WO2018133793A1 - 凝血因子XIa抑制剂及其用途 - Google Patents

凝血因子XIa抑制剂及其用途 Download PDF

Info

Publication number
WO2018133793A1
WO2018133793A1 PCT/CN2018/072988 CN2018072988W WO2018133793A1 WO 2018133793 A1 WO2018133793 A1 WO 2018133793A1 CN 2018072988 W CN2018072988 W CN 2018072988W WO 2018133793 A1 WO2018133793 A1 WO 2018133793A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
atoms
independently
mmol
Prior art date
Application number
PCT/CN2018/072988
Other languages
English (en)
French (fr)
Inventor
左应林
王建成
林继华
张英勋
曹生田
吴方园
杨雯
许娟
王晓军
张英俊
Original Assignee
广东东阳光药业有限公司
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 广东东阳光药业有限公司 filed Critical 广东东阳光药业有限公司
Priority to CN201880004854.3A priority Critical patent/CN110062757B/zh
Publication of WO2018133793A1 publication Critical patent/WO2018133793A1/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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/529Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/08Bridged systems
    • 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
    • C07D487/08Bridged systems

Definitions

  • the present invention belongs to the field of medicine, relates to a blood coagulation factor XIa inhibitor and uses thereof, and particularly to a macrocyclic compound having a blood coagulation factor XIa and/or a plasma kallikrein and a pharmaceutical composition thereof; wherein the macrocyclic compound And pharmaceutical compositions thereof for use in the treatment or prevention of thromboembolic disorders.
  • Procoagulant hemostasis
  • anticoagulation antithrombotic
  • Procoagulant and anticoagulation are two opposing mechanisms in the human blood system that contradict each other and maintain relative balance. This precise and harmonious process maintains the integrity of the circulatory system.
  • blood coagulation and anticoagulant function are out of balance, and coagulation occurs, which may cause thrombosis or embolism, which may lead to thromboembolism such as myocardial infarction, stroke, deep vein thrombosis, pulmonary embolism, etc. disease.
  • thromboembolic disease is the most serious disease in cardiovascular disease and the first killer of human health.
  • antithrombotic drugs have been researched and developed for the characteristics and causes of thrombosis, including inhibition of thrombosis (anticoagulation, for example, warfarin, heparin, low molecular weight heparin, etc.) And drugs that inhibit platelet aggregation (eg, aspirin, clopidogrel, etc.) and thrombolytic drugs.
  • the former mainly inhibits the formation and enlargement of thrombus, and the latter mainly dissolves the formed thrombus, thereby eliminating the harm caused by thrombotic diseases to humans.
  • the use of clinically applied anticoagulant drugs is still limited by various risks (eg, bleeding), and it is increasingly important to discover and develop safe and effective oral anticoagulant drugs for the prevention and treatment of extensive thromboembolic disorders. .
  • Factor XIa coagulation factor XIa
  • Factor XIa is a plasma serine protease involved in the regulation of blood coagulation, which is initiated by the in vivo binding of tissue factor (TF) to factor VII (FVII) to produce factor VIIa (FVIIa).
  • the TF:FVIIa complex produced therein activates Factor IX (FIX) and Factor X (FX), thereby causing the production of Factor Xa (FXa).
  • TFPI tissue factor pathway inhibitor
  • the resulting FXa catalyzes the conversion of prothrombin to a small amount of thrombin, and the catalytic amount of thrombin feedbacks the activation factors V, VIII and XI, thereby further expanding the coagulation process.
  • TFPI tissue factor pathway inhibitor
  • the thrombin burst caused by this process converts fibrinogen to fibrin, which polymerizes to form a structural framework for blood clots and activates platelets that are key cellular components of coagulation (Hoffman, M., Blood Reviews, 2003, 17: S1-S5). Therefore, Factor XIa plays a key role in expanding the amplification loop of the coagulation process, and thus it can serve as an attractive target for anti-thrombotic therapy.
  • Plasma prekallikrein is a chymase of trypsin-like serine protease which is present in plasma at a concentration of 35 ⁇ g/mL to 50 ⁇ g/mL.
  • the gene structure of plasma kallikrein is similar to that of factor XI; in general, the amino acid sequence of plasma kallikrein (PK) has 58% homology with factor XI.
  • Plasma kallikrein is thought to play a role in many inflammatory conditions.
  • the major inhibitor of plasma kallikrein is a serpin C1 esterase inhibitor.
  • HAE hereditary angioedema
  • DX-88 vascular permeability
  • a plasma kallikrein inhibitor For the treatment of hereditary angioedema and the prevention of blood loss in on-pumpcardiothoracic surgery, Expert Opin. Biol. Ther. 8, pp. 1187-99).
  • Kallikrein kinin system is a complex endogenous multi-enzyme system involved in the regulation of cardiovascular, renal, nervous system and other physiological functions, and heart disease, kidney disease, inflammatory response, cancer There is a close relationship between the occurrence of diseases. In recent years, research on the cardiovascular system has progressed rapidly. Many clinical studies and basic experiments have confirmed that the occurrence of diabetes, hypertension, heart failure, myocardial infarction and left ventricular hypertrophy is associated with decreased activity of KKS.
  • FXI factor XI
  • HK high molecular weight kininogen
  • PK prokallikrein
  • the present invention provides a macrocyclic compound suitable for a serine protease, particularly a selective inhibitor of factor XIa and/or plasma kallikrein, an analog thereof, and a pharmaceutical composition comprising the macrocyclic compound, the compound Or a thromboembolic disease in which the pharmaceutical composition is effective for the treatment and prevention.
  • the present invention provides a compound which is a stereoisomer, a geometric isomer, a tautomer, an oxynitride of a compound of formula (I) or a compound of formula (I), a hydrate, solvate, metabolite, pharmaceutically acceptable salt or prodrug,
  • Ring A is a C 7-12 carbocyclic group, a C 8-12 aryl group, a heterocyclic group consisting of 7 to 12 atoms or a heteroaryl group of 7 to 12 atoms;
  • Ring B is a C 5-12 carbocyclic group, a C 6-12 aryl group, a heterocyclic group consisting of 5 to 12 atoms or a heteroaryl group of 5 to 12 atoms;
  • Ring C is a C 3-12 carbocyclic group, a C 6-12 aryl group, a heterocyclic group consisting of 3 to 12 atoms or a heteroaryl group of 5 to 12 atoms;
  • Each R 3 is independently a C 1-6 alkyl group, a C 3-12 carbocyclic group, a C 6-12 aryl group, a heterocyclic group consisting of 3 to 12 atoms, or a heteroaryl group of 5 to 12 atoms; Wherein each R 3 is independently optionally substituted by 1, 2, 3, 4 or 5 R 3a ;
  • Each R 4 is independently H, hydrazine, halogen, hydroxy, amino, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, deuterated C 1-6 alkyl, halo C 1 -6 alkoxy, -(CR 7a R 7b ) r -OR 8 , -(CR 7a R 7b ) r -NR 5a R 5b , -(CR 7a R 7b ) r -CN, or halogenated C 1-6 Or an alkyl group; or any two of the R 4 and the atom to which they are attached form a C 3-8 cycloalkyl group, a C 6-10 aryl group, a heterocyclic group of 3 to 8 atoms, or 5 to 6 atoms.
  • any two R 9 together with the atoms to which they are attached form a C 3-6 cycloalkyl group or a heterocyclic group consisting of 3 to 6 atoms;
  • R 9 , R 1a , R 2a and R 3a are independently independently substituted by 1, 2, 3, 4 or 5 R 10 ;
  • R 5 , R 9 and the atoms attached thereto form a heterocyclic group consisting of 3-8 atoms;
  • Each of R 6 and R 8 is independently hydrogen, deuterium, C 1-6 alkyl, deuterated C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy substituted C 1-6 Alkyl, amino-substituted C 1-6 alkyl, cyano substituted C 1-6 alkyl, C 1-6 alkoxy C 1-6 alkyl, halo C 1-6 alkyl, C 6- a 10 aryl group, a C 3-10 cycloalkyl group, a heterocyclic group of 3 to 10 atoms or a heteroaryl group of 5 to 10 atoms;
  • R 7 , R 7a and R 7b is independently hydrogen, C 1-6 alkyl, deuterated C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl or halo C 1-6 alkyl;
  • Each m, n and t are independently 0, 1, 2, 3 or 4;
  • p 1, 2, 3 or 4;
  • Each r is independently 0, 1, 2, 3 or 4.
  • Ring C is a heterocyclic group consisting of 5-7 atoms or a heteroaryl group consisting of 5-6 atoms.
  • Ring C is of the following substructure:
  • each of Z, Z 1 and Z 2 is independently CH 2 or NH;
  • each of Z, Z 1 and Z 2 is independently CH or N;
  • Z 4 is CH 2 or NH
  • Each Z 3 and Z 5 are independently CH 2 , NH, S or O;
  • q 0, 1, or 2.
  • ring C is
  • the compound of formula (I) of the present invention is a stereoisomer, geometric isomer, tautomer, oxynitride of a compound of formula (II) or a compound of formula (II). a hydrate, solvate, metabolite, pharmaceutically acceptable salt or prodrug,
  • each Z and Z 1 are independently CH 2 or NH; or When it is a double bond, each Z and Z 1 are independently CH or N;
  • Ring A, Ring B, R 1 , R 2 , R 3 , R 4 , X, Y, n, m, p and t all have the meanings as described in the present invention.
  • each R 3 is independently C 1-4 alkyl, C 3-8 carbocyclyl, C 6-10 aryl, heterocyclyl consisting of 3-8 atoms, or 5-10 atoms. a heteroaryl group;
  • each R 3 is independently optionally substituted by 1, 2, 3, 4 or 5 R 3a .
  • each R 3 is independently methyl, ethyl, phenyl, pyrrolyl, pyrazolyl, imidazolyl, imidazolinyl, triazolyl, pyridyl, pyrimidinyl, fluorenyl, Carbazolyl, benzofuranyl, benzothienyl, benzothiazolyl, benzimidazolyl, benzisoxazolyl, benzisothiazolyl, quinolyl, isoquinolinyl or quinazolinyl ;
  • each R 3 is independently optionally substituted by 1, 2, 3, 4 or 5 R 3a .
  • the compound of formula (I) of the present invention is a stereoisomer, geometric isomer, tautomer, oxynitride of a compound of formula (III) or a compound of formula (III). a hydrate, solvate, metabolite, pharmaceutically acceptable salt or prodrug,
  • Z when or When it is a single bond, Z is CH 2 or NH; or When it is a double bond, Z is CH or N;
  • f 0, 1, 2, 3, 4 or 5;
  • g 0, 1, 2, 3, 4, 5, 6, 7, or 8;
  • Ring A, Ring B, R 1 , R 2 , R 3a , R 4 , R 5 , R 9 , n, m and t all have the meanings as described in the present invention.
  • Ring A is a C 7-12 bicyclic carbocyclyl, a C 8-12 bicyclic aryl, a bicyclic heterocyclyl consisting of 7-12 atoms, or a bicyclic heteroaryl consisting of 7-12 atoms.
  • Ring A is of the following substructure:
  • each of T 1 , T 2 , T 3 , T 4 , T 5 , T 6 , T 7 and T 8 are independently -CH- or -N-;
  • Each k and j are independently 0, 1, 2, 3 or 4.
  • Ring A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • Ring B is C 5-8 carbocyclyl, C 6-10 aryl, heterocyclyl consisting of 5-8 atoms, or heteroaryl consisting of 5-10 atoms;
  • Each R 2 is independently optionally substituted by 1, 2, 3 or 4 R 2a ;
  • R 2a , R 5a , R 5b , R 5c , R 5d , R 6 , R 7 , R 7a , R 7b , R 8 and r all have the meanings as described in the present invention.
  • Ring B is of the following substructure:
  • Each of Q 2 , Q 3 , Q 4 , Q 5 , Q 7 and Q 8 is independently CH or N;
  • s 0, 1, 2 or 3;
  • R 2a , R 5a , R 5b , R 5c , R 5d , R 6 , R 7 , R 7a , R 7b , R 8 and r all have the meanings as described in the present invention.
  • Ring B is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • each R 2 is independently optionally substituted by 1, 2, 3 or 4 R 2a ; R 2a has the meanings as described herein.
  • Each R 4 is independently H, hydrazine, F, Cl, Br, I, hydroxy, amino, methyl, ethyl, propyl, butyl, trifluoromethyl, difluoromethyl, 1,2-difluoro Ethyl, trifluoromethoxy, difluoromethoxy, -OMe, -OEt, -O(t-Bu), -CH 2 OH, -CH 2 CH 2 OH, -CN, -CH 2 CN, - CH 2 NH 2 , -CH 2 CH 2 NH 2 , -CH 2 OMe, -CH 2 CH 2 OMe, -NHMe or -N(Me) 2 ; alternatively, any two R 4 and the atoms to which they are attached form a C 3-6 cycloalkyl group or a heterocyclic group consisting of 3 to 6 atoms;
  • any two R 9 together with the atoms to which they are attached form a C 3-6 cycloalkyl group or a heterocyclic group consisting of 3 to 6 atoms;
  • R 9 , R 1a , R 2a and R 3a are independently independently substituted by 1, 2, 3, 4 or 5 R 10 ;
  • R 5a , R 5b , R 5c , R 6 , R 7a , R 7b , R 8 , R 10 and r all have the meanings as described in the present invention.
  • Each R 4 is independently H, hydrazine, F, Cl, Br, I, hydroxy, amino, methyl, ethyl, propyl, butyl, vinyl, propenyl, allyl, deuterated methyl, three Fluoromethyl, difluoromethyl, 1,2-difluoroethyl, -OMe, -OEt, -CH 2 OH, -CH 2 CH 2 OH, -CN, -CH 2 CN, -CH 2 NH 2 , -CH 2 CH 2 NH 2 , -CH 2 OMe, -CH 2 CH 2 OMe, -NHMe or -N(Me) 2 ; alternatively, any two R 4 and the atoms to which they are attached form a cyclopropyl group, a ring Butyl, epoxyalkyl or azetidinyl;
  • any two R 9 together with the atom to which they are attached form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, alkylene oxide, epoxybutyl, azetidinyl, tetrahydrofuranyl, pyrrole
  • R 9 , R 1a , R 2a and R 3a are independently independently substituted by 1, 2, 3, 4 or 5 R 10 ;
  • R 10 has the meanings as described in the present invention.
  • Each of R 6 and R 8 is independently hydrogen, deuterium, C 1-4 alkyl, deuterated C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, hydroxy substituted C 1-4 Alkyl, amino substituted C 1-4 alkyl, cyano substituted C 1-4 alkyl, C 1-4 alkoxy C 1-4 alkyl, halo C 1-4 alkyl, C 6- a 10 aryl group, a C 3-6 cycloalkyl group, a heterocyclic group of 3 to 6 atoms or a heteroaryl group of 5 to 6 atoms;
  • R 7 , R 7a and R 7b is independently hydrogen, C 1-4 alkyl, deuterated C 1-4 alkyl or halo C 1-4 alkyl;
  • R 6 and R 8 is independently hydrogen, deuterium, methyl, ethyl, propyl, butyl, deuterated methyl, vinyl, propynyl, hydroxymethyl, hydroxyethyl, methoxymethyl , methoxyethyl, ethoxymethyl, tert-butoxymethyl, tert-butoxyethyl, ethoxyethyl, isopropoxyethyl, trifluoromethyl, difluoromethyl, 1,2-difluoroethyl, 2,2-difluoroethyl, phenyl, cyclopropyl, cyclohexyl, azetidinyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, piperazinyl, pyridine , pyrrolyl, pyrimidinyl, pyrazolyl, pyrazinyl, furyl, thiazolyl, oxazolyl
  • R 7 , R 7a and R 7b is independently hydrogen, methyl, ethyl, propyl, butyl, deuterated methyl, trifluoromethyl, difluoromethyl, 1,2-difluoroethyl or 2,2-difluoroethyl;
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of any of the invention.
  • the pharmaceutical compositions of the present invention further comprise at least one of a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, and vehicle.
  • the invention relates to the use of a compound or pharmaceutical composition for the manufacture of a medicament for the prevention, treatment or alleviation of a thromboembolic disorder.
  • the use of the invention wherein the thromboembolic disease is an arterial cardiovascular thromboembolic disease, a venous cardiovascular thromboembolic disease, and a thromboembolic disease in the ventricular or peripheral circulation.
  • the use of the invention wherein the thromboembolic disease is angina pectoris, acute coronary syndrome, atrial fibrillation, myocardial infarction, transient ischemic attack, stroke, atherosclerosis, peripheral Occlusive arterial disease, venous thrombosis, deep vein thrombosis, thrombophlebitis, arterial embolism, coronary thrombosis, cerebral arterial thrombosis, cerebral embolism, renal embolism, pulmonary embolism, and due to medical implants, instruments, or The blood in the procedure is exposed to the artificial surface to promote thrombosis caused by thrombosis.
  • the thromboembolic disease is angina pectoris, acute coronary syndrome, atrial fibrillation, myocardial infarction, transient ischemic attack, stroke, atherosclerosis, peripheral Occlusive arterial disease, venous thrombosis, deep vein thrombosis, thrombophlebitis, arterial embolism, coronary
  • the invention relates to the use of a compound or pharmaceutical composition for the manufacture of a medicament for the treatment of disseminated intravascular coagulation (DIC) disease.
  • DIC disseminated intravascular coagulation
  • the invention relates to the use of a compound or pharmaceutical composition for the preparation of a medicament, wherein the medicament is for inhibiting the activity of Factor XIa and/or plasma kallikrein.
  • the compounds or pharmaceutical compositions of the invention are used to prevent, treat or ameliorate thromboembolic disorders.
  • the compound or pharmaceutical composition of the present invention is for use in preventing, treating or ameliorating a thromboembolic disease, wherein the thromboembolic disease is an arterial cardiovascular thromboembolic disease, venous cardiovascular thromboembolism Sexual disease, and thromboembolic disease in the ventricular or peripheral circulation.
  • a thromboembolic disease is an arterial cardiovascular thromboembolic disease, venous cardiovascular thromboembolism Sexual disease, and thromboembolic disease in the ventricular or peripheral circulation.
  • the compounds or pharmaceutical compositions of the invention are used to prevent, treat or ameliorate a thromboembolic disease, wherein the thromboembolic disease is angina pectoris, acute coronary syndrome, atrial fibrillation, myocardium Infarction, transient ischemic attack, stroke, atherosclerosis, peripheral occlusive arterial disease, venous thrombosis, deep vein thrombosis, thrombophlebitis, arterial embolism, coronary thrombosis, cerebral arterial thrombosis, cerebral embolism , renal embolism, pulmonary embolism, and thrombosis caused by exposure of blood in a medical implant, device, or procedure to an artificial surface to promote thrombosis.
  • the thromboembolic disease is angina pectoris, acute coronary syndrome, atrial fibrillation, myocardium Infarction, transient ischemic attack, stroke, atherosclerosis, peripheral occlusive arterial disease, venous thrombosis, deep vein thro
  • the compounds or pharmaceutical compositions of the invention are useful for inhibiting the activity of Factor XIa and/or plasma kallikrein.
  • the invention also relates to a method of preventing, treating or ameliorating a thromboembolic disease using a compound or pharmaceutical composition of the invention.
  • the method of the present invention wherein the thromboembolic disease is an arterial cardiovascular thromboembolic disease, a venous cardiovascular thromboembolic disease, and a thromboembolic disease in the ventricular or peripheral circulation.
  • the method of the present invention wherein the thromboembolic disease is angina pectoris, acute coronary syndrome, atrial fibrillation, myocardial infarction, transient ischemic attack, stroke, atherosclerosis, Peripheral occlusive arterial disease, venous thrombosis, deep vein thrombosis, thrombophlebitis, arterial embolism, coronary thrombosis, cerebral arterial thrombosis, cerebral embolism, renal embolism, pulmonary embolism, and medical implants, instruments Or blood in the procedure is exposed to an artificial surface to promote thrombosis caused by thrombosis.
  • the thromboembolic disease is angina pectoris, acute coronary syndrome, atrial fibrillation, myocardial infarction, transient ischemic attack, stroke, atherosclerosis, Peripheral occlusive arterial disease, venous thrombosis, deep vein thrombosis, thrombophlebitis, arterial embolism, coronar
  • the invention also relates to a method of inhibiting the activity of Factor XIa and/or plasma kallikrein using a compound or pharmaceutical composition of the invention.
  • &quot refers to one or more components, i.e., more than one component may be considered for use or use in embodiments of the embodiments.
  • subject refers to an animal. Typically the animal is a mammal. Subjects, for example, also refer to primates (eg, humans, males or females), cattle, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds, and the like. In certain embodiments, the subject is a primate. In other embodiments, the subject is a human.
  • primates eg, humans, males or females
  • the subject is a primate. In other embodiments, the subject is a human.
  • patient refers to a person (including adults and children) or other animal. In some embodiments, “patient” refers to a human.
  • Stereoisomer refers to a compound that has the same chemical structure but differs in the way the atoms or groups are spatially aligned. Stereoisomers include enantiomers, diastereomers, conformational isomers (rotomers), geometric isomers (cis/trans) isomers, atropisomers, etc. .
  • “Chirality” is a molecule that has properties that cannot overlap with its mirror image; “non-chiral” refers to a molecule that can overlap with its mirror image.
  • Enantiomer refers to two isomers of a compound that are not superimposable but are mirror images of each other.
  • Diastereomer refers to a stereoisomer that has two or more centers of chirality and whose molecules are not mirror images of each other. Diastereomers have different physical properties such as melting point, boiling point, spectral properties and reactivity. The mixture of diastereomers can be separated by high resolution analytical procedures such as electrophoresis and chromatography, such as HPLC.
  • optically active compounds Many organic compounds exist in optically active forms, i.e., they have the ability to rotate a plane of plane polarized light.
  • the prefixes D and L or R and S are used to indicate the absolute configuration of the molecule with respect to one or more of its chiral centers.
  • the prefixes d and l or (+) and (-) are symbols for specifying the rotation of plane polarized light caused by the compound, wherein (-) or l indicates that the compound is left-handed.
  • Compounds prefixed with (+) or d are dextrorotatory.
  • a particular stereoisomer is an enantiomer and a mixture of such isomers is referred to as a mixture of enantiomers.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which can occur when there is no stereoselectivity or stereospecificity in a chemical reaction or process.
  • any asymmetric atom (e.g., carbon, etc.) of the compounds disclosed herein may exist in racemic or enantiomerically enriched form, such as the (R)-, (S)- or (R, S)-configuration presence.
  • each asymmetric atom has at least 50% enantiomeric excess in the (R)- or (S)-configuration, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess.
  • the compounds of the invention may be one of the possible isomers or mixtures thereof, such as racemates and mixtures of diastereomers (depending on the number of asymmetric carbon atoms) The form exists.
  • Optically active (R)- or (S)-isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituent may be in the E or Z configuration; if the compound contains a disubstituted cycloalkyl group, the substituent of the cycloalkyl group may have a cis or trans configuration.
  • the resulting mixture of any stereoisomers can be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, for example, by chromatography, depending on the difference in physicochemical properties of the components. Method and / or step crystallization.
  • racemate of any of the resulting end products or intermediates can be resolved into the optical antipodes by methods known to those skilled in the art by known methods, for example, by obtaining the diastereomeric salts thereof. Separation. Racemic products can also be separated by chiral chromatography, such as high performance liquid chromatography (HPLC) using a chiral adsorbent.
  • HPLC high performance liquid chromatography
  • enantiomers can be prepared by asymmetric synthesis, for example, see Jacques, et al., Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Principles of Asymmetric Synthesis (2 nd Ed. Robert) E.
  • tautomer or "tautomeric form” refers to structural isomers having different energies that are interconvertible by a low energy barrier. If tautomerism is possible (as in solution), the chemical equilibrium of the tautomers can be achieved.
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by recombination of some bonding electrons.
  • keto-enol tautomerization is the interconversion of a pentane-2,4-dione and a 4-hydroxypent-3-en-2-one tautomer.
  • Another example of tautomerization is phenol-keto tautomerization.
  • a specific example of phenol-keto tautomerization is the interconversion of pyridin-4-ol and pyridine-4(1H)-one tautomers. All tautomeric forms of the compounds of the invention are within the scope of the invention unless otherwise indicated.
  • the compounds of the present invention may be optionally substituted with one or more substituents, such as the compounds of the above formula, or specific examples, subclasses, and inclusions of the present invention.
  • substituents such as the compounds of the above formula, or specific examples, subclasses, and inclusions of the present invention.
  • a class of compounds may be used interchangeably.
  • substituted means that one or more hydrogen atoms in a given structure are replaced by a particular substituent. Unless otherwise indicated, an optional substituent group can be substituted at each substitutable position of the group.
  • substituents When more than one position in the given formula can be substituted by one or more substituents selected from a particular group, the substituents may be substituted at the various positions, either identically or differently.
  • substituents mean one or more substituents, and the number of specific substituents is determined by the number of positions at which the substituent group can be substituted.
  • the present invention is described in the "C 4 alkylene group optionally substituted independently with one or more identical or different R 9 substituted" indicates that the group may be an alkylene or substituted with one or more R 9, particularly Said alkylene group may be independently and optionally substituted by 1, 2, 3, 4, 5, 6, 7 or 8 R 9 ; when said alkylene group is more than one R 9 When substituted, the R 9 may be the same or different.
  • each alkylamino group, arylamino group, alkoxy group, aryloxy group, hydroxyl group, mercapto group, alkyl group, haloalkyl group, carbocyclic group, heterocyclic group, aryl group, heteroaryl group, alkyl group in the substituent Sulfonyl, aminosulfonyl, hydroxyalkyl, aminoalkyl, alkylacyl, aminoacyl and alkylthio have the meanings as described herein and may be further monosubstituted or identical by the substituents described herein Or different multiple substitutions.
  • each R 9 , R 1a , R 2a and R 3a are independently hydrogen, deuterium, nitro or C 1-6 alkyl
  • R 9 may be hydrogen, deuterium, nitro or C 1-6 alkyl
  • R 1a may also be hydrogen, deuterium, nitro or C 1-6 alkyl
  • R 2a and R 3a may also be hydrogen, deuterium, nitro or C 1-6 alkane. base.
  • C 1 - 6-alkyl refers particularly disclosed independently methyl, ethyl, C 3 alkyl, C 4 alkyl, C 5 and C 6 alkyl group.
  • C 1-4 alkyl specifically refers to independently disclosed methyl, ethyl, C 3 alkyl (ie propyl, including n-propyl and isopropyl), C 4 alkyl (ie butyl, including n-butyl) Base, isobutyl, sec-butyl and tert-butyl).
  • linking substituents are described.
  • the Markush variable recited for that group is understood to be a linking group.
  • the definition of the Markush group for the variable is "alkyl” or "aryl”
  • the “alkyl” or “aryl” respectively represent the attached An alkylene group or an arylene group.
  • the ring A in the structure of the formula (I) of the present invention may be a heteroaryl group, and the heteroaryl group herein represents an arylene group having two linking sites attached to the rest of the molecule, more specifically, for example
  • ring A is a quinolyl group, and the quinolyl group here represents a quinolinyl group.
  • C 1-6 alkoxy C 1-6 alkyl C 1-6 alkyl represents a C 1-6 alkylene group, including but not limited to, methylene, ethylene and the like.
  • alkyl or "alkyl group” as used herein, denotes a saturated straight or branched chain monovalent hydrocarbon group containing from 1 to 20 carbon atoms, wherein the alkyl group may be any Optionally, it is substituted with one or more substituents described herein. Unless otherwise specified, an alkyl group contains from 1 to 20 carbon atoms. In some embodiments, the alkyl group contains from 1 to 12 carbon atoms; in other embodiments, the alkyl group contains from 1 to 6 carbon atoms; in still other embodiments, the alkyl group contains 1 - 4 carbon atoms; in other embodiments, the alkyl group contains 1-3 carbon atoms.
  • C 1 - 6 alkyl denotes an alkyl group containing 1 to 6 carbon atoms.
  • alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), n-propyl (n-Pr, -CH 2 CH 2 CH 3 ), isopropyl (i-Pr, -CH(CH 3 ) 2 ), n-butyl (n-Bu, -CH 2 CH 2 CH 2 CH 3 ), isobutyl (i-Bu, -CH 2 CH) (CH 3 ) 2 ), sec-butyl (s-Bu, -CH(CH 3 )CH 2 CH 3 ), tert-butyl (t-Bu, -C(CH 3 ) 3 ), n-pentyl (-CH) 2 CH 2 CH 2 CH 2 CH 3 ), 2-pentyl (-CH(CH 3 )CH 2 CH 2 CH 3 ), 3-pentyl (-CH(CH 2 CH 3 ) 2 ), 2-methyl -2-butyl (-C(CHCH
  • alkenyl denotes a straight or branched chain monovalent hydrocarbon radical containing from 2 to 12 carbon atoms, wherein there is at least one carbon-carbon sp 2 double bond, wherein the alkenyl group may be optionally one or Substituted by a plurality of substituents described herein, which include the positioning of "cis” and “tans", or the positioning of "E” and "Z".
  • the alkenyl group contains 2-8 carbon atoms; in other embodiments, the alkenyl group contains 2-6 carbon atoms; in still other embodiments, the alkenyl group comprises 2 - 4 carbon atoms.
  • alkynyl means a straight or branched chain monovalent hydrocarbon radical containing from 2 to 12 carbon atoms, wherein at least one carbon-carbon sp triple bond, wherein the alkynyl group may be optionally one or more Substituted by the substituents described in the present invention.
  • the alkynyl group contains 2-8 carbon atoms; in other embodiments, the alkynyl group contains 2-6 carbon atoms; in still other embodiments, the alkynyl group comprises 2 - 4 carbon atoms.
  • alkynyl groups include, but are not limited to, ethynyl, propynyl, and the like.
  • alkylene means a saturated divalent hydrocarbon group derived by removing two hydrogen atoms from a saturated linear or branched hydrocarbon group. Unless otherwise specified, an alkylene group contains from 1 to 12 carbon atoms. In some embodiments, the alkylene group contains 1-6 carbon atoms; in other embodiments, the alkylene group contains 1-4 carbon atoms; in still other embodiments, the alkylene group The group contains 1-3 carbon atoms; in other embodiments, the alkylene group contains 1-2 carbon atoms.
  • Such examples include methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), isopropylidene (-CH(CH 3 )CH 2 -), n-butylene (-CH 2 ) CH 2 CH 2 CH 2 -) and so on.
  • alkenylene means a divalent hydrocarbyl group derived by removing two hydrogen atoms from an alkenyl group. Unless otherwise specified, an alkenylene group contains from 1 to 12 carbon atoms. In some embodiments, the alkenylene group contains 1-6 carbon atoms; in other embodiments, the alkenylene group contains 1-4 carbon atoms; in still other embodiments, the alkenylene group The group contains 1-3 carbon atoms. Examples of such include propenylene, butenylene, and the like.
  • alkoxy denotes an alkyl group attached to the remainder of the molecule through an oxygen atom, wherein the alkyl group has the meaning as described herein.
  • the alkoxy group contains from 1 to 6 carbon atoms; in other embodiments, the alkoxy group contains from 1 to 4 carbon atoms; in still other embodiments, the alkoxy group The group contains 1-3 carbon atoms.
  • the alkoxy group can be optionally substituted with one or more substituents described herein.
  • alkoxy groups include, but are not limited to, methoxy (MeO, -OCH 3 ), ethoxy (EtO, -OCH 2 CH 3 ), 1-propoxy (n-PrO, n- Propyloxy, -OCH 2 CH 2 CH 3 ), and the like.
  • alkoxyalkyl denotes an alkyl group substituted by one or more alkoxy groups, wherein alkyl and alkoxy have the meanings as described herein.
  • the alkoxyalkyl group can be optionally substituted with one or more substituents described herein.
  • the alkoxyalkyl group is a C 1-4 alkoxy C 1-4 alkyl group; in other embodiments, the alkoxyalkyl group is a C 1-2 alkoxy group C 1-3 Alkyl; In still other embodiments, the alkoxyalkyl group is a C1-3 alkoxy C1-3 alkyl group.
  • Such examples include, but are not limited to, methoxymethyl, methoxyethyl, isopropoxymethyl, tert-butoxymethyl, tert-butoxyethyl, and the like.
  • the alkoxyacyl, alkanoyl, or aminoacyl group may be optionally substituted with one or more substituents described herein.
  • alkyl sulfonyl denotes an alkyl group, an alkoxy group, an alkylamino group or an amino group (-NH 2) group bonded through a sulfonyl group (-SO 2 -) is attached to the remainder of the molecule wherein the alkyl, alkoxy, alkylamino group has the meaning as described herein.
  • the alkylsulfonyl, alkoxysulfonyl, alkylaminosulfonyl, aminosulfonyl group may be optionally substituted with one or more substituents described herein. Examples of alkylsulfonyl groups include, but are not limited to, methylsulfonyl (-SO 2 CH 3 ), ethylsulfonyl (-SO 2 CH 2 CH 3 ), and the like.
  • haloalkyl denotes an alkyl, alkenyl or alkoxy group substituted by one or more halogen atoms, examples of which include, but are not limited to, Trifluoromethyl, difluoromethyl 2,2-difluoroethyl, trifluoromethoxy, and the like.
  • haloalkyl or “deuterated alkoxy” denotes an alkyl or alkoxy group substituted by one or more deuterium atoms, examples of which include, but are not limited to, deuterated methyl (- CD 3 ), deuterated methoxy (-OCD 3 ), and the like.
  • carbocyclic or “carbocyclyl” refers to a monovalent or polyvalent, non-aromatic, saturated or partially unsaturated monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 ring carbon atoms.
  • the carbocycle comprises from 3 to 10 ring carbon atoms; in other embodiments, the carbocycle comprises from 3 to 8 ring carbon atoms; in still other embodiments, the carbocycle comprises from 3 to 6 rings.
  • carbocyclic groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1-cyclopentyl-1-enyl, 1-cyclopentyl-2-alkenyl, 1-cyclopentyl-3-alkenyl, 1-cyclohexyl-1-alkenyl, 1-cyclohexyl-2-alkenyl, 1-cyclohexyl-3-alkenyl, cyclohexadienyl, cycloheptyl , cyclooctyl, cyclodecyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like. Wherein the carbocyclic group may be optionally substituted by one or more substituents described herein.
  • cycloalkyl refers to a monovalent or polyvalent, non-aromatic, saturated monocyclic, bicyclic or tricyclic system containing from 3 to 12 ring carbon atoms.
  • the cycloalkyl group contains 3-10 ring carbon atoms; in other embodiments, the cycloalkyl group contains 3-8 ring carbon atoms; in still other embodiments, the cycloalkyl group comprises 3- 6 ring carbon atoms.
  • Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • the cycloalkyl group can be optionally substituted with one or more substituents described herein.
  • heterocyclyl and “heterocycle” are used interchangeably herein to refer to a saturated or partially unsaturated monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 ring atoms, wherein at least one ring atom is selected. From nitrogen, sulfur and oxygen atoms, and any one of the heterocyclic ring systems is non-aromatic.
  • the sulfur atom of the ring can be optionally oxidized to an S-oxide.
  • the nitrogen atom of the ring can be optionally oxidized to an N-oxygen compound.
  • the heterocyclic group is a heterocyclic group consisting of 5 to 12 atoms; in other embodiments, the heterocyclic group is a heterocyclic group of 5 to 8 atoms; in still other embodiments, The heterocyclic group is a heterocyclic group consisting of 5 to 7 atoms; and in some embodiments, the heterocyclic group is a heterocyclic group of 5 to 6 atoms.
  • the heterocyclic group may also be a bicyclic heterocyclic group; in some embodiments, the heterocyclic group is a bicyclic heterocyclic group consisting of 7 to 12 atoms; in other embodiments, the heterocyclic group is composed of 7 to 10 atoms. Bicyclic heterocyclic groups; In still other embodiments, the heterocyclic group is a bicyclic heterocyclic group consisting of 8-10 atoms.
  • heterocyclic groups include, but are not limited to, oxiranyl, azetidinyl, oxetanyl, pyrrolidinyl, pyrazolidinyl, dihydrothienyl, 1,3-dioxo Pentyl, dithiocyclopentyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, 1,2-dihydropyridyl, morpholinyl, thiomorpholinyl, hexahydropyrimidinyl, 1 , 6-dihydropyrimidinyl, 1,2-dihydropyrimidinyl, 1,2-dihydropyrazinyl, 1,3-oxazinyl, piperazinyl, oxazolidinyl, dioxoalkyl, Dithiaalkyl, thiamethane, homopiperazinyl, homopiperidinyl, oxetanyl,
  • sulfur atom in the heterocyclic group being oxidized examples include, but are not limited to, a sulfolane group, a 1,1-dioxothiomorpholinyl group, and a 1,1-dioxo-1,2-thiomorpholinyl group.
  • a heterocyclic group is a heterocyclic group consisting of 5-6 atoms, and refers to a saturated or partially unsaturated monocyclic ring containing 5 or 6 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen and sulfur. And oxygen atoms.
  • the heterocyclic group of 5-6 atoms include, but are not limited to, pyrrolidinyl, pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuran.
  • Base tetrahydrothiophenyl, oxazolidinyl, piperidinyl, 1,2-dihydropyridyl, morpholinyl, thiomorpholinyl, hexahydropyrimidinyl, 1,6-dihydropyrimidinyl, 1 , 2-dihydropyrimidinyl, 1,2-dihydropyrazinyl, 1,3-oxazinyl, piperazinyl, 1,2,3,6-tetrahydropyridyl, 1,2,3, 4-tetrahydropyridyl, 1,2,3,4-tetrahydropyrimidinyl, 2,5-dihydro-1H-pyrrolyl and the like.
  • the heterocyclic group consisting of 5-6 atoms may be optionally substituted by one or more substituents described in the present invention.
  • unsaturated as used in the present invention means that the group contains one or more unsaturations.
  • heteroatom refers to O, S, N, P, and Si, including any form of oxidation states of N, S, and P; forms of primary, secondary, tertiary, and quaternary ammonium salts; or nitrogen atoms in heterocycles. a form in which hydrogen is substituted, for example, N (like N in 3,4-dihydro-2H-pyrrolyl), NH (like NH in pyrrolidinyl) or NR (like in N-substituted pyrrolidinyl) NR).
  • halogen means fluorine (F), chlorine (Cl), bromine (Br) or iodine (I).
  • aryl denotes monocyclic, bicyclic and tricyclic hydrocarbyl groups containing from 6 to 14 ring atoms, or from 6 to 12 ring atoms, or from 8 to 12 ring atoms, or from 6 to 10 ring atoms, Wherein at least one ring is aromatic and one or more attachment points in the aromatic system are attached to the remainder of the molecule.
  • aryl can be used interchangeably with the terms “aromatic ring” or "aromatic ring”.
  • the aryl group includes an aromatic ring and an aromatic ring, or a ring system in which an aromatic ring is fused with a non-aromatic carbocyclic ring.
  • aryl group may include phenyl, naphthyl, anthracenyl, 1,2,3,4-tetrahydronaphthyl, 2,3-dihydro-1H-indenyl, bicyclo[4,2,0 ] ⁇ -1(6), 2,4-trienyl.
  • heteroaryl denotes a monocyclic, bicyclic, and tricyclic ring system having 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 6 ring atoms, wherein at least one ring is aromatic, and At least one ring contains one or more heteroatoms wherein one or more attachment points in the heteroaryl system are attached to the remainder of the molecule.
  • heteroaryl can be used interchangeably with the terms “heteroaryl ring” or "heteroaromatic compound”.
  • the heteroaryl group includes a heteroaryl ring and an aromatic ring, a heteroaryl ring and a heteroaryl ring, or a ring system in which a heteroaryl ring is fused with a non-aromatic carbocyclic or heterocyclic ring.
  • a heteroaryl group of 5-10 atoms comprises 1, 2, 3 or 4 heteroatoms independently selected from O, S and N.
  • the heteroaryl is a heteroaryl group of 7 to 12 atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N;
  • the heteroaryl group may be a single ring system or a bicyclic system containing two rings.
  • the heteroaryl is a heteroaryl group of 7-10 atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N; 7-10 atoms
  • the heteroaryl group may be a single ring system or a bicyclic system containing two rings.
  • heteroaryl groups include, but are not limited to, furyl, imidazolyl (eg, 1H-imidazol-1-yl), isoxazolyl, oxazolyl, pyrrolyl, 1,3,4-oxo Azyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (eg 3-pyridazinyl), thiazolyl, 2- Thienyl, 3-thienyl, pyrazolyl (eg 2-pyrazolyl, 1H-pyrazol-1-yl), pyrazinyl, 1,3,5-triazinyl, triazolyl, tetrazolyl Etc.; also includes the following bicyclic rings, but is by no means limited to these bicyclic rings: benzimidazolyl, benzofuranyl, dihydrobenzofuranyl, benzothi
  • i atoms typically describes the number of ring atoms in the molecule in which the number of ring atoms is i.
  • a piperidinyl group is a heterocyclic alkyl group composed of 6 atoms
  • a quinolyl group or a 5,6,7,8-tetrahydroquinolyl group is a heteroaryl group composed of 10 atoms.
  • bicyclic carbocyclyl denote carbocyclyl, aryl, heteroaryl and heterocyclyl consisting of two rings
  • the carbocyclic, aryl, heteroaryl and heterocyclic groups described have the meanings as described herein.
  • the bicyclic carbocyclic group and the bicyclic heterocyclic group include a fused ring, a spiro ring and a bridged ring composed of two rings.
  • the bicyclic aryl, bicyclic heteroaryl is a ring system in which an aromatic ring (aryl or heteroaryl) and a non-aromatic ring (carbocyclic or heterocyclic) are fused, wherein
  • the non-aromatic ring may be saturated or unsaturated, for example, 1,2,3,4-tetrahydronaphthyl, 2,3-dihydro-1H-indenyl, 5,6,7,8 -tetrahydroquinolinyl, 3,4-dihydro-2H-pyrano[3,2-b]pyridinyl, 2,3-dihydro-[1,4]dioxin[2,3- b] pyridyl, 2,3-dihydrobenzo[b][1,4]dioxin, 6,7-dihydro-5H-cyclopenta[3,2-b]pyridinyl, 2, 3-dihydrofuran [3,2-b]pyridine, etc.; in
  • the system is, for example, a naphthyl group, a quinolyl group, an oxazolyl group or the like.
  • bicyclic aryl or bicyclic heteroaryl groups can be found in the foregoing examples of aryl and heteroaryl definitions.
  • the alkyl acyloxy group can be optionally substituted with one or more substituents described herein.
  • alkylamino includes “N-alkylamino” and “N,N-dialkylamino", wherein the amino groups are each independently substituted with one or two alkyl groups.
  • the alkyl group having a suitable meaning as described in the present invention may be a monoalkylamino group or a dialkylamino group, and examples thereof include, but are not limited to, N-methylamino group, N- Ethylamino, N,N-dimethylamino, N,N-diethylamino and the like.
  • amino-substituted alkyl includes C 1-10 straight or branched alkyl groups substituted with one or more amino groups.
  • the aminoalkyl group is a C1-6 "lower aminoalkyl group” substituted with one or more amino groups, examples of which include, but are not limited to, aminomethyl, ammonia Ethyl, aminopropyl, aminobutyl and aminohexyl groups.
  • hydroxy substituted alkyl includes C 1-10 straight or branched alkyl groups substituted with one or more hydroxy groups.
  • the hydroxyalkyl group is a C 1-6 "lower hydroxyalkyl group" substituted with one or more hydroxy groups, examples of which include, but are not limited to, hydroxymethyl, hydroxy Ethyl, hydroxypropyl, hydroxybutyl and hydroxyhexyl, and the like.
  • cyano-substituted alkyl includes C1-10 straight or branched alkyl groups substituted by one or more cyano groups.
  • the hydroxyalkyl group is a C 1-6 "lower cyanoalkyl group” substituted with one or more cyano groups, examples of which include, but are not limited to, cyano Base, cyanoethyl and the like.
  • the ring system formed by the substituent (R e ) n1 bonded to the central ring by a bond represents that n1 substituents R e may be substituted at any substitutable position on the ring, wherein n1 is at most The sum of the number of sites on the ring that can be replaced.
  • n1 may It is 1, 2, 3, 4, 5, 6, or 7; wherein, when n1 is greater than 1, each of the R e may be the same or different.
  • the system in which the substituent (R 9 ) g is bonded to the macrocycle by a bond represents that the substituents R 9 may be substituted at any substitutable position on the alkylene group.
  • R 9 in formula d can be a butylene group in the ring Substituting at any position that can be substituted, including but not limited to the case shown by formula d1; wherein g, R 9 , A, B, R 1 , R 2 , R 5 , m and n in formula d, d1 have The meaning of the invention.
  • the attachment point can be attached to the remainder of the molecule at any attachable position on the ring.
  • ring A a in formula b is a quinoline ring (as shown in formula b1)
  • any position on the A 2 ring or B 2 ring that may be attached may serve as a point of attachment.
  • the remainder of the molecule attached to the linkage is interchangeable.
  • the A a ring in formula c is a quinoline ring represented by formula c1
  • the quinoline ring may be attached to the remainder of the molecule through the E 1 and E 2 ends, and the E 1 and E 2 ends are linked.
  • protecting group refers to a class of substituents which, when reacted with other functional groups, are generally used to block or protect the particular functionality of the functional group.
  • protecting group of an amino group refers to a substituent attached to an amino group to block or protect the functionality of an amino group in a compound.
  • Suitable amino protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl. (BOC, Boc), benzyloxycarbonyl (CBZ, Cbz) and 9-fluorenylmethoxycarbonyl (Fmoc).
  • hydroxy protecting group refers to a substituent used to block or protect a hydroxyl group
  • suitable protecting groups include acetyl and silyl groups.
  • Carboxy protecting group means a substituent of a carboxy group used to block or protect the functionality of a carboxy group.
  • Typical carboxy protecting groups include -CH 2 CH 2 SO 2 Ph, cyanoethyl, 2-(trimethylsilane Ethyl, 2-(trimethylsilyl)ethoxymethyl, 2-(p-toluenesulfonyl)ethyl, 2-(p-nitrophenylsulfonyl)ethyl, 2-(diphenyl Phosphine) ethyl, nitroethyl, and the like.
  • a general description of protecting groups can be found in the literature: T W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991; and PJ Kocienski, Protecting Groups, Thieme, Stuttgart, 2005.
  • prodrug means that a compound can be converted to a compound of the invention in vivo. Such transformation is affected by the hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue.
  • the prodrug-like compound of the present invention may be an ester.
  • the ester may be used as a prodrug such as a phenyl ester, an aliphatic (C 1-24 ) ester, an acyloxymethyl ester, or a carbonate. , carbamates and amino acid esters.
  • a compound of the invention comprises a hydroxyl group, i.e., it can be acylated to give a compound in the form of a prodrug.
  • Other prodrug forms include phosphates, such as those obtained by phosphorylation of a hydroxy group on the parent.
  • Metal product refers to a product obtained by metabolism of a specific compound or a salt thereof in vivo. Metabolites of a compound can be identified by techniques well known in the art, and the activity can be characterized by experimental methods as described herein. Such a product may be obtained by administering a compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage and the like. Accordingly, the invention includes metabolites of a compound, including metabolites produced by intimate contact of a compound of the invention with a mammal for a period of time.
  • Solvent-forming solvents include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol.
  • hydrate means that the solvent molecule is an association formed by water.
  • Room temperature in the present invention means that the temperature is from 10 ° C to 40 ° C. In some embodiments, “room temperature” refers to a temperature of from 20 °C to 30 °C; in other embodiments, “room temperature” refers to 25 °C.
  • treating any disease or condition, in some embodiments thereof, refers to ameliorating a disease or condition (i.e., slowing or preventing or alleviating the progression of a disease or at least one of its clinical symptoms). In other embodiments, “treating” refers to alleviating or ameliorating at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, “treating” refers to modulating a disease or condition from the body (eg, stabilizing a detectable symptom) or physiologically (eg, stabilizing the body's parameters) or both. In other embodiments, “treating” refers to preventing or delaying the onset, onset, or exacerbation of a disease or condition.
  • thromboembolic disease refers to a disease caused by two pathological processes of thrombosis and thromboembolism, which is also called a thrombotic disease.
  • thrombosis refers to a pathological process in which blood forms a part of an embolus in the blood vessel or in the inner membrane of the heart under certain conditions, causing partial or complete occlusion of the blood vessel and a blood supply disorder at the corresponding site.
  • Thromboembolism is a pathological process in which blood clots fall off from the site of formation and partially or completely block blood vessels during blood flow, causing ischemia or ischemia, hypoxia, necrosis, congestion and edema.
  • thromboembolic diseases include, but are not limited to, arterial cardiovascular thromboembolic disease, venous cardiovascular thromboembolic disease, and thromboembolic disease in the chamber of the heart. More specific examples of such diseases include, but are not limited to, myocardial infarction, angina pectoris (including unstable colic), acute coronary syndrome, reocclusion, and angioplasty or restenosis after aortic coronary venous shunt , stroke, transient ischemic attack, peripheral arterial occlusive disease, arterial thrombosis, coronary thrombosis, cerebral arterial thrombosis, cerebral embolism, renal embolism, pulmonary embolism, thrombophlebitis, venous thrombosis or deep vein thrombosis Form, and so on.
  • DIC diffuse intravascular coagulation
  • the "pharmaceutically acceptable salts" of the present invention can be synthesized from the parent compound, basic or acidic moiety by conventional chemical methods.
  • such salts can be obtained by reacting the free acid form of these compounds with a stoichiometric amount of a suitable base such as a hydroxide, carbonate, bicarbonate, or the like of Na, Ca, Mg or K.
  • a suitable base such as a hydroxide, carbonate, bicarbonate, or the like of Na, Ca, Mg or K.
  • the free base form of these compounds is prepared by reaction with a stoichiometric amount of a suitable acid. This type of reaction is usually carried out in water or an organic solvent or a mixture of the two.
  • a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile.
  • a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile.
  • the pharmaceutically acceptable salt may be a pharmaceutically acceptable acid addition salt which may be formed by the action of a compound of the invention with an inorganic acid and/or an organic acid, for example, with a mineral acid such as hydrochloric acid, hydrobromic acid, nitric acid, phosphoric acid or sulfuric acid.
  • a mineral acid such as hydrochloric acid, hydrobromic acid, nitric acid, phosphoric acid or sulfuric acid.
  • Salt formed formed; and organic acids such as acetic acid, trifluoroacetic acid, propionic acid, malonic acid, oxalic acid, maleic acid, fumaric acid, malic acid, citric acid, gluconic acid, mandelic acid, tartaric acid, stearic acid a salt formed of acid, succinic acid, sulfosalicylic acid, lactic acid, benzoic acid, benzenesulfonic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid or naphthalene disulfonic acid.
  • organic acids such as acetic acid, trifluoroacetic acid, propionic acid, malonic acid, oxalic acid, maleic acid, fumaric acid, malic acid, citric acid, gluconic acid, mandelic acid, tartaric acid, stearic acid a salt formed of acid, succinic acid, sulfosalicylic
  • the pharmaceutically acceptable salt can be a pharmaceutically acceptable base addition salt formed by the action of a compound of the invention with an inorganic base and/or an organic base.
  • Inorganic bases from which salts can be derived include, for example, ammonium salts and metals of Groups I to XII of the Periodic Table.
  • the salt is derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include the ammonium, potassium, sodium, calcium, and magnesium salts.
  • Organic bases from which salts can be derived include primary, secondary and tertiary amines, and substituted amines include naturally occurring substituted amines, cyclic amines, basic ion exchange resins and the like.
  • Certain organic amines include, for example, isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine, and tromethamine. .
  • the compounds disclosed in the present invention may also exist in the form of their hydrates or in the form of their solvents (e.g., ethanol, DMSO, etc.), and may be used for crystallization.
  • the compounds disclosed herein may form solvates either intrinsically or by design with pharmaceutically acceptable solvents, including water; thus, the compounds of the invention include both solvated and unsolvated forms.
  • any structural formula given by the present invention is also intended to indicate that these compounds are not isotopically enriched and isotopically enriched.
  • Isotopically enriched compounds have the structure depicted by the general formula given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • Exemplary isotopes that may be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O , 18 O, 18 F, 31 P, 32 P, 35 S, 36 Cl and 125 I.
  • the compounds of the invention include isotopically enriched compounds of the invention, for example, those in which a radioisotope such as 3 H, 14 C and 18 F is present, or in which a non-radioactive isotope is present, such as 2 H and 13 C.
  • a radioisotope such as 3 H, 14 C and 18 F
  • a non-radioactive isotope such as 2 H and 13 C.
  • isotopically enriched compounds can be used for metabolic studies (using 14 C), reaction kinetic studies (using, for example, 2 H or 3 H), detection or imaging techniques such as positron emission tomography (PET) or including drugs or Single photon emission computed tomography (SPECT) of substrate tissue distribution assays, or may be used in patient radiation therapy.
  • 18 F enriched compounds are particularly desirable for PET or SPECT studies.
  • Isotopically enriched compounds of the invention can be prepared by conventional techniques familiar to those skilled in the art or by the use of suitable isotopically labeled reagents in place of the previously used unlabeled reagents as described in the Examples and Preparations of the present invention.
  • substitution of heavier isotopes may provide certain therapeutic advantages resulting from higher metabolic stability. For example, increased in vivo half-life or reduced dose requirements or improved therapeutic index.
  • the indole in the present invention is considered to be a substituent of the compound of the present invention.
  • Isotopic enrichment factors can be used to define the concentration of such heavier isotopes, particularly ruthenium.
  • isotopic enrichment factor refers to the ratio between the isotope abundance and the natural abundance of a given isotope.
  • a substituent of a compound of the invention is designated as hydrazine
  • the compound has at least 3500 for each of the specified hydrazine atoms (52.5% of ruthenium incorporation at each of the specified ruthenium atoms), at least 4,000 (60% of ruthenium incorporation), At least 4,500 (67.5% of cerium incorporation), at least 5,000 (75% of cerium incorporation), at least 5,500 (82.5% of cerium incorporation), at least 6,000 (90% of cerium incorporation), at least 6333.3 (95%) Iridium enrichment factor with at least 6466.7 (97% cerium incorporation), at least 6600 (99% cerium incorporation) or at least 6633.3 (99.5% cerium incorporation).
  • the present invention can include pharmaceutically acceptable solvates wherein the solvent of crystallization may be isotopically substituted, for example D 2 O, acetone -d 6, DMSO-d 6 solvate of those.
  • the inventors of the present application have extensively studied and synthesized a series of macrocyclic compounds, and first discovered the following through FXIa enzyme inhibition activity screening, plasma kallikrein inhibition activity screening, metabolic screening, anticoagulant activity experiments, and other experiments.
  • the compound represented by the formula (I) has strong anti-FXIa activity and/or plasma kallikrein inhibitory activity, excellent drug metabolism properties and physicochemical properties, and is particularly suitable as an anticoagulant drug for treating thromboembolic diseases.
  • the present invention provides a macrocyclic compound represented by the formula (I), wherein the ring A is a bicyclic system, preferably, the ring A is a heterocyclic ring-containing fused bicyclic ring (including a fused bicyclic hetero ring and a fused bicyclic heteroaryl) Ring);
  • the ring A is a bicyclic system, preferably, the ring A is a heterocyclic ring-containing fused bicyclic ring (including a fused bicyclic hetero ring and a fused bicyclic heteroaryl) Ring);
  • the compounds involved in the ring A ring have good pharmacological activity data, and can effectively treat thromboembolic diseases.
  • the present invention relates to a compound which is a stereoisomer, a geometric isomer, a tautomer, an oxynitride, a hydrate, or a compound of the formula (I). a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug,
  • ring A, ring B, ring C, X, Y, R 1 , R 2 , R 3 , R 4 , n, m, p and t all have the meanings as described in the present invention.
  • the -8 alkylene group and the C 4-8 alkenylene group are each independently optionally substituted by one or more of the same or different R 9 ; wherein R 5 and R 9 have the meanings as described in the present invention.
  • the compound of formula (I) of the present invention is a compound of formula (Ia) or a stereoisomer, geometric isomer, tautomer, oxynitride of a compound of formula (Ia). a hydrate, solvate, metabolite, pharmaceutically acceptable salt or prodrug,
  • ring A, ring B, ring C, R 1 , R 2 , R 3 , R 4 , R 5 , R 9 , g, n, m, p and t all have the meanings as described in the present invention.
  • Ring C is carbocyclyl, aryl, heterocyclyl or heteroaryl.
  • Ring C is a C 3-12 carbocyclyl, a C 6-12 aryl, a heterocyclic group consisting of 3-12 atoms, or a heteroaryl group of 5-12 atoms.
  • Ring C is a C 5-6 carbocyclic group, a C 6 aryl group, a heterocyclic group consisting of 5-7 atoms, or a heteroaryl group consisting of 5-6 atoms.
  • Ring C is of the following substructure:
  • each of Z, Z 1 and Z 2 is independently CH 2 or NH;
  • each of Z, Z 1 and Z 2 is independently CH or N;
  • Z 4 is CH 2 or NH
  • Each Z 3 and Z 5 are independently CH 2 , NH, S or O;
  • q 0, 1, or 2.
  • the substructure for or More specifically, the formula (C1) is
  • each of the above substructures C1, C1-a, C1-b, C1-c, C1-d, C1-e, C1-f or C1-g passes through an N atom and structure Connected with Independently a single bond or a double bond; Z has the meanings described herein.
  • ring C is Where ring C passes through the N atom and structure Connected.
  • the compound of formula (I) according to the invention may be a stereoisomer, a geometric isomer, a tautomer of a compound of formula (II) or a compound of formula (II).
  • Body nitrogen oxides, hydrates, solvates, metabolites, pharmaceutically acceptable salts or prodrugs,
  • each Z and Z 1 are independently CH 2 or NH; or When it is a double bond, each Z and Z 1 are independently CH or N;
  • Ring A, Ring B, X, Y, R 1 , R 2 , R 3 , R 4 , n, m, p and t all have the meanings as described herein.
  • the compound of formula (I) according to the invention may be a compound of formula (IIa), or a stereoisomer, geometric isomer, tautomer of a compound of formula (IIa) a construct, an oxynitride, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug,
  • each Z and Z 1 are independently CH 2 or NH; or When it is a double bond, each Z and Z 1 are independently CH or N; ring A, ring B, R 1 , R 2 , R 3 , R 4 , R 5 , R 9 , g, n, m, p and t All have the meanings described in the present invention.
  • each R 3 is independently halo, C 1-6 alkyl, C 3-12 carbocyclyl, C 6-12 aryl, heterocyclyl consisting of 3-12 atoms, or 5-12 A heteroaryl group consisting of atoms; wherein each R 3 is independently optionally substituted by 1, 2, 3, 4 or 5 identical or different R 3a ; wherein R 3a has the meanings as described herein.
  • each R 3 is independently C 1-4 alkyl, C 3-8 carbocyclyl, C 6-10 aryl, heterocyclyl consisting of 3-8 atoms, or 5-10 atoms. a heteroaryl group; wherein each R 3 is independently optionally substituted by 1, 2, 3, 4 or 5 R 3a ; R 3a has the meanings as described herein.
  • each R 3 is independently
  • each of E 1 , E 2 and E 3 is independently CH or N;
  • Each of E 4 and E 5 is independently CH 2 , O, S or NH;
  • Each e is independently 0, 1, 2 or 3;
  • each R 3 is independently optionally substituted by 1, 2, 3, 4 or 5 R 3a ; R 3a has the meanings as described herein.
  • each R 3 is independently methyl, ethyl, F, Cl, Br, phenyl, pyrrolyl, pyrazolyl, imidazolyl, imidazolinyl, triazolyl, pyridyl, pyrimidinyl , mercapto, carbazolyl, benzofuranyl, benzothienyl, benzothiazolyl, benzimidazolyl, benzisoxazolyl, benzisothiazolyl, quinolyl, isoquinolyl Or quinazolinyl;
  • each R 3 is independently optionally substituted by 1, 2, 3, 4 or 5 R 3a ; R 3a has the meanings as described herein.
  • the compound of formula (I) according to the invention may be a stereoisomer, a geometric isomer, a tautomer of a compound of formula (IIIa) or a compound of formula (IIIa).
  • Body nitrogen oxides, hydrates, solvates, metabolites, pharmaceutically acceptable salts or prodrugs,
  • each Z and Z 1 when or When it is a single bond, each Z and Z 1 are independently CH 2 or NH; or When it is a double bond, each Z and Z 1 are independently CH or N; ring A, ring B, R 1 , R 2 , R 4 , R 5 , R 9 , R 3a , E 1 , E 2 , E 3 , n, m, f, g, and t all have the meanings described herein.
