CN108690016B - Pyrazolopyridines and uses thereof - Google Patents

Pyrazolopyridines and uses thereof Download PDF

Info

Publication number
CN108690016B
CN108690016B CN201810315504.0A CN201810315504A CN108690016B CN 108690016 B CN108690016 B CN 108690016B CN 201810315504 A CN201810315504 A CN 201810315504A CN 108690016 B CN108690016 B CN 108690016B
Authority
CN
China
Prior art keywords
radical
alkyl
compound
group
independently
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201810315504.0A
Other languages
Chinese (zh)
Other versions
CN108690016A (en
Inventor
王晓军
左应林
余川
阳传文
王建成
李静
曹生田
吴方园
张英俊
S·戈尔德曼
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Guangdong HEC Pharmaceutical
Original Assignee
Sunshine Lake Pharma Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sunshine Lake Pharma Co Ltd filed Critical Sunshine Lake Pharma Co Ltd
Publication of CN108690016A publication Critical patent/CN108690016A/en
Application granted granted Critical
Publication of CN108690016B publication Critical patent/CN108690016B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • 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
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers

Abstract

The invention relates to a pyrazolopyridine compound and application thereof, and further relates to a pharmaceutical composition containing the compound. The compounds of the invention or the pharmaceutical compositions may be used as soluble guanylate cyclase stimulators.

Description

Pyrazolopyridines and uses thereof
Technical Field
The invention belongs to the technical field of medicines, particularly relates to a pyrazolopyridine compound and application thereof, and further relates to a pharmaceutical composition containing the compound. The compound or the pharmaceutical composition may be used as a soluble guanylate cyclase stimulator (sGC stimulator).
Background
Guanosine monophosphate (cGMP) is a second messenger with intracellular message transmission; it forms the NO/cGMP system together with Nitric Oxide (NO) which is released from endothelial tissue and transports hormones and mechanical signals. Guanylate cyclase catalyzes the biosynthesis of cGMP from Guanosine Triphosphate (GTP). The presently known representatives of this family can be divided into two groups according to structural features and ligand type: a particulate guanylate cyclase activatable by natriuretic peptides, and a soluble guanylate cyclase activatable by NO. Soluble guanylate cyclase consists of two subunits, and each heterodimer is very likely to contain one heme, which is part of the regulatory center. NO can bind to the iron atom of heme, thereby significantly increasing the activity of the enzyme. Conversely, heme-free enzymes are not activated by NO. Carbon monoxide (CO) can also attach to the central iron atom of heme, but the activation of CO is significantly lower than that of NO.
By the formation of cGMP, and the resulting modulation of phosphodiesterases, ion channels and protein kinases, guanylate cyclase plays an important role in various physiological processes, in particular in the relaxation and proliferation of smooth muscle cells, in platelet aggregation and platelet adhesion and in neuronal signal transmission, and in diseases which are based on disturbances of the above processes. Under pathophysiological conditions, the NO/cGMP system can be inhibited, which can lead to diseases such as hypertension, platelet activation, cell proliferation, endothelial dysfunction, arteriosclerosis, angina pectoris, heart failure, myocardial infarction, thrombosis, stroke and sexual dysfunction.
Based on the expected higher effectiveness and fewer side effects, the treatment of these diseases by modulation of the cGMP signaling pathway through NO-independent pathways is very promising. Therapeutic stimulation of soluble guanylate cyclase by NO-based compounds such as organic nitrates activates soluble guanylate cyclase by attack of the central iron atom of heme by NO formed by biotransformation. In addition to side effects, the development of tolerance is also one of the drawbacks of such treatment methods.
Soluble guanylate cyclase (sGC), widely present in mammalian cytosol, is a relatively high content in the lung and brain and is a key signal transducer in the Nitric Oxide (NO) -sGC-cyclic guanosine monophosphate (cGMP) signaling pathway, and sGC, when activated in vivo, catalyzes the conversion of GTP to cGMP. cGMP is an important secondary messenger molecule that initiates a series of downstream cascade reactions by activating downstream effector molecules, such as Phosphodiesterase (PDE), cyclic nucleotide gated ion Channel (CNG), and protein kinase g (pkg), etc., and plays important physiological functions in the gastrointestinal system, blood circulation system, and nervous system, such as promoting vasodilation of blood vessels and smooth muscles, inhibiting platelet aggregation, vascular remodeling, apoptosis and inflammation generation, and participating in neurotransmission.
sGC is a sensor and receptor for NO, and contains two subunits, α and β, each having 3 domains, including a heme domain, a central domain, and a catalytic domain, wherein the heme domains of the two subunits share a heme. After NO binds to the heme of sGC, sGC is activated, catalyzing the conversion of its substrate GTP to the secondary signal molecule cGMP, opening the PKG signaling pathway, leading to vasodilation. sGC plays an important role as a receptor of NO in the cardiovascular system and the nervous system, and disorder of NO signaling causes disorder of physiological functions, thereby causing various diseases. Therefore, sGC stimulators are receiving increasing attention as novel drugs that can directly activate sGC.
The sGC stimulator has a dual action mechanism on sGC in vivo, and can directly activate sGC when the concentration of NO is low; when NO has a certain level, the NO can act synergistically with NO, so that sGC is activated to catalyze the conversion of a substrate Guanosine Triphosphate (GTP) into a secondary messenger molecule guanosine cyclophosphate (cGMP), and further the sGC is involved in regulating a plurality of important physiological processes, such as promotion of vasodilation and smooth muscle relaxation; inhibiting platelet aggregation, vascular remodeling, etc. The sGC stimulator activates sGC and also mediates other signal pathways such as TGF-beta and the like, thereby playing roles in resisting fibrosis, tumors and the like. Therefore, the NO-sGC-cGMP pathway becomes an effective target for treating various cardiovascular diseases, such as pulmonary hypertension, acute heart failure, angina pectoris, myocardial infarction-induced vascular remodeling and the like.
Over the years, a plurality of researchers develop medicaments such as NO donors, phosphodiesterase inhibitors, NO-independent sGC stimulators and the like, wherein the NO-independent sGC stimulators can avoid the defects of easy tolerance, weak action specificity, short duration and the like of the traditional NO donor medicaments and are paid extensive attention.
Summary of the invention
The present invention provides a novel pyrazolopyridine compound as a sGC stimulator (sGC stimulator), a pharmaceutical composition thereof, and use of the compound or the pharmaceutical composition for the preparation of a medicament for the treatment and/or prevention of sGC mediated diseases, such as heart failure, sclerosis, systemic sclerosis, sickle cell anemia, cardiac achalasia, pulmonary fibrosis, idiopathic pulmonary fibrosis, cystic fibrosis, pulmonary hypertension, and the like.
In one aspect, the invention relates to a compound that is a compound of formula (I) or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, ester, pharmaceutically acceptable salt, or prodrug of a compound of formula (I),
Figure BDA0001623638800000021
wherein the content of the first and second substances,
E 1 is N or CR 1 ;E 2 Is N or CR 2 ;E 3 Is N or CR 3 ;E 4 Is N or CR 4
L is- (CR) a R b ) t -、-(CR a R b ) f -O-、-(CR a R b ) f -S-、-(CR a R b ) f -S(=O)-、-(CR a R b ) f -S(=O) 2 -、-(CR a R b ) f -N(R c )-、-(CR a R b ) f -C(=O)N(R c )-、-C(=O)N(R c )-(CR a R b ) f -or- (CR) a R b ) f -C(=O)-;
t is 1,2, 3 or 4;
f is 0, 1,2, 3 or 4;
each R a And R b Independently H, D, F, Cl, Br, I, CN, amino, hydroxyl, sulfhydryl, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Alkylamino, halogeno C 1-6 Alkoxy radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclyl or C 1-5 A heteroaryl group; or, R a And R b Together form a carbonyl group; or, R a 、R b And together with the C atom to which they are attached form a carbocyclic ring of 3 to 8 atoms or a heterocyclic ring of 3 to 8 atoms;
R c is H, D, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group;
ring C1 being C 6-10 Aryl radical, C 1-9 Heteroaryl group, C 3-10 Carbocyclic radical or C 2-9 A heterocyclic group;
each R 0 Independently H, D, F, Cl, Br, I, CN, NO 2 Amino, hydroxy, mercapto, oxo, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino, acyl, sulfonyl, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl or C 1-5 Heteroaryl C 1-6 An alkyl group; wherein, the amino, the hydroxyl, the mercapto and the C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino, acyl, sulfonyl, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl and C 1-5 Heteroaryl C 1-6 Alkyl is unsubstituted or independently optionally substituted by 1,2, 3 or 4 groups selected from D, F, Cl, Br, I, CN, NO 2 Amino, hydroxy, mercapto, oxo, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl and C 1-5 Heteroaryl, or a substituted heteroaryl;
each R 6 And R 7 Independently H, D, F, Cl, Br, I, CN, NO 2 Amino, hydroxyl, mercapto, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino radical, hydroxy radical C 1-6 Alkyl, cyano C 1-6 Alkyl, amino C 1-6 Alkyl, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, acyl, sulfonyl or C 1-6 An alkoxy group;
each R 1 、R 2 、R 3 And R 4 Independently H, D, F, Cl, Br, I, CN, NO 2 Mercapto group, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 3-10 A cycloalkyl group, a,C 2-10 Heterocyclic group, C 6-10 Aryl radical, C 1-9 Heteroaryl group, C 3-10 Cycloalkyl radical C 1-6 Alkyl radical, C 2-10 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl C 1-6 Alkyl, -N ═ NR 8 、-N=CR d R e 、-(CR d R e ) j CN、-(CR d R e ) j NR 8a R 8b 、-(CR d R e ) j C(=O)(CR f R g ) k NR 8a R 8b 、-(CR d R e ) j C(=O)(CR f R g ) k OR 9 、-(CR d R e ) j OR 9 、-(CR d R e ) j S(=O) 2 (CR f R g ) k OR 9 、-(CR d R e ) j S(=O) 2 (CR f R g ) k NR 8a R 8b 、-(CR d R e ) j N(R 8 )(CR d R e ) p C(=O)(CR f R g ) k OR 9 、-(CR d R e ) j N(R 8 )C(=O)(CR f R g ) k NR 8a R 8b 、-(CR d R e ) j N(R 8 )C(=O)R 10 、-(CR d R e ) j N(R 8 )S(=O) 2 R 11 、-(CR d R e ) j S(=O) 2 R 11 、-(CR d R e ) j OS(=O) 2 R 11 、-(CR d R e ) j OC(=O)(CR f R g ) k OR 9 、-(CR d R e ) j OC(=O)R 10 Or- (CR) d R e ) j C(=O)R 10 (ii) a The R is 1 、R 2 、R 3 And R 4 Unsubstituted or optionally independently substituted by 1,2, 3 or 4R x Substitution;
R 5 d, F, Cl, Br, I, CN, mercapto, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl radical, C 1-5 Heteroaryl C 1-6 Alkyl, - (CR) h R i ) g CN、-(CR h R i ) g NR 8c R 8d 、-(CR h R i ) g C(=O)(CR j R k ) h NR 8c R 8d 、-(CR h R i ) g C(=O)(CR j R k ) h OR 9a 、-(CR h R i ) g OR 9a 、-(CR h R i ) g S(=O) 2 (CR j R k ) h OR 9a 、-(CR h R i ) g S(=O) 2 (CR j R k ) h NR 8c R 8d 、-(CR h R i ) g N(R 8e )(CR h R i ) i C(=O)(CR j R k ) h OR 9a 、-(CR h R i ) g N(R 8e )C(=O)R 10a 、-(CR h R i ) g N(R 8e )C(=O)(CR j R k ) h NR 8c R 8d 、-(CR h R i ) g N(R 8e )S(=O) 2 R 11a 、-(CR h R i ) g S(=O) 2 R 11a 、-(CR h R i ) g OS(=O) 2 R 11a 、-(CR h R i ) g OC(=O)(CR j R k ) h OR 9a 、-(CR h R i ) g OC(=O)R 10a Or- (CR) h R i ) g C(=O)R 10a (ii) a The R is 5 Unsubstituted or optionally substituted by 1,23 or 4R y Substitution;
each R 8 And R 8e Independently H, D, C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy radical C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl or C 1-5 Heteroaryl C 1-6 An alkyl group;
each R 8a 、R 8b 、R 8c And R 8d Independently H, D, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, carboxyl C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy radical C 1-6 Alkyl radical, C 1-6 Alkoxyacyl group, C 1-6 Alkyl acyl radical, C 1-6 Alkylamino acyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl acyl, C 2-5 Heterocyclyl acyl radical, C 6-10 Aryl acyl radical, C 1-5 Heteroaryl acyl, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl or C 1-5 Heteroaryl C 1-6 An alkyl group; or, R 8a 、R 8b And together with the N atom to which they are attached form a heterocyclic ring of 3 to 10 atoms or a heteroaromatic ring of 5 to 10 atoms; or, R 8c 、R 8d And together with the N atom to which they are attached form a heterocyclic ring of 3 to 10 atoms or a heteroaromatic ring of 5 to 10 atoms;
each R d 、R e 、R f 、R g 、R h 、R i 、R j And R k Independently H, D, F, Cl, Br, I, CN, amino, hydroxyl, mercapto, or a salt thereof,C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino, acyl, sulfonyl, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group; or, R d And R e 、R f And R g 、R h And R i 、R j And R k Each independently and together with the atoms to which they are attached form a carbonyl group, a carbocyclic ring of 3 to 6 atoms, or a heterocyclic ring of 3 to 6 atoms;
each R 11 And R 11a Independently H, D, amino, hydroxy, mercapto, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino radical, hydroxy radical C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, carboxyl C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group;
each R 10 And R 10a Independently H, D, amino, hydroxy, mercapto, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, carboxyl C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group;
each R 9 And R 9a Independently H, D, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl, acyl, sulfonyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, carboxyl C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl or C 1-5 Heteroaryl C 1-6 An alkyl group;
each R x And R y Independently oxo, H, D, F, Cl, Br, I, CN, amino, hydroxy, mercapto, carboxyl, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino, acyl, sulfonyl, C 1-6 Alkyl acyl radical, C 1-6 Alkylsulfonyl radical, C 1-6 Alkylamido radical, C 1-6 Alkylsulfonylamino group, C 1-6 Alkoxyamido group, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group;
n is 0, 1,2, 3 or 4;
each g, h, i, j, k and p is independently 0, 1,2, 3 or 4;
with the proviso that the compounds do not include the following:
1- (4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidin-5-yl) cyclopropanecarboxylic acid ethyl ester, 1- (4-chloro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b]Pyridin-3-yl) pyrimidin-5-yl) cyclopropanecarboxylic acid ethyl ester, 2- (4-chloro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b]Pyridin-3-yl) pyrimidin-5-yl) acetic acid ethyl ester, N 4 - (2, 4-dimethoxybenzyl) -2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidine-4, 5-diamine, 2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b]Pyridin-3-yl) pyrimidine-4, 5, 6-triamine, 2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b]Pyridin-3-yl) pyrimidine-4, 5-diamine, (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidin-5-yl) carbamic acid methyl ester, (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyri-dineAzolo [3,4-b]Pyridin-3-yl) pyrimidin-5-yl) (methyl) carbamic acid methyl ester, (4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidin-5-yl) carbamic acid methyl ester, (4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidin-5-yl) (methyl) carbamic acid methyl ester and (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidin-5-yl) (2,2, 2-trifluoroethyl) carbamic acid methyl ester.
In some embodiments, ring C1 is:
Figure BDA0001623638800000041
Figure BDA0001623638800000051
in some embodiments, the compound of formula (I) according to the present invention is a compound of formula (II) or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, ester, pharmaceutically acceptable salt or prodrug of a compound of formula (II),
Figure BDA0001623638800000052
in some embodiments, R 5 D, F, Cl, Br, I, CN, mercapto, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-4 Alkyl radical, C 2-5 Heterocyclyl radical C 1-4 Alkyl radical, C 6-10 Aryl radical C 1-4 Alkyl radical, C 1-5 Heteroaryl C 1-4 Alkyl, - (CR) h R i ) g CN、-NR 8c R 8d 、-C(=O)NR 8c R 8d 、-C(=O)OR 9a 、-OR 9a 、-S(=O) 2 OR 9a 、-S(=O) 2 NR 8c R 8d 、-N(R 8e )(CR h R i ) i C(=O)(CR j R k ) h OR 9a 、-N(R 8e )C(=O)R 10a 、-N(R 8e )C(=O)NR 8c R 8d 、-N(R 8e )S(=O) 2 R 11a 、-S(=O) 2 R 11a 、-OS(=O) 2 R 11a 、-OC(=O)OR 9a 、-OC(=O)R 10a or-C (═ O) R 10a (ii) a The R is 5 Unsubstituted or optionally substituted by 1,2, 3 or 4R y And (4) substitution.
In other embodiments, R 5 D, F, Cl, Br, I, CN, mercapto, methyl, ethyl, propyl, butyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, trifluoromethyl, difluoromethyl, 2-difluoroethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclobutyl, azetidinyl, pyrrolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, morpholinyl, oxomorpholinyl, thiomorpholinyl, 4-dioxothiomorpholinyl, tetrahydrofuranyl, oxazolidinyl, thiazolidinyl, 1-dioxoisothiazolidinyl, oxo-1, 3-oxazinidinyl, phenyl, pyridinyl, pyrimidinyl, pyrazolyl, imidazolyl, pyrrolyl, thiazolyl, oxazolyl, triazolyl, tetrazolyl, thienyl, furyl, - (CR R) h R i ) g CN、-NR 8c R 8d 、-C(=O)NR 8c R 8d 、-C(=O)OR 9a 、-OR 9a 、-S(=O) 2 OR 9a 、-S(=O) 2 NR 8c R 8d 、-N(R 8e )C(=O)(CR j R k ) h OR 9a 、-N(R 8e )C(=O)R 10a 、-N(R 8e )C(=O)NR 8c R 8d 、-N(R 8e )S(=O) 2 R 11a 、-S(=O) 2 R 11a or-C (═ O) R 10a (ii) a The R is 5 Unsubstituted or optionally substituted by 1,2, 3 or 4R y And (4) substitution.
In some embodiments, each R is 1 、R 2 、R 3 And R 4 Independently H, D, F, Cl, Br, I, CN, NO 2 Mercapto group, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-4 Alkyl radical, C 2-5 Heterocyclyl radical C 1-4 Alkyl radical, C 6-10 Aryl radical C 1-4 Alkyl radical, C 1-5 Heteroaryl C 1-4 Alkyl, - (CR) d R e ) j CN、-NR 8a R 8b 、-C(=O)NR 8a R 8b 、-C(=O)OR 9 、-OR 9 、-S(=O) 2 OR 9 、-S(=O) 2 NR 8a R 8b 、-N(R 8 )(CR d R e ) p C(=O)(CR f R g ) k OR 9 、-N(R 8 )C(=O)NR 8a R 8b 、-N(R 8 )C(=O)R 10 、-N(R 8 )S(=O) 2 R 11 、-S(=O) 2 R 11 、-OS(=O) 2 R 11 、-OC(=O)OR 9 、-OC(=O)R 10 or-C (═ O) R 10 (ii) a The R is 1 、R 2 、R 3 And R 4 Unsubstituted or optionally independently substituted by 1,2, 3 or 4R x And (4) substitution.
In other embodiments, each R is 1 、R 2 、R 3 And R 4 Independently H, D, F, Cl, Br, I, CN, NO 2 Mercapto, methyl, ethyl, propyl, butyl, trifluoromethyl, 2-difluoroethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclobutyl, azetidinyl, pyrrolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, morpholinyl, oxomorpholinyl, thiomorpholinyl, 4-dioxothiomorpholinyl, tetrahydrofuranyl, oxazolidinyl, thiazolidinyl, 1-dioxoisothiazolidinyl, oxo-1, 3-oxazinylalkyl, phenyl, pyridinyl, pyrimidinyl, pyrazolyl, imidazolyl, pyrrolyl, thiazolyl, oxazolyl, triazolyl, tetrazolyl, thienyl, furyl, -NR 8a R 8b 、-C(=O)NR 8a R 8b 、-C(=O)OR 9 、-OR 9 、-S(=O) 2 OR 9 、-S(=O) 2 NR 8a R 8b 、-N(R 8 )(CR d R e ) p C(=O)(CR f R g ) k OR 9 、-N(R 8 )C(=O)NR 8a R 8b 、-N(R 8 )C(=O)R 10 、-N(R 8 )S(=O) 2 R 11 、-S(=O) 2 R 11 or-C (═ O) R 10 (ii) a The R is 1 、R 2 、R 3 And R 4 Unsubstituted or optionally independently substituted by 1,2, 3 or 4R x And (4) substitution.
In some embodiments, each R is 8a 、R 8b 、R 8c And R 8d Independently H, D, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, carboxyl C 1-4 Alkyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy radical C 1-4 Alkyl radical, C 1-4 Alkoxyacyl group, C 1-4 Alkyl acyl radical, C 1-4 Alkylamino acyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl acyl, C 2-5 Heterocyclyl acyl radical, C 6-10 Aryl acyl radical, C 1-5 Heteroaryl acyl, C 3-6 Cycloalkyl radical C 1-4 Alkyl radical, C 2-5 Heterocyclyl radical C 1-4 Alkyl radical, C 6-10 Aryl radical C 1-4 Alkyl or C 1-5 Heteroaryl C 1-4 An alkyl group; or, R 8a 、R 8b And together with the N atom to which they are attached form a heterocyclic ring of 3 to 6 atoms or a heteroaromatic ring of 5 to 6 atoms; or, R 8c 、R 8d And together with the N atom to which they are attached form a heterocyclic ring of 3 to 6 atoms or a heteroaromatic ring of 5 to 6 atoms;
each R 9 And R 9a Independently H, D, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl, acyl, sulfonyl, hydroxy C 1-4 Alkyl radicalAmino group C 1-4 Alkyl, cyano C 1-4 Alkyl, carboxyl C 1-4 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-6 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-3 Alkyl radical, C 2-5 Heterocyclyl radical C 1-3 Alkyl radical, C 6-10 Aryl radical C 1-3 Alkyl or C 1-5 Heteroaryl C 1-3 An alkyl group.
