IMINO-UREA DERIVATIVES

Information

  • Patent Application
  • 20200325109
  • Publication Number
    20200325109
  • Date Filed
    April 27, 2018
    6 years ago
  • Date Published
    October 15, 2020
    3 years ago
Abstract
The present invention relates to the field of medicines, and in particular to an imino-urea derivative containing a furan structure and a composition thereof. The derivative and the composition thereof can be used in the manufacture of a medicament for treating a disease pathologically characterized by indoleamine 2,3-dioxygenase-mediated tryptophan metabolic pathway. The present invention also relates to a method for preparing the derivative and an intermediate thereof.
Description
TECHNICAL FIELD

The invention relates to the field of medicines, in particular to an imino-urea derivative and a preparation method and a use thereof.


BACKGROUND ART

Indoleamine 2,3-dioxygenase, a monomeric enzyme containing heme, is capable of catalyzing the oxicracking of indole ring of L-tryptophan to form kynurenine. The high expression of indoleamine 2,3-dioxygenase results in depletion of local tryptophan in cells, which induces T cell arrest in G1 phase, thereby inhibiting T cell proliferation. On the other hand, the degradation of indoleamine 2,3-dioxygenase-dependent tryptophan leads to an increase in kynurenine level and also induces oxygen free radical-mediated T cell apoptosis. Thirdly, the up-regulation of dendritic cell indoleamine 2,3-dioxygenase expression enhances local regulatory T cell (Treg)-mediated immunosuppression by degrading local tryptophan, thereby promoting peripheral immune tolerance of body to tumor specific antigen. Indoleamine 2,3-dioxygenase has become the most important small molecule regulatory target for anti-tumor immunotherapy.


It has been found in studies that indoleamine 2,3-dioxygenase is associated with many physiological processes in human body. In 1998, Munn et al. revealed that the fetus was able to survive in the mother body with different genotype during pregnancy without being rejected because the placental plasmoditrophoblast cells synthesized indoleamine 2,3-dioxygenase, which inhibited the rejection reaction of maternal T cells against the fetus through blood flow. After further subcutaneous implantation of the sustained-release capsule containing indoleamine 2,3-dioxygenase inhibitor 1-methyltryptophan in pregnant mice, they found that the embryo was rejected and aborted (Munn D H, Zhou M, Attwood J T, et al. Prevention of allogeneic fetal rejection by tryptophan catabolism. Scienice, 1998, 281 (5380): 1191-3). In addition, some diseases caused by abnormal immune responses, such as transplant rejection and autoimmune diseases, are also closely related to indoleamine 2,3-dioxygenase.


Although great progress has been made in treatment of tumors in recent years, the clinical efficacy is still unsatisfactory. Immune escape is one of the main biological mechanisms of tumorigenesis and tumor metastasis, and has become an important factor affecting the therapeutic effect of tumors. Indoleamine 2,3-dioxygenase, as an immune regulatory enzyme, can effectively inhibit T cell function, enhance Treg cell function, and induce NK cell dysfunction, while tumor cells can use these inherent immune regulation mechanisms in body to escape from the identification and killing of immune system (Jia Yunlong, Wang Yu, Chinese Journal of Cancer Biotherapy, 2004, 21 (6): 693-7). In order to enable tumor patients to get optimum benefit from treatment, it is imperative to rationally adjust the treatment strategy for tumor immune escape. The indoleamine 2,3-dioxygenase inhibitor of the present invention can effectively regulate the immune system of patient, block the immune escape of tumor cells, and has a good therapeutic effect on most spontaneous tumors. Based on the regulation of immune system, in addition to treating tumors, the indoleamine 2,3-dioxygenase inhibitor of the present invention can also treat other diseases related to immunity, such as chronic infection and AIDS.


Indoleamine 2,3-dioxygenase is also closely related to neurological diseases. It can lower the level of 5-hydroxytryptamine and cause mental illnesses such as depression and anxiety. It can also cause accumulation of neurotoxic metabolites such as quinolinic acid in brain, which is closely related to the occurrence of neurodegenerative diseases such as Alzheimer's disease. Indoleamine 2,3-dioxygenase can influence brain function by at least two mechanisms: 1) during an inflammatory response, the catabolism of tryptophan can result in a decrease in circulating tryptophan concentrations, thereby resulting in a decrease in 5-hydroxytryptamine level, which leads to depression; 2) indoleamine 2,3-dioxygenase catabolizes tryptophan into products that enter the kynurenine pathway, resulting in the accumulation of kynurenine and neurotoxic quinolinic acid (Kong Linglei, Kuang Chunxiang, Yang Qing, Chinese Journal of Medicinal Chemistry, 2009, 19(2): 147-154).


Contents of the Invention:

The present invention provides a compound, or a pharmaceutically acceptable salt thereof, a composition containing the compound or a pharmaceutically acceptable salt thereof, and a method for inhibiting the activity of indoleamine 2,3-dioxygenase (IDO) by using the compound or a pharmaceutically acceptable salt thereof, or a method for treating a disease pathologically characterized by indoleamine 2,3-dioxygenase-mediated tryptophan metabolic pathway by using the compound or a pharmaceutically acceptable salt thereof, and a use of the compound or a pharmaceutically acceptable salt in manufacture of a medicament for inhibiting the activity of indoleamine 2,3-dioxygenase, or for treating a disease pathologically characterized by indoleamine 2,3-dioxygenase-mediated tryptophan metabolic pathway.


The compound or a pharmaceutically acceptable salt thereof has excellent activity for inhibiting indoleamine 2,3-dioxygenase, and the activity is significantly superior to other IDO inhibitors. In addition, by measuring the body weight of mice before and after the administration of the compound or a pharmaceutically acceptable salt thereof, it is found that as compared to other IDO inhibitors, the compound of the present invention or a pharmaceutically acceptable salt thereof can significantly improve the life quality of mice during tumor treatment and significantly reduce side effects. In clinical practice, the compound of the present or a pharmaceutically acceptable salt thereof will improve not only patient's life quality, but also significantly improve patient compliance with medications and medicament effectiveness. The compound of the present invention or a pharmaceutically acceptable salt thereof can significantly improve the learning and memory impairment in animals and enhance learning acquisition ability and spatial memory ability, has positive therapeutic significance for neurodegenerative diseases such as Alzheimer's syndrome, and is superior to other IDO inhibitors. The compound of the present invention or a pharmaceutically acceptable salt thereof can promote the function of DC in stimulation of T cell proliferation, so that it can be used for treating tumor diseases, autoimmune diseases, transplant rejection, and infectious diseases, and it is superior to other IDO inhibitors.







DETAILED DESCRIPTION OF THE INVENTION

In some embodiments, the invention provides a compound represented by Formula I0, or a pharmaceutically acceptable salt thereof:




embedded image


wherein, R1, R2 are each independently selected from the group consisting of: H, substituted or unsubstituted C1-10 alkyl, aldehyde group, substituted or unsubstituted carbonyl, cyano, CF3, substituted or unsubstituted C1-10 alkoxy, substituted or unsubstituted sulfonyl, substituted or unsubstituted C3-10 cycloalkyl, substituted or unsubstituted C2-10 alkenyl, substituted or unsubstituted C6-20 aryl, substituted or unsubstituted C3-14 heteroaryl;


R3, R4 are each independently a mono-substituent selected from the group consisting of: H, substituted or unsubstituted C1-10 alkyl, substituted or unsubstituted C3-10 cycloalkyl, cyano, substituted or unsubstituted C1-10 alkoxy, substituted or unsubstituted sulfonyl, substituted or unsubstituted C6-20 aryl, substituted or unsubstituted C3-14 heteroaryl; or


R3, R4 are each independently selected from di-substituents, thereby forming the following groups together with the C atom at a- or b-position: C═O, C═NH or




embedded image


wherein C represents the C atom at a- or b-position, m is an integer selected from 0 to 6, such as 0 or 1 or 2 or 3 or 4 or 5 or 6; further, the group formed by R3, R4 together with the C atom at a- or b-position is C═CH2,




embedded image


n is an integer selected from 0 to 6, such as 0, 1, 2, 3, 4, 5 or 6; preferably n is 0, 1, 2 or 3.


In some embodiments, the present invention provides the above-described Compound represented by Formula I0, or a pharmaceutically acceptable salt thereof, wherein R1, R2 are each independently selected from the group consisting of: C1-6 alkyl, carbonyl, C1-6 alkoxy, sulfonyl, amidino, sulfinyl, which is optionally substituted by one or more substituents selected from group consisting of: halogen, hydroxy, carboxy, carbonyl, aldehyde group, cyano, amino, aryl, heteroaryl, C1-6 alkyl, C3-12 cycloalkyl, C2-6 alkenyl, and C3-12 cycloalkenyl, wherein, the carboxy, carbonyl, aldehyde group, cyano, amino, aryl, heteroaryl, C3-12 cycloalkyl, C2-6 alkenyl, C3-12 cycloalkenyl as the substituents of C1-6 alkyl, carbonyl, C1-6 alkoxy, sulfonyl, amidino or sulfinyl is optionally substituted by one or more substituents selected from group consisting of: H, halogen, C1-6 alkyl, carbonyl, C1-6 alkoxy, sulfinyl and sulfonyl, the halogen is selected from the group consisting of F, Cl, Br and I.


In some embodiments, the present invention provides the above-described Compound represented by Formula I0 or a pharmaceutically acceptable salt thereof, wherein R1, R2 are each independently selected from the group consisting of: H, methyl, ethyl, propyl, isopropyl, R5C(O)—, R5S(O)x-; R5 is selected from the group consisting of C1-10 alkyl, C3-12 cycloalkyl, substituted C1-10 alkyl and substituted C3-12 cycloalkyl, wherein the substituted C1-10 alkyl or substituted C3-12 cycloalkyl is substituted by hydroxy, cyano, C1-6 alkyl, C3-12 cycloalkyl, C1-6 alkoxy, aryl or heteroaryl, wherein x is 1 or 2;


R3, R4 are each independently a mono-substituent selected from the group consisting of: H, substituted or unsubstituted C1-10 alkyl, substituted or unsubstituted C3-10 cycloalkyl, cyano, substituted or unsubstituted C1-10 alkoxy, substituted or unsubstituted sulfonyl, substituted or unsubstituted C6-20 aryl, substituted or unsubstituted C3_14 heteroaryl; or


R3, R4 are each independently selected from di-substituents, thereby forming the following groups together with the C atom at a- or b-position: C═O, C═NH, C═CH2,




embedded image


In some embodiments, the present invention provides the above-described Compound represented by Formula I0 or a pharmaceutically acceptable salt thereof, wherein R1, R2 are each independently selected from the group consisting of: H, methyl, ethyl, propyl, isopropyl, R5C(O)—, R5S(O)x—; R5 is selected from the group consisting of C1-10 alkyl, C3-12 cycloalkyl, substituted C1-6 alkyl, and substituted C3-8 cycloalkyl, the substituted C1-6 alkyl or the substituted C3-8 cycloalkyl is substituted by hydroxy, cyano, C1-6 alkyl, C3-12 cycloalkyl, C1-6 alkoxy, aryl or heteroaryl; x is 2; R3, R4 are both H.


In some embodiments, the present invention provides the above-described Compound represented by Formula I0 or a pharmaceutically acceptable salt thereof, wherein when R2, R3, and R4 in the Formula I0 are H respectively, the compound is represented by Formula I:




embedded image


wherein, R1 is selected from the group consisting of: H, amino, sulfonyl, nitro, carbonyl, amidino, C1-6 alkyl, substituted amidino, substituted C1-6 alkyl, substituted C1-6 alkoxy, substituted carbonyl, substituted sulfonyl and substituted sulfinyl, wherein the substituted amidino, the substituted C1-6 alkyl, the substituted C1-6 alkoxy, the substituted carbonyl, the substituted sulfonyl or the substituted sulfinyl is substituted by halogen, hydroxy, carboxy, carbonyl, aldehyde group, cyano, amino, aryl, heteroaryl, C3-12 cycloalkyl, C2-6 alkenyl, C3-12 cycloalkenyl; n is an integer selected from 0 to 6, such as 0, 1, 2, 3, 4, 5 or 6; preferably, n is 1 or 2.