  • the compound of formula (I) according to the invention may be a stereoisomer, a geometric isomer, a tautomer of a compound of formula (IIIb) or a compound of formula (IIIb).
  • Body nitrogen oxides, hydrates, solvates, metabolites, pharmaceutically acceptable salts or prodrugs,
  • each Z and Z 1 when or When it is a single bond, each Z and Z 1 are independently CH 2 or NH; or When it is a double bond, each Z and Z 1 are independently CH or N; ring A, ring B, R 1 , R 2 , R 4 , R 5 , R 9 , R 3a , E 1 , E 2 , E 3 , n, m, f, g, and t all have the meanings described herein.
  • the compound of formula (I) according to the invention may be a stereoisomer, a geometric isomer, a tautomer of a compound of formula (III) or a compound of formula (III).
  • Body nitrogen oxides, hydrates, solvates, metabolites, pharmaceutically acceptable salts or prodrugs,
  • each Z and Z 2 are independently CH 2 or NH; or When it is a double bond, each Z and Z 2 is independently CH or N; ring A, ring B, R 1 , R 2 , R 4 , R 5 , R 9 , R 3a , n, m, f, g and t All have the meanings described in the present invention.
  • the compound of formula (I) according to the invention may be a compound of formula (IIIc), or a stereoisomer, geometric isomer, tautomer of a compound of formula (IIIc) a construct, an oxynitride, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug,
  • each Z and Z 2 are independently CH 2 or NH; or When it is a double bond, each Z and Z 2 is independently CH or N; ring A, ring B, R 1 , R 2 , R 4 , R 5 , R 9 , R 3a , n, m, f, g and t All have the meanings described in the present invention.
  • the substructure for among them, Through N atoms and structures Connected.
  • the substructure for among them, Through N atoms and structures Connected.
  • Ring A is a C 7-12 carbocyclyl, a C 8-12 aryl, a heterocyclic group consisting of 7-12 atoms, or a heteroaryl group of 7-12 atoms.
  • Ring A is a C 7-12 bicyclic carbocyclyl, a C 8-12 bicyclic aryl, a bicyclic heterocyclyl consisting of 7-12 atoms, or a bicyclic heteroaryl consisting of 7-12 atoms.
  • Ring A is of the following substructure:
  • each of T 1 , T 2 , T 3 , T 4 , T 5 , T 6 , T 7 and T 8 are independently -CH- or -N-;
  • Each k and j are independently 0, 1, 2, 3 or 4.
  • Ring A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • Ring B is carbocyclyl, aryl, heterocyclyl or heteroaryl.
  • Ring B is a C 5-8 carbocyclic group, a C 6-10 aryl group, a heterocyclic group consisting of 5-8 atoms, or a heteroaryl group consisting of 5-10 atoms.
  • Ring B is of the following substructure:
  • Each of Q 2 , Q 3 , Q 4 , Q 5 , Q 7 and Q 8 is independently CH or N;
  • s 0, 1, 2 or 3.
  • Ring B is
  • each R 1 is independently optionally substituted by 1, 2, 3 or 4 identical or different R 1a ;
  • R 1a , R 5a , R 5b , R 6 , R 7a , R 7b , R 8 and r all have the meanings as described in the present invention.
  • each R 1 is independently optionally substituted by 1, 2, 3 or 4 identical or different R 1a ;
  • R 1a , R 5a , R 5b , R 6 , R 7a , R 7b , R 8 and r all have the meanings as described in the present invention.
  • Each R 2 is independently optionally substituted by 1, 2, 3 or 4 identical or different R 2a ;
  • R 2a , R 5a , R 5b , R 5c , R 5d , R 6 , R 7 , R 7a , R 7b , R 8 and r all have the meanings as described in the present invention.
  • Each R 2 is independently optionally substituted by 1, 2, 3 or 4 identical or different R 2a ;
  • R 2a , R 5a , R 5b , R 5c , R 5d , R 6 , R 7 , R 7a , R 7b , R 8 and r all have the meanings as described in the present invention.
  • each R 2 is independently optionally substituted by 1, 2, 3 or 4 identical or different R 2a ; R 2a has the meanings as described herein.
  • each R 4 is independently H, deuterium, halogen, hydroxy, amino, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, deuterated C 1-6 alkane Base, deuterated C 1-6 alkoxy, halo C 1-6 alkoxy, -(CR 7a R 7b ) r -OR 8 , -(CR 7a R 7b ) r -NR 5a R 5b , -( CR 7a R 7b ) r CN, or halogenated C 1-6 alkyl;
  • R 5a , R 5b , R 7a , R 7b , R 8 and r all have the meanings as described in the present invention.
  • any two R 4 and the atoms to which they are attached form a C 3-8 cycloalkyl group, a C 6-10 aryl group, a heterocyclic group consisting of 3-8 atoms, or 5-6 atoms.
  • each R 4 is independently H, hydrazine, F, Cl, Br, I, hydroxy, amino, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, hydrazine C 1-4 alkyl, deuterated C 1-4 alkoxy, halo C 1-4 alkoxy, -(CR 7a R 7b ) r -OR 8 , -(CR 7a R 7b ) r -NR 5a R 5b , -(CR 7a R 7b ) r CN, or halogenated C 1-4 alkyl;
  • R 5a , R 5b , R 7a , R 7b , R 8 and r all have the meanings as described in the present invention.
  • any two R 4 and the atoms to which they are attached form a C 3-6 cycloalkyl group, a C 6 aryl group, a heterocyclic group consisting of 3-6 atoms, or 5-6 atoms. a heteroaryl group; the C 3-6 cycloalkyl group, a C 6 aryl group, a heterocyclic group composed of 3 to 6 atoms, and a heteroaryl group composed of 5 to 6 atoms, optionally independently one or more Substituted by R 10 ; said R 10 has the meanings as described in the present invention.
  • each R 4 is independently H, hydrazine, F, Cl, Br, I, hydroxy, amino, methyl, ethyl, propyl, butyl, trifluoromethyl, difluoromethyl, 1,2-difluoroethyl, trifluoromethoxy, difluoromethoxy, -OMe, -OEt, -O(t-Bu), -CH 2 OH, -CH 2 CH 2 OH, -CN, -CH 2 CN, -CH 2 NH 2 , -CH 2 CH 2 NH 2 , -CH 2 OMe, -CH 2 CH 2 OMe, -NHMe or -N(Me) 2 .
  • any two R 4 and the atoms to which they are attached form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, alkylene oxide, oxetanyl, tetrahydrofuranyl, pyrrolidine , piperidinyl, piperazinyl, morpholinyl or azetidinyl; said cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, alkylene oxide, oxetanyl, tetrahydrofuran
  • the base, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl and azetidinyl are optionally independently substituted by one or more R 10 ; said R 10 has the meanings as described herein.
  • any two R 9 together with the atoms to which they are attached form a C 3-6 cycloalkyl group or a heterocyclic group consisting of 3 to 6 atoms;
  • each R 9 , R 1a , R 2a and R 3a are independently independently substituted by 1, 2, 3, 4 or 5 identical or different R 10 ;
  • R 5a , R 5b , R 5c , R 6 , R 7a , R 7b , R 8 , R 10 and r all have the meanings as described in the present invention.
  • any two R 9 together with the atoms to which they are attached form a C 3-6 cycloalkyl group or a heterocyclic group consisting of 3 to 6 atoms;
  • each R 9 , R 1a , R 2a and R 3a are independently independently substituted by 1, 2, 3, 4 or 5 identical or different R 10 ;
  • R 5a , R 5b , R 5c , R 6 , R 7a , R 7b , R 8 , R 10 and r all have the meanings as described in the present invention.
  • any two R 9 together with the atom to which they are attached form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, alkylene oxide, epoxybutyl, azetidinyl, tetrahydrofuranyl, pyrrole
  • each R 9 , R 1a , R 2a and R 3a is independently optionally substituted by 1, 2, 3, 4 or 5 identical or different R 10 ; said R 10 has the meanings as described herein.
  • R 5 , R 9 and the atoms attached thereto form a heterocyclic group consisting of 3-8 atoms.
  • R 5 , R 9 and the atoms attached thereto form a heterocyclic group of 3 to 6 atoms.
  • each R 6 and R 8 are independently hydrogen, deuterium, C 1-6 alkyl, deuterated C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy Substituted C 1-6 alkyl, amino substituted C 1-6 alkyl, cyano substituted C 1-6 alkyl, C 1-6 alkoxy C 1-6 alkyl, halo C 1-6 An alkyl group, a C 6-10 aryl group, a C 3-10 cycloalkyl group, a heterocyclic group of 3 to 10 atoms or a heteroaryl group of 5 to 10 atoms.
  • each R 6 and R 8 are independently hydrogen, deuterium, C 1-4 alkyl, deuterated C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, hydroxy Substituted C 1-4 alkyl, amino substituted C 1-4 alkyl, cyano substituted C 1-4 alkyl, C 1-4 alkoxy C 1-4 alkyl, halo C 1-4 An alkyl group, a C 6-10 aryl group, a C 3-6 cycloalkyl group, a heterocyclic group composed of 3 to 6 atoms or a heteroaryl group of 5 to 6 atoms.
  • each R 6 and R 8 are independently hydrogen, deuterium, methyl, ethyl, propyl, butyl, deuterated methyl, vinyl, propynyl, hydroxymethyl, hydroxyethyl , methoxymethyl, methoxyethyl, ethoxymethyl, tert-butoxymethyl, tert-butoxyethyl, ethoxyethyl, isopropoxyethyl, trifluoromethyl , difluoromethyl, 1,2-difluoroethyl, 2,2-difluoroethyl, phenyl, cyclopropyl, cyclohexyl, azetidinyl, tetrahydrofuranyl, pyrrolidinyl, piperidine Base, piperazinyl, pyridyl, pyrrolyl, pyrimidinyl, pyrazolyl, pyrazinyl, furyl, thiazolyl,
  • each R 7 , R 7a , and R 7b are, independently, hydrogen, C 1-6 alkyl, deuterated C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, or Halogenated C 1-6 alkyl.
  • each R 7 , R 7a , and R 7b is independently hydrogen, C 1-4 alkyl, deuterated C 1-4 alkyl, or halo C 1-4 alkyl.
  • each R 7 , R 7a and R 7b are independently hydrogen, methyl, ethyl, propyl, butyl, deuterated methyl, trifluoromethyl, difluoromethyl, 1, 2-difluoroethyl or 2,2-difluoroethyl.
  • each m, n, and t are independently 0, 1, 2, 3, or 4.
  • p is 1, 2, 3 or 4.
  • each r is independently 0, 1, 2, 3 or 4.
  • f is 0, 1, 2, 3, 4 or 5;
  • g is 0, 1, 2, 3, 4, 5, 6, 7, or 8.
  • the present invention relates to a compound which is one of the following structures:
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of any of the invention.
  • the pharmaceutical compositions of the present invention further comprise at least one of a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, and vehicle.
  • the invention relates to the use of a compound or pharmaceutical composition for the manufacture of a medicament for the prevention, treatment or alleviation of a thromboembolic disorder.
  • the compounds or pharmaceutical compositions of the invention are used to prevent, treat or ameliorate thromboembolic disorders.
  • the invention also relates to a method of using a compound or pharmaceutical composition of the invention to prevent, treat or ameliorate a thromboembolic disorder.
  • the thromboembolic disease of the invention is an arterial cardiovascular thromboembolic disease, a venous cardiovascular thromboembolic disease, and a thromboembolic disease in the ventricular or peripheral circulation.
  • the thromboembolic disease is angina pectoris, acute coronary syndrome, atrial fibrillation, myocardial infarction, transient ischemic attack, stroke, atherosclerosis, peripheral occlusive arterial disease, venous thrombosis , deep vein thrombosis, thrombophlebitis, arterial embolism, coronary thrombosis, cerebral arterial thrombosis, cerebral embolism, renal embolism, pulmonary embolism, and exposure to blood from artificial surfaces in medical implants, instruments, or procedures Thereby promoting thrombosis caused by thrombosis.
  • the invention relates to the use of a compound or pharmaceutical composition for the manufacture of a medicament for the treatment of disseminated intravascular coagulation (DIC) disease.
  • DIC disseminated intravascular coagulation
  • the invention relates to the use of a compound or pharmaceutical composition for the preparation of a medicament for inhibiting the activity of factor XIa and/or plasma kallikrein.
  • the compounds or pharmaceutical compositions of the invention are used to inhibit the activity of Factor XIa and/or plasma kallikrein.
  • the invention also relates to a method of inhibiting the activity of Factor XIa and/or plasma kallikrein using a compound or pharmaceutical composition of the invention.
  • the present invention encompasses the use of the compounds and their pharmaceutically acceptable salts for the manufacture of pharmaceutical products for the treatment of thromboembolic disorders in patients, including those described herein.
  • the invention comprises a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention in combination with at least one pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle.
  • the invention also encompasses a method of treating or ameliorating, or susceptibility to, a thromboembolic disease in a patient comprising treating a patient with a therapeutically effective amount of a compound of the invention.
  • the thromboembolic diseases of the present invention include myocardial infarction, angina pectoris, re-occlusion and angioplasty or restenosis after aortic coronary venous shunt, stroke, transient ischemic attack, peripheral arterial occlusive disease, pulmonary embolism Or deep vein thrombosis.
  • the salt is a pharmaceutically acceptable salt.
  • pharmaceutically acceptable includes that the substance or composition must be chemically or toxicologically relevant to the other components of the formulation and to the mammal being treated.
  • Salts of the compounds of the invention also include the intermediates used in the preparation or purification of the compounds of the invention or the isolated enantiomers of the compounds of the invention, but are not necessarily pharmaceutically acceptable salts.
  • the desired salt can be prepared by any suitable method provided in the literature, for example, using a mineral acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid or the like.
  • a mineral acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid or the like.
  • organic acids such as acetic acid, trifluoroacetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid and salicylic acid; pyranoic acid such as glucitol Acids and galacturonic acids; alpha-hydroxy acids such as citric acid and tartaric acid; amino acids such as aspartic acid and glutamic acid; aromatic acids such as benzoic acid and cinnamic acid; sulfonic acids such as p-toluenesulfonic acid, Ethane sulfonic acid, and so on.
  • organic acids such as acetic acid, trifluoroacetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid and salicylic acid
  • pyranoic acid such as glucitol Acids and galacturonic acids
  • the desired salt can be prepared by a suitable method, for example, using an inorganic base or an organic base such as ammonia (primary ammonia, secondary ammonia, tertiary ammonia), alkali metal hydroxide or alkaline earth. Metal hydroxide, and so on.
  • Suitable salts include, but are not limited to, organic salts derived from amino acids such as glycine and arginine, ammonia such as primary, secondary and tertiary ammonia, and cyclic ammonia such as piperidine, morpholine and piperazine Etc., and inorganic salts are obtained from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • the pharmaceutical composition of the present invention comprises the formula (I) or formula (Ia) or formula (II) or formula (IIa) or formula (III) or formula (IIIa) or formula (IIIb) or formula A compound represented by (IIIc), a compound listed in the present invention, or a compound of Examples 1-11, and a pharmaceutically acceptable carrier, adjuvant, or excipient.
  • the amount of the compound in the composition of the present invention is effective for treating or alleviating a thromboembolic disease in a patient, or effectively inhibiting the activity of factor XIa and/or plasma kallikrein.
  • the pharmaceutical composition of the present invention comprises any one of the compounds of the present invention, or a stereoisomer, tautomer, pharmaceutically acceptable salt or solvate thereof; In some embodiments, the pharmaceutical compositions of the present invention comprise any combination of any of the compounds described herein.
  • the pharmaceutical compositions of the present invention further comprise additional therapeutic agents.
  • the other therapeutic agent is selected from the group consisting of an antiarrhythmic agent, an antihypertensive agent, an anticoagulant, an antiplatelet agent, a thrombin inhibitor, a thrombolytic agent, a fibrinolytic agent, a calcium channel blocker, Potassium channel blockers, cholesterol/lipid lowering agents, or combinations thereof.
  • the present invention provides a pharmaceutical composition wherein the other therapeutic agent is an antihypertensive agent selected from the group consisting of an ACE inhibitor, an AT-1 receptor antagonist, a beta adrenergic receptor antagonist, ETA receptor antagonist, dual ETA/AT-1 receptor antagonist, renin inhibitor (alliskerin) and vasopressin inhibitor), antiarrhythmic agent (selected from IKur inhibition)
  • Anticoagulant which is selected from the group consisting of thrombin inhibitors, antithrombin-III activators, heparin cofactor II activators, other factor XIa inhibitors, other kallikrein inhibitors, plasmin Pro-activator inhibitor (PAI-1) antagonist, thrombin-activated fibrinolysis inhibitor (TAFI) inhibitor, factor VIIa inhibitor, factor IXa inhibitor and factor Xa inhibitor) or anti-platelet agent GPIIb/IIIa blocker, GP Ib/IX blocker, protease activated receptor 1 (PAR-1) antagonist, protease
  • the other therapeutic agents included in the pharmaceutical compositions of the invention are antiplatelet agents or a combination thereof.
  • the anti-platelet agent includes, but is not limited to, clopidogrel and/or aspirin or a combination thereof.
  • the pharmaceutical composition of the present invention comprises other therapeutic agents: warfarin, unfractionated heparin, low molecular weight heparin, synthetic pentasaccharide, hirudin, argatroban, aspirin, bupro Fen, naproxen, sulindac, indomethacin, mefima, dipyridamol, dioxicam, diclofenac, sulfinpyrazone, piroxicam, ticlopidine, clopidogrel, Tirofiban, eptifibatide, abciximab, melagatran, ximelagatran, hirudin sulfate, tissue plasminogen activator, modified tissue plasminogen activator Agent, anipase, urokinase, and streptokinase or a combination thereof.
  • other therapeutic agents warfarin, unfractionated heparin, low molecular weight heparin, synthetic pentasaccharide, hirudin, argat
  • pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable prodrugs, salts, esters, ester salts, or any other agent which can be administered, directly or indirectly, depending on the needs of the patient.
  • compositions of the present invention further comprise a pharmaceutically acceptable carrier, adjuvant, or excipient, as used herein, including any solvent, diluent, or other Liquid excipients, dispersing or suspending agents, surfactants, isotonic agents, thickeners, emulsifiers, preservatives, solid binders or lubricants, etc., are suitable for the particular target dosage form.
  • a pharmaceutically acceptable carrier including any solvent, diluent, or other Liquid excipients, dispersing or suspending agents, surfactants, isotonic agents, thickeners, emulsifiers, preservatives, solid binders or lubricants, etc.
  • Substances which may be used as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, aluminum, aluminum stearate, lecithin, serum proteins such as human serum albumin, buffer substances such as phosphate, glycine, sorbic acid, sorbus Potassium acid, a partial glyceride mixture of saturated vegetable fatty acids, water, salt or electrolyte, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salt, colloidal silicon, magnesium trisilicate, polyethylene Pyrrolidone, polyacrylate, wax, polyethylene-polyoxypropylene-blocking polymer, lanolin, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as carboxymethyl Cellulose sodium, ethyl cellulose and cellulose acetate; gum powder; malt; gelatin; talcum powder; excipients such as cocoa
  • the compounds of the present invention can be administered in the form of oral preparations, such as tablets, capsules (each of which includes a sustained release or timed release formulation), pills, powders, granules, elixirs, elixirs, suspensions, syrups. , and emulsifiers. They can also be administered intravenously (bolus or infusion), intraperitoneally, subcutaneously or intramuscularly. All dosage forms used are well known to those of ordinary skill in the pharmaceutical arts. They can be administered alone, but will generally be administered with a pharmaceutical carrier selected based on the mode of administration chosen and standard pharmaceutical practice.
  • the dosage regimen of the compounds of the invention will vary with various factors known, such as the pharmacokinetic profile of the particular agent and its mode and route of administration; the race, age, sex, health, medical condition and weight of the recipient; The nature and extent of the symptoms; the type of treatment being treated; the frequency of treatment; the route of administration, the kidney and liver function of the patient, and the desired effect.
  • a physician or veterinarian can make a decision and prescribe an effective amount of the drug to prevent, counteract or prevent the development of a thromboembolic disease.
  • the daily oral dose of each active ingredient used ranges from about 0.001 to 1000 mg/kg body weight, preferably between about 0.01 and 100 mg/kg body weight. . Moreover, most preferably, it is between about 1.0 and 20 mg/kg body weight per day.
  • the most preferred dosage range during a conventional rate of infusion is from about 1 to about 10 mg/kg body weight per minute.
  • the compounds of the invention may be administered once daily, or may be administered in two, three or four times daily.
  • the compounds of the invention may be administered in intranasal form via topical use of a suitable intranasal vehicle or by transdermal routes using transdermal patches.
  • a suitable intranasal vehicle or by transdermal routes using transdermal patches.
  • the dosage administered throughout the administration is continuous rather than intermittent.
  • the compound is administered in admixture with a suitable pharmaceutical diluent, excipient, or carrier (herein referred to as a pharmaceutical carrier), depending on the form of administration and conventional pharmaceutical practice, in the form of an oral tablet.
  • a suitable pharmaceutical diluent excipient, or carrier
  • a pharmaceutical carrier depending on the form of administration and conventional pharmaceutical practice, in the form of an oral tablet.
  • the active drug component can be combined with an oral, non-toxic, pharmaceutically acceptable inert carrier such as lactose, starch, sucrose, glucose, methylcellulose , magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol, etc.; for oral administration in liquid form, the oral pharmaceutical component can be combined with any oral, non-toxic, pharmaceutically acceptable inert carrier Combine, such as ethanol, glycerin, water, etc. Moreover, suitable binders, lubricants, decomposition agents, and colorants can also be added to the mixture as needed or necessary.
  • an oral, non-toxic, pharmaceutically acceptable inert carrier such as lactose, starch, sucrose, glucose, methylcellulose , magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol, etc.
  • any oral, non-toxic, pharmaceutically acceptable inert carrier Combine such as ethanol, glycerin,
  • Suitable binders include starch, gelatin, natural sugars (such as glucose or beta-lactose), corn sweeteners, natural and synthetic gums (such as acacia), tragacanth, or sodium alginate, carboxymethyl fibers. , polyethylene glycol, wax and so on.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like.
  • Decomposers include, but are not limited to, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
  • the compounds of the invention may also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles.
  • Liposomes can be formed by different phospholipids, such as cholesterol, stearylamine, or phosphatidylcholine.
  • the compounds of the invention are also coupled to a soluble polymer that acts as a targeted pharmaceutical carrier.
  • soluble polymer include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropyl methacrylate-phenol, polyhydroxyethylaspartamide phenol, or polyethylene oxide substituted with palmitoyl residues-poly Amino acid.
  • the compounds of the invention can be coupled to a class of biodegradable polymers for accomplishable controlled drug release, for example, polylactic acid, polyglycolic acid, copolymers of polylactic acid and polyglycolic acid, Crosslinked or amphiphilic blocking copolymers of esters, polyhydroxybutyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and hydrogels.
  • biodegradable polymers for accomplishable controlled drug release for example, polylactic acid, polyglycolic acid, copolymers of polylactic acid and polyglycolic acid, Crosslinked or amphiphilic blocking copolymers of esters, polyhydroxybutyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and hydrogels.
  • Each unit dose of a dosage form (pharmaceutical composition) suitable for administration may contain from about 1 mg to about 100 mg of the active ingredient.
  • the weight of the active ingredient will generally comprise from about 0.5% to about 95% by weight based on the total weight of the composition.
  • Gelatin capsules may contain the active ingredient as well as powder carriers such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like.
  • Compressed tablets can be made using similar diluents. Tablets and capsules can be made as a product of sustained release to provide a continuous release of the drug over a period of time.
  • Compressed tablets may be sugar coated or coated with a film to mask any unpleasant taste and to isolate the tablet from the air, or an enteric coating for selective decomposition in the gastrointestinal tract.
  • Liquid dosage forms for oral administration may contain coloring and flavoring to enhance patient acceptance.
  • water, a suitable oil, saline, hydrated dextrose (glucose), and related sugar solutions, as well as glycols such as propylene glycol or polyethylene glycol are suitable carriers for parenteral solutions.
  • the solution for parenteral administration preferably contains a water-soluble salt of the active ingredient, a suitable stabilizer, and possibly a buffer material.
  • the antioxidant is a suitable stabilizer such as sodium hydrogen sulfite, sodium sulfite, or vitamin C, either alone or in combination, or citric acid and a salt thereof, and sodium EDTA.
  • parenteral solutions also contain preservatives such as benzalkonium, methyl- or propyl-p-hydroxybenzoate, and chlorobutanol.
  • a daily dose may be from about 0.1 to 100 mg of a compound of the invention and from about 1 to 7.5 mg of a second anticoagulant combination.
  • the compound of the invention will generally be from about 5 to 10 mg per dosage unit, and the amount of the second anti-aggregating agent will be from about 1 to 5 mg per dosage unit.
  • anticoagulant reagents include, but are not limited to, apixaban, rivaroxaban, edoxaban, betrixaban, dabigatran (dabigatran), bemiparin, enoxaparin sodium, tinzaparin sodium, danaparoid sodium, pentosan sodium, nadroparin calcium, aspartame sodium, palparin sodium and the like.
  • the compounds of the invention may be used alone or in combination with other therapeutic agents, either sequentially or sequentially.
  • the other therapeutic agent is selected from the group consisting of a factor Xa inhibitor (eg, apixaban, rivaroxaban, betrixaban, edoxaban), an anticoagulant, an antiplatelet agent, thrombin inhibition Agents (eg, dabigatran), thrombolytic agents, and fibrinolytic agents.
  • the other therapeutic agent is at least one agent selected from the group consisting of warfarin, unfractionated heparin, low molecular weight heparin, synthetic pentasaccharide, hirudin, argatroban, aspirin, ibuprofen ( Ibuprofen), naproxen, sulindac, indomethacin, mefenamate, droxicam, diclofenac, sulfinpyrazone ), piroxicam, ticlopidine, clopidogrel, tirofiban, eptifibatide, abciximab, melagatran ), desulfatohirudin, tissue plasminogen activator, modified tissue plasminogen activator, anistreplase, urokinase, and streptokinase.
  • the additional therapeutic agent is at least one anti-platelet agent.
  • the antiplatelet agent is clopidogrel and/or aspirin or
  • the compounds of the invention are administered in combination with an anti-platelet agent.
  • a typical daily dose may be from about 0.01 to 300 mg of the compound of the invention per kg of body weight of the patient and from about 50 to 150 mg of the anti-platelet agent, preferably about 0.1 to 4 mg of a compound of the invention and about 1 to 3 mg of an anti-platelet agent.
  • the usual daily dose may be from about 0.1 to 100 mg of the compound of the present invention per kg of the patient's body weight, and in the presence of a thrombolytic agent, generally when administered alone with the thrombolytic agent.
  • the dosage of the thrombolytic agent can be reduced by about 50-80%.
  • each of the typical daily dosages and typical dosage forms may be decreased relative to the usual dose when administered alone.
  • one active ingredient can be an enteric coating.
  • an enteric coating By coating an active ingredient with an enteric coating, it is possible to not only minimize contact between the combined active ingredients, but it is also possible to control the release of one of these ingredients in the gastrointestinal tract so that one of these components It is not released in the stomach but released in the small intestine.
  • One of the active ingredients may also be coated with a material which affects its sustained release in the gastrointestinal tract and which may also be used to reduce physical contact between the combined active ingredients.
  • the sustained release component may also be additionally coated with an enteric coating. This ingredient is only released in the intestines.
  • Still another method involves the formulation of a combination product in which one component is coated with a continuous and/or enteric release polymer and the other component is also a polymer such as a low viscosity grade hydroxyl group.
  • HPMC Propylmethylcellulose
  • Polymer coating creates an additional barrier to reaction with other components.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof or a hydrate thereof can be effectively used for preventing, treating or ameliorating a thromboembolic disease in a patient;
  • the thromboembolic disorder includes an arterial cardiovascular thromboembolic disorder, venous cardiovascular thromboembolism Symptoms, arterial cerebrovascular thromboembolic disorders, and venous cerebrovascular thromboembolic disorders.
  • thromboembolic disorders include, but are not limited to, unstable angina, acute coronary syndrome, atrial fibrillation, first myocardial infarction, recurrent myocardial infarction, ischemic sudden death, transient ischemic attack, stroke, atherosclerosis Hardening, peripheral occlusive arterial disease, venous thrombosis, deep vein thrombosis, thrombophlebitis, arterial embolism, coronary thrombosis, cerebral arterial thrombosis, cerebral embolism, renal embolism, pulmonary embolism, and medical implants, The blood in the device or procedure is exposed to an artificial surface to promote thrombosis caused by thrombosis.
  • the compounds or pharmaceutical compositions provided herein can also be used to treat, prevent or treat inflammatory diseases; including, but not limited to, sepsis, acute respiratory distress syndrome, or systemic inflammatory response syndrome.
  • the compound or pharmaceutical composition provided by the present invention can be used for preventing, treating or ameliorating a disease associated with plasma kallikrein; wherein the disease associated with plasma kallikrein includes, but is not limited to, visual acuity Damage, diabetic retinopathy, diabetic macular edema, hereditary angioedema, diabetes, pancreatitis, kidney disease, cardiomyopathy, neuropathy, inflammatory bowel disease, arthritis, inflammation, septic shock, hypotension, cancer Adult respiratory distress syndrome, disseminated intravascular coagulation and cardiopulmonary bypass.
  • the disease associated with plasma kallikrein includes, but is not limited to, visual acuity Damage, diabetic retinopathy, diabetic macular edema, hereditary angioedema, diabetes, pancreatitis, kidney disease, cardiomyopathy, neuropathy, inflammatory bowel disease, arthritis, inflammation, septic shock, hypotension, cancer Adult respiratory distress syndrome, disseminated intravascular coagulation and cardiopulmonary bypass.
  • the compounds of the present invention can be prepared by the methods described herein, unless otherwise stated, wherein the substituents are as defined in (I) or Formula (Ia) or Formula (II) or Formula (IIa) or Formula (III) or formula (IIIa) or formula (IIIb) or formula (IIIc).
  • the following reaction schemes and Examples 1-11 are used to further illustrate the contents of the present invention.
  • the reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Arco Chemical Company and Alfa Chemical Company and were used without further purification unless otherwise indicated.
  • the general reagents were purchased from Shantou Xiqiao Chemical Plant, Guangdong Guanghua Chemical Reagent Factory, Guangzhou Chemical Reagent Factory, Tianjin Haoyuyu Chemical Co., Ltd., Qingdao Tenglong Chemical Reagent Co., Ltd., and Qingdao Ocean Chemical Plant.
  • Anhydrous tetrahydrofuran, dioxane, toluene and diethyl ether are obtained by refluxing with sodium metal.
  • Anhydrous dichloromethane and chloroform were obtained by reflux drying of calcium hydride.
  • Ethyl acetate, petroleum ether, n-hexane, N,N-dimethylacetamide and N,N-dimethylformamide were previously dried over anhydrous sodium sulfate.
  • reaction is generally carried out under a positive pressure of nitrogen or argon or on a dry solvent (unless otherwise indicated), the reaction bottle is stoppered with a suitable rubber stopper, and the substrate is driven through a syringe. The glassware is dried.
  • the column is a silica gel column.
  • Silica gel 300-400 mesh
  • Nuclear magnetic resonance spectroscopy data was determined by Bruker Avance 400 NMR spectrometer or Bruker Avance III HD 600 NMR spectrometer with CDC1 3 , DMSO-d 6 , CD 3 OD or acetone-d 6 as solvent (reported in ppm) TMS (0 ppm) or chloroform (7.25 ppm) was used as a reference standard.
  • MS mass spectrometry
  • the purity of the compound is characterized by: Agilent 1260 preparative high performance liquid chromatography (Pre-HPLC) or Calesep Pump 250 preparative high performance liquid chromatography (Pre-HPLC) (column model: NOVASEP, 50/80 mm, DAC) at 210 nm /254nm with UV detection.
  • Grubbs 2nd generation catalyst Grubb second generation catalyst, benzylidene-1,3-bis(2,4,6-trimethylphenyl)-2-(imidazoline carbene) (tricyclohexylphosphine) ruthenium dichloride
  • Dess-Martin oxidant (1,1,1-triacetoxy)-1,1-dihydro-1,2-phenyliodo-3(1H)-one
  • PEG8000 polyethylene glycol 8000
  • Compound 1c can be prepared by the method described in Synthesis Scheme 1.
  • HATU 2-(7-oxobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate
  • DBU monocarb-7-ene
  • Compound 2f can be prepared by the method described in Synthesis Scheme 2.
  • Compound 2a is first subjected to a Michael addition reaction with compound 2c in a solvent such as tetrahydrofuran or the like, and then subjected to a compound 2d under basic conditions (for example, under the action of pyridine or 4-dimethylaminopyridine).
  • a compound 2d under basic conditions (for example, under the action of pyridine or 4-dimethylaminopyridine).
  • Acetylation gives compound 2e.
  • Compound 2e is subjected to ring closure under basic conditions (e.g., sodium methoxide or sodium hydride) to give compound 2f.
  • basic conditions e.g., sodium methoxide or sodium hydride
  • the intermediate compound 2e can also be produced by the following method: Compound 2a is first added in the presence of trimethylchlorosilane (TMSCl) and sodium iodide (NaI) in a solvent (for example, acetonitrile and water). The reaction gives compound 2b; compound 2b and compound 2c undergo a substitution reaction under basic conditions (for example, under the action of triethylamine), and then under basic conditions (for example, in pyridine or 4-dimethylaminopyridine) The compound 2d is acetylated to give the compound 2e.
  • TMSCl trimethylchlorosilane
  • NaI sodium iodide
  • Compound 3h can be prepared by the method described in Synthetic Scheme 3.
  • Compound 3a is reacted with a vinyl Grignard reagent (such as vinylmagnesium bromide) to obtain compound 3b; compound 3b is subjected to an oxidation reaction (such as reaction with Jones (Jone's) reagent or Dess-Martin oxidant, etc.) to obtain compound 3c.
  • Compound 3c is first subjected to a Michael addition reaction with compound 3e in a solvent such as tetrahydrofuran or the like, and then acetylated by compound 3f under basic conditions (for example, under the action of pyridine or 4-dimethylaminopyridine).
  • Compound 3g is obtained; compound 3g is subjected to ring closure under basic conditions (e.g., under sodium methoxide, sodium hydride, etc.) to afford compound 3h.
  • the intermediate compound 3g can also be produced by the following method: the compound 3c is first subjected to an addition reaction in the presence of TMSCl and NaI in a solvent (for example, acetonitrile and water) to obtain a compound 3d; the compound 3d is further reacted with the compound 3e.
  • the substitution reaction occurs under basic conditions (for example, under the action of triethylamine), and then acetylated by 3f under basic conditions (for example, under the action of pyridine or 4-dimethylaminopyridine) to obtain a compound 3g. .
  • the intermediate compound 4m can be produced by the method described in Synthesis Scheme 4.
  • the aldehyde 4a and (S)-2-methylpropyl-2-sulfinamide are condensed in a solvent such as dichloromethane in the presence of anhydrous copper sulfate to give compound 4b.
  • Compound 4b is reacted with allyl magnesium bromide under the action of indium trichloride catalyzed to obtain compound 4c; compound 4c is subjected to acidic conditions (for example, under the action of HCl), and (S)-2-A is removed.
  • Base propyl-2-sulfinyl group gives compound 4d; compound 4d is amino protected to give compound 4e; compound 4e and compound 4f are in a catalyst (such as Pd(dppf)Cl 2 -CH 2 Cl 2 ) and a base (such as cesium carbonate)
  • a catalyst such as Pd(dppf)Cl 2 -CH 2 Cl 2
  • a base such as cesium carbonate
  • the Suzuki coupling reaction is carried out in a solvent such as dioxane and water to obtain a compound 4g; the compound 4g is subjected to a reduction reaction (for example, a nitro group is reduced in a zinc powder-ammonium chloride system) to obtain a compound 4h.
  • Compound 4h is substituted with compound 4i under basic conditions (such as pyridine) to obtain compound 4j; compound 4j is subjected to ring-closing metathesis reaction under the action of a catalyst (such as Grubbs 2 generation catalyst) to obtain macrocyclic compound 4k; compound 4k Hydrogenation by catalysis (e.g., Pd/C catalysis) affords compound 41; compound 41 is deprotected (e.g., the Boc protecting group is removed under acidic conditions such as trifluoroacetic acid) to afford compound 4m.
  • a catalyst such as Grubbs 2 generation catalyst
  • the intermediate compound 5i can be produced by the method described in Synthesis Scheme 5.
  • Compound 4e and compound 5b are subjected to Suzuki coupling reaction in a solvent such as dioxane and water in the presence of a catalyst such as Pd(dppf)Cl 2 -CH 2 Cl 2 ) and a base such as cesium carbonate.
  • a catalyst such as Pd(dppf)Cl 2 -CH 2 Cl 2
  • a base such as cesium carbonate.
  • Compound 5c; compound 5c is reacted by reduction (such as reduction of nitro group to amino group in zinc powder-ammonium chloride system) to obtain compound 5d;
  • compound 5d and compound 4i are substituted under basic conditions (such as pyridine) to obtain compound 5f.
  • Compound 5f is subjected to a ring-closing metathesis reaction in the presence of a catalyst (such as Grubbs 2nd generation catalyst) to obtain a macrocyclic compound 5g; compound 5g is catalyzed (e.g., Pd/C catalyzed) hydrogenation reduction to obtain compound 5h; compound 5h is deprotected (for example, Removal of the Boc protecting group under acidic conditions such as trifluoroacetic acid affords compound 5i.
  • a catalyst such as Grubbs 2nd generation catalyst
  • the intermediate compound 6j can be produced by the method described in Synthesis Scheme 6.
  • Compound 4e and compound 6b are subjected to Suzuki coupling reaction in a solvent such as dioxane and water in the presence of a catalyst such as Pd(dppf)Cl 2 -CH 2 Cl 2 ) and a base such as cesium carbonate.
  • a catalyst such as Pd(dppf)Cl 2 -CH 2 Cl 2
  • a base such as cesium carbonate.
  • the intermediate compound 7i can be produced by the method described in Synthesis Scheme 7.
  • Compound 4e and compound 7b are in the presence of a palladium (II) catalyst (such as Pd(OAC) 2 ), a phosphorus ligand (such as n-butylbis(1-adamantyl)phosphine), and a base (such as potassium carbonate).
  • a palladium (II) catalyst such as Pd(OAC) 2
  • a phosphorus ligand such as n-butylbis(1-adamantyl)phosphine
  • a base such as potassium carbonate
  • a coupling reaction occurs in a solvent (such as N, N-dimethylformamide) to obtain a compound 7c; a compound 7c is reduced (for example, a nitro group is reduced to an amino group in a zinc powder-ammonium chloride system) to obtain a compound 7d; Compound 7d and compound 4i are subjected to a substitution reaction in a solvent (such as dichloromethane) under basic conditions (such as pyridine) to obtain compound 7f; and compound 7f is subjected to a ring-closing metathesis reaction under the action of a catalyst (such as Grubbs 2 generation catalyst).
  • a solvent such as N, N-dimethylformamide
  • a compound 7c is reduced (for example, a nitro group is reduced to an amino group in a zinc powder-ammonium chloride system) to obtain a compound 7d
  • Compound 7d and compound 4i are subjected to a substitution reaction in a solvent (such as dichloromethane) under basic conditions (such
  • the ring compound 7g; the compound 7g is hydrogenated by catalytic (e.g., Pd/C catalysis) reduction to give the compound 7h, and then deprotected (for example, the Boc protecting group is removed under acidic conditions such as trifluoroacetic acid) to obtain the compound 7i.
  • catalytic e.g., Pd/C catalysis
  • deprotected for example, the Boc protecting group is removed under acidic conditions such as trifluoroacetic acid
  • Phosphorus oxybromide (8.9 g, 31 mmol) and sodium carbonate (4.3 g, 31 mmol) were added to a solution of 1B (2.1 g, 10 mmol) in anhydrous acetonitrile (45 mL). Stir at 92 ° C for 2 hours. Then it was cooled to room temperature, and the solvent was evaporated under reduced pressure. The residue was taken up in EtOAc (EtOAc)EtOAc.
  • Step 6 (S)-N-((S)-1-(4-bromoquinolin-2-yl)but-3-en-1-yl)-2-methylpropane-2-sulfinamide ( 1F)
  • Step 8 (S)-(1-(4-Bromoquinolin-2-yl)but-3-en-1-yl)carbamic acid tert-butyl ester (1H)
  • Step 9 (S)-(1-(4-(4-Amino-2-nitrophenyl)quinolin-2-yl)but-3-en-1-yl)carbamic acid tert-butyl ester (1I)
  • Step 10 (S)-(1-(4-(4-(methoxy)amino)-2-nitrophenyl)quinolin-2-yl)but-3-en-1-yl)carbamic acid Tert-butyl ester (1J)
  • Step 11 (S)-(1-(4-(4-(methoxy)amino)-2-aminophenyl)quinolin-2-yl)but-3-en-1-yl)carbamic acid Butyl ester (1K)
  • Zinc powder (4.26 g, 65.20 mmol) and ammonium chloride (3.49 g, 65.20 mmol) were added to a solution containing 1 J (3.21 g, 6.52 mmol) in methanol (65 mL) and the mixture was stirred at room temperature for 2.5 hours. .
  • the reaction was stopped, filtered, and the filter cake was washed with methanol (10 mL ⁇ 3), and the filtrate was concentrated to give residue.
  • the residue was dissolved in dichloromethane (100mL), washed with water (30mL ⁇ 2) and brine (30mL) The aqueous solution was dried with EtOAc (EtOAc m.
  • Step 12 (S)-(1-(4-(4-(methoxy)amino)-2-((R)-2-methylbut-3-enoylamino)phenyl)quinoline-2 -yl)but-3-en-1-yl)carbamic acid Butyl ester (1L)
  • Step 13 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-8,16-diazatetracyclo[13.7.1.0 2.7 .0 17, 22 ] icosane -1(23),2(3),4,6,11(12),15,17(22),18,20-nonenylene-5-yl)methyl carbamate (1M)
  • P-toluenesulfonic acid monohydrate (0.84 g, 4.28 mmol) was placed in a two-necked flask at room temperature, heated to 80 ° C, and dried under reduced pressure for about 1 hour.
  • a solution of 1 L (2.12 g, 3.89 mmol) in dry methylene chloride (100 mL) was then taken and then cooled to room temperature and stirred for 45 min.
  • a solution of Grubbs 2 Generation Catalyst (1.05 g, 1.22 mmol) in dry dichloromethane (20 mL) was then slowly taken and then warmed to reflux and stirred overnight. The reaction was quenched and cooled to room temperature.
  • Step 14 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-8,16-diazatetracyclo[13.7.1.0 2.7 .0 17, 22 ] icosane -1(23),2(3),4,6,15,17(22),18,20-octadec-5-yl)carbamate (1N)
  • Step 15 N - ((10R, 14S) -14- amino-10-methyl-9-oxo-8,16-diaza- tetracyclo [13.7.1.0 2.7 .0 17,22] tricosane -1(23),2(3),4,6,15,17(22),18,20-octadec-5-yl)carbamate (1O)
  • Trifluoroacetic acid (4.6 g, 3.0 mL, 40.00 mmol) was added to 1N (0.67 g, 1.28 mmol) in dry dichloromethane (30 mL) The reaction was quenched, EtOAc (EtOAc m. The combined organic layers were dried with EtOAc EtOAcjjjjjjjjj
  • Step 16 1-(6-Bromo-3-chloro-2-fluorophenyl)-3-iodopropan-1-one (1P)
  • Trimethylchlorosilane (0.78 g, 0.62 mL, 7.13 mmol) was added to a system containing sodium iodide (1.10 g, 7.3 mmol) and acetonitrile (5 mL) at room temperature, stirred for 10 min, then water was added (0.50g), after stirring for 5 minutes, adding 1-(6-bromo-3-chloro-2-fluorophenyl)prop-2-en-1-one (prepared by the synthesis method of intermediate 2 of patent WO 2014022766) (1.25 g, 4.74 mmol), stirring was continued for 2 hours. The reaction was stopped, water (5 mL) was added and the mixture was extracted with ethyl acetate (20mL ⁇ 3). The organic phase was combined, the organic phase was dried with anhydrous sodium sulfate, filtered, and the filtrate was evaporated to crystals crystals crystals crystals Liquid (1.35 g, 72.7%).
  • Step 17 N-((10R,14S)-14-(3-(6-bromo-3-chloro-2-fluorophenyl)-3-oxopropylamino)-10-methyl-9-oxo -8,16-diazatetracyclic [13.7.1.0 2.7 .0 17,22] tricosa-1 (23), 2 (3), 4,6,15,17 (22), 18,20- eight-5-yl) carbamic acid Methyl ester (1Q)
  • Step 18 N-((10R,14S)-14-(N-(3-(6-bromo-3-chloro-2-fluorophenyl)-3-oxopropyl)-2-(diethoxy) Phosphoryl)acetamido)-10-methyl-9- Oxo-8,16-diaza- tetracyclo [13.7.1.0 2.7 .0 17,22] tricosa-1 (23), 2 (3), 4,6,15,17 (22), 18 Methyl 20-octadec-5-yl)carbamate (1R)
  • Step 19 N-((10R,14S)-14-(4-(6-Bromo-3-chloro-4-fluorophenyl)-6-oxo-1,2,3,6-tetrahydropyridine- 1-yl)-10-methyl-9-oxo-8,16-diazo Oxatetracyclo [13.7.1.0 2,7 .0 17,22] tricosa-1 (23), 2 (7), 3,5,15,17 (22), 18,20- eight 5-ene Methyl carbamate
  • the organic layer was dried (MgSO4) (mjjjjjjj
  • Step 8 (S,E)-N-((4-Bromo-6,7-dihydro-5H-cyclopenta[b]pyridin-2-yl)methylene)-2-methylpropane- 2-sulfinamide (2H)
  • Step 9 (S)-N-((S)-1-(4-bromo-6,7-dihydro-5H-cyclopenta[b]pyridin-2-yl)but-3-ene-1 -yl)-2-methylpropane-2-sulfinamide (2I)
  • Step 10 (S)-1-(4-Bromo-6,7-dihydro-5H-cyclopenta[b]pyridin-2-yl)but-3-en-1-amine (2J)
  • Step 11 (S)-(1-(4-Bromo-6,7-dihydro-5H-cyclopenta[b]pyridin-2-yl)but-3-en-1-yl)carbamic acid Butyl ester (2K)
  • Step 12 (S)-(1-(4-(4-Amino-2-nitrophenyl)-6,7-dihydro-5H-cyclopenta[b]pyridin-2-yl)- Tert-butyl 3-en-1-yl)carbamate (2L)
  • Step 13 N-[4-[2-[(S)-1-(tert-Butoxycarbonylamino)but-3-enyl]-6,7-dihydro-5H-cyclopenta[b]pyridine 4-yl]-3-nitro-phenyl]amino Methyl formate (2M)
  • Step 14 N-[3-Amino-4-[2-[(S)-1-(tert-butoxycarbonylamino)but-3-enyl]-6,7-dihydro-5H-cyclopentane [b]pyridin-4-yl]phenyl]carbamate Methyl ester (2N)
  • Zinc powder (10.03 g, 153.3 mmol) and ammonium chloride (8.36 g, 156 mmol) were added to a solution of 2M (7.32 g, 15.2 mmol) in methanol (200 mL) at room temperature overnight.
  • the reaction was terminated, the reaction solution was suction filtered with celite, and the filter cake was washed with methanol (30mL ⁇ 3), the solvent was evaporated under reduced pressure, and the residue was dissolved in dichloromethane (500 mL)
  • the organic layer was dried over anhydrous sodium sulfate and filtered and evaporated to dryness crystals
  • Step 15 N-[4-[2-[(1S)-1-(tert-Butoxycarbonylamino)but-3-enyl]-6,7-dihydro-5H-cyclopenta[b]pyridine 4-yl]-3-[[(2S)-2-methylbutyl) Methyl 3-enoyl]amino]phenyl]carbamate (2O)
  • Step 16 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-8,16-diazatetracyclo[13.6.1.0 2.7 .0 17, 21 ]tetracosane -1(22),2(3),4,6,11(12),15,17(21)-hepten-5-yl)carbamic acid methyl ester (2P)
  • p-toluenesulfonic acid monohydrate (0.297 g, 1.56 mmol) was weighed into a two-necked bottle, and the two vials were placed in an 80 ° C oil bath and dried under reduced pressure for 0.5 hour. The oil bath was heated to dryness, cooled to room temperature, and a solution of EtOAc (EtOAc) A solution of Grubbs 2 generation catalyst (0.369 g, 0.435 mmol) in dry dichloromethane (50 mL) was then added dropwise to the system and the mixture was stirred at 45 ° C overnight.
  • EtOAc EtOAc
  • Step 17 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-8,16-diazatetracyclo[13.6.1.0 2.7 .0 17, 21 ]tetracosane -1(22),2(3),4,6,15,17(21)-hexaen-5-yl)carbamic acid methyl ester (2Q)
  • Step 18 N - ((10R, 14S) -14- amino-10-methyl-9-oxo-8,16-diaza- tetracyclo [13.6.1.0 2.7 .0 17,21] behenic -1(22),2(3),4,6,15,17(21)-hexaen-5-yl)carbamic acid methyl ester (2R)
  • Step 19 N-((10R,14S)-14-(3-(6-Bromo-3-chloro-4-fluorophenyl)-3-oxopropylamino)-10-methyl-9-oxo -8,16-diazatetracyclic [13.6.1.0 2.7 .0 17,21 ]Tetradane-1(22),2(3),4,6,15,17(21)-hexaen-5-yl)carbamic acid methyl ester (2S )
  • Step 20 N-((10R,14S)-14-(N-(3-(6-bromo-3-chloro-4-fluorophenyl)-3-oxopropyl)-2-(diethoxy) Phosphoryl)acetamido)-10-methyl-9- Oxo-8,16-diazatetracyclo[13.6.1.0 2.7 .0 17,21 ]tetracosane-1(22), 2(3), 4,6,15,17(21)-six Methyl ene-5-yl)carbamate (2T)
  • Step 21 N-((10R,14S)-14-(4-(6-bromo-3-chloro-2-fluorophenyl)-6-oxo-1,2,3,6-tetrahydropyridine- 1-yl)-10-methyl-9-oxo-8,16-diazo Heterotetracycline [13.6.1.0 2,7 .0 17,21 ]tetracosane-1(22), 2(7),3,5,15,17(21)-hexa-5-yl)amino Methyl formate
  • Step 5 (S)-N-((4-Bromo-5,6,7,8-tetrahydroquinolin-2-yl)methylene)-2-methylpropane-2-sulfinamide (3E )
  • Step 6 (S)-N-((S)-1-(4-Bromo-5,6,7,8-tetrahydroquinolin-2-yl)but-3-en-1-yl)-2 -methylpropane-2-sulfinamide (3F)
  • Step 8 (S)-(1-(4-Bromo-5,6,7,8-tetrahydroquinolin-2-yl)but-3-en-1-yl)carbamic acid tert-butyl ester (3H)
  • Step 9 (S)-(1-(4-(4-Amino-2-nitrophenyl)-5,6,7,8-tetrahydroquinolin-2-yl)but-3-ene-1 -yl)-tert-butyl carbamate (3I)
  • Step 10 (S)-(1-(4-(4-(methoxy)amino)-2-nitrophenyl)-5,6,7,8-tetrahydroquinolin-2-yl) Tert-butyl 3--3-en-1-ylcarbamate (3J)
  • Step 11 (S)-(1-(4-(4-(methoxy)amino)-2-aminophenyl)-5,6,7,8-tetrahydroquinolin-2-yl)- Tert-butyl 3-en-1-yl)carbamate (3K)
  • Zinc powder (4.51 g, 69.03 mmol) and ammonium chloride (3.71 g, 69.0 mmol) were added to a solution containing 3J (3.41 g, 6.87 mmol) in methanol (60 mL) at room temperature. hour.
  • the reaction was stopped, filtered, and the filter cake was washed with methanol (10 mL ⁇ 3), and the filtrate was concentrated.
  • the residue was dissolved in dichloromethane (100 mL), washed with water (30mL ⁇ 2) and brine (30mL)
  • the sodium was dried, filtered, and evaporated tolulululululululululululululululululululululululululululululululu
  • Step 12 (S)-(1-(4-(4-(methoxy)amino)-2-((R)-2-methylbut-3-enoylamino)phenyl)-5,6 ,7,8-tetrahydroquinolin-2-yl)but-3-en-1-yl) Tert-butyl carbamate (3L)
  • Step 13 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-8,16-diazatetracyclo[13.7.1.0 2.7 .0 17, 22 ] icosane -1(23),2(7),3,5,11(12),15,17(22)-hepten-5-yl)carbamic acid methyl ester (3M)
  • Pentyl p-toluenesulfonic acid monohydrate (0.45 g, 2.30 mmol) was placed in a two-necked flask at room temperature, heated to 80 ° C, and dried under reduced pressure for about 1 hour. After cooling to room temperature, a solution of 3 L (1.15 g, 2.10 mmol) in dry dichloromethane (100 mL) was then poured into the system and stirred for 30 min. A solution of Grubbs 2 Generation Catalyst (0.54 g, 0.63 mmol) in dry dichloromethane (20 mL) was then taken and evaporated. The reaction was quenched and cooled to room temperature.
  • Step 14 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-8,16-diazatetracyclo[13.7.1.0 2.7 .0 17, 22 ] icosane -1(23),2(7),3,5,15,17(22)-hexaen-5-yl)carbamic acid methyl ester (3N)
  • Step 15 N - ((10R, 14S) -14- amino-10-methyl-9-oxo-8,16-diaza- tetracyclo [13.7.1.0 2.7 .0 17,22] tricosane -1(23),2(7),3,5,15,17(22)-hexaen-5-yl)carbamic acid methyl ester (3O)
  • Trifluoroacetic acid (4.61 g, 3.0 mL, 40.02 mmol) was added to 3N (0.69 g, 1.33 mmol) in dry dichloromethane (30 mL) The reaction was quenched, EtOAc (EtOAc m. The combined organic layers were dried with EtOAc EtOAc EtOAcjjjjjjj
  • Step 16 N-((10R,14S)-14-(3-(6-Bromo-3-chloro-2-fluorophenyl)-3-oxopropylamino)-10-methyl-9-oxo -8,16-diazatetracyclic [13.7.1.0 2.7 .0 17,22] tricosa-1 (23), 2 (7), 3,5,15,17 (22) - Six-5-yl) carbamate (3P )
  • Step 17 N-((10R,14S)-14-(N-(3-(6-bromo-3-chloro-2-fluorophenyl)-3-oxopropyl)-2-(diethoxy) Phosphoryl)acetamido)-10-methyl-9- Oxo-8,16-diaza- tetracyclo [13.7.1.0 2.7 .0 17,22] tricosa-1 (23), 2 (7), 3,5,15,17 (22) - six Methyl ene-5-yl)carbamate (3Q)
  • Step 18 N-((10R,14S)-14-(4-(6-Bromo-3-chloro-4-fluorophenyl)-6-oxo-1,2,3,6-tetrahydropyridine- 1-yl)-10-methyl-9-oxo-8,16-diazo Oxatetracyclo [13.7.1.0 2,7 .0 17,22] tricosa-1 (23), 2 (7), 3,5,15,17 (22) - Six-5-yl) amino Methyl formate
  • Hexylphosphine (8.30 g, 29.6 mmol), (dba) 3 Pd 2 (9.00 g, 9.83 mmol), 4,4,5,5-tetramethyl-2-vinyl-1,3,2-dioxaboron Pentacyclo(36.0 mL, 212 mmol) and N,N-dimethylformamide (900 mL) were then stirred and stirred at 100 ° C overnight.
  • the mixture was cooled to room temperature, filtered, and evaporated, evaporated, mjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjj
  • Step 10 (S)-N-((S)-1-(8-Bromo-3,4-dihydro-2H-pyrano[3,2-b]pyridin-6-yl)but-3- En-1-yl)-2-methylpropane-2-sulfinamide (4J)
  • Step 11 (S)-1-(8-Bromo-3,4-dihydro-2H-pyrano[3,2-b]pyridin-6-yl)but-3-en-1-amine (4K )
  • Step 12 (S)-(1-(8-Bromo-3,4-dihydro-2H-pyrano[3,2-b]pyridin-6-yl)but-3-en-1-yl) Tert-butyl carbamate (4L)
  • Step 13 (S)-(1-(8-(4-Amino-2-nitrophenyl)-3,4-dihydro-2H-pyrano[3,2-b]pyridine-6-yl Tert-butyl 3-buten-1-yl)carbamate (4M)
  • Step 14 (S)-(1-(8-(4-(methoxy)amino)-2-nitrophenyl)-3,4-dihydro-2H-pyrano[3,2-b Pyridine-6-yl)but-3-en-1-yl)carbamate Tert-butyl acid ester (4N)
  • Step 15 (S)-(1-(8-(4-(methoxy)amino)-2-aminophenyl)-3,4-dihydro-2H-pyrano[3,2-b] Pyridin-6-yl)but-3-en-1-yl)carbamate Tert-butyl acid ester (4O)
  • Step 16 (S)-(1-(8-(4-(methoxy)amino)-2-((R)-2-methylbut-3-enoylamino)phenyl)-3,4 -dihydro-2H-pyrano[3,2-b]pyridine-6- Tert-butyl 3-buten-1-yl)carbamate (4P)
  • Step 17 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-21-oxa-8,16-diazatetracyclo[13.7.1.0 2,7 .0 17,22 ]Twenty Trioxane-1(23), 2(3),5,6,11(12),15,17(22)-hepten-5-yl)carbamic acid methyl ester (4Q)
  • Step 18 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-21-oxa-8,16-diazatetracyclo[13.7.1.0 2,7 .0 17,22 ]Twenty Trioxane-1(23), 2(3),5,6,15,17(22)-hexaen-5-yl)carbamic acid methyl ester (4R)
  • Step 19 N-((10R,14S)-14-Amino-10-methyl-9-oxo-21-oxa-8,16-diazatetracyclo[13.7.1.0 2,7 .0 17 ,22 ]tetracosane -1(23),2(3),5,6,15,17(22)-hexaen-5-yl)carbamic acid methyl ester (4S)
  • Step 20 N-((10R,14S)-14-(3-(6-bromo-3-chloro-2-fluorophenyl)-3-oxopropylamino)-10-methyl-9-oxo -21-oxa-8,16-diazatetracyclo [13.7.1.0 2,7 .0 17,22] tricosa-1 (23), 2 (3), 5,6,15,17 (22) - Six-5-yl) carbamate (4T)
  • Step 21 N-((10R,14S)-14-(N-(3-(6-bromo-3-chloro-2-fluorophenyl)-3-oxopropyl)-2-(diethoxy) Phosphoryl)acetamido)-10-methyl-9- -21- oxo-oxa-8,16-diaza- tetracyclo [13.7.1.0 2,7 .0 17,22] tricosa-1 (23), 2 (3), 5,6,15 ,17(22)-hexaen-5-yl)carbamic acid methyl ester (4U)
  • Step 22 N-((10R,14S)-14-(4-(6-Bromo-3-chloro-4-fluorophenyl)-6-oxo-1,2,3,6-tetrahydropyridine- 1-yl)-10-methyl-9-oxo-21-oxa 8,16-diaza- tetracyclo [13.7.1.0 2,7 .0 17,22] tricosa-1 (23), 2 (7), 3,5,15,17 (22) - six Methyl ene-5-yl)carbamate
  • Potassium hydroxide 38 g, 677.3 mmol was weighed into a flask, and water (260 mL), 2-bromopyridin-3-ol (100 g, 574.71 mmol), EDTA-2Na (4.36 g, 13.0 mmol) and formaldehyde were added thereto.
  • Aqueous solution (38%, 156 mL) was warmed to 90 ° C for 5 hours. The reaction was stopped, cooled to room temperature, neutralized with acetic acid, and extracted with ethyl acetate (100 mL ⁇ 2). The organic phase was washed with water (50 mL) and brine (50 mL) Yellow solid (101 g, 86.14%).
  • 5D (23.06 g, 152.7 mmol) was weighed into a flask, and dichloromethane (500 mL) and m-chloroperoxybenzoic acid (31 g, 152.7 mmol) were added thereto, and reacted at room temperature for 24 hours. The reaction was quenched and EtOAc (EtOAc)EtOAc.
  • Step 9 (S,E)-N-((7-Bromo-2,3-dihydrofuro[3,2-b]pyridin-5-yl)methylene)-2-methylpropane-2 - sulfinamide (5I)
  • Step 10 (S)-N-((S)-1-(7-Bromo-2,3-dihydrofuro[3,2-b]pyridin-5-yl)but-3-ene-1- Base)-2-methylpropane-2-sulfinamide (5J)
  • Step 11 (S)-1-(7-Bromo-2,3-dihydrofuro[3,2-b]pyridin-5-yl)but-3-en-1-amine (5K)
  • Step 12 (S)-(1-(7-Bromo-2,3-dihydrofuro[3,2-b]pyridin-5-yl)but-3-en-1-yl)carbamic acid tert-butyl Ester (5L)
  • Step 13 (S)-(1-(7-(4-Amino-2-nitrophenyl)-2,3-dihydrofuro[3,2-b]pyridin-5-yl)butene-3 -en-1-yl)carbamic acid tert-butyl ester (5M)
  • Step 14 (S)-(1-(7-(4-(methoxy)amino)-2-nitrophenyl)-2,3-dihydrofuro[3,2-b]pyridine-5 -yl)but-3-en-1-yl)carbamic acid Butyl ester (5N)
  • Step 15 (S)-(1-(7-(4-(methoxy)amino)-2-aminophenyl)-2,3-dihydrofuro[3,2-b]pyridine-5- Tert-but-3-en-1-yl)carbamic acid Butyl ester (5O)
  • Zinc powder (15.36 g, 234.8 mmol) and ammonium chloride (12.58 g, 784.2 mmol) were added to a solution of 5N (11.4 g, 23.5 mmol) in methanol (300 mL) at room temperature for 12 hours at room temperature.
  • the reaction was terminated, the reaction solution was suction filtered with celite, and the filter cake was washed with methanol (50mL ⁇ 3), the solvent was evaporated under reduced pressure, and the residue was dissolved in dichloromethane (300 mL). Washed with saturated brine (300 mL), dried over anhydrous sodium sulfate.
  • Step 16 (S)-(1-(7-(4-(methoxy)amino)-2-((S)-2-methylbut-3-enoylamino)phenyl)-2,3 -dihydrofuro[3,2-b]pyridin-5-yl) Tert-butyl 3--3-en-1-ylcarbamate (5P)
  • Step 17 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-20-oxa-8,16-diazatetracyclo[13.6.1.0 2.7 .0 17,21 ] twenty Dioxane-1(22),2(3),4,6,11(12),15,17(21)-hepten-5-yl)carbamic acid methyl ester (5Q)
  • Step 18 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-20-oxa-8,16-diazatetracyclo[13.6.1.0 2.7 .0 17,21 ] twenty Dioxane-1(22),2(3),4,6,15,17(21)-hexaen-5-yl)carbamic acid methyl ester (5R)
  • Step 19 N-((10R,14S)-14-Amino-10-methyl-9-oxo-20-oxa-8,16-diazatetracyclo[13.6.1.0 2.7 .0 17,21 Tetane -1(22),2(3),4,6,15,17(21)-hexaen-5-yl)carbamic acid methyl ester (5S)
  • Step 20 N-((10R,14S)-14-(3-(6-bromo-3-chloro-2-fluorophenyl)-3-oxopropylamino)-10-methyl-9-oxo -20-oxa-8,16-diazatetracyclo [13.6.1.0 2.7 .0 17,21 ]Tetradane-1(22),2(3),4,6,15,17(21)-hexaen-5-yl)carbamic acid methyl ester (5T )
  • Step 21 N-((10R,14S)-14-(N-(3-(6-bromo-3-chloro-2-fluorophenyl)-3-oxopropyl)-2-(diethoxy) Phosphoryl)acetamido)-10-methyl-9- Oxo-20-oxa-8,16-diazatetracyclo[13.6.1.0 2.7 .0 17,21 ]tetracosane-1 (22), 2 (3), 4, 6, 15, 17 (21)-hexaen-5-yl)carbamic acid methyl ester (5U)
  • Step 22 N-((10R,14S)-14-(4-(6-bromo-3-chloro-2-fluorophenyl)-6-oxo-1,2,3,6-tetrahydropyridine- 1-yl)-10-methyl-9-oxo-20-oxa -8,16-diazatetracyclo[13.6.1.0 2,7 .0 17,21 ]docosane-1(22),2(7),3,5,15,17(21)-six Methyl ene-5-yl)carbamate
  • Step 3 8-Iodo-2,3-dihydro-[1,4]dioxa[2,3-b]pyridine-6-carbaldehyde (6C)
  • Step 4 (S,E)-N-((8-iodo-2,3-dihydro-[1,4]dioxa[2,3-b]pyridin-6-yl)methylene)- 2-methylpropane-2-sulfinamide (6D)
  • Step 5 (S)-N-((S)-1-(8-iodo-2,3-dihydro-[1,4]dioxa[2,3-b]pyridin-6-yl) 3--3-en-1-yl)-2-methylpropane-2-sulfinamide (6E)
  • Step 6 (S)-1-(8-iodo-2,3-dihydro-[1,4]dioxa[2,3-b]pyridin-6-yl)but-3-ene-1- Amine (6F)
  • Step 7 (S)-(1-(8-iodo-2,3-dihydro-[1,4]dioxino[2,3-b]pyridin-6-yl)but-3-ene-1 -yl)-tert-butyl carbamate (6G)
  • Step 8 (S)-(1-(8-(4-Amino-2-nitrophenyl)-2,3-dihydro-[1,4]dioxa[2,3-b]pyridine- 6-yl)but-3-en-1-yl)carbamic acid tert-butyl ester (6H)
  • Step 9 (S)-(1-(8-(4-(methoxy)amino)-2-nitrophenyl)-2,3-dihydro-[1,4]dioxin[2, 3-b]pyridin-6-yl)but-3-en-1-yl)carbamate Tert-butyl acid ester (6I)
  • Step 10 (S)-(1-(8-(4-(methoxy)amino)-2-aminophenyl)-2,3-dihydro-[1,4]dioxo[2,3 -b]pyridine-6-yl)but-3-en-1-yl)amino Tert-butyl formate (6J)
  • Step 11 (S)-(1-(8-(4-(methoxy)amino)-2-((R)-2-methylbut-3-enoylamino)phenyl)-2,3 -dihydro-[1,4]dioxa-[2,3-b]pyridine-6- Tert-butyl 3-buten-1-yl)carbamate (6K)
  • Step 12 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-18,21-dioxa-8,16-diazatetracyclo[ 13.7.1.0 2,7 .0 17,22] Tridecane-1(23), 2(7),3,5,11(12),15,17(22)-hepten-5-yl)carbamic acid methyl ester (6L)
  • Step 13 N-((10R,14S)-14-(tert-Butoxycarbonylamino)-10-methyl-9-oxo-18,21-dioxa-8,16-diazatetracyclo[ 13.7.1.0 2,7 .0 17,22] Tridecane-1(23),2(7),3,5,15,17(22)-hexaen-5-yl)carbamic acid methyl ester (6M)
  • Step 14 N-((10R,14S)-14-Amino-10-methyl-9-oxo-18,21-dioxa-8,16-diazatetracyclo[13.7.1.0 2,7 .0 17,22 ]tetracosane -1(23),2(7),3,5,15,17(22)-hexaen-5-yl)carbamic acid methyl ester (6N)
  • Step 15 N-((10R,14S)-14-(3-(6-Bromo-3-chloro-2-fluorophenyl)-3-oxopropylamino)-10-methyl-9-oxo -18,21-dioxa-8,16-diaza Tetracyclo [13.7.1.0 2,7 .0 17,22] tricosa-1 (23), 2 (7), 3,5,15,17 (22) - Six-5-yl) carbamic acid Methyl ester (6O)
  • Step 16 N-((10R,14S)-14-(N-(3-(6-bromo-3-chloro-2-fluorophenyl)-3-oxopropyl)-2-(diethoxy) Phosphoryl)acetamido)-10-methyl-9- Oxo-dioxa-8,16-diaza -18,21- tetracyclo [13.7.1.0 2,7 .0 17,22] tricosa-1 (23), 2 (7), 3, 5,15,17(22)-hexaen-5-yl)carbamic acid Methyl ester (6P)
  • Step 17 N-((10R,14S)-14-(4-(6-Bromo-3-chloro-4-fluorophenyl)-6-oxo-1,2,3,6-tetrahydropyridine- 1-yl)-10-methyl-9-oxo-18,21-dioxo Heteroaryl 8,16-diaza tetracyclo [13.7.1.0 2,7 .0 17,22] tricosa-1 (23), 2 (7), 3,5,15,17 (22) - Methyl hexene-5-yl)carbamate
  • Step 3 (S)-2-Methyl-N-((S)-1-(1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazole-3- Butyr-3-en-1-yl)propane-2-sulfinamide (7C)
  • Step 4 (S)-N-((S)-1-(1H-indazol-3-yl)but-3-en-1-yl)-2-methylpropane-2-sulfinamide (7D )
  • Step 5 (4-(3-((S)-1-((S)-1,1-dimethylethylsulfonylamino)but-3-en-1-yl)-1H-carbazole Methyl -1-yl)-3-nitrophenyl)carbamate (7E)

