WO2019120212A1 - Ido抑制剂 - Google Patents

Ido抑制剂 Download PDF

Info

Publication number
WO2019120212A1
WO2019120212A1 PCT/CN2018/122015 CN2018122015W WO2019120212A1 WO 2019120212 A1 WO2019120212 A1 WO 2019120212A1 CN 2018122015 W CN2018122015 W CN 2018122015W WO 2019120212 A1 WO2019120212 A1 WO 2019120212A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
solution
pharmaceutically acceptable
acceptable salt
Prior art date
Application number
PCT/CN2018/122015
Other languages
English (en)
French (fr)
Inventor
周明
陆剑宇
胡国平
丁照中
黎健
陈曙辉
Original Assignee
基石药业(苏州)有限公司
拓石药业(上海)有限公司
基石药业
南京明德新药研发股份有限公司
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 基石药业(苏州)有限公司, 拓石药业(上海)有限公司, 基石药业, 南京明德新药研发股份有限公司 filed Critical 基石药业(苏州)有限公司
Priority to CN201880074353.2A priority Critical patent/CN111356692B/zh
Publication of WO2019120212A1 publication Critical patent/WO2019120212A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/554Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one sulfur as ring hetero atoms, e.g. clothiapine, diltiazem
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D243/00Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms
    • C07D243/06Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4
    • C07D243/08Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4 not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention relates to a novel compound as an IDO inhibitor, and specifically discloses a compound of the formula (I) and a pharmaceutically acceptable salt thereof.
  • the invention further relates to the use of a compound of formula (I) and a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of cancer.
  • Malignant tumors are one of the major diseases that endanger human life today.
  • humans have developed a variety of treatments and treatments including the most commonly used chemotherapy, surgery, radiotherapy and targeted therapy.
  • These therapies delay the development of tumors to a certain extent and prolong the life of patients.
  • the above treatments still fail to achieve the desired inhibitory effect.
  • the toxic side effects of the above therapies are also key factors limiting their application.
  • immunotherapy has been developed as an emerging therapy for the treatment of malignant tumors, characterized by mobilizing the host's natural defense mechanisms to counter the growth of tumors.
  • the main way is to activate the host's original immune system, enhance the host's collective immune response to tumor cells, and use the immune system's ability to achieve accurate and effective killing of tumors and curb their development.
  • IDO Indoleeamine-pyrrole-2,3-dioxygenase
  • ferrene which functions to catalyze tryptophan to kennel The rate-limiting step of the amino acid transition.
  • IDO is overexpressed in a variety of tumor cells. Over-expressed IDO can rapidly deplete tryptophan in the tumor microenvironment, so that T cells infiltrating into the tumor are arrested in the middle of G1 phase due to lack of tryptophan, thereby inhibiting T cell proliferation and blocking T cell signaling. Lost. Therefore, inhibition of overexpression of IDO function within the tumor helps to activate the collective immune system and counteract tumor growth.
  • IDO inhibitors have good application prospects in the pharmaceutical industry as a drug, but there is currently no IDO inhibitor listed.
  • the clinical IDO inhibitors are only NLG-0919 developed by New Link Genetics and INCB-24360 from Incyte and BMS-985205 from Bristol-Myers Squibb.
  • existing clinical IDO inhibitors have problems such as CYP inhibition (NLG 0919), large doses, short half-lives, and high number of administrations (INCB-24360). Therefore, IDO inhibitors without the above disadvantages are still unmet medical needs.
  • NLG-0919, INCB-24360 and BMS-986205 are as follows:
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof,
  • Ring A is selected from a 5-membered heteroaryl group having at least 1 N atom;
  • Y is selected from C and N;
  • R 1 is selected from H, C 1-6 alkyl, C 1-6 heteroalkyl, and the C 1-6 alkyl, C 1-6 heteroalkyl is optionally substituted by 1, 2 or 3 R;
  • R 2 , R 3 , R 4 and R 5 are each independently selected from H and C 1-6 alkyl;
  • R 4 and R 5 are bonded together to form a 3- to 4-membered cycloalkyl group, and the 3- to 4-membered cycloalkyl group is optionally substituted by 1, 2 or 3 R;
  • R 6 , R 7 and R 8 are each independently selected from the group consisting of H, halogen, OH, CN, NH 2 and C 1-3 alkyl, and the C 1-3 alkyl group is optionally 1, 2 or 3 R 9 Replace
  • R 9 is independently selected from the group consisting of CN, F, Cl, Br, and I;
  • R is independently selected from the group consisting of halogen, OH, CN, NH 2 , C 1-4 alkyl, C 1-4 heteroalkyl, C 3-6 cycloalkyl, 3-6 -membered heterocycloalkyl, phenyl, and a 5- to 6-membered heteroaryl group, said C 1-4 alkyl group, C 1-4 heteroalkyl group, C 3-6 cycloalkyl group, 3 to 6 membered heterocycloalkyl group, phenyl group and 5 to 6 membered hetero
  • the aryl group is optionally substituted by 1, 2 or 3 R';
  • R' is independently selected from the group consisting of F, Cl, Br, I, OH, CN, NH 2 , CH 3 , CH 3 O, CH 3 CH 2 , CH 3 CH 2 O, CH(CH 3 ) 2 ,
  • the number of heteroatoms or heteroatoms is independently selected from 1, 2 or 3.
  • the above R is selected from the group consisting of F, Cl, Br, I, OH, CN, NH 2 , CH 3 , CH 3 O, CH 3 CH 2 , CH 3 CH 2 O, CH(CH 3 ) 2 , N(CH 3 ) 2 , CF 3 , CH 2 F, CHF 2 ,
  • R 1 above is selected from the group consisting of H, CH 3 , CH 3 CH 2 , CH(CH 3 ) 2 , CH 3 O, CH 3 CH 2 O, and N(CH 3 ) 2 , said CH 3 , CH 3 CH 2 , CH(CH 3 ) 2 , CH 3 O, CH 3 CH 2 O and N(CH 3 ) 2 are optionally substituted by 1, 2 or 3 R.
  • R 1 is selected from the group consisting of H, CH 3 , CH 3 CH 2 , CH(CH 3 ) 2 , CH 3 O, CH 3 CH 2 O, N(CH 3 ) 2 , CH 2 OH , CF 3 , CH 2 F, CHF 2 and
  • R 2 , R 3 , R 4 and R 5 are each independently selected from the group consisting of H, CH 3 , CH 3 CH 2 and CH(CH 3 ) 2 .
  • R 6 , R 7 and R 8 are each independently selected from the group consisting of F, Cl, Br, I, OH, CN, NH 2 , CH 3 , CH 3 CH 2 , CH(CH 3 ) 2 , CF 3 , CH 2 F and CHF 2 .
  • Ring A is selected from the group consisting of pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl.
  • the above R is selected from the group consisting of F, Cl, Br, I, OH, CN, NH 2 , CH 3 , CH 3 O, CH 3 CH 2 , CH 3 CH 2 O, CH(CH 3 ) 2 , N(CH 3 ) 2 , CF 3 , CH 2 F, CHF 2 , Other variables are as defined above.
  • R 1 above is selected from the group consisting of H, CH 3 , CH 3 CH 2 , CH(CH 3 ) 2 , CH 3 O, CH 3 CH 2 O, and N(CH 3 ) 2 , said CH 3 , CH 3 CH 2 , CH(CH 3 ) 2 , CH 3 O, CH 3 CH 2 O and N(CH 3 ) 2 are optionally substituted by 1, 2 or 3 R, and other variables are as defined above.
  • R 1 is selected from the group consisting of H, CH 3 , CH 3 CH 2 , CH(CH 3 ) 2 , CH 3 O, CH 3 CH 2 O, N(CH 3 ) 2 , CH 2 OH , CF 3 , CH 2 F, CHF 2 and Other variables are as defined above.
  • R 2 , R 3 , R 4 and R 5 are each independently selected from the group consisting of H, CH 3 , CH 3 CH 2 and CH(CH 3 ) 2 , and other variables are as defined above.
  • R 6 , R 7 and R 8 are each independently selected from the group consisting of F, Cl, Br, I, OH, CN, NH 2 , CH 3 , CH 3 CH 2 , CH(CH 3 ) 2 , CF 3 , CH 2 F and CHF 2 , other variables are as defined above.
  • the structural unit Selected from Other variables are as defined above.
  • Ring A is selected from the group consisting of pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl, and other variables are as defined above.
  • the structural unit Selected from Other variables are as defined above.
  • the structural unit Selected from Other variables are as defined above.
  • the above compound is selected from the group consisting of
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are as defined above.
  • the present invention also provides a compound or a pharmaceutically acceptable salt thereof, selected from the group consisting of
  • the present invention also provides the use of the above compound or a pharmaceutically acceptable salt thereof for the preparation of a medicament for treating cancer.
  • the present invention provides a novel structure of a selective IDO inhibitor having such excellent in vitro IDO enzyme activity; since the molecular weight is small, it is expected to have better solubility and permeability, thereby making it more medicinal.
  • the compound of the invention has good metabolic stability, good oral bioavailability and excellent pharmacokinetic properties.
  • pharmaceutically acceptable salt refers to a salt of a compound of the invention prepared from a compound having a particular substituent found in the present invention and a relatively non-toxic acid or base.
  • a base addition salt can be obtained by contacting a neutral amount of such a compound with a sufficient amount of a base in a neat solution or a suitable inert solvent.
  • Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amine or magnesium salts or similar salts.
  • an acid addition salt can be obtained by contacting a neutral form of such a compound with a sufficient amount of an acid in a neat solution or a suitable inert solvent.
  • pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, hydrogencarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, Hydrogen sulfate, hydroiodic acid, phosphorous acid, etc.; and an organic acid salt, such as acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, Similar acids such as fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, and me
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound containing an acid group or a base by conventional chemical methods.
  • such salts are prepared by reacting these compounds in water or an organic solvent or a mixture of the two via a free acid or base form with a stoichiometric amount of a suitable base or acid.
  • the compounds provided herein also exist in the form of prodrugs.
  • Prodrugs of the compounds described herein are readily chemically altered under physiological conditions to convert to the compounds of the invention.
  • prodrugs can be converted to the compounds of the invention by chemical or biochemical methods in an in vivo setting.
  • Certain compounds of the invention may exist in unsolvated or solvated forms, including hydrated forms.
  • the solvated forms are equivalent to the unsolvated forms and are included within the scope of the invention.
  • the compounds of the invention may exist in specific geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including the cis and trans isomers, the (-)- and (+)-p-enantiomers, the (R)- and (S)-enantiomers, and the diastereomeric a conformation, a (D)-isomer, a (L)-isomer, and a racemic mixture thereof, and other mixtures, such as enantiomerically or diastereomeric enriched mixtures, all of which belong to It is within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in the substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the invention.
  • enantiomer or “optical isomer” refer to stereoisomers that are mirror images of one another.
  • cis-trans isomer or “geometric isomer” is caused by the inability to freely rotate a single bond due to a double bond or a ring-forming carbon atom.
  • diastereomer refers to a stereoisomer in which the molecule has two or more chiral centers and the molecules are in a non-mirrored relationship.
  • wedge-shaped dashed keys Represents the absolute configuration of a solid center with straight solid keys
  • straight dashed keys Indicates the relative configuration of the stereocenter, using wavy lines Indicates a wedge solid key Or wedge-shaped dotted key Or with wavy lines Represents a straight solid key And straight dashed keys
  • tautomer or “tautomeric form” mean that the different functional isomers are in dynamic equilibrium at room temperature and can be rapidly converted into each other. If tautomers are possible (as in solution), the chemical equilibrium of the tautomers can be achieved.
  • proton tautomers also known as prototropic tautomers
  • prototropic tautomers include interconversions by proton transfer, such as keto-enol isomerization and imine-enes. Amine isomerization.
  • the valence tautomer includes the mutual transformation of some of the bonding electrons.
  • keto-enol tautomerization is the interconversion between two tautomers of pentane-2,4-dione and 4-hydroxypent-3-en-2-one.
  • the terms "enriched in one isomer”, “isomer enriched”, “enriched in one enantiomer” or “enantiomeric enriched” refer to one of the isomers or pairs
  • the content of the oligo is less than 100%, and the content of the isomer or enantiomer is 60% or more, or 70% or more, or 80% or more, or 90% or more, or 95% or more, or 96% or more, or 97% or more, 98% or more, 99% or more, 99.5% or more, 99.6% or more, 99.7% or more, 99.8% or more, or greater than or equal to 99.9%.
  • the term “isomer excess” or “enantiomeric excess” refers to the difference between the two isomers or the relative percentages of the two enantiomers. For example, if one of the isomers or enantiomers is present in an amount of 90% and the other isomer or enantiomer is present in an amount of 10%, the isomer or enantiomeric excess (ee value) is 80%. .
  • optically active (R)- and (S)-isomers as well as the D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If an enantiomer of a compound of the invention is desired, it can be prepared by asymmetric synthesis or by derivatization with a chiral auxiliary wherein the resulting mixture of diastereomers is separated and the auxiliary group cleaved to provide pure The desired enantiomer.
  • a diastereomeric salt is formed with a suitable optically active acid or base, followed by conventional methods well known in the art.
  • the diastereomers are resolved and the pure enantiomer is recovered.
  • the separation of enantiomers and diastereomers is generally accomplished by the use of chromatography using a chiral stationary phase, optionally in combination with chemical derivatization (eg, formation of an amino group from an amine). Formate).
  • the compounds of the present invention may contain unnatural proportions of atomic isotopes on one or more of the atoms that make up the compound.
  • radiolabeled compounds can be used, such as tritium (3 H), iodine -125 (125 I) or C-14 (14 C).
  • hydrogen can be replaced by heavy hydrogen to form a deuterated drug.
  • the bond composed of barium and carbon is stronger than the bond composed of common hydrogen and carbon.
  • deuterated drugs have reduced side effects and increased drug stability. Enhance the efficacy and prolong the biological half-life of the drug. Alterations of all isotopic compositions of the compounds of the invention, whether radioactive or not, are included within the scope of the invention.
  • pharmaceutically acceptable carrier refers to any formulation or carrier medium that is capable of delivering an effective amount of an active substance of the present invention, does not interfere with the biological activity of the active substance, and has no toxic side effects to the host or patient, including water, oil, Vegetables and minerals, cream bases, lotion bases, ointment bases, etc. These bases include suspending agents, tackifiers, transdermal enhancers and the like. Their formulations are well known to those skilled in the cosmetic or topical pharmaceutical arts.
  • excipient generally refers to the carrier, diluent and/or vehicle required to formulate an effective pharmaceutical composition.
  • an "effective amount” or “therapeutically effective amount” with respect to a pharmaceutical or pharmacologically active agent refers to a sufficient amount of a drug or agent that is non-toxic but that achieves the desired effect.
  • an "effective amount” of an active substance in a composition refers to the amount required to achieve the desired effect when used in combination with another active substance in the composition. The determination of the effective amount will vary from person to person, depending on the age and general condition of the recipient, and also on the particular active substance, and a suitable effective amount in a case can be determined by one skilled in the art based on routine experimentation.
  • active ingredient refers to a chemical entity that is effective in treating a target disorder, disease or condition.
  • substituted means that any one or more hydrogen atoms on a particular atom are replaced by a substituent, and may include variants of heavy hydrogen and hydrogen, as long as the valence of the particular atom is normal and the substituted compound is stable. of.
  • any variable eg, R
  • its definition in each case is independent.
  • the group may optionally be substituted with at most two R, and each case has an independent option.
  • combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
  • linking group When the number of one linking group is 0, such as -(CRR) 0 -, it indicates that the linking group is a single bond.
  • one of the variables When one of the variables is selected from a single bond, it means that the two groups to which it is attached are directly linked. For example, when L represents a single bond in A-L-Z, the structure is actually A-Z.
  • hetero denotes a hetero atom or a hetero atomic group (ie, a radical containing a hetero atom), including atoms other than carbon (C) and hydrogen (H), and radicals containing such heteroatoms, including, for example, oxygen (O).
  • ring means substituted or unsubstituted cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, cycloalkynyl, heterocycloalkynyl, aryl or heteroaryl.
  • the rings include monocyclic, bicyclic, and polycyclic systems, wherein the bicyclic and polycyclic systems include bicyclic, spiro, bicyclic, and bridged rings.
  • the number of atoms on the ring is usually defined as the number of elements of the ring. For example, "5 to 7-membered ring" means 5 to 7 atoms arranged in a circle. Unless otherwise specified, the ring optionally contains from 1 to 3 heteroatoms.
  • 5- to 7-membered ring includes, for example, phenyl, pyridine, and piperidinyl; on the other hand, the term “5- to 7-membered heterocyclic” includes pyridyl and piperidinyl, but does not include phenyl.
  • ring also includes ring systems containing at least one ring, each of which "ring” independently conforms to the above definition.
  • alkyl is used to mean a straight or branched saturated hydrocarbon group, and in some embodiments, the alkyl group is a C 1-12 alkyl group; in other embodiments The alkyl group is a C 1-6 alkyl group; in other embodiments, the alkyl group is a C 1-3 alkyl group. It may be monosubstituted (such as -CH 2 F) or polysubstituted (such as -CF 3 ), and may be monovalent (such as methyl), divalent (such as methylene) or polyvalent (such as methine). .
  • alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, s-butyl) And t-butyl), pentyl (including n-pentyl, isopentyl and neopentyl), hexyl and the like.
  • alkenyl is used to indicate a straight or branched hydrocarbon group containing one or more carbon-carbon double bonds, and the carbon-carbon double bond may be located at any position of the group.
  • the alkenyl group is a C 2-8 alkenyl group; in other embodiments, the alkenyl group is a C 2-6 alkenyl group; in other embodiments, the alkenyl group is C 2-4 alkenyl. It may be monosubstituted or polysubstituted, and may be monovalent, divalent or multivalent.
  • alkenyl groups include, but are not limited to, ethenyl, propenyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl and the like.
  • alkynyl is used to indicate a straight or branched hydrocarbon group containing one or more carbon-carbon triple bonds, and the carbon-carbon triple bond may be located at any position of the group.
  • the alkynyl group is a C 2-8 alkynyl group; in other embodiments, the alkynyl group is a C 2-6 alkynyl group; in other embodiments, the alkynyl group is C 2-4 alkynyl. It may be monosubstituted or polysubstituted, and may be monovalent, divalent or multivalent. Examples of alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, and the like.
  • heteroalkyl by itself or in conjunction with another term, denotes a stable straight or branched alkyl radical or a combination thereof consisting of a number of carbon atoms and at least one heteroatom or heteroatom. Things.
  • the heteroatoms are selected from the group consisting of B, O, N, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen heteroatoms are optionally quaternized.
  • the heteroalkyl group is a C1-6 heteroalkyl group; in other embodiments, the heteroalkyl group is a C1-3 heteroalkyl group.
  • a heteroatom or heteroatom can be located at any internal position of a heteroalkyl group, including the position at which the alkyl group is attached to the rest of the molecule, but the terms "alkoxy”, “alkylamino” and “alkylthio” (or thioalkyl) Oxyl) is a conventional expression and refers to those alkyl groups which are attached to the remainder of the molecule through an oxygen atom, an amino group or a sulfur atom, respectively.
  • alkoxy represents attached through an oxygen bridge
  • C 1-6 alkoxy groups include C 1, C 2, C 3 , C 4, C 5 , and C 6 alkoxy groups.
  • the alkoxy group is a C 1-3 alkoxy group.
  • alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentyloxy, and S- Pentyloxy.
  • heteroalkenyl by itself or in conjunction with another term, denotes a stable straight or branched alkenyl radical or a combination thereof consisting of a number of carbon atoms and at least one heteroatom or heteroatom. Things.
  • the heteroatoms are selected from the group consisting of B, O, N, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen heteroatoms are optionally quaternized.
  • the heteroalkenyl group is a C 2-6 heteroalkenyl group; in other embodiments, the heteroalkyl group is a C 2-4 heteroalkenyl group.
  • the hetero atom or heteroatom group may be located at any internal position of the heteroalkenyl group, including the position at which the alkenyl group is attached to the rest of the molecule, but the terms "alkenyloxy”, “alkenylamino” and “alkenylthio” are customary. By expression, it is meant those alkenyl groups which are attached to the remainder of the molecule through an oxygen atom, an amino group or a sulfur atom, respectively.
  • heteroalkynyl by itself or in conjunction with another term, denotes a stable straight or branched alkynyl radical or a combination thereof consisting of a number of carbon atoms and at least one heteroatom or heteroatom. Things.
  • the heteroatoms are selected from the group consisting of B, O, N, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen heteroatoms are optionally quaternized.
  • the heteroalkynyl group is a C 2-6 heteroalkynyl group; in other embodiments, the heteroalkyl group is a C 2-4 heteroalkynyl group.
  • hetero atom or heteroatom group may be located at any internal position of the heteroalkynyl group, including the position at which the alkynyl group is attached to the rest of the molecule, but the terms "alkynyloxy", “alkynylamino” and “alkynylthio” are customary. By expression, it is meant those alkynyl groups attached to the remainder of the molecule through an oxygen atom, an amino group or a sulfur atom, respectively.
  • heteroalkynyl groups include, but are not limited to, Up to two heteroatoms can be continuous, for example
  • cycloalkyl includes any stable cyclic alkyl group including monocyclic, bicyclic or tricyclic systems wherein the bicyclic and tricyclic systems include spiro, co and ring.
  • the cycloalkyl group is a C 3-8 cycloalkyl group; in other embodiments, the cycloalkyl group is a C 3-6 cycloalkyl group; in other embodiments, the The cycloalkyl group is a C 5-6 cycloalkyl group. It may be monosubstituted or polysubstituted, and may be monovalent, divalent or multivalent.
  • cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, [2.2.2]bicyclooctane, [4.4.0] Dicyclodecane and the like.
  • cycloalkenyl includes any stable cyclic alkenyl group containing one or more carbon-carbon double bonds at any position of the group, including monocyclic, bicyclic or tricyclic systems, wherein Bicyclic and tricyclic systems include spiro, parallel and bridged rings, but any ring of this system is non-aromatic.
  • the cycloalkenyl group is a C 3-8 cycloalkenyl group; in other embodiments, the cycloalkenyl group is a C 3-6 cycloalkenyl group; in other embodiments, the The cycloalkenyl group is a C 5-6 cycloalkenyl group.
  • cycloalkenyl groups include, but are not limited to, cyclopentenyl, cyclohexenyl, and the like.
  • cycloalkynyl includes any stable cyclic alkynyl group containing one or more carbon-carbon triple bonds at any position of the group, including monocyclic, bicyclic or tricyclic systems, wherein Bicyclic and tricyclic systems include spiro, parallel and bridging rings. It may be monosubstituted or polysubstituted, and may be monovalent, divalent or multivalent.
  • heterocycloalkyl by itself or in conjunction with other terms, denotes a cyclized “heteroalkyl”, respectively, which includes monocyclic, bicyclic, and tricyclic systems, wherein the bicyclic and tricyclic systems include spiro rings, And ring and bridge ring. Further, in the case of the "heterocycloalkyl group", a hetero atom may occupy a position where a heterocycloalkyl group is bonded to the rest of the molecule.
  • the heterocycloalkyl group is a 4-6 membered heterocycloalkyl group; in other embodiments, the heterocycloalkyl group is a 5-6 membered heterocycloalkyl group.
  • heterocycloalkyl groups include, but are not limited to, azetidinyl, oxetanyl, thioheterobutyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrothiophenyl (including tetrahydrothiophene) -2-yl and tetrahydrothiophen-3-yl, etc.), tetrahydrofuranyl (including tetrahydrofuran-2-yl, etc.), tetrahydropyranyl, piperidinyl (including 1-piperidinyl, 2-piperidinyl and 3-piperidinyl, etc.), piperazinyl (including 1-piperazinyl and
  • heterocyclenyl by itself or in conjunction with other terms, denotes a cyclized “heteroalkenyl”, respectively, which includes monocyclic, bicyclic, and tricyclic systems, wherein the bicyclic and tricyclic systems include spiro rings, Rings and bridge rings, but any ring of this system is non-aromatic.
  • a heteroatom can occupy the position of attachment of the heterocyclenyl group to the rest of the molecule.
  • the heterocycloalkenyl is 4 to 6 membered heterocycloalkenyl; in other embodiments, the heterocycloalkenyl is 5 to 6 membered heterocycloalkenyl.
  • heterocycloalkenyl groups include, but are not limited to,
  • heterocycloalkynyl by itself or in conjunction with other terms, denotes a cyclized “heteroalkynyl” group, respectively, which includes monocyclic, bicyclic, and tricyclic systems, wherein the bicyclic and tricyclic systems include spiro rings, And ring and bridge ring.
  • a hetero atom may occupy a position where a heterocyclic alkynyl group is bonded to the rest of the molecule.
  • the heterocycloalkynyl group is a 4 to 6 membered heterocycloalkynyl group; in other embodiments, the heterocycloalkynyl group is a 5 to 6 membered heterocycloalkynyl group.
  • halo or “halogen”, by itself or as part of another substituent, denotes a fluorine, chlorine, bromine or iodine atom. Further, the term “haloalkyl” is intended to include both monohaloalkyl and polyhaloalkyl.
  • halo(C 1 -C 4 )alkyl is intended to include, but is not limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like. Wait. Unless otherwise specified, examples of haloalkyl include, but are not limited to, trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl.
  • aromatic ring and “aryl” are used interchangeably and the term “aryl ring” or “aryl” means a polyunsaturated carbocyclic ring system which may be monocyclic, bicyclic or poly A ring system in which at least one ring is aromatic, and each ring in the bicyclic and polycyclic ring system is fused together or covalently linked. It may be mono- or poly-substituted, may be monovalent, divalent or multivalent, in some embodiments, the aryl is a C 6-12 aryl; in other embodiments, the aryl It is a C 6-10 aryl group.
  • Non-limiting examples of aryl groups include phenyl, naphthyl (including 1-naphthyl and 2-naphthyl, etc.), biphenyl (including 4-biphenyl, etc.).
  • the substituent of any of the above aryl ring systems is selected from the group of acceptable substituents described herein.
  • heteroaryl ring and “heteroaryl” are used interchangeably and the term “heteroaryl” means 1, 2, 3 or 4 independently selected from B, N, O and An aryl (or aromatic ring) of a hetero atom of S, which may be a monocyclic, bicyclic or tricyclic ring system in which the nitrogen atom may be substituted or unsubstituted (ie, N or NR, wherein R is H or has been herein Other substituents are defined, and are optionally quaternized, and the nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O) p , p is 1 or 2).
  • a heteroaryl group can be attached to the remainder of the molecule through a heteroatom.
  • the heteroaryl is a 5-10 membered heteroaryl; in other embodiments, the heteroaryl is a 5-6 membered heteroaryl.
  • Non-limiting examples of such heteroaryl groups include pyrrolyl (including N-pyrrolyl, 2-pyrrolyl, and 3-pyrrolyl, etc.), pyrazolyl (including 2-pyrazolyl and 3-pyrazolyl, etc.) , imidazolyl (including N-imidazolyl, 2-imidazolyl, 4-imidazolyl and 5-imidazolyl, etc.), oxazolyl (including 2-oxazolyl, 4-oxazolyl and 5-oxazolyl) Etiso), triazolyl (1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl, 1H-1,2,4-triazolyl and 4H-1,2,4 - triazolyl, etc.
  • aralkyl is intended to include those groups in which an aryl group is attached to an alkyl group.
  • the aralkyl group is a C6-10 aryl- C1-4 alkyl group.
  • the aralkyl group is a C 6-10 aryl-C 1-2 alkyl group. Examples of aralkyl groups include, but are not limited to, benzyl, phenethyl, naphthylmethyl, and the like.
  • Aryloxy and "arylthio” mean those groups wherein the carbon atom (e.g., methylene) in the aralkyl group has been replaced by an oxygen or sulfur atom, and in some embodiments, the aryloxy group is C 6-10 aryl-OC 1-2 alkyl; in other embodiments, the aryloxy group is C 6-10 aryl-C 1-2 alkylene-O-. In some embodiments, the arylthio group is a C 6-10 aryl-SC 1-2 alkyl group; in other embodiments, the arylthio group is a C 6-10 aryl-C 1-2 alkylene group. Base-S-. Examples of aryloxy and arylthio groups include, but are not limited to, phenoxymethyl, 3-(1-naphthyloxy)propyl, phenylthiomethyl, and the like.
  • heteroarylkyl is meant to include those heteroaryl groups attached to an alkyl group, in some embodiments, the heteroaryl group is a 5-8 membered heteroaryl -C 1 -4 alkyl; in other embodiments, the heteroarylalkyl group is a 5-6 membered heteroaryl-C 1-2 alkyl group.
  • heteroarylalkyl include, but are not limited to, pyrrolylmethyl, pyrazolylmethyl, pyridylmethyl, pyrimidinylmethyl, and the like.
  • Heteroaryloxy and “heteroarylthio” mean those groups wherein a carbon atom (e.g., methylene) in a heteroaralkyl group has been replaced by an oxygen or sulfur atom, respectively, and in some embodiments, the hetero The aryloxy group is a 5-8 membered heteroaryl-OC 1-2 alkyl group; in other embodiments, the heteroaryloxy group is a 5-6 membered heteroaryl-C 1-2 alkylene-O- group.
  • the heteroarylthio group is a 5-8 membered heteroaryl-SC 1-2 alkyl group; in other embodiments, the heteroarylthio group is a 5-6 membered heteroaryl-C 1 -2 alkylene-S-.
  • heteroaryloxy and heteroarylthio groups include, but are not limited to, pyrroleoxymethyl, pyrazolyloxymethyl, 2-pyridyloxymethyl, pyrrolethiomethyl, pyrazolethiomethyl, 2-pyridinethiomethyl Wait.
  • leaving group refers to a functional group or atom which may be substituted by another functional group or atom by a substitution reaction (for example, an affinity substitution reaction).
  • substituent groups include triflate; chlorine, bromine, iodine; sulfonate groups such as mesylate, tosylate, p-bromobenzenesulfonate, p-toluenesulfonic acid Esters and the like; acyloxy groups such as acetoxy, trifluoroacetoxy and the like.
  • the present invention employs the following abbreviations: eq for equivalents, equivalents; DCM for methylene chloride; PE for petroleum ether; DMF for N,N-dimethylformamide; DMSO for dimethyl sulfoxide; EtOAc for acetic acid Ester; MeOH stands for methanol; BOC stands for t-butoxycarbonyl is an amine protecting group; THF stands for tetrahydrofuran; Boc 2 O stands for di-tert-butyl dicarbonate; NCS stands for N-chlorosuccinimide; PMB Represents p-methoxybenzylamine; THP stands for 2-tetrahydrofuranyl; Pd(t-Bu 3 P) 2 stands for bis(tri-tert-butylphosphine)palladium; Na 2 SO 4 stands for anhydrous sodium sulfate; T 3 P stands for C Phosphoric acid anhydride; LiOH.H 2 O stands for lithium hydroxide hydrate; BH
  • Triethylamine (17.14 g, 169.42 mmol, 23.58 mL) was added to a solution of compound 3-b (8.2 g, 56.47 mmol) in dichloromethane. After cooling the solution to 0 ° C, the compound 1-d (9.21 g, 56.47 mmol, 5.90 mL) was added dropwise, and after the addition was completed, the reaction was carried out at 25 ° C for 12 hours, and the color of the mixture changed from colorless to brown. After adding 100 ml of dichloromethane, the mixture was washed three times with saturated aqueous ammonium chloride solution, 200 ml each time, dried over anhydrous sodium sulfate, filtered and concentrated to give compound 3-c.
  • Triethylamine (9.98 g, 98.60 mmol, 13.72 mL) and p-methoxybenzylamine (6.76 g, 49.30 mmol, 6.38 mL) were added to a solution of compound 3-c (11.6 g, 49.30 mmol) in tetrahydrofuran (110 mL). After reacting at 70 ° C for 12 hours, the color of the solution remained brown. Diluted with 300 ml of ethyl acetate, washed three times with saturated brine, 200 ml each time, dried over anhydrous sodium sulfate, filtered, and evaporated. 1) Separation and purification to obtain the product 3-d.
  • the aqueous layer was adjusted to pH 9 with a saturated sodium carbonate solution, and then extracted twice with ethyl acetate (50 mL), and the combined organic layer was washed once with 50 mL of brine, dried over anhydrous sodium sulfate 3-g.
  • Triethylamine ((18.96 g, 187.38 mmol, 26.08 mL) was added to a solution of compound 4-b (12.07 g, 62.46 mmol) in dichloromethane (100 mL). After the solution was cooled to 0 ° C, compound 1-d was added dropwise. (10.18g, 62.46mmol, 6.53mL), after the addition was completed, reacted at 25 ° C for 12 hours, the color of the mixture changed from colorless to black. Dilute with 30 ml of dichloromethane, and the mixture was washed with saturated aqueous solution of ammonium chloride. (20 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to give compound 4-c.
  • the aqueous layer was saturated with sodium carbonate The pH was adjusted to 10, then extracted with ethyl acetate (20 mL ⁇ 2), and the combined organic layer was washed twice with 20 mL of brine, dried over anhydrous sodium sulfate
  • the compound 4-i (177.00 mg, 372.63 ⁇ mol) was dissolved in a methanolic hydrochloric acid solution (4M, 5 mL) and reacted at 25 ° C for 2 hours.
  • the crude product obtained was purified by preparative high performance liquid chromatography (formic acid). Get the product 4.
  • Triethylamine (9.73 g, 96.12 mmol, 13.38 mL) and p-methoxybenzylamine (6.59 g, 48.06 mmol, 6.22 mL) were added to a solution of compound 5-d (9.96 g, 48.06 mmol) in THF (100 mL). After reacting for 12 hours at 70 ° C in a nitrogen atmosphere, the color of the solution remained black.
  • Triethylamine (63.16 g, 624.17 mmol, 86.88 mL) was added to a solution of compound 6-a (50 g, 312.08 mmol, 49.02 mL) in dichloromethane. After cooling the solution to 0 ° C, the compound 1-d (50.88 g, 312.08 mmol, 32.61 mL) was added dropwise. After the dropwise addition, the reaction was carried out at 25 ° C for 12 hours, and the color of the mixture was yellow.
  • the compound 6-b (67 g, 267.66 mmol) was added to a 4M aqueous solution of dioxane (600 mL), and the mixture was reacted at 25 ° C for 1 hour, and the color of the solution remained yellow.
  • the crude product 6-c obtained was directly concentrated.
  • test compound is added with DMSO to form a high concentration storage solution.
  • IDO1 enzyme solution containing components: 50 mM phosphate buffer solution, 0.1% Tween 20, 2% glycerol, 20 mM L-ascorbic acid, 200 U/ml catalase (sigma), 20 ⁇ M Blue, 120nM IDO1 enzyme;
  • test plate Place the test plate on the microplate reader, set the temperature to 25 ° C, and read OD 320 at 10 min and 60 min respectively.
  • the compounds of the present invention have a significant inhibitory effect on IDO protease activity in HeLa cells.
  • the healthy adult female Balb/c mice used in this study were purchased from Shanghai Lingchang Biotechnology Co., Ltd.
  • mice Twelve female Balb/c mice were divided into 2 groups according to 6 rats in each group.
  • the blood of three animals was collected at each time point by means of cross-harvesting. 40 ⁇ L of whole blood was collected before administration and 0.0833 (intravenous only), 0.25, 0.5, 1, 2, 4, 8, and 24 hours after administration. Whole blood was placed in an anticoagulant tube, and centrifuged at 3000 g for 15 minutes at 4 ° C to prepare plasma and stored at -80 °C. The concentration of the drug in the plasma was determined by LC/MS-MS.
  • the compound of the present invention has good metabolic stability, good oral bioavailability and excellent comprehensive pharmacokinetic properties.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本发明公开了新的一类作为IDO抑制剂的化合物,具体公开了式(I)所示化合物及其药学上可接受的盐。

