completed as monitored by TLC. The reaction liquid was introduced into 100 mL of a saturated sodium chloride solution, and the mixture was extracted with DCM (30 mL×3). The organic phases were combined and dried over anhydrous sodium sulfate. The mixture was filtered, and the filtrate was concentrated to dryness to give a crude product, which was purified by silica gel column chromatography to give 610 mg of compound 58a-2.


Compound 58a was prepared referring to the method of Example 1 to give 104.0 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 5.53 (s, 1H), 4.87-4.79 (m, 1H), 3.22 (dd, J=6.9 Hz, 13.2 Hz, 2H), 2.67-2.52 (m, 6H), 2.15 (t, J=7.2 Hz, 2H), 1.65-1.19 (m, 63H), 1.15 (s, 6H), 0.88 (t, J=6.9 Hz, 9H); ESI-MS m/z: 737.60 [M+H]+.


Example 96: Synthesis of Compound 59a



embedded image


In a 100 mL sealed tube, 4-dimethylamino-1-butanol (3.0 g, 25.5 mmol, 1.0 eq.) and thiourea (8.4 g, 110.0 mmol, 4.3 eq.) were added, then an aqueous HBr solution (48%, 60 mL) was added, and the mixture was heated to 120° C. and stirred for overnight, which was directly used in the next step without purification.


The system described above was cooled to 0° C., NaOH (10.2 g, 255.0 mmol, 10.0 eq.) was added in batches, and the mixture was heated to 120° C. and reacted for 2 h. The mixture was cooled to room temperature and extracted with dichloromethane. The organic phases were combined, dried over anhydrous sodium sulfate, and filtered to remove the drying agent, and the solvent was removed to give 2.45 g of a colorless oily compound 59a-3.


In a 100 mL three-neck flask, compound 59a-6 (1.5 g, 5.8 mmol, 1.0 eq.), 2-heptyl-1-nonanol (1.40 g, 5.8 mmol, 1.0 eq.), EDCI (1.66 g, 8.7 mmol, 1.5 eq.), and DMAP (0.35 g, 2.9 mmol, 0.5 eq.) were dissolved in 20 ml of dichloromethane, and the mixture was stirred at room temperature overnight. The mixture was washed with water (3×15 ml). The organic phase was subjected to rotary evaporation under reduced pressure, directly mixed with silica gel, and purified by silica gel column chromatography to give 3.3 g of compound 59a-7.


In a 100 mL three-neck flask, compound 59a-7 (2.8 g, 5.8 mmol, 1.0 eq.) was dissolved in 20 mL of dichloromethane, HCl/dioxane (4 M, 14 mL) was added, and the mixture was stirred at room temperature overnight. The organic solvent was removed by rotary evaporation, the reaction liquid was adjusted to neutrality with a saturated sodium bicarbonate solution, and the mixture was extracted with DCM (3×20 ml). The organic phase was collected and dried over anhydrous Na2SO4. The organic phase was purified by silica gel column chromatography to give 1.95 g of compound 8.


In a 40 mL sealed tube, compound 59a-8 (680 mg, 1.8 mmol, 1.0 eq.), KI (365 mg, 2.2 mmol, 1.2 eq.), K2CO3 (745 mg, 5.4 mmol, 3.0 eq.), and 59a-5 (668.9 mg, 1.8 mmol, 1.0 eq.) were dissolved in 10 mL of acetonitrile. The mixture was heated to 80° C. and stirred for reaction overnight. After the reaction was completed, the mixture was cooled to room temperature and filtered, and the filter cake was washed with acetonitrile (5 ml×2). The filtrate was collected and concentrated by rotary evaporation to dryness. The residue was purified by column chromatography to give 580 mg of a yellow oily compound 59a-9.


In a 20 mL sealed tube, compound 59a-9 (200 mg, 0.29 mmol, 1.0 eq.) was dissolved in 5 mL of DCM. After the reaction system was cooled to 5° C., TEA (59.5 mg, 0.59 mmol, 2.0 eq.) and BTC (87.3 mg, 0.29 mmol, 1.0 eq.) were added dropwise. The temperature was kept at 5° C., the mixture was stirred for reaction for 1 h and concentrated to remove DCM, and 5 mL of tetrahydrofuran was added. In another 20 mL sealed tube, compound 59a-3 (101.9 mg, 0.76 mmol, 2.6 eq.) was dissolved in 5 mL of tetrahydrofuran. After the mixture was cooled to 0° C., NaH (60%, 58.0 mg, 1.45 mmol, 5.0 eq.) was added. After stirring at 0° C. for 1 h, the above prepared THF mixed solution was added dropwise, and the mixture was reacted for another 1 h. The reaction liquid was poured into 10 mL of ice water, and the mixture was extracted with ethyl acetate. The organic phases were combined, dried over anhydrous sodium sulfate, and filtered to remove the drying agent, and the solvent was removed to give a crude product. The crude product was purified by Prep-HPLC (Column: XSelect C18 (30×150 mm, 5 μm); Eluent A: H2O/ACN 60/40, 10 mM NH4HCO3+1% NH3·H2O; Eluent B: IPA/ACN 90/10; Flow rate: 60 mL/min; Gradient program: 65%-85% B in 0-12 min) to give 126.9 mg of a light yellow oily compound 59a.



1H NMR (300 MHz, CDCl3) δ: 4.06 (t, J=6.6 Hz, 2H), 3.96 (d, J=5.7 Hz, 2H), 3.36-3.15 (m, 4H), 2.90 (t, J=6.9 Hz, 2H), 2.41 (s, 2H), 2.31-2.24 (m, 10H), 1.64-1.16 (m, 61H), 0.93-0.77 (m, 15H); ESI-MS m/z: 839.65 [M+H]+.