In some embodiments, the present invention provides the above-described Compound represented by Formula I0 or a pharmaceutically acceptable salt thereof, wherein when R1 in Formula I is H, the compound is represented by Formula II,




embedded image


wherein n is an integer selected from 0 to 6, such as 0, 1, 2, 3, 4, 5 or 6.


In some embodiments, the present invention provides the above-described Compound represented by Formula I0 or a pharmaceutically acceptable salt thereof, wherein when R1 in Formula I is R0-substituted carbonyl, the compound is represented by Formula III,




embedded image


wherein, R0 is selected from the group consisting of H, C1-6 alkyl, substituted C1-6 alkyl and substituted C1-6 alkoxy, wherein the substituted C1-6 alkyl or the substituted C1-6 alkoxy is substituted by halogen, hydroxy, carboxy, carbonyl, aldehyde group, cyano, amino, aryl, heteroaryl, C3-12 cycloalkyl, C2-6 alkenyl, C3-12 cycloalkenyl, n is an integer selected from 0 to 6, such as 0, 1, 2, 3, 4, 5 or 6.


Further, R0 is selected from the group consisting of C1-6 alkyl, substituted C1-6 alkyl and substituted C1-6 alkoxy, wherein the substituted C1-6 alkyl or the substituted C1-6 alkoxy is substituted by halogen, hydroxy, carboxy, carbonyl, aldehyde group, cyano, amino, aryl, heteroaryl, C3-12 cycloalkyl, C2-6 alkenyl, C3-12 cycloalkenyl.


In some embodiments, the present invention provides the above-described Compound represented by Formula I0, or a pharmaceutically acceptable salt thereof, wherein when R1 in the Formula I0 is H, the compound is represented by Formula IV:




embedded image


wherein, R2 is selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, R5C(O)—, R5S(O)m—; R5 is selected from the group consisting of H, C1-10 alkyl, C3-12 cycloalkyl, substituted C1-10 alkyl and substituted C3-12 cycloalkyl, wherein the substituted C1-10 alkyl and the substituted C3-12 cycloalkyl is substituted by hydroxy, cyano, CF3, C1-6 alkyl, C3-10 cycloalkyl, alkoxy, aryl or heteroaryl; m is selected from 1 or 2;


R3 and R4 are each independently selected from the group consisting of H, C1-10 alkyl, C3-12 cycloalkyl, substituted C1-10 alkyl, substituted C3-12 cycloalkyl, substituted C1-10 alkoxy, substituted sulfonyl and substituted C3-14 heteroaryl, wherein the substituted C1-10 alkyl, the substituted C3-12 cycloalkyl, the substituted C1-10 alkoxy, the substituted sulfonyl or the substituted C3-14 heteroaryl is substituted by hydroxy, cyano, halogen, C1-6 alkyl, C3-10 cycloalkyl, alkoxy, aryl or heteroaryl;


wherein, n is an integer selected from 0 to 6, such as 0, 1, 2, 3, 4, 5 or 6;


further, R2 is selected from the group consisting of H, R5C(O)—, R5S(O)m—; R5 is selected from the group consisting of H, C1-10 alkyl, C3-12 cycloalkyl, substituted C1-10 alkyl, and substituted C3-12 cycloalkyl, wherein the substituted C1-10 alkyl or the substituted C3-12 cycloalkyl is substituted by hydroxy, cyano, CF3, C1-6 alkyl, C3-10 cycloalkyl, C1-6 alkoxy, aryl or heteroaryl; m is selected from 1 or 2;


further, R2 is selected from the group consisting of R5C(O)— and R5S(O)x—; R5 is selected from the group consisting of C1-6 alkyl, C3-8 cycloalkyl, substituted C1-6 alkyl and substituted C3-8 cycloalkyl, wherein the substituted C1-6 alkyl or the substituted C3-8 cycloalkyl is substituted by hydroxy, cyano, C1-6 alkyl, C3-8 cycloalkyl, C1-6 alkoxy, aryl or heteroaryl; x is 2; R3, R4 are both H.


In some embodiments, the invention provides a compound as follows, or a pharmaceutically acceptable salt thereof:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


From the general formula or the synthesis method of general formula of the present invention, compounds not limited to these specific compounds can be derived, and under the guidance of the general formula or the synthesis method of general formula of the present invention, specific compounds that can be obtained by those skilled in the art without the need of creative labor are all within the scope of the invention.


In some embodiments, the present invention provides a method for synthesizing the above-described compound represented by Formula I0,




embedded image


i.e., General Synthesis Method I, steps of which are as follows:


1) Oxidation of compound 1a to obtain compound 2a:




embedded image


an oxidizing agent used above includes, but is not limited to, at least one of hydrogen peroxide, ozone or peracetic acid;


2) Reaction of a compound represented by formula 1 with a compound represented by formula 2a under alkaline condition to obtain a compound represented by formula 2:




embedded image


an alkali used above includes, but is not limited to, alkali metal hydroxides, preferably sodium hydroxide, potassium hydroxide, barium hydroxide;


3) Reaction of the compound represented by formula 2 with a compound represented by formula 3a to obtain the compound represented by formula I0:




embedded image


wherein, R1, R2 are each independently selected from the group consisting of: H, substituted or unsubstituted C1-10 alkyl, aldehyde group, substituted or unsubstituted carbonyl, cyano, CF3, substituted or unsubstituted C1-10 alkoxy, substituted or unsubstituted sulfonyl, substituted or unsubstituted C3-10 cycloalkyl, substituted or unsubstituted C2-10 alkenyl, substituted or unsubstituted C6-20 aryl, substituted and unsubstituted C3-14 heteroaryl;


R3, R4 are each independently a mono-substituent selected from the group consisting of: H, substituted or unsubstituted C1-10 alkyl, substituted or unsubstituted C3-10 cycloalkyl, cyano, substituted or unsubstituted C1-10 alkoxy, substituted or unsubstituted sulfonyl, substituted or unsubstituted C6-20 aryl, and substituted or unsubstituted C3-14 heteroaryl;


or R3, R4 are each independently selected from di-substituents, thereby forming the following groups together with the C atom at a- or b-position: C═O, C═NH or




embedded image


wherein C represents the C atom at a- or b-position, m is an integer selected from 0 to 6;


n is an integer selected from 0 to 6.


In some embodiments, the present invention provides a method for synthesizing the above-described compound represented by Formula I,




embedded image


i.e., General Synthesis Method IA, steps of which are as follows:

    • 1) Oxidation of compound 1a to obtain compound 2a;




embedded image




    • 2) Reaction of a compound represented by formula 1 with compound 2a under alkaline condition to obtain a compound represented by formula 2;







embedded image




    • 3) Reaction of the compound represented by formula 2 with a compound represented by formula 3a to obtain the compound represented by formula I,







embedded image


wherein R1 is selected from the group consisting of: H, amino, sulfonyl, nitro, carbonyl, amidino, C1-6 alkyl, substituted amidino, substituted C1-6 alkyl, substituted C1-6 alkoxy, substituted carbonyl, substituted sulfonyl, and substituted sulfinyl, wherein the substituted amidino or the substituted C1-6 alkyl, the substituted C1-6 alkoxy, the substituted carbonyl, the substituted sulfonyl or the substituted sulfinyl is substituted by halogen, hydroxy, carboxy, carbonyl, aldehyde group, cyano, amino, aryl, heteroaryl, C3-12 cycloalkyl, C2-6 alkenyl, C3-12 cycloalkenyl; n is an integer selected from 0 to 6;


an oxidizing agent used above is preferably, but not limited to, at least one of hydrogen peroxide, ozone or peroxyacetic acid;


the alkali is preferably selected from, but not limited to, alkali metal hydroxides, preferably sodium hydroxide, potassium hydroxide or barium hydroxide.


In some embodiments, the present invention provides a method for synthesizing the above compound represented by Formula II,




embedded image


i.e., General Synthesis Method II, steps of which are as follows:


1) Reaction of a compound represented by formula 2 with compound 3a′ to obtain a compound represented by formula IIa:




embedded image


2) Deprotection of the compound represented by formula IIa under acidic condition, followed by reaction under alkaline condition to obtain the compound represented by formula II,




embedded image


wherein n is 0, 1, 2, 3 or 4,


the alkali is selected from, but not limited to, hydroxides of alkali metal or alkaline earth metal, preferably sodium hydroxide, potassium hydroxide or barium hydroxide.


In some embodiments, the present invention provides a method for synthesizing the above-described compound represented by Formula II′,




embedded image


i.e., General Synthesis Method III, steps of which are as follows:




embedded image


n is 0, 1, 2, 3 or 4,


the compound represented by Formula IIa is reacted under acidic condition to give the compound represented by Formula II′.


In some embodiments, the present invention provides a method for synthesizing the above compound represented by Formula III,




embedded image


i.e., General Synthesis Method IV, steps of which are as follows:


1) Reaction of a compound represented by formula II with a compound represented by formula 4a to give a compound represented by formula IIIa,




embedded image


wherein R3 is selected from the group consisting of H, OH, CN, CH3-mXm, nitro, C1-9 alkyl, C1-9 alkoxy, C3-9 cycloalkoxy, C3-12 cycloalkyl, C1-6 heteroalkyl, 3- to 12-membered heterocycloalkyl, aryl, heteroaryl, substituted C1-6 alkyl, substituted C1-9 alkoxy, substituted aryl, substituted heteroaryl and substituted carbonyl, wherein the substituted C1-6 alkyl, the substituted C1-9 alkoxy, the substituted aryl, the substituted heteroaryl or the substituted carbonyl is substituted by halogen, hydroxy, carboxy, carbonyl, aldehyde group, cyano, amino, sulfonyl, aryl, heteroaryl, C3-12 cycloalkyl, C3-12 cycloalkenyl, m is 1, 2 or 3; preferably, m is 2 or 3;


2) Reaction of the compound represented by formula IIIa under alkaline condition to give the compound represented by formula III,




embedded image


Specifically, R3 is selected from the group consisting of H, OH, CN, CH3-mXm, nitro, C1-9 alkyl, C3-9 cycloalkoxy, C3-12 cycloalkyl, C1-6 heteroalkyl, 3- to 12-membered heterocycloalkyl, aryl, heteroaryl, substituted C1-6 alkyl, substituted C1-9 alkoxy, substituted aryl and substituted heteroaryl, wherein the substituted C1-6 alkyl, the substituted C1-9 alkoxy, the substituted aryl or the substituted heteroaryl is substituted by halogen, hydroxy, carboxy, carbonyl, aldehyde group, cyano, amino, sulfonyl, aryl, heteroaryl, C3-12 cycloalkyl, C3-12 cycloalkenyl. Further, R3 is selected from the group consisting of H, OH, CN, CF3, CHCl2, CH2Cl, nitro, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, 1,1-dimethylpropyl, 2,2-dimethylpropyl, 1,2-dimethylpropyl, 1-ethylpropyl, hexyl, pentylmethyl, pentylethyl, pentylpropyl, pentylbutyl, hexylmethyl, hexylethyl, hexylpropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopropylmethyl, cyclopropylethyl, cyclopropylpropyl, cyclobutylmethyl, cyclobutylethyl, cyclobutylpropyl, cyclopentylmethyl, cyclopentylethyl, cyclopentylpropyl, cyclohexylmethyl, cyclohexylethyl, cyclohexylpropyl, p-methoxybenzyl (PMB), benzyl (Bn); or


R3 is any one selected from the group consisting of substituted furan, pyrrole, thiophene, pyrazole, imidazole, oxazole, thiophene, isoxazole, isothiazole, pyridine, pyran, thiopyran, pyridazine, pyridine, pyrazine, and piperazine, or is a di-substituent at benzene ring, i.e., forms a benzofuran, benzopyrrole, benzopiperazine together with benzene ring; or


the group




embedded image


in the Formula 4a is selected from the group consisting of carbazole, acridine, phenazine or phenothiazine.


The synthesis method provided by the present invention is only one way to realize each synthesized target compound and its intermediate, wherein each step and number, such as 1a, 2a, 3a, 4a, 1, 2, 3, etc., are independent, and the preparation thereof is not limited to the method of the present invention.


