CN110204537B - 1,2, 5-oxadiazole derivatives as inhibitors of indoleamine 2, 3-dioxygenase - Google Patents

1,2, 5-oxadiazole derivatives as inhibitors of indoleamine 2, 3-dioxygenase Download PDF

Info

Publication number
CN110204537B
CN110204537B CN201910617997.8A CN201910617997A CN110204537B CN 110204537 B CN110204537 B CN 110204537B CN 201910617997 A CN201910617997 A CN 201910617997A CN 110204537 B CN110204537 B CN 110204537B
Authority
CN
China
Prior art keywords
reaction
compound
synthesis
give
solution
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201910617997.8A
Other languages
Chinese (zh)
Other versions
CN110204537A (en
Inventor
余强
丁炬平
郝岩
殷时杰
潘慧平
汤木林
徐永梅
任峰
陈春麟
高珍妮
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shengshi Taike Biopharmaceutical Technology (Suzhou) Co.,Ltd.
Original Assignee
Cgenetech Suzhou China Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cgenetech Suzhou China Co Ltd filed Critical Cgenetech Suzhou China Co Ltd
Priority to CN201910617997.8A priority Critical patent/CN110204537B/en
Publication of CN110204537A publication Critical patent/CN110204537A/en
Application granted granted Critical
Publication of CN110204537B publication Critical patent/CN110204537B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/12Ophthalmic agents for cataracts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/02Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D271/081,2,5-Oxadiazoles; Hydrogenated 1,2,5-oxadiazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Abstract

The invention belongs to the technical field of 1,2, 5-oxadiazole derivatives, and particularly relates to a 1,2, 5-oxadiazole derivative serving as an inhibitor of indoleamine 2, 3-dioxygenase or a pharmaceutically acceptable salt thereof. The 1,2, 5-oxadiazole derivative used as an IDO inhibitor or a pharmaceutically acceptable salt thereof has a structure shown in the following formula I:

Description

1,2, 5-oxadiazole derivatives as inhibitors of indoleamine 2, 3-dioxygenase
Technical Field
The invention belongs to the technical field of 1,2, 5-oxadiazole derivatives, and particularly relates to a 1,2, 5-oxadiazole derivative used as an inhibitor of indoleamine 2, 3-dioxygenase.
Background
Tryptophan (Trp) is an essential amino acid required for the biosynthesis of proteins, nicotinic acid and the neurotransmitter 5-hydroxytryptamine (serotonin). The enzyme indoleamine 2, 3-dioxygenase (also known as INDO or IDO) catalyzes the first and rate-limiting step in the degradation of L-tryptophan to N-formyl-kynurenine. In human cells, Trp depletion by IDO activity is a well-known gamma interferon (IFN- γ) -induced antimicrobial effector mechanism. IFN- γ stimulation induces activation of IDO, which leads to Trp depletion, thus inhibiting the growth of Trp-dependent intracellular pathogens such as Toxoplasma murine (Toxoplasma gondii) and Chlamydia trachomatis (Chlamydia trachomatis).
Research has shown that IDO also plays a role in many processes, such as it may play a role in tumor rejection (Daubener et al, 1999, adv.exp.med.biol., 467: 517-24; Taylor et al, 1991, FASEB j., 5: 2516-22); immunosuppression (Logan et al, 2002, Immunology, 105: 478-87). Several lines of evidence also suggest that IDO plays a role in mammalian pregnancy, tumor resistance (tumor resistance), chronic infections, and autoimmune diseases.
Therefore, effective inhibition of IDO can elevate virus-specific T cell levels in mouse HIV models while reducing the number of virus-infected macrophages (Portula et al, 2005, Blood, 106: 2382-90).
Currently, there has been much development of small molecule inhibitors of IDO for the treatment or prevention of IDO-related diseases (such as those mentioned above). Oxadiazole and other heterocyclic IDO inhibitors are reported, for example, in US 2006/0258719 and US 2007/0185165. Compounds having indoleamine-2, 3-dioxygenase (IDO) inhibitory activity are reported in WO 2004/094409; and U.S. patent application publication No. 2004/0234623 relates to a method of treating a patient with cancer or an infection by administering an inhibitor of indoleamine-2, 3-dioxygenase in combination with other therapeutic modalities.
As another example, Incyte's high throughput screening has resulted in a series of N-hydroxyamidine derivatives and in China has laid out the corresponding patent application CN200980134351.9 which discloses 1,2, 5-dioxazoles of formula I as inhibitors of indoleamine 2, 3-dioxygenase.
Figure GDA0002151341970000011
However, at present, the research and development of indoleamine-2, 3-dioxygenase inhibitors are in the early stage, and the disclosed IDO inhibitors cannot meet the requirements of clinical production by combining various performances such as inhibitory activity, pharmacokinetics and safety.
Therefore, there is an urgent need in the market to develop new structures of inhibitors of IDO enzyme activity.
Disclosure of Invention
The present invention aims to provide a 1,2, 5-oxadiazole derivative useful as an inhibitor of indoleamine 2, 3-dioxygenase having both excellent IDO inhibitory activity and permeability.
In order to achieve the above objects, one of the technical aspects of the present invention provides a 1,2, 5-oxadiazole derivative useful as an IDO inhibitor of the following formula i or a pharmaceutically acceptable salt thereof,
Figure GDA0002151341970000021
wherein R1 and R2 are independently selected from hydrogen, halogen, cyano, substituted or unsubstituted C1-C10 alkyl, substituted or unsubstituted C3-C10 cycloalkyl, substituted or unsubstituted C1-C10 haloalkyl, substituted or unsubstituted C1-C10 hydroxyalkyl, or substituted or unsubstituted C1-C10 alkoxy;
r is-X-Z-A, and X is selected from oxygen atom or methylene;
z is selected from a covalent bond or the following divalent groups: a substituted or unsubstituted C1-C4 alkylene group, an imide group, a substituted or unsubstituted C1-C4 alkyleneoxy group, or a substituted or unsubstituted C1-C4 alkyleneamino group;
a is selected from substituted or unsubstituted C3-C14 heterocycloalkyl, substituted or unsubstituted C3-C14 heteroaryl, or Q-NH (SO)2)NH2Wherein Q is selected from C2-C6 alkylene;
and when Z is methylene, A is selected from substituted or unsubstituted C3-C14 heterocycloalkyl, substituted or unsubstituted C3-C14 heteroaryl.
Preferably, R1 and R2 are independently selected from halogen, cyano, unsubstituted C5-7 cycloalkyl or unsubstituted C1-4 alkoxy;
x is a methylene group, and X is a methylene group,
z is selected from methylene, imide, methyleneoxy, or methyleneamino;
a is selected from the group consisting of substituted or unsubstituted imidazolyl, substituted or unsubstituted thienyl, substituted or unsubstituted pyrazolyl, substituted or unsubstituted pyrazol-4-yl, substituted or unsubstituted 1-methylpyrazol-4-yl, substituted or unsubstituted pyridyl, substituted or unsubstituted pyridonyl, or substituted or unsubstituted cyclobut-3-en-1, 2-dionyl.
Preferably, the "substituted" refers to having one or more substituents selected from the group consisting of:
halogen, hydroxy, cyano, amino, C1-C4 alkyl, - (SO)2)NH2、-(SO2)CH3C1-C4 alkylamino, phenyl, ammoximino, five-to six-membered heterocycle, wherein the heteroatom can be nitrogen, oxygen, sulfur.
Further preferably, R1 is bromo and R2 is fluoro.
Preferably, the 1,2, 5-oxadiazole derivative or a pharmaceutically acceptable salt thereof used as an IDO inhibitor provided by the invention is selected from compounds of the following structures:
Figure GDA0002151341970000031
Figure GDA0002151341970000041
preferably, the present invention provides a 1,2, 5-oxadiazole derivative useful as an IDO inhibitor of formula x below, or a pharmaceutically acceptable salt thereof,
Figure GDA0002151341970000042
r1 and R2 are independently selected from hydrogen, halogen, cyano, substituted or unsubstituted C1-C10 alkyl, substituted or unsubstituted C3-C10 cycloalkyl, substituted or unsubstituted C1-C10 haloalkyl, substituted or unsubstituted C1-C10 hydroxyalkyl, or substituted or unsubstituted C1-C10 alkoxy;
a is selected from substituted or unsubstituted C5-C7 heterocycloalkyl wherein the substituent is- (SO)2)NH2Or is- (SO)2)CH3
Further preferably, said 1,2, 5-oxadiazole derivative of formula x, or a pharmaceutically acceptable salt thereof, useful as an IDO inhibitor is selected from the compounds of the following structures: .
Figure GDA0002151341970000051
Preferably, the present invention provides a 1,2, 5-oxadiazole derivative of formula XI useful as an IDO inhibitor, or a pharmaceutically acceptable salt thereof,
Figure GDA0002151341970000052
Ⅺ(CGT-9018)。
the compounds of the present invention are intended to include all possible geometric isomers. Cis and trans geometric isomers of the compounds of the present invention are described, which may be separated as mixtures of isomers or as individual isomers. The bonds in the structural diagrams represented by wavy lines are intended to indicate that the structure represents the cis or trans isomer, or a mixture of cis and trans isomers in any proportion.
The compounds of the present invention also include tautomeric forms. The tautomeric form results from the exchange of a single bond with an adjacent double bond with concomitant proton migration.
The compounds of the invention may also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same number of atoms but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium.
The invention also includes salts of the compounds described herein. As used herein, "salt" refers to a derivative of the disclosed compound in which the parent compound is modified by converting an acid or base moiety present into its salt form. Examples of salts include, but are not limited to: inorganic acid (such as HCl, HBr, H2SO4) or organic acid (such as acetic acid, benzoic acid, trifluoroacetic acid) salts of bases such as amines; bases (such as Li, Na, K, Mg, Ca) or organic (such as trialkylammonium) salts of acid groups such as carboxylic acids, and the like. The salts of the present invention can be synthesized from the parent compound, which contains a basic or acidic moiety, by conventional chemical methods. Such salts can be prepared, in general, by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent or in a mixture of the two; generally, nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or Acetonitrile (ACN) are preferred.
The "pharmaceutically acceptable salts" of the present invention include a subset of the "salts" described above, which are conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. See the list of suitable salts: remington's Pharmaceutical Sciences, 17 th edition, Mack Publishing Company, Easton, Pa., 1985, p. 1418, and Journal of Pharmaceutical Science, 66, 2(1977), each of which is incorporated herein by reference in its entirety. The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
Composition comprising a metal oxide and a metal oxide
Another embodiment of the present invention provides a pharmaceutical composition comprising one or more 1,2, 5-oxadiazole derivatives or pharmaceutically acceptable salts thereof for use as IDO inhibitors as described above as an active ingredient in combination with at least one pharmaceutically acceptable carrier.
In preparing the compositions of the present invention, the active ingredient is typically mixed with an excipient, diluted with an excipient, or enclosed within a carrier, e.g., in the form of a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it may be a solid, semi-solid, or liquid material that serves as a vehicle, carrier, or medium for the active ingredient. Thus, the composition may be in the form of: tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
In preparing the formulations, the active compound may be comminuted to give the appropriate particle size and then combined with the other ingredients. If the active compound is substantially insoluble, it may be comminuted to a particle size of less than 200 mesh. If the active compound is substantially soluble in water, the particle size may be adjusted by comminution to give a substantially uniform distribution in the formulation, for example about 40 mesh.
Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starch, acacia gum, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulation may further comprise: lubricants, such as talc, magnesium stearate and mineral oil; a humectant; emulsifying and suspending agents; preservatives, such as methyl benzoate and hydroxypropyl benzoate; sweetening agents and flavoring agents. The compositions of the present invention may be formulated so as to provide immediate, sustained or delayed release of the active ingredient after administration to the patient, using methods known in the art.
The compositions may be formulated in unit dosage forms, each dose containing from about 5 to about 100mg, more usually from about 10 to about 30mg, of the active ingredient. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in admixture with suitable pharmaceutical excipients.
The effective dose of the active compound can vary widely and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will generally be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
For preparing solid compositions such as tablets, the principal active ingredient is mixed with pharmaceutical excipients to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention. When referring to these preformulation compositions as homogeneous, the active ingredient is generally uniformly distributed throughout the composition such that the composition may be readily subdivided into equally effective unit dosage forms, such as tablets, pills and capsules. The solid pre-formulation is then subdivided into unit dosage forms of the type described above containing, for example, from 0.1 to about 500mg of the active ingredient of the invention.
The tablets or pills of the present invention may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, a tablet or pill may comprise an inner dosage and an outer dosage component, the latter being in the form of a shell of the former. The two components may be separated by an enteric layer which serves to resist disintegration in the stomach, to allow the inner component to pass intact into the duodenum, or to delay release. A variety of materials may be used for such enteric layers or coatings, such materials including a variety of polymeric acids, and mixtures of polymeric acids with materials such as shellac, cetyl alcohol and cellulose acetate.
Liquid forms in which the compounds and compositions of the present invention may be incorporated for oral or injectable administration include: aqueous solutions, suitably flavored syrups, aqueous or oily suspensions and flavored emulsions with edible oils (e.g., cottonseed oil, sesame oil, coconut oil or peanut oil), as well as elixirs and similar pharmaceutical vehicles.
Compositions for inhalation or insufflation include: solutions and suspensions in pharmaceutically acceptable aqueous or organic solvents or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described above. In certain embodiments, the composition is administered by the oral or nasal respiratory route to achieve a local or systemic effect. The composition may be atomized using an inert gas. The nebulized solution may be inhaled directly from the nebulizing device, or the nebulizing device may be connected to a mask or intermittent positive pressure ventilator. Solutions, suspensions or powder compositions can be administered orally or nasally from a device that delivers the formulation in an appropriate manner.
The amount of compound or composition administered to a patient varies with the following factors: the drug to be administered, the purpose of administration (e.g., prophylaxis or treatment), the condition of the patient, the mode of administration, and the like. In therapeutic applications, the compositions may be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective dosages will depend on the disease state being treated and the judgment of the attending clinician, which will depend on factors such as the severity of the disease, the age, weight and general condition of the patient.
The composition to be administered to the patient may be in the form of a pharmaceutical composition as described above. These compositions may be sterilized by conventional sterilization techniques, or may be filter sterilized. Aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the preparation of the compound is generally from 3 to 11, more preferably from 5 to 9, most preferably from 7 to 8. It will be appreciated that the use of certain of the aforementioned excipients, carriers or stabilizers may result in the formation of a pharmaceutical salt.
The therapeutic dosage of the compounds of the invention may vary depending on the following factors: for example, the particular use of the treatment, the mode of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of a compound of the invention in a pharmaceutical composition may vary depending on a number of factors: including dosage, chemical properties (e.g., hydrophobicity), and route of administration. For example, for parenteral administration, the compounds of the invention may be provided in a physiologically buffered aqueous solution containing from about 0.1 to about 10% w/v of the compound. Some typical dosage ranges are from about 1. mu.g/kg to about 1g/kg body weight/day. In certain embodiments, the dosage range is from about 0.01mg/kg to about 100mg/kg body weight/day. The dosage will likely depend on such variables as the type and extent of progression of the disease or disorder, the general health status of the particular patient, the relative biological potency of the selected compound, the excipient formulation and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
The compounds of the present invention may also be formulated in combination with one or more additional active ingredients, which may include any drug, such as antiviral agents, vaccines, antibodies, immune promoters, immunosuppressive agents, anti-inflammatory agents, and the like.
Use of
Another embodiment of the present invention provides a use of the 1,2, 5-oxadiazole derivative or the pharmaceutically acceptable salt thereof or the pharmaceutical composition containing any of them as described above for the manufacture of a medicament for inhibiting immunosuppression in a patient.
The invention further provides that immunosuppression (e.g., IDO-mediated immunosuppression) in a patient can be effectively inhibited by administering to the patient an effective amount of a compound or composition described herein. IDO-mediated immunosuppression is known to be associated with: for example, cancer, tumor growth, metastasis, viral infection, viral replication, and the like.
The invention further provides methods of treating diseases associated with IDO activity or expression, including aberrant activity and/or overexpression, in an individual (e.g., patient) by administering to the individual in need of such treatment a therapeutically effective amount or dose of a compound of the present invention or a pharmaceutical composition thereof. Examples of diseases may include any disease, disorder or condition that is directly or indirectly associated with IDO enzyme expression or activity (e.g., overexpression or aberrant activity). IDO-related diseases also include any disease, disorder or condition that can be prevented, alleviated or cured by modulating the activity of an enzyme. Examples of IDO-related diseases include: cancer; viral infections, such as HIV infection, HCV infection; depression; neurodegenerative disorders such as alzheimer's disease and huntington's disease; trauma; age-related cataracts; organ transplantation (e.g., organ transplant rejection) and autoimmune diseases, including asthma, rheumatoid arthritis, multiple sclerosis, allergic inflammation, inflammatory bowel disease, psoriasis, and systemic lupus erythematosus. Examples of cancers that can be treated by the methods herein include cancers of the colon, pancreas, breast, prostate, lung, brain, ovary, cervix, testis, kidney, head and neck; lymphoma; leukemia; melanoma, and the like.
Preferably, the medicament is for use in the treatment of cancer, viral infections, depression, neurodegenerative disorders, trauma, age-related cataracts, organ transplant rejection or autoimmune diseases in a patient.
Preferably, the cancer is selected from the group consisting of colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, brain cancer, ovarian cancer, cervical cancer, testicular cancer, kidney cancer, head and neck cancer, lymphatic cancer, and leukemia.
Preferably, the medicament is for treating melanoma in a patient.
Another embodiment of the present invention provides a use of the 1,2, 5-oxadiazole derivative or the pharmaceutically acceptable salt thereof or the pharmaceutical composition containing any of them as an IDO inhibitor for the preparation of a medicament for the treatment of obesity and/or ischemia.
The term "cell" as used herein is intended to mean a cell in vitro, ex vivo or in vivo. In certain embodiments, the ex vivo cell may be a portion of a tissue sample excised from an organism (e.g., a mammal). In certain embodiments, the in vitro cell can be a cell in cell culture. In certain embodiments, an in vivo cell is a cell that lives in an organism (e.g., a mammal).