Example 97: Synthesis of Compound 60a



embedded image


Compound 60a was prepared referring to the method of Example 96 to give 157.3 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.86 (p, J=6.3 Hz, 1H), 4.06 (t, J=6.9 Hz, 2H), 3.37-3.17 (m, 4H), 2.90 (t, J=6.9 Hz, 2H), 2.39-2.25 (m, 12H), 1.79-1.18 (m, 70H), 0.93-0.83 (m, 15H); ESI-MS m/z: 895.80 [M+H]+.


Example 98: Synthesis of Compound 61a



embedded image


Compound 61a was prepared referring to the method of Example 96 to give 184.0 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.06 (t, J=6.9 Hz, 2H), 3.96 (d, J=5.7 Hz, 2H), 3.35-3.18 (m, 4H), 2.91 (t, J=7.2 Hz, 2H), 2.43-2.27 (m, 12H), 1.87-1.77 (m, 2H), 1.61-1.18 (m, 57H), 0.93-0.84 (m, 15H); ESI-MS m/z: 825.55 [M+H].


Example 99: Synthesis of Compound 38b



embedded image


Compound 38b was prepared referring to the method of Example 96 to give 228 mg of an oily product.



1H NMR (300 MHz, CDClb) δ: 4.86 (p, J=6.3 Hz, 1H), 3.78 (s, 2H), 3.33-3.18 (m, 4H), 2.90 (t, J=6.6 Hz, 2H), 2.38-2.25 (m, 12H), 1.69-1.44 (m, 16H), 1.34-1.17 (m, 48H), 0.90-0.85 (m, 15H); ESI-MS m/z: 853.65 [M+H]+.


Example 100: Synthesis of Compound 39b



embedded image


Compound 39b was prepared referring to the method of Example 96 to give 117.9 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.86 (p, J=6.3 Hz, 1H), 3.78 (s, 2H), 3.36-3.17 (m, 4H), 2.92 (t, J=7.2 Hz, 2H), 2.46 (t, J=7.2 Hz, 2H), 2.34-2.22 (m, 10H), 1.90-1.80 (m, 3H), 1.62-1.49 (m, 14H), 1.34-1.17 (m, 51H), 0.93-0.86 (m, 15H); ESI-MS m/z: 881.65 [M+H]+.


Example 101: Synthesis of Compound 40b



embedded image


Compound 40b was prepared referring to the method of Example 96 to give 75.3 mg of an oily product.



1H NMR (300 MHz, CDCb) δ: 4.86 (p, J=6.3 Hz, 1H), 3.78 (s, 2H), 3.36-3.17 (m, 4H), 2.91 (t, J=6.6 Hz, 2H), 2.48-2.28 (m, 12H), 1.89-1.60 (m, 10H), 1.51-1.16 (m, 60H), 0.93-0.87 (m, 15H); ESI-MS m/z: 895.70 [M+H]+.


Example 102: Synthesis of Compound 41b



embedded image


Compound 41b was prepared referring to the method of Example 96 to give 95.1 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.86 (p, J=6.4 Hz, 1H), 4.09 (t, J=7.6 Hz, 2H), 3.31-3.24 (m, 4H), 2.90 (t, J=7.6 Hz, 2H), 2.40-2.26 (m, 12H), 1.63-1.50 (m, 21H), 1.32-1.16 (m, 49H), 0.90-0.86 (m, 15H); ESI-MS m/z: 895.80 [M+H]+.


Example 103: Synthesis of Compound 42b



embedded image


Compound 42b was prepared referring to the method of Example 96 to give 63.6 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.09 (p, J=7.6 Hz, 2H), 3.96 (d, J=6.0 Hz, 2H), 3.31-3.24 (m, 4H), 2.90 (t, J=7.2 Hz, 2H), 2.57-2.26 (m, 12H), 1.67-1.43 (m, 20H), 1.32-1.16 (m, 41H), 0.90-0.86 (m, 15H); ESI-MS m/z: 839.70 [M+H]+.


Example 104: Synthesis of Compound 43b



embedded image


Compound 43b was prepared referring to the method of Example 96 to give 130.5 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 3.96 (d, J=6.0 Hz, 2H), 3.89 (s, 2H), 3.35-3.18 (m, 4H), 2.91 (t, J=7.2 Hz, 2H), 2.44-2.28 (m, 12H), 1.87-1.79 (m, 2H), 1.77-1.60 (m, 10H), 1.34-1.17 (m, 47H), 0.90-0.85 (m, 9H), 0.52-0.32 (m, 4H); ESI-MS m/z: 823.60 [M+H]+.


Example 105: Synthesis of Compound 44b



embedded image


Compound 44b was prepared referring to the method of Example 96 to give 79.4 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 3.95 (d, J=5.7 Hz, 2H), 3.90 (s, 2H), 3.35-3.19 (m, 4H), 2.90 (t, J=7.2 Hz, 2H), 2.67 (s, 4H), 2.34-2.27 (m, 4H), 1.72-1.51 (m, 16H), 1.29-1.21 (m, 49H), 0.90-0.83 (m, 9H), 0.46-0.36 (in, 4H); ESI-MS m/z: 837.75 [M+H]+.


Example 106: Synthesis of Compound 45b



embedded image


Compound 45b was prepared referring to the method of Example 96 to give 41 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.13 (s, 2H), 3.96 (d, J=6.0 Hz, 2H), 3.31-3.24 (m, 4H), 2.90 (t, J=6.8 Hz, 2H), 2.54-2.26 (m, 10H), 1.67-1.45 (m, 18H), 1.29-1.21 (m, 45H), 0.90-0.82 (m, 9H), 0.32-0.29 (m, 4H); ESI-MS m/z: 837.70 [M+H]+.


