POLYAMIDE COMPOUNDS, METHOD FOR PREPARING SAME, AND MEDICAL USE THEREOF

Information

  • Patent Application
  • 20250230129
  • Publication Number
    20250230129
  • Date Filed
    March 22, 2023
    2 years ago
  • Date Published
    July 17, 2025
    5 days ago
Abstract
The present invention relates to the field of medicines, particularly to synthetic polyamide compounds represented by formulas IA, IB, and IC, or pharmaceutically acceptable salts and stereoisomers thereof, a composition containing same, a method for preparing same, and use thereof in the field of medicines.
Description
TECHNICAL FIELD

The present invention relates to the field of medicines, particularly to a synthetic polyamide compound, or a pharmaceutically acceptable salt and a stereoisomer thereof, a composition comprising same, and use thereof in the field of medicines.


BACKGROUND

Opioid receptors are an important class of G protein-coupled receptors and are targets for binding to endogenous opioid peptides and opioid drugs. After being activated, opioid receptors play a regulatory role in nervous system immunity and endocrine system. Opioid drugs are the strongest and most commonly used central analgesics at present. Opioid receptors present in the central nervous system include μ, δ, κ receptors and the like.


κ-opioid receptor (KOR) is composed of 380 amino acids. It is expressed in sensory neurons, dorsal root ganglion cells, and primary afferent neuron terminals, and is involved in important physiological activities such as pain perception, neuroendocrine, emotional behavior, and cognition.


KOR agonists, as medicaments, have good application prospects in the pharmaceutical industry. For example, CN108290926A discloses a class of phenylpropanamide derivatives as KOR agonists, and CN 101627049 A also discloses a class of synthetic peptide amides as KOR agonists, wherein the compound D-Phe-D-Phe-D-Leu-D-Lys-[ω(4-aminopiperidine-4-carboxylic acid)]-OH (development code: CR845) has been clinically studied.


Although some KOR agonists already exist in the art, there is still a need for novel KOR agonists with improved activity and/or druggability.


SUMMARY

The present invention provides a novel κ-opioid receptor (KOR receptor) agonist compound, which surprisingly exhibits excellent effects and functions. In particular, such novel amide bond-containing compounds not only have excellent KOR receptor agonistic potency (high affinity for the κ-opioid receptor), but also have very good hydrophilicity and thus less ability to penetrate the blood brain barrier and lower ability to enter the brain. In some embodiments, the compounds of the present invention also have higher selectivity for the κ-opioid receptor than for the μ- and δ-opioid receptors. In some embodiments, the compounds of the present invention also have lower addiction, better physicochemical properties (e.g., solubility, physical and/or chemical stability), improved pharmacokinetic properties (e.g., lower inhibition of cytochrome P450 isoenzymes, improved bioavailability, suitable half-life and duration of action), improved safety (lower toxicity and/or fewer side effects (e.g., side effects on the central nervous system, respiratory depression, sedation, causing hallucinations, antidiuresis, nausea, constipation, dependence, etc.)), good patient compliance, and/or less susceptibility to developing tolerance, and other superior druggability properties. In some embodiments, the compounds of the present invention have improved safety, i.e., have lower acute toxicity and cardiotoxicity. In some embodiments, the compounds of the present invention have an improved safety window (e.g., have a greater safe dose range of administration, or are less likely to have side effects at the same dose). In some embodiments, the compounds of the present invention have improved pharmacokinetic properties (e.g., improved bioavailability and longer half-life, etc.).


One aspect of the present invention provides a compound of formula IA, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:




embedded image




    • wherein,

    • R1A is H or —COOH, preferably H; R2A is —NRaAC(═O)ORbA or —NRaAS(═O)2ORbA, preferably —NRaAC(═O)ORbA, wherein RaA is H or C1-6 alkyl substituted with one or more groups selected from amino, C1-6 alkylamino, C1-6 alkoxy, halogen, hydroxyl, nitro, cyano, NH2C(═O)—, and C1-6 alkoxy, preferably H or C1-6 alkyl substituted with amino, C1-6 alkylamino, or C1-6 alkoxy, and more preferably H or C1-6 alkyl substituted with amino or C1-6 alkylamino; RbA is selected from C1-6 alkyl, C6-14 aryl, 5- to 14-membered heteroaryl, C3-8 cycloalkyl, and 3- to 8-membered heterocyclyl, preferably from C1-6 alkyl and 3- to 8-membered heterocyclyl, more preferably from C1-6 alkyl and







embedded image


and even more preferably from C1-6 alkyl and




embedded image


the group RbA described above is optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, wherein ReA and RfA are each independently —(CH2)n1A— and —(CH2)n1A′—, n1A and n1A′ are each independently selected from 0, 1, 2, and 3, preferably 2, and n1A and n1A′ are not both 0; WA is selected from —NH—C(═O)—, —NH—S(═O)2—, —NR5A—, —O—, —S—, and —S(═O)2—, preferably from —O— and —S(═O)2—, R5A is selected from H, C1-6 alkyl, amidino, and HOOC—(CH2)n3A—, wherein n3A is selected from 1, 2, and 3;

    • or R1A and R2A, together with the carbon atom to which they are both attached form an optionally substituted 9- to 10-membered bicyclic moiety, preferably, the bicyclic moiety, together with the piperidine ring attached thereto, forms a structure selected from:




embedded image




    • when any one of Q1A-Q4A is N, the rest are C, or Q1A-Q4A are all C;

    • W1A and W2A are each independently —C(═O)—NH—, —NH—C(═O)—, —S(═O)2—NH—, —NH—S(═O)2—, —S—, —O—, —NR6A—, —NR6A—CH2—, —(CH2)n2A— optionally substituted with one or more groups selected from —NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, or absent, wherein R6A is selected from H, C1-6 alkyl, amidino, and HOOC—(CH2)n3A—; preferably, W1A and W2A cannot be simultaneously absent;

    • W3A is —(CH2)n2A— optionally substituted with one or more groups selected from NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy;

    • n2A is selected from 0, 1, 2, and 3;

    • more preferably, the bicyclic moiety, together with the piperidine ring attached thereto, forms a structure selected from:







embedded image


embedded image


more preferably from




embedded image


preferably from




embedded image




    • R3A is selected from H or —(CH2)mANRcARdA;

    • RcA and RdA are each independently selected from H, C1-6 alkyl, amidino, and C1-6 alkoxycarbonyl;

    • R4A is selected from halogen, NO2, C1-6 alkyl, C1-6 haloalkyl, cyano, NH2C(═O)—, and C1-6 alkoxy;

    • mA and nA are each independently 0, 1, 2, 3, 4, or 5.





Another aspect of the present invention provides a method for preparing the compound of the present invention, which is selected from the following methods:

    • method I:
    • method I comprises the following steps:




embedded image




    • method II:

    • method II comprises the following steps:







embedded image




    • wherein R1A′ is R1A or R1A in which NH2 is protected with an amino-protecting group, R2A′ is R2A or R2A in which NH2 is protected with an amino-protecting group, R3A′ is R3A or R3A in which NH2 is protected with an amino-protecting group, R1A, R2A, R3A, R4A, and nA are as defined above, and RxA is an amino-protecting group;

    • the amino-protecting groups are preferably each independently selected from tert-butoxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trichloroethoxycarbonyl, trimethylsilylethoxycarbonyl, benzyloxycarbonyl, p-toluenesulfonyl, p-nitrobenzenesulfonyl, tert-butyl, trifluoroacetyl, methoxycarbonyl, tert-butylsulfinyl, or ethoxycarbonyl.





The present invention further provides a compound of the following general formula, a stereoisomer thereof, or a salt thereof,




embedded image




    • wherein R1A′ is R1A or R1A in which NH2 is protected with an amino-protecting group, R2A′ is R2A or R2A in which NH2 is protected with an amino-protecting group, R3A′ is R3A or R3A in which NH2 is protected with an amino-protecting group, R1A, R2A, R3A, R4A, and nA are as defined above, and RxA is an amino-protecting group; the amino-protecting groups are preferably each independently selected from tert-butoxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trichloroethoxycarbonyl, trimethylsilylethoxycarbonyl, benzyloxycarbonyl, p-toluenesulfonyl, p-nitrobenzenesulfonyl, tert-butyl, trifluoroacetyl, methoxycarbonyl, tert-butylsulfinyl, or ethoxycarbonyl.





One aspect of the present invention provides a compound of formula IB, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:




embedded image




    • wherein,

    • R1B is selected from H, C1-6 alkyl (e.g., methyl), C1-6 alkylcarbonyl (e.g., methylcarbonyl), C1-6 alkoxycarbonyl (e.g., methoxycarbonyl), C6-14 aryl, C6-14 arylcarbonyl, C6-14 aryloxycarbonyl, C3-8 cycloalkyl, C3-8 cycloalkylcarbonyl, C3-8 cycloalkoxycarbonyl, 5- to 14-membered heteroaryl, 5- to 14-membered heteroarylcarbonyl, 5- to 14-membered heteroaryloxycarbonyl, 3- to 8-membered heterocyclyl, 3- to 8-membered heterocyclylcarbonyl, and 3- to 8-membered heterocyclyloxycarbonyl, preferably selected from H, C1-6 alkyl (e.g., methyl), C1-6 alkylcarbonyl (e.g., methylcarbonyl), and C1-6 alkoxycarbonyl (e.g., methoxycarbonyl), and more preferably selected from C1-6 alkyl (e.g., methyl) and C1-6 alkylcarbonyl (e.g., methylcarbonyl), the above substituents are each optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy;

    • R2B and R3B are each independently selected from H, C1-6 alkyl (e.g., methyl and isopropyl), amidino, and C1-6 alkoxycarbonyl (e.g., methoxycarbonyl);

    • R4B is selected from halogen, NO2, C1-6 alkyl, C1-6 haloalkyl, cyano, NH2C(═O)—, and C1-6 alkoxy;

    • mB and nB are each independently 0, 1, 2, 3, 4, or 5, for example, mB is 3, and/or nB is 0.





Another aspect of the present invention provides a method for preparing the compound of the present invention, which comprises the following steps:




embedded image




    • wherein, R1B, R4B, mB, and nB are as defined above, and RxB and RyB are amino-protecting groups, preferably independently selected from tert-butoxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trichloroethoxycarbonyl, trimethylsilylethoxycarbonyl, benzyloxycarbonyl, p-toluenesulfonyl, p-nitrobenzenesulfonyl, tert-butyl, trifluoroacetyl, methoxycarbonyl, or ethoxycarbonyl.





Another aspect of the present invention provides a compound of formula iB-1, a stereoisomer thereof, or a salt thereof,




embedded image




    • wherein R1B, R4B, mB, and nB are as defined above, and RxB and RyB are amino-protecting groups, preferably independently selected from tert-butoxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trichloroethoxycarbonyl, trimethylsilylethoxycarbonyl, benzyloxycarbonyl, p-toluenesulfonyl, p-nitrobenzenesulfonyl, tert-butyl, trifluoroacetyl, methoxycarbonyl, or ethoxycarbonyl.





One aspect of the present invention provides a compound of formula IC, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:




embedded image




    • wherein,

    • ring A is C3-8 cycloalkyl, C6-14 aryl, or 5- to 14-membered heteroaryl, preferably phenyl;

    • Y is selected from CH or N;

    • G is selected from —S—, —O—, —CR4CR5C—, —NR6C—, —S(═O)2—, —S(═O)(═NR6C′)—, and







embedded image


preferably —O—, —CR4CR5C—, —NR6C—, —S(═O)2—, —S(═O)(═NR6C′)—, and




embedded image




    • R1C is selected from H or —(CH2)tNRaCRbC;

    • R2C is selected from H, amino, hydroxyl, C1-6 alkyl, C1-6 alkylamino, and C1-6 aminoalkyl, wherein the alkyl is optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy;

    • when Y is CH, R3C is selected from H, hydroxyl, C1-6 alkyl, 3- to 8-membered heterocyclyl, and C1-6 alkoxy, and when Y is N, R3C is selected from H, C1-6 alkyl, C3-8 cycloalkyl, C3-8 cycloalkyl-(CH2)mC—, 3- to 8-membered heterocyclyl, 3- to 8-membered heterocyclyl-(CH2)mC—, and —(CH2)mCNR10R11, wherein the alkyl, cycloalkyl, heterocyclyl, and alkoxy are optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, C1-6 alkoxy, and C1-6 alkylamino;

    • R4C and R5C are each independently selected from H, C1-6 alkyl, C1-6 alkyl-O—, hydroxyl, —C(O)OR7, —NR8R9, —NRcCC(O)NR8R9, C1-6 alkylamino, 3- to 8-membered heterocyclyl-(CH2)mC—, halogen, cyano, —NRcCS(═O)2NR8R9, —NRcCC(O)ORdC, —NRcCS(═O)2ORdC, —NRcCC(O)R7′, and —NH(CH2)mCNR8R9, wherein the alkyl and heterocyclyl are optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy;

    • or CR4CR5C forms a 3- to 8-membered heterocyclic or 9- to 10-membered bicyclic moiety, wherein the 3- to 8-membered heterocyclic or 9- to 10-membered bicyclic moiety, together with the piperidine ring attached thereto, forms a structure selected from:







embedded image




    • when any one of Q1C-Q4C is N, the rest are C, or Q1C-Q4C are all C;

    • W1C and W2C are each independently —C(═O)—NH—, —NH—C(═O)—, —S(═O)2—NH—, —NH—S(═O)2—, —S—, —O—, —NR12—, —NR12—CH2—, —(CH2)n2C— optionally substituted with one or more groups selected from —NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, or absent, R12 is selected from H, C1-6 alkyl, amidino, and HOOC—(CH2)n3C—; preferably, W1C and W2C cannot be simultaneously absent;

    • W3C is —(CH2)n2C— optionally substituted with one or more groups selected from NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, or absent; preferably —(CH2)n2C— optionally substituted with one or more groups selected from NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy;

    • W4C and W5C are —(CH2)n2C— optionally substituted with one or more groups selected from NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, or absent;

    • n2C is selected from 0, 1, 2, and 3;

    • more preferably, the 3- to 8-membered heterocyclic or 9- to 10-membered bicyclic moiety, together with the piperidine ring attached thereto, forms a structure selected from:







embedded image


embedded image


more preferably from




embedded image




    • RaC and RbC are each independently selected from H, C1-6 alkyl, amidino, and C1-6 alkoxycarbonyl;

    • RcC is H or C1-6 alkyl substituted with one or more groups selected from amino, C1-6 alkylamino, C1-6 alkoxy, halogen, hydroxyl, nitro, cyano, NH2C(═O)—, and C1-6 alkoxy, preferably H or C1-6 alkyl substituted with amino, C1-6 alkylamino, or C1-6 alkoxy, and more preferably H or C1-6 alkyl substituted with amino or C1-6 alkylamino;

    • RdC is selected from C1-6 alkyl, C6-14 aryl, 5- to 14-membered heteroaryl, C3-8 cycloalkyl, and 3- to 8-membered heterocyclyl, preferably from C1-6 alkyl and 3- to 8-membered heterocyclyl, more preferably from C1-6 alkyl and







embedded image


and even more preferably from C1-6 alkyl and




embedded image


the group RdC described above is optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, wherein ReC and RfC are each independently —(CH2)n1C— and —(CH2)n1C′—, wherein n1C and n1C′ are each independently selected from 0, 1, 2, and 3, preferably 2, and n1C and n1C′ are not both 0; WC is selected from —NH—C(═O)—, —NH—S(═O)2—, —NR12—, —O—, —S—, and —S(═O)2—, preferably from —O— and —S(═O)2—, wherein R12 is selected from H, C1-6 alkyl, amidino, and HOOC—(CH2)n3C—, wherein n3C is selected from 1, 2, and 3;

    • R6C and R6C′ are independently selected from H, C1-6 alkyl, C1-6 alkylcarbonyl, C1-6 alkyl-S(═O)2—, C1-6 alkoxycarbonyl, C6-14 aryl, C6-14 arylcarbonyl, C6-14 aryloxycarbonyl, C3-8 cycloalkyl, C3-8 cycloalkylcarbonyl, C3-8 cycloalkoxycarbonyl, 5- to 14-membered heteroaryl, 5- to 14-membered heteroarylcarbonyl, 5- to 14-membered heteroaryloxycarbonyl, 3- to 8-membered heterocyclyl, 3- to 8-membered heterocyclylcarbonyl, and 3- to 8-membered heterocyclyloxycarbonyl, the above substituents are each optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy;
    • R7 and R7′ are each independently selected from H, C1-6 alkyl, C3-8 cycloalkyl, 3- to 8-membered heterocyclyl, C6-14 aryl, and 5- to 14-membered heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy;
    • R8 and R9 are each independently H or C1-6 alkyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy; R10 and R11 are each independently H or C1-6 alkyl, or R10 and R11, together with the nitrogen atom to which they are both attached form 3- to 8-membered heterocyclyl, wherein the alkyl and heterocyclyl are optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy;
    • p and t are each independently selected from 0, 1, 2, 3, 4, or 5;
    • mC is independently selected from 1, 2, 3, and 4 at each occurrence;
    • R0 is selected from H, halogen, NO2, cyano, NH2C(═O)—, C1-6 alkoxy, and C1-6 alkyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy.


Another aspect of the present invention provides a method for preparing the compound of the present invention, which is selected from the following methods:

    • method I:
    • method I comprises the following steps:




embedded image




    • method II:

    • method II comprises the following steps:







embedded image


Preferably, a compound of formula iiC-2 is prepared by a method selected from:

    • method III:
    • method III comprises the following steps: subjecting a compound of formula iiC-4-a and a compound of formula iiC-5-a to a condensation reaction, a hydrolysis reaction, an optional removal reaction of an amino-protecting group, and an amino protection reaction to give the compound of formula iiC-2,




embedded image




    • method IV:

    • method IV comprises the following steps: subjecting a compound of formula iiC-4-b and a compound of formula iiC-5-b to a condensation reaction, a hydrolysis reaction, an optional removal reaction of an amino-protecting group, and an amino-protection reaction to give the compound of formula iiC-2







embedded image




    • method V:

    • method V comprises the following steps: subjecting a compound of formula iiC-4-c and a compound of formula iiC-5-c to a condensation reaction, a hydrolysis reaction, an optional removal reaction of an amino-protecting group, and an amino protection reaction to give the compound of formula iiC-2,







embedded image




    • method VI:

    • method VI comprises the following steps: subjecting a compound of formula iiC-4-d and a compound of formula iiC-5-d to a condensation reaction, a hydrolysis reaction, an optional removal reaction of an amino-protecting group, and an amino protection reaction to give the compound of formula iiC-2,







embedded image




    • preferably, the formula iC-2







embedded image




    • preferably, iiC-2 is selected from:







embedded image


embedded image




    • wherein R0′ is R0 or R0 in which NH2 is protected with an amino-protecting group, R1C′ and R1C″ are R1C or R1C in which NH2 is protected with an amino-protecting group, R2C′ and R2C″ are R2C or R2C in which NH2 is protected with an amino-protecting group, R3C′ and R3C″ are R3C or R3C in which NH2 or the cyclic imino is protected with an amino-protecting group, G′ is G or G in which NH2 and/or the cyclic imino is protected with an amino-protecting group and/or the carboxyl is protected with a carboxyl-protecting group, Rw is a carboxyl-protecting group, ring A, Y, R0, R1C, R2C, R3C, G, and p are as defined above, and Rs, Rt, and Ru are each independently selected from H and amino-protecting groups;

    • preferably, the amino-protecting groups are each independently selected from tert-butoxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trichloroethoxycarbonyl, trimethylsilylethoxycarbonyl, benzyloxycarbonyl, p-toluenesulfonyl, p-nitrobenzenesulfonyl, tert-butyl, trifluoroacetyl, methoxycarbonyl, tert-butylsulfinyl, 1-phenylethyl, or ethoxycarbonyl; the carboxyl-protecting groups are each independently selected from C1-6 alkyl, allyl, benzyl, 2,4-dimethoxybenzyl, p-methoxybenzyl, methoxyethoxymethyl, pentafluorophenyl, and 4-p-methylbenzyloxybenzyl.





Another aspect of the present invention provides a compound of the following general formula, a stereoisomer thereof, or a salt thereof,




embedded image




    • preferably, formula iiC-2-X is selected from:







embedded image


embedded image




    • wherein R0′ is R0 or R0 in which NH2 is protected with an amino-protecting group, R1C is R1C or R1C in which NH2 is protected with an amino-protecting group, R2C′ is R2C or R2C in which NH2 is protected with an amino-protecting group, R3C′ is R3C or R3C in which NH2 or the cyclic imino is protected with an amino-protecting group, G′ is G or G in which NH2 and/or the cyclic imino is protected with an amino-protecting group and/or the carboxyl is protected with a carboxyl-protecting group, ring A, Y, R0, R1C, R2C, R3C, G, and p are as defined above, Rs and Rt are each independently selected from H and amino-protecting groups, and Rv is selected from H and carboxyl-protecting groups;

    • preferably, the amino-protecting groups are each independently selected from tert-butoxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trichloroethoxycarbonyl, trimethylsilylethoxycarbonyl, benzyloxycarbonyl, p-toluenesulfonyl, p-nitrobenzenesulfonyl, tert-butyl, trifluoroacetyl, methoxycarbonyl, tert-butylsulfinyl, 1-phenylethyl, or ethoxycarbonyl; the carboxyl-protecting groups are each independently selected from C1-6 alkyl, allyl, benzyl, 2,4-dimethoxybenzyl, p-methoxybenzyl, methoxyethoxymethyl, pentafluorophenyl, and 4-p-methylbenzyloxybenzyl.





Another aspect of the present invention provides a pharmaceutical composition comprising a prophylactically or therapeutically effective amount of the compound of the present invention, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient, as well as optionally other therapeutic agents.


Another aspect of the present invention provides use of the compound of the present invention, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof, or the pharmaceutical composition, in the preparation of a medicament for agonizing κ-opioid receptors.


Another aspect of the present invention provides a method for preventing and/or treating a related disease mediated by κ-opioid receptor, comprising administering an effective amount of the compound of the present invention, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof, or the pharmaceutical composition.


Another aspect of the present invention provides the use of the compound of the present invention, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof, or the pharmaceutical composition, in the preparation of a medicament, particularly, the medicament is used for preventing and/or treating a related disease mediated by κ-opioid receptor.


The related disease mediated by the κ-opioid receptor in the present disclosure is selected from pain, inflammation, pruritus, edema, hyponatremia, hypokalemia, ileus, cough, and glaucoma, preferably pain. The pain disclosed herein is selected from neuropathic pain, trunk pain, visceral pain, skin pain, arthritis pain, kidney stone pain, uterine cramp, dysmenorrhea, endometriosis, dyspepsia, post-surgical pain, post-medical treatment pain, ocular pain, otitis pain, breakthrough cancer pain, and pain associated with a GI disorder.







DETAILED DESCRIPTION
Terminology

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by those skilled in the art to which the claimed subject matter belongs.


Unless otherwise stated, the present invention employs conventional methods of mass spectrometry, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques, pharmacology, etc. within the skill of the art. Unless specific definitions are provided, nomenclature and laboratory operations and techniques related to the chemistry, such as analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry, described herein are known to those skilled in the art. In general, the foregoing techniques and procedures can be performed by conventional methods that are well known in the art and described in various general and more specific documents, and these documents are cited and discussed in the present specification.


The term “alkyl” refers to an aliphatic hydrocarbon group, which may be branched or linear alkyl. According to the structure, the alkyl may be a monovalent group or a divalent group (i.e., alkylene). In the present invention, the alkyl is preferably alkyl having 1 to 8 carbon atoms, more preferably “lower alkyl” having 1 to 6 carbon atoms, and even more preferably alkyl having 1 to 4 carbon atoms. Typical alkyl groups include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, and the like. It should be understood that the “alkyl” mentioned herein includes all possible configurations and conformations of the alkyl. For example, the “propyl” mentioned herein includes n-propyl and isopropyl, “butyl” includes n-butyl, isobutyl, and tert-butyl, “pentyl” includes n-pentyl, isopropyl, neopentyl, tert-pentyl, and pent-3-yl, and the like. In some embodiments, the alkyl is optionally substituted with one or more (such as 1 to 3) suitable substituents.


The term “aryl” refers to an all-carbon monocyclic or polycyclic aromatic group having a conjugated π-electron system. For example, the term “C6-14 aryl” as used herein refers to an aromatic group containing 6 to 14 carbon atoms. In some embodiments, the C6-14 aryl is preferably C6-10 aryl. Examples of aryl groups may include phenyl, naphthyl, anthryl, and the like. In some embodiments, the aryl is optionally substituted with one or more (such as 1 to 3) suitable substituents.


The term “heteroaryl” refers to a monocyclic or polycyclic aromatic group containing one or more identical or different heteroatoms, such as oxygen, nitrogen, or sulfur. For example, the term “5- to 14-membered heteroaryl” as used herein refers to heteroaryl having 5 to 14 ring atoms. Further, the heteroaryl refers to a monocyclic, bicyclic, or tricyclic aromatic group, which has 5, 6, 8, 9, 10, 11, 12, 13, or 14 ring atoms, in particular 1 or 2 or 3 or 4 or 5 or 6 or 9 or 10 carbon atoms, and contains at least one heteroatom that may be identical or different, such as oxygen, nitrogen, or sulfur. In addition, the heteroaryl may be a benzo-fused group. In particular, the heteroaryl is selected from thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, etc., and benzo derivatives thereof; or pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, etc., and benzo derivatives thereof. In some embodiments, the heteroaryl is optionally substituted with one or more (such as 1 to 3) suitable substituents.


The term “cycloalkyl” refers to a saturated or unsaturated non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring group (e.g., monocyclic rings such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and cyclononyl, or bicyclic rings, including spiro, fused, or bridged systems, such as bicyclo[1.1.1]pentyl, bicyclo[2.2.1]heptyl, bicyclo[3.2.1]octyl, or bicyclo[5.2.0]nonyl, decahydronaphthyl, etc.). Non-limiting examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc. In some embodiments, the cycloalkyl is optionally substituted with one or more (such as 1 to 3) suitable substituents.


The term “heterocyclyl” refers to a saturated or unsaturated monocyclic or polycyclic group, which has, for example, 3 to 12 ring atoms or 3 to 8 ring atoms in the ring, the ring atoms including carbon atoms and one or more (e.g., one, two, three, or four) heteroatoms selected from nitrogen, oxygen, and sulfur; the heterocyclyl may be linked to the rest of a molecule via any one of the carbon atoms or the nitrogen atom (if present). The carbon atom in the heterocyclyl may be optionally substituted with oxo (═O). The sulfur atom of the ring may be optionally oxidized to an S-oxide, such as SO or SO2. The nitrogen atom of the ring may be optionally oxidized to an N-oxide. For example, 3- to 8-membered heterocyclyl is a group having 3 to 8 ring atoms including carbon atoms and heteroatoms, including, but not limited to, oxiranyl, aziridinyl, azetidinyl, oxetanyl, thietanyl, tetrahydrofuranyl, dioxolyl, pyrrolidinyl, pyrrolidonyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, tetrahydropyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, pyrimidindionyl, 2-oxopyrrolidinyl, 3,5-dioxopiperidinyl, sulfolanyl, 1,1-dioxothiomorpholinyl, and 1,1-dioxotetrahydrothiopyranyl. In some embodiments, the heterocyclyl is optionally substituted with one or more (such as 1 to 3) suitable substituents. In some embodiments, the 3- to 8-membered heterocyclyl is 3- to 8-membered heterocyclyl containing one heteroatom selected from oxygen, nitrogen, and sulfur; in some embodiments, the 3- to 8-membered heterocyclyl




embedded image


wherein WA, ReA, RfA, WC, ReC, and RfC are as defined in the present specification.


The term “alkoxy” refers to alkyl-O—, wherein the alkyl is as defined herein. Typical alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, pentyloxy, hexyloxy, and the like.


The term “aryloxy” refers to aryl-O—, wherein the aryl is as defined herein.


The term “heteroaryloxy” refers to heteroaryl-O—, wherein the heteroaryl is as defined herein.


The term “cycloalkoxy” refers to cycloalkyl-O—, wherein the cycloalkyl is as defined herein.


The term “heterocyclyloxy” refers to heterocyclyl-O—, wherein the heterocyclyl is as defined herein.


The term “carbonyl” refers to an organic functional group, —C(═O)—, which is formed by linking two atoms, carbon and oxygen, via a double bond. The term “alkoxycarbonyl” refers to alkoxy-C(═O)—, i.e., the group is linked to the rest of a compound via carbonyl. The term “alkylcarbonyl” refers to alkyl-C(═O)—, i.e., the group is linked to the rest of a compound via carbonyl. Similarly, the term “arylcarbonyl” refers to aryl-C(═O)—, i.e., the group is linked to the rest of a compound via carbonyl; the term “aryloxycarbonyl” refers to aryloxy-C(═O)—, i.e., the group is linked to the rest of a compound via carbonyl; and so on.


The term “cyano” refers to a group, —CN, where a carbon atom and a nitrogen atom are linked via a triple bond.




embedded image


The term “amidino” refers to


The term “halo-” or “halogen” refers to fluorine, chlorine, bromine, and iodine.


The term “haloalkyl” refers to alkyl in which at least one hydrogen is replaced by a halogen atom. In certain embodiments, if two or more hydrogen atoms are replaced by halogen atoms, the halogen atoms are identical or different from each other. Examples of the haloalkyl include, but are not limited to, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, difluoroethyl, trifluoroethyl, and the like.


The term “aminoalkyl” refers to alkyl substituted with amino.


The term “alkylamino” refers to amino substituted with alkyl.