The term "contacting" as used herein means bringing the specified moieties together in an in vitro system or in an in vivo system. For example, "contacting" an IDO enzyme with a compound of the present invention includes administering a compound of the present invention to an individual or patient (e.g., a human) having IDO, and, for example, introducing a compound of the present invention into a sample containing cells or purified preparations containing IDO enzyme.
The terms "individual" or "patient" are used interchangeably herein to refer to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, pigs, cows, sheep, horses or primates, most preferably humans.
The phrase "therapeutically effective amount" as used herein, refers to an amount of an active compound or pharmaceutical agent that elicits the biological or medical response in a tissue, system, animal, subject, or human that is being sought by a researcher, veterinarian, medical doctor, or other clinician.
The term "treatment" as used herein means: 1) preventing diseases; for example, preventing a disease, condition, or disorder in an individual who is susceptible to the disease, condition, or disorder but has not experienced or exhibited pathology or symptomatology of the disease; 2) inhibiting the disease; for example, inhibiting a disease, condition, or disorder (i.e., arresting the further development of the pathology and/or symptomatology) in an individual who is experiencing or presenting with the pathology or symptomatology of the disease, condition, or disorder; or 3) ameliorating the disease; for example, the disease, condition, or disorder is alleviated (i.e., the pathology and/or symptomatology is reversed) in an individual who is experiencing or presenting with the pathology or symptomatology of the disease, condition, or disorder.
Preferably, the medicament also includes antiviral, chemotherapeutic or other anticancer agents, immune-boosting agents, immunosuppressive agents, radiation, anti-tumor and antiviral vaccines, cytokine therapy (e.g., IL2, GM-CSF, etc.), and/or tyrosine kinase inhibitors. The medicament can be used for treating IDO-related diseases, disorders or conditions. These agents may be combined with the compounds of the present invention in a single dosage form, or the agents may be administered as separate dosage forms, simultaneously or sequentially.
Suitable antiviral agents contemplated for use in combination with the compounds of the present invention may include nucleoside and Nucleotide Reverse Transcriptase Inhibitors (NRTI), non-nucleoside reverse transcriptase inhibitors (NNRTD, protease inhibitors and other antiviral agents.
Examples of suitable NRTIs include: zidovudine (AZT); didanosine (ddl); zalcitabine (ddC); stavudine (d 4T); lamivudine (3 TC); abacavir (1592U 89); adefovir dipivoxil [ di (POM) -PMEA ]; lobbucavir (BMS-180194); BCH-10652; emtricitabine [ (-) -FTC ]; beta-L-FD 4 (also known as beta-L-D4C and named beta-L-2 ', 3' -dieoxy-5-fluoro-cydene); DAPD, ((-) - β -D-2, 6, -diamino-purine dioxolane); and lodenosine (FddA). Typical suitable NNRTIs include nevirapine (BI-RG-587); delavirdine (BHAP, U-90152); efavirenz (DMP-266); PNU-142721; AG-1549; MKC-442(1- (ethoxy-methyl) -5- (1-methylethyl) -6- (phenylmethyl) - (2, 4(1H, 3H) -pyrimidinedione), and (+) -Calophyllum plant extract (calanolide A) (NSC-675451) and B.typical suitable protease inhibitors include saquinavir (Ro 31-8959), ritonavir (ABT-538), indinavir (MK-639), nelfinavir (AG-1343), amprenavir (141W94), lacinavir (BMS-234475), DMP-450, BMS-2322623, ABT-378, and AG-1549. other antiviral agents include hydroxyurea, ribavirin, IL-2, IL-12, pentafuside and YISSum project No. 11607.
Suitable chemotherapeutic or other anti-cancer agents include, for example, alkylating agents (including but not limited to nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas, and triazenes), such as uramustine, nitrogen mustards (chlormethine), cyclophosphamide (cytoxan), ifosfamide, melphalan, chlorambucil, pipobroman, triethylenemelamine, thiotepa, busulfan, carmustine, lomustine, streptozocin, dacarbazine, and temozolomide.
In the treatment of melanoma, agents suitable for use in combination with the compounds of the present invention include: dacarbazine (DTIC), optionally, as well as other chemotherapeutic agents such as carmustine (BCNU) and cisplatin; "Dartmouth protocol" consisting of DTIC, BCNU, cisplatin, and tamoxifen; a combination of cisplatin, vinblastine and DTIC; or temozolomide. In the treatment of melanoma, the compounds according to the invention may also be combined with immunotherapeutic agents including cytokines such as interferon alpha, interleukin 2 and Tumour Necrosis Factor (TNF).
In the treatment of melanoma, the compounds of the present invention may also be used in combination with vaccine therapy. Anti-melanoma vaccines are similar in some respects to antiviral vaccines used to prevent diseases caused by viruses, such as polio, measles and mumps. Attenuated melanoma cells or portions of melanoma cells (referred to as antigens) may be injected into a patient to stimulate the body's immune system, thereby destroying the melanoma cells.
Melanoma restricted to the arms or legs can also be treated with combinations of agents comprising one or more compounds of the present invention using hyperthermic isolated limb perfusion technology. This treatment temporarily isolates the circulation of the relevant limb from the rest of the body and injects high doses of chemotherapeutic agents into the arteries supplying the limb, thereby providing high doses to the tumor area without exposing internal organs to these doses, which could otherwise cause serious side effects. Typically, the fluid is warmed to 102 ° to 104 ° f. Melphalan is the most commonly used drug in this chemotherapy procedure. It can be administered with another agent known as Tumor Necrosis Factor (TNF) (see section for cytokines).
Suitable chemotherapeutic or other anti-cancer agents include, for example: antimetabolites (including but not limited to folate antagonists, pyrimidine analogs, purine analogs, and adenosine deaminase inhibitors), such as methotrexate, 5-fluorouracil, floxuridine, cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, pentostatin, and gemcitabine.
Suitable chemotherapeutic or other anti-cancer agents also include, for example: certain natural products and derivatives thereof (e.g., vinca alkaloids, antitumor antibiotics, enzymes, lymphokines, and epipodophyllotoxins), such as vinblastine, vincristine, vindesine, bleomycin, actinomycin D, daunorubicin, doxorubicin, epirubicin, idarubicin, cytarabine, paclitaxel (TAXOLTM), plicamycin, deoxycoformycin, mitomycin-C, L-asparaginase, interferons (especially IFN-a), etoposide, and teniposide.
Other cytotoxic agents include navelbene (navelbene), CPT-11, anastrozole, letrozole, capecitabine, reloxafine, cyclophosphamide, ifosfamide and droloxifene.
Also suitable are cytotoxic agents, such as: epipodophyllotoxin; an anti-tumor enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes, such as cisplatin and carboplatin; a biological response modifier; a growth inhibitor; an anti-hormone therapeutic agent; folinic acid; tegafur; and hematopoietic growth factors.
Other anti-cancer agents include antibody therapeutics such as antibodies to trastuzumab (Herceptin), costimulatory molecules such as CTLA-4, 4-1BB and PD-1, or cytokines such as IL-10, TGF- β, etc.
Other anti-cancer agents also include those that block immune cell migration, such as antagonists of chemokine receptors, including CCR2 and CCR 4.
Other anti-cancer agents also include those that enhance the immune system, such as adjuvants or adoptive T cell transfer.
Anti-cancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses.
Methods for safe and effective administration of most such chemotherapeutic agents are known to those skilled in the art. In addition, their administration is described in the standard literature. For example, in the "Physicians' Desk Reference" (PDR, e.g., 1996edition, Medical Economics Company, Montvale, NJ), the administration of a variety of chemotherapeutic agents is described, the disclosure of which is incorporated herein by Reference as if fully set forth.
Interpretation of terms
Unless stated to the contrary, the following terms used in the specification and claims have the following meanings.
The term "alkyl" refers to a saturated aliphatic hydrocarbon group, including straight and branched chain groups of 1 to 20 carbon atoms. Preferably an alkyl group containing 1 to 10 carbon atoms, more preferably an alkyl group containing 1 to 6 carbon atoms, most preferably an alkyl group containing 1 to 4 carbon atoms, most preferably a methyl group. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2, 3-dimethylpentyl, 2, 4-dimethylpentyl, 2-dimethylpentyl, 3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2, 3-dimethylhexyl, 2, 4-dimethylhexyl, 2, 5-dimethylhexyl, 2-dimethylhexyl, 3-dimethylhexyl, 4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2-dimethylpentyl, 2-dimethylhexyl, 3-dimethylpentyl, 2-ethylhexyl, 3-dimethylhexyl, 2-ethylhexyl, 2-dimethylhexyl, 2-ethylhexyl, 2-dimethylhexyl, 2-dimethylhexyl, 2-dimethylhexyl, 2-ethylhexyl, 2-ethyl, 2-2, 2-2, 2-2, or, 2, 2-diethylpentyl, n-decyl, 3-diethylhexyl, 2-diethylhexyl, and various branched isomers thereof. More preferred are lower alkyl groups having 1 to 6 carbon atoms, non-limiting examples of which include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl and the like. The alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halo, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate.
The term "cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms, more preferably the cycloalkyl ring comprises 3 to 10 carbon atoms, most preferably the cycloalkyl ring comprises 3 to 6 carbon atoms, most preferably cyclopropyl or cyclopentyl. Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl and the like, with cyclopropyl, cyclopentyl being preferred. Polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups. Cycloalkyl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate.
The term "heterocycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising 3 to 20 ring atoms, one or more of which is selected from nitrogen, oxygen, or a heteroatom of S (O) m (where m is an integer from 0 to 2), but does not include the ring portion of-O-O-, -O-S-, or-S-S-, the remaining ring atoms being carbon. Preferably 3 to 12 ring atoms of which 1 to 4 are heteroatoms, more preferably a heterocycloalkyl ring comprising 3 to 10 ring atoms, and even more preferably a heterocycloalkyl ring comprising 5 to 6 ring atoms. Non-limiting examples of monocyclic heterocycloalkyl include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl, tetrahydrofuranyl and the like. Polycyclic heterocycloalkyl groups include spiro, fused and bridged heterocycloalkyl groups. The heterocycloalkyl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, amino, haloalkyl, hydroxyalkyl, carboxyl, or carboxylate.
The term "aryl" refers to a 6 to 14 membered all carbon monocyclic or fused polycyclic (i.e., rings which share adjacent pairs of carbon atoms) group having a conjugated pi-electron system, preferably 6 to 10 membered, more preferably phenyl and naphthyl, most preferably phenyl. The aryl ring may be fused to a heteroaryl, heterocycloalkyl, or cycloalkyl ring, where the ring that is attached to the parent structure is an aryl ring.
Heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate.
The term "alkoxy" refers to-O- (alkyl) and-O- (unsubstituted cycloalkyl), wherein alkyl and cycloalkyl are as defined above. Non-limiting examples include methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy, and the like. Alkoxy groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, amino, haloalkyl, hydroxyalkyl, carboxy or carboxylate.
"haloalkyl" means an alkyl group substituted with one or more halogens wherein the alkyl group is as defined above. "hydroxy" refers to an-OH group. "hydroxyalkyl" refers to an alkyl group substituted with a hydroxy group, wherein alkyl is as defined above. "halogen" means fluorine, chlorine, bromine or iodine, preferably fluorine or iodine.
"amino" refers to-NH 2. "cyano" means-CN. "nitro" means-NO 2. "oxo" refers to ═ O. "carboxy" refers to-C (O) OH. "carboxylate" refers to-C (O) O (alkyl) or (cycloalkyl), wherein alkyl and cycloalkyl are as defined above.
"optional" or "optionally" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs or does not. For example, "a heterocycloalkyl group optionally substituted with an alkyl group" means that the alkyl group may, but need not, be present, and the description includes the case where the heterocycloalkyl group is substituted with an alkyl group and the case where the heterocycloalkyl group is not substituted with an alkyl group.
"substituted" means that one or more, preferably up to 5, more preferably 1 to 3, hydrogen atoms in a group are independently substituted with a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (experimentally or theoretically) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable in combination with carbon atoms having unsaturated (e.g., olefinic) bonds.
Synthesis method
The compounds of the present invention can be prepared by a variety of methods known to those skilled in the art of organic synthesis. The compounds of the present invention may be synthesized using the methods described hereinafter, as well as synthetic methods known in the art of synthetic organic chemistry or modifications thereof as recognized by those skilled in the art.
The compounds of the present invention can be prepared from readily available starting materials using the following general methods and procedures. It is to be understood that where typical or preferred process conditions are given (i.e., reaction temperature, time, molar ratios of reactants, solvents, pressures, etc.); other process conditions may also be used unless otherwise specified. Optimum reaction conditions may vary with the particular reactants or solvents used, but such conditions can be determined by one skilled in the art by routine optimization methods.
The process described herein may be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectrophotometry, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C), infrared spectroscopy, spectrophotometry (e.g., ultraviolet-visible), or mass spectrometry, or by chromatography, such as High Performance Liquid Chromatography (HPLC) or thin layer chromatography. The compound obtained by the reaction may be purified by any suitable method known in the art. For example, chromatography (medium pressure), HPLC or preparative thin layer chromatography, distillation, sublimation, trituration or recrystallization on a suitable adsorbent (e.g., silica gel, alumina, etc.).
The preparation of the compounds may involve protection and deprotection of different chemical groups. The need for protection and deprotection, and the choice of suitable protecting groups, can be readily determined by those skilled in the art. Protecting group chemistry can be found, for example, in Wuts and Greene, Greene's Protective Groups in Organic Synthesis, 4 th edition, John Wiley & Sons: new York, 2006, which is incorporated herein by reference in its entirety.
The reactions of the methods described herein can be carried out in a suitable solvent, which can be readily selected by one skilled in the art of organic synthesis. Suitable solvents are substantially unreactive with the starting materials (reactants), intermediates, or products at the temperatures at which the reaction is carried out, i.e., at temperatures in the range from the freezing temperature of the solvent to the boiling temperature of the solvent. The specific reaction may be carried out in one solvent or a mixture of more than one solvent. Depending on the reaction step, a solvent suitable for the particular reaction step may be selected. Suitable solvents include water, alkanes (such as pentanes, hexanes, heptanes, cyclohexane, and the like, or mixtures thereof), aromatic solvents (such as benzene, toluene, xylene, and the like), alcohols (such as methanol, ethanol, isopropanol, and the like), ethers (such as dialkyl ethers, methyl tert-butyl ether (MTBE), Tetrahydrofuran (THF), esters (such as ethyl acetate, butyl acetate, and the like), halogenated solvents (such as Dichloromethane (DCM), chloroform, dichloroethane, tetrachloroethane), Dimethylformamide (DMF), dimethyl sulfoxide (DMSO), acetone, Acetonitrile (ACN), Hexamethylphosphoramide (HMPA), and N-methylpyrrolidinone (NMP)), such solvents can be used in their aqueous or anhydrous forms.
Racemic mixtures of the compounds can be resolved by any of a variety of methods known in the art. One example method includes fractional recrystallization, wherein a "chiral resolving acid" is used, which is an optically active salt-forming organic acid. Resolving agents suitable for use in fractional recrystallization processes are, for example: optically active acids such as tartaric acid in the D and L forms, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or different optically active camphorsulfonic acids. The racemic mixture can also be resolved by elution on a column packed with an optically active resolving agent, for example dinitrobenzoylphenylglycine. One skilled in the art can determine suitable elution solvent compositions.
For example, using the following reaction routes and techniques, a general synthetic route to compounds of the invention of formula I is shown below:
Figure GDA0002151341970000161
among them, compound I-a is a known structure, the synthesis of which has been reported in the literature (WO2014066834, CN 106565696). I-A and I-B react to generate I-C, and corresponding ammoxim compounds I-D are generated under the alkaline condition; compounds I-B sometimes have protecting groups, in which case the protecting group must eventually be removed to form I-D.
Amino protecting groups are often used in organic synthesis to prevent unwanted amino reactions while performing the desired transformations. The amino protecting group allows for easy covalent attachment to, and selective cleavage from, a nitrogen atom. Different amino protecting groups are known to those skilled in the art and are broadly classified as alkoxycarbonyl groups (such as ethoxycarbonyl, tert-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), 9-fluorenylmethoxycarbonyl (Fmoc) and the like), acyl groups (such as acetyl (Ac), benzoyl (Bz) and the like), sulfonyl groups (such as methanesulfonyl, trifluoromethanesulfonyl and the like), aralkyl groups (such as benzyl, diphenylmethyl, triphenylmethyl (trityl) and the like), alkenylalkyl groups (such as allyl, isoprenyl and the like), diarylmethylene (diarylmethyl) (such as (C6H5)2C ═ N and the like) and silyl groups (such as tert-butyldimethylsilyl, triisopropylsilyl and the like). The chemistry of the amino protecting group can be found in: wuts and Greene, Greene's Protective Groups in Organic Synthesis, 4 th edition, page 696-: new York, 2006.
Compared with the prior art, the invention has the beneficial effects that:
provides a compound I with a novel structure and a general formula;
experimental results show that some compounds in the examples provided by the invention have excellent IDO inhibitory activity and permeability. The compound provided by the embodiment of the invention is expected to be used as a tumor molecular immunotherapy medicament for treating cancers.
Detailed Description
The present invention is further illustrated in more detail by the following specific examples. The following examples are provided for illustrative purposes and should not be construed as limiting the invention in any way. One skilled in the art will readily recognize that various noncritical parameters may be changed or modified to achieve substantially the same results. The example compounds were found to be IDO inhibitors according to one or more of the assays provided herein.
EXAMPLE 1 Synthesis of CGT-9009
Figure GDA0002151341970000171
Step 1 Synthesis of MC-105-2
LiAlH is reacted at room temperature4(1.2g, 31.6mmol, 1.58eq) was added portionwise to a solution of compound MC-105-1(2.84g, 20mmol, 1.0eq) in THF (20 mL). The reaction was stirred at room temperature for 20 minutes. The reaction was quenched with a saturated solution of ammonium chloride (2mL) and stirred for 10 min. The resulting mixture was filtered and the filtrate was concentrated to give the crude compound MC-105-2(2.5g, 97% yield) as a brown oil.
Step 2 Synthesis of MC-105-3
MsCl (2mL) was slowly added dropwise to a solution of compound MC-105-2(2.5g,19.5mmol,1.0eq) and triethylamine (2.5mL) in dichloromethane (25mL) at room temperature. The reaction was quenched with a saturated solution of sodium bicarbonate (30mL) after half an hour at room temperature. The quenched reaction was stirred at room temperature for 1 hour and washed with saturated sodium chloride (50mL X2). The organic phase was dried and concentrated to a crude brown oily intermediate (3.5g, 87% yield.) the crude intermediate (1.04g,5.0mmol,1.0eq) was dissolved directly in DMSO (10mL) and then sodium azide (1g) was added. The reaction mixture was stirred at 80 ℃ for 1 hour. The reaction was then cooled to room temperature and diluted with ethyl acetate (50 mL). The resulting white mixture was washed with saturated sodium chloride solution (50mL X3). The organic phase was concentrated by drying. The residue was purified by silica gel chromatography (EtOAc/PE ═ 1/20) to give the compound MC-105-3(320mg, 42% yield) as a brown oil.
Step 3 Synthesis of MC-105-4
The compound MC-105-3(320mg,2.0mmol) and Pd/C (50mL) were mixed into MeOH (10mL) at room temperature. Then, the reaction solution was reacted under a hydrogen atmosphere for 16 hours. After the reaction, the reaction mixture was filtered, and the filtrate was concentrated to give MC-105-4(260mg, 100% yield) as a brown oily compound.
Step 4 Synthesis of MC-105-5
The reaction was completed after the compound MC-105-4(254mg,2.0mmol,1.86eq) and the compound MC-101-007(400mg,1.07mmol,1.0eq) were reacted in THF (5mL) at 35 ℃ for 10 minutes. The reaction solution was directly concentrated. The residue was purified by silica gel chromatography (EtOAc/PE ═ 1/3) to give the compound MC-105-5(450mg, 93% yield) as a brown oil.
Step 5 Synthesis of CGT-9009
Figure GDA0002151341970000181
Sodium hydroxide (56mg,1.44mmol,8.0eq) was added to a solution of compound MC-105-5(80mg,0.18mmol,1.0eq) in tetrahydrofuran (3mL) and water (1mL) at room temperature. The reaction was carried out at room temperature for 1 hour. After completion of the reaction, the reaction mixture was diluted with 25mL of water, and the resulting mixture was extracted with ethyl acetate (25mL of X3). The organic phase was dried and the residue was purified by HPLC to give CGT-9009(20mg, 27% yield) as a white solid
1H NMR(400MHz,DMSO-d6)δ11.45(s,1H),8.89(s,1H),7.36-7.34(d,J=4.8Hz,1H),7.19(t,J=8.8Hz,1H),7.10(d,J=6.4Hz,1H),6.98-6.96(m,1H),6.91(s,1H),6.76(t,J=4.4Hz,1H),6.34-6.31(m,1H),3.50-3.45(m,2H),3.10(d,J=6.8Hz,2H).
19F NMR(376.5MHz,DMSO-d6)δ-117.798
HPLC:@254nm:99.61%,@214nm:99.76%.
LCMS:M+1=428.0
Example 2 Synthesis of CGT-9016
Figure GDA0002151341970000191
Step 1 Synthesis of MC-113-1
Tert-butanol (451mg,6.09mmol,10.50eq) was added to a solution of chlorosulfonic isocyanate (818mg,5.8mmol,10.0eq) in dichloromethane (5mL) at 0 deg.C and reacted under these conditions for 1 hour. This solution was then added to a solution of compound MC-149-3(250mg,0.58mmol,1.0eq) and triethylamine (2mL) in dichloromethane (10mL) which had been cooled to 0 ℃ beforehand. The reaction mixture was reacted at 0 ℃ for 2 hours and then quenched with saturated sodium bicarbonate (20 mL). The resulting mixture was extracted with dichloromethane (20 mL). The organic phase was dried and concentrated to give MC-113-1 as a yellow solid (500mg, extra heavy).
Step 2 of synthesizing CGT-9016
A solution of compound MC-113-1(500mg,0.58mmol,1.0eq) in TFA (2mL) and dichloromethane (5mL) was reacted at 25 ℃ for 1 hour and concentrated to give the intermediate. The intermediate was dissolved in tetrahydrofuran (5mL) and 2N sodium hydroxide solution (2mL) was added. The reaction mixture was reacted at room temperature for 1 hour. After the reaction was complete the reaction mixture was diluted with water. The resulting mixture was extracted with ethyl acetate (20mLX 2). The organic phase was concentrated by drying. The residue was purified by HPLC to give CGT-9016(8mg, 2%) as a white solid.
LCMS:M+1=484.0
1H NMR(400MHz,DMSO-d6)δ8.48(s,0.45H),7.13-7.11(m,1H),7.06-7.02(m,1H),6.86-6.82(m,1H),3.71-3.62(m,6H),3.48-3.46(m,2H),3.28-3.19(m,2H).
19F NMR(376.5MHz,DMSO-d6)δ-117.84.
EXAMPLE 3 Synthesis of CGT-9019
Step 1 Synthesis of MC-154-3
Compound MC-154-1(372mg,1mmol,1.0eq) was dissolved in THF (4mL) at room temperature. Cesium carbonate (656mg,2mmol,2.0eq), MC-154-2(217mg,1mmol,1.0eq) and water (2mL) were then added to the solution. The reaction mixture was reacted at 20 ℃ for 0.5 hour. The reaction was then diluted with water (10 mL). The resulting mixture was extracted with ethyl acetate (20mL X2). The organic phase was concentrated by drying. The residue was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/5-1/3) to give MC-154-3(300mg, 55%) as a pale brown solid.
Figure GDA0002151341970000201
Step 2 Synthesis of MC-154-4
A solution of compound MC-154-3(300mg,0.55mmol,1.0eq) and TFA (2mL) in dichloromethane (5mL) was reacted at 30 ℃ for 1 hour. The reaction mixture was then concentrated to give MC-154-4(310mg) as a pale brown oil.
Step 3 Synthesis of MC-154-5
Tert-butanol (428mg,5.78mmol,10.50eq) was added to a solution of chlorosulfonic isocyanate (776mg,5.5mmol,10.0eq) in dichloromethane (5mL) at 0 deg.C and reacted under these conditions for 1 hour. This solution was then added to a solution of compound MC-154-4(310mg,0.55mmol,1.0eq) and triethylamine (1mL) in dichloromethane (5mL) which had been cooled to 0 ℃ beforehand. The reaction mixture was reacted at 0 ℃ for 2 hours and then quenched with saturated sodium bicarbonate (20 mL). The resulting mixture was extracted with dichloromethane (20 mL). The organic phase was dried and concentrated to give MC-154-5(350mg, 79%) as a yellow solid.
Step 4 of synthesizing CGT-9019
A solution of compound MC-154-5(250mg,0.48mmol,1.0eq) in TFA (2mL) and dichloromethane (5mL) was reacted at 25 ℃ for 1 hour and concentrated to give the intermediate. The intermediate was dissolved in tetrahydrofuran (5mL) and 2N sodium hydroxide solution (2mL) was added. The reaction mixture was reacted at room temperature for 1 hour. After the reaction was complete the reaction mixture was diluted with water. The resulting mixture was extracted with ethyl acetate (20mLX 2). The organic phase was concentrated by drying. The residue was purified by HPLC to give CGT-9019(2.9mg, 2%) as a white solid.
LCMS:M+1=497.0
1H NMR(400MHz,DMSO-d6)δ11.62(s,1H),8.86(s,1H),8.17-8.14(m,1H),7.20-7.11(m,2H),6.83-6.81(m,1H),6.58-6.46(m,4H),3.88-3.86(m,2H),3.27-3.22(m,2H),2.97-2.93(m,2H).
19F NMR(376.5MHz,DMSO-d6)δ-118.03.
EXAMPLE 4 Synthesis of CGT-9020
Figure GDA0002151341970000211
Step 1 Synthesis of MC-159-3
Sodium hydride (123mg,3.08mmol,1.05eq) was added to a solution of compound MC-159-1(210mg,3.08mmol,1.05eq) in tetrahydrofuran (3mL) at 0 ℃. The mixture was stirred at room temperature for 0.5 hour, and then a solution of compound MC-159-2(702mg,2.94mmol,1.0eq) in tetrahydrofuran (1mL) was added at 0 ℃. After the addition was completed, the reaction solution was stirred at room temperature for 16 hours, and then quenched with water (20 mL). The compound was extracted with ethyl acetate (20mL X3), and the organic phase was washed with water (20mL X2) and saturated aqueous sodium chloride (20mL), then dried and filtered, and the filtrate was concentrated under reduced pressure to give the desired product MC-159-3(550mg) as a brown oil.
Step 2 Synthesis of MC-159-4
Trifluoroacetic acid (6mL) was added to a solution of the compound MC-159-3(550mg) in 1, 2-dichloroethane (20mL) at room temperature, and the reaction mixture was stirred at room temperature for 2 hours and then concentrated under reduced pressure to give the desired product MC-159-4(720mg) as a brown oil.
Step 3 Synthesis of MC-159-5
Cesium carbonate (3.0g,9.21mmol,6.5eq) was added to a mixed solution of the compound MC-159-4(720mg,3.08mmol,2.2eq) and the compound MC-101-007(530mg,1.42mmol,1.0eq) in water (2mL) and tetrahydrofuran (20mL) at room temperature, and the reaction was stirred at room temperature for 16 hours. After completion of the reaction, the reaction was quenched with water (30mL), the compound was extracted with ethyl acetate (30mL X2), the organic phase was washed with water (20mL) and saturated aqueous sodium chloride (20mL), then dried and filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent gradient: EA/PE. RTM. 1/2) to give MC-159-005(230mg, 37% yield) as a yellow solid.
Step 4 of synthesizing CGT-9020
Potassium carbonate (218mg) was added to a solution of the compound MC-159-5(230mg,0.526mmol,1.0eq) in methanol (2.5mL) at room temperature, and the reaction mixture was heated to 40 ℃ for reaction for 2 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, diluted with ethyl acetate (10mL), stirred, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent gradient: MeOH/DCM ═ 1/100) to give the desired compound CGT-9020(150mg, 70% yield) as a yellow solid.
LCMS:M+1=412.0
1H NMR(400MHz,CDCl3)δ10.45(br,1H),7.51(br,1H),7.36(d,J=2.0Hz,1H),7.19(dd,J=5.6Hz,2.4Hz,1H),7.03(t,J=8.4Hz,1H),6.93-6.89(m,2H),6.30(s,1H),5.96(t,J=6.0Hz,1H),4.41(t,J=5.6Hz,2H),3.78(dd,J=11.2Hz,6.0Hz,2H).
EXAMPLE 5 Synthesis of CGT-9021
Figure GDA0002151341970000221
Step 1 Synthesis of MC-164A-1
Compound SM1(1.0g,10.52mmol,1.0eq.) was dissolved in DMF (50mL), cooled to 0 degrees for half an hour, and a solution of SM2(2.8g,11.5mmol,1.1eq.) in DMF (10mL) was added dropwise at 0 degrees. The mixture was stirred at room temperature for 16 hours. The reaction was checked by LCMS, and after completion of the reaction, the reaction solution was diluted with water, extracted with ethyl acetate, and the organic phase was concentrated by drying to obtain a residue, which was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/4-pure ethyl acetate) to obtain a white solid compound MC-164A-1(1.4g, 55%) and a compound MC164B-1(400mg, 15%).
Step 2 Synthesis of MC-164A-2
To a solution of compound MC-164A-2(500mg,2.10mmol,1.0eq.) in dichloromethane (5mL) was added 2mL of trifluoroacetic acid at room temperature. The reaction solution was reacted at room temperature for 2 hours. After the completion of the reaction, the reaction mixture was directly concentrated to give the crude compound MC-164A-2(260mg, trifluoroacetate salt) as a pale yellow oil.
Step 3 Synthesis of MC-164A-3
The compounds MC-101-007(260mg,0.70mmol,1.0eq.), MC-164A-2(260mg,1.88mmol,2.7eq.) and triethylamine (425mg,4.20,6.0eq.) were dissolved in tetrahydrofuran (20 mL). After 4 hours at 40 ℃ reaction time, the reaction was checked by LCMS, and after completion of the reaction, the residue obtained by concentrating the reaction mixture was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/1) to obtain MC-164A-3(200mg, 61%) as a pale yellow solid.
Step 4 of synthesizing CGT-9021
Compound MC-164A-3(200mg,0.43mmol,1.0eq) was dissolved in tetrahydrofuran (10mL) at room temperature with the addition of 5mL sodium hydroxide (1N) solution. The reaction was carried out at room temperature for 1 hour. After the reaction was completed, the reaction solution was diluted with water, and the resulting mixture was extracted with ethyl acetate. The organic phase was dried and concentrated to give a pale yellow oil, and the residue was purified by high performance preparative chromatography to give the pale yellow solid compound CGT-9021(72mg, 38% yield).
1H NMR(400MHz,DMSO-d6):δ11.41(s,1H),8.87(s,1H),7.51-7.49(dd,J=1.6Hz,1H),7.41-7.37(m,1H),7.20-7.16(t,J=8.8Hz,1H),7.11-7.10(dd,J=2.8Hz,1H),6.76-6.67(m,1H),6.39-6.35(m,2H),6.18-6.15(m,1H),4.11-4.08(m,2H),3.55-3.52(m,2H).
19F NMR(376.5MHz,DMSO-d6)δ-117.82,-118.39
HPLC:@254nm:99.89%,@214nm:99.75%.
LCMS:M+1=439.2
EXAMPLE 6 Synthesis of CGT-9022
Figure GDA0002151341970000231
Step 1 Synthesis of MC-164B-2
To a solution of compound MC-164B-1(400mg,1.68mmol,1.0eq.) in dichloromethane (5mL) was added 2mL of trifluoroacetic acid at room temperature. The reaction solution was reacted at room temperature for 2 hours. After the reaction, the reaction mixture was directly concentrated to give a pale yellow oily crude compound MC-164B-2(200mg, trifluoroacetate salt).
Step 2 Synthesis of MC-164B-3
The compounds MC-101-007(200mg,0.54mmol,1.0eq.), MC-164A-2(200mg,1.45mmol,2.7eq.) and triethylamine (328mg,3.24,6.0eq.) were dissolved in tetrahydrofuran (20 mL). After 4 hours at 40 ℃ reaction time, the reaction was checked by LCMS, and after completion of the reaction, the residue obtained by concentrating the reaction mixture was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/5) to obtain MC-164B-3(200mg, 80%) as a pale yellow solid.
Step 3 of synthesizing CGT-9022
Compound MC-164B-3(200mg,0.43mmol,1.0eq) was dissolved in tetrahydrofuran (10mL) at room temperature with the addition of 5mL sodium hydroxide (1N) solution. The reaction was carried out at room temperature for 1 hour. After the reaction was completed, the reaction solution was diluted with water, and the resulting mixture was extracted with ethyl acetate. The organic phase was dried and concentrated to give a pale yellow oil, and the residue was purified by high performance preparative chromatography to give the pale yellow solid compound CGT-9022(35mg, 18% yield).
1H NMR(400MHz,DMSO-d6):δ11.48(s,1H),8.87(s,1H),8.17-8.16(d,J=3.2Hz,1H),7.77-7.70(m,1H),7.18-7.09(m,2H),7.00-6.97(t,J=6Hz,1H),6.84-6.75(m,2H),6.40-6.37(t,t=6Hz,2H),4.46-4.43(m,2H),3.63-3.59(m,2H).
19F NMR(376.5MHz,DMSO-d6)δ-117.80,-118.35
HPLC:@254nm:99.60%,@214nm:99.86%.
LCMS:M+1=437.1
Example 7 Synthesis of CGT-9023
Figure GDA0002151341970000241
Step 1 Synthesis of MC-160-3
The compound MC-160-2(2.6g,14.98mmol,1.0eq) was dissolved in N, N-dimethylformamide (20mL) at room temperature and a mixture of diisopropylethylamine (5.8g,44.91mmol,3.0eq) was added and the mixture was stirred for 3 minutes, after which HATU (6.8g,17.96mmol,1.2eq) and the compound MC-160-1(2.0g,14.96mmol,1.0eq) were added to the system. The reaction mixture was reacted at room temperature for 2 hours, then diluted with ethyl acetate (200mL), washed with saturated aqueous sodium chloride (200mL X6), dried and concentrated, and the residue was purified by silica gel column chromatography (eluent gradient: MeOH/DCM ═ 1/100) to give the objective compound MC-160-3(1.3g, 34% yield) as a purple solid
Step 2 Synthesis of MC-160-4
Trifluoroacetic acid (2.5mL) was added to a solution of compound MC-160-3(250mg) in dichloromethane (2.5mL) at room temperature and stirred for 2 h. After completion of the reaction, the reaction mixture was concentrated under reduced pressure to give MC-160-4(280mg) as a brown oily target compound.
Step 3 Synthesis of MC-160-5
To a mixed solution of the compound MC-160-4(280mg,0.984mmol,1.0eq) and the compound MC-101-007(366mg,0.984mmol,1.0eq) in water (1mL) and tetrahydrofuran (3mL) was added cesium carbonate (962mg,2.952mmol,3.0eq) at room temperature and the reaction was stirred for 16 hours. The reaction was quenched with water (15mL) and extracted with ethyl acetate (15mL X2), the organic phase was washed with water (15mL) and saturated aqueous sodium chloride (15mL), then dried and filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent gradient: EA/PE ═ 1/2) to give MC-160-005(145mg, 31% yield) as a yellow solid.
Step 4 of synthesizing CGT-9023
Potassium carbonate (150mg) was added to a solution of the compound MC-160-5(145mg,0.30mmol,1.0eq) in methanol (2mL) at room temperature, followed by heating to 40 ℃ for 2 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was washed with ethyl acetate EA (10 mL). The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent gradient: MeOH/DCM ═ 1/100) to give the desired compound CGT-9023(25mg, 18% yield) as a yellow solid.
LCMS:M-1=453.1
1H NMR(400MHz,CDCl3)δ11.64(s,1H),10.20(brs,1H),8.90(s,1H),7.86(s,1H),7.46(s,1H),7.20(t,J=8.8Hz,1H),7.14(dd,J=6.0Hz,2.8Hz,1H),6.85-6.81(m,1H),6.55(t,J=5.6Hz,1H),4.04(d,J=5.2Hz,2H),3.79(s,3H).
Example 8 Synthesis of CGT-9024
Figure GDA0002151341970000261
Step 1 Synthesis of MC-160-6
A borane-tetrahydrofuran (1N,2.5mL) solution was added to a solution of the compound MC-160-3(250mg,0.984mmol,1.0eq) in tetrahydrofuran (5mL) at room temperature and stirred for 3 hours. After completion of the reaction, concentrated hydrochloric acid (36%, 5mL) was added to the reaction mixture at 0 ℃ and the reaction was stirred for 1 hour. After completion of the reaction, the reaction mixture was concentrated under reduced pressure to give MC-160-6(250mg) as a brown solid.
Step 2 Synthesis of MC-160-7
Cesium carbonate (960mg,2.952mmol,3.0eq) was added to a mixed solution of the compound MC-160-6(250mg,0.984mmol,1.0eq) and the compound MC-101-007(366mg,0.984mmol,1.0eq) in water (1mL) and tetrahydrofuran (5mL) at room temperature, and the reaction was stirred at room temperature for 16 hours. After completion of the reaction, the reaction was quenched with water (15mL), the compound was extracted with ethyl acetate (15mL X2), the organic phase was washed with water (10mL) and saturated aqueous sodium chloride (10mL), then dried and filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent gradient: EA/PE ═ 1/2) to give the desired compound MC-160-7(120mg, 26% yield) as a yellow solid.
Step 3 of synthesizing CGT-9024
Potassium carbonate (120mg) was added to a solution of the compound MC-160-7(120mg,0.258mmol,1.0eq) in methanol (2mL) at room temperature, and the reaction solution was heated to 40 ℃ for reaction for 2 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, diluted with ethyl acetate (10mL), stirred, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent gradient: MeOH/DCM ═ 1/100) to give the desired compound CGT-9024(25mg, 22% yield) as a yellow solid.
LCMS:M+1=441.2
1H NMR(400MHz,CDCl3)δ11.52(s,1H),8.91(s,1H),7.59(br,1H),7.33(br,1H),7.11(dd,J=6.0Hz,2.4Hz,1H),6.80-6.75(m,1H),6.33(t,J=5.6Hz,1H),3.78(s,3H),3.49-3.44(m,2H),3.26(br,2H).
Example 9 Synthesis of CGT-9026
Figure GDA0002151341970000271
Step 1 Synthesis of MC-157-2