Abstract

提供了一类具有抑制凝血因子XIa和/或血浆激肽释放酶活性的大环化合物及其药物组合物;其中,所述大环化合物及其药物组合物可用于治疗或预防血栓栓塞性疾病。

Description

凝血因子XIa抑制剂及其用途 发明领域
本发明属于药物领域,涉及凝血因子XIa抑制剂及其用途,具体涉及一类具有抑制凝血因子XIa和/或血浆激肽释放酶的大环化合物及其药物组合物;其中,所述大环化合物及其药物组合物可用于治疗或预防血栓栓塞性疾病。
背景技术
促凝血(止血)和抗凝血(抗栓)是人体血液***中两种对立的机制,互为矛盾并保持相对平衡,这一精准和谐的过程维持着循环***的完整性。当体内抗凝纤溶***功能逐渐降低,血液中凝血与抗凝血功能失去平衡时则出现凝血,从而引起血栓或栓塞,进而导致诸如心肌梗死、中风、深度静脉血栓、肺栓塞等血栓栓塞性疾病。血栓栓塞性疾病是心血管疾病中危害最严重的疾病,是人类健康的第一杀手。
随着血栓形成机制的进一步阐明,针对血栓形成的特点和原因,研究和开发了很多抗血栓形成的新药,既有抑制血栓形成(抗凝血,例如,华法林、肝素、低分子肝素等)和抑制血小板聚集(例如,阿司匹林、氯格雷等)的药物,又有溶解血栓的药物。前者主要是对血栓的形成和增大具有抑制作用,后者主要是将已形成的血栓进行溶解,从而消除血栓性疾病对人类造成的危害。目前,临床应用的抗凝血药物的使用仍受各种风险(例如,出血)所限制,发现和研发用于预防和治疗广泛血栓栓塞障碍的安全且有效的口服抗凝血药物变得日益重要。
通过抑制凝血因子XIa(FXIa)可以抑制凝血酶的产生,从而达到抗凝血的治疗效果。因子XIa为涉及凝血调节的血浆丝氨酸蛋白酶,凝血是在体内由组织因子(TF)结合至因子VII(FVII)产生因子VIIa(FVIIa)而引发的。其中所产生的TF:FVIIa复合物活化因子IX(FIX)和因子X(FX),从而引起因子Xa(FXa)的产生。在此路径被组织因子途径抑制因子(TFPI)阻断之前,所产生的FXa催化凝血酶原转化成少量凝血酶,催化量的凝血酶反馈活化因子V、VIII和XI,从而使凝血过程进一步扩展(Gailani,D.等人,Arterioscler.Thromb.Vasc.Biol.,2007,27:2507-2513)。这一过程所引起的凝血酶爆发会使纤维蛋白原转化成纤维蛋白,纤维蛋白聚合形成血液凝块的结构构架,且活化作为凝血的关键细胞组分的血小板(Hoffman,M.,Blood Reviews,2003,17:S1-S5)。因此,因子XIa在扩展凝血过程的放大回路中起关键作用,也因此它可以作为有吸引力的抗抗血栓治疗的标靶。
血浆激肽释放酶原(plasma prekallikrein)是一种胰蛋白酶类丝氨酸蛋白酶的酶原,其以35μg/mL至50μg/mL的浓度存在于血浆中。血浆激肽释放酶原的基因结构类似于因子XI;总体而言,血浆激肽释放酶(plasma kallikrein,PK)的氨基酸序列与因子XI具有58%的同源性。人们认为,血浆激肽释放酶在许多炎性病症中起作用。血浆激肽释放酶的主要抑制剂为丝酶抑制蛋白(serpin)C1酯酶抑制剂。C1酯酶抑制剂遗传缺陷的患者患有遗传性血管水肿(HAE),从而引起面部、手部、咽喉、胃肠道和生殖器的间歇性肿胀;在急性发作期间形成的水泡中含有大量血浆激肽释放酶,其裂解高分子量的激肽原,释放缓激肽,从而导致血管渗透性的增加。已有研究表明,大蛋白血浆激肽释放酶抑制剂可以通过防止能引起血管渗透性增加的血管舒缓激肽的释放来有效治疗HAE(A.Lehmann,Ecallantide(DX-88),a plasma kallikrein inhibitor for thetreatment of hereditary angioedema and the prevention of blood loss in on-pumpcardiothoracic surgery,Expert Opin.Biol.Ther.8,第1187-99页)。
激肽释放酶-激肽***(kallikrein kinin system,KKS)是一个复杂的内源性多酶***,参与调控心血管、肾脏、神经***等的生理功能,与心脏病、肾病、炎症反应、癌症等疾病的发生有着密切关系。近年来在心血管***方面的研究进展很快,许多临床研究和基础实验已证实糖尿病、高血压、心力衰竭、心肌梗死及左心室肥厚等疾病的发生与KKS的活性降低有关。有研究发现,在心肌梗死患者的血中,因子XI (FXI)、高分子量激肽原(HK)和激肽释放酶原(prekallikrein,PK)的水平显著升高,表明它们对心肌梗死的发生可能起一定的作用(Merlo C.,et al.Elevated levels of plasma prekallikrein,high molecular weight kininoen and factor XI in coronary heart disease[J].Atherosclerosis,2002,161(2):261-267.)。因而深入研究KKS的作用为研究心血管相关疾病的发病机制和治疗手段提供了又一新的途径。
发明内容
本发明提供一种适用于丝氨酸蛋白酶,特别是因子XIa和/或血浆激肽释放酶的选择性抑制剂的大环化合物、其类似物以及包含所述大环化合物的药物组合物,该类化合物或药物组合物可以有效治疗和预防的血栓栓塞性疾病。
一方面,本发明提供一种化合物,其为如式(I)所示的化合物或式(I)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
Figure PCTCN2018072988-appb-000001
其中,
环A为C 7-12碳环基、C 8-12芳基、7-12个原子组成的杂环基或7-12个原子组成的杂芳基;
环B为C 5-12碳环基、C 6-12芳基、5-12个原子组成的杂环基或5-12个原子组成的杂芳基;
环C为C 3-12碳环基、C 6-12芳基、3-12个原子组成的杂环基或5-12个原子组成的杂芳基;
X为C 4-8亚烷基或C 4-8亚烯基,其中,所述亚烷基和亚烯基中一个或多个碳原子独立任选地被选自-O-、-S-、-C(=O)-、-S(=O)-、-S(=O) 2-或-N(R 5)-的基团所替代;X中的C 4-8亚烷基和C 4-8亚烯基独立任选地被一个或多个R 9所取代;
Y为-C(=O)N(R 5)-或-N(R 5)C(=O)-;
各R 1独立地为氧代(=O)、H、氘、卤素、-(CR 7aR 7b) rCN、C 1-6烷基、氘代C 1-6烷基、卤代C 1-6烷基、C 2-6烯基、C 2-6炔基、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=O)R 6、-(CR 7aR 7b) rC(=O)OR 8、-(CR 7aR 7b) rC(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O)R 6、-(CR 7aR 7b) rS(=O)OR 8、-(CR 7aR 7b) rS(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-12环烷基、-(CR 7aR 7b) r-C 6-12芳基、-(CR 7aR 7b) r-(3-12个原子组成的杂环基)或-(CR 7aR 7b) r-(5-12个原子组成的杂芳基);其中,各R 1独立任选地被1、2、3或4个R 1a所取代;
各R 2独立地为H、氘、卤素、-(CR 7aR 7b) rCN、C 1-6烷基、氘代C 1-6烷基、卤代C 1-6烷基、C 2-6烯基、C 2-6炔基、R 8O-C 2-6亚烯基、R 8OC(=O)-C 2-6亚烯基、R 8O-C 2-6亚炔基、R 8OC(=O)-C 2-6亚炔基、-N=C=S、-N=C=O、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=G)R 6、-(CR 7aR 7b) rN(R 5c)C(=G)R 6、-(CR 7aR 7b) rOC(=G)R 6、-(CR 7aR 7b) rC(=G)OR 8、-(CR 7aR 7b) rN(R 5c)C(=G)OR 8、-(CR 7aR 7b) rOC(=G)OR 8、-(CR 7aR 7b) rC(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)=C(R 7)NR 5aR 5b、-(CR 7aR 7b) rS(=O)R 6、-(CR 7aR 7b) rN(R 5c)S(=O)R 6、-(CR 7aR 7b) rS(=O)OR 8、-(CR 7aR 7b) rN(R 5c)S(=O)OR 8、-(CR 7aR 7b) rS(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rN(R 5c)S(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-12环烷基、-(CR 7aR 7b) r-C 6-12芳基、-(CR 7aR 7b) r-(3-12个原子组成的杂环基)、-(CR 7aR 7b) r-(5-12个原子组成的杂芳基)或D-甘露糖基;其中,各G独立地为O、S或NR 5d;各R 2独立任选地被1、2、3或4个R 2a所取代;
各R 3独立地为C 1-6烷基、C 3-12碳环基、C 6-12芳基、3-12个原子组成的杂环基或5-12个原子组成的杂 芳基;其中,各R 3独立任选地被1、2、3、4或5个R 3a所取代;
各R 4独立地为H、氘、卤素、羟基、氨基、C 1-6烷基、C 2-6烯基、C 2-6炔基、氘代C 1-6烷基、卤代C 1-6烷氧基、-(CR 7aR 7b) r-OR 8、-(CR 7aR 7b) r-NR 5aR 5b、-(CR 7aR 7b) r-CN、或卤代C 1-6烷基;或者,任意两个R 4和与它们相连的原子共同形成C 3-8环烷基、C 6-10芳基、3-8个原子组成的杂环基或5-6个原子组成的杂芳基;
各R 9、R 1a、R 2a和R 3a独立地为氧代(=O)、氢、氘、卤素、-(CR 7aR 7b) rCN、硝基、C 1-6烷基、卤代C 1-6烷基、C 2-6烯基、C 2-6炔基、R 8O-C 2-6亚烯基、R 8OC(=O)-C 2-6亚烯基、R 8O-C 2-6亚炔基、R 8OC(=O)-C 2-6亚炔基、-(CR 7aR 7b) r-OR 8、-(CR 7aR 7b) r-NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=O)R 6、-(CR 7aR 7b) rOC(=O)R 6、-(CR 7aR 7b) rCOOH、-(CR 7aR 7b) rC(=O)OC 1-6烷基、-(CR 7aR 7b) rC(=O)C 1-6烷基、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-12环烷基、-(CR 7aR 7b) r-C 6-12芳基、-(CR 7aR 7b) r-(3-12个原子组成的杂环基)或-(CR 7aR 7b) r-(5-12个原子组成的杂芳基);
或者,任意两个R 9与和它们相连的原子共同形成C 3-6环烷基或3-6个原子组成的杂环基;
其中,各R 9、R 1a、R 2a和R 3a独立任选地被1、2、3、4或5个R 10所取代;
各R 5、R 5a、R 5b、R 5c和R 5d独立地为氢、氰基、羟基、C 1-6烷基、C 1-6烷氧基-C(=O)-、C 6-10芳基、C 3-10环烷基、3-10个原子组成的杂环基或5-10个原子组成的杂芳基;
或者,R 5、R 9和与它们相连的原子共同形成3-8个原子组成的杂环基;
各R 6和R 8独立地为氢、氘、C 1-6烷基、氘代C 1-6烷基、C 2-6烯基、C 2-6炔基、羟基取代的C 1-6烷基、氨基取代的C 1-6烷基、氰基取代的C 1-6烷基、C 1-6烷氧基C 1-6烷基、卤代C 1-6烷基、C 6-10芳基、C 3-10环烷基、3-10个原子组成的杂环基或5-10个原子组成的杂芳基;
各R 7、R 7a和R 7b独立地为氢、C 1-6烷基、氘代C 1-6烷基、C 2-6烯基、C 2-6炔基或卤代C 1-6烷基;
各R 10独立地为氧代(=O)、H、D、卤素、CN、氨基、羟基、硝基、羧基、C 1-4烷基、C 2-4烯基、C 2-4炔基、氘代C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4烷氧基C 1-4烷基、羟基取代的C 1-4烷基、氰基取代的C 1-4烷基、氨基取代的C 1-4烷基、卤代C 1-4烷基、C 1-4烷基酰基、C 1-4烷基磺酰基、C 1-4烷氧基酰基、C 1-4烷氨基酰基、氨基酰基、氨基磺酰基、C 1-4烷氧基磺酰基、C 1-4烷氨基磺酰基、C 1-4酰氧基、C 6-10芳基、C 3-6环烷基、3-6个原子组成的杂环基或5-10个原子组成的杂芳基;
各m、n和t独立地为0、1、2、3或4;
p为1、2、3或4;
各r独立地为0、1、2、3或4。
在一些实施方案中,环C为5-7个原子组成的杂环基或5-6个原子组成的杂芳基。
在另一些实施方案中,环C为以下子结构式:
Figure PCTCN2018072988-appb-000002
其中,环C通过N原子与
Figure PCTCN2018072988-appb-000003
相连;
Figure PCTCN2018072988-appb-000004
Figure PCTCN2018072988-appb-000005
独立地为代表单键或双键;
Figure PCTCN2018072988-appb-000006
Figure PCTCN2018072988-appb-000007
为单键时,各Z、Z 1和Z 2独立地为CH 2或NH;
Figure PCTCN2018072988-appb-000008
Figure PCTCN2018072988-appb-000009
为双键时,各Z、Z 1和Z 2独立地为CH或N;
Z 4为CH 2或NH;
各Z 3和Z 5独立地为CH 2、NH、S或O;
q为0、1或2。
在又一些实施方案中,环C为
Figure PCTCN2018072988-appb-000010
Figure PCTCN2018072988-appb-000011
其中,环C通过N原子与
Figure PCTCN2018072988-appb-000012
相连。
在一些实施方案中,本发明式(I)化合物为如式(II)所示的化合物或式(II)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
Figure PCTCN2018072988-appb-000013
其中,各
Figure PCTCN2018072988-appb-000014
Figure PCTCN2018072988-appb-000015
独立地为单键或双键;
Figure PCTCN2018072988-appb-000016
Figure PCTCN2018072988-appb-000017
为单键时,各Z和Z 1独立地为CH 2或NH;当
Figure PCTCN2018072988-appb-000018
Figure PCTCN2018072988-appb-000019
为双键时,各Z和Z 1独立地为CH或N;
环A、环B、R 1、R 2、R 3、R 4、X、Y、n、m、p和t均具有本发明所描述的含义。
在一些实施方案中,各R 3独立地为C 1-4烷基、C 3-8碳环基、C 6-10芳基、3-8个原子组成的杂环基或5-10个原子组成的杂芳基;
其中,各R 3独立任选地被1、2、3、4或5个R 3a所取代。
在另一些实施方案中,各R 3独立地为甲基、乙基、苯基、吡咯基、吡唑基、咪唑基、咪唑啉基、***基、吡啶基、嘧啶基、吲哚基、吲唑基、苯并呋喃基、苯并噻吩基、苯并噻唑基、苯并咪唑基、苯并异噁唑基、苯并异噻唑基、喹啉基、异喹啉基或喹唑啉基;
其中,各R 3独立任选地被1、2、3、4或5个R 3a所取代。
在一些实施方案中,本发明式(I)化合物为如式(III)所示的化合物或式(III)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
Figure PCTCN2018072988-appb-000020
其中,各
Figure PCTCN2018072988-appb-000021
Figure PCTCN2018072988-appb-000022
独立地为单键或双键;
Figure PCTCN2018072988-appb-000023
为单键时,Z为CH 2或NH;当
Figure PCTCN2018072988-appb-000025
Figure PCTCN2018072988-appb-000026
为双键时,Z为CH或N;
f为0、1、2、3、4或5;
g为0、1、2、3、4、5、6、7或8;
环A、环B、R 1、R 2、R 3a、R 4、R 5、R 9、n、m和t均具有本发明所描述的含义。
在一些实施方案中,环A为C 7-12双环碳环基、C 8-12双环芳基、7-12个原子组成的双环杂环基或7-12个原子组成的双环杂芳基。
在一些实施方案中,环A为以下子结构式:
Figure PCTCN2018072988-appb-000027
其中,各T 1、T 2、T 3、T 4、T 5、T 6、T 7和T 8独立地为-CH-或-N-;
各V 1、V 2、V 3和V 4独立地为-CH 2-、-NH-、-O-、-S-、-S(=O)-、-S(=O) 2-或-C(=O)-;
各k和j独立地为0、1、2、3或4。
在一些实施方案中,环A为
Figure PCTCN2018072988-appb-000028
Figure PCTCN2018072988-appb-000029
Figure PCTCN2018072988-appb-000030
在一些实施方案中,环B为C 5-8碳环基、C 6-10芳基、5-8个原子组成的杂环基或5-10个原子组成的杂芳基;
各R 2独立地为H、氘、F、Cl、Br、I、-(CR 7aR 7b) rCN、C 1-4烷基、氘代C 1-4烷基、卤代C 1-4烷基、C 2-4烯基、C 2-4炔基、R 8O-C 2-4亚烯基、R 8OC(=O)-C 2-4亚烯基、R 8O-C 2-4亚炔基、R 8OC(=O)-C 2-4亚炔基、-N=C=S、-N=C=O、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=G)R 6、-(CR 7aR 7b) rN(R 5c)C(=G)R 6、-(CR 7aR 7b) rOC(=G)R 6、-(CR 7aR 7b) rC(=G)OR 8、-(CR 7aR 7b) rN(R 5c)C(=G)OR 8、-(CR 7aR 7b) rOC(=G)OR 8、-(CR 7aR 7b) rC(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)=C(R 7)NR 5aR 5b、-(CR 7aR 7b) rS(=O)R 6、-(CR 7aR 7b) rN(R 5c)S(=O)R 6、-(CR 7aR 7b) rS(=O)OR 8、-(CR 7aR 7b) rN(R 5c)S(=O)OR 8、-(CR 7aR 7b) rS(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rN(R 5c)S(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-8环烷基、-(CR 7aR 7b) r-C 6-10芳基、-(CR 7aR 7b) r-(3-8个原子组成的杂环基)、-(CR 7aR 7b) r-(5-10个原子组成的杂芳基)或D-甘露糖基;其中,各G独立地为O、S或NR 5d
各R 2独立任选地被1、2、3或4个R 2a所取代;
其中,R 2a、R 5a、R 5b、R 5c、R 5d、R 6、R 7、R 7a、R 7b、R 8和r均具有本发明所描述的含义。
在另一些实施方案中,环B为以下子结构式:
Figure PCTCN2018072988-appb-000031
Figure PCTCN2018072988-appb-000032
其中,各Q 1、Q 6和Q 9独立地为CH 2、NH、O、S、C(=O)、S(=O)或S(=O) 2
各Q 2、Q 3、Q 4、Q 5、Q 7和Q 8独立地为CH或N;
s为0、1、2或3;
各R 2独立地为H、氘、F、Cl、Br、I、-(CR 7aR 7b) rCN、C 1-3烷基、氘代C 1-3烷基、卤代C 1-3烷基、C 2-4烯基、R 8O-C 2-6亚烯基、R 8OC(=O)-C 2-6亚烯基、R 8O-C 2-6亚炔基、R 8OC(=O)-C 2-6亚炔基、-N=C=S、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=O)R 6、-(CR 7aR 7b) rOC(=G)R 6、-(CR 7aR 7b) rN(R 5c)C(=O)R 6、-(CR 7aR 7b) rC(=O)OR 8、-(CR 7aR 7b) rN(R 5c)C(=O)OR 8、-(CR 7aR 7b) rC(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=S)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=N-R 5d)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rN(R 5c)S(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-6环烷基、-(CR 7aR 7b) r-C 6-10芳基、-(CR 7aR 7b) r-(3-6个原子组成的杂环基)或-(CR 7aR 7b) r-(5-10个原子组成的杂芳基);其中,各R 2独立任选地被1、2、3或4个R 2a所取代;
R 2a、R 5a、R 5b、R 5c、R 5d、R 6、R 7、R 7a、R 7b、R 8和r均具有本发明所描述的含义。
在又一些实施方案中,环B为
Figure PCTCN2018072988-appb-000033
Figure PCTCN2018072988-appb-000034
Figure PCTCN2018072988-appb-000035
各R 2独立地为H、氘、F、Cl、Br、I、CN、-CH 2-CN、-CH 2CH 2-CN、甲基、乙基、丙基、丁基、氘代甲基、二氘代甲基、一氘代甲基、三氟甲基、二氟甲基、2,2-二氟乙基、1,2-二氟乙基、2,2,2-三氟乙基、乙烯基、丙烯基、烯丙基、HO-乙烯基、MeO-乙烯基、HOC(=O)-乙烯基、MeOC(=O)-乙烯基、-N=C=S、-OH、-OMe、-O(i-Pr)、-O(t-Bu)、-CH 2-OH、-CH 2CH 2-OH、-CH 2-OMe、-CH 2CH 2-OMe、-CH 2-O(i-Pr)、-CH 2-O(t-Bu)、苯基-O-、吡啶基-O-、嘧啶基-O-、吡嗪基-O-、哒嗪基-O-、吡咯基-O-、吡唑基-O-、噻唑基-O-、噁唑基-O-、噁二唑基-O-、咪唑基-O-、噻二唑基-O-、-NH 2、-NHMe、-N(Me) 2、-CH 2-NH 2、-CH 2CH 2-NH 2、-CH 2-NHMe、-CH 2-N(Me) 2、苯基-NH-、吡啶基-NH-、嘧啶基-NH-、吡嗪基-NH-、哒嗪基-NH-、吡咯基-NH-、吡唑基-NH-、噻唑基-NH-、噁唑基-NH-、噁二唑基-NH-、咪唑基-NH-、噻二唑基-NH-、-C(=O)Me、-C(=O)Et、-CH 2-C(=O)Me、-CH 2-C(=O)Et、-NHC(=O)Me、-C(=O)OH、-C(=O)OMe、-NHC(=O)OMe、-NHC(=O)OH、-NHC(=O)OEt、-NHC(=O)O(t-Bu)、-C(=O)NH 2、-C(=O)NHMe、-NHC(=O)NH 2、-NHC(=S)NH 2、-NHC(=NH)NH 2、-S(=O) 2Me、-NHS(=O) 2Me、-S(=O) 2OH、-S(=O) 2OMe、-NHS(=O) 2OH、-NHS(=O) 2OMe、-S(=O) 2NH 2、-NHS(=O) 2NH 2、环丙基、环丙基甲基、环丁基、环戊基、环己基、四氢吡喃基、二氧六环基、哌啶基、哌嗪基、吗啉基、四氢呋喃基、四氢吡咯基、环氧丙基、环氧丁基、氮杂环丁基、二氢吡啶基、二氢吡喃基、苯基、苄基、吡啶基、嘧啶基、吡嗪基、哒嗪基、吡咯基、吡唑基、噻唑基、噁唑基、噁二唑基、咪唑基、噻二唑基、***基或四唑基;
其中,各R 2独立任选地被1、2、3或4个R 2a所取代;R 2a具有本发明所描述的含义。
在一些实施方案中,各R 1独立地为氧代(=O)、H、氘、F、Cl、Br、I、-CN、甲基、乙基、丙基、丁基、三氟甲基、-OH、-OMe、-CH 2OMe、-CH 2OH、-NH 2、-NHMe、-N(Me) 2、-C(=O)Me、-C(=O)OH、-C(=O)OMe、-C(=O)NH 2、-S(=O) 2Me、-S(=O) 2OH、-S(=O) 2OMe、-S(=O) 2NH 2、环丙基、环丙基甲基、环丁基、环戊基、环丁基、苯基、吡啶基、嘧啶基、吡嗪基、哒嗪基、吡咯基、咪唑基、吡唑基、***基、四唑基、环氧烷 基、环氧丁基、氮杂环丁基、四氢呋喃基、哌啶基、吡咯烷基、哌嗪基或吗啉基;其中,各R 1独立任选地被1、2、3或4个R 1a所取代;
各R 4独立地为H、氘、F、Cl、Br、I、羟基、氨基、甲基、乙基、丙基、丁基、三氟甲基、二氟甲基、1,2-二氟乙基、三氟甲氧基、二氟甲氧基、-OMe、-OEt、-O(t-Bu)、-CH 2OH、-CH 2CH 2OH、-CN、-CH 2CN、-CH 2NH 2、-CH 2CH 2NH 2、-CH 2OMe、-CH 2CH 2OMe、-NHMe或-N(Me) 2;或者,任意两个R 4和与它们相连的原子共同形成C 3-6环烷基或3-6个原子组成的杂环基;
各R 9、R 1a、R 2a和R 3a独立地为氧代(=O)、氢、氘、F、Cl、Br、I、-(CR 7aR 7b) rCN、硝基、C 1-4烷基、卤代C 1-4烷基、C 2-4烯基、C 2-4炔基、R 8O-C 2-4亚烯基、R 8OC(=O)-C 2-4亚烯基、R 8O-C 2-4亚炔基、R 8OC(=O)-C 2-4亚炔基、-(CR 7aR 7b) r-OR 8、-(CR 7aR 7b) r-NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=O)R 6、-(CR 7aR 7b) rOC(=O)R 6、-(CR 7aR 7b) rCOOH、-(CR 7aR 7b) rC(=O)OC 1-4烷基、-(CR 7aR 7b) rC(=O)C 1-4烷基、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-6环烷基、-(CR 7aR 7b) r-C 6-10芳基、-(CR 7aR 7b) r-(3-6个原子组成的杂环基)或-(CR 7aR 7b) r-(5-6个原子组成的杂芳基);
或者,任意两个R 9与和它们相连的原子共同形成C 3-6环烷基或3-6个原子组成的杂环基;
其中,各R 9、R 1a、R 2a和R 3a独立任选地被1、2、3、4或5个R 10所取代;
R 5a、R 5b、R 5c、R 6、R 7a、R 7b、R 8、R 10和r均具有本发明所描述的含义。
在另一些实施方案中,各R 1独立地为氧代(=O)、H、氘、F、Cl、Br、I、氰基、甲基、乙基、丙基、丁基、-OH、-OMe、-CH 2OMe、-CH 2OH、-NH 2、-NHMe、-N(Me) 2、-C(=O)Me、-C(=O)OH、-C(=O)OMe、-C(=O)NH 2、-S(=O) 2Me、-S(=O) 2OH、-S(=O) 2OMe或-S(=O) 2NH 2;其中,各R 1独立任选地被1、2、3或4个R 1a所取代;
各R 4独立地为H、氘、F、Cl、Br、I、羟基、氨基、甲基、乙基、丙基、丁基、乙烯基、丙烯基、烯丙基、氘代甲基、三氟甲基、二氟甲基、1,2-二氟乙基、-OMe、-OEt、-CH 2OH、-CH 2CH 2OH、-CN、-CH 2CN、-CH 2NH 2、-CH 2CH 2NH 2、-CH 2OMe、-CH 2CH 2OMe、-NHMe或-N(Me) 2;或者,任意两个R 4和与它们相连的原子共同形成环丙基、环丁基、环氧烷基或氮杂环丁基;
各R 9、R 1a、R 2a和R 3a独立地为氧代(=O)、氢、氘、F、Cl、Br、I、-CN、甲基、乙基、丙基、丁基、三氟甲基、二氟甲基、2,2-二氟乙基、-CH 2CN、-CH 2CH 2CN、乙烯基、丙烯基、烯丙基、乙炔基、丙炔基、-OH、-OMe、-O(i-Pr)、-O(t-Bu)、-CH 2-OH、-CH 2CH 2-OH、-CH 2-OMe、-CH 2CH 2-OMe、-CH 2-O(i-Pr)、-CH 2-O(t-Bu)、-NH 2、-NHMe、-N(Me) 2、-CH 2-NH 2、-CH 2CH 2-NH 2、-CH 2-NHMe、-CH 2-N(Me) 2、-NHC(=O)Me、-CH 2COOH、-COOH、-C(=O)OMe、-CH 2C(=O)OMe、-C(=O)Me、-CH 2C(=O)Me、环丙基、环丁基、环戊基、环己基、环氧烷基、环氧丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基、四氢吡喃基、二氢吡喃基、苯基、苄基、吡啶基、嘧啶基、吡嗪基、呋喃基、噻吩基、吡咯基、吡唑基、咪唑基、噁唑基、噻唑基、噁二唑基、噻二唑基、***基或四唑基;
或者,任意两个R 9与和它们相连的原子共同形成环丙基、环丁基、环戊基、环己基、环氧烷基、环氧丁基、氮杂环丁基、四氢呋喃基、吡咯烷基、哌啶基或哌嗪基;
其中,各R 9、R 1a、R 2a和R 3a独立任选地被1、2、3、4或5个R 10所取代;
R 10均具有本发明所描述的含义。
在一些实施方案中,其中,各R 5、R 5a、R 5b、R 5c和R 5d独立地为氢、氰基、羟基、C 1-4烷基、C 1-4烷氧基-C(=O)-、C 6-10芳基、C 3-6环烷基、3-6个原子组成的杂环基或5-6个原子组成的杂芳基;
各R 6和R 8独立地为氢、氘、C 1-4烷基、氘代C 1-4烷基、C 2-4烯基、C 2-4炔基、羟基取代的C 1-4烷基、氨基取代的C 1-4烷基、氰基取代的C 1-4烷基、C 1-4烷氧基C 1-4烷基、卤代C 1-4烷基、C 6-10芳基、C 3-6环烷基、 3-6个原子组成的杂环基或5-6个原子组成的杂芳基;
各R 7、R 7a和R 7b独立地为氢、C 1-4烷基、氘代C 1-4烷基或卤代C 1-4烷基;
各R 10独立地为氧代(=O)、H、D、F、Cl、Br、I、CN、氨基、羟基、硝基、羧基、C 1-3烷基、C 2-4烯基、C 2-4炔基、氘代C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4烷氧基C 1-4烷基、羟基取代的C 1-4烷基、氰基取代的C 1-4烷基、氨基取代的C 1-4烷基、卤代C 1-4烷基、C 1-4烷基酰基、C 1-4烷基磺酰基、C 1-4烷氧基酰基、C 1-4烷氨基酰基、苯基、C 3-6环烷基、3-6个原子组成的杂环基或5-6个原子组成的杂芳基。
在另一些实施方案中,各R 5、R 5a、R 5b、R 5c和R 5d独立地为氢、氰基、羟基、甲基、乙基、丙基、丁基、甲氧基-C(=O)-、乙氧基-C(=O)-、异丙氧基-C(=O)-、叔丁氧基-C(=O)-、苯基、环丙基、环丁基、环戊基、环己基、环氧烷基、环氧丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基、四氢吡喃基、二氢吡喃基、苯基、苄基、吡啶基、嘧啶基、吡嗪基、呋喃基、噻吩基、吡咯基、吡唑基、咪唑基、噁唑基、噻唑基、噁二唑基、噻二唑基、***基或四唑基;
各R 6和R 8独立地为氢、氘、甲基、乙基、丙基、丁基、氘代甲基、乙烯基、丙炔基、羟基甲基、羟基乙基、甲氧基甲基、甲氧基乙基、乙氧基甲基、叔丁氧基甲基、叔丁氧基乙基、乙氧基乙基、异丙氧基乙基、三氟甲基、二氟甲基、1,2-二氟乙基、2,2-二氟乙基、苯基、环丙基、环己基、氮杂环丁基、四氢呋喃基、吡咯烷基、哌啶基、哌嗪基、吡啶基、吡咯基、嘧啶基、吡唑基、吡嗪基、呋喃基、噻唑基、噁唑基、噁二唑基、噻二唑基、咪唑基、***基或四唑基;
各R 7、R 7a和R 7b独立地为氢、甲基、乙基、丙基、丁基、氘代甲基、三氟甲基、二氟甲基、1,2-二氟乙基或2,2-二氟乙基;
各R 10独立地为氧代(=O)、H、D、F、Cl、Br、I、CN、氨基、羟基、硝基、羧基、甲基、乙基、丙基、丁基、氘代甲基、甲氧基、甲氨基、二甲氨基、甲氧基甲基、甲氧基乙基、叔丁氧基甲基、叔丁氧基乙基、羟基甲基、羟基乙基、CN取代的甲基、CN取代的乙基、氨基甲基、氨基乙基、三氟甲基、二氟甲基、1,2-二氟乙基或2,2-二氟乙基、甲基酰基、乙基酰基、甲基磺酰基、甲氧基酰基、叔丁氧基酰基、甲氨基酰基、二甲氨基酰基、苯基、环丙基、环丁基、环戊基、环己基、环氧丙烷基、环氧丁基、氮杂环丁基、四氢呋喃基、吡咯烷基、哌啶基、哌嗪基、吗啉基、呋喃基、噻吩基、吡咯基、吡唑基、咪唑基、噁二唑基、***基、四唑基、吡啶基、嘧啶基或吡嗪基。
一方面,本发明涉及一种药物组合物,其包含本发明任一所述的化合物。在一些实施方案中,本发明所述的药物组合物进一步包含药学上可接受的载体、赋形剂、稀释剂、辅剂和媒介物的至少一种。
另一方面,本发明涉及所述的化合物或药物组合物在制备药物中的用途,其中,所述的药物用于预防、治疗或减轻血栓栓塞性疾病。
在一些实施方案中,本发明所述的用途,其中所述血栓栓塞性疾病为动脉心血管血栓栓塞性疾病、静脉心血管血栓栓塞性疾病、及心室或外周循环中的血栓栓塞性疾病。
在另一些实施方案中,本发明所述的用途,其中所述血栓栓塞性疾病为心绞痛、急性冠状动脉综合征、心房颤动、心肌梗塞、短暂性缺血发作、中风、动脉粥样硬化、外周闭塞性动脉疾病、静脉血栓形成、深静脉血栓形成、血栓性静脉炎、动脉栓塞、冠状动脉血栓形成、脑动脉血栓形成、脑栓塞、肾栓塞、肺栓塞、以及因医疗植入物、器械或程序中的血液暴露于人造表面从而促使血栓形成而引起的血栓形成。
一方面,本发明涉及所述的化合物或药物组合物在制备药物中的用途,其中,所述的药物用于治疗弥散性血管内凝血(DIC)疾病。
另一方面,本发明涉及所述的化合物或药物组合物在制备药物中的用途,其中,所述的药物用于抑制因子XIa和/或血浆激肽释放酶的活性。
一方面,本发明所述的化合物或药物组合物用于预防、治疗或减轻血栓栓塞性疾病。
在一些实施方案中,本发明所述的化合物或药物组合物用于预防、治疗或减轻血栓栓塞性疾病,其中,所述血栓栓塞性疾病为动脉心血管血栓栓塞性疾病、静脉心血管血栓栓塞性疾病、及心室或外周循环中的血栓栓塞性疾病。
在另一些实施方案中,本发明所述的化合物或药物组合物用于预防、治疗或减轻血栓栓塞性疾病,其中,所述血栓栓塞性疾病为心绞痛、急性冠状动脉综合征、心房颤动、心肌梗塞、短暂性缺血发作、中风、动脉粥样硬化、外周闭塞性动脉疾病、静脉血栓形成、深静脉血栓形成、血栓性静脉炎、动脉栓塞、冠状动脉血栓形成、脑动脉血栓形成、脑栓塞、肾栓塞、肺栓塞、以及因医疗植入物、器械或程序中的血液暴露于人造表面从而促使血栓形成而引起的血栓形成。
另一方面,本发明所述的化合物或药物组合物用于抑制因子XIa和/或血浆激肽释放酶的活性。
一方面,本发明还涉及一种使用本发明所述的化合物或药物组合物预防、治疗或减轻血栓栓塞性疾病的方法。
在一些实施方案中,本发明所述的方法,其中,所述血栓栓塞性疾病为动脉心血管血栓栓塞性疾病、静脉心血管血栓栓塞性疾病、及心室或外周循环中的血栓栓塞性疾病。
在另一些实施方案中,本发明所述的方法,其中,所述血栓栓塞性疾病为心绞痛、急性冠状动脉综合征、心房颤动、心肌梗塞、短暂性缺血发作、中风、动脉粥样硬化、外周闭塞性动脉疾病、静脉血栓形成、深静脉血栓形成、血栓性静脉炎、动脉栓塞、冠状动脉血栓形成、脑动脉血栓形成、脑栓塞、肾栓塞、肺栓塞、以及因医疗植入物、器械或程序中的血液暴露于人造表面从而促使血栓形成而引起的血栓形成。
另一方面,本发明还涉及一种使用本发明所述的化合物或药物组合物抑制因子XIa和/或血浆激肽释放酶的活性的方法。
前面所述内容只概述了本发明的某些方面,但并不限于这些方面。这些方面及其他的方面的内容将在下面作更加具体完整的描述。
详细说明书
定义和一般术语
现在详细描述本发明的某些实施方案,其实例由随附的结构式和化学式说明。本发明意图涵盖所有的替代、修改和等同技术方案,它们均包括在如权利要求定义的本发明范围内。本领域技术人员应认识到,许多与本文所述类似或等同的方法和材料能够用于实践本发明。本发明绝不限于本文所述的方法和材料。在所结合的文献、专利和类似材料的一篇或多篇与本申请不同或相矛盾的情况下(包括但不限于所定义的术语、术语应用、所描述的技术,等等),以本申请为准。
应进一步认识到,本发明的某些特征,为清楚可见,在多个独立的实施方案中进行了描述,但也可以在单个实施例中以组合形式提供。反之,本发明的各种特征,为简洁起见,在单个实施方案中进行了描述,但也可以单独或以任意适合的子组合提供。
除非另外说明,本发明所使用的所有科技术语具有与本发明所属领域技术人员的通常理解相同的含义。本发明涉及的所有专利和公开出版物通过引用方式整体并入本发明。
除非另外说明,应当应用本文所使用的下列定义。出于本发明的目的,化学元素与元素周期表CAS版,和《化学和物理手册》,第75版,1994一致。此外,有机化学一般原理可参考"Organic Chemistry",Thomas Sorrell,University Science Books,Sausalito:1999,和"March's Advanced Organic Chemistry”by Michael B.Smith and Jerry March,John Wiley&Sons,New York:2007中的描述,其全部内容通过引用并入本文。
除非另有说明或者上下文中有明显的冲突,本文所使用的冠词“一”、“一个(种)”和“所述”旨在包括“至少一个”或“一个或多个”。因此,本文所使用的这些冠词是指一个或多于一个(即至少一个)宾语的冠词。例 如,“一组分”指一个或多个组分,即可能有多于一个的组分被考虑在所述实施方案的实施方式中采用或使用。
本发明所使用的术语“受试对象”是指动物。典型地所述动物是哺乳动物。受试对象,例如也指灵长类动物(例如人类,男性或女性)、牛、绵羊、山羊、马、犬、猫、兔、大鼠、小鼠、鱼、鸟等。在某些实施方案中,所述受试对象是灵长类动物。在其他实施方案中,所述受试对象是人。
本发明所使用的术语“患者”是指人(包括成人和儿童)或者其他动物。在一些实施方案中,“患者”是指人。
术语“包含”为开放式表达,即包括本发明所指明的内容,但并不排除其他方面的内容。
“立体异构体”是指具有相同化学构造,但原子或基团在空间上排列方式不同的化合物。立体异构体包括对映异构体、非对映异构体、构象异构体(旋转异构体)、几何异构体(顺/反)异构体、阻转异构体,等等。
“手性”是具有与其镜像不能重叠性质的分子;而“非手性”是指与其镜像可以重叠的分子。
“对映异构体”是指一个化合物的两个不能重叠但互成镜像关系的异构体。
“非对映异构体”是指有两个或多个手性中心并且其分子不互为镜像的立体异构体。非对映异构体具有不同的物理性质,如熔点、沸点、光谱性质和反应性。非对映异构体混合物可通过高分辨分析操作如电泳和色谱,例如HPLC来分离。
本发明所使用的立体化学定义和规则一般遵循S.P.Parker,Ed.,McGraw-Hill Dictionary of Chemical Terms(1984)McGraw-Hill Book Company,New York;and Eliel,E.and Wilen,S.,“Stereochemistry of Organic Compounds”,John Wiley&Sons,Inc.,New York,1994。
许多有机化合物以光学活性形式存在,即它们具有使平面偏振光的平面发生旋转的能力。在描述光学活性化合物时,使用前缀D和L或R和S来表示分子关于其一个或多个手性中心的绝对构型。前缀d和l或(+)和(-)是用于指定化合物所致平面偏振光旋转的符号,其中(-)或l表示化合物是左旋的。前缀为(+)或d的化合物是右旋的。一种具体的立体异构体是对映异构体,这种异构体的混合物称作对映异构体混合物。对映异构体的50:50混合物称为外消旋混合物或外消旋体,当在化学反应或过程中没有立体选择性或立体特异性时,可出现这种情况。
本发明公开化合物的任何不对称原子(例如,碳等)都可以以外消旋或对映体富集的形式存在,例如(R)-、(S)-或(R,S)-构型形式存在。在某些实施方案中,各不对称原子在(R)-或(S)-构型方面具有至少50%对映体过量,至少60%对映体过量,至少70%对映体过量,至少80%对映体过量,至少90%对映体过量,至少95%对映体过量,或至少99%对映体过量。
依据起始物料和方法的选择,本发明化合物可以以可能的异构体中的一个或它们的混合物,例如外消旋体和非对应异构体混合物(这取决于不对称碳原子的数量)的形式存在。光学活性的(R)-或(S)-异构体可使用手性合成子或手性试剂制备,或使用常规技术拆分。如果化合物含有一个双键,取代基可能为E或Z构型;如果化合物中含有二取代的环烷基,环烷基的取代基可能有顺式或反式构型。
所得的任何立体异构体的混合物可以依据组分物理化学性质上的差异被分离成纯的或基本纯的几何异构体,对映异构体,非对映异构体,例如,通过色谱法和/或分步结晶法。
可以用已知的方法将任何所得终产物或中间体的外消旋体通过本领域技术人员熟悉的方法拆分成光学对映体,如,通过对获得的其非对映异构的盐进行分离。外消旋的产物也可以通过手性色谱来分离,如,使用手性吸附剂的高效液相色谱(HPLC)。特别地,对映异构体可以通过不对称合成制备,例如,可参考Jacques,et al.,Enantiomers,Racemates and Resolutions(Wiley Interscience,New York,1981);Principles of Asymmetric Synthesis(2 nd Ed.Robert E.Gawley,Jeffrey Aubé,Elsevier,Oxford,UK,2012);Eliel,E.L.Stereochemistry of Carbon Compounds(McGraw-Hill,NY,1962);Wilen,S.H.Tables of Resolving Agents  and Optical Resolutions p.268(E.L.Eliel,Ed.,Univ.of Notre Dame Press,Notre Dame,IN 1972);Chiral Separation Techniques:A Practical Approach(Subramanian,G.Ed.,Wiley-VCH Verlag GmbH&Co.KGaA,Weinheim,Germany,2007)。
术语“互变异构体”或“互变异构形式”是指具有不同能量的可通过低能垒(low energy barrier)互相转化的结构异构体。若互变异构是可能的(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(protontautomer)(也称为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。价键互变异构体(valence tautomer)包括通过一些成键电子的重组来进行的互相转化。酮-烯醇互变异构的具体实例是戊烷-2,4-二酮和4-羟基戊-3-烯-2-酮互变异构体的互变。互变异构的另一个实例是酚-酮互变异构。酚-酮互变异构的一个具体实例是吡啶-4-醇和吡啶-4(1H)-酮互变异构体的互变。除非另外指出,本发明化合物的所有互变异构体形式都在本发明的范围之内。
术语“任选”或“任选地”是指随后描述的事件或情形可以但不一定出现,即,该描述包括其中所述事件或情形出现的情况以及不出现的情况。例如,“任选地被1、2、3或4个…所取代”包括该基团被1个、或2个、或3个、或4个所述的取代基所取代的情况,以及该基团不被所述取代基取代的情况。进一步地,当该基团被1个以上所述取代基取代时,所述取代基之间是相互独立,即,所述的1个以上的取代基可以是互不相同的,也可以是相同的。
像本发明所描述的,本发明的化合物可以任选地被一个或多个取代基所取代,如上面的通式化合物,或者像实施例里面特殊的例子,子类,和本发明所包含的一类化合物。应了解“任选取代的”这个术语与“取代或非取代的”这个术语可以交换使用。一般而言,术语“取代的”表示所给结构中的一个或多个氢原子被具体取代基所取代。除非其他方面表明,一个任选的取代基团可以在基团各个可取代的位置进行取代。当所给出的结构式中不只一个位置能被选自具体基团的一个或多个取代基所取代,那么取代基可以相同或不同地在各个位置取代。其中,所述的“一个或多个取代基”表示一个或一个以上的取代基,具体取代基的个数由被取代基团可被取代的位置的个数所决定。例如,本发明中的描述“C 4亚烷基独立任选地被一个或多个相同或不同的R 9所取代”表示所述亚烷基可以被一个或一个以上的R 9所取代,具体的说,所述的亚烷基可以独立任选地被1、2、3、4、5、6、7或8个R 9所取代;当所述的亚烷基被一个以上的R 9所取代时,所述R 9可以是相同的,也可以是不同的。
本发明中,所述的取代基可以是,但并不限于,氢、氘、氧代(=O)、卤素、氰基、硝基、羟基、巯基、氨基、芳氨基、氨基烷基、烷基、烷基硫基、羟基烷基、卤代烷基、碳环基、杂环基、芳基、杂芳基、-C(=O)R、-OR a、-COOR a、-SO 2R a、-NR bR c、-CONR bR c、-SO 2NR bR c、-C(NR bR c)=NR d或=NR d;其中,R、R a、R b、R c和R d各自独立的为氢、氰基、氨基、烷氨基、芳氨基、烷基硫基、烷氧基、芳氧基、羟基、巯基、烷基、卤代烷基、碳环基、杂环基、芳基、杂芳基、烷基磺酰基、氨基磺酰基、羟基烷基、氨基烷基、烷基酰基或氨基酰基。其中,所述取代基中的各烷氨基、芳氨基、烷氧基、芳氧基、羟基、巯基、烷基、卤代烷基、碳环基、杂环基、芳基、杂芳基、烷基磺酰基、氨基磺酰基、羟基烷基、氨基烷基、烷基酰基、氨基酰基和烷基硫基具有本发明所描述的含义,且均可进一步被本发明所描述的取代基单取代或相同或不同的多取代。
另外,需要说明的是,除非以其他方式明确指出,在本发明中所采用的描述方式“各…独立地为”与“…各自独立地为”和“…独立地为”可以互换,均应做广义理解,其既可以是指在不同基团中,相同符号之间所表达的具体选项之间互相不影响,也可以表示在相同的基团中,相同符号之间所表达的具体选项之间互相不影响。例如,描述“各R 9、R 1a、R 2a和R 3a独立地为...”表示所述的R 9、R 1a、R 2a和R 3a之间是各自独立,其具体选择的基团是互不影响的,比如,“各R 9、R 1a、R 2a和R 3a独立地为氢、氘、硝基或C 1-6烷基”表示R 9可以为氢、氘、硝基或C 1-6烷基,R 1a也可以为氢、氘、硝基或C 1-6烷基,同样的,R 2a和R 3a也均可 以是氢、氘、硝基或C 1-6烷基。
在本说明书的各部分,本发明公开化合物的取代基按照基团种类或范围公开。特别指出,本发明包括这些基团种类和范围的各个成员的每一个独立的次级组合。例如,术语“C 1- 6烷基”特别指独立公开的甲基、乙基、C 3烷基、C 4烷基、C 5烷基和C 6烷基。“C 1-4烷基”特指独立公开的甲基、乙基、C 3烷基(即丙基,包括正丙基和异丙基)、C 4烷基(即丁基,包括正丁基、异丁基、仲丁基和叔丁基)。
在本发明的各部分,描述了连接取代基。当该结构清楚地需要连接基团时,针对该基团所列举的马库什变量应理解为连接基团。例如,如果该结构需要连接基团并且针对该变量的马库什基团定义列举了“烷基”或“芳基”,则应该理解,该“烷基”或“芳基”分别代表连接的亚烷基基团或亚芳基基团。例如,本发明式(I)所示结构中的环A可以为杂芳基,则此处的杂芳基代表有两个连接位点与分子其余部分相连的亚芳基,更具体的,例如,环A为喹啉基,则此处的喹啉基代表亚喹啉基。又如,“C 1-6烷氧基C 1-6烷基”中的C 1-6烷基代表C 1-6亚烷基,包括但不限于,亚甲基、亚乙基等。
本发明使用的术语“烷基”或“烷基基团”,表示含有1至20个碳原子的,饱和的直链或支链一价烃基基团,其中,所述烷基基团可以任选地被一个或多个本发明所描述的取代基所取代。除非另外详细说明,烷基基团含有1-20个碳原子。在一些实施方案中,烷基基团含有1-12个碳原子;在另一些实施方案中,烷基基团含有1-6个碳原子;在又一些实施方案中,烷基基团含有1-4个碳原子;在另一些实施方案中,烷基基团含有1-3个碳原子。术语“C 1- 6烷基”表示包含1-6个碳原子的烷基基团。
烷基基团的实例包含,但并不限于,甲基(Me、-CH 3),乙基(Et、-CH 2CH 3),正丙基(n-Pr、-CH 2CH 2CH 3),异丙基(i-Pr、-CH(CH 3) 2),正丁基(n-Bu、-CH 2CH 2CH 2CH 3),异丁基(i-Bu、-CH 2CH(CH 3) 2),仲丁基(s-Bu、-CH(CH 3)CH 2CH 3),叔丁基(t-Bu、-C(CH 3) 3),正戊基(-CH 2CH 2CH 2CH 2CH 3),2-戊基(-CH(CH 3)CH 2CH 2CH 3),3-戊基(-CH(CH 2CH 3) 2),2-甲基-2-丁基(-C(CH 3) 2CH 2CH 3),3-甲基-2-丁基(-CH(CH 3)CH(CH 3) 2),3-甲基-1-丁基(-CH 2CH 2CH(CH 3) 2),2-甲基-1-丁基(-CH 2CH(CH 3)CH 2CH 3),正己基(-CH 2CH 2CH 2CH 2CH 2CH 3),2-己基(-CH(CH 3)CH 2CH 2CH 2CH 3),3-己基(-CH(CH 2CH 3)(CH 2CH 2CH 3)),2-甲基-2-戊基(-C(CH 3) 2CH 2CH 2CH 3),3-甲基-2-戊基(-CH(CH 3)CH(CH 3)CH 2CH 3),4-甲基-2-戊基(-CH(CH 3)CH 2CH(CH 3) 2),3-甲基-3-戊基(-C(CH 3)(CH 2CH 3) 2),2-甲基-3-戊基(-CH(CH 2CH 3)CH(CH 3) 2),2,3-二甲基-2-丁基(-C(CH 3) 2CH(CH 3) 2),3,3-二甲基-2-丁基(-CH(CH 3)C(CH 3) 3),正庚基,正辛基,等等。
术语“烯基”表示含有2-12个碳原子的直链或支链一价烃基,其中至少有一个碳-碳sp 2双键,其中,所述烯基基团可以任选地被一个或多个本发明所描述的取代基所取代,其包括“cis”和“tans”的定位,或者"E"和"Z"的定位。在一些实施方案中,烯基基团包含2-8个碳原子;在另一些实施方案中,烯基基团包含2-6个碳原子;在又一些实施方案中,烯基基团包含2-4个碳原子。烯基基团的实例包括,但并不限于,乙烯基(-CH=CH 2)、烯丙基(-CH 2CH=CH 2)、烯丙基(-CH=CHCH 3)等等。
术语“炔基”表示含有2-12个碳原子的直链或支链一价烃基,其中至少有一个碳-碳sp三键,其中,所述炔基基团可以任选地被一个或多个本发明所描述的取代基所取代。在一些实施方案中,炔基基团包含2-8个碳原子;在另一些实施方案中,炔基基团包含2-6个碳原子;在又一些实施方案中,炔基基团包含2-4个碳原子。炔基基团的实例包括,但并不限于,乙炔基、丙炔基等等。
术语“亚烷基”表示从饱和的直链或支链烃基中去掉两个氢原子所得到的饱和的二价烃基基团。除非另外详细说明,亚烷基基团含有1-12个碳原子。在一些实施方案中,亚烷基基团含有1-6个碳原子;在另一些实施方案中,亚烷基基团含有1-4个碳原子;在又一些实施方案中,亚烷基基团含有1-3个碳原子;在另一些实施方案中,亚烷基基团含有1-2个碳原子。这样的实例包括亚甲基(-CH 2-),亚乙基(-CH 2CH 2-),亚异丙基(-CH(CH 3)CH 2-),亚正丁基(-CH 2CH 2CH 2CH 2-)等等。
术语“亚烯基”表示从烯基基团中去掉两个氢原子所得到的二价烃基基团。除非另外详细说明,亚烯基 基团含有1-12个碳原子。在一些实施方案中,亚烯基基团含有1-6个碳原子;在另一些实施方案中,亚烯基基团含有1-4个碳原子;在又一些实施方案中,亚烯基基团含有1-3个碳原子。这样的实例包括亚丙烯基、亚丁烯基等等。
术语“烷氧基”表示烷基基团通过氧原子与分子其余部分相连,其中,烷基基团具有如本发明所述的含义。在一些实施方案中,烷氧基基团含有1-6个碳原子;在另一些实施方案中,烷氧基基团含有1-4个碳原子;在又一些实施方案中,烷氧基基团含有1-3个碳原子。所述烷氧基基团可以任选地被一个或多个本发明所描述的取代基所取代。烷氧基基团的实例包括,但并不限于,甲氧基(MeO、-OCH 3),乙氧基(EtO、-OCH 2CH 3),1-丙氧基(n-PrO、n-丙氧基、-OCH 2CH 2CH 3),等等。
术语“烷氧基烷基”表示烷基基团被一个或多个烷氧基基团所取代,其中,烷基和烷氧基具有本发明所描述的含义。所述烷氧基烷基可以任选地被一个或多个本发明所描述的取代基所取代。在一些实施方案中,烷氧基烷基为C 1-4烷氧基C 1-4烷基;在另一些实施方案中,烷氧基烷基为C 1-2烷氧基C 1-3烷基;在又一些实施方案中,烷氧基烷基为C 1-3烷氧基C 1-3烷基。这样的实例包括但不限于,甲氧基甲基、甲氧基乙基、异丙氧甲基、叔丁氧基甲基、叔丁氧基乙基等。
术语“烷酰基”、“烷基酰基”或“烷基甲酰基”表示烷基基团通过羰基(-C(=O)-)与分子其余部分相连,其中,烷基基团具有如本发明所述的含义。所述烷酰基基团可以任选地被一个或多个本发明所描述的取代基所取代。烷酰基基团的实例包括,但并不限于,乙酰基(-C(=O)CH 3),丙酰基(-C(=O)CH 2CH 3),丁酰基(-C(=O)CH 2CH 2CH 3)等等。
术语“烷氧基酰基”、“烷氨基酰基”、“氨基酰基”表示烷氧基、烷氨基或氨基(-NH 2)基团通过羰基(-C(=O)-)与分子其余部分相连,其中,烷氧基和烷氨基基团具有如本发明所述的含义。所述烷氧基酰基、烷氨基酰基、氨基酰基基团可以任选地被一个或多个本发明所描述的取代基所取代。烷氧基酰基、烷氨基酰基的实例包括,但并不限于,甲氧基酰基(-C(=O)OCH 3),乙氧基酰基(-C(=O)OCH 2CH 3),甲氨基酰基(-C(=O)NHCH 3),二甲氨基酰基(-C(=O)N(CH 3) 2)等等。
术语“烷基磺酰基”、“烷氧基磺酰基”、“烷氨基磺酰基”、“氨基磺酰基”表示烷基、烷氧基、烷氨基或氨基(-NH 2)基团通过磺酰基(-SO 2-)与分子其余部分相连,其中,烷基、烷氧基、烷氨基基团具有如本发明所述的含义。所述烷基磺酰基、烷氧基磺酰基、烷氨基磺酰基、氨基磺酰基基团可以任选地被一个或多个本发明所描述的取代基所取代。烷基磺酰基基团的实例包括,但并不限于,甲基磺酰基(-SO 2CH 3),乙基磺酰基(-SO 2CH 2CH 3),等等。
术语“卤代烷基”,“卤代烯基”或“卤代烷氧基”表示烷基,烯基或烷氧基基团被一个或多个卤素原子所取代,这样的实例包含,但并不限于,三氟甲基、二氟甲基2,2-二氟乙基、三氟甲氧基等。
术语“氘代烷基”或“氘代烷氧基”表示烷基或烷氧基基团被一个或多个氘原子所取代,这样的实例包含,但并不限于,氘代甲基(-CD 3)、氘代甲氧基(-OCD 3)等。
术语“碳环”或“碳环基”都是指含有3-12个环碳原子的,单价或多价的非芳香性的饱和或部分不饱和单环、双环或三环体系。在一些实施方案中,碳环包含3-10个环碳原子;在另一些实施方案中,碳环包含3-8个环碳原子;在又一些实施方案中,碳环包含3-6个环碳原子;在一些实施方案中,碳环包含7-12个环碳原子;在另一些实施方案中,碳环包含5-8个环碳原子。碳环基团的实例包括,但并不限于,环丙基、环丁基、环戊基、环己基、1-环戊基-1-烯基、1-环戊基-2-烯基、1-环戊基-3-烯基、1-环己基-1-烯基、1-环己基-2-烯基、1-环己基-3-烯基、环己二烯基、环庚基、环辛基、环壬基、环癸基、环十一烷基、环十二烷基,等等。其中,所述的碳环基团可以任选地被一个或多个本发明所描述的取代基所取代。
术语“环烷基”是指含有3-12个环碳原子的,单价或多价的非芳香性的饱和单环、双环或三环体系。在一些实施方案中,环烷基包含3-10个环碳原子;在另一些实施方案中,环烷基包含3-8个环碳原子;在又 一些实施方案中,环烷基包含3-6个环碳原子。环烷基基团的实例包括,但并不限于,环丙基、环丁基、环戊基、环己基等。所述环烷基基团可以任选的被一个或多个本发明所描述的取代基所取代。
术语“杂环基”和“杂环”在此处可交换使用,都是指包含3-12个环原子的饱和或部分不饱和的单环、双环或三环体系,其中至少一个环原子选自氮、硫和氧原子,且所述杂环体系中任意一个环都是非芳香性的。除非另外说明,杂环基可以是碳基或氮基,且-CH 2-基团可以任选地被-C(=O)-替代。环的硫原子可以任选地被氧化成S-氧化物。环的氮原子可以任选地被氧化成N-氧化合物。在一些实施方案中,杂环基为5-12个原子组成的杂环基;在另一些实施方案中,杂环基为5-8个原子组成的杂环基;在又一些实施方案中,杂环基为5-7个原子组成的杂环基;还在一些实施方案中,杂环基为5-6个原子组成的杂环基。杂环基还可以是双环杂环基;在一些实施方案中,杂环基是7-12个原子组成的双环杂环基;在另一些实施方案中,杂环基是7-10个原子组成的双环杂环基;在又一些实施方案中,杂环基是8-10个原子组成的双环杂环基。
杂环基的实例包括,但不限于:环氧乙烷基、氮杂环丁基、氧杂环丁基、吡咯烷基、吡唑烷基、二氢噻吩基、1,3-二氧环戊基、二硫环戊基、四氢吡喃基、四氢噻喃基、哌啶基、1,2-二氢吡啶基、吗啉基、硫代吗啉基、六氢嘧啶基、1,6-二氢嘧啶基、1,2-二氢嘧啶基、1,2-二氢吡嗪基、1,3-噁嗪烷基、哌嗪基、噁唑烷基、二噁烷基、二噻烷基、噻噁烷基、高哌嗪基、高哌啶基、氧杂环庚烷基、硫杂环庚烷基、吲哚啉基、1,2,3,4-四氢异喹啉基、1,3-苯并二噁茂基、2-氧杂-5-氮杂双环[2.2.1]庚-5-基。杂环基中-CH 2-基团被-C(=O)-取代的实例包括,但不限于,2-氧代吡咯烷-1-基、氧代-1,3-噻唑烷基、2-氧代噁唑烷-3-基、2-氧代哌啶-1-基、3-氧代吗啉基、2-氧代哌嗪-1-基、2-氧代吡啶-1(2H)-基、3,5-二氧代哌啶基、6-氧代嘧啶-1(6H)-基、2-氧代嘧啶-1(2H)-基、2-氧代四氢嘧啶-1(2H)-基、2-氧代吡嗪-2(1H)-基和嘧啶二酮基。杂环基中硫原子被氧化的实例包括,但不限于,环丁砜基、1,1-二氧代硫代吗啉基和1,1-二氧代-1,2-硫代吗啉基。所述的杂环基基团可以是取代或非取代的,其中取代基可以是,但并不限于,氟、氯、溴、氧代(=O)、氰基、硝基、羧基、羟基、氨基、氨基甲基、氨基酰基、甲氨基、苯氨基、羟基甲基、甲基磺酰基、氨基磺酰基、乙酰基、甲氧基、苯氧基、三氟甲氧基、甲基、乙基、丙基、异丙基、正丁基、叔丁基、环丙基、环戊基、环己基、四氢呋喃基、四氢噻吩基、四氢吡咯基、咪唑基、咪唑啉基、哌啶基、哌嗪基、吗啉基、噻吩基、噻唑基、呋喃基、吡咯基、苯基、吡啶基、嘧啶基、-C(=NH)NH 2或三氟甲基等。
在一些实施方案中,杂环基为5-6个原子组成的杂环基,是指包含5或6个环原子的饱和或部分不饱和的单环,其中至少一个环原子选自氮、硫和氧原子。5-6个原子组成的杂环基的实例包括,但不限于:吡咯烷基,吡咯啉基,吡唑啉基,吡唑烷基,咪唑啉基,咪唑烷基,四氢呋喃基,二氢呋喃基,四氢噻吩基,噁唑烷基,哌啶基,1,2-二氢吡啶基,吗啉基,硫代吗啉基,六氢嘧啶基,1,6-二氢嘧啶基,1,2-二氢嘧啶基,1,2-二氢吡嗪基,1,3-噁嗪烷基,哌嗪基,1,2,3,6-四氢吡啶基,1,2,3,4-四氢吡啶基,1,2,3,4-四氢嘧啶基,2,5-二氢-1H-吡咯基等。5-6个原子组成的杂环基中的-CH 2-基团可以被-C(=O)-取代,或其中的硫原子可以被氧化成S-氧化物。并且,所述的5-6个原子组成的杂环基基团可以任选地被一个或多个本发明所描述的取代基所取代。
在本发明中所使用的术语“不饱和的”表示基团中含有一个或多个不饱和度。
术语“杂原子”是指O、S、N、P和Si,包括N、S和P任何氧化态的形式;伯、仲、叔胺和季铵盐的形式;或者杂环中氮原子上的氢被取代的形式,例如,N(像3,4-二氢-2H-吡咯基中的N),NH(像吡咯烷基中的NH)或NR(像N-取代的吡咯烷基中的NR)。
术语“卤素”是指氟(F)、氯(Cl)、溴(Br)或碘(I)。
术语“芳基”表示含有6-14个环原子,或6-12个环原子,或8-12个环原子,或6-10个环原子的单环、双环和三环的烃基基团,其中,至少一个环是芳香族的,并且所述芳香体系中有一个或多个附着点与分子的其余部分相连。术语“芳基”可以和术语“芳香环”或“芳环”交换使用。所述芳基基团包括芳香环与芳香环、 或芳香环与非芳香性的碳环稠合而成的环体系。芳基基团的实例可以包括苯基、萘基、蒽基、1,2,3,4-四氢萘基、2,3-二氢-1H-茚基、二环[4,2,0]辛-1(6),2,4-三烯基。所述芳基基团可以是取代或非取代的,其中取代基可以是,但并不限于,氟、氯、溴、氧代(=O)、氰基、硝基、羧基、羟基、氨基、氨基甲基、氨基酰基、甲氨基、苯氨基、羟基甲基、甲基磺酰基、氨基磺酰基、乙酰基、甲氧基、苯氧基、三氟甲氧基、甲基、乙基、丙基、异丙基、正丁基、叔丁基、环丙基、环戊基、环己基、四氢呋喃基、四氢噻吩基、四氢吡咯基、咪唑基、咪唑啉基、哌啶基、哌嗪基、吗啉基、噻吩基、噻唑基、呋喃基、吡咯基、苯基、吡啶基、嘧啶基、-C(=NH)NH 2或三氟甲基等。
术语“杂芳基”表示含有5-12个环原子,或5-10个环原子,或5-6个环原子的单环、双环和三环体系,其中至少一个环是芳香族的,且至少一个环包含一个或多个杂原子,其中所述杂芳体系中有一个或多个附着点与分子其余部分相连。术语“杂芳基”可以与术语“杂芳环”或“杂芳族化合物”交换使用。所述杂芳基包括杂芳环与芳香环、杂芳环与杂芳环、或杂芳环与非芳香性的碳环或杂环稠合而成的环体系。在一些实施方案中,5-10个原子组成的杂芳基包含1、2、3或4个独立选自O、S和N的杂原子。在一些实施方案中,杂芳基为7-12个原子组成的杂芳基,其包含1、2、3或4个独立选自O、S和N的杂原子;7-12个原子组成的杂芳基可以是单环体系,也可以是包含两个环的双环体系。在另一些实施方案中,杂芳基为7-10个原子组成的杂芳基,其包含1、2、3或4个独立选自O、S和N的杂原子;7-10个原子组成的杂芳基可以是单环体系,也可以是包含两个环的双环体系。所述杂芳基基团可以是取代或非取代的,其中取代基可以是,但并不限于,氟、氯、溴、氧代(=O)、氰基、硝基、羧基、羟基、氨基、氨基甲基、氨基酰基、甲氨基、苯氨基、羟基甲基、甲基磺酰基、氨基磺酰基、乙酰基、甲氧基、苯氧基、三氟甲氧基、甲基、乙基、丙基、异丙基、正丁基、叔丁基、环丙基、环戊基、环己基、四氢呋喃基、四氢噻吩基、四氢吡咯基、咪唑基、咪唑啉基、哌啶基、哌嗪基、吗啉基、噻吩基、噻唑基、呋喃基、吡咯基、苯基、吡啶基、嘧啶基、胍基(-NHC(=NH)NH 2)、-N=C=S、-C(=NH)NH 2或三氟甲基等。
杂芳基基团的实例包括,但并不限于,呋喃基、咪唑基(如1H-咪唑-1-基)、异噁唑基、噁唑基、吡咯基、1,3,4-噁二唑基、2-吡啶基、3-吡啶基、4-吡啶基、2-嘧啶基、4-嘧啶基、5-嘧啶基、哒嗪基(如3-哒嗪基)、噻唑基、2-噻吩基、3-噻吩基、吡唑基(如2-吡唑基、1H-吡唑-1-基)、吡嗪基、1,3,5-三嗪基、***基、四唑基等;也包括以下的双环,但绝不限于这些双环:苯并咪唑基、苯并呋喃基、二氢苯并呋喃基、苯并噻吩基、吲哚基(如2-吲哚基)、异吲哚啉基、吲唑基(如1H-吲唑-1-基等)、喹啉基(如2-喹啉基、3-喹啉基、4-喹啉基)、异喹啉基(如1-异喹啉基、3-异喹啉基或4-异喹啉基)、5,6,7,8-四氢喹啉基、3,4-二氢-2H-吡喃并[3,2-b]吡啶基、2,3-二氢-[1,4]二噁英并[2,3-b]吡啶基、2,3-二氢苯并[b][1,4]二噁英基、6,7-二氢-5H-环戊烷并[3,2-b]吡啶基、2,3-二氢呋喃[3,2-b]吡啶、喹啉酮基、二氢喹啉酮基,等等。
术语“i个原子组成的”,其中i是整数,典型地描述分子中成环原子的数目,在所述分子中成环原子的数目是i。例如,哌啶基是6个原子组成的杂环烷基,而喹啉基或5,6,7,8-四氢喹啉基是10个原子组成的杂芳基基团。
术语“双环碳环基”、“双环芳基”、“双环杂芳基”、“双环杂环基”表示由两个环组成的碳环基、芳基、杂芳基和杂环基,所述的碳环基、芳基、杂芳基和杂环基具有本发明所描述的含义。其中,双环碳环基、双环杂环基包括两个环组成的稠环、螺环和桥环。在一些实施方案中,双环芳基、双环杂芳基为芳香性环(芳基或杂芳基)和非芳香性环(碳环或杂环)稠合而成的环体系,其中,所述非芳香性环可以是饱和的,也可以是不饱和的,例如,1,2,3,4-四氢萘基、2,3-二氢-1H-茚基、5,6,7,8-四氢喹啉基、3,4-二氢-2H-吡喃并[3,2-b]吡啶基、2,3-二氢-[1,4]二噁英并[2,3-b]吡啶基、2,3-二氢苯并[b][1,4]二噁英基、6,7-二氢-5H-环戊烷并[3,2-b]吡啶基、2,3-二氢呋喃[3,2-b]吡啶等;在另一些实施方案中,双环芳基或双环杂芳基为两个芳香性环(芳基或杂芳基)稠合而成的环体系,例如,萘基、喹啉基、吲唑基等。更多的双环芳基或双环杂芳基 的例子可参见前述的芳基和杂芳基定义中实例。
术语“羧基”,无论是单独使用还是和其他术语连用,如“羧烷基”,表示-CO 2H;术语“羰基”,无论是单独使用还是和其他术语连用,如“氨基酰基”或“酰氧基”,表示-(C=O)-。
术语“酰氧基”,无论是单独使用还是和其他术语连用,如“烷基酰氧基”,表示-(C=O)O-;术语“烷基酰氧基”表示烷基通过酰氧基(-(C=O)O-)与分子其余部分相连,其中烷基基团具有如本发明所述的含义。所述烷基酰氧基基团可以任选地被一个或多个本发明描述的取代基所取代。
术语“烷氨基”、“烷基氨基”包括“N-烷基氨基”和“N,N-二烷基氨基”,其中氨基基团分别独立地被一个或两个烷基基团所取代。所述烷基基团具有如本发明所述的含义合适的烷基氨基基团可以是单烷基氨基或二烷基氨基,这样的实例包括,但并不限于,N-甲氨基,N-乙氨基,N,N-二甲氨基,N,N-二乙氨基等等。
术语“氨基取代的烷基”包括被一个或多个氨基所取代的C 1-10直链或支链烷基基团。其中一些实施例是,氨基烷基是被一个或多个氨基基团所取代的C 1-6“较低级的氨基烷基”,这样的实例包括,但并不限于,氨甲基,氨乙基,氨丙基,氨丁基和氨己基等。
术语“羟基取代的烷基”包括被一个或多个羟基所取代的C 1-10直链或支链烷基基团。其中一些实施例是,羟基烷基是被一个或多个羟基基团所取代的C 1-6“较低级的羟基烷基”,这样的实例包括,但并不限于,羟基甲基,羟基乙基,羟基丙基,羟基丁基和羟基己基等。
术语“氰基取代的烷基”包括被一个或多个氰基所取代的C 1-10直链或支链烷基基团。其中一些实施例是,羟基烷基是被一个或多个氰基基团所取代的C 1-6“较低级的氰基烷基”,这样的实例包括,但并不限于,氰基甲基,氰基乙基等。
如本发明所描述,取代基(R e) n1由一个键连接到中心的环上形成的环体系代表n1个取代基R e可以在环上任何可取代的位置进行取代,其中,n1最大为环上可被取代的位点的数目的总和。例如,当式a中环A a为喹啉环(如式a1所示)时,代表A 1环或B 1环上任何可能被取代的位置均可被n1个R e取代,此处,n1可为1、2、3、4、5、6或7;其中,当n1大于1时,所述的各个R e可以是相同的,也可以是不同的。
Figure PCTCN2018072988-appb-000036
如本发明所描述,取代基(R 9) g由一个键连接到大环上形成的体系代表g个取代基R 9可以在亚烷基上任何可以取代的位置进行取代。例如,式d中的R 9可以在环中的亚丁基
Figure PCTCN2018072988-appb-000037
上任何可以取代的位置进行取代,包括但不限于式d1所示的情况;其中,式d、d1中的g、R 9、A、B、R 1、R 2、R 5、m和n具有本发明所述的含义。
Figure PCTCN2018072988-appb-000038
如本发明所描述,附着点可以在环上任何可连接的位置与分子其余部分连接。例如,当式b中的环A a为喹啉环(如式b1所示)时,代表A 2环或B 2环上任何可能被连接的位置均可作为连接的点。
Figure PCTCN2018072988-appb-000039
如本发明所描述,当某基团通过两个连接位点与分子其余部分相连时,与所述连接位点相连的分子其余部分是可以互换的。例如,当式c中的A a环为式c1所示的喹啉环时,所述的喹啉环可以通过E 1和E 2端与分子其余部分相连,且E 1和E 2端所连接的部分可以互换;又如,式c4中的-C(=O)NH-基团通过E 3和E 4端与分子其余部分相连,且E 3和E 4端所连接的部分可以互换。更具体一点的例子是,当式c1中的喹啉环与环B a相连时,所述的喹啉环可以通过E 1或E 2端与环B a相连(如式c2和c3所示)。
Figure PCTCN2018072988-appb-000040
术语“保护基团”或“PG”是指一类取代基,它们与其他官能团起反应的时候,通常用来阻断或保护该官能团的特殊功能性。例如,“氨基的保护基团”是指一个取代基与氨基基团相连来阻断或保护化合物中氨基的功能性,合适的氨基保护基团包括乙酰基,三氟乙酰基,叔丁氧羰基(BOC,Boc),苄氧羰基(CBZ,Cbz)和9-芴亚甲氧羰基(Fmoc)。相似地,“羟基保护基团”是指羟基的取代基用来阻断或保护羟基的功能性,合适的保护基团包括乙酰基和甲硅烷基。“羧基保护基团”是指羧基的取代基用来阻断或保护羧基的功能性,一般的羧基保护基包括-CH 2CH 2SO 2Ph,氰基乙基,2-(三甲基硅烷基)乙基,2-(三甲基硅烷基)乙氧基甲基,2-(对甲苯磺酰基)乙基,2-(对硝基苯磺酰基)乙基,2-(二苯基膦基)乙基,硝基乙基,等等。对于保护基团一般的描述可参考文献:T W.Greene,Protective Groups in Organic Synthesis,John Wiley&Sons,New York,1991;and P.J.Kocienski,Protecting Groups,Thieme,Stuttgart,2005.
本发明所使用的术语“前药”,代表一个化合物在体内可以转化为本发明所述的化合物。这样的转化受前体药物在血液中水解或在血液或组织中经酶转化为母体结构的影响。本发明前体药物类化合物可以是酯,在现有的发明中酯可以作为前体药物的有苯酯类,脂肪族(C 1-24)酯类,酰氧基甲基酯类,碳酸酯,氨基甲酸酯类和氨基酸酯类。例如本发明里的一个化合物包含羟基,即可以将其酰化得到前体药物形式的化合物。其他的前体药物形式包括磷酸酯,如这些磷酸酯类化合物是经母体上的羟基磷酸化得到的。关于前体药物完整的讨论可以参考以下文献:T.Higuchi and V.Stella,Pro-drugs as Novel Delivery Systems,Vol.14of the A.C.S.Symposium Series,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,J.Rautio et al.,Prodrugs:Design and Clinical Applications,Nature Review Drug Discovery,2008,7,255-270,and S.J.Hecker et al.,Prodrugs of Phosphates and Phosphonates,Journal of Medicinal Chemistry,2008,51,2328-2345。
“代谢产物”是指具体的化合物或其盐在体内通过代谢作用所得到的产物。一个化合物的代谢产物可以通过所属领域公知的技术来进行鉴定,其活性可以通过如本发明所描述的那样采用试验的方法进行表征。这样的产物可以是通过给药化合物经过氧化,还原,水解,酰氨化,脱酰氨作用,酯化,脱脂作用,酶裂解等等方法得到。相应地,本发明包括化合物的代谢产物,包括将本发明的化合物与哺乳动物充分接触一段时间所产生的代谢产物。
本发明的“溶剂化物”是指一个或多个溶剂分子与本发明的化合物所形成的缔合物。形成溶剂化物的溶剂包括,但并不限于,水,异丙醇,乙醇,甲醇,二甲亚砜,乙酸乙酯,乙酸和氨基乙醇。术语“水合物”是指溶剂分子是水所形成的缔合物。
本发明中“室温”指的是温度由10℃到40℃。在一些实施例中,“室温”指的是温度由20℃到30℃;在另一些实施例中,“室温”指的是25℃。
如本发明所使用的“治疗”任何疾病或病症,在其中一些实施方案中指改善疾病或病症(即减缓或阻止 或减轻疾病或其至少一种临床症状的发展)。在另一些实施方案中,“治疗”指缓和或改善至少一种身体参数,包括可能不为患者所察觉的身体参数。在另一些实施方案中,“治疗”指从身体上(例如稳定可察觉的症状)或生理学上(例如稳定身体的参数)或上述两方面调节疾病或病症。在另一些实施方案中,“治疗”指预防或延迟疾病或病症的发作、发生或恶化。
本发明所使用的“血栓栓塞性疾病”是指由血栓形成和血栓栓塞两种病理过程所引起的疾病,又称血栓性疾病。血栓形成是指在一定条件下,血液有形成份在血管内或心脏内膜局部形成栓子,造成血管部分或完全堵塞,相应部位血液供应障碍的病理过程。血栓栓塞为血栓由形成部位脱落,在随血液流动过程中,部分或全部堵塞血管,引起血管或***缺血、缺氧、坏死、淤血及水肿的病理过程。血栓栓塞性疾病的实例包括,但并不限于,动脉心血管血栓栓塞疾病、静脉心血管血栓栓塞疾病和在心脏的腔室中的血栓栓塞疾病。此类疾病更具体的实例包括,但并不限于,心肌梗塞、心绞痛(包括不稳定绞痛)、急性冠状动脉综合征、再阻塞和血管形成术或主动脉冠状功脉分流术后的再狭窄、中风、短暂的局部缺血发作、周围动脉闭塞性疾病、动脉血栓、冠状动脉血栓形成、脑动脉血栓形成、脑栓塞、肾栓塞、肺栓塞、血栓性静脉炎、静脉血栓形成或深静脉血栓形成,等等。
本发明所使用的“弥散性血管内凝血(DIC)”是指在多种疾病基础上发生的凝血***激活,导致小血管内广泛微血栓形成,消耗大量凝血因子并继发纤溶亢进,进而引起全身性出血及微循环衰竭的临床综合征。广泛的微血栓的形成会导致多器官功能衰竭,激活纤溶***并使凝血因子耗竭,从而继发出血。因此弥散性血管内凝血是一种凝血与出血并发的综合症。目前,主要的治疗方法是在积极控制原发病的基础上进行抗凝、替代、抗血小板聚集及对症支持治疗等。因此,可以使用本发明公开的化合物治疗弥散性血管内凝血。
本发明的“药学上可接受的盐”,即可药用盐可以用常规化学方法由母体化合物、碱性或酸性部分来合成。一般而言,该类盐可以通过使这些化合物的游离酸形式与化学计量量的适宜碱(如Na、Ca、Mg或K的氢氧化物、碳酸盐、碳酸氢盐等)反应,或者通过使这些化合物的游离碱形式与化学计量量的适宜酸反应来进行制备。该类反应通常在水或有机溶剂或二者的混合物中进行。一般地,在适当的情况中,需要使用非水性介质如***、乙酸乙酯、乙醇、异丙醇或乙腈。在例如“Remington′s Pharmaceutical Sciences”,第20版,Mack Publishing Company,Easton,Pa.,(1985);和“药用盐手册:性质、选择和应用(Handbook of Pharmaceutical Salts:Properties,Selection,and Use)”,Stahl and Wermuth(Wiley-VCH,Weinheim,Germany,2002)中可找到另外一些适宜盐的列表。
可药用盐可以是可药用的酸加成盐,可由本发明的化合物与无机酸和/或有机酸作用所形成,例如,与无机酸,如盐酸、氢溴酸、硝酸、磷酸或硫酸等形成的盐;与有机酸,如乙酸、三氟乙酸、丙酸、丙二酸、草酸、马来酸、富马酸、苹果酸、柠檬酸、葡糖酸、扁桃酸、酒石酸、硬脂酸、琥珀酸、磺基水杨酸、乳酸、苯甲酸、苯磺酸、甲磺酸、乙磺酸、甲苯磺酸或萘二磺酸等形成的盐。
可药用盐可以是可药用的碱加成盐,可由本发明的化合物与无机碱和/或有机碱作用所形成。可以由其衍生得到盐的无机碱包括,例如铵盐和周期表的I族至XII族的金属。在某些实施方案中,该盐衍生自钠、钾、铵、钙、镁、铁、银、锌和铜;特别适合的盐包括铵、钾、钠、钙和镁盐。可以由其衍生得到盐的有机碱包括伯胺、仲胺和叔胺,取代的胺包括天然存在的取代的胺、环状胺、碱性离子交换树脂等。某些有机胺包括,例如,异丙胺、苄星青霉素(benzathine)、胆碱盐(cholinate)、二乙醇胺、二乙胺、赖氨酸、葡甲胺(meglumine)、哌嗪和氨丁三醇。
另外,本发明公开的化合物、包括它们的盐,也可以以它们的水合物形式或包含其溶剂(例如乙醇,DMSO,等等)的形式存在,并可用于结晶。本发明公开化合物可以与药学上可接受的溶剂(包括水)固有地或通过设计形成溶剂化物;因此,本发明化合物包括溶剂化的和未溶剂化的形式。
本发明给出的任何结构式也意欲表示这些化合物未被同位素富集的形式以及同位素富集的形式。同位素富集的化合物具有本发明给出的通式描绘的结构,除了一个或多个原子被具有所选择原子量或质量数的原子替换。可引入本发明化合物中的示例性同位素包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,如 2H, 3H, 11C, 13C, 14C, 15N, 17O, 18O, 18F, 31P, 32P, 35S, 36Cl和 125I。
另一方面,本发明所述化合物包括同位素富集的本发明所定义的化合物,例如,其中存在放射性同位素,如 3H, 14C和 18F的那些化合物,或者其中存在非放射性同位素,如 2H和 13C。该类同位素富集的化合物可用于代谢研究(使用 14C)、反应动力学研究(使用例如 2H或 3H)、检测或成像技术,如正电子发射断层扫描术(PET)或包括药物或底物组织分布测定的单光子发射计算机断层成像术(SPECT),或可用于患者的放疗中。 18F富集的化合物对PET或SPECT研究而言是特别理想的。同位素富集的本发明化合物可以通过本领域技术人员熟悉的常规技术或本发明中的实施例和制备过程所描述使用合适的同位素标记试剂替代原来使用过的未标记试剂来制备。
此外,较重同位素特别是氘(即, 2H或D)的取代可提供某些治疗优点,这些优点是由代谢稳定性更高带来的。例如,体内半衰期增加或剂量需求降低或治疗指数得到改善带来的。应当理解,本发明中的氘被看做本发明化合物的取代基。可以用同位素富集因子来定义该类较重同位素特别是氘的浓度。本发明所使用的术语“同位素富集因子”是指所指定同位素的同位素丰度和天然丰度之间的比例。如果本发明化合物的取代基被指定为氘,该化合物对各指定的氘原子而言具有至少3500(各指定氘原子处52.5%的氘掺入)、至少4000(60%的氘掺入)、至少4500(67.5%的氘掺入),至少5000(75%的氘掺入),至少5500(82.5%的氘掺入)、至少6000(90%的氘掺入)、至少6333.3(95%的氘掺入)、至少6466.7(97%的氘掺入)、至少6600(99%的氘掺入)或至少6633.3(99.5%的氘掺入)的同位素富集因子。本发明可药用的溶剂化物包括其中结晶溶剂可以是同位素取代的例如D 2O、丙酮-d 6、DMSO-d 6的那些溶剂化物。
除非其他方面表明,本发明的化合物的所有互变异构形式都包含在本发明的范围之内。另外,除非其他方面表明,本发明所描述的化合物的结构式包括一个或多个不同的原子的富集同位素。