In other embodiments, each R is 8a 、R 8b 、R 8c And R 8d Independently H, D, methyl, ethyl, propyl, butyl, hydroxymethyl, hydroxyethyl, hydroxypropyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, trifluoromethoxy, chloroethyl, 2,2, 2-trifluoroethyl, 2, 2-difluoroethyl, 2-chloro-1-methylethyl, methoxymethyl, methoxyethyl, methoxyacyl, ethoxyacyl, propoxoyl, methylacyl, ethylacoyl, methylaminoacyl, ethylaminoacyl, cyclopropylacyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholinyl, phenyl, pyridinyl, pyrimidinyl, cyclopropylacyl, cyclopentanoyl, cyclohexylacyl, cyclopropyloxy, tetrahydrofuranoyl, tetrahydropyranoyl, piperidinoyl, piperazinyl, morpholinyl, tetrahydrothienyl, pyrrolidinylacyl, phenylacyl, pyridinylacyl, pyrimidinyl, pyrrolanoyl, imidazolyl, etc, Pyrazolylacyl, thienylacyl, furylacyl, cyclopropylmethyl, cyclobutylmethyl, tetrahydropyranylmethyl, tetrahydropyranylethyl, tetrahydrofuranylmethyl, tetrahydrofuranylethyl, piperidinylmethyl, piperidinylethyl, morpholinylmethyl, phenylmethyl, phenylethyl, pyridylmethyl, pyridylethyl or C 2-5 Heterocyclyl radical C 1-3 An alkyl group; or, R 8a 、R 8b And the N atom to which they are attached form an azetidine, pyrrolidine, oxazolidine, thiazolidine, isothiazolidine, piperidine, morpholine, piperazine, thiomorpholine, 1, 3-oxazinane, pyrrole, pyrazole, imidazole or triazole group; or, R 8c 、R 8d And with themThe linked N atoms together form an azetidine, pyrrolidine, oxazolidine, thiazolidine, isothiazolidine, piperidine, morpholine, piperazine, thiomorpholine, 1, 3-oxazinane, pyrrole, pyrazole, imidazole or triazolyl group;
each R 9 And R 9a Independently H, D, methyl, ethyl, propyl, butyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, trifluoromethyl, chloroethyl, 2,2, 2-trifluoroethyl, 2, 2-difluoroethyl, 2-chloro-1-methylethyl, hydroxymethyl, hydroxyethyl, hydroxypropyl, aminomethyl, aminoethyl, cyanomethyl, cyanoethyl, carboxymethyl, carboxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, tetrahydrofuryl, morpholinyl, piperidinyl, piperazinyl, phenyl, pyridinyl, pyrimidinyl, cyclopropylmethyl, cyclobutylmethyl, phenylmethyl, phenylethyl, pyridylmethyl, pyridylethyl or C 2-5 Heterocyclyl radical C 1-3 An alkyl group.
In some embodiments, each R is 8 And R 8e Independently H, D, methyl, ethyl, propyl, butyl, hydroxymethyl, hydroxyethyl, hydroxypropyl, aminomethyl, cyanomethyl, cyanoethyl, difluoromethyl, 2-difluoroethyl, methoxymethyl, methoxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, tetrahydrofuranyl, piperidinyl, morpholinyl, tetrahydropyranyl, phenyl, pyridinyl, pyrimidinyl, cyclopropylmethyl, cyclobutylmethyl, morpholinylmethyl, piperidinylmethyl, tetrahydropyranyl ethyl, phenylmethyl, phenylethyl, pyridylmethyl or pyrimidinylmethyl;
each R 10 And R 10a Independently H, D, amino, hydroxy, mercapto, methyl, ethyl, propyl, butyl, trifluoromethyl, chloroethyl, 2,2, 2-trifluoroethyl, 2, 2-difluoroethyl, 2-chloro-1-methylethyl, methoxy, ethoxy, propoxy, trifluoromethoxy, methylamino, dimethylamino, hydroxymethyl, hydroxyethyl, hydroxypropyl, aminomethyl, aminoethyl, cyanomethyl, cyanoethyl, carboxymethyl, carboxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydropyranylPyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, morpholinyl, piperidinyl, piperazinyl, phenyl, pyrrolyl, thienyl, furanyl, pyridinyl, or pyrimidinyl;
each R x And R y Independently oxo, H, D, F, Cl, Br, I, CN, amino, hydroxy, mercapto, carboxyl, methyl, ethyl, propyl, butyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, hydroxymethyl, hydroxyethyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, trifluoromethyl, chloroethyl, 2,2, 2-trifluoroethyl, 2, 2-difluoroethyl, 2-chloro-1-methylethyl, methoxy, ethoxy, propoxy, trifluoromethoxy, methylamino, dimethylamino, C 1-3 Alkyl acyl radical, C 1-3 Alkylsulfonyl radical, C 1-3 Alkylamido radical, C 1-3 Alkylsulfonylamino, methoxycarbonylamino, C 2-3 Alkoxyamido, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, C 2-5 Heterocyclyl, phenyl or C 1-5 A heteroaryl group.
In another aspect, the invention relates to a pharmaceutical composition comprising a compound of the invention; it further comprises at least one of a pharmaceutically acceptable carrier, excipient, diluent, adjuvant and vehicle.
In one aspect, the present invention relates to the use of a compound according to the present invention or a pharmaceutical composition according to the present invention for the preparation of a medicament for the treatment and/or prevention of: heart failure, angina pectoris, hypertension, pulmonary hypertension, ischemia, vascular disease, renal disease, thromboembolic disease, male sexual dysfunction, systemic sclerosis, sickle cell anemia, cardiac achalasia, fibrotic disease and/or arteriosclerosis.
In another aspect, the invention relates to the use of a compound according to the invention or a pharmaceutical composition according to the invention for the preparation of a medicament for use as a soluble guanylate cyclase stimulator.
Detailed description of the invention
Definitions and general terms
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated by the accompanying structural and chemical formulas. The invention is intended to cover alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. Those skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated documents, patents, and similar materials differ or contradict this application (including but not limited to defined terminology, application of terminology, described techniques, and the like), this application controls.
It will be further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications referred to herein are incorporated by reference in their entirety.
The following definitions as used herein should be applied unless otherwise indicated. For the purposes of the present invention, the chemical elements are in accordance with the CAS version of the periodic Table of the elements, and the handbook of chemistry and Physics, 75 th edition, 1994. In addition, general principles of Organic Chemistry can be referred to as described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausaltito: 1999, and "March's Advanced Organic Chemistry" by Michael B.Smith and Jerry March, John Wiley & Sons, New York:2007, the entire contents of which are incorporated herein by reference.
The articles "a," "an," and "the" as used herein are intended to include "at least one" or "one or more" unless otherwise indicated or clearly contradicted by context. Thus, as used herein, the articles refer to articles of one or more than one (i.e., at least one) object. For example, "a component" refers to one or more components, i.e., there may be more than one component contemplated for use or use in embodiments of the described embodiments.
The term "patient" as used herein refers to humans (including adults and children) or other animals. In some embodiments, "patient" refers to a human.
The term "comprising" is open-ended, i.e. includes the elements indicated in the present invention, but does not exclude other elements.
"stereoisomers" refers to compounds having the same chemical structure but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric isomers (cis/trans), atropisomers, and the like.
"enantiomer" refers to two isomers of a compound that are not overlapping but are in mirror image relationship to each other.
"diastereomer" refers to a stereoisomer having two or more chiral centers and whose molecules are not mirror images of each other. Diastereomers have different physical properties, such as melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may be separated by high resolution analytical procedures such as electrophoresis and chromatography, e.g., HPLC.
The stereochemical definitions and rules used in the present invention generally follow the general definitions of 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.
Any asymmetric atom (e.g., carbon, etc.) of a compound disclosed herein can exist in racemic or enantiomerically enriched forms, such as the (R) -, (S) -or (R, S) -configuration. In certain embodiments, each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R) -or (S) -configuration.
Any resulting mixture of stereoisomers may be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, depending on differences in the physicochemical properties of the components, for example, by chromatography and/or fractional crystallization.
The term "tautomer" or "tautomeric form" refers to structural isomers having different energies that can interconvert by a low energy barrier (low energy barrier). If tautomerism is possible (e.g., in solution), then the chemical equilibrium of the tautomer can be reached. For example, proton tautomers (also known as proton transfer tautomers) include interconversions by proton migration, such as keto-enol isomerization and imine-enamine isomerization. Valence tautomers (valenctautomers) include interconversion by recombination of some of the bonding electrons. A specific example of keto-enol tautomerism is the tautomerism of the pentan-2, 4-dione and 4-hydroxypent-3-en-2-one tautomers. Another example of tautomerism is phenol-ketone tautomerism. One specific example of phenol-ketone tautomerism is the tautomerism of pyridin-4-ol and pyridin-4 (1H) -one tautomers. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
The compounds of the invention may be optionally substituted with one or more substituents, as described herein, in compounds of the general formula above, or as specifically exemplified, sub-classes, and classes of compounds encompassed by the invention.
It is understood that the term "optionally substituted" may be used interchangeably with the term "substituted or unsubstituted". In general, the term "substituted" means that one or more hydrogen atoms in a given structure are replaced with a particular substituent. "optionally substituted" means that the given structure or group is unsubstituted or substituted with one or more specific substituents. Unless otherwise indicated, an optional substituent group may be substituted at each substitutable position of the substituted group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, the substituents may be substituted at each position, identically or differently. Wherein the substituent may be, but is not limited to, oxo (═ O), hydrogen, deuterium, cyano, nitro, halogen, hydroxy, mercapto, amino, alkyl, haloalkyl, hydroxyalkyl, cyanoalkyl, aminoalkyl, alkoxy, haloalkoxy, acyl, acyloxy, sulfonyl, sulfinyl, carboxy, cycloalkyl, cycloalkylalkyl, cycloalkyloxy, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl, arylalkyl, aryloxy, heteroaryl, heteroarylalkyl, heteroaryloxy, and the like.
In addition, unless otherwise explicitly indicated, the descriptions of the terms "… independently" and "… independently" and "… independently" used in the present invention are interchangeable, and should be understood in a broad sense, which means that the specific items expressed between the same symbols do not affect each other in different groups, or that the specific items expressed between the same symbols in the same groups do not affect each other. Likewise, the above broad understanding should be made with respect to "independent" in describing manner "… independently optionally.
The terms "optional" or "optionally" mean that the subsequently described event or circumstance may, but need not, occur, i.e., that the description includes instances where said event or circumstance occurs and instances where it does not. For example, "optionally substituted with 1,2, 3, or 4 substituents independently selected from …" includes the case where the group is substituted with 1, or 2, or 3, or 4 of the substituents described, as well as the case where the group is not substituted with the substituents described. Further, when the group is substituted with 1 or more of the substituents, the substituents are independent of each other, that is, the 1 or more substituents may be different from each other or the same.
In the various parts of this specification, substituents of the disclosed compounds are disclosed in terms of group type or range. It is specifically intended that the invention include all such individual members of the group classes and rangesEach independent sub-combination. For example, the term "C 1 -C 6 Alkyl "or" C 1-6 Alkyl "means in particular independently disclosed methyl, ethyl, C 3 Alkyl radical, C 4 Alkyl radical, C 5 Alkyl and C 6 An alkyl group; "C 1-4 Alkyl "refers specifically to independently disclosed C 1 Alkyl (i.e. methyl), C 2 Alkyl (i.e. ethyl), C 3 Alkyl (i.e. propyl, including n-propyl and isopropyl), C 4 Alkyl (i.e., butyl, including n-butyl, isobutyl, sec-butyl, and tert-butyl).
In each of the parts of the invention, linking substituents are described. Where the structure clearly requires a linking group, the markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for the variable recites "alkyl" or "aryl," it is understood that the "alkyl" or "aryl" represents an attached alkylene group or arylene group, respectively.
The term "alkyl" or "alkyl group" as used herein, denotes a saturated, straight or branched chain, monovalent hydrocarbon group containing 1 to 20 carbon atoms, wherein the alkyl group may be optionally substituted with one or more substituents as described herein. In some embodiments, the alkyl group contains 1 to 12 carbon atoms; in other embodiments, the alkyl group contains 1-6 carbon atoms, i.e., C 1-6 An alkyl group; in still other embodiments, the alkyl group contains 1-4 carbon atoms, i.e., C 1-4 An alkyl group; in still other embodiments, the alkyl group contains 1 to 3 carbon atoms, i.e., C 1-3 An alkyl group. In some embodiments, C is described herein 1-6 The alkyl group may be C 1-4 An alkyl group; in other embodiments, C is described herein 1-6 The alkyl group may be C 1-3 An alkyl group.
Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl, tert-butyl), n-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, n-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2, 3-dimethyl-2-butyl, 3, 3-dimethyl-2-butyl, n-heptyl, n-octyl, and the like.
In some specific structures, when an alkyl group is explicitly indicated as a linking group, then the alkyl group represents a linked alkylene group, e.g., "C 3-10 Cycloalkyl radical C 1-6 Alkyl group "," C 2-10 Heterocyclyl radical C 1-6 Alkyl group "," C 6-10 Aryl radical C 1-6 Alkyl group "," C 1-9 Heteroaryl C 1-6 Alkyl "or the like 1-6 Alkyl is understood to mean C 1-6 An alkylene group.
The term "alkylene" refers to a saturated divalent hydrocarbon radical resulting from the removal of two hydrogen atoms from a saturated straight or branched chain hydrocarbon radical. Unless otherwise specified, the alkylene group contains 1 to 12 carbon atoms. In some embodiments, the alkylene group contains 1 to 6 carbon atoms; in other embodiments, the alkylene group contains 1 to 4 carbon atoms; in still other embodiments, the alkylene group contains 1 to 3 carbon atoms; in still other embodiments, the alkylene group contains 1 to 2 carbon atoms. Examples of this include methylene (-CH) 2 -), ethylene (including-CH) 2 CH 2 -or-CH (CH) 3 ) -), isopropylidene (including-CH (CH) 3 )CH 2 -or-C (CH) 3 ) 2 -) and the like. Wherein said alkylene may be optionally substituted with one or more substituents as described herein.
The term "alkenyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein at least one carbon-carbon sp is present 2 A double bond, wherein the alkenyl group may be optionally substituted with one or more substituents described herein, including the positioning of "cis" and "tans", or the positioning of "E" and "Z". In some embodiments, alkenyl groups contain 2 to 8 carbon atoms; in other embodiments, alkenyl groups contain 2 to 6 carbon atoms; in still other embodiments, alkenyl groupsThe radicals contain 2 to 4 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl (-CH ═ CH) 2 ) Allyl (-CH) 2 CH=CH 2 ) And so on.
The term "alkynyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one carbon-carbon sp triple bond, wherein the alkynyl radical may be optionally substituted with one or more substituents as described herein. In one embodiment, alkynyl groups contain 2-8 carbon atoms; in another embodiment, alkynyl groups contain 2-6 carbon atoms; in yet another embodiment, alkynyl groups contain 2-4 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl (-C.ident.CH), propargyl (-CH) 2 C.ident.CH), 1-propynyl (-C.ident.C-CH) 3 ) And so on.
The term "alkoxy" means an alkyl group attached to the rest of the molecule through an oxygen atom, wherein the alkyl group has the meaning as described herein. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-butoxy, and the like.
The term "haloalkyl" or "haloalkoxy" means an alkyl or alkoxy group substituted with one or more halogen atoms, examples of which include, but are not limited to, trifluoromethyl, trifluoromethoxy, chloroethyl (e.g., 2-chloroethyl), 2,2, 2-trifluoroethyl, 2, 2-difluoroethyl, 2-chloro-1-methylethyl, and the like.
The term "amino" denotes the group-NH 2
The term "carboxyl" denotes the group-COOH.
The terms "hydroxy", "cyano", "nitro" and "mercapto" denote the radicals-OH, -CN, -NO, respectively 2 -SH. The term "oxo" represents a group ═ O.
The term "alkylamino" denotes the group-NH 2 Substituted by one or two alkyl groups, wherein the alkyl groups have the meaning as described herein. Examples of alkylamino groups include, but are not limited to, methylamino, dimethylamino, and the like.
The terms "hydroxyalkyl",cyanoalkyl and aminoalkyl represent, respectively, one or more hydroxy (-OH), cyano (-CN) or amino (-NH) 2 ) Substituted alkyl, said alkyl having the meaning described herein. In some embodiments, the "hydroxyalkyl", "cyanoalkyl" and "aminoalkyl" groups of the present invention are each "hydroxy C 1-6 Alkyl group and cyano group C 1-6 Alkyl group and amino group C 1-6 Alkyl ", i.e. substituted by one or more hydroxy (-OH), cyano (-CN) or amino (-NH) groups 2 ) Substituted C 1-6 An alkyl group. In other embodiments, "hydroxy C 1-6 Alkyl group and cyano group C 1-6 Alkyl group and amino group C 1-6 Alkyl is independently of each other' hydroxy C 1-4 Alkyl group and cyano group C 1-4 Alkyl "or" amino C 1-4 Alkyl groups ". Examples include, but are not limited to, hydroxymethyl, hydroxyethyl (e.g., 2-hydroxyethyl), aminomethyl, aminoethyl (e.g., 2-aminoethyl), cyanomethyl, cyanoethyl (e.g., 2-cyanoethyl), and the like.
The terms "hydroxyalkoxy", "cyanoalkoxy", "aminoalkoxy" represent, respectively, one or more hydroxy (-OH), cyano (-CN) or amino (-NH) 2 ) Substituted alkoxy, said alkoxy having the meaning described in the present invention. Examples include, but are not limited to, hydroxymethoxy, hydroxyethoxy, cyanomethoxy, aminomethoxy and the like.
The terms "alkoxyalkyl", "alkylaminoalkyl" denote alkyl substituted by one or more alkoxy or alkylamino groups, respectively, all having the meaning described herein. Examples include, but are not limited to, methoxymethyl, methoxyethyl, methylaminomethyl, methylaminoethyl, and the like.
The term "carbocyclyl" or "carbocycle" denotes a monovalent or polyvalent saturated or partially unsaturated monocyclic, bicyclic or tricyclic ring system containing 3 to 12 ring carbon atoms; wherein the carbocyclic group is non-aromatic and does not contain any aromatic rings. In some embodiments, carbocyclyl contains 3 to 10 ring carbon atoms, e.g., C 3-10 A carbocyclic group; in other embodiments, carbocyclyl contains 3 to 8 ring carbon atoms, e.g., C 3-8 A carbocyclic group; in still other embodiments, carbocyclyl contains 3-6 ring carbon atoms, e.g., C 3-6 A carbocyclic group. Examples of carbocyclyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like; wherein, said C 3-6 Carbocyclyl includes, but is not limited to, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. The carbocyclyl group may be optionally substituted with one or more substituents described herein.
The term "cycloalkyl" denotes a saturated monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 ring carbon atoms. In some embodiments, cycloalkyl groups contain 3 to 10 ring carbon atoms, e.g., C 3-10 A cycloalkyl group; in other embodiments, cycloalkyl groups contain 3 to 8 ring carbon atoms, e.g., C 3-8 A cycloalkyl group; in still other embodiments, cycloalkyl groups contain 3-6 ring carbon atoms, e.g., C 3-6 A cycloalkyl group. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like; wherein, said C 3-6 Cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. The cycloalkyl group may be optionally substituted with one or more substituents described herein.
The term "cycloalkylalkyl" denotes a cycloalkyl group attached to the rest of the molecule through an alkyl group, wherein cycloalkyl and alkyl have the meaning described herein. In the present invention, the mode "C 3-10 Cycloalkyl radical C 1-6 Alkyl "or" C 3-10 Cycloalkyl radical C 1-4 Alkyl "etc. represents C 3-10 Cycloalkyl radicals through C 1-6 Alkyl or C 1-4 The alkyl group is attached to the rest of the molecule. The cycloalkylalkyl group may be optionally substituted with one or more substituents described herein. Examples include, but are not limited to, cyclopropylmethyl, cyclopropylethyl, cyclopentylmethyl, cyclohexylmethyl, cyclohexylethyl, and the like.
The term "heterocyclyl" is meant to encompass 3 to 12A saturated or partially unsaturated monocyclic, bicyclic or tricyclic ring system of ring atoms, wherein at least one ring atom is selected from nitrogen, sulfur and oxygen atoms; wherein the heterocyclic group is non-aromatic and does not contain any aromatic ring. Unless otherwise specified, heterocyclyl may be carbon-or nitrogen-based, and-CH 2 -the group may optionally be replaced by-C (═ O) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atoms of the ring may optionally be oxidized to the N-oxide. The term "heterocyclyl" may be used interchangeably with the term "heterocycle". The heterocyclyl group may be optionally substituted with one or more substituents described herein.
In some embodiments, heterocyclyl is C 2-9 Heterocyclyl, meaning that heterocyclyl contains from 2 to 9 ring carbon atoms and at least one ring heteroatom selected from O, S and N; in other embodiments, heterocyclyl is C 2-7 Heterocyclyl, meaning that heterocyclyl contains from 2 to 7 ring carbon atoms and at least one ring heteroatom selected from O, S and N; in other embodiments, heterocyclyl is C 2-5 Heterocyclyl, meaning heterocyclyl contains 2 to 5 ring carbon atoms and at least one ring heteroatom selected from O, S and N. Examples of heterocyclyl groups include, but are not limited to:
Figure BDA0001623638800000121
Figure BDA0001623638800000122
oxirane, thietanyl, azetidinyl, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, oxazolidinyl, tetrahydrofuranyl, dihydrothienyl, dihydropyranyl, piperidinyl, morpholinyl, tetrahydropyrimidinyl, oxazinanyl, thiomorpholinyl, piperazinyl, and the like. In heterocyclic radicals of-CH 2 Examples of the substitution of the-group by-C (═ O) -include, but are not limited to, 2-oxopyrrolidinyl, 2-piperidinonyl, 3-morpholinonyl, 3-thiomorpholinonyl, oxotetrahydropyrimidinyl and the like. In some embodiments, C of the invention 2-5 Examples of heterocyclyl groups include, but are not limited to: azetidinyl, pyrrolidinyl (i.e., tetrahydropyrrolyl), pyrazolidinyl, piperazinylPyridyl, piperazinyl, morpholinyl, oxomorpholinyl, thiomorpholinyl, 4-dioxothiomorpholinyl, tetrahydrofuranyl, oxazolidinyl (i.e., tetrahydrooxazolyl), thiazolidinyl (i.e., tetrahydrothiazolyl), 1-dioxoisothiazolidinyl (e.g.,
Figure BDA0001623638800000123
) Oxo-1, 3-oxazinalkyl (e.g.,
Figure BDA0001623638800000124
) An oxazinoalkyl group (for example,
Figure BDA0001623638800000125
) Dihydropyridines (e.g., 1, 2-dihydropyridines)
Figure BDA0001623638800000126
) Dihydropyrimidines (e.g., 1, 6-dihydropyrimidines)
Figure BDA0001623638800000127
) Dihydropyrazines (e.g., 1, 2-dihydropyrazines)
Figure BDA0001623638800000128
) Tetrahydropyridine, tetrahydropyrimidine, tetrahydropyrazine, or the like.
The term "heterocyclylalkyl" means that the heterocyclyl group is attached to the rest of the molecule through an alkyl group, where heterocyclyl and alkyl have the meaning described herein. In the present invention, the group "C 2-9 Heterocyclyl radical C 1-6 Alkyl "etc. represents C 2-9 Cycloalkyl radicals through C 1-6 The alkyl group is attached to the rest of the molecule. The heterocyclylalkyl group may be optionally substituted with one or more substituents described herein. Examples include, but are not limited to, tetrahydropyranylmethyl, tetrahydropyranylethyl, tetrahydrofuranylmethyl, tetrahydrofuranylethyl, pyrrolidinylmethyl, piperidinylmethyl, piperidinylethyl, morpholinylmethyl, morpholinylethyl and the like.
The term "halogen" refers to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I).
The term "aryl" denotes a monocyclic, bicyclic and tricyclic carbocyclic ring system containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring is aromatic and has one or more attachment points to the rest of the molecule. The term "aryl" may be used interchangeably with the terms "aromatic ring" or "aromatic ring". Examples of the aryl group may include phenyl, 2, 3-dihydro-1H-indenyl, naphthyl and anthryl. The aryl group may be optionally substituted with one or more substituents described herein. Unless otherwise stated, the group "C 6-14 Aryl "represents an aryl group containing from 6 to 14 ring carbon atoms.