The expression “C1-C6” or “C1-6” encompasses a range of 1 to 6 carbon atoms and should be understood as also encompassing any sub-range therein as well as each point value, including, for example, C1-C6, C2-C6, C3-C6, C4-C6, C5-C6, C1-C5, C2-C5, C3-C5, C4-C5, C1-C4, C2-C4, C3-C4, C1-C3, C2-C3, C1-C2, etc., as well as C1, C2, C3, C4, C5, C6, etc. Similarly, the expression “C3-C8” or “C3-8” encompasses a range of 3 to 8 carbon atoms and should be understood as also encompassing any sub-range therein as well as each point value, including, for example, C3-C6, etc., as well as C3, C4, C5, C6, C7, C8, etc. The expression “3- to 8-membered” should be understood as encompassing any sub-range therein as well as each point value, for example, 3- to 7-membered, 3- to 6-membered, 3- to 5-membered, and 4- to 8-membered, as well as 3-membered, 4-membered, 5-membered, 6-membered, 7-membered, 8-membered, etc. Similarly, the expression “5- to 14-membered” should be understood as encompassing any sub-range therein as well as each point value, for example, 5-membered, 6-membered, 7-membered, 8-membered, 9-membered, 10-membered, 11-membered, 12-membered, 13-membered, 14-membered, etc. Other similar expressions herein should also be understood in a similar manner.


The term “optionally substituted” or “optionally substituted with . . . ” means that the group mentioned may or may not be substituted.


The term “optionally substituted” or “substituted” means that the group mentioned may be substituted with one or more additional groups, and the additional groups are each independently selected from, for example, alkyl, alkenyl, alkynyl, haloalkyl, alkoxy, alkylthio, halogen, sulfhydryl, hydroxyl, nitro, amino, cyano, carboxyl, oxo, cycloalkyl, heterocyclyl, aryl, heteroaryl, NH2C(═O)—, alkylamino, and the like.


The expression “one or more” in “substituted with one or more groups selected from . . . ” or “optionally substituted with one or more groups selected from . . . ” refers to 1 to 5, or 1 to 4, or 1 to 3, or 1 to 2 group substitutions, provided that the valence requirements are met.


In addition, it should be understood that the terms “C6-14 arylcarbonyl” and “C6-14 aryloxycarbonyl” refer to a group in which the aryl is C6-14 aryl; the terms “C3-8 cycloalkylcarbonyl” and “C3-8 cycloalkyloxycarbonyl” refer to a group in which the cycloalkyl is C3-8 cycloalkyl; the terms “5- to 14-membered heteroarylcarbonyl” and “5- to 14-membered heteroaryloxycarbonyl” refer to a group in which the heteroaryl is 5- to 14-membered heteroaryl; the terms “3- to 8-membered heterocyclylcarbonyl” and “3- to 8-membered heterocyclyloxycarbonyl” refer to a group in which the heterocyclyl is 3- to 8-membered heterocyclyl; and the term “C1-6 alkoxycarbonyl” refers to a group in which the alkyl is C1-6 alkyl. Similarly, “C1-6 aminoalkyl” refers to a group in which the alkyl is C1-6 alkyl.


The term “pharmaceutically acceptable carrier” refers to those substances that do not have a significant irritating effect on organisms and do not impair the biological activity and properties of the active compound. The “pharmaceutically acceptable carrier” includes, but is not limited to, a glidant, a sweetener, a diluent, a preservative, a dye colorant, a flavoring agent, a surfactant, a wetting agent, a dispersant, a disintegrant, a stabilizer, a solvent, or an emulsifier.


The names of the compounds of the present disclosure, “compound *-0” and “compound *-100” (wherein * is the compound number), are stereoisomers of “compound *”, respectively. For example, compound 3 is a racemate, and compounds 3-0 and 3-100 are stereoisomers thereof, respectively.


“*” in the present invention refers to a linking site. For example, when G of the present disclosure is




embedded image


it indicates that




embedded image


is




embedded image


In addition, it should be understood that the expression “G is selected from —CR4CR5C—” means that the C atom is a ring atom, i.e.,




embedded image


is




embedded image


similarly, “G is selected from —S(═O)(═NR6C′)—” means that the S atom is a ring atom, i.e.,




embedded image


is




embedded image


It should also be understood that the structure




embedded image


mentioned in the present disclosure refers to an aromatic ring, for example, when any one of Q1A/Q1C-Q4A/Q4C is N and the rest are C, the structure is a pyridine ring, and when Q1A/Q1C-Q4A/Q4C are all C, the structure is a benzene ring. It should also be understood that the expression “the rest are C” in the description described above means that the ring atoms are C, and in practice, means that “the rest are CH” according to valence bond theory.


The compounds of the present invention or salts thereof may contain one or more stereogenic centers, and each stereogenic center is independently present in an R or S configuration, and thus, enantiomers, diastereomers, and other stereoisomeric forms may be generated. These forms may be defined, in terms of absolute stereochemistry, as (R)- or (S)-, or as (D)- or (L)- for amino acids. The present invention is intended to encompass all such possible isomers or mixtures thereof, e.g., substantially pure enantiomers, diastereomers, racemates, or mixtures thereof. In certain embodiments, preferred compounds are those isomer compounds that exhibit superior biological activity. Purified or partially purified isomers and stereoisomers, or racemic or diastereomeric mixtures of the compounds of the present invention are also encompassed within the scope of the present invention. Purification and separation of such materials may be accomplished by standard techniques known in the art.


In some embodiments, the compounds of the present disclosure are racemic. In some embodiments, the compounds of the present disclosure are single enantiomers. In some embodiments, the compounds of the present disclosure are substantially free of other isomers. In some embodiments, the compounds of the present disclosure are single isomers substantially free of other isomers. In some embodiments, the compounds of the present disclosure comprise 25% or less of other isomers, or comprise 20% or less of other isomers, or comprise 15% or less of other isomers, or comprise 10% or less of other isomers, or comprise 5% or less of other isomers, or comprise 1% or less of other isomers.


In some embodiments, the compounds of the present disclosure have a stereochemical purity of at least 75%, or have a stereochemical purity of at least 80%, or have a stereochemical purity of at least 85%, or have a stereochemical purity of at least 90%, or have a stereochemical purity of at least 95%, or have a stereochemical purity of at least 96%, or have a stereochemical purity of at least 97%, or have a stereochemical purity of at least 98%, or have a stereochemical purity of at least 99%.


In some embodiments, asymmetric carbon atoms in the compounds of the present disclosure may all be present in racemic or enantiomerically enriched forms, such as an (R)-, (S)-, or (R,S)-configuration. In certain embodiments, the asymmetric carbon atom in the (S)- or (R)-configuration in the compound of the present disclosure has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess.


As used herein, “substantially pure” or “substantially free of other isomers” means that the product contains, by weight, less than 10%, preferably less than 5%, preferably less than 4%, preferably less than 3%, preferably less than 2%, or preferably less than 1% of other isomers relative to the preferred isomer. It should be understood that when the present disclosure mentions or indicates that the configuration of a compound is an absolute configuration, it means that the compound is in the substantially pure absolute configuration; similarly, when the present disclosure mentions or indicates that a compound is a single enantiomer, it means that the compound is in the substantially pure enantiomer form.


The following detailed description is intended to illustrate non-limiting embodiments and to enable others skilled in the art to more fully understand the technical schemes of the present invention, its principles, and its practical applications.


Compound

The present disclosure provides a compound of formula IA, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:




embedded image


In some embodiments, R1A is H or —COOH, preferably H; R2A is —NRaAC(═O)ORbA or —NRaAS(═O)2ORbA, preferably —NRaAC(═O)ORbA, wherein RaA is H or C1-6 alkyl substituted with one or more groups selected from amino, C1-6 alkylamino, C1-6 alkoxy, halogen, hydroxyl, nitro, cyano, NH2C(═O)—, and C1-6 alkoxy, preferably H or C1-6 alkyl substituted with amino, C1-6 alkylamino, or C1-6 alkoxy, and more preferably H or C1-6 aminoalkyl; RbA is selected from C1-6 alkyl, C6-14 aryl, 5- to 14-membered heteroaryl, C3-8 cycloalkyl, and 3- to 8-membered heterocyclyl (preferably




embedded image


preferably from C1-6 alkyl and 3- to 8-membered heterocyclyl, more preferably from C1-6 alkyl and




embedded image


more preferably from C1-6 alkyl and




embedded image


and even more preferably from C1-6 alkyl and




embedded image


the group RbA described above is optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, wherein ReA and RfA are each independently —(CH2)n1A— and —(CH2)n1A′—, and in some embodiments, are —(CH2)n1A— and —(CH2)n1A′—, respectively, wherein n1A and n1A′ are each independently selected from 0, 1, 2, and 3, preferably 2, and n1A and n1A′ are not both 0; WA is selected from —NH—C(═O)—, —NH—S(═O)2—, —NR5A—, —O—, —S—, and —S(═O)2—, preferably from —O— and —S(═O)2—, wherein RaA is selected from H, C1-6 alkyl, amidino, and HOOC—(CH2)n3A—, wherein n3A is selected from 1, 2, and 3. In some embodiments, RaA is H. In some embodiments, RaA is C1-6 alkyl substituted with amino, C1-6 alkylamino, or C1-6 alkoxy, such as methoxyethyl, methylaminoethyl, aminopropyl, aminoethyl, or the like. In some embodiments, RaA is H or C1-6 aminoalkyl. In some embodiments, RbA is C1-6 alkyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, such as methyl, ethyl, or propyl, preferably methyl.


In some embodiments, RbA is




embedded image


optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, wherein WA is selected from —NH—C(═O)—, —NH—S(═O)2—, —NR5A—, —O—, —S—, and —S(═O)2—, preferably from —O— and —S(═O)2—; R5A is selected from H, C1-6 alkyl, amidino, and HOOC—(CH2)n3A—, wherein n3A is selected from 1, 2, and 3. In some embodiments, R1A and R2A, together with the carbon atom to which they are both attached form an optionally substituted 9- to 10-membered bicyclic moiety (specifically a 9- to 10-membered fused ring). Preferably, the bicyclic moiety, together with the piperidine ring attached thereto, forms a structure selected from:




embedded image




    • when any one of Q1A-Q4A is N, the rest are CH, or Q1A-Q4A are all CH,

    • W1A and W2A are each independently —C(═O)—NH—, —NH—C(═O)—, —S(═O)2—NH—, —NH—S(═O)2—, —S—, —O—, —NR6A—, —NR6A—CH2—, —(CH2)n2A— optionally substituted with one or more groups selected from —NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, or absent, wherein R6A is selected from H, C1-6 alkyl, amidino, and HOOC—(CH2)n3A—, n3A is selected from 1, 2, and 3, preferably 1; preferably, W1A and W2A cannot be simultaneously absent;

    • W3A is —(CH2)n2A— optionally substituted with one or more groups selected from NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, wherein n2A is selected from 0, 1, 2, and 3.





More preferably, the bicyclic moiety, together with the piperidine ring attached thereto, forms a structure selected from:




embedded image


embedded image


more preferably from




embedded image


and even more preferably from




embedded image


In some embodiments, R3A is selected from H or —(CH2)mANRcAR1A, wherein RcA and RdA are each independently selected from H, C1-6 alkyl (e.g., methyl, isopropyl, etc.), amidino, and C1-6 alkoxycarbonyl (e.g., methoxycarbonyl, etc.), and mA is selected from 0, 1, 2, 3, 4, or 5. In some embodiments, R3A is H. In some embodiments, RcA and RdA are each independently selected from H and C1-6 alkoxycarbonyl (e.g., methoxycarbonyl, etc.). In some embodiments, RcA and RdA are each H. In some embodiments, RcA is H, and RdA is C1-6 alkoxycarbonyl, preferably methoxycarbonyl. In some embodiments, mA is selected from 0, 1, 2, or 3, preferably 3.


In some embodiments, R4A is selected from halogen, NO2, C1-6 alkyl, C1-6 haloalkyl, cyano, NH2C(═O)—, and C1-6 alkoxy, preferably from F, Cl, NO2, CH3, CF3, cyano, and NH2C(═O)—.


In some embodiments, nA is 0, 1, 2, 3, 4, or 5, preferably 0, 1, 2, or 3, and more preferably 0.


In some embodiments, formula IA is as shown in formula IIA, preferably formula IIIA, even more preferably formula IVA, and still more preferably formula VA:




embedded image


Preferably, RbA in formula IA, formula IIA, formula IIIA, formula IVA, or formula VA is selected from C1-6 alkyl and




embedded image


preferably




embedded image


wherein WA is selected from —O— and —S(═O)2—.


In some embodiments, formula IA is as shown in formula VIA:




embedded image


Preferably, RaA in formula VIA is H or C1-6 alkyl substituted with amino, C1-6 alkylamino, or C1-6 alkoxy, more preferably H or C1-6 aminoalkyl.


In some embodiments, the compound of the present invention is a compound of formula IIIA, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein RaA is H; RbA is selected from C1-6 alkyl,




embedded image


ReA and RfA are —(CH2)n1A— and —(CH2)n1A′—, respectively, n1A and n1A′ are each independently selected from 0, 1, 2, and 3, preferably 2, and n1A and n1A′ are not both 0; WA is selected from —NH—C(═O)—, —NH—S(═O)2—, —NR5A—, —O—, —S—, and —S(═O)2—, wherein R5A is selected from H, C1-6 alkyl, amidino, and HOOC—(CH2)n3A—, wherein n3A is selected from 1, 2, and 3; RcA and RdA are each independently selected from H, C1-6 alkyl, amidino, and C1-6 alkoxycarbonyl.


In some embodiments, the compound of the present invention is a compound of formula IIIA, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein RaA is H; RbA is selected from C1-6 alkyl and




embedded image


wherein WA is selected from —O— and —S(═O)2—; RcA and RbA are each independently selected from H, C1-6 alkyl, amidino, and C1-6 alkoxycarbonyl.


In some embodiments, the compound of the present invention is a compound of formula IVA, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein RaA is H; RbA is selected from C1-6 alkyl and




embedded image


wherein WA is selected from —O— and —S(═O)2—.


In some embodiments, the compound of the present invention is selected from compounds shown below, stereoisomers thereof, or pharmaceutically acceptable salts thereof:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


The present disclosure further provides a compound of the following general formula, a stereoisomer thereof, or a salt thereof, which is an intermediate for preparing the compounds of the present disclosure described above,




embedded image




    • wherein, R1A′ is R1A or R1A in which NH2 is protected with an amino-protecting group, R2A′ is R2A or R2A in which NH2 is protected with an amino-protecting group, R3A′ is R3A or R3A in which NH2 is protected with an amino-protecting group, R1A, R2A, R3A, R4A, and nA are as defined above, and RxA is an amino-protecting group;

    • the amino-protecting groups are preferably each independently selected from tert-butoxycarbonyl (Boc), 9-fluorenylmethoxycarbonyl (Fmoc), allyloxycarbonyl (Alloc), trichloroethoxycarbonyl (Troc), trimethylsilylethoxycarbonyl (Teoc), benzyloxycarbonyl (CBz), p-toluenesulfonyl (Tosyl), p-nitrobenzenesulfonyl (Nosyl), tert-butyl (t-Bu), trifluoroacetyl (Tfa), methoxycarbonyl, tert-butylsulfinyl, or ethoxycarbonyl.





Preparation Method

In some embodiments, the compound of the present invention may be prepared by a method selected from:

    • method I:
    • method I comprises the following steps:




embedded image




    • method II:

    • method II comprises the following steps:







embedded image




    • wherein, R1A′ is R1A or R1A in which NH2 is protected with an amino-protecting group, R2A′ is R2A or R2A in which NH2 is protected with an amino-protecting group, R3A′ is R3A or R3A in which NH2 is protected with an amino-protecting group, R1A, R2A, R3A, R4A, and nA are as defined above, and RxA is an amino-protecting group;

    • the amino-protecting groups are preferably each independently selected from tert-butoxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trichloroethoxycarbonyl, trimethylsilylethoxycarbonyl, benzyloxycarbonyl, p-toluenesulfonyl, p-nitrobenzenesulfonyl, tert-butyl, trifluoroacetyl, methoxycarbonyl, tert-butylsulfinyl, or ethoxycarbonyl.





Compound

The present disclosure provides a compound of formula IB, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:




embedded image




    • R1B is selected from H, C1-6 alkyl (e.g., methyl), C1-6 alkylcarbonyl (e.g., methylcarbonyl), C1-6 alkoxycarbonyl (e.g., methoxycarbonyl), C6-14 aryl, C6-14 arylcarbonyl, C6-14 aryloxycarbonyl, C3-8 cycloalkyl, C3-8 cycloalkylcarbonyl, C3-8 cycloalkoxycarbonyl, 5- to 14-membered heteroaryl, 5- to 14-membered heteroarylcarbonyl, 5- to 14-membered heteroaryloxycarbonyl, 3- to 8-membered heterocyclyl, 3- to 8-membered heterocyclylcarbonyl, and 3- to 8-membered heterocyclyloxycarbonyl, preferably from H, C1-6 alkyl (e.g., methyl), C1-6 alkylcarbonyl (e.g., methylcarbonyl), and C1-6 alkoxycarbonyl (e.g., methoxycarbonyl), and more preferably from C1-6 alkyl (e.g., methyl) and C1-6 alkylcarbonyl (e.g., methylcarbonyl), the above substituents are each optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy;

    • R2B and R3B are each independently selected from H, C1-6 alkyl (e.g., methyl and isopropyl), amidino, and C1-6 alkoxycarbonyl (e.g., methoxycarbonyl);

    • R4B is selected from halogen, NO2, C1-6 alkyl, C1-6 haloalkyl, cyano, NH2C(═O)—, and C1-6 alkoxy, preferably from F, Cl, NO2, CH3, CF3, cyano, and NH2C(═O)—;

    • mB and nB are each independently 0, 1, 2, 3, 4, or 5, for example, mB is 3, and/or nB is 0.





In some embodiments, R1B is selected from H, C1-6 alkyl, C1-6 alkylcarbonyl, C1-6 alkoxycarbonyl, C6-14 aryl, C6-14 arylcarbonyl, C6-14 aryloxycarbonyl, C3-8 cycloalkyl, C3-8 cycloalkylcarbonyl, C3-8 cycloalkoxycarbonyl, 5- to 14-membered heteroaryl, 5- to 14-membered heteroarylcarbonyl, 5- to 14-membered heteroaryloxycarbonyl, 3- to 8-membered heterocyclyl, 3- to 8-membered heterocyclylcarbonyl, and 3- to 8-membered heterocyclyloxycarbonyl, preferably from C1-6 alkyl and C1-6 alkylcarbonyl, preferably from H, C1-6 alkyl, C1-6 alkylcarbonyl, and C1-6 alkoxycarbonyl, and more preferably from C1-6 alkyl and C1-6 alkylcarbonyl, the above substituents are each optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy. In some embodiments, R1B is H. In some embodiments, R1B is C1-6 alkyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, preferably methyl, ethyl, propyl, or butyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, and more preferably methyl. In some embodiments, R1B is C1-6 alkylcarbonyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, preferably methylcarbonyl, ethylcarbonyl, propylcarbonyl, or butylcarbonyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, and more preferably methylcarbonyl.


In some embodiments, R2B and R3B are each independently selected from H, C1-6 alkyl, amidino, and C1-6 alkoxycarbonyl. In some embodiments, R2B and R3B are each independently selected from H and C1-6 alkoxycarbonyl. In some embodiments, R2B and R3B are both H. In some embodiments, R2B is H, and R3B is C1-6 alkoxycarbonyl, preferably methoxycarbonyl.


In some embodiments, R4B is selected from halogen, NO2, C1-6 alkyl, C1-6 haloalkyl, cyano, NH2C(═O)—, and C1-6 alkoxy, preferably from F, Cl, NO2, CH3, CF3, cyano, and NH2C(═O)—.


In some embodiments, mB and nB are each independently 0, 1, 2, 3, 4, or 5. In some embodiments, mB is 0, 1, 2, 3, 4, or 5, preferably 3. In some embodiments, nB is 0, 1, 2, or 3, preferably 0.


In some embodiments, formula IB is preferably as shown in formula IIB,




embedded image


further preferably formula IIIB,




embedded image


and still further preferably formula IVB,




embedded image


In some embodiments, in the general formulas described above, R1B is selected from C1-6 alkyl and C1-6 alkylcarbonyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, preferably C1-6 alkyl or C1-6 alkylcarbonyl, and more preferably methyl or methylcarbonyl.


In some embodiments, the compound of the present invention is selected from compounds shown below, stereoisomers thereof, or pharmaceutically acceptable salts thereof:




embedded image


embedded image


The present disclosure further provides a compound of formula iB-1, a stereoisomer thereof, or a salt thereof, which is an intermediate for preparing the compound of the present invention,




embedded image




    • wherein R1B, R4B, mB, and nB are as defined above, and RxB and RyB are amino-protecting groups, which are preferably each independently selected from tert-butoxycarbonyl (Boc), 9-fluorenylmethoxycarbonyl (Fmoc), allyloxycarbonyl (Alloc), trichloroethoxycarbonyl (Troc), trimethylsilylethoxycarbonyl (Teoc), benzyloxycarbonyl (CBz), p-toluenesulfonyl (Tosyl), p-nitrobenzenesulfonyl (Nosyl), tert-butyl (t-Bu), trifluoroacetyl (Tfa), methoxycarbonyl, or ethoxycarbonyl.





Similarly, the present disclosure further provides a compound of formula iB-4, a stereoisomer thereof, or a salt thereof,




embedded image




    • wherein R1B, R2B, R3B, R4B, mB, nB, and RxB are as defined above.





Preparation Method

In some embodiments, the compound of the present invention may be prepared by the following method, which comprises the following steps:




embedded image




    • wherein, R1B, R4B, mB, and nB are as defined above, and RxB and RyB are amino-protecting groups, which are preferably each independently selected from tert-butoxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trichloroethoxycarbonyl, trimethylsilylethoxycarbonyl, benzyloxycarbonyl, p-toluenesulfonyl, p-nitrobenzenesulfonyl, tert-butyl, trifluoroacetyl, methoxycarbonyl, or ethoxycarbonyl.





Compound

The present disclosure provides a compound of formula IC, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:




embedded image


In some embodiments, ring A is C3-8 cycloalkyl, C6-14 aryl (preferably monocyclic aryl), or 5- to 14-membered heteroaryl (preferably monocyclic heteroaryl), such as cyclopropyl, cyclopentyl, phenyl, pyridinyl, and pyrimidinyl. In some embodiments, ring A is phenyl.


In some embodiments, Y is selected from CH or N. In some embodiments, Y is N.


In some embodiments, G is selected from —S—, —O—, —CR4CR5C—, —NR6C—, —S(═O)2—, —S(═O)(═NR6C′)—, and




embedded image


In some embodiments, G is selected from —O—, —CR4CR5C—, —NR6C—, —S(═O)2—, —S(═O)(═NR6C′)—, and




embedded image


In some embodiments, G is —O—. In some embodiments, G is —S(═O)2—.


In some embodiments, G is —S(═O)(═NR6C′)—, wherein R6C′ is selected from H, C1-6 alkyl (e.g., methyl), C1-6 alkylcarbonyl (e.g., methylcarbonyl), C1-6 alkoxycarbonyl (e.g., methoxycarbonyl), C6-14 aryl, C6-14 arylcarbonyl, C6-14 aryloxycarbonyl, C3-8 cycloalkyl, C3-8 cycloalkylcarbonyl, C3-8 cycloalkoxycarbonyl, 5- to 14-membered heteroaryl, 5- to 14-membered heteroarylcarbonyl, 5- to 14-membered heteroaryloxycarbonyl, 3- to 8-membered heterocyclyl, 3- to 8-membered heterocyclylcarbonyl, and 3- to 8-membered heterocyclyloxycarbonyl, preferably from H, C1-6 alkyl (e.g., methyl), C1-6 alkylcarbonyl (e.g., methylcarbonyl), and C1-6 alkoxycarbonyl (e.g., methoxycarbonyl), and more preferably from C1-6 alkyl (e.g., methyl) and C1-6 alkylcarbonyl (e.g., methylcarbonyl), the above substituents are each optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy;


In some embodiments, G is —CR4CR5C—, wherein R4C and R5C are each independently selected from H, C1-6 alkyl, C1-6 alkyl-O—, hydroxyl, —C(O)OR7, —NR8R9, —NRcCC(O)NR8R9, C1-6 alkylamino, 3- to 8-membered heterocyclyl-(CH2)mC—, halogen, cyano, —NRcCS(═O)2NR8R9, —NRcCC(O)ORdC, —NRcCS(═O)2ORdC, —NRcCC(O)R7′, and —NH(CH2)mCNR8R9, wherein the alkyl and heterocyclyl are optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy; R7 and R7′ are each independently selected from H, C1-6 alkyl, C3-8 cycloalkyl, 3- to 8-membered heterocyclyl, C6-14 aryl, and 5- to 14-membered heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy; R8 and R9 are each independently H or C1-6 alkyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy; RcC is H or C1-6 alkyl substituted with one or more groups selected from amino, C1-6 alkylamino, C1-6 alkoxy, halogen, hydroxyl, nitro, cyano, NH2C(═O)—, and C1-6 alkoxy, preferably H or C1-6 alkyl substituted with amino, C1-6 alkylamino, or C1-6 alkoxy, and more preferably H or C1-6 alkyl substituted with amino or C1-6 alkylamino; RdC is selected from C1-6 alkyl, C6-14 aryl, 5- to 14-membered heteroaryl, C3-8 cycloalkyl, and 3- to 8-membered heterocyclyl, preferably from C1-6 alkyl and 3- to 8-membered heterocyclyl, more preferably from C1-6 alkyl and




embedded image


and even more preferably from C1-6 alkyl and




embedded image


the group RdC described above is optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, wherein ReC and RfC are each independently —(CH2)n1C— and —(CH2)n1C′—, and in some embodiments, are —(CH2)n1C— and —(CH2)n1C′—, respectively, wherein n1C and n1C′ are each independently selected from 0, 1, 2, and 3, preferably 2, and n1C and n1C′ are not both 0; WC is selected from —NH—C(═O)—, —NH—S(═O)2—, —NR12—, —O—, —S—, and —S(═O)2—, preferably from —O— and —S(═O)2—, wherein R12 is selected from H, C1-6 alkyl, amidino, and HOOC—(CH2)n3C—, wherein n3C is selected from 1, 2, and 3; mC is selected from 1, 2, 3, and 4;

    • or CR4CR5C forms a 3- to 8-membered heterocyclic or 9- to 10-membered bicyclic moiety (particularly a 9- to 10-membered fused ring), wherein the 3- to 8-membered heterocyclic or 9- to 10-membered bicyclic moiety, together with the piperidine ring attached thereto, forms a structure selected from:




embedded image




    • when any one of Q1C-Q4C is N, the rest are CH, or Q1C-Q4C are all CH;

    • W1C and W2C are each independently —C(═O)—NH—, —NH—C(═O)—, —S(═O)2—NH—, —NH—S(═O)2—, —S—, —O—, —NR12—, —NR12—CH2—, —(CH2)n2C— optionally substituted with one or more groups selected from —NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, or absent, wherein R12 is selected from H, C1-6 alkyl, amidino, and HOOC—(CH2)n3C—; preferably, W1C and W2C cannot be simultaneously absent;

    • W3C is —(CH2)n2C— optionally substituted with one or more groups selected from NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, or absent; preferably —(CH2)n2C— optionally substituted with one or more groups selected from NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy;

    • W4C and W5C are —(CH2)n2C— optionally substituted with one or more groups selected from NH2, —OH, halogen, nitro, cyano, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, or absent;

    • n2C is selected from 0, 1, 2, and 3;

    • more preferably, the 3- to 8-membered heterocyclic or 9- to 10-membered bicyclic moiety, together with the piperidine ring attached thereto, forms a structure selected from:







embedded image


embedded image


In some embodiments, G is —CR4CR5C—, wherein R4C is H, and R5C is —NRcCC(O)ORdC, wherein RcC and RdC are as defined above.


In some embodiments, G is —CR4CR5C—, wherein R4C is —NR8R9, and R5C is —C(O)OR7, wherein R7, R8, and R9 are as defined above. In some embodiments, R7 is H or C1-6 alkyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, e.g., H or C1-6 alkyl, such as H or methyl. In some embodiments, R8 and R9 are both H.


In some embodiments, G is —S(═O)(═NR6C′)—, wherein R6C′ is as defined above.


In some embodiments, R1C is selected from H or —(CH2)tNRaCRbC, wherein RaC and RbC are each independently selected from H, C1-6 alkyl (e.g., methyl, isopropyl, etc.), amidino, and C1-6 alkoxycarbonyl (e.g., methoxycarbonyl, etc.), and t is selected from 0, 1, 2, 3, 4, or 5. In some embodiments, R1C is H. In some embodiments, RaC and RbC are each independently selected from H and C1-6 alkoxycarbonyl (e.g., methoxycarbonyl, etc.). In some embodiments, RaC and RbC are each H. In some embodiments, t is 0, 1, 2, or 3, preferably 3.


In some embodiments, R2C is selected from H, amino, hydroxyl, C1-6 alkyl, C1-6 alkylamino, and C1-6 aminoalkyl, wherein the alkyl is optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy.