SM (0.9g,8.03mmol,1.0eq) was added to SOCl2(3.82g,32.11mmol) and stirred at 70 ℃ for 30min, the reaction was concentrated and spin dried to give MC18-157-2(1.0g, yield 95%).
Step 2 Synthesis of MC-157-4
MC-157-2(1.0g,7.66mmol,1.0eq), MC-157-3(1.56g,8.42mmol,1.1eq) and sodium bicarbonate (1.4g,16.67mmol) were added to DMF (30mL) and stirred at 100 deg.C overnight. The reaction mixture was diluted with 300mL of ethyl acetate, washed with 100mL of saturated brine, and dried over anhydrous sodium sulfate to give a solid, which was purified by column (petroleum ether: ethyl acetate: 1: 0 to 10:1) to give MC-157-4(1.1g,4.56mmol, yield 59%) as a yellow solid.
Step 3 Synthesis of MC-157-5
MC-157-4(300mg,1.24mmol,1.0eq) was added to HCl (6N,30mL) and stirred at 110 deg.C overnight. The reaction solution was spin-dried to give MC-157-5(300mg, crude, 11.24mmol) as a white solid.
Step 4 Synthesis of MC-157-7
MC-157-5(300mg,1.24mmol, crude), MC-157-6(200mg,0.54mmol) and Cs2CO3(703mg,2.15mmol) were added to THF (30mL) and stirred overnight at room temperature the reaction was filtered, the filtrate was dried and purified over a column (petroleum ether: ethyl acetate ═ 20:1 to 1:1) to give MC-157-7(100mg,0.229mmol, yield 42%) as a white solid.
Step 5 of synthesizing CGT-9026
MC-157-7(100mg,0.229mmol,1.0eq) was added to NaOH (1N1mL) and stirred for one hour at ambient temperature. The reaction was extracted with ethyl acetate (20 mL. multidot.2), dried and then purified on a preparative plate (EA) to give CGT-9026(77.6mg,0.189mmol, yield 82%) as a white solid.
LCMS:M+1=412.1
1H NMR(400MHz,DMSO-d6)δ11.47(s,1H),8.88(s,1H),7.47(s,2H),7.20-7.16(m,1H),7.12-7.10(m,1H),6.77-6.73(m,1H),6.23-6.20(t,J=12HZ,1H),3.21-3.20(m,2H),2.75-2.71(t,J=16HZ,2H)。
EXAMPLE 10 Synthesis of CGT-9027
Figure GDA0002151341970000281
Step 1 Synthesis of MC-158-1
MC-157-4(200mg,0.83mmol,1.0eq) was dissolved in 10mL tetrahydrofuran and sodium hydride (20mg) was added at 0 deg.C. Methyl iodide (59mg,1.66mmol,2.0eq) was then added and the reaction was allowed to proceed for one hour at ambient temperature. The reaction mixture was added with 25mL of water, and extracted three times with 35mL of ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and then spin-dried to give MC-158-1(150mg, 49%) as a yellow solid.
Step 2 Synthesis of MC-158-2
MC-158-1(150mg,0.59mmol,1.0eq) was added to 25mL of hydrochloric acid (6M) and the reaction stirred at 110 ℃ for 16 h. The reaction was directly spin-dried to give MC-158-2(150mg, crop) as a yellow solid.
Step 3 Synthesis of MC-158-3
SM1(200mg,0.53mmol,1.0eq), MC-158-2(150mg, loud), and cesium carbonate (1.0g,3.07mmol,6.0eq) were dissolved in 15mL of a mixed solvent of tetrahydrofuran and water at a ratio of 5:1, and the reaction was stirred at room temperature for 30 minutes. 50mL of water was added, and the mixture was extracted three times with 25mL of ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate to give the crude product, which was purified using a preparative plate (petroleum ether: ethyl acetate 1:1) to give MC-158-3(110mg, 46%) as a yellow solid.
Step 4 of synthesizing CGT-9027
MC-158-3(110mg,0.24mmol,1.0eq) was dissolved in 6mL of a mixed solvent of tetrahydrofuran and water at a mixing ratio of 5:1, sodium hydroxide (30mg,0.73mmol) was added, and the reaction solution was stirred at room temperature for reaction for 30 minutes. The reaction was taken up in 20mL of water, extracted three times with 25mL of ethyl acetate, the organic phase washed with saturated brine, dried over anhydrous sodium sulfate and dried in vacuo to give the crude product which was isolated by hplc (mobile phase 0.1% FA/CH3CN/H2O) to afford CGT-9027(55.3mg, 53%) as a white solid.
LCMS:M+1=426.1
1H NMR(400MHz,DMSO-d6)δ11.47(s,1H),8.88(s,1H),7.51(s,1H),7.28(s,1H),7.20-7.16(t,J=16Hz,1H),7.12-7.10(m,1H),6.78-6.70(m,1H),6.21(m,1H),3.76(s,3H),2.71-2.67(m,2H),1.23(s,2H)。
EXAMPLE 11 Synthesis of CGT-9028
Figure GDA0002151341970000291
Step 1 Synthesis of MC-163-2
MC-163-1(2g,17.68mmol,1.0eq), p-toluenesulfonic acid (336mg,1.768mmol,0.1eq) were dissolved in 40mL of tetrahydrofuran, followed by addition of DHP (1.6g,19.448mmol,1.1eq) and reaction at ambient temperature for 16 hours. The reaction mixture was extracted three times with 25mL of water and 35mL of ethyl acetate, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and dried to give a crude product, which was purified by column (petroleum ether: ethyl acetate 20:1 to 5:1) to give MC-158-1 as an oil (3.5g, 99%).
Step 2 Synthesis of MC-163-3
MC-163-3(450mg,2.28mmol,1.0eq) was dissolved in MeOH (5mL), Pd/C (100mg) was added and stirred at room temperature under H2 for 16hrs, the reaction was filtered and rotary dried to give MC-163-3(400mg, crude) as a yellow solid.
Step 3 Synthesis of MC-163-4
MC-163-3(400mg,2.4mmol,1.0eq), SM (420mg,2.4mmol) was dissolved in DMF (20mL), followed by the addition of HATU (1.82g,4.8mmol), DIEA (1.1g,7.2mmol). reaction was carried out at ambient temperature for 16 hours. The reaction mixture was extracted three times with 25mL of water and 25mL of ethyl acetate, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and dried to give the crude product, which was purified by column (petroleum ether: ethyl acetate 2: 1) to give MC-163-4(870mg, 95%) as a yellow solid.
Step 4 Synthesis of MC-163-5
MC-163-4(870mg,2.7mmol,1.0eq) was dissolved in DCM (15mL), TFA (2.5g,0.73mmol) was added and the reaction stirred at 40 deg.C for 16 h. The reaction solution was spin-dried in vacuo to give crude MC-163-5(2 g).
Step 5 Synthesis of MC-163-6
MC-163-5(2g, crede) was dissolved in tetrahydrofuran (50mL), cesium carbonate (10g,18.8mmol) was added to adjust the pH to >7, then SM1(700mg,1.88mmol) was added and the reaction stirred at ambient temperature for half an hour. The reaction mixture was extracted three times with 50mL of water and 25mL of ethyl acetate, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and dried by spinning to give a crude product, which was then prepared by hplc to give MC-163-6(30mg) as a yellow solid.
Step 6 of synthesizing CGT-9028
MC-163-6(30mg,0.06mmol) was dissolved in THF/H2O 5:1(6mL), then sodium hydroxide (13mg,0.3mmol) was added and the reaction stirred at ambient temperature for half an hour. The reaction was filtered and purified by hplc to give CGT-9028(2.3mg, 8% yield) as a white solid
LCMS:M+1=441.1
1H NMR(400MHz,DMSO-d6)δ8.48(s,2H),8.19(s,1H),7.85-7.83(m,1H),7.50-7.46(m,1H),7.18-7.14(t,J=16Hz,1H),6.91(s,1H),3.71(s,2H)。
EXAMPLE 12 Synthesis of CGT-9029
Figure GDA0002151341970000301
Step 1 Synthesis of MC-165-2
MC-165-1(2.64g,10mmol,1.0eq), SM1(1.75g,10mmol,1.0eq) were dissolved in DMF (30mL), and HATU (5.7g,15mmol,1.5eq) was added and the reaction stirred at ambient temperature for 16 h. The reaction was taken up in 50mL of water and extracted with ethyl acetate (50 mL. times.3.) the organic phase was washed with brine, dried over anhydrous sodium sulfate and dried in vacuo to yield MC-158-1(600mg, 26%) as a yellow solid.
Step 2 Synthesis of MC-165-3
MC-165-2(600mg,2.5mmol,1.0eq) was added to 4M dioxane hydrochloride and stirred at ambient temperature for 16 h. The reaction was spun down in vacuo and taken up three times with 50ml of methanol to give MC-165-3(400mg, crede) as a white solid.
Step 3 Synthesis of MC-165-4
MC-165-3(100mg,0.56mmol,1.0eq), SM2(229mg,0.62mmol,1.1eq) were dissolved in DMF (20mL) and Et3N (170mg,1.68mmol) was added at 0 deg.C and the reaction stirred at 0 deg.C for 1 h. 30mL of water was added to the reaction solution, and the mixture was extracted three times with 50mL of ethyl acetate. The organic phase was washed with brine, dried over anhydrous sodium sulfate and rotary dried in vacuo to afford MC-165-4(150mg, 50%) as a red solid.
Step 4 of synthesizing CGT-9029
MC-165-4(150mg,0.32mmol,1.0eq) was dissolved in DMF (5mL) and five mL of 10% aqueous potassium hydroxide was added. The reaction was stirred at 50 ℃ for 2 hours. The reaction mixture was filtered and the filtrate was purified by HPLC (mobile phase 0.1% FA/CH3CN/H2O) to give CGT-9029(22.6mg, 15%) as a white solid.
LCMS:M+1=439.2
1H NMR(400MHz,DMSO-d6)δ11.88(s,1H),8.89(s,1H),8.31(s,2H),7.22-7.17(t,J=20HZ,1H),7.15-7.13(m,1H),6.82-6.80(m,1H),6.74(s,2H),6.57(s,1H),4.11-4.09(d,J=12HZ,2H)
Example 13 Synthesis of CGT-9003
Figure GDA0002151341970000311
The compound MC-001-3(200mg,0.52mmol,1.0eq) was dissolved in THF (5mL) at room temperature. Then, an aqueous cesium carbonate solution (2N,5mL), SM1(290mg,0.78mmol,1.5eq) was added to the solution. The reaction mixture was reacted at 25 ℃ for 1 hour. Then, aqueous sodium hydroxide (2N, 2mL) was added to the reaction mixture, the reaction mixture was reacted at room temperature for 1 hour, and the reaction mixture was diluted with water (20mL) and extracted with ethyl acetate (20mL X2). The organic phase was concentrated by drying. The residue was purified by HPLC to give CGT-9003(120mg, 35%) as a white solid.
1H NMR(400MHz,DMSO-d6):δ11.49(br,2H),8.91(s,2H),7.19-7.10(m,4H),6.76-6.72(m,2H),6.36-6.32(m,2H),3.47-3.44(m,4H).
19F NMR(376.5MHz,DMSO-d6):δ-117.83,-118.37.
HPLC:@254nm:99.33%,@214nm:99.67%.
LCMS:M+1=659.0
EXAMPLE 14 Synthesis of CGT-9010
Figure GDA0002151341970000321
Step 1 Synthesis of MC-149-2
Compound MC-149-1(632mg,1.7mmol,1.0eq) was dissolved in THF (5mL) at room temperature. Cesium carbonate (1.7g,5.1mmol,3.0eq), SM1(408mg,2.0mmol,1.2eq) and water (5mL) were then added to the solution. The reaction mixture was reacted at 25 ℃ for 0.5 hour. The reaction was then diluted with water (30 mL). The resulting mixture was extracted with ethyl acetate (30mL X2). The organic phase was concentrated by drying. The residue was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/5-1/3) to give MC-001-2(750mg, 83%) as a pale brown solid.
Step 2 Synthesis of MC-149-3
A solution of compound MC-149-2(750mg,1.4mmol,1.0eq) and TFA (2mL) in dichloromethane (5mL) was reacted at 25 ℃ for 2 h. The reaction was then concentrated to give a light brown oily compound. The oily intermediate was dissolved in methylene chloride (10mL), and potassium carbonate (2g) was added to the solution. The reaction mixture was stirred at room temperature for 30 minutes, then filtered, and the filtrate was concentrated to give a crude yellow solid compound MC-149-3(600mg, 99%).
Step 3 of synthesizing CGT-9010
Compound MC-149-3(150mg,0.35mmol,1.0eq) was dissolved in THF (4mL) at room temperature. Then, an aqueous cesium carbonate solution (2N,4mL) and SM2(143mg,0.39mmol,1.1eq) were added to the solution. The reaction mixture was reacted at 25 ℃ for 1 hour. Then, aqueous sodium hydroxide (2N, 2mL) was added to the reaction mixture. The reaction was continued at room temperature for 1 hour and then diluted with water (20 mL). The resulting mixture was extracted with ethyl acetate (20mL X2). The organic phase was concentrated by drying. The residue was purified by HPLC to give CGT-9010(7mg, 2%) as a white solid.
1H NMR(400MHz,DMSO-d6):δ11.50(s,2H),8.88(s,2H),7.21-7.11(m,4H),6.81-6.76(m,2H),6.24-6.20(m,2H),3.65-3.61(m,4H),3.44-3.38(m,4H).
19F NMR(376.5MHz,DMSO-d6):δ-117.82.
HPLC:@254nm:89.80%,@214nm:94.31%.
LCMS:M+1=702.9
EXAMPLE 15 Synthesis of CGT-9012
Figure GDA0002151341970000331
Step 1 Synthesis of MC-117-3
Compound MC-117-1(500mg,1.34mmol,1.0eq) was dissolved in THF (5mL) at room temperature. Cesium carbonate (657mg,2.02mmol,1.5eq), MC-117-2(404mg,2.02mmol,1.5eq) and water (2mL) were then added to the solution. The reaction mixture was reacted at room temperature for 2 hours. The reaction was then diluted with water (20 mL). The resulting mixture was extracted with ethyl acetate (20mL X3). The organic phase was concentrated by drying. The residue was purified by silica gel chromatography (ethyl acetate/petroleum ether ═ 1/10) to give MC-117-3(630mg, 89%)
Step 2 Synthesis of MC-117-4
A solution of compound MC-117-3(630mg,1.20mol,1.0eq) in TFA (10mL) was reacted at room temperature for 3 hours. The reaction was then concentrated to give MC-117-4 as a light brown oil (870mg, extra heavy).
Step 3 Synthesis of MC-117-5
Tert-butanol (787mg,10.62mol,10.5eq) was added to a solution of chlorosulfonic isocyanate (1.43g,10.11mmol,10.0eq) in dichloromethane (10mL) at 0 deg.C and reacted under these conditions for 1 hour. This solution was then added to a solution of compound MC-117-4(430mg,1.01mmol,1.0eq) and triethylamine (614mg,6.07mmol,6.0eq) in dichloromethane (15mL) which had been cooled to 0 ℃ beforehand. The reaction mixture was reacted at 0 ℃ for 2 hours and then quenched with water (30 mL). The resulting mixture was extracted three times with dichloromethane (30mL), and the organic phase was washed twice with saturated brine (20 mL). The organic phase was dried and concentrated to give MC-117-5(400mg, 65%) as a yellow solid.
Step 4 Synthesis of MC-117-6
A solution of compound MC-117-5(400mg,0.662mol,1.0eq) in TFA (10mL) was reacted at room temperature for 3 hours. The reaction mixture was then concentrated to give MC-117-6(320mg, 96%) as a pale brown oil.
Step 5 of synthesizing CGT-9012
Compound MC-117-6(320mg,0.635mmol,1.0eq) was dissolved in methanol (5mL) and potassium carbonate (263mg,1.904mmol,3.0eq) was added. The reaction mixture was reacted at 40 ℃ for 3 hours. After the reaction was complete, the reaction mixture was diluted with water (20 mL). The resulting mixture was extracted with ethyl acetate (20mLX 3). The organic phase was concentrated by drying. The residue was purified by HPLC to give CGT-9012(92mg, 30%)
1H NMR(400MHz,DMSO-d6):δ11.5(br,1H),8.90(br,1H),7.17(t,J=8.8Hz,1H),7.13(dd,J=6.0Hz,2.4Hz,1H),6.80-6.76(m,2H),6.20(t,J=6.0Hz,1H),3.86-3.83(m,1H),3.38-3.20(m,4H),1.87-1.79(m,3H),1.70-1.68(m,1H).
HPLC:@254nm:98.04%,@214nm:98.03%.
LCMS:M+1=478.0
Example 16 Synthesis of CGT-9013
Figure GDA0002151341970000341
Step 1 Synthesis of MC-118-1
A mixture of the compound MC-101-007(500mg,1.34mol,1.0eq), (S) -1-N-tert-butoxycarbonyl-2- (aminoethyl) pyrrolidine (500mg,2.50mmol,1.8eq), cesium carbonate (873mg,2.68mmol,2.0eq) and tetrahydrofuran/water (10/10mL) was stirred at 20 ℃ for 3 hours. After completion of the reaction, the reaction mixture was extracted with ethyl acetate (50mLX 2). The organic phase was dried and the residue obtained after concentration was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/10) to give MC-118-1(660mg, 93%) as a white solid.
Step 2 Synthesis of MC-118-2
To a solution of compound MC-118-1(660mg,1.26mol,1.0eq) in dichloromethane (10mL) was added 5mL of trifluoroacetic acid at room temperature. The reaction was then allowed to react overnight at room temperature. After the reaction, the reaction mixture was concentrated to give a crude product, MC-118-2(800mg, trifluoroacetate) as a pale yellow solid.
Step 3 Synthesis of MC-118-3
Tert-butanol (549mg, 7.41mmol, 10.5eq) was added dropwise to a solution of the compound chlorosulfonyl isocyanate (998mg,7.06mmol,10.0eq) in dichloromethane (10mL) under ice bath conditions. After the reaction was carried out under ice-bath conditions for 1 hour, the reaction solution was added dropwise to an ice-bath protected solution of compound MC-118-2(300mg, 0.706mmol, 1.0eq) and triethylamine (714mg, 7.06mmol,10.0eq) in dichloromethane (10 mL). The reaction solution was reacted for 2 hours under ice bath protection. After the reaction was completed, the reaction solution was quenched and diluted with cold water (50 mL). The mixture was then extracted with dichloromethane (50mLX 2). The organic phase was dried and concentrated to give the crude compound MC-118-3(500mg, crude) as a pale yellow solid.
Step 4 Synthesis of MC-118-4
To a solution of compound MC-118-3(500mg, crude) in dichloromethane (10mL) at room temperature was added 5mL of trifluoroacetic acid. The reaction solution was reacted at room temperature overnight. After the reaction, the reaction mixture was directly concentrated to give a pale yellow oily crude compound MC-118-4(400mg, trifluoroacetate salt).
Step 5 of synthesizing CGT-9013
To a solution of compound MC-118-4(400mg, trifluoroacetate) in methanol (10mL) was added 5mL of sodium hydroxide (1N) solution. The reaction was carried out at 25 ℃ for 1 hour. After completion of the reaction, the reaction mixture was diluted with water (50mL), and the resulting mixture was extracted with ethyl acetate (50 mL. times.3). The organic phase was dried and concentrated to give a pale yellow oil, and the residue was purified by high performance preparative chromatography to give the compound CGT-9013 as a white solid (32mg, 8% yield).
1H NMR(400MHz,DMSO-d6):δ11.49(s,1H),8.86(s,1H),7.20-7.11(m,2H),6.80-6.76(m,3H),6.19(t,J=6.0Hz,1H),3.85-3.83(m,1H),3.38-3.19(m,4H),1.86-1.78(m,3H),1.71-1.68(m,1H).
19F NMR(376.5MHz,DMSO-d6)δ-117.89,-118.40
HPLC:@254nm:99.48%,@214nm:99.14%.
LCMS:M-1=478.0
Example 17 Synthesis of CGT-9014
Figure GDA0002151341970000361
Step 1 Synthesis of MC-123-2
MsCl (139mg, 1.21mmol, 1.2eq) was added dropwise to a solution of compound MC-123-1(430mg, 1.01mmol,1.0eq) and triethylamine (614mg,6.07mmol,6.0eq) in dichloromethane (20mL) at zero degrees. The reaction mixture was reacted for 2 hours under ice bath conditions. The reaction was then diluted with cold water (30 mL). The resulting mixture was extracted three times with dichloromethane (30 mL). The organic phase was washed twice with saturated brine (30mL), dried and concentrated to give MC-123-2(400mg, 78%) as a pale yellow solid.
Step 2 of synthesizing CGT-9014
Potassium carbonate (329mg,2.38mmol,3.0eq) was added to a solution of compound MC-123-1(400mg,0.79mmol,1.0eq) in methanol (5 mL). The reaction mixture was reacted at 40 ℃ for 3 hours. After the reaction was complete, the reaction mixture was diluted with water (20 mL). The resulting mixture was extracted with ethyl acetate (20mLX 3). The organic phase was concentrated by drying. The residue was purified by HPLC to give CGT-9014(101.4mg, 26%)
1H NMR(400MHz,DMSO-d6):δ11.53(br,1H),8.89(br,1H),7.18(t,J=8.8Hz,1H),7.11(dd,J=6.0Hz,2.4Hz,1H),6.80-6.76(m,1H),6.27(t,J=6.0Hz,1H),3.90-3.87(m,1H),3.40-3.22(m,4H),2.91(s,3H),1.97-1.70(m,4H).
19F NMR(376.5MHz,DMSO-d6):δ-117.94,-118.38.
HPLC:@254nm:99.57%,@214nm:99.58%.
LCMS:M+1=479.0
EXAMPLE 18 Synthesis of CGT-9015
Figure GDA0002151341970000362
Step 1 Synthesis of MC-124-1
MsCl (0.11mL, 1.41mmol, 1.5eq) was added dropwise to a solution of compound MC-118-2(400mg, 0.94mmol, 1.0eq) and triethylamine (0.65mL, 4.70mmol, 5.0eq) in dichloromethane (5mL) at 0 deg.C. The reaction mixture was reacted for 2 hours under ice bath conditions. The reaction was then diluted with cold water (50 mL). The resulting mixture was extracted with dichloromethane (50mLX2) and the organic phase was concentrated by drying to give the crude compound MC-124-1(350mg, crude) as a pale yellow solid.
Step 2 of synthesizing CGT-9015
5mL of 1N sodium hydroxide solution was added to a solution of compound MC-124-1(350mg, crude) in methanol (10mL) at room temperature, and the reaction was allowed to react at 25 ℃ for 2 hours. After completion of the reaction, the reaction mixture was diluted with water (30mL), and the resulting mixture was extracted with ethyl acetate (50 mL. times.3). The organic phase is then directly dried and concentrated. The resulting residue was purified by high performance preparative chromatography to give the compound CGT-9015(82mg, 24% yield) as a white solid.
1H NMR(400MHz,DMSO-d6)δ11.50(brs,1H),8.87(brs,1H),7.20-7.12(m,2H),6.79(dd,J=7.2Hz,3.2Hz,1H),6.28-6.25(t,J=6.0Hz,1H),3.91-3.89(m,1.0H),3.40-3.18(m,4H),2.91(s,3H),1.95-1.81(m,3H),1.74-1.69(m,1H).