The term "arylalkyl" means that the aryl group is attached to the rest of the molecule through an alkyl group, where aryl and alkyl have the meaning described herein. For example, the group "C 6-14 Aryl radical C 1-6 Alkyl "represents C 6-14 Aryl radicals through C 1-6 The alkyl group is attached to the rest of the molecule. The arylalkyl group can be optionally substituted with one or more substituents described herein. Examples include, but are not limited to, benzyl, phenylethyl, naphthylmethyl, and the like.
The term "heteroaryl" denotes monocyclic, bicyclic and tricyclic ring systems containing 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 ring heteroatoms selected from nitrogen, oxygen, sulfur, and wherein the heteroaryl has one or more attachment points to the rest of the molecule. when-CH is present in the heteroaryl group 2 When it is a group, -CH 2 -the group may optionally be replaced by-C (═ O) -. Unless otherwise indicated, the heteroaryl group may be attached to the rest of the molecule (e.g., the main structure in the general formula) via any reasonable site (which may be C in CH, or N in NH). The term "heteroaryl" may be used interchangeably with the terms "heteroaromatic ring" or "heteroaromatic compound". In some embodiments, heteroaryl is C 1-9 Heteroaryl, meaning that heteroaryl contains 1 to 9 ring carbon atoms and at least one member selected from the group consisting of O, S anda ring heteroatom of N; in other embodiments, heteroaryl is C 1-7 Heteroaryl, meaning that heteroaryl contains 1 to 7 ring carbon atoms and at least one ring heteroatom selected from O, S and N; in other embodiments, heteroaryl is C 1-6 Heteroaryl, meaning that heteroaryl contains 1 to 6 ring carbon atoms and at least one ring heteroatom selected from O, S and N; in other embodiments, heteroaryl is C 1-5 Heteroaryl, meaning that heteroaryl contains 1 to 5 ring carbon atoms and at least one ring heteroatom selected from O, S and N; in other embodiments, heteroaryl is C 1-4 Heteroaryl, meaning that heteroaryl contains 1 to 4 ring carbon atoms and at least one ring heteroatom selected from O, S and N; in other embodiments, heteroaryl is C 1-3 Heteroaryl, meaning that heteroaryl contains 1 to 3 ring carbon atoms and at least one ring heteroatom selected from O, S and N. Examples include, but are not limited to, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyrazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, thienyl, thiazolyl, and the like; the following bicyclic rings are also included, but by no means limited to these: benzimidazolyl, benzofuranyl, benzothienyl, indolyl, oxoindolyl, indolinyl, imidazopyridinyl, pyrazolopyridinyl, pyrazolopyrimidinyl, quinolinyl, isoquinolinyl, quinazolinyl and the like. The heteroaryl group may be optionally substituted with one or more substituents described herein.
The term "heteroarylalkyl" means that the heteroaryl group is attached to the rest of the molecule through an alkyl group, wherein heteroaryl and alkyl have the meaning described herein. The heteroarylalkyl group may be optionally substituted with one or more substituents described herein. Examples include, but are not limited to, pyridylmethyl, pyrrolylethyl, quinolinylmethyl and the like.
The term "x-y atomic constituent" (x, y are each any natural number other than 0, and y > x) means that the cyclic group is composed of x-y ring atoms including carbon atoms and/or heteroatoms such as O, N, S, P, wherein "x-y" includes x, y, and any natural number between x and y. For example, "3-8 atoms", "3-10 atoms", or "3-6 atoms" means that the cyclic group consists of 3-8, 3-10, or 3-6 ring atoms, including carbon atoms or heteroatoms such as O, N, S, P. As another example, "heteroaryl of 6 to 10 atoms" includes heteroaryl of 6, 7, 8, 9, or 10 atoms.
The term "acyl" denotes — C (═ O) -R, where the substituent R is attached to the rest of the molecule through a carbonyl group (-C (═ O) -), where R is a substituent described herein, including, but not limited to, alkyl, alkoxy, hydroxy, amino, cycloalkyl, heterocyclyl, aryl, heteroaryl, and the like. Wherein alkyl, alkoxy, hydroxy, amino, cycloalkyl, heterocyclyl, aryl and heteroaryl have the meaning as indicated in the present invention, and such examples include, but are not limited to, acetyl (-C (═ O) CH 3 ) Carboxy (-C (═ O) OH), methoxyacyl (-C (═ O) OCH 3 ) Carbamoyl (-O-CO) NH 2 ) Phenylformyl, and the like. The term "sulfonyl" denotes-S (═ O) 2 -R, wherein the substituent R is substituted by a sulfonyl (-S (═ O) 2 -) is attached to the rest of the molecule, wherein R is a substituent described herein, including, but not limited to, alkyl, alkoxy, hydroxy, amino, cycloalkyl, heterocyclyl, aryl, heteroaryl, and the like. Wherein alkyl, alkoxy, hydroxy, amino, cycloalkyl, heterocyclyl, aryl and heteroaryl have the meaning as indicated in the present invention, and such examples include, but are not limited to, sulfonic acid groups (-S (═ O) 2 OH), methylsulfonyl (-S (═ O) 2 CH 3 ) Methoxysulfonyl (-S (═ O) 2 OCH 3 ) Aminosulfonyl (-S (═ O) 2 NH 2 ) Phenylsulfonyl, and the like.
The term "sulfinyl" denotes-S (═ O) -R, where the substituent R is attached to the rest of the molecule through sulfinyl (-S (═ O) -), where R is a substituent described herein, including, but not limited to, alkyl, alkoxy, hydroxy, amino, cycloalkyl, heterocyclyl, aryl, heteroaryl, and the like. Wherein the alkyl, alkoxy, hydroxy, amino, cycloalkyl, heterocyclyl, aryl and heteroaryl groupsRadicals have the meaning as indicated in the present invention, and such examples include, but are not limited to, sulfinyl (-O) OH), methylsulfinyl (-S (-O) CH 3 ) Phenylsulfinyl, and the like.
The terms "alkanoyl", "alkylsulfonyl" and "alkanoyl" denote the radicals-C (═ O) -alkyl and-S (═ O), respectively 2 -an alkyl group, wherein the alkyl group has the meaning as described herein. Examples include, but are not limited to, acetyl (-C (═ O) CH 3 ) Methylsulfonyl (-S (═ O) 2 CH 3 ) And the like.
The terms "alkylamido", "alkylsulfonylamino", "alkoxyamido" denote the groups-NH-C (═ O) -alkyl, -NH-S (═ O) 2 -alkyl and-NH-C (═ O) -alkoxy (i.e., -NH-C (═ O) -O-alkyl), wherein both the alkyl and alkoxy have the meaning described herein. Examples include, but are not limited to, methylamido (-NH-C (═ O) CH 3 ) Methyl sulfonyl amino (-NH-S (═ O) 2 CH 3 ) Methoxyamido (-NH-C (═ O) -O-CH) 3 ) And the like.
The terms "alkoxyacyl", "alkylaminoacyl", "cycloalkylacyl", "heterocyclylacyl", "arylacyl", "heteroarylacyl" denote the radicals-C (═ O) -alkoxy, -C (═ O) -alkylamino, -C (═ O) -cycloalkyl, -C (═ O) -heterocyclyl, -C (═ O) -aryl, -C (═ O) -heteroaryl, respectively, where alkoxy, alkylamino, cycloalkyl, heterocyclyl, aryl, heteroaryl all have the meanings described in the present invention. Examples include, but are not limited to, methoxyacyl, ethoxyacyl, propoxycarbonyl, methylaminoacyl, ethylaminoacyl, cyclopropylacyl, cyclopentylacyl, cyclohexylacyl, cyclopropyloxyacyl, tetrahydrofuranoyl, tetrahydropyranoyl, piperidinoyl, piperazinoylyl, morpholinoyl, tetrahydrothienylacyl, pyrrolidinylacyl, phenylacyl, pyridinylacyl, pyrimidinylacyl, pyrrolylacyl, imidazoloyl, pyrazolyl, thienylacyl, or furanylacyl, and the like.
As described herein, substituent (R) n The ring system formed by a ring linked to the center by a bond represents that the n substituents R may be substituted at any substitutable position on the ring. For example, formula a represents any possible substituted position on the ring of C1 optionally substituted by n R 0 And (4) substitution. When the C1 ring is a bicyclic structure, R 0 Substitutions may be made at any substitutable position on any one of the rings.
Figure BDA0001623638800000141
The group "- (CR) as described in the present invention a R b ) f The two attachment sites on-O- "are interchangeable with one another and can be attached to the rest of the molecule. For example, when L is a group of formula b as described herein, L (i.e., - (CR) a R b ) f -O-) can be attached to the rest of the molecule (e.g., the pyrazolopyridine structure in formula (I) via the E-terminus or the E' -terminus.
Figure BDA0001623638800000142
As described herein, at "R 8a 、R 8b Together with the N atom to which they are attached form a heterocyclic ring of 3 to 6 atoms or a heteroaromatic ring of 5 to 6 atoms, or "R 8c 、R 8d And the N atom to which they are attached, form a 3-6 atom-containing heterocyclic ring or a 5-6 atom-containing heteroaromatic ring ", both of which have the meanings described in the present invention, and examples thereof can be found in the description of the above-mentioned" heterocyclic group "and" heteroaryl group ". Specifically, the heterocyclic ring with 3 to 6 atoms or the heteroaromatic ring with 5 to 6 atoms includes, but is not limited to, the following groups: azetidine, pyrrolidine (i.e.,
Figure BDA0001623638800000151
) An oxazolidine (i.e.,
Figure BDA0001623638800000152
) A thiazolidine (i.e.,
Figure BDA0001623638800000153
) An isothiazolidine (i.e.,
Figure BDA0001623638800000154
) The dioxoisothiazolidine (i.e.,
Figure BDA0001623638800000155
) The amount of piperidine (i.e.,
Figure BDA0001623638800000156
) The compound of formula (i) morpholine (i.e.,
Figure BDA0001623638800000157
) An oxygen-substituted morpholine (for example,
Figure BDA0001623638800000158
) The amount of piperazine (i.e.,
Figure BDA0001623638800000159
) A compound of the formula (i.e.,
Figure BDA00016236388000001510
)1, 3-oxazinane (i.e.,
Figure BDA00016236388000001511
) The pyrrole (i.e.,
Figure BDA00016236388000001512
) The base of the compound is pyrazole (i.e.,
Figure BDA00016236388000001513
) An imidazole (i.e.,
Figure BDA00016236388000001514
) Triazoles (including but not limited to
Figure BDA00016236388000001515
) Tetrazoles (including but not limited to
Figure BDA00016236388000001516
) Dihydropyridines (e.g., 1, 2-dihydropyridines)
Figure BDA00016236388000001517
) Dihydropyrimidines (e.g., 1, 6-dihydropyrimidines)
Figure BDA00016236388000001518
) Dihydropyrazines (e.g., 1, 2-dihydropyrazines)
Figure BDA00016236388000001519
) Tetrahydropyridine, tetrahydropyrimidine, tetrahydropyrazine, or the like.
As described herein, "R" is d And R e 、R f And R g 、R h And R i 、R j And R k Each independently and taken together with the atoms to which they are attached form a carbonyl group, a carbocyclic ring of 3-6 atoms, or a heterocyclic ring of 3-6 atoms, refers to R d And R e May form together with the atoms to which they are attached a carbonyl group, a carbocyclic ring of 3 to 6 atoms or a heterocyclic ring of 3 to 6 atoms, R f And R g May form together with the atoms to which they are attached a carbonyl group, a carbocyclic ring of 3 to 6 atoms or a heterocyclic ring of 3 to 6 atoms, R h And R i May form together with the atoms to which they are attached a carbonyl group, a carbocyclic ring of 3 to 6 atoms or a heterocyclic ring of 3 to 6 atoms, R j And R k Can form a carbonyl group, a carbocyclic ring of 3 to 6 atoms or a heterocyclic ring of 3 to 6 atoms together with the atoms to which they are attached, and the carbocyclic and heterocyclic rings formed are independent of each other and do not affect each other.
The term "pharmaceutically acceptable" refers to molecular entities and compositions that are physiologically tolerable when administered to humans and do not typically produce an allergic or similar untoward reaction, such as gastrointestinal upset, dizziness and the like. Preferably, the term "pharmaceutically acceptable" as used herein refers to those approved by a federal regulatory agency or a state government or listed in the U.S. pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
The term "sGC stimulator" refers to a compound or agent that has a stimulating effect on soluble guanylate cyclase (sGC), and in some embodiments, "stimulators" and "agonists" may be used interchangeably.
The term "carrier" refers to a diluent, adjuvant, excipient, or matrix with which the compound is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water and aqueous solutions (e.g., saline solution, aqueous dextrose solution, aqueous glycerol solution) are preferably employed as carriers, particularly injectable solutions. Suitable Pharmaceutical carriers are described in e.w. martin, "Remington's Pharmaceutical Sciences".
The term "prodrug", as used herein, represents a compound that is converted in vivo to a compound of formula (I). Such conversion is effected by hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue. The prodrug compound of the invention can be ester, and in the prior invention, the ester can be used as the prodrug and comprises phenyl ester and aliphatic (C) 1-24 ) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, a compound of the present invention contains a hydroxy group, i.e., it can be acylated to provide the compound in prodrug form. Other prodrug forms include phosphate esters, such as those obtained by phosphorylation of a hydroxyl group on the parent. For a complete discussion of prodrugs, reference may be made to the following: T.Higuchi and V.Stella, Pro-drugs as Novel Delivery Systems, Vol.14 of the A.C.S.Symphosis Series, Edward B.Roche, ed., Bioreversible Carriers in Drug designs, American Pharmaceutical Association and Pergamon Press,1987, J.Rautio et al, Prodrugs in Design and Clinical Applications, Nature Review Drug Discovery,2008,7,255-stry,2008,51,2328-2345。
"metabolite" refers to the product of a particular compound or salt thereof obtained by metabolism in vivo. Metabolites of a compound can be identified by techniques well known in the art, and its activity can be characterized by assay methods as described herein. Such products may be obtained by administering the compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage, and the like. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting a compound of the present invention with a mammal for a sufficient period of time.
As used herein, "pharmaceutically acceptable salts" refers to both organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable salts are well known in the art, as are: berge et al, description of the scientific acceptable salts in detail in J. pharmaceutical Sciences,1977,66:1-19. Pharmaceutically acceptable non-toxic acid forming salts include, but are not limited to, inorganic acid salts such as hydrochloride, hydrobromide, phosphate, sulfate, perchlorate; organic acid salts such as acetate, oxalate, maleate, tartrate, citrate, succinate, malonate; or by other methods described in the literature, such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, cyclopentylpropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, malates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, palmitates, pamoates, pectinates, persulfates, 3-phenylpropionates, picrates, pivalatesSalts, propionates, stearates, thiocyanates, p-toluenesulfonates, undecanoates, pentanoates, and the like. Salts obtained by reaction with a suitable base include alkali metals, alkaline earth metals, ammonium and N + (C 1-4 Alkyl radical) 4 A salt. The present invention also contemplates quaternary ammonium salts formed from any compound containing a group of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. Alkali or alkaline earth metals that can form salts include sodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C 1-8 Sulfonates and aromatic sulfonates.
"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that is water.
"ester" in the context of the present invention refers to an in vivo hydrolysable ester formed from a compound containing a hydroxy or carboxy group. Such esters are, for example, pharmaceutically acceptable esters which are hydrolysed in the human or animal body to yield the parent alcohol or acid. The compounds of formula (I) of the present invention contain a carboxyl group and may form in vivo hydrolysable esters with appropriate groups including, but not limited to, alkyl, arylalkyl and the like.
By "nitroxide" herein is meant that when a compound contains several amine functional groups, 1 or more than 1 nitrogen atom can be oxidized to form an N-oxide. Specific examples of N-oxides are N-oxides of tertiary amines or N-oxides of nitrogen-containing heterocyclic nitrogen atoms. The corresponding amines can be treated with an oxidizing agent such as hydrogen peroxide or a peracid (e.g., peroxycarboxylic acid) to form the N-oxide (see Advanced Organic Chemistry, Wiley Interscience, 4 th edition, Jerry March, pages). In particular, the N-oxide may be prepared by the method of L.W.Deady (Syn.Comm.1977,7,509-514) in which an amine compound is reacted with m-chloroperoxybenzoic acid (MCPBA), for example, in an inert solvent such as dichloromethane.
As used herein, the terms "compound of the present invention", "compound described in the present invention" or the like refer to a compound represented by formula (I) or formula (II) or formula (IIa) in the present invention.
The term "treating" or "treatment" as used herein refers, in some embodiments, to ameliorating a disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one clinical symptom thereof). In other embodiments, "treating" or "treatment" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" or "treatment" refers to modulating the disease or disorder, either physically (e.g., stabilizing a perceptible symptom) or physiologically (e.g., stabilizing a parameter of the body), or both. In other embodiments, "treating" or "treatment" refers to preventing or delaying the onset, occurrence, or worsening of a disease or disorder.
Any formulae given herein are also intended to represent the non-isotopically enriched forms as well as the isotopically enriched forms of these compounds. Isotopically enriched compounds have the structure depicted by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Exemplary isotopes that can be incorporated into 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。
in another aspect, the compounds of the invention include isotopically enriched compounds as defined herein, e.g. wherein a radioisotope, e.g. is present 3 H, 14 C and 18 those compounds of F, or in which a non-radioactive isotope is present, e.g. 2 H and 13 C. the isotopically enriched compounds can be used for metabolic studies (use) 14 C)、Reaction kinetics study (using, for example 2 H or 3 H) Detection or imaging techniques such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution determination, or may be used in radiotherapy of a patient. 18 F-enriched compounds are particularly desirable for PET or SPECT studies. Isotopically enriched compounds of the present invention can be prepared by conventional techniques known to those skilled in the art or by the procedures and examples of preparation of the present invention by using a suitable isotopically labelled reagent in place of the original used unlabelled reagent.
In addition, heavier isotopes are, in particular, deuterium (i.e., 2 substitution of H or D) may provide certain therapeutic advantages resulting from greater metabolic stability. For example, increased in vivo half-life or decreased dosage requirements or improved therapeutic index. It is to be understood that deuterium in the present invention is to be considered as a substituent of a compound of formula (I) or formula (II) or formula (IIa). The concentration of such heavier isotopes, particularly deuterium, can be defined by isotopic enrichment factors. The term "isotopic enrichment factor" as used herein refers to the ratio between the isotopic and natural abundance of a given isotope. If a substituent of a compound of the invention is designated as deuterium, the compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). Pharmaceutically acceptable solvates of the invention include those in which the crystallization solvent may be isotopically substituted, e.g. D 2 O, acetone-d 6 、DMSO-d 6 Those solvates of (a).
Unless otherwise indicated, all tautomeric forms of the compounds of the invention are included within the scope of the invention. In addition, unless otherwise indicated, the structural formulae of the compounds described herein include isotopically enriched concentrations of one or more different atoms.
Abbreviations for any protecting groups, amino acids and other compounds used in the present invention are based on their commonly used, accepted abbreviations unless otherwise indicated, or refer to IUPAC-IUBCommission on Biochemical Nomenclature (see biochem.1972, 11: 942-944).
Description of the Compounds of the invention
The present invention provides a novel pyrazolopyridine compound useful as a stimulator of sGC and a pharmaceutical composition thereof, and the use of the compound or the pharmaceutical composition for the preparation of a medicament for the treatment and/or prevention of sGC mediated diseases such as heart failure, sclerosis, systemic sclerosis, sickle cell anemia, cardiac achalasia, pulmonary fibrosis, idiopathic pulmonary fibrosis, cystic fibrosis, pulmonary hypertension and the like.
In one aspect, the invention relates to a compound that is a compound of formula (I) or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, ester, pharmaceutically acceptable salt, or prodrug of a compound of formula (I),
Figure BDA0001623638800000181
wherein, L, C1, R 0 、R 5 、R 6 、R 7 、E 1 、E 2 、E 3 、E 4 And n have the meaning described in the present invention;
wherein the compounds of the present invention do not include the following compounds:
1- (4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidin-5-yl) cyclopropanecarboxylic acid ethyl ester, 1- (4-chloro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b]Pyridin-3-yl) pyrimidin-5-yl) cyclopropanecarboxylic acid ethyl ester, 2- (4-chloro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b]Pyridin-3-yl) pyrimidin-5-yl) acetic acid ethyl ester, N 4 - (2, 4-dimethoxybenzyl) -2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazoleAnd [3,4-b ]]Pyridin-3-yl) pyrimidine-4, 5-diamine, 2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b]Pyridin-3-yl) pyrimidine-4, 5, 6-triamine, 2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b]Pyridin-3-yl) pyrimidine-4, 5-diamine, (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidin-5-yl) carbamic acid methyl ester, (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidin-5-yl) (methyl) carbamic acid methyl ester, (4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidin-5-yl) carbamic acid methyl ester, (4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidin-5-yl) (methyl) carbamic acid methyl ester and (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidin-5-yl) (2,2, 2-trifluoroethyl) carbamic acid methyl ester.
In some embodiments, E 1 Is N or CR 1 ;E 2 Is N or CR 2 ;E 3 Is N or CR 3 ;E 4 Is N or CR 4
In some embodiments, L is- (CR) a R b ) t -、-(CR a R b ) f -O-、-(CR a R b ) f -S-、-(CR a R b ) f -S(=O)-、-(CR a R b ) f -S(=O) 2 -、-(CR a R b ) f -N(R c )-、-(CR a R b ) f -C(=O)N(R c )-、-C(=O)N(R c )-(CR a R b ) f -or- (CR) a R b ) f -C(=O)-;
t is 1,2, 3 or 4;
f is 0, 1,2, 3 or 4.
In some embodiments, each R is a And R b Independently H, D, F, Cl, Br, I, CN, amino, hydroxyl, sulfydryl and C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Alkylamino, halogeno C 1-6 Alkoxy, acyl, sulfonyl, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group; or, R a And R b Together form a carbonyl group; or, R a 、R b And together with the C atom to which they are attached form a carbocyclic ring of 3 to 8 atoms or a heterocyclic ring of 3 to 8 atoms.
In some embodiments, R c Is H, D, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl, acyl, sulfonyl, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group.
In some embodiments, each R is a And R b Independently H, D, F, Cl, Br, I, CN, amino, hydroxyl, mercapto, methyl, ethyl, propyl, butyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy radical, C 1-4 Alkylamino, halogeno C 1-4 Alkoxy radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclyl, phenyl or C 1-5 A heteroaryl group; or, R a And R b Together form a carbonyl group; or, R a 、R b And together with the C atom to which they are attached form a carbocyclic ring of 3 to 6 atoms or a heterocyclic ring of 3 to 6 atoms.
In some embodiments, R c H, D, methyl, ethyl, propyl, butyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclyl, phenyl or C 1-5 A heteroaryl group.
In some embodiments, L is-CH 2 -、-CH(CH 3 )-、-(CH 2 ) 2 -、-(CH 2 ) 3 -、
Figure BDA0001623638800000191
Figure BDA0001623638800000192
-O-、-S-、-NH-、
Figure BDA0001623638800000193
In some embodiments, ring C1 is aryl, heteroaryl, carbocyclyl, or heterocyclyl.
In some embodiments, ring C1 is C 6-10 Aryl radical, C 1-9 Heteroaryl group, C 3-10 Carbocyclic radical or C 2-9 A heterocyclic group.
In some embodiments, ring C1 is C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Carbocyclic radical or C 2-5 A heterocyclic group.