In some embodiments, when Y is CH, R3C is selected from H, hydroxyl, C1-6 alkyl, 3- to 8-membered heterocyclyl, and C1-6 alkoxy, and when Y is N, R3C is selected from H, C1-6 alkyl, C3-8 cycloalkyl, C3-8 cycloalkyl-(CH2)mC—, 3- to 8-membered heterocyclyl, 3- to 8-membered heterocyclyl-(CH2)mC—, and —(CH2)mCNR10R11, wherein the alkyl, cycloalkyl, heterocyclyl, and alkoxy are optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, C1-6 alkoxy, and C1-6 alkylamino; R10 and R11 are each independently H or C1-6 alkyl, or R10 and R11, together with the nitrogen atom to which they are both attached form 3- to 8-membered heterocyclyl (e.g.,




embedded image


wherein the alkyl and heterocyclyl are optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy; mC is independently selected from 1, 2, 3, and 4 at each occurrence.


In some embodiments, R0 is selected from H, halogen NO2, cyano, NH2C(═O)—, C1-6 alkoxy, and C1-6 alkyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy, preferably from H, F, Cl, NO2, CH3, CF3, cyano, NH2C(═O)—, and C1-6 aminoalkyl. In some embodiments, R4C is H.


In some embodiments, p is 0, 1, 2, 3, 4, or 5, preferably 0, 1, 2, or 3, and more preferably 0.


In some embodiments, formula IC is as shown in formula IIC-1 or IIC-2:




embedded image


In some embodiments, in IIC-1, R2C is C1-6 aminoalkyl or amino, and R3C is H or C1-6 alkyl; in some embodiments, R2C is amino, and R3C is H.


In some embodiments, in formula IIC-2, R2C is C1-6 aminoalkyl or amino, and R3C is H or C1-6 alkyl, or R2C is H or C1-6 alkyl, and R3C is C1-6 aminoalkyl or 3- to 8-membered heterocyclyl containing nitrogen.


In some embodiments, formula IC is as shown in formula IIIC or formula IVC:




embedded image


In some embodiments, in the general formulas described above, one or more of the following criteria are satisfied:

    • (1) R1C is —(CH2)tNRaCRbC, preferably —(CH2)3NH2;
    • (2) ring A is phenyl; and
    • (3) p is 0.


In some embodiments, in the general formulas, G is selected from one of the following:

    • (1) G is —S(═O)(═NR6C′)—
    • (2) G is —CR4CR5C—, wherein R4C is H, and R5C is —NRcC(O)ORdC; and
    • (3) G is —CR4CR5C—, wherein R4C is —NR8R9, and R5C is —C(O)OR7, wherein R7 is selected from H, C1-6 alkyl, C3-8 cycloalkyl, 3- to 8-membered heterocyclyl, C6-14 aryl, and 5- to 14-membered heteroaryl, wherein the alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy; and R7 is preferably H or C1-6 alkyl optionally substituted with one or more groups selected from halogen, hydroxyl, amino, nitro, cyano, C1-6 alkyl, C1-6 haloalkyl, NH2C(═O)—, and C1-6 alkoxy.


In some embodiments, formula IC is as shown in formula IVC-1, IVC-2, or IVC-3:




embedded image


In some embodiments, the compound of the present disclosure is selected from compounds shown below, stereoisomers thereof, or pharmaceutically acceptable salts thereof:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image




embedded image


The present disclosure further provides a compound of the following general formula, a stereoisomer thereof, or a salt thereof, which is an intermediate for preparing the compounds of the present disclosure described above:




embedded image




    • wherein, R0′ is R0 or R0 in which NH2 is protected with an amino-protecting group, R1C is R1C or R1C in which NH2 is protected with an amino-protecting group, R2C′ is R2C or R2C in which NH2 is protected with an amino-protecting group, R3C′ is R3C or R3C in which NH2 or the cyclic imino is protected with an amino-protecting group, G′ is G or G in which NH2 and/or the cyclic imino is protected with an amino-protecting group and/or the carboxyl is protected with a carboxyl-protecting group, ring A, Y, R0, R1C, R2C, R3C, G, and p are as defined above, and Rv is selected from H and carboxyl-protecting groups.





In some embodiments, formula iiC-2-X is selected from:




embedded image




    • wherein Rs and Rt are each independently selected from H and amino-protecting groups.





In some embodiments, the amino-protecting groups are each independently selected from tert-butoxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trichloroethoxycarbonyl, trimethylsilylethoxycarbonyl, benzyloxycarbonyl, p-toluenesulfonyl, p-nitrobenzenesulfonyl, tert-butyl, trifluoroacetyl, methoxycarbonyl, tert-butylsulfinyl, 1-phenylethyl, or ethoxycarbonyl.


In some embodiments, the carboxyl-protecting groups are each independently selected from C1-6 alkyl, allyl, benzyl, 2,4-dimethoxybenzyl, p-methoxybenzyl, methoxyethoxymethyl, pentafluorophenyl, and 4-p-methylbenzyloxybenzyl.


Preparation Method

In some embodiments, the compound of the present invention may be prepared by a method selected from:

    • method I:
    • method I comprises the following steps: subjecting a compound of formula iC-2 and a compound of formula iC-3-a or iC-3-b or iC-3-c to a condensation reaction to generate a compound of formula iC-1, and then subjecting the compound of formula iC-1 to a protecting group removal reaction to generate a compound of formula IC,




embedded image




    • method II:

    • method II comprises the following steps: subjecting a compound of formula iiC-2 and a compound of formula iiC-3 to a condensation reaction to generate a compound of formula iiC-1, and then subjecting the compound of iiC-1 to a protecting group removal reaction to generate the compound of formula IC,







embedded image


Preferably, a compound of formula iiC-2 is prepared by a method selected from:

    • method III:
    • method III comprises the following steps: subjecting a compound of formula iiC-4-a and a compound of formula iiC-5-a to a condensation reaction, a hydrolysis reaction, an optional removal reaction of an amino-protecting group, and an amino protection reaction to give the compound of formula iiC-2,




embedded image




    • method IV:

    • method IV comprises the following steps: subjecting a compound of formula iiC-4-b and a compound of formula iiC-5-b to a condensation reaction, a hydrolysis reaction, an optional removal reaction of an amino-protecting group, and an amino protection reaction to give the compound of formula iiC-2,







embedded image




    • method V:

    • method V comprises the following steps: subjecting a compound of formula iiC-4-c and a compound of formula iiC-5-c to a condensation reaction, a hydrolysis reaction, an optional removal reaction of an amino-protecting group, and an amino protection reaction to give the compound of formula iiC-2,







embedded image




    • method VI:

    • method VI comprises the following steps: subjecting a compound of formula iiC-4-d and a compound of formula iiC-5-d to a condensation reaction, a hydrolysis reaction, an optional removal reaction of an amino-protecting group, and an amino protection reaction to give the compound of formula iiC-2,







embedded image




    • preferably, the formula iC-2 is







embedded image




    • preferably, iiC-2 is selected from:







embedded image


embedded image




    • wherein, R0′ is R0 or R0 in which NH2 is protected with an amino-protecting group, R1C and R1C′ are R1C or R1C in which NH2 is protected with an amino-protecting group, R2C′ and R2C″ are R2C or R2C in which NH2 is protected with an amino-protecting group, R3C′ and R3C″ are R3C or R3C in which NH2 or the cyclic imino is protected with an amino-protecting group, G′ is G or G in which NH2 and/or the cyclic imino is protected with an amino-protecting group and/or the carboxyl is protected with a carboxyl-protecting group, Rw is a carboxyl-protecting group, ring A, Y, R0, R1C, R2C, R3C, G, and p are as defined above, and Rs, Rt, and Ru are each independently selected from H and amino-protecting groups;

    • preferably, the amino-protecting groups are each independently selected from tert-butoxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trichloroethoxycarbonyl, trimethylsilylethoxycarbonyl, benzyloxycarbonyl, p-toluenesulfonyl, p-nitrobenzenesulfonyl, tert-butyl, trifluoroacetyl, methoxycarbonyl, tert-butylsulfinyl, 1-phenylethyl, or ethoxycarbonyl; the carboxyl-protecting groups are each independently selected from C1-6 alkyl, allyl, benzyl, 2,4-dimethoxybenzyl, p-methoxybenzyl, methoxyethoxymethyl, pentafluorophenyl, and 4-p-methylbenzyloxybenzyl.





In method III, method IV, method V, and method VI described above, the condensation reaction refers to a condensation reaction of iiC-4 (specifically iiC-4-a, iiC-4-b, iiC-4-c, and iiC-4-d) with iiC-5 (specifically iiC-5-a, iiC-5-b, iiC-5-c, and iiC-5-d), respectively to form a condensation product; the hydrolysis reaction is intended to remove the carboxyl-protecting group Rw in the condensation product, but according to different hydrolysis conditions, the amino-protecting group (if present) in the condensation product may be partially or completely removed while the carboxyl-protecting group Rw in the condensation product is removed, and the product after the hydrolysis reaction is referred to as a hydrolysis product. In some embodiments, the hydrolysis product is the compound of iiC-2. In some embodiments, the hydrolysis product is further subjected to an amino protection reaction to give the compound of formula iiC-2. In some embodiments, the hydrolysis product is further subjected to a removal reaction of an amino-protecting group and an amino protection reaction to give the compound of formula iiC-2.


EXAMPLES

Examples of the present invention are described in detail below. The following examples are exemplary and illustrative only, and should not be construed as limiting the present invention. Unless otherwise indicated, the proportions, percentages, and the like referred to herein are calculated by weight.


Reagents or instruments without specified manufacturers used herein are conventional products that are commercially available. Specifically, intermediate 2 (the structure is shown below) was purchased from HANGZHOU TAIJIA BIOTECH CO., LTD.




embedded image


In addition, with respect to the purification of the final product, in each example, it is mentioned as “purified by reversed-phase flash chromatography”, “purified by preparative HPLC”, “purified by reversed-phase column chromatography”, “purified by high-performance liquid chromatography”, and “purified by a reversed-phase column”. In some examples, the mobile phase conditions used for the purification are specifically indicated as “solvent ACN and solvent H2O (FA, 0.1%)”, but it should be understood that the same or similar mobile phase conditions are also used in other examples. Under such chromatographic conditions, the compounds in certain examples are formed as formate salts. However, it should be understood by those skilled in the art that the free compounds may be formed by conventional neutralization reaction conditions in the art.


Abbreviations





    • ACN: acetonitrile

    • BTC: (trichloromethyl)carbonate

    • CDI: N,N-carbonyl diimidazole

    • DCM: dichloromethane

    • DIEA: diethylamine

    • DIPEA: N,N-diisopropylethylamine

    • DMAP: 4-dimethylaminopyridine

    • DMF: dimethylformamide

    • HATU: 2-(7-azabenzotriazole)-N,N,N′,N′-tetramethyluronium hexafluorophosphate

    • HOBT: 1-hydroxybenzotriazole

    • TBTU: O-benzotriazole-N,N,N′,N′-tetramethyluronium tetrafluoroborate

    • TEA: triethylamine

    • TFA: trifluoroacetic acid

    • TFE: 2,2,2-trifluoroethanol

    • THF: tetrahydrofuran





Example 1. Synthesis of Compound 1



embedded image


embedded image


1.1 Synthesis of Compound 1-1

Methyl 4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate hydrochloride (3.7 g, 12.552 mmol, 1.00 equiv) was dissolved in DMF (37 mL) at room temperature, and DIEA (4.87 g, 37.656 mmol, 3 equiv), (2R)-2-{[(benzyloxy)carbonyl]amino}-6-[(tert-butoxycarbonyl)amino]hexanoic acid (5.73 g, 15.062 mmol, 1.2 equiv) and HATU (5.73 g, 15.062 mmol, 1.2 equiv) were separately added. The resulting mixture was stirred at room temperature overnight. The resulting mixture was extracted with dichloromethane (2×200 mL). The combined organic layer was washed with brine (2×300 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with dichloromethane and methanol (20:1) to give methyl 1-[(2R)-2-{[(benzyloxy)carbonyl]amino}-6-[(tert-butoxycarbonyl)amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (8.97 g, 94.63%, purity: 82.0%) as an off-white solid.


LC-MS-1-1 (ES, m/z): [M+1]=621


1.2 Synthesis of Compound 1-3

tert-Butyl (2R)-2-amino-4-methylpentanoate hydrochloride (5 g, 22.347 mmol, 1.00 equiv) and DIEA (8.66 g, 67.041 mmol, 3 equiv) were dissolved in DMF (50.00 mL, 646.052 mmol, 28.91 equiv), and (2R)-2-{[(9H-fluoren-9-ylmethoxy)carbonyl]amino}-3-phenylpropionic acid (10.39 g, 26.816 mmol, 1.2 equiv) and HATU (10.20 g, 26.816 mmol, 1.2 equiv) were added. The resulting mixture was stirred at room temperature overnight. The resulting mixture was extracted with ethyl acetate (1×500 mL). The combined organic layer was washed with brine (5×300 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with petroleum ether/ethyl acetate (4:1) to give tert-butyl (2R)-2-[(2R)-2-{[(9H-fluoren-9-ylmethoxy)carbonyl]amino}-3-phenylpropionylamino]-4-methylpentanoate (13.94 g, 97.60%, purity: 87.0%) as a white solid.


LC-MS-1-3 (ES, m/z): [M+1]=557


1.3 Synthesis of Compound 1-2

Methyl 1-[(2R)-2-{[(benzyloxy)carbonyl]amino}-6-[(tert-butoxycarbonyl)amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (8.97 g, 14.450 mmol, 1.00 equiv) was dissolved in methanol (162.35 mL), and Pd/C (922.69 mg, w/t, 10%) was added. The resulting mixture was stirred at room temperature overnight under hydrogen atmosphere. The resulting mixture was filtered, and the filter cake was washed with methanol (3×20 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography to give methyl 1-[(2R)-2-amino-6-[(tert-butoxycarbonyl)amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (5.65 g, 76.53%, purity: 95.0%).


LC-MS-1-2 (ES, m/z): [M+1]=487


1.4 Synthesis of Compound 1-4

tert-Butyl (2R)-2-[(2R)-2-{[(9H-fluoren-9-ylmethoxy)carbonyl]amino}-3-phenylpropionylamino]-4-methylpentanoate (13.94 g, 25.040 mmol) was dissolved in DCM (100 mL, 1573.005 mmol, 62.82 equiv.), and TFA (1.00 mL, 1346.304 mmol, 53.77 equiv) was added. The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated under reduced pressure. The crude product was recrystallized from dichloromethane/1 M hydrochloric acid in water, washed with water (1×400 mL), and filtered. The solid was collected to give (2R)-2-[(2R)-2-{[(9H-fluoren-9-ylmethoxy)carbonyl]amino}-3-phenylpropionylamino]-4-methylpentanoic acid (9.43 g, 74.48%, purity: 98.8%).


LC-MS-1-4 (ES, m/z): [M+1]=501


1.5 Synthesis of Compound 1-5

(2R)-2-[(2R)-2-{[(9H-Fluoren-9-ylmethoxy)carbonyl]amino}-3-phenylpropionylamino]-4-methylpentanoic acid (4.78 g, 9.539 mmol, 1.1 equiv) and DIEA (3.36 g, 26.016 mmol, 3 equiv) were dissolved in DMF (40 mL, 516.870 mmol, 59.60 equiv), and methyl 1-[(2R)-2-amino-6-[(tert-butoxycarbonyl)amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (4.22 g, 8.672 mmol, 1.00 equiv) and HATU (3.96 g, 10.406 mmol, 1.2 equiv) were added. The resulting mixture was stirred at room temperature overnight. The resulting mixture was extracted with ethyl acetate (1×400 mL). The combined organic layer was washed with brine (3×200 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with petroleum ether/ethyl acetate (1:2), and the fractions were concentrated to dryness by rotary evaporation to give methyl 4-[(tert-butoxycarbonyl)amino]-1-[(2R)-6-[(tert-butoxycarbonyl)amino]]-2-[(2R)-2-[(2R)-2-{[(9H-fluoren-9-ylmethoxy) carbonyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylate (8.39 g, 98.12%, purity: 98.3%).


LC-MS-1-5 (ES, m/z): [M+1]=969


1.6 Synthesis of Compound 1-6

Methyl 4-[(tert-butoxycarbonyl)amino]-1-[(2R)-6-[(tert-butoxycarbonyl)amino]-2-[(2R)-2-[(2R)-2]—{[(9H-fluoren-9-ylmethoxy)carbonyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylate (8.93 g, 9.214 mmol, 1.00 equiv) was dissolved in DCM (80 mL), and piperidine (8.90 mL, 104.487 mmol, 11.34 equiv) was added portionwise. The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography to give methyl 1-[(2R)-2-[(2R)-2-[(2R)-2-amino-3-phenylpropionylamino]-4-methylpentanoylamino]-6-[(tert-butoxycarbonyl)amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (5.74 g, 80.98%, purity: purity: 97.0%).


LC-MS-1-6 (ES, m/z): [M+1]=747


1.7 Synthesis of Compound 1-7

Carbonylimidazole (47.76 mg, 0.295 mmol, 1.1 equiv) was dissolved in N,N-dimethylformamide (1.60 mL), and methyl 1-[(2R)-2-[(2R)-2-[(2R)-2-amino-3-phenylpropionylamino]-4-methylpentanoylamino]-6-[(tert-butoxycarbonyl) amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (200 mg, 0.268 mmol, 1.00 equiv), N-methylbenzylamine (32.45 mg, 0.268 mmol, 1 equiv) and triethylamine (54.19 mg, 0.536 mmol, 2 equiv) were added under stirring. The resulting mixture was stirred at room temperature overnight and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography, and the fractions were concentrated to dryness by rotary evaporation to give methyl 1-[(2R)-2-[(2R)-2-[(2R)-2-{[benzyl(methyl)carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]-6-[(tert-butoxycarbonyl)amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (136 mg, 55.67%, purity: 66.7%).


LC-MS-1-7 (ES, m/z): [M+1]=894


1.8 Synthesis of Compound 1-8

Methyl 1-[(2R)-2-[(2R)-2-[(2R)-2-{[benzyl(methyl)carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]-6-[(tert-butoxycarbonyl)amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (136 mg, 0.152 mmol, 1.00 equiv) was dissolved in dioxane (10.00 mL), and a 4 M solution of HCl (gas) in 1,4-dioxane (15.0 mL) was added portionwise at room temperature. The resulting mixture was stirred at room temperature for 4 h. The reaction solution was concentrated under reduced pressure to give methyl 4-amino-1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-{[benzyl(methyl)carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylate (115 mg, crude).


LC-MS-1-8 (ES, m/z): [M+1]=694


1.9 Synthesis of Compound 1

Methyl 4-amino-1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-{[benzyl(methyl)carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylate (115 mg, 0.166 mmol, 1.00 equiv) was dissolved in a mixed solvent THE (10 mL)/water (2 mL), and LiOH·H2O (13.933 mg, 0.332 mmol, 2.00 equiv) was added. The resulting mixture was stirred at room temperature for 1 h. The mixture was acidified with 1 M HCl (aq.) to pH=7. The resulting mixture was purified by reversed-phase flash chromatography to give 4-amino-1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-{[benzyl(methyl)carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylic acid (60.0 mg, 49.23%, purity: 95.2%).


LC-MS-1 (ES, m/z): [M+1]=680



1H NMR-1 (400 MHz, Deuterium Oxide) δ 8.38 (s, 1H), 7.36-7.22 (m, 6H), 7.18 (dd, J=7.6, 2.0 Hz, 2H), 7.03-6.95 (m, 2H), 4.76-4.72 (m, 1H), 4.50 (ddd, J=9.0, 5.7, 3.0 Hz, 1H), 4.43 (d, J=16.5 Hz, 1H), 4.38-4.25 (m, 2H), 3.88-3.47 (m, 4H), 3.11 (dd, J=13.9, 5.8 Hz, 1H), 2.97-2.85 (m, 3H), 2.77 (d, J=2.9 Hz, 3H), 2.27-2.06 (m, 2H), 1.93-1.42 (m, 9H), 1.42-1.23 (m, 2H), 0.92-0.77 (m, 6H).


Example 2. Synthesis of Compound 2



embedded image


LC-MS-2-0 (ES, m/z): [M+1]=710



1H NMR (400 MHz, Deuterium Oxide) δ 8.30 (s, 1H), 7.51-7.20 (m, 8H), 7.18-6.96 (m, 2H), 5.52 (ddd, J=19.0, 10.7, 4.7 Hz, 1H), 4.45 (dt, 1=8.9, 6.2 Hz, 1H), 4.30 (p, =4.7 Hz, 1H), 3.75 (dp, =18.4, 6.9 Hz, 2H), 3.68-3.38 (m, 4H), 3.10 (ddd, J=14.6, 9.2, 6.3 Hz, 1H), 2.97 (dt, =13.9, 10.1 Hz, 1H), 2.87 (q, J=7.8 Hz, 2H), 2.53 (d, J=5.4 Hz, 3H), 2.30-2.05 (m, 2H), 1.96-1.43 (m, 9H), 1.32 (d, J=20.6 Hz, 2H), 0.85 (dd, J=16.8, 5.6 Hz, 6H).


LC-MS-2-100 (ES, m/z): [M+1]=710



1H NMR (400 IVI-z, Deuterium Oxide) δ 8.35 (s, 2H), 7.50-7.10 (m, 8H), 7.03-6.72 (m, 2H), 5.53 (dd, J=11.1, 4.2 Hz, 1H), 4.86-4.74 (m, 1H), 4.72-4.59 (m, 1H), 4.35 (dt, J=9.3, 5.2 Hz, 1H), 4.06-3.66 (m, 3H), 3.68-3.37 (m, 3H), 3.34-3.14 (m, 1H), 3.07-2.73 (m, 3H), 2.43 (d, J=6.2 Hz, 3H), 2.31-2.01 (m, 2H), 1.97-1.21 (m, 11H), 0.85 (dd, J=16.8, 5.6 Hz, 6H).


Example 3. Synthesis of Compound 3



embedded image


LC-MS-3 (ES, m/z): [M+1]=681



1H NMR-3 (400 MHz, Deuterium Oxide) δ 8.34 (s, 2H), 7.34-7.19 (m, 6H), 7.11 (dt, J=7.3, 2.2 Hz, 2H), 7.09-7.01 (m, 2H), 4.53 (td, J=6.9, 6.1, 3.1 Hz, 1H), 4.33 (ddd, J=7.0, 4.9, 1.9 Hz, 1H), 4.30-4.21 (m, 1H), 3.88-3.46 (m, 4H), 3.01-2.82 (m, 5H), 2.75 (ddd, J=14.1, 7.1, 4.5 Hz, 1H), 2.28-2.04 (m, 2H), 1.95-1.55 (m, 6H), 1.57-1.26 (m, 5H), 0.82 (dd, J=19.0, 5.2 Hz, 6H).


Example 4. Synthesis of Compound 4



embedded image


4.1 Synthesis of Compound 4-1

Methyl 1-[(2R)-2-[(2R)-2-[(2R)-2-amino-3-phenylpropionylamino]-4-methylpentanoylamino]-6-[(tert-butoxycarbonyl)amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (120 mg, 0.161 mmol, 1.00 equiv) was dissolved in DMF (7 mL), and 3-[(tert-butoxycarbonyl)amino]-3-phenylpropionic acid (51.15 mg, 0.193 mmol, 1.2 equiv), HATU (73.30 mg, 0.193 mmol, 1.2 equiv) and DIEA (62.29 mg, 0.483 mmol, 3 equiv) were added. The resulting mixture was stirred overnight, diluted with water (30 mL), and extracted with EA (3×20 mL). The combined organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give methyl 4-[(tert-butoxycarbonyl)amino]-1-[(2R)-6-[(tert-butoxycarbonyl)amino]-2-[(2R)-2-[(2R)-2-{3-[(tert-butoxy carbonyl)amino]-3-phenylpropionylamino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylate (256 mg, crude).


LC-MS-4-1 (ES, m/z): [M+1]=994


4.2 Synthesis of Compound 4-2

Methyl 4-[(tert-butoxycarbonyl)amino]-1-[(2R)-6-[(tert-butoxycarbonyl)amino]-2-[(2R)-2-[(2R)-2-{3-[(tert-butoxycarbonyl)amino]-3-phenylpropionylamino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylate (265 mg, 0.267 mmol, 1.00 equiv) was dissolved in dioxane (5.3 mL), and a 4 M solution of HCl (gas) in 1,4-dioxane (8 mL) was added at room temperature. The resulting mixture was stirred at room temperature for 2 h and concentrated under reduced pressure to give methyl 4-amino-1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-(3-amino-3-phenylpropionylamino)-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylate (101 mg, crude).


LC-MS-4-2 (ES, m/z): [M+1]=694


4.3 Synthesis of Compounds 4-0/100

Methyl 4-amino-1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-(3-amino-3-phenylpropionylamino)-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylate (80 mg, 0.115 mmol, 1.00 equiv) was dissolved in THF (3 mL), and H2O (0.8 mL, 44.4075.15 mmol) and LiOH (5.52 mg, 0.230 mmol, 2 equiv) were added. The resulting mixture was stirred at room temperature for 4 h. The mixture was acidified with HCl (1 N) to pH 7. The resulting mixture was diluted with water (100 mL). The resulting mixture was extracted with ethyl acetate (3×100 mL). The combined organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography. The fractions were concentrated to dryness by rotary evaporation to give 4-amino-1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-((3R)-3-amino-3-phenylpropionylamino)-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylic acid (11 mg, 13.43%, purity: 95.3%) and 4-amino-1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-[(3S)-3-amino-3-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylic acid (11 mg, 13.74%, purity: 97.5%).


LC-MS-4-0 (ES, m/z): [M+1]=680


Compound 4-0: 1H NMR (300 MHz, Deuterium Oxide) δ 8.34 (s, 2H), 7.22 (t, J=41.9 Hz, 10H), 4.27 (d, J=80.8 Hz, 4H), 3.64 (d, J=47.2 Hz, 4H), 2.89 (s, 6H), 2.13 (s, 2H), 1.90-1.48 (m, 6H), 1.37 (s, 5H), 0.76 (d, J=20.6 Hz, 6H).


LC-MS-4-100 (ES, m/z): [M+1]=680


Compound 4-100: 1H NMR (300 MHz, Deuterium Oxide) δ 8.34 (s, 2H), 7.50-6.77 (m, 10H), 4.59-4.16 (m, 4H), 3.67 (d, J=40.1 Hz, 4H), 3.05-2.62 (m, 6H), 2.12 (d, J=27.4 Hz, 2H), 1.96-1.52 (m, 6H), 1.54-1.16 (m, 5H), 0.93-0.66 (m, 6H).


Example 5. Synthesis of Compound 5



embedded image


LC-MS-5-0 (ES, m/z): [M+1]=680


Compound 5-0: 1H NMR (300 MHI-z, Deuterium Oxide) δ 8.37 (s, 1H), 7.37-7.08 (m, 10H), 4.90 (t, 1=7.1 Hz, 1H), 4.48-4.39 (m, 2H), 4.20 (t, 1=7.3 Hz, 1H), 3.86-3.45 (m, 4H), 3.06-2.78 (m, 4H), 2.78-2.50 (in, 2H), 2.26-2.02 (m, 2H), 1.93-1.53 (m, 6H), 1.51-1.19 (m, 5H), 0.80 (dd, J=17.5, 5.1 Hz, 6H).


LC-MS-5-100 (ES, m/z): [M+1]=680


Compound 5-100: 1H NMR (300 MHz, Deuterium Oxide) δ 7.39-7.01 (m, 10H), 4.94 (t, J=7.1 Hz, 1H), 4.44 (t, J=7.4 Hz, 2H), 4.24 (d, J=8.8 Hz, 1H), 3.66 (d, J=58.6 Hz, 4H), 2.99-2.56 (m, 6H), 2.14 (d, J=20.9 Hz, 2H), 1.92-1.55 (m, 6H), 1.40 (d, J=33.6 Hz, 5H), 0.81 (dd, J=15.7, 4.9 Hz, 6H).


Example 6. Synthesis of Compound 6



embedded image


LC-MS-6-0 (ES, m/z): [M+1]=734



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 2H), 7.48-6.81 (m, 10H), 4.66-4.48 (m, 1H), 4.06 (q, J=6.6, 5.5 Hz, 1H), 3.91-3.44 (m, 6H), 3.25 (dd, J=13.1, 4.9 Hz, 1H), 3.16-2.48 (m, 9H), 2.35-1.97 (m, 2H), 1.98-1.18 (m, 15H), 0.91 (dd, J=18.5, 5.3 Hz, 6H).


LC-MS-6-100 (ES, m/z): [M+1]=734



1H NMR (400 MHz, Deuterium Oxide) δ 8.38 (s, 3H), 7.45-6.86 (m, 10H), 4.68-4.57 (m, 1H), 4.10 (d, J=2.8 Hz, 1H), 3.96-3.42 (m, 5H), 3.42-3.17 (m, 1H), 3.02 (dt, J=12.5, 5.7 Hz, 1H), 2.97-2.77 (m, 4H), 2.71-2.48 (m, 1H), 2.14 (dd, J=27.5, 13.5 Hz, 2H), 2.02-1.49 (m, 13H), 1.39-1.23 (m, 2H), 1.08-0.79 (m, 6H).


Example 7. Synthesis of Compound 7



embedded image


LC-MS-7 (ES, m/z): [M+1]=695



1H NMR (400 MHz, Deuterium Oxide) δ 8.35 (s, 2H), 7.53-6.99 (m, 9H), 4.39 (ddd, J=8.8, 5.6, 3.0 Hz, 1H), 4.32-3.97 (m, 5H), 3.94-3.63 (m, 2H), 3.58-3.29 (m, 2H), 3.04 (dt, J=13.9, 5.1 Hz, 1H), 2.93-2.63 (m, 3H), 2.39-1.92 (m, 2H), 1.97-1.18 (m, 11H), 0.79 (dd, J=19.1, 5.4 Hz, 6H).