本发明的化合物的描述
本申请的发明人经过广泛的研究,合成了一系列大环化合物,并通过FXIa酶抑制活性筛选、血浆激肽释放酶抑制活性筛选、代谢筛选、抗凝血活性实验以及其它实验,首次发现以下通式(I)表示的化合物具有很强的抗FXIa活性和/或血浆激肽释放酶抑制活性、优良的药物代谢性质以及理化性质,特别适合作为抗凝血药物用于治疗血栓栓塞性疾病。
本发明提供一种式(I)所示的大环化合物,其中环A为双环体系,优选地,环A为含杂原子的稠合双环(包括稠合双环杂环和稠合的双环杂芳环);目前该类化合物没有相关文献报道,且涉及的A环为双环的化合物具有良好的药理活性数据,可以有效治疗血栓栓塞性疾病。
本发明涉及一种化合物,其为如式(I)所示的化合物或式(I)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
Figure PCTCN2018072988-appb-000041
其中,环A、环B、环C、X、Y、R 1、R 2、R 3、R 4、n、m、p和t均具有本发明所描述的含义。
在一些实施方案中,X为C 4-8亚烷基或C 4-8亚烯基,其中,所述亚烷基和亚烯基中一个或多个碳原子独立任选地被选自-O-、-S-、-C(=O)-、-S(=O)-、-S(=O) 2-或-N(R 5)-的基团所替代;X中的C 4-8亚烷基和C 4-8亚烯基独立任选地被一个或多个相同或不同的R 9所取代;其中,R 5和R 9具有本发明所述描述的含义。
在一些实施方案中,X为C 4亚烷基或C 4亚烯基,其中,所述C 4亚烷基和C 4亚烯基中一个或多个碳原子独立任选地被选自-O-、-S-、-C(=O)-、-S(=O)-、-S(=O) 2-或-NH-的基团所替代;X中的C 4亚烷基和C 4亚烯基独立任选地被一个或多个相同或不同的R 9所取代;其中,R 9具有本发明所述描述的含义。
在另一些实施方案中,X为亚正丁基(-CH 2CH 2CH 2CH 2-)或亚丁-2-烯基(-CH 2CH=CHCH 2-),其中,所述亚正丁基和亚丁-2-烯基中一个或多个碳原子独立任选地被选自-O-、-S-、-C(=O)-、-S(=O)-、-S(=O) 2-或-NH-的基团所替代;X中的亚正丁基和亚丁-2-烯基独立任选地被一个或多个相同或不同的R 9所取代;其中,R 9具有本发明所述描述的含义。
在一些实施方案中,Y为-C(=O)N(R 5)-或-N(R 5)C(=O)-;其中,R 5和R 9具有本发明所述描述的含义。
在一些实施方案中,本发明式(I)化合物为式(Ia)所示的化合物或式(Ia)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
Figure PCTCN2018072988-appb-000042
其中,环A、环B、环C、R 1、R 2、R 3、R 4、R 5、R 9、g、n、m、p和t均具有本发明所描述的含义。
在一些实施方案中,环C为碳环基、芳基、杂环基或杂芳基。
在一些实施方案中,环C为C 3-12碳环基、C 6-12芳基、3-12个原子组成的杂环基或5-12个原子组成的杂芳基。
在另一些实施方案中,环C为C 5-6碳环基、C 6芳基、5-7个原子组成的杂环基或5-6个原子组成的杂芳基。
在一些实施方案中,环C为以下子结构式:
Figure PCTCN2018072988-appb-000043
其中,环C通过N原子与结构
Figure PCTCN2018072988-appb-000044
相连;
Figure PCTCN2018072988-appb-000045
Figure PCTCN2018072988-appb-000046
独立地为单键或双键;
Figure PCTCN2018072988-appb-000047
Figure PCTCN2018072988-appb-000048
为单键时,各Z、Z 1和Z 2独立地为CH 2或NH;
Figure PCTCN2018072988-appb-000049
Figure PCTCN2018072988-appb-000050
为双键时,各Z、Z 1和Z 2独立地为CH或N;
Z 4为CH 2或NH;
各Z 3和Z 5独立地为CH 2、NH、S或O;
q为0、1或2。
在另一些实施方案中,子结构式
Figure PCTCN2018072988-appb-000051
Figure PCTCN2018072988-appb-000052
Figure PCTCN2018072988-appb-000053
更具体的,式(C1)为
Figure PCTCN2018072988-appb-000054
其中,上述各子结构式C1、C1-a、C1-b、C1-c、C1-d、C1-e、C1-f或C1-g通过N原子与结构
Figure PCTCN2018072988-appb-000055
相连;各
Figure PCTCN2018072988-appb-000056
Figure PCTCN2018072988-appb-000057
独立地为单键或双键;Z具有本发明所描述的含义。
在一些实施方案中,环C为
Figure PCTCN2018072988-appb-000058
Figure PCTCN2018072988-appb-000059
Figure PCTCN2018072988-appb-000060
其中,环C通过N原子与结构
Figure PCTCN2018072988-appb-000061
相连。
在一些实施方案中,本发明所述的式(I)化合物可为如式(II)所示的化合物或式(II)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
Figure PCTCN2018072988-appb-000062
其中,各
Figure PCTCN2018072988-appb-000063
Figure PCTCN2018072988-appb-000064
独立地为单键或双键;
Figure PCTCN2018072988-appb-000065
Figure PCTCN2018072988-appb-000066
为单键时,各Z和Z 1独立地为CH 2或NH;当
Figure PCTCN2018072988-appb-000067
Figure PCTCN2018072988-appb-000068
为双键时,各Z和Z 1独立地为CH或N;
环A、环B、X、Y、R 1、R 2、R 3、R 4、n、m、p和t均具有本发明所描述的含义。
在一些实施方案中,本发明所述的式(I)化合物可为如式(IIa)所示的化合物,或式(IIa)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
Figure PCTCN2018072988-appb-000069
其中,各
Figure PCTCN2018072988-appb-000070
Figure PCTCN2018072988-appb-000071
独立地为单键或双键;
Figure PCTCN2018072988-appb-000072
Figure PCTCN2018072988-appb-000073
为单键时,各Z和Z 1独立地为CH 2或NH;当
Figure PCTCN2018072988-appb-000074
Figure PCTCN2018072988-appb-000075
为双键时,各Z和Z 1独立地为CH或N;环A、环B、R 1、R 2、R 3、R 4、R 5、R 9、g、n、m、p和t均具有本发明所描述的含义。
在一些实施方案中,各R 3独立地为卤素、C 1-6烷基、C 3-12碳环基、C 6-12芳基、3-12个原子组成的杂环基或5-12个原子组成的杂芳基;其中,各R 3独立任选地被1、2、3、4或5个相同或不同的R 3a所取代;其中,R 3a具有本发明所描述的含义。
在一些实施方案中,各R 3独立地为C 1-4烷基、C 3-8碳环基、C 6-10芳基、3-8个原子组成的杂环基或5-10个原子组成的杂芳基;其中,各R 3独立任选地被1、2、3、4或5个R 3a所取代;R 3a具有本发明所描述的含义。
在一些实施方案中,各R 3独立地为
Figure PCTCN2018072988-appb-000076
其中,各E 1、E 2和E 3独立地为CH或N;
各E 4和E 5独立地为CH 2、O、S或NH;
各e独立地为0、1、2或3;
其中,各R 3独立任选地被1、2、3、4或5个R 3a所取代;R 3a具有本发明所描述的含义。
在一些实施方案中,各R 3独立地为甲基、乙基、F、Cl、Br、苯基、吡咯基、吡唑基、咪唑基、咪唑啉基、***基、吡啶基、嘧啶基、吲哚基、吲唑基、苯并呋喃基、苯并噻吩基、苯并噻唑基、苯并咪唑基、苯并异噁唑基、苯并异噻唑基、喹啉基、异喹啉基或喹唑啉基;
其中,各R 3独立任选地被1、2、3、4或5个R 3a所取代;R 3a具有本发明所描述的含义。
在一些实施方案中,本发明所述的式(I)化合物可为如式(IIIa)所示的化合物或式(IIIa)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
Figure PCTCN2018072988-appb-000077
其中,各
Figure PCTCN2018072988-appb-000078
Figure PCTCN2018072988-appb-000079
独立地为单键或双键;
Figure PCTCN2018072988-appb-000080
Figure PCTCN2018072988-appb-000081
为单键时,各Z和Z 1独立地为CH 2或NH;当
Figure PCTCN2018072988-appb-000082
Figure PCTCN2018072988-appb-000083
为双键时,各Z和Z 1独立地为CH或N;环A、环B、R 1、R 2、R 4、R 5、R 9、R 3a、E 1、E 2、E 3、n、m、f、g和t均具有本发明所描述的含义。
在一些实施方案中,本发明所述的式(I)化合物可为如式(IIIb)所示的化合物或式(IIIb)所示化合物 的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
Figure PCTCN2018072988-appb-000084
其中,各
Figure PCTCN2018072988-appb-000085
Figure PCTCN2018072988-appb-000086
独立地为单键或双键;
Figure PCTCN2018072988-appb-000087
Figure PCTCN2018072988-appb-000088
为单键时,各Z和Z 1独立地为CH 2或NH;当
Figure PCTCN2018072988-appb-000089
Figure PCTCN2018072988-appb-000090
为双键时,各Z和Z 1独立地为CH或N;环A、环B、R 1、R 2、R 4、R 5、R 9、R 3a、E 1、E 2、E 3、n、m、f、g和t均具有本发明所描述的含义。
在一些实施方案中,本发明所述的式(I)化合物可为如式(III)所示的化合物或式(III)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
Figure PCTCN2018072988-appb-000091
其中,各
Figure PCTCN2018072988-appb-000092
Figure PCTCN2018072988-appb-000093
独立地为单键或双键;
Figure PCTCN2018072988-appb-000094
Figure PCTCN2018072988-appb-000095
为单键时,各Z和Z 2独立地为CH 2或NH;当
Figure PCTCN2018072988-appb-000096
Figure PCTCN2018072988-appb-000097
为双键时,各Z和Z 2独立地为CH或N;环A、环B、R 1、R 2、R 4、R 5、R 9、R 3a、n、m、f、g和t均具有本发明所描述的含义。
在一些实施方案中,本发明所述的式(I)化合物可为如式(IIIc)所示的化合物,或式(IIIc)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
Figure PCTCN2018072988-appb-000098
其中,各
Figure PCTCN2018072988-appb-000099
Figure PCTCN2018072988-appb-000100
独立地为单键或双键;
Figure PCTCN2018072988-appb-000101
Figure PCTCN2018072988-appb-000102
为单键时,各Z和Z 2独立地为CH 2或NH;当
Figure PCTCN2018072988-appb-000103
Figure PCTCN2018072988-appb-000104
为双键时,各Z和Z 2独立地为CH或N;环A、环B、R 1、R 2、R 4、R 5、R 9、R 3a、n、m、f、g和t均具有本发明所描述的含义。
在一些实施方案中,子结构式
Figure PCTCN2018072988-appb-000105
Figure PCTCN2018072988-appb-000106
Figure PCTCN2018072988-appb-000107
其中,
Figure PCTCN2018072988-appb-000108
通过N原子与结构
Figure PCTCN2018072988-appb-000109
相连。
在一些实施方案中,子结构式
Figure PCTCN2018072988-appb-000110
Figure PCTCN2018072988-appb-000111
Figure PCTCN2018072988-appb-000112
其中,
Figure PCTCN2018072988-appb-000113
通过N原子与结构
Figure PCTCN2018072988-appb-000114
相连。
在一些实施方案中,环A为C 7-12碳环基、C 8-12芳基、7-12个原子组成的杂环基或7-12个原子组成的杂芳基。
在一些实施方案中,环A为C 7-12双环碳环基、C 8-12双环芳基、7-12个原子组成的双环杂环基或7-12个原子组成的双环杂芳基。
在一些实施方案中,环A为以下子结构式:
Figure PCTCN2018072988-appb-000115
其中,各T 1、T 2、T 3、T 4、T 5、T 6、T 7和T 8独立地为-CH-或-N-;
各V 1、V 2、V 3和V 4独立地为-CH 2-、-NH-、-O-、-S-、-S(=O)-、-S(=O) 2-或-C(=O)-;
各k和j独立地为0、1、2、3或4。
在一些实施方案中,环A为
Figure PCTCN2018072988-appb-000116
Figure PCTCN2018072988-appb-000117
在一些实施方案中,环B为碳环基、芳基、杂环基或杂芳基。
在一些实施方案中,环B为C 5-8碳环基、C 6-10芳基、5-8个原子组成的杂环基或5-10个原子组成的杂芳基。
在一些实施方案中,环B为以下子结构式:
Figure PCTCN2018072988-appb-000118
Figure PCTCN2018072988-appb-000119
其中,各Q 1、Q 6和Q 9独立地为CH 2、NH、O、S、C(=O)、S(=O)或S(=O) 2
各Q 2、Q 3、Q 4、Q 5、Q 7和Q 8独立地为CH或N;
s为0、1、2或3。
在一些实施方案中,环B为
Figure PCTCN2018072988-appb-000120
Figure PCTCN2018072988-appb-000121
Figure PCTCN2018072988-appb-000122
在一些实施方案中,各R 1独立地为氧代(=O)、H、氘、卤素、-(CR 7aR 7b) rCN、C 1-6烷基、氘代C 1-6烷基、卤代C 1-6烷基、C 2-6烯基、C 2-6炔基、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=O)R 6、-(CR 7aR 7b) rC(=O)OR 8、-(CR 7aR 7b) rC(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O)R 6、-(CR 7aR 7b) rS(=O)OR 8、-(CR 7aR 7b) rS(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-12环烷基、-(CR 7aR 7b) r-C 6-12芳基、-(CR 7aR 7b) r-(3-12个原子组成的杂环基)或-(CR 7aR 7b) r-(5-12个原子组成的杂芳基);
其中,各R 1独立任选地被1、2、3或4个相同或不同的R 1a所取代;
R 1a、R 5a、R 5b、R 6、R 7a、R 7b、R 8和r均具有本发明所描述的含义。
在另一些实施方案中,各R 1独立地为氧代(=O)、H、氘、卤素、-(CR 7aR 7b) rCN、C 1-3烷基、氘代C 1-3烷基、卤代C 1-3烷基、C 2-4烯基、C 2-4炔基、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=O)R 6、-(CR 7aR 7b) rC(=O)OR 8、-(CR 7aR 7b) rC(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-6环烷基、-(CR 7aR 7b) r-C 6-10芳基、-(CR 7aR 7b) r-(3-6个原子组成的杂环基)或-(CR 7aR 7b) r-(5-10个原子组成的杂芳基);
其中,各R 1独立任选地被1、2、3或4个相同或不同的R 1a所取代;
R 1a、R 5a、R 5b、R 6、R 7a、R 7b、R 8和r均具有本发明所描述的含义。
在另一些实施方案中,各R 1独立地为氧代(=O)、H、氘、F、Cl、Br、I、-CN、-CH 2-CN、-CH 2CH 2-CN、甲基、乙基、丙基、丁基、三氟甲基、-OH、-OMe、-CH 2OMe、-CH 2OH、-NH 2、-CH 2NH 2、-CH(NH 2) 2、-NHMe、-N(Me) 2、-C(=O)Me、-C(=O)OH、-C(=O)OMe、-C(=O)NH 2、-S(=O) 2Me、-S(=O) 2OH、-S(=O) 2OMe、-S(=O) 2NH 2、环丙基、环丙基甲基、环丁基、环戊基、环丁基、苯基、吡啶基、嘧啶基、吡嗪基、哒嗪基、吡咯基、咪唑基、吡唑基、***基、四唑基、环氧烷基、环氧丁基、氮杂环丁基、四氢呋喃基、哌啶基、吡咯烷基、哌嗪基或吗啉基;其中,各R 1独立任选地被1、2、3或4个相同或不同的R 1a所取代;R 1a具有本发明所描述的含义。
在一些实施方案中,各R 2独立地为H、氘、卤素、-(CR 7aR 7b) rCN、C 1-6烷基、氘代C 1-6烷基、卤代C 1-6烷基、C 2-6烯基、C 2-6炔基、R 8O-C 2-6亚烯基、R 8OC(=O)-C 2-6亚烯基、R 8O-C 2-6亚炔基、R 8OC(=O)-C 2-6亚炔基、-N=C=S、-N=C=O、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=G)R 6、-(CR 7aR 7b) rN(R 5c)C(=G)R 6、-(CR 7aR 7b) rOC(=G)R 6、-(CR 7aR 7b) rC(=G)OR 8、-(CR 7aR 7b) rN(R 5c)C(=G)OR 8、-(CR 7aR 7b) rOC(=G)OR 8、-(CR 7aR 7b) rC(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)=C(R 7)NR 5aR 5b、-(CR 7aR 7b) rS(=O)R 6、-(CR 7aR 7b) rN(R 5c)S(=O)R 6、-(CR 7aR 7b) rS(=O)OR 8、-(CR 7aR 7b) rN(R 5c)S(=O)OR 8、-(CR 7aR 7b) rS(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rN(R 5c)S(=O) 2OR 8、 -(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-12环烷基、-(CR 7aR 7b) r-C 6-12芳基、-(CR 7aR 7b) r-(3-12个原子组成的杂环基)、-(CR 7aR 7b) r-(5-12个原子组成的杂芳基)或D-甘露糖基;其中,各G独立地为O、S或NR 5d
各R 2独立任选地被1、2、3或4个相同或不同的R 2a所取代;
其中,R 2a、R 5a、R 5b、R 5c、R 5d、R 6、R 7、R 7a、R 7b、R 8和r均具有本发明所描述的含义。
在另一些实施方案中,各R 2独立地为H、氘、F、Cl、Br、I、-(CR 7aR 7b) rCN、C 1-4烷基、氘代C 1-4烷基、卤代C 1-4烷基、C 2-4烯基、C 2-4炔基、R 8O-C 2-4亚烯基、R 8OC(=O)-C 2-4亚烯基、R 8O-C 2-4亚炔基、R 8OC(=O)-C 2-4亚炔基、-N=C=S、-N=C=O、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=G)R 6、-(CR 7aR 7b) rN(R 5c)C(=G)R 6、-(CR 7aR 7b) rOC(=G)R 6、-(CR 7aR 7b) rC(=G)OR 8、-(CR 7aR 7b) rN(R 5c)C(=G)OR 8、-(CR 7aR 7b) rOC(=G)OR 8、-(CR 7aR 7b) rC(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)=C(R 7)NR 5aR 5b、-(CR 7aR 7b) rS(=O)R 6、-(CR 7aR 7b) rN(R 5c)S(=O)R 6、-(CR 7aR 7b) rS(=O)OR 8、-(CR 7aR 7b) rN(R 5c)S(=O)OR 8、-(CR 7aR 7b) rS(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rN(R 5c)S(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-8环烷基、-(CR 7aR 7b) r-C 6-10芳基、-(CR 7aR 7b) r-(3-8个原子组成的杂环基)、-(CR 7aR 7b) r-(5-10个原子组成的杂芳基)或D-甘露糖基;其中,各G独立地为O、S或NR 5d
各R 2独立任选地被1、2、3或4个相同或不同的R 2a所取代;
其中,R 2a、R 5a、R 5b、R 5c、R 5d、R 6、R 7、R 7a、R 7b、R 8和r均具有本发明所描述的含义。
在又一些实施方案中,各R 2独立地为H、氘、F、Cl、Br、I、-(CR 7aR 7b) rCN、C 1-3烷基、氘代C 1-3烷基、卤代C 1-3烷基、C 2-4烯基、R 8O-C 2-6亚烯基、R 8OC(=O)-C 2-6亚烯基、R 8O-C 2-6亚炔基、R 8OC(=O)-C 2-6亚炔基、-N=C=S、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=O)R 6、-(CR 7aR 7b) rOC(=G)R 6、-(CR 7aR 7b) rN(R 5c)C(=O)R 6、-(CR 7aR 7b) rC(=O)OR 8、-(CR 7aR 7b) rN(R 5c)C(=O)OR 8、-(CR 7aR 7b) rC(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=S)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=N-R 5d)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rN(R 5c)S(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-6环烷基、-(CR 7aR 7b) r-C 6-10芳基、-(CR 7aR 7b) r-(3-6个原子组成的杂环基)或-(CR 7aR 7b) r-(5-10个原子组成的杂芳基);其中,各R 2独立任选地被1、2、3或4个相同或不同的R 2a所取代;R 2a、R 5a、R 5b、R 5c、R 5d、R 6、R 7a、R 7b、R 8和r均具有本发明所描述的含义。
在一些实施方案中,各R 2独立地为H、氘、F、Cl、Br、I、CN、-CH 2-CN、-CH 2CH 2-CN、甲基、乙基、丙基、丁基、氘代甲基、二氘代甲基、一氘代甲基、三氟甲基、二氟甲基、2,2-二氟乙基、1,2-二氟乙基、2,2,2-三氟乙基、乙烯基、丙烯基、烯丙基、HO-乙烯基、MeO-乙烯基、HOC(=O)-乙烯基、MeOC(=O)-乙烯基、-N=C=S、-OH、-OMe、-O(i-Pr)、-O(t-Bu)、-CH 2-OH、-CH 2CH 2-OH、-CH 2-OMe、-CH 2CH 2-OMe、-CH 2-O(i-Pr)、-CH 2-O(t-Bu)、苯基-O-、吡啶基-O-、嘧啶基-O-、吡嗪基-O-、哒嗪基-O-、吡咯基-O-、吡唑基-O-、噻唑基-O-、噁唑基-O-、噁二唑基-O-、咪唑基-O-、噻二唑基-O-、-NH 2、-NHMe、-N(Me) 2、-CH 2-NH 2、-CH 2CH 2-NH 2、-CH 2-NHMe、-CH 2-N(Me) 2、苯基-NH-、吡啶基-NH-、嘧啶基-NH-、吡嗪基-NH-、哒嗪基-NH-、吡咯基-NH-、吡唑基-NH-、噻唑基-NH-、噁唑基-NH-、噁二唑基-NH-、咪唑基-NH-、噻二唑基-NH-、-C(=O)Me、-C(=O)Et、-CH 2-C(=O)Me、-CH 2-C(=O)Et、-NHC(=O)Me、-C(=O)OH、-C(=O)OMe、-NHC(=O)OMe、-NHC(=O)OH、-NHC(=O)OEt、-NHC(=O)O(t-Bu)、-C(=O)NH 2、-C(=O)NHMe、-NHC(=O)NH 2、-NHC(=S)NH 2、-NHC(=NH)NH 2、-S(=O) 2Me、-NHS(=O) 2Me、-S(=O) 2OH、-S(=O) 2OMe、-NHS(=O) 2OH、-NHS(=O) 2OMe、-S(=O) 2NH 2、-NHS(=O) 2NH 2、环丙基、环丙基甲基、环丁基、环戊基、 环己基、四氢吡喃基、二氧六环基、哌啶基、哌嗪基、吗啉基、四氢呋喃基、四氢吡咯基、环氧丙基、环氧丁基、氮杂环丁基、二氢吡啶基、二氢吡喃基、苯基、苄基、吡啶基、嘧啶基、吡嗪基、哒嗪基、吡咯基、吡唑基、噻唑基、噁唑基、噁二唑基、咪唑基、噻二唑基、***基或四唑基;
其中,各R 2独立任选地被1、2、3或4个相同或不同的R 2a所取代;R 2a具有本发明所描述的含义。
在一些实施方案中,各R 4独立地为H、氘、卤素、羟基、氨基、C 1-6烷基、C 2-6烯基、C 2-6炔基、氘代C 1-6烷基、氘代C 1-6烷氧基、卤代C 1-6烷氧基、-(CR 7aR 7b) r-OR 8、-(CR 7aR 7b) r-NR 5aR 5b、-(CR 7aR 7b) rCN、或卤代C 1-6烷基;
其中,R 5a、R 5b、R 7a、R 7b、R 8和r均具有本发明所描述的含义。
在一些实施方案中,任意两个R 4和与它们相连的原子共同形成C 3-8环烷基、C 6-10芳基、3-8个原子组成的杂环基或5-6个原子组成的杂芳基;所述的C 3-8环烷基、C 6-10芳基、3-8个原子组成的杂环基和5-6个原子组成的杂芳基任选独立地被一个或多个R 10所取代;所述R 10具有本发明所描述的含义。
在一些实施方案中,各R 4独立地为H、氘、F、Cl、Br、I、羟基、氨基、C 1-4烷基、C 2-4烯基、C 2-4炔基、氘代C 1-4烷基、氘代C 1-4烷氧基、卤代C 1-4烷氧基、-(CR 7aR 7b) r-OR 8、-(CR 7aR 7b) r-NR 5aR 5b、-(CR 7aR 7b) rCN、或卤代C 1-4烷基;
其中,R 5a、R 5b、R 7a、R 7b、R 8和r均具有本发明所描述的含义。
在一些实施方案中,任意两个R 4和与它们相连的原子共同形成C 3-6环烷基、C 6芳基、3-6个原子组成的杂环基或5-6个原子组成的杂芳基;所述的C 3-6环烷基、C 6芳基、3-6个原子组成的杂环基和5-6个原子组成的杂芳基任选独立地被一个或多个R 10所取代;所述R 10具有本发明所描述的含义。
在一些实施方案中,各R 4独立地为H、氘、F、Cl、Br、I、羟基、氨基、甲基、乙基、丙基、丁基、三氟甲基、二氟甲基、1,2-二氟乙基、三氟甲氧基、二氟甲氧基、-OMe、-OEt、-O(t-Bu)、-CH 2OH、-CH 2CH 2OH、-CN、-CH 2CN、-CH 2NH 2、-CH 2CH 2NH 2、-CH 2OMe、-CH 2CH 2OMe、-NHMe或-N(Me) 2
在一些实施方案中,任意两个R 4和与它们相连的原子共同形成环丙基、环丁基、环戊基、环己基、环氧烷基、氧杂环丁基、四氢呋喃基、吡咯烷基、哌啶基、哌嗪基、吗啉基或氮杂环丁基;所述的环丙基、环丁基、环戊基、环己基、环氧烷基、氧杂环丁基、四氢呋喃基、吡咯烷基、哌啶基、哌嗪基、吗啉基和氮杂环丁基任选独立地被一个或多个R 10所取代;所述R 10具有本发明所描述的含义。
在一些实施方案中,各R 9、R 1a、R 2a和R 3a独立地为氧代(=O)、氢、氘、卤素、-(CR 7aR 7b) rCN、硝基、C 1-6烷基、卤代C 1-6烷基、C 2-6烯基、C 2-6炔基、R 8O-C 2-6亚烯基、R 8OC(=O)-C 2-6亚烯基、R 8O-C 2-6亚炔基、R 8OC(=O)-C 2-6亚炔基、-(CR 7aR 7b) r-OR 8、-(CR 7aR 7b) r-NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=O)R 6、-(CR 7aR 7b) rOC(=O)R 6、-(CR 7aR 7b) rCOOH、-(CR 7aR 7b) rC(=O)OC 1-6烷基、-(CR 7aR 7b) rC(=O)C 1-6烷基、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-12环烷基、-(CR 7aR 7b) r-C 6-12芳基、-(CR 7aR 7b) r-(3-12个原子组成的杂环基)或-(CR 7aR 7b) r-(5-12个原子组成的杂芳基);
或者,任意两个R 9与和它们相连的原子共同形成C 3-6环烷基或3-6个原子组成的杂环基;
其中,各R 9、R 1a、R 2a和R 3a独立任选地被1、2、3、4或5个相同或不同的R 10所取代;
R 5a、R 5b、R 5c、R 6、R 7a、R 7b、R 8、R 10和r均具有本发明所描述的含义。
在一些实施方案中,各R 9、R 1a、R 2a和R 3a独立地为氧代(=O)、氢、氘、F、Cl、Br、I、-(CR 7aR 7b) rCN、硝基、C 1-4烷基、卤代C 1-4烷基、C 2-4烯基、C 2-4炔基、R 8O-C 2-4亚烯基、R 8OC(=O)-C 2-4亚烯基、R 8O-C 2-4亚炔基、R 8OC(=O)-C 2-4亚炔基、-(CR 7aR 7b) r-OR 8、-(CR 7aR 7b) r-NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=O)R 6、-(CR 7aR 7b) rOC(=O)R 6、-(CR 7aR 7b) rCOOH、-(CR 7aR 7b) rC(=O)OC 1-4烷基、-(CR 7aR 7b) rC(=O)C 1-4烷基、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、 -(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-6环烷基、-(CR 7aR 7b) r-C 6-10芳基、-(CR 7aR 7b) r-(3-6个原子组成的杂环基)或-(CR 7aR 7b) r-(5-6个原子组成的杂芳基);
或者,任意两个R 9与和它们相连的原子共同形成C 3-6环烷基或3-6个原子组成的杂环基;
其中,各R 9、R 1a、R 2a和R 3a独立任选地被1、2、3、4或5个相同或不同的R 10所取代;
R 5a、R 5b、R 5c、R 6、R 7a、R 7b、R 8、R 10和r均具有本发明所描述的含义。
在一些实施方案中,各R 9、R 1a、R 2a和R 3a独立地为氧代(=O)、氢、氘、F、Cl、Br、I、-CN、甲基、乙基、丙基、丁基、三氟甲基、二氟甲基、2,2-二氟乙基、-CH 2CN、-CH 2CH 2CN、乙烯基、丙烯基、烯丙基、乙炔基、丙炔基、-OH、-OMe、-O(i-Pr)、-O(t-Bu)、-CH 2-OH、-CH 2CH 2-OH、-CH 2-OMe、-CH 2CH 2-OMe、-CH 2-O(i-Pr)、-CH 2-O(t-Bu)、-NH 2、-NHMe、-N(Me) 2、-CH 2-NH 2、-CH 2CH 2-NH 2、-CH 2-NHMe、-CH 2-N(Me) 2、-NHC(=O)Me、-CH 2COOH、-COOH、-C(=O)OMe、-CH 2C(=O)OMe、-C(=O)Me、-CH 2C(=O)Me、环丙基、环丁基、环戊基、环己基、环氧烷基、环氧丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基、四氢吡喃基、二氢吡喃基、苯基、苄基、吡啶基、嘧啶基、吡嗪基、呋喃基、噻吩基、吡咯基、吡唑基、咪唑基、噁唑基、噻唑基、噁二唑基、噻二唑基、***基或四唑基;
或者,任意两个R 9与和它们相连的原子共同形成环丙基、环丁基、环戊基、环己基、环氧烷基、环氧丁基、氮杂环丁基、四氢呋喃基、吡咯烷基、哌啶基或哌嗪基;
其中,各R 9、R 1a、R 2a和R 3a独立任选地被1、2、3、4或5个相同或不同的R 10所取代;所述R 10具有本发明所描述的含义。
在一些实施方案中,各R 5、R 5a、R 5b、R 5c和R 5d独立地为氢、氰基、羟基、C 1-6烷基、C 1-6烷氧基-C(=O)-、C 6-10芳基、C 3-10环烷基、3-10个原子组成的杂环基或5-10个原子组成的杂芳基。
在一些实施方案中,各R 5、R 5a、R 5b、R 5c和R 5d独立地为氢、氰基、羟基、C 1-4烷基、C 1-4烷氧基-C(=O)-、C 6-10芳基、C 3-6环烷基、3-6个原子组成的杂环基或5-6个原子组成的杂芳基。
在一些实施方案中,各R 5、R 5a、R 5b、R 5c和R 5d独立地为氢、氰基、羟基、甲基、乙基、丙基、丁基、甲氧基-C(=O)-、乙氧基-C(=O)-、异丙氧基-C(=O)-、叔丁氧基-C(=O)-、苯基、环丙基、环丁基、环戊基、环己基、环氧烷基、环氧丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基、四氢吡喃基、二氢吡喃基、苯基、苄基、吡啶基、嘧啶基、吡嗪基、呋喃基、噻吩基、吡咯基、吡唑基、咪唑基、噁唑基、噻唑基、噁二唑基、噻二唑基、***基或四唑基。
在一些实施方案中,R 5、R 9和与它们相连的原子共同形成3-8个原子组成的杂环基。
在另一些实施方案中,R 5、R 9和与它们相连的原子共同形成3-6个原子组成的杂环基。
在一些实施方案中,各R 6和R 8独立地为氢、氘、C 1-6烷基、氘代C 1-6烷基、C 2-6烯基、C 2-6炔基、羟基取代的C 1-6烷基、氨基取代的C 1-6烷基、氰基取代的C 1-6烷基、C 1-6烷氧基C 1-6烷基、卤代C 1-6烷基、C 6-10芳基、C 3-10环烷基、3-10个原子组成的杂环基或5-10个原子组成的杂芳基。
在一些实施方案中,各R 6和R 8独立地为氢、氘、C 1-4烷基、氘代C 1-4烷基、C 2-4烯基、C 2-4炔基、羟基取代的C 1-4烷基、氨基取代的C 1-4烷基、氰基取代的C 1-4烷基、C 1-4烷氧基C 1-4烷基、卤代C 1-4烷基、C 6-10芳基、C 3-6环烷基、3-6个原子组成的杂环基或5-6个原子组成的杂芳基。
在另一些实施方案中,各R 6和R 8独立地为氢、氘、甲基、乙基、丙基、丁基、氘代甲基、乙烯基、丙炔基、羟基甲基、羟基乙基、甲氧基甲基、甲氧基乙基、乙氧基甲基、叔丁氧基甲基、叔丁氧基乙基、乙氧基乙基、异丙氧基乙基、三氟甲基、二氟甲基、1,2-二氟乙基、2,2-二氟乙基、苯基、环丙基、环己基、氮杂环丁基、四氢呋喃基、吡咯烷基、哌啶基、哌嗪基、吡啶基、吡咯基、嘧啶基、吡唑基、吡嗪基、呋喃基、噻唑基、噁唑基、噁二唑基、噻二唑基、咪唑基、***基或四唑基。
在一些实施方案中,各R 7、R 7a和R 7b独立地为氢、C 1-6烷基、氘代C 1-6烷基、C 2-6烯基、C 2-6炔基或 卤代C 1-6烷基。
在一些实施方案中,各R 7、R 7a和R 7b独立地为氢、C 1-4烷基、氘代C 1-4烷基或卤代C 1-4烷基。
在另一些实施方案中,各R 7、R 7a和R 7b独立地为氢、甲基、乙基、丙基、丁基、氘代甲基、三氟甲基、二氟甲基、1,2-二氟乙基或2,2-二氟乙基。
在一些实施方案中,各R 10独立地为氧代(=O)、H、D、卤素、CN、氨基、羟基、硝基、羧基、C 1-4烷基、C 2-4烯基、C 2-4炔基、氘代C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4烷氧基C 1-4烷基、羟基取代的C 1-4烷基、氰基取代的C 1-4烷基、氨基取代的C 1-4烷基、卤代C 1-4烷基、C 1-4烷基酰基、C 1-4烷基磺酰基、C 1-4烷氧基酰基、C 1-4烷氨基酰基、氨基酰基、氨基磺酰基、C 1-4烷氧基磺酰基、C 1-4烷氨基磺酰基、C 1-4酰氧基、C 6-10芳基、C 3-6环烷基、3-6个原子组成的杂环基或5-10个原子组成的杂芳基。
在一些实施方案中,各R 10独立地为氧代(=O)、H、D、F、Cl、Br、I、CN、氨基、羟基、硝基、羧基、C 1-3烷基、C 2-4烯基、C 2-4炔基、氘代C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4烷氧基C 1-4烷基、羟基取代的C 1-4烷基、氰基取代的C 1-4烷基、氨基取代的C 1-4烷基、卤代C 1-4烷基、C 1-4烷基酰基、C 1-4烷基磺酰基、C 1-4烷氧基酰基、C 1-4烷氨基酰基、苯基、C 3-6环烷基、3-6个原子组成的杂环基或5-6个原子组成的杂芳基。
在另一些实施方案中,各R 10独立地为氧代(=O)、H、D、F、Cl、Br、I、CN、氨基、羟基、硝基、羧基、甲基、乙基、丙基、丁基、氘代甲基、甲氧基、甲氨基、二甲氨基、甲氧基甲基、甲氧基乙基、叔丁氧基甲基、叔丁氧基乙基、羟基甲基、羟基乙基、CN取代的甲基、CN取代的乙基、氨基甲基、氨基乙基、三氟甲基、二氟甲基、1,2-二氟乙基或2,2-二氟乙基、甲基酰基、乙基酰基、甲基磺酰基、甲氧基酰基、叔丁氧基酰基、甲氨基酰基、二甲氨基酰基、苯基、环丙基、环丁基、环戊基、环己基、环氧丙烷基、环氧丁基、氮杂环丁基、四氢呋喃基、吡咯烷基、哌啶基、哌嗪基、吗啉基、呋喃基、噻吩基、吡咯基、吡唑基、咪唑基、噁二唑基、***基、四唑基、吡啶基、嘧啶基或吡嗪基。
在一些实施方案中,各m、n和t独立地为0、1、2、3或4。
在一些实施方案中,p为1、2、3或4。
在一些实施方案中,各r独立地为0、1、2、3或4。
在一些实施方案中,f为0、1、2、3、4或5;
在一些实施方案中,g为0、1、2、3、4、5、6、7或8。
本发明涉及一种化合物,其为以下其中之一的结构:
Figure PCTCN2018072988-appb-000123
Figure PCTCN2018072988-appb-000124
Figure PCTCN2018072988-appb-000125
或上述结构的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药。
一方面,本发明涉及一种药物组合物,其包含本发明任一所述的化合物。在一些实施方案中,本发明所述的药物组合物进一步包含药学上可接受的载体、赋形剂、稀释剂、辅剂和媒介物的至少一种。
另一方面,本发明涉及所述的化合物或药物组合物在制备药物中的用途,其中,所述的药物用于预防、治疗或减轻血栓栓塞性疾病。
一方面,本发明所述的化合物或药物组合物用于预防、治疗或减轻血栓栓塞性疾病。
另一方面,本发明还涉及一种使用本发明所述的化合物或药物组合物预防、治疗或减轻血栓栓塞性疾病的方法。
在一些实施方案中,本发明所述的血栓栓塞性疾病为动脉心血管血栓栓塞性疾病、静脉心血管血栓栓塞性疾病、及心室或外周循环中的血栓栓塞性疾病。
在另一些实施方案中,所述血栓栓塞性疾病为心绞痛、急性冠状动脉综合征、心房颤动、心肌梗塞、短暂性缺血发作、中风、动脉粥样硬化、外周闭塞性动脉疾病、静脉血栓形成、深静脉血栓形成、血栓性静脉炎、动脉栓塞、冠状动脉血栓形成、脑动脉血栓形成、脑栓塞、肾栓塞、肺栓塞、以及因医疗植入物、器械或程序中的血液暴露于人造表面从而促使血栓形成而引起的血栓形成。
一方面,本发明涉及所述的化合物或药物组合物在制备药物中的用途,其中,所述的药物用于治疗弥散性血管内凝血(DIC)疾病。
另一方面,本发明涉及所述的化合物或药物组合物在制备药物中的用途,其中,所述的药物用于抑制 因子XIa和/或血浆激肽释放酶的活性。
一方面,本发明所述的化合物或药物组合物用于抑制因子XIa和/或血浆激肽释放酶的活性。
另一方面,本发明还涉及一种使用本发明所述的化合物或药物组合物抑制因子XIa和/或血浆激肽释放酶的活性的方法。
本发明包含所述的化合物及其药学上可接受的盐的应用,用于生产医药产品治疗患者血栓栓塞性疾病,包括那些本发明所描述的疾病。本发明包含药物组合物,该药物组合物包括本发明所述的化合物与至少一种药学上可接受的载体,赋形剂,稀释剂,辅剂,媒介物的结合所需的有效治疗量。
本发明同样包含治疗或减轻患者血栓栓塞性疾病,或对此病症敏感的方法,该方法包含使用本发明化合物的治疗有效量对患者进行治疗。
本发明所述血栓栓塞性疾病包括心肌梗塞、心绞痛、再阻塞和血管形成术或主动脉冠状功脉分流术后的再狭窄、中风、短暂的局部缺血发作、周围动脉闭塞性疾病、肺栓塞或深静脉血栓形成。
除非其他方面表明,本发明化合物所有的立体异构体,几何异构体,互变异构体,氮氧化物,水合物,溶剂化物,代谢产物,盐和药学上可接受的前药都属于本发明的范围。
具体地说,盐是药学上可接受的盐。术语“药学上可接受的”包括物质或组合物必须是适合化学或毒理学地,与组成制剂的其他组分和用于治疗的哺乳动物有关。
本发明化合物的盐还包括用于制备或纯化本发明化合物的中间体或本发明化合物分离的对映异构体的盐,但不一定是药学上可接受的盐。
如果本发明的化合物是碱性的,则想得到的盐可以通过文献上提供的任何合适的方法制备得到,例如,使用无机酸,如盐酸,氢溴酸,硫酸,硝酸和磷酸等等。或者使用有机酸,如乙酸,三氟乙酸,马来酸,琥珀酸,扁桃酸,富马酸,丙二酸,丙酮酸,草酸,羟乙酸和水杨酸;吡喃糖酸,如葡萄糖醛酸和半乳糖醛酸;α-羟酸,如柠檬酸和酒石酸;氨基酸,如天门冬氨酸和谷氨酸;芳香族酸,如苯甲酸和肉桂酸;磺酸,如对甲苯磺酸,乙磺酸,等等。
如果本发明的化合物是酸性的,则想得到的盐可以通过合适的方法制备得到,如,使用无机碱或有机碱,如氨(伯氨,仲氨,叔氨),碱金属氢氧化物或碱土金属氢氧化物,等等。合适的盐包括,但并不限于,从氨基酸得到的有机盐,如甘氨酸和精氨酸,氨,如伯氨、仲氨和叔氨,和环状氨,如哌啶,吗啉和哌嗪等,和从钠,钙,钾,镁,锰,铁,铜,锌,铝和锂得到无机盐。
本发明化合物的药物组合物,制剂,给药和用途
根据另一方面,本发明的药物组合物的特点包括式(I)或式(Ia)或式(II)或式(IIa)或式(III)或式(IIIa)或式(IIIb)或式(IIIc)所示的化合物,本发明所列出的化合物,或实施例1-11的化合物,和药学上可接受的载体,辅剂,或赋形剂。本发明的组合物中化合物的量能有效地治疗或减轻患者血栓栓塞性疾病,或有效抑制因子XIa和/或血浆激肽释放酶的活性。
在一些实施方案中,本发明所述的药物组合物,其包括本发明所述的任一化合物或其立体异构体、互变异构体、药学上可接受的盐或溶剂化合物;在另一些实施方案中,本发明所述的药物组合物包括本发明所述的任一化合物它们的任意组合。
在一些实施方案中,本发明所述的药物组合物进一步包含其他治疗剂。其中,所述的其他治疗剂选自抗心律不齐剂、抗高血压剂、抗凝血剂、抗血小板剂、凝血酶抑制剂、血栓溶解剂、纤维蛋白溶解剂、钙通道阻断剂、钾通道阻断剂、胆固醇/脂质降低剂或它们的组合。
在另一些实施方案中,本发明提供的药物组合物,其中,其他治疗剂为抗高血压剂(其选自ACE抑制剂、AT-1受体拮抗剂、β肾上腺素能受体拮抗剂、ETA受体拮抗剂、双重ETA/AT-1受体拮抗剂、肾素抑制剂(阿斯科因(alliskerin))及血管胜肽酶抑制剂)、抗心律不齐剂(其选自IKur抑制剂)、抗凝血剂(其选 自凝血酶抑制剂、抗凝血酶-III活化剂、肝素辅因子II活化剂、其他因子XIa抑制剂、其他激肽释放酶抑制剂、纤维蛋白溶酶原活化剂抑制剂(PAI-1)拮抗剂、凝血酶可活化纤维蛋白溶解抑制剂(TAFI)抑制剂、因子VIIa抑制剂、因子IXa抑制剂及因子Xa抑制剂)或抗血小板剂(选自GPIIb/IIIa阻断剂、GP Ib/IX阻断剂、蛋白酶活化受体1(PAR-1)拮抗剂、蛋白酶活化受体4(PAR-4)拮抗剂、***素E2受体EP3拮抗剂、胶原受体拮抗剂、磷酸二酯酶-III抑制剂、P2Y受体拮抗剂、P2Y 12拮抗剂、凝血脂素受体拮抗剂、环氧合酶-1抑制剂及阿司匹林)或它们的任意组合。
在另一些实施方案中,本发明所述的药物组合物包含的其他治疗剂为抗血小板剂或其组合。其中,所述的抗血小板剂包括但不限于氯吡格雷和/或阿司匹林或它们的组合。在又一些实施方案中,本发明所述的药物组合物包含的其他治疗剂为华法林、未分段肝素、低分子量肝素、合成五糖、水蛭素、阿加曲班、阿司匹林、布洛芬、萘普生、舒林酸、吲哚美辛、美非玛特、双嘧达莫(dipyridamol)、屈昔康、双氯芬酸、磺吡酮、吡罗昔康、噻氯匹定、氯吡格雷、替罗非班、依替巴肽、阿昔单抗、美拉加群、希美加群(ximelagatran)、去硫酸水蛭素、组织纤维蛋白溶酶原活化剂、经修饰组织纤维蛋白溶酶原活化剂、阿尼普酶、尿激酶及链激酶或它们的组合。
本发明的化合物存在自由形态,或合适的、作为药学上可接受的衍生物。根据本发明,药学上可接受的衍生物包括,但并不限于,药学上可接受的前药,盐,酯,酯类的盐,或能直接或间接地根据患者的需要给药的其他任何加合物或衍生物,本发明其他方面所描述的化合物,其代谢产物或它的残留物。
像本发明所描述的,本发明药学上可接受的组合物进一步包含药学上可接受的载体,辅剂,或赋形剂,这些像本发明所应用的,包括任何溶剂,稀释剂,或其他液体赋形剂,分散剂或悬浮剂,表面活性剂,等渗剂,增稠剂,乳化剂,防腐剂,固体粘合剂或润滑剂,等等,适合于特有的目标剂型。如以下文献所描述的:In Remington:The Science and Practice of Pharmacy,21st edition,2005,ed.D.B.Troy,Lippincott Williams&Wilkins,Philadelphia,and Encyclopedia of Pharmaceutical Technology,eds.J.Swarbrick and J.C.Boylan,1988-1999,Marcel Dekker,New York,综合此处文献的内容,表明不同的载体可应用于药学上可接受的组合物的制剂和它们公知的制备方法。除了任何常规的载体媒介与本发明的化合物不相容的范围,例如所产生的任何不良的生物效应或与药学上可接受的组合物的任何其他组分以有害的方式产生的相互作用,它们的用途也是本发明所考虑的范围。
可作为药学上可接受载体的物质包括,但并不限于,离子交换剂,铝,硬脂酸铝,卵磷脂,血清蛋白,如人血清蛋白,缓冲物质如磷酸盐,甘氨酸,山梨酸,山梨酸钾,饱和植物脂肪酸的部分甘油酯混合物,水,盐或电解质,如硫酸鱼精蛋白,磷酸氢二钠,磷酸氢钾,氯化钠,锌盐,胶体硅,三硅酸镁,聚乙烯吡咯烷酮,聚丙烯酸脂,蜡,聚乙烯-聚氧丙烯-阻断聚合体,羊毛脂,糖,如乳糖,葡萄糖和蔗糖;淀粉如玉米淀粉和土豆淀粉;纤维素和它的衍生物如羧甲基纤维素钠,乙基纤维素和乙酸纤维素;树胶粉;麦芽;明胶;滑石粉;辅料如可可豆脂和栓剂蜡状物;油如花生油,棉子油,红花油,麻油,橄榄油,玉米油和豆油;二醇类化合物,如丙二醇和聚乙二醇;酯类如乙基油酸酯和乙基月桂酸酯;琼脂;缓冲剂如氢氧化镁和氢氧化铝;海藻酸;无热原的水;等渗盐;林格(氏)溶液;乙醇,磷酸缓冲溶液,和其他无毒的合适的润滑剂如月桂硫酸钠和硬脂酸镁,着色剂,释放剂,包衣衣料,甜味剂,调味剂和香料,防腐剂和抗氧化剂。
本发明化合物可以以口服剂的形式被施用,如片剂,胶囊(其中的每一个都包括持续释放或者定时释放的配方),丸剂,粉剂,粒剂,酏剂,酊剂,悬浮剂,糖浆剂,和乳化剂。它们也可以以静脉内(大丸剂或者输液),腹膜内,皮下或者肌肉内的形式施用。所有使用的剂量形式都是药学领域的普通技术人员所熟知的。它们可以单独施用,但一般将基于所选择的施用方式和标准的药学实践选择一种药学载体一起施用。
本发明化合物的给药方案将随已知的各种因素而不同,如特定试剂的药动学特征及其模式和施用途 径;接受者的种族,年龄,性别,健康状况,医疗状况和体重;症状的性质和程度;并行的治疗的种类;治疗的频率;施药的途径,病人的肾和肝功能,和希望达到的效果。一个医师或者兽医可以作出决定并开出有效量的药物来预防、抵销或者阻止血栓栓塞疾病的发展。
根据一般的指导原则,为了达到指定的效果,所使用的每一种活性成分的日口服剂量的范围为大约0.001到1000mg/kg体重之间,优选地,在大约0.01到100mg/kg体重之间。而且,最优选地,在大约1.0到20mg/kg体重/天之间。对于静脉内的施用,在常规速率的输液过程中最优选的剂量范围为大约1到大约10mg/kg体重/分钟。本发明化合物可以以每日一次来施用,或者可以以每日分两次,三次或者四次进行施用。
本发明的化合物可以经过合适的鼻内载体的局部使用以鼻内形式施用,或者通过使用经皮药贴以经皮途径施用。当以经皮传递***的形式施用时,在整个用药期间施用的剂量是连续的而不是间歇的。
典型地,该化合物与根据施用的形式和常规的药学实践来选择的合适的药物稀释剂,赋形剂,或者载体(在此是指药物载剂)混和施用,施用方式可以是口服片剂,胶囊,酏剂,糖浆等等。
例如,对于以片剂或者胶囊形式口服施用,活性药物组分可以和一种口服的、非毒性的、药学上可接受的惰性载剂结合,如乳糖,淀粉,蔗糖,葡萄糖、甲基纤维素,硬脂酸镁,磷酸二钙,硫酸钙,甘露醇,山梨醇等等;对于以液体形式口服施用,口服药物组分可以和任何口服的、非毒性的、药学上可以接受的惰性载剂结合,如乙醇,甘油,水等等。而且,当需要或必需时,合适的粘合剂、滑润剂、分解试剂以及着色剂也可以加入到混合物中。合适的粘合剂包括淀粉、明胶、天然糖(如葡萄糖或者β-乳糖),玉米甜味剂,天然的和合成的树胶(如***胶),黄芪胶,或者藻酸钠,羧甲基纤维素,聚乙烯乙二醇,蜡等等。在这些剂型中应用的润滑剂包括油酸钠,硬脂酸钠,硬脂酸镁,苯甲酸钠,乙酸钠,氯化钠等等。分解剂包括,但不限于,淀粉,甲基纤维素,琼脂,膨润土,黄原胶,等等。
本发明化合物也可以以脂质体传递***的形式施用,如小的单层的囊泡,大的单层的囊泡以及多层囊泡。脂质体可以通过不同的磷脂形成,如胆固醇,硬脂胺,或者磷脂酰胆碱。
本发明化合物也与可溶性的聚合物偶联,该多聚物作为靶向的药物载剂。这样的多聚物包括聚乙烯吡咯烷酮,吡喃共聚物,聚羟基丙基甲基丙烯酸胺-酚,聚羟基乙基天冬酰胺酚,或者用棕榈酰残基取代的聚乙烯氧化物-聚赖氨酸。而且,本发明化合物可以与一类生物可降解的聚合物偶联,用于完成可控制的药物释放,例如,聚乳酸,聚羟基乙酸,聚乳酸和聚羟基乙酸的共聚物,聚ε己内酯,聚羟基丁酸,聚原酸酯,聚缩醛,聚二氢吡喃,聚氰基丙烯酸酯,和水凝胶的交联的或者两亲性的阻断共聚物。
适于施用的剂型(药物组合物)的每一单位剂量,可以含有大约1mg到大约100mg的活性成分。在这些药物组合物中,活性成分的重量一般将占组合物的总重量的大约0.5-95%。
明胶胶囊可以含有活性成分以及粉末载剂,如乳糖,淀粉,纤维素衍生物,硬脂酸镁,硬脂酸,等等。可以使用类似的稀释剂制作压缩片剂。可以制造片剂和胶囊作为可持续释放的产物来提供在一段时间内连续释放的药物。压缩的片剂可以加糖衣或者包上一层薄膜来掩盖任何不愉快的味道而且使片剂与空气隔绝,或者加上肠溶性的包被以用于在肠胃消化道中选择性地分解。
口服施用的液体剂型可以含有着色剂和调味料以提高病人的接受度。
通常,水,一种合适的油,盐水,水合的右旋糖(葡萄糖),和相关的糖溶液以及二醇(如丙二醇或者聚乙二醇)是不经肠道的溶液的合适载剂。不经肠道施用的溶液优选含有活性成分的水溶性盐,合适的稳定剂,以及可能必要的缓冲液物质。抗氧化剂是合适的稳定剂,如亚硫酸氢钠,亚硫酸钠,或者维生素C,既可以单独也可以组合使用,也可以用柠檬酸和其盐以及EDTA钠盐。此外,不经肠道的溶液也含有防腐剂,如洁尔灭,甲基-或者丙基-对羟基苯甲酸酯,和氯代丁醇。
本发明的化合物可以与其它的抗凝试剂组合,例如,对于每千克病人体重,一种日剂量可以是大约0.1 到100mg的本发明化合物和大约1到7.5mg的第二抗凝剂组合。对于一种片剂剂型,本发明的化合物一般可以是每个剂量单位有大约5到10mg,而且第二抗凝集剂的量是每个剂量单位有大约从1到5mg。其中,其它的抗凝试剂具体包括,但不限于,阿哌沙班(apixaban)、利伐沙班(rivaroxaban)、依度沙班(edoxaban)、贝曲西班(betrixaban)、达比加群(dabigatran)、贝米肝素、依诺肝素钠、亭扎肝素钠、达那肝素钠、戊聚糖钠、那屈肝素钙、阿地肝素钠、帕肝素钠等等。
本发明的化合物可以单独、或与其他治疗剂同时或依序的联合使用。其中,所述的其他治疗剂选自因子Xa抑制剂(例如,阿哌沙班、利伐沙班、贝曲西班、依度沙班)、抗凝血剂、抗血小板剂、凝血酶抑制剂(例如,达比加群)、血栓溶解剂及纤维蛋白溶解剂。优选地,其他治疗剂为至少一种选自以下之药剂:华法林、未分段肝素、低分子量肝素、合成五糖、水蛭素、阿加曲班(argatroban)、阿司匹林、布洛芬(ibuprofen)、萘普生(naproxen)、舒林酸(sulindac)、吲哚美辛(indomethacin)、美非玛特(mefenamate)、屈昔康(droxicam)、双氯芬酸(diclofenac)、磺吡酮(sulfinpyrazone)、吡罗昔康(piroxicam)、噻氯匹定(ticlopidine)、氯吡格雷、替罗非班(tirofiban)、依替巴肽(eptifibatide)、阿昔单抗(abciximab)、美拉加群(melagatran)、去硫酸水蛭素(desulfatohirudin)、组织纤维蛋白溶酶原活化剂、经修饰组织纤维蛋白溶酶原活化剂、阿尼普酶(anistreplase)、尿激酶(urokinase)及链激酶(streptokinase)。优选地,其他治疗剂为至少一种抗血小板剂。优选地,抗血小板剂为氯吡格雷和/或阿司匹林或其组合
根据一般的指导原则,本发明化合物与一种抗血小板试剂组合施用,一般的日剂量可以是每公斤病人体重大约0.01到300mg的本发明化合物和大约50到150mg的抗血小板试剂,优选大约0.1到4mg的本发明化合物和大约1到3mg的抗血小板试剂。
当本发明化合物与溶栓剂组合施用时,一般的日剂量可以是每公斤病人体重大约0.1到100mg的本发明化合物,而且在溶栓剂存在的条件下,与溶栓剂单独施用时的一般剂量相比,当溶栓剂与本发明化合物一起施用时,溶栓剂的剂量可以降低大约50-80%。
当两个或者多个前述的第二治疗剂与本发明化合物一起施用时,一般地,考虑到联合施用时治疗剂的附加的或者协同的效果,在典型的日剂量和典型的剂型中的每一个组分的量,相对于单独施用时的通常剂量,可以有所下降。
特别地,当作为一个单一的剂量单位提供时,存在着组合的活性成分之间发生化学反应的可能性。由于这一原因,当本发明的化合物和第二治疗剂在一个单一的剂量单位中被联合时,它们的配制方法要使活性成分间的物理接触最小化(即是,减少),尽管活性成分组合在一个单一的剂量单位内。例如,一种活性成分可以是肠溶衣包被。通过肠溶衣包被一种活性成分,有可能不仅仅使联合的活性成分间的接触最小化,而且还有可能控制这些成分中的一种在胃肠道中的释放以便这些组分的一种并不在胃中释放而在小肠中释放。活性成分的一种也可以包裹上影响其在胃肠道中的持续释放而且也可用于减少联合的活性成分间的物理接触的材料进一步,持续释放的组分也可以额外地用肠溶衣包被以便于这一成分只在肠道中发生释放。还有另一个方法涉及联合产物的配方,其中的一个组分用一种持续的和/或肠溶释放的聚合物包被,而且另一个组分也用多聚物如一种低粘性级别的羟基丙基甲基纤维素(HPMC)或者其它的合适的在该领域内已知的材料包被,以达到进一步分离活性成分的目的。聚合物包被对与其它组分的反应形成了一种额外的阻碍。
一旦了解本发明内容,这些以及其它的使本发明的联合产物的组分间的接触最小化的方法对于本领域技术人员是很明显的,无论它们是以单一剂型施用或者以分离的形式施用,但是是在相同的时间或以相同的方式施用。
本发明涉及的化合物或者其药用盐或其水合物能有效用于预防、治疗或减轻患者血栓栓塞性疾病;所述血栓栓塞性病症包含动脉心血管血栓栓塞性病症、静脉心血管血栓栓塞性病症、动脉脑血管血栓栓塞性 病症及静脉脑血管血栓栓塞性病症。血栓栓塞性病症的实例包含,但不限于,不稳定型心绞痛、急性冠状动脉症候群、心房颤动、首次心肌梗塞、复发性心肌梗塞、缺血性猝死、短暂性缺血发作、中风、动脉粥样硬化、周边闭塞性动脉疾病、静脉血栓形成、深静脉血栓形成、血栓性静脉炎、动脉栓塞、冠状动脉血栓形成、脑动脉血栓形成、脑栓塞、肾栓塞、肺栓塞及因医疗植入物、装置或程序中的血液暴露于人造表面从而促使血栓形成而引起的血栓形成。
本发明提供的化合物或药物组合物还可以用于治疗、预防或治疗炎性疾病;所述炎性疾病包括但不限于败血症、急性呼吸窘迫症候群或全身性发炎反应症候群。
本发明提供的化合物或药物组合物可以用于预防、治疗或减轻与血浆激肽释放酶相关的疾病;其中,所述与血浆激肽释放酶相关的疾病包括,但不限于,视觉敏锐度受损、糖尿病性视网膜病、糖尿病性黄斑部水肿、遗传性血管水肿、糖尿病、胰脏炎、肾病、心肌病、神经病变、发炎性肠病、关节炎、发炎、败血性休克、低血压、癌症、成人呼吸窘迫症候群、散播性血管内凝血及心肺搭桥术。
一般合成过程
在本说明书中,如果在化学名称和化学结构间存在任何差异,结构是占优的。一般地,本发明的化合物可以通过本发明所描述的方法制备得到,除非有进一步的说明,其中取代基的定义如(I)或式(Ia)或式(II)或式(IIa)或式(III)或式(IIIa)或式(IIIb)或式(IIIc)所示。下面的反应方案和实施例1-11用于进一步举例说明本发明的内容。
所属领域的技术人员将认识到:本发明所描述的化学反应可以用来合适地制备许多本发明的其他化合物,且用于制备本发明的化合物的其它方法都被认为是在本发明的范围之内。例如,根据本发明那些非例证的化合物的合成可以成功地被所属领域的技术人员通过修饰方法完成,如适当的保护干扰基团,通过利用其他已知的试剂(除了本发明所描述的),或将反应条件做一些常规的修改。另外,本发明所公开的反应或已知的反应条件也公认地适用于本发明其他化合物的制备。
下面所描述的实施例,除非其他方面表明,所有的温度定为摄氏度。试剂购买于商品供应商如Aldrich Chemical Company,Arco Chemical Company和Alfa Chemical Company,使用时都没有经过进一步纯化,除非其他方面表明。一般的试剂从汕头西陇化工厂,广东光华化学试剂厂,广州化学试剂厂,天津好寓宇化学品有限公司,青岛腾龙化学试剂有限公司,和青岛海洋化工厂购买得到。
无水四氢呋喃,二氧六环,甲苯,***是经过金属钠回流干燥得到。无水二氯甲烷和氯仿是经过氢化钙回流干燥得到。乙酸乙酯,石油醚,正己烷,N,N-二甲基乙酰胺和N,N-二甲基甲酰胺是经无水硫酸钠事先干燥使用。
以下反应一般是在氮气或氩气正压下或在无水溶剂上套一干燥管(除非其他方面表明),反应瓶都塞上合适的橡皮塞,底物通过注射器打入。玻璃器皿均是经过干燥的。
色谱柱是使用硅胶柱。硅胶(300-400目)购于青岛海洋化工厂。核磁共振光谱数据通过Bruker Avance400核磁共振谱仪或Bruker Avance III HD 600核磁共振谱仪来测定,以CDC1 3,DMSO-d 6,CD 3OD或丙酮-d 6为溶剂(报导以ppm为单位),用TMS(0ppm)或氯仿(7.25ppm)作为参照标准。当出现多重峰的时候,将使用下面的缩写:s(singlet,单峰),d(doublet,双峰),t(triplet,三重峰),m(multiplet,多重峰),br(broadened,宽峰),dd(doublet of doublets,四重峰),dt(doublet of triplets,双三重峰),ddd(doublet of doublet of doublets,双双二重峰),ddt(doublet of doublet of triplets,双双三重峰),dddd(doublet of doublet of doublet of doublets,双双双二重峰)。偶合常数,用赫兹(Hz)表示。
低分辨率质谱(MS)数据测定的条件是:Agilent 6120Quadrupole HPLC-MS(柱子型号:Zorbax SB-C18,2.1x 30mm,3.5μm,6min,流速为0.6mL/min,流动相:5%-95%(含0.1%甲酸的CH 3CN)在(含0.1%甲酸的H 2O)中的比例)),在210/254nm用UV检测,用电喷雾电离模式(ESI)。
化合物纯度的表征方式为:Agilent 1260制备型高效液相色谱(Pre-HPLC)或Calesep Pump 250制备型高效液相色谱(Pre-HPLC)(柱子型号:NOVASEP,50/80mm,DAC),在210nm/254nm用UV检测。
下面简写词的使用贯穿本发明:
Grubbs 2代催化剂  格拉布第二代催化剂,苯亚甲基-1,3-双(2,4,6-三甲苯基)-2-(咪唑啉卡宾)(三环己基磷)二氯化钌
(dba) 3Pd 2,(dba) 3Pd(0) 2  三(二亚苄基茚丙酮)二钯(0)
Pd(dppf)Cl 2-CH 2Cl 2  1,1-双(二苯基膦)二茂铁二氯化钯二氯甲烷络合物
Dess-Martin氧化剂  戴斯-马丁氧化剂,(1,1,1-三乙酰氧基)-1,1-二氢-1,2-苯碘酰-3(1H)-酮
Boc  叔丁氧羰基
Boc酸酐  二碳酸二叔丁酯
EDTA-2Na  乙二胺四乙酸二钠
SEM  2-(三甲基硅烷基)乙氧甲基
PtO 2-3H 2O  二氧化铂(三水)
CDC1 3  氘代氯仿
CD 3OD  氘代甲醇
DMSO-d 6  氘代二甲基亚砜
DMSO  二甲基亚砜
g  克
mg  毫克
mol  摩尔
mmol  毫摩尔
mL  毫升
μL  微升
MPa  兆帕
mass%  质量百分含量或者质量分数或者质量百分比
Pd/C  钯/碳
FXIa  因子XIa
HEPES  4-羟乙基哌嗪乙磺酸
NaCl  氯化钠
KCl  氯化钾
PEG8000  聚乙二醇8000
PT  凝血酶原时间
APTT  部分活化凝血酶原时间
下列反应方案描述了制备本发明公开化合物的步骤。其中,A、B、X、Y、Z、Z 1、R 1、R 2、R 3、R 4、R 3a、m、n、t、f具有本发明所描述的含义;L为Cl、Br或I;PG代表保护基团,合适的保护基团可以参考文献:T W.Greene,Protective Groups in Organic Synthesis,John Wiley&Sons,New York,1991;and P.J.Kocienski,Protecting Groups,Thieme,Stuttgart,2005。
合成方案1
Figure PCTCN2018072988-appb-000126
化合物1c可以通过合成方案1所描述的方法制备得到。化合物1a和1b在2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(HATU)和1,8-二氮杂二环十一碳-7-烯(DBU)的存在下,在溶剂(如乙腈)中反应,得到嘧啶酮类化合物1c。
合成方案2
Figure PCTCN2018072988-appb-000127
化合物2f可以通过合成方案2描述的方法制备得到。化合物2a先与化合物2c在溶剂(如四氢呋喃等)中发生麦克尔(Michael)加成反应,然后再在碱性条件下(例如,在吡啶或4-二甲氨基吡啶的作用下)被化合物2d乙酰化,得到化合物2e。化合物2e在碱性条件下(如甲醇钠或氢化钠等的作用下)关环得到化合物2f。
其中,中间体化合物2e还可以通过以下方法制备得到:化合物2a先在三甲基氯硅烷(TMSCl)和碘化钠(NaI)的存在下、在溶剂(例如,乙腈和水)中发生加成反应得到化合物2b;化合物2b与化合物2c在碱性条件下(例如,在三乙胺的作用下)发生取代反应,然后再在碱性条件下(例如,在吡啶或4-二甲氨基吡啶的作用下)被化合物2d乙酰化,得到化合物2e。
合成方案3
Figure PCTCN2018072988-appb-000128
化合物3h可以通过合成方案3所描述的方法制备得到。化合物3a通过与乙烯基格氏试剂(如乙烯基溴化镁)发生加成反应得化合物3b;化合物3b经氧化反应(如与琼斯(Jone's)试剂或戴斯-马丁氧化剂反 应等)得到化合物3c;化合物3c先与化合物3e在溶剂(如四氢呋喃等)中发生Michael加成反应,然后再在碱性条件下(例如,在吡啶或4-二甲氨基吡啶的作用下)被化合物3f乙酰化,得到化合物3g;化合物3g在碱性条件下(如在甲醇钠,氢化钠等作用下)关环得到化合物3h。
其中,中间体化合物3g还可以通过以下方法制备得到:化合物3c先在TMSCl和NaI的存在下、在溶剂(例如,乙腈和水)中发生加成反应得到化合物3d;化合物3d再与化合物3e在碱性条件下(例如,在三乙胺的作用下)发生取代反应,然后再在碱性条件下(例如,在吡啶或4-二甲氨基吡啶的作用下)被3f乙酰化,得到化合物3g。
合成方案4
Figure PCTCN2018072988-appb-000129
中间体化合物4m可以通过合成方案4所描述的方法制备得到。醛4a和(S)-2-甲基丙基-2-亚磺酰胺在无水硫酸铜的存在下,在溶剂(如二氯甲烷)中发生缩合反应得到化合物4b。化合物4b与烯丙基溴化镁在三氯化铟催化的作用下发生加成反应得到化合物4c;化合物4c在酸性条件下(例如,在HCl作用下),脱去(S)-2-甲基丙基-2-亚磺酰基得到化合物4d;对化合物4d进行氨基保护得到化合物4e;化合物4e与化合物4f在催化剂(如Pd(dppf)Cl 2-CH 2Cl 2)和碱(如碳酸铯)的存在下,于溶剂(如二氧六环和水)中进行铃木偶联反应得到化合物4g;化合物4g发生还原反应(如在锌粉-氯化铵体系中硝基被还原)得到化合物4h;化合物4h与化合物4i在碱性条件下(如吡啶的作用下)发生取代反应得到化合物4j;化合物4j在催化剂(如Grubbs2代催化剂)作用下经关环复分解反应得大环化合物4k;化合物4k经催化(如Pd/C催化)氢化还原得到化合物4l;化合物4l脱保护(例如,在三氟乙酸等酸性条件下脱去Boc保护基)得到化合物4m。
合成方案5
Figure PCTCN2018072988-appb-000130
Figure PCTCN2018072988-appb-000131
中间体化合物5i可以通过合成方案5所描述的方法制备得到。化合物4e和化合物5b在催化剂(如Pd(dppf)Cl 2-CH 2Cl 2)和碱(如碳酸铯)的存在下,于溶剂(如二氧六环和水)中发生铃木偶联反应得化合物5c;化合物5c经还原(如在锌粉-氯化铵体系中硝基被还原成氨基)反应得到化合物5d;化合物5d和化合物4i在碱性(如吡啶)条件下发生取代反应得到化合物5f;化合物5f在催化剂(如Grubbs2代催化剂)的存在下经关环复分解反应得到大环化合物5g;化合物5g经催化(如Pd/C催化)氢化还原得到化合物5h;化合物5h脱保护(例如,在三氟乙酸等酸性条件下脱去Boc保护基)得到化合物5i。
合成方案6
Figure PCTCN2018072988-appb-000132
中间体化合物6j可以通过合成方案6所描述的方法制备得到。化合物4e和化合物6b在催化剂(如Pd(dppf)Cl 2-CH 2Cl 2)和碱(如碳酸铯)的存在下,于溶剂(如二氧六环和水)中发生铃木偶联反应得化合物6c;化合物6c与氯甲酸甲酯在碱性条件下(如吡啶存在下)发生缩合反应得到化合物6d;化合物6d经还原(如在锌粉-氯化铵体系中硝基被还原成氨基)反应得到化合物6e;化合物6e和化合物4i在碱性(如吡啶)条件下发生取代反应得到化合物6g;化合物6g在催化剂(如Grubbs2代催化剂)的存在下经关环复分解反应得到大环化合物6h;化合物6h经催化(如Pd/C催化)氢化还原得到化合物6i;化合物6i脱保护(例如,在三氟乙酸等酸性条件下脱去Boc保护基)得到化合物6j。
合成方案7
Figure PCTCN2018072988-appb-000133
Figure PCTCN2018072988-appb-000134
中间体化合物7i可以通过合成方案7所描述的方法制备得到。化合物4e和化合物7b在钯(II)催化剂(如Pd(OAC) 2)、磷配体(如正丁基二(1-金刚烷基)膦)和碱(如碳酸钾)的存在下,在溶剂(如N,N-二甲基甲酰胺)中发生偶联反应得到化合物7c;化合物7c经还原反应(例如,在锌粉-氯化铵体系中硝基被还原成氨基)得到化合物7d;化合物7d和化合物4i在碱性条件(如吡啶)下,于溶剂(如二氯甲烷)中发生取代反应得到化合物7f;化合物7f在催化剂(如Grubbs2代催化剂)作用下经关环复分解反应得到大环化合物7g;化合物7g经催化(如Pd/C催化)氢化还原得化合物7h,再脱保护(例如,在三氟乙酸等酸性条件下脱去Boc保护基)得到化合物7i。
下面的实施例可以对本发明做进一步的描述,然而,这些实施例不应作为对本发明的范围的限制。
实施例
实施例1
N-((10R,14S)-14-(4-(6-溴-3-氯-4-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22),18,20-八烯-5-基)氨基甲酸甲酯
Figure PCTCN2018072988-appb-000135
步骤1:2-(苯基氨基)马来酸二甲酯(1A)
Figure PCTCN2018072988-appb-000136
在室温下,将丁炔二羧酸二甲酯(15.26g,107.40mmol)缓慢滴加到苯胺(10.00g,107.40mmol)的无水甲醇(100mL)溶液中,滴加完毕后,体系升温到90℃,回流搅拌12小时。停止反应,冷却至室温,浓缩得到粗产品,经硅胶柱层析(乙酸乙酯/石油醚(v/v)=1/10)纯化,得到黄色油状物(25.00g,99.0%)。MS(ESI,pos.ion)m/z:236.0[M+1] +.
步骤2:4-羟基喹啉-2-甲酸甲酯(1B)
Figure PCTCN2018072988-appb-000137
室温下,将多聚磷酸(31.25g,318.82mmol)加到1A(25.00g,106.27mmol)中,然后体系升温到120℃搅拌1小时。停止反应,冷却至室温,用20%的碳酸钠溶液(300mL)淬灭过量的酸,过滤,用水洗涤,再用二氯甲烷(2000mL)将滤饼溶解,有机相用水(100mL×3)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,得到粗产品,经乙酸乙酯和石油醚重结晶得到灰色固体(12.00g,52.8%)。
1H NMR(400MHz,DMSO-d 6)δ(ppm)12.07(s,1H),8.08(d,J=7.4Hz,1H),7.94(d,J=8.4Hz,1H),7.76–7.65(m,1H),7.38(t,J=7.5Hz,1H),6.66(s,1H),3.96(s,3H).
步骤3:4-溴喹啉-2-甲酸甲酯(1C)
Figure PCTCN2018072988-appb-000138
在室温下,将三溴氧磷(8.9g,31mmol)和碳酸钠(4.3g,31mmol)加到1B(2.1g,10mmol)的无水乙腈(45mL)溶液中,氮气保护下,体系升温到92℃搅拌2小时。然后冷却至室温,减压蒸除溶剂。剩余物用水溶解,过滤,滤饼用水(10mL×2)洗涤,然后在60℃下真空干燥,用乙酸乙酯和石油醚重结晶,得灰色固体(2.70g,98.0%)。
MS(ESI,pos.ion)m/z:399.1(M+1).
步骤4:4-溴喹啉-2-甲醛(1D)
Figure PCTCN2018072988-appb-000139
氮气保护下,将1C(0.60g,2.25mmol)的干燥四氢呋喃溶液(15mL)冷却到-78℃,然后滴加二异丁基氢化铝(12.09mL,12.09mmol,1mol/L的甲苯溶液),在该温度下反应4小时。将3mol/L的稀盐酸(20mL)缓慢滴加到体系中,然后升温至室温。加入饱和碳酸氢钠(20mL)溶液,体系用乙酸乙酯(150mL×2)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩,得到的粗产品经硅胶柱层析(乙酸乙酯/石油醚(v/v)=1/10)纯化,得到白色固体(0.51g,96.2%)。
步骤5:(S,E)-N-((4-溴喹啉-2-基)亚甲基)-2-甲基丙烷-2-亚磺酰胺(1E)
Figure PCTCN2018072988-appb-000140
在室温下,将硫酸铜(0.045g,0.28mmol)和1D(0.03g,0.13mmol)加入到(S)-(-)-叔丁基亚磺酰胺(0.015g,0.13mmol)的干燥二氯甲烷(5mL)溶液中,加料完毕后,室温搅拌3小时。过滤,用二氯甲烷洗涤,滤液浓缩,得到粗产品,经硅胶柱层析(乙酸乙酯/石油醚(v/v)=1/10)纯化,得到亮黄色固体(0.036g,83.5%)。
MS(ESI,pos.ion)m/z:339.0[M+1] +.
步骤6:(S)-N-((S)-1-(4-溴喹啉-2-基)丁-3-烯-1-基)-2-甲基丙烷-2-亚磺酰胺(1F)
Figure PCTCN2018072988-appb-000141
氮气氛围下,将三氯化铟(0.022g,0.097mmol)的干燥四氢呋喃(1mL)溶液冷却到-5℃,然后向体系内缓慢滴加烯丙基溴化镁(0.13mL,0.13mmol,1mol/L的四氢呋喃溶液),滴加完毕后,升温到室温搅拌1小时,再加入1E(0.033g,0.0973mmol)的无水乙醇(1mL)溶液,滴加完毕后,继续搅拌2-3小时。停止反应,减压浓缩,得到残渣加入乙酸乙酯(200mL)和水(50mL),分液,收集有机相,水相再用乙酸乙酯(50mL×2)萃取,合并有机相,用饱和食盐水(100mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩得到粗产品,经硅胶柱层析(乙酸乙酯/石油醚(v/v)=1/15)纯化,得到亮黄色油状产物(0.0060g,16.0%)。
MS(ESI,pos.ion)m/z:383.3[M+1] +.
步骤7:(S)-1-(4-溴喹啉-2-基)丁-3-烯-1-胺(1G)
Figure PCTCN2018072988-appb-000142
在室温下,将6mol/L的盐酸溶液(2.7mL,16.32mmol)缓慢滴加到1F(0.37g,0.96mmol)的甲醇(6mL)溶液中,滴加完毕后,室温下继续搅拌3小时。停止反应,减压除去甲醇,然后再加入水(3mL),用乙酸乙酯(5mL)洗涤,有机相作废液处理,水相用饱和碳酸钠水溶液调至弱碱性,用乙酸乙酯(10mL×3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,得到黄色固体(0.200g,75.2%)。
1H NMR(600MHz,CDCl 3)δ(ppm)8.18–8.14(m,1H),8.06–8.03(m,1H),7.84–7.79(m,1H),7.74(t,J=7.4Hz,1H),7.60(t,J=7.5Hz,1H),5.85–5.76(m,1H),5.19(d,J=17.3Hz,1H),5.15(d,J=10.3Hz),4.24(dd,J=8.1,4.9Hz,1H),2.73–2.64(m,1H),2.50–2.43(m,1H).
步骤8:(S)-(1-(4-溴喹啉-2-基)丁-3-烯-1-基)氨基甲酸叔丁酯(1H)
Figure PCTCN2018072988-appb-000143
在室温下,将二碳酸二叔丁酯(0.16g,0.17mL,0.72mmol)滴加到溶有1G(0.20g,0.72mmol)和三乙胺(0.074g,0.10mL,0.72mmol)的干燥二氯甲烷(10mL)溶液中,滴加完毕后,室温下继续搅拌16小时。停止反应,加入二氯甲烷(20mL),然后用水(15mL×2)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩,粗产品经硅胶柱层析(乙酸乙酯/石油醚(v/v)=1/10)纯化,得到黄色固体(0.26g,95.9%)。
MS(ESI,pos.ion)m/z:376.9[M+1] +.
步骤9:(S)-(1-(4-(4-氨基-2-硝基苯基)喹啉-2-基)丁-3-烯-1-基)氨基甲酸叔丁酯(1I)
Figure PCTCN2018072988-appb-000144
在室温下,向3-硝基-4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯胺(0.22g,0.83mmol)和 1H(0.26g,0.69mmol)的体系中依次加入乙二醇二甲醚(15mL)和2mol/L的碳酸钠水溶液(1.73mL,3.46mmol),再加入四(三苯基膦)钯(0)(0.080g,0.069mmol),氮气氛围下,体系升温到90℃搅拌4小时。停止反应,冷却至室温,浓缩,残渣经硅胶柱层析(乙酸乙酯/石油醚(v/v)=1/10)纯化,得到红色固体(0.21g,70.0%)。
MS(ESI,pos.ion)m/z:435.3[M+1] +.
步骤10:(S)-(1-(4-(4-(甲氧基酰氨基)-2-硝基苯基)喹啉-2-基)丁-3-烯-1-基)氨基甲酸叔丁酯(1J)
Figure PCTCN2018072988-appb-000145
在-78℃下,将氯甲酸甲酯(0.024g,0.020mL,0.25mmol)的二氯甲烷溶液(5mL)缓慢滴加到化合物1I(0.10g,0.23mmol)和吡啶(0.055g,0.056mL,0.69mmol)的二氯甲烷(20mL)溶液中,滴加完毕后,在-78℃下继续搅拌2小时。停止反应,向体系中加入饱和氯化铵溶液(5mL),搅拌5分钟后,升温到室温继续搅拌10分钟。加入二氯甲烷(50mL),体系用水(10mL×2)和饱和食盐水(20mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩后,经硅胶柱层析(乙酸乙酯/石油醚(v/v)=1/10)纯化,得到暗红色晶体(0.103g,90.9%)。