In some embodiments, ring C1 is of the subformula:
Figure BDA0001623638800000194
each X 1 、X 2 、X 3 、X 4 And X 5 Independently is N or CH;
each Y is 1 、Y 2 、Y 3 And Y 4 Independently is CH 2 、C(=O)、NH、S、S(=O)、S(=O) 2 Or O;
d is 0, 1,2, 3 or 4.
In some embodiments, ring C1 is of the subformula:
Figure BDA0001623638800000195
wherein each X 1 、X 2 、X 3 And X 5 Independently is N or CH;
Y 1a is CH 2 NH, S or O;
Y 2 is CH 2 、C(=O)、NH、S、S(=O)、S(=O) 2 Or O;
d is 0, 1,2, 3 or 4.
In some embodiments, ring C1 is
Figure BDA0001623638800000201
Figure BDA0001623638800000202
In some embodiments, the compound of the present invention, which is a compound of formula (IIa) or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, ester, pharmaceutically acceptable salt, or prodrug of a compound of formula (IIa),
Figure BDA0001623638800000203
wherein R is 0 、R 3 、R 4 、R 5 、R 6 、R 7 、X 1 、X 2 、X 3 、X 4 、X 5 L and n all have the meanings described in the present invention.
In some embodiments, each R is 0 Independently H, D, F, Cl, Br, I, CN, NO 2 Amino, hydroxy, mercapto, oxo, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino, acyl, sulfonyl, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl or C 1-5 Heteroaryl C 1-6 An alkyl group; wherein, the amino, the hydroxyl, the mercapto and the C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino, acyl, sulfonyl, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl and C 1-5 Heteroaryl C 1-6 Alkyl is unsubstituted or independently optionally substituted by 1,2, 3 or 4 groups selected from D, F, Cl, Br, I, CN, NO 2 Amino, hydroxy, mercapto, oxo, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl and C 1-5 Heteroaryl group is substituted.
In some embodiments, each R is 0 Independently H, D, F, Cl, Br, I, CN, NO 2 Amino, hydroxy, mercapto, oxo, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy, halo C 1-4 Alkoxy radical, C 1-4 Alkylamino radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group;
wherein, the C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy, halo C 1-4 Alkoxy radical, C 1-4 Alkylamino radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl and C 1-5 Heteroaryl is unsubstituted or independently optionally substituted by 1,2, 3 or 4 substituents selected from D, F, Cl, Br, I, CN, NO 2 Amino, hydroxy, mercapto, oxo, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy, halo C 1-4 Alkoxy radical, C 1-4 Alkylamino radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl and C 1-5 Heteroaryl group is substituted.
In some embodiments, each R is 0 Independently H, D, F, Cl, Br, I, CN, NO 2 Amino, hydroxy, mercapto, oxo, methyl, ethylA group selected from the group consisting of phenyl, pyridyl, pyrimidinyl, and trifluoromethyl, including trifluoromethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, piperidinyl, morpholinyl, piperazinyl, and cyclopropyl;
wherein said methyl, ethyl, propyl, butyl, difluoromethyl, methoxy, ethoxy, propoxy, tert-butoxy, methylamino, dimethylamino, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, piperidinyl, morpholinyl, piperazinyl, epoxypropyl, azetidinyl, phenyl, pyridinyl and pyrimidinyl are unsubstituted or independently optionally substituted with 1,2, 3 or 4 substituents selected from D, F, Cl, Br, I, CN, NO 2 Amino, hydroxy, mercapto, oxo, methyl, ethyl, propyl, butyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, trifluoromethyl, methoxy, ethoxy, propoxy, trifluoromethoxy, C 1-3 Alkylamino, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, C 2-5 Heterocyclyl, phenyl and C 1-5 Heteroaryl group is substituted.
In some embodiments, the compound of the present invention, which is a compound of formula (II) or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, ester, pharmaceutically acceptable salt, or prodrug of a compound of formula (II),
Figure BDA0001623638800000211
wherein R is a 、R b 、R 3 、R 4 、R 5 、R 6 、R 7 All have the meaning described herein.
In some embodiments, each R is 6 And R 7 Independently H, D, F, Cl, Br, I, CN, NO 2 Amino, hydroxy, mercapto, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl, haloC 1-6 Alkoxy radical, C 1-6 Alkylamino radical, hydroxy radical C 1-6 Alkyl, cyano C 1-6 Alkyl, amino C 1-6 Alkyl, hydroxy C 1-6 Alkoxy, amino C 1-6 Alkoxy, acyl, sulfonyl or C 1-6 An alkoxy group.
In some embodiments, each R is 1 、R 2 、R 3 And R 4 Independently H, D, F, Cl, Br, I, CN, NO 2 Mercapto group, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 3-10 Cycloalkyl radical, C 2-10 Heterocyclic group, C 6-10 Aryl radical, C 1-9 Heteroaryl group, C 3-10 Cycloalkyl radical C 1-6 Alkyl radical, C 2-10 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl C 1-6 Alkyl, -N ═ NR 8 、-N=CR d R e 、-(CR d R e ) j CN、-(CR d R e ) j NR 8a R 8b 、-(CR d R e ) j C(=O)(CR f R g ) k NR 8a R 8b 、-(CR d R e ) j C(=O)(CR f R g ) k OR 9 、-(CR d R e ) j OR 9 、-(CR d R e ) j S(=O) 2 (CR f R g ) k OR 9 、-(CR d R e ) j S(=O) 2 (CR f R g ) k NR 8a R 8b 、-(CR d R e ) j N(R 8 )(CR d R e ) p C(=O)(CR f R g ) k OR 9 、-(CR d R e ) j N(R 8 )C(=O)(CR f R g ) k NR 8a R 8b 、-(CR d R e ) j N(R 8 )C(=O)R 10 、-(CR d R e ) j N(R 8 )S(=O) 2 R 11 、-(CR d R e ) j S(=O) 2 R 11 、-(CR d R e ) j OS(=O) 2 R 11 、-(CR d R e ) j OC(=O)(CR f R g ) k OR 9 、-(CR d R e ) j OC(=O)R 10 Or- (CR) d R e ) j C(=O)R 10 (ii) a The R is 1 、R 2 、R 3 And R 4 Unsubstituted or optionally independently substituted by 1,2, 3 or 4R x Substitution;
wherein, R is d 、R e 、R f 、R g 、R 8 、R 8a 、R 8b 、R 9 、R 10 、R 11 、R x J, k and p have the meanings described in the present invention.
In some embodiments, R 5 D, F, Cl, Br, I, CN, mercapto, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl radical, C 1-5 Heteroaryl C 1-6 Alkyl, - (CR) h R i ) g CN、-(CR h R i ) g NR 8c R 8d 、-(CR h R i ) g C(=O)(CR j R k ) h NR 8c R 8d 、-(CR h R i ) g C(=O)(CR j R k ) h OR 9a 、-(CR h R i ) g OR 9a 、-(CR h R i ) g S(=O) 2 (CR j R k ) h OR 9a 、-(CR h R i ) g S(=O) 2 (CR j R k ) h NR 8c R 8d 、-(CR h R i ) g N(R 8e )(CR h R i ) i C(=O)(CR j R k ) h OR 9a 、-(CR h R i ) g N(R 8e )C(=O)R 10a 、-(CR h R i ) g N(R 8e )C(=O)(CR j R k ) h NR 8c R 8d 、-(CR h R i ) g N(R 8e )S(=O) 2 R 11a 、-(CR h R i ) g S(=O) 2 R 11a 、-(CR h R i ) g OS(=O) 2 R 11a 、-(CR h R i ) g OC(=O)(CR j R k ) h OR 9a 、-(CR h R i ) g OC(=O)R 10a Or- (CR) h R i ) g C(=O)R 10a (ii) a The R is 5 Unsubstituted or optionally substituted by 1,2, 3 or 4R y Substitution;
wherein, R is h 、R i 、R j 、R k 、R 8c 、R 8d 、R 8e 、R 9a 、R 10a 、R 11a 、R y G, h and i have the meanings given in the description of the invention.
In some embodiments, each R is 8 And R 8e Independently H, D, C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy radical C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl or C 1-5 Heteroaryl C 1-6 An alkyl group.
In some embodiments, each R is 8a 、R 8b 、R 8c And R 8d Independently H, D, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, carboxyl C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy radical C 1-6 Alkyl radical, C 1-6 Alkoxyacyl group, C 1-6 Alkyl acyl radical, C 1-6 Alkylamino acyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl acyl, C 2-5 Heterocyclyl acyl radical, C 6-10 Aryl acyl radical, C 1-5 Heteroaryl acyl, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl or C 1-5 Heteroaryl C 1-6 An alkyl group; or, R 8a 、R 8b And together with the N atom to which they are attached form a heterocyclic ring of 3 to 10 atoms or a heteroaromatic ring of 5 to 10 atoms; or, R 8c 、R 8d And together with the N atom to which they are attached form a heterocyclic ring of 3 to 10 atoms or a heteroaromatic ring of 5 to 10 atoms.
In some embodiments, each R is d 、R e 、R f 、R g 、R h 、R i 、R j And R k Independently H, D, F, Cl, Br, I, CN, amino, hydroxyl, sulfydryl and C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, carboxyl C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino, acyl, sulfonyl, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group; or, R d And R e 、R f And R g 、R h And R i 、R j And R k Each independently and together with the atoms to which they are attached form a carbonyl group, a carbocyclic ring of 3 to 6 atoms, or a heterocyclic ring of 3 to 6 atoms.
In some embodiments, each R is 11 And R 11a Independently H, D, amino, hydroxy, mercapto, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino radical, hydroxy radical C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, carboxyl C 1-6 Alkyl radical, C 3-6 Cycloalkyl, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group.
In some embodiments, each R is 10 And R 10a Independently H, D, amino, hydroxy, mercapto, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino radical, hydroxy radical C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, carboxyl C 1-6 Alkyl radical, C 3-6 Cycloalkyl, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group.
In some embodiments, each R is 9 And R 9a Independently H, D, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, halo C 1-6 Alkyl, acyl, sulfonyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, carboxyl C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical C 1-6 Alkyl or C 1-5 Heteroaryl C 1-6 An alkyl group.
In some embodiments, each R is x Independently oxo, H, D, F, Cl, Br, I, CN, amino, hydroxy, mercapto, carboxyl, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino, acyl, sulfonyl, C 1-6 Alkyl acyl radical, C 1-6 Alkylsulfonyl radical, C 1-6 Alkylamido radical, C 1-6 Alkylsulfonylamino group, C 1-6 Alkoxyamido group, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group.
In some embodiments, each R is y Independently oxo, H, D, F, Cl, Br, I, CN, amino, hydroxy, mercapto, carboxyl, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl, cyano C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 Alkoxy radical, C 1-6 Alkylamino, acyl, sulfonyl, C 1-6 Alkyl acyl, C 1-6 Alkylsulfonyl radical, C 1-6 Alkylamido radical, C 1-6 Alkylsulfonylamino group, C 1-6 Alkoxyamido group, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group.
In some embodiments, n is 0, 1,2, 3, or 4.
In some embodiments, each g, h, i, j, k, and p is independently 0, 1,2, 3, or 4.
In some embodiments, R 5 D, F, Cl, Br, I, CN, mercapto, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-4 Alkyl radical, C 2-5 Heterocyclyl radical C 1-4 Alkyl radical, C 6-10 Aryl radical C 1-4 Alkyl radical, C 1-5 Heteroaryl C 1-4 Alkyl, - (CR) h R i ) g CN、-NR 8c R 8d 、-C(=O)NR 8c R 8d 、-C(=O)OR 9a 、-OR 9a 、-S(=O) 2 OR 9a 、-S(=O) 2 NR 8c R 8d 、-N(R 8e )(CR h R i ) i C(=O)(CR j R k ) h OR 9a 、-N(R 8e )C(=O)R 10a 、-N(R 8e )C(=O)NR 8c R 8d 、-N(R 8e )S(=O) 2 R 11a 、-S(=O) 2 R 11a 、-OS(=O) 2 R 11a 、-OC(=O)OR 9a 、-OC(=O)R 10a or-C (═ O) R 10a (ii) a The R is 5 Unsubstituted or optionally substituted by 1,2, 3 or 4R y Substitution;
wherein, R is h 、R i 、R j 、R k 、R 8e 、R 8c 、R 8d 、R 9a 、R 10a 、R 11a 、R y G, i and h have the meanings given in the description of the invention.
In some embodiments, R 5 D, F, Cl, Br, I, CN, mercapto, methyl, ethyl, propyl, butyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, trifluoromethyl, difluoromethyl, 2-difluoroethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclobutyl, azetidinyl, pyrrolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, morpholinyl, oxomorpholinyl (including, but not limited to, 3-oxomorpholin-4-yl)
Figure BDA0001623638800000231
) Thiomorpholinyl, 4-dioxothiomorpholinyl, tetrahydrofuranyl, oxazolidinyl, thiazolidinyl, 1-dioxoisothiazolidinyl
Figure BDA0001623638800000241
Oxo-1, 3-oxazinyl
Figure BDA0001623638800000242
Phenyl, pyridyl, pyrimidinyl, pyrazolyl, imidazolyl, pyrrolyl, thiazolyl, oxazolyl, triazolyl, tetrazolyl, thienyl, furyl, - (CR) h R i ) g CN、-NR 8c R 8d 、-C(=O)NR 8c R 8d 、-C(=O)OR 9a 、-OR 9a 、-S(=O) 2 OR 9a 、-S(=O) 2 NR 8c R 8d 、-N(R 8e )C(=O)(CR j R k ) h OR 9a 、-N(R 8e )C(=O)R 10a 、-N(R 8e )C(=O)NR 8c R 8d 、-N(R 8e )S(=O) 2 R 11a 、-S(=O) 2 R 11a or-C (═ O) R 10a (ii) a The R is 5 Unsubstituted or optionally substituted by 1,2, 3 or 4R y Substitution;
wherein, R is h 、R i 、R j 、R k 、R 8e 、R 8c 、R 8d 、R 9a 、R 10a 、R 11a 、R y G and h have the meanings given in the description of the invention.
In some embodiments, each R is 6 And R 7 Independently H, D, F, Cl, Br, I, CN, NO 2 Amino, hydroxy, mercapto, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl, halo C 1-4 Alkoxy radical, C 1-4 Alkylamino radical, hydroxy radical C 1-4 Alkyl, cyano C 1-4 Alkyl, amino C 1-4 Alkyl, hydroxy C 1-4 Alkoxy, amino C 1-4 Alkoxy or C 1-4 An alkoxy group.
In some embodiments, each R is 6 And R 7 Independently H, D, F, Cl, Br, I, CN, NO 2 Amino, hydroxy, mercapto, methyl, ethyl, propyl, butyl, trifluoromethyl, trifluoromethoxy, methylamino, dimethylamino, hydroxymethyl, hydroxyethyl, cyanomethyl, cyanoethyl, aminomethyl, aminoethyl, hydroxymethoxy, hydroxyethoxy, amino C 1-3 Alkoxy, methoxy, ethoxy, propoxy or butoxy.
In some embodiments, each R is 1 、R 2 、R 3 And R 4 Independently H, D, F, Cl, Br, I, CN, NO 2 Mercapto group, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-4 Alkyl radical, C 2-5 Heterocyclyl radical C 1-4 Alkyl radical, C 6-10 Aryl radical C 1-4 Alkyl radical, C 1-5 Heteroaryl C 1-4 Alkyl, - (CR) d R e ) j CN、-NR 8a R 8b 、-C(=O)NR 8a R 8b 、-C(=O)OR 9 、-OR 9 、-S(=O) 2 OR 9 、-S(=O) 2 NR 8a R 8b 、-N(R 8 )(CR d R e ) p C(=O)(CR f R g ) k OR 9 、-N(R 8 )C(=O)NR 8a R 8b 、-N(R 8 )C(=O)R 10 、-N(R 8 )S(=O) 2 R 11 、-S(=O) 2 R 11 、-OS(=O) 2 R 11 、-OC(=O)OR 9 、-OC(=O)R 10 or-C (═ O) R 10 (ii) a The R is 1 、R 2 、R 3 And R 4 Unsubstituted or optionally independently substituted by 1,2, 3 or 4R x Substitution;
wherein, R is d 、R e 、R f 、R g 、R 8 、R 8a 、R 8b 、R 9 、R 10 、R 11 、R x J, k and p have the meanings described in the present invention.
In some embodiments, each R is 1 、R 2 、R 3 And R 4 Independently H, D, F, Cl, Br, I, CN, NO 2 Mercapto, methyl, ethyl, propyl, butyl, trifluoromethyl, 2-difluoroethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclobutyl, azetidinyl, pyrrolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, morpholinyl, oxomorpholinyl, thiomorpholinyl, 4-dioxothiomorpholinyl, tetrahydrofuranyl, oxazolidinyl, thiazolidinyl, 1-dioxoisothiazolidinyl (i.e., a salt thereof with a thiol group in the molecule of formula I)
Figure BDA0001623638800000243
) Oxo-1, 3-oxazinyl (i.e. phenyl)
Figure BDA0001623638800000244
)、
Figure BDA0001623638800000245
Phenyl, pyridyl, pyrimidinyl, pyrazolyl, imidazolyl, pyrrolyl, thiazolyl, oxazolyl, triazolyl, tetrazolyl, thienyl, furyl, -NR 8a R 8b 、-C(=O)NR 8a R 8b 、-C(=O)OR 9 、-OR 9 、-S(=O) 2 OR 9 、-S(=O) 2 NR 8a R 8b 、-N(R 8 )(CR d R e ) p C(=O)(CR f R g ) k OR 9 、-N(R 8 )C(=O)NR 8a R 8b 、-N(R 8 )C(=O)R 10 、-N(R 8 )S(=O) 2 R 11 、-S(=O) 2 R 11 or-C (═ O) R 10 (ii) a The R is 1 、R 2 、R 3 And R 4 Unsubstituted or optionally independently substituted by 1,2, 3 or 4R x Substitution;
wherein, R is d 、R e 、R f 、R g 、R 8 、R 8a 、R 8b 、R 9 、R 10 、R 11 、R x K and p have the meanings described in the present invention.
In some embodiments, each R is 8 And R 8e Independently H, D, C 1-4 Alkyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy radical C 1-4 Alkyl radical, C 3-4 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl, C 3-6 Cycloalkyl radical C 1-6 Alkyl radical, C 2-5 Heterocyclyl radical C 1-4 Alkyl radical, C 6-10 Aryl radical C 1-4 Alkyl or C 1-5 Heteroaryl C 1-4 An alkyl group.
In some embodiments, each R is 8 And R 8e Independently H, D, methyl, ethyl, propyl, butyl, hydroxymethyl, hydroxyethyl, hydroxypropyl, aminomethyl, cyanomethyl, cyanoethyl, difluoromethyl, 2-difluoroethyl, methoxymethyl, methoxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, tetrahydrofuranyl, piperidinyl, morpholinyl, tetrahydropyranyl, phenyl, pyridinyl, pyrimidinyl, cyclopropylmethyl, cyclobutylmethyl, morpholinylmethyl, piperidinylmethyl, tetrahydropyranyl ethyl, phenylmethyl, phenylethyl, pyridylmethyl or pyrimidinylmethyl.
In some embodiments, each R is 8a 、R 8b 、R 8c And R 8d Independently H, D, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, carboxyl C 1-4 Alkyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy radical C 1-4 Alkyl radical, C 1-4 Alkoxyacyl group, C 1-4 Alkyl acyl radical, C 1-4 Alkylamino acyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl acyl, C 2-5 Heterocyclyl acyl radical, C 6-10 Aryl acyl radical, C 1-5 Heteroaryl acyl, C 3-6 Cycloalkyl radical C 1-4 Alkyl radical, C 2-5 Heterocyclyl radical C 1-4 Alkyl radical, C 6-10 Aryl radical C 1-4 Alkyl or C 1-5 Heteroaryl C 1-4 An alkyl group; or, R 8a 、R 8b And together with the N atom to which they are attached form a heterocyclic ring of 3 to 6 atoms or a heteroaromatic ring of 5 to 6 atoms; or, R 8c 、R 8d And together with the N atom to which they are attached form a heterocyclic ring of 3 to 6 atoms or a heteroaromatic ring of 5 to 6 atoms.
In some embodiments, each R is 8a 、R 8b 、R 8c And R 8d Independently H, D, methyl, ethyl, propyl, butyl, hydroxymethyl, hydroxyethyl (including 2-hydroxyethyl and 1-hydroxyethyl), hydroxypropyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, trifluoromethoxy, chloroethyl (including 2-chloroethyl and 1-chloroethyl), 2,2, 2-trifluoroethyl, 2, 2-difluoroethyl, 2-chloro-1-methylethyl, methoxymethyl, methoxyethyl, methoxyacyl, ethoxyacyl, propoxoyl, methylacyl, ethylacoyl, methylaminoacyl, ethylaminoacyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholinyl, phenyl, pyridinyl, pyrimidinyl, cyclopropylacyl, cyclopentylacyl, cyclohexylacyl, cyclopropyloyl, tetrahydrofuranoyl, tetrahydropyranoyl, piperidinoylacyl, piperazinoylmorpholinoylacyl, tetrahydrothienylacyl, pyrrolidinylacyl, phenylacyl, pyridinoacyl, piperazinoylacyl, morpholinoylacyl, tetrahydrothienylacyl, pyrrolidinylacyl, phenylacyloyl, pyridinoacyl, and mixtures thereof, Pyrimidinylacyl, pyrrolylacyl, imidazolylacyl, pyrazolylacyl, thienylacyl, furylacyl, cyclopropylmethyl, cyclobutylmethyl, tetrahydropyranylmethyl, tetrahydropyranylethyl, tetrahydrofuranylmethyl, tetrahydrofuranylethyl, piperidinylmethyl, piperidinylethyl, morpholinylmethyl, phenylmethyl, phenylethyl, pyridylmethyl, pyridylethyl or C 2-5 Heterocyclyl radical C 1-3 An alkyl group; alternatively, the first and second electrodes may be,
R 8a 、R 8b and the N atom to which they are attached form an azetidine, pyrrolidine, oxazolidine, thiazolidine, isothiazolidine, oxoisothiazolidine (e.g., dioxoisothiazolidine), piperidine, morpholine, piperazine, thiomorpholine, 1, 3-oxazinane, pyrrole, pyrazole, imidazole, or triazolyl group; alternatively, the first and second electrodes may be,
R 8c 、R 8d and the N atom to which they are attached form an azetidine, pyrrolidine, oxazolidine, thiazolidine, isothiazolidine, oxoisothiazolidine (e.g., dioxoisothiazolidine), piperidine, morpholine, piperazine, thiomorpholine, 1, 3-oxazinane, pyrrole, pyrazole, imidazole, or triazole group.
In some embodimentsIn the scheme, each R d 、R e 、R f 、R g 、R h 、R i 、R j And R k Independently H, D, F, Cl, Br, I, CN, amino, hydroxyl, sulfydryl and C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, carboxyl C 1-4 Alkyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy, halo C 1-4 Alkoxy radical, C 1-4 Alkylamino, acyl, sulfonyl, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group; or, R d And R e 、R f And R g 、R h And R i 、R j And R k Each independently and together with the atoms to which they are attached form a carbonyl group, a carbocyclic ring of 3-6 atoms, or a heterocyclic ring of 3-6 atoms.