Example 8. Synthesis of Compound 8



embedded image


8.1 Synthesis of Compound 8-1

tert-Butyl (2R)-2-amino-3-phenylpropionate (480 mg, 2.169 mmol, 1.00 equiv) and triethylamine (1.85 g, 18.282 mmol, 8.43 equiv) were dissolved in acetonitrile (19 mL, 361.469 mmol, 166.65 equiv). The resulting mixture was stirred at 0° C. for 30 min under nitrogen atmosphere. N,N′-Carbonyldiimidazole (1.2 g, 7.401 mmol, 3.41 equiv) and 2,2,2-trifluoro-N-[2-(methylamino)-2-phenylethyl]acetamide (12.80 g, 51.991 mmol, 23.97 equiv) were added to the mixture described above. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography, and the fractions were concentrated to dryness by rotary evaporation to give tert-butyl (2R)-2-({methyl[1-phenyl-2-(2,2,2-trifluoroacetamido)ethyl]carbamoyl}amino)-3-phenylpropionate (855 mg, 79.87%, purity: 100%).


LC-MS-8-1 (ES, m/z): [M+1]=494


8.2 Synthesis of Compound 8-2

tert-Butyl (2R)-2-({methyl[1-phenyl-2-(2,2,2-trifluoroacetamido)ethyl]carbamoyl}amino)-3-phenylpropionate (67 mg, 0.136 mmol, 1 equiv) was dissolved in dichloromethane (7.70 mL), and trifluoroacetic acid (7.70 mL, 78.592 mmol, 45.09 equiv) was added dropwise. The resulting mixture was stirred in the air for 3 h. The resulting mixture was concentrated under reduced pressure to give (2R)-2-({methyl[1-phenyl-2-(2,2,2-trifluoroacetamido)ethyl]carbamoyl}amino)-3-phenylpropionic acid (600 mg, crude). The crude mixture was directly used in the next step without further purification.


LC-MS-8-2 (ES, m/z): [M+1]=438


8.3 Synthesis of Compound 8-3

{4-[(4-Methylphenyl)methoxy]phenyl}methyl (2R)-2-[(2R)-2-amino-4-methylpentanoylamino]-6-[(tert-butoxycarbonyl)amino]hexanoate (1445.79 mg, 2.538 mmol, 3 equiv), (2R)-2-({methyl[1-phenyl-2-(2,2,2-trifluoroacetamido)ethyl]carbamoyl}amino)-3-phenylpropionic acid (2600 mg, 5.944 mmol, 9.15 equiv) and HOBT (222 mg, 1.643 mmol, 2.53 equiv) were dissolved in DMF (50 mL, 64.609 mmol, 99.49 equiv) at room temperature, and TBTU (528 mg, 1.644 mmol) was added. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere to give {4-[(4-methylphenyl)methoxy]phenyl}methyl (2R)-6-[(tert-butoxycarbonyl)amino]-2-[(2R)-4-methyl-2-[(2R)-2-({methyl[1-phenyl-2-(2,2,2-trifluoroacetamido)ethyl]carbamoyl}amino)-3-phenylpropionylamino]pentanoylamino]hexanoate (2.6 g, crude).


8.4 Synthesis of Compound 8-4

{4-[(4-Methylphenyl)methoxy]phenyl}methyl (2R)-6-[(tert-butoxycarbonyl)amino]-2-[(2R)-4-methyl-2-[(2R)-2-({methyl[1-phenyl-2-(2,2,2-trifluoroacetamido)ethyl]carbamoyl}amino)-3-phenylpropionylamino]pentanoylamino]hexanoate (2.8 g, 3.150 mmol, 1 equiv) was dissolved in DCM (20 mL) at room temperature, and trifluoroacetic acid (10 mL) was added. The resulting mixture was stirred at room temperature for 2 h under air atmosphere to give (2R)-6-amino-2-[(2R)-4-methyl-2-[(2R)-2-({methyl[1-phenyl-2-(2,2,2-trifluoroacetamido)ethyl]carbamoyl}amino)-3-phenylpropionylamino]pentanoylamino]hexanoic acid (470 mg, crude).


LC-MS-8-4 (ES, m/z): [M+1]=679


8.5 Synthesis of Compound 8-5

(2R)-6-Amino-2-[(2R)-4-methyl-2-[(2R)-2-({methyl[1-phenyl-2-(2,2,2-trifluoroacetamido)ethyl]carbamoyl}amino)-3-phenylpropionylamino]pentanoylamino]hexanoic acid (470 mg, 0.692 mmol, 1 equiv), H2O (10 mL) and lithium hydroxide (33.17 mg, 1.384 mmol, 2 equiv) were added to a 50 mL round-bottom flask at room temperature. The resulting mixture was stirred at room temperature for 3 h. The resulting mixture was concentrated under reduced pressure to give (2R)-6-amino-2-[(2R)-2-[(2R)-2-{[(2-amino-1-phenylethyl) (methyl)carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoic acid (500 mg, crude).


LC-MS-8-5 (ES, m/z): [M+1]=583


8.6 Synthesis of Compound 8-6

(2R)-6-Amino-2-[(2R)-2-[(2R)-2-{[(2-amino-1-phenylethyl)(methyl)carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoic acid (350 mg, 0.601 mmol, 1 equiv) and TEA (182.33 mg, 1.803 mmol, 3 equiv) were dissolved in THF (10 mL), and di-tert-butyl dicarbonate (262.1202 mg, 1 equiv) was added. The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated under reduced pressure and purified by reversed-phase flash chromatography to give (2R)-6-[(tert-butoxycarbonyl)amino]-2-[(2R)-2-[(2R)-2-[({2-[(tert-butoxycarbonyl)amino]]-1-phenylethyl}(methyl)carbamoyl) amino]-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoic acid (260 mg, 55.29%, purity: 100%).


LC-MS-8-6 (ES, m/z): [M+1]=783


8.7 Synthesis of Compound 8-7

(2R)-6-[(tert-Butoxycarbonyl)amino]-2-[(2R)-2-[(2R)-2-[({2-[(tert-butoxycarbonyl)amino]-1-phenylethyl}(methyl)carbamoyl)amino]-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoic acid (60 mg, 0.077 mmol, 1 equiv), 3-methyl-1-(piperidin-4-yl)urea (24.10 mg) and TEA (23.26 mg, 0.231 mmol, 3 equiv) were dissolved in DMF (5 mL, 64.609 mmol, 843.12 equiv) at room temperature under nitrogen atmosphere, and HATU (43.71 mg, 0.115 mmol, 3 equiv) was added. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. The resulting mixture was purified by reversed-phase flash chromatography to give tert-butyl N-[(5R)-5-[(2R)-2-[(2R)-2-[({2-[(tert-butoxycarbonyl)amino]-1-phenylethyl)}(methyl)carbamoyl)amino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-{4-[(methylcarbamoyl)amino]piperidin-1-yl}-6-oxohexyl]carbamate (62 mg, 87.74%, purity: 100%).


LC-MS-8-7 (ES, m/z): [M+1]=922


8.8 Synthesis of Compound 8

tert-Butyl N-[(5R)-5-[(2R)-2-[(2R)-2-[({2-[(tert-butoxycarbonyl)amino]-1-phenylethyl}(methyl)carbamoyl) amino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-{4-[(methylcarbamoyl)amino]piperidin-1-yl}-6-oxohexyl]carbamate (62 mg, 0.067 mmol, 1 equiv) was dissolved in dioxane (10 mL), and a solution of HCl (gas) in 1,4-dioxane (5 mL) was added. The resulting mixture was stirred at room temperature for 2 h and The resulting mixture was concentrated under reduced pressure and purified by reversed-phase flash chromatography to give (2R)-2-[(2R)-2-{[(2-amino-1-phenylethyl)(methyl)carbamoyl)amino}-3-phenylpropionylamino]-N-[(2R)-6-amino-1-{4-[(methylcarbamoyl)amino]piperidin-1-yl}-1-oxohexyl-2-yl]-4-methylpentanamide (13.6 mg, 26.73%, purity: 95.0%).


LC-MS-8 (ES, m/z): [M+1]=722



1H NMR (400 MHz, Deuterium Oxide) δ 8.33 (s, 2H), 7.50-7.15 (m, 8H), 7.10 (d, J=7.2 Hz, 2H), 5.49 (dd, J=10.8, 5.2 Hz, 1H), 4.42 (dd, J=9.1, 6.2 Hz, 1H), 4.37-4.25 (m, 1H), 4.24-4.00 (m, 1H), 3.95-3.40 (m, 7H), 3.26-3.02 (m, 2H), 2.99-2.69 (m, 4H), 2.70-2.42 (m, 6H), 2.02-1.75 (m, 6H), 1.73-1.43 (m, 4H), 1.42-1.13 (m, 7H), 0.80 (ddd, J=15.3, 6.0, 2.9 Hz, 6H).


Example 9. Synthesis of Compound 9



embedded image


LC-MS-9 (ES, m/z): [M+1]=671.20



1H NMR (300 MHz, Deuterium Oxide) δ 8.37 (s, 1H), 7.43-6.81 (m, 10H), 4.49 (s, 2H), 4.46-3.55 (m, 6H), 3.60-2.94 (m, 5H), 3.03-2.52 (m, 6H), 1.83-1.10 (m, 9H), 0.78 (dd, J=14.0, 5.0 Hz, 6H).


Example 10. Synthesis of Compound 10



embedded image


LC-MS-10 (ES, m/z): [M+1]=699.20



1H NMR (300 MHz, Deuterium Oxide) δ 7.42-6.89 (m, 10H), 5.51 (dd, J=10.6, 4.8 Hz, 1H), 4.43 (dd, J=9.0, 6.3 Hz, 1H), 4.34-4.12 (m, 2H), 4.10-3.71 (m, 2H), 3.65-3.44 (m, 3H), 3.32 (d, J=19.3 Hz, 2H), 3.18 (d, J=17.1 Hz, 2H), 3.09 (dd, J=13.8, 6.1 Hz, 1H), 3.00-2.93 (m, 1H), 2.85 (t, J=7.6 Hz, 2H), 2.57 (d, J=31.2 Hz, 4H), 1.82-1.43 (m, 7H), 1.42-1.16 (m, 2H), 0.82 (dd, J=12.3, 5.0 Hz, 6H).


Example 11. Synthesis of Compound 11



embedded image


LC-MS-11 (ES, m/z): [M+1]=695



1H NMR (400 MHz, Deuterium Oxide) δ 8.39 (s, 1H), 7.40-7.19 (m, 6H), 7.08 (ddt, J=27.2, 5.4, 1.7 Hz, 4H), 4.59-4.47 (m, 1H), 4.30 (ddd, J=28.1, 8.0, 5.5 Hz, 2H), 4.14 (dd, J=47.0, 13.4 Hz, 1H), 3.88 (dd, J=36.1, 13.4 Hz, 1H), 3.70-3.50 (m, 4H), 3.24 (q, J=13.9, 12.7 Hz, 1H), 2.99-2.70 (m, 7H), 2.01-1.80 (m, 2H), 1.76-1.55 (m, 4H), 1.48 (q, J=9.6, 8.5 Hz, 3H), 1.41-1.17 (m, 4H), 0.83 (ddd, J=16.8, 5.1, 2.1 Hz, 6H).


Example 12. Synthesis of Compound 12



embedded image


12.1 Synthesis of Compound 12-1

tert-Butyl 4-aminopiperidine-1-carboxylate (500 mg, 2.496 mmol, 1 equiv) was dissolved in DCM, and TEA (505.25 mg, 4.992 mmol, 2 equiv) was added. Methyl chloroformate (353.84 mg, 3.744 mmol, 1.5 equiv) was added portionwise at 0° C. under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 2 h and The reaction was quenched with water at room temperature. The resulting mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography and eluted with PE/EA (5:1). The fractions were concentrated to dryness by rotary evaporation to give tert-butyl 4-[(methoxycarbonyl)amino]piperidine-1-carboxylate (522 mg, 80.94%, purity: 100%).


LC-MS-12-1 (ES, m/z): [M+1]=259.32


12.2 Synthesis of Compound 12-2

tert-Butyl 4-[(methoxycarbonyl)amino]piperidine-1-carboxylate (552 mg, 2.137 mmol, 1 equiv) was added to a 4 M solution of HCl (gas) in 1,4-dioxane (5 mL) at room temperature. The resulting mixture was stirred at room temperature for 2 h and The reaction was quenched with water at room temperature. The resulting mixture was concentrated under reduced pressure to give methyl N-(piperidin-4-yl)carbamate (455 mg, crude). The crude product was directly used in the next step without further purification.


LC-MS-12-2 (ES, m/z): [M+1]=159.20


12.3 Synthesis of Compound 12-3

(2R)-6-[(tert-Butoxycarbonyl)amino]-2-[(2R)-2-[(2R)-2-[({2-[(tert-butoxycarbonyl)amino]-1-phenylethyl}(methyl)carbamoyl)amino]-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoic acid (70 mg, 0.089 mmol, 1 equiv), TEA (27.14 mg, 0.267 mmol, 3 equiv) and methyl N-(piperidin-4-yl)carbamate (21.22 mg, 0.134 mmol, 1.5 equiv) were added to DMF (1 mL), and HATU (50.99 mg, 0.134 mmol, 1.5 equiv) was added portionwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for another 2 h. The reaction was quenched with water at room temperature. The desired product could be detected by LCMS. The reaction solution was purified by reversed-phase flash chromatography to give tert-butyl N-[(5R)-5-[(2R)-2-[(2R)-2-[({2-[(tert-butoxycarbonyl)amino]-1-phenylethyl}(methyl)carbamoyl)amino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-{4-[(methoxycarbonyl)amino]piperidin-1-yl}-6-oxohexyl]carbamate (70 mg, 83.00%, purity: 100%).


LC-MS-12-3 (ES, m/z): [M+1]=924.1


12.4 Synthesis of Compound 12

tert-Butyl N-[(5R)-5-[(2R)-2-[(2R)-2-[({2-[(tert-butoxycarbonyl)amino]-1-phenylethyl}(methyl)carbamoyl)amino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-{4-[(methoxycarbonyl)amino]piperidin-1-yl}-6-oxohexyl]carbamate (70 mg, 0.076 mmol, 1 equiv) was added to DCM (4 mL) at room temperature under nitrogen atmosphere, and TFA (1 mL) was added dropwise. The resulting mixture was stirred at room temperature for another 3 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography, and the fractions were concentrated to dryness by rotary evaporation to give methyl N-{1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-{[(2-amino-1-phenylethyl)(methyl)carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidin-4-yl}carbamate (42.1 mg, 72.07%, purity: 93.9%).


LC-MS-12 (ES, m/z): [M+1]=723.15



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 2H), 7.58-6.75 (m, 10H), 5.52 (s, 1H), 4.79-4.73 (m, 1H), 4.50-3.99 (m, 3H), 3.87 (d, J=32.6 Hz, 1H), 3.57 (s, 6H), 3.35-3.03 (m, 2H), 3.02-2.68 (m, 4H), 2.48 (dd, J=39.8, 5.2 Hz, 3H), 1.90 (s, 2H), 1.74-1.07 (m, 11H), 1.01-0.61 (m, 6H).


Example 13. Synthesis of Compound 13



embedded image


13.1 Synthesis of Compound 13-1

Methyl 1-[(2R)-2-[(2R)-2-[(2R)-2-amino-3-phenylpropionylamino]-4-methylpentanoylamino]-6-[(tert-butoxycarbonyl)amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (100 mg, 0.134 mmol, 1.00 equiv) was added to a solution of tert-butyl N-[2-(benzylamino)ethyl]carbamate (100 mg, 0.399 mmol, 2.98 equiv), TEA (40.64 mg, 0.402 mmol, 3 equiv) and N,N′-carbonyldiimidazole (23.88 mg, 0.147 mmol) in ACN (3 mL)/DMF (3 mL) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. The resulting mixture was purified by reversed-phase flash chromatography, and the fractions were concentrated to dryness by rotary evaporation to give methyl 1-[(2R)-2-[(2R)-2-[(2R)-2-{[benzyl({2-[(tert-butoxycarbonyl)amino]ethyl})carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]-6-[(tert-butoxycarbonyl)amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (120 mg, 87.59%, purity: 100%).


LC-MS-13-1 (ES, m/z): [M+1]=1024.28


13.2 Synthesis of Compound 13-2

Methyl 1-[(2R)-2-[(2R)-2-[(2R)-2-{[benzyl({2-[(tert-butoxycarbonyl)amino]ethyl})carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]-6-[(tert-butoxycarbonyl)amino]hexanoyl]-4-[(tert-butoxycarbonyl)amino]piperidine-4-carboxylate (120 mg, 0.117 mmol, 1 equiv) was dissolved in DCM (1 mL, 5.612 mmol), and TFA (1 mL) was added. The solution was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography to give methyl 4-amino-1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-{[2-aminoethyl(benzyl)carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylate (75 mg, 88.47%, purity: 100%).


LC-MS-13-2 (ES, m/z): [M+1]=723.93



1H NMR (400 MHz, Deuterium Oxide) δ 7.46-7.09 (m, 6H), 6.97 (dt, J=14.3, 5.4 Hz, 4H), 4.52-4.36 (m, 2H), 4.31 (d, J=17.5 Hz, 1H), 4.19 (p, J=4.9, 4.1 Hz, 1H), 4.07 (d, J=14.1 Hz, 1H), 3.92 (d, J=14.4 Hz, 1H), 3.73 (dt, J=12.4, 2.2 Hz, 3H), 3.64 (s, 1H), 3.60-3.31 (m, 3H), 3.30-3.16 (m, 1H), 3.10-2.89 (m, 3H), 2.87-2.57 (m, 3H), 2.36-2.06 (m, 2H), 1.93-1.47 (m, 6H), 1.48-1.18 (m, 5H), 0.92-0.40 (m, 6H).


13.3 Synthesis of Compound 13

Methyl 4-amino-1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-{[(2-aminoethyl)(benzyl)carbamoyl]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylate (75 mg, 0.104 mmol, 1 equiv) was dissolved in THF (1 mL) and H2O (1 mL), and lithium hydroxide (5 mg, 0.209 mmol, 2.01 equiv) was added. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase flash chromatography, and the fractions were concentrated to dryness by rotary evaporation to give 4-amino-1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-{[aminoethyl(benzyl)carbamoyl]]amino}-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidine-4-carboxylic acid (25.5 mg, 29.28%, purity: 84.5%).


LC-MS-13 (ES, m/z): [M+1]=709.25



1H NMR (300 MHz, Deuterium Oxide) δ 8.33 (s, 2H), 7.33-7.15 (m, 6H), 7.00 (ddt, J=12.9, 5.2, 2.6 Hz, 4H), 4.46-4.21 (m, 4H), 3.76-3.36 (m, 6H), 3.13-2.51 (m, 6H), 2.25-1.88 (m, 2H), 1.96-1.49 (m, 6H), 1.50-1.10 (m, 5H), 0.78 (dd, J=14.4, 5.4 Hz, 6H).


Example 14. Synthesis of Compound 14



embedded image


LC-MS-14 (ES, m/z): [M+1]=723.25



1H NMR (300 MHz, Deuterium Oxide) δ 8.33 (s, 3H), 7.45-6.79 (m, 10H), 5.62-5.41 (m, 1H), 4.49-4.19 (m, 2H), 4.05-3.35 (m, 8H), 3.35-2.59 (m, 5H), 2.45 (dd, J=30.9, 2.3 Hz, 3H), 2.20 (t, J=16.0 Hz, 2H), 2.00-1.10 (m, 11H), 0.81 (dt, J=11.9, 5.9 Hz, 6H).


Example 15. Synthesis of Compound 15



embedded image


LC-MS-15 (ES, m/z): [M+1]=751



1H NMR (400 MHz, Deuterium Oxide) δ 8.32 (s, 2H), 7.52-6.72 (m, 10H), 5.70-5.31 (m, 2H), 4.48-4.09 (m, 3H), 4.11-3.34 (m, 7H), 3.29-3.14 (m, 1H), 3.14-2.68 (m, 5H), 2.49 (d, J=5.1 Hz, 2H), 2.44-2.28 (m, 1H), 2.17 (d, J=18.8 Hz, 2H), 1.93-1.34 (m, 10H), 1.20 (dd, J=9.0, 6.3 Hz, 8H), 1.00-0.59 (m, 6H).


Example 16. Synthesis of Compound 16



embedded image


16.1 Synthesis of Compound 16-1

Compound 1-3 (500 mg, 0.898 mmol, 1 equiv) was dissolved in DCM (5 mL), and then piperidine (0.5 mL) was added dropwise to the system described above. The resulting mixture was stirred at room temperature for 3 h. The reaction solution was then concentrated to dryness by rotary evaporation to give a yellow crude product. The resulting crude product was purified by reversed-phase column chromatography (conditions were as follows: C18 chromatographic column, mobile phase: solvent ACN and solvent H2O (FA, 0.1%), gradient 10% to 50% for 10 min, UV254 nanometer detector), and the fractions were concentrated to dryness by rotary evaporation to give compound 16-1 (250 mg, 83.22%, purity: 99%).


LC-MS-16-1 (ES, m/z): [M+1]=335


16.2 Synthesis of Compound 16-3

CDI (791.55 mg, 4.882 mmol, 1.1 equiv) was added portionwise to a solution of compound 16-1 (1482 mg, 4.438 mmol, 1 equiv) and compound 16-2 (711 mg, 4.438 mmol, 1.00 equiv) in ACN (2 mL)/DMF (0.4 mL) at 0° C. under nitrogen atmosphere. The resulting mixture was stirred at room temperature overnight. The reaction solution was then concentrated to dryness by rotary evaporation to give a crude product as a yellow oil. The resulting crude product was purified by reversed-phase column chromatography (conditions were as follows: C18 chromatographic column, mobile phase: solvent ACN and solvent H2O (FA, 0.1%), gradient 10% to 50% for 10 min, UV254 nanometer detector), and the fractions were concentrated to dryness by rotary evaporation to give compound 16-3 (950 mg, 35.05%, purity: 99%).


LC-MS-16-3 (ES, m/z): [M+1]=611


16.3 Synthesis of Compound 16-4

Compound 16-3 (760 mg, 1.244 mmol, 1 equiv) was dissolved in DCM (5 mL), and then TFA (5 mL) was added dropwise to the system described above. The resulting mixture was stirred at room temperature for 2 h. The reaction solution was then concentrated to dryness by rotary evaporation to give compound 16-4 (800 mg, crude).


LC-MS-16-4 (ES, m/z): [M+1]=455


16.4 Synthesis of Compound 16-5

Compound 16-4 (600 mg, 1.320 mmol, 1 equiv) was dissolved in THF (20 mL) at room temperature, and then TEA (267.13 mg, 2.640 mmol, 2 equiv) and Boc2O (432.11 mg, 1.980 mmol, 1.5 equiv) were added dropwise to the system described above. The resulting mixture was stirred at room temperature for 2 h. The reaction solution was then concentrated to dryness by rotary evaporation to give a crude compound. The resulting crude product was purified by silica gel column chromatography (PE/EA (1/1)), and the fractions were concentrated to dryness by rotary evaporation to give compound 16-5 (600 mg, 81.95%, purity: 99%).


LC-MS-16-5 (ES, m/z): [M+1]=555


16.5 Synthesis of Compound 16-7

Compound 16-5 (600 mg, 1.320 mmol, 1 equiv), compound 16-6 (1975.52 mg, 4.328 mmol, 4 equiv), HOBt (292.33 mg, 2.164 mmol, 2 equiv), DMF (50 mL), DIPEA (279.33 mg, 2.164 mmol, 2 equiv) and TBTU (694.64 mg, 2.164 mmol, 2 equiv) were added to a 100 mL single-necked flask at room temperature. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. The reaction solution was filtered. The filter residue was washed with methanol and diethyl ether, and concentrated to dryness by rotary evaporation to give compound 16-7 (2.6 g, crude).


16.6 Synthesis of Compound 16-8

Compound 16-7 (2.6 g, 2.618 mmol, 1 equiv), DCM (25 mL, 393.265 mmol, 150.24 equiv) and TFA (25 mL, 336.577 mmol, 128.58 equiv) were added to a 100 mL single-necked flask at room temperature. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. The reaction solution was filtered. The filter residue was washed with TFA/DCM (v/v, 1/1), and the filtrate was concentrated to dryness by rotary evaporation to give compound 16-8 (1 g, crude).


LC-MS-16-8 (ES, m/z): [M+1]=583


16.7 Synthesis of Compound 16-9

Compound 16-8 (1 g, 1.716 mmol, 1 equiv) was dissolved in THF (20 mL) at room temperature, and then TEA (0.35 g, 3.432 mmol, 2 equiv) and Boc2O (0.75 g, 3.432 mmol, 2 equiv) were added dropwise to the system described above. The resulting mixture was stirred at room temperature for 2 h. The reaction solution was then concentrated to dryness by rotary evaporation to give a crude product of compound 10. The resulting crude product was purified by silica gel column chromatography and eluted with PE/EA (1/1), and the fractions were concentrated to dryness by rotary evaporation to give compound 16-9 (700 mg, 52.10%, purity: 99%).


LC-MS-16-9 (ES, m/z): [M+1]=783


16.8 Synthesis of Compound 16-11

Compound 16-9 (60 mg, 0.076 mmol, 1 equiv), compound 16-10 (18.18 mg, 0.115 mmol, 1.5 equiv), TEA (15.51 mg, 0.154 mmol, 2 equiv), DMF (2 mL) and HATU (43.71 mg, 0.115 mmol, 1.5 equiv) were added to a 50 mL single-necked flask at room temperature. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. The reaction solution was purified by reversed-phase column chromatography (conditions were as follows: C18 chromatographic column, mobile phase: solvent ACN and solvent H2O (0.1% FA), gradient 10% to 50% for 10 min, UV254 nanometer detector), and the fractions were concentrated to dryness by rotary evaporation to give compound 16-11 (50 mg, 70.68%, purity: 99%).


LC-MS-16-11 (ES, m/z): [M+1]=923


16.9 Synthesis of Compound 16

Compound 16-11 (60 mg, 0.065 mmol, 1 equiv), DCM (4 mL) and TFA (4 mL) were added to a 50 mL single-necked flask at room temperature. The resulting mixture was stirred at room temperature for 2 h. The reaction solution was concentrated to dryness by rotary evaporation. The crude product was purified by reversed-phase column chromatography (conditions were as follows: C18 chromatographic column, mobile phase: solvent ACN and solvent H2O (0.1% FA), gradient 10% to 50% for 10 min, UV254 nanometer detector), and the fractions were concentrated to dryness by rotary evaporation to give compound 16 (25 mg, 53.21%, purity: 99.8%).


LC-MS-16 (ES, m/z): [M+1]=723



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 2H), 7.49-7.17 (m, 6H), 7.06 (q, J=14.0, 8.1 Hz, 4H), 4.58-4.33 (m, 3H), 4.33-3.99 (m, 2H), 3.87 (dd, J=36.1, 13.2 Hz, 1H), 3.74-3.37 (m, 6H), 3.21 (t, J=13.4 Hz, 1H), 3.13-2.68 (m, 7H), 2.02-1.77 (m, 2H), 1.80-1.16 (m, 11H), 0.82 (dd, J=18.2, 5.7 Hz, 6H).


Example 17. Synthesis of Compound 17

According to the similar preparation procedures as in Example 16, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-17 (ES, m/z): [M+1]=721



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 2H), 7.32 (d, J=7.3 Hz, 2H), 7.30-7.19 (m, 4H), 7.11 (d, J=7.0 Hz, 1H), 7.05 (dd, J=10.4, 6.5 Hz, 4H), 4.49 (d, J=16.9 Hz, 2H), 4.45-4.35 (m, 1H), 4.27 (s, 1H), 3.83 (t, J=13.7 Hz, 1H), 3.58-3.41 (m, 2H), 3.26-3.13 (m, 1H), 3.02 (did, J=19.7, 11.6, 4.7 Hz, 3H), 2.96-2.87 (m, 3H), 2.87-2.75 (m, 2H), 2.62-2.56 (m, 2H), 2.54 (s, 1H), 1.87 (dd, J=19.6, 8.9 Hz, 2H), 1.70-1.56 (m, 4H), 1.50-1.43 (m, 2H), 1.40-1.28 (m, 3H), 0.89-0.72 (m, 6H).


Example 18. Synthesis of Compound 18



embedded image


LC-MS-18 (ES, m/z): [M+1]=709



1H NMR (400 MHz, Deuterium Oxide) δ 8.32 (s, 1H), 8.29 (s, 1H), 8.08 (d, J=4.9 Hz, 1H), 7.46-7.35 (m, 1H), 7.29 (dd, J=8.1, 5.1 Hz, 1H), 7.10 (dd, J=4.4, 2.2 Hz, 3H), 6.96 (t, J=4.3 Hz, 2H), 4.48 (s, 1H), 4.46-4.32 (m, 2H), 4.22 (t, J=7.0 Hz, 1H), 3.85-3.22 (m, 7H), 3.05-2.87 (m, 3H), 2.83 (t, J=7.7 Hz, 2H), 2.73 (ddd, J=14.7, 9.6, 5.7 Hz, 1H), 2.27-1.94 (m, 2H), 1.88-1.09 (m, 11H), 0.86-0.64 (m, 6H).