Example 107: Synthesis of Compound 46b



embedded image


In a 100 mL three-neck round-bottom flask, compound 46b-1 (1.0 g, 4.60 mmol, 1.0 eq.), pyridine (0.74 mL, 9.2 mmol, 2.0 eq.), DMAP (112.4 mg, 0.92 mmol, 0.2 eq.), and p-nitrophenyl chloroformate (1.11 g, 5.52 mmol, 1.2 eq.) were dissolved in 20 mL of dichloromethane at room temperature under nitrogen atmosphere. After the reaction system was stirred at room temperature for 1 h, 2-nonyl-1-decanol (3.93 g, 13.8 mmol, 3.0 eq.) and DIEA (2.40 mL, 13.8 mmol, 3.0 eq.) were added to the reaction system. The reaction system was stirred at room temperature overnight. The reaction was monitored by TLC, and a new spot of the product was found. The mixture was diluted with DCM (20 mL) and washed once with a saturated sodium bicarbonate solution (10 mL). The organic phase was washed 2 times with a saturated NaCl solution (40 mL), dried over anhydrous Na2SO4, filtered, and concentrated. The crude product was purified by silica gel column chromatography to give a light yellow oily compound 46b-2 (1.68 g).


Compound 46b was prepared referring to the method of Example 96 to give 78.5 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.68 (p, J=6.0 Hz, 1H), 4.11 (t, J=6.6 Hz, 2H), 3.78 (s, 2H), 3.36-3.22 (m, 4H), 2.90 (t, J=6.9 Hz, 2H), 2.47-2.29 (m, 10H), 1.70-1.56 (m, 18H), 1.29-1.21 (m, 50H), 0.90-0.86 (m, 15H); ESI-MS m/z: 897.70 [M+H]+.


Example 108: Synthesis of Compound 47b



embedded image


Compound 47b was prepared referring to the method of Example 107 to give 189.8 mg of an oily product.



1H NMR (400 MHz, CDCl3) δ: 4.12 (t, J=6.4 Hz, 2H), 4.02 (d, J=5.6 Hz, 2H), 3.78 (s, 2H), 3.32-3.24 (m, 4H), 2.92 (t, J=7.2 Hz, 2H), 2.47-2.30 (m, 10H), 1.85-1.84 (m, 2H), 1.66-1.40 (m, 7H), 1.26-1.18 (m, 48H), 0.93-0.88 (m, 15H); ESI-MS m/z: 827.55 [M+H]+.


Example 109: Synthesis of Compound 48b



embedded image


Compound 48b was prepared referring to the method of Example 107 to give 129.4 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.12 (t, J=6.6 Hz, 2H), 4.02 (d, J=5.7 Hz, 2H), 3.78 (s, 2H), 3.36-3.21 (m, 4H), 2.90 (t, J=6.9 Hz, 2H), 2.36-2.26 (m, 10H), 1.63-1.41 (m, 13H), 1.26-1.18 (m, 46H), 0.89-0.86 (m, 15H); ESI-MS m/z: 841.65 [M+H]+.


Example 110: Synthesis of Compound 49b



embedded image


Compound 49b was prepared referring to the method of Example 107 to give 135.6 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.12 (t, J=6.3 Hz, 2H), 4.02 (d, J=5.7 Hz, 2H), 3.89 (s, 2H), 3.31-3.22 (m, 4H), 2.90 (t, J=6.9 Hz, 2H), 2.34-2.26 (m, 10H), 1.65-1.48 (m, 13H), 1.27-1.19 (m, 46H), 0.90-0.86 (m, 9H), 0.45-0.35 (m, 4H); ESI-MS m/z: 839.50 [M+H]+.


Example 111: Synthesis of Compound 50b



embedded image


Compound 50b was prepared referring to the method of Example 107 to give 123.1 mg of an oily product.



1H NMR (300 MHz, CDCl3) δ: 4.12 (t, J=6.6 Hz, 2H), 4.02 (t, J=6.6 Hz, 2H), 3.84 (s, 2H), 3.34-3.19 (m, 4H), 2.90 (t, J=6.6 Hz, 2H), 2.46-2.20 (m, 10H), 1.84 (m, 2H), 1.65-1.44 (m, 13H), 1.27-1.19 (m, 46H), 0.92-0.86 (m, 15H); ESI-MS m/z: 855.60 [M+H]+.


The compounds in Table below were synthesized using the methods of the above examples, or using similar methods of the corresponding intermediates.

