MS(ESI,pos.ion)m/z:492.9[M+1] +.
步骤11:(S)-(1-(4-(4-(甲氧基酰氨基)-2-氨基苯基)喹啉-2-基)丁-3-烯-1-基)氨基甲酸叔丁酯(1K)
Figure PCTCN2018072988-appb-000146
在室温下,将锌粉(4.26g,65.20mmol)和氯化铵(3.49g,65.20mmol)加入含有1J(3.21g,6.52mmol)的甲醇(65mL)溶液中,体系在室温下搅拌2.5小时。停止反应,过滤,滤饼用甲醇(10mL×3)洗涤,滤液浓缩得残渣,残渣用二氯甲烷(100mL)溶解,用水(30mL×2)和饱和食盐水(30mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩,得粗产品,经硅胶柱层析(乙酸乙酯/石油醚(v/v)=1/3)纯化,得到黄色晶体(1.03g,34.2%)。
MS(ESI,pos.ion)m/z:463.3[M+1] +.
步骤12:(S)-(1-(4-(4-(甲氧基酰氨基)-2-((R)-2-甲基丁-3-烯酰氨基)苯基)喹啉-2-基)丁-3-烯-1-基)氨基甲酸叔 丁酯(1L)
Figure PCTCN2018072988-appb-000147
在0℃下,将含有(R)-2-甲基丁-3-烯酰氯(0.95g,8.04mmol)的二氯甲烷溶液(10mL)缓慢加入到溶有1K(0.95g,2.06mmol)和吡啶(0.66g,0.67mL,8.24mmol)的干燥二氯甲烷(100mL)溶液中,然后在0℃下搅拌30分钟,升温到室温再搅拌5小时。停止反应,向体系中再加入二氯甲烷(100mL),再用水(50mL×2)和饱和食盐水(50mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩得粗产品,用硅胶柱层析(乙酸乙酯/石油醚(v/v)=1/3)纯化,得到淡黄色固体(0.476g,42.4%)。
MS(ESI,pos.ion)m/z:545.9[M+1] +.
步骤13:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-8,16-二氮杂四环[13.7.1.0 2.7.0 17,22]二十三烷 -1(23),2(3),4,6,11(12),15,17(22),18,20-九烯-5-基)氨基甲酸甲酯(1M)
Figure PCTCN2018072988-appb-000148
在室温下,将一水合对甲苯磺酸(0.84g,4.28mmol)放置到两口瓶中,加热到80℃,减压干燥约1小时。然后将溶有1L(2.12g,3.89mmol)的干燥二氯甲烷(100mL)溶液注射到冷却到室温的体系内,搅拌45分钟。再将Grubbs 2代催化剂(1.05g,1.22mmol)的干燥二氯甲烷(20mL)溶液缓慢注射到体系中,升温至回流搅拌过夜。停止反应,冷却至室温,向体系中加入二氯甲烷(100mL)和饱和碳酸氢钠水溶液(50mL),收集有机相,水相再用二氯甲烷(30mL×3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩,得到粗产品经硅胶柱层析(乙酸乙酯/石油醚(v/v)=1/3)纯化,得到淡黄色固体(1.15g,57.2%)。MS(ESI,pos.ion)m/z:517.3[M-13] +.
步骤14:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-8,16-二氮杂四环[13.7.1.0 2.7.0 17,22]二十三烷 -1(23),2(3),4,6,15,17(22),18,20-八烯-5-基)氨基甲酸甲酯(1N)
Figure PCTCN2018072988-appb-000149
在室温下,将10%的Pd/C(0.123g)加到1M(1.15g,2.17mmol)的无水甲醇(100mL)溶液中,用氢气置换2-3次,在氢气氛围下,室温搅拌过夜。停止反应,过滤,滤液浓缩得到固体,经硅胶柱层析(二氯甲烷/甲醇(v/v)=10/1)纯化,得到白色固体(1.01g,87.5%)。
MS(ESI,pos.ion)m/z:519.5[M-13] +.
步骤15:N-((10R,14S)-14-氨基-10-甲基-9-氧代-8,16-二氮杂四环[13.7.1.0 2.7.0 17,22]二十三烷 -1(23),2(3),4,6,15,17(22),18,20-八烯-5-基)氨基甲酸甲酯(1O)
Figure PCTCN2018072988-appb-000150
在室温下,将三氟乙酸(4.6g,3.0mL,40.00mmol)加到1N(0.67g,1.28mmol)的干燥二氯甲烷溶液(30mL)中,然后室温搅拌6小时。停止反应,减压浓缩得到暗红色固体,加入乙酸乙酯(50mL)和饱和碳酸氢钠溶液(10mL),搅拌10分钟,收集有机相,水相再用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩,经硅胶柱层析(二氯甲烷/甲醇(v/v)=20/1)纯化,得到白色固体(0.52g,96.75%)。
步骤16:1-(6-溴-3-氯-2-氟苯基)-3-碘丙烷-1-酮(1P)
Figure PCTCN2018072988-appb-000151
在室温下,将三甲基氯硅烷(0.78g,0.62mL,7.13mmol)加到含有碘化钠(1.10g,7.3mmol)和乙腈(5mL)的体系中,搅拌10分钟后,再加入水(0.50g),搅拌5分钟后,加入1-(6-溴-3-氯-2-氟苯基)丙-2-烯-1-酮(参照专利WO 2014022766中间体2的合成方法制备得到)(1.25g,4.74mmol),继续搅拌2小时。停止反应,向体系中加入水(5mL),用乙酸乙酯(20mL×3)萃取。合并有机相,有机相用无水硫酸钠干燥,过滤,滤液浓缩,得到红色油状液体,再经柱层析(乙酸乙酯/石油醚(v/v)=1/10)提纯,得到红色油状液体(1.35g,72.7%)。
1H NMR(600MHz,CDCl 3)δ(ppm)7.36–7.32(m,1H),3.48(t,J=7.0Hz,1H),3.41(dd,J=10.7,3.8Hz,1H).
步骤17:N-((10R,14S)-14-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙氨基)-10-甲基-9-氧代-8,16-二氮杂四环 [13.7.1.0 2.7.0 17,22]二十三烷-1(23),2(3),4,6,15,17(22),18,20-八烯-5-基)氨基甲酸甲酯(1Q)
Figure PCTCN2018072988-appb-000152
在室温下,将三乙胺(0.24g,0.33mL,2.37mmol)加到1O(0.20g,0.48mmol)的四氢呋喃(20mL)溶液中,再加入1P(0.19g,0.48mmol)的四氢呋喃(10mL)溶液,加入完毕后,室温搅拌5小时。停止反应,加入乙酸乙酯(50mL)和水(20mL),收集有机相,水相用乙酸乙酯(15mL×3)萃取,合并有机相并用 无水硫酸钠干燥,过滤,滤液浓缩,得到固体,再用中性氧化铝柱层析(乙酸乙酯/石油醚(v/v)=1/1)纯化,得到白色固体(0.26g,79.8%)。
MS(ESI,pos.ion)m/z:683.4[M-13] +.
步骤18:N-((10R,14S)-14-(N-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙基)-2-(二乙氧基磷酰基)乙酰氨基)-10-甲基-9- 氧代-8,16-二氮杂四环[13.7.1.0 2.7.0 17,22]二十三烷-1(23),2(3),4,6,15,17(22),18,20-八烯-5-基)氨基甲酸甲酯 (1R)
Figure PCTCN2018072988-appb-000153
在室温下,将吡啶(0.021g,0.021mL,0.27mmol)和4-二甲氨基吡啶(0.006g,0.049mmol)加到1Q(0.029g,0.042mmol)的二氯甲烷(10mL)溶液中,再加入(2-氯-2-氧代乙基)磷酸二乙酯(0.019g,0.088mmol)的二氯甲烷(10mL)溶液,加入完毕后,在微波下,升温到40℃搅拌2小时。停止反应,冷却至室温,加入乙酸乙酯(50mL)和水(20mL),收集有机相,水相用乙酸乙酯(15mL×3)萃取,合并有机相并用无水硫酸钠干燥,过滤,滤液浓缩,得到固体,再用硅胶柱层析(二氯甲烷/甲醇(v/v)=20/1)纯化,得到白色固体(0.02g,54.7%)。
MS(ESI,pos.ion)m/z:861.5[M-13] +.
步骤19:N-((10R,14S)-14-(4-(6-溴-3-氯-4-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-8,16-二氮 杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22),18,20-八烯-5-基)氨基甲酸甲酯
在室温下,将60%的氢化钠(0.015g,0.38mmol)置于两口瓶中,用氮气保护后,用注射器注入溶有1R(0.078g,0.091mmol)的干燥四氢呋喃(5mL)溶液,室温搅拌30分钟。停止反应,加入二氯甲烷(50mL)和饱和氯化铵溶液(15mL),收集有机相,水相用二氯甲烷(20mL×3)萃取,合并有机相,无水硫酸钠干燥。过滤,浓缩得淡黄色固体,经中性氧化铝柱层析(乙酸乙酯/石油醚(v/v)=3/2)纯化,得到灰白色固体(0.034g,53.1%)。
MS(ESI,pos.ion):m/z:707.4[M+1] +
1H NMR(600MHz,CDCl 3)δ(ppm)8.22(s,1H),8.02(d,J=7.9Hz,1H),7.87(d,J=8.4Hz,1H),7.66(d,J=8.9Hz,1H),7.56–7.53(m,1H),7.48–7.43(m,1H),7.43–7.38(m,2H),7.33(s,1H),7.29(d,J=7.7Hz,1H),6.51(d,J=4.8Hz,1H),4.84–4.77(m,1H),3.02(s,3H),3.00–2.90(m,2H),2.70–2.64(m,1H),2.46(td,J=14.8,7.4Hz,2H),2.24–2.18(m,1H),2.03–1.99(m,2H),1.68-1.63(m,2H),0.93(d,J=6.9Hz,3H),0.84(s,2H).
实施例2
N-((10R,14S)-14-(4-(6-溴-3-氯-2-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-8,16-二氮杂四环[13.6.1.0 2,7.0 17,21]二十二烷-1(22),2(7),3,5,15,17(21)-六烯-5-基)氨基甲酸甲酯
Figure PCTCN2018072988-appb-000154
步骤1:6,7-二氢-5H-环戊烷并[b]吡啶1-氧化物(2A)
Figure PCTCN2018072988-appb-000155
称取6,7-二氢-5H-环戊烷并[b]吡啶(50g,419.60mmol)于烧瓶中,加入二氯甲烷(500mL),冰浴条件下分批加入间氯过氧苯甲酸(95g,467.93mmol,85mass%),5分钟后移至室温下搅拌过夜。终止反应,旋干溶剂,残余物经硅胶柱层析分离纯化(石油醚/乙酸乙酯(v/v)=10/1),得白色固体(52.8g,93.1%)。
步骤2:6,7-二氢-5H-环戊烷并[b]吡啶-2-甲腈(2B)
Figure PCTCN2018072988-appb-000156
称取2A(55.2g,408mmol)于四口烧瓶中,向其中加入二氯甲烷(700mL),室温条件下加入三甲基氰硅烷(56.5mL,452mmol),搅拌20分钟后,逐滴滴加二乙氨基甲酰氯(57mL,450mmol),室温反应64小时。终止反应,向体系中加入10%碳酸钾溶液(700mL),搅拌20分钟,分出有机相,水相用二氯甲烷萃取(40mL×3),合并有机相并用无水硫酸钠干燥,过滤,旋干,残余物经硅胶柱层析分离纯化(石油醚/乙酸乙酯(v/v)=20/1),得白色固体(23.1g,39.2%)。
步骤3:(6,7-二氢-5H-环戊烷并[b]吡啶-2-基)甲醇(2C)
Figure PCTCN2018072988-appb-000157
称取2B(28.56g,198.1mmol)于高压釜中,向其中加入10%的Pd/C(4.1g),并加入2mol/L的盐酸溶液(400mL)和甲醇(200mL),通入氢气,在2.6MPa、室温条件下反应。当压力表上指针停留在2.4MPa不再降低时,停止反应。向反应瓶中加入碳酸钠固体至不再生成气泡,旋蒸除去甲醇,水相用二氯甲烷和甲醇(v/v=10/1)混合溶剂萃取(30mL×6),有机相经无水硫酸钠干燥,过滤,旋干,残留物经硅胶柱层析分离纯化(石油醚/乙酸乙酯(v/v)=10/1),得白色固体(16.88g,57.11%)。
MS(ESI,pos.ion)m/z:150.3[M+1] +.
步骤4:2-(羟甲基)-6,7-二氢-5H-环戊烷并[b]吡啶1-氧化物(2D)
Figure PCTCN2018072988-appb-000158
称取2C(16.364g,109.69mmol)于烧瓶中,向其中加入二氯甲烷(400mL),室温条件下分批加入间氯过氧苯甲酸(24.5g,120.65mmol,85mass%),体系在室温下搅拌1小时。终止反应,减压蒸除溶剂,残留物经硅胶柱层析分离纯化(石油醚/乙酸乙酯(v/v)=1/2),得白色固体(17.69g,97.63%)。
步骤5:4-溴-2-(溴甲基)-6,7-二氢-5H-环戊烷并[b]吡啶(2E)
Figure PCTCN2018072988-appb-000159
称取2D(4.18g,25.3mmol)于烧瓶中,向其中加入N,N-二甲基甲酰胺(70mL)和甲苯(70mL),室温下分批加入三溴氧磷(29.16g,101.7mmol),体系室温搅拌反应1小时。终止反应,向体系中加入水(70mL),并加入碳酸钠固体至没有气泡生成,乙酸乙酯萃取(50mL×4),合并有机相,无水硫酸钠干燥,过滤,旋干,残余物经硅胶柱层析分离纯化(石油醚/乙酸乙酯(v/v)=200/1),得白色固体(3.19g,43.3%)。
步骤6:(4-溴-6,7-二氢-5H-环戊烷并[b]吡啶-2-基)甲醇(2F)
Figure PCTCN2018072988-appb-000160
称取2E(10.28g,35.33mmol)于烧瓶中,室温下向反应瓶中加入碳酸钠(60g,3740.2mmol)、丙酮(200mL)和水(200mL),体系升温至75℃搅拌过夜。终止反应,冷却至室温,减压旋出丙酮,乙酸乙酯萃取(30mL×8),合并有机相,无水硫酸钠干燥,过滤,残留物经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=20/1),得淡黄色固体(5.14g,63.8%)。
MS(ESI,pos.ion)m/z:229.1[M+1] +.
步骤7:4-溴-6,7-二氢-5H-环戊烷并[b]吡啶-2-甲醛(2G)
Figure PCTCN2018072988-appb-000161
称取2F(5.14g,22.5mmol)于烧瓶中,向其中加入二氯甲烷(200mL)和Dess-Martin氧化剂(11.5g,27.1mmol),室温下搅拌1小时。向体系中加入饱和硫代硫酸钠溶液(50mL)和饱和碳酸氢钠溶液(50mL),搅拌至溶液澄清,乙酸乙酯(50mL×2)萃取,合并有机相,无水硫酸钠干燥,过滤,旋干,残余物经硅胶柱层析分离纯化(石油醚/乙酸乙酯(v/v)=50/1),得到白色固体(4.01g,78.7%)。
步骤8:(S,E)-N-((4-溴-6,7-二氢-5H-环戊烷并[b]吡啶-2-基)亚甲基)-2-甲基丙烷-2-亚磺酰胺(2H)
Figure PCTCN2018072988-appb-000162
室温条件下,将(S)-(-)-叔丁基亚磺酰胺(3.80g,31.38mmol)和硫酸铜(6.69g,41.9mmol)加入到2G(4.73g,20.9mmol)的二氯甲烷(150mL)溶液中,混合体系加热回流反应8小时。终止反应,反应液经硅藻土抽滤,滤液旋干,残渣经硅胶柱层析(石油醚/乙酸乙酯(v/v)=10/1)纯化,得黄色油状物(6.46g,93.8%)。
MS(ESI,pos.ion)m/z:330.2[M+1] +.
步骤9:(S)-N-((S)-1-(4-溴-6,7-二氢-5H-环戊烷并[b]吡啶-2-基)丁-3-烯-1-基)-2-甲基丙烷-2-亚磺酰胺(2I)
Figure PCTCN2018072988-appb-000163
在-5℃下,将烯丙基溴化镁(40mL,40mmol,1.0mol/L的四氢呋喃溶液)逐滴滴入三氯化铟(8.73g,39.5mmol)的四氢呋喃(150mL)溶液中,滴加完毕后移至室温下反应1小时。向瓶中逐滴滴加2H(6.46g,19.6mmol)的乙醇(60mL)溶液,室温条件下搅拌3小时。终止反应,反应液经过硅藻土抽滤,并用乙酸乙酯洗涤(30mL×3),旋干滤液,残余物用乙酸乙酯溶解后,再用水洗涤(30mL×4),有机层用无水硫酸钠干燥,过滤,旋干,残余物经硅胶柱层析(石油醚/乙酸乙酯(v/v)=3/1)纯化,得白色固体(7.18g,98.6%)。
MS(ESI,pos.ion)m/z:372.2[M+1] +.
步骤10:(S)-1-(4-溴-6,7-二氢-5H-环戊烷并[b]吡啶-2-基)丁-3-烯-1-胺(2J)
Figure PCTCN2018072988-appb-000164
将2I(7.18g,19.3mmol)加入烧瓶中,再加入甲醇(75mL)及6mol/L的盐酸溶液(103mL,618.7mmol),混合体系在室温下搅拌1小时。减压旋出甲醇,然后向瓶中加入饱和碳酸氢钠溶液至碱性,二氯甲烷(80mL×2)萃取,有机相用无水硫酸钠干燥,过滤,滤液旋干,得棕黄色液体(6.6g,100%)。
步骤11:(S)-(1-(4-溴-6,7-二氢-5H-环戊烷并[b]吡啶-2-基)丁-3-烯-1-基)氨基甲酸叔丁酯(2K)
Figure PCTCN2018072988-appb-000165
称取2J(6.6g,25mmol)于烧瓶中,向其中加入二氯甲烷(150mL)和三乙胺(7mL,50.2mmol)。室温条件下逐滴滴加二碳酸二叔丁酯(6.2mL,27mmol),滴毕,室温反应6小时。终止反应,向体系中加入二氯甲烷(150mL),体系分别用水(300mL×2)和饱和食盐水(200mL)洗涤,有机相经无水硫酸钠干燥,过滤,旋干,残余物经硅胶柱层析(石油醚/乙酸乙酯(v/v)=100/1)纯化,得白色固体(7.82g,86%)。MS(ESI,pos.ion)m/z:368.3[M+1] +.
步骤12:(S)-(1-(4-(4-氨基-2-硝基苯基)-6,7-二氢-5H-环戊烷并[b]吡啶-2-基)丁-3-烯-1-基)氨基甲酸叔丁酯 (2L)
Figure PCTCN2018072988-appb-000166
称取2K(7.82g,21.3mmol)、3-硝基-4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯胺(8.59g,32.5mmol)和2mol/L的碳酸钠溶液(22mL,44mmol)于双口烧瓶中,向其中迅速加入三环己基膦(0.92g,3.19mmol),然后用氮气置换掉体系中含有的氧气,然后迅速加入(dba) 3Pd(0) 2(68.73mg,2.13mmol),然后再用氮气置换掉体系中含有的氧气,将反应瓶置于100℃油浴下搅拌反应过夜。终止反应,冷却至室温,反应液经硅藻土抽滤,往滤液中加入乙酸乙酯(400mL),有机相分别用水(200mL×3)和饱和食盐水(300mL)洗涤,无水硫酸钠干燥,过滤,残留物经硅胶柱层析(石油醚/乙酸乙酯(v/v)=10/1)纯化,得棕红色油状物(6.83g,75.6%)。
MS(ESI,pos.ion)m/z:425.3[M+1] +.
步骤13:N-[4-[2-[(S)-1-(叔丁氧羰基氨基)丁-3-烯基]-6,7-二氢-5H-环戊烷并[b]吡啶-4-基]-3-硝基-苯基]氨基 甲酸甲酯(2M)
Figure PCTCN2018072988-appb-000167
将2L(6.83g,16.1mmol)和吡啶(4mL,49.7mmol)加入两口烧瓶中,向其中加入二氯甲烷(170mL),冷至-78℃,向体系中缓慢滴加氯甲酸甲酯(1.4mL,18mmol)的二氯甲烷(80mL)溶液,滴加完毕后搅拌反应3小时。终止反应,向体系中加入饱和氯化铵溶液(16mL)并搅拌5分钟,然后升至室温搅拌10分钟。向体系中加入二氯甲烷(200mL),体系分别用水(300mL)和饱和食盐水(200mL)洗涤,无水硫酸钠干燥,过滤,滤液旋干,得暗红色固体(7.32g,94.3%),所得产品直接用于下一步反应。
步骤14:N-[3-氨基-4-[2-[(S)-1-(叔丁氧羰基氨基)丁-3-烯基]-6,7-二氢-5H-环戊烷并[b]吡啶-4-基]苯基]氨基甲 酸甲酯(2N)
Figure PCTCN2018072988-appb-000168
室温下,将锌粉(10.03g,153.3mmol)、氯化铵(8.36g,156mmol)加入到2M(7.32g,15.2mmol)的甲醇(200mL)溶液中,室温下反应过夜。终止反应,反应液经硅藻土抽滤,滤饼用甲醇洗涤(30mL×3),滤液减压旋出溶剂,残余物用二氯甲烷(500mL)溶解,依次用水(300mL×2)和饱和食盐水(300mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液旋干,得淡黄色晶体(6.58g,95.9%)。
MS(ESI,pos.ion)m/z:453.3[M+1] +.
步骤15:N-[4-[2-[(1S)-1-(叔丁氧羰基氨基)丁-3-烯基]-6,7-二氢-5H-环戊烷并[b]吡啶-4-基]-3-[[(2S)-2-甲基丁 -3-烯酰基]氨基]苯基]氨基甲酸甲酯(2O)
Figure PCTCN2018072988-appb-000169
在0℃下,将2N(0.99g,2.2mmol)溶于二氯甲烷(15mL)中,向反应瓶中加入吡啶(0.72mL,8.9mmol),再逐滴加入(R)-2-甲基丁-3-烯酰氯(0.4g,3mmol)的二氯甲烷(10mL)溶液,滴加完毕,在该条件下反应1小时,然后再移至室温下继续反应3小时。终止反应,向反应瓶中加入水(200mL),用二氯甲烷萃取(30mL×3),无水硫酸钠干燥,过滤,旋干滤液,残留物经硅胶柱层析(石油醚/乙酸乙酯(v/v)=10/1)纯化,得淡黄色固体(0.74g,63%)。
MS(ESI,pos.ion)m/z:535.4[M+1] +.
步骤16:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-8,16-二氮杂四环[13.6.1.0 2.7.0 17,21]二十二烷 -1(22),2(3),4,6,11(12),15,17(21)-七烯-5-基)氨基甲酸甲酯(2P)
Figure PCTCN2018072988-appb-000170
室温下,称取一水合对甲苯磺酸(0.297g,1.56mmol)于两口瓶中,将两口瓶置于80℃油浴中减压干燥0.5小时。关闭油浴加热,冷却至室温,将2O(0.74g,1.4mmol)的干燥二氯甲烷(100mL)溶液注射到反应体系中,并在室温条件下搅拌20分钟。然后再将Grubbs 2代催化剂(0.369g,0.435mmol)的干燥二氯甲烷(50mL)溶液滴加到体系中,体系在45℃下回流搅拌过夜。终止反应,冷却至室温,将反应液转移至单口瓶中,减压旋蒸除去溶剂,残余物经硅胶柱层析分离纯化(石油醚/乙酸乙酯(v/v)=2/1),得棕色固体(0.55g,76%)。
MS(ESI,pos.ion)m/z:507.2[M+1] +.
步骤17:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-8,16-二氮杂四环[13.6.1.0 2.7.0 17,21]二十二烷 -1(22),2(3),4,6,15,17(21)-六烯-5-基)氨基甲酸甲酯(2Q)
Figure PCTCN2018072988-appb-000171
室温条件下,称取2P(0.55g,1.1mmol)于高压釜中,向其中加入甲醇(50mL)和10%的Pd/C(0.065mg)。通入氢气,将高压釜加压到5MPa,并置于50℃油浴中加热搅拌过夜。终止反应,冷却至室温,反应液经硅藻土过滤,滤饼用甲醇洗涤(30mL×3)。滤液经减压旋蒸除去溶剂,得到棕黄色固体(0.56g,100%)。
MS(ESI,pos.ion)m/z:509.2[M+1] +.
步骤18:N-((10R,14S)-14-氨基-10-甲基-9-氧代-8,16-二氮杂四环[13.6.1.0 2.7.0 17,21]二十二烷 -1(22),2(3),4,6,15,17(21)-六烯-5-基)氨基甲酸甲酯(2R)
Figure PCTCN2018072988-appb-000172
在室温下,称取2Q(0.56g,1.1mmol)于烧瓶中,向其中加入干燥二氯甲烷(30mL)和三氟乙酸(2.5mL,34mmol),室温条件下搅拌8小时。终止反应,减压旋蒸除去溶剂,向反应瓶中加入乙酸乙酯(30mL)和饱和碳酸钠溶液(20mL),室温下搅拌10分钟。分出有机相,水相再用乙酸乙酯萃取(30mL×3),合并有机相并用无水硫酸钠干燥,过滤,直接旋干得棕红色固体(0.383g,85%)。
MS(ESI,pos.ion)m/z:409.2[M+1] +.
步骤19:N-((10R,14S)-14-(3-(6-溴-3-氯-4-氟苯基)-3-氧代丙氨基)-10-甲基-9-氧代-8,16-二氮杂四环 [13.6.1.0 2.7.0 17,21]二十二烷-1(22),2(3),4,6,15,17(21)-六烯-5-基)氨基甲酸甲酯(2S)
Figure PCTCN2018072988-appb-000173
称取2R(0.383g,0.938mmol)于烧瓶中,向其中加入四氢呋喃(20mL)和三乙胺(0.7mL,5mmol),然后再加入1P(0.55g,1.41mmol)的四氢呋喃(10mL)溶液,加入完毕后室温下搅拌过夜。终止反应,减压旋蒸除去溶剂,残余物用二氯甲烷(30mL)溶解后倒入分液漏斗中,加入水(300mL),分出有机相,水相再用二氯甲烷进行萃取(30mL×3),合并有机相并用无水硫酸钠干燥,过滤,旋干,得到棕红色固体(0.4g,60%),所得产品直接用于下一步反应。
MS(ESI,pos.ion)m/z:672.7[M+1] +.
步骤20:N-((10R,14S)-14-(N-(3-(6-溴-3-氯-4-氟苯基)-3-氧代丙基)-2-(二乙氧基磷酰基)乙酰氨基)-10-甲基-9- 氧代-8,16-二氮杂四环[13.6.1.0 2.7.0 17,21]二十二烷-1(22),2(3),4,6,15,17(21)-六烯-5-基)氨基甲酸甲酯(2T)
Figure PCTCN2018072988-appb-000174
室温下,分别将(2-氯-2-氧代乙基)磷酸二乙酯(0.652g,3.04mmol)、4-二甲氨基吡啶(0.088g,0.72mmol)和吡啶(0.24mL,2.98mmol)加到2S(0.4g,0.6mmol)的二氯甲烷(15mL)溶液中,加入完毕后在50℃下微波反应3小时。停止反应,将反应液倒入分液漏斗中,并加入二氯甲烷(50mL)和水(100mL),分出有机相,水相再用二氯甲烷萃取(30mL×5),合并有机相并用无水硫酸钠干燥,过滤,旋干,得棕褐色固体(0.5g,100%)。
MS(ESI,pos.ion)m/z:851.4[M+1] +.
步骤21:N-((10R,14S)-14-(4-(6-溴-3-氯-2-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-8,16-二氮 杂四环[13.6.1.0 2,7.0 17,21]二十二烷-1(22),2(7),3,5,15,17(21)-六烯-5-基)氨基甲酸甲酯
称取60%的氢化钠(0.087g,3.6mmol)于烧瓶中,氮气保护下向瓶中加入四氢呋喃(5.0mL),搅拌下再向瓶中加入2T(0.5g,0.6mmol)的四氢呋喃(20mL)溶液,室温条件下搅拌1小时。向反应瓶中加入水(20mL)淬灭反应。减压旋蒸除去溶剂,残余物用二氯甲烷(50mL)溶解后倒入分液漏斗中,向其中加入水(200mL),分出有机相,水相再用二氯甲烷进行萃取(30mL×5),合并有机相并用无水硫酸钠干燥,过滤,旋干,残留物经硅胶柱层析分离纯化(石油醚/乙酸乙酯(v/v)=3/1),得淡黄色固体(0.278g,70%)。MS(ESI,pos.ion)m/z:695.8[M+1] +
1H NMR(400MHz,CDCl 3)δ(ppm)7.94(s,1H),7.90(s,1H),7.52(d,J=7.9Hz,1H),7.37(d,J=8.7Hz,1H),7.32-7.25(m,3H),5.93(s,1H),5.23(brs,1H),3.92-3.86(m,1H),3.71(s,3H),3.40-3.32(m,2H),3.19-3.13(m, 5H),2.74-2.65(m,4H),2.38-2.29(m,2H),1.69-1.51(m,2H),1.05-0.96(m,2H).
实施例3
N-((10R,14S)-14-(4-(6-溴-3-氯-4-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯
Figure PCTCN2018072988-appb-000175
步骤1:4-羟基-5,6,7,8-四氢喹啉-2-甲酸甲酯(3A)
Figure PCTCN2018072988-appb-000176
在室温下,将浓盐酸(200mL)和4-羟基喹啉-2-甲酸甲酯(50.00g,246.08mmol)加入到高压釜中,将混合体系搅拌均匀后,加入PtO 2-3H 2O(5.00g,17.79mmol),密封体系,用氢气置换气体后,充入氢气,并加压到3.4MPa左右,室温搅拌两天。中间气体压力会下降,充入约3-4次氢气后,至体系压力基本保持不变,停止反应。减压浓缩,除去多余的浓盐酸和水分,得到褐色固体,用甲醇溶解,溶液过滤,滤液浓缩得到白色固体(47.60g,93.3%)。
MS(ESI,pos.ion)m/z:208.3[M+1] +.
步骤2:4-溴-5,6,7,8-四氢喹啉-2-甲酸甲酯(3B)
Figure PCTCN2018072988-appb-000177
在室温下,将碳酸钾(102.00g,730.63mmol)加入到溶有3A(50.30g,243.00mmol)的乙腈(500mL)溶液中,体系加热到95℃,分批加入三溴氧磷(210.00g,732.52mmol),加入完毕后,体系回流反应4小时。停止反应,静置冷却至室温,浓缩上清液,下层沉淀与浓缩后的残渣用饱和碳酸氢钠中和,再用二氯甲烷(500mL×4)萃取,有机相用无水硫酸钠干燥,过滤,滤液浓缩得到褐色固体,用硅胶柱层析(乙酸乙酯/石油醚(v/v)=1/5)纯化,得到灰色固体(30.00g,45.8%)。
MS(ESI,pos.ion)m/z:270.2[M+1] +.
步骤3:(4-溴-5,6,7,8-四氢喹啉-2-基)甲醇(3C)
Figure PCTCN2018072988-appb-000178
在室温下将硼氢化钠(3.20g,83.00mmol)分批次加入到溶有3B(7.40g,27.4mmol)的无水乙醇(100mL)溶液中,然后升温至回流搅拌3小时。停止反应,冷却至室温,向体系中加入饱和氯化铵溶液(30mL)和乙酸乙酯(500mL),继续搅拌0.5小时,加无水硫酸钠干燥,过滤,滤液浓缩,得到粗产品,经硅胶柱 层析(乙酸乙酯/石油醚(v/v)=1/1)纯化,得到白色固体(5.83g,87.9%)。
1H NMR(400MHz,CDCl 3)δ(ppm)7.29(s,1H),4.65(s,2H),2.90(t,J=5.8Hz,2H),2.75(t,J=5.8Hz,2H),1.96–1.74(m,4H).
步骤4:4-溴-5,6,7,8-四氢喹啉-2-甲醛(3D)
Figure PCTCN2018072988-appb-000179
将3C(5.08g,21.00mmol)溶解在二氯甲烷(100mL)中,冰浴冷却,搅拌下分批加入Dess-Martin氧化剂(9.08g,21.40mmol),加毕,升温至室温反应8小时。停止反应,过滤,用二氯甲烷(50mL)洗涤滤饼,合并有机层,用饱和硫代硫酸钠溶液(25mL)和饱和碳酸氢钠溶液(25mL)淬灭反应,分出有机层,水层用二氯甲烷(50mL)萃取,合并有机层,用饱和食盐水(25mL)洗涤,无水硫酸钠干燥,过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=20/1)得白色固体(4.56g,90.5%)。 1H NMR(400MHz,CDCl 3)δ(ppm)9.96(s,1H),7.96(s,1H),3.02(t,J=5.6Hz,2H),2.83(t,J=5.8Hz,2H),1.94-1.87(m,4H).
步骤5:(S)-N-((4-溴-5,6,7,8-四氢喹啉-2-基)亚甲基)-2-甲基丙烷-2-亚磺酰胺(3E)
Figure PCTCN2018072988-appb-000180
在室温下,将无水硫酸铜(6.02g,38.03mmol)和(S)-(-)-叔丁基亚磺酰胺(2.50g,20.61mmol)加入到3D(4.52g,8.81mmol)的二氯甲烷(100mL)溶液中,得到的混合体系在室温下搅拌过夜。停止搅拌,过滤,滤饼用二氯甲烷洗涤(50mL×2),滤液减压浓缩得到残渣,用硅胶柱层析(石油醚/乙酸乙酯(v/v)=10/1)纯化,得到亮黄色固体(6.40g,99.0%)。
1H NMR(400MHz,DMSO-d 6)δ(ppm)8.37(s,1H),8.06(s,1H),2.91(s,2H),2.75(s,2H),1.91–1.67(m,4H),1.19(s,9H).
步骤6:(S)-N-((S)-1-(4-溴-5,6,7,8-四氢喹啉-2-基)丁-3-烯-1-基)-2-甲基丙烷-2-亚磺酰胺(3F)
Figure PCTCN2018072988-appb-000181
在-5℃下,将烯丙基溴化镁(41.0mL,41.0mmol,1.0mol/L的四氢呋喃溶液)缓慢滴加到三氯化铟(9.10g,41.12mmol)的四氢呋喃(100mL)溶液中,滴加完毕后,体系升温到室温搅拌1小时。向体系中滴加3E(7.06g,20.61mmol)的无水乙醇溶液(60mL),滴加完毕后,室温反应过夜。过滤,用二氯甲烷(150mL×3)洗涤,滤液浓缩。向残渣中加入的二氯甲烷(500mL),再用水(100mL×2)和饱和食盐水(150mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩,再用硅胶柱层析(石油醚/乙酸乙酯(v/v)=5/1)提纯,得到白色固体(5.91g,74.0%)。
1H NMR(400MHz,CDCl 3)δ(ppm)7.29(s,1H),5.78–5.65(m,1H),5.07(d,J=4.7Hz,1H),5.04(s,1H),4.89(d,J=6.9Hz,1H),4.36(q,J=6.6Hz,1H),2.88(d,J=3.8Hz,2H),2.71(s,2H),2.54(dd,J=6.8Hz,2H),1.84-1.82(m,4H),1.25(s,9H).
步骤7:(S)-1-(4-溴-5,6,7,8-四氢喹啉-2-基)丁-3-烯-1-胺(3G)
Figure PCTCN2018072988-appb-000182
在室温下,将6mol/L的盐酸溶液(82.5mL,495.00mmol)加入到3F(5.92g,15.41mmol)的甲醇溶液(60mL)中,混合体系在室温下搅拌1小时。停止反应,反应液浓缩除去甲醇,水溶液用乙酸乙酯(15mL×2)萃取,合并有机相,用饱和碳酸氢钠水溶液调成碱性,用二氯甲烷萃取(100mL×3),收集有机相,用无水硫酸钠干燥,过滤,滤液浓缩得到白色固体(4.30g,99.6%)。
MS(ESI,pos.ion)m/z:281.1[M+1] +
1H NMR(400MHz,DMSO-d 6)δ(ppm)8.39(s,2H),7.65(s,1H),5.68-5.62(m,1H),5.09(d,J=4.7Hz,1H),5.05(s,1H),4.38(t,J=6.9Hz,1H),2.87(s,2H),2.70(s,2H),1.80(s,4H).
步骤8:(S)-(1-(4-溴-5,6,7,8-四氢喹啉-2-基)丁-3-烯-1-基)氨基甲酸叔丁酯(3H)
Figure PCTCN2018072988-appb-000183
在室温下,将二碳酸二叔丁酯(4.30g,4.5mL,19.03mmol)缓慢滴加到溶有3G(4.30g,15.31mmol)和三乙胺(2.22g,3.0mL,21.03mmol)的干燥二氯甲烷(100mL)溶液中,滴加完毕后,体系室温搅拌6小时。向体系中加入二氯甲烷(100mL),用水(50mL×2)和饱和食盐水(50mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩得到残渣,经硅胶柱层析(石油醚/乙酸乙酯(v/v)=10/1)提纯,得白色固体(5.84g,100%)。
MS(ESI,pos.ion)m/z:381.3[M+1] +.
步骤9:(S)-(1-(4-(4-氨基-2-硝基苯基)-5,6,7,8-四氢喹啉-2-基)丁-3-烯-1-基)氨基甲酸叔丁酯(3I)
Figure PCTCN2018072988-appb-000184
在室温下,将3-硝基-4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯胺(6.92g,26.03mmol)、3H(7.04g,18.52mmol)和三环己基膦(0.78g,2.78mmol)溶于N,N-二甲基甲酰胺(100mL)中,向体系中加入2mol/L的碳酸钠水溶液(19.0mL,38.02mmol)和(dba) 3Pd(0) 2(1.71g,1.91mmol),再用氮气置换掉进入的氧气,体系升温到90℃搅拌14小时。停止反应,冷却至室温,过滤,加入乙酸乙酯(500mL),用水(150mL×3)和饱和食盐水(150mL)洗涤,有机相用无水硫酸钠干燥。过滤,滤液浓缩,得到粗产品,用硅胶柱层析(石油醚/乙酸乙酯(v/v)=3/1)提纯,得到褐色固体(4.35g,53.7%)。
MS(ESI,pos.ion)m/z:439.3[M+1] +.
步骤10:(S)-(1-(4-(4-(甲氧基酰氨基)-2-硝基苯基)-5,6,7,8-四氢喹啉-2-基)丁-3-烯-1-基)氨基甲酸叔丁酯(3J)
Figure PCTCN2018072988-appb-000185
在-78℃下,将氯甲酸甲酯(1.00g,0.82mL,10.59mmol)的二氯甲烷溶液(50mL)缓慢滴加到3I(4.22g,9.62mmol)和吡啶(2.31g,2.3mL,29.03mmol)的二氯甲烷(100mL)溶液中,滴加完毕后,在-78℃下继续搅拌2小时。停止反应,向体系中加入饱和氯化铵溶液(10mL),搅拌5分钟后,升温到室温继续搅拌10分钟。加入二氯甲烷(100mL),体系用水(50mL×2)和饱和食盐水(30mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩后,经硅胶柱层析(石油醚/乙酸乙酯(v/v)=5/1)提纯,得暗红色晶体(4.35g,91.0%)。
MS(ESI,pos.ion)m/z:497.5[M+1] +.
步骤11:(S)-(1-(4-(4-(甲氧基酰氨基)-2-氨基苯基)-5,6,7,8-四氢喹啉-2-基)丁-3-烯-1-基)氨基甲酸叔丁酯(3K)
Figure PCTCN2018072988-appb-000186
在室温下,将锌粉(4.51g,69.03mmol)和氯化铵(3.71g,69.0mmol)加到含有3J(3.41g,6.87mmol)的甲醇(60mL)溶液中,体系在室温下搅拌2.5小时。停止反应,过滤,滤饼用甲醇(10mL×3)洗涤,滤液浓缩,残渣用二氯甲烷(100mL)溶解,用水(30mL×2)和饱和食盐水(30mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩,得粗产品,用硅胶柱层析(石油醚/乙酸乙酯(v/v)=3/1)提纯,得到黄色晶体(2.02g,63.0%)。
MS(ESI,pos.ion)m/z:467.3[M+1] +.
步骤12:(S)-(1-(4-(4-(甲氧基酰氨基)-2-((R)-2-甲基丁-3-烯酰氨基)苯基)-5,6,7,8-四氢喹啉-2-基)丁-3-烯-1-基) 氨基甲酸叔丁酯(3L)
Figure PCTCN2018072988-appb-000187
在0℃下,将含有(R)-2-甲基丁-3-烯酰氯(1.54g,13.01mmol)的二氯甲烷溶液(10mL)缓慢加入到溶有3K(2.02g,4.33mmol)和吡啶(1.41g,1.4mL,17.02mmol)的干燥二氯甲烷(100mL)中,在0℃下搅拌30分钟后,升温到室温,继续搅拌5小时。停止反应,向体系中再加入二氯甲烷(100mL),再用水(50mL×2)和饱和食盐水(50mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩得粗产品,用硅胶柱层析(石油醚/乙酸乙酯(v/v)=3/1)提纯,得到淡黄色固体(1.15g,48.4%)。
MS(ESI,pos.ion)m/z:549.3[M+1] +.
步骤13:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-8,16-二氮杂四环[13.7.1.0 2.7.0 17,22]二十三烷 -1(23),2(7),3,5,11(12),15,17(22)-七烯-5-基)氨基甲酸甲酯(3M)
Figure PCTCN2018072988-appb-000188
在室温下,将一水合对甲苯磺酸(0.45g,2.30mmol)放置到两口瓶中,加热到80℃,减压干燥约1小时。冷却到室温,然后将溶有3L(1.15g,2.10mmol)的干燥二氯甲烷(100mL)溶液注射到体系中,搅拌30分钟。再将Grubbs 2代催化剂(0.54g,0.63mmol)的干燥二氯甲烷(20mL)溶液注射到体系中,升温至回流,搅拌过夜。停止反应,冷却至室温,向体系中加入二氯甲烷(100mL)和饱和碳酸氢钠水溶液(50mL),收集有机相,水相再用二氯甲烷(30mL×3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩,得到粗产品经硅胶柱层析(石油醚/乙酸乙酯(v/v)=1/1)提纯,得到淡黄色固体(0.74g,67.8%)。MS(ESI,pos.ion)m/z:521.6[M+1] +.
步骤14:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-8,16-二氮杂四环[13.7.1.0 2.7.0 17,22]二十三烷 -1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯(3N)
Figure PCTCN2018072988-appb-000189
在室温下,将10%的Pd/C(0.07g)加到溶有3M(0.69g,1.33mmol)的无水甲醇(20mL)溶液中,用氢气置换2-3次,在氢气氛围下,室温搅拌过夜,停止反应。过滤,滤液浓缩得到淡黄色固体(0.695g,100.0%)。
MS(ESI,pos.ion)m/z:523.4[M+1] +.
步骤15:N-((10R,14S)-14-氨基-10-甲基-9-氧代-8,16-二氮杂四环[13.7.1.0 2.7.0 17,22]二十三烷 -1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯(3O)
Figure PCTCN2018072988-appb-000190
在室温下,将三氟乙酸(4.61g,3.0mL,40.02mmol)加到3N(0.69g,1.33mmol)的干燥二氯甲烷溶液(30mL)中,然后室温搅拌8小时。停止反应,减压浓缩得到暗红色固体,加入乙酸乙酯(50mL)和饱和碳酸氢钠溶液(10mL),搅拌10分钟,收集有机相,水相再用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩,经硅胶柱层析(二氯甲烷/甲醇(v/v)=20/1)纯化,得到白色固体(0.56g,99.8%)。
MS(ESI,pos.ion)m/z:423.5[M+1] +.
步骤16:N-((10R,14S)-14-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙氨基)-10-甲基-9-氧代-8,16-二氮杂四环 [13.7.1.0 2.7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯(3P)
Figure PCTCN2018072988-appb-000191
在室温下,将三乙胺(0.73g,1.0mL,7.21mmol)加到3O(0.56g,1.33mmol)的四氢呋喃(20mL)溶液中,再加入1P(0.52g,1.33mmol)的四氢呋喃(10mL)溶液,体系室温搅拌过夜。加入乙酸乙酯(50mL)和水(20mL),分液,收集有机相,水相用乙酸乙酯(15mL×3)萃取,合并有机相用无水硫酸钠干燥,过滤,滤液浓缩,得到固体,再用中性氧化铝柱层析(石油醚/乙酸乙酯(v/v)=1/1)提纯,得到褐色固体(0.26g,28.7%)。
MS(ESI,pos.ion)m/z:687.0[M+1] +.
步骤17:N-((10R,14S)-14-(N-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙基)-2-(二乙氧基磷酰基)乙酰氨基)-10-甲基-9- 氧代-8,16-二氮杂四环[13.7.1.0 2.7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯(3Q)
Figure PCTCN2018072988-appb-000192
室温下,将吡啶(0.098g,0.1mL,1.24mmol)和4-二甲氨基吡啶(0.025g,0.20mmol)加到3P(0.13g,0.19mmol)的二氯甲烷(10mL)溶液中,再加入(2-氯-2-氧代乙基)磷酸二乙酯(0.11g,0.47mmol)的二氯甲烷(10mL)溶液,微波升温到40℃室温搅拌2小时。停止反应,冷却室温,加入乙酸乙酯(50mL)和水(20mL),收集有机相,水相用乙酸乙酯(15mL×3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩,得到白色固体(0.16g,99.54%)。
MS(ESI,pos.ion)m/z:865.0[M+1] +.
步骤18:N-((10R,14S)-14-(4-(6-溴-3-氯-4-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-8,16-二氮 杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯
Figure PCTCN2018072988-appb-000193
在室温下,将60%氢化钠(0.066g,1.65mmol)置于两口瓶中,用氮气保护后,用注射器注入3Q(0.72g,0.83mmol)的干燥四氢呋喃(5mL)溶液,室温搅拌30分钟。停止反应,加入二氯甲烷(50mL)和饱和氯化铵(15mL)溶液,收集有机相,水相用二氯甲烷(20mL×3)萃取,合并有机相,浓缩得淡黄色固体,经中性氧化铝柱层析(石油醚/乙酸乙酯(v/v)=3/2)提纯,得到灰白色固体(0.42g,71.0%)。
MS(ESI,pos.ion)m/z:711.6[M+1] +
1H NMR(400MHz,CDCl 3)δ(ppm)8.00(s,1H),7.44(d,J=5.8Hz,2H),7.38(d,J=8.5Hz,1H),7.08(d,J=8.4Hz,1H),6.80(s,1H),5.95(s,1H),5.62(dd,J=11.6,4.3Hz,1H),4.68–4.55(m,1H),3.98-3.91(m,1H),3.59(s,3H),2.94-2.72(m,4H),2.61-2.55(m,1H),2.44–2.37(m,2H),2.09-1.99(m,2H),1.98-1.88(m,2H),1.86-1.70(m,4H),1.64–1.45(m,4H),0.97(d,J=6.9Hz,3H).
实施例4
N-((10R,14S)-14-(4-(6-溴-3-氯-4-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-21-氧杂-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯
Figure PCTCN2018072988-appb-000194
步骤1:6-(羟甲基)-2-乙烯基吡啶-3-醇(4A)
Figure PCTCN2018072988-appb-000195
氮气保护下,在四口瓶中依次加入2-溴-6-(羟甲基)吡啶-3-醇(40g,196.1mmol)、碳酸钠(42g,396.3mmol)的水溶液(200mL)、三环己基膦(8.30g,29.6mmol)、(dba) 3Pd 2(9.00g,9.83mmol)、4,4,5,5-四甲基-2-乙烯基-1,3,2-二氧硼戊环(36.0mL,212mmol)和N,N-二甲基甲酰胺(900mL),然后加热至100℃搅拌过夜。冷至室温,过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得黄色油状物(28g,94.5%)。
MS(ESI,pos.ion)m/z:152.2[M+1] +.
步骤2:(5-(烯丙氧基)-6-乙烯基吡啶-2-基)甲醇(4B)
Figure PCTCN2018072988-appb-000196
在0℃下,将2A(28g,185.2mmol)和碳酸钾(50g,361.8mmol)加入到N,N-二甲基甲酰胺(300mL)中,然后缓慢滴加烯丙基溴(16.5mL,191mmol),滴加完毕后移至室温下反应2小时。加入水淬灭反应,用乙酸乙酯(200mL×3)萃取。合并有机相,依次用水(200mL×2)、饱和食盐水(200mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=4/1),得淡黄色油状物(31g,87.5%)。
MS(ESI,pos.ion)m/z:192.2[M+1] +.
步骤3:(2H-吡喃并[3,2-b]吡啶-6-基)甲醇(4C)
Figure PCTCN2018072988-appb-000197
氮气保护下,将4B(11g,57.5mmol)溶于无水甲苯(300mL)中,加入Grubbs 2代催化剂(10.0g,11.8mmol),在室温下搅拌30分钟后,加热至60℃搅拌3小时。冷至室温,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得黑色固体(5g,53.3%)。
MS(ESI,pos.ion)m/z:164.0[M+1] +.
步骤4:(3,4-二氢-2H-吡喃并[3,2-b]吡啶-6-基)甲醇(4D)
Figure PCTCN2018072988-appb-000198
将4C(14g,85.8mmol)溶于无水甲醇(150mL)中,然后加入10%的Pd/C(5.0g,4.7mmol),在氢气氛围下室温搅拌6小时。经硅藻土过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得棕黄色固体(10g,70.6%)。
MS(ESI,pos.ion)m/z:166.2[M+1] +.
步骤5:6-(羟甲基)-3,4-二氢-2H-吡喃并[3,2-b]吡啶5-氧化物(4E)
Figure PCTCN2018072988-appb-000199
将4D(3.50g,21.2mmol)溶于二氯甲烷(100mL)中,降温至0℃,加入间氯过氧苯甲酸(4.70g,27.2mmol),然后移至室温下反应5小时。减压蒸去溶剂,粗产品经硅胶柱层析纯化(乙酸乙酯),得淡黄色固体(3.10g,80.8%)。
步骤6:8-溴-6-(溴甲基)-3,4-二氢-2H-吡喃并[3,2-b]吡啶(4F)
Figure PCTCN2018072988-appb-000200
将4E(1.0g,5.5mmol)溶于N,N-二甲基甲酰胺(60mL)和甲苯(30mL)中,缓慢加入三溴氧磷(3.2g,11mmol),在室温下搅拌30分钟后,再加入三溴氧磷(3.2g,11mmol),然后继续在室温下搅拌2小时。用乙酸乙酯(100mL×2)萃取,合并有机相,依次用饱和碳酸氢钠溶液(50mL)和水(50mL)洗涤,合并有机相,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色油状物(1.30g,77%)。
MS(ESI,pos.ion)m/z:308.0[M+1] +.
步骤7:(8-溴-3,4-二氢-2H-吡喃并[3,2-b]吡啶-6-基)甲醇(4G)
Figure PCTCN2018072988-appb-000201
将4F(5.0g,16mmol)溶于丙酮(100mL)中,然后加入20%的碳酸钠水溶液(80mL),加热至70℃搅拌10小时。冷至室温,用乙酸乙酯(200mL×2)萃取。合并有机相,依次用水(50mL)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得白色固体(2.5g,63%)。
MS(ESI,pos.ion)m/z:244.1[M+1] +.
步骤8:8-溴-3,4-二氢-2H-吡喃并[3,2-b]吡啶-6-甲醛(4H)
Figure PCTCN2018072988-appb-000202
将4G(5.15g,21.1mmol)溶于二氯甲烷(150mL)中,然后加入Dess-Martin氧化剂(10.00g,23.58mmol),体系在室温下搅拌1小时。减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=4/1),得白色固体(4.2g,82%)。
MS(ESI,pos.ion)m/z:242.05[M+1] +.
步骤9:(S,E)-N-((8-溴-3,4-二氢-2H-吡喃并[3,2-b]吡啶-6-基)亚甲基)-2-甲基丙烷-2-亚磺酰胺(4I)
Figure PCTCN2018072988-appb-000203
将4H(6.60g,27.3mmol)溶于二氯甲烷(150mL)中,加入(S)-(-)-叔丁基亚磺酰胺(5.00g,41.3mmol)和无水硫酸铜(22.0g,138mmol),体系在室温下搅拌2天。停止反应,硅藻土过滤,用二氯甲烷洗涤滤饼。收集滤液,依次用水(100mL×2)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=4/1),得白色固体(9.0g,96%)。
MS(ESI,pos.ion)m/z:347.0[M+1] +.
步骤10:(S)-N-((S)-1-(8-溴-3,4-二氢-2H-吡喃并[3,2-b]吡啶-6-基)丁-3-烯-1-基)-2-甲基丙烷-2-亚磺酰胺(4J)
Figure PCTCN2018072988-appb-000204
在-20℃左右时,将烯丙基溴化镁(80.0mL,80.0mmol,1.0mol/L的四氢呋喃溶液)缓慢滴加到溶有三氯化铟(18.0g,81.4mmol)的无水四氢呋喃(100mL)中,滴加完毕,体系升温到室温搅拌1小时。冷至-20℃,向体系中滴加溶有4I(9.50g,27.5mmol)的乙醇(120mL)溶液,滴加完毕后,继续反应2小时。过滤,用乙酸乙酯(300mL×2)萃取。合并有机相,依次用水(200mL×2)、饱和食盐水(200mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得黄色油状物(10.5g,98.5%)。
MS(ESI,pos.ion)m/z:389.15[M+1] +.
步骤11:(S)-1-(8-溴-3,4-二氢-2H-吡喃并[3,2-b]吡啶-6-基)丁-3-烯-1-胺(4K)
Figure PCTCN2018072988-appb-000205
将4J(10.5g,27.1mmol)溶于无水甲醇(50mL)中,然后加入4mol/L的盐酸(60mL,240mmol)溶液,体系在室温下搅拌0.5小时。减压蒸去溶剂,用饱和碳酸氢钠溶液中和后用乙酸乙酯(500mL×2)萃取。合并有机相,依次用水(200mL)、饱和食盐水(100mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色油状物(7.60g,99.0%)。粗产物直接用于下一步反应。
MS(ESI,pos.ion)m/z:283.15[M+1] +.
步骤12:(S)-(1-(8-溴-3,4-二氢-2H-吡喃并[3,2-b]吡啶-6-基)丁-3-烯-1-基)氨基甲酸叔丁酯(4L)
Figure PCTCN2018072988-appb-000206
将4K(7.60g,26.8mmol)溶于二氯甲烷(150mL)中,然后加入三乙胺(8.00mL,57.4mmol)和Boc酸酐(7.10mL,31mmol),在室温下搅拌3小时。用二氯甲烷(200mL×2)萃取,合并有机相,依次用水(100mL×2)、饱和食盐水(100mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色油状物(10.0g,97.2%)。MS(ESI,pos.ion)m/z:383.15[M+1] +.
步骤13:(S)-(1-(8-(4-氨基-2-硝基苯基)-3,4-二氢-2H-吡喃并[3,2-b]吡啶-6-基)丁-3-烯-1-基)氨基甲酸叔丁酯 (4M)
Figure PCTCN2018072988-appb-000207
氮气保护下,在两口瓶中依次加入4L(10.0g,26.1mmol)、碳酸钠(6.00g,56.6mmol)的水(28mL)溶液、三环己基膦(1.10g,3.92mmol)、(dba) 3Pd 2(2.50g,2.73mmol)、3-硝基-4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯胺(10.0g,37.9mmol)和N,N-二甲基甲酰胺(300mL),加热至90℃搅拌过夜。冷至室温,用乙酸乙酯(500mL×3)萃取,合并有机相,依次用水(200mL×2)、饱和食盐水(200mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得黄色油状物(8.00g,69.6%)。
MS(ESI,pos.ion)m/z:441.3[M+1] +.
步骤14:(S)-(1-(8-(4-(甲氧基酰氨基)-2-硝基苯基)-3,4-二氢-2H-吡喃并[3,2-b]吡啶-6-基)丁-3-烯-1-基)氨基甲 酸叔丁酯(4N)
Figure PCTCN2018072988-appb-000208
在-70℃下,将氯甲酸甲酯(1.55mL,20.1mmol)缓慢滴加到4M(7.50g,17.0mmol)和吡啶(4.50mL,55.9mmol)的二氯甲烷(100mL)溶液中,体系在-70℃下搅拌2小时。用饱和氯化铵溶液淬灭反应,然后用二氯甲烷(200mL×2)萃取。合并有机相,依次用水(100mL)、饱和食盐水(100mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色固体(7.2g,85%)。
MS(ESI,pos.ion)m/z:499.3[M+1] +.
步骤15:(S)-(1-(8-(4-(甲氧基酰氨基)-2-氨基苯基)-3,4-二氢-2H-吡喃并[3,2-b]吡啶-6-基)丁-3-烯-1-基)氨基甲 酸叔丁酯(4O)
Figure PCTCN2018072988-appb-000209
将4N(7.20g,14.4mmol)溶于甲醇(200mL)中,加入锌粉(9.50g,145mmol)和氯化铵(8.0g,150mmol),体系在室温下搅拌过夜。过滤,减压蒸去溶剂,用二氯甲烷(100mL×2)萃取。合并有机相,依次用水(100mL)、饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色固体(5.50g,81.3%)。
MS(ESI,pos.ion)m/z:469.3[M+1] +.
步骤16:(S)-(1-(8-(4-(甲氧基酰氨基)-2-((R)-2-甲基丁-3-烯酰氨基)苯基)-3,4-二氢-2H-吡喃并[3,2-b]吡啶-6- 基)丁-3-烯-1-基)氨基甲酸叔丁酯(4P)
Figure PCTCN2018072988-appb-000210
在0℃下,将4O(3.0g,6.4mmol)溶于二氯甲烷(20mL)中,加入吡啶(2.10mL,26.0mmol)和(R)-2-甲基丁-3-烯酰氯(1.0g,8.4mmol),继续在0℃下搅拌1小时后,移至室温下搅拌3小时。用二氯甲烷(100mL×3)萃取,合并有机相,依次用水(50mL×2)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得淡黄色泡沫状固体(2.3g,65%)。
MS(ESI,pos.ion)m/z:551.2[M+1] +.
步骤17:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-21-氧杂-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十 三烷-1(23),2(3),5,6,11(12),15,17(22)-七烯-5-基)氨基甲酸甲酯(4Q)
Figure PCTCN2018072988-appb-000211
在室温下,将一水合对甲苯磺酸(650mg,3.42mmol)放置到两口瓶中,加热到80℃,减压干燥约30分钟。然后将4P(1.70g,3.09mmol)的干燥二氯甲烷(90mL)溶液注射到冷却到45℃的体系中,继续在45℃下搅拌30分钟。再将Grubbs 2代催化剂(1.05g,1.24mmol)的干燥二氯甲烷(50mL)溶液缓慢注射到体系中,滴加完毕后在45℃下搅拌过夜。冷至室温,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得棕色固体(650mg,40.3%)。
MS(ESI,pos.ion)m/z:523.2[M+1] +.
步骤18:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-21-氧杂-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十 三烷-1(23),2(3),5,6,15,17(22)-六烯-5-基)氨基甲酸甲酯(4R)
Figure PCTCN2018072988-appb-000212
将4Q(990mg,1.894mmol)溶于甲醇(150mL)中,加入10%的Pd/C(200mg,0.188mmol),在4.0MPa的氢气氛围下搅拌过夜。经硅藻土过滤,减压蒸去溶剂,得棕色固体(893mg,89.86%)。
MS(ESI,pos.ion)m/z:525.2[M+1] +.
步骤19:N-((10R,14S)-14-氨基-10-甲基-9-氧代-21-氧杂-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷 -1(23),2(3),5,6,15,17(22)-六烯-5-基)氨基甲酸甲酯(4S)
Figure PCTCN2018072988-appb-000213
将4R(890mg,1.70mmol)溶于二氯甲烷(10mL)中,加入三氟乙酸(2.0mL,27mmol),室温下搅拌过夜。用饱和碳酸氢钠溶液中和反应液,然后用二氯甲烷(50mL×3)萃取。合并有机相,依次用水(20mL×2)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得棕色固体(700mg,97.43%)。MS(ESI,pos.ion)m/z:425.0[M+1] +.
步骤20:N-((10R,14S)-14-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙氨基)-10-甲基-9-氧代-21-氧杂-8,16-二氮杂四环 [13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(3),5,6,15,17(22)-六烯-5-基)氨基甲酸甲酯(4T)
Figure PCTCN2018072988-appb-000214
将4S(700mg,1.65mmol)溶于四氢呋喃(10mL)中,加入三乙胺(1.20mL,8.63mmol),再加入1P(800mg,2.0439mmol)的四氢呋喃(10mL)溶液,然后在室温下搅拌5小时。用乙酸乙酯(50mL×3)萃取,合并有机相,依次用水(20mL×2)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得棕色固体(500mg,44.07%)。
MS(ESI,pos.ion)m/z:689.8[M+1] +.
步骤21:N-((10R,14S)-14-(N-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙基)-2-(二乙氧基磷酰基)乙酰氨基)-10-甲基-9- 氧代-21-氧杂-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(3),5,6,15,17(22)-六烯-5-基)氨基甲酸甲酯 (4U)
Figure PCTCN2018072988-appb-000215
在微波管中加入4T(400mg,0.581mmol)、(2-氯-2-氧代乙基)磷酸二乙酯(700mg,3.26mmol)、吡啶(0.25mL,3.1mmol)、4-二甲氨基吡啶(70mg,0.573mmol)和二氯甲烷(15mL),然后在微波下加热至50℃反应3小时。冷却至室温,用二氯甲烷(50mL×2)萃取,合并有机相,依次用水(50mL)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色固体(420mg,83.41%)。
MS(ESI,pos.ion)m/z:867.5[M+1] +.
步骤22:N-((10R,14S)-14-(4-(6-溴-3-氯-4-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-21-氧杂 -8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯
在氮气保护下,将4U(500mg,0.577mmol)的四氢呋喃(100mL)溶液加入到60%氢化钠(140mg,3.50mmol)中,然后在室温下搅拌1小时。加水(10mL)淬灭反应,用乙酸乙酯(50mL×2)萃取。合并有机相,依次用水(50mL)、饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得淡黄色固体,经制备色谱进一步纯化得白色固体(170mg,41.36%)。
MS(ESI,pos.ion)m/z:713.4[M+1] +
1H NMR(400MHz,CDCl 3)δ(ppm)9.58(s,1H),7.96(s,1H),7.59(s,2H),7.40(d,J=8.7Hz,1H),7.33(d,J=7.9Hz,2H),6.00(s,1H),5.05(s,2H),4.39(s,2H),3.80(s,4H),3.28(s,2H),2.73(s,2H),2.64–2.39(m,1H), 2.28(s,2H),2.06-1.76(m,2H),1.68-1.52(m,2H),1.28(s,3H),1.10-0.97(m,2H).
实施例5
N-((10R,14S)-14-(4-(6-溴-3-氯-2-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-20-氧杂-8,16-二氮杂四环[13.6.1.0 2,7.0 17,21]二十二烷-1(22),2(7),3,5,15,17(21)-六烯-5-基)氨基甲酸甲酯
Figure PCTCN2018072988-appb-000216
步骤1:2-溴-6-(羟甲基)吡啶-3-醇(5A)
Figure PCTCN2018072988-appb-000217
称取氢氧化钾(38g,677.3mmol)于烧瓶中,向其中加入水(260mL)、2-溴吡啶-3-醇(100g,574.71mmol)、EDTA-2Na(4.36g,13.0mmol)和甲醛水溶液(38%,156mL),体系升温至90℃反应5小时。停止反应,冷却至室温,加入乙酸中和反应,乙酸乙酯(100mL×2)萃取,有机相依次用水(50mL)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥,旋干,得浅黄色固体(101g,86.14%),所得产品直接用于下一步反应。
步骤2:(2-(三甲基硅基)呋喃并[3,2-b]吡啶-5-基)甲醇(5B)
Figure PCTCN2018072988-appb-000218
称取5A(1.0g,4.9mmol)、双(三苯基膦)二氯化钯(II)(80mg,0.1140mmol)和碘化亚铜(40mg,0.21mmol)于双口瓶中,氮气置换三次,向其中加入四氢呋喃(20mL)、三乙胺(3.4mL,25mmol)和三甲基硅基乙炔(1.0mL,7mmol),体系升温至80℃反应24小时。反应结束后,冷至室温,经硅藻土过滤,旋转蒸发除去溶剂,残留物经硅胶柱层析(石油醚/乙酸乙酯(v/v)=2/1),纯化得黄色固体(0.50g,46%)。
MS(ESI,pos.ion)m/z:222.2[M+1] +
1H NMR(400MHz,CDCl 3)δ(ppm)7.76(d,J=8.0Hz,1H),7.16(d,J=7.1Hz,1H),7.13(s,1H),4.87(s,2H),0.39(s,9H).
步骤3:呋喃并[3,2-b]吡啶-5-基甲醇(5C)
Figure PCTCN2018072988-appb-000219
称取5B(10.0g,45.2mmol)于烧瓶中,向其中加入甲醇(100mL),搅拌下加入氢氧化钠(2.17g,54.3mmol)的水(100mL)溶液,室温反应24小时。加入水淬灭反应,旋转蒸发除去甲醇,乙酸乙酯(80mL×2)萃取,合并有机相,无水硫酸钠干燥。过滤,减压蒸除溶剂,残渣用硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=5/1),得淡黄色油状物(3.0g,45%)。
步骤4:(2,3-二氢呋喃并[3,2-b]吡啶-5-基)甲醇(5D)
Figure PCTCN2018072988-appb-000220
称取5C(1.0g,6.71mmol)于高压釜中,向其中加入乙醇(15mL)、冰乙酸(5mL)和10%的Pd/C(0.6g),在氢气氛围下(3MPa),室温反应5小时。停止反应,经硅藻土过滤,甲醇洗涤,滤液旋干,得无色油状物(0.6g,58%),所得产品直接用于下一步反应。
MS(ESI,pos.ion)m/z:152.25[M+1] +.
步骤5:5-(羟甲基)-2,3-二氢呋喃并[3,2-b]吡啶4-氧化物(5E)
Figure PCTCN2018072988-appb-000221
称取5D(23.06g,152.7mmol)于烧瓶中,向其中加入二氯甲烷(500mL)和间氯过氧苯甲酸(31g,152.7mmol),室温反应24小时。停止反应,旋转蒸发除去溶剂,残留物经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得白色固体(25g,98%)。
步骤6:7-溴-5-(溴甲基)-2,3-二氢呋喃并[3,2-b]吡啶(5F)
Figure PCTCN2018072988-appb-000222
称取5E(27g,161.52mmol)于烧瓶中,向其中加入N,N-二甲基甲酰胺(1L)、甲苯(0.5L)和三溴氧磷(185g,645mmol),室温反应2小时。加水(500mL)淬灭反应,加入固体碳酸钠中和至中性,乙酸乙酯(500mL×2)萃取,合并有机相,水洗(500mL),饱和食盐水洗涤(200mL),无水硫酸钠干燥,过滤,旋干,粗产品直接用于下一步反应。
步骤7:(7-溴-2,3-二氢呋喃并[3,2-b]吡啶-5-基)甲醇(5G)
Figure PCTCN2018072988-appb-000223
称取5F(15g,51.202mmol)于烧瓶中,向其中加入丙酮(200mL)、水(200mL)和碳酸钾(30g,217.1mmol),体系升温至70℃反应过夜。终止反应,冷却至室温,旋转蒸发除去丙酮,二氯甲烷萃取(100mL×8),合并有机相,无水硫酸钠干燥,旋干,残留物经硅胶柱层析(石油醚/乙酸乙酯(v/v)=3/1),得白色固体(11.2g,95.1%)。
MS(ESI,pos.ion)m/z:232.1[M+1] +.
步骤8:7-溴-2,3-二氢呋喃并[3,2-b]吡啶-5-甲醛(5H)
Figure PCTCN2018072988-appb-000224
称取5G(5.6g,24mmol)于烧瓶中,向其中加入二氯甲烷(120mL)和Dess-Martin氧化剂(15.5g,36.5mmol),室温下搅拌1小时。停止反应,浓缩反应液,残余物经硅胶柱层析分离(石油醚/乙酸乙酯(v/v)=10/1)纯化,得白色固体(4.83g,87%)。
MS(ESI,pos.ion)m/z:228.0[M+1] +
1H NMR(400MHz,CDCl 3)δ(ppm)9.92(d,J=7.6Hz,1H),9.88(s,1H),7.95(s,1H),7.79(d,J=8.3Hz,1H),7.10(d,J=8.3Hz,1H),4.88(t,J=9.0Hz,2H),4.79(t,J=8.9Hz,1H),3.50(t,J=8.9Hz,2H),3.41(t,J=8.9Hz,2H).
步骤9:(S,E)-N-((7-溴-2,3-二氢呋喃并[3,2-b]吡啶-5-基)亚甲基)-2-甲基丙烷-2-亚磺酰胺(5I)
Figure PCTCN2018072988-appb-000225
室温下,将(S)-(-)-叔丁基亚磺酰胺(8.93g,73.7mmol)和硫酸铜(17.81g,111.6mmol)加入到5H(8.16g,35.8mmol)的二氯甲烷(300mL)溶液中,得到的混合体系在室温下搅拌18小时。终止反应,反应液经硅藻土抽滤,滤液旋干,得棕红色油状物(18.66g,100%)。
步骤10:(S)-N-((S)-1-(7-溴-2,3-二氢呋喃并[3,2-b]吡啶-5-基)丁-3-烯-1-基)-2-甲基丙烷-2-亚磺酰胺(5J)
Figure PCTCN2018072988-appb-000226
在-5℃条件下,将烯丙基溴化镁(175mL,175mmol,1mol/L的四氢呋喃溶液)逐滴滴入三氯化铟(25.43g,115.0mmol)的四氢呋喃(300mL)溶液中,滴加完毕后移至室温下反应1小时。向反应液中逐滴滴加5I(18.66g,56.34mmol)的乙醇(170mL)溶液,滴加完毕后在该温度下反应2小时。终止反应,反应液经过硅藻土抽滤,并用乙酸乙酯洗涤(30mL×3),旋干滤液,残余物用乙酸乙酯(50mL)溶解后,用水洗涤(30mL),有机层用无水硫酸钠干燥,过滤,旋干,残留物经硅胶柱层析(石油醚/乙酸乙酯(v/v)=1/2)纯化,得棕黄色油状物(16.1g,76.6%)。
MS(ESI,pos.ion)m/z:374.0[M+1] +.
步骤11:(S)-1-(7-溴-2,3-二氢呋喃并[3,2-b]吡啶-5-基)丁-3-烯-1-胺(5K)
Figure PCTCN2018072988-appb-000227
称取5J(16g,42.86mmol)于烧瓶中,向瓶中加入甲醇(200mL)及6mol/L的盐酸溶液(230mL,1380mmol),混合体系在室温下搅拌1小时。停止反应,减压旋出甲醇,然后向瓶中加入饱和碳酸钠溶液至碱性,二氯甲烷萃取(200mL×2),干燥,过滤,旋干得到棕红色液体(11.21g,97.18%)。
MS(ESI,pos.ion)m/z:270.05[M+1] +.
步骤12:(S)-(1-(7-溴-2,3-二氢呋喃并[3,2-b]吡啶-5-基)丁-3-烯-1-基)氨基甲酸叔丁酯(5L)
Figure PCTCN2018072988-appb-000228
称取5K(11.21g,41.65mmol)于烧瓶中,向其中加入二氯甲烷(300mL)和三乙胺(15mL,108mmol),然后向体系中逐滴滴加二碳酸二叔丁酯(17mL,73.9mmol),滴加完毕后室温条件下反应5小时。向体系中加入二氯甲烷(200mL),分别用水(300mL)和饱和食盐水(300mL)洗涤,有机相用无水硫酸钠干燥,过滤,旋干,得黄色油状物(22.16g,100%)。
MS(ESI,pos.ion)m/z:370.2[M+1] +.
步骤13:(S)-(1-(7-(4-氨基-2-硝基苯基)-2,3-二氢呋喃并[3,2-b]吡啶-5-基)丁-3-烯-1-基)氨基甲酸叔丁酯(5M)
Figure PCTCN2018072988-appb-000229
称取5L(22.16g,60.02mmol)、3-硝基-4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯胺(10g,37.86mmol)、N,N-二甲基甲酰胺(250mL)和2mol/L的碳酸钠溶液(50mL,100mmol)于烧瓶中,向其中迅速加入三环己基膦(1.9g,6.8mmol),用氮气置换掉体系中含有的氧气,然后迅速加入(dba) 3Pd 2(4.3g,4.7mmol),再用氮气置换掉体系中含有的氧气,体系于100℃下反应12小时。终止反应,反应液经硅藻土抽滤,向滤液中加入乙酸乙酯(400mL),有机相分别用水(250mL×3)和饱和食盐水(250mL)洗涤,无水硫酸钠干燥,过滤,残留物经硅胶柱层析(石油醚/乙酸乙酯(v/v)=5/1)分离,得棕黄色油状物(7.8g,30%)。
MS(ESI,pos.ion)m/z:427.2[M+1] +.