Example 19. Synthesis of Compound 19



embedded image


LC-MS-19 (ES, m/z): [M+1]=710



1H NMR (400 MHz, Deuterium Oxide) δ 8.95 (d, J=2.4 Hz, 1H), 8.37 (s, 2H), 8.35 (s, 1H), 7.09 (dq, J=45.5, 3.8, 3.3 Hz, 5H), 4.61 (d, J=18.1 Hz, 2H), 4.52-4.40 (m, 2H), 4.31 (t, J=6.7 Hz, 1H), 3.94-3.24 (m, 6H), 3.05 (dt, J=22.4, 6.0 Hz, 3H), 2.91 (t, J=7.7 Hz, 2H), 2.78 (ddd, J=14.1, 10.2, 7.5 Hz, 1H), 2.14 (ddd, J=35.9, 12.0, 5.5 Hz, 2H), 1.94-1.21 (m, 11H), 0.97-0.69 (m, 6H).


Example 20. Synthesis of Compound 20

According to the similar preparation procedures as in Example 16, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-20 (ES, m/z): [M+1]=713



1H NMR (400 MHz, Deuterium Oxide) δ 8.36 (s, 2H), 7.33 (s, 1H), 7.33-7.23 (m, 2H), 7.27-7.21 (m, 2H), 7.10-7.00 (m, 5H), 4.74 (s, 1H), 4.54-4.44 (m, 2H), 4.39 (d, J=17.5 Hz, 1H), 4.28 (s, 1H), 4.20 (d, J=15.1 Hz, 1H), 4.05 (s, 1H), 3.98-3.76 (m, 1H), 3.72 (d, J=14.2 Hz, 1H), 3.63-3.55 (m, 1H), 3.48 (dd, J=13.9, 7.6 Hz, 2H), 3.36 (s, 2H), 3.32-3.21 (m, 1H), 3.02 (dq, J=20.9, 5.7 Hz, 6H), 2.98-2.80 (m, 1H), 2.73-2.64 (m, 2H), 1.68 (d, J=6.1 Hz, 1H), 1.62 (s, 5H), 1.47 (s, 4H), 1.35 (d, J=12.0 Hz, 1H), 0.85-0.80 (d, J=5.5 Hz, 6H).


Example 21. Synthesis of Compound 21



embedded image


21.1 Synthesis of Compound 21-2

Compound 21-1 (34.53 mg, 0.256 mmol, 4 equiv) was added to a solution of intermediate 3 (50 mg, 0.064 mmol, 1 equiv) in DMF (10 mL) at room temperature under nitrogen atmosphere. After the resulting mixture was stirred for 2 h, HATU (36.42 mg, 0.096 mmol, 1.5 equiv) and DIPEA (16.51 mg, 0.128 mmol, 2 equiv) were added dropwise at room temperature. The resulting mixture was extracted with EA (3×100 mL). The organic layer was washed with brine (3×10 mL) and dried over Na2SO4. The resulting filtrate was concentrated under reduced pressure. The resulting crude product was purified by reversed-phase column chromatography (conditions were as follows: C18 chromatographic column, mobile phase: solvent ACN and solvent H2O (0.1% FA), gradient 10% to 50% for 10 min, UV200 nanometer detector) to give compound 21-2 (58 mg, crude).


LC-MS-21-2 (ES, m/z): [M+1]=900


21.2 Synthesis of Compound 21

A mixed solution of compound 21-2 (40 mg, 0.044 mmol, 1 equiv) in TFA (1 mL)/DCM (4 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting mixture filtrate was concentrated under reduced pressure. The resulting crude product was purified by reversed-phase column chromatography (conditions were as follows: C18 chromatographic column, mobile phase: solvent ACN and solvent H2O (0.1% FA), gradient 10% to 50% for 10 min, UV200 nanometer detector), and the fractions were concentrated to dryness by rotary evaporation to give compound 21 (18.6 mg, 57.47%, purity: 96.0%).


LC-MS-21 (ES, m/z): [M+1]=700



1H NMR (400 MHz, Deuterium Oxide) δ 8.36 (s, 2H), 7.31 (d, J=7.3 Hz, 3H), 7.24 (q, J=3.6, 3.1 Hz, 3H), 7.05 (dd, J=12.4, 5.8 Hz, 4H), 4.74 (s, 2H), 4.49 (dt, J=13.3, 4.4 Hz, 2H), 4.39 (d, J=17.5 Hz, 1H), 4.28 (s, 3H), 4.19 (d, J=14.8 Hz, 1H), 3.88 (q, J=15.4, 14.7 Hz, 1H), 3.70 (d, J=15.5 Hz, 1H), 3.67-3.51 (m, 1H), 3.48 (s, 1H), 3.28 (d, J=16.0 Hz, 1H), 3.19 (s, 2H), 3.09-2.96 (m, 3H), 2.87 (dt, J=27.6, 8.3 Hz, 3H), 1.69 (t, J=7.4 Hz, 2H), 1.62 (q, J=7.5 Hz, 2H), 1.47 (s, 4H), 1.35 (dt, J=16.9, 8.0 Hz, 2H), 0.83 (dt, J=19.1, 3.9 Hz, 6H).


Example 22. Synthesis of Compound 22



embedded image


LC-MS-22 (ES, m/z): [M+1]=784



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 1H), 8.11 (s, 1H), 7.61 (t, J=7.9 Hz, 1H), 7.40 (d, J=3.4 Hz, 1H), 7.31-7.19 (m, 5H), 7.18-6.99 (m, 5H), 4.53-4.38 (m, 2H), 4.36 (s, 3H), 4.28 (t, J=7.6 Hz, 1H), 4.00 (d, J=13.8 Hz, 1H), 3.89 (s, 1H), 3.61 (d, J=13.3 Hz, 1H), 3.55 (s, 1H), 3.47 (ddd, J=21.8, 14.8, 6.7 Hz, 3H), 3.18-2.99 (m, 3H), 2.95-2.82 (m, 5H), 2.17 (s, 3H), 1.99 (d, J=17.9 Hz, 3H), 1.92 (s, 2H), 1.62 (p, J=6.8 Hz, 2H), 1.47 (d, J=7.3 Hz, 1H), 1.37 (s, 4H), 1.20 (t, J=7.3 Hz, 1H), 0.82 (dt, J=16.5, 6.7 Hz, 6H), 0.67 (d, J=6.2 Hz, 1H).


Example 23. Synthesis of Compound 23



embedded image


LC-MS-23 (ES, m/z): [M+1]=840.46



1H NMR (400 MHz, Deuterium Oxide) δ 8.29 (s, 2H), 7.31-7.08 (m, 6H), 7.00 (dd, J=35.0, 7.4 Hz, 4H), 4.80 (s, 1H), 4.48-3.95 (m, 6H), 3.54-3.35 (m, 3H), 3.28-2.66 (m, 12H), 2.15 (s, 4H), 1.79 (s, 3H), 1.65-1.15 (m, 11H), 0.83-0.55 (m, 6H).


Example 24. Synthesis of Compound 24

According to the similar preparation procedures as in Example 16, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-24 (ES, m/z): [M+1]=793



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 2H), 7.31 (d, J=7.4 Hz, 2H), 7.30-7.21 (m, 3H), 7.12 (d, J=6.6 Hz, 1H), 7.04 (t, J=9.5 Hz, 4H), 4.48 (d, J=17.6 Hz, 3H), 4.39 (d, J=17.8 Hz, 1H), 4.27 (s, 1H), 4.20 (d, J=12.5 Hz, 1H), 4.07 (d, J=13.1 Hz, 2H), 3.55 (s, 5H), 3.20 (d, J=13.5 Hz, 1H), 2.96 (s, 6H), 2.89 (s, 1H), 1.86 (s, 4H), 1.60 (s, 6H), 1.46 (d, J=6.9 Hz, 1H), 1.35 (s, 6H), 0.81-0.66 (dd, J=17.8, 5.7 Hz, 6H).


Example 25. Synthesis of Compound 25



embedded image


LC-MS-25 (ES, m/z): [M+1]=853



1H NMR (400 MHz, Deuterium Oxide) δ 8.38 (s, 2H), 7.32 (d, J=7.3 Hz, 3H), 7.32-7.20 (m, 3H), 7.05 (d, J=17.7 Hz, 1H), 7.05 (s, 3H), 4.27 (d, J=7.3 Hz, 1H), 3.53 (s, 4H), 3.41 (s, 1H), 3.31 (s, 1H), 3.03 (dd, J=15.0, 8.3 Hz, 3H), 2.89 (q, J=12.2, 9.9 Hz, 3H), 2.49 (t, J=12.5 Hz, 2H), 2.19-2.09 (m, 2H), 1.82 (s, 2H), 1.63 (s, 8H), 1.51-1.43 (m, 4H), 0.82 (dd, J=18.6, 5.9 Hz, 6H).


Example 26. Synthesis of Compound 26

According to the similar preparation procedures as in Example 16, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-26 (ES, m/z): [M+1]=758



1H NMR (400 MHz, Deuterium Oxide) δ 8.39 (s, 2H), 7.40-7.18 (m, 6H), 7.16-7.00 (m, 4H), 4.56-4.47 (m, 2H), 4.47-4.35 (m, 2H), 4.30 (dq, J=10.1, 5.2, 4.5 Hz, 1H), 4.20 (d, J=12.0 Hz, 1H), 4.10 (td, J=13.3, 11.5, 6.9 Hz, 1H), 3.58 (dd, J=15.1, 6.5 Hz, 1H), 3.54-3.43 (m, 1H), 3.37 (dp, J=11.2, 5.1 Hz, 1H), 3.28-3.14 (m, 1H), 3.10-2.94 (m, 3H), 2.92 (dd, J=7.8, 4.0 Hz, 1H), 2.90-2.77 (m, 2H), 2.56 (dd, J=11.6, 4.2 Hz, 3H), 2.00 (s, 1H), 1.94 (d, J=8.6 Hz, 1H), 1.75-1.55 (m, 4H), 1.55-1.40 (m, 4H), 1.40-1.27 (m, 3H), 0.82 (ddd, J=17.6, 6.3, 2.6 Hz, 4H), 0.74 (dd, J=6.8, 3.3 Hz, 1H), 0.67 (t, J=5.4 Hz, 1H).


Example 27. Synthesis of Compound 27



embedded image


LC-MS-27 (ES, m/z): [M+1]=736



1H NMR (400 MHz, Deuterium Oxide) δ 8.33 (s, 1H), 7.52-7.10 (m, 6H), 6.98 (q, J=4.8, 3.9 Hz, 4H), 4.57-4.26 (m, 4H), 4.21 (dd, J=9.8, 4.8 Hz, 1H), 3.71 (tt, J=13.0, 5.6 Hz, 3H), 3.63-3.31 (m, 3H), 3.25 (td, J=12.2, 8.6 Hz, 1H), 3.17-2.90 (m, 3H), 2.91-2.58 (m, 3H), 2.39-1.98 (m, 3H), 1.94-1.15 (m, 12H), 0.96-0.44 (m, 6H).


Example 28. Synthesis of Compound 28



embedded image


LC-MS-28 (ES, m/z): [M+1]=722



1H NMR (400 MHz, Deuterium Oxide) δ 8.38 (s, 2H), 7.41-7.18 (m, 6H), 7.14-6.91 (m, 4H), 4.58-4.22 (m, 5H), 3.95-3.35 (m, 7H), 3.16-2.78 (m, 7H), 2.61 (s, 3H), 2.33-2.01 (m, 3H), 1.94-1.25 (m, 12H), 0.84 (dd, J=18.5, 5.4 Hz, 6H).


Example 29. Synthesis of Compound 29



embedded image


LC-MS-29 (ES, m/z): [M+1]=734



1H NMR (400 MHz, Deuterium Oxide) δ 8.36 (s, 2H), 7.32 (d, J=7.5 Hz, 3H), 7.23 (dt, J=7.7, 4.0 Hz, 3H), 7.04 (d, J=7.1 Hz, 4H), 4.51 (s, 1H), 4.42 (dd, J=22.2, 16.8 Hz, 1H), 4.31-4.20 (m, 3H), 4.09-3.97 (m, 2H), 3.94 (d, J=10.0 Hz, 2H), 3.57 (s, 1H), 3.49 (dd, J=9.9, 5.5 Hz, 1H), 3.33 (dd, J=28.5, 14.0 Hz, 1H), 3.06 (d, J=7.0 Hz, 1H), 3.01 (s, 3H), 2.95-2.80 (m, 3H), 1.88 (d, J=9.0 Hz, 2H), 1.83-1.72 (m, 1H), 1.72-1.60 (m, 4H), 1.48 (s, 3H), 1.34 (s, 3H), 0.83 (dd, J=19.5, 5.7 Hz, 6H).


Example 30. Synthesis of Compound 30



embedded image


LC-MS-30-0 (ES, m/z): [M+1]=749



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 1H), 7.68-6.76 (m, 10H), 5.47 (d, J=11.3 Hz, 1H), 4.61-4.18 (m, 2H), 3.78-3.24 (m, 6H), 3.21-2.76 (m, 3H), 2.70-2.33 (m, 5H), 2.11 (dd, J=14.2, 7.6 Hz, 2H), 1.95-1.15 (m, 14H), 1.15-0.39 (m, 6H).


LC-MS-30-100 (ES, m/z): [1M+1]=749



1H NMR (400 MHI-z, Deuterium Oxide) δ 8.37 (s, 1H), 7.57-6.76 (in, 10H), 5.51 (d, J=11.1 Hz, 1H), 4.35 (d, J=6.8 Hz, 1H), 3.75-3.12 (m, 6H), 3.06-2.67 (m, 3H), 2.46 (d, J=17.8 Hz, 5H), 2.12 (dd, J=13.7, 7.4 Hz, 2H), 2.00-1.18 (m, 12H), 0.85 (dd, J=17.1, 5.1 Hz, 6H).


Example 31. Synthesis of Compound 31



embedded image


LC-MS-31 (ES, m/z): [M+1]=709



1H NMR (400 MHz, Deuterium Oxide) δ 8.34 (s, 2H), 7.76-6.68 (m, 10H), 5.15-4.80 (m, 2H), 4.56-3.67 (m, 6H), 3.61 (d, J=16.4 Hz, 4H), 3.30-2.97 (m, 4H), 3.11-2.70 (m, 4H), 2.19-1.79 (m, 2H), 1.76-1.06 (m, 11H), 0.95-0.39 (m, 6H).


Example 32. Synthesis of Compound 32

According to the similar preparation procedures as in Example 8, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-32 (ES, m/z): [M+1]=782



1H NMR (400 MHz, Deuterium Oxide) δ 8.51 (s, 1H), 8.36 (s, 1H), 7.93 (s, 1H), 7.57 (s, 1H), 7.35 (d, J=7.2 Hz, 2H), 7.31-7.26 (m, 4H), 7.22 (d, J=7.7 Hz, 2H), 7.13 (d, J=7.4 Hz, 1H), 6.89 (d, J=6.8 Hz, 1H), 5.57-5.49 (m, 1H), 4.49-4.41 (m, 1H), 4.33 (s, 2H), 4.24-4.16 (m, 1H), 3.94 (s, 1H), 3.69-3.50 (m, 1H), 3.49 (s, 3H), 3.24 (dd, J=13.7, 5.2 Hz, 1H), 3.07 (s, 1H), 3.00-2.78 (m, 2H), 2.53 (d, J=8.6 Hz, 3H), 2.43 (d, J=2.8 Hz, 3H), 1.87 (s, 2H), 1.68-1.58 (m, 2H), 1.57 (s, 6H), 1.52 (s, 1H), 1.35 (s, 3H), 0.90-0.78 (m, 2H), 0.83 (s, 5H).


Example 33. Synthesis of Compound 33

According to the similar preparation procedures as in Example 8, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-33 (ES, m/z): [M+1]=793



1H NMR (400 MHz, Deuterium Oxide) δ 8.36 (s, 2H), 7.59-7.10 (m, 9H), 6.89 (d, J=6.7 Hz, 1H), 5.54 (dd, J=11.4, 4.5 Hz, 1H), 4.56-4.25 (m, 2H), 4.25-4.03 (m, 1H), 3.96-3.78 (m, 3H), 3.72-3.42 (m, 5H), 3.34-3.05 (m, 2H), 3.05-2.73 (m, 4H), 2.49 (d, J=39.1 Hz, 3H), 1.87 (s, 4H), 1.57 (d, J=31.1 Hz, 9H), 1.34 (dd, J=24.6, 11.5 Hz, 4H), 0.85 (dt, J=13.0, 7.0 Hz, 6H).


Example 34. Synthesis of Compound 34



embedded image


LC-MS-34 (ES, m/z): [M+1]=712



1H NMR (400 MHz, Deuterium Oxide) δ 8.26 (s, 1H), 7.28-7.18 (m, 3H), 7.15 (td, J=4.4, 1.9 Hz, 3H), 7.01-6.90 (m, 4H), 4.67 (s, 1H), 4.47-4.31 (m, 3H), 4.31-4.18 (m, 2H), 3.81 (s, 1H), 3.51 (dt, J=15.1, 5.8 Hz, 1H), 3.35 (ddd, J=28.0, 13.3, 7.3 Hz, 2H), 3.02-2.87 (m, 4H), 2.80 (dddd, J=23.1, 13.9, 9.1, 4.7 Hz, 4H), 2.03-1.77 (m, 4H), 1.56 (ddt, J=20.7, 8.8, 4.7 Hz, 4H), 1.44-1.20 (m, 5H), 0.74 (ddd, J=15.8, 5.2, 2.4 Hz, 6H).


Example 35. Synthesis of Compound 35

According to the similar preparation procedures as in Example 16, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-35 (ES, m/z): [M+1]=675



1H NMR (400 MHz, Deuterium Oxide) δ 8.29 (s, 2H), 7.18 (dtd, J=29.2, 8.0, 7.1, 2.7 Hz, 6H), 7.08-6.88 (m, 4H), 4.49-4.11 (m, 4H), 3.62-3.23 (m, 5H), 3.16-2.71 (m, 8H), 1.88-1.14 (m, 14H), 0.78-0.53 (m, 6H).


Example 36. Synthesis of Compound 36

According to the similar preparation procedures as in Example 8, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-36 (ES, m/z): [M+1]=700



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 1H), 7.43-7.17 (m, 8H), 7.13 (d, J=7.4 Hz, 1H), 6.91-6.85 (m, 1H), 5.56-5.49 (m, 1H), 4.45 (dd, J=9.0, 6.2 Hz, 1H), 4.29 (d, J=8.4 Hz, 2H), 4.17 (d, J=15.9 Hz, 1H), 3.94-3.84 (m, 1H), 3.67 (dd, J=25.3, 12.6 Hz, 1H), 3.61-3.45 (m, 2H), 3.35 (t, J=11.9 Hz, 1H), 3.28 (s, 1H), 3.20 (s, 5H), 3.18-3.06 (m, 1H), 3.01-2.72 (m, 3H), 2.54 (s, 2H), 2.43 (s, 1H), 1.63 (s, 1H), 1.61-1.49 (m, 4H), 1.34 (t, J=7.9 Hz, 1H), 0.85 (ddd, J=15.5, 9.9, 5.4 Hz, 6H).


Example 37. Synthesis of Compound 37

According to the similar preparation procedures as in Example 8, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-37 (ES, m/z): [M+1]=652



1H NMR (400 MHz, Deuterium Oxide) δ 8.36 (s, 2H), 7.54-6.66 (m, 10H), 5.52 (dd, J=11.4, 5.7 Hz, 1H), 4.64-4.13 (m, 2H), 3.91-3.30 (m, 10H), 3.35-2.73 (m, 4H), 2.48 (d, J=40.5 Hz, 3H), 1.95-1.22 (m, 9H), 0.84 (dt, J=15.4, 7.6 Hz, 6H).


Example 38. Synthesis of Compound 38



embedded image


LC-MS-38 (ES, m/z): [M+1]=651



1H NMR (400 MHz, Deuterium Oxide) δ 8.38 (s, 1H), 7.57-6.68 (m, 10H), 5.52 (dd, J=11.0, 4.5 Hz, 1H), 4.70-4.10 (m, 2H), 4.13-3.32 (m, 6H), 3.27-2.73 (m, 8H), 2.73-2.30 (m, 3H), 1.90-1.41 (m, 9H), 1.44-1.01 (m, 3H), 1.01-0.42 (m, 6H).


Example 39. Synthesis of Compound 39

According to the similar preparation procedures as in Example 16, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-39 (ES, m/z): [M+1]=652



1H NMR (400 MHz, Deuterium Oxide) δ 7.68-6.57 (m, 10H), 4.50-3.83 (m, 4H), 3.77-3.21 (m, 10H), 3.16-2.57 (m, 6H), 1.76-1.03 (m, 9H), 0.98-0.23 (m, 6H).


Example 40. Synthesis of Compound 40



embedded image


LC-MS-40 (ES, m/z): [M+1]=651



1H NMR (400 MHz, Deuterium Oxide) δ 8.36 (s, 1H), 7.53-7.15 (m, 6H), 7.08-6.79 (m, 4H), 4.58-4.15 (m, 5H), 3.96-3.34 (m, 6H), 3.34-2.77 (m, 10H), 1.77-21.22 (m, 9H), 0.80 (dd, J=19.9, 5.3 Hz, 6H).


Example 41. Synthesis of Compound 41



embedded image


LC-MS-41 (ES, m/z): [M+1]=723



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 2H), 7.32 (q, J=7.1 Hz, 3H), 7.26-7.20 (m, 3H), 7.06 (t, J=4.7 Hz, 4H), 4.54-4.46 (m, 2H), 4.40 (d, J=17.4 Hz, 2H), 4.28 (d, J=6.8 Hz, 1H), 4.01 (s, 2H), 3.67-3.54 (m, 4H), 3.48 (dt, J=15.2, 6.8 Hz, 1H), 3.28-3.11 (m, 3H), 2.95 (dqd, J=44.1, 15.2, 14.4, 6.9 Hz, 6H), 2.67 (t, J=13.1 Hz, 1H), 1.87-1.59 (m, 6H), 1.48 (t, J=7.5 Hz, 3H), 1.26 (dt, J=13.0, 6.8 Hz, 3H), 0.83 (dq, J=18.4, 3.4 Hz, 6H).


Example 42. Synthesis of Compound 42



embedded image


LC-MS-42 (ES, m/z): [M+1]=666



1H NMR (400 MHz, Deuterium Oxide) δ 8.38 (s, 1H), 7.32 (q, J=7.0 Hz, 3H), 7.23 (q, J=3.8 Hz, 3H), 7.05 (t, J=5.2 Hz, 4H), 4.53-4.45 (m, 2H), 4.40 (d, J=17.5 Hz, 1H), 4.28 (t, J=6.9 Hz, 1H), 4.21-4.09 (m, 1H), 3.84 (d, J=16.9 Hz, 1H), 3.57 (t, J=8.5 Hz, 5H), 3.45 (dq, J=15.4, 5.4, 5.0 Hz, 1H), 3.23 (d, J=13.5 Hz, 1H), 3.03 (dd, J=13.0, 6.4 Hz, 1H), 2.97 (d, J=6.6 Hz, 2H), 2.91-2.79 (m, 1H), 2.86 (s, 1H), 1.93 (d, J=12.6 Hz, 1H), 1.86 (d, J=15.1 Hz, 1H), 1.48 (s, 2H), 1.49-1.44 (m, 2H), 1.24 (q, J=7.0 Hz, 4H), 0.88-0.77 (m, 6H).


Example 43. Synthesis of Compound 43



embedded image



1H NMR (400 MHz, Deuterium Oxide) δ 8.35 (s, 2H), 7.35 (dd, J=12.2, 5.2 Hz, 3H), 7.30-6.81 (m, 7H), 5.70-5.42 (m, 1H), 4.46-4.28 (m, 2H), 4.12-3.83 (m, 2H), 3.61 (d, J=11.6 Hz, 3H), 3.50 (q, J=11.8, 9.2 Hz, 2H), 3.30-3.02 (m, 4H), 2.91 (h, J=10.1, 9.2 Hz, 3H), 2.70-2.50 (m, 3H), 2.43 (d, J=4.9 Hz, 1H), 2.00-1.63 (m, 5H), 1.60-1.44 (m, 3H), 1.37-0.98 (m, 3H), 0.95-0.50 (m, 6H).


Example 44. Synthesis of Compound 44



embedded image


LC-MS-44 (ES, m/z): [M+1]=706



1H NMR (400 MHz, Deuterium Oxide) δ 8.28 (s, 2H), 7.28-7.09 (m, 6H), 7.07-6.90 (m, 4H), 4.49-4.10 (m, 5H), 4.04-3.79 (m, 2H), 3.59-3.28 (m, 2H), 3.13 (ddt, J=36.4, 14.4, 7.9 Hz, 3H), 2.99-2.67 (m, 6H), 2.68-2.52 (m, 2H), 2.13-1.94 (m, 3H), 1.86-1.02 (m, 12H), 0.84-0.51 (m, 6H).


Example 45. Synthesis of Compound 45

According to the similar preparation procedures as in Example 16, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-45 (ES, m/z): [M+1]=708



1H NMR (400 MHz, Deuterium Oxide) δ 8.29 (s, 1H), 7.29-7.07 (m, 6H), 7.07-6.91 (m, 4H), 4.46-4.25 (m, 3H), 4.25-3.93 (m, 2H), 3.90-3.58 (m, 2H), 3.58-3.29 (m, 2H), 3.13 (t, J=13.6 Hz, 1H), 3.02-2.64 (m, 7H), 1.79 (dd, J=19.2, 5.9 Hz, 5H), 1.64-1.08 (m, 11H), 0.79-0.54 (m, 6H).


Example 46. Synthesis of Compound 46

According to similar preparation procedures as in Example 16, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-46 (ES, m/z): [M+1]=709



1H NMR (400 MHz, Deuterium Oxide) δ 8.34 (d, J=2.0 Hz, 2H), 7.33-7.17 (m, 6H), 7.01 (dq, J=13.1, 5.3, 4.5 Hz, 4H), 4.52-4.41 (m, 2H), 4.35 (dd, J=17.6, 4.9 Hz, 1H), 4.25 (dd, J=8.8, 5.9 Hz, 1H), 3.61 (d, J=3.0 Hz, 6H), 3.50 (t, J=6.6 Hz, 3H), 3.38 (s, 1H), 3.01 (dt, J=14.3, 4.5 Hz, 1H), 2.96-2.76 (m, 5H), 1.68-1.52 (m, 4H), 1.43 (q, J=10.1, 8.0 Hz, 3H), 1.35-1.25 (m, 2H), 0.84-0.74 (m, 6H).


Example 47. Synthesis of Compound 47

According to similar preparation procedures as in Example 16, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-47 (ES, m/z): [M+1]=693



1H NMR (400 MHz, Deuterium Oxide) δ 8.33 (s, 2H), 7.31-7.14 (m, 6H), 7.00 (dd, J=11.1, 5.8 Hz, 4H), 4.47 (dd, J=13.2, 7.5 Hz, 2H), 4.38-4.22 (m, 2H), 3.68-3.51 (m, 5H), 3.38 (tdt, J=24.0, 18.3, 9.2 Hz, 5H), 3.05-2.79 (m, 6H), 2.03 (t, J=6.9 Hz, 3H), 1.70-1.53 (m, 4H), 1.43 (hept, J=6.6, 5.9 Hz, 3H), 1.36-1.22 (m, 2H), 0.79 (dd, J=18.2, 5.9 Hz, 6H).


Example 48. Synthesis of Compound 48



embedded image


LC-MS-48 (ES, m/z): [M+1]=673.43



1H NMR (400 MHz, Deuterium Oxide) δ 7.28-7.14 (m, 5H), 4.70 (s, 1H), 4.65 (s, 2H), 4.43 (dd, J=9.5, 5.7 Hz, 1H), 4.28-4.21 (m, 1H), 3.56 (s, 2H), 3.35 (dt, J=14.1, 7.2 Hz, 2H), 3.13-2.89 (m, 2H), 2.87 (s, 2H), 2.86 (d, J=7.6 Hz, 2H), 2.85-2.75 (m, 2H), 1.98 (dd, J=13.5, 6.8 Hz, 1H), 1.87 (d, J=9.5 Hz, 1H), 1.55 (dq, J=15.0, 7.6 Hz, 6H), 1.45 (s, 5H), 0.77 (dd, J=17.0, 5.1 Hz, 7H), 0.66-0.59 (m, 1H), 0.33 (d, J=7.8 Hz, 2H).


Example 49. Synthesis of Compound 49



embedded image


LC-MS-49 (ES, m/z): [M+1]=700



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 2H), 7.31 (t, J=7.3 Hz, 2H), 7.23 (dd, J=17.4, 7.3 Hz, 3H), 4.76 (d, J=6.2 Hz, 1H), 4.69 (s, 1H), 4.49 (dd, J=9.7, 5.4 Hz, 1H), 4.30 (d, J=8.6 Hz, 1H), 3.74 (s, 2H), 3.56 (ddd, J=29.1, 13.5, 7.1 Hz, 2H), 3.34 (ddt, J=16.1, 10.9, 5.8 Hz, 1H), 3.14 (dd, J=14.4, 6.6 Hz, 2H), 2.93 (dq, J=14.1, 6.5 Hz, 6H), 2.22-2.11 (m, 1H), 2.09 (s, 1H), 1.72 (s, 5H), 1.62 (t, J=7.7 Hz, 2H), 1.47 (s, 10H), 1.37 (s, 1H), 0.94 (s, 1H), 0.84 (dd, J=17.7, 5.3 Hz, 7H).