Unless otherwise specified, the solvent used in each step of the above or below reactions of the present invention is a conventional solvent in the art, and the selection principle is that the solvent is able to dissolve the reactants but not participate in the reaction, extract the product or allow the corresponding product to crystallize therein so as to be separated from the impurity. Examples of the solvent include water, halogenated alkanes, alkylamines, aliphatic hydrocarbons, esters, alcohols, aromatic hydrocarbons, ethers, heterocyclic solvents. Specifically, the solvent is selected from, but not limited to, the following solvents: methanol, ethanol, propanol, isopropyl, diethyl ether, ethyl acetate, acetic acid, cyclohexane, dichloromethane, chloroform, tetrahydrofuran, pyridine, diethyl amine, triethylamine, dimethylformamide, toluene, and mixtures of at least two of them.


Unless otherwise specified, in each of the above or below reactions of the present invention, when a reactant is in an excessive amount, the termination of the reaction may be carried out by adding a substance which can react with the excessive reactant.


Unless otherwise specified, in each of the above or below reactions of the present invention, the purification method of the product in each step of the reactions is selected from the group consisting of extraction, crystallization, solvent removal, column chromatography, the operations thereof are all conventional techniques in the art, and a skilled in the art can handle them according to specific conditions.


The numbers used in the general formula of the present invention are used for conveniently describing the general formula, and they may be modified into other numbers in specific embodiments, such as 1, 2, 3, etc., for convenience of description, and are the expressions of general formula and general reaction equations that do not affect the essence of the structural formula and its reaction equations.


For the compounds encompassed by the Formulas I to III and specific representative substances thereof, their chiral or cis-trans isomers and mixtures of these isomers in any ratio also fall within the scope of the compounds encompassed by the Formulas I to III and specific representative substances thereof.


In some embodiments, the present invention provides a pharmaceutical composition comprising the above compounds, i.e., the compounds encompassed by the Formulas I to III and the above specific compounds, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable pharmaceutic adjuvants.


The term “pharmaceutically acceptable salt” as used in the present invention refers to an addition salt formed with a pharmaceutically acceptable acid or alkali, or a solvate thereof. Such pharmaceutically acceptable salts include salts of acids, wherein the acids include hydrochloric acid, sulfuric acid, nitric acid, phosphoric acid, hydrobromic acid, hydroiodic acid, formic acid, acetic acid, p-toluenesulfonic acid, sulfinic acid, methanesulfonic acid, benzoic acid, fumaric acid, citric acid, tartaric acid, maleic acid, fatty acid. Addition salts of non-toxic pharmaceutically acceptable alkali include salts of alkali, and these alkali include sodium, potassium, calcium, magnesium, aluminum, ammonium.


In some embodiments, the present invention provides the above-described compound or a pharmaceutically acceptable salt thereof, which is capable of inhibiting the activity of indoleamine 2,3-dioxygenase, and can be used in the manufacture of a medicament for treating a disease pathologically characterized by indoleamine 2,3-dioxygenase-mediated tryptophan metabolic pathway; the medicament is used for treating a cancer, an infectious disease, a neurodegenerative disease, a depression, an anxiety or an age-related cataract;


wherein the cancer is selected from the group consisting of lung cancer, liver cancer, colon cancer, pancreatic cancer, breast cancer, prostate cancer, brain cancer, ovarian cancer, cervical cancer, testicular cancer, renal cancer, head and neck cancer, lymphoma, melanoma or leukemia;


the neurodegenerative disease refers to Alzheimer's disease;


the infectious disease refers to an infection caused by a bacterium, a fungus, a virus or a parasite.


In some embodiments, the present invention provides a method for inhibiting the activity of indoleamine 2,3-dioxygenase by using the above-described compound or a pharmaceutically acceptable salt thereof, or for treating a disease pathologically characterized by indoleamine 2,3-dioxygenase-mediated tryptophan metabolic pathway, the method comprising administering a therapeutically effective amount of the compound or a pharmaceutically acceptable salt thereof. The disease is selected from the group consisting of cancer, infectious disease, neurodegenerative disease, depression, anxiety, or age-related cataract; wherein the cancer is selected from the group consisting of lung cancer, liver cancer, colon cancer, pancreatic cancer, breast cancer, prostate cancer, brain cancer, ovarian cancer, cervical cancer, testicular cancer, kidney cancer, head and neck cancer, lymphoma, melanoma or leukemia; the neurodegenerative disease refers to Alzheimer's disease; the infectious disease refers to an infection caused by a bacterium, a fungus, a virus or a parasite.


The activity test results according to the examples of the present invention show that the compounds obtained by the present invention have excellent activity for inhibiting indoleamine 2,3-dioxygenase, and the activity is significantly superior to the compound INCB024360. In vivo test results show that the compounds of the present invention have a high inhibition rate on tumors, and the therapeutic effect on tumors is significantly better than that of the compound INCB024360 and other IDO inhibitors. Moreover, by measuring the body weights of the mice before and after administration, it is found that as compared to other IDO inhibitors, the indoleamine 2,3-dioxygenase inhibitors of the present invention can significantly reduce the side effects during the period of tumor treatment, significantly improve the life quality of the mice, and in clinical practice, improve not only patient's life quality, but also significantly improve patient compliance with medications and medicament effectiveness.


The compound of the invention can significantly improve the learning and memory impairment, enhance the learning acquisition ability and the spatial memory ability in animals, and has positive therapeutic significance for neurodegenerative diseases such as Alzheimer's syndrome, and the effect thereof is superior to other IDO inhibitors.


Through the T cell proliferation reaction experiment, it is found that the compound of the present invention can promote the function of DC in stimulation of T cell proliferation, so that it can be used for treating tumor diseases, autoimmune diseases, transplant rejection, and infectious diseases, and it is obviously superior to other IDO inhibitors.


When the indoleamine 2,3-dioxygenase inhibitor of the present invention is used in the manufacture of a medicament for treating a disease pathologically characterized by indoleamine 2,3-dioxygenase-mediated tryptophan metabolic pathway, it shows the following technical advantages:


(1) The antitumor effect is remarkable. The compound of the present invention has activity for significantly inhibiting indoleamine 2,3-dioxygenase, and the in vivo test shows that the tumor inhibition rate of the compound of the present invention is significantly higher than those of the positive control drug cyclophosphamide and the compound INCB024360.


(2) The side effect is lowered. The compound of the present invention is an indoleamine 2,3-dioxygenase inhibitor, which reverses the inhibiting of T cells proliferation and regulates the immune function of body by inhibiting the activity of indoleamine 2,3-dioxygenase, thereby completing the monitoring and killing effects of the human immune system on tumor cells. Based on this special mechanism of action, this compound does not adversely affect the growth of normal cells of the human body while inhibiting the growth of tumor cells, thus significantly reducing the side effects. Moreover, it has a significant therapeutic effect on autoimmune diseases, transplant rejection, and infectious diseases associated with T cell proliferation.


(3) The treatment of Alzheimer's and other neurodegenerative diseases is significantly effective, the learning and memory impairment of animals can significantly be improved, and the learning acquisition ability and the spatial memory ability can significantly be enhanced.


Specific Models for Carrying Out the Invention:

The invention is further described below in conjunction with specific embodiments, but the invention is not limited thereto. In addition, under the guidance of the general formula or the synthesis method of general formula (General Synthesis Method I, General Synthesis Method IA, General Synthesis Method II, General Synthesis Method III, General Synthesis Method IV) and specific embodiments of the present invention, the specific compounds obtained by those skilled in the art without the need of creative labor are all within the scope of the invention.


Example 1



embedded image


Compound 1 (903 mg, 10 mmol) was dissolved in acetone (10 mL), then potassium carbonate (2.76 g, 20 mmol) was added at room temperature, stirred at room temperature for 0.5 h, added dropwise with benzenesulfonyl chloride, stirred at room temperature overnight, quenched with water, and the resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 900 mg of a white powder.




embedded image


Compound 2 (230 mg, 1 mmol) was dissolved in acetonitrile (5 mL), stirred at room temperature for 0.5 h, then added with compound 2a (320 mg, 2 mmol), heated at 60° C. for 24 h, and the resultant was directly evaporated to dryness under reduced pressure, and subjected to purification by pre-HPLC to obtain 210 mg of the desired product.




embedded image


Compound 3 (171 mg, 0.5 mmol) was dissolved in dichloromethane (10 mL), stirred at room temperature for 5 min, added with trifluoroacetic acid (5 mL), reacted at room temperature for 2 h, and then the resultant was directly evaporated to dryness under reduced pressure to give the desired product 4 which was directly used as a crude product in the next step.




embedded image


Compound 4 (120 mg, 0.5 mmol) was dissolved in tetrahydrofuran, the compound 4a (120 mg, 0.5 mmol) was added, the mixture was stirred at room temperature for 10 min, 1N sodium hydroxide (1 mL) was added dropwise, and the mixture was stirred at room temperature for 10 min. The reaction solution was concentrated and subjected to purification by pre-HPLC to give 5 mg of the desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.45 (s, 1H), 8.89 (s, 1H), 7.76-7.77 (m, 2H), 7.48-7.52 (m, 3H), 7.10-7.20 (m, 3H), 6.75-6.77 (m, 2H), 6.27 (s, 1H), 3.73-3.77 (m, 2H), 2.52-2.69 (m, 2H).


HPLC purity: @214 nm 99.5%, @254 nm 100%.


LC-MS: m/z 541 [M+1].


Referring to the General Synthesis Method I and Example 1, the following compounds were synthesized.















Example No.
Substituent
Substance No.
MS


















Example 2


embedded image


XSD3-059
574.0





Example 3


embedded image


XSD3-060
558.0





Example 4


embedded image


XSD3-061
546.1





Example 5


embedded image


XSD3-062
478.0





Example 6


embedded image


3050
456.1





Example 7


embedded image


3051
600.2





Example 8
R1 is ethylcarbonyl, n is 1, R2, R3, R4 is H.
XSD3-052
456.1





Example 9


embedded image


XSD3-056
442.1









Example 10



embedded image


Compound 1a (682 mg, 2 mmol) was dissolved in trifluoroacetic acid (13 mL), then 30% H2O2 was added dropwise at room temperature, the mixture was reacted overnight at 50° C., and the reaction solution changed from turbid to clear yellow. After the reaction was completed, the reaction was quenched with a saturated sodium sulfite solution. After KI starch test paper showed clolorless, the resultant was extracted with ethyl acetate (50 mL*2), and the organic phase was dried over anhydrous sodium sulfate, concentrated, and then subjected to purification by column chromatography (petroleum ether:ethyl acetate=1:1) to obtain a pale yellow solid 2a (500 mg, yield 67%).




embedded image


Ethylenediamine 1 (30 mg, 0.5 mmol) was added to a solution of Compound 2a (170 mg, 0.5 mmol) in tetrahydrofuran (10 mL), then 1N NaOH (0.4 mL) was added, the reaction solution was stirred at room temperature for 0.5 h, and the reaction solution was directly used to prepare and obtain Compound 2 (120 mg).




embedded image


A mixture solution of Compound 2 (120 mg, 0.33 mmol) and Compound 3a (100 mg, 0.33 mmol) in methanol (10 mL) was stirred at room temperature overnight, and the reaction solution was concentrated and subjected to purification by column chromatography (petroleum ether:ethyl acetate=1:1) to obtain Compound 3 (42 mg, 23%).




embedded image


Trifluoroacetic acid (0.6 mL) was added to a solution of Compound 3 (31 mg, 0.05 mmol) in dichloromethane (3 mL), and stirred at room temperature overnight. The reaction solution was concentrated and subjected to purification by pre-HPLC to obtain compound XSD3-047 (9 mg, yield 68%). For the desired compound: 1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.4 (s, 1H), 8.88 (s, 1H), 7.22-7.11 (m, 2H), 6.71-6.5 (m, 3H), 6.38-6.21 (m, 2H), 3.37 (m, 3H), 3.01 (m, 2H). MS: m/z 401.2 [M+1].