Description

IDO抑制剂
本申请主张如下优先权:
CN201711375318.8,申请日2017年12月19日。
技术领域
本发明涉及新的作为IDO抑制剂的化合物,具体公开了式(I)所示化合物及其药学上可接受的盐。本发明还涉及式(I)所示化合物及其药学上可接受的盐在制备治疗肿瘤药物中的应用。
背景技术
恶性肿瘤是现如今危害人类生命安全的主要疾病之一。近百年来,人类为了对抗恶性肿瘤,发展出了多种包括最常用的化疗,手术,放疗以及靶向疗法在内的诊疗手段与治疗方法。这些疗法在一定程度上延缓的肿瘤的发展,延长了患者生命。但是由于恶性肿瘤的无限制生长,浸润与转移的特性,以上疗法依旧无法达到理想的抑制效果。同时,以上的疗法的毒副作用也是限制其应用的关键因素。
近年来,免疫疗法作为恶性肿瘤治疗的新兴疗法被发展起来,其特点是通过调动宿主的天然防御机制来抵抗肿瘤的生长。其主要途径主要通过激活宿主原有的免疫系统,增强宿主集体对肿瘤细胞的免疫反应,利用免疫系统能力达到精准有效的杀伤肿瘤,遏制其发展。
吲哚胺-吡咯-2,3-双加氧酶(Indoleeamine-pyrrole-2,3-dioxygenase,IDO)是一种含铁红素的单体蛋白,其功能在于催化了色氨酸向犬尿氨酸转变的决速步骤。IDO在多种肿瘤细胞内有过表达。过度表达的IDO可以迅速消耗肿瘤微环境内色氨酸,使得浸润至肿瘤内部的T细胞由于缺乏色氨酸而停滞于G1期中期,从而抑制了T细胞增殖并使T细胞信号传导受阻,功能丧失。因此,抑制肿瘤内部过表达的IDO功能有助于激活集体免疫系统,抵制肿瘤生长。
IDO抑制剂作为药物在医药行业具有良好的应用前景,但是目前尚无IDO抑制剂上市。处于临床的IDO抑制剂只有New Link Genetics公司开发的NLG-0919与Incyte公司的INCB-24360及百时美施贵宝的BMS-985205。但是,现有临床IDO抑制剂存在诸如CYP抑制(NLG 0919)、给药剂量大、半衰期短、给药次数多(INCB-24360)等问题。因此,无上述缺点的IDO抑制剂依旧是未被满足的医疗需求。
NLG-0919,INCB-24360及BMS-986205的具体结构如下:
Figure PCTCN2018122015-appb-000001
发明内容
本发明提供式(I)化合物或其药学上可接受的盐,
Figure PCTCN2018122015-appb-000002
其中,
Figure PCTCN2018122015-appb-000003
为单键或双键;
环A选自5元杂芳基,所述5元杂芳基上至少含有1个N原子;
Y选自C和N;
R 1选自H、C 1-6烷基、C 1-6杂烷基,所述C 1-6烷基、C 1-6杂烷基任选被1、2或3个R取代;
R 2、R 3、R 4和R 5分别独立地选自H和C 1-6烷基;
或者R 4和R 5连接在一起,形成3~4元环烷基,所述3~4元环烷基任选被1、2或3个R取代;
R 6、R 7和R 8分别独立地选自H、卤素、OH、CN、NH 2和C 1~3烷基,所述C 1~3烷基任选被1、2或3个R 9取代;
R 9分别独立地选自CN、F、Cl、Br和I;
R分别独立地选自卤素、OH、CN、NH 2、C 1-4烷基、C 1-4杂烷基、C 3-6环烷基、3~6元杂环烷基、苯基和5~6元杂芳基,所述C 1-4烷基、C 1-4杂烷基、C 3~6环烷基、3~6元杂环烷基、苯基和5~6元杂芳基任选被1、2或3个R’取代;
R’分别独立地选自F、Cl、Br、I、OH、CN、NH 2、CH 3、CH 3O、CH 3CH 2、CH 3CH 2O、CH(CH 3) 2
N(CH 3) 2、CF 3、CH 2F和CHF 2
所述5元杂芳基、5~6元杂芳基、3~6元杂环烷基、C 1-6杂烷基和C 1-4杂烷基分别包含1、2、独立地选自-C(=O)-、N、-NH-、-O-、-S-;
以上任何一种情况下,杂原子或杂原子团的数目分别独立地选自1、2或3。
在本发明的一些方案中,上述R选自F、Cl、Br、I、OH、CN、NH 2、CH 3、CH 3O、CH 3CH 2、CH 3CH 2O、CH(CH 3) 2、N(CH 3) 2、CF 3、CH 2F、CHF 2
Figure PCTCN2018122015-appb-000004
在本发明的一些方案中,上述R 1选自H、CH 3、CH 3CH 2、CH(CH 3) 2、CH 3O、CH 3CH 2O和N(CH 3) 2,所述CH 3、CH 3CH 2、CH(CH 3) 2、CH 3O、CH 3CH 2O和N(CH 3) 2任选被1、2或3个R取代。
在本发明的一些方案中,上述R 1选自H、CH 3、CH 3CH 2、CH(CH 3) 2、CH 3O、CH 3CH 2O、N(CH 3) 2、CH 2OH、CF 3、CH 2F、CHF 2
Figure PCTCN2018122015-appb-000005
在本发明的一些方案中,上述R 2、R 3、R 4和R 5分别独立地选自H、CH 3、CH 3CH 2和CH(CH 3) 2
在本发明的一些方案中,上述R 6、R 7和R 8分别独立地选自F、Cl、Br、I、OH、CN、NH 2、CH 3、CH 3CH 2、CH(CH 3) 2、CF 3、CH 2F和CHF 2
在本发明的一些方案中,上述结构单元
Figure PCTCN2018122015-appb-000006
选自
Figure PCTCN2018122015-appb-000007
Figure PCTCN2018122015-appb-000008
在本发明的一些方案中,上述结构单元
Figure PCTCN2018122015-appb-000009
选自
Figure PCTCN2018122015-appb-000010
Figure PCTCN2018122015-appb-000011
在本发明的一些方案中,上述环A选自吡咯基、吡唑基、咪唑基、三唑基、噁唑基、异噁唑基、噻唑基和异噻唑基。
在本发明的一些方案中,上述结构单元
Figure PCTCN2018122015-appb-000012
选自
Figure PCTCN2018122015-appb-000013
在本发明的一些方案中,上述结构单元
Figure PCTCN2018122015-appb-000014
选自
Figure PCTCN2018122015-appb-000015
Figure PCTCN2018122015-appb-000016
在本发明的一些方案中,上述R选自F、Cl、Br、I、OH、CN、NH 2、CH 3、CH 3O、CH 3CH 2、CH 3CH 2O、CH(CH 3) 2、N(CH 3) 2、CF 3、CH 2F、CHF 2
Figure PCTCN2018122015-appb-000017
其他变量如上述所定义。
在本发明的一些方案中,上述R 1选自H、CH 3、CH 3CH 2、CH(CH 3) 2、CH 3O、CH 3CH 2O和N(CH 3) 2,所述CH 3、CH 3CH 2、CH(CH 3) 2、CH 3O、CH 3CH 2O和N(CH 3) 2任选被1、2或3个R取代,其他变量如上述所定义。
在本发明的一些方案中,上述R 1选自H、CH 3、CH 3CH 2、CH(CH 3) 2、CH 3O、CH 3CH 2O、N(CH 3) 2、CH 2OH、CF 3、CH 2F、CHF 2
Figure PCTCN2018122015-appb-000018
其他变量如上述所定义。
在本发明的一些方案中,上述R 2、R 3、R 4和R 5分别独立地选自H、CH 3、CH 3CH 2和CH(CH 3) 2,其他变量如上述所定义。
在本发明的一些方案中,上述R 6、R 7和R 8分别独立地选自F、Cl、Br、I、OH、CN、NH 2、CH 3、CH 3CH 2、CH(CH 3) 2、CF 3、CH 2F和CHF 2,其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2018122015-appb-000019
选自
Figure PCTCN2018122015-appb-000020
Figure PCTCN2018122015-appb-000021
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2018122015-appb-000022
选自
Figure PCTCN2018122015-appb-000023
Figure PCTCN2018122015-appb-000024
Figure PCTCN2018122015-appb-000025
其他变量如上述所定义。
在本发明的一些方案中,上述环A选自吡咯基、吡唑基、咪唑基、三唑基、噁唑基、异噁唑基、噻唑基和异噻唑基,其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2018122015-appb-000026
选自
Figure PCTCN2018122015-appb-000027
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2018122015-appb-000028
选自
Figure PCTCN2018122015-appb-000029
Figure PCTCN2018122015-appb-000030
其他变量如上述所 定义。
在本发明的一些方案中,上述化合物选自
Figure PCTCN2018122015-appb-000031
其中,R 1、R 2、R 3、R 4、R 5、R 6、R 7和R 8如上述所定义。
本发明还提供化合物或其药学上可接受的盐,选自
Figure PCTCN2018122015-appb-000032
Figure PCTCN2018122015-appb-000033
本发明还提供上述化合物或其药学上可接受的盐在制备治疗癌症药物中的应用。
本发明还有一些方案是由上述各变量任意组合而来。
技术效果
本发明提供一种新型结构的选择性IDO抑制剂,具有此优异的体外IDO酶活性;由于分子量较小,预计会有更好的溶解度和渗透性,从而成药性更强。本发明化合物小鼠体内代谢稳定性好,具有较好的口服生物利用度,药代动力学性质优良。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。这里所采用的术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物的中性形式接触的方式获得碱加成盐。药学上可接受的碱加成盐包括钠、钾、钙、铵、有机胺或镁盐或类似的盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物的中性形式接触的方式获得酸加成盐。药学上可接受的酸加成盐的实例包括无机酸盐,所述无机酸包括例如盐酸、氢溴酸、硝酸、碳酸,碳酸氢根,磷酸、磷酸一氢根、磷酸二氢根、硫酸、硫酸氢根、氢碘酸、亚磷酸等;以及有机酸盐,所述有机酸包括如乙酸、丙酸、异丁酸、马来酸、丙二酸、苯甲酸、琥珀酸、辛二酸、反丁烯二酸、乳酸、扁桃酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸、酒石酸和甲磺酸等类似的酸;还包括氨基酸(如精氨酸等)的盐,以及如葡糖醛酸等有机酸的盐。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换成任一碱或酸加成盐。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与化学计量的适当的碱或酸反应来制备。
除了盐的形式,本发明所提供的化合物还存在前药形式。本文所描述的化合物的前药容易地在生理条件下发生化学变化从而转化成本发明的化合物。此外,前体药物可以在体内环境中通过化学或生化方法被转换到本发明的化合物。
本发明的某些化合物可以以非溶剂化形式或者溶剂化形式存在,包括水合物形式。一般而言,溶剂化形式与非溶剂化的形式相当,都包含在本发明的范围之内。
本发明的化合物可以存在特定的几何或立体异构体形式。本发明设想所有的这类化合物,包括顺式和反式异构体、(-)-和(+)-对对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。烷基等取代基中可存在另外的不对称碳原子。所有这些异构体以及它们的混合物,均包括在本发明的范围之内。
除非另有说明,术语“对映异构体”或者“旋光异构体”是指互为镜像关系的立体异构体。
除非另有说明,术语“顺反异构体”或者“几何异构体”系由因双键或者成环碳原子单键不能自由旋转而引起。
除非另有说明,术语“非对映异构体”是指分子具有两个或多个手性中心,并且分子间为非镜像的关系的立体异构体。
除非另有说明,“(D)”或者“(+)”表示右旋,“(L)”或者“(-)”表示左旋,“(DL)”或者“(±)”表示外消旋。
除非另有说明,用楔形实线键
Figure PCTCN2018122015-appb-000034
和楔形虚线键
Figure PCTCN2018122015-appb-000035
表示一个立体中心的绝对构型,用直形实线键
Figure PCTCN2018122015-appb-000036
和直形虚线键
Figure PCTCN2018122015-appb-000037
表示立体中心的相对构型,用波浪线
Figure PCTCN2018122015-appb-000038
表示楔形实线键
Figure PCTCN2018122015-appb-000039
或楔形虚线键
Figure PCTCN2018122015-appb-000040
或用波浪线
Figure PCTCN2018122015-appb-000041
表示直形实线键
Figure PCTCN2018122015-appb-000042
和直形虚线键
Figure PCTCN2018122015-appb-000043
本发明的化合物可以存在特定的。除非另有说明,术语“互变异构体”或“互变异构体形式”是指在室温下,不同官能团异构体处于动态平衡,并能很快的相互转化。若互变异构体是可能的(如在溶液中),则可以达到互变异构体的化学平衡。例如,质子互变异构体(proton tautomer)(也称质子转移互变异构体(prototropic tautomer))包括通过质子迁移来进行的互相转化,如酮-烯醇异构化和亚胺-烯胺异构化。价键异构体(valence tautomer)包括一些成键电子的重组来进行的相互转化。其中酮-烯醇互变异构化的具体实例是戊烷-2,4-二酮与4-羟基戊-3-烯-2-酮两个互变异构体之间的互变。
除非另有说明,术语“富含一种异构体”、“异构体富集”、“富含一种对映体”或者“对映体富集”指其中一种异构体或对映体的含量小于100%,并且,该异构体或对映体的含量大于等于60%,或者大于等于70%,或者大于等于80%,或者大于等于90%,或者大于等于95%,或者大于等于96%,或者大于等于97%,或者大于等于98%,或者大于等于99%,或者大于等于99.5%,或者大于等于99.6%,或者大于等于99.7%,或者大于等于99.8%,或者大于等于99.9%。
除非另有说明,术语“异构体过量”或“对映体过量”指两种异构体或两种对映体相对百分数之间的差值。例如,其中一种异构体或对映体的含量为90%,另一种异构体或对映体的含量为10%,则异构体或对映体过量(ee值)为80%。
可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本发明某化合物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的常规方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和 非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。本发明的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚( 3H),碘-125( 125I)或C-14( 14C)。又例如,可用重氢取代氢形成氘代药物,氘与碳构成的键比普通氢与碳构成的键更坚固,相比于未氘化药物,氘代药物有降低毒副作用、增加药物稳定性、增强疗效、延长药物生物半衰期等优势。本发明的化合物的所有同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。术语“药学上可接受的载体”是指能够递送本发明有效量活性物质、不干扰活性物质的生物活性并且对宿主或者患者无毒副作用的任何制剂或载体介质代表性的载体包括水、油、蔬菜和矿物质、膏基、洗剂基质、软膏基质等。这些基质包括悬浮剂、增粘剂、透皮促进剂等。它们的制剂为化妆品领域或局部药物领域的技术人员所周知。
术语“赋形剂”通常是指配制有效的药物组合物所需要载体、稀释剂和/或介质。
针对药物或药理学活性剂而言,术语“有效量”或“治疗有效量”是指无毒的但能达到预期效果的药物或药剂的足够用量。对于本发明中的口服剂型,组合物中一种活性物质的“有效量”是指与该组合物中另一种活性物质联用时为了达到预期效果所需要的用量。有效量的确定因人而异,取决于受体的年龄和一般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
术语“活性成分”、“治疗剂”,“活性物质”或“活性剂”是指一种化学实体,它可以有效地治疗目标紊乱、疾病或病症。
“任选”或“任选地”指的是随后描述的事件或状况可能但不是必需出现的,并且该描述包括其中所述事件或状况发生的情况以及所述事件或状况不发生的情况。
术语“被取代的”是指特定原子上的任意一个或多个氢原子被取代基取代,可以包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧(即=O)时,意味着两个氢原子被取代。氧取代不会发生在芳香基上。
术语“任选被取代的”是指可以被取代,也可以不被取代,除非另有规定,取代基的种类和数目在化学上可以实现的基础上可以是任意的。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。因此,例如,如果一个基团被0-2个R所取代,则所述基团可以任选地至多被两个R所取代,并且每种情况下的R都有独立的选项。此外,取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