19F NMR(376.5MHz,DMSO-d6)δ-117.85,-118.39
HPLC:@254nm:99.38%,@214nm:99.49%.
LCMS:M+1=479.1
EXAMPLE 19 Synthesis of CGT-9017
Figure GDA0002151341970000371
A solution of compound MC-153-1(200mg,0.33mmol,1.0eq) in TFA (2mL) and dichloromethane (5mL) was reacted at 25 ℃ for 1 hour and concentrated to give the intermediate. The intermediate was dissolved in tetrahydrofuran (5mL) and 2N sodium hydroxide solution (2mL) was added. The reaction mixture was reacted at room temperature for 1 hour. After the reaction was complete the reaction mixture was diluted with water. The resulting mixture was extracted with ethyl acetate (20mLX 2). The organic phase was concentrated by drying. The residue was purified by HPLC to give CGT-9017(23mg, 7%) as a white solid.
LCMS:M+1=478.1
1H NMR(400MHz,DMSO-d6)δ11.52(s,1H),8.90(s,1H),7.20-7.10(m,2H),6.78-6.73(m,3H),6.40-6.37(m,1H),3.32-3.09(m,5H),2.90-2.86(m,1H),2.55-2.53(m,1H),1.94-1.90(m,1H),1.59-1.54(m,1H).
19F NMR(376.5MHz,DMSO-d6)δ-117.72,-118.40.
EXAMPLE 20 Synthesis of CGT-9018
Figure GDA0002151341970000381
Step 1 Synthesis of MC-116-1
Compounds SM1(1.61g,10.0mmol,1.0eq), triphenylphosphine (2.75g,10.5,1.05eq) and SM2(1.71g,1.71mmol,1.05eq.) were dissolved in dry tetrahydrofuran (50mL) and cooled to 0 degrees by the addition of DEAD (1.82g,10.5mmol,1.05 eq.). The mixture was stirred at room temperature for 16 hours. The reaction was detected by dot plate chromatography, and after completion of the reaction, the residue obtained after concentration of the reaction mixture was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/2) to obtain MC-116-1(2.83g, 92%) as a white solid.
Step 2 Synthesis of MC-116-2
Hydrazine monohydrate (0.146mL,4.71mmol,1.03eq.) was added to a solution of compound MC-116-1(1.4g,4.57mol,1.0eq.) in ethanol (10mL) at room temperature. The reaction was then allowed to react overnight at room temperature. The reaction was detected by dot plate, after completion of the reaction, the reaction solution was filtered, and the residue obtained by concentrating the filtrate was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/2) to give MC-116-2(448mg, 55%) as a colorless oily compound.
Step 3 Synthesis of MC-116-3
The compounds MC-101-007(200mg,0.54mmol,1.0eq.) and MC-116-2(200mg,1.13mmol,2.1eq.) were dissolved in tetrahydrofuran (10 mL). After 4 hours at 40 ℃ reaction time, the reaction was checked by LCMS, and after completion of the reaction, the residue obtained by concentrating the reaction mixture was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/5) to obtain MC-116-3(280mg, 83%) as a white solid.
Step 4 Synthesis of MC-116-4
To a solution of compound MC-116-3(50mg,0.10mmol,1.0eq.) in dichloromethane (5mL) at room temperature was added 1mL of trifluoroacetic acid. The reaction solution was reacted at room temperature overnight. After the reaction, the reaction mixture was directly concentrated to give a pale yellow oily crude compound MC-116-4(200mg, trifluoroacetate salt).
Step 5 Synthesis of MC-116-5
Tert-butanol (389mg,5.25mmol,10.5eq.) was added dropwise to a solution of the compound chlorosulfonyl isocyanate (708mg,5.0mmol,10.0eq.) in dichloromethane (5mL) under ice bath conditions. After the reaction was carried out in ice bath for 1 hour, the reaction solution was added dropwise to a solution of ice-bath protected compound MC-116-4(200mg,0.50mmol,1.0eq) and triethylamine (531mg,5.25mmol,10.5eq) in dichloromethane (5 mL). The reaction solution was reacted for 2 hours under ice bath protection. After the reaction was completed, the reaction solution was quenched and diluted with cold water (50 mL). The mixture was then extracted with dichloromethane (50mL X2). The organic phase was dried and concentrated to give the crude compound MC-116-5(500mg, crude) as a pale yellow solid.
Step 6 of synthesizing CGT-9018
To a solution of compound MC-116-5(500mg, crude) in dichloromethane (10mL) at room temperature was added 2mL of trifluoroacetic acid. The reaction solution was reacted at room temperature for 2 hours. After the reaction, the reaction mixture was directly concentrated to give a pale yellow oily crude compound, which was dissolved in tetrahydrofuran (10mL) and 5mL of sodium hydroxide (1N) solution was added. The reaction was carried out at 25 ℃ for 1 hour. After completion of the reaction, the reaction mixture was diluted with water (50mL), and the resulting mixture was extracted with ethyl acetate (50 mL. times.3). The organic phase was dried and concentrated to give a pale yellow oil, and the residue was purified by high performance preparative chromatography to give the compound CGT-9018(21mg, 5% yield) as a white solid.
1H NMR(400MHz,DMSO-d6):δ11.49(s,1H),9.78(s,1H),8.95(s,1H),7.22-7.17(t,J=8.8Hz,1H),7.12-7.10(m,1H),6.79-6.75(m,1H),6.10-6.54(m,3H),3.96-3.91(t,J=6.0Hz,2H),3.16-3.12(m,2H).
19F NMR(376.5MHz,DMSO-d6)δ-117.22,-118.20
HPLC:@254nm:98.59%,@214nm:98.59%.
LCMS:M+1=454.1。
EXAMPLE 21 Synthesis of CGT-9201
Figure GDA0002151341970000391
Step 1 Synthesis of CGT-9201-1
Cesium carbonate (880.4mg,2.68mmol,2.0eq) was added to a solution of the compound MC-101-007(500mg,1.34mmol,1.0eq), N-tert-butoxycarbonyl-1, 2-ethylenediamine (428.8mg,2.0mmol,2.0eq) in tetrahydrofuran/water (5/1mL) at room temperature. The reaction was stirred at 20 ℃ for 0.5 hour. Water (10mL) was added to the reaction solution, and the resulting mixture was extracted with ethyl acetate (25mL × 3). The organic phase was washed with saturated brine (20mL × 2), dried, filtered and concentrated. The obtained residue was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/3) to obtain CGT-9201-1(517mg, yield 79.3%) as a yellow solid.
Step 2 Synthesis of CGT-9201-2
To a solution of compound CGT-9201-1(570mg,1.17mmol,1.0eq) in dichloromethane (20mL) was added 3mL of trifluoroacetic acid at room temperature. The reaction was carried out at room temperature for 1 hour, and the reaction solution was adjusted to pH 9 with aqueous sodium bicarbonate solution and then directly concentrated by extraction with dichloromethane (30X 2) to give a crude pale yellow solid compound CGT-9201-2(325mg, yield 71.9%).
Step 3 Synthesis of CGT-9201-3
CGT-9201-2(200mg,0.52mmol) and triethylamine (263mg,2.6mmol) were dissolved in ethanol (10ml) and stirred for 5 minutes then the compound diethyl squarate (105.5mg,0.62mmol) was dissolved in ethanol (5ml) and added dropwise to the mixture then stirring was continued for 3 hours finally the compound methylamine hydrochloride (49.5mg,0.78mmol) was added the reaction was stirred for 15 hours then dried and concentrated to give the crude product as a pale yellow oil (230 mg).
Step 4 of synthesizing CGT-9201
Potassium carbonate (82.8mmol,0.6mmol) was added to a solution of the compound CGT-9201-3(200mg,0.40mmol)) in methanol (5mL) at room temperature. The reaction was carried out at room temperature for 3 hours. After the reaction is complete the solvent is concentrated. The residue was purified by HPLC to give CGT-9201(20mg, 11%) as a white solid.
LCMS:M+1=468.1,470.1(LCMS:MC18-71-008-1)
1H NMR(400MHz,DMSO-d6)δ11.47(s,1H),8.88(s,1H),7.19(t,J=4.4Hz,1H),7.12(m,1H),6.76(m,1H),6.31(t,J=2.2Hz,1H),3.69(m,2H),3.42(m,2H),3.14(s,3H).(HNMR:MC18-71-008-1)。
EXAMPLE 22 Synthesis of CGT-9030
Figure GDA0002151341970000411
Step 1 Synthesis of MC-105-10
The reaction was completed after the compound MC-105-4(254mg,2.0mmol,1.58eq) and the compound MC-101-009(400mg,1.26mmol,1.0eq) were reacted in THF (5mL) at 35 ℃ for 10 minutes. The reaction solution was directly concentrated. The residue was purified by silica gel chromatography (EtOAc/PE ═ 1/3) to give the compound MC-105-10(420mg, 83% yield) as a brown oil.
Step 2 of synthesizing CGT-9030
Sodium hydroxide (56mg,1.44mmol,8.0eq) was added to a solution of compound MC-105-10(71.7mg,0.18mmol,1.0eq) in tetrahydrofuran (3mL) and water (1mL) at room temperature. The reaction was carried out at room temperature for 1 hour. After completion of the reaction, the reaction mixture was diluted with 25mL of water, and the resulting mixture was extracted with ethyl acetate (25 mL. times.3). The organic phase was dried and concentrated, and the residue was purified by HPLC to give CGT-9030(21mg, 27% yield) as a white solid.
LCMS:M+1=373.1。
EXAMPLE 23 Synthesis of CGT-9031
Figure GDA0002151341970000412
Step 1 Synthesis of MC-164A-7
The compounds MC-101-018(227mg,0.70mmol,1.0eq.), MC-164A-2(260mg,1.88mmol,2.7eq.) and triethylamine (425mg,4.20,6.0eq.) were dissolved in tetrahydrofuran (20 mL). After 4 hours at 40 ℃ reaction time, the reaction was checked by LCMS, and after completion of the reaction, the residue obtained by concentrating the reaction mixture was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/1) to obtain MC-164A-7(170mg, 58%) as a pale yellow solid.
Step 2 of synthesizing CGT-9031
Compound MC-164A-7(170mg,0.41mmol,1.0eq) was dissolved in tetrahydrofuran (10mL) at room temperature with the addition of 5mL sodium hydroxide (1N) solution. The reaction was carried out at room temperature for 1 hour. After the reaction was completed, the reaction solution was diluted with water, and the resulting mixture was extracted with ethyl acetate. The organic phase was dried and concentrated to give a pale yellow oil, and the residue was purified by high performance preparative chromatography to give the pale yellow solid compound CGT-9031(69mg, 43% yield).
LC-MS:M+1=389.1。
EXAMPLE 24 Synthesis of CGT-9032
Figure GDA0002151341970000421
Step 1 Synthesis of MC-164B-15
The compounds MC-101-013(195mg,0.54mmol,1.0eq.), MC-164B-2(200mg,1.45mmol,2.7eq.) and triethylamine (328mg,3.24,6.0eq.) were dissolved in tetrahydrofuran (20 mL). After 5 hours at 40 ℃ reaction, the reaction was checked by LCMS, and after completion of the reaction, the residue obtained by concentrating the reaction mixture was purified by silica gel chromatography (ethyl acetate/petroleum ether: 1/5) to obtain MC-164B-15(195mg, 80%) as a pale yellow solid.
Step 2 of synthesizing CGT-9032
Compound MC-164B-15(195mg,0.43mmol,1.0eq) was dissolved in tetrahydrofuran (10mL) at room temperature with the addition of 5mL sodium hydroxide (1N) solution. The reaction was carried out at room temperature for 1 hour. After the reaction was completed, the reaction solution was diluted with water, and the resulting mixture was extracted with ethyl acetate. The organic phase was dried and concentrated to give a pale yellow oil, and the residue was purified by high performance preparative chromatography to give the pale yellow solid compound CGT-9032(38mg, 21% yield).
LCMS:M+1=427.1。
EXAMPLE 25 Indolylamine 2, 3-dioxygenase (IDO) enzyme Activity assay
The assay buffer of IDO1 was prepared by dissolving 400. mu. M L-tryptophan (Cat # T8941-25G, Sigma),20mM ascorbic acid (Cat # A4034-100G, Sigma), 20. mu.M methylene blue (Cat # M9140-25G, Sigma) and 1000U/ml catalase in 100mM PBS buffer (pH 6.5)
To 98. mu.L of IDO1 assay buffer was added 1. mu.L of the test compound (100X) at the desired assay concentration and 1. mu.L of 100 ng/. mu.L of IDO1 enzyme (made by Meidicy). IDO1 and assay buffer required preheating to 37 ℃. The reaction is carried out for 30min in a constant temperature water tank at 37 ℃.50 μ L of 30% trichloroacetic acid (Cat # T0699-100ML, Sigma) was added. Reacting for 30min in a constant temperature water tank at 52 ℃. Centrifuge at 12000g for 10min at room temperature. 100 μ L of the supernatant was taken in a 96 well assay plate and mixed with 100 μ L of Ehrlich's Reagent (4-dimethylaminobenzaldehyde (Cat #156477-25g, Sigma)). The absorbance was measured at 480nm using an M5 microplate reader.
Data analysis
Inhibition ratio%positive―ODsample)/(ODpositive―ODnegative)*100%
IC50 curve fitting was performed using software Graphpad Prism 6 and using the calculation formula log (inhibitor) vs. normalized response and calculated IC50 values, as shown in table 1 below.
Example 26IDO inhibitor assay for the enzyme Activity of IDO1 induced by Hela cells
Hela human cervical cancer cells (Cat # CCL-2, ATCC) were collected in the logarithmic growth phase, counted, adjusted to a cell concentration of 5000 cells per well, and inoculated into 96-well culture plates with 100. mu.L of cell suspension per well. Cells were incubated at 37 ℃ in a 100% relative humidity, 5% CO2 incubator for 24 hours. Candidate compounds and controls were diluted to the set final concentration of action with the medium DMEM medium (Cat #11995-040, GIBCO) at 50ng/mL IFN-gamma for a total of 200. mu.L/well change to Hela cells and re-well tested. Cells were incubated at 37 ℃ in a 100% relative humidity, 5% CO2 incubator for 48 hours. The inhibition efficiency of IDO1 cell level was measured by taking 140. mu.L/well of cell culture supernatant. Add 15. mu.L/well of trichloroacetic acid. The reaction was carried out in a constant temperature water bath at 52 ℃ for 30 minutes. Centrifuge at 12000g for 10min at room temperature. 100 μ L/well of the supernatant was transferred to a 96-well assay plate, and 100 μ L/well of Erwinia solution (Ehrlich's Reagent, 4-dimethylaminobenzaldehyde (Cat #156477-25g, Sigma)) was mixed and the absorbance was measured at 480nm using an M5 microplate reader.
Data analysis
Inhibition rate (OD)positive―ODsample)/(ODpositive―ODnegative)*100%
IC50 curve fitting was performed using software Graphpad Prism 6 and using the calculation formula log (inhibitor) vs. normalized response-variable slope and the IC50 values were calculated.
In table 1 below, IDO and Hela cell IC50 data are provided for the compounds provided in examples 1-24, and the results calculated are given in table 1 below.
TABLE 1 enzyme inhibitory Activity data and Hela cell inhibitory Activity data for each Compound
Figure GDA0002151341970000441
Figure GDA0002151341970000451
As can be seen from the data in table 1 above, the compounds provided by the present invention all have excellent enzymatic and cytological IDO inhibitory activity.
EXAMPLE 26 drug permeation Performance test
The compounds provided in examples 1-24 and the control compound CGT-9002 were evaluated for their bidirectional transmembrane transport and efflux properties using the MDCKII-hMDR1 cell monolayer model. The corresponding structural formula of CGT-9002 is a compound of the formula A, wherein R1 is NH2, R2 is Br, and R3 is F.
Figure GDA0002151341970000452
Cell culture: during the experiment, MDCKII-hMDR1 cells were cultured in high-sugar DMEM (Hyclone) containing 10% fetal bovine serum (GIBCO) and streptomycin (100 units/mL each) at 37 ℃ and containing 5% CO2The incubator of (2) for cultivation.
Prior to the start of the transport experiment, pre-warmed HBSS buffer (137mM NaCl, 4.17mM NaHCO) was used3,0.34mM Na2HPO4,5.37mM KCl,0.44mM KH2PO4,1.26mM CaCl2,0.49mM MgCl2,0.41mM MgSO45.55mM D-Glucose, 10mM HEPES, pH 7.4)) MDCKII-hMDR1 cells were washed 3 times in a monolayer and placed at 37 deg.CAfter 30min incubation, the TEER value was determined with a cell resistance meter (Millicell-ERS2) to confirm the integrity and tightness of the cell monolayer.
In transport experiments, MDCKII-hMDR1 cells were cultured at 1X 105The density of each well was inoculated on a 12-well Transwell plate (Corning Costar, cat #3401, 1.12 cm)20.4 μm pore size), the seeded MDCKII-hMDR1 cells will form a complete cell monolayer after 5 days, including P-gp efflux transporter expression and trans-epithelial electrical resistance (TEER) formation. During this period, the culture medium was changed every other day, and the volumes of the culture medium on the Apical Side (Apical Side, A) and basal Side (basal Side, B) of the Transwell filter were 0.5mL and 1.5mL, respectively. The transport assay was started when the HBSS buffer on the A-or B-side of the Transwell filter was replaced with the test compound and was terminated after a final incubation period of 2 hours at 37 ℃.
50 μ L of each sample solution was taken on both sides of the Transwell filter and mixed with 100 μ L acetonitrile containing the internal standard compound (tolbutamide), and for the sample to be diluted (e.g., 10-fold dilution), 5 μ L of the sample solution was diluted and mixed with 45 μ L HBSS buffer, 100 μ L acetonitrile containing the internal standard compound was added and mixed, and then centrifuged at 13000rpm for 10min at 4 ℃, and finally 10 μ L of the supernatant was extracted for LC-MS/MS sample analysis. For each compound, the transport experiments in the a → B and B → a directions were in triple tubes, i.e. n ═ 3.
LC-MS/MS sample analysis
Sample analysis was performed by high performance liquid chromatography coupled with an API4000 triple quadrupole mass spectrometer (Applied Biosystems) by tandem, wherein the API4000 triple quadrupole mass spectrometer was equipped with an electrospray ionization (ESI) source. Ultra-pure nitrogen GAS was used as the curtain GAS, GAS1, assist GAS (GAS2), and collision GAS. Data were collected by Analyst 1.5 software.
And (3) data analysis:
the apparent permeability coefficient (Papp) was calculated from the rate of permeation of the compound through a monolayer of MDCKII-hMDR1 cells, and the magnitude of the value correlated with the in vivo absorption of the compound, and after the concentrations of the compounds on the A-side and B-side were determined by the LC-MS/MS method, the A → B or A → B formula was calculated according to the following formulaPapp value in B → A direction: papp(A→B)or Papp(B→A)=(dQ/dt)/(A*C0)=(C2h*V)/(t*A*C0)
In the above formula, dQ/dt is the permeation rate, i.e. the amount of compound permeated over the dt time, C0For the initial concentration of drug on the dosing side, a is the surface area of the cell monolayer, i.e. the membrane area.
The determination criteria for transmembrane transport permeation rate are as follows:
low permeability: papp(A→B)<1x 10-6cm/s
Middle permeation: papp(A→B)1-10x 10-6cm/s
High permeability: papp(A→B)>10x 10-6cm/s
In obtaining P of the compoundapp(A→B)And Papp(B→A)After the value is obtained, the Efflux Ratio (ER) of the compound can be calculated by the following formula:
Efflux Ratio(ER)=Papp(B→A)/Papp(A→B)
when the efflux ratio is more than or equal to 2, the compound is considered as a substrate of an efflux transporter.
Results of the experiment
The statistical results of the tests are shown in Table 2 below.
TABLE 2 statistics of cell transport assay results for compounds provided in examples 1-24 of the present invention
Figure GDA0002151341970000471
Figure GDA0002151341970000481
Figure GDA0002151341970000491
As can be seen from the data provided in table 2 above, some of the example compounds of the present invention are significantly superior to the reference compound (CGT-9002), have good permeability, not only to meet the requirement for drug absorption, but also to deliver enough drug to the target organ, and in addition to simple passive diffusion across the lipid layer, P-glycoprotein plays an important role in the selective accumulation and distribution of the drug in the target organ.
While the basic principles and embodiments of the present invention have been described above, the present invention is not limited to the above-described embodiments, and those skilled in the art can make various changes and modifications without departing from the spirit of the present invention, and these changes and modifications fall within the scope of the present invention.