embedded image


51b





Example 112



[M + H]+: 766.65








embedded image


52b





Example 113



[M + H]+: 794.80








embedded image


53b





Example 114



[M + H]+: 764.70








embedded image


54b





Example 115



[M + H]+: 780.85








embedded image


55b





Example 116



[M + H]+: 808.90








embedded image


56b





Example 117



[M + H]+: 724.60








embedded image


57b





Example 118



[M + H]+: 724.60








embedded image


58b





Example 119



[M + H]+: 737.60








embedded image


59b





Example 120



[M + H]+: 752.65








embedded image


62a





Example 121



[M + H]+: 738.60








embedded image


63a





Example 122



[M + H]+: 780.65








embedded image


64a





Example 123



[M + H]+: 822.70








embedded image


65b





Example 124



[M + H]+: 766.60








embedded image


66a





Example 125



[M + H]+: 794.70








embedded image


67a





Example 126



[M + H]+: 808.70








embedded image


68a





Example 127



[M + H]+: 850.75








embedded image


69a





Example 128



[M + H]+: 752.60








embedded image


70a





Example 129



[M + H]+: 738.55








embedded image


60b





Example 130



[M + H]+: 738.75








embedded image


61b





Example 131



[M + H]+: 724.70








embedded image


62b





Example 132



[M + H]+: 710.80








embedded image


63b





Example 133



[M + H]+: 766.95








embedded image


64b





Example 134



[M + H]+: 752.65








embedded image


65b





Example 135



[M + H]+: 822.75








embedded image


66b





Example 136



[M + H]+: 864.85








embedded image


67b





Example 137



[M + H]+: 738.55








embedded image


68b





Example 138



[M + H]+: 724.70








embedded image


69b





Example 139



[M + H]+: 766.70








embedded image


70b





Example 140



[M + H]+: 752.75








embedded image


71b





Example 141



[M + H]+: 710.70








embedded image


72b





Example 142



[M + H]+: 752.70








embedded image


73b





Example 143



[M + H]+: 724.70








embedded image


74b





Example 144



[M + H]+: 724.65








embedded image


75b





Example 145



[M + H]+: 710.65








embedded image


76b





Example 146



[M + H]+: 156








embedded image


77b





Example 147



[M + H]+: 738.80








embedded image


78b





Example 148



[M + H]+: 808.70








embedded image


79b





Example 149



[M + H]+: 766.70








embedded image


80b





Example 150



[M + H]+: 752.65








embedded image


81b





Example 151



[M + H]+: 766.65








embedded image


82b





Example 152



[M + H]+: 752.75








embedded image


83b





Example 153



[M + H]+: 724.65








embedded image


84b





Example 154



[M + H]+: 766.65








embedded image


85b





Example 155



[M + H]+: 794.85








embedded image


86b





Example 156



[M + H]+: 836.70








embedded image


87b





Example 157



[M + H]+: 752.65








embedded image


88b





Example 158



[M + H]+: 808.70








embedded image


89b





Example 159



[M + H]+: 780.65








embedded image


90b





Example 160



[M + H]+: 822.75








embedded image


91b





Example 161



[M + H]+: 850.80








embedded image


92b





Example 162



[M + H]+: 738.65








embedded image


93b





Example 163



[M + H]+: 752.70








embedded image


94b





Example 164



[M + H]+: 738.65








embedded image


95b





Example 165



[M + H]+: 780.90








embedded image


96b





Example 166



[M + H]+: 752.90








embedded image


97b





Example 167



[M + H]+: 822.75








embedded image


98b





Example 168



[M + H]+: 794.60








embedded image


99b





Example 169



[M + H]+: 766.65








embedded image


100b





Example 170



[M + H]+: 780.70








embedded image


101b





Example 171



[M + H]: 794.75








embedded image


102b





Example 172



[M + H]+: 724.70








embedded image


103b





Example 173



[M + H]+: 738.65








embedded image


104b





Example 174



[M + H]+: 752.75








embedded image


105b





Example 175



[M + H]+: 738.60








embedded image


106b





Example 176



[M + H]+: 780.70








embedded image


107b





Example 177



[M + H]+: 752.65








embedded image


108b





Example 178



[M + H]+: 766.60








embedded image


109b





Example 179



[M + H]: 794.75








embedded image


110b





Example 180



[M + H]+: 738.70








embedded image


111b





Example 181



[M + H]+: 710.60








embedded image


112b





Example 182



[M + H]+: 724.85








embedded image


113b





Example 183



[M + H]+: 738.85








embedded image


114b





Example 184



[M + H]+: 794.75








embedded image


115b





Example 185



[M + H]+: 808.80








embedded image


116b





Example 186



[M + H]+: 780.75








embedded image


117b





Example 187



[M + H]+: 752.90









Compounds 1c-283c were synthesized using the method as described in the above Examples, or similar methods using corresponding intermediates.


PHARMACOLOGICAL EXPERIMENTS
Assay Example 1: Preparation of Nanoparticles

Materials used for lipid nanoparticle assembly include: (1) ionizable lipid compounds: e.g., ionizable lipids designed and synthesized in the present disclosure or DLin-MC3-DMA (purchased from AVT) as a control; (2) structure lipids: e.g., Cholesterol (purchased from Sigma-Aldrich); (3) phospholipids: e.g., DSPC i.e., 1,2-distearoyl-SN-glycero-3-phosphocholine (Distearoylphosphatidylcholine, purchased from AVT); (4) polyethylene glycolated lipids: e.g. DMG-PEG2000 i.e., dimyristoylglycero-polyethylene glycol 2000 (1,2-dimyristoyl-rac-glycero-3-methoxypolyethylene glycol-2000, purchased from AVT); (5) active ingredients of nucleic acid fragments: e.g. Luciferase mRNA, siRNA, CRISPR Cas 9 mRNA, etc. (manufactured in-house). The names of materials of the lipid nanoparticle assembly and their structural formulae are detailed in Table 4.











TABLE 4






Structure



No.
name
Structural formula







1
DLin- MC3- DMA (MC3)


embedded image







2
Cholesterol


embedded image







3
DSPC


embedded image







4
DMG- PEG2000


embedded image











Lipid nanoparticles were prepared by (1) dissolving and mixing ionizable lipid compounds, cholesterol, phospholipids and polyethylene glycolated lipids in ethanol at (molar percentages) 50%, 38.5%, 10% and 1.5%, respectively; (2) dissolving the mRNA active ingredient in 25 mM sodium acetate solution (pH=4.5); (3) using an automated high-throughput microfluidic system to mix the organic phase containing the lipid mixture and the aqueous phase containing the mRNA component in the flow ratio range of 1:1 to 1:4 at a mixing speed of 10 mi/min to 18 mL/min; (4) the prepared lipid nanoparticles (N/P ratio=6) were diluted with phosphate buffer solution and the nanoparticle solutions were ultrafiltered to the original preparation volume using ultrafiltration tubes (purchased from Millipore) with a cut-off molecular weight of 30 kDa; and (5) the obtained nanoparticles were filtered through a sterile 0.2 μm filter membrane and then stored in a sealed glass vial at low temperature.