当一个连接基团的数量为0时,比如-(CRR) 0-,表示该连接基团为单键。
当其中一个变量选自单键时,表示其连接的两个基团直接相连,比如A-L-Z中L代表单键时表示该结构实际上是A-Z。
当一个取代基为空缺时,表示该取代基是不存在的,比如A-X中X为空缺时表示该结构实际上是A。
除非另有规定,术语“杂”表示杂原子或杂原子团(即含有杂原子的原子团),包括碳(C)和氢(H)以外的原子以及含有这些杂原子的原子团,例如包括氧(O)、氮(N)、硫(S)、硅(Si)、锗(Ge)、铝(Al)、硼(B)、-O-、-S-、-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)、-S(=O) 2-,以及任选被取代的-C(=O)N(H)-、-N(H)-、-C(=NH)-、-S(=O) 2N(H)-或-S(=O)N(H)-。
除非另有规定,“环”表示被取代或未被取代的环烷基、杂环烷基、环烯基、杂环烯基、环炔基、杂环炔基、芳基或杂芳基。所述的环包括单环、双环和多环体系,其中双环和多环体系包括联环、螺环、并环和桥环。环上原子的数目通常被定义为环的元数,例如,“5~7元环”是指环绕排列5~7个原子。除非另有规定,该环任选地包含1~3个杂原子。因此,“5~7元环”包括例如苯基、吡啶和哌啶基;另一方面,术语“5~7元杂环基”包括吡啶基和哌啶基,但不包括苯基。术语“环”还包括含有至少一个环的环系,其中的每一个“环”均独立地符合上述定义。
除非另有规定,术语“烷基”用于表示直链或支链的饱和的碳氢基团,在一些实施方案中,所述烷基为C 1-12烷基;在另一些实施方案中,所述烷基为C 1-6烷基;在另一些实施方案中,所述烷基为C 1-3烷基。其可以是单取代(如-CH 2F)或多取代的(如-CF 3),可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。烷基的实例包括但不限于甲基(Me),乙基(Et),丙基(包括n-丙基和异丙基),丁基(包括n-丁基,异丁基,s-丁基和t-丁基),戊基(包括n-戊基,异戊基和新戊基)、己基等。
除非另有规定,“烯基”用于表示直链或支链的包含一个或多个碳-碳双键的碳氢基团,碳-碳双键可以位于该基团的任何位置上。在一些实施方案中,所述烯基为C 2-8烯基;在另一些实施方案中,所述烯基为C 2-6烯基;在另一些实施方案中,所述烯基为C 2-4烯基。其可以是单取代或多取代的,可以是一价、二价或者多价。烯基的实例包括但不限于乙烯基,丙烯基,丁烯基,戊烯基,己烯基,丁间二烯基,戊间二烯基,己间二烯基等。
除非另有规定,“炔基”用于表示直链或支链的包含一个或多个碳-碳三键的碳氢基团,碳-碳三键可以位于该基团的任何位置上。在一些实施方案中,所述炔基为C 2-8炔基;在另一些实施方案中,所述炔基为C 2-6炔基;在另一些实施方案中,所述炔基为C 2-4炔基。其可以是单取代或多取代的,可以是一价、二价或者多价。炔基的实例包括但不限于乙炔基,丙炔基,丁炔基,戊炔基等。
除非另有规定,术语“杂烷基”本身或者与另一术语联合,表示由一定数目碳原子和至少一个杂原子或杂原子团组成的,稳定的直链或支链的烷基原子团或其组合物。在一些实施方案中,杂原子选自B、O、N和S,其中氮和硫原子任选地被氧化,氮杂原子任选地被季铵化。在另一些实施方案中,杂原子团选自-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)、-S(=O) 2-、-C(=O)N(H)-、-N(H)-、-C(=NH)-、-S(=O) 2N(H)-和-S(=O)N(H)-。在一些实施方案中,所述杂烷基为C 1-6杂烷基;在另一些实施方案中,所述杂烷基为C 1-3杂烷基。杂原子或杂原子团可以位于杂烷基的任何内部位置,包括该烷基与分子其余部分的连接位置,但术语“烷氧基”、“烷氨基”和“烷硫基”(或硫代烷氧基)属于惯用表达,是指分别通过一个氧原子、氨基或硫原子连接到分子的其余部分的那些烷基基团。杂烷基的实例包括但不限于-OCH 3、-OCH 2CH 3、-OCH 2CH 2CH 3、-OCH 2(CH 3) 2、-CH 2-CH 2-O-CH 3、-NHCH 3、-N(CH 3) 2、-NHCH 2CH 3、-N(CH 3)(CH 2CH 3)、-CH 2-CH 2-NH-CH 3、-CH 2-CH 2-N(CH 3)-CH 3、-SCH 3、-SCH 2CH 3、-SCH 2CH 2CH 3、-SCH 2(CH 3) 2、-CH 2-S-CH 2-CH 3、-CH 2-CH 2、-S(=O)-CH 3、-CH 2-CH 2-S(=O) 2-CH 3、-CH=CH-O-CH 3、-CH 2-CH=N-OCH 3和–CH=CH-N(CH 3)-CH 3。至多两个杂原子可以是连续的,例如-CH 2-NH-OCH 3
“烷氧基”代表通过氧桥连接的具有特定数目碳原子的上述烷基,除非另有规定,C 1-6烷氧基包括C 1、C 2、C 3、C 4、C 5和C 6的烷氧基。在一些实施方案中,所述烷氧基为C 1-3烷氧基。烷氧基的实例包括但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、仲丁氧基、叔丁氧基、正戊氧基和S-戊氧基。
除非另有规定,术语“杂烯基”本身或者与另一术语联合,表示由一定数目碳原子和至少一个杂原子或杂原子团组成的,稳定的直链或支链的烯基原子团或其组合物。在一些实施方案中,杂原子选自B、O、N和S,其中氮和硫原子任选地被氧化,氮杂原子任选地被季铵化。在另一些实施方案中,杂原子团选自-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)、-S(=O) 2-、-C(=O)N(H)-、-N(H)-、-C(=NH)-、-S(=O) 2N(H)-和-S(=O)N(H)-。在一些实施方案中,所述杂烯基为C 2-6杂烯基;在另一些实施方案中,所述杂烷基为C 2-4杂烯基。杂原子或杂原子团可以位于杂烯基的任何内部位置,包括该烯基与分子其余部分的连接位置,但术语“烯基氧基”、“烯基氨基”和“烯基硫基”属于惯用表达,是指分别通过一个氧原子、氨基或硫原子连接到分子的其余部分的那些烯基基团。杂烯基的实例包括但不限于-O-CH=CH 2、-O-CH=CHCH 3、-O-CH=C(CH 3) 2、-CH=CH-O-CH 3、-O-CH=CHCH 2CH 3、-CH 2-CH=CH-OCH 3、-NH-CH=CH 2、-N(CH=CH 2)-CH 3、-CH=CH-NH-CH 3、-CH=CH-N(CH 3) 2、-S-CH=CH 2、-S-CH=CHCH 3、-S-CH=C(CH 3) 2、-CH 2-S-CH=CH 2、-S(=O)-CH=CH 2和-CH=CH-S(=O) 2-CH 3。至多两个杂原子可以是连续的,例如-CH=CH-NH-OCH 3
除非另有规定,术语“杂炔基”本身或者与另一术语联合,表示由一定数目碳原子和至少一个杂原子或杂原子团组成的,稳定的直链或支链的炔基原子团或其组合物。在一些实施方案中,杂原子选自B、O、N和S,其中氮和硫原子任选地被氧化,氮杂原子任选地被季铵化。在另一些实施方案中,杂原子团选自-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)、-S(=O) 2-、-C(=O)N(H)-、-N(H)-、-C(=NH)-、-S(=O) 2N(H)-和-S(=O)N(H)-。在一些实施方案中,所述杂炔基为C 2-6杂炔基;在另一些实施方案中,所述杂烷基为C 2-4杂炔基。杂原子或杂原子团可以位于杂炔基的任何内部位置,包括该炔基与分子其余部分的连接位置,但术语“炔基氧基”、“炔基氨基”和“炔基硫基”属于惯用表达,是指分别通过一个氧原子、氨基或硫原子连接到分子的其余部分的那些炔基基团。杂炔基的实例包括但不限于
Figure PCTCN2018122015-appb-000044
Figure PCTCN2018122015-appb-000045
Figure PCTCN2018122015-appb-000046
至多两个杂原子可以是连续的,例如
Figure PCTCN2018122015-appb-000047
除非另有规定,“环烷基”包括任何稳定的环状烷基,其包括单环、双环或者三环体系,其中双环和三环体系包括螺环、并环和桥环。在一些实施方案中,所述环烷基为C 3-8环烷基;在另一些实施方案中,所述环烷基为C 3-6环烷基;在另一些实施方案中,所述环烷基为C 5-6环烷基。其可以是单取代或多取代的,可以是一价、二价或者多价。这些环烷基的实例包括,但不限于,环丙基、环丁基、环戊基、环己基、环庚基、降冰片烷基、[2.2.2]二环辛烷、[4.4.0]二环癸烷等。
除非另有规定,“环烯基”包括任何稳定的环状烯基,在该基团的任何位点含有一个或多个碳-碳双键,其包括单环、双环或者三环体系,其中双环和三环体系包括螺环、并环和桥环,但是此体系的任意环都是非芳香性的。在一些实施方案中,所述环烯基为C 3-8环烯基;在另一些实施方案中,所述环烯基为C 3-6环烯基;在另一些实施方案中,所述环烯基为C 5-6环烯基。其可以是单取代或多取代的,可以是一价、二价或者多价。这些环烯基的实例包括,但不限于,环戊烯基、环己烯基等。
除非另有规定,“环炔基”包括任何稳定的环状炔基,在该基团的任何位点含有一个或多个碳-碳三键,其包含单环、双环或者三环体系,其中双环和三环体系包括螺环、并环和桥环。其可以是单取代或多取代的,可以是一价、二价或者多价。
除非另有规定,术语“杂环烷基”本身或者与其他术语联合分别表示环化的“杂烷基”,其包括单环、双环和三环体系,其中双环和三环体系包括螺环、并环和桥环。此外,就该“杂环烷基”而言,杂原子可以占据杂环烷基与分子其余部分的连接位置。在一些实施方案中,所述杂环烷基为4~6元杂环烷基;在另一些实施方案中,所述杂环烷基为5~6元杂环烷基。杂环烷基的实例包括但不限于氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢噻吩基(包括四氢噻吩-2-基和四氢噻吩-3-基等)、四氢呋喃基(包括四氢呋喃-2-基等)、四氢吡喃基、哌啶基(包括1-哌啶基、2-哌啶基和3-哌啶基等)、哌嗪基(包括1-哌嗪基和2-哌嗪基等)、吗啉基(包括3-吗啉基和4-吗啉基等)、二噁烷基、二噻烷基、异噁唑烷基、异噻唑烷基、1,2-噁嗪基、1,2-噻嗪基、六氢哒嗪基、高哌嗪基、高哌啶基或氧杂环庚烷基。
除非另有规定,术语“杂环烯基”本身或者与其他术语联合分别表示环化的“杂烯基”,其包括单环、双环和三环体系,其中双环和三环体系包括螺环、并环和桥环,但是此体系的任意环都是非芳香性的。此外,就该“杂环烯基”而言,杂原子可以占据杂环烯基与分子其余部分的连接位置。在一些实施方案中,所述杂环烯基为4~6元杂环烯基;在另一些实施方案中,所述杂环烯基为5~6元杂环烯基。杂环烯基的实例包括但不限于
Figure PCTCN2018122015-appb-000048
Figure PCTCN2018122015-appb-000049
除非另有规定,术语“杂环炔基”本身或者与其他术语联合分别表示环化的“杂炔基”,其包括单环、双环和三环体系,其中双环和三环体系包括螺环、并环和桥环。此外,就该“杂环炔基”而言,杂原子可以占据杂环炔基与分子其余部分的连接位置。在一些实施方案中,所述杂环炔基为4~6元杂环炔基;在另一些实施方案中,所述杂环炔基为5~6元杂环炔基。除非另有规定,术语“卤代素”或“卤素”本身或作为另一取代基的一部分表示氟、氯、溴或碘原子。此外,术语“卤代烷基”意在包括单卤代烷基和多卤代烷基。例如,术语“卤代(C 1-C 4)烷基”意在包括但不仅限于三氟甲基、2,2,2-三氟乙基、4-氯丁基和3-溴丙基等等。除非另有规定,卤代烷基的实例包括但不仅限于:三氟甲基、三氯甲基、五氟乙基,和五氯乙基。
除非另有规定,本发明术语“芳环”和“芳基”可以互换使用,术语“芳环”或“芳基”表示多不饱和的碳环体系,它可以是单环、双环或多环体系,其中至少一个环是芳香性的,所述双环和多环体系中的各个环稠合在一起或共价连接。其可以是单取代或多取代的,可以是一价、二价或者多价,在一些实施方案中,所述芳基为C 6-12芳基;在另一些实施方案中,所述芳基为C 6-10芳基。芳基的非限制性实施例包括苯基、萘基(包括1-萘基和2-萘基等)、联苯基(包括4-联苯基等)。、上述任意一个芳基环系的取代基选自本发明所述的可接受的取代基。
除非另有规定,本发明术语“杂芳环”和“杂芳基”可以互换使用,术语“杂芳基”是指含有1、2、3或4个独立选自B、N、O和S的杂原子的芳基(或芳环),其可以是单环、双环或三环体系,其中氮原子可以是 被取代的或未取代的(即N或NR,其中R是H或本文已经定义过的其他取代基),且任选地被季铵化,氮和硫杂原子可任选被氧化(即NO和S(O) p,p是1或2)。杂芳基可通过杂原子连接到分子的其余部分。在一些实施方案中,所述杂芳基为5-10元杂芳基;在另一些实施方案中,所述杂芳基为5-6元杂芳基。所述杂芳基的非限制性实施例包括吡咯基(包括N-吡咯基、2-吡咯基和3-吡咯基等)、吡唑基(包括2-吡唑基和3-吡唑基等)、咪唑基(包括N-咪唑基、2-咪唑基、4-咪唑基和5-咪唑基等)、噁唑基(包括2-噁唑基、4-噁唑基和5-噁唑基等)、三唑基(1H-1,2,3-三唑基、2H-1,2,3-三唑基、1H-1,2,4-三唑基和4H-1,2,4-三唑基等)、四唑基、异噁唑基(3-异噁唑基、4-异噁唑基和5-异噁唑基等)、噻唑基(包括2-噻唑基、4-噻唑基和5-噻唑基等)、呋喃基(包括2-呋喃基和3-呋喃基等)、噻吩基(包括2-噻吩基和3-噻吩基等)、吡啶基(包括2-吡啶基、3-吡啶基和4-吡啶基等)、吡嗪基、嘧啶基(包括2-嘧啶基和4-嘧啶基等)、苯并噻唑基(包括5-苯并噻唑基等)、嘌呤基、苯并咪唑基(包括2-苯并咪唑基等)、吲哚基(包括5-吲哚基等)、异喹啉基(包括1-异喹啉基和5-异喹啉基等)、喹喔啉基(包括2-喹喔啉基和5-喹喔啉基等)、喹啉基(包括3-喹啉基和6-喹啉基等)、吡嗪基、2-苯基-4-噁唑基、嘌呤基、苯基并噁唑基。上述任意一个杂芳基环系的取代基选自本发明所述的可接受的取代基。
除非另有规定,术语“芳烷基”意在包括芳基附着于烷基的那些基团,在一些实施方案中,所述芳烷基为C 6-10芳基-C 1-4烷基;在另一些实施方案中,所述芳烷基为C 6-10芳基-C 1-2烷基。芳烷基的实例包括但不限于苄基、苯乙基、萘甲基等。“芳氧基”和“芳硫基”分别表示芳烷基中的碳原子(如亚甲基)已经被氧或硫原子代替的那些基团,在一些实施方案中,所述芳氧基为C 6-10芳基-O-C 1-2烷基;在另一些实施方案中,芳氧基为C 6-10芳基-C 1-2亚烷基-O-。在一些实施方案中,所述芳硫基为C 6-10芳基-S-C 1-2烷基;在另一些实施方案中,芳硫基为C 6-10芳基-C 1-2亚烷基-S-。芳氧基和芳硫基的实例包括但不限于苯氧基甲基、3-(1-萘氧基)丙基,苯硫基甲基等。
除非另有规定,术语“杂芳烷基”意在包括杂芳基附着于烷基的那些基团,在一些实施方案中,所述杂芳烷基为5-8元杂芳基-C 1-4烷基;在另一些实施方案中,所述杂芳烷基为5-6元杂芳基-C 1-2烷基。杂芳烷基的实例包括但不限于吡咯基甲基、吡唑基甲基、吡啶基甲基、嘧啶基甲基等。“杂芳氧基”和“杂芳硫基”分别表示杂芳烷基中的碳原子(如亚甲基)已经被氧或硫原子代替的那些基团,在一些实施方案中,所述杂芳氧基为5-8元杂芳基-O-C 1-2烷基;在另一些实施方案中,杂芳氧基为5-6元杂芳基-C 1-2亚烷基-O-。在一些实施方案中,所述杂芳硫基为5-8元杂芳基-S-C 1-2烷基;在另一些实施方案中,杂芳硫基为5-6元杂芳基-C 1-2亚烷基-S-。杂芳氧基和杂芳硫基的实例包括但不限于吡咯氧甲基、吡唑氧甲基、2-吡啶氧甲基、吡咯硫甲基、吡唑硫甲基、2-吡啶硫甲基等。
术语“离去基团”是指可以被另一种官能团或原子通过取代反应(例如亲和取代反应)所取代的官能团或原子。例如,代表性的离去基团包括三氟甲磺酸酯;氯、溴、碘;磺酸酯基,如甲磺酸酯、甲苯磺酸酯、对溴苯磺酸酯、对甲苯磺酸酯等;酰氧基,如乙酰氧基、三氟乙酰氧基等等。
本发明采用下述缩略词:eq代表当量、等量;DCM代表二氯甲烷;PE代表石油醚;DMF代表N,N-二甲基甲酰胺;DMSO代表二甲亚砜;EtOAc代表乙酸乙酯;MeOH代表甲醇;BOC代表叔丁氧羰基是一种胺保护基团;THF代表四氢呋喃;Boc 2O代表二-叔丁基二碳酸酯;NCS代表N-氯代丁二酰亚胺;PMB代表对甲氧基苄胺;THP代表2-四氢呋喃基;Pd(t-Bu 3P) 2代表二(三叔丁基膦)钯;Na 2SO 4代 表无水硫酸钠;T 3P代表丙基磷酸酐;LiOH.H 2O代表水合氢氧化锂;BH3-Me2S代表硼烷二甲硫醚溶液;t-BuONa代表叔丁醇钠;TEA代表三乙胺;MeCN代表乙腈;K 2CO 3代表碳酸钾;NH 4Cl代表氯化铵;H 2O代表水;NaOH代表氢氧化钠;BH 3-Me 2S代表硼烷二甲硫醚溶液;Pd(OH) 2/C代表氢氧化钯/碳;N 2代表氮气;HCl代表盐酸;FA代表甲酸;n-BμLi代表正丁基锂;MeI代表碘甲烷;LiHMDS代表六甲基二硅基胺基锂的四氢呋喃溶液。
具体实施方式
下面通过实施例对本发明进行详细描述,但并不意味着对本发明任何不利限制。本发明的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。对本领域的技术人员而言,在不脱离本发明精神和范围的情况下针对本发明具体实施方式进行各种变化和改进将是显而易见的。
实施例1
Figure PCTCN2018122015-appb-000050
Figure PCTCN2018122015-appb-000051
化合物1-c的合成
将化合物1-a(10g,59.88mmol,6.62mL)溶解在MeCN(100mL)中,并在20℃下分批加入1-b(10.80g,239.52mmol,15.67mL),K 2CO 3(24.83g,179.64mmol).所得混合物在氮气保护下,20℃下搅拌12小时。将反应物过滤后,滤液减压浓缩,得到化合物1-c。
化合物1-e的合成
将化合物1-c(6g,45.74mmol)溶解于DCM(60mL)中,并在20℃下往溶液中加入TEA(13.89g,137.22mmol,19.10mL).并在0℃下加入1-d(7.46g,45.74mmol,4.78mL),所得混合物在20℃下氮气保护下搅拌1小时.将反应液用DCM(75mL×2)萃取除去有机杂质.将所得有机相混合并用饱和NH 4Cl(50mL×3),洗涤Na 2SO 4干燥,过滤旋干后得到粗品产物1-e。