步骤14:(S)-(1-(7-(4-(甲氧基酰氨基)-2-硝基苯基)-2,3-二氢呋喃并[3,2-b]吡啶-5-基)丁-3-烯-1-基)氨基甲酸叔 丁酯(5N)
Figure PCTCN2018072988-appb-000230
将5M(8.2g,19mmol)和吡啶(4.8mL,60mmol)加入双口瓶中,向其中加入二氯甲烷(200mL),冷至-78℃,向体系中缓慢滴加氯甲酸甲酯(1.7mL,22mmol)的二氯甲烷(100mL)溶液,滴加完毕后在-78℃下搅拌12小时。终止反应,向体系中加入饱和氯化铵(20mL)并搅拌5分钟,然后升至室温搅拌10分钟。向体系中加入二氯甲烷(200mL),体系分别用水(200mL)和饱和食盐水(200mL)洗涤,无水硫酸钠干燥,过滤,滤液旋干,得棕红色油状物(11.4g,100%)。
MS(ESI,pos.ion)m/z:485.3[M+1] +.
步骤15:(S)-(1-(7-(4-(甲氧基酰氨基)-2-氨基苯基)-2,3-二氢呋喃并[3,2-b]吡啶-5-基)丁-3-烯-1-基)氨基甲酸叔 丁酯(5O)
Figure PCTCN2018072988-appb-000231
室温下,将锌粉(15.36g,234.8mmol)和氯化铵(12.58g,784.2mmol)加入到5N(11.4g,23.5mmol)的甲醇(300mL)溶液中,室温条件下反应12小时。终止反应,反应液经硅藻土抽滤,滤饼用甲醇洗涤(50mL×3),滤液减压旋出溶剂,残余物用二氯甲烷溶解(300mL),有机相分别用水(300mL×2)和饱和食 盐水(300mL)洗涤,无水硫酸钠干燥,过滤,旋干得淡黄色晶体(7.8g,73%)。
MS(ESI,pos.ion)m/z:455.2[M+1] +.
步骤16:(S)-(1-(7-(4-(甲氧基酰氨基)-2-((S)-2-甲基丁-3-烯酰氨基)苯基)-2,3-二氢呋喃并[3,2-b]吡啶-5-基)丁 -3-烯-1-基)氨基甲酸叔丁酯(5P)
Figure PCTCN2018072988-appb-000232
在0℃下,将5O(5.2g,11mmol)溶于二氯甲烷(80mL)中,向反应瓶中加入吡啶(3.7mL,46mmol)。将(R)-2-甲基丁-3-烯酰氯(2.24g,18.9mmol)的二氯甲烷溶液(80mL)逐滴滴加入反应瓶中,滴加完毕后,继续搅拌1小时,然后再移至室温条件下继续反应1小时。终止反应,向反应瓶中加入水(300mL),用二氯甲烷萃取(40mL×10),合并有机相,无水硫酸钠干燥,过滤,旋干,残留物经硅胶柱层析(石油醚/乙酸乙酯(v/v)=5/1)纯化,得淡黄色泡沫状固体(4.5g,73%)。
MS(ESI,pos.ion)m/z:537.2[M+1] +.
步骤17:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-20-氧杂-8,16-二氮杂四环[13.6.1.0 2.7.0 17,21]二十 二烷-1(22),2(3),4,6,11(12),15,17(21)-七烯-5-基)氨基甲酸甲酯(5Q)
Figure PCTCN2018072988-appb-000233
室温下,称取一水合对甲苯磺酸(1.75g,9.20mmol)于两口瓶中,将两口瓶置于60℃油浴中减压干燥1小时。关闭加热,将5P(4.5g,8.4mmol)的干燥二氯甲烷(250mL)溶液注射到冷却至室温的体系中,搅拌10分钟。再将Grubbs 2代催化剂(2.2g,2.6mmol)的干燥二氯甲烷(50mL)溶液滴加到体系中,滴加完毕后体系在45℃下回流搅拌12小时。终止反应,冷却至室温,减压旋蒸除去溶剂,残余物经硅胶柱层析分离(石油醚/乙酸乙酯(v/v)=1/1),得棕色固体(1.92g,45%)。
MS(ESI,pos.ion)m/z:509.4[M+1] +.
步骤18:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-20-氧杂-8,16-二氮杂四环[13.6.1.0 2.7.0 17,21]二十 二烷-1(22),2(3),4,6,15,17(21)-六烯-5-基)氨基甲酸甲酯(5R)
Figure PCTCN2018072988-appb-000234
室温下,称取5Q(1.38g,2.71mmol)加入高压釜中,向其中加入甲醇(100mL)和10%Pd/C(0.3g),充入氢气到5MPa,室温下搅拌8小时。终止反应,反应液经硅藻土过滤,滤饼用甲醇洗涤(30mL×3),滤液经减压蒸发除去溶剂,得到棕褐色固体(1.4g,100%)。
MS(ESI,pos.ion)m/z:511.3[M+1] +.
步骤19:N-((10R,14S)-14-氨基-10-甲基-9-氧代-20-氧杂-8,16-二氮杂四环[13.6.1.0 2.7.0 17,21]二十二烷 -1(22),2(3),4,6,15,17(21)-六烯-5-基)氨基甲酸甲酯(5S)
Figure PCTCN2018072988-appb-000235
在室温条件下,称取5R(1.4g,2.7mmol)于烧瓶中,加入干燥二氯甲烷(80mL)和三氟乙酸(6.2mL,83mmol),室温条件下搅拌10小时。终止反应,减压旋蒸除去溶剂,向反应瓶中加入乙酸乙酯(30mL)和饱和碳酸钠溶液(20mL),室温条件下搅拌10分钟。分出有机相,水相再用乙酸乙酯萃取(40mL×8),合并有机相并用无水硫酸钠干燥,过滤,滤液旋干,得棕红色固体(1.07g,95%)。
MS(ESI,pos.ion)m/z:411.3[M+1] +.
步骤20:N-((10R,14S)-14-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙氨基)-10-甲基-9-氧代-20-氧杂-8,16-二氮杂四环 [13.6.1.0 2.7.0 17,21]二十二烷-1(22),2(3),4,6,15,17(21)-六烯-5-基)氨基甲酸甲酯(5T)
Figure PCTCN2018072988-appb-000236
称取5S(1.07g,2.61mmol)于烧瓶中,向其中加入四氢呋喃(60mL)和三乙胺(2mL,14.4mmol),再加入1P(1.53g,3.91mmol)的四氢呋喃(40mL)溶液,加入完毕后室温下搅拌13小时。终止反应,减压旋蒸除去溶剂,残余物用二氯甲烷(30mL)溶解后倒入分液漏斗中,加入水(300mL),分出有机相,水相再用二氯甲烷萃取(30mL×4),合并有机相并用无水硫酸钠干燥,过滤,滤液旋干,得棕红色固体(1.2g,68%)。
MS(ESI,pos.ion)m/z:674.8[M+1] +.
步骤21:N-((10R,14S)-14-(N-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙基)-2-(二乙氧基磷酰基)乙酰氨基)-10-甲基-9- 氧代-20-氧杂-8,16-二氮杂四环[13.6.1.0 2.7.0 17,21]二十二烷-1(22),2(3),4,6,15,17(21)-六烯-5-基)氨基甲酸甲酯 (5U)
Figure PCTCN2018072988-appb-000237
室温下,分别将吡啶(0.36mL,4.5mmol)、4-二甲氨基吡啶(0.13g,1.1mmol)和(2-氯-2-氧代乙基)磷酸二乙酯(1g,4.6603mmol)加到5T(0.6g,0.9mmol)的二氯甲烷(20mL)溶液中,然后微波加热到50℃反应3小时。终止反应,冷却至室温,加入二氯甲烷(50mL)和水(100mL),分出有机相,水相再用二氯甲烷萃取(30mL×5),合并有机相并用无水硫酸钠干燥,过滤,滤液旋干,得棕褐色固体(0.8g,100%)。
MS(ESI,pos.ion)m/z:853.6[M+1] +.
步骤22:N-((10R,14S)-14-(4-(6-溴-3-氯-2-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-20-氧杂 -8,16-二氮杂四环[13.6.1.0 2,7.0 17,21]二十二烷-1(22),2(7),3,5,15,17(21)-六烯-5-基)氨基甲酸甲酯
称取60%氢化钠(0.05g,1mmol)于烧瓶中,氮气保护下向瓶中加入四氢呋喃(5mL),搅拌下再向瓶中加入5U(0.2g,0.2mmol)的四氢呋喃(15mL)溶液,室温条件下搅拌1小时。向反应瓶中加入水(15mL)淬灭反应,减压旋蒸除去溶剂,残余物用二氯甲烷(50mL)溶解后倒入分液漏斗中,向其中加入水(200mL),分出有机相,水相再用二氯甲烷萃取(30mL×3),合并有机相并用无水硫酸钠干燥,过滤,旋干,残留物经硅胶柱层析分离(石油醚/乙酸乙酯(v/v)=3/1),得棕红色固体(0.073g,40%)。
MS(ESI,pos.ion)m/z:698.5[M+1] +
1H NMR(400MHz,CDCl 3)δ(ppm)8.73(s,1H),7.84-7.80(s,2H),7.57-7.50(m,2H),7.37(d,J=8.8Hz,1H),7.31(s,1H),7.24(s,1H),5.95(s,1H),,5.25(s,1H),5.11-4.71(m,6H),4.01(s,1H),3.81(s,1H),3.69(s,3H),2.87-2.81(m,1H),2.70-2.61(m,2H),2.51-2.45(m,1H),1.97–1.75(m,2H),1.65-1.56(m,1H),1.04(d,J=5.0Hz,3H).
实施例6
N-((10R,14S)-14-(4-(6-溴-3-氯-4-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-18,21-二氧杂-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯
Figure PCTCN2018072988-appb-000238
步骤1:2-((2-溴-6-(羟甲基)-4-碘吡啶-3-基)氧基)乙醇(6A)
Figure PCTCN2018072988-appb-000239
将2-溴-6-(羟甲基)-4-碘吡啶-3-醇(30g,90.93mmol)和碳酸钾(25g,180.88mmol)溶于N,N-二甲基甲酰胺(200mL)中,然后加入2-溴乙醇(11mL,155mmol),加热至100℃搅拌3小时。冷至室温,用1mol/L的稀盐酸调至pH=4,用乙酸乙酯(300mL×2)萃取。合并有机相,依次用水(200mL)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得白色固体(16.0g,47.1%)。
MS(ESI,pos.ion)m/z:374.0[M+1] +.
步骤2:(8-碘-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6-基)甲醇(6B)
Figure PCTCN2018072988-appb-000240
将6A(650mg,1.74mmol)、粉末状氢氧化钾(200mg,3.56mmol)和18-冠醚-6(700mg,2.65mmol)溶于甲苯(50mL)中,然后加热至135℃搅拌2小时。用1mol/L的稀盐酸调至pH=4,用乙酸乙酯(50mL×2)萃取。合并有机相,依次用水(40mL)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1)白色固体(140mg,27.5%)。
MS(ESI,pos.ion)m/z:294.1[M+1] +
1H NMR(400MHz,CDCl 3)δ(ppm)7.35(s,1H),4.59(s,2H),4.48(dd,J=5.1,2.9Hz,2H),4.38(dd,J=5.1,2.9Hz,2H),2.81(s,1H).
步骤3:8-碘-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6-甲醛(6C)
Figure PCTCN2018072988-appb-000241
将6B(6.10g,20.8mmol)溶于二氯甲烷(150mL)中,然后加入Dess-Martin氧化剂(10.00g,23.58mmol),在室温下搅拌1小时。减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1)得白色固体(6.08g,100%)。
MS(ESI,pos.ion)m/z:292.1[M+1] +.
步骤4:(S,E)-N-((8-碘-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6-基)亚甲基)-2-甲基丙烷-2-亚磺酰胺(6D)
Figure PCTCN2018072988-appb-000242
将6C(6.10g,20.96mmol)溶于二氯甲烷(100mL)中,加入(S)-(-)-叔丁基亚磺酰胺(2.80g,23.1mmol)和无水硫酸铜(10.00g,62.65mmol),然后在室温下搅拌2天。停止反应,经硅藻土过滤,用二氯甲烷洗涤滤饼。收集滤液,减压蒸去溶剂,得棕色固体(8.20g,99.2%)。
MS(ESI,pos.ion)m/z:395.3[M+1] +.
步骤5:(S)-N-((S)-1-(8-碘-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6-基)丁-3-烯-1-基)-2-甲基丙烷-2-亚磺酰胺(6E)
Figure PCTCN2018072988-appb-000243
在-20℃下,将烯丙基溴化镁(2.54mL,2.54mmol,1.0mol/L四氢呋喃溶液)缓慢滴加到溶有三氯化铟(565mg,2.55mmol)的四氢呋喃(9.0mL)溶液中,滴加完毕后体系升温到室温,搅拌0.5小时。在-20℃下,向体系中滴加溶有6D(500mg,1.27mmol)的乙醇(6mL)溶液,滴加完毕后,继续在此温度下搅拌3小时。停止反应,用乙酸乙酯(50mL×2)萃取,合并有机相,依次用水(20mL×2)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得白色固体(432mg,78.1%)。
MS(ESI,pos.ion)m/z:437.3[M+1] +.
步骤6:(S)-1-(8-碘-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6-基)丁-3-烯-1-胺(6F)
Figure PCTCN2018072988-appb-000244
将6E(432mg,0.99mmol)溶于甲醇(7mL)中,然后加入6mol/L的盐酸溶液(3mL,9.3mmol),在室温下搅拌0.5小时。减压蒸去溶剂,得白色固体(320mg,97.0%)。粗产物直接用于下一步反应。
MS(ESI,pos.ion)m/z:332.9[M+1] +.
步骤7:(S)-(1-(8-碘-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6-基)丁-3-烯-1-基)氨基甲酸叔丁酯(6G)
Figure PCTCN2018072988-appb-000245
将6F(320mg,0.96mmol)溶于二氯甲烷(20mL)中,加入三乙胺(0.30mL,2.2mmol)和二碳酸二叔丁酯(0.25mL,1.1mmol),然后在室温下搅拌2小时。停止反应,用二氯甲烷(50mL×2)萃取。合并有机相,依次用水(20mL×2)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色油状物(400mg,96.1%)。
MS(ESI,pos.ion)m/z:432.9[M+1] +.
步骤8:(S)-(1-(8-(4-氨基-2-硝基苯基)-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6-基)丁-3-烯-1-基)氨基甲酸叔丁酯 (6H)
Figure PCTCN2018072988-appb-000246
氮气保护下,向两口瓶中依次加入3-硝基-4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯胺(410mg,1.397mmol)、[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(100mg,0.109mmol)、碳酸铯(600mg,1.8415mmol)、6G(400mg,0.926mmol)、1,4-二氧六环(20.0mL)和水(5.0mL),然后加热至50℃搅拌5小时。停止反应,冷却至室温,用乙酸乙酯(50mL×2)萃取。合并有机相,依次用水(40mL×2)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得黄色固体(255mg,62.3%)。
MS(ESI,pos.ion)m/z:443.5[M+1] +.
步骤9:(S)-(1-(8-(4-(甲氧基酰氨基)-2-硝基苯基)-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6-基)丁-3-烯-1-基)氨基甲 酸叔丁酯(6I)
Figure PCTCN2018072988-appb-000247
在-70℃下,将氯甲酸甲酯(0.050mL,0.65mmol)缓慢滴加到6H(255mg,0.576mmol)和吡啶(0.14mL,1.7mmol)的二氯甲烷(10mL)溶液中,然后在-70℃下搅拌1.5小时。停止反应,用饱和氯化铵溶液淬灭反应,然后用二氯甲烷(50mL×2)萃取。合并有机相,依次用水(40mL)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色固体(285mg,98.8%)。
MS(ESI,pos.ion)m/z:501.5[M+1]+.
步骤10:(S)-(1-(8-(4-(甲氧基酰氨基)-2-氨基苯基)-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6-基)丁-3-烯-1-基)氨基 甲酸叔丁酯(6J)
Figure PCTCN2018072988-appb-000248
将6I(285mg,0.57mmol)溶于甲醇(20mL)中,然后加入锌粉(373mg,5.702mmol)和氯化铵(305mg,5.702mmol),在室温下搅拌2小时。停止反应,过滤,减压蒸去溶剂,得黄色油状物(250mg,93.3%)。MS(ESI,pos.ion)m/z:471.5[M+1] +
步骤11:(S)-(1-(8-(4-(甲氧基酰氨基)-2-((R)-2-甲基丁-3-烯酰氨基)苯基)-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6- 基)丁-3-烯-1-基)氨基甲酸叔丁酯(6K)
Figure PCTCN2018072988-appb-000249
在0℃下,将6J(3.8g,8.1mmol)溶于二氯甲烷(5mL)中,然后加入吡啶(2.6mL,32mmol)和(R)-2-甲基丁-3-烯酰氯(1.1g,9.3mmol),继续在0℃下搅拌1小时,移至室温下搅拌3小时。停止反应,用二氯甲烷(30mL×3)萃取,合并有机相,依次用水(20mL×2)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得淡黄色固体(1.95g,44%)。
MS(ESI,pos.ion)m/z:553.1[M+1] +
步骤12:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-18,21-二氧杂-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22] 二十三烷-1(23),2(7),3,5,11(12),15,17(22)-七烯-5-基)氨基甲酸甲酯(6L)
Figure PCTCN2018072988-appb-000250
在室温下,将一水合对甲苯磺酸(155mg,0.815mmol)放置到两口瓶中,加热到80℃,减压干燥约30分钟。然后将溶有6K(1.85g,3.35mmol)的干燥二氯甲烷(15mL)溶液注射到冷却到室温的体系内, 搅拌30分钟。再将Grubbs 2代催化剂(860mg,1.01mmol)的干燥二氯甲烷(15mL)溶液注射到体系中去,体系升温至回流搅拌过夜。停止反应,冷至室温,用二氯甲烷(50mL×3)萃取。合并有机相,依次用水(50mL×2)、饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得黄色固体(170mg,9.68%)。
MS(ESI,pos.ion)m/z:525.3[M+1] +.
步骤13:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-18,21-二氧杂-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22] 二十三烷-1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯(6M)
Figure PCTCN2018072988-appb-000251
将6L(180mg,0.343mmol)溶于甲醇(50mL)中,然后加入10%Pd/C(50mg,0.047mmol),在3MPa的氢气氛围下搅拌5小时。停止反应,经硅藻土过滤,减压蒸去溶剂,得棕色固体(156mg,86.34%)。所得产品直接用于下一步反应。
MS(ESI,pos.ion)m/z:527.3[M+1] +.
步骤14:N-((10R,14S)-14-氨基-10-甲基-9-氧代-18,21-二氧杂-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷 -1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯(6N)
Figure PCTCN2018072988-appb-000252
将6M(156mg,0.296mmol)溶于二氯甲烷(10mL)中,然后加入三氟乙酸(1mL,13.46mmol),室温下搅拌过夜。停止反应,用饱和碳酸氢钠溶液中和反应液,然后用二氯甲烷(50mL×3)萃取。合并有机相,依次用水(20mL×2)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得棕色固体(110mg,90.3%)。
MS(ESI,pos.ion)m/z:427.3[M+1] +.
步骤15:N-((10R,14S)-14-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙氨基)-10-甲基-9-氧代-18,21-二氧杂-8,16-二氮杂 四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯(6O)
Figure PCTCN2018072988-appb-000253
将6N(110mg,0.258mmol)溶于四氢呋喃(10mL)中,加入三乙胺(0.18mL,1.3mmol),再加入1P(110mg,0.281mmol)的四氢呋喃(10mL)溶液,在室温下搅拌过夜。用乙酸乙酯(50mL×3)萃取,合并有机相,依次用水(20mL×2)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得棕色固体(90mg,50.57%)
MS(ESI,pos.ion)m/z:691.1[M+1] +.
步骤16:N-((10R,14S)-14-(N-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙基)-2-(二乙氧基磷酰基)乙酰氨基)-10-甲基-9- 氧代-18,21-二氧杂-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸 甲酯(6P)
Figure PCTCN2018072988-appb-000254
在微波管中加入6O(90mg,0.1304mmol)、(2-氯-2-氧代乙基)磷酸二乙酯(200mg,0.932mmol)、吡啶(0.1mL,1.24mmol)、4-二甲氨基吡啶(20mg,0.164mmol)和二氯甲烷(15mL),然后微波加热至50℃反应3小时。停止反应,冷却至室温,用二氯甲烷(50mL×2)萃取。合并有机相,依次用水(50mL)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,减压蒸去溶剂,得黄色固体(80mg,70.7%)。
MS(ESI,pos.ion)m/z:869.5[M+1] +.
步骤17:N-((10R,14S)-14-(4-(6-溴-3-氯-4-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-18,21-二氧 杂-8,16-二氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-5-基)氨基甲酸甲酯
在氮气保护下,将6P(100mg,0.115mmol)的四氢呋喃(15mL)溶液加入到60%氢化钠(30mg,0.75mmol)中,然后体系在室温下搅拌1小时。停止反应,加水淬灭反应,用乙酸乙酯(50mL×2)萃取。合并有机相,依次用水(50mL)、饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得淡黄色固体,再经制备色谱纯化得白色固体(10mg,12.2%)。
MS(ESI,pos.ion)m/z:715.2[M+1] +
1H NMR(400MHz,CDCl 3)δ(ppm)8.57(s,1H),7.53-7.49(m,2H),7.39(d,J=8.6Hz,1H),7.32-7.28(m,3H),6.06(s,1H),5.38-5.36(m,2H),4.99(s,2H),4.66-4.56(m,2H),4.41-4.33(m,2H),4.07(s,1H),3.88(s, 1H),3.78(s,3H),2.74-2.64(m,2H),2.38–2.27(m,1H),1.87-1.80(m,2H),1.52-1.41(m,2H),1.03(d,J=5.6Hz,3H).
实施例7
N-((10R,14S)-14-(4-(6-溴-3-氯-2-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-1,8,22-三氮杂四环[13.6.1.0 2,7.0 16,21]二十二烷-2(7),3,5,15(22),16(21),17,19-七烯-5-基)氨基甲酸甲酯
Figure PCTCN2018072988-appb-000255
步骤1:1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑-3-甲醛(7A)
Figure PCTCN2018072988-appb-000256
冰浴下,将1H-吲唑-3-甲醛(10.0g,68.4mmol)溶于二氯甲烷(50mL)中,加入四丁基溴化铵(230mg,0.7134mmol)、50%的氢氧化钾水溶液(50mL)、2-(氯甲氧基)乙基-三甲基-硅烷(13.5mL,76.3mmol),体系继续在冰浴下搅拌1小时,然后移至室温下搅拌过夜。停止反应,用二氯甲烷(200mL×2)萃取,合并有机相,依次用水(100mL×2)和饱和食盐水(100mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色油状物(17.0g,89.9%)。
MS(ESI,pos.ion)m/z:277.2[M+1] +.
步骤2:(S,E)-2-甲基-N-((1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑-3-基)亚甲基)丙烷-2-亚磺酰胺(7B)
Figure PCTCN2018072988-appb-000257
将7A(2.50g,9.04mmol)溶于二氯甲烷(100mL)中,加入(S)-(-)-叔丁基亚磺酰胺(1.30g,10.7mmol)和无水硫酸铜(5.80g,36.3mmol),然后在室温下搅拌过夜。停止反应,经硅藻土过滤,用二氯甲烷洗涤滤饼。收集滤液,依次用水(20mL×2)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得棕色固体(3.16g,92.0%)。
MS(ESI,pos.ion)m/z:380.25[M+1] +.
步骤3:(S)-2-甲基-N-((S)-1-(1-((2-(三甲基硅基)乙氧基)甲基)-1H-吲唑-3-基)丁-3-烯-1-基)丙烷-2-亚磺酰胺 (7C)
Figure PCTCN2018072988-appb-000258
在-20℃下,将烯丙基溴化镁(16.5mL,16.5mmol,1.0mol/L的四氢呋喃溶液)缓慢滴加到溶有三氯化 铟(3.60g,16.3mmol)的四氢呋喃(50mL)中,然后体系升温到室温搅拌1小时。冷至-20℃,向体系中滴加7B(3.00g,7.90mmol)的乙醇(100mL)溶液,滴加完毕后,继续反应3小时。过滤,用乙酸乙酯(100mL×2)萃取,合并有机相,依次用水(100mL×2)、饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得淡黄色油状物(1.96g,58.8%)。MS(ESI,pos.ion)m/z:422.3[M+1] +.
步骤4:(S)-N-((S)-1-(1H-吲唑-3-基)丁-3-烯-1-基)-2-甲基丙烷-2-亚磺酰胺(7D)
Figure PCTCN2018072988-appb-000259
将7C(615mg,1.46mmol)溶于四氢呋喃(20mL)溶液中,加入乙二胺(1.00mL)和四丁基氟化铵(15.0mL,15.0mmol,1.0mol/L的四氢呋喃溶液),然后加热至60℃搅拌3小时。继续补加四丁基氟化铵(15.0mL,15.0mmol,1.0mol/L的四氢呋喃溶液),在60℃下继续搅拌过夜。停止反应,冷至室温,用乙酸乙酯(100mL×3)萃取。合并有机相,依次用水(50mL×2)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得淡黄色油状物(320mg,75.3%)。
MS(ESI,pos.ion)m/z:292.25[M+1] +.
步骤5:(4-(3-((S)-1-((S)-1,1-二甲基乙基亚磺酰氨基)丁-3-烯-1-基)-1H-吲唑-1-基)-3-硝基苯基)氨基甲酸甲酯 (7E)
Figure PCTCN2018072988-appb-000260
氮气保护下,向四口瓶中依次加入7D(20.8g,71.4mmol)、磷酸钾(31.14g,146.7mmol)、碘化亚铜(1.40g,7.35mmol)、N-(4-碘-3-硝基-苯基)氨基甲酸甲酯(32.0g,99.4mmol)、N,N'-二甲基乙烷-1,2-二胺(0.80mL,9.0mmol)和N,N-二甲基甲酰胺(20mL),然后加热至130℃搅拌48小时。冷至室温,用乙酸乙酯(100mL×3)萃取,合并有机相,依次用水(50mL×2)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得黄色油状物(6.0g,17.0%)。
MS(ESI,pos.ion)m/z:486.1[M+1] +.
步骤6:(S)-(4-(3-(1-氨基丁-3-烯-1-基)-1H-吲唑-1-基)-3-硝基苯基)氨基甲酸甲酯(7F)
Figure PCTCN2018072988-appb-000261
将7E(500mg,1.03mmol)溶于甲醇(10mL)中,加入6mol/L的盐酸(4mL,24mmol)溶液,在室温下搅拌4小时。停止反应,减压蒸去溶剂,用饱和碳酸氢钠溶液中和后,用乙酸乙酯(50mL×3)萃取。合并有机相,依次用水(50mL)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得 黄色油状物(390mg,99.3%)。
MS(ESI,pos.ion)m/z:382.2[M+1] +.
步骤7:(S)-(4-(3-(1-(叔丁氧羰基氨基)丁-3-烯-1-基)-1H-吲唑-1-基)-3-硝基苯基)氨基甲酸甲酯(7G)
Figure PCTCN2018072988-appb-000262
将7F(390mg,1.023mmol)溶于无水乙醇(20mL)中,加入盐酸胍(20mg,0.2094mmol)和二碳酸二叔丁酯(0.35mL,1.5mmol),氮气保护下加热至40℃搅拌1小时。停止反应,冷却至室温,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=4/1),得棕黄色固体(105mg,21.3%)。
MS(ESI,pos.ion)m/z:503.9[M+23] +.
步骤8:(S)-(4-(3-(1-(叔丁氧羰基氨基)丁-3-烯-1-基)-1H-吲唑-1-基)-3-氨基苯基)氨基甲酸甲酯(7H)
Figure PCTCN2018072988-appb-000263
将7G(3.10g,6.44mmol)溶于甲醇(100mL)中,然后加入锌粉(5.00g,76.4mmol)和氯化铵(4.00g,74.8mmol),在室温下搅拌过夜。停止反应,过滤,减压蒸去溶剂,用二氯甲烷(100mL×2)萃取。合并有机相,依次用水(100mL)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色固体(2.91g,100%)。
MS(ESI,pos.ion)m/z:452.4[M+1] +.
步骤9:(S)-(4-(3-(1-(叔丁氧羰基氨基)丁-3-烯-1-基)-1H-吲唑-1-基)-3-((R)-2-甲基丁-3-烯酰氨基)苯基)氨基甲 酸甲酯(7I)
Figure PCTCN2018072988-appb-000264
在0℃下,将7H(3.00g,6.64mmol)溶于二氯甲烷(20mL)中,加入吡啶(2.20mL,27.2mmol)和(R)-2-甲基丁-3-烯酰氯(1.30g,11.0mmol),体系继续在0℃下搅拌1小时,移至室温下搅拌2.5小时。停止反应,用二氯甲烷(100mL×3)萃取。合并有机相,依次用水(50mL×2)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得淡黄色固体(2.71g,76.4%)。
MS(ESI,pos.ion)m/z:534.2[M+1] +.
步骤10:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-1,8,22-三氮杂四环[13.6.1.0 2,7.0 16,21]二十二烷 -2(7),3,5,11(12),15(22),16(21),17,19-八烯-5-基)氨基甲酸甲酯(7J)
Figure PCTCN2018072988-appb-000265
在室温下,将一水合对甲苯磺酸盐(1.16g,6.10mmol)放置到两口瓶中,加热到70℃,减压干燥约2小时。然后将溶有7I(2.71g,5.08mmol)的干燥二氯甲烷(100mL)溶液注射到冷却到45℃的体系内,继续在45℃下搅拌2小时。再将Grubbs 2代催化剂(1.30g,1.53mmol)的干燥二氯甲烷(50mL)溶液缓慢注射到体系中去,滴加完毕后在45℃回流搅拌过夜。冷却至室温,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得棕色固体(230mg,8.96%)。
MS(ESI,pos.ion)m/z:406.1[M-100+1] +
步骤11:N-((10R,14S)-14-(叔丁氧羰基氨基)-10-甲基-9-氧代-1,8,22-三氮杂四环[13.6.1.0 2,7.0 16,21]二十二烷 -2(7),3,5,15(22),16(21),17,19-七烯-5-基)氨基甲酸甲酯(7K)
Figure PCTCN2018072988-appb-000266
将7J(230mg,0.455mmol)溶于甲醇(50mL)中,加入10%Pd/C(100mg,0.094mmol),在3.5MPa的氢气氛围下搅拌过夜。停止反应,经硅藻土过滤,减压蒸去溶剂,得黄色固体(150mg,64.96%)。MS(ESI,pos.ion)m/z:508.2[M+1] +
步骤12:N-((10R,14S)-14-氨基-10-甲基-9-氧代-1,8,22-三氮杂四环[13.6.1.0 2,7.0 16,21]二十二烷 -2(7),3,5,15(22),16(21),17,19-七烯-5-基)氨基甲酸甲酯(7L)
Figure PCTCN2018072988-appb-000267
将7K(550mg,1.084mmol)溶于二氯甲烷(10mL)中,加入三氟乙酸(2.00mL,26.9mmol),室温下搅拌1小时。停止反应,减压蒸去溶剂,得棕色固体(440mg,99.7%),所得产品直接用于下一步反应。MS(ESI,pos.ion)m/z:408.9[M+1] +.
步骤13:N-((10R,14S)-14-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙氨基)-10-甲基-9-氧代-1,8,22-三氮杂四环 [13.6.1.0 2,7.0 16,21]二十二烷-2(7),3,5,15(22),16(21),17,19-七烯-5-基)氨基甲酸甲酯(7M)
Figure PCTCN2018072988-appb-000268
将7L(440mg,1.08mmol)溶于四氢呋喃(10mL)中,加入三乙胺(2.00mL,14.4mmol),然后再加入1P(500mg,1.278mmol)的四氢呋喃(10mL)溶液,体系在室温下搅拌5小时。用乙酸乙酯(50mL×3)萃取,合并有机相,依次用水(20mL×2)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得棕色固体(400mg,55.2%)。
MS(ESI,pos.ion)m/z:672.1[M+1] +.
步骤14:N-((10R,14S)-14-(N-(3-(6-溴-3-氯-2-氟苯基)-3-氧代丙基)-2-(二乙氧基磷酰基)乙酰氨基)-10-甲基-9- 氧代-1,8,22-三氮杂四环[13.6.1.0 2,7.0 16,21]二十二烷-2(7),3,5,15(22),16(21),17,19-七烯-5-基)氨基甲酸甲酯 (7N)
Figure PCTCN2018072988-appb-000269
在微波管中加入7M(400mg,0.596mmol)、(2-氯-2-氧代乙基)磷酸二乙酯(500mg,2.33mmol)、吡啶(0.200mL,2.49mmol)、4-二甲氨基吡啶(80mg,0.655mmol)和二氯甲烷(15mL),然后在微波下加热至50℃反应3小时。停止反应,用二氯甲烷(50mL×2)萃取。合并有机相,依次用水(50mL)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,减压蒸去溶剂,得黄色固体(400mg,79.0%)。
MS(ESI,pos.ion)m/z:850.05[M+1] +.
步骤15:N-((10R,14S)-14-(4-(6-溴-3-氯-2-氟苯基)-6-氧代-1,2,3,6-四氢吡啶-1-基)-10-甲基-9-氧代-1,8,22-三氮 杂四环[13.6.1.0 2,7.0 16,21]二十二烷-2(7),3,5,15(22),16(21),17,19-七烯-5-基)氨基甲酸甲酯
在氮气保护下,将7N(400mg,0.471mmol)的四氢呋喃(20mL)溶液加入到60%的氢化钠(90mg,2.25mmol)中,然后在室温下搅拌1小时。停止反应,加水淬灭反应。用乙酸乙酯(50mL×2)萃取,合并有机相,依次用水(50mL)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1)得淡黄色固体,再经制备色谱纯化得白色固体(60mg,18.3%)。
MS(ESI,pos.ion)m/z:696.2[M+1] +
1H NMR(400MHz,CDCl 3)δ(ppm)9.95(s,1H),7.95(d,J=1.9Hz,1H),7.91(d,J=8.1Hz,1H),7.80(d,J=7.4Hz,1H),7.70(dd,J=8.5,5.1Hz,2H),7.52(t,J=7.7Hz,1H),7.38(d,J=8.7Hz,1H),7.35–7.28(m,2H), 7.18(s,1H),6.37-6.33(m,1H),6.16(s,1H),3.78(s,3H),3.76–3.68(m,1H),3.49-3.42(m,1H),2.55-2.46(m,2H),2.35–2.18(m,2H),2.01–1.86(m,3H),1.78(s,1H),1.32(d,J=7.2Hz,3H).
实施例8
(9R,13S)-13-(4-(5-氯-2-(1H-四氮唑-1-基)苯基)-6-氧代-嘧啶-1-基)-3,9-二甲基-17,20-二氧杂-3,4,7,15-四氮杂四环[12.7.1.0 2,6.0 16,21]二十二烷-1(22),2(6),4,14,16(21)-五烯-8-酮
Figure PCTCN2018072988-appb-000270
步骤1:((1S)-1-(8-(1-甲基-4-硝基-1H-吡唑-5-基)-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6-基)丁-3-烯-1-基)氨基甲 酸叔丁酯(8A)
Figure PCTCN2018072988-appb-000271
在两口瓶中加入1-甲基-4-硝基-1H-吡唑(1.11g,8.73mmol)、特戊酸(210mg,2.06mmol)、醋酸钯(II)(300mg,1.336mmol)、正丁基二(1-金刚烷基)膦(725mg,2.022mmol)、碳酸钾(2.80g,20.3mmol)和N,N-二甲基甲酰胺(100mL),再加入6G(2.91g,6.73mmol)的N,N-二甲基甲酰胺(10mL)溶液,体系加热至120℃搅拌5小时。停止反应,冷至室温,用乙酸乙酯(100mL×3)萃取。合并有机相,依次用水(50mL×2)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得棕色油状物(2.90g,99.8%)。
MS(ESI,pos.ion)m/z:432.1[M+1] +.
步骤2:((1S)-1-(8-(4-氨基-1-甲基-1H-吡唑-5-基)-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6-基)丁-3-烯-1-基)氨基甲 酸叔丁酯(8B)
Figure PCTCN2018072988-appb-000272
将8A(3.10g,7.19mmol)溶于甲醇(50mL)中,加入锌粉(5.00g,76.4mmol)和氯化铵(3.85g,72.0mmol),在室温下搅拌过夜。停止反应,过滤,减压蒸去溶剂,用二氯甲烷(100mL×2)萃取。合并有机相,依次用水(100mL)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色固体(2.60g,90.1%)。
MS(ESI,pos.ion)m/z:402.1[M+1] +.
步骤3:((1S)-1-(8-(1-甲基-4-((R)-2-甲基丁-3-烯酰氨基)-1H-吡唑-5-基)-2,3-二氢-[1,4]二噁并[2,3-b]吡啶-6- 基)丁-3-烯-1-基)氨基甲酸叔丁酯(8C)
Figure PCTCN2018072988-appb-000273
在0℃下,将8B(2.534g,6.311mmol)溶于二氯甲烷(20mL)中,加入吡啶(2.0mL,25mmol)和(R)-2-甲基丁-3-烯酰氯(1.20g,10.1mmol),继续在0℃下搅拌1小时后,移至室温下搅拌3小时。停止反应,用二氯甲烷(100mL×3)萃取。合并有机相,依次用水(50mL×2)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得淡黄色泡沫状固体(1.085g,35.55%)。
MS(ESI,pos.ion)m/z:484.3[M+1] +.
步骤4:((9R,13S)-3,9-二甲基-8-氧代-17,20-二氧杂-3,4,7,15-四氮杂四环[12.7.1.0 2,6.0 16,21]二十二烷 -1(22),2(6),4,10(11),14,16(21)-六烯-13-基)氨基甲酸叔丁酯(8D)
Figure PCTCN2018072988-appb-000274
在室温下,将一水合对甲苯磺酸(512mg,2.69mmol)放置到两口瓶中,加热到70℃,减压干燥约1.5小时。然后将8C(1.085g,2.24mmol)的干燥二氯甲烷(100mL)溶液注射到冷却到45℃的体系内,继续在45℃下搅拌1小时。再将Grubbs 2代催化剂(570mg,0.671mmol)的干燥二氯甲烷(50mL)溶液缓慢注射到体系中,滴加完毕后在45℃下搅拌回流过夜。停止反应,冷却至室温,减压蒸去溶剂,粗产品经硅胶柱层析纯化(甲醇/乙酸乙酯(v/v)=10/1),得棕色固体(140mg,13.70%)。
MS(ESI,pos.ion)m/z:456.1[M+1] +.
步骤5:((9R,13S)-3,9-二甲基-8-氧代-17,20-二氧杂-3,4,7,15-四氮杂四环[12.7.1.0 2,6.0 16,21]二十二烷 -1(22),2(6),4,14,16(21)-五烯-13-基)氨基甲酸叔丁酯(8E)
Figure PCTCN2018072988-appb-000275
将8D(140mg,0.307mmol)溶于甲醇(10mL)中,加入10%的Pd/C(65mg),在5.0MPa的氢气氛围下,加热至50℃搅拌过夜。停止反应,冷却至室温,经硅藻土过滤,减压蒸去溶剂,得黄色油状物(130mg,92.45%)
MS(ESI,pos.ion)m/z:458.1[M+1] +.
步骤6:(9R,13S)-13-氨基-3,9-二甲基-17,20-二氧杂-3,4,7,15-四氮杂四环[12.7.1.0 2,6.0 16,21]二十二烷 -1(22),2(6),4,14,16(21)-五烯-8-酮(8F)
Figure PCTCN2018072988-appb-000276
将8E(130mg,0.284mmol)溶于二氯甲烷(10mL)中,加入三氟乙酸(1mL,13.46mmol),室温下搅拌1小时。停止反应,用饱和碳酸氢钠溶液中和反应液,然后用二氯甲烷(50mL×3)萃取。合并有机相,依次用水(20mL×2)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得棕色固体(130mg,97.0%)。
MS(ESI,pos.ion)m/z:358.2[M+1] +.
步骤7:4-氯-2-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯胺(8G)
Figure PCTCN2018072988-appb-000277
向两口瓶中依次加入2-溴-4-氯-苯胺(30g,145.30mmol)、联硼酸频那醇酯(41g,161.5mmol)、醋酸钾(43g,438.1mmol)、[1,1'-双(二苯基膦)二茂铁]二氯化钯(II)二氯甲烷络合物(3.56g,4.36mmol)和1,4-二氧六环(700mL),氮气置换三次,然后加热至115℃反应5小时。冷至室温,经硅藻土过滤。滤液减压蒸去溶剂,残留物经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=20/1),得白色固体(26.2g,71.1%)。
步骤8:4-氯-2-(6-甲氧基嘧啶-4-基)苯胺(8H)
Figure PCTCN2018072988-appb-000278
在室温下,将8G(4.56g,8.99mmol)、4-氯-6-甲氧基嘧啶(1.0g,6.91mmol)、碳酸钠(1.47g,13.9mmol)和[1,1'-双(二苯基膦)二茂铁]二氯化钯(II)二氯甲烷络合物(0.576g,0.691mmol)装入反应器中,用氮气置换掉空气,再注射入乙二醇二甲醚(45mL)、乙醇(3mL)和水(3mL),加料完毕后,升温到90℃搅拌14小时。过滤,滤液加乙酸乙酯(500mL)萃取,再用水(150mL×3)和饱和食盐水(150mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩,得到粗产品,经硅胶柱层析(石油醚/乙酸乙酯(v/v)=3/1)提纯,得褐色固体(1.32g,81.0%)。
MS(ESI,pos.ion)m/z:236.0[M+1] +.
步骤9:4-(5-氯-2-(1H-四氮唑-1-基)苯基)-6-甲氧基嘧啶(8I)
Figure PCTCN2018072988-appb-000279
称取8H(0.50g,2.1mmol)于烧瓶中,向其中加入醋酸(5mL)和原甲酸三甲酯(0.7mL,6mmol),搅拌半小时。加入叠氮化钠(0.41g,6.3mmol),然后在室温下反应24小时。反应完毕,加入乙酸乙酯(20mL)和水(20mL)稀释,乙酸乙酯萃取(20mL×2),合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,旋干,残留物经硅胶柱层析(石油醚/乙酸乙酯(v/v)=2/1)纯化,得淡黄色固体(0.60g,98.0%)。
步骤10:6-(5-氯-2-(1H-四氮唑-1-基)苯基)嘧啶-4-醇(8J)
Figure PCTCN2018072988-appb-000280
称取8I(0.70g,2.4mmol)于烧瓶中,向其中加入乙腈(25mL)、碘化钠(3.5g,23mmol)和三甲基氯硅烷(3.0mL,35mmol),室温反应24小时。加入饱和硫代硫酸钠溶液(10mL)淬灭反应。乙酸乙酯(30mL×2)萃取,无水硫酸钠干燥,过滤,旋干,残留物经硅胶柱层析(乙酸乙酯)纯化,得黄色固体(0.65g,98.0%)。
MS(ESI,pos.ion)m/z:275.0[M+H] +.
步骤11:(9R,13S)-13-(4-(5-氯-2-(1H-四氮唑-1-基)苯基)-6-氧代-嘧啶-1-基)-3,9-二甲基-17,20-二氧杂-3,4,7,15- 四氮杂四环[12.7.1.0 2,6.0 16,21]二十二烷-1(22),2(6),4,14,16(21)-五烯-8-酮
在单口瓶中加入8J(120mg,0.437mmol)、8F(150mg,0.42mmol)、1,8-二氮杂二环十一碳-7-烯(0.10mL,0.67mmol)、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(190mg,0.50mmol)和乙腈(10mL),然后在室温下搅拌2天。停止反应,减压蒸去溶剂,粗产品经硅胶柱层析纯化(乙酸乙酯)得黄色固体,再经制备色谱纯化得白色固体(6mg,2.33%)。
MS(ESI,pos.ion)m/z:615.1[M+1] +
1H NMR(400MHz,CD 3OD)δ(ppm)9.45(s,1H),8.75(s,1H),7.91(d,J=1.5Hz,1H),7.78(d,J=8.5Hz,1H),7.69(d,J=8.5Hz,1H),7.48(s,1H),7.10(s,1H),6.49(s,1H),5.84(s,1H),4.63–4.47(m,2H),4.43-4.36(m,2H),3.92(s,3H),2.91(s,1H),2.68-2.58(m,1H),2.19-2.11(m,1H),2.07-2.02(m,1H),2.00–1.79(m,2H),1.72–1.46(m,2H),1.02(s,3H).
实施例9
(9R,13S)-13-(4-(5-氯-2-(1H-四氮唑-1-基)苯基)-6-氧代-嘧啶-1-基)-3,9-二甲基-17,20-二氧杂-3,4,7-三氮杂四环[12.7.1.0 2,6.0 16,21]二十二烷-1(22),2(6),4,14,16(21)-五烯-8-酮
Figure PCTCN2018072988-appb-000281
步骤1:3-溴-4,5-二羟基苯甲醛(9A)
Figure PCTCN2018072988-appb-000282
在冰浴下,将3,4-二羟基苯甲醛(500mg,3.62mmol)溶于冰乙酸(10mL)中,然后滴入溴素(0.2mL4.00mmol)的冰乙酸(1mL)溶液,体系在室温下搅拌过夜。停止反应,加入冰水淬灭反应,灰色固体析出,过滤,用冰水洗涤,得灰色固体(260mg,33.1%)。
MS(ESI,pos.ion)m/z:217.0[M+1] +,219.0[M+1] +.
步骤2:8-溴-2,3-二氢苯并[b][1,4]二噁烷-6-甲醛(9B)
Figure PCTCN2018072988-appb-000283
将9A(27g,124.4mmol)和碳酸钾(43g,311.1mmol)溶于N,N-二甲基甲酰胺(350mL)中,然后加入1,2-二溴乙烷(32mL,371mmol),体系加热至110℃搅拌4小时。冷至室温,用乙酸乙酯(300mL×2)萃取。合并有机相,依次用水(200mL×2)和饱和食盐水(200mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色油状物(30g,99.2%)。
MS(ESI,pos.ion)m/z:242.9[M+1] +.
步骤3:(S,E)-N-((8-溴-2,3-二氢苯并[b][1,4]二噁烷-6-基)亚甲基)-2-甲基丙烷-2-亚磺酰胺(9C)
Figure PCTCN2018072988-appb-000284
将9B(555mg,2.28mmol)溶于二氯甲烷(20mL)中,加入(S)-(-)-叔丁基亚磺酰胺(300mg,2.48mmol)和碳酸铯(1.1g,3.4mmol),然后在室温下搅拌过夜。停止反应,经硅藻土过滤,用二氯甲烷洗涤滤饼。收集滤液,依次用水(20mL×2)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得黄色油状物(700mg,88.6%)。
MS(ESI,pos.ion)m/z:348.0[M+1] +.
步骤4:(S)-N-((S)-1-(8-溴-2,3-二氢苯并[b][1,4]二噁烷-6-基)丁-3-烯-1-基)-2-甲基丙烷-2-亚磺酰胺(9D)
Figure PCTCN2018072988-appb-000285
将9C(10.2g,29.5mmol)溶于饱和的溴化钠(200mL)溶液中,然后加入铟粉(13.5g,117mmol)和烯丙基溴(10.5mL,120mmol),在室温下搅拌过夜。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得黄色油状物(8.1g,71%).
MS(ESI,pos.ion)m/z:390.0[M+1] +.
步骤5:(S)-1-(8-溴-2,3-二氢苯并[b][1,4]二噁烷-6-基)丁-3-烯-1-胺(9E)
Figure PCTCN2018072988-appb-000286
将9D(9.0g,23mmol)溶于甲醇(100mL)中,加入4mol/L的稀盐酸(17mL,68mmol),在室温下搅拌0.5小时。停止反应,减压蒸去溶剂,用饱和碳酸氢钠溶液中和后用乙酸乙酯(100mL×5)萃取。合并有机相,依次用水(100mL)和饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色固体(5.0g,76%),粗产品直接用于下一步反应。
步骤6:(S)-(1-(8-溴-2,3-二氢苯并[b][1,4]二噁烷-6-基)丁-3-烯-1-基)氨基甲酸叔丁酯(9F)
Figure PCTCN2018072988-appb-000287
将9E(5.0g,17.6mmol)溶于二氯甲烷(100mL)中,加入三乙胺(4.9mL,35mmol)和二碳酸二叔丁酯(4.5mL,19mmol),然后在室温下搅拌3小时。停止反应,加水(50mL),用二氯甲烷(50mL×2)萃取。合并有机相,用无水硫酸钠干燥,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=4/1),得黄色油状物(3.0g,44%)。
1H NMR(400MHz,CDCl 3)δ(ppm)7.02(d,J=1.6Hz,1H),6.75(d,J=1.3Hz,1H),5.72-5.62(m,1H),5.18–5.03(m,2H),4.84(s,1H),4.60(s,1H),4.38–4.31(m,2H),4.26(dd,J=5.1,2.5Hz,2H),2.45(s,2H),1.43(s,9H).
步骤7:((1S)-1-(8-(1-甲基-4-硝基-1H-吡唑-5-基)-2,3-二氢苯并[b][1,4]二噁烷-6-基)丁-3-烯-1-基)氨基甲酸叔 丁酯(9G)
Figure PCTCN2018072988-appb-000288
在两口瓶中加入1-甲基-4-硝基-1H-吡唑(1.60g,12.6mmol)、特戊酸(300mg,2.94mmol)、醋酸钯(II)(440mg,1.960mmol)、正丁基二(1-金刚烷基)膦(1.0g,2.8mmol)、碳酸钾(4.0g,29mmol)和N,N-二甲基甲酰胺(30mL),再加入9F(3.73g,9.71mmol)的N,N-二甲基甲酰胺(20mL)溶液,然后加热至120℃搅拌5小时。停止反应,冷至室温,用乙酸乙酯(100mL×3)萃取。合并有机相,依次用水(50mL×2)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得棕色油状物(3.35g,80.2%)。
1H NMR(600MHz,CDCl 3)δ(ppm)8.18(s,1H),6.97(s,1H),6.74(d,J=12.4Hz,1H),5.72-5.66(m,1H),5.18–5.04(m,2H),4.92(s,1H),4.68(s,1H),4.31–4.18(m,4H),3.73(d,J=16.6Hz,3H),2.51(s,2H),1.41(d,J=7.7Hz,9H).
步骤8:((1S)-1-(8-(4-氨基-1-甲基-1H-吡唑-5-基)-2,3-二氢苯并[b][1,4]二噁烷-6-基)丁-3-烯-1-基)氨基甲酸叔 丁酯(9H)
Figure PCTCN2018072988-appb-000289
将9G(3.35g,7.78mmol)溶于甲醇(50mL)中,加入锌粉(5.1g,78mmol)和氯化铵(4.16g,77.8mmol),在室温下搅拌过夜。停止反应,过滤,减压蒸去溶剂,用二氯甲烷(100mL×2)萃取。合并有机相,依次用水(100mL)、饱和食盐水(50mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得黄色固体(2.90g,93.0%)。
MS(ESI,pos.ion)m/z:401.1[M+1] +.
步骤9:((1S)-1-(8-(1-甲基-4-((R)-2-甲基丁-3-烯酰氨基)-1H-吡唑-5-基)-2,3-二氢苯并[b][1,4]二噁烷-6-基)丁 -3-烯-1-基)氨基甲酸叔丁酯(9I)
Figure PCTCN2018072988-appb-000290
在0℃下,将9H(2.9g,7.2mmol)溶于二氯甲烷(20mL)中,加入吡啶(2.3mL,28mmol)和(R)-2-甲基丁-3-烯酰氯(1.40g,11.8mmol),体系继续在0℃下搅拌1小时后,移至室温下搅拌2小时。停止反应,用二氯甲烷(100mL×3)萃取。合并有机相,依次用水(50mL×2)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得***泡沫状固体(1.93g,55%)。
MS(ESI,pos.ion)m/z:483.2[M+1] +.
步骤10:((9R,13S)-3,9-二甲基-8-氧代-17,20-二氧杂-3,4,7-三氮杂四环[12.7.1.0 2,6.0 16,21]二十二烷 -1(22),2(6),4,10(11),14,16(21)-六烯-13-基)氨基甲酸叔丁酯(9J)
Figure PCTCN2018072988-appb-000291
在室温下,将一水合对甲苯磺酸(915mg,4.81mmol)放置到两口瓶中,加热到70℃,减压干燥约1.5小时。将9I(1.93g,4.00mmol)的干燥二氯甲烷(100mL)溶液注射到冷却到45℃的体系内,继续在45℃ 下搅拌1小时。再将Grubbs 2代催化剂(1.0g,1.2mmol)的干燥二氯甲烷(50mL)溶液缓慢注射到体系中,滴加完毕后在45℃搅拌回流过夜。停止反应,冷却至室温,减压蒸去溶剂,粗产品经硅胶柱层析纯化(乙酸乙酯)得棕色固体(500mg,27.5%)。
MS(ESI,pos.ion)m/z:455.2[M+1] +.
步骤11:((9R,13S)-3,9-二甲基-8-氧代-17,20-二氧杂-3,4,7-三氮杂四环[12.7.1.0 2,6.0 16,21]二十二烷 -1(22),2(6),4,14,16(21)-五烯-13-基)氨基甲酸叔丁酯(9K)
Figure PCTCN2018072988-appb-000292
将9J(600mg,1.32mmol)溶于甲醇(50mL)中,加入10%Pd/C(300mg),在5.0MPa的氢气氛围下加热至50℃搅拌过夜。停止反应,冷却至室温,经硅藻土过滤,减压蒸去溶剂,得淡黄色固体(500mg,82.97%)。
MS(ESI,pos.ion)m/z:457.1[M+1] +.
步骤12:(9R,13S)-13-氨基-3,9-二甲基-17,20-二氧杂-3,4,7-三氮杂四环[12.7.1.0 2,6.0 16,21]二十二烷 -1(22),2(6),4,14,16(21)-五烯-8-酮(9L)
Figure PCTCN2018072988-appb-000293
将9K(500mg,1.095mmol)溶于二氯甲烷(10mL)中,加入三氟乙酸(4mL,53.85mmol),室温下搅拌1小时。停止反应,用饱和碳酸氢钠溶液中和反应液,然后用二氯甲烷(50mL×3)萃取。合并有机相,依次用水(20mL×2)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得棕色固体(390mg,99.9%)。
MS(ESI,pos.ion)m/z:357.1[M+1] +.
步骤13:(9R,13S)-13-(4-(5-氯-2-(1H-四氮唑-1-基)苯基)-6-氧代-嘧啶-1-基)-3,9-二甲基-17,20-二氧杂-3,4,7- 三氮杂四环[12.7.1.0 2,6.0 16,21]二十二烷-1(22),2(6),4,14,16(21)-五烯-8-酮
在单口瓶中加入8J(230mg,0.837mmol)、9L(280mg,0.786mmol)、1,8-二氮杂二环十一碳-7-烯(0.20mL,1.3mmol)、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(370mg,0.973mmol)和乙腈(20mL),然后在室温下搅拌1天。停止反应,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得黄色固体,再经制备色谱进一步纯化,得白色固体(20mg,4.15%)。
MS(ESI,pos.ion)m/z:614.2[M+1] +
1H NMR(400MHz,CD 3OD)δ(ppm)9.46(s,1H),8.45(s,1H),7.93(s,1H),7.78(d,J=7.0Hz,1H),7.71(d,J=7.8Hz,1H),7.48(s,1H),6.98(s,1H),6.70(s,1H),6.57(s,1H),5.59(d,J=9.6Hz,1H),4.39-4.35(m,4H),3.89(s,3H),3.26-3.20(m,1H),2.91(s,1H),2.49–2.31(m,1H),2.26–2.10(m,1H),2.01–1.90(m,1H),1.85–1.70(m,1H),1.16(d,J=6.1Hz,3H).
实施例10
(10R,14S)-14-(4-(5-氯-2-(1H-四氮唑-1-基)苯基)-6-氧代-嘧啶-1-基)-4-氟-10-甲基-18,21-二氧杂-8-氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-9-酮
Figure PCTCN2018072988-appb-000294
步骤1:(S)-(1-(8-(2-氨基-5-氟苯基)-2,3-二氢苯并[b][1,4]二噁烷-6-基)丁-3-烯-1-基)氨基甲酸叔丁酯(10A)
Figure PCTCN2018072988-appb-000295
氮气保护下,在两口瓶中依次加入9F(4.85g,12.7mmol)、碳酸钠(2.70g,25.5mmol)的水(10mL)溶液、三环己基膦(530mg,1.89mmol)、(dba) 3Pd 2(580mg,0.633mmol)、4-氟-2-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯胺(4.50g,19.0mmol)和N,N-二甲基甲酰胺(100mL),然后加热至90℃搅拌5小时。停止反应,冷至室温,用乙酸乙酯(500mL×3)萃取。合并有机相,依次用水(200mL×2)和饱和食盐水(200mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得黄色油状物(2.80g,53.5%)。
MS(ESI,pos.ion)m/z:437.1[M+23] +.
步骤2:((S)-1-(8-(5-氟-2-((R)-2-甲基丁-3-烯酰氨基)苯基)-2,3-二氢苯并[b][1,4]二噁烷-6-基)丁-3-烯-1-基)氨 基甲酸叔丁酯(10B)
Figure PCTCN2018072988-appb-000296
在0℃下,将10A(2.8g,6.8mmol)溶于二氯甲烷(20mL)中,加入吡啶(2.2mL,27mmol)和(R)-2-甲基丁-3-烯酰氯(1.15g,9.70mmol),体系继续在0℃下搅拌1小时后,移至室温下搅拌2小时。停止反应,用二氯甲烷(100mL×3)萃取。合并有机相,依次用水(50mL×2)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=4/1),得棕色油状物(1.76g,52%)。
MS(ESI,pos.ion)m/z:519.1[M+23] +.
步骤3:((10R,14S)-4-氟-10-甲基-18,21-二氧杂-8-氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷 -1(23),2(7),3,5,11(12),15,17(22)-七烯-14-基)氨基甲酸叔丁酯(10C)
Figure PCTCN2018072988-appb-000297
在室温下,将一水合对甲苯磺酸(770mg,4.05mmol)放置到两口瓶中,加热到70℃,减压干燥约1.5小时。然后将10B(1.67g,3.36mmol)的干燥二氯甲烷(100mL)溶液注射到冷却到45℃的体系中,继续在45℃下搅拌1小时。再将Grubbs 2代催化剂(860mg,1.013mmol)的干燥二氯甲烷(50mL)溶液缓慢注射到体系中,滴加完毕后体系在45℃搅拌回流过夜。停止反应,冷却至室温,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=2/1),得棕色固体(280mg,17.8%)。
MS(ESI,pos.ion)m/z:491.1[M+23] +.
步骤4:((10R,14S)-4-氟-10-甲基-18,21-二氧杂-8-氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)- 六烯-14-基)氨基甲酸叔丁酯(10D)
Figure PCTCN2018072988-appb-000298
将10C(280mg,0.598mmol)溶于甲醇(50mL)中,加入10%的Pd/C(150mg,0.141mmol),在4.0MPa的氢气氛围下搅拌过夜。停止反应,经硅藻土过滤,减压蒸去溶剂,得淡黄色固体(260mg,92.46%)。MS(ESI,pos.ion)m/z:493.1[M+23] +.
步骤5:(10R,14S)-14-氨基-4-氟-10-甲基-18,21-二氧杂-8-氮杂四环[13.7.1.0 2,7.0 17,22]二十三烷 -1(23),2(7),3,5,15,17(22)-六烯-9-酮(10E)
Figure PCTCN2018072988-appb-000299
将10D(260mg,0.553mmol)溶于二氯甲烷(10mL)中,加入三氟乙酸(2mL,26.9mmol),室温下搅拌1小时。停止反应,用饱和碳酸氢钠溶液中和反应液,然后用二氯甲烷(50mL×3)萃取。合并有机相,依次用水(20mL×2)、饱和食盐水(40mL)洗涤,无水硫酸钠干燥。过滤,减压蒸去溶剂,得棕色固体(200mg,97.71%)。
MS(ESI,pos.ion)m/z:354.1[M-17+1] +.
步骤6:(10R,14S)-14-(4-(5-氯-2-(1H-四氮唑-1-基)苯基)-6-氧代-嘧啶-1-基)-4-氟-10-甲基-18,21-二氧杂-8-氮 杂四环[13.7.1.0 2,7.0 17,22]二十三烷-1(23),2(7),3,5,15,17(22)-六烯-9-酮
在单口瓶中加入8J(180mg,0.655mmol)、10E(200mg,0.540mmol)、1,8-二氮杂二环十一碳-7-烯(0.13 mL,0.87mmol)、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(250mg,0.658mmol)和乙腈(20mL),然后在室温下搅拌2天。停止反应,减压蒸去溶剂,粗产品经硅胶柱层析纯化(石油醚/乙酸乙酯(v/v)=1/1),得黄色固体,再经制备色谱进一步纯化后,得白色固体(15mg,4.42%)。
MS(ESI,pos.ion)m/z:628.0[M+1] +
1H NMR(400MHz,CDCl 3)δ(ppm)8.81(s,1H),8.04(s,1H),7.69(s,1H),7.65(d,J=8.4Hz,1H),7.50(d,J=8.4Hz,1H),7.43(dd,J=8.6,5.4Hz,1H),7.30(d,J=2.8Hz,1H),7.16–7.04(m,1H),6.95(s,1H),6.66(s,1H),6.55(d,J=8.2Hz,2H),5.56(d,J=9.5Hz,1H),4.29(s,4H),2.53–2.41(m,1H),2.16–2.00(m,2H),1.97–1.87(m,1H),1.74(s,2H),1.69-1.62(m,2H),1.17(d,J=6.9Hz,3H).
实施例11
(9R,13S)-13-(4-(5-氯-2-氰基苯基)-6-氧代-嘧啶-1-基)-3,9-二甲基-17,20-二氧杂-3,4,7,15-四氮杂四环[12.7.1.0 2,6.0 16,21]二十二烷-1(22),2(6),4,14,16(21)-五烯-8-酮
Figure PCTCN2018072988-appb-000300
步骤1:(9R,13S)-13-(4-(5-氯-2-碘苯基)-6-氧代-嘧啶-1-基)-3,9-二甲基-17,20-二氧杂-3,4,7,15-四氮杂四环 [12.7.1.0 2,6.0 16,21]二十二烷-1(22),2(6),4,14,16(21)-五烯-8-酮(11A)
Figure PCTCN2018072988-appb-000301
在单口瓶中加入6-(5-氯-2-碘苯基)嘧啶-4-醇(参照专利WO 2015116886中间体5的合成方法制备得到)(215mg,0.647mmol)、8F(242.60mg,0.679mmol)、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(390mg,1.03mmol)、乙腈(60mL)和1,8-二氮杂二环十一碳-7-烯(0.60mL,4.0mmol),然后在室温下搅拌过夜。减压蒸去溶剂,粗产品经柱层析纯化(乙酸乙酯),得白色固体(250mg,54.73%)。
MS(ESI,pos.ion)m/z:673.0(M+1).
步骤2:(9R,13S)-13-(4-(5-氯-2-氰基苯基)-6-氧代-嘧啶-1-基)-3,9-二甲基-17,20-二氧杂-3,4,7,15-四氮杂四环 [12.7.1.0 2,6.0 16,21]二十二烷-1(22),2(6),4,14,16(21)-五烯-8-酮
氮气保护下,将氰化亚铜(92mg,1.03mmol)溶于二甲基亚砜(10mL)中,加热至150℃,滴加11A(460mg,0.684mmol)的二甲基亚砜(20mL)溶液,然后继续在150℃下搅拌2小时。冷却至室温,加入氨水(3mL)淬灭反应。用乙酸乙酯(30mL×3)萃取。合并有机相,减压蒸去溶剂,粗产品经柱层析纯化(乙酸乙酯),得白色固体(300mg,76.72%)。
MS(ESI,pos.ion)m/z:572.3(M+1);
1H NMR(400MHz,DMSO-d 6)δ(ppm)9.11(d,J=21.9Hz,1H),8.07–7.95(m,2H),7.78(dd,J=8.3,2.0Hz, 1H),7.46(d,J=13.5Hz,1H),7.13(s,1H),6.97(d,J=10.6Hz,1H),5.78(d,J=9.0Hz,1H),4.56–4.14(m,4H),3.84(s,3H),2.30(t,J=30.7Hz,1H),1.86(d,J=36.4Hz,2H),1.72–1.13(m,4H),0.89(d,J=25.3Hz,3H).
生物活性测试
实施例A  本发明化合物对人FXIa酶抑制实验
本发明化合物对人源凝血因子XIa(FXIa)活性的抑制作用通过以下方法测定。
人凝血因子XIa(FXIa)的酶活性通过对FXIa特异性的生色底物的转化来测定。该测定如下述在微孔滴定板上进行。
1实验材料:
酶:因子XIa蛋白酶(来源:Enzyme research laboratories,货号:HFXIa 1111a)
底物:S-2366(来源:Chromogenix,货号:82109039)
缓冲液:50mM HEPES、145mM NaCl、5mM KCl、0.1%PEG8000、pH=7.5
2实验步骤:
2.1 S-2366底物储备液(10mM)的配制:取一瓶S-2366显色底物(25mg),加入4.64mL无菌去离子水混匀,分装,于4℃冰箱中避光保存。
S-2366底物工作液(1.875mM)的配制:临用前将S-2366底物储备液用缓冲液稀释5.33倍。
2.2 FXIa酶工作液(125mU/mL)的配制:临用前将原装母液(102.6U/mL)用缓冲液稀释到125mU/mL。
2.3化合物工作液的配制:取10μL化合物储备液(20μM的DMSO溶液),加入90μL缓冲液配制成浓度为2μM、DMSO含量为10%的化合物溶液。取20μL的上述化合物溶液,用含10%DMSO的缓冲液2倍梯度稀释成浓度为20000、10000、5000、2500、1250、625、312.5、156.25、78.13、39.06nM的工作液,置于冰上备用。
2.4加样及酶学反应(双复孔):准备384孔板,加入4μL化合物工作液(对照组加入4μL DMSO)和12.8μL FXIa酶工作液(终浓度为80mU/mL),封板膜封板,微型振荡器摇匀后,25℃孵育10分钟。加入3.2μL S-2366底物工作液(终浓度为300μM),于25℃ 405nm处动力学测试吸光度值,做△A-时间曲线,计算斜率为反应速率,按照下列公式计算出抑制率。以抑制率为纵坐标,采用Graph Pad Prism 5软件绘制酶活性量效曲线,计算出IC 50值。
抑制率%=(V 0-V i)/V 0×1 00(式中:V 0为对照孔反应速率,V i为待测样品反应速率)
实验结果如表1所示。
表1 本发明化合物对人FXIa活性的抑制作用
实施例编号 FXIa IC 50(nM)
实施例5 44.7
实施例6 28.9
实施例8 10.4
实施例9 14.1
实施例10 21.7
结论:本发明化合物对人源凝血因子XIa活性有明显的抑制作用。
实施例B  本发明化合物体外抗凝作用测试
本发明化合物体外抗凝作用通过延长兔和人血浆的凝血时间来体现,可以通过以下方法测定。
1.各浓度化合物溶液的配制
取4μL各化合物工作液(10mM),用二甲亚砜3倍稀释成各个浓度的工作液。
2.血浆样品的制备
兔血浆的制备:取若干只兔,耳缘静脉注射3%戊巴比妥溶液(30mg/kg)麻醉,用含0.2mL 3.8%枸橼酸钠的真空采血管腹主动脉采血至2mL,收集多管,上下颠倒混匀数次,静置10分钟,于3000rpm离心10分钟,吸取各管血浆,将所有血浆混至同一离心管,按每管1.6mL分装,迅速置入-80℃冰箱保存备用。
人血浆的制备:健康人群空腹,肘正中静脉采血,用含0.2mL 3.8%枸橼酸钠的真空采血管采血至2mL,男性每人采50mL全血,女性每人采30mL全血,真空采血管采血完上下颠倒混匀数次,静置10分钟,于3000rpm离心10分钟,吸取各管血浆。将所有男性血浆混至同一离心管,女性血浆混至另一离心管,男女血浆按1:1制成混合血浆后,按每管1.3mL分装,迅速置入-80℃冰箱。
3.加样及测定凝血时间PT和APTT
准备好1.5mL EP管,每管加入180μL血浆标本;向各管血标本中分别加入4μL相应浓度的化合物,对照组加入4μL二甲亚砜溶液,震荡混匀,37℃孵育5分钟;用Sysmex CA1500全自动血凝仪测定PT以及APTT;绘制量效曲线,对曲线进行拟合,由此计算出使凝血时间加倍的测试化合物的浓度(CT 2)。本发明化合物对人血浆的抗凝作用测试结果见表2。
表2 本发明化合物体外抗凝作用测试
实施例编号 APTT CT 2(μM)
实施例8 0.20
结论:本发明化合物体外对人血浆有明显的抗凝血作用。
在本说明书的描述中,参考术语“一个实施例”、“一些实施例”、“示例”、“具体示例”、或“一些示例”等的描述意指结合该实施例或示例描述的具体特征、结构、材料或者特点包含于本发明的至少一个实施例或示例中。在本说明书中,对上述术语的示意性表述不必须针对的是相同的实施例或示例。而且,描述的具体特征、结构、材料或者特点可以在任一个或多个实施例或示例中以合适的方式结合。此外,在不相互矛盾的情况下,本领域的技术人员可以将本说明书中描述的不同实施例或示例以及不同实施例或示例的特征进行结合和组合。
尽管上面已经示出和描述了本发明的实施例,可以理解的是,上述实施例是示例性的,不能理解为对本发明的限制,本领域的普通技术人员在本发明的范围内可以对上述实施例进行变化、修改、替换和变型。