The preparation method of lipid nanoparticles includes microfluidic mixing systems, but is not limited to this method, which also includes T-type mixers and ethanol injection method, and the like.


Assay Example 2: Characterization of Physical Properties of Lipid Nanoparticles

The particle size and particle size dispersity index (PDI) of the prepared lipid nanoparticles were measured using a Zetasizer Pro (purchased from Malvern Instruments Ltd) and a DynaPro NanoStar (purchased from Wyatt) dynamic light scattering instrument. The degree of RNA encapsulation by lipid nanoparticles was characterized by the Encapsulation Efficiency %, which reflects the degree of binding of lipid nanoparticles to RNA fragments. This parameter was measured by the method of Quant-it™ RiboGreen RNA Assay (purchased from Invitrogen). Lipid nanoparticle samples were diluted in TE buffer (10 mM Tris-HCl, 1 mM EDTA, pH=7.5). A portion of the sample solution was removed, to which 0.5% Triton (Triton X-100) was added, and then allowed to stand at 37° C. for 30 minutes. Immediately after the addition of RIBOGREEN® reaction solution, the fluorescence values were read on a Varioskan LUX multifunctional microplate reader (purchased from Thermofisher) at 485 nm for absorption and 528 nm for emission to give the encapsulation efficiency values.


Assay Example 3: Animal Experiment

The delivery effect and safety of nanoparticles encapsulated with luciferase mRNA (Trilink, L-7202) in mice were evaluated. The test mice were SPF-grade C57BL/6 mice, female, 6-8 weeks old, weighing 18-22 g, and were purchased from SPF (Beijing) Biotechnology Co., Ltd. All animals were acclimatized for more than 7 days prior to the experiment, and had free access to food and water during the experiment. The conditions include alternating light and dark for 12/12 h, the indoor temperature of 20-26° C. and the humidity of 40-70%. The mice were randomly grouped. The lipid nanoparticles encapsulated with luciferase mRNA prepared above were injected into mice by intravenous administration at a single dose of 0.5 mg/kg mRNA, and the mice were subjected to in vivo bioluminescence assay using a Small Animal In Vivo imaging System (IVIS LUMINA III, purchased from PerkinElmer) at 6 h after administration. The assay was performed as follows: D-luciferin solution was prepared in saline at a concentration of 15 mg/mL, and each mouse was given the substrate by intraperitoneal injection. At ten minutes after administration of the substrate, the mice were anesthetized in an anesthesia chamber with isoflurane at a concentration of 2.5%. The anesthetized mice were placed in IVIS for fluorescence imaging, and data acquisition and analysis were performed on the concentrated distribution area of fluorescence.


The in vivo delivery efficiency of lipid nanoparticle carriers was expressed as the mean values of fluorescence intensity and total photon count in different animals within the same subject group, as shown in Table 5-Table 11. Higher values of fluorescence intensity and total photon count indicate higher in vivo delivery efficiency of this mRNA fragment by lipid nanoparticles. The lipid nanoparticles containing the cationic lipids of the present disclosure have good in vivo delivery efficiency.













TABLE 5









Total photon






count in vivo at



Particle
Particle-size
Encapsulation
6 hours after



size
distribution
efficiency
administration


Compound
(nm)
(PDI)
(%)
(Total Flux)



















 1a
113.08
0.022
94.61
3.52E+10


 3a
89.72
0.021
94.23
1.03E+11


 4a
103.7
0.005
92.87
3.25E+10


 6a
90.67
0.018
87.22
2.88E+10


 7a
103.32
0.018
90.92
5.26E+10


 9a
103.03
0.031
89.64
2.06E+10


10a
131.09
0.032
89.69
7.08E+10


11a
98.01
0.021
89.36
2.05E+10


12a
104.85
0.021
85.24
1.39E+10


13a
110.20
0.018
92.96
5.01E+10


14a
90.99
0.012
91.67
1.09E+11


15a
95.94
0.021
90.77
4.15E+10


19a
109.37
0.035
87.90
4.73E+10


20a
102.13
0.032
87.83
1.18E+11


21a
110.00
0.016
88.37
8.84E+10


22a
99.48
0.010
97.25
1.74E+10


23a
99.25
0.011
96.30
9.84E+10


24a
109.53
0.052
88.89
3.07E+10


25a
86.99
0.051
95.62
1.33E+11


26a
106.37
0.050
91.18
1.11E+11


27a
136.43
0.016
93.52
4.31E+10


28a
110.30
0.018
93.90
4.30E+10


29a
88.59
0.012
97.30
5.46E+10


MC3
96.83
0.04
95.62
8.04E+09




















TABLE 6









Total photon






count in vivo at



Particle
Particle-size
Encapsulation
6 hours after



size
distribution
efficiency
administration


Compound
(nm)
(PDI)
(%)
(Total Flux)



















 1b
88.59
0.031
89.89
9.34E+10


 2b
104.61
0.013
80.17
8.31E+10


 3b
102.32
0.015
74.43
1.41E+11


 6b
99.55
0.029
80.01
7.42E+10


 7b
101.67
0.030
88.68
2.00E+10


10b
114.72
0.052
72.72
3.15E+10


12b
104.60
0.031
91.04
1.19E+10


13b
99.02
0.018
89.27
1.81E+11


14b
99.73
0.013
83.54
1.05E+11


15b
120.27
0.015
69.69
8.22E+10


16b
101.90
0.028
89.09
1.28E+11


17b
109.87
0.030
87.72
1.74E+11


18b
100.46
0.037
89.34
3.91E+10


19b
109.53
0.036
83.01
6.13E+10


20b
107.83
0.032
87.69
4.82E+10


21b
94.58
0.051
92.37
4.95E+10


22b
99.44
0.050
93.03
8.78E+10


23b
115.70
0.031
93.14
3.92E+10


24b
83.05
0.019
94.52
7.06E+10


MC3
96.83
0.04
95.62
8.04E+09




















TABLE 7









Total photon






count in vivo at



Particle
Particle-size
Encapsulation
6 hours after



size
distribution
efficiency
administration


Compound
(nm)
(PDI)
(%)
(Total Flux)



