In some embodiments, each R is d 、R e 、R f 、R g 、R h 、R i 、R j And R k Independently H, D, F, Cl, Br, I, CN, amino, hydroxy, mercapto, methyl, ethyl, hydroxymethyl, hydroxyethyl, aminomethyl, aminoethyl, cyanomethyl, cyanoethyl, trifluoromethyl, difluoromethyl, monofluoromethyl, chloroethyl, methoxy, ethoxy, trifluoromethoxy, or difluoromethoxy; or, R d And R e 、R f And R g 、R h And R i 、R j And R k Each independently and together with the atoms to which they are attached form a carbonyl, cyclopropane, cyclobutane, cyclopentane, cyclohexane, propylene oxide, aziridine, butylene oxide, tetrahydrofuran, tetrahydropyran, piperidine, morpholine, or piperazine.
In some embodiments, each R is 11 And R 11a Independently H, D, amino, hydroxy, mercapto, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy, halo C 1-4 Alkoxy radical, C 1-4 Alkylamino radical, hydroxy radical C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, carboxyl C 1-4 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group.
In some embodiments, each R is 11 And R 11a Independently H, D, amino, hydroxy, mercapto, methyl, ethyl, propyl, butyl or trifluoromethyl.
In some embodiments, each R is 10 And R 10a Independently H, D, amino, hydroxy, mercapto, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy, halo C 1-4 Alkoxy radical, C 1-4 Alkylamino radical, hydroxy radical C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, carboxyl C 1-4 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group.
In some embodiments, each R is 10 And R 10a Independently H, D, amino, hydroxy, mercapto, methyl, ethyl, propyl, butyl, trifluoromethyl, chloroethyl, 2,2, 2-trifluoroethyl, 2, 2-difluoroethyl, 2-chloro-1-methylethyl, methoxy, ethoxy, propoxy, trifluoromethoxy, methylamino, dimethylamino, hydroxymethyl, hydroxyethyl, hydroxypropyl, aminomethyl, aminoethyl, cyanomethyl, cyanoethyl, carboxymethyl, carboxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydropyranyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, morpholinyl, piperidinyl, piperazinyl, phenyl, pyrrolyl, thienyl, furyl, pyridinyl or pyrimidinyl.
In some embodiments, each R is 9 And R 9a Independently H, D, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, halo C 1-4 Alkyl, acyl, sulfonyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, aryl, heteroaryl, and heteroaryl,Carboxy group C 1-4 Alkyl radical, C 3-6 Cycloalkyl radical, C 2-6 Heterocyclic group, C 6-10 Aryl radical, C 1-5 Heteroaryl group, C 3-6 Cycloalkyl radical C 1-3 Alkyl radical, C 2-5 Heterocyclyl radical C 1-3 Alkyl radical, C 6-10 Aryl radical C 1-3 Alkyl or C 1-5 Heteroaryl C 1-3 An alkyl group.
In some embodiments, each R is 9 And R 9a Independently H, D, methyl, ethyl, propyl, butyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, trifluoromethyl, chloroethyl, 2,2, 2-trifluoroethyl, 2, 2-difluoroethyl, 2-chloro-1-methylethyl, hydroxymethyl, hydroxyethyl, hydroxypropyl, aminomethyl, aminoethyl, cyanomethyl, cyanoethyl, carboxymethyl, carboxyethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, tetrahydrofuryl, morpholinyl, piperidinyl, piperazinyl, phenyl, pyridinyl, pyrimidinyl, cyclopropylmethyl, cyclobutylmethyl, phenylmethyl, phenylethyl, pyridylmethyl, pyridylethyl or C 2-5 Heterocyclyl radical C 1-3 An alkyl group.
In some embodiments, each R is x Independently oxo, H, D, F, Cl, Br, I, CN, amino, hydroxy, mercapto, carboxyl, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy, halo C 1-4 Alkoxy radical, C 1-4 Alkylamino, acyl, sulfonyl, C 1-4 Alkyl acyl radical, C 1-4 Alkylsulfonyl radical, C 1-4 Alkylamido radical, C 1-4 Alkylsulfonylamino group, C 1-4 Alkoxyamido group, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group.
In some embodiments, each R is x Independently oxo (═ O), H, D, F, Cl, Br, I, CN, amino, hydroxy, mercapto, carboxyl, methyl, ethyl, propyl, butyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, alkynyl,Hydroxymethyl, hydroxyethyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, trifluoromethyl, chloroethyl, 2,2, 2-trifluoroethyl, 2, 2-difluoroethyl, 2-chloro-1-methylethyl, methoxy, ethoxy, propoxy, trifluoromethoxy, methylamino, dimethylamino, C 1-3 Alkyl acyl radical, C 1-3 Alkylsulfonyl radical, C 1-3 Alkylamido radical, C 1-3 Alkylsulfonylamino, methoxycarbonylamino, C 2-3 Alkoxyamido, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, C 2-5 Heterocyclyl, phenyl or C 1-5 A heteroaryl group.
In some embodiments, each R is y Independently oxo, H, D, F, Cl, Br, I, CN, amino, hydroxy, mercapto, carboxyl, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, hydroxy C 1-4 Alkyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, halo C 1-4 Alkyl radical, C 1-4 Alkoxy, halo C 1-4 Alkoxy radical, C 1-4 Alkylamino, acyl, sulfonyl, C 1-4 Alkyl acyl radical, C 1-4 Alkylsulfonyl radical, C 1-4 Alkylamido radical, C 1-4 Alkylsulfonylamino group, C 1-4 Alkoxyamido group, C 3-6 Cycloalkyl radical, C 2-5 Heterocyclic group, C 6-10 Aryl or C 1-5 A heteroaryl group.
In some embodiments, each R is y Independently oxo, H, D, F, Cl, Br, I, CN, amino, hydroxy, mercapto, carboxyl, methyl, ethyl, propyl, butyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, hydroxymethyl, hydroxyethyl, amino C 1-4 Alkyl, cyano C 1-4 Alkyl, trifluoromethyl, chloroethyl, 2,2, 2-trifluoroethyl, 2, 2-difluoroethyl, 2-chloro-1-methylethyl, methoxy, ethoxy, propoxy, trifluoromethoxy, methylamino, dimethylamino, C 1-3 Alkyl acyl radical, C 1-3 Alkylsulfonyl radical, C 1-3 Alkylamido radical, C 1-3 Alkylsulfonylamino, methoxycarbonylamino, C 2-3 Alkoxyamido, cyclopropyl, cyclobutyl, cyclopentylCyclohexyl radical, C 2-5 Heterocyclyl, phenyl or C 1-5 A heteroaryl group.
In some embodiments, a compound of the present invention comprises a structure of one of the following or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, ester, pharmaceutically acceptable salt, or prodrug of the structure:
Figure BDA0001623638800000271
Figure BDA0001623638800000281
Figure BDA0001623638800000291
Figure BDA0001623638800000301
Figure BDA0001623638800000311
in another aspect, the invention relates to a pharmaceutical composition comprising a compound of any of the invention.
In some embodiments, the pharmaceutical composition of the present invention further comprises at least one of a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, and vehicle.
In one aspect, the present invention relates to the use of a compound according to the present invention or a pharmaceutical composition according to the present invention for the preparation of a medicament for the treatment and/or prevention of: heart failure, angina pectoris, hypertension, pulmonary hypertension, ischemia, vascular disease, renal disease, thromboembolic disease, male sexual dysfunction, systemic sclerosis, sickle cell anemia, cardiac achalasia, fibrotic disease and/or arteriosclerosis, etc.
In another aspect, the invention relates to the use of a compound according to the invention or a pharmaceutical composition according to the invention for the preparation of a medicament for use as a soluble guanylate cyclase stimulator.
The invention encompasses the use of the compounds of the invention and pharmaceutically acceptable salts thereof for the manufacture of a pharmaceutical product for the treatment of sGC mediated diseases in a patient, including those described herein. The present invention encompasses pharmaceutical compositions comprising therapeutically effective amounts of a compound represented by formula (I) or formula (II) or formula (IIa) in combination with at least one pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle.
The invention also encompasses methods of treating or ameliorating an sGC-mediated disease, or a condition responsive thereto, in a subject, comprising treating the subject with a therapeutically effective amount of a compound represented by formula (I) or formula (II) or formula (IIa).
Unless otherwise indicated, all hydrates, solvates and pharmaceutically acceptable salts of the compounds of the invention are within the scope of the invention.
In particular, the salts are pharmaceutically acceptable salts. The term "pharmaceutically acceptable" includes substances or compositions which must be compatible with chemical or toxicological considerations, in connection with the other ingredients comprising the formulation and the mammal being treated.
Salts of the compounds of the invention also include, but are not necessarily pharmaceutically acceptable salts of, intermediates used in the preparation or purification of compounds of formula (I) or formula (II) or formula (IIa) or isolated enantiomers of compounds of formula (I) or formula (II) or formula (IIa).
Salts of the compounds of the present invention may be prepared by any suitable method provided in the literature, for example, using inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric, and phosphoric acids, and the like. Or using organic acids such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid and salicylic acid; pyranonic acids, such as glucuronic acid and galacturonic acid; 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, ethanesulfonic acid, and the like.
The biological activity of the compounds of the present invention can be assessed by using any conventionally known method. Suitable detection methods are well known in the art. For example, the sGC activating activity, pharmacokinetic activity and/or liver microsome stability of the compound of the present invention and the like can be measured by an appropriate conventional method. The detection methods provided by the present invention are presented by way of example only and are not limiting of the invention. The compounds of the invention are active in at least one of the detection methods provided herein. For example, the compound has an activating effect on a recombinant guanylate cyclase reporter cell line, for example, the compound can effectively activate the activity of a CHO-K1-Rat sGC monoclonal cell strain, namely, the compound has better sGC activating activity. In another example, the compound of the invention has better in vivo pharmacokinetic properties, better absorption and exposure and higher bioavailability.
Pharmaceutical compositions, formulations, administration and uses of the compounds of the invention
According to another aspect, a pharmaceutical composition of the invention is characterized by comprising a pyrazolopyridine compound of formula (I) or formula (II) or formula (IIa), a compound of the invention as set forth, or a compound of examples 1-27, and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The amount of compound in the composition of the invention is effective to treat or alleviate sGC-associated diseases in a patient.
As described herein, the pharmaceutically acceptable compositions of the present invention further comprise a pharmaceutically acceptable carrier, adjuvant, or excipient, as used herein, including any solvent, diluent, or other liquid excipient, dispersant or suspending agent, surfactant, isotonic agent, thickening agent, emulsifier, preservative, solid binder or lubricant, and the like, as appropriate for the particular target dosage form. As described in the following documents: 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. Annu 1999, Marcel Dekker, New York, taken together with The disclosure of this document, indicates that different carriers can be used In The preparation of pharmaceutically acceptable compositions and their well known methods of preparation. Except insofar as any conventional carrier vehicle is incompatible with the compounds of the invention, e.g., any adverse biological effect produced or interaction in a deleterious manner with any other component of a pharmaceutically acceptable composition, its use is contemplated by the present invention.
Substances which may serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-blocking polymers, lanolin, sugars, such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; gum powder; malt; gelatin; talc powder; adjuvants such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic salt; ringer's solution; ethanol, phosphate buffered solutions, and other non-toxic suitable lubricants such as sodium lauryl sulfate and magnesium stearate, coloring agents, releasing agents, coating materials, sweetening, flavoring and perfuming agents, preservatives and antioxidants.
The pharmaceutical compositions of the present invention may be administered directly or in the form of pharmaceutical compositions or medicaments, together with suitable carriers or excipients, as is well known in the art. The methods of treatment of the present invention may comprise administering to a subject in need thereof an effective compound of the present invention. In some embodiments, the subject is a mammalian subject, and in some preferred embodiments, the subject is a human subject.
Effective amounts of the compounds, pharmaceutical compositions or medicaments of the present invention can be readily determined by routine experimentation, as can the most effective and convenient route of administration and the most appropriate formulation.
Pharmaceutical dosage forms of the compounds of the invention may be provided in the form of immediate release, controlled release, sustained release or targeted drug release systems. For example, common dosage forms include solutions and suspensions, (micro) emulsions, ointments, gels and patches, liposomes, tablets, dragees, soft or hard shell capsules, suppositories, ovules, implants, amorphous or crystalline powders, aerosols and freeze-dried preparations. Depending on the route of administration used, Special devices may be required to administer or administer the drug, such as syringes and needles, inhalers, pumps, injection pens, applicators or Special bottles (Special flash). Pharmaceutical dosage forms often consist of a drug, excipients, and a container/closure system. One or more excipients (also known as inactive ingredients) may be added to the compounds of the present invention to improve or facilitate the manufacture, stability, administration, and safety of the drug, and may provide a means to obtain the desired drug release profile. Thus, the type of excipient added to a drug may depend on various factors, such as the physical and chemical properties of the drug, the route of administration, and the preparation steps. Pharmaceutically acceptable excipients exist in the art and include those listed in various pharmacopoeias. (see U.S. Pharmacopoeia (USP), Japanese Pharmacopoeia (JP), European Pharmacopoeia (EP) and British Pharmacopoeia (BP)), the United states food and Drug Administration (the U.S. food and Drug Administration, www.fda.gov) Drug Evaluation and Research Center (Center for Drug Evaluation and Research, CEDR) publications, such as "Guide for Inactive ingredients" (Inactive Ingredient Guide,1996), the Handbook of Pharmaceutical Additives (Handbook of Pharmaceutical Additives,2002, Union Information Resources (synergy Information Resources, Inc., Endocument; c.).
Pharmaceutical dosage forms of the compounds of the invention may be manufactured by any of the methods well known in the art, for example, by means of conventional mixing, sieving, dissolving, melting, granulating, dragee-making, tabletting, suspending, extruding, spray-drying, grinding, emulsifying, (nano/micro) encapsulating, coating or lyophilizing processes. As noted above, the compositions of the present invention may include one or more physiologically acceptable inactive ingredients that facilitate processing of the active molecule into a formulation for pharmaceutical use.
Appropriate formulations will depend on the desired route of administration. For example, for intravenous injection, the compositions may be formulated in aqueous solution, with physiologically compatible buffers, if necessary, including, for example, phosphate, histidine or citrate for adjusting the pH of the formulation, and tonicity agents such as sodium chloride or dextrose. For transmucosal or nasal administration, a semi-solid, liquid formulation or patch, possibly containing a penetration enhancer, may be preferred; such penetrants are generally known in the art. For oral administration, the compounds may be formulated in liquid or solid dosage forms and as immediate release or controlled/sustained release formulations. Suitable dosage forms for oral ingestion by a subject include tablets, pills, dragees, hard and soft shell capsules, liquids, gels, syrups, slurries, suspensions and emulsions. The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
Solid oral dosage forms can be obtained using excipients which include fillers, disintegrants, binders (dry and wet), dissolution retardants, lubricants, glidants, anti-adherents, cationic exchange resins, wetting agents, antioxidants, preservatives, coloring and flavoring agents. These excipients may be of synthetic or natural origin. Examples of such excipients include cellulose derivatives, citric acid, dicalcium phosphate, gelatin, magnesium carbonate, magnesium/sodium lauryl sulfate, mannitol, polyethylene glycol, polyvinylpyrrolidone, silicates, silica, sodium benzoate, sorbitol, starch, stearic acid or salts thereof, sugars (i.e., dextrose, sucrose, lactose, etc.), talc, tragacanth mucilage (tragacanth mucilage), vegetable oils (hydrogenated), and waxes. Ethanol and water may be used as granulation aids. In some cases, it is desirable to coat the tablets with, for example, a taste-masking film, a gastric acid-resistant film, or a delayed-release film. Natural and synthetic polymers are often used to coat tablets in combination with colorants, sugars, and organic solvents or water to produce sugar coated pills. Where capsules are preferred over tablets, the pharmaceutical powders, suspensions or solutions may be delivered in the form of compatible hard or soft shell capsules.
In some embodiments, the compounds of the present invention may be administered topically, e.g. by means of a skin patch, a semi-solid or liquid formulation, such as a gel, (micro) emulsion, ointment, solution, (nano/micro) suspension or foam. Skin and underlying tissue penetration of the drug may be regulated by: for example, the use of penetration enhancers; using appropriate selection and combination of lipophilic, hydrophilic and amphiphilic excipients, including water, organic solvents, waxes, oils, synthetic and natural polymers, surfactants, emulsifiers; by adjusting the pH value; and the use of complexing agents. Other techniques, such as iontophoresis (ionophoresi), may also be used to modulate skin penetration of the compounds of the present invention. Transdermal or topical administration will be preferred, for example, in situations where topical administration with minimal systemic exposure is desired.
For administration by inhalation or nasal administration, the compounds for use according to the invention are conveniently administered from pressurized packs or nebulizers in the form of solutions, suspensions, emulsions or semi-solid aerosols, usually with the aid of propellants, such as halocarbons derived from methane and ethane, carbon dioxide or any other suitable gas. For topical aerosols, hydrocarbons such as butane, isobutene and pentane are suitable. In the case of a pressurized aerosol, the appropriate dosage unit can be determined by providing a valve to deliver a metered dose. Capsules and cartridges of, for example, gelatin may be formulated for use in an inhaler or insufflator. These generally comprise a powder mix of the compound with a suitable powder base such as lactose or starch.
Compositions formulated for parenteral administration by injection are generally sterile and may be presented in unit dosage form, for example, in ampoules, syringes, injection pens, or multi-dose containers, the latter typically containing a preservative. The compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as buffers, tonicity agents, viscosity enhancing agents, surfactants, suspending and dispersing agents, antioxidants, biocompatible polymers, chelating agents, and preservatives. Depending on the injection site, the carrier may contain water, synthetic or vegetable oils and/or organic co-solvents. In some cases, e.g., for lyophilized products or concentrates, the parenteral formulation will be reconstituted or diluted prior to administration. Depot formulations (depot formulations) that provide controlled or sustained release of the compounds of the invention may include injectable suspensions of nano/micron sized particles or nano/micron sized or non-micronized crystals. Other well known matrices in the art, polymers such as poly (lactic acid), poly (glycolic acid) or copolymers thereof, may be used as the controlled/sustained release matrix. Other depot type (depot) delivery systems may be provided in the form of implants and pumps requiring incisions.
Suitable carriers for the compounds of the invention for intravenous injection are well known in the art and include aqueous based solutions containing a base (e.g., sodium hydroxide) for forming the ionic compound; sucrose or sodium chloride as tonicity agent; such as a buffer containing phosphate or histidine. A co-solvent such as polyethylene glycol may be added. These water-based systems are effective in dissolving the compounds of the present invention and produce low toxicity after systemic administration. The proportions of the components of the solution system can be varied considerably without destroying the solubility and toxicity characteristics. In addition, the nature of the components may be varied. For example, low toxicity surfactants such as polysorbates or poloxamers (poloxamers) may be used, polyethylene glycols or other co-solvents may be used, biocompatible polymers such as polyvinylpyrrolidone may be added, and other sugars and polyols may be used in place of dextrose.
The compounds of the invention may act systemically and/or locally. They may be administered in a suitable manner, for example, by oral administration, parenteral administration, pulmonary administration, nasal administration, sublingual administration, lingual administration, buccal administration, rectal administration, dermal administration, transdermal administration, conjunctival administration, ear canal administration or administration as a graft or stent. The compounds of the invention are preferably administered orally or parenterally.
Suitable modes of administration for oral administration are as follows: modes of administration for the rapid release and/or release in an improved way of action according to the prior art of the compounds of the invention, which comprise the compounds of the invention in crystalline and/or amorphous and/or dissolved form, for example tablets (uncoated tablets or tablets coated, for example, with a gastric juice-resistant or dissolution-delaying or insoluble coating which controls the release of the compounds of the invention), tablets or films/wafers which disintegrate rapidly in the oral cavity, films/lyophilisates, capsules (for example hard or soft capsules), sugar-coated tablets, granules, pills, powders, emulsions, suspensions, aerosols or solutions.
Parenteral administration may bypass the absorption step (e.g., intravenous, intraarterial, cardiac, intraspinal or lumbar) or include absorption (e.g., intramuscular, subcutaneous, intradermal, transdermal or intraperitoneal). Administration forms suitable for parenteral administration include preparations for injection and infusion in the form of solutions, suspensions, emulsions, lyophilisates or sterile powders.
For other routes of administration, suitable examples are pharmaceutical forms for inhalation (including powder inhalers, nebulizers), nasal drops, solutions or sprays, tablets for lingual, sublingual or buccal administration, films/sheets or capsules, suppositories, otic or ophthalmic preparations, vaginal capsules, aqueous suspensions (lotions, concussors), lipophilic suspensions, ointments, creams, transdermal therapeutic systems (e.g. patches), emulsions (Milch), pastes, foams, spray powders, implants or stents.
A therapeutically effective amount of a compound of the present invention should be present in the above-described pharmaceutical formulations at a concentration of about 0.1 to 99.5%, preferably about 0.5 to 95% by weight of the total mixture.
The above pharmaceutical preparations may contain other pharmaceutically active ingredients in addition to the compound of the present invention.
The therapeutically effective dose can first be estimated using various methods well known in the art. The initial dose for animal studies can be based on the effective concentration established in cell culture assays. Dosage ranges suitable for human individuals can be determined, for example, using data obtained from animal studies and cell culture assays. In certain embodiments, the compounds of the present invention may be prepared as medicaments for oral administration. An exemplary dose of the compounds of the invention in a medicament for oral administration is from about 0.01 to about 100mg/kg (where kg denotes the body weight of the subject). In some embodiments, the agent comprises from about 0.01 to about 20mg/kg (where kg represents the weight of the subject), or optionally from about 0.01 to about 10mg/kg (where kg represents the weight of the subject), or optionally from about 0.01 to about 5.0mg/kg (where kg represents the weight of the subject). In certain embodiments, the compounds of the present invention are administered parenterally in an effective dose of about 0.001-1mg/kg, preferably about 0.01-0.5mg/kg (where kg represents the body weight of the subject).
The dosing regimen for a medicament typically for oral administration is three times a week, twice a week, once a week, three times a day, twice a day, or once a day. In certain embodiments, the compounds of the present invention are administered as the active ingredient in a total amount of about 0.001 to about 50, preferably 0.001 to 10mg/kg body weight per 24 hours, optionally in the form of multiple single doses, in order to achieve the desired result. A single dose may preferably contain a compound of the invention in an amount of from about 0.001 to about 30, especially from 0.001 to 3mg/kg body weight.
An effective or therapeutically effective amount or dose of an agent (e.g., a compound of the invention) refers to the amount of the agent or compound that causes improvement in the symptoms or prolongation of survival of the individual. Toxicity and therapeutic efficacy of the molecules can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining LD 50 (dose lethal to 50% of the population) and ED 50 (a dose therapeutically effective for 50% of the population). The dose ratio of toxic to therapeutic effects is the therapeutic index, which can be expressed as LD 50 /ED 50 . Agents that exhibit high therapeutic indices are preferred.
An effective or therapeutically effective amount is that amount of the compound or pharmaceutical composition that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician. The dosage is preferably selected to include an ED with minimal or no toxicity 50 In the circulating concentration range of (2). The dosage may vary within this range depending upon the dosage form employed and/or the route of administration utilized. The correct formulation, route of administration, dosage and time between administrations should be selected according to methods known in the art, taking into account the specificity of the individual condition.