Example 11



embedded image


1,3-Diaminopropane (35 mg, 0.5 mmol) was added to a solution of Compound 1 (170 mg, 0.5 mmol, the synthesis method of which was referred to the synthesis of Compound 2a in XSD3-047 Final report) in tetrahydrofuran (10 mL), the reaction solution was stirred at room temperature for 0.5 h, and the reaction solution was directly used to prepare and obtain the desired product of 130 mg.




embedded image


A mixture solution of Compound 2 (130 mg, 0.33 mmol) and Compound 1a (100 mg, 0.33 mmol) in methanol (10 mL) was stirred at room temperature overnight, the reaction solution was concentrated and subjected to purification by column chromatography (petroleum ether:ethyl acetate=1:1) to obtain Compound 3 (78 mg, 45%).




embedded image


Trifluoroacetic acid (0.6 mL) was added to a solution of Compound 3 (31 mg, 0.05 mmol) in dichloromethane (3 mL), stirred at room temperature overnight, then adjusted with 1N NaOH solution to pH=12, stirred continuously for 20 min, and the reaction was monitored by LCMS. After the reaction was completed, the reaction solution was adjusted to neutral with 0.5 N HCl, and the reaction solution was concentrated and subjected to purification by pre-HPLC to obtain the desired product (9 mg, yield 68%). For the desired product: 1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.4 (s, 0.6 H), 8.88 (s, 1H), 7.6 (s, 1H), 7.4-6.7 (m, 6H), 6.7 (s, 1H), 6.24 (s, 1H), 3.19-3.14 (m, 4H), 1.75 (m, 2H). MS: m/z 415.2 [M+1].


Example 12



embedded image


A mixture solution of Compound 1 (384 mg, 1 mmol, the preparation method of which was referred to that in the Example of 3047) and Compound 1a (100 mg, 1.1 mmol) in tetrahydrofuran (10 mL) were stirred at room temperature for 48 h, and the reaction solution was directly used to prepare and obtain the desired product 2 (89 mg, yield 20%).




embedded image


Compound 2 (89 mg, 0.2 mmol) was dissolved in THF (10 mL), then adjusted with 1N NaOH solution to pH=12, and stirred continuously for 20 min. The reaction was monitored by LCMS. After the reaction was completed, 0.5N HCl was used to adjust the reaction solution to neutral, and the reaction solution was concentrated and subjected to purification by pre-HPLC to obtain the desired product (13 mg, yield 15%). For the desired compound: 1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.47 (s, 0.6 H), 8.92 (s, 1H), 7.50-7.09 (m, 5H), 6.70 (s, 1H), 6.28 (s, 1H), 2.72-2.50 (m, 4H), 2.50 (m, 3H). MS: m/z 416.2 [M+1].


Example 13



embedded image


Compound 1 (102 mg, 0.016 mmol, the synthesis method of which was referred to XSD3-047 Final Report) was dissolved in THF (10 mL), then 1N NaOH solution (0.1 mL) was added at room temperature, stirred at room temperature for 0.2 h, and the reaction was monitored by LCMS. After the reaction was completed, the pH was adjusted to neutral with 0.5 N HCl solution, to obtain 52 mg of desired compound. For the desired compound: 1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.45 (s, 1H), 8.89 (s, 1H), 8.24 (m, 1H), 7.76-7.77 (m, 2H), 7.14-7.20 (m, 2H), 7.18 (s, 1H), 6.31 (s, 1H), 3.33-3.67 (m, 4H), 1.44-1.48 (m, 18H). MS: m/z 601.2 [M+1].


Example 14



embedded image


Compound 1 (52 mg, 0.1 mmol, the synthesis method of which was referred to XSD3-048 Final Report) was dissolved in THF (10 mL), then TFA (0.5 mL) was added at room temperature, stirred at room temperature for 12 h, and the reaction was monitored by LCMS. After purification by pre-HPLC, the desired compound of 15 mg was obtained. For the desired compound: 1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.44 (s, 1H), 10.85 (s, 1H), 8.91 (s, 1H), 8.49 (s, 1H), 7.18-7.09 (m, 3H), 6.78-6.75 (m, 1H), 6.28-6.25 (m, 1H), 3.33-3.19 (m, 4H), 1.85-1.78 (m, 2H), 1.5 (s, 3H). LC-MS characterization result: m/z 515 [M+1].


Example 15



embedded image


Compound 1 (100 mg, 0.23 mmol) was dissolved in acetone (10 mL), then added with potassium carbonate (0.276 g, 2.0 mmol), stirred at room temperature for 0.5 h, added dropwise with benzenesulfonyl chloride, stirred at room temperature overnight. The reaction was added with water for quenching, and the resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain a crude product which was directly used in the next step.




embedded image


Compound 2 (120 mg, 0.5 mmol) was dissolved in tetrahydrofuran/water, added dropwise with 1N sodium hydroxide (1 mL), and stirred at room temperature for 10 min. The reaction solution was concentrated and subjected to purification by pre-HPLC to obtain 20 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.45 (s, 1H), 8.90 (s, 1H), 7.75-7.77 (m, 2H), 7.46-7.52 (m, 3H), 7.12-7.20 (m, 1H), 7.10-7.11 (m, 1H), 6.75-6.78 (m, 2H), 6.19 (s, 1H), 4.25 (m, 2H), 3.11-3.19 (m, 4H), 1.67-1.70 (m, 2H).


HPLC purity: @214 nm 99.2%, @254 nm 99.3%.


LC-MS: m/z 555 [M+1].


Example 16



embedded image


Compound 1 (106 mg, 0.25 mmol) was dissolved in acetone (10 mL), added with potassium carbonate (138 mg, 1 mmol), added dropwise with Compound 1a (49 mg, 0.25 mmol), stirred at room temperature overnight, quenched with water, and the resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to give 100 mg of a crude product.




embedded image


Compound 2 (100 mg) was dissolved in methanol, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min. The reaction solution was concentrated and subjected to purification by pre-HPLC to obtain 12 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.45 (s, 1H), 8.90 (s, 1H), 7.10-7.21 (m, 2H), 6.76-7.17 (m, 4H), 6.28 (m, 1H), 3.33-3.40 (m, 4H), 2.76 (m, 2H), 0.98-1.86 (m, 11H).


HPLC purity: @214 nm 93.7%, @254 nm 97.6%.


LC-MS: m/z 563 [M+1].


Example 17



embedded image


Compound 1 (110 mg, 0.25 mmol) was dissolved in acetone (10 mL), added with potassium carbonate (138 mg, 1 mmol), added dropwise with Compound 1a (47.5 mg, 0.25 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, concentrated to obtain 100 mg of a crude product.




embedded image


Compound 2 (100 mg) was dissolved in methanol, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min, and the reaction solution was concentrated and subjected to purification by pre-HPLC to prepare 15 mg of desired product.



1H-NMR (400 MHz, CDCl3): δ (ppm): 9.96 (s, 1H), 7.77-7.79 (m, 2H), 6.18-7.32 (m, 9H), 3.31-3.42 (m, 4H), 2.40 (s, 3H), 1.76 (m, 2H).


HPLC purity: @214 nm 98.6%, @254 nm 98.9%.


LC-MS: m/z 571 [M+1].


Example 18



embedded image


Compound 1 (96 mg, 10 mmol) and Compound 1a (48 mg, 0.25 mmol) were dissolved in acetonitrile (10 mL), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate and concentrated to give 100 mg of a crude product.




embedded image


Compound 2 (100 mg) was dissolved in methanol, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min. The reaction solution was concentrated and subjected to purification by pre-HPLC to prepare 15 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.76 (s, 1H), 11.45 (s, 1H), 8.68-9.04 (m, 3H), 7.09-7.20 (m, 2H), 6.73-6.77 (m, 1H), 6.33-6.36 (m, 1H), 3.53-3.54 (m, 2H), 3.44-3.46 (m, 2H), 1.18 (m, 1H), 1.00-1.02 (m, 2H), 0.90-0.92 (m, 2H).


HPLC purity: @214 nm 98.8%, @254 nm 99.8%.


LC-MS: m/z 471 [M+1].


Example 19



embedded image


Compound 1 (96 mg, 0.25 mmol) and Compound 1a (48 mg, 0.25 mmol) were dissolved in acetonitrile (10 mL), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 100 mg of a crude product.




embedded image


Compound 2 (100 mg) was dissolved in methanol, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min. The reaction solution was concentrated and subjected to purification by pre-HPLC to prepare 12 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.48 (s, 1H), 11.28 (s, 1H), 8.75-8.92 (m, 3H), 7.09-7.20 (m, 2H), 6.73-6.77 (m, 1H), 6.35-6.38 (m, 1H), 3.53-3.54 (m, 2H), 3.44-3.46 (m, 2H), 3.24-3.28 (m, 2H), 2.13-2.20 (m, 3H), 1.77-1.98 (m, 2H).


HPLC purity: @214 nm 97.9%, @254 nm 98.6%.


LC-MS: m/z 485 [M+1].


Example 20



embedded image


Compound 1 (500 mg, 5.5 mmol) was dissolved in 3 mL of dichloromethane, and O-(7-azabenzotriazole)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (128 mg, 0.336 mmol), cyclohexanecarboxylic acid (800 mg, 6.25 mmol) and diisopropylethylamine (1.16 g, 896 mmol) were added in sequence, the reaction was carried out at room temperature overnight under the protection of nitrogen gas, quenched with water, the resultant was extracted with ethyl acetate (50 mL×5), the extract liquors were combined and washed with a saturated aqueous solution of sodium chloride and dried over anhydrous sodium sulfate, evaporated to dryness under reduced pressure to obtain 40 mg of product, which was directly used in the next step.




embedded image


Compound 2 (530 mg, 2.65 mmol) was dissolved in acetonitrile (5 mL), added with compound 2a (850 mg, 5.3 mmol), stirred at room temperature for 24 h, and directly evaporated to dryness under reduced pressure to give 300 mg of product that was directly used in the next step.




embedded image


Compound 3 (300 mg, 0.99 mmol) was dissolved in dichloromethane (10 mL), stirred at room temperature for 5 min, added with trifluoroacetic acid (5 mL), reacted at room temperature for 2 h, and the resultant was directly evaporated to dryness under reduced pressure to give 200 mg of the desired product 4, which was a crude product and directly used in the next step.




embedded image


Compound 4 (200 mg, 0.99 mmol) was dissolved in tetrahydrofuran, added with Compound 4a (250 mg, 1.0 mmol), stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min. The reaction solution was concentrated and subjected to purification by pre-HPLC to prepare 3 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.90 (s, 1H), 11.55 (s, 1H), 8.67-9.12 (m, 3H), 7.10-7.20 (m, 2H), 6.73-6.77 (m, 1H), 6.37 (s, 1H), 3.33-3.54 (m, 4H), 2.16-2.21 (m, 1H), 1.56-1.81 (m, 6H), 1.15-1.36 (m, 1H).


HPLC purity: @214 nm 97.7%, @254 nm 98.0%.


LC-MS: m/z511 [M+1].


Example 21



embedded image


Compound 1 (35 mg, 0.2 mmol) and Compound 2 (90 mg, 0.22 mmol) were dissolved in 10 mL of acetonitrile and stirred at 90° C. for 16 h. The reaction solution was directly evaporated to dryness under reduced pressure to give Compound 3, which was a crude product and directly used in the next step (100 mg).




embedded image


Compound 3 (100 mg, 0.1 mmol) was dissolved in a solution of 1.5 N NaOH (2 mL) in THF (5 mL), stirred at room temperature for 0.5 hour, and the resultant was subjected to purification by pre-HPLC to prepare the desired compound (30 mg, yield: 30%).


HNMR (DMSO, 400M): 11.40 (s, 1H), 11.33 (s, 1H), 9.09 (s, 1H), 8.93 (s, 1H), 8.7 (s, 1H), 7.09-7.20 (m, 2H), 6.73-6.77 (m, 1H), 6.36 (s, 1H) 3.45-3.55 (m, 4H), 2.31-2.33 (m, 2H), 0.95-0.99 (m, 1H), 0.47-0.51 (m, 2H), 0.16-0.20 (m, 2H).


HPLC purity: @214 nm 98.8%, @254 nm 98.9%.


LC-MS: m/z 483.1 [M+1].


Example 22



embedded image


Compound 1 (44 mg, 0.2 mmol) and Compound 2 (90 mg, 0.22 mmol) were dissolved in 10 mL of acetonitrile and stirred at 90° C. for 16 h. The reaction solution was directly evaporated to dryness under reduced pressure to give Compound 3, which was a crude product and used directly in the next step (100 mg).




embedded image


Compound 3 (100 mg, 0.1 mmol) was dissolved in a solution of 1N NaOH (4 mL) in tetrahydrofuran (5 mL), stirred at room temperature for 1 hour, and subjected to purification by pre-HPLC to prepare the desired compound (35 mg, yield: 35%).