化合物1-f的合成
将化合物1-e(8.49g,38.37mmol)溶解于THF(90mL)中,并在20℃下往溶液中加入TEA(11.65g,115.11mmol,16.02mL)和1-m(5.26g,38.37mmol,4.97mL).所得混合物在70℃下搅拌12小时.通过液相质谱检 测至反应完成.将反应液用EtOAc(135mL)萃取除去有机杂质.将所得有机相混合并用饱和NH4Cl(45mL×3),洗涤Na 2SO4干燥,将粗品通过柱层析(石油醚/乙酸乙酯=1:0到0:1)纯化得到1-f。
化合物1-g的合成
将化合物1-f(3g,8.37mmol)溶解于H 2O(30mL)和MeOH(30mL)中,并在20℃下往溶液中加入LiOH.H 2O(3.51g,83.69mmol).所得混合物在20℃下搅拌12小时.将反应物减压浓缩后,用EtOAc(30mL×3)萃取.分液,水相冷却到0℃加入2M HCl调节pH=7,过滤得到粗品1-g。
化合物1-h的合成
将化合物1-g(4.6g,13.92mmol)溶解于DMF(50mL)中,并在20℃下往溶液中加入T 3P(13.29g,20.88mmol,12.42mL)和TEA(4.23g,41.77mmol,5.81mL).所得混合物在氮气保护下20℃搅拌12小时.将反应物加入水(100ml)后,用EtOAc(50mL×3)萃取.合并有机相用饱和食盐水洗(50mL×3),Na 2SO 4干燥,过滤浓缩后得到粗品1-h。
化合物1-i的合成
将化合物1-h(637mg,2.04mmol)溶解于THF(10mL)中,并在0℃下往溶液中加入BH 3-Me 2S(10M,1.02mL).所得混合物在氮气保护下25℃搅拌2小时.将反应物加入水(20ml)后加入1M HCl调节pH=1,用EtOAc(10mL×3)萃取.分液.水相加入2M NaOH调节pH=10,用EtOAc(10mL×3)萃取,合并有机相用饱和食盐水洗(10mL×3),Na 2SO 4干燥,过滤浓缩后得到粗品1-i。
化合1-j的合成
将化合物1-i(170mg,569.70μmol)溶解于MeOH(5mL)中,并在20℃下往溶液中加入Pd(OH) 2/C(40.00mg,纯度20%),所得混合物在氢气保护下45℃搅拌1小时.将反应物过滤浓缩后得到粗品1-j.
化合物1-l的合成
将化合物1-k(200mg,633.73μmol),化合物1-j(112.96mg,633.73μmol),Pd(PtBu 3) 2(32.38mg,63.37μmol,0.1eq)和tBuONa(182.70mg,1.90mmol,3eq)溶于THF(5mL),用N 2置换三次,所得棕色混合物在N 2氛围70℃搅拌2小时.将混合物倒入水30mL和EtOAc(30mL)30mL中.有机相分离后用水30mL洗涤,Na 2SO 4干燥,过滤浓缩得到化合物1-l.
化合物1的合成
将化合物1-l(280mg,678.07μmol)溶于MeOH(10mL)后加入HCl/MeOH(4M,847.59μL).所得混合物在25℃下搅拌3小时。将混合物倒入水30mL和EtOAc(30mL)中,分离得到有机相用水30mL洗涤,Na 2SO 4干燥,过滤浓缩后得到粗品,粗品通过制备色谱纯化(甲酸体系)得到化合物1。
1H NMR(400MHz,氘代氯仿)δ=7.99(s,1H),6.96(s,1H),6.26(d,J=1.3Hz,1H),4.07(m,4H),3.70-3.59(m,2H),3.57-3.46(m,2H),3.10(q,J=7.2Hz,2H),1.06(t,J=7.1Hz,3H);
实施例2
Figure PCTCN2018122015-appb-000052
化合物2-a的合成
将化合物1-j(500mg,2.81mmol)溶解于THF(10mL)中,并在25℃下往溶液中加入TEA(851.52mg,8.42mmol,1.17mL)和Boc 2O(642.80mg,2.95mmol,676.63μL)。所得混合物在25℃下氮气保护下搅拌12小时.向溶液中加入饱和NH4Cl(15mL),将混合液用EtOAc(15mL×3)萃取除去有机杂质.将所得有机相混合并用饱和NH 4Cl(15mL×3)洗涤Na 2SO 4干燥,过滤旋干后得到粗品产物2-a。
化合物2-b的合成
将化合物2-a(500mg,1.80mmol)溶解于THF(5mL)中,用氮气置换三次,并在-70℃下往溶液中缓慢加入n-BμLi(2.5M,3.59mL)。所得混合物在-70℃下搅拌1小时.然后在此温度下加入MeI(5.10g,35.92mmol,2.24mL)。所得混合溶液在氮气保护下,25℃下反应11小时。将反应液加入H 2O(15ml),用EtOAc(15mL×3)萃取除去有机杂质.将所得有机相混合并用饱和食盐水(15mL×3),洗涤Na 2SO 4干燥,将粗品通过制备色谱(甲酸体系).纯化,得到2-b.
化合物2-c的合成
将化合物2-b(100mg,326.35μmol)溶解于MeOH(5mL)中,并在25℃下往溶液中加入HCl/MeOH(4M,5mL).所得混合物在25℃下搅拌1小时.将反应物减压浓缩后,得到粗品2-c。
化合物2-d的合成
将化合物2-c(50mg,242.36μml),1-k(69.53mg,220.33μmol),t-BuONa(63.52mg,660.98μmol)溶解于THF(5mL)中,氮气置换三次后,往溶液中加入Pd(t-Bu 3P) 2(11.26mg,22.03μmol,0.1eq).所得混合物在氮气保护下70℃搅拌3小时.向反应物中加入EtOAc(15ml).过滤,滤液用饱和食盐水洗(10mL×2),Na 2SO 4干燥,过滤浓缩后得到粗品2-d.
化合物2的合成
向化合物2-d(77mg,174.61μmol)中加入HCl/MeOH(4M,2mL).所得混合溶液在25℃搅拌1小时.通过液相质谱检测至反应完成.减压浓缩后得到粗品。粗品通过制备色谱纯化后,得到化合物2.
1H NMR(400MHz,DMSO-d 6)δ=13.05(br s,1H),8.25(s,1H),6.87(s,1H),6.43(s,1H),3.95(t,J=6.1Hz,2H),3.88(s,2H),3.58-3.45(m,2H),3.32-3.23(m,2H),1.32-1.17(m,6H),1.08(t,J=7.0Hz,3H)
实施例3
Figure PCTCN2018122015-appb-000053
化合物3-b的合成
向化合物1-a(10g,59.88mmol,6.62mL)的乙腈溶液(100mL)中加入碳酸钾(24.83g,179.64mmol)和化合物3-a(10.62g,179.64mmol,15.43mL),在25℃下反应12小时,悬浊液一直保持无色。加入200毫升乙酸乙酯后过滤,滤液浓缩得到化合物3-b。
化合物物3-c的合成
向化合物3-b(8.2g,56.47mmol)的二氯甲烷溶液(100mL)中加入三乙胺(17.14g,169.42mmol,23.58mL)。将溶液冷却到0℃后滴加化合物1-d(9.21g,56.47mmol,5.90mL),滴加完后在25℃下反应12小时,混合物的颜色从无色变为棕色。加入100毫升二氯甲烷,混合物用饱和的氯化铵水溶液洗涤3次,每次200毫升,无水硫酸钠干燥,过滤,浓缩得到化合物3-c。
化合物物3-d的合成
向化合物3-c(11.6g,49.30mmol)的四氢呋喃溶液(110mL)中加入三乙胺(9.98g,98.60mmol,13.72mL)和对甲氧基苄氨(6.76g,49.30mmol,6.38mL),在70℃下反应12小时后,溶液的颜色保持棕色。加入300毫升乙酸乙酯稀释,用饱和食盐水洗3次,每次200毫升,无水硫酸钠干燥,过滤,浓缩得到的粗品通过快速硅胶柱(洗脱液:石油醚/乙酸乙酯=1:1)分离纯化得到产品3-d。
化合物物3-e的合成
向化合物3-d(13.5g,36.24mmol)的甲醇(70mL)和水(70mL)的溶液中加入一水合氢氧化锂(15.21g,362.44mmol),在25℃下反应12小时后,混合物的颜色保持黄色。加入140毫升水后用乙酸乙酯萃取两次,每次100mL,用1M的盐酸溶液将水层pH调到7,过滤,滤饼用30mL水洗涤一次,然后真空干燥得到化合物3-e。
化合物物3-f的合成
向化合物3-e(4g,11.61mmol)的N,N-二甲基甲酰胺(60mL)溶液中加入三乙胺(3.53g,34.84mmol,4.85mL)和三正丙基磷酸酐(11.09g,17.42mmol,10.36mL),在氮气保护和25℃下反应12小时后,溶液的颜色保持黄色。加入150毫升乙酸乙酯稀释,用饱和氯化铵溶液洗3次,每次150毫升,再用饱和食盐水溶液洗涤(2次,每次150毫升,无水硫酸钠干燥,过滤,浓缩得到化合物3-f。
化合物物3-g的合成
在0℃下,向化合物3-f(3.7g,11.34mmol)的四氢呋喃(50mL)溶液中加入硼烷二甲硫醚(10M,5.67mL),在25℃下反应12小时后,溶液的颜色保持黄色。慢慢加入50毫升水淬灭反应,然后用1M的盐酸溶液将混合物pH调到3,用乙酸乙酯萃取两次,每次50mL。水层用饱和的碳酸钠溶液将pH调到9,然后用乙酸乙酯萃取两次,每次50mL,联合的有机层用50mL饱和食盐水溶液洗涤一次,无水硫酸钠干燥、过滤,浓缩得到化合物3-g。
化合物物3-h的合成
向化合物3-g(2.45g,7.84mmol)的甲醇(30mL)溶液中加入Pd(OH) 2/C(250mg,纯度20%),经三次氢气置换后,在氢气氛围(15psi)、25℃下反应12小时后,经硅藻土过滤,滤液浓缩得到化合物3-h。
化合物物3-i的合成
向化合物1-k(300mg,950.59μmol)和3-h(201.06mg,1.05mmol)的四氢呋喃(10mL)溶液中加入叔丁醇钠(274.07mg,2.85mmol)和二(三叔丁基磷)钯(48.58mg,95.06μmol,),在氮气氛围、70℃下反应2小时后, 溶液颜色从红色变为黑色。加入20毫升乙酸乙酯稀释,经硅藻土过滤,滤液用饱和氯化钠溶液洗2次,每次20毫升,无水硫酸钠干燥,过滤,浓缩得到化合物3-i(500mg,粗品)。产品直接用于下一步。
化合物3的合成
将化合物3-i(450mg,1.05mmol)溶于盐酸甲醇溶液(4M,15mL)中,在25℃下反应1小时后,溶液颜色保持黄色。浓缩得到的粗品通过制备级高效液相色谱(甲酸体系)制备纯化得到化合物3.
1H NMR(400MHz,DMSO-d 6)δ=13.01(s,1H),8.15(s,1H),6.84(s,1H),6.24(s,1H),4.04-3.94(m,5H),3.49-3.40(m,2H),3.39-3.35(m,2H),0.84(d,J=6.8Hz,6H).
实施例4
Figure PCTCN2018122015-appb-000054
化合物物4-b的合成
在25℃下向化合物1-a(10g,59.88mmol,6.62mL)的乙腈溶液(100mL)中加入碳酸钾(24.83g,179.64mmol)。然后化合物4-a(10.62g,179.64mmol,15.43mL)加入混合液中,在25℃下反应12小时,悬浊液一直保持无色。加入200毫升二氯甲烷后过滤,滤液加入饱和氯化铵洗涤萃取,然后加入无水硫酸钠干燥,浓缩得到化合物4-b。
化合物物4-c的合成
向化合物4-b(12.07g,62.46mmol)的二氯甲烷溶液(100mL)中加入三乙胺((18.96g,187.38mmol,26.08mL)。将溶液冷却到0℃后滴加化合物1-d(10.18g,62.46mmol,6.53mL),滴加完后在25℃下反应12小时,混合物的颜色从无色变为黑色。加入30毫升二氯甲烷稀释,混合物用饱和的氯化铵水溶液洗涤(20mL×3),无水硫酸钠干燥,过滤,浓缩得到化合物4-c。
化合物物4-d的合成
向化合物4-c(10.31g,36.39mmol)的四氢呋喃溶液(100mL)中加入三乙胺(7.36g,72.77mmol,10.13mL)和对甲氧基苄氨(4.99g,36.39mmol,4.71mL),在70℃下反应12小时后,溶液的颜色保持黑色。直接浓缩得到的粗品通过快速硅胶柱(洗脱液:石油醚/乙酸乙酯=0:1)分离纯化得到产品4-d。
化合物物4-e的合成
向化合物4-d(11.34g,26.97mmol)的甲醇(50mL)和水(50mL)的溶液中加入一水合氢氧化锂(11.32g,269.67mmol,10eq),在25℃下反应12小时后,混合物的颜色保持无色。用乙酸乙酯萃取(30mL×3),用1M的盐酸溶液将水层pH调到7,过滤,然后将滤饼真空干燥得到化合物4-e。
化合物物4-f的合成
向化合物4-e(6.69g,17.05mmol)的N,N-二甲基甲酰胺(50mL)溶液中加入三乙胺(5.17g,51.14mmol,7.12mL)和三正丙基磷酸酐((16.27g,25.57mmol,15.21mL),在氮气保护和25℃下反应12小时后,溶液的颜色保持黄色。加入30毫升乙酸乙酯稀释萃取,用饱和食盐水溶液洗涤(60mL×3),无水硫酸钠干燥,过滤,浓缩得到化合物4-f。
化合物物4-g的合成
在0℃下,向化合物4-f(5.19g,13.87mmol)的四氢呋喃(60mL)溶液中加入硼烷二甲硫醚((10M,6.93mL),在25℃下反应12小时后,溶液的颜色保持黄色。慢慢加入60毫升水淬灭反应,然后用1M的盐酸溶液将混合物pH调到3,用甲基叔丁基醚萃取两次,每次20mL。水层用饱和的碳酸钠溶液将pH调到10,然后用乙酸乙酯萃取(20mL×2),合并有机层用20mL饱和食盐水溶液洗1次,无水硫酸钠干燥,过滤,浓缩得到化合物4-g。
化合物物4-h的合成
向化合物4-g(200mg,554.83μmol)的甲醇(10mL)溶液中加入氢氧化钯(38.96mg,277.42μmol),经三次氢气置换后,在氢气环境下(15psi)和50℃下反应2小时后,用20M甲醇稀释后经硅藻土过滤,滤液浓缩得到4-h。
化合物物4-i的合成
向化合物1-k(130.13mg,412.33μmol)和4-h(109.00mg,453.56μmol)的四氢呋喃(10mL)溶液中加入叔丁醇 钠(118.88mg,1.24mmol)和二(三叔丁基磷)钯(21.07mg,41.23μmol),在氮气保护和70℃下反应3小时后,溶液颜色变为黑色。加入20毫升乙酸乙酯稀释,经硅藻土过滤,滤液用饱和氯化钠溶液洗2次,每次20毫升,无水硫酸钠干燥,过滤,浓缩得到化合物4-i。
化合物物4的合成
将化合物4-i(177.00mg,372.63μmol)溶于盐酸甲醇溶液(4M,5mL)中,在25℃下反应2小时后,浓缩得到的粗品通过制备级高效液相色谱(甲酸体系)制备纯化得到产品4.
1H NMR(400MHz,DMSO-d 6)δ=13.04(s,1H),8.13(s,1H),7.34-7.24(m,3H),7.17(d,J=6.5Hz,2H),6.90(s,1H),6.26(s,1H),4.17(s,2H),4.06(t,J=5.2Hz,2H),3.97(t,J=5.7Hz,2H),3.76-3.63(m,2H),3.39(t,J=5.6Hz,2H).
实施例5
Figure PCTCN2018122015-appb-000055
Figure PCTCN2018122015-appb-000056
化合物物5-c的合成
在25℃下向化合物5-b(7.16g,106.08mmol)的乙腈溶液(10mL)中加入碳酸钾(33.83g,244.80mmol)。然后化合物5-a(10g,81.60mmol,8.70mL)加入混合液中,在25℃下反应12小时,悬浊液一直保持无色。浓缩得到5-c。
化合物物5-d的合成
向化合物5-c(6.92g,59.07mmol)的二氯甲烷溶液(20mL)中加入三乙胺(17.93g,177.21mmol,24.67mL)。将溶液冷却到0℃后滴加化合物1-d(9.63g,59.07mmol,6.17mL),滴加完后在25℃下反应12小时,混合物的颜色从无色变为黑色。加入20毫升二氯甲烷稀释,混合物用饱和的氯化铵溶液洗3次,每次20毫升,无水硫酸钠干燥,过滤,浓缩得到物5-d。
化合物物5-e的合成
向化合物5-d(9.96g,48.06mmol)的四氢呋喃溶液(100mL)中加入三乙胺(9.73g,96.12mmol,13.38mL)和对甲氧基苄氨(6.59g,48.06mmol,6.22mL),在氮气氛围、70℃下反应12小时后,溶液的颜色保持黑色。直接浓缩得到的粗品通过快速硅胶柱(洗脱液:石油醚/乙酸乙酯=0:1)分离纯化得到5-e。
化合物物5-f的合成
向化合物5-e(4.72g,13.70mmol)的甲醇(50mL)和水(50mL)的溶液中加入一水合氢氧化锂(5.75g,137.04mmol),在25℃下反应12小时后,混合物的颜色保持无色。用乙酸乙酯萃取2次,每次20mL,在0℃下用1M的盐酸溶液将水层pH调到7,过滤,然后将滤饼真空干燥得到化合物5-f。
化合物物5-g的合成
向化合物5-f(1.5g,4.74mmol)的N,N-二甲基甲酰胺(10mL)溶液中加入三乙胺(1.44g,14.22mmol,1.98mL,)和三正丙基磷酸酐(4.53g,7.11mmol,4.23mL),在氮气氛围、25℃下反应12小时后,溶液的颜色保持黄色。加入20毫升乙酸乙酯稀释萃取,用饱和氯化钠溶液洗2次,每次20毫升,无水硫酸钠干燥,过滤,浓缩得到化合物5-g。
化合物物5-h的合成
在0℃下,向化合物5-g(1.17g,3.92mmol)的四氢呋喃(10mL)溶液中加入硼烷二甲硫醚(10M,1.96mL),在25℃下反应12小时后,溶液的颜色保持黄色。慢慢加入10毫升水淬灭反应,然后用1M的5mL盐酸溶液将混合物pH调到3,用甲基叔丁基醚萃取两次,每次10mL。水层用饱和的碳酸钠溶液将pH调到10,然后用乙酸乙酯萃取(20mL×2),合并有机层用20mL饱和的氯化钠溶液洗1次,无水硫酸钠干燥,过滤,浓缩得到化合物5-h。
化合物物5-i的合成
向化合物5-h(552mg,1.94mmol)的甲醇(10mL)溶液中加入氢氧化钯(136.30mg,纯度25%),经三次氢气置换后,在氢气氛围(15psi)、50℃下反应2小时后,经硅藻土过滤,将滤液浓缩得到化合物5-i。
化合物物5-j的合成