30a
107.40
0.12
86.94
8.32E+10


31a
115.56
0.11
86.08
7.04E+10


32a
129.00
0.10
83.51
1.24E+10


33a
111.60
0.17
86.98
1.09E+11


34a
116.67
0.12
95.73
3.14E+10


35a
126.63
0.22
95.00
3.01E+10


36a
104.80
0.15
92.41
2.51E+10


37a
110.67
0.15
92.14
3.54E+10


38a
114.03
0.13
94.51
1.39E+10


39a
144.83
0.08
93.59
2.24E+10


40a
120.90
0.18
95.08
2.03E+11


41a
114.03
0.13
94.51
1.11E+11


42a
104.13
0.17
95.54
3.47E+10


43a
105.63
0.15
97.03
2.23E+10


44a
92.67
0.20
96.84
1.06E+11


45a
114.73
0.15
94.63
1.22E+11


46a
110.83
0.16
93.03
9.06E+10


47a
104.47
0.21
93.23
3.10E+10


48a
81.22
0.16
96.13
8.16E+10


49a
100.56
0.14
94.85
2.28E+10


50a
78.03
0.12
98.67
2.86E+10


51a
92.08
0.15
94.81
3.11E+10


25b
94.39
0.15
95.59
1.11E+11


26b
113.77
0.14
94.58
6.53E+10


27b
91.10
0.18
97.83
5.59E+10


28b
94.55
0.16
96.19
4.18E+10


29b
112.43
0.10
96.28
3.52E+10


30b
115.83
0.08
95.02
4.03E+10


31b
118.73
0.07
95.73
1.87E+10


32b
116.50
0.16
95.31
3.52E+10


33b
85.80
0.15
94.85
8.23E+10


34b
85.20
0.19
97.61
2.78E+10


35b
100.40
0.21
96.35
3.77E+10


36b
112.50
0.19
96.10
3.84E+10


37b
83.72
0.16
95.15
3.72E+10


MC3
96.83
0.04
95.62
8.04E+09




















TABLE 8









Total photon






count in vivo at



Particle
Particle-size
Encapsulation
6 hours after



size
distribution
efficiency
administration


Compound
(nm)
(PDI)
(%)
(Total Flux)



















52a
79.40
0.09
93.30
2.58E+10


53a
124.00
0.23
90.96
9.04E+10


54a
87.37
0.14
91.34
1.32E+10


55a
126.17
0.16
73.85
1.96E+10


56a
86.37
0.09
82.84
3.23E+10


57a
79.17
0.17
95.41
3.42E+10


58a
187.50
0.18
68.54
2.72E+10




















TABLE 9









Total photon






count in vivo at



Particle
Particle-size
Encapsulation
6 hours after



size
distribution
efficiency
administration


Compound
(nm)
(PDI)
(%)
(Total Flux)



















60a
92.74
0.15
97.44
1.75E+10


61a
111.90
0.09
95.87
1.32E+10


38b
102.50
0.11
98.29
1.49E+10


39b
102.67
0.07
93.58
7.16E+10


40b
97.10
0.11
96.74
3.70E+10


43b
121.89
0.08
95.26
1.29E+10


44b
92.63
0.15
97.45
1.16E+10


46b
98.91
0.09
97.78
2.44E+10


47b
122.40
0.07
97.27
2.84E+10


50b
98.36
0.13
98.50
1.01E+10




















TABLE 10









Total photon






count in vivo at



Particle
Particle-size
Encapsulation
6 hours after



size
distribution
efficiency
administration


Compound
(nm)
(PDI)
(%)
(Total Flux)



















51b
91.96
0.12
95.92
7.60E+10


52b
105.97
0.06
90.84
1.01E+11


53b
125.03
0.42
91.92
1.00E+11


54b
81.37
0.17
96.69
5.71E+10


55b
90.37
0.11
94.65
1.02E+11


56b
98.59
0.16
93.87
8.35E+10


57b
98.23
0.18
94.15
3.86E+10


58b
112.93
0.15
92.88
6.17E+10


59b
127.23
0.12
80.27
1.04E+11


62a
98.07
0.17
89.88
1.20E+10


63a
80.23
0.17
95.56
1.17E+11


64a
86.99
0.15
96.72
9.82E+10


65a
95.88
0.16
95.93
6.53E+10


66a
105.77
0.11
92.88
9.31E+10


67a
111.33
0.06
91.57
6.07E+10


68a
103.93
0.04
94.47
7.55E+10


69a
107.20
0.34
96.65
7.41E+10


70a
86.08
0.15
96.69
9.33E+10




















TABLE 11









Total photon






count in vivo at



Particle
Particle-size
Encapsulation
6 hours after



size
distribution
efficiency
administration


Compound
(nm)
(PDI)
(%)
(Total Flux)



