HNMR (DMSO, 400M): 11.40 (s, 1H), 11.33 (s, 1H), 9.18 (s, 1H), 9.02 (s, 1H), 8.78 (s, 1H, 7.10-7.17 (m, 2H), 6.73-6.76 (m, 1H), 6.35-6.38 (m, 1H) 3.46-3.65 (m, 4H), 2.26-2.38 (m, 2H), 1.47-1.69 (m, 5H), 0.80-1.22 (m, 6H),


HPLC purity: @214 nm 96.5%, @254 nm 96.5%.


LC-MS: m/z527.2 [M+1].


Example 23



embedded image


Compound 1 (35 mg, 0.26 mmol) and Compound 2 (90 mg, 0.22 mmol) were dissolved in 10 mL of acetonitrile and stirred at 90° C. for 20 h. The reaction solution was directly evaporated to dryness under reduced pressure to give Compound 4, which was a crude product and used directly in the next step (110 mg).




embedded image


Compound 3 (100 mg, 0.2 mmol) was dissolved in a solution of 1N NaOH (1.5 mL) in tetrahydrofuran (5 mL), stirred at room temperature for 0.5 hour, and the resultant was subjected to purification by pre-HPLC to prepare the desired compound (15 mg, yield: 15%).


HNMR (DMSO, 400M): 12.50 (s, 1H), 11.50 (br, 1H), 9.19 (s, 1H), 8.65-8.91 (m, 2H) 7.12-7.21 (m, 2H), 6.73-6.77 (m, 1H), 6.36 (s, 1H) 3.24-3.36 (m, 4H), 1.77-1.84 (m, 3H), 0.90-1.02 (m, 4H).


HPLC purity: @214 nm 98.6%, @254 nm 98.3%.


LC-MS: m/z481.1 [M+1].


Example 24



embedded image


Compound 1 (42 mg, 0.2 mmol) and Compound 2 (90 mg, 0.22 mmol) were dissolved in 10 mL of acetonitrile and stirred at 90° C. for 20 h. The reaction solution was directly evaporated to dryness under reduced pressure to give Compound 4, which was a crude product and used directly in the next step (120 mg).




embedded image


Compound 3 (110 mg, 0.2 mmol) was dissolved in a solution of 1N NaOH (5 mL) in tetrahydrofuran (5 mL), stirred at room temperature for 1 hour, and the resultant was subjected to purification by pre-HPLC to prepare the desired compound (35 mg, yield: 35%).


HNMR (DMSO, 400M): 11.44 (s, 1H), 11.35 (s, 1H), 9.06 (s, 1H), 8.92 (s, 1H), 8.64 (s, 3H), 7.16-7.21 (t, 1H), 7.09-7.11 (q, 1H), 6.74-6.78 (m, 1H), 6.26-6.29 (m, 1H), 3.23-3.34 (m, 4H), 2.49-2.50 (m, 1H), 1.61-1.81 (m, 7H), 1.17-1.32 (m, 5H).


HPLC purity: @214 nm 99.4%, @254 nm 99.8%.


LC-MS: m/z527.2 [M+1].


Example 25



embedded image


In two parallel pots, Compound 1 (500 mg, 8.77 mmol) was dissolved in DCM, added with compound 1a (3.3 g, 21.93 mmol), HATU (4.01 g, 5.52 mmol), and DIEA (3.71 g, 56.5 mmol), stirred at room temperature overnight, quenched with water, the resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and subjected to purification by column chromatography to obtain 128 mg of product.




embedded image


Compound 2 (128 mg, 0.743 mmol) was dissolved in ACN, added with compound 2a (300 mg, 0.752 mmol), stirred at 90° C. overnight. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 300 mg of crude product.




embedded image


Compound 3 (300 mg) was dissolved in THF/H2O, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), and stirred at room temperature for 10 min. After the hydrolysis was completed when monitored by LCMS, 1M HCl was added dropwise to neutral, the reaction solution was extracted with EA, and the extract was concentrated and subjected to purification by pre-HPLC (acid method) to prepare 25 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.341 (s, 2H), 8.649-8.923 (m, 4H), 6.274-7.188 (m, 4H), 3.255-3.324 (m, 4H), 1.694-2.067 (m, 9H).


HPLC purity: @214 nm 99.05%, @254 nm 98.1%.


LC-MS: m/z497[M+1].


Example 26



embedded image


Compound 1 (1.0 g, 10 mmol) and Compound 2 (2.7 mg, 30 mmol) were dissolved in 20 mL of DMF and stirred at room temperature, then added with HATU (3.8 g, 10 mmol), DIEA (6.5 g, 50 mmol) and stirred for 10 h. The reaction solution was added to 60 mL of water, extracted with ethyl acetate, and the extract was evaporated to dryness under reduced pressure and subjected to purification by column chromatography to obtain Compound 3 (0.4 g, yield: 24%).




embedded image


Compound 3 (35 mg, 0.2 mmol) and Compound 4 (90 mg, 0.22 mmol) were dissolved in 10 mL of acetonitrile and stirred at 90° C. for 20 h, and the reaction solution was directly evaporated to dryness under reduced pressure to give Compound 5, which was a crude product and used directly in the next step (120 mg).




embedded image


Compound 3 (110 mg, 0.2 mmol) was dissolved in a solution of 1 N NaOH (4 mL) in tetrahydrofuran (5 mL), stirred at room temperature for 1 hour, and the resultant was subjected to purification by pre-HPLC to obtain the desired compound (15 mg, yield: 15%).


HNMR (DMSO, 400M): 11.43 (s, 1H), 11.27 (s, 1H), 9.06 (s, 1H), 8.92 (s, 1H), 8.64 (s, 3H), 7.16-7.21 (t, 1H), 7.09-7.11 (q, 1H), 6.74-6.78 (m, 1H), 6.26-6.29 (m, 1H), 3.23-3.35 (m, 4H), 2.32-2.34 (m, 2H), 1.80-1.84 (m, 2H), 0.95-1.01 (m, 1H), 0.49-0.51 (m, 5H), 0.18-0.19 (m, 2H).


HPLC purity: @214 nm 96.5%, @254 nm 97.9%.


LC-MS: m/z497.2 [M+1].


Example 27



embedded image


Compound 1 (45 mg, 0.2 mmol) and Compound 2 (90 mg, 0.22 mmol) were dissolved in 10 mL of acetonitrile and stirred at 90° C. for 20 h, and the reaction solution was directly evaporated to dryness under reduced pressure to give Compound 4, which was a crude product and used directly in the next step (120 mg crude, y>99%).




embedded image


Compound 3 (110 mg, 0.2 mmol) was dissolved in a solution of 1N NaOH (4 mL) in tetrahydrofuran (5 mL) and stirred at room temperature for 1 hour, and the resultant was subjected to purification by pre-HPLC to prepare the desired compound (55 mg y=50%).


HNMR (DMSO, 400M): 11.53 (s, 1H), 11.44 (s, 1H), 9.13 (s, 1H), 8.92 (s, 1H), 8.67 (s, 1H), 7.16-7.21 (t, 1H), 7.09-7.11 (q, 1H), 6.74-6.78 (m, 1H), 6.26-6.29 (m, 1H), 3.23-3.34 (m, 4H), 2.27-2.28 (m, 2H), 1.66-1.84 (m, 8H), 0.91-1.17 (m, 5H).


LC-MS: m/z 583.2 [M+1].


Example 28



embedded image


Compound 1 (106 mg, 0.25 mmol) was dissolved in acetone (10 mL), added with potassium carbonate (138 mg, 1 mmol), added dropwise with Compound 1a (43.5 mg, 0.25 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 100 mg of a crude product.




embedded image


Compound 2 (100 mg) was dissolved in methanol, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min, and the reaction solution was concentrated and subjected to purification by pre-HPLC to prepare 9 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.48 (s, 1H), 11.44 (s, 1H), 8.92-8.95 (m, 3H), 7.27-7.34 (m, 5H), 7.09-7.17 (m, 2H), 6.74-6.76 (m, 1H), 6.32-6.35 (m, 1H), 3.75-3.77 (m, 2H), 3.50-3.56 (m, 2H), 3.44-3.46 (m, 2H).


HPLC purity: @214 nm 97.9%, @254 nm 99.2%.


LC-MS: m/z 521 [M+1].


Example 29



embedded image


Compound 1 (110 mg, 0.25 mmol) was dissolved in acetone (10 mL), added with potassium carbonate (138 mg, 1 mmol), added dropwise with Compound 1a (43.5 mg, 0.25 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 100 mg of a crude product.




embedded image


Compound 2 (100 mg) was dissolved in methanol, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min, and the reaction solution was concentrated and subjected to purification by pre-HPLC to prepare 9 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.64 (s, 1H), 11.42 (s, 1H), 9.03-9.07 (m, 3H), 7.27-7.34 (m, 5H), 7.09-7.17 (m, 2H), 6.74-6.76 (m, 1H), 6.32-6.35 (m, 1H), 3.75-3.77 (m, 2H), 3.14-3.34 (m, 4H), 1.80-1.84 (m, 2H).


HPLC purity: @214 nm 96.0%, @254 nm 96.0%.


LC-MS: m/z 535 [M+1].


Example 30



embedded image


Compound 1 (330 mg, 0.75 mmol) was dissolved in acetone (30 mL), added with potassium carbonate (414 mg, 3 mmol), added dropwise with Compound 1a (130.5 mg, 0.75 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 300 mg of a crude product.




embedded image


Compound 2 (300 mg) was dissolved in methanol, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min, and the reaction solution was concentrated and subjected to purification by pre-HPLC to obtain 59 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.64 (s, 1H), 11.42 (s, 1H), 9.03-9.07 (m, 4H), 7.32-7.35 (m, 2H), 7.15-7.32 (m, 3H), 7.08-7.11 (m, 1H) 6.74-6.77 (m, 1H), 6.25-6.28 (m, 1H), 3.75-3.77 (m, 2H), 3.14-3.34 (m, 4H), 1.80-1.84 (m, 2H).


HPLC purity: @214 nm 99.90%, @254 nm 99.70%.


LC-MS: m/z569[M+1].


Example 31



embedded image


Compound 1 (330 mg, 0.75 mmol) was dissolved in acetone (30 mL), added with potassium carbonate (414 mg, 3 mmol), added dropwise with Compound 1a (130.5 mg, 0.75 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 300 mg of a crude product.




embedded image


Compound 2 (300 mg) was dissolved in methanol, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min, and the reaction solution was concentrated and subjected to purification by pre-HPLC to obtain 35 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.64 (s, 1H), 11.42 (s, 1H), 9.03-9.07 (m, 4H), 7.32-7.35 (m, 2H), 7.15-7.32 (m, 3H), 7.08-7.11 (m, 1H) 6.74-6.77 (m, 1H), 6.25-6.28 (m, 1H), 3.75-3.77 (m, 2H), 3.14-3.34 (m, 4H), 1.80-1.84 (m, 2H).


HPLC purity: @214 nm 99.40%, @254 nm 99.25%.


LC-MS: m/z553[M+1].


Example 32



embedded image


Compound 1 (330 mg, 0.75 mmol) was dissolved in tetrahydrofuran (10 mL), added with potassium carbonate (414 mg, 3 mmol), stirred at room temperature for 20 min, added dropwise with a solution of Compound 1a (184 mg, 1 mmol) in tetrahydrofuran (1 mL), stirred at room temperature for 2 h, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 500 mg of a crude product.




embedded image


Compound 2 (500 mg) was dissolved in tetrahydrofuran (20 mL), stirred at room temperature for 5 min, added dropwise with 1N sodium hydroxide (5 mL), stirred at room temperature for 30 min. The reaction solution was extracted with water and ethyl acetate, and dried over anhydrous sodium sulfate, and concentrated to obtain a crude product. The crude product was dissolved in acetonitrile, and subjected to purification by pre-HPLC to prepare obtain 29 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.91 (s, 1H), 11.43 (s, 1H), 9.04 (s, 1H), 8.90 (s, 1H), 8.70 (s, 2H), 7.25-7.23 (m, 2H), 7.20-7.16 (m, 1H), 7.11-7.09 (m, 1H), 6.91-6.88 (m, 2H), 6.78-6.75 (m, 1H), 6.25 (m, 1H), 3.73 (s, 3H), 3.68 (s, 2H), 3.28-3.23 (m, 4H), 1.83-1.80 (m, 2H).