Claims (4)

1. A1, 2, 5-oxadiazole derivative or a pharmaceutically acceptable salt thereof for use as an IDO inhibitor,
a compound selected from the following structures:
Figure FDA0003226121590000011
2. a pharmaceutical composition comprising one or more 1,2, 5-oxadiazole derivatives or pharmaceutically acceptable salts thereof for use as IDO inhibitors of claim 1 as an active ingredient in combination with at least one pharmaceutically acceptable carrier.
3. Use of the 1,2, 5-oxadiazole derivative or the pharmaceutically acceptable salt thereof, or the pharmaceutical composition containing any of them according to claim 1 as an IDO inhibitor in the manufacture of a medicament for inhibiting immunosuppression in a patient.
4. The use according to claim 3, for the treatment of viral infections, depression, neurodegenerative disorders, trauma, age-related cataracts, organ transplant rejection, autoimmune diseases, melanoma, obesity or ischemia.
CN201910617997.8A 2019-07-10 2019-07-10 1,2, 5-oxadiazole derivatives as inhibitors of indoleamine 2, 3-dioxygenase Active CN110204537B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910617997.8A CN110204537B (en) 2019-07-10 2019-07-10 1,2, 5-oxadiazole derivatives as inhibitors of indoleamine 2, 3-dioxygenase