Example 50. Synthesis of Compound 50



embedded image


LC-MS-50 (ES, m/z): [M+1]=726



1H NMR (400 MHz, Deuterium Oxide) δ 8.29 (s, 2H), 7.18-7.12 (m, 3H), 6.98 (d, J=5.5 Hz, 2H), 6.91 (td, J=8.0, 6.9, 3.2 Hz, 4H), 4.45-4.34 (m, 2H), 4.31-4.17 (m, 2H), 3.75-3.57 (m, 3H), 3.51 (dq, J=13.0, 6.3, 5.7 Hz, 2H), 3.35 (dtt, J=21.5, 10.9, 6.0 Hz, 1H), 2.97 (dt, J=14.0, 4.6 Hz, 1H), 2.90 (q, J=5.4 Hz, 2H), 2.83 (t, J=7.7 Hz, 2H), 2.75 (ddd, J=13.5, 9.2, 3.4 Hz, 1H), 2.15-2.06 (m, 1H), 2.01 (dd, J=13.5, 7.7 Hz, 1H), 1.75 (dt, J=13.3, 5.6 Hz, 1H), 1.68-1.59 (m, 2H), 1.56 (s, 1H), 1.53 (d, J=7.5 Hz, 1H), 1.40 (q, J=9.4, 8.3 Hz, 3H), 1.35-1.22 (m, 2H), 0.74 (dd, J=18.9, 5.1 Hz, 6H).


Example 51. Synthesis of Compound 51

According to similar preparation procedures as in Example 16, the target compound was synthesized, with reactant compound




embedded image


replaced by




embedded image


specifically, as shown below:




embedded image


LC-MS-51 (ES, m/z): [M+1]=829



1H NMR (400 MHz, Deuterium Oxide) δ 8.34 (s, 2H), 7.34-7.22 (m, 3H), 7.21 (dt, J=6.3, 2.6 Hz, 3H), 7.07-6.97 (m, 4H), 4.52-4.42 (m, 2H), 4.36 (d, J=17.5 Hz, 1H), 4.25 (dd, J=8.8, 5.7 Hz, 1H), 3.74 (d, J=13.5 Hz, 2H), 3.63-3.37 (m, 4H), 3.28 (ddd, J=17.5, 11.7, 4.4 Hz, 2H), 3.16 (td, J=8.7, 8.2, 4.1 Hz, 1H), 3.04 (ddt, J=20.9, 12.1, 5.0 Hz, 2H), 2.99-2.77 (m, 7H), 1.71-1.63 (m, 1H), 1.60 (ddd, J=14.5, 9.7, 5.6 Hz, 3H), 1.44 (s, 3H), 1.35 (ddd, J=30.9, 14.8, 7.2 Hz, 1H), 0.79 (dd, J=18.9, 5.7 Hz, 6H).


Example 1A. Synthesis of Compound 1A



embedded image


1.1 Synthesis of Compound 1A-1

Benzyl 4-aminopiperidine-1-carboxylate (100 mg, 0.427 mmol, 1.00 equiv) and TEA (0.2 mL, 2.693 mmol, 6.31 equiv) were dissolved in DCM (15 mL), and methyl chloroformate (60.49 mg, 0.640 mmol, 1.5 equiv) was added portionwise. The mixture was stirred at room temperature for 2 h. The reaction was then quenched with a saturated ammonium chloride solution and extracted with DCM (15 mL×3). The organic phases were combined, dried, and concentrated to dryness by rotary evaporation to give a crude product. The crude product was purified by silica gel column chromatography and eluted with PE/EA (1:1). The fractions were concentrated to dryness by rotary evaporation to give benzyl 4-[(methoxycarbonyl)amino]piperidine-1-carboxylate.


LC-MS-1A-1 (ES, m/z): [M+1]=293


1.2 Synthesis of Compound 1A-2

Pd/C (18 mg, w/t, 10%) was added to a stirred solution of benzyl 4-[(methoxycarbonyl)amino]piperidine-1-carboxylate (180 mg, 0.616 mmol, 1.00 equiv) in THF (12 mL). The mixture was stirred for 13 h under hydrogen atmosphere. The resulting mixture was washed with MeOH (7×50 mL). The filtrate was concentrated under reduced pressure to give methyl N-(piperidin-4-yl)carbamate (100 mg, crude).


LC-MS-1A-2 (ES, m/z): [M+1]=159


1.3 Synthesis of Compound 1A-3

DIPEA (1.48 mg, 0.123 mmol, 3 equiv), intermediate 2 (31 mg, 0.041 mmol, 1.00 equiv) and HATU (23.45 mg, 0.061 mmol, 1.5 equiv) were added to a stirred solution of methyl N-(piperidin-4-yl)carbamate (19.51 mg, 0.123 mmol, 3.00 equiv) in DMF (10 mL). The reaction was carried out at room temperature for 3 h under nitrogen atmosphere. Water (12 mL) was added to quench the reaction, and the resulting mixture was extracted with ethyl acetate (3×10 mL). The organic phases were combined, dried, and concentrated to dryness by rotary evaporation to give a crude product. The crude product was purified by reversed-phase chromatography and eluted with CH3CN/H2O (0.1% FA) (6:1), and the fractions were concentrated to dryness by rotary evaporation to give tert-butyl N-[(5R)-5-[(2R))-2-[(2R)-2-[(2R)-2-[(tert-butoxycarbonyl)amino]-3-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-{4-[(methoxycarbonyl))amino]piperidin-1-yl}-6-oxohexyl]carbamate.


LC-MS-1A-3 (ES, m/z): [M+1]=894


1.4 Synthesis of Compound 1A

tert-Butyl N-[(5R)-5-[(2R)-2-[(2R)-2-[(2R)-2-[(tert-butoxycarbonyl)amino]-3-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-{4-[(methoxycarbonyl)amino]piperidin-1-yl}-6-oxohexyl]carbamate (350 mg, 0.391 mmol, 1 equiv) was dissolved in 4 M HCl (gas)/1,4-dioxane (5 mL). The resulting mixture was reacted at room temperature for 4 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by preparative HPLC, and the fractions were concentrated to dryness by rotary evaporation to give methyl N-{1-[(2R)-6-amino-2-[(2R)-2-[(2R)-2-[(2R)-2-amino-3]-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanoylamino]hexanoyl]piperidin-4-yl}carbamate (169.1 mg, 62.01%, purity: 99.2%).


LC-MS-1A (ES, m/z): [M+1]=693



1H NMR (400 MHz, Deuterium Oxide) δ 8.36 (s, 2H), 7.45-6.86 (m, 10H), 4.63-4.47 (m, 2H), 4.27-4.06 (m, 3H), 3.85 (dd, J=37.5, 14.1 Hz, 1H), 3.55 (d, J=3.6 Hz, 4H), 3.32-2.69 (m, 8H), 2.14-1.71 (m, 2H), 1.73-1.13 (m, 11H), 0.82 (dq, J=20.0, 2.6 Hz, 6H).


Example 2A. Synthesis of Compound 2A



embedded image


2.1 Synthesis of Compound 2A-1

Lithium bis(trimethylsilyl)amide (19.97 mL, 119.348 mmol, 11.95 equiv) was added dropwise to a stirred solution of indene (1.16 g, 9.986 mmol, 1.00 equiv) in THE (12.00 mL, 148.116 mmol, 14.83 equiv) at 0° C. under nitrogen atmosphere. The resulting mixture was stirred at 0° C. for 1 h under nitrogen atmosphere. tert-Butyl N,N-bis(2-chloroethyl)carbamate (2.42 g, 9.994 mmol, 1.00 equiv) was added dropwise to the mixture described above at 0° C. The resulting mixture was stirred at 0° C. for another 1 h and concentrated under reduced pressure to give a crude product. The crude product was purified by silica gel column chromatography and eluted with petroleum ether/ethyl acetate (9:1), and the fractions were concentrated to dryness by rotary evaporation to give tert-butyl spiro[indene-1,4′-piperidine]-1′-carboxylate (2.22 g, 77.90%, purity: 99%).


LC-MS-2A-1 (ES, m/z): [M+1]=286


2.2 Synthesis of Compound 2A-2

Borane dimethyl sulfide complex 2M (THF) (19.45 mL, 38.895 mmol, 5 equiv) was added to a stirred solution of tert-butyl spiro[indene-1,4′-piperidine]-1′-carboxylate (2.22 g, 7.779 mmol, 1.00 equiv) in THF (22 mL) at 0° C. under nitrogen atmosphere. The resulting mixture was stirred at 0° C. for 4 h under nitrogen atmosphere. Sodium hydroxide (2 M) (23.34 mL, 46.674 mmol, 6 equiv) and hydrogen peroxide (6.66 mL, 285.878 mmol, 36.75 equiv) were added dropwise to the mixture described above at 0° C. The resulting mixture was stirred at 0° C. for another 30 min and then extracted with ethyl acetate (1×150 mL). The combined organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with petroleum ether/ethyl acetate (1:1), and the fractions were concentrated to dryness by rotary evaporation to give tert-butyl 2-hydroxy-2,3-dihydrospiro[indene-1,4′-piperidine]-1′-carboxylate (959 mg, 40.63%, purity: 99%).


LC-MS-2A-2 (ES, m/z): [M+1]=304


2.3 Synthesis of Compound 2A-3

Dess-Martin reagent (2180.81 mg, 5.142 mmol, 2 equiv) was added portionwise to a solution of tert-butyl 2-hydroxy-2,3-dihydrospiro[indene-1,4′-piperidine]-1′-carboxylate (780 mg, 2.571 mmol, 1.00 equiv) in dichloromethane (16 mL) at room temperature. The resulting mixture was stirred at room temperature for 12 h. The resulting mixture was filtered, and the filter cake was washed with dichloromethane (1×20 mL). The combined organic layer was washed with aqueous sodium bicarbonate solution (1×15 mL) and brine (1×15 mL) and extracted with ethyl acetate, and the organic phases were combined, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with petroleum ether/ethyl acetate (4:1), and the fractions were concentrated to dryness by rotary evaporation to give tert-butyl 2-oxo-3H-spiro[indene-1,4′-piperidine]-1′-carboxylate (550 mg, 70.98%, purity: 99%).


LC-MS-2A-3 (ES, m/z): [M+1]=302


2.4 Synthesis of Compound 2A-4

tert-Butanesulfinamide (2.08 g, 17.121 mmol, 3 equiv) was added portionwise to a stirred mixture of tert-butyl 2-oxo-3H-spiro[indene-1,4′-piperidine]-1′-carboxylate (1.72 g, 5.707 mmol, 1.00 equiv) and ethyl titanate (4 mL) at room temperature. The resulting mixture was stirred at 85° C. for 2 h. The reaction was quenched with water at room temperature. The resulting mixture was filtered, and the filter cake was washed with ethyl acetate (3×10 mL). The filtrate was concentrated under reduced pressure. The resulting mixture was extracted with ethyl acetate (2×50 mL). The combined organic phase (1×40 mL) was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with petroleum ether/ethyl acetate (2:1), and the fractions were concentrated to dryness by rotary evaporation to give tert-butyl (2E)-2-[(2-methylpropane-2-sulfinyl)imino]-3H-spiro[indene-1,4′-piperidine]-1′-carboxylate (450 mg, 19.49%, purity: 99%).


LC-MS-2A-4 (ES, m/z): [M+1]=405


2.5 Synthesis of Compound 2A-5

BH3·THF (1.73 mL, 1.730 mmol, 2 equiv) was added portionwise to a stirred solution of tert-butyl (2E)-2-[(2-methylpropane-2-sulfinyl)imino]-3H-spiro[indene-1,4′-piperidine]-1′-carboxylate (350 mg, 0.865 mmol) in tetrahydrofuran (5 mL) at 0° C. under nitrogen atmosphere. The resulting mixture was stirred at 0° C. for 2 h under nitrogen atmosphere. The reaction was quenched with methanol at room temperature. The resulting mixture was concentrated under reduced pressure to give tert-butyl 2-[(2-methylpropane-2-sulfinyl)amino]-2,3-dihydrospiro[indene-1,4′-piperidine]-1′-carboxylate (380 mg, crude).


LC-MS-2A-5 (ES, m/z): [M+1]=407


2.6 Synthesis of Compound 2A-6

TFA (0.3 mL) was added to a stirred solution of tert-butyl 2-[(2-methylpropane-2-sulfinyl)amino]-2,3-dihydrospiro[indene-1,4′-piperidine]-1′-carboxylate (100 mg, 0.246 mmol) in dichloromethane (1.7 mL) at room temperature. The resulting mixture was stirred at room temperature for 1.5 h. The resulting mixture was concentrated in vacuo to give N-{2,3-dihydrospiro[indene-1,4′-piperidin]-2-yl}-2-methylpropane-2-sulfinamide (100 mg, crude).


LC-MS-2A-6 (ES, m/z): [M+1]=307


2.7 Synthesis of Compound 2A-7

Diisopropylethylamine (115.01 mg, 0.891 mmol, 3 equiv), intermediate 2 (223.64 mg, 0.297 mmol, 1.00 equiv) and HATU (135.34 mg, 0.3156 mmol, 1.2 equiv) were added portionwise to a solution of N-{2,3-dihydrospiro[indene-1,4′-piperidin]-2-yl}-2-methylpropane-2-sulfinamide (100.00 mg, 0.327 mmol, 1.1 equiv) in dimethylformamide (2 mL) at room temperature. The resulting mixture was stirred at room temperature overnight. The resulting solution was purified by reversed-phase flash chromatography, and the fractions were concentrated to dryness by rotary evaporation to give tert-butyl N-[(5R)-5-[(2R)-2-[(2R)-2-[(2R)-2-[(tert-butoxycarbonyl)amino]-3-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-{2-[(2-methylpropane-2-sulfinyl)amino]-2,3-dihydrospiro[indene-1,4′-piperidin]-1′-yl}-6-oxohexyl]carbamate (70 mg, 22.64%, purity: 99%).


LC-MS-2A-7 (ES, m/z): [M+1]=1042


2.8 Synthesis of Compound 2A

4 M HCl (gas)/1,4-dioxane (2 mL) was added portionwise to a stirred solution of tert-butyl N-[(5R)-5-[(2R)-2-[(2R)-2-[(2R)-2-[(tert-butoxycarbonyl)amino]-3-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-{2-[(2-methylpropane-2-sulfinyl)amino]-2,3-dihydrospiro[indene-1,4′-piperidin]-1′-yl}-6-oxohexyl]carbamate (70 mg, 0.067 mmol, 1.00 equiv) in dioxane (2 mL) at room temperature. The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated under reduced pressure. The crude product (50 mg) was purified by preparative HPLC to give (2R)—N-[(2R)-6-amino-1-{2-amino-2,3-dihydrospiro[indene-1,4′-piperidin]-1′-yl}-1-oxohex-2-yl]-2-[(2R)-2-[(2R)-2-amino-3-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanamide (24.7 mg, 41.65%, purity: 83.3%).


LC-MS-2A (ES, m/z): [M+1]=738



1H NMR-2 (400 MHz, Deuterium Oxide) δ 8.26 (s, 3H), 7.48-6.89 (m, 14H), 4.52 (ddt, J=25.9, 13.0, 7.1 Hz, 2H), 4.14 (dq, J=17.7, 5.4, 4.2 Hz, 2H), 3.99 (q, J=8.3, 7.1 Hz, 2H), 3.86-3.23 (m, 3H), 3.16-2.65 (m, 7H), 2.25-1.18 (m, 13H), 0.97-0.56 (m, 6H).


Example 3A. Synthesis of Compound 3A



embedded image


3.1 Synthesis of Compound 3A-1

Thionyl chloride (1.27 g, 10.636 mmol, 2.0 equiv) was added portionwise to a stirred solution of (3-bromopyridin-2-yl)methanol (1 g, 5.318 mmol, 1.00 equiv) in DCM at 0° C. The resulting mixture was stirred at 0° C. for 4 h. The mixture was neutralized with saturated NaHCO3 (aq.) to pH 7. The crude product was purified by silica gel column chromatography and eluted with PE/EA (5:1), and the fractions were concentrated to dryness by rotary evaporation to give 3-bromo-2-(chloromethyl)pyridine (700 mg, 64.4%, purity: 99%).


LC-MS-3A-1 (ES, m/z): [M+1]=205


3.2 Synthesis of Compound 3A-2

LDA (10 mL, 73.747 mmol, 10.15 equiv) was added dropwise to a solution of 1-tert-butyl 4-methylpiperidine-1,4-dicarboxylate (2.65 g, 10.897 mmol, 1.5 equiv) in TEE (50 mL) at −30° C. under nitrogen atmosphere. After 30 min, a solution of 3-bromo-2-(chloromethyl)pyridine (1.5 g, 7.265 mmol, 1 equiv) in THF was added dropwise to the solution described above. The resulting mixture was stirred at room temperature overnight. The reaction was quenched with saturated NH4Cl (aq.) at room temperature. The resulting mixture was extracted with ethyl acetate. The organic phases were combined and concentrated to dryness by rotary evaporation. The crude product was purified by silica gel column chromatography and eluted with PE/EA (1:1), and the fractions were concentrated to dryness by rotary evaporation to give 1-tert-butyl 4-methyl 4-[(3-bromopyridin-2-yl)methyl]piperidine-1,4-dicarboxylate (3.7 g, 82.4%, purity: 99%).


LC-MS-3A-2 (ES, m/z): [M+1]=413


3.3 Synthesis of Compound 3A-3

LiOH (0.37 g, 8.709 mmol, 3 equiv) was added to a solution of 1-tert-butyl 4-methyl 4-[(3-bromopyridin-2-yl)methyl]piperidine-1,4-dicarboxylate (1.2 g, 2.903 mmol, 1.00 equiv) in methanol at 70° C. The mixture was stirred at 70° C. for 6 h. The reaction solution was cooled, and then the pH of the mixture was adjusted to 6 with HCl (aq.). The resulting mixture was extracted with ethyl acetate. The organic phases were combined, dried, and concentrated to dryness by rotary evaporation. The crude product was purified by silica gel column chromatography and eluted with PE/EA (1:1), and the fractions were concentrated to dryness by rotary evaporation to give 4-[(3-bromopyridin-2-yl)methyl]-1-(tert-butoxycarbonyl)piperidine-4-carboxylic acid (630 mg, 54.34%, purity: 99%).


LC-MS-3A-3 (ES, m/z): [1M+1]=399


3.4 Synthesis of Compound 3A-4

DIPEA (786.87 mg, 6.087 mmol, 3 equiv) and HATU (1543.30 mg, 4.058 mmol, 2 equiv) were added to a stirred solution of N,O-dimethylhydroxylamine hydrochloride (200 mg, 2.029 mmol, 1 equiv) and 4-[(3-bromopyridin-2-yl)methyl]-1-(tert-butoxycarbonyl)piperidine-4-carboxylic acid (1604.22 mg, 4.058 mmol, 2 equiv) in DMF at room temperature under nitrogen atmosphere. The mixture was stirred at 0° C. for 3 h under nitrogen atmosphere. The reaction solution was quenched with water and extracted with ethyl acetate. The organic phases were combined, dried, and concentrated to dryness by rotary evaporation. The crude product was purified by silica gel column chromatography and eluted with PE/EA (1:1), and the fractions were concentrated to dryness by rotary evaporation to give tert-butyl 4-[(3-bromopyridin-2-yl)methyl]-4-(methoxy(methyl)carbamoyl)piperidine-1-carboxylate (120 mg, 13.52%, purity: 99%).


LC-MS-3A-4 (ES, m/z): [M+1]=442


3.5 Synthesis of Compound 3A-5

Butyl lithium (1.74 mL, 27.130 mmol, 10 equiv) was added to a mixture of tert-butyl 4-[(3-bromopyridin-2-yl)methyl]-4-(methoxy(methyl)carbamoyl)piperidine-1-carboxylate (1.2 g, 2.713 mmol, 1 equiv) in THF at −78° C. under nitrogen atmosphere. The mixture was stirred at −78° C. for 3 h under nitrogen atmosphere. The reaction was quenched with a saturated NH4Cl (aq.) (2 mL) solution at room temperature. The resulting mixture was extracted with ethyl acetate. The organic phases were combined, dried, and concentrated to dryness by rotary evaporation. The crude product was purified by silica gel column chromatography and eluted with PE/EA (1:1), and the fractions were concentrated to dryness by rotary evaporation to give tert-butyl 5-oxo-7H-spiro[cyclopenta[b]pyridine-6,4′-piperidine]-1′-carboxylate (590 mg, 71.93%, purity: 99%).


LC-MS-3A-5 (ES, m/z): [M+1]=303


3.6 Synthesis of Compound 3A-6

tert-Butanesulfinamide (30.06 mg, 0.249 mmol, 3 equiv) was added dropwise to a stirred solution of tert-butyl 5-oxo-7H-spiro[cyclopenta[b]pyridine-6,4′-piperidine]-1′-carboxylate (25 mg, 0.083 mmol, 1 equiv) under air atmosphere. The mixture was stirred at 110° C. for 4 h under nitrogen atmosphere. The reaction solution was cooled to room temperature, and then the reaction was quenched with water. The resulting mixture was extracted with ethyl acetate. The organic phases were combined, dried, and concentrated to dryness by rotary evaporation. The crude product was purified by silica gel column chromatography and eluted with PE/EA (1:1), and the fractions were concentrated to dryness by rotary evaporation to give tert-butyl (5Z)-5-[(2-methylpropane-2-sulfinyl)imino]-7H-spiro[cyclopenta[b]pyridine-6,4′-piperidine]-1′-carboxylate (10 mg, 29.82%, purity: 99%).


LC-MS-3A-6 (ES, m/z): [M+1]=413


3.7 Synthesis of Compound 3A-7

A solution of tert-butyl (5Z)-5-[(2-methylpropane-2-sulfinyl)imino]-7H-spiro[cyclopenta[b]pyridine-6,4′-piperidine]-1′-carboxylate (20 mg, 0.049 mmol, 1 equiv) and BH3-THF (0.2 mL, 2.327 mmol, 47.19 equiv) in THE was stirred at 0° C. for 3 h under nitrogen atmosphere. The reaction was quenched with MeOH (2 mL) at room temperature. The reaction solution was purified by reversed-phase flash chromatography under the following conditions: column: silica gel column; mobile phase: MeCN/aqueous solution, gradient 10% to 100% within 10 min; detector: UV 220 nm. The fractions were concentrated to dryness by rotary evaporation to give tert-butyl 5-[(2-methylpropane-2-sulfinyl)amino]-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4′-piperidine]-1′-carboxylate (10 mg, 49.75%, purity: 99%).


LC-MS-3A-7 (ES, m/z): [M+1]=408


3.8 Synthesis of Compound 3A-8

tert-Butyl 5-[(2-methylpropane-2-sulfinyl)amino]-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4′-piperidine]-1′-carboxylate (180 mg, 0.4421 mmol, 1 equiv) and TFA (1 mL, 13.463 mmol, 30.48 equiv) were dissolved in DCM at room temperature under air atmosphere and stirred for 3 h. The resulting mixture was concentrated under reduced pressure to give N-{5,7-dihydrospiro[cyclopenta[b]pyridine-6,4′-piperidin]-5-yl}-2-methylpropane-2-sulfinamide (66 mg, crude), which was directly used in the next step without further purification.


LC-MS-3A-8 (ES, m/z): [M+1]=308


3.9 Synthesis of Compound 3A-9

A solution of N-{5,7-dihydrospiro[cyclopenta[b]pyridine-6,4′-piperidin]-5-yl}-2-methylpropane-2-sulfinamide (66 mg, 0.107 mmol, 1 equiv) and intermediate 2 (126.70 mg, 0.086 mmol, 0.8 equiv) in DMF was stirred at room temperature for 1 min under air atmosphere. DIPEA (41.62 mg, 0.323 mmol, 1.5 equiv) and HATU (163.24 mg, 0.430 mmol, 2 equiv) were added to the mixture described above at room temperature. The mixture was stirred at room temperature for 3 h under nitrogen atmosphere. The reaction solution was purified by reversed-phase flash chromatography under the following conditions: column: silica gel; mobile phase: MeCN/aqueous solution, gradient 10% to 50% within 10 min; detector: UV 220 nm. The fractions were concentrated to dryness by rotary evaporation to give tert-butyl N-[(5R)-5-[(2R)-2-[(2R)-2-[(2R)-2-[(tert-butoxycarbonyl)amino]-3-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-{5-[(2-methylpropane-2-sulfinyl)amino]-5,7-dihydrospiro[cyclopenta[c]pyridine-6,4′-piperidin]-1′-yl}-6-oxohexyl]carbamate (30 mg, 13.39%, purity: 99%).


LC-MS-3A-9 (ES, m/z): [M+1]=1043


3.10 Synthesis of Compound 3A

4 M HCl (gas)/1,4-dioxane (5 mL) was added dropwise to tert-butyl N-[(5R)-5-[(2R)-2-[(2R)-2-[(2R)-2-[(tert-butoxycarbonyl)amino]-3-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-{5-[(2-methylpropane-2-sulfinyl)amino]-5,7-dihydrospiro[cyclopenta[c]pyridine-6,4′-piperidin]-1′-yl}-6-oxohexyl]carbamate (110 mg, 0.105 mmol, 1 equiv). The resulting mixture was stirred at room temperature for 3 h under nitrogen atmosphere and concentrated under reduced pressure to give a crude product. The crude product (100 mg) was purified by preparative HPLC under the following conditions (column: C18 silica gel; mobile phase: acetonitrile/aqueous solution, gradient 10% to 50% within 10 min; detector: UV 220 nm), and the fractions were concentrated to dryness by rotary evaporation to give (2R)—N-[(2R)-6-amino-1-{5-amino-5,7-dihydrospiro[cyclopenta[c]pyridine-6,4′-piperidin]-1′-yl}-1-oxohex-2-yl]-2-[(2R)-2-amino-3-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanamide (20.1 mg, 18.2%, purity: 70.3%).


LC-MS-3A (ES, m/z): [M+1]=739



1H NMR (400 MHz, Deuterium Oxide) δ 8.40 (s, 1H), 8.27 (s, 3H), 7.81 (dd, J=8.2, 4.4 Hz, 1H), 7.41-6.90 (m, 11H), 4.58-4.38 (m, 2H), 4.36-4.16 (m, 2H), 4.10 (s, 2H), 3.98-3.71 (m, 1H), 3.39 (d, J=12.4 Hz, 1H), 3.23-2.72 (m, 9H), 1.81-1.20 (m, 13H), 0.79 (dt, J=20.4, 5.1 Hz, 6H).


Example 4A. Synthesis of Compound 4A



embedded image


4.1 Synthesis of Compound 4A-1

Sodium bis(trimethylsilyl)amide (1426.37 mg, 7.778 mmol) was to a stirred solution of 6-chloropyridin-2-amine (500 mg, 3.889 mmol, 1.00 equiv) in tetrahydrofuran (10 mL) at 0° C. under nitrogen atmosphere. Di-tert-butyl dicarbonate (933.69 mg, 4.278 mmol, 1.1 equiv) was added dropwise to the mixture described above. The resulting mixture was stirred at room temperature overnight. The reaction was quenched with water/ice at 0° C. The resulting mixture was extracted with ethyl acetate (3×10 mL). The combined organic layer was washed with brine (1×10 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, purified by silica gel column chromatography, and eluted with petroleum ether/ethyl acetate (10:1), and the fractions were concentrated to dryness by rotary evaporation to give tert-butyl N-(6-chloropyridin-2-yl)carbamate (800 mg, 89.95%, purity: 99%).


LC-MS-4A-1 (ES, m/z): [M+1]=229


4.2 Synthesis of Compound 4A-2

In a two-necked round-bottom flask, tetramethylethylenediamine (3353.91 mg, 28.862 mmol, 2.2 equiv) was dissolved in tetrahydrofuran (78.43 mL). n-Butyllithium (1848.71 mg, 28.862 mmol, 2.2 equiv) was added dropwise to the mixture described above at −78° C. The mixture was stirred at −20° C. for another 30 min. Benzyl 4-oxopiperidine-1-carboxylate (4590.26 mg, 19.678 mmol, 1.5 equiv) was added dropwise to the mixture described above at −78° C. The mixture was stirred at −78° C. for another 1 h. tert-Butyl N-(6-chloropyridin-2-yl)carbamate (3000 mg, 13.119 mmol, 1.00 equiv) was added dropwise to the mixture described above at −50° C. The resulting mixture was stirred at 40° C. overnight. The resulting mixture was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography and eluted with petroleum ether/ethyl acetate (2:1) to give benzyl 7′-chloro-2′-oxo-1′H-spiro[piperidine-4,4′-pyrido[2,3-d][1,3]oxazine]-1-carboxylate (3000 mg, 58.97%, purity: 99%).


LC-MS-4A-2 (ES, m/z): [M+1]=388


4.3 Synthesis of Compound 4A-3

Benzyl 7′-chloro-2′-oxo-1′H-spiro[piperidine-4,4′-pyrido[2,3-d][1,3]oxazine]-1-carboxylate (230 mg, 0.612 mmol, 1.00 equiv) was dissolved in methanol (20 mL), and Pd/C (12.62 mg, w/t, 10%) was added. The system was purged 3 times with hydrogen. The resulting mixture was stirred at room temperature overnight under hydrogen atmosphere. The resulting mixture was filtered, and the filter cake was washed with methanol (3×10 mL). The filtrate was concentrated under reduced pressure to give spiro[piperidine-4,4′-pyrido[2,3-d][1,3]oxazin]-2′(1′H)-one (110 mg, crude).