HPLC purity: @214 nm 99.2%, @254 nm 99.7%.


LC-MS: m/z563.2[M+1].


Example 33



embedded image


Compound 1 (150 mg, 0.47 mmol) was dissolved in DCM, added with Et3N, stirred at room temperature for 1 h, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 100 mg of crude product.




embedded image


Compound 2 (100 mg, 1.08 mmol) was dissolved in ACN, added with compound 2a (120 mg, 1.06 mmol), stirred at 90° C. overnight. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 100 mg of crude product.




embedded image


Compound 3 (100 mg) was dissolved in THF/H2O, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min, and the reaction solution was concentrated and subjected to purification by pre-HPLC to obtain 8 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.579 (s, 1H), 11.480 (s, 1H), 8.718-8.917 (m, 3H), 6.286-7.187 (m, 4H), 3.255-3.324 (m, 4H), 2.499-2.507 (m, 1H), 1.570-1.843 (m, 8H).


HPLC purity: @214 nm 97.3%, @254 nm 98.1%.


LC-MS: m/z497[M+1].


Example 34



embedded image


Compound 1 (500 mg, 8.77 mmol) was dissolved in DCM, added with compound 1a (3.3 g, 21.93 mmol), HATU (4.01 g, 5.52 mmol), and DIEA (3.71 g, 56.5 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and subjected to purification by column chromatography to obtain 120 mg of desired product.




embedded image


Compound 2 (120 mg, 1.08 mmol) was dissolved in ACN, added with compound 2a (100 mg, 1.06 mmol), stirred at 90° C. overnight. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 100 mg of crude product.




embedded image


Compound 3 (100 mg) was dissolved in THF/H2O, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min, and the reaction solution was concentrated and subjected to purification by pre-HPLC to give 12 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 12.041 (s, 1H), 8.649-8.923 (m, 3H), 6.274-7.188 (m, 4H), 3.255-3.324 (m, 4H), 2.509-2.683 (m, 2H), 1.594-2.067 (m, 6H).


HPLC purity: @214 nm 95.9%, @254 nm 95.4%.


LC-MS: m/z497[M+1].


Example 35



embedded image


Compound 1 (1 g, 8.77 mmol) was dissolved in DCM, added with 1a (6.3 g, 21.93 mmol), HATU (4.01 g, 10.52 mmol), and DIEA (5.71 g, 56.5 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and subjected to purification by column chromatography to give 200 mg of product.




embedded image


Compound 2 (200 mg, 1.08 mmol) was dissolved in ACN, added with compound 2a (420 mg, 1.06 mmol), stirred at 90° C. overnight. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to give 100 mg of a crude product.




embedded image


Compound 3 (100 mg) was dissolved in THF/H2O, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), and stirred at room temperature for 10 min, and the reaction solution was concentrated and subjected to purification by pre-HPLC to prepare 12 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.579 (s, 1H), 11.480 (s, 1H), 8.718-8.917 (m, 3H), 6.286-7.187 (m, 4H), 3.255-3.324 (m, 4H), 2.499-2.507 (m, 1H), 1.570-1.843 (m, 10H).


HPLC purity: @214 nm 91.2%, @254 nm 96.8%.


LC-MS: m/z511 [M+1].


Example 36



embedded image


Compound 1 (500 mg, 8.77 mmol) was dissolved in DCM, added with compound 1a (3.3 g, 21.93 mmol), HATU (4.01 g, 10.52 mmol), and DIEA (5.71 g, 56.5 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and subjected to purification by column chromatography to obtain 120 mg of desired product.




embedded image


Compound 2 (120 mg, 1.08 mmol) was dissolved in ACN, added with compound 2a (100 mg, 1.06 mmol), stirred at 90° C. overnight. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 100 mg of a crude product.




embedded image


Compound 3 (100 mg) was dissolved in THF/H2O, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min, and the reaction solution was concentrated and subjected to purification by pre-HPLC to give 12 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.441 (s, 2H), 8.649-8.923 (m, 3H), 6.274-7.188 (m, 4H), 3.255-3.324 (m, 4H), 1.694-2.067 (m, 11H).


HPLC purity: @214 nm 99.1%, @254 nm 99.3%.


LC-MS: m/z511 [M+1].


Example 37



embedded image


Compound 1 (110 mg, 0.25 mmol) was dissolved in acetone (10 mL), added with potassium carbonate (138 mg, 1 mmol), added dropwise with Compound 1a (113 mg, 1 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 100 mg of a crude product.




embedded image


Compounds 2 and 2a (100 mg) were dissolved in methanol, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min. The reaction solution was concentrated and subjected to purification by pre-HPLC to give the desired products XSD3-087 (15 mg) and XSD3-087-01 (17 mg).


XSD3-087 1H-NMR (400 MHz, DMSO): δ (ppm): 11.41 (s, 1H), 8.90 (m, 1H), 6.18-7.32 (s, 1H), 7.10-7.21 (m, 2H), 7.10-7.12 (m, 2H), 6.76-6.79 (m, 1H), 6.23 (m, 1H), 3.12-3.23 (m, 4H), 6.76-6.79 (m, 1H), 2.67 (s, 3H), 1.72-1.75 (m, 2H).


HPLC purity: @214 nm 93.5%, @254 nm 94.6%.


LC-MS: m/z492 [M+1].


XSD3-087-01 1H-NMR (400 MHz, DMSO): δ (ppm): 11.43 (s, 1H), 8.90 (m, 1H), 8.01 (m, 2H), 7.10-7.20 (s, 2H), 6.85 (m, 1H), 6.23 (m, 1H), 3.72-3.76 (m, 2H), 3.40 (s, 3H), 3.24-3.26 (m, 2H), 2.93 (s, 3H), 1.87-1.91 (m, 2H).


HPLC purity: @214 nm 98%, @254 nm 98.6%.


LC-MS: m/z570 [M+3].


Example 38



embedded image


Compound 1 (100 mg, 0.21 mmol) was dissolved in acetone (10 mL), added with potassium carbonate (78 mg, 0.55 mmol), added dropwise with Compound 1a (41 mg, 0.23 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 100 mg of a crude product.




embedded image


Compound 2 (100 mg) was dissolved in THF/H2O, stirred at room temperature for 10 min, added dropwise with 1N sodium hydroxide (1 mL), stirred at room temperature for 10 min. The reaction solution was concentrated and subjected to pre-HPLC to prepare 5 mg of desired product of 5 mg.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.421 (s, 1H), 8.897 (s, 1H), 7.153-7.197 (m, 2H), 7.105-7.120 (m, 1H), 6.746-7.098 (m, 1H), 6.220 (s, 1H), 4.078 (s, 2H), 3.104-3.129 (m, 4H), 2.735-2.761 (m, 2H), 1.535-1.853 (m, 7H), 0.976-1.202 (m, 6H).


HPLC purity: @214 nm 99.8%, @254 nm 99.3%.


LC-MS: m/z575[M+1].


Example 39



embedded image


Compound 1 (1.0 g, 3.6 mmol) was dissolved in acetone (16 mL), added with potassium carbonate (993 mg, 7.2 mmol), added dropwise with Compound 1a (254 mg, 2.7 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 300 mg of a crude product.




embedded image


Compound 2a (100 mg) was dissolved in acetonitrile, added with compound 2 (40 mg, 0.27 mmol), stirred at 90° C. for 12 h, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 80 mg of a crude product.




embedded image


Compound 3 (80 mg) was dissolved in DMF, stirred at room temperature for 10 min, added with potassium carbonate (132 mg, 0.96 mmol), stirred at room temperature for 12 h, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, concentrated to obtain 60 mg of a crude product, which was subjected to purification by pre-HPLC to obtain 14.2 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.53 (s, 1H), 8.91 (s, 1H), 8.58-8.27 (m, 3H), 7.38-7.12 (m, 2H), 6.78 (s, 1H), 6.28 (s, 1H), 3.65 (s, 3H), 3.28-3.24 (m, 4H), 1.79 (s, 2H).


HPLC purity: @214 nm 94.5%, @254 nm 95.4%.


LC-MS: m/z 473 [M+1].


Example 40



embedded image


Compound 1 (1.0 g, 3.6 mmol) was dissolved in acetone (16 mL), added with potassium carbonate (993 mg, 7.2 mmol), added dropwise with Compound 1a (294 mg, 2.7 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 400 mg of a crude product.




embedded image


Compound 2a (100 mg) was dissolved in acetonitrile, added with Compound 2 (42 mg, 0.27 mmol), stirred at 90° C. for 12 h, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 66 mg of a crude product.




embedded image


Compound 3 (66 mg) was dissolved in DMF, stirred at room temperature for 10 min, added with potassium carbonate (108 mg, 0.78 mmol), stirred at room temperature for 12 h, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 50 mg of a crude product, which was subjected to purification by pre-HPLC to prepare 26.5 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.47 (s, 1H), 11.24 (s, 1H), 8.92 (s, 1H), 8.62 (s, 2H), 7.21-7.09 (m, 2H), 6.79-6.75 (m, 1H), 6.29-6.25 (m, 1H), 4.25-4.20 (m, 2H), 3.32-3.24 (m, 4H), 1.85-1.80 (m, 2H), 1.28-1.24 (m, 3H).


HPLC purity: @214 nm 97.8%, @254 nm 98.6%.


LC-MS: m/z 487 [M+1].


Example 41



embedded image


Compound 1 (1.0 g, 3.6 mmol) was dissolved in acetone (16 mL), added with potassium carbonate (993 mg, 7.2 mmol), added dropwise with Compound 1a (732 mg, 6.0 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 360 mg of a crude product.




embedded image


Compound 2a (100 mg) was dissolved in acetonitrile, added with Compound 2 (45 mg, 0.27 mmol), stirred at 90° C. for 12 h, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 51 mg of a crude product.




embedded image


Compound 3 (51 mg) was dissolved in DMF, stirred at room temperature for 10 min, added with potassium carbonate (108 mg, 0.78 mmol), stirred at room temperature for 12 h, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 50 mg of a crude product, which was subjected to purification by pre-HPLC to obtain 10.6 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.48 (s, 1H), 11.18 (s, 1H), 8.92 (s, 1H), 8.60 (s, 2H), 7.21-7.09 (m, 2H), 6.79-6.75 (m, 1H), 6.30-6.25 (m, 1H), 4.98-4.91 (m, 1H), 3.32-3.24 (m, 4H), 1.83-1.78 (m, 2H), 1.28-1.27 (m, 6H).


HPLC purity: @214 nm 95.2%, @254 nm 98.0%.


LC-MS: m/z 501 [M+1].


Example 42



embedded image


Compound 1 (1.0 g, 3.6 mmol) was dissolved in acetone (16 mL), added with potassium carbonate (993 mg, 7.2 mmol), added dropwise with Compound 1a (540 mg, 3.6 mmol), stirred at room temperature overnight, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 330 mg of a crude product.




embedded image


Compound 2a (100 mg) was dissolved in acetonitrile, added with Compound 2 (50 mg, 0.27 mmol), stirred at 90° C. for 12 h, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 90 mg of a crude product.




embedded image


Compound 3 (90 mg) was dissolved in DMF, stirred at room temperature for 10 min, added with potassium carbonate (132 mg, 0.96 mmol), stirred at room temperature for 12 h, quenched with water. The resultant was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated to obtain 70 mg of a crude product, which was subjected to purification by pre-HPLC to obtain 32.5 mg of desired product.



1H-NMR (400 MHz, DMSO-d6): δ (ppm): 11.51 (s, 1H), 11.24 (s, 1H), 8.91 (s, 1H), 8.55-8.27 (m, 3H), 7.21-7.09 (m, 2H), 6.79-6.75 (m, 1H), 6.29-6.25 (m, 1H), 4.25-4.206 (m, 2H), 3.32-3.24 (m, 4H), 1.80 (s, 2H), 1.57 (s, 2H), 1.28 (s, 4H), 0.88-0.85 (m, 3H).