The dose and interval may be adjusted individually to provide plasma levels of the active moiety sufficient to achieve the desired effect; i.e. the Minimum Effective Concentration (MEC). The MEC for each compound will vary, but can be estimated, for example, from in vitro (in vitro) data and animal experiments. The dosage necessary to obtain MEC will depend on the individual characteristics and route of administration. In the case of topical administration or selective uptake, the effective local concentration of the drug may not be related to the plasma concentration.
The amount of the agent or composition administered can depend on a variety of factors, including the sex, age and weight of the subject being treated, the severity of the affliction, the mode of administration and the judgment of the prescribing physician.
The compositions of the present invention may be provided in a pack or dispenser device containing one or more unit dosage forms containing the active ingredient, as desired. For example, the package or device may comprise a metal or plastic foil (such as a blister pack) or glass and rubber stopper. The packaging or dispensing device may be accompanied by instructions for administration. Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of a specified condition.
The compounds of the invention may be used alone or, if desired, in combination with other active compounds. The invention also provides a combination comprising at least one compound according to the invention and one or more further active substances, in particular drugs for the treatment and/or prophylaxis of the diseases mentioned in the invention.
The compounds of the invention are useful as stimulators of soluble guanylate cyclase and have the same or improved therapeutic properties, e.g. the same or improved in vivo properties, pharmacokinetic and pharmacodynamic behaviour and/or dose-activity relationship and/or safety properties, compared to the compounds known from the prior art. They are suitable for the treatment and/or prophylaxis of diseases in humans and animals.
The compounds of the invention can cause vasodilation, inhibit platelet aggregation and lower blood pressure, and increase coronary blood flow. These effects are mediated by direct stimulation of soluble guanylate cyclase and increased intracellular cGMP. In addition, the compound of the present invention enhances the effect of substances that increase cGMP levels, including EDRF (endothelial cell-derived relaxation factor), NO donor, protoporphyrin IX, arachidonic acid or phenylhydrazine derivative, and the like.
The compounds of the invention are suitable for the treatment and/or prophylaxis of cardiovascular, pulmonary, thromboembolic and fibrotic diseases.
The compounds of the invention can be used as medicaments for the treatment and/or prophylaxis of the following diseases: cardiovascular diseases such as hypertension, acute and chronic heart failure, coronary heart disease, stable and unstable angina, peripheral vascular and cardiovascular disease, arrhythmias, atrial and ventricular arrhythmias and impaired conduction (e.g., I-III degree atrioventricular conduction block (AB block I-III)), supraventricular tachyarrhythmias, atrial fibrillation, atrial flutter, ventricular fibrillation, ventricular flutter, ventricular tachyarrhythmias, torsade de pointes, atrial and ventricular premature beats, AV-binding premature beats, sick sinus syndrome, syncope, AV-nodal reentry tachycardia, woolp-pak syndrome; acute Coronary Syndrome (ACS), autoimmune heart disorders (pericarditis, endocarditis, valsolis, aortic inflammation, cardiomyopathy), shock such as cardiogenic shock, septic and anaphylactic shock, aneurysms, boxer cardiomyopathy (premature ventricular contractions (PVC)); thromboembolic and ischemic disorders, such as myocardial ischemia, myocardial infarction, stroke, cardiac hypertrophy, transient and ischemic attacks, preeclampsia, inflammatory cardiovascular diseases, coronary and peripheral arterial spasm, edema formation (such as that caused by pulmonary edema, cerebral edema, renal edema, or heart failure), peripheral circulatory disorders, reperfusion injury, arterial and venous thrombosis, microalbuminuria, myocardial insufficiency, endothelial dysfunction; prevention of restenosis, e.g., after thrombolytic therapy, Percutaneous Transluminal Angioplasty (PTA), transluminal coronary angioplasty (PTCA), heart transplantation and bypass surgery, as well as microvascular and macrovascular injury (vasculitis), increased fibrinogen and Low Density Lipoprotein (LDL) levels and increased concentration of plasminogen activator inhibitor 1 (PAI-1); erectile dysfunction and female sexual dysfunction.
Within the scope of the present invention, the term heart failure also includes more specific or related types of diseases, such as acute decompensated heart failure, right heart failure, left heart failure, global failure, ischemic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, idiopathic cardiomyopathy, congenital heart defects, heart valve defects, heart failure associated with heart valve defects, mitral stenosis, mitral insufficiency, aortic stenosis, aortic insufficiency, tricuspid stenosis, tricuspid insufficiency, pulmonary stenosis, pulmonary insufficiency, combined heart valve defects, myocardial inflammation (myocarditis), chronic myocarditis, acute myocarditis, viral myocarditis, diabetic heart failure, alcoholic toxic cardiomyopathy, heart storage diseases and diastolic and systolic heart failure.
In addition, the compounds of the present invention are also useful in the treatment and/or prevention of arteriosclerosis, impaired lipid metabolism, hypolipidaemia, dyslipidaemia, hypertriglyceridaemia, hyperlipidaemia, hypercholesterolaemia, betalipoproteinaemia, sitosterolemia, xanthoma, dangill's disease, obesity, adiposity, and in the treatment and/or prevention of combined hyperlipidaemia and metabolic syndrome.
The compounds of the invention are also useful for the treatment and/or prevention of primary and secondary raynaud's phenomenon, impairment of microcirculation function, claudication, peripheral and autonomic neuropathy, diabetic microangiopathy, diabetic retinopathy, diabetic ulcers of the extremities, gangrene, CREST syndrome, erythtoma (erythmitose), onychomycosis, rheumatism and for promoting wound healing.
The compounds of the invention are additionally suitable for the treatment of the following diseases: urological disorders, such as Benign Prostate Syndrome (BPS), Benign Prostatic Hyperplasia (BPH), Benign Prostatic Enlargement (BPE), Bladder Outlet Obstruction (BOO), lower urinary tract syndrome (LUTS, including feline urinary tract syndrome (FUS)); genitourinary disorders including overactive bladder (OAB) and (IC), incontinence (UI) (e.g. mixed, urge, stress or overflow urinary incontinence (MUI, UUI, SUI, OUI)), pelvic pain, benign and malignant disorders of the male and female genitourinary organs.
The compounds of the invention are furthermore suitable for the treatment and/or prophylaxis of renal diseases, in particular acute and chronic renal insufficiency and acute and chronic renal failure. Within the scope of the present invention, the term renal insufficiency includes its acute and chronic symptoms as well as essential or related renal diseases, such as: inadequate renal blood flow, dialysis hypotension, obstructive urinary tract disease, glomerulopathy, glomerulonephritis, acute glomerulonephritis, glomerulosclerosis, tubulointerstitial disease, nephropathy (such as primary and congenital kidney diseases), nephritis, immune kidney diseases (such as kidney graft rejection and immune complex-induced kidney diseases), toxicant-induced nephropathy, contrast-induced nephropathy, diabetic and non-diabetic nephropathy, pyelonephritis, renal cysts, nephrosclerosis, hypertensive nephrosclerosis and nephrotic syndrome, which diseases diagnostically may have the following characteristics: abnormally reduced creatinine and/or water excretion, abnormally increased urine, nitrogen, potassium and/or creatinine blood concentrations, altered renal enzyme activity (e.g., glutamate synthase), altered urine permeability or urine volume, increased microalbuminuria, macroproteinuria, injury on the glomeruli and arterioles, tubular dilation, hyperphosphatemia, and/or dialysis need. The invention also encompasses the use of the compounds according to the invention for the treatment and/or prophylaxis of the sequelae of renal insufficiency, such as pulmonary edema, heart failure, uremia, anemia, electrolyte disorders (e.g. hypercalcemia, hyponatremia) as well as bone disorders and disorders of carbohydrate metabolism.
In addition, the compounds of the invention are also suitable for the treatment and/or prevention of asthma, Pulmonary Arterial Hypertension (PAH) and other forms of Pulmonary Hypertension (PH), including left heart disease, HIV, sickle cell anemia, thromboembolism (CTEPH), Sarkoidose, COPD or pulmonary hypertension associated with pulmonary fibrosis, Chronic Obstructive Pulmonary Disease (COPD), Acute Respiratory Distress Syndrome (ARDS), Acute Lung Injury (ALI), alpha-1-antitrypsin deficiency (AATD), pulmonary fibrosis, emphysema (e.g. smoking-induced emphysema) and Cystic Fibrosis (CF).
The compounds described herein are also useful in the control of central nervous system disorders characterized by disturbances of the NO/cGMP system. They are particularly suitable for use in improving perception, attention, learning or memory following cognitive impairment including cognitive impairment accompanying situational/disease/syndrome such as mild cognitive impairment, learning and memory decline accompanying the aging, memory loss accompanying the aging, vascular dementia, craniocerebral trauma, stroke, dementia occurring after stroke (post-stroke dementia), post-traumatic craniocerebral trauma, general attention deficit disorder, attention deficit disorder in childhood learning and memory problems, alzheimer's disease, lewy body dementia, frontal lobe degenerative dementia (including pick's syndrome, parkinson's disease, progressive nuclear palsy), corticobasal degenerative dementia, Amyotrophic Lateral Sclerosis (ALS), huntington's disease, demyelination, multiple sclerosis, thalamus degeneration, creutzfeldt-jakob disease dementia, HIV dementia, schizophrenia with dementia, or coxsackie psychosis. They are also suitable for the treatment and/or prophylaxis of central nervous system disorders, such as anxiety states, stress and depression, central nervous-related sexual dysfunction and sleep disorders, and of pathological disorders for controlling the intake of food, stimulants and addictive substances.
The compounds of the present invention are additionally suitable for controlling cerebral blood flow and thus may be used as effective agents for controlling migraine. They are also suitable for the prevention and control of cerebral infarction sequelae, such as stroke, cerebral ischemia and craniocerebral trauma. The compounds of the invention are also useful for controlling pain and tinnitus conditions.
In addition, the compounds of the present invention have anti-inflammatory effects and are therefore useful as anti-inflammatory agents for the treatment and/or prevention of sepsis (SIRS), multiple organ failure (MODS, MOF), inflammatory disorders of the kidney, chronic enteritis (IBD, crohn's disease, UC), pancreatitis, peritonitis, rheumatoid disease, inflammatory skin diseases and inflammatory eye diseases.
Furthermore, the compounds of the present invention may also be useful in the treatment and/or prevention of autoimmune diseases.
The compounds according to the invention are also suitable for the treatment and/or prophylaxis of fibrotic diseases of internal organs such as the lung, heart, kidney, bone marrow and in particular of the liver, and also of dermal fibrosis and fibrotic eye diseases. Within the scope of the present invention, the term fibrotic disease specifically includes the following terms: liver fibrosis, cirrhosis, lung fibrosis, myocardial endocardial fibrosis, kidney disease, glomerulonephritis, interstitial kidney fibrosis, fibrotic damage from diabetes, myelofibrosis and similar fibrotic diseases, scleroderma (also known as systemic sclerosis), maculopathy, keloids, hypertrophic scars (and after surgical procedures), nevi, diabetic retinopathy, proliferative vitreoretinopathy and connective tissue diseases (e.g. sarcoidosis).
The compounds of the invention are also suitable for controlling post-operative scarring, for example due to glaucoma surgery.
The compounds of the invention can also be used cosmetically in the case of aged and keratinized skin.
In addition, the compounds of the invention are suitable for the treatment and/or prophylaxis of hepatitis, neoplasms, osteoporosis, glaucoma and gastroparesis.
The compounds of the present invention are also suitable for the treatment and/or prevention of gastrointestinal disorders, e.g., gastrointestinal inflammation, gastrointestinal cancer, gastrointestinal dysfunction, etc.; and esophageal motility disorders. Gastrointestinal disorders include, for example, Irritable Bowel Syndrome (IBS), non-ulcer dyspepsia, chronic intestinal pseudo-obstruction, functional dyspepsia, colonic pseudo-obstruction, duodenal reflux, gastroesophageal reflux disease (GERD), inflammatory intestinal obstruction (e.g., post-operative ileus), gastroparesis, heartburn (high acidity in the gastrointestinal tract), constipation (e.g., constipation associated with the use of drugs such as opioids, osteoarthritic drugs, osteoporotic drugs; post-operative constipation; constipation associated with neurological disorders). Esophageal motility disorders include, for example, achalasia of the cardia (also known as achalasia), diffuse esophageal spasm, and nutcracker esophagus.
The invention further provides the use of a compound of the invention for the treatment and/or prophylaxis of diseases, in particular of the abovementioned diseases.
The present invention further provides the use of a compound of the invention for the treatment and/or prevention of: heart failure, angina pectoris, hypertension, pulmonary hypertension, ischemia, vascular disease, renal disease, thromboembolic disease, male sexual dysfunction, systemic sclerosis, sickle cell anemia, cardiac achalasia, fibrotic disease and/or arteriosclerosis.
The invention further provides the use of a compound of the invention for the preparation of a medicament for the treatment and/or prophylaxis of diseases, in particular of the abovementioned diseases.
The invention further provides the use of a compound of the invention in the manufacture of a medicament for the treatment and/or prevention of: heart failure, angina pectoris, hypertension, pulmonary hypertension, ischemia, vascular disease, renal disease, thromboembolic disease, male sexual dysfunction, systemic sclerosis, sickle cell anemia, cardiac achalasia, fibrotic disease and/or arteriosclerosis.
The present invention further provides a method for the treatment and/or prophylaxis of diseases, in particular of the abovementioned diseases, with an effective amount of at least one compound according to the invention.
The present invention further provides a method for the treatment and/or prophylaxis of the following diseases with an effective amount of at least one compound according to the invention: heart failure, angina pectoris, hypertension, pulmonary hypertension, ischemia, vascular disease, renal disease, thromboembolic disease, male sexual dysfunction, systemic sclerosis, sickle cell anemia, cardiac achalasia, fibrotic disease and/or arteriosclerosis.
The compounds of the invention can be used alone or, if desired, in combination with other active substances. The invention further provides medicaments, in particular for the treatment and/or prophylaxis of the abovementioned diseases, which comprise at least one compound according to the invention and one or more further active substances. Preferred examples of suitable active substance combinations include:
(1) organic nitrate and NO donors, such as sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SIN-1, and inhaled NO;
(2) compounds which inhibit cyclic guanosine monophosphate (cGMP) cleavage, for example Phosphodiesterase (PDE)1,2 and/or 5 inhibitors, in particular PDE5 inhibitors such as sildenafil, vardenafil and tadalafil;
(3) an agent having an anticoagulant effect, for example, an agent preferably selected from the group consisting of a platelet aggregation inhibitor group, an anticoagulant group, or a plasminogen group;
(4) the blood pressure lowering active substance, for example, is preferably an active substance selected from the group consisting of: calcium antagonists, angiotensin II antagonists, ACE inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid receptor antagonists, and diuretics groups;
(5) active substances which alter lipid metabolism, for example, preferably selected from the group of thyroid receptor agonists, cholesterol synthesis inhibitors (for example, inhibitors of the group of preferably HMG-CoA reductase inhibitors or squalene synthesis inhibitors), ACAT inhibitors, CETP inhibitors, MTP inhibitors, PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, lipase inhibitors, bile acid adsorbents, bile acid reabsorption inhibitors and lipoprotein (a) antagonists; and/or
(6) The agent having antithrombotic activity preferably means a compound selected from the group of platelet aggregation inhibitors, anticoagulants or plasminogen agents.
In some embodiments of the invention, the compounds of the invention are administered in combination with a platelet aggregation inhibitor (e.g., preferably aspirin, clopidogrel, ticlopidine, rivaroxaban, or dipyridamole).
In some embodiments of the invention, the compounds of the invention are administered in combination with a thrombin inhibitor (e.g., ximegadine, dabigatran, melagatran, bivalirudin, or cricet).
In some embodiments of the invention, a compound of the invention is administered in combination with a GPIIb/IIIa receptor antagonist (e.g., tirofiban or abciximab).
In some embodiments of the invention, the compounds of the invention are administered in combination with a factor Xa inhibitor: the Xa factor inhibitor is preferably rivaroxaban, DU-176b, apixaban, otamixaban, feditaban, razasban, fondaparinux sodium, epidoparin, PMD-3112, YM-150, KFA-1982, EMD-503982, MCM-17, MLN-1021, DX9065a, DPC906, JTV803, SSR-126512, or SSR-128428.
In some embodiments of the invention, the compounds of the invention are administered in combination with heparin or with a Low Molecular Weight (LMW) heparin derivative.
In some embodiments of the invention, the compounds of the invention are administered in combination with a vitamin K antagonist (e.g., coumarin).
In some embodiments of the invention, the compounds of the invention are administered in combination with a calcium antagonist (e.g., nifedipine, amlodipine, verapamil or diltiazem).
In some embodiments of the invention, the compounds of the invention are administered in combination with an alpha-1 receptor blocker (e.g., and preferably prazosin).
In some embodiments of the invention, the compounds of the invention are administered in combination with a beta blocker, preferably propranolol, atenolol, timolol, pinolol, allylalol, oxprenolol, penbutolol, blanalol, metipranolol, nadolol, mepindolol, carabronolol, sotalol, metoprolol, betaxolol, celiprolol, bisoprolol, carteolol, esmolol, labetalol, carvedilol, adaprolol, landiolol, nebivolol, epalol, or bucindolol.
In some embodiments of the invention, the compounds of the invention are administered in combination with an angiotensin II antagonist (e.g., losartan, candesartan, valsartan, telmisartan, or embsartan).
In some embodiments of the invention, the compounds of the invention are administered in combination with an ACE inhibitor, preferably enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinapril, perindopril, or trandolapril.
In some embodiments of the invention, a compound of the invention is administered in combination with an endothelin antagonist (e.g., bosentan, darussan, ambrisentan, or sitaxsentan).
In some embodiments of the invention, a compound of the invention is administered in combination with a renin inhibitor (e.g., aliskiren, SPP-600, or SPP-800).
In some embodiments of the invention, the compounds of the invention are administered in combination with a mineralocorticoid receptor antagonist (e.g., spironolactone or eplerenone).
In some embodiments of the invention, the compounds of the invention are administered in combination with: loop diuretics (e.g., furosemide, torasemide, bumetanide, and piretanide), potassium sparing diuretics (e.g., amiloride and triamterene), aldosterone antagonists (e.g., spironolactone, potassium entestopropionate, and eplerenone), and thiazide diuretics (e.g., hydrochlorothiazide, chlorothiadone, empanamide, and indapamide).
In some embodiments of the invention, a compound of the invention is administered in combination with a CETP inhibitor (e.g., dacipratropium, BAY60-5521, anacetrapib, or CETP vaccine (CETi-1)).
In some embodiments of the invention, a compound of the invention is administered in combination with a thyroid receptor agonist (e.g., D-thyroxine, 3,5,3' -liothyronine (T3), CGS23425, or acitinic (CGS 26214)).
In some embodiments of the invention, the compounds of the invention are administered in combination with an HMG-CoA reductase inhibitor selected from the group of statins, preferably lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin or pitavastatin.
In some embodiments of the invention, a compound of the invention is administered in combination with a squalene synthesis inhibitor (e.g., BMS-188494 or TAK-475).
In some embodiments of the invention, the compounds of the invention are administered in combination with an ACAT inhibitor (e.g., avasimibe, melinamide, paratubb, ibrutinib, or SMP-797).
In some embodiments of the invention, a compound of the invention is administered in combination with an MTP inhibitor (e.g., Enptota brand, BMS-201038, R-103757, or JTT-130).
In some embodiments of the invention, a compound of the invention is administered in combination with a PPAR-gamma agonist (e.g., pioglitazone or rosiglitazone).
In some embodiments of the invention, the compounds of the invention are administered in combination with a PPAR-delta agonist (e.g., GW501516 or BAY 68-5042).
In some embodiments of the invention, the compounds of the invention are administered in combination with a cholesterol absorption inhibitor (e.g., ezetimibe, tiquinan, or pamabrin).
In some embodiments of the invention, the compounds of the invention are administered in combination with a lipase inhibitor, a preferred example of which is orlistat.
In some embodiments of the invention, a compound of the invention is administered in combination with a polymeric bile acid absorber (e.g., cholestyramine, colestipol, colesevelam, or colestimide).
In some embodiments of the invention, a compound of the invention is administered in combination with a bile acid reabsorption inhibitor (e.g., an ASBT (IBAT) inhibitor such as AZD-7806, S-8921, AK-105, bali-1741, SC-435, or SC-635).
In some embodiments of the invention, a compound of the invention is administered in combination with a lipoprotein ester antagonist (e.g., Gemcabennecalcum (CI-1027) or niacin).
General synthetic procedure
In this specification, a structure is dominant if there is any difference between the chemical name and the chemical structure.
In general, the compounds of the invention can be prepared by the methods described herein, unless otherwise indicated, wherein the substituents are as defined in formula (I) or formula (II) or formula (IIa). The following reaction schemes and examples serve to further illustrate the context of the invention.
Those skilled in the art will recognize that: the chemical reactions described herein may be used to suitably prepare other compounds of the invention, and other methods for preparing compounds of the invention are considered to be within the scope of the invention. For example, the synthesis of those non-exemplified compounds according to the present invention can be successfully accomplished by those skilled in the art by modification, such as appropriate protection of interfering groups, by the use of other known reagents other than those described herein, or by some routine modification of the reaction conditions. In addition, the reactions disclosed herein or known reaction conditions are also recognized as being applicable to the preparation of other compounds of the present invention.
In the examples described below, all temperatures are given in degrees celsius unless otherwise indicated. Unless otherwise indicated, reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Arco Chemical Company and Alfa Chemical Company and were used without further purification; general reagents were purchased from Shantou West Long chemical plant, Guangdong Guanghua chemical plant, Guangzhou chemical plant, Tianjin HaoYu Chemicals Co., Ltd, Qingdao Tenglong chemical reagent Co., and Qingdao maritime chemical plant.
The anhydrous tetrahydrofuran, dioxane, toluene and ether are obtained through reflux drying of metal sodium. The anhydrous dichloromethane and chloroform are obtained by calcium hydride reflux drying. Ethyl acetate, petroleum ether, N-hexane, N, N-dimethylacetamide and N, N-dimethylformamide were used as they were previously dried over anhydrous sodium sulfate.
The following reactions are generally carried out under positive pressure of nitrogen or argon or by sleeving a dry tube over an anhydrous solvent (unless otherwise indicated), the reaction vial being stoppered with a suitable rubber stopper and the substrate being injected by syringe. Glassware was dried.
The column chromatography is performed using a silica gel column. Silica gel (300 and 400 meshes) was purchased from Qingdao oceanic chemical plants. NMR spectral data were measured by Bruker Avance 400 NMR spectrometer or Bruker Avance III HD 600 NMR spectrometer, CDC1 3 ,DMSO-d 6 ,CD 3 OD or Acetone-d 6 As solvent (reported in ppm) TMS (0ppm) or chloroform (7.25ppm) was used as reference standard. When multiple peaks occur, the following abbreviations will be used: s (singleton, singlet), d (doublet ), t (triplet, triplet), m (multiplet ), q (quatet, quartet), br (broadpede, broad), dd (doublet of doublets, doublet), dt (doublet of triplets, doublet), dq (doublet of quatts, doublet), ddd (doublet of doublet of doublets, doublet of doublets), ddt (doublet of doublet of triplets, doublet of doublet), dddd (doublet of doublet of doublet). Coupling constant J, expressed in Hertz (Hz).
Low resolution Mass Spectral (MS) data were measured by an Agilent 6320 series LC-MS spectrometer equipped with a G1312A binary pump and a G1316A TCC (column temperature maintained at 30 ℃), a G1329A autosampler and a G1315B DAD detector were applied for analysis, and an ESI source was applied to the LC-MS spectrometer.