LC-MS-4A-3 (ES, m/z): [M+1]=220


4.4 Synthesis of Compound 4A-4

Spiro[piperidine-4,4′-pyrido[2,3-d][1,3]oxazin]-2′(1′H)-one (44 mg, 0.138 mmol, 1.00 equiv) and intermediate 2 (100 mg, 0.067 mmol, 0.5 equiv) were dissolved in DMF (2 mL), and diisopropylethylamine (34 mg, 0.4 mmol, 3.00 equiv) and HATU (76 mg, 0.207 mmol, 1.50 equiv) were added portionwise. The mixture was stirred at room temperature overnight under nitrogen atmosphere. Water (10 mL) was added to quench the reaction, and the resulting mixture was extracted with ethyl acetate (3×10 mL). The organic phases were combined, dried, and concentrated in vacuo to give tert-butyl ((10R,13R,16R,19R)-16-benzyl-13-isobutyl-2,2-dimethyl-4,12,15,18-tetraoxo-10-(2′-oxo-1′,2′-dihydrospiro[piperidine-4,4′-pyrido[2,3-d][1,3]oxazine]-1-carbonyl)-20-phenyl-3-oxa-5,11,14,17-tetraazaeicosan-19-yl)carbamate (120 mg, crude).


LC-MS-4A-4 (ES, m/z): [M+1]=955


4.5 Synthesis of Compound 4A

tert-Butyl ((10R,13R,16R,19R)-16-benzyl-13-isobutyl-2,2-dimethyl-4,12,15,18-tetraoxo-10-(2′-oxo-1′,2′-dihydrospiro[piperidine-4,4′-pyrido[2,3-d][1,3]oxazine]-1-carbonyl)-20-phenyl-3-oxa-5,11,14,17-tetraazaeicosan-19-yl)carbamate (120 mg, 0.126 mmol, 1.00 equiv) and a 4 M solution of HCl (gas) in 1,4-dioxane (4 mL) were added to a 50 mL round-bottom flask. The resulting mixture was stirred at room temperature for 1 h. The resulting mixture was concentrated under reduced pressure and purified by reverse flash chromatography, and the fractions were concentrated to dryness by rotary evaporation to give (2R)—N-[(2R)-6-amino-1-oxo-1-{2′-oxo-1′H-spiro[piperidine-4,4′-pyrido[2,3-d][1,3]oxazin]-1-yl}hex-2-yl]-2-[(2R)-2-[(2R)-2-amino-3-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanamide (7 mg, 7.31%, purity: 99.2%).


LC-MS-4A (ES, m/z): [M+1]=755



1H NMR (400 MHz, Deuterium Oxide) δ 8.36 (s, 2H), 8.10 (dd, J=10.1, 5.0 Hz, 1H), 7.58 (dd, J=15.2, 7.7 Hz, 1H), 7.24 (h, J=7.1 Hz, 6H), 7.12 (q, J=7.7 Hz, 5H), 4.67 (s, 1H), 4.55 (q, J=7.5 Hz, 1H), 4.33 (d, J=13.6 Hz, 1H), 4.25-4.16 (m, 1H), 4.07-3.82 (m, 2H), 3.66-3.43 (m, 1H), 3.09 (q, J=13.2 Hz, 1H), 3.04-2.82 (m, 6H), 2.15 (d, J=27.4 Hz, 2H), 2.05-1.82 (m, 2H), 1.78-1.55 (m, 4H), 1.57-1.23 (m, 5H), 0.84 (dt, J=20.5, 5.2 Hz, 6H).


Example 5A. Synthesis of Compound 5A



embedded image


LC-MS-5A (ES, m/z): [M+1]=753



1H NMR (300 MHz, Deuterium Oxide) δ 8.55-8.39 (m, 1H), 8.35 (s, 2H), 7.97-7.74 (m, 1H), 7.56 (d, J=5.1 Hz, 1H), 7.37-7.00 (m, 10H), 4.85-4.70 (m, 2H), 4.58-4.46 (m, 1H), 4.33-4.13 (m, 2H), 4.05-3.79 (m, 2H), 3.71-3.54 (m, 2H), 3.50-3.26 (m, 1H), 3.02-2.82 (m, 6H), 1.95-1.17 (m, 13H), 0.83 (dd, J=14.9, 6.7 Hz, 6H).


Example 6A. Synthesis of Compound 6A



embedded image


LC-MS-6A (ES, m/z): [M+1]=753



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 2H), 8.23-7.98 (m, 1H), 7.77 (dd, J=27.3, 7.8 Hz, 1H), 7.47-6.96 (m, 11H), 4.85-4.77 (m, 1H), 4.58 (t, J=7.5 Hz, 1H), 4.34-4.01 (m, 3H), 3.86 (dd, J=53.3, 14.0 Hz, 1H), 3.46 (q, J=12.7, 12.2 Hz, 1H), 3.25-2.68 (m, 9H), 2.10-1.58 (m, 8H), 1.47 (d, J=6.9 Hz, 5H), 1.13-0.44 (m, 6H).


Example 7A. Synthesis of Compound 7A



embedded image


LC-MS-7A (ES, m/z): [M+1]=753



1H NMR (400 MHz, Deuterium Oxide) δ 8.38 (s, 2H), 7.88-7.37 (m, 2H), 7.42-7.00 (m, 10H), 6.90-6.47 (m, 1H), 4.57 (t, J=7.4 Hz, 2H), 4.37-3.97 (m, 3H), 3.86 (dd, J=44.1, 13.8 Hz, 1H), 3.63-3.34 (m, 3H), 3.20-2.66 (m, 9H), 2.28-1.11 (m, 15H), 0.84 (dd, J=20.1, 5.4 Hz, 6H).


Example 8A. Synthesis of Compound 8A



embedded image


8.1 Synthesis of Compound 8A-1

Benzyl 4-aminopiperidine-1-carboxylate (500 mg, 2.134 mmol, 1 equiv) was dissolved in toluene (5 mL), and diphosgene (260 μL, 1.314 mmol, 0.62 equiv) was added. The resulting mixture was stirred at 90° C. for 3 h, and then tetrahydropyran-4-ol (205 μL, 2.007 mmol, 0.94 equiv) was added dropwise. The resulting mixture was stirred at 90° C. overnight under nitrogen atmosphere. The reaction solution was cooled to room temperature and then poured into ice water, and the resulting mixture was extracted with EtOAc (3×10 mL). The combined organic layer was washed with ethyl acetate (3×10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The crude product was directly used in the next step without further purification.


8.2 Synthesis of Compound 8A-2

Compound 8A-1 (433 mg, 1.195 mmol, 1 equiv) and Pd/C (100 mg, w/t, 10%) were added to methanol (10 mL, 246.988 mmol, 206.73 equiv), and the mixture was stirred at room temperature overnight under hydrogen atmosphere. The resulting mixture was filtered, and the filter cake was washed with MeOH (3×10 mL). The filtrate was concentrated under reduced pressure, and the filtrate was concentrated to dryness by rotary evaporation to give oxacyclohexan-4-yl N-(piperidin-4-yl)carbamate (448 mg, crude).


8.3 Synthesis of Compound 8A-3

Oxacyclohexan-4-yl N-(piperidin-4-yl)carbamate (113 mg, 0.495 mmol, 1.5 equiv) was dissolved in DMF (10 mL) at room temperature under nitrogen atmosphere, and intermediate 2 (248.79 mg, 0.330 mmol, 1 equiv) was added. The resulting mixture was stirred, and then DIPEA (85.30 mg, 0.660 mmol, 2 equiv) and HATU (188.21 mg, 0.495 mmol, 1.5 equiv) were added. After the addition was completed, the resulting mixture was stirred overnight. Water was added to quench the reaction, and the resulting mixture was extracted with EtOAc (3×20 mL). The combined organic layer was washed with diethyl ether (3×20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography under the following conditions: column: silica gel; mobile phase: MeCN aqueous solution, gradient 10% to 50% within 10 min; detector: UV 254 nm. The fractions were concentrated to dryness by rotary evaporation to give compound 8A-3 (104 mg, 32.69%, purity: 99%).


8.4 Synthesis of Compound 8A

Compound 8A-3 (104 mg, 0.108 mmol, 1 equiv) and TFA (4 mL) were added to DCM (16 mL) at room temperature under nitrogen atmosphere and stirred for 3 h. The resulting mixture was concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase flash chromatography under the following conditions: column: silica gel; mobile phase: MeCN aqueous solution, gradient 10% to 50% within 10 min; detector: UV 254 nm. The fractions were concentrated to dryness by rotary evaporation to give compound 8A (19.7 mg, 22.73%, purity: 95.0%).


LC-MS-8A (ES, m/z): [M+1]=764



1H NMR (400 MHz, Deuterium Oxide) δ 8.36 (s, 1H), 7.26 (h, J=6.6 Hz, 6H), 7.13 (t, J=7.1 Hz, 4H), 4.55 (dt, J=8.0, 4.0 Hz, 2H), 4.30-3.99 (m, 2H), 3.98-3.72 (m, 4H), 3.56 (d, J=10.5 Hz, 3H), 3.22 (q, J=13.9 Hz, 1H), 2.96 (t, J=6.7 Hz, 3H), 2.90 (td, J=7.6, 2.7 Hz, 3H), 1.85 (s, 4H), 1.74-1.52 (m, 6H), 1.52-1.17 (m, 7H), 0.93-0.75 (m, 6H).


Example 9A. Synthesis of Compound 9A



embedded image


LC-MS-9A (ES, m/z): [M+1]=812.25



1H NMR (300 MHz, Deuterium Oxide) δ 8.41 (s, 2H), 7.38-7.16 (m, 10H), 4.92 (s, 1H), 4.60 (s, 2H), 4.26 (s, 2H), 4.02 (d, J=38.6 Hz, 2H), 3.67 (s, 1H), 3.41-2.90 (m, 12H), 2.28 (s, 4H), 1.93 (d, J=21.3 Hz, 2H), 1.71-1.28 (m, 11H), 0.95-0.83 (m, 6H).


Example 10A. Synthesis of Compound 10A



embedded image


10.1 Synthesis of Compound 10A-3

Compound 10A-2 (1 g) was added to a 100 mL round-bottom flask, and stirred and swollen in DCM (40 mL) for 30 min at room temperature. The resulting mixture was filtered, and the solid was washed with DCM and dried. The solid was dissolved in DMF (40 mL), followed by addition of compound 10A-1 (2 g, 8.761 mmol, 1.00 equiv), DCC (5.42 g, 26.283 mmol, 3 equiv) and HOBT (2.37 g, 17.522 mmol, 2 equiv). The resulting mixture was stirred at room temperature for 3 h, and finally, DMAP (0.54 g, 4.380 mmol, 0.5 equiv) was added and stirred overnight at room temperature. The resulting mixture was filtered and washed with MeOH, and the solid was collected to give compound 10A-3 (3 g, crude) as a white solid. The resulting mixture was directly used in the next step without further purification.


10.2 Synthesis of Compound 10A-4

A solution of compound 10A-3 (2.9 g, 5.560 mmol, 1 equiv) in DMF (40 mL) and piperidine (10 mL) was stirred at room temperature for 6 h under nitrogen atmosphere. The resulting mixture was filtered and washed with MeOH, and the solid was collected and concentrated to dryness by rotary evaporation. The resulting white crude product (2 g) was directly used in the next step.


10.3 Synthesis of Compound 10A-6

Under nitrogen atmosphere, compound 10A-5 (3.54 g, 10.021 mmol, 1.5 equiv) was added to a solution of compound 10A-4 (2 g, 6.681 mmol, 1 equiv) in DMF (10 mL) at room temperature, and DIEA (1.30 g, 10.021 mmol, 1.5 equiv), HOBT (1.35 g, 10.021 mmol, 1.5 equiv) and TBTU (3.22 g, 10.021 mmol, 1.5 equiv) were added at room temperature. After stirring at room temperature for 2 h, the resulting mixture was filtered and washed with MeOH, and the solid was collected and concentrated under reduced pressure to give compound 10A-6 (2.9 g, crude).


10.4 Synthesis of Compound 10A-7

A mixed solution of compound 10A-6 (2 g, 3.151 mmol, 1 equiv) in TFA (10 mL, 134.631 mmol, 42.73 equiv) and DCM (40 mL, 629.224 mmol, 199.71 equiv) was stirred at room temperature for 4 h under nitrogen atmosphere. The resulting mixture filtrate was concentrated under reduced pressure and concentrated to dryness by rotary evaporation to give compound 10A-7 (1.6 g, crude).


LC-MS-10A-7 (ES, m/z): [M+1]=424


10.5 Synthesis of Compound 10A-9

Compound 10A-8 (229.60 mg, 0.728 mmol, 1.5 equiv) was added to a solution of compound 10A-7 (206 mg, 0.485 mmol, 1 equiv) in DMF (10 mL) at room temperature under nitrogen atmosphere, and HATU (276.78 mg, 0.728 mmol, 1.5 equiv) and DIPEA (125.44 mg, 0.970 mmol, 2 equiv) were added at room temperature. After the resulting mixture was stirred for 4 h, water was added to quench the reaction. The resulting mixture was extracted with EA (3×100 mL). The organic layer was washed with EA (3×10 mL), dried over Na2SO4, and filtered. The resulting solution was concentrated under reduced pressure. The resulting crude product was purified by reversed-phase column chromatography (conditions were as follows: C18 chromatographic column, mobile phase: solvent ACN and solvent H2O (0.1% FA), gradient 10% to 50% for 10 min, UV254 nanometer detector), and the fractions were concentrated to dryness by rotary evaporation to give compound 10A-9 (165 mg, 47.10%).


LC-MS-10A-9 (ES, m/z): [M+1]=721


10.6 Synthesis of Compound 10A-10

A solution of compound 10A-9 (165 mg, 0.229 mmol, 1 equiv) in piperidine (1 mL) and DCM (10 mL) was stirred at room temperature for 3 h under nitrogen atmosphere. The resulting solution was concentrated under reduced pressure. The resulting crude product was purified by reversed-phase column chromatography under the following conditions: C18 chromatographic column, mobile phase: solvent ACN and solvent H2O (0.1% FA), gradient 10% to 50% for 10 min, UV254 nanometer detector. The fractions were concentrated to dryness by rotary evaporation to give compound 10A-10 (100 mg, 87.5%).


LC-MS-10A-10 (ES, m/z): [M+1]=499


10.7 Synthesis of Compound 10A-13

Compound 10A-12 (1.93 g, 4.981 mmol, 1.28 equiv) was added to a solution of compound 10A-11 (1 g, 3.880 mmol, 1 equiv) in DMF (10 mL) at room temperature under nitrogen atmosphere, and HATU (2.1 g, 8.712 mmol, 2.25 equiv) and DIPEA (1.8 g, 13.927 mmol, 3.59 equiv) were added dropwise at room temperature and stirred overnight. The resulting mixture was quenched with water and extracted with EA (3×100 mL). The organic phases were combined, dried, concentrated to dryness by rotary evaporation, and filtered. The resulting solution was concentrated under reduced pressure. The resulting crude product was purified by reversed-phase column chromatography (conditions were as follows: C18 chromatographic column, mobile phase: solvent ACN and solvent H2O (0.1% FA), gradient 10% to 50% for 10 min, UV254 nanometer detector), and the fractions were concentrated to dryness by rotary evaporation to give compound 10A-13 (1.86 g, 81.16%, purity: 99%).


LC-MS-10A-13 (ES, m/z): [M+1]=590


10.8 Synthesis of Compound 10A-14

A mixed solution of compound 10A-13 (1.86 g, 3.149 mmol, 1 equiv) in TFA (6 mL, 61.209 mmol, 19.44 equiv)/DCM (24 mL, 377.534 mmol, 119.90 equiv) was stirred at room temperature for 3 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure to give compound 10A-14 (2.7 g, crude).


LC-MS-10A-14 (ES, m/z): [M+1]=535


10.9 Synthesis of Compound 10A-15

Compound 10A-14 (74.90 mg, 0.140 mmol, 1 equiv) was added to a solution of compound 10A-10 (70 mg, 0.140 mmol, 1 equiv) in DMF (10 mL) at room temperature under nitrogen atmosphere, and HATU (53.27 mg, 0.140 mmol, 1.5 equiv) and DIPEA (24.14 mg, 0.187 mmol, 1.25 equiv) were added at room temperature and stirred for 4 h. The reaction solution was quenched with water and extracted with EA (3×100 mL). The organic phases were combined, dried, and concentrated to dryness by rotary evaporation. The resulting crude product was purified by reversed-phase column chromatography (conditions were as follows: C18 chromatographic column, mobile phase: solvent ACN and solvent H2O (0.1% FA), gradient 10% to 50% for 10 min, UV254 nanometer detector), and the fractions were concentrated to dryness by rotary evaporation to give compound 10A-15 (118 mg, 83.1%, purity: 99%).


LC-MS-10A-15 (ES, m/z): [M+1]=1017


10.10 Synthesis of Compound 10A-16

A solution of compound 10A-15 (118 mg, 0.116 mmol, 1 equiv) in piperidine (1 mL) and DCM (10 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting solution was concentrated under reduced pressure to give compound 10A-16 (94 mg, crude), which was directly used in the next step.


LC-MS-10A-16 (ES, m/z): [M+1]=916


10.11 Synthesis of Compound 10A

A mixed solution of compound 10A-16 (94 mg, 0.118 mmol, 1 equiv) in TFA (1 mL)/DCM (5 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting mixture filtrate was concentrated under reduced pressure. The resulting crude product was purified by reversed-phase column chromatography (conditions were as follows: C18 chromatographic column, mobile phase: solvent ACN and solvent H2O (0.1% FA), gradient 10% to 50% for 10 min, UV254 nanometer detector), and the fractions were concentrated to dryness by rotary evaporation to give compound 10A (0.0233 g, 28.22%, purity: 99.0%).


LC-MS-10A (ES, m/z): [M+1]=694



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 1H), 7.30 (s, 3H), 7.30-7.19 (m, 4H), 7.14 (dd, J=6.8, 4.0 Hz, 3H), 4.61 (t, J=6.7 Hz, 1H), 4.53 (t, J=7.5 Hz, 1H), 4.37 (d, J=13.1 Hz, 1H), 4.22 (t, J=7.9 Hz, 1H), 4.00 (t, J=13.1 Hz, 2H), 3.91 (d, J=5.7 Hz, 1H), 3.64 (s, 3H), 3.21 (s, 2H), 3.13 (d, J=13.5 Hz, 1H), 2.94 (ddd, J=15.8, 9.8, 4.5 Hz, 6H), 2.66 (s, 1H), 1.81 (q, J=7.3 Hz, 3H), 1.74 (d, J=14.4 Hz, 2H), 1.46 (d, J=8.1 Hz, 4H), 1.26 (dd, J=14.7, 7.1 Hz, 3H), 0.85 (dt, J=20.3, 5.0 Hz, 6H).


Example 11A. Synthesis of Compound 11A



embedded image


LC-MS-11A (ES, m/z): [M+1]=680



1H NMR (400 MHz, Deuterium Oxide) δ 8.35 (s, 1H), 7.24 (ddd, J=11.2, 7.7, 5.5 Hz, 6H), 7.10 (t, J=9.0 Hz, 4H), 4.69 (s, 2H), 4.58 (t, J=7.1 Hz, 1H), 4.48 (t, J=7.4 Hz, 1H), 4.36 (s, 1H), 4.20 (t, J=8.0 Hz, 1H), 4.08 (s, 1H), 4.00 (d, J=13.6 Hz, 1H), 3.68 (d, J=6.7 Hz, 1H), 3.64 (s, 1H), 3.47-3.39 (m, 2H), 3.14 (t, J=13.3 Hz, 1H), 3.06-2.98 (m, 2H), 2.90 (s, 2H), 2.85 (dd, J=17.8, 6.6 Hz, 2H), 2.65 (s, 1H), 1.75 (s, 2H), 1.51 (s, 3H), 1.43 (s, 3H), 1.24 (dd, J=12.7, 7.0 Hz, 3H), 0.82 (dt, J=20.9, 5.0 Hz, 6H).


Example 12A. Synthesis of Compound 12A



embedded image


LC-MS-12A (ES, m/z): [M+1]=637



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 1H), 7.32-7.17 (m, 6H), 7.12 (dd, J=7.2, 3.8 Hz, 4H), 4.68 (s, 1H), 4.61-4.46 (m, 1H), 3.88 (t, J=6.9 Hz, 2H), 3.61 (s, 2H), 3.56 (s, 4H), 3.21 (q, J=13.1 Hz, 3H), 2.98-2.88 (m, 4H), 1.93 (s, 1H), 1.84 (d, J=13.1 Hz, 1H), 1.43 (s, 2H), 1.30 (s, 2H), 1.23 (t, J=7.0 Hz, 4H), 0.85-0.80 (d, J=4.9 Hz, 6H).


Example 1B. Synthesis of Compound 1B



embedded image


1.1 Synthesis of Compound 1B-2

Intermediate 2 (750 mg, 0.995 mmol, 1.00 equiv) and DIEA (192.85 mg, 1.492 mmol, 1.5 equiv) were dissolved in DMF (30 mL) at room temperature under nitrogen atmosphere, and HATU (567.37 mg, 1.492 mmol, 1.5 equiv) and compound 1B3-1 (200 mg, 1.492 mmol, 1.5 equiv) were added portionwise. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. The resulting crude product was purified by a reversed-phase column under the following conditions: C18 column, mobile phase: solvent ACN and solvent H2O (0.1% formic acid), gradient 10% to 50% for 10 min, UV200 nanometer detector. The resulting fractions were concentrated to dryness by rotary evaporation to give tert-butyl N-[(5R)-5-[(2R)-2-[(2R)-2-[(2R)-2-[(tert-butoxycarbonyl)amino]-3]-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-(1-imino-1-oxo-1λ6-thiomorpholin-4-yl)-6-oxohexyl]carbamate (600 mg, 69.32%, purity: 99%) as a pale yellow solid.


LC-MS-1B-2 (ES, m/z): [M+1]=870


1.2 Synthesis of Compound 1B

tert-Butyl N-[(5R)-5-[(2R)-2-[(2R)-2-[(2R)-2-[(tert-butoxycarbonyl)amino]-3]-phenylpropionylamino]-3-phenylpropionylamino]-4-methylpentanoylamino]-6-(1-imino-1-oxo-1λ6-thiomorpholin-4-yl)-6-oxohexyl]carbamate (80 mg, 0.092 mmol, 1.00 equiv) and a 4 M solution of HCl in 1,4-dioxane (8 mL) were added to a 25 mL round-bottom flask. The resulting mixture was stirred at room temperature for 2 h. The resulting mixture was concentrated under reduced pressure. The resulting crude product was purified by a reversed-phase column under the following conditions: C18 column, mobile phase: solvent ACN and solvent H2O (0.1% formic acid), gradient 10% to 50% for 10 min, detector: UV200 nm to give (R)—N—((R)-6-amino-1-(1-imino-1-oxo-1λ6-thiomorpholine)-1-oxohex-2-yl)-2-((R)-2-((R)-2-amino-3-phenylpropionylamino)-3-phenylpropionylamino)-4-methylpentanamide (10.2 mg, 15.90%, purity: 95.9%).


LC-MS-1B (ES, m/z): [M+1]=670



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 2H)(HCOOH), 7.63-7.01 (m, 10H), 4.56 (t, J=7.5 Hz, 1H), 4.37-3.87 (m, 5H), 3.85-3.67 (m, 1H), 3.52 (t, J=12.4 Hz, 1H), 3.40-3.07 (m, 4H), 3.11-2.81 (m, 6H), 1.64 (tt, J=15.1, 7.8 Hz, 4H), 1.52-1.17 (m, 5H), 0.85 (dd, J=21.2, 5.0 Hz, 6H).


Example 2B. Synthesis of Compound 2B



embedded image


According to similar preparation procedures as in Example 1B, the target compound was synthesized, with the difference being that reactant compound




embedded image


was replaced with




embedded image


LC-MS-2B (ES, m/z): [M+1]=684.20



1H NMR (400 MHz, Deuterium Oxide) δ 8.30 (d, J=6.9 Hz, 1H), 7.51-7.05 (m, 10H), 4.62 (dt, J=15.2, 7.6 Hz, 2H), 4.33-4.12 (m, 3H), 4.12-3.90 (m, 1H), 3.87-3.66 (m, 1H), 3.62-3.36 (m, 4H), 3.36-3.04 (m, 3H), 2.98 (dq, J=24.6, 7.8 Hz, 4H), 2.73 (s, 3H), 1.92-1.60 (m, 4H), 1.57-1.21 (m, 5H), 0.87 (dd, J=20.8, 5.0 Hz, 6H).


Example 3B. Synthesis of Compound 3B



embedded image


According to similar preparation procedures as in Example 1B, the target compound was synthesized, with the difference being that reactant compound




embedded image


was replaced with




embedded image


LC-MS-3B (ES, m/z): [M+1]=712.20



1H NMR (400 MHz, Deuterium Oxide) δ 8.37 (s, 1H), 7.43-7.01 (m, 10H), 4.63-4.50 (m, 1H), 4.43 (s, 1H), 4.33-3.98 (m, 4H), 3.98-3.26 (m, 6H), 3.12-2.70 (m, 6H), 2.18-1.91 (m, 3H), 1.74-1.27 (m, 9H), 0.98-0.72 (m, 6H).


Example 4B. Synthesis of Compound 4B



embedded image


According to similar preparation procedures as in Example 1B, the target compound was synthesized, with the difference being that reactant compound




embedded image


was replaced with




embedded image


LC-MS-4B (ES, m/z): [M+1]=728.10



1H NMR (300 MHz, Deuterium Oxide) δ 8.34 (s, 2H), 7.38-7.07 (m, 10H), 4.63-4.04 (m, 5H), 3.96 (q, J=6.5 Hz, 1H), 3.86-3.30 (m, 8H), 3.22 (s, 1H), 3.12-2.59 (m, 6H), 1.90-1.09 (m, 9H), 0.89 (dd, J=13.9, 5.0 Hz, 6H).


The present specification illustrates the preparation of the compounds described above, and it can be understood by those skilled in the art that other compounds not illustrated in the present invention can also be prepared by reference to the general methods and specific examples described above.


Comparative Example 1B



embedded image


This compound was prepared by reference to the method in Example 5 of CN108290926A.


Biological Test Evaluation

The present invention is further described below using test examples, but these examples are not intended to limit the scope of the present invention.


The following CR845 in the present disclosure was prepared by reference to the method in Example 2 of CN 101627049 A.




embedded image


Test Example 1. In Vitro Receptor Binding Assay
1.1 Objective

The affinities of the compounds for μ, Kappa, and delta receptors were detected by using an in vitro isotope labeling method.


1.2 Experimental Materials

Cell lines: CHO-K1-μ, CHO-K1-Kappa, and CHO-K1-delta stably transfected cell strains (Nanjing GenScript Biotech Co., Ltd.)









TABLE 1







Related information about labeled ligands and preparation








Name of labeled ligands














No.
Name
Source
Cat. No.
Batch No.
Suitable receptor
Storage condition
















1

3H-DAMGO

PE
NET902250
2139100
Mu OR
−20° C.


2

3H-U69593

PE
NET952250
2126416
Kappa
−20° C.


3

3H-DADLE

PE
NET648250
2060549
Delta
−20° C.





Note:


PE: PerkinElmer Inc.;













TABLE 2







Related information about non-labeled compounds













Name of non-
Molecular



Suitable
Storage


labeled compounds
weight
Source
Cat. No.
Batch No.
receptor
condition
















DAMGO
513.59
Abcam
Ab120674
Apn113800-1-1
Mu OR
2-8° C.


U69593
356.50
Sigma
U103-25MG
0000102364
Kappa
2-8° C.


DADLE
569.66
Abcam
Ab120673
APN11299-1-3
Delta
2-8° C.





Note:


5-HT: 5-hydroxytryptamine.













TABLE 3







Information about reagents and consumables









Name
Cat. No./batch No.
Manufacturer





Absolute ethanol
20150422
Yonghua Technology Co., Ltd.


Tris
T1378-1KG/BCBQ2051V
SIGMA


EDTA
T20110308
Sinopharm (Hushi)


Magnesium chloride
E010094/GF290086
Sun Chemical Technology




(Shanghai) Co., Ltd.


MICROSCINT PS
6013631/94-19221
Perkin Elmer


scintillation solution


PEI
306185-100G/07518TE
SIGMA


UniFilter-96 GF/C plate
6005174
Perkin Elmer





Note:


Tris: tris(hydroxymethyl)aminomethane; EDTA: ethylenediaminetetraacetic acid; PEI: polyetherimide.













TABLE 4







Instrument information









Name
Model
Manufacturer





PH meter
PHS-3C
Shanghai REX Sensor




Technology Co., Ltd.


Precision electronic
MS105DU
METTLER TOLEDO


balance


Liquid scintillation
Microbeta 2450
Perkin Elmer Inc.


instrument


High-speed centrifuge
J26xpi
Beckman Coulter


High-speed dispersion
IKA-T10
IKA


machine









1.3 Experimental Methods
1.3.1 Buffer Preparation

A: (for the preparation of μ, Kappa, and delta receptor membranes): 11.7 mg of EDTA and 380.84 mg of MgCl2 were weighed out, and 50 mM Tris-HCl buffer was added to reach a total volume of 400 mL, and the solution was adjusted to pH 7.4 to make final concentrations of 0.1 mM for EDTA and 10 mM for MgCl2.


B: preparation of compound: the theoretical sample weighing amount was calculated according to the designed concentration and the required volume. 5.0×10−3 M was used as an initial preparation dose, and the solution was dissolved in DMSO; the mixture was sequentially diluted with DMSO to 5.0×10−4 M-5.0×10−9 M; the diluted DMSO solution was diluted with Buffer to a working concentration, 5.0×10−5 M-5.0×10−11 M; and the final concentration of DMSO in the working solution was 1% (the final concentration of DMSO in the reaction system was 0.2%). The test sample was prepared and then stored at 4° C. After the test was completed, the test sample was discarded.