1H-NMR (400 MHz, CDCl3): δ (ppm): 7.23-7.20 (m, 1H), 7.10 (s, 1H), 7.04-7.00 (m, 1H), 6.92-6.88 (m, 1H), 5.85 (s, 1H), 4.09-4.06 (m, 2H), 3.58-3.53 (m, 2), 3.38-3.35 (m, 2H), 1.98-1.97 (m, 2H), 1.70-1.63 (m, 2H), 1.38-1.35 (m, 4H), 0.91-0.88 (m, 3H).


HPLC purity: @214 nm 98.1%, @254 nm 97.8%.


LC-MS: m/z 529[M+1]


Referring to Synthetic Method I and the above examples, the following compounds were synthesized.















Example No.
Substituent
Substance No.
MS







Example 43


embedded image


XSD3-075
560.3





Example 44
R2 is ethylcarbonyl, n is 1, R1, R3, R4 are H.
XSD3-063
456.3


Example 45
R2 is isopropylcarbonyl, n is 1, R1, R3, R4 are H.
XSD3-064
470.2


Example 46
R2 is methylcarbonyl, n is 2, R1, R3, R4 are H.
XSD3-086
456.1


Example 47
R2 is cyano, n is 1, R1, R3, R4 are H.
XSD3-073
425.1


Example 48
R2 is cyano, n is 2, R1, R3, R4 are H.
XSD3-085
439.1





Example 49


embedded image


XSD3-071
510.2





Example 50


embedded image


XSD3-077
469.1





Example 51


embedded image


XSD3-083
524.2





Example 52
R3 is 1,2-ethanedily n is 1, R1, R2, R4 are H.
XSD3-090
426.1


Example 53
R3 is 1,2-ethanedily n is 2, R1, R2, R4 are H.
XSD3-091
440.1





Example 54


embedded image


XSD3-099
554.1









Example 55: Determination of the Activity for Inhibiting Indoleamine 2,3-Dioxygenase and IC50

The construction of plasmid containing the human indoleamine 2,3-dioxygenase gene, the expression in E. coli, the extraction and the purification were performed according to the method reported by Littlejohn et al. (Takikawa O, Kuroiwa T, Yamazaki F, et al. J. Biol. Chem. 1988, 263, 2041-2048). In a 96-well plate, 50 mM potassium phosphate buffer (pH 6.5), 20 mM ascorbate, 20 μM methylene blue and purified human indoleamine 2,3-dioxygenase protein were mixed, and 200 μM L-tryptophan and inhibitor were added to the mixture. The reaction was carried out at 37° C. for 60 minutes, then the reaction was terminated by adding 30% trichloroacetic acid, and the mixture was incubated at 65° C. for 15 minutes to hydrolyze N-formyl-kynurenine into kynurenine, and centrifuged at 3400 g for 5 min to remove the precipitated protein. The supernatant was transferred to a new 96-well plate, a solution of 2% (w/v) p-dimethylaminobenzaldehyde in acetic acid was added, the reaction was performed by incubation at 25° C. for 10 minutes, and the reading was carried out on a spectrophotometer at 480 nm. The control wells were those without indoleamine 2,3-dioxygenase inhibitor or without indoleamine 2,3-dioxygenase, and used as the necessary nonlinear regression parameters to determine IC50 of each compound. The nonlinear regression and the determination of IC50 values were carried out by using GraphPad PRism 4 software. Those compounds with an IC50 of less than 10 μM were considered as effective inhibitors in the assay. The compounds of examples of the present invention had excellent activity for inhibiting indoleamine 2,3-dioxygenase.









TABLE 1







IC50 values of compounds










Structural formula No.
IC50 (nM)














3047
37



3048
20



3049
22



3051
23



3053
16



INCB024360
102







Note: INCB024360 was an indoleamine 2,3-dioxygenase inhibitor with a structure of





embedded image








The IC50 values of the following compounds were determined by the method described in Example 55, and the specific results are shown in Table 2:









TABLE 2







IC50 values of compounds










Structural formula No.
IC50 (nM)














XSD3-058
57



XSD3-079
95



XSD3-093
44



XSD3-093-1
56



XSD3-093-2
61



XSD3-093-3
76



XSD3-074
63



XSD3-088
72



XSD3-089
67



XSD3-076
102



XSD3-081
89



XSD3-069
345



XSD3-052
343



XSD3-065
117.9



XSD3-066
131.5



XSD3-067
142



XSD3-068
539



XSD3-070
345



XSD3-072
502



XSD3-078
145



XSD3-080
87



XSD3-082
101



XSD3-084
160



XSD3-092
399










Example 56: In Vivo Antitumor Activity Test of Indoleamine 2,3-Dioxygenase Inhibitors
1. Animal Grouping and Test Method

LLC cells in logarithmic growth phase were taken, the cell viability thereof was detected by trypan blue staining method, the viable cell concentration was adjusted to 1×107 cells/ml, and subcutaneously injected at a dose of 0.2 ml/mouse into homologous C57BL6 mice. Once tumors were established, the mice were randomly divided into model group, cyclophosphamide (CTX) group, compound INCB024360 group, compound 3047 group according to their tumor weights and body weights, there were 10 mice in each group, wherein the CTX group was intraperitoneally injected with a dose of 150 mg·kg−1, the compound INCB024360 group and the compound 3047 group were subjected to intragastric administration, the model group was given the same volume of normal saline at the same time, and the administration frequency for each group was once per day. The test was terminated after 21 days of administration.


24 Hours after the last administration, the animals were weighed and sacrificed, the tumors were taken and weighted, and average tumor inhibition rate (I) was calculated according to the following formula: I=(1−average tumor weight of the drug-administered group/average tumor weight of the model group)×100%


2. Data Statistics and Processing Method

The experimental data were analyzed by spss16.0, and one-way ANOVA, and difference with p<0.05 was statistically significant.


3. Test Results and Discussion









TABLE 3







Inhibition results of compounds on LLC tumors in mice











Number of





animals

Tumor inhibition


Group
(mouse)
Tumor weight (g)
rate (%)





Model group
10
1.514 ± 0.312
/


CTX group
10
0.701 ± 0.314##
53.7


INCB024360
10
0.653 ± 0.178##
56.9


group


Compound
10

0.370 ± 0.209##※

75.6


3047 group





In comparison with the model group,



##P < 0.01;



In comparison with the CTX group and the INCB024360 group,



P < 0.05;







As can be seen from Table 3, the tumor weight of each drug-administered group was significantly different from that of the model group (P<0.01); the compound 3047 group was significantly different from the cyclophosphamide group and the INCB024360 group (P<0.05). This result indicates that the compound of the present invention is superior to the existing chemotherapeutic drug cyclophosphamide and the compound INCB024360 in therapeutic effect of tumors.









TABLE 4







Effects of compounds on body weight of mice












Average body
Average body




weight before test
weight after test



Group
(g)
(g)







Model group
21 ± 4.2
28 ± 3.2



CTX group
22 ± 3.1
19 ± 3.3



Compound
22 ± 3.4

29 ± 5.2##




3047 group







In comparison with the cyclophosphamide group,




##p < 0.01







As can be seen from Table 4, the compound 3047 group showed no significant difference in body weight in comparison with the model group, but showed a significant difference in comparison with the CTX group. This result indicates that the compound of the present invention increased the body weight in mice while controlling tumor growth, that is, the side effect was reduced, and the life quality of mice was significantly improved. Clinically, it means that the life quality of patients can be improved and the patient compliance with medications and medicament effectiveness can be significantly enhanced.


In addition, we also tested mouse colon cancer cell Colon26, mouse liver cancer cell Hepa1-6, mouse breast cancer cell 4T1 and other cell lines, and the results showed that the compounds of the present invention had significant inhibitory effects on these tumors.


The in vivo antitumor activity of the following compounds was determined by the method of Example 56, and the specific results are shown in the following table:









TABLE 5







Inhibition results of compound on LLC tumors in mice













Animal

Tumor




number
Tumor
inhibition



Group
(mouse)
weight (g)
rate (%)







Model group
10
1.445 ± 0.217
/



Compound
10
0.400 ± 0.112##
72.3



XSD3-058 group



Compound
10
0.384 ± 0.092##
73.4



XSD3-079 group







In comparison with the model group,




##P < 0.01.







As can be seen from Table 5, the tumor weight of each drug-administered group was significantly different from that of the model group (P<0.01). This result indicates that the compound of the present invention has significant therapeutic effect on tumor.


The effect of each compound on the body weight of mice was examined. It was found that there was no significant difference in body weight between the compound XSD3-058 group and the compound XSD3-079 group compared with the model group. This result indicates that the compounds of the present invention can increase the body weight in mice while controlling tumor growth, reduce the side effects of the drug and significantly improve the quality of life of the mouse. Clinically, the compounds of the present invention can improve the quality of life of patients and significantly enhance the patient compliance with medications and medicament effectiveness.


Example 57: Morris Water Maze Detection of Behavioral Changes in Mice with Alzheimer
1. Animal Grouping and Test Methods

In the present invention, 9-month-old mice were selected to establish AD models according to the method of Richardson et al. by single injection of aggregated A01-42 in the bilateral hippocampal CA3 region of rats, and then divided into a model group, a compound INCB024360 group, a compound 3047 group, 10 mice per group, five male and five female. Mouse behavioral analysis was performed using a Morris water maze (Ethovision XT monitoring and analysis software, Morris water maze system, from Noldus, Netherlands). The water maze test process was divided into two parts, hidden platform acquisition test for consecutive 5 days, and spatial probe test on the sixth day, the mice were administered according to the test groups and the design doses before each test. The mice were trained 4 times a day, each time the mice were dived in different areas, the water maze was divided into areas 1, 2, 3 and 4 according to the south, east, north and west, and the platform was the 5th area located in the 4th area. Each swimming time was 60 s, the interval for each training was about 1 h, and when a mouse did not find the platform, the latent period was calculated as 60 s. The hidden platform acquisition test was used to detect the learning ability of mice, while the spatial probe test was used to detect the spatial memory ability of mice.


2. Data Statistics and Processing Method

Using the statistical analysis software SPSS16.0, the escape latent period of the hidden platform acquisition test was analyzed by variance of multiple measures; and the swimming time in each quadrant and the number of crossing target in the spatial probe test were analyzed by one-way analysis of variance. The data were expressed as mean±standard deviation, and the significant level of difference was set as bilateral P=0.05.


3. Test Results and Discussion









TABLE 6







Latent period (s) of each group of animals for searching platform in hidden platform test












Group
1st Day
2nd Day
3th Day
4th Day
5th Day





Model group
54.0 ± 5.9
52.8 ± 1.9
43.5 ± 6.7
39.8 ± 6.9   
38.4 ± 5.6     


INCB024360
53.2 ± 2.3
51.4 ± 4.8
38.6 ± 5.5
28.7 ± 3.9# 
19.9 ± 6.7##  


group


Compound
52.3 ± 5.2

44.0 ± 4.4#

   22.1 ± 4.5##※
10.5 ± 5.7##※
5.4 ± 6.0##※※


3047 group





In comparison with the model group,



#P < 0.05,




##P < 0.01,



In comparison with the INCB024360 group,



P < 0.05,




※※P < 0.01.














TABLE 7







Time and number of stays of each group of animals in platform areas










Time of stay (s)
Number of stays (times)











Group
4th Area
5th Area
4th Area
5th Area





Model group
14.9 ± 3.8
1.9 ± 0.8   
4.8 ± 0.7 
1.0 ± 0.3   


INCB024360
16.2 ± 2.4
3.9 ± 0.7# 
8.4 ± 1.0# 
2.1 ± 0.4## 


group


Compound
   25.9 ± 5.2##※
5.9 ± 0.7##※
13.0 ± 1.5##※
3.8 ± 0.6##※


3047 group





In comparison with the model group,



#P < 0.05,




##P < 0.01,



In comparison with the INCB024360 group,



P < 0.05.







As can be seen from Tables 6 and 7, Compound 3047 can significantly improve learning and memory impairment in animals, significantly improve learning ability and spatial memory ability, and is superior to compound INCB024360, indicating that the compound of the present invention has great development value in the treatment of Alzheimer's syndrome.