Low resolution Mass Spectrometry (MS) data were measured by Agilent 6120 series LC-MS spectrometer equipped with a G1311A quaternary pump and a G1316A TCC (column temperature maintained at 30 ℃), with a G1329A autosampler and a G1315D DAD detector applied for analysis, and an ESI source applied to the LC-MS spectrometer.
Both spectrometers were equipped with an Agilent Zorbax SB-C18 column, 2.1X 30mm, 5 μm. The injection volume is determined by the sample concentration; the flow rate is 0.6 mL/min; peaks of HPLC were recorded by UV-Vis wavelength at 210nm and 254 nm. The mobile phases were 0.1% formic acid in acetonitrile (phase a) and 0.1% formic acid in ultrapure water (phase B). Gradient elution conditions are shown in table 1:
TABLE 1 gradient elution conditions for low resolution mass spectrometry mobile phase
Time (min) A(CH 3 CN,0.1%HCOOH) B(H 2 O,0.1%HCOOH)
0-3 5-100 95-0
3-6 100 0
6-6.1 100-5 0-95
6.1-8 5 95
The following acronyms are used throughout the invention:
CDC1 3 deuterated chloroform
DMSO-d 6 Deuterated dimethyl sulfoxide
DMSO dimethyl sulfoxide
g
mg of
mol mole of
mmol millimole
h hours
min for
mL of
Microliter of μ L
The following reaction scheme describes the steps for preparing the compounds disclosed herein. Wherein, unless otherwise stated, L 1 Is F, Cl, Br, I or other suitable leaving group, L 2 Is H or a leaving group such as Cl, Br, etc., L 3 Is a leaving group such as Cl, Br, etc.; each of L, ring C1, R 0 、R 6 、R 7 、R 8a 、R 8c 、R 8d 、R 8e 、R 9 、R 10a And n has the meaning as described in the present invention.
Reaction scheme
Reaction scheme 1
Figure BDA0001623638800000431
Intermediate compound A8a can be prepared according to the procedure described in scheme 1: reacting 2, 6-dichloro-5-fluoro-pyridine-3-nitrile under the action of concentrated sulfuric acid to obtain a compound A1, and reacting under the action of zinc and acetic acid to obtain 2-chloro-5-fluoronicotinamide A2. Compound a2 is reacted with phosphorus oxychloride to give compound A3, which is reduced (e.g., by hydrazine hydrate) to give pyrazolopyridine compound a 4. Compound A4 is diazotized with a suitable reagent (e.g., boron trifluoride etherate and isoamyl nitrite) and then reacted with an iodine-containing reagent (e.g., sodium iodide, etc.) to provide iodo compound A5 a. Compound A5a is reacted with 1- (bromomethyl) -2-fluoro-benzene in the presence of a base (e.g., cesium carbonate, etc.) to give compound A6a, which is then reacted with cuprous cyanide to give cyano-substituted compound A7a, and finally reacted with sodium methoxide, ammonium chloride, and acetic acid to give formamidine compound A8 a.
Reaction scheme 2
Figure BDA0001623638800000432
Intermediate compound A8 can be prepared by the method described in scheme 2: the compound A5 and the compound A9 undergo substitution reaction to obtain a compound A6, then undergo cyanation reaction to obtain a cyano-substituted compound A7, and finally undergo addition reaction under the action of a suitable reagent to obtain a formamidine compound A8.
Reaction scheme 3
Figure BDA0001623638800000441
Compound a11 can be prepared by the method described in reaction scheme 3: the compound A8 and phenyl azomalononitrile react to obtain a compound A9, and the compound A9 is subjected to hydrogenation reduction reaction under the action of a metal palladium catalyst to obtain an intermediate compound A10. Reaction of compound a10 with a suitable reagent can afford compound a 11. For example, when R is 8c Is tetrahydropyranyl ethyl
Figure BDA0001623638800000442
When the compound A11 can be prepared from the compound A10 and tetrahydropyranone
Figure BDA0001623638800000443
Reacting under the action of acetic acid and sodium cyanoborohydride.
Reaction scheme 4
Figure BDA0001623638800000444
Compounds a13, a20, a21 can be prepared by the methods described in reaction scheme 4: compound A8 was reacted with compound a12 to give compound a 13. Substitution of compound a13 with a suitable reagent (e.g., phosphorus oxychloride, etc.) affords chloro compound a 19. Compound A19 with substrate R 8a NH 2 Under the action of a proper reagent (for example, under the action of a base such as triethylamine), the reaction can obtain the compound A21.
Compound A13 may also be reacted with the substrate R 9 L a (wherein L a A leaving group such as Cl, Br, etc.) under the action of a suitable reagent (e.g., under the action of a base such as cesium carbonate, etc.) to give compound a 20.
Compound A20 can also be prepared from compound A19 and a suitable substrate (e.g., R) 9 ONa, etc.).
Reaction scheme 5
Figure BDA0001623638800000451
Compound a16 can be prepared according to the procedure described in reaction scheme 5: directly reacting compound A8 with ethyl ethoxymethylene cyanoacetate to obtain compound A14, hydrolyzing with base (such as sodium hydroxide) to obtain compound A15, and reacting with compound NHR 8c R 8d Condensing under the action of a condensing agent to obtain a compound A16.
Reaction scheme 6
Figure BDA0001623638800000452
Compounds A17 and A18 can be prepared as described in scheme 6: the compound A10 and the compound S-a are subjected to condensation reaction under the action of a proper reagent (a condensation reagent or alkali) to obtain a compound A17, and then the compound A17 and the compound R are subjected to condensation reaction 8e -L 3 The reaction yielded compound a 18.
The following examples may further illustrate the present invention, however, these examples should not be construed as limiting the scope of the present invention. Examples of preparation of intermediates
Intermediate 15-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine-3-formamidine
Figure BDA0001623638800000453
Step 1)2, 6-dichloro-5-fluoronicotinamide
2, 6-dichloro-5-fluoro-pyridine-3-carbonitrile (24.8g,130mmol) was weighed into a 500-mL flask, concentrated sulfuric acid (125mL) was added thereto, and the temperature was raised to 62 ℃ for reaction for 1 hour. After cooling to room temperature, the reaction mixture was poured into ice-water (800mL), extracted with ethyl acetate (300 mL. times.4), and the organic phase was washed successively with water (100mL), a saturated sodium bicarbonate solution (100mL) and a saturated brine (50mL), and dried over anhydrous sodium sulfate. Filtration and spin-drying of the filtrate gave a pale yellow solid (27.0g, 99.5%).
Step 2) 2-chloro-5-fluoronicotinamide
2, 6-dichloro-5-fluoronicotinamide (27.0g,129mmol) was weighed into a 500mL flask, methanol (130mL), zinc powder (26.8g,410mmol) and glacial acetic acid (23.0mL,402mmol) were added thereto, and the temperature was raised to 80 ℃ for reaction for 24 hours. Cool to room temperature, filter through celite to remove zinc dust, wash with methanol (50mL), combine filtrates, remove solvent by rotary evaporation, and purify the residue by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 1/1) to give a pale yellow solid (17.0g, 75.4%).
MS(ESI,pos.ion)m/z:175.0(M+1).
Step 3) 2-chloro-5-fluoro-pyridine-3-carbonitrile
2-chloro-5-fluoronicotinamide (6.4g,37mmol) was weighed into a 500mL flask, chloroform (100mL) and phosphorus oxychloride (24.0mL,264mmol) were added thereto, and the temperature was raised to 80 ℃ for reaction for 8 hours. After cooling to room temperature, the reaction was quenched by addition of saturated sodium bicarbonate solution (100mL), extracted with dichloromethane (50 mL. times.3), and the organic phases were combined, washed successively with saturated sodium bicarbonate solution (50mL) and saturated brine (30mL), and dried over anhydrous sodium sulfate. The filtrate was spun dry and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 50/1) to give a white solid (3.80g, 66.0%).
1 H NMR(400MHz,CDCl 3 )δ(ppm)8.51(d,J=2.9Hz,1H),7.79(dd,J=6.8,2.9Hz,1H).
Step 4) 5-fluoro-1H-pyrazolo [3,4-b ] pyridin-3-amine
2-chloro-5-fluoro-pyridine-3-carbonitrile (8.0g,51mmol) was weighed into a 250 mL flask, ethylene glycol (80mL) and hydrazine hydrate (24mL,494mmol) were added thereto, and the reaction was refluxed at 180 ℃ overnight. Cooled to room temperature, water (50mL) was added, stirred at room temperature for 10 min, and filtered. The filter cake was washed successively with water (30mL) and tetrahydrofuran (30mL, -10 ℃ C.) and dried in vacuo to give a yellow solid (5.0g, 64.0%).
MS(ESI,pos.ion)m/z:153.10(M+1).
Step 5) 5-fluoro-3-iodo-1H-pyrazolo [3,4-b ] pyridine
5-fluoro-1H-pyrazolo [3,4-b ] pyridin-3-amine (5.0g,33mmol) was weighed into a 500mL flask, tetrahydrofuran (150mL) was added thereto, cooled to 0 ℃, boron trifluoride diethyl ether (8.1mL,66mmol) was added thereto, cooling was continued to-10 ℃, isoamylnitrite (5.7mL,42mmol) was added thereto, stirred for 30 minutes, cooled diethyl ether (100mL) was added thereto, and filtered to obtain a diazonium salt. Another 250 mL reaction flask was charged with sodium iodide (6.4g,43mmol) and acetone (150mL), cooled to 0 deg.C, added with the diazonium salt prepared above, and allowed to warm to room temperature for 30 minutes. The reaction solution was poured into ice water (300mL), extracted with ethyl acetate (50 mL. times.3), and the organic phase was washed successively with a sodium thiosulfate solution (50mL) and a saturated brine (50mL), dried over anhydrous sodium sulfate, filtered and dried to give a yellow solid (8.0g, 93.0%) which was used directly in the next reaction.
Step 6) 5-fluoro-1- (2-fluorobenzyl) -3-iodo-1H-pyrazolo [3,4-b ] pyridine
5-fluoro-3-iodo-1H-pyrazolo [3,4-b ] pyridine (10.0g,38.0mmol) was weighed in a 500-mL flask, and N, N-dimethylformamide (150mL), cesium carbonate (13.6g,41.7mmol) and 1- (bromomethyl) -2-fluoro-benzene (5.0mL,41.5mmol) were added thereto and reacted at room temperature for 2 hours. The reaction was poured into saturated sodium chloride solution (500mL), extracted with ethyl acetate (50 mL. times.3), and the organic phase was washed with saturated brine (50mL), dried over anhydrous sodium sulfate, filtered, and spun dry to give a yellow solid (10.0g, 70.9%) which was used in the next reaction without further purification.
Step 7) 5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine-3-carbonitrile
5-fluoro-1- (2-fluorobenzyl) -3-iodo-1H-pyrazolo [3,4-b ] pyridine (10.0g,26.9mmol) was weighed in a 500-mL flask, and dimethyl sulfoxide (150mL) and cuprous cyanide (3.0g,33mmol) were added thereto, and the mixture was heated to 150 ℃ for reaction for 2 hours. Cool to room temperature, quench the reaction with ammonia (70mL) and water (400mL), stir for 10 min, extract with ethyl acetate (300mL × 5), combine the organic phases, spin dry, and purify the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 10/1) to give a yellow solid (5.0g, 69.0%).
MS(ESI,pos.ion)m/z:271.0(M+1).
Step 8) 5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine-3-carboxamidine
Sodium methoxide (0.66g,12mmol) was weighed into a 250 mL flask, and methanol (100mL) and 5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine-3-carbonitrile (2.9g,11mmol) were added thereto, followed by stirring at room temperature for 2 hours. Ammonium chloride (0.72g,13mmol) and glacial acetic acid (2.5mL,44mmol) were added and the reaction was refluxed at 90 ℃ overnight. Cooled to room temperature and the solvent removed by rotary evaporation. Water (30mL) and ethyl acetate (30mL) were added, and a 2mol/L aqueous solution of sodium hydroxide was added to adjust the pH to 10. Extraction with ethyl acetate (30 mL. times.3), combining the organic phases, drying over anhydrous sodium sulfate, filtration, spin-drying gave a yellow oil (3.0g, 97.0%) which was used in the next reaction without further purification.
MS(ESI,pos.ion)m/z:288.1(M+1).
Intermediate 22- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine
Figure BDA0001623638800000471
Step 1)2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) -5- (phenylazo) pyrimidine-4, 6-diamine
5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine-3-carboxamidine (3.0g,10.0mmol) was weighed in a 250 mL flask, N-dimethylformamide (30.0mL) and triethylamine (2.0mL,14.0mmol) were added thereto, the temperature was raised to 85 ℃, phenylazopropionitrile (2.2g,13mmol) was added, and the temperature was raised to 100 ℃ for 4 hours. Cool to room temperature, stand overnight, add water (150mL), stir for 1 hour, filter, wash the filter cake sequentially with water (20mL) and methanol (20mL), and dry in vacuo to afford a yellow solid (4.0g, 84.0%).
MS(ESI,pos.ion)m/z:458.1(M+1).
Step 2)2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine
2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) -5- (phenylazo) pyrimidine-4, 6-diamine (4.0g,10mmol) was weighed in a 250 mL flask, N, N-dimethylformamide (80mL) and 10% palladium on carbon (0.6g) were added thereto, and reacted at room temperature for 8 hours, the reaction solution was filtered through celite, washed with N, N-dimethylformamide (20mL), and the filtrate was spin-dried to obtain a brown-yellow solid (3.0g, 81.0%).
MS(ESI,pos.ion)m/z:369.4(M+1).
Examples
Example 14- (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) morpholin-3-one
Figure BDA0001623638800000481
Step 1)2- (2-chloroethoxy) acetyl chloride
2- (2-chloroethoxy) acetic acid (1.0g,7.2mmol) was dissolved in dichloromethane (10.0mL), oxalyl chloride (1.2mL,14mmol) was added, and the mixture was stirred at room temperature for 4 hours. The solvent was distilled off under reduced pressure and the crude product was used directly in the next reaction.
1 H NMR(400MHz,CDCl 3 )δ(ppm)4.49(s,2H),3.84(t,J=5.5Hz,2H),3.68–3.59(m,2H).
Step 2)2- (2-chloroethoxy) -N- (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) acetamide
2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine (500mg,1.36mmol) was dissolved in dichloromethane (20mL), and a solution of pyridine (0.35mL,4.3mmol) and 2- (2-chloroethoxy) acetyl chloride (250mg,1.59mmol) in dichloromethane (5mL) was added, followed by stirring at room temperature for 1 hour. It was extracted with dichloromethane (70 mL. times.2). The organic phases were combined, washed successively with water (30mL) and saturated brine (40mL), and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent under reduced pressure gave a yellow solid (600mg, 90.4%).
MS(ESI,pos.ion)m/z:489.1(M+1).
Step 3)4- (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) morpholin-3-one
2- (2-chloroethoxy) -N- (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) acetamide (1.0g,2.0mmol) was dissolved in anhydrous tetrahydrofuran (50mL) at 0 deg.C, 60% sodium hydride (90mg,2.25mmol) was added and stirring continued at 0 deg.C for 2 hours. The reaction was quenched with water and extracted with ethyl acetate (50 mL. times.3). The organic phases were combined, washed successively with water (20mL) and saturated brine (20mL), and dried over anhydrous sodium sulfate. Filtration, evaporation of the solvent under reduced pressure, purification of the crude product by silica gel column chromatography (ethyl acetate) gave a yellow solid which was recrystallized from ethyl acetate to give a pale yellow solid (150mg, 16.0%).
MS(ESI,pos.ion)m/z:453.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.90(dd,J=8.9,2.7Hz,1H),8.66(s,1H),7.36(dd,J=13.4,7.2Hz,1H),7.18(ddd,J=18.3,16.2,8.7Hz,3H),6.45(s,4H),5.80(s,2H),4.16(s,2H),4.04(d,J=5.2Hz,2H),3.48(s,2H).
Example 21- ((4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrrolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) amino) -2-methylpropan-2-ol
Figure BDA0001623638800000491
Step 1)2- (benzyloxy) -2-methylpropionaldehyde
2- (benzyloxy) -2-methylpropan-1-ol (0.45g,2.5mmol) was added to dichloromethane (20mL) followed by dess-Martin oxidant (2.1g,5.0mmol) and stirred at room temperature for 2 h. After dilution with dichloromethane (60mL), the mixture was washed with saturated aqueous sodium bicarbonate (80mL), water (80mL) and saturated brine (80mL), the organic phase was dried over anhydrous sodium sulfate, filtered with suction, and spin-dried, and the residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 15/1) to give a colorless oil (0.37g, 83.0%).
Step 2) N 5 - (2- (benzyloxy) -2-methylPhenylpropyl) -2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b]Pyridin-3-yl) pyrimidine-4, 5, 6-triamines
2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine (0.10g,0.27mmol) and 2- (benzyloxy) -2-methylpropanal (0.053g,0.30mmol) were added to methanol (15mL), acetic acid (0.078mL,1.4mmol) was added, and after stirring at room temperature for 1 hour, sodium cyanoborohydride (0.085g,1.4mmol) was added and stirring continued at room temperature overnight. The solvent was distilled off, a saturated aqueous sodium bicarbonate solution (50mL) was added to the residue, ethyl acetate was extracted (30mL × 2), the organic phase was washed with water (50mL) and a saturated brine (50mL), dried over anhydrous sodium sulfate, filtered with suction, dried by spinning, and the residue was separated by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 100/1) to give a yellowish brown solid (0.090g, 62.0%).
Step 3)1- ((4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrrolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) amino) -2-methylpropan-2-ol
Will N 5 - (2- (benzyloxy) -2-methylpropanyl) -2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidine-4, 5, 6-triamine (0.090g,0.17mmol) was added to methanol (8mL), 10% palladium on carbon (0.10g) was added, and the mixture was stirred under hydrogen at 50 ℃ overnight. The filtrate was suction filtered, the filtrate was spin-dried, and the residue was separated by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 100/1) to give a pale yellow solid (0.059g, 79.0%).
MS(ESI,pos.ion)m/z:441.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.85(dd,J=8.8,2.6Hz,1H),8.65(s,1H),7.36(dd,J=13.1,6.6Hz,1H),7.18(ddd,J=19.0,16.4,8.7Hz,3H),6.28(s,4H),5.77(s,2H),4.72(s,1H),3.51(s,1H),2.66(s,2H),1.15(s,6H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.29(s),-136.77(s).
Example 32- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) -N 5 - (1- (1-methyl-1H-pyrazol-3-yl) ethyl) pyrimidine-4, 5, 6-triamine
Figure BDA0001623638800000501
Step 1) 1-methyl-1H-pyrazole-3-carbaldehyde
1H-pyrazole-3-carbaldehyde (4.02g,41.8mmol), potassium carbonate (11.5g,83.2mmol) and N, N-dimethylformamide (35mL) were charged in a 100mL two-necked flask, followed by methyl iodide (3.89mL,62.5mmol), and the mixture was stirred at room temperature overnight. Water was added, extraction was performed with ethyl acetate (40mL × 6), the organic phase was washed with water (80mL) and saturated brine (80mL) in turn, dried over anhydrous sodium sulfate, suction filtered, and dried, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 10/1) to give a pale yellow oil (2.37g, 51.4%).
MS(ESI,pos.ion)m/z:111.1(M+1);
1 H NMR(600MHz,CDCl 3 )δ(ppm)9.97(s,1H),7.44(d,J=2.2Hz,1H),6.81(d,J=2.3Hz,1H),4.03(s,3H).
Step 2)1- (1-methyl-1H-pyrazol-3-yl) ethanol
1-methyl-1H-pyrazole-3-carbaldehyde (0.32g,2.9mmol) and tetrahydrofuran (10.0mL) were added dropwise to a 50mL two-necked flask, and a 3.0mol/L tetrahydrofuran solution of methylmagnesium bromide (1.3mL,3.9mmol) was added dropwise at 0 ℃ and then stirred at room temperature overnight. The reaction was quenched by addition of saturated aqueous ammonium chloride, dried by evaporation, the residue dissolved in tetrahydrofuran, filtered and the filtrate dried by evaporation to give a white solid (0.16g, 44%).
MS(ESI,pos.ion)m/z:127.1(M+1).
Step 3)1- (1-methyl-1H-pyrazol-3-yl) acetaldehyde
1- (1-methyl-1H-pyrazol-3-yl) ethanol (0.15g,1.2mmol) and methylene chloride (20.0mL) were charged into a 50mL single-neck flask, and dess-martin oxidant (1.0g,2.4mmol) was added thereto at room temperature, followed by stirring at room temperature for 2 hours. Suction was performed, the filter cake was washed with dichloromethane, the filtrate was spun dry, and the residue was purified by neutral alumina column chromatography (petroleum ether/ethyl acetate (v/v) ═ 6/1) to give a colorless oil (0.13g, 88%).
MS(ESI,pos.ion)m/z:125.2(M+1).
Step 4)2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b]Pyridin-3-yl) -N 5 - (1- (1-methyl-1H-pyrazol-3-yl) ethyl) pyrimidine-4, 5, 6-triamine
2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine (0.17g,0.46mmol) and methanol (20mL) were added to a 50mL two-necked flask, followed by 1- (1-methyl-1H-pyrazol-3-yl) acetaldehyde (0.086g,0.69mmol) and acetic acid (0.26mL,4.5mmol), followed by stirring at room temperature for 1 hour, sodium cyanoborohydride (0.15g,2.4mmol) was added, and stirring was continued at room temperature overnight. The solvent was distilled off, the residue was added to a saturated sodium bicarbonate solution, extracted with ethyl acetate (30mL × 2), the organic phase was washed successively with water (30mL) and saturated brine (30mL), dried over anhydrous sodium sulfate, suction-filtered, spin-dried, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 60/1) to give a pale yellow solid (0.091g, 41%).
MS(ESI,pos.ion)m/z:477.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.87(dd,J=8.9,2.7Hz,1H),8.64(s,1H),7.53(d,J=1.7Hz,1H),7.40–7.31(m,1H),7.28–7.18(m,1H),7.18–7.10(m,2H),6.27(d,J=1.9Hz,1H),6.02(s,4H),5.77(s,2H),4.20–4.09(m,1H),3.67(d,J=8.6Hz,1H),2.51(s,3H),1.38(d,J=6.6Hz,3H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.30(s),-136.76(s).
Example 42- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) -N 5 - (tetrahydrofuran-3-yl) pyrimidine-4, 5, 6-triamines
Figure BDA0001623638800000511
In a 50mL two-necked flask was added 2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine (0.10g,0.27mmol), dihydro-3 (2H) -furanone (0.035g,0.41mmol), and methanol (10mL), followed by acetic acid (0.16mL,2.8mmol), stirring at room temperature for 1 hour, followed by addition of sodium cyanoborohydride (0.085g,1.4mmol), and stirring at room temperature was continued overnight. The solvent was distilled off, a saturated aqueous sodium bicarbonate solution was added, extraction was performed with ethyl acetate (30mL × 2), the organic phase was washed with water (30mL × 2) and a saturated brine (30mL × 2) in succession, dried over anhydrous sodium sulfate, suction-filtered, spin-dried, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 100/1) to give a pale yellow solid (0.068g, 57%).