向化合物5-j(310mg,1.89mmol)和1-k(541.57mg,1.72mmol)的四氢呋喃(10mL)溶液中加入叔丁醇钠(494.75mg,5.15mmol)和二(三叔丁基磷)钯(87.70mg,171.60μmol),在氮气氛围、70℃下反应3小时后,溶液颜色保持红色。加入20毫升乙酸乙酯稀释,经硅藻土过滤,滤液用饱和氯化钠溶液洗2次,每次20毫升,无水硫酸钠干燥,过滤,浓缩得到化合物5-j。
化合物物5的合成
将化合物5-j(358mg,897.45μmol)溶于盐酸甲醇溶液(4M)和甲醇溶液(10mL,)中,在25℃下反应1小时后,溶液颜色保持无色。浓缩得到的粗品通过制备级高效液相色谱(甲酸体系)制备纯化得到化合物5. 1H NMR(400MHz,DMSO-d 6)δ=13.06(s,1H),8.11(s,1H),6.88(s,1H),6.22(s,1H),3.99(t,J=4.8Hz,2H),3.90(t,J=5.8Hz,2H),3.70-3.45(m,4H),2.71(s,3H).
实施例6
Figure PCTCN2018122015-appb-000057
Figure PCTCN2018122015-appb-000058
化合物物6-b的合成
向化合物6-a(50g,312.08mmol,49.02mL)的二氯甲烷溶液(500mL)中加入三乙胺(63.16g,624.17mmol,86.88mL)。将溶液冷却到0℃后滴加化合物1-d(50.88g,312.08mmol,32.61mL),滴加完后在25℃下反应12小时,混合物的颜色为黄色。加入30毫升二氯甲烷萃取,混合物用饱和的氯化铵溶液洗2次,每次20毫升,无水硫酸钠干燥,过滤,浓缩得到化合物6-b。
化合物物6-c的合成
把化合物6-b(67g,267.66mmol)加入4M盐酸二氧六环溶液(600mL)中,在25℃下反应1小时后,溶液的颜色保持黄色。直接浓缩得到的粗品产品6-c。
化合物物6-d的合成
向化合物6-c(58.82g,391.61mmol)(粗品)的甲醇(50mL)的溶液中加入三乙胺(118.88g,1.17mol,163.52mL),在25℃下反应24小时后,混合物的颜色保持黄色。直接浓缩得到的产品6-d。
化合物物6-e的合成
向化合物6-d(47.59mg,316.86μmol),化合物1-k(50mg,158.43μmol)混合的N,N-二甲基甲酰胺(5mL)溶液中加入叔丁醇钠(45.68mg,475.30μmol)和二(三叔丁基磷)钯(8.10mg,15.84μmol),在氮气氛围、70℃下反应3小时后,溶液的颜色保持红色。加入20毫升乙酸乙酯萃取,经硅藻土过滤,滤液用饱和氯化钠溶液洗2次,每次10毫升,饱和氯化铵溶液洗2次,每次10毫升,无水硫酸钠干燥,过滤,浓缩得到的粗品通过制备级高效液相色谱(甲酸体系)制备纯化得到产品6-e。
化合物物6的合成
将化合物6-e(38mg,98.73μmol)溶于盐酸甲醇溶液(4M,5mL)和甲醇溶液(5mL)中,在25℃下反应1小时后,溶液颜色保持无色。浓缩得到的粗品通过制备级高效液相色谱(甲酸体系)制备纯化得到化合物6. 1H NMR(400MHz,DMSO-d 6)δ=13.01(s,1H),8.12(s,1H),7.38(s,1H),6.84(s,1H),6.20(s,1H),4.02(t,J=5.2Hz,2H),3.92(t,J=5.7Hz,2H),3.49(t,J=5.2Hz,2H),3.30-3.24(m,2H)。
实施例7
Figure PCTCN2018122015-appb-000059
化合物7-b的合成
将化合物7-a(1g,3.35mmol)溶解在THF(10mL)中,氮气置换三次,并在-70℃下缓慢加入LiHMDS(1M,16.76mL),在此温度下反应一小时后,依然在此温度下缓慢加入MeI(9.51g,67.02mmol,4.17mL,).所得混合物在氮气保护下,25℃下搅拌11小时.将反应物加入水(20ml)后,用EtOAc(15mL×3)萃取.合并有机相用饱和食盐水洗(15mL×3),Na 2SO 4干燥,过滤浓缩后得到粗品7-b.
化合物物7-c的合成
将化合物7-b(500mg,1.60mmol)溶解于MeOH(10mL)中,并在25℃下往溶液中加入氢氧化钯/C(50mg).氢气置换三次,所得混合物在50℃下氢气保护下搅拌12小时.通过液相质谱检测至反应完成.将反应物过滤浓缩后得到粗品7-c。
化合物物7-d的合成
将化合物7-c(120mg,624.09μmol),1-d(179.05mg,567.36μmol,1eq),t-BuONa(163.58mg,1.70mmol)溶解于THF(5mL)中,氮气置换三次,并在25℃下往溶液中加入Pd(t-Bu 3P) 2(29.00mg,56.74μmol).所得混合物在70℃下搅拌3小时.通过质谱检测至反应完成.将反应液加入EtOAc(15ml),过滤,滤液用饱和食盐水洗(10mL×2),Na 2SO 4干燥,过滤浓缩后得到粗品7-d.
化合物7的合成
将化合物(140mg,327.90μmol)溶解于HCl/MeOH(4M,5mL)中,所得混合物在25℃下搅拌1小时.通过质谱检测至反应完成.将反应物减压浓缩后,粗品通过制备色谱(甲酸体系)纯化得到化合物7.
1H NMR(400MHz,DMSO-d 6)δ=13.06(br s,1H),8.09(s,1H),6.88(s,1H),6.26-6.20(m,1H),4.28-4.16(m,1H),4.10-3.99(m,1H),3.68-3.41(m,2H),3.30-3.13(m,2H),2.72(qd,J=6.9,13.9Hz,1H),2.59-2.52(m,1H),1.35(d,J=6.2Hz,3H),0.92(t,J=7.1Hz,3H)。
实施例8
Figure PCTCN2018122015-appb-000060
化合物8-b的合成
将化合物8-a(5g,32.55mmol,HCl)和TEA(11.53g,113.93mmol,15.86mL)溶解于DCM(50mL)中,然后在氮气氛围0℃下,于30分钟之内逐滴加入1-d(5.31g,32.55mmol,3.40mL),所得混合物在25℃搅拌5.5小时。通过加入饱和氯化铵溶液(80mL)淬灭反应,然后用DCM(2×80mL)萃取。合并有机相用饱和食盐水洗涤(70mL×3),Na 2SO 4干燥,过滤浓缩后得到粗品,即为8-b。
化合物8-c的合成
将化合物8-b(5.4g,26.06mmol)和1-m(3.57g,26.06mmol,3.37mL,1eq)溶解于THF(40mL)中,然后加入TEA(5.27g,52.11mmol,7.25mL,2eq)。将反应液加热至70℃搅拌6小时。往反应液中加入水(40mL),浓缩反应液除去大部分THF。将混合液用乙酸乙酯(150mL)稀释,分液。有机相用氯化铵洗涤(80 mL×3)、Na 2SO 4干燥,过滤浓缩后得到粗品。所得粗品用柱层析(洗脱液,石油醚/乙酸乙酯=5/1至0/1)纯化得到8-c。
化合物8-d的合成
将化合物8-c(6.16g,17.88mmol)溶解于MeOH(60mL)和H 2O(60mL)中,然后加入LiOH.H 2O(3.00g,71.54mmol,4eq)。将反应液在25℃下搅拌12小时。通过液相质谱检测显示反应结束。将反应液浓缩除去MeOH。使用3M HCl调节溶液的pH至8,此时大量固体析出,过滤并用冰水(10mL)洗涤滤饼,将其真空干燥即得8-d。
化合物8-e的合成
将化合物8-d(4.8g,14.53mmol)溶解于DMF(30mL)中,然后在氮气氛围下加入T 3P(13.87g,21.79mmol,12.96mL)和TEA(4.41g,43.58mmol,6.07mL,3eq)。将反应液在50℃搅拌2小时。通过液相质谱检测显示目标产物形成。向反应液中加入水(70mL),用EtOAc(70mL×)萃取,合并有机相用饱和食盐水(50mL×3)洗涤,用Na 2SO 4干燥,浓缩得粗产品8-e。
化合物8-f的合成
将化合物8-e(3.9g,12.48mmol)溶解于THF(12.48mL)中,然后在0℃、氮气氛围下,于30分钟内逐滴加入BH 3-Me 2S(10M,12.48mL)。将反应液在25℃搅拌9.5小时。通过液相质谱检测显示目标产物形成。向反应液中加入水(70mL),用3M HCl调节反应液pH=1,用EtOAc(70mL×3)萃取;然后使用4M氢氧化钠调节水相pH=10,用EtOAc(70mL×3)萃取,将合并有机相用饱和食盐水(60mL×3)洗涤、Na 2SO 4干燥,浓缩得粗产品8-f。
化合物8-g的合成
将化合物8-f(1.3g,4.36mmol)溶解于MeOH(10mL)中,然后在氮气氛围下加入钯/碳(200mg,4.36mmol)和乙酸(315.00mg,5.25mmol,0.3mL),将混合液使用氢气置换三次,所得混合液在氢气氛围(50psi)、25℃搅拌3小时。通过液相质谱检测显示目标产物形成。将混合液过滤浓缩得粗产品8-g。
化合物8-h的合成
将化合物8-g(100mg,419.63μmol,CH3COOH)和1-k(120.15mg,377.67μmol)溶解于THF(4mL)中,然后在氮气氛围下加入t-BuONa(80.65mg,839.26μmol)和Pd(t-Bu3P)2(21.45mg,41.96μmol),用氮气置换三次,将混合液在70℃搅拌4小时。通过液相质谱检测显示目标产物形成。将混合液浓缩除去溶剂,加入饱和食盐水(10mL),并用乙酸乙酯(10mL×2)萃取。将合并的有机相用饱和食盐水(10mL×3)洗涤,用Na2SO4干燥、过滤,浓缩得残留物,其用制备色谱纯化得8-h。
化合物8的合成
将化合物8-h(110mg,266.39μmol)溶解于HCl/MeOH(2mL)中,之后将反应液在25℃搅拌2小时。将混合液浓缩获得粗产品。将粗品用饱和食盐水(20mL)稀释,之后用饱和Na 2CO 3溶液调节溶液pH=10。用乙酸乙酯(40mL×2)萃取。将有机相用饱和食盐水(40mL×2)洗涤,Na 2SO 4干燥,过滤,浓缩得粗产品。将粗产品使用大板纯化(PE:EtOAc=3:1)得化合物8。
1H NMR(400MHz,DMSO-d 6)δ=8.11(s,1H),6.89(s,1H),6.51(d,J=1.3Hz,1H),3.97-3.90(m,4H),3.54(br d,J=5.4Hz,2H),1.09(s,6H)。
实施例9
Figure PCTCN2018122015-appb-000061
化合物9-b的合成
将化合物9-a(800mg,2.68mmol)溶解在THF(10mL)中,氮气置换三次,并在-70℃下缓慢加入n-BμLi(2.5M,5.36mL),在此温度下反应一小时后,依然在此温度下缓慢加入9-e(5.04g,26.81mmol,2.02mL),将所得混合物在氮气保护下,25℃下搅拌11小时。将反应物加入水(30mL)后,用EtOAc(15mL×3)萃取,将有机相合并,用饱和食盐水(15mL×3)洗涤,Na 2SO 4干燥,过滤浓缩后得到粗品,粗品通过制备色谱(甲酸体系)纯化,纯化回来的溶液直接用EtOAc(20mL×3)萃取,合并有机相用饱和食盐水(3×20mL)洗涤,Na 2SO 4干燥,过滤浓缩后得到产品9-b。
化合物9-c的合成
将化合物9-b(180mg,554.81μmol,1eq)溶解于MeOH(5mL)中,并在25℃下往溶液中加入氢氧化钯/碳(20mg,纯度20%),氢气置换三次,所得混合物在50℃下氢气保护下搅拌12小时。将反应物过滤浓缩后得到粗品9-c。
化合物9-d的合成
将化合物9-c(73mg,357.34μmol),1-d(102.52mg,324.85μmol),t-BuONa(93.66mg,974.55μmol)溶解于THF(5mL)中,氮气置换三次,并在25℃下往溶液中加入Pd(t-Bu 3P) 2(16.60mg,32.49μmol),所得混合物在70℃下搅拌12小时。向反应液中加入EtOAc(15mL,过滤,滤液用饱和食盐水(10mL×2)洗涤,Na 2SO 4干燥,过滤浓缩得粗品9-d。
化合物9的合成
将化合物9-d(120mg,273.37μmol)溶解于HCl/MeOH(4M,3mL)中,所得混合物在25℃下搅拌1小时。通过液相质谱检测至反应完成,将反应物减压浓缩后,粗品通过制备色谱纯化得到化合物9。
1H NMR(400MHz,DMSO-d 6)δ=13.02(br s,1H),8.17(s,1H),6.85(s,1H),6.26(s,1H),4.13-3.97(m,2H),3.89(s,2H),3.62-3.47(m,2H),3.29-3.15(m,2H),2.08(s,1H),1.31-1.15(m,4H),1.07(t,J=7.0Hz,3H)。
实施例10
Figure PCTCN2018122015-appb-000062
化合物10的合成
将化合物10-a(100mg,432.01μmol)和1-j(115.51mg,648.01μmol)溶于THF(5.00mL)中,再加入Pd(t-Bu 3P) 2(22.08mg,43.20μmol)和t-BuONa(83.04mg,864.02μmol),所得混合物用氮气置换后,加热至70℃搅拌3小时。向反应液中加入水(50mL)中并搅拌3分钟,水相用EtOAc(50mL×3)萃取,合并有机相用饱和食盐水(50mL)洗涤,Na 2SO4干燥,过滤后减压浓缩得到粗品,粗品通过制备色谱纯化得到10。
1H NMR(400MHz,DMSO-d 6)δppm 8.31(s,1H),8.06(s,1H)7.52(s,1H),5.83(s,1H),3.94-3.91(m,2H),3.83-3.80(m,2H),3.58-3.56(m,4H),3.14-3.08(m,2H),1.02(t,J=7.2Hz,3H).
实施例11
Figure PCTCN2018122015-appb-000063
化合物11的合成
将化合物1(20mg,60.82μmol)溶于DCM(5mL)后加入NCS(8.12mg,60.82μmol),所得白色悬浊液在30℃搅拌3小时。在25℃下将混合用水(30mL)淬灭反应,然后用DCM(30mL)萃取,合并有机相用水(30mL)洗涤,Na 2SO 4干燥,过滤浓缩得到粗品,粗品通过高效液相色谱制备纯化得化合物11。 1H NMR(400MHz,CHLOROFORM-d)δ=8.03(s,1H),6.35(s,1H),6.51-6.17(m,1H),4.05(s,4H),3.68-3.56(m,2H),3.55-3.44(m,2H),3.10(d,J=7.2Hz,2H),1.08(t,J=7.1Hz,3H)
实施例12
Figure PCTCN2018122015-appb-000064
化合物12的合成
将化合物1(20mg,60.82μmol)溶于DCM(5mL)后加入NCS(8.12mg,60.82μmol),将所得白色悬浊液在30℃搅拌3小时。在25℃下用水(30mL)淬灭反应,然后用DCM(30mL)萃取,合并有机相用水(30mL)洗涤,,Na 2SO 4干燥,过滤浓缩得到粗品,粗品通过高效液相色谱制备纯化得到化合物12。 1H NMR(400MHz,CHLOROFORM-d)δ=8.22-8.04(m,1H),7.55(d,J=1.0Hz,1H),3.76-3.67(m,1H),3.74-3.66(m,3H),3.66-3.56(m,4H),3.46-3.41(m,2H),1.31(t,J=7.2Hz,3H)
实验例1化合物对IDO1酶抑制活性的评价
实验目的:
检测本发明化合物对IDO 1酶活性的抑制效应。
实验方法:
I.化合物溶液配制
1)待测化合物加DMSO配成高浓度储存溶液。
2)用DMSO稀释阳性化合物储存液,配成100倍液。
3)在工作板中第一列加入8μL上述溶液为最高浓度,3倍稀释11个浓度,配成100倍溶液。
4)取以上板中溶液0.5μL到检测板。
II. 所用试剂配制
1)2倍IDO1酶溶液,包含的组分有:50mM磷酸盐缓冲溶液,0.1%吐温20,2%甘油,20mM L-抗坏血酸,200U/ml过氧化氢酶(catalase,sigma),20μM亚甲蓝,120nM IDO1酶;
2)2倍底物(L-色氨酸)溶液:终浓度为0.1mM,用水将底物稀释至2倍(0.2mM)。
III. IDO1酶反应
1)按表1每孔加入0.5μL 100倍化合物溶液。所有对照孔加入0.5μL100%DMSO。
2)按表1每孔加入25μL 2倍IDO1酶溶液。100%效果对照孔加入25μL不含有IDO1酶的反应液。
3)将检测板1000rpm离心1min以混匀。
4)将检测板放置室温下孵育30min。
5)按表1每孔加入25μL上述-底物(L-色氨酸)溶液。
6)将检测板1000rpm离心1分钟以混匀。
7)将检测板置于酶标仪,设置温度为25℃,分别在10min,60min读OD 320
实验数据分析结果:见表1。
表1体外筛选试验结果
受试化合物 IC 50(nM)
1 730
2 400
5 700
10 290
结论:本发明的化合物对Hela细胞内的IDO蛋白酶活性具有明显的抑制作用。
实验例2:化合物药代动力学的评价
试验动物
本研究使用的健康的成年雌性Balb/c小鼠,均购自上海灵畅生物科技有限公司。
药物的配制
静脉注射组给药溶液的配制
精确称量适量化合物,加入一定量的PEG400超声2分钟后得到澄清溶液,然后加入适量的纯水。得到终浓度为1mg/mL的澄清溶液,给药溶媒为60%PEG400+40%纯水。静脉注射组溶液在给药前用2um的滤膜进行过滤。
口服给组给药溶液的配制
精确称量适量化合物,加入一定量的PEG400超声2分钟后得到澄清溶液,然后加入适量的纯水。得到终浓度为10mg/mL的混悬溶液,给药溶媒为60%PEG400+40%纯水。
给药
12只雌性Balb/c小鼠,按照每组6只,分为2组。
第一组,静脉注射给予2mg/kg的化合物1;第二组,灌胃给予50mg/kg化合物1。
样品采集
采用交叉采血方式,每个时间点采集三只动物的血液。分别于给药前及给药后0.0833(仅静注组)、0.25、0.5、1、2、4、8、24小时采集40μL全血。将全血至于抗凝管中,在4℃条件先,3000g离心15分钟,制备血浆并保存于-80℃。应用LC/MS-MS测定血浆中的药物浓度。
实验结果:
见表2。
表2化合物药代动力学的评价实验结果
Figure PCTCN2018122015-appb-000065
结论:本发明化合物小鼠体内代谢稳定性好,具有较好的口服生物利用度,综合药代动力学性质优。