Claims (32)

  1. 一种化合物,其为如式(I)所示的化合物或式(I)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
    Figure PCTCN2018072988-appb-100001
    其中,
    环A为C 7-12碳环基、C 8-12芳基、7-12个原子组成的杂环基或7-12个原子组成的杂芳基;
    环B为C 5-12碳环基、C 6-12芳基、5-12个原子组成的杂环基或5-12个原子组成的杂芳基;
    环C为C 3-12碳环基、C 6-12芳基、3-12个原子组成的杂环基或5-12个原子组成的杂芳基;
    X为C 4-8亚烷基或C 4-8亚烯基,其中,所述亚烷基和亚烯基中一个或多个碳原子独立任选地被选自-O-、-S-、-C(=O)-、-S(=O)-、-S(=O) 2-或-N(R 5)-的基团所替代;X中的C 4-8亚烷基和C 4-8亚烯基任选地被一个或多个R 9所取代;
    Y为-C(=O)N(R 5)-或-N(R 5)C(=O)-;
    各R 1独立地为=O、H、氘、卤素、-(CR 7aR 7b) rCN、C 1-6烷基、氘代C 1-6烷基、卤代C 1-6烷基、C 2-6烯基、C 2-6炔基、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=O)R 6、-(CR 7aR 7b) rC(=O)OR 8、-(CR 7aR 7b) rC(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O)R 6、-(CR 7aR 7b) rS(=O)OR 8、-(CR 7aR 7b) rS(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-12环烷基、-(CR 7aR 7b) r-C 6-12芳基、-(CR 7aR 7b) r-(3-12个原子组成的杂环基)或-(CR 7aR 7b) r-(5-12个原子组成的杂芳基);其中,各R 1独立任选地被1、2、3或4个R 1a所取代;
    各R 2独立地为H、氘、卤素、-(CR 7aR 7b) rCN、C 1-6烷基、氘代C 1-6烷基、卤代C 1-6烷基、C 2-6烯基、C 2-6炔基、R 8O-C 2-6亚烯基、R 8OC(=O)-C 2-6亚烯基、R 8O-C 2-6亚炔基、R 8OC(=O)-C 2-6亚炔基、-N=C=S、-N=C=O、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=G)R 6、-(CR 7aR 7b) rN(R 5c)C(=G)R 6、-(CR 7aR 7b) rOC(=G)R 6、-(CR 7aR 7b) rC(=G)OR 8、-(CR 7aR 7b) rN(R 5c)C(=G)OR 8、-(CR 7aR 7b) rOC(=G)OR 8、-(CR 7aR 7b) rC(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)=C(R 7)NR 5aR 5b、-(CR 7aR 7b) rS(=O)R 6、-(CR 7aR 7b) rN(R 5c)S(=O)R 6、-(CR 7aR 7b) rS(=O)OR 8、-(CR 7aR 7b) rN(R 5c)S(=O)OR 8、-(CR 7aR 7b) rS(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rN(R 5c)S(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-12环烷基、-(CR 7aR 7b) r-C 6-12芳基、-(CR 7aR 7b) r-(3-12个原子组成的杂环基)、-(CR 7aR 7b) r-(5-12个原子组成的杂芳基)或D-甘露糖基;其中,各G独立地为O、S或NR 5d;各R 2独立任选地被1、2、3或4个R 2a所取代;
    各R 3独立地为C 1-6烷基、C 3-12碳环基、C 6-12芳基、3-12个原子组成的杂环基或5-12个原子组成的杂芳基;其中,各R 3独立任选地被1、2、3、4或5个R 3a所取代;
    各R 4独立地为H、氘、卤素、羟基、氨基、C 1-6烷基、C 2-6烯基、C 2-6炔基、氘代C 1-6烷基、卤代C 1-6烷氧基、-(CR 7aR 7b) r-OR 8、-(CR 7aR 7b) r-NR 5aR 5b、-(CR 7aR 7b) r-CN或卤代C 1-6烷基;或者,任意两个R 4和与它们相连的原子共同形成C 3-8环烷基、C 6-10芳基、3-8个原子组成的杂环基或5-6个原子组成的杂芳基;
    各R 9、R 1a、R 2a和R 3a独立地为=O、氢、氘、卤素、-(CR 7aR 7b) rCN、硝基、C 1-6烷基、卤代C 1-6烷基、C 2-6烯基、C 2-6炔基、R 8O-C 2-6亚烯基、R 8OC(=O)-C 2-6亚烯基、R 8O-C 2-6亚炔基、R 8OC(=O)-C 2-6亚炔基、-(CR 7aR 7b) r-OR 8、-(CR 7aR 7b) r-NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=O)R 6、-(CR 7aR 7b) rOC(=O)R 6、-(CR 7aR 7b) rCOOH、-(CR 7aR 7b) rC(=O)OC 1-6烷基、-(CR 7aR 7b) rC(=O)C 1-6烷基、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、 -(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-12环烷基、-(CR 7aR 7b) r-C 6-12芳基、-(CR 7aR 7b) r-(3-12个原子组成的杂环基)或-(CR 7aR 7b) r-(5-12个原子组成的杂芳基);
    或者,任意两个R 9与和它们相连的原子共同形成C 3-6环烷基或3-6个原子组成的杂环基;
    其中,各R 9、R 1a、R 2a和R 3a独立任选地被1、2、3、4或5个R 10所取代;
    各R 5、R 5a、R 5b、R 5c和R 5d独立地为氢、氰基、羟基、C 1-6烷基、C 1-6烷氧基-C(=O)-、C 6-10芳基、C 3-10环烷基、3-10个原子组成的杂环基或5-10个原子组成的杂芳基;
    或者,R 5、R 9和与它们相连的原子共同形成3-8个原子组成的杂环基;
    各R 6和R 8独立地为氢、氘、C 1-6烷基、氘代C 1-6烷基、C 2-6烯基、C 2-6炔基、羟基取代的C 1-6烷基、氨基取代的C 1-6烷基、氰基取代的C 1-6烷基、C 1-6烷氧基C 1-6烷基、卤代C 1-6烷基、C 6-10芳基、C 3-10环烷基、3-10个原子组成的杂环基或5-10个原子组成的杂芳基;
    各R 7、R 7a和R 7b独立地为氢、C 1-6烷基、氘代C 1-6烷基、C 2-6烯基、C 2-6炔基或卤代C 1-6烷基;
    各R 10独立地为=O、H、D、卤素、CN、氨基、羟基、硝基、羧基、C 1-4烷基、C 2-4烯基、C 2-4炔基、氘代C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4烷氧基C 1-4烷基、羟基取代的C 1-4烷基、氰基取代的C 1-4烷基、氨基取代的C 1-4烷基、卤代C 1-4烷基、C 1-4烷基酰基、C 1-4烷基磺酰基、C 1-4烷氧基酰基、C 1-4烷氨基酰基、氨基酰基、氨基磺酰基、C 1-4烷氧基磺酰基、C 1-4烷氨基磺酰基、C 1-4酰氧基、C 6-10芳基、C 3-6环烷基、3-6个原子组成的杂环基或5-10个原子组成的杂芳基;
    各m、n和t独立地为0、1、2、3或4;
    p为1、2、3或4;
    各r独立地为0、1、2、3或4。
  2. 根据权利要求1所述的化合物,其中,环C为5-7个原子组成的杂环基或5-6个原子组成的杂芳基。
  3. 根据权利要求1或2所述的化合物,其中,环C为以下子结构式:
    Figure PCTCN2018072988-appb-100002
    其中,环C通过N原子与
    Figure PCTCN2018072988-appb-100003
    相连;
    Figure PCTCN2018072988-appb-100004
    Figure PCTCN2018072988-appb-100005
    独立地为单键或双键;
    Figure PCTCN2018072988-appb-100006
    Figure PCTCN2018072988-appb-100007
    为单键时,各Z、Z 1和Z 2独立地为CH 2或NH;
    Figure PCTCN2018072988-appb-100008
    Figure PCTCN2018072988-appb-100009
    为双键时,各Z、Z 1和Z 2独立地为CH或N;
    Z 4为CH 2或NH;
    各Z 3和Z 5独立地为CH 2、NH、S或O;
    q为0、1或2。
  4. 根据权利要求1-3任意一项所述的化合物,其中,环C为
    Figure PCTCN2018072988-appb-100010
    Figure PCTCN2018072988-appb-100011
    其中,环C通过N原子与
    Figure PCTCN2018072988-appb-100012
    相连。
  5. 根据权利要求1-4任意一项所述的化合物,其为如式(II)所示的化合物或式(II)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
    Figure PCTCN2018072988-appb-100013
    其中,各
    Figure PCTCN2018072988-appb-100014
    Figure PCTCN2018072988-appb-100015
    独立地为单键或双键;
    Figure PCTCN2018072988-appb-100016
    Figure PCTCN2018072988-appb-100017
    为单键时,各Z和Z 1独立地为CH 2或NH;当
    Figure PCTCN2018072988-appb-100018
    Figure PCTCN2018072988-appb-100019
    为双键时,各Z和Z 1独立地为CH或N。
  6. 根据权利要求1-5任意一项所述的化合物,其中,各R 3独立地为C 1-4烷基、C 3-8碳环基、C 6-10芳基、3-8个原子组成的杂环基或5-10个原子组成的杂芳基;
    其中,各R 3独立任选地被1、2、3、4或5个R 3a所取代。
  7. 根据权利要求1-6任意一项所述的化合物,其中,各R 3独立地为甲基、乙基、苯基、吡咯基、吡唑基、咪唑基、咪唑啉基、***基、吡啶基、嘧啶基、吲哚基、吲唑基、苯并呋喃基、苯并噻吩基、苯并噻唑基、苯并咪唑基、苯并异噁唑基、苯并异噻唑基、喹啉基、异喹啉基或喹唑啉基;
    其中,各R 3独立任选地被1、2、3、4或5个R 3a所取代。
  8. 根据权利要求1-7任意一项所述的化合物,其为如式(III)所示的化合物或式(III)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
    Figure PCTCN2018072988-appb-100020
    其中,各
    Figure PCTCN2018072988-appb-100021
    Figure PCTCN2018072988-appb-100022
    独立地为单键或双键;
    Figure PCTCN2018072988-appb-100023
    Figure PCTCN2018072988-appb-100024
    为单键时,Z为CH 2或NH;当
    Figure PCTCN2018072988-appb-100025
    Figure PCTCN2018072988-appb-100026
    为双键时,Z为CH或N;
    f为0、1、2、3、4或5;
    g为0、1、2、3、4、5、6、7或8。
  9. 根据权利要求1-8任意一项所述的化合物,其中,环A为C 7-12双环碳环基、C 8-12双环芳基、7-12个原子组成的双环杂环基或7-12个原子组成的双环杂芳基。
  10. 根据权利要求1-9任意一项所述的化合物,其中,环A为以下子结构式:
    Figure PCTCN2018072988-appb-100027
    其中,各T 1、T 2、T 3、T 4、T 5、T 6、T 7和T 8独立地为-CH-或-N-;
    各V 1、V 2、V 3和V 4独立地为-CH 2-、-NH-、-O-、-S-、-S(=O)-、-S(=O) 2-或-C(=O)-;
    各k和j独立地为0、1、2、3或4。
  11. 根据权利要求1-10任意一项所述的化合物,其中,环A为
    Figure PCTCN2018072988-appb-100028
    Figure PCTCN2018072988-appb-100029
    Figure PCTCN2018072988-appb-100030
  12. 根据权利要求1-11任意一项所述的化合物,其中,环B为C 5-8碳环基、C 6-10芳基、5-8个原子组成的杂环基或5-10个原子组成的杂芳基;
    各R 2独立地为H、氘、F、Cl、Br、I、-(CR 7aR 7b) rCN、C 1-4烷基、氘代C 1-4烷基、卤代C 1-4烷基、C 2-4烯基、C 2-4炔基、R 8O-C 2-4亚烯基、R 8OC(=O)-C 2-4亚烯基、R 8O-C 2-4亚炔基、R 8OC(=O)-C 2-4亚炔基、-N=C=S、-N=C=O、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=G)R 6、-(CR 7aR 7b) rN(R 5c)C(=G)R 6、-(CR 7aR 7b) rOC(=G)R 6、-(CR 7aR 7b) rC(=G)OR 8、-(CR 7aR 7b) rN(R 5c)C(=G)OR 8、-(CR 7aR 7b) rOC(=G)OR 8、-(CR 7aR 7b) rC(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=G)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)=C(R 7)NR 5aR 5b、-(CR 7aR 7b) rS(=O)R 6、-(CR 7aR 7b) rN(R 5c)S(=O)R 6、-(CR 7aR 7b) rS(=O)OR 8、-(CR 7aR 7b) rN(R 5c)S(=O)OR 8、-(CR 7aR 7b) rS(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rN(R 5c)S(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-8环烷基、-(CR 7aR 7b) r-C 6-10芳基、-(CR 7aR 7b) r-(3-8个原子组成的杂环基)、-(CR 7aR 7b) r-(5-10个原子组成的杂芳基)或D-甘露糖基;其中,各G独立地为O、S或NR 5d
    各R 2独立任选地被1、2、3或4个R 2a所取代。
  13. 根据权利要求1-12任意一项所述的化合物,其中,环B为以下子结构式:
    Figure PCTCN2018072988-appb-100031
    Figure PCTCN2018072988-appb-100032
    其中,各Q 1、Q 6和Q 9独立地为CH 2、NH、O、S、C(=O)、S(=O)或S(=O) 2
    各Q 2、Q 3、Q 4、Q 5、Q 7和Q 8独立地为CH或N;
    s为0、1、2或3;
    各R 2独立地为H、氘、F、Cl、Br、I、-(CR 7aR 7b) rCN、C 1-3烷基、氘代C 1-3烷基、卤代C 1-3烷基、C 2-4烯基、R 8O-C 2-6亚烯基、R 8OC(=O)-C 2-6亚烯基、R 8O-C 2-6亚炔基、R 8OC(=O)-C 2-6亚炔基、-N=C=S、-(CR 7aR 7b) rOR 8、-(CR 7aR 7b) rNR 5aR 5b、-(CR 7aR 7b) rC(=O)R 6、-(CR 7aR 7b) rOC(=G)R 6、-(CR 7aR 7b) rN(R 5c)C(=O)R 6、-(CR 7aR 7b) rC(=O)OR 8、-(CR 7aR 7b) rN(R 5c)C(=O)OR 8、-(CR 7aR 7b) rC(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=O)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=S)NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=N-R 5d)NR 5aR 5b、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rN(R 5c)S(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)S(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-6环烷基、-(CR 7aR 7b) r-C 6-10芳基、-(CR 7aR 7b) r-(3-6个原子组成的杂环基)或-(CR 7aR 7b) r-(5-10个原子组成的杂芳基);其中,各R 2独立任选地被1、2、3或4个R 2a所取代。
  14. 根据权利要求1-13任意一项所述的化合物,其中,环B为
    Figure PCTCN2018072988-appb-100033
    Figure PCTCN2018072988-appb-100034
    Figure PCTCN2018072988-appb-100035
    各R 2独立地为H、氘、F、Cl、Br、I、CN、-CH 2-CN、-CH 2CH 2-CN、甲基、乙基、丙基、丁基、氘代甲基、二氘代甲基、一氘代甲基、三氟甲基、二氟甲基、2,2-二氟乙基、1,2-二氟乙基、2,2,2-三氟乙基、乙烯基、丙烯基、烯丙基、HO-乙烯基、MeO-乙烯基、HOC(=O)-乙烯基、MeOC(=O)-乙烯基、-N=C=S、-OH、-OMe、-O(i-Pr)、-O(t-Bu)、-CH 2-OH、-CH 2CH 2-OH、-CH 2-OMe、-CH 2CH 2-OMe、-CH 2-O(i-Pr)、-CH 2-O(t-Bu)、苯基-O-、吡啶基-O-、嘧啶基-O-、吡嗪基-O-、哒嗪基-O-、吡咯基-O-、吡唑基-O-、噻唑基-O-、噁唑基-O-、噁二唑基-O-、咪唑基-O-、噻二唑基-O-、-NH 2、-NHMe、-N(Me) 2、-CH 2-NH 2、-CH 2CH 2-NH 2、-CH 2-NHMe、-CH 2-N(Me) 2、苯基-NH-、吡啶基-NH-、嘧啶基-NH-、吡嗪基-NH-、哒嗪基-NH-、吡咯基-NH-、吡唑基-NH-、噻唑基-NH-、噁唑基-NH-、噁二唑基-NH-、咪唑基-NH-、噻二唑基-NH-、-C(=O)Me、-C(=O)Et、-CH 2-C(=O)Me、-CH 2-C(=O)Et、-NHC(=O)Me、-C(=O)OH、-C(=O)OMe、-NHC(=O)OMe、-NHC(=O)OH、-NHC(=O)OEt、-NHC(=O)O(t-Bu)、-C(=O)NH 2、-C(=O)NHMe、-NHC(=O)NH 2、-NHC(=S)NH 2、-NHC(=NH)NH 2、-S(=O) 2Me、-NHS(=O) 2Me、-S(=O) 2OH、-S(=O) 2OMe、-NHS(=O) 2OH、-NHS(=O) 2OMe、-S(=O) 2NH 2、-NHS(=O) 2NH 2、环丙基、环丙基甲基、环丁基、环戊基、环己基、四氢吡喃基、二氧六环基、哌啶基、哌嗪基、吗啉基、四氢呋喃基、四氢吡咯基、环氧丙基、环氧丁基、氮杂环丁基、二氢吡啶基、二氢吡喃基、苯基、苄基、吡啶基、嘧啶基、吡嗪基、哒嗪基、吡咯基、吡唑基、噻唑基、噁唑基、噁二唑基、咪唑基、噻二唑基、***基或四唑基;
    其中,各R 2独立任选地被1、2、3或4个R 2a所取代。
  15. 根据权利要求1-14所述的化合物,其中,各R 1独立地为=O、H、氘、F、Cl、Br、I、-CN、甲基、乙基、丙基、丁基、三氟甲基、-OH、-OMe、-CH 2OMe、-CH 2OH、-NH 2、-NHMe、-N(Me) 2、-C(=O)Me、-C(=O)OH、-C(=O)OMe、-C(=O)NH 2、-S(=O) 2Me、-S(=O) 2OH、-S(=O) 2OMe、-S(=O) 2NH 2、环丙基、环丙基甲基、环丁基、环戊基、环丁基、苯基、吡啶基、嘧啶基、吡嗪基、哒嗪基、吡咯基、咪唑基、吡唑基、***基、四唑基、环氧烷基、环氧丁基、氮杂环丁基、四氢呋喃基、哌啶基、吡咯烷基、哌嗪基或吗啉基;其中,各R 1独立任选地被1、2、3或4个R 1a所取代;
    各R 4独立地为H、氘、F、Cl、Br、I、羟基、氨基、甲基、乙基、丙基、丁基、三氟甲基、二氟甲基、 1,2-二氟乙基、三氟甲氧基、二氟甲氧基、-OMe、-OEt、-O(t-Bu)、-CH 2OH、-CH 2CH 2OH、-CN、-CH 2CN、-CH 2NH 2、-CH 2CH 2NH 2、-CH 2OMe、-CH 2CH 2OMe、-NHMe或-N(Me) 2;或者,任意两个R 4和与它们相连的原子共同形成C 3-6环烷基或3-6个原子组成的杂环基;
    各R 9、R 1a、R 2a和R 3a独立地为=O、氢、氘、F、Cl、Br、I、-(CR 7aR 7b) rCN、硝基、C 1-4烷基、卤代C 1-4烷基、C 2-4烯基、C 2-4炔基、R 8O-C 2-4亚烯基、R 8OC(=O)-C 2-4亚烯基、R 8O-C 2-4亚炔基、R 8OC(=O)-C 2-4亚炔基、-(CR 7aR 7b) r-OR 8、-(CR 7aR 7b) r-NR 5aR 5b、-(CR 7aR 7b) rN(R 5c)C(=O)R 6、-(CR 7aR 7b) rOC(=O)R 6、-(CR 7aR 7b) rCOOH、-(CR 7aR 7b) rC(=O)OC 1-4烷基、-(CR 7aR 7b) rC(=O)C 1-4烷基、-(CR 7aR 7b) rS(=O) 2R 6、-(CR 7aR 7b) rOS(=O) 2R 6、-(CR 7aR 7b) rN(R 5c)S(=O) 2R 6、-(CR 7aR 7b) rS(=O) 2OR 8、-(CR 7aR 7b) rS(=O) 2NR 5aR 5b、-(CR 7aR 7b) r-C 3-6环烷基、-(CR 7aR 7b) r-C 6-10芳基、-(CR 7aR 7b) r-(3-6个原子组成的杂环基)或-(CR 7aR 7b) r-(5-6个原子组成的杂芳基);
    或者,任意两个R 9与和它们相连的原子共同形成C 3-6环烷基或3-6个原子组成的杂环基;
    其中,各R 9、R 1a、R 2a和R 3a独立任选地被1、2、3、4或5个R 10所取代。
  16. 根据权利要求1-15任意一项所述的化合物,其中,各R 1独立地为=O、H、氘、F、Cl、Br、I、氰基、甲基、乙基、丙基、丁基、-OH、-OMe、-CH 2OMe、-CH 2OH、-NH 2、-NHMe、-N(Me) 2、-C(=O)Me、-C(=O)OH、-C(=O)OMe、-C(=O)NH 2、-S(=O) 2Me、-S(=O) 2OH、-S(=O) 2OMe或-S(=O) 2NH 2;其中,各R 1独立任选地被1、2、3或4个R 1a所取代;
    各R 4独立地为H、氘、F、Cl、Br、I、羟基、氨基、甲基、乙基、丙基、丁基、乙烯基、丙烯基、烯丙基、氘代甲基、三氟甲基、二氟甲基、1,2-二氟乙基、-OMe、-OEt、-CH 2OH、-CH 2CH 2OH、-CN、-CH 2CN、-CH 2NH 2、-CH 2CH 2NH 2、-CH 2OMe、-CH 2CH 2OMe、-NHMe或-N(Me) 2;或者,任意两个R 4和与它们相连的原子共同形成环丙基、环丁基、环氧烷基或氮杂环丁基;
    各R 9、R 1a、R 2a和R 3a独立地为=O、氢、氘、F、Cl、Br、I、-CN、甲基、乙基、丙基、丁基、三氟甲基、二氟甲基、2,2-二氟乙基、-CH 2CN、-CH 2CH 2CN、乙烯基、丙烯基、烯丙基、乙炔基、丙炔基、-OH、-OMe、-O(i-Pr)、-O(t-Bu)、-CH 2-OH、-CH 2CH 2-OH、-CH 2-OMe、-CH 2CH 2-OMe、-CH 2-O(i-Pr)、-CH 2-O(t-Bu)、-NH 2、-NHMe、-N(Me) 2、-CH 2-NH 2、-CH 2CH 2-NH 2、-CH 2-NHMe、-CH 2-N(Me) 2、-NHC(=O)Me、-CH 2COOH、-COOH、-C(=O)OMe、-CH 2C(=O)OMe、-C(=O)Me、-CH 2C(=O)Me、环丙基、环丁基、环戊基、环己基、环氧烷基、环氧丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基、四氢吡喃基、二氢吡喃基、苯基、苄基、吡啶基、嘧啶基、吡嗪基、呋喃基、噻吩基、吡咯基、吡唑基、咪唑基、噁唑基、噻唑基、噁二唑基、噻二唑基、***基或四唑基;
    或者,任意两个R 9与和它们相连的原子共同形成环丙基、环丁基、环戊基、环己基、环氧烷基、环氧丁基、氮杂环丁基、四氢呋喃基、吡咯烷基、哌啶基或哌嗪基;
    其中,各R 9、R 1a、R 2a和R 3a独立任选地被1、2、3、4或5个R 10所取代。
  17. 根据权利要求1-16任意一项所述的化合物,其中,各R 5、R 5a、R 5b、R 5c和R 5d独立地为氢、氰基、羟基、C 1-4烷基、C 1-4烷氧基-C(=O)-、C 6-10芳基、C 3-6环烷基、3-6个原子组成的杂环基或5-6个原子组成的杂芳基;
    各R 6和R 8独立地为氢、氘、C 1-4烷基、氘代C 1-4烷基、C 2-4烯基、C 2-4炔基、羟基取代的C 1-4烷基、氨基取代的C 1-4烷基、氰基取代的C 1-4烷基、C 1-4烷氧基C 1-4烷基、卤代C 1-4烷基、C 6-10芳基、C 3-6环烷基、3-6个原子组成的杂环基或5-6个原子组成的杂芳基;
    各R 7、R 7a和R 7b独立地为氢、C 1-4烷基、氘代C 1-4烷基或卤代C 1-4烷基;
    各R 10独立地为=O、H、D、F、Cl、Br、I、CN、氨基、羟基、硝基、羧基、C 1-3烷基、C 2-4烯基、C 2-4炔基、氘代C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4烷氧基C 1-4烷基、羟基取代的C 1-4烷基、氰基取代的 C 1-4烷基、氨基取代的C 1-4烷基、卤代C 1-4烷基、C 1-4烷基酰基、C 1-4烷基磺酰基、C 1-4烷氧基酰基、C 1-4烷氨基酰基、苯基、C 3-6环烷基、3-6个原子组成的杂环基或5-6个原子组成的杂芳基。
  18. 根据权利要求1-17任意一项所述的化合物,其中,各R 5、R 5a、R 5b、R 5c和R 5d独立地为氢、氰基、羟基、甲基、乙基、丙基、丁基、甲氧基-C(=O)-、乙氧基-C(=O)-、异丙氧基-C(=O)-、叔丁氧基-C(=O)-、苯基、环丙基、环丁基、环戊基、环己基、环氧烷基、环氧丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基、四氢吡喃基、二氢吡喃基、苯基、苄基、吡啶基、嘧啶基、吡嗪基、呋喃基、噻吩基、吡咯基、吡唑基、咪唑基、噁唑基、噻唑基、噁二唑基、噻二唑基、***基或四唑基;
    各R 6和R 8独立地为氢、氘、甲基、乙基、丙基、丁基、氘代甲基、乙烯基、丙炔基、羟基甲基、羟基乙基、甲氧基甲基、甲氧基乙基、乙氧基甲基、叔丁氧基甲基、叔丁氧基乙基、乙氧基乙基、异丙氧基乙基、三氟甲基、二氟甲基、1,2-二氟乙基、2,2-二氟乙基、苯基、环丙基、环己基、氮杂环丁基、四氢呋喃基、吡咯烷基、哌啶基、哌嗪基、吡啶基、吡咯基、嘧啶基、吡唑基、吡嗪基、呋喃基、噻唑基、噁唑基、噁二唑基、噻二唑基、咪唑基、***基或四唑基;
    各R 7、R 7a和R 7b独立地为氢、甲基、乙基、丙基、丁基、氘代甲基、三氟甲基、二氟甲基、1,2-二氟乙基或2,2-二氟乙基;
    各R 10独立地为=O、H、D、F、Cl、Br、I、CN、氨基、羟基、硝基、羧基、甲基、乙基、丙基、丁基、氘代甲基、甲氧基、甲氨基、二甲氨基、甲氧基甲基、甲氧基乙基、叔丁氧基甲基、叔丁氧基乙基、羟基甲基、羟基乙基、CN取代的甲基、CN取代的乙基、氨基甲基、氨基乙基、三氟甲基、二氟甲基、1,2-二氟乙基或2,2-二氟乙基、甲基酰基、乙基酰基、甲基磺酰基、甲氧基酰基、叔丁氧基酰基、甲氨基酰基、二甲氨基酰基、苯基、环丙基、环丁基、环戊基、环己基、环氧丙烷基、环氧丁基、氮杂环丁基、四氢呋喃基、吡咯烷基、哌啶基、哌嗪基、吗啉基、呋喃基、噻吩基、吡咯基、吡唑基、咪唑基、噁二唑基、***基、四唑基、吡啶基、嘧啶基或吡嗪基。
  19. 一种化合物,其为以下其中之一的结构:
    Figure PCTCN2018072988-appb-100036
    Figure PCTCN2018072988-appb-100037
    Figure PCTCN2018072988-appb-100038
    或上述结构的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药。
  20. 一种药物组合物,其包含权利要求1-19任意一项所述的化合物;其中,所述药物组合物进一步包含药学上可接受的载体、赋形剂、稀释剂和媒介物的至少一种。
  21. 如权利要求1-19任意一项所述的化合物或如权利要求20所述的药物组合物在制备药物中的用途,其中,所述的药物用于预防、治疗或减轻血栓栓塞性疾病。
  22. 根据权利要求21所述的用途,其中,所述血栓栓塞性疾病为动脉心血管血栓栓塞性疾病、静脉心血管血栓栓塞性疾病、及心室或外周循环中的血栓栓塞性疾病。
  23. 根据权利要求21所述的用途,其中,所述血栓栓塞性疾病为心绞痛、急性冠状动脉综合征、心房颤动、心肌梗塞、短暂性缺血发作、中风、动脉粥样硬化、外周闭塞性动脉疾病、静脉血栓形成、深静脉血栓形成、血栓性静脉炎、动脉栓塞、冠状动脉血栓形成、脑动脉血栓形成、脑栓塞、肾栓塞、肺栓塞、以及因医疗植入物、器械或程序中的血液暴露于人造表面从而促使血栓形成而引起的血栓形成。
  24. 如权利要求1-19任意一项所述的化合物或如权利要求20所述的药物组合物在制备药物中的用途,其中,所述的药物用于抑制因子XIa和/或血浆激肽释放酶的活性。
  25. 如权利要求1-19任意一项所述的化合物或如权利要求20所述的药物组合物用于预防、治疗或减轻血栓栓塞性疾病。
  26. 根据权利要求25所述的化合物或组合物,其中,所述血栓栓塞性疾病为动脉心血管血栓栓塞性疾病、静脉心血管血栓栓塞性疾病、及心室或外周循环中的血栓栓塞性疾病。
  27. 根据权利要求26所述的化合物或组合物,其中,所述血栓栓塞性疾病为心绞痛、急性冠状动脉综合征、心房颤动、心肌梗塞、短暂性缺血发作、中风、动脉粥样硬化、外周闭塞性动脉疾病、静脉血栓形成、深静脉血栓形成、血栓性静脉炎、动脉栓塞、冠状动脉血栓形成、脑动脉血栓形成、脑栓塞、肾栓塞、肺栓塞、以及因医疗植入物、器械或程序中的血液暴露于人造表面从而促使血栓形成而引起的血栓形成。
  28. 如权利要求1-19任意一项所述的化合物或如权利要求20所述的药物组合物用于抑制因子XIa和/ 或血浆激肽释放酶的活性。
  29. 一种使用如权利要求1-19任意一项所述的化合物或如权利要求20所述的药物组合物预防、治疗或减轻血栓栓塞性疾病的方法。
  30. 根据权利要求29所述的方法,其中,所述血栓栓塞性疾病为动脉心血管血栓栓塞性疾病、静脉心血管血栓栓塞性疾病、及心室或外周循环中的血栓栓塞性疾病。
  31. 根据权利要求30所述的方法,其中,所述血栓栓塞性疾病为心绞痛、急性冠状动脉综合征、心房颤动、心肌梗塞、短暂性缺血发作、中风、动脉粥样硬化、外周闭塞性动脉疾病、静脉血栓形成、深静脉血栓形成、血栓性静脉炎、动脉栓塞、冠状动脉血栓形成、脑动脉血栓形成、脑栓塞、肾栓塞、肺栓塞、以及因医疗植入物、器械或程序中的血液暴露于人造表面从而促使血栓形成而引起的血栓形成。
  32. 一种使用如权利要求1-19任意一项所述的化合物或如权利要求20所述的药物组合物抑制因子XIa和/或血浆激肽释放酶的活性的方法。
PCT/CN2018/072988 2017-01-18 2018-01-17 凝血因子XIa抑制剂及其用途 WO2018133793A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201880004854.3A CN110062757B (zh) 2017-01-18 2018-01-17 凝血因子XIa抑制剂及其用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710036621 2017-01-18
CN201710036621.9 2017-01-18