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910617997.8A CN110204537B (en) 2019-07-10 2019-07-10 1,2, 5-oxadiazole derivatives as inhibitors of indoleamine 2, 3-dioxygenase

Publications (2)

Publication Number Publication Date
CN110204537A CN110204537A (en) 2019-09-06
CN110204537B true CN110204537B (en) 2021-10-08

Family

ID=67796970

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910617997.8A Active CN110204537B (en) 2019-07-10 2019-07-10 1,2, 5-oxadiazole derivatives as inhibitors of indoleamine 2, 3-dioxygenase

Country Status (1)

Country Link
CN (1) CN110204537B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111138425A (en) * 2020-01-06 2020-05-12 中国药科大学 Triazole derivative and preparation method and application thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2824100T3 (en) * 2008-07-08 2018-04-16 Incyte Holdings Corp 1,2,5-oxadiazoles as inhibitors of indolamine-2,3-dioxygenase
WO2017106062A1 (en) * 2015-12-15 2017-06-22 Merck Sharp & Dohme Corp. Novel compounds as indoleamine 2,3-dioxygenase inhibitors
CN109942565B (en) * 2017-12-20 2021-03-26 海创药业股份有限公司 Indoleamine-2, 3-dioxygenase inhibitor and preparation method and application thereof
CN108586378B (en) * 2018-01-22 2020-06-19 南京华威医药科技集团有限公司 Indoleamine 2, 3-bis-oxidase inhibitor and preparation method and application thereof
CN110343098B (en) * 2018-04-04 2023-05-26 中国科学院上海药物研究所 Oxadiazole compound, preparation method, pharmaceutical composition and application thereof

Also Published As

Publication number Publication date
CN110204537A (en) 2019-09-06

Similar Documents

Publication Publication Date Title
JP6145491B2 (en) IDO inhibitor
ES2944573T3 (en) TLR7/8 antagonists and uses thereof
ES2601226T3 (en) Imidazole derivatives as IDO inhibitors
TWI453204B (en) 1,2,5-oxadiazoles as inhibitors of indoleamine 2,3-dioxygenase
BR112016009786B1 (en) indoleamine 2,3-dioxigenase inhibitor compounds and their synthesis processes
US20200369648A1 (en) Tlr7/8 antagonists and uses thereof
JP2009538352A (en) 2-Carbocycloamino-4-imidazolylpyrimidines as agents for inhibiting cell proliferation
US20230110550A1 (en) Quinazoline Compounds, Preparation Method, Use, and Pharmaceutical Composition Thereof
WO2017088784A1 (en) Deuterium-modified brigatinib derivatives, pharmaceutical compositions comprising same, and use thereof
CN107021929A (en) One class Novel IDO inhibitor, preparation method, medical composition and its use
US20230103494A1 (en) Novel spiropyrrolidine derived antiviral agents
TW202321251A (en) Novel spiropyrrolidine derived antiviral agents
CN110204537B (en) 1,2, 5-oxadiazole derivatives as inhibitors of indoleamine 2, 3-dioxygenase
WO2018028491A1 (en) Indoleamine2,3-dioxygenase inhibitors and uses thereof in pharmacy
WO2018195649A1 (en) Substituted tetrahydropyridine derivatives as ido-1 inhibitors and uses thereof
NZ616457B2 (en) Fused imidazole derivatives useful as ido inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
CP03 Change of name, title or address
CP03 Change of name, title or address

Address after: 215123 units 101 and 102, C11 building, phase I project of biomedical industrial park, No. 218 Xinghu street, Suzhou Industrial Park, China (Jiangsu) pilot Free Trade Zone, Suzhou, Jiangsu Province

Patentee after: Shengshi Taike Biopharmaceutical Technology (Suzhou) Co.,Ltd.

Address before: 215123 unit 101, C11 building, bio nano Park, 218 Xinghu street, industrial park, Suzhou, Jiangsu

Patentee before: CGENE TECH (SUZHOU, CHINA) Co.,Ltd.