63b
165.53
0.09
84.27
2.72E+10


64b
143.67
0.09
89.27
3.36E+10


65b
80.60
0.16
97.75
1.25E+11


66b
81.83
0.17
97.18
4.35E+10


67b
88.28
0.23
96.30
1.71E+11


68b
88.33
0.20
96.31
2.88E+11


69b
99.05
0.13
93.85
1.52E+11


70b
96.20
0.09
96.91
1.15E+11


71b
95.59
0.17
95.95
1.70E+11


72b
92.45
0.12
95.13
7.01E+10


73b
119.37
0.14
92.71
4.73E+10


74b
93.18
0.17
94.92
7.13E+10


75b
91.60
0.19
95.99
7.05E+10


76b
91.27
0.18
94.97
9.41E+10


77b
93.16
0.16
95.88
9.72E+10


78b
98.88
0.12
92.27
5.51E+10


79b
99.27
0.15
92.85
1.62E+11


80b
93.39
0.18
93.59
8.94E+10


81b
107.43
0.19
92.87
5.20E+10


82b
99.57
0.16
90.71
5.03E+10


83b
100.67
0.10
95.70
1.62E+10


84b
123.77
0.11
89.17
6.68E+10


85b
92.14
0.17
95.44
9.61E+10


86b
92.26
0.13
94.74
1.01E+11


87b
92.25
0.22
94.16
1.35E+11


88b
98.16
0.12
94.68
4.51E+10


89b
122.20
0.08
84.88
5.51E+10


90b
109.57
0.08
91.21
4.72E+10


91b
96.43
0.12
91.95
8.66E+10


92b
99.90
0.15
92.47
3.74E+10


93b
141.73
0.10
90.17
2.45E+10


94b
125.60
0.11
93.55
2.23E+10


95b
84.62
0.15
95.41
2.44E+10


96b
97.75
0.15
92.10
3.77E+10


97b
85.56
0.14
93.79
9.01E+10


98b
87.99
0.18
93.04
1.07E+11


99b
85.13
0.15
92.70
4.78E+10


100b 
101.50
0.12
91.14
8.51E+10


101b 
126.00
0.10
81.76
5.39E+10


102b 
92.12
0.16
93.92
7.52E+10


103b 
96.82
0.11
94.07
5.95E+10


104b 
135.57
0.09
91.51
3.20E+10


105b 
89.73
0.16
94.38
9.24E+10


106b 
88.22
0.15
94.89
1.42E+11


107b 
88.13
0.16
94.05
1.98E+11


108b 
86.61
0.16
94.16
1.38E+11


109b 
95.32
0.13
93.00
1.90E+11


110b 
133.93
0.21
85.48
1.36E+11


111b 
187.13
0.21
63.26
3.98E+10


112b 
175.67
0.17
69.74
1.31E+10


113b 
147.03
0.09
88.21
2.09E+10


114b 
103.40
0.13
93.55
4.53E+10


115b 
100.25
0.12
93.91
3.71E+10


116b 
120.13
0.09
90.71
1.96E+10


117b 
151.17
0.09
72.51
1.73E+10









Assay Example 4: Evaluation of Delivery Efficiency and Safety In Vitro

The delivery effect and safety of nanoparticles encapsulated with luciferase mRNA were evaluated at the cellular level in vitro. The cells used in the assay were human embryonic kidney cells 293 (HEK293T cells) cultured in DMEM (Dulbecco's Modified Eagle Medium) (purchased from Thermo Fisher) containing 10% fetal bovine serum and 5% penicillin-streptomycin double antibiotics at a indoor temperature of 37° C. and a CO2 concentration of 5%. The cells were uniformly dispersed and spread in 48-well plates, and incubated in the incubator for 24 h. Then a solution of the lipid nanoparticles encapsulated with luciferase mRNA were added. After 24 h, the cells were lysed, and the intracellular expression intensity and relative light units (RLU) of luciferase in each type of lipid nanoparticles were measured with a luciferase detection reagent (purchased from Promega). The higher the intensity of expression, the higher the delivery efficiency of the lipid material at the cellular level. Meanwhile, CCK-8 reagent (purchased from DOJINDO) was used in cytotoxicity testing for the parallel lipid nanoparticle-treated cell groups after 24 hours. In the test, the group of cells to which only PBS was added was used as a negative control. The procedure was as follows: after the addition of CCK-8 solution, the cells were left to stand in an incubator at 37° C. for 4 h. The absorbance values were read on a multifunctional microplate reader at an absorbance band of 450 nm. The ratio of the absorbance value of the nanoparticle-treated cells to that of the negative control was used as a characterization parameter for cell viability.


The delivery effects and the toxicity data of nanoparticles at the cellular level in vitro are shown in Table 12.











TABLE 12






Fluorescence intensity of cells
Cell viability


Compound
(RLU)
(%)

















20a
6.55E+06
105.11


MC3
2.95E+06
97.84









Assay Example 5: Evaluation of Clearance Rate of Ionizable Lipids In Vivo

The 6-8 weeks old C57 WT female mice (n=3) were injected by tail vein administration of 100 μL of unloaded LNP (0.1 mg/mL) formed from the ionizable lipid, cholesterol, DSPC, and DMG·PEG 2000 at a ratio of 50:38.5:10:1.5. The experimental animals were executed by removing the cervical vertebrae and dissected for the liver at 24 h and 72 h after administration, respectively, while the same batch of non-dosed experimental animal livers were used as a blank control group. Water was added to the liver sample for homogenization, and the protein was precipitated. The content of the ionizable lipid in the sample was quantitatively analyzed using LC-MS/MS in comparison to a calibration standard prepared from matched blank liver tissues.


Clearance rate datas for ionizable lipids are shown in Table 13.











TABLE 13






Percentage of cationic
Percentage of cationic



lipids in the liver in the
lipids in the liver in the



total dose, 24 h after
total dose, 72 h after


Compound
administration (%)
administration (%)

















 1a
<1
0


 4a
<1
0


 1b
15 ± 4.2
<1


 3b
12 ± 3.8
<1


10b
 5 ± 3.1
<1


12b
<1
0


13b
 8 ± 2.7
<1


30a
4
<2


26b
8
<1


22b
2
<1


39b
<1
<1


56a
<1
<1


Control
92 ± 3.5
89 ± 4.7


group









The control compound is of the following structure:




embedded image


Datas in Table 13 show that the geminal dialkyl compound in the control group degrade slowly in the liver, and about 90% of it still remains in the liver 72 hours after administration. The compounds of the present invention has a faster degradation rate, and can be degraded in the liver in about 72 hours.