Publications (1)

Publication Number Publication Date
WO2018133793A1 true WO2018133793A1 (zh) 2018-07-26

Family

ID=62908170

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/072988 WO2018133793A1 (zh) 2017-01-18 2018-01-17 凝血因子XIa抑制剂及其用途

Country Status (2)

Country Link
CN (1) CN110062757B (zh)
WO (1) WO2018133793A1 (zh)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020210613A1 (en) * 2019-04-11 2020-10-15 Bristol-Myers Squibb Company Novel synthetic options towards the manufacture of (6r,10s)-10- {4-[5-chloro-2-(4-chloro-1h-1,2,3-triazol-1-yl)phenyl]-6-oxo-1(6h)- pyrimidinyl}- 1-(difluoromethyl)-6-methyl-1,4,7,8,9,10-hexahydro-11,15 -(metheno)pyrazolo [4,3-b] [1,7] diazacyclotetradecin-5(6h)-one
WO2020211781A1 (zh) 2019-04-16 2020-10-22 南京明德新药研发有限公司 作为XIa因子抑制剂的大环衍生物
WO2021013209A1 (zh) 2019-07-23 2021-01-28 南京明德新药研发有限公司 作为XIa因子抑制剂的大环衍生物
CN113004286A (zh) * 2019-12-20 2021-06-22 成都康弘药业集团股份有限公司 作为血浆激肽释放酶抑制剂的三环类化合物及其用途
WO2021136390A1 (zh) * 2019-12-31 2021-07-08 上海京新生物医药有限公司 凝血因子XIa抑制剂
CN114984820A (zh) * 2022-05-27 2022-09-02 吉林大学 一种采血管晃动式达到血液混合的摇匀台
CN115215867A (zh) * 2021-04-21 2022-10-21 上海美悦生物科技发展有限公司 FXIa抑制剂及其药物组合物、制备方法和用途

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103702999A (zh) * 2011-08-05 2014-04-02 百时美施贵宝公司 作为因子xia抑制剂的环状p1连接体
CN104507924A (zh) * 2012-08-03 2015-04-08 百时美施贵宝公司 二氢吡啶酮p1作为凝血因子xia抑制剂
CN104520289A (zh) * 2012-08-03 2015-04-15 百时美施贵宝公司 作为凝血因子xia抑制剂的二氢吡啶酮p1
WO2015116882A1 (en) * 2014-01-31 2015-08-06 Bristol-Myers Squibb Company Macrocyclic factor xia inhibitors condensed with heterocycles
WO2016053455A1 (en) * 2014-10-01 2016-04-07 Bristol-Myers Squibb Company Pyrimidinones as factor xia inhibitors
CN105980384A (zh) * 2014-01-31 2016-09-28 百时美施贵宝公司 作为因子xia抑制剂的具有杂环p2'基团的大环化合物

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103702999A (zh) * 2011-08-05 2014-04-02 百时美施贵宝公司 作为因子xia抑制剂的环状p1连接体
CN104507924A (zh) * 2012-08-03 2015-04-08 百时美施贵宝公司 二氢吡啶酮p1作为凝血因子xia抑制剂
CN104520289A (zh) * 2012-08-03 2015-04-15 百时美施贵宝公司 作为凝血因子xia抑制剂的二氢吡啶酮p1
WO2015116882A1 (en) * 2014-01-31 2015-08-06 Bristol-Myers Squibb Company Macrocyclic factor xia inhibitors condensed with heterocycles
CN105980384A (zh) * 2014-01-31 2016-09-28 百时美施贵宝公司 作为因子xia抑制剂的具有杂环p2'基团的大环化合物
WO2016053455A1 (en) * 2014-10-01 2016-04-07 Bristol-Myers Squibb Company Pyrimidinones as factor xia inhibitors

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020210613A1 (en) * 2019-04-11 2020-10-15 Bristol-Myers Squibb Company Novel synthetic options towards the manufacture of (6r,10s)-10- {4-[5-chloro-2-(4-chloro-1h-1,2,3-triazol-1-yl)phenyl]-6-oxo-1(6h)- pyrimidinyl}- 1-(difluoromethyl)-6-methyl-1,4,7,8,9,10-hexahydro-11,15 -(metheno)pyrazolo [4,3-b] [1,7] diazacyclotetradecin-5(6h)-one
CN114375290A (zh) * 2019-04-11 2022-04-19 百时美施贵宝公司 制备(6r,10s)-10-{4-[5-氯-2-(4-氯-1h-1,2,3-***-1-基)苯基]-6-氧代基-1(6h)-嘧啶基}-1-(二氧甲基)-6-甲基-1,4,7,8,9,10-六氢-11,15-(亚甲桥基)吡唑并[4,3-b][1,7]二氮杂环十四炔-5(6h)-酮的新颖合成选项
EP4234555A3 (en) * 2019-04-11 2024-01-03 Bristol-Myers Squibb Company Novel synthetic options towards the manufacture of (6r,10s)-10- {4-[5-chloro-2-(4-chloro-1h-1,2,3-triazol-1-yl)phenyl]-6-oxo-1(6h)- pyrimidinyl}- 1-(difluoromethyl)-6-methyl-1,4,7,8,9,10-hexahydro-11,15 -(metheno)pyrazolo [4,3-b] [1,7] diazacyclotetradecin-5(6h)-one
WO2020211781A1 (zh) 2019-04-16 2020-10-22 南京明德新药研发有限公司 作为XIa因子抑制剂的大环衍生物
JP7286001B2 (ja) 2019-07-23 2023-06-02 メッドシャイン ディスカバリー インコーポレイテッド 第XIa因子阻害剤としての大環状誘導体
WO2021013209A1 (zh) 2019-07-23 2021-01-28 南京明德新药研发有限公司 作为XIa因子抑制剂的大环衍生物
CN114008047A (zh) * 2019-07-23 2022-02-01 南京明德新药研发有限公司 作为XIa因子抑制剂的大环衍生物
JP2022541927A (ja) * 2019-07-23 2022-09-28 メッドシャイン ディスカバリー インコーポレイテッド 第XIa因子阻害剤としての大環状誘導体
EP4006029A4 (en) * 2019-07-23 2023-06-21 Medshine Discovery Inc. MACROCYCLIC DERIVATIVES AS FACTOR XIA INHIBITORS
CN114008047B (zh) * 2019-07-23 2023-03-21 南京明德新药研发有限公司 作为XIa因子抑制剂的大环衍生物
CN113004286A (zh) * 2019-12-20 2021-06-22 成都康弘药业集团股份有限公司 作为血浆激肽释放酶抑制剂的三环类化合物及其用途
CN113004286B (zh) * 2019-12-20 2022-08-12 成都康弘药业集团股份有限公司 作为血浆激肽释放酶抑制剂的三环类化合物及其用途
WO2021136390A1 (zh) * 2019-12-31 2021-07-08 上海京新生物医药有限公司 凝血因子XIa抑制剂
CN115215867A (zh) * 2021-04-21 2022-10-21 上海美悦生物科技发展有限公司 FXIa抑制剂及其药物组合物、制备方法和用途
CN115215867B (zh) * 2021-04-21 2023-12-26 上海美悦生物科技发展有限公司 FXIa抑制剂及其药物组合物、制备方法和用途
CN114984820B (zh) * 2022-05-27 2023-09-29 吉林大学 一种采血管晃动式达到血液混合的摇匀台
CN114984820A (zh) * 2022-05-27 2022-09-02 吉林大学 一种采血管晃动式达到血液混合的摇匀台

Also Published As

Publication number Publication date
CN110062757B (zh) 2022-03-04
CN110062757A (zh) 2019-07-26

Similar Documents

Publication Publication Date Title
WO2018133793A1 (zh) 凝血因子XIa抑制剂及其用途
KR102599788B1 (ko) 단백질 키나제의 조절물질로서 키랄 디아릴 매크로사이클
TW202214605A (zh) 免疫調節劑、組合物及其方法
ES2800313T3 (es) Compuestos de heteroarilo bicíclico sustituidos
JP2016526540A (ja) 置換ベンジルピラゾール
TW201041872A (en) Amino ester derivatives, salts thereof and methods of use
KR102587619B1 (ko) Nik 억제제로서의 시아노인돌린 유도체
TW200829589A (en) Compound for inhibiting mitotic progression
CN110041253B (zh) 吡啶类n-氧化衍生物及其制备方法和应用
TW202140449A (zh) 用作選擇性Aurora A抑制劑的新型雜環化合物
CN107108586A (zh) 多环类间变性淋巴瘤激酶抑制剂
KR20220088375A (ko) 피롤아미드 화합물 및 그 용도
US9663498B2 (en) Aromatic heterocyclic compounds and their application in pharmaceuticals
EP3484874B1 (en) Monocyclic heteroaryl substituted compounds
CN110312717A (zh) 取代的稠合杂芳三环化合物作为激酶抑制剂及其应用
CN104804017B (zh) 噁唑烷酮类化合物及其在药物中的应用
JP7149854B2 (ja) Pi3k beta阻害薬としての二環式ピリジン、ピラジンおよびピリミジン誘導体
JP2017538765A (ja) PI3Kβ阻害剤としての複素環連結イミダゾピリダジン誘導体
CN113493453B (zh) 稠合芳香环类衍生物、其制备方法及其在医药上的应用
EP4294798A1 (en) Factor xiia inhibitors
KR20220153595A (ko) AhR 신호전달의 조절에 유용한 피리도피리미딘 유도체
JPWO2006106804A1 (ja) 環内窒素原子にアシル基が結合した三環系スピロ化合物
CN104530046A (zh) 二氮杂螺类化合物及其在药物中的应用
JP2019532998A (ja) Pfkfb阻害剤としての置換キノキサリン誘導体
WO2017181993A1 (zh) 一种新的喜巴辛类似物及其在医药中的应用

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: 18741028

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: 18741028

Country of ref document: EP

Kind code of ref document: A1