The effect of the following compounds on behavior of mice with Alzheimer were tested by the method of Example 57. The specific results are shown in the following table:









TABLE 8







Latent period (s) of each group of animals for


searching platform in hidden platform test












Group
1st Day
2nd Day
3th Day
4th Day
5th Day





Model group
59.7 ± 4.6
59.8 ± 1.8
50.6 ± 5.5
42.8 ± 4.9
38.7 ± 5.2  


Compound
45.3 ± 3.7#
 41.3 ± 3.2#
25.4 ± 4.6##
15.5 ± 4.9##
7.3 ± 6.1##


XSD3-058 group


Compound
40.5 ± 5.5#
39.3 ± 44#
20.3 ± 4.2##
17.7 ± 5.2##
9.9 ± 5.2##


XSD3-079 group





In comparison with the model group,



#P < 0.05,




##P < 0.01.














TABLE 9







Time and number of stays of each group of animals in platform areas










Time of stay (s)
Number of stays (times)











Group
4th Area
5th Area
4th Area
5th Area





Model group
15.8 ± 3.1
2.0 ± 0.9
4.1 ± 0.9 
1.2 ± 0.4


Compound
25.2 ± 4.2##
5.9 ± 0.6##
13.3 ± 1.8##
3.5 ± 0.9##


XSD3-058 group


Compound
26.7 ± 3.3##
5.8 ± 0.7##
14.4 ± 1.3##
3.9 ± 0.5##


XSD3-079 group





In comparison with the model group,



##P < 0.01.







As can be seen from Tables 8 and 9, the compounds of the present invention can significantly improve learning and memory impairment in animals, and significantly improve learning ability and spatial memory ability, and the results indicate that the compounds of the present invention have great development value in the treatment of Alzheimer's syndrome.


Example 58: DC-Stimulated T Cell Proliferative Response after Drug Treatment

Dendritic cell (DC) is the most powerful antigen-presenting cell (APC), which can effectively activate naive T cells for proliferation, which is the most important difference between DC and other APCs. DC is the initiator of the immune response. DC has become one of the hotspots in immunology research today due to its key role in CD4+, CD8+ T cell immune response. Currently, the research of DC is mainly focused on prevention and treatment effect on tumor diseases, autoimmune diseases, transplant rejection, and anti-infection.


1. Isolation and Culture of Human Peripheral Blood Dendritic Cells

The human peripheral blood leukocyte layer was taken and diluted with same volume of 0.01 mol/L PBS. PBMC was routinely isolated with a lymphocyte separation medium, and the cell concentration was adjusted to 3×106 ml−1 in a complete RPMI1640 medium; the cells were added to a 6-well plate, 3 ml per well, and cultured in a 5% CO2, 37° C. incubator for 2 hours, washed with PBS for 3 times to remove non-adherent cells, then added with a culture medium containing IL-4 (100 U/ml), GM-CSF (150 ng/ml) and TNF-α (500 U/ml), and then routinely cultured, the medium was changed for half every other day. After 8 days of culture, the cells were used for identification and experiment.


2. Preparation of T Cells

The human PBMC layer was separated by the method in the step 1. The macrophages were removed by the adherence method, the B cells were removed by the nylon wool fiber syringe method, and the obtained T cells were adjusted to a cell concentration of 1×106 cells/ml.


3. Preparation of DC

The mature DCs with a purity of 99% were centrifuged, added with RPMI1640 to adjust the cell concentration to 1×105, 4×104, 2×104/ml, and added to a 96-well plate, two wells for each concentration, 100 μl/well. Compound INCB024360 and Compound 3047 were separately added, and cultured for 2 days.


4. T Cell Proliferation Assay (MLR)

T cells were added to DCs of each of the above drug-added groups, 100 μl/well, cultured in a 5% CO2, 37° C. incubator for 72 hours; for each well, 100 μl of the culture solution was gently sucked out 6 hours before the end of the culture, and 10 μl of MTT (5 mg/ml) was added, further cultured in incubator for 6 hours, then 100 μl of 0.01 mol/L HCl-10% SDS was added, stood at 37° C. overnight, and the absorbance value (A570 nm) was determined by a microplate to show the T cell proliferation level.


5. Experimental Results and Analysis









TABLE 10







Effect of Compound 3047 on DC-stimulated T cell proliferation









T:DC (absorbance value)










Group
10:1
25:1
50:1





Control
0.801 ± 0.003   
0.782 ± 0.002   
0.755 ± 0.005   


group


INCB024360
0.830 ± 0.002# 
0.804 ± 0.004# 
0.799 ± 0.003# 


group


Compound
0.864 ± 0.006##※
0.834 ± 0.004##※
0.825 ± 0.007##※


3047 group





In comparison with the control group,



#P < 0.05,




##P < 0.01,



In comparison with the INCB024360 group,



P < 0.05.







As can be seen from Table 10, compared with the control group, the numbers of T cells in the compound 3047 group and the INCB024360 group were significantly increased, showing significant differences (#P<0.05, ##P<0.01); and compared with the compound INCB024360 group, the compound 3047 group had a more significant effect on the proliferation of T cells, showing significant difference (*P<0.05). This indicates that the compound of the present invention has a significant effect on promoting DC-stimulated T cell proliferation, and the effect is significantly better than that of the compound INCB024360, and thereby can be used for the treatment of tumor diseases, autoimmune diseases, transplant rejection and infectious diseases.


The DC-stimulated T cell proliferative responses after treatment with the following compounds were determined by the method of Example 58, and the specific results are shown in the following table:









TABLE 11







Effect of each compound on DC-stimulated T cell proliferation









T:DC (absorbance value)










Group
10:1
25:1
50:1





Control group
0.810 ± 0.004
0.793 ± 0.004
0.760 ± 0.002


Compound
0.880 ± 0.003##
0.849 ± 0.002##
0.831 ± 0.002##


XSD3-058 group


Compound
0.876 ± 0.004##
0.850 ± 0.003##
0.837 ± 0.004##


XSD3-079 group





In comparison with the control group,



##P < 0.01.







As can be seen from Table 11, compared with the control group, the number of T cells in each compound group was significantly increased, showing a significant difference (##P<0.01), indicating that the compound of the present invention can significantly promote the DC-stimulated T cell proliferation, and thereby can be used for the treatment of IDO-related diseases such as tumor diseases, autoimmune diseases, transplant rejection and infectious diseases.

Claims
  • 1. A compound represented by Formula I0, or a pharmaceutically acceptable salt thereof:
  • 2. The compound or a pharmaceutically acceptable salt thereof according to claim 1, characterized in that R1, R2 are each independently selected from the group consisting of the following substituents: C1-6 alkyl, carbonyl, C1-6 alkoxy, sulfonyl, amidino, sulfinyl, which is substituted by one or more substituents selected from group consisting of: halogen, hydroxy, carboxy, carbonyl, aldehyde group, cyano, amino, aryl, heteroaryl, C1-6 alkyl, C3-12 cycloalkyl, C2-6 alkenyl, C3-12 cycloalkenyl; wherein, the carboxy, carbonyl, aldehyde group, cyano, amino, aryl, heteroaryl, C3-12 cycloalkyl, C2-6 alkenyl, C3-12 cycloalkenyl as the substituents of C1-6 alkyl, carbonyl, C1-6 alkoxy, sulfonyl, amidino or sulfinyl are optionally substituted by one or more substituents selected from group consisting of: H, halogen, C1-6 alkyl, carbonyl, C1-6 alkoxy, sulfinyl and sulfonyl; the halogen is selected from the group consisting of F, Cl, Br and I.
  • 3. The compound or a pharmaceutically acceptable salt thereof according to claim 1, characterized in that R1, R2 are each independently selected from the group consisting of the following substituents: H, methyl, ethyl, propyl, isopropyl, R5C(O)—, R5S(O)x—; R5 is selected from the group consisting of C1-10 alkyl, C3-12 cycloalkyl, substituted C1-10 alkyl and substituted C3-12 cycloalkyl, wherein the substituted C1-10 alkyl or substituted C3-12 cycloalkyl is substituted by hydroxy, cyano, C1-6 alkyl, C3-12 cycloalkyl, C1-6 alkoxy, aryl or heteroaryl, wherein x is 1 or 2.
  • 4. The compound or a pharmaceutically acceptable salt thereof according to claim 3, characterized in that R1, R2 are each independently selected from the group consisting of the following substituents: H, methyl, ethyl, propyl, isopropyl, R5C(O)—, R5S(O)x—; R5 is selected from the group consisting of C1-10 alkyl, C3-12 cycloalkyl, substituted C1-6 alkyl and substituted C3-8 cycloalkyl, wherein the substituted C1-6 alkyl or the substituted C3-8 cycloalkyl is substituted by hydroxy, cyano, C1-6 alkyl, C3-12 cycloalkyl, C1-6 alkoxy, aryl or heteroaryl; x is 2; R3, R4 are both H.
  • 5. The compound or a pharmaceutically acceptable salt thereof according to claim 1, characterized in that when R2, R3, and R4 in the Formula o are H respectively, the compound is represented by Formula I:
  • 6. (canceled)
  • 7. The compound or a pharmaceutically acceptable salt thereof according to claim 5, characterized in that R1 is selected from the group consisting of H, NH2, CN, methyl, ethyl, propyl, isopropyl, butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methoxycarbonyl, tert-butoxycarbonyl, fluorenylmethoxycarbonyl, allyloxycarbonyl, benzyl, 5-azaindene methylcarbonyl.
  • 8. (canceled)
  • 9. The compound or a pharmaceutically acceptable salt thereof according to claim 5, characterized in that when R1 in Formula I is H, the compound is represented by Formula II,
  • 10. The compound or a pharmaceutically acceptable salt thereof according to claim 9, characterized in that the n is 0, 1, 2 or 3.
  • 11. The compound or a pharmaceutically acceptable salt thereof according to claim 5, characterized in that when R1 in Formula I is R0-substituted carbonyl, the compound is represented by Formula III,
  • 12. (canceled)
  • 13. The compound or a pharmaceutically acceptable salt thereof according to claim 1, characterized in that when R1 in the Formula I0 is H, the compound is represented by Formula IV:
  • 14. (canceled)
  • 15. The compound or a pharmaceutically acceptable salt thereof according to claim 1, characterized in that the compound is selected from the group consisting of:
  • 16. A synthesis method for preparing a compound represented by the Formula I0 or a pharmaceutically acceptable salt thereof according to claim 1,
  • 17. The synthesis method according to claim 16, characterized in that the compound represented by formula 2 is obtained by reacting a compound represented by formula 1 with compound 2a under an alkaline condition,
  • 18. The synthesis method according to claim 17, characterized in that the compound 2a is obtained by oxidizing compound 1a,
  • 19. A synthesis method for preparing a compound represented by the following formula II or a pharmaceutically acceptable salt thereof according to claim 9,
  • 20. A synthesis method for preparing a compound represented by the following formula,
  • 21. A synthesis method for preparing a compound represented by Formula III or a pharmaceutically acceptable salt thereof according to claim 11,
  • 22-24. (canceled)
  • 25. A pharmaceutical composition, comprising the compound or a pharmaceutically acceptable salt thereof according to claim 1, and one or more pharmaceutically acceptable pharmaceutic adjuvants.
  • 26. A method for treatment of a disease, the method comprising administering to a patient in need of such treatment a therapeutically effective amount of at least one of the compound or a pharmaceutically acceptable salt thereof according to claim 1, wherein the disease pathologically characterized by indoleamine 2,3-dioxygenase-mediated tryptophan metabolic pathway, or the disease is a cancer, an infectious disease, a neurodegenerative disease, a depression, an anxiety or an age-related cataract.
  • 27. (canceled)
  • 28. The method according to claim 26, wherein the cancer is selected from the group consisting of lung cancer, liver cancer, colon cancer, pancreatic cancer, breast cancer, prostate cancer, brain cancer, ovarian cancer, cervical cancer, testicular cancer, renal cancer, head and neck cancer, lymphoma, melanoma or leukemia.
  • 29. The method according to claim 26, wherein the neurodegenerative disease is Alzheimer's disease.
  • 30. The method according to claim 26, wherein the infectious disease is an infection caused by a bacterium, a fungus, a virus or a parasite.
Priority Claims (1)
Number Date Country Kind
201710288663.1 Apr 2017 CN national
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2018/084923 4/27/2018 WO 00