While the present disclosure has been fully described by way of its embodiments, it is worth noting that various variations and modifications are apparent to those skilled in the art. Such variations and modifications should all be included within the scope of the claims appended to this disclosure.

Claims
  • 1. A compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof:
  • 2. The compound of formula (I) of claim 1, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, which has a structure of formula (II) or formula (III):
  • 3. The compound of formula (II) or formula (III) of claim 2, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,
  • 4. The compound of formula (II) of claim 2, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,
  • 5. The compound of formula (II) of claim 4, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein a=2;b=5;c=2, 3, 4, 5, or 6; alternatively c=6;d=0, 1, 2, 3, or 4; alternatively d=0;c+d=5 or 6, alternatively 6;M1 and M2 are —C(O)O—;R1 is C7-12 alkyl, alternatively C8-12 alkyl, alternatively C8-9 alkyl, or alternatively C9 alkyl, which is optionally substituted with 1 R1s;R1s is independently H or C1-10 alkyl, alternatively H or C1-9 alkyl, alternatively C6-8 alkyl, alternatively C7-8 alkyl;R2 is C7-11 linear alkyl, alternatively C10-11 linear alkyl, alternatively C11 linear alkyl, which is optionally substituted with 1 C1-3 alkyl, alternatively optionally substituted with 1 methyl;R4 and R5 are independently C1-3 alkyl, alternatively methyl;or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene;alternatively, R1 is
  • 6. The compound of formula (II) of claim 2, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,
  • 7. The compound of formula (II) of claim 6, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein a=2;b=5;c=2, 3, 4, 5, or 6; alternatively c=6;d=0, 1, 2, 3, or 4; alternatively d=0;c+d=5 or 6, alternatively 6;M1 is —C(O)O—, M2 is —C(O)O— or —C)O)S—; alternatively M1 abd M2 are —C(O))—;R1 is C7-12 alkyl, alternatively C8-12 alkyl, alternatively C8-9 alkyl, or alternatively C9 alkyl, which is optionally substituted with 1 R1s;R1s is independently H or C1-10 alkyl, alternatively H or C1-9 alkyl, alternatively C6-8 alkyl, alternatively C7-8 alkyl;R2 is C7-11 linear alkyl, alternatively C10-11 linear alkyl, alternatively C11 linear alkyl, which is optionally substituted with 1 C1-3 alkyl, alternatively optionally substituted with 1 methyl;R4 and R5 are independently C1-3 alkyl, alternatively methyl;or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively cyclopropylene;alternatively, R1 is
  • 8. The compound of formula (II) of claim 2, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,
  • 9. The compound of formula (II) of claim 8, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein, a=2;b=5 or 6, alternatively b=6;c=4, 5, or 6; alternatively c=4 or 6; alternatively c=6;d=0, 1, or 2; alternatively d=0 or 2;c+d=6;M1 is —C(O)O—, and M2 is —OC(O)O—;R1 is C8-11 alkyl, alternatively C9-10 alkyl, alternatively C9 alkyl, which is optionally substituted with 1 R1s;R1, is C7-9 alkyl, alternatively C7 alkyl;R2 is C8-11 linear alkyl, alternatively C9-10 linear alkyl, alternatively C9 linear alkyl;R4 and R5 are independently C1-3 alkyl, alternatively methyl;or, R4 and R5 are taken together with the carbon atom to which they are attached to form C3-4 cycloalkylene, alternatively not to form a ring;alternatively, R1 is
  • 10. The compound of formula (III) of claim 2, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,
  • 11. The compound of formula (II) of claim 2, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,
  • 12. The compound of formula (II) of claim 2, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof,
  • 13. A compound of formula (II) or formula (III), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof:
  • 14. The compound of formula (I) of claim 1, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein the compound is selected from the compounds in Table (I).
  • 15. A pharmaceutical composition, comprising the compound of claim 1, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof and a pharmaceutically acceptable excipient(s).
  • 16. A nanoparticle composition, comprising a lipid component, and optionally a load, wherein the lipid component comprises the compound of claim 1, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, and the load is selected from one or more of therapeutic, prophylactic, and diagnostic agents.
  • 17. The nanoparticle composition of claim 16, wherein the therapeutic, prophylactic, or diagnostic agents is a nucleic acid; alternatively, the nucleic acid is selected from one or more of ASO, RNA, and DNA;alternatively, the RNA is selected from one or more of small interfering RNA (siRNA), short hairpin RNA (shRNA), antisense RNA (aRNA), messenger RNA (mRNA), long non-coding RNA (lncRNA), microRNA (miRNA), small activating RNA (saRNA), multimeric coding nucleic acid (MCNA), polymeric coding nucleic acid (PCNA), guide RNA (gRNA), CRISPRRNA (crRNA), and nucleases, alternatively mRNA, more alternatively modified mRNA.
  • 18. A method for treating, diagnosing, or preventing a disease in a subject, comprising administering to the subject the nanoparticle composition of claim 16.
  • 19. A method for delivering a load in a subject, comprising administering to the subject the nanoparticle composition of claim 16; wherein the load is selected from one or more of therapeutic, prophylactic, and diagnostic agents.
  • 20. A method for preparing a compound of formula (II), comprising: reacting a compound of formula (IIA) with a compound of formula (IIB) to give the compound of formula (II),
Priority Claims (4)
Number Date Country Kind
202310720951.5 Jun 2023 CN national
202310723466.3 Jun 2023 CN national
202410115561.X Jan 2024 CN national
202410437163.X Apr 2024 CN national