1.3.2 Preparation of Receptor Membranes

CHO-μ, Kappa, delta, etc. cells were taken out of a refrigerator at −80° C., thawed spontaneously, and centrifuged at 1000 g for 10 min at 4° C. The precipitate was collected, and the supernatant was discarded. Buffer was added to the precipitate, and the mixture was homogenized for 20-30 s and then centrifuged at 50000 g for 15 min at 4° C. The supernatant was carefully discarded, and Buffer was added into the precipitate again. The cells were mixed well, and the mixture was centrifuged at 50000 g for 15 min at 4° C. The procedures were repeated three times. The sample was stored at −80° C.


1.3.3 Competitive Receptor Binding Assay
3.3.1 μ Receptor Affinity Assay

Step 1: 50 μL of a vehicle (1% DMSO) was added into a total binding tube (TB), 50 μL of DAMGO (with a final concentration of 1.0×10−5 M) was added into a non-specific binding tube (NB), and 50 μL of a test compound was added into each compound binding tube (CB).


Step 2: 100 μL of a buffer (homogenate A) was added into each reaction tube.


Step 3: the prepared membrane was suspended in the homogenate A to obtain a 10 mg/mL membrane suspension for later use.


Step 4: 50 μL of radioligand [3H] DAMGO was added into each reaction tube to reach a final concentration of 2 nM.


Step 5: 50 μL of the membrane solution was added into each reaction tube.


Step 6: each reaction tube was incubated at 25° C. for 90 min. After the reaction was completed, the binding ligands were rapidly filtered under reduced pressure. The UniFilter GF/C plate was saturated with 0.5% PEI solution 1 h in advance, fully washed with cold Tris buffer, filtered under vacuum, then placed into a thermostatic dryer, and dried for 30 min. The filter plate was taken out, and MICROSCINT PS scintillation solution was added at 40 μL/well.


Step 7: the scintillation vial was put into a liquid scintillation counter for counting.


3.3.2 Competitive Kappa Receptor Binding Assay

Step 1: 50 μL of a vehicle (1% DMSO) was added into a total binding tube (TB), 50 μL of U69593 (with a final concentration of 1.0×10−5 M) was added into a non-specific binding tube (NB), and 50 μL of a test compound was added into each compound binding tube (CB).


Step 2: 100 μL of a buffer (homogenate A) was added into each reaction tube.


Step 3: the prepared membrane was suspended in the homogenate A to obtain a 15 mg/mL membrane suspension for later use.


Step 4: 50 μL of radioligand 3H-U69593 was added into each reaction tube to reach a final concentration of 2 nM.


Step 5: 50 μL of the membrane solution was added into each reaction tube.


Step 6: each reaction tube was incubated at 25° C. for 90 min. After the reaction was completed, the binding ligands were rapidly filtered under reduced pressure. The UniFilter GF/C plate was saturated with 0.5% PEI solution 1 h in advance, fully washed with cold Tris buffer, filtered under vacuum, then placed into a thermostatic dryer, and dried for 30 min. The filter plate was taken out, and MICROSCINT PS scintillation solution was added at 40 μL/well.


Step 7: the filter plate was put into a liquid scintillation counter for counting.


3.3.3 Competitive Delta Receptor Binding Assay

Step 1: 50 μL of a vehicle (1% DMSO) was added into a total binding tube (TB), 50 μL of DADLE (with a final concentration of 1.0×10−5 M) was added into a non-specific binding tube (NB), and 50 μL of a test compound was added into each compound binding tube (CB).


Step 2: 100 μL of a buffer (homogenate A) was added into each reaction tube.


Step 3: the prepared membrane was suspended in the homogenate A to obtain a 10 mg/mL membrane suspension for later use.


Step 4: 50 μL of radioligand 3H-DADLE was added into each reaction tube to reach a final concentration of 4 nM.


Step 5: 50 μL of the membrane solution was added into each reaction tube.


Step 6: each reaction tube was incubated at 25° C. for 90 mp. After the reaction was completed, the binding ligands were rapidly filtered under reduced pressure. The UniFilter GF/C plate was saturated with 0.5% PEI solution 1 h in advance, fully washed with cold Tris buffer, filtered under vacuum, then placed into a thermostatic dryer, and dried for 30 min. The filter plate was taken out, and MICROSCINT PS scintillation solution was added at 40 μL/well.


Step 7: the filter plate was put into a liquid scintillation counter for counting.


1.4 Data Analysis

Based on the effect values of different concentration test points of the compound samples, the GraphPad Prism software was used to fit the action curve of the compound samples to the receptors, and the Ki values were calculated. The results are shown in Table 5.









TABLE 5







Receptor binding Ki values













κ receptor
μ receptor
δ receptor



Compound
Ki (nM)
Ki (nM)
Ki (nM)
















 2-0
1.15
>1000
>1000



 2-100
32.51
>1000
>1000



 3
3.31
>1000
>1000



 4-0
2.80
>1000
>1000



 4-100
59.93
>1000
>1000



 8
0.09
>1000
>1000



 9
11.15
>1000
>1000



10
0.86
>1000
>1000



11
0.94
>1000
>1000



12
0.53
>1000
>1000



13
0.13
>1000
>1000



13-2
0.13
>1000
>1000



14
1.10
>1000
>1000



15
0.83
>1000
>1000



16
0.26
>1000
>1000



17
0.44
>1000
>1000



18
0.21
>1000
>1000



19
0.90
>1000
>1000



20
0.58
>1000
>1000



21
0.33
>1000
>1000



22
0.35
>1000
>1000



23
0.36
>1000
>1000



24
0.36
>1000
>1000



25
0.45
>1000
>1000



26
0.31
>1000
>1000



27
0.88
>1000
>1000



28
3.31
>1000
>1000



29
0.63
>1000
>1000



30
1.61
>1000
>1000



31
3.49
>1000
>1000



32
0.58
>1000
>1000



33
0.56
>1000
>1000



34
0.60
>1000
>1000



35
0.18
>1000
>1000



36
0.98
>1000
>1000



37
0.41
>1000
>1000



38
0.17
>1000
>1000



39
0.27
>1000
>1000



40
0.58
>1000
>1000



41
0.57
>1000
>1000



42
0.47
>1000
>1000



43
2.00
>1000
>1000



44
0.55
>1000
>1000



45
0.52
>1000
>1000



46
0.29
>1000
>1000



47
0.49
>1000
>1000



48
3.79
>1000
>1000



49
0.93
>1000
>1000



50
0.27
>1000
>1000



51
0.35
>1000
>1000



55
10.63
>1000
>1000



69
14.37
>1000
>1000



 1A
0.40
>1000
>1000



 2A
0.34
>1000
>1000



 3A
0.26
>1000
>1000



 4A
0.30
>1000
>1000



 5A
0.34
>1000
>1000



 6A
0.43
>1000
>1000



 7A
0.60
>1000
>1000



 8A
0.31
>1000
>1000



 9A
0.37
>1000
>1000



10A
0.48
>1000
>1000



11A
0.48
>1000
>1000



12A
0.34
>1000
>1000



 1B
0.42
>1000
>1000



 2B
0.91
>1000
>1000



 3B
1.05
>1000
>1000



 4B
1.88
>1000
>1000










It can be seen that the compounds of the present invention have excellent selectivity for κ-opioid receptors and high affinity for the κ-opioid receptors.


Test Example 2. In Vitro Functional Assay
2.1 Objective

Using the Cisbio HTRF cAMP-Gi kit, changes in the cAMP concentration of the κ-opioid receptor (Kappa) signaling pathway were detected with a microplate reader, and EC50 values of the compounds were calculated to evaluate agonistic effects of the compounds on the κ-opioid receptors.


2.2 Experimental Materials





    • Cell line: CHO-K1-OPRK1 stably transfected cell strain (Nanjing GenScript Biotech Co., Ltd.)

    • Cell culture conditions: F12, 10% FBS, 200 μg/mL Zeocin, 100 μg/mL Hygromycin B

    • Reagents and consumables:

    • F12 (meilunbio, MA0229)

    • FBS (BOVOGEN, SFBS)

    • Zeocin (invitrogen, R25001)

    • Hygromycin B (invitrogen, 10687010)

    • PBS (meilunbio, MA0015)

    • Pancreatin (Gibco, 25200-072)

    • 96 cell plate (cisbio, 66PL96025)

    • cAMP-Gi kit (cisbio, 62AM9PEB)

    • CO2 incubator (Thermo, 311)

    • Centrifuge (Shanghai Anting Scientific Instrument Factory, TGL-16C)

    • Cell counter (Countstar, IC1000)

    • Microplate reader (PerkinElmer, EnVision)





2.3 Experimental Methods

(1) A reaction buffer (1× Stimulation buffer) required for the experiment was prepared as follows: 5× Stimulation buffer and ddH2O in the Cisbio cAMP-Gi kit were diluted according to a ratio of 1:4 for later use.


(2) Preparation of compound: the compound was diluted with DMSO to obtain a 5 mM stock solution, followed by a 3.16-fold dilution to obtain 10 gradients, and then the prepared compound was diluted with the Stimulation buffer to the corresponding concentrations (2.5×) for later use.


(3) Cell preparation: CHO-K1-OPRK1 cells on the culture dish were digested with the pancreatin, and the cells were eluted with the medium and collected into a 5 mL centrifuge tube. The cells were centrifuged at 1000 rpm for 5 min, and the supernatant was discarded. 3 mL of PBS was added, and the resulting mixture was gently pipetted and mixed well using a pipette. The cells were centrifuged again at 1000 rpm for 5 min, and the supernatant was discarded. The cells were resuspended in 1× Stimulation buffer and counted using a Countstar cell counter, and the cell density was adjusted to 6×105 cells/mL for later use.


(4) Cell addition: the cell suspension was added to the assay plate at 5 μL/well (i.e., about 3000 cells/well).


(5) Compound addition: the compound diluted with the Stimulation buffer was added to the assay plate described above at 4 μL/well.

    • (6) Reaction and incubation: after gentle shaking, the assay plate was incubated at 37° C. for 20 min.


(7) Agonist Forskolin addition: a 10× adenylate cyclase agonist (with a final concentration of 1 μM Forskolin) solution was added at 1 μL/well.

    • (8) Reaction and incubation: after gentle shaking, the assay plate was incubated at 37° C. for 45 min.


(9) Detection reagent addition: cAMP-cryptate and Anti-cAMP-d2 were separately diluted according to a ratio of 1:20 with Lysis & detection buffer in the Cisbio cAMP-Gi detection kit, and the diluted cAMP-cryptate and Anti-cAMP-d2 were separately added to the assay plate at 5 μL/well. After shaking, the assay plate was left to stand at room temperature for 60 min.


(10) Experimental readings: the plate was read on Envision, readings from 665 nm and 615 nm channels were measured, and the ratio of 665 nm/615 nm readings was calculated.


2.4. Data Analysis

Based on the agonistic effect values of different concentration test points of the compound samples, the GraphPad Prism software was used to fit the agonistic effect curve of the compound samples to the κ-opioid receptors, and the EC50 values were calculated. The experimental results are shown in Table 6.









TABLE 6







In vitro agonistic effects on k-opioid receptors










Compound
EC50 (nM)














 4-0
3.62



 8
0.90



10
1.1



11
5.1



12
0.17



13
0.52



13-2
0.13



14
8.20



15
0.63



16
0.079



17
0.30



18
0.18



19
0.87



20
0.52



21
0.68



22
0.36



23
0.34



24
0.27



25
0.46



26
0.25



27
0.87



29
0.058



31
1.23



32
0.65



33
0.81



34
0.71



35
0.17



36
0.35



37
0.39



38
0.54



39
0.35



40
0.13



41
0.57



42
0.46



43
6.13



44
0.09



45
0.16



46
0.22



47
0.24



48
12.01



49
3.06



50
0.43



 1A
0.05



 2A
0.36



 3A
0.40



 4A
0.20



 5A
0.30



 6A
0.95



 7A
<0.1



 8A
0.075



 9A
<0.1



10A
0.59



11A
0.21



12A
0.07



 1B
0.52



 2B
0.30



 3B
1.00



 4B
0.65










It can be seen that the compounds of the present invention have relatively strong agonistic effects on the κ-opioid receptors.


Test Example 3. Sedation Safety Window Assay
3.1 Acetic Acid Writhing Test in Mice
3.1.1 Objective

The analgesic effects of the compounds after single administration were investigated by an acetic acid writhing test in mice, and the ED50 values were calculated.


3.1.2 Test System
3.1.2.1 Experimental Animal Information





    • Species and strain: ICR mice

    • Grade: normal

    • Number and sex: male, 100 mice (as the case may be)

    • Age range: 6-8 weeks old

    • Weight range: 23-32 g

    • Supplier: SPF (Beijing) Biotechnology Co., Ltd.

    • Animal use license: SYXK (Jiangsu) 2019-053

    • Treatment of the remaining animals: the remaining animals were handed over to the department of the experimental system for treatment.





3.1.2.2 Instruments and Reagents








TABLE 7







Main instruments and reagents









Name
Model
Manufacturer





Precision electronic
BS224s
Sartorius Scientific Instruments


balance

(Beijing) Co., Ltd.


Electronic balance
YP6001N
Shanghai Precision Instrument




Co., Ltd.


Hot/cold plate
35100
UGO Basile, Italy


Acetic acid
20160408
Shanghai Lingfeng Chemical




Reagents Co., Ltd.









3.1.3 Test Method

The acetic acid writhing test in mice is a classic visceral pain test model. The peritoneum is stimulated to cause a lasting pain response by intraperitoneal injection of a 0.6% aqueous acetic acid solution in the mice, further causing a writhing reaction of the mice. The analgesic drugs can inhibit the writhing reaction of the mice. The mice were adapted for 15 min after intravenous injection of the drug at the tail, then a 0.6% acetic acid solution (0.1 mL/10 g) was intraperitoneally injected. A vehicle control group (0.9% sodium chloride injection) was set. The times of writhing of the mice within 15 min after the injection of acetic acid were observed and recorded, and the writhing inhibition rate of the test compound was calculated to preliminarily evaluate the analgesic effect of single administration.







Writhing


inhibition


rate


%

=


(


times


of


vehicle


group

-

times


of


administration


group


)

×
100


%
/
times



of


vehicle



group
.






3.1.4 Data Processing

The experimental data were expressed as mean±standard deviation (Mean±SD). The one-way ANOVA was carried out by using the GraphPad Prism 8 statistical software, pairwise comparisons were carried out by using the Dunnett t test, and ED50 values were calculated using a non-linear fitting method. The differences were considered statistically significant when P<0.05.


3.2 Autonomous Activity Test in Mice
3.2.1 Objective

The effects of the compounds on the autonomic activity of the mice after single administration were determined, and ED50 values were calculated.


3.2.2 Test System
3.2.2.1 Experimental Animal Information





    • Species and strain: ICR mice

    • Grade: normal

    • Number and sex: male, 200 mice (as the case may be)

    • Age range: 6-8 weeks old

    • Weight range: 23-35 g

    • Supplier: SPF (Beijing) Biotechnology Co., Ltd.

    • Animal use license: SYXK (Jiangsu) 2019-053

    • Treatment of the remaining animals: the remaining animals were handed over to the department of the experimental system for treatment.





3.2.2.2 Instruments and Reagents








TABLE 8







Main instruments and reagents









Name
Model
Manufacturer





Precision electronic balance
BS224s
Sartorius Scientific Instruments




(Beijing) Co., Ltd.


Electronic balance
YP6001N
Shanghai Precision Instrument Co.,




Ltd.


Dahua L series hard disk drive
DH/DVR1604LE-L
Zhejiang Dahua Technology Co., Ltd.


video recorder


TopScan Version 3.00 (animal
Version 3.00
Clever Sys, Inc.


video analysis system)









3.2.3. Test Method

The autonomous activity test in mice is a classical test model for evaluating the inhibition of compounds on the central nervous system. The mice were intravenously injected with a test sample solution with a corresponding concentration (freshly prepared before use) at the tail, and the mice in the blank group were given a vehicle (0.9% sodium chloride injection). After the administration, the mice were immediately placed into an autonomous activity test box (a black polyethylene box with the specification of 29 cm×29 cm×30 cm) to be recorded for 30 min. Video analysis was carried out after the recording was completed, and the autonomous activity of the mice after the administration was evaluated.


3.2.4 Data Processing

The experimental data were expressed as mean±standard deviation (Mean±SD). The one-way ANOVA was carried out by using the GraphPad Prism 8 statistical software, pairwise comparisons were carried out by using the Dunnett t test, and ED50 values were calculated using a non-linear fitting method. The differences were considered statistically significant when P<0.05.


3.3 Safety Window

Sedation is the most significant side effect of peripheral Kappa receptor agonists, and the sedation safety window can be obtained according to the sedation side effect ED50 (autonomic activity test)/efficacy ED50 (acetic acid writhing test), with a larger safety window indicating higher safety and a lower probability of the clinical sedation side effect at the same dose. The experimental results are shown in Table 9.









TABLE 9







Safety window











Acetic acid writhing
Autonomous activity
Safety


Compound
in mice ED50 (mg/kg)
in mice ED50 (mg/kg)
window













 4-0
1.14
7.34
6.44


11
1.82
9.60
5.27


13
0.23
2.83
12.30


13-2
0.17
2.75
16.18


16
0.27
1.37
5.13


21
0.50
1.42
2.82


27
0.88
4.59
5.22


29
0.41
1.35
3.28


 1A
0.38
1.08
2.84


 3A
0.45
1.41
3.15


 4A
0.41
1.25
3.05


 8A
0.16
5.93
37.06


10A
1.61
5.52
3.43


 1B
0.20
0.50
2.50


 2B
0.31
3.33
10.74


 3B
0.42
3.25
7.74


Comparative
0.25
0.59
2.36


Example 1B









It can be seen that the compounds of the present invention have a very large safety window, i.e., have a greater safe dose range of administration, or are less likely to have side effects at the same dose.


Test Example 4. PK and Blood-Brain Barrier Permeability in Mice
4.1 Objective

The compounds of the present disclosure were investigated for pharmacokinetic characteristics and blood-brain barrier permeability in mice.


4.2 Test Devices








TABLE 10







Device information









Name
Model
Manufacturer





AB triple quadrupole liquid
5500+
Sciex


chromatography-mass


spectrometry instrument


Mettler-Toledo analytical
XSE105
Mettler-Toledo Group


balance


Benchtop high-speed refrigerated
TL-18M
Shanghai Centrifuge Institute Co.,


centrifuge

Ltd.


Eppendorf micropipette
20/200/1000 μL
Eppendorf, Germany


Digital display constant-
SHA-B(A)
Jinyi Instrument Technology


temperature oscillator

Company Limited


Benchtop ultrasonic cleaner
CPX8800H
Emerson, USA


Vortex oscillator
Vortex genie 2
Scientific Industries, USA


Acetonitrile
Chromatography grade
Tedia, USA


Methanol
Chromatography grade
Tedia, USA


Watsons purified water
600 mL
Watsons Group


Formic acid
095324
MREDA, USA









4.3 Test Method and Data Processing

Healthy male ICR mice were randomly grouped, 6 animals per group. For each test compound, 3 groups of animals were used. The first group of animals was used for collecting blood and brain 5 min after the administration, and the second group of animals and the third group of animals were used for cross blood collection at subsequent time points; each test compound was intravenously administered (1 mg/kg) to corresponding groups of mice, and blood sampling via submaxillary veins of the mice was carried out at different time points. Drug contents in plasma and brain tissues at different time points were measured by an LC-MS/MS method, the brain-to-blood ratio B/P was calculated by using EXCEL software, and pharmacokinetic parameters were calculated by using DAS3.0 software. The experimental results are shown in Table 11.









TABLE 11







Pharmacokinetic parameters















AUC(0-∞)
Clearance
Vd




Compound
Cmax (ng/mL)
(μg/L*h)
(L/h/kg)
(L/kg)
(h)
B/P
















10
3224.52
4010.04
0.25
0.29
0.82
0.007


12
2264.57
3681.68
0.28
0.54
1.42
0.008


13
2538.43
1351.65
0.74
0.38
0.35
0.015


13-2
2929.28
1685.68
0.60
0.63
0.72
BQL**


16
4729.52
2503.55
0.40
0.51
0.87
0.010


21
3461.88
2667.60
0.38
0.63
1.15
0.010


27
2316.41
1163.46
0.88
0.63
0.50
0.010


29
4707.93
3343.19
0.31
0.32
0.74
0.010


CR845
1621.13
652.67
1.62
0.60
0.26
0.015





**below the lower limit of quantitation, indicating that the test substance was undetectable in the brain tissue.






It can be seen that the compounds of the present invention have excellent pharmacokinetic properties. Particularly, compared to CR845, the compounds of the present invention have comparable or lower brain-to-blood ratios B/P, which can selectively act on the peripheral Kappa receptors, and the compounds of the present invention have significantly improved pharmacokinetic characteristics and significantly longer half-lives.


Test Example 5. Toxicity Test of Single Administration in Mice
5.1 Objective

Safety data were supplemented by researching the toxic responses of the compounds of the present invention after a single intravenous administration in mice.


5.2 Test System





    • Species and strain: ICR mice

    • Grade: SPF grade

    • Sex: male

    • Weight range: 24-28 g

    • Supplier: SPF (Beijing) Biotechnology Co., Ltd.

    • Animal production license: SCXK (Beijing) 2019-0010

    • Animal use license: SYXK (Jiangsu) 2019-0053

    • Treatment of the remaining animals: the remaining animals were handed over to the department of the test system for treatment.





5.3 Instruments and Reagents








TABLE 12







Main instruments and reagents









Name
Model
Manufacturer





Precision electronic
BS224s
Sartorius Scientific Instruments


balance

(Beijing) Co., Ltd.


Electronic balance
YP6001N
Shanghai Precision Instrument




Co., Ltd.


Sodium chloride
2204141334
Shandong Cisen Pharmaceutical


injection

Co., Ltd.









5.4 Test Method

The ICR mice were randomly grouped, 10 mice per group. The mice were intravenously administrated via the tail at 10 mL/kg. The animals were observed for state and mortality and recorded. The administration was carried out sequentially according to the recommended dose of AOT425 software until the LD50 values were determined.


5.5 Data Processing

The LD50 values were calculated using the AOT425 software. The results are shown in Table 13.









TABLE 13







Acute toxicity










Example No.
LD50 (mg/kg)














8A
175



2B
151.6



3B
150










It can be seen that the compounds of the present invention have relatively low single administration toxicity, and thus have improved safety.


Test Example 6. Research on Effect of Compounds on hERG Potassium Ion Channel
6.1 Objective

In this experiment, the inhibitory effects of the compounds on the hERG channel were investigated by using the manual patch-clamp technique.


6.2 Test System

Non-cardiac drugs may cause prolongation of myocardial action potential duration by inhibiting hERG (IKr) channels, thereby increasing the likelihood of life-threatening torsades de pointes (TdP) ventricular arrhythmias. In this experiment, an HEK293 cell line without endogenous IKr current was used as a host cell. The cell line is widely applied to the detection of hERG.


HEK293 cells stably expressing the hERG channel were cultured in a 35 mm culture dish and placed in a 37° C. C/5% CO2 incubator for at least 24 h for later use. The hERG cell line was routinely cultured and passaged in DMEM containing 10% fetal bovine serum and 250 μg/mL of G418.


6.3 Liquid Preparation

The extracellular fluid used in the whole-cell patch-clamp experiment was composed of (mM): NaCl 145; MgCl2 1; KCl 4; Glucose 10; HEPES 10; CaCl2 2; the pH value was adjusted to 7.4 with NaOH, and osmotic pressure value was adjusted to 300 mOsm with sucrose.


The intracellular fluid was composed of (mM): KCl 140; MgCl2 1; EGTA 5; HEPES 10 and Na2ATP 4; the pH value was adjusted to 7.2 with KOH, and osmotic pressure value was adjusted to 290 mOsm with sucrose.









TABLE 14







Main reagent concentrations









Reagent
Extracellular fluid (mM)
Intracellular fluid (mM)












CaCl2
2



MgCl2
1
1


KCl
4
140


NaCl
145



Glucose
10



HEPES
10
10


EGTA

5


Na2ATP

4


pH
7.4
7.2









6.4 Test Method

Electrophysiological recording: one culture dish was taken out for each experiment, washed twice with the extracellular fluid and placed on the stage of an inverted microscope. The whole-cell patch-clamp experiment was carried out at room temperature, and the tip resistance of the borosilicate glass microelectrode was 3-5 MΩ.


Voltage stimulation protocol and current recording: after the whole-cell recording mode, the membrane potential was clamped at −80 mV, the cells were given a depolarization voltage stimulation of +50 mV every 30 s for 2 s, and repolarization was carried out to −50 mV for 3 s, causing an hERG tail current. Before the depolarization voltage stimulation, the cells were given a repolarization voltage of −50 mV for 50 ins, and the current recorded at this voltage served as the baseline for calculation of the hERG tail current. Only cells that met the recording criteria could be used for detection of the test compounds. Before the addition of the compounds, the hERG tail current in the extracellular fluid was stably recorded for at least 3 min. After perfusion administration, when the hERG tail current amplitude change was less than 5%, it was considered that the drug action reached a steady state. If the current did not reach steady state within 6 min, the compound detection at this concentration was terminated.


6.5 Data Processing

The raw data were recorded by using Clampex 10.2, and data acquisition and analysis were carried out by using the pCLAMP 10.1 software program. 4-5 sweeps with the current in a steady state before the addition of the compounds were selected, and the average value of peak values was calculated to serve as a control current amplitude. 4-5 sweeps with the current in a steady state after the addition of the compounds were selected, and the average value of peak values w as calculated to serve as the residual amplitude after the current w as inhibited. The inhibition rate of the test compound on the hERG current was calculated according to the following equation:





% inhibition rate={1−(current residual amplitude)/(control current amplitude)}×100


The inhibition rate of the test compounds on the hERG current at a concentration of 10 μM was obtained according to the calculation method described above. The results are shown in Table 15.









TABLE 15







Cardiotoxicity










Example No.
% inhibition rate







 4
0.54% ± 1.72%



12
2.13% ± 2.96%



13
0.80% ± 0.77%



 1A
−0.94% ± 0.76% 



 2A
1.00% ± 1.86%



 4A
0.90% ± 3.50%



 5A
4.15% ± 0.07%



 6A
1.04% ± 0.53%



 1B
1.20% ± 0.39%



 3B
1.13% ± 0.00%










It can be seen that the compounds of the present invention have relatively low cardiotoxicity, and thus have improved safety.


The above examples merely illustrate the activity data of certain representative compounds of the present invention, and other compounds provided by the present invention also have similar effects when tested by the same methods.

Claims
  • 1-24. (canceled)
  • 25. A compound of formula IA, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
  • 26. The compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 25, being a compound of formula IIA, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
  • 27. The compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 25, being a compound of formula IIIA, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
  • 28. The compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 25, being a compound of formula IVA, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
  • 29. The compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 25, being a compound of formula VA, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
  • 30. The compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 25, wherein RbA is selected from C1-6 alkyl or 3- to 8-membered heterocyclyl; preferably from C1-6 alkyl or
  • 31. The compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 25, being a compound of formula VIA, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
  • 32. The compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 25, being selected from the following compounds, stereoisomers thereof, or pharmaceutically acceptable salts thereof:
  • 33. A compound of formula IB, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
  • 34. The compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 33, being selected from the following compounds, stereoisomers thereof, or pharmaceutically acceptable salts thereof:
  • 35. A compound of formula IC, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
  • 36. The compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 35, being a compound of the following general formula, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
  • 37. The compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 35, being selected from the following compounds, stereoisomers thereof, or pharmaceutically acceptable salts thereof:
  • 38. A pharmaceutical composition comprising the compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 25, and a pharmaceutically acceptable carrier or excipient, as well as optionally other therapeutic agents.
  • 39. A pharmaceutical composition comprising the compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 33, and a pharmaceutically acceptable carrier or excipient, as well as optionally other therapeutic agents.
  • 40. A pharmaceutical composition comprising the compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 35, and a pharmaceutically acceptable carrier or excipient, as well as optionally other therapeutic agents.
  • 41. A method for preventing and/or treating a subject having a disease or conditions mediated by κ-opioid receptor comprising administering to a patient the compound, the stereoisomer thereof, or the pharmaceutically acceptable salt thereof according to claim 25.
  • 42. The method according to claim 41, wherein, the disease is selected from pain, inflammation, pruritus, edema, hyponatremia, hypokalemia, ileus, cough, and glaucoma, preferably pain; preferably, the pain selected from neuropathic pain, trunk pain, visceral pain, skin pain, arthritis pain, kidney stone pain, uterine cramp, dysmenorrhea, endometriosis, dyspepsia, post-surgical pain, post-medical treatment pain, ocular pain, otitis pain, breakthrough cancer pain, and pain associated with a GI disorder.
  • 43. A method for preventing and/or treating a subject having a disease or conditions mediated by κ-opioid receptor comprising administering to a patient the pharmaceutical composition according to claim 38.
  • 44. The method according to claim 43, wherein, the disease is selected from pain, inflammation, pruritus, edema, hyponatremia, hypokalemia, ileus, cough, and glaucoma, preferably pain; preferably, the pain selected from neuropathic pain, trunk pain, visceral pain, skin pain, arthritis pain, kidney stone pain, uterine cramp, dysmenorrhea, endometriosis, dyspepsia, post-surgical pain, post-medical treatment pain, ocular pain, otitis pain, breakthrough cancer pain, and pain associated with a GI disorder.
Priority Claims (3)
Number Date Country Kind
202210291100.9 Mar 2022 CN national
202210291141.8 Mar 2022 CN national
202210291169.1 Mar 2022 CN national
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2023/083063 3/22/2023 WO