Claims (14)

  1. 式(I)化合物或其药学上可接受的盐,
    Figure PCTCN2018122015-appb-100001
    其中,
    Figure PCTCN2018122015-appb-100002
    为单键或双键;
    环A选自5元杂芳基,所述5元杂芳基上至少含有1个N原子;
    Y选自C和N;
    R 1选自H、C 1-6烷基、C 1-6杂烷基,所述C 1-6烷基、C 1-6杂烷基任选被1、2或3个R取代;
    R 2、R 3、R 4和R 5分别独立地选自H和C 1-6烷基;
    或者R 4和R 5连接在一起,形成3~4元环烷基,所述3~4元环烷基任选被1、2或3个R取代;
    R 6、R 7和R 8分别独立地选自H、卤素、OH、CN、NH 2和C 1~3烷基,所述C 1~3烷基任选被1、2或3个R 9取代;
    R 9分别独立地选自CN、F、Cl、Br和I;
    R分别独立地选自卤素、OH、CN、NH 2、C 1-4烷基、C 1-4杂烷基、C 3-6环烷基、3~6元杂环烷基、苯基和5~6元杂芳基,所述C 1-4烷基、C 1-4杂烷基、C 3~6环烷基、3~6元杂环烷基、苯基和5~6元杂芳基任选被1、2或3个R’取代;
    R’分别独立地选自F、Cl、Br、I、OH、CN、NH 2、CH 3、CH 3O、CH 3CH 2、CH 3CH 2O、CH(CH 3) 2、N(CH 3) 2、CF 3、CH 2F和CHF 2
    所述5元杂芳基、5~6元杂芳基、3~6元杂环烷基、C 1-6杂烷基和C 1-4杂烷基分别包含1、2、独立地选自-C(=O)-、N、-NH-、-O-、-S-;
    以上任何一种情况下,杂原子或杂原子团的数目分别独立地选自1、2或3。
  2. 根据权利要求1所述的化合物或其药学上可接受的盐,其中,R选自F、Cl、Br、I、OH、CN、NH 2、CH 3、CH 3O、CH 3CH 2、CH 3CH 2O、CH(CH 3) 2、N(CH 3) 2、CF 3、CH 2F、CHF 2
    Figure PCTCN2018122015-appb-100003
  3. 根据权利要求1或2所述的化合物或其药学上可接受的盐,其中,R 1选自H、CH 3、CH 3CH 2、CH(CH 3) 2、CH 3O、CH 3CH 2O和N(CH 3) 2,所述CH 3、CH 3CH 2、CH(CH 3) 2、CH 3O、CH 3CH 2O和N(CH 3) 2任选被1、2或3个R取代。
  4. 根据权利要求3所述的化合物或其药学上可接受的盐,其中,R 1选自H、CH 3、CH 3CH 2、CH(CH 3) 2、 CH 3O、CH 3CH 2O、N(CH 3) 2、CH 2OH、CF 3、CH 2F、CHF 2
    Figure PCTCN2018122015-appb-100004
  5. 根据权利要求1所述的化合物或其药学上可接受的盐,其中,R 2、R 3、R 4和R 5分别独立地选自H、CH 3、CH 3CH 2和CH(CH 3) 2
  6. 根据权利要求1所述的化合物或其药学上可接受的盐,其中,R 6、R 7和R 8分别独立地选自F、Cl、Br、I、OH、CN、NH 2、CH 3、CH 3CH 2、CH(CH 3) 2、CF 3、CH 2F和CHF 2
  7. 根据权利要求1所述的化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2018122015-appb-100005
    选自
    Figure PCTCN2018122015-appb-100006
  8. 根据权利要求7所述的化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2018122015-appb-100007
    选自
    Figure PCTCN2018122015-appb-100008
    Figure PCTCN2018122015-appb-100009
  9. 根据权利要求1所述的化合物或其药学上可接受的盐,其中,环A选自吡咯基、吡唑基、咪唑基、三唑基、噁唑基、异噁唑基、噻唑基和异噻唑基。
  10. 根据权利要求9所述的化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2018122015-appb-100010
    选自
    Figure PCTCN2018122015-appb-100011
  11. 根据权利要求9所述的化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2018122015-appb-100012
    选自
    Figure PCTCN2018122015-appb-100013
  12. 根据权利要求1~6任意一项或权利要求9所述的化合物或药学上可接受的盐,化合物选自
    Figure PCTCN2018122015-appb-100014
    其中,R 1、R 2、R 3、R 4、R 5、R 6、R 7和R 8如权利要求1~6或9所定义。
  13. 化合物或其药学上可接受的盐,选自
    Figure PCTCN2018122015-appb-100015
  14. 根据权利要求1-13任意一项所述的化合物或其药学上可接受的盐在制备治疗癌症药物中的应用。
PCT/CN2018/122015 2017-12-19 2018-12-19 Ido抑制剂 WO2019120212A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201880074353.2A CN111356692B (zh) 2017-12-19 2018-12-19 Ido抑制剂

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201711375318 2017-12-19
CN201711375318.8 2017-12-19

Publications (1)

Publication Number Publication Date
WO2019120212A1 true WO2019120212A1 (zh) 2019-06-27

Family

ID=66993093

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/122015 WO2019120212A1 (zh) 2017-12-19 2018-12-19 Ido抑制剂

Country Status (3)

Country Link
CN (1) CN111356692B (zh)
TW (1) TW201927777A (zh)
WO (1) WO2019120212A1 (zh)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017079519A1 (en) * 2015-11-06 2017-05-11 Incyte Corporation Heterocyclic compounds as pi3k-gamma inhibitors
CN106905256A (zh) * 2017-03-06 2017-06-30 中国药科大学 苯并五元杂环类ido1抑制剂、其制备方法及应用

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6927216B2 (en) * 2000-10-03 2005-08-09 Bristol-Myers Squibb Pharma Company Cyclic sulfonyl compounds as inhibitors of metalloproteases
AU2002323406A1 (en) * 2001-08-30 2003-03-18 Merck And Co., Inc. Tyrosine kinase inhibitors
WO2006062982A2 (en) * 2004-12-07 2006-06-15 Locus Pharmaceuticals, Inc. Urea inhibitors of map kinases
BR112015000675B1 (pt) * 2012-07-13 2022-07-12 UCB Biopharma SRL Derivados de imidazopiridina como moduladores da atividade de tnf
DK3125883T3 (da) * 2014-04-04 2020-10-19 Iomet Pharma Ltd Indolderivater til anvendelse i medicin
CN114539284A (zh) * 2016-03-16 2022-05-27 库拉肿瘤学公司 经取代的menin-mll抑制剂及使用方法

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017079519A1 (en) * 2015-11-06 2017-05-11 Incyte Corporation Heterocyclic compounds as pi3k-gamma inhibitors
CN106905256A (zh) * 2017-03-06 2017-06-30 中国药科大学 苯并五元杂环类ido1抑制剂、其制备方法及应用

Also Published As

Publication number Publication date
CN111356692A (zh) 2020-06-30
CN111356692B (zh) 2021-09-03
TW201927777A (zh) 2019-07-16

Similar Documents

Publication Publication Date Title
CN112300195B (zh) 四并环类化合物及其制备方法和应用
WO2021175199A1 (zh) 一类芳香杂环类化合物及其在药物中的应用
CN110446712B (zh) 作为A2A受体抑制剂的[1,2,4]三唑并[1,5-c]嘧啶衍生物
CN113321654B (zh) 作为激酶抑制剂的稠合吡啶酮类化合物
CN110156786A (zh) 嘧啶并环化合物及其制备方法和应用
WO2019154365A1 (zh) 一种atr抑制剂及其应用
CN112955432B (zh) 稠合芳香环类衍生物、其制备方法及其在医药上的应用
WO2022100624A1 (zh) 氧取代氨基碳酸酯噻吩类化合物及其用途
WO2020035065A1 (zh) 作为ret抑制剂的吡唑衍生物
WO2023280280A1 (zh) 作为KRas G12D抑制剂的稠环化合物
WO2019062803A1 (zh) 作为a2a受体抑制剂的并环类衍生物
WO2019137201A1 (zh) 杂芳基并四氢吡啶类化合物、其制备方法、药物组合物及应用
CN111171049A (zh) 酪氨酸激酶抑制剂及其用途
WO2021104488A1 (zh) 作为jak抑制剂的三并杂环类化合物及其应用
CN114728975A (zh) 唑稠合的哒嗪-3(2h)-酮衍生物
WO2021129817A1 (zh) 具有果糖激酶(khk)抑制作用的嘧啶类化合物
WO2022063308A1 (zh) 一类1,7-萘啶类化合物及其应用
WO2018036470A1 (zh) 作为pde4抑制剂的并环类化合物
CN113874379B (zh) 作为Cdc7抑制剂的四并环类化合物
CN112574278A (zh) 作为蛋白降解剂杂环类化合物及其制备方法和医药应用
WO2023143352A1 (zh) 一种含氮杂环化合物、其制备方法及应用
WO2019134662A1 (zh) 作为csf-1r抑制剂的杂环化合物及其应用
WO2023011573A1 (zh) 芳香乙炔类衍生物及其制备方法和用途
CN111356692B (zh) Ido抑制剂
WO2022222911A1 (zh) 嘧啶酮化合物及其用途

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18891674

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18891674

Country of ref document: EP

Kind code of ref document: A1