MS(ESI,pos.ion)m/z:439.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.88(dd,J=8.9,2.7Hz,1H),8.65(s,1H),7.36(dd,J=13.1,7.3Hz,1H),7.27–7.19(m,1H),7.19–7.11(m,2H),6.14(s,4H),5.78(s,2H),3.94(q,J=7.6Hz,1H),3.75(s,1H),3.66(ddd,J=14.2,8.4,5.3Hz,2H),3.52(dd,J=8.8,3.1Hz,1H),3.46(d,J=6.9Hz,1H),1.90(dt,J=14.6,7.2Hz,1H),1.77(ddd,J=12.0,7.8,4.6Hz,1H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.28(s,),-136.69(s).
Example 5N 5 - (1-cyclohexylethyl) -2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) pyrimidine-4, 5, 6-triamines
Figure BDA0001623638800000521
In a 50mL two-necked flask were added 2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine (0.12g,0.33mmol) and methanol (11.0mL), followed by acetylcyclohexane (0.067mL,0.49mmol) and acetic acid (0.19mL,3.30mmol), stirred at room temperature for half an hour, added sodium cyanoborohydride (0.20g,3.2mmol), and then further stirred at room temperature overnight. The solvent was distilled off, a saturated aqueous sodium bicarbonate solution (30mL) was added, extraction was performed with ethyl acetate (30mL × 2), the organic phase was washed successively with water (30mL) and saturated brine (30mL), dried over anhydrous sodium sulfate, suction-filtered, spin-dried, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 100/1) to give a pale yellow solid (460mg, 30.0%).
MS(ESI,pos.ion)m/z:479.2(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.88(dd,J=8.9,2.7Hz,1H),8.64(s,1H),7.36(dd,J=11.9,5.5Hz,1H),7.25–7.11(m,3H),5.94(s,4H),5.77(s,2H),3.20(d,J=9.2Hz,1H),2.81(dd,J=12.9,6.5Hz,1H),1.92–1.79(m,2H),1.72(d,J=2.8Hz,2H),1.64(d,J=8.8Hz,1H),1.38–1.28(m,2H),1.17(d,J=9.0Hz,2H),1.05(dd,J=23.0,13.2Hz,2H),0.94(d,J=6.4Hz,3H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.30(s),-136.76(s).
Example 62- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3, 4-b)]Pyridin-3-yl) -N 5 - (1- (tetrahydro-2H-pyran-4-yl) ethyl) pyrimidine-4, 5, 6-triamine
Figure BDA0001623638800000522
In a 50mL single-necked flask were added 2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine (0.11g,0.30mmol), 1- (tetrahydro-2H-pyran-4-yl) ethanone (0.050g,0.39mmol), and methanol (10mL), acetic acid (0.17mL,3.0mmol) was added at 0 deg.C, and after stirring at room temperature for 1 hour, was cooled to 0 deg.C, sodium cyanoborohydride (0.094g,1.5mmol) was added, followed by stirring at room temperature overnight. The solvent was evaporated, a saturated aqueous sodium bicarbonate solution (30mL) was added, extraction was performed with ethyl acetate (30mL × 2), the organic phase was washed with water (30mL) and a saturated brine (30mL), dried over anhydrous sodium sulfate, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 80/1, 0.5% triethylamine) to give a pale yellow solid (0.036g, 25.0%).
MS(ESI,pos.ion)m/z:481.3(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.89(dd,J=8.9,2.7Hz,1H),8.65(s,1H),7.36(dd,J=13.4,7.2Hz,1H),7.28–7.20(m,1H),7.16(td,J=14.2,7.4Hz,2H),5.97(s,4H),5.77(s,2H),3.97–3.84(m,2H),3.25(d,J=9.7Hz,2H),2.83(m,1H),1.76(d,J=12.7Hz,1H),1.68(d,J=13.1Hz,1H),1.63–1.51(m,1H),1.32(m,2H),1.25(m,1H),0.95(d,J=6.4Hz,3H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.29(s),-136.77(s).
Example 7N- (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) tetrahydrofuran-2-carboxamide
Figure BDA0001623638800000531
2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine (0.10g,0.27mmol), tetrahydrofuran-2-carboxylic acid (0.046g,0.40mmol), and N, N-dimethylformamide (10.0mL) were added to a 50mL two-necked flask, and 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (0.15g,0.39mmol) and N, N-diisopropylethylamine (0.13mL,0.79mmol) were added at 0 deg.C and stirred at room temperature overnight. After quenching with water, ethyl acetate extraction (30mL × 2), the organic phase was washed successively with water (30mL) and saturated brine (30mL), dried over anhydrous sodium sulfate, filtered with suction, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 80/1, 0.5% triethylamine) to give a white solid (0.067g, 53.0%).
MS(ESI,pos.ion)m/z:467.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.88(dd,J=8.7,2.8Hz,1H),8.79(s,1H),8.73(s,1H),7.42–7.33(m,1H),7.27–7.12(m,3H),6.62(s,4H),5.85(s,2H),4.46(m,1H),4.00(m,1H),3.83(m,1H),2.22–2.05(m,2H),1.98–1.79(m,2H).
Example 8N- (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) cyclobutylcarboxamide
Figure BDA0001623638800000541
2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine (0.15g,0.41mmol), dichloromethane (15mL), and pyridine (0.066mL,0.82mmol) were added to a 50mL two-necked flask, and cyclobutylformyl chloride (0.056mL,0.49mmol) was added at 0 ℃ and stirred at room temperature for 2 hours. Dichloromethane (50mL) was added, followed by washing with water (30mL) and saturated brine (30mL), followed by drying over anhydrous sodium sulfate, suction filtration and spin-drying, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 30/1) to give a pale yellow solid (0.11g, 60.0%).
MS(ESI,pos.ion)m/z:451.2(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.91(dd,J=8.9,2.8Hz,1H),8.69–8.64(m,1H),8.50(s,1H),7.37(td,J=7.9,2.2Hz,1H),7.19(m,3H),6.07(s,4H),5.80(s,2H),3.27(m,1H),2.34–2.23(m,2H),2.16–2.05(m,2H),1.91(m,1H),1.77(m,1H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.24(s),-136.42(s).
Example 9N- (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) cyclopropylcarboxamide
Figure BDA0001623638800000542
2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine (50mg,0.136mmol) was dissolved in dichloromethane (10mL), followed by addition of pyridine (55. mu.L, 0.684mmol) and cyclopropylcarbonyl chloride (25. mu.L, 0.276mmol) and stirring at room temperature for 1 hour. The solvent was evaporated under reduced pressure and the crude product was purified by preparative chromatography to give a white solid (33mg, 55.7%).
MS(ESI,pos.ion)m/z:437.2(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)9.18(s,1H),8.93–8.74(m,2H),7.25(ddd,J=32.5,30.0,7.1Hz,7H),5.90(s,2H),1.80(d,J=4.7Hz,1H),0.90–0.74(m,4H).
Example 10N- (4, 6-diamino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) morpholine-4-carboxamide
Figure BDA0001623638800000551
2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine (106mg,0.288mmol) was placed in a 50-mL flask, and pyridine (10mL) and 4-morpholinecarbonyl chloride (51. mu.L, 0.437mmol) were added thereto and reacted at room temperature for 6 hours. Extraction with ethyl acetate (50mL × 3), washing of the organic phase with water (100mL × 2) and saturated brine (100mL × 1), respectively, drying over anhydrous sodium sulfate, filtration, spin-drying, and separation of the residue by silica gel column chromatography (ethyl acetate/methanol (v/v) ═ 5/1) gave an off-white solid (100mg, 72.0%).
MS(ESI,pos.ion)m/z:482.2(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.81(d,J=6.9Hz,2H),7.61(s,1H),7.41(s,2H),7.38(dd,J=13.2,6.3Hz,2H),7.20(ddd,J=23.1,16.7,8.4Hz,4H),5.91(s,2H),3.63(d,J=4.3Hz,8H).
Example 115-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-ol
Figure BDA0001623638800000552
Step 1) 1-ethoxy-2-fluoro-1, 3-dioxopropane-2-sodium salt
A100 mL single-neck flask was charged with sodium ethoxide (1.41g,20.7mmol) and tetrahydrofuran (30mL), and a solution of ethyl formate (3.3mL,41mmol) and ethyl fluoroacetate (2.0mL,20.7mmol) in tetrahydrofuran (10.4mL) was added dropwise at 0 deg.C and stirred at room temperature overnight. The solvent was evaporated and dried in vacuo to give a pale yellow solid (3.06g, 94.7%).
Step 2) 5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-ol
In a 100mL single-necked flask, 5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine-3-carboxamidine (0.71g,2.5mmol), 1-ethoxy-2-fluoro-1, 3-dioxopropane-2-sodium salt (0.50g,3.2mmol) and ethanol (20mL) were added, and the mixture was heated to reflux for 3 hours. Cooling to room temperature, evaporation of solvent, addition of water (20mL), adjustment of pH 5.0 with 1mol/L hydrochloric acid, suction filtration, water washing, vacuum drying of the solid and purification of the crude product by preparative chromatography gave a white solid (0.82g, 93.0%).
MS(ESI,pos.ion)m/z:358.0(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)13.44(s,1H),8.79(s,1H),8.43(d,J=9.0Hz,1H),8.24(s,1H),7.47–7.31(m,2H),7.29–7.20(m,1H),7.16(t,J=7.5Hz,1H),5.86(s,2H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.18(s),-133.86(s),-151.37(s).
Example 123- (4-ethoxy-5-fluoropyrimidin-2-yl) -5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine
Figure BDA0001623638800000561
Step 1)3- (4-chloro-5-fluoropyrimidin-2-yl) -5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine
Figure BDA0001623638800000562
In a 100mL single-neck flask were added 5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-ol (0.97g,2.7mmol) and phosphorus oxychloride (15mL), and the mixture was heated to reflux for 3 hours. After cooling to room temperature, the solvent was evaporated, ice water (50mL) was added to the residue, the pH was adjusted to 7 with saturated aqueous sodium bicarbonate, the mixture was filtered, washed with water (15mL), and the solid was dried under vacuum to give a pale yellow solid (0.92g, 90.0%).
MS(ESI,pos.ion)m/z:376.0(M+1).
Step 2)3- (4-ethoxy-5-fluoropyrimidin-2-yl) -5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine
In a 50mL single neck flask was added 3- (4-chloro-5-fluoropyrimidin-2-yl) -5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine (0.060g,0.16mmol) and ethanol (10mL), followed by sodium ethoxide (0.016g,0.24mmol), and the mixture was stirred at room temperature overnight. The solvent was distilled off, water (20mL) was added to the residue, extraction was performed with ethyl acetate (20mL × 3), the organic phase was washed with water (10mL) and saturated brine (10mL) in succession, dried over anhydrous sodium sulfate, suction-filtered, and spin-dried, and the residue was purified by silica gel column chromatography (dichloromethane/ethyl acetate (v/v) ═ 100/1) to give a white solid (0.053g, 86.0%).
MS(ESI,pos.ion)m/z:386.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.77–8.68(m,2H),8.48(d,J=6.1Hz,1H),7.37(dd,J=13.0,6.0Hz,1H),7.28–7.19(m,2H),7.15(t,J=7.5Hz,1H),5.86(s,2H),4.65(q,J=7.0Hz,2H),1.46(t,J=7.0Hz,3H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.07(s),-134.73(s),-154.34(s).
Example 132- (4- (5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-yl) piperazin-1-yl) ethanol
Figure BDA0001623638800000571
Step 1) 5-fluoro-3- (5-fluoro-4- (piperazin-1-yl) pyrimidin-2-yl) -1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine
In a 50mL two-necked flask were added 3- (4-chloro-5-fluoropyrimidin-2-yl) -5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine (0.15g,0.40mmol), piperazine (0.17g,2.0mmol), N-dimethylformamide (10mL), and triethylamine (0.21mL,1.6mmol), and the mixture was heated to 80 ℃ for 6 hours. After cooling to room temperature, water (20mL) was added, extraction was performed with ethyl acetate (20mL × 3), the organic phase was washed with water (10mL) and saturated brine (10mL), dried over anhydrous sodium sulfate, suction-filtered, and spin-dried, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 100/1, 0.5% triethylamine) to give a pale yellow solid (0.15g, 88%).
Step 2)2- (4- (5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-yl) piperazin-1-yl) ethanol
In a 50mL single-necked flask were added 5-fluoro-3- (5-fluoro-4- (piperazin-1-yl) pyrimidin-2-yl) -1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine (0.21g,0.49mmol), acetonitrile (10mL), 2-bromoethanol (0.053mL,0.75mmol), and potassium carbonate (0.14g,1.0mmol), and the mixture was heated to 68 ℃ for reaction for 8 hours. After cooling to room temperature, water (20mL) was added, extraction was performed with ethyl acetate (20mL × 3), the organic phase was washed with water (20mL) and saturated brine (20mL), dried over anhydrous sodium sulfate, suction-filtered, and spin-dried, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 100/1, 0.5% triethylamine) to give a white solid (0.052g, 22.0%).
LC-MS(ESI,pos.ion)m/z:470.2(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.72(m,1H),8.48–8.34(m,2H),7.42–7.32(m,1H),7.19(m,3H),5.84(s,2H),4.50(s,1H),3.81(m,4H),3.56(t,J=4.1Hz,2H),2.61(m,4H),2.48(t,J=4.5Hz,2H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.11(s),-135.17(s),-144.90(s).
Example 142 methyl- ((5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-yl) amino) acetate
Figure BDA0001623638800000572
In a 50mL single-necked flask were added 3- (4-chloro-5-fluoropyrimidin-2-yl) -5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine (0.12g,0.32mmol), glycine methyl ester hydrochloride (0.060g,0.48mmol) and N, N-dimethylformamide (8mL), and triethylamine (0.14mL,1.1mmol) was further added, and the mixture was heated to 80 ℃ for reaction overnight. After cooling to room temperature, water was added, extraction was performed with ethyl acetate (30mL × 2), the organic phase was washed with water (20mL) and saturated brine (10mL), dried over anhydrous sodium sulfate, filtered with suction, and dried by spinning, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 100/1, 0.5% triethylamine) to give a pale yellow solid (0.11g, 80%).
Example 152- ((5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-yl) amino) acetic acid
Figure BDA0001623638800000581
Methyl 2- ((5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-yl) amino) acetate (0.10g,0.23mmol), tetrahydrofuran (2mL), water (2mL) and methanol (2mL) were added to a 50mL single-neck flask, and sodium hydroxide (0.019g,0.48mmol) was further added and stirred at room temperature for 2 hours. The solvent was distilled off, water (5mL) was added, pH was adjusted to 4 with 1mol/L hydrochloric acid, ethyl acetate was extracted (10mL × 2), the organic phase was washed successively with water (5mL) and saturated brine (5mL), dried over anhydrous sodium sulfate, suction filtered, spun dried, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20/1) to give a white solid (0.042g, 43%).
MS(ESI,pos.ion)m/z:415.0(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.70(s,1H),8.49(dd,J=8.6,2.3Hz,1H),8.31(d,J=3.3Hz,1H),8.13(s,1H),7.36(dd,J=13.2,5.9Hz,1H),7.27–7.11(m,3H),5.82(s,2H),4.13(d,J=5.7Hz,2H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.16(s),-135.48(s),-155.49(s).
Example 163- ((5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-yl) amino) propan-1-ol
Figure BDA0001623638800000582
In a 50mL single-necked flask were added 3- (4-chloro-5-fluoropyrimidin-2-yl) -5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine (0.10g,0.27mmol), N-dimethylformamide (6mL), aminopropanol (0.10mL,1.3mmol) and triethylamine (0.11mL,0.79mmol), and the mixture was heated to 100 ℃ for 5 hours. After cooling to room temperature, ethyl acetate (60mL) was added, followed by washing with water (20mL) and saturated brine (20mL), the organic phase was dried over anhydrous sodium sulfate, filtered with suction, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 60/1, 0.5% triethylamine) to give a white solid (0.065g, 59.0%).
MS(ESI,pos.ion)m/z:415.2(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.71(d,J=1.0Hz,1H),8.56(dd,J=8.6,2.7Hz,1H),8.24(d,J=3.7Hz,1H),7.83(t,J=5.3Hz,1H),7.42–7.33(m,1H),7.22(dd,J=15.8,7.7Hz,2H),7.15(t,J=7.4Hz,1H),5.83(s,2H),4.60(t,J=5.1Hz,1H),3.61(dd,J=13.0,6.5Hz,2H),3.54(dd,J=11.3,5.9Hz,2H),1.87–1.77(m,2H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.14(s),-135.51(s),-155.29(s).
Example 175-fluoro-3- (5-fluoro-4- ((tetrahydrofuran-3-yl) oxy) pyrimidin-2-yl) -1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine
Figure BDA0001623638800000591
In a 50mL two-necked flask was added tetrahydrofuran-3-ol (0.032mL,0.40mmol) and tetrahydrofuran (8.0mL), cooled to 0 deg.C, 60% sodium hydride (0.016g,0.40mmol) was added, and after half an hour 3- (4-chloro-5-fluoropyrimidin-2-yl) -5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine (0.10g,0.27mmol) was added, which was then warmed to room temperature and stirred for 4 hours. After quenching with water (20mL), ethyl acetate extraction (20mL × 2) was performed, the organic phase was washed with water (10mL) and saturated brine (10mL), dried over anhydrous sodium sulfate, suction-filtered, spin-dried, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 100/1, 0.5% triethylamine) to give a white solid (0.051g, 45.0%).
MS(ESI,pos.ion)m/z:428.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.75(d,J=2.8Hz,2H),8.50(dd,J=8.5,2.7Hz,1H),7.38(dd,J=14.3,6.6Hz,1H),7.24(dd,J=12.9,5.7Hz,2H),7.16(t,J=7.5Hz,1H),5.93–5.88(m,1H),5.87(s,2H),4.05–3.96(m,2H),3.91(m,1H),3.83(m,1H),2.40(m,1H),2.25–2.15(m,1H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.03(s),-134.66(s),-153.93(s).
Example 182- ((5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-yl) oxy) ethanol
Figure BDA0001623638800000592
Step 1)2- ((5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-yl) oxy) ethyl acetate
Figure BDA0001623638800000601
5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-ol (50mg,0.14mmol) was weighed into a 50mL flask, N-dimethylformamide (5.0mL), cesium carbonate (91mg,0.28mmol) and 2-bromoethyl acetate (31. mu.L, 0.28mmol) were added thereto, and the reaction flask was placed in an 80 ℃ oil bath and stirred for 10 hours. Cooled to room temperature, water was added, extracted with ethyl acetate (30mL × 3), dried over anhydrous sodium sulfate, filtered, spun-dried, and the residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 1/1) to give an off-white solid (44mg, 71.1%).
MS(ESI,pos.ion)m/z:444.1(M+1).
Step 2)2- ((5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-yl) oxy) ethanol
2- ((5-fluoro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-4-yl) oxy) ethyl acetate (330mg,0.744mmol) was weighed into a 50mL flask, to which ethanol (12.0mL), water (4.0mL) and a sodium hydroxide solution (305mg,2.29mmol,30 mass%) were added and stirred at room temperature for 1 hour. The solvent was removed by evaporation under reduced pressure, 1mol/L hydrochloric acid was added to the reaction flask to adjust pH to 5, ethyl acetate was extracted (30mL × 4), dried over anhydrous sodium sulfate, filtered, spun-dried, and the residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 1/2) to give a white solid (158mg, 53.0%).
MS(ESI,pos.ion)m/z:402.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.75(m,2H),8.53(dd,J=8.4,2.5Hz,1H),7.37(dd,J=14.3,7.5Hz,1H),7.28–7.19(m,2H),7.15(t,J=7.5Hz,1H),5.86(s,2H),5.00(t,J=5.3Hz,1H),4.69–4.59(m,2H),3.84(dd,J=9.6,4.9Hz,2H).
Example 194-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) -N-methylpyrimidine-5-carboxamide
Figure BDA0001623638800000602
Step 1) 4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carboxylic acid ethyl ester
Figure BDA0001623638800000611
5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine-3-carboxamidine (200mg,0.696mmol) and ethyl 2-cyano-3-ethoxyacrylate (240mg,1.42mmol) were dissolved in toluene (20mL), and then heated to 110 ℃ and stirred for 5 hours. The solvent was evaporated under reduced pressure and the crude product was used in the next step without purification.
MS(ESI,pos.ion)m/z:411.1(M+1).
Step 2) 4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carboxylic acid
Figure BDA0001623638800000612
Ethyl 4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carboxylate (2.3g,5.6mmol) was dissolved in methanol (20mL), and a 2mol/L solution of sodium hydroxide (14mL,28mmol) was added and stirred at room temperature overnight. Methanol was evaporated under reduced pressure, the pH was adjusted to 4 with 1mol/L dilute hydrochloric acid, filtered, and the filter cake was washed with water (10mL) and dried to give a yellow solid (2.01g, 94.0%).
MS(ESI,pos.ion)m/z:383.2(M+1).
Step 3) 4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) -N-methylpyrimidine-5-carboxamide
4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carboxylic acid (160mg,0.419mmol), methylamine hydrochloride (70mg,1.04mmol) were dissolved in N, N-dimethylformamide (50mL), followed by addition of 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (240mg,0.631mmol) and N, N-diisopropylethylamine (0.30mL,1.8mmol) at 0 deg.C, then allowed to stir at room temperature overnight. Water was added thereto, followed by extraction with ethyl acetate (50 mL. times.2), and the organic phases were combined, washed successively with water (40mL) and saturated brine (40mL), and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent under reduced pressure gave a crude product which was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 15/1) as a white solid (73mg, 44.1%).
MS(ESI,pos.ion)m/z:396.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.75(dd,J=57.6,27.8Hz,3H),8.17(t,J=99.3Hz,2H),7.23(dd,J=53.9,30.6Hz,4H),5.84(s,2H),2.76(d,J=22.5Hz,3H).
Example 204-amino-N-cyclopropyl-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carboxamide
Figure BDA0001623638800000621
4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carboxylic acid (500mg,1.308mmol), cyclopropylamine (0.23mL,3.3mmol) were dissolved in N, N-dimethylformamide (50mL), and then 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (745mg,1.96mmol) and N, N-diisopropylethylamine (0.90mL,5.4mmol) were added at 0 deg.C, followed by stirring overnight at room temperature. Water was added thereto, followed by extraction with ethyl acetate (50 mL. times.2), and the organic phases were combined, washed with water (40mL) and saturated brine (40mL) in this order, and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent under reduced pressure gave a crude product which was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 1/3) as a white solid (147mg, 26.7%).
MS(ESI,pos.ion)m/z:422.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.89–8.55(m,4H),8.07(s,2H),7.36(dd,J=13.3,6.0Hz,1H),7.30–7.09(m,3H),5.83(s,2H),2.91–2.76(m,1H),0.72(q,J=6.7Hz,2H),0.65–0.49(m,2H).
Example 21 methyl (4, 6-diamino-2- (1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) carbamate
Figure BDA0001623638800000622
Step 1)4- (bromomethyl) benzo [ d ] [1,3] dioxide
Benzo [ d ] [1,3] dioxo-4-ylmethanol (223mg,1.47mmol) was dissolved in dichloromethane (10mL) and phosphorus tribromide (0.17mL,1.8mmol) was added dropwise at 0 ℃. After the addition was completed, the mixture was stirred at room temperature for 3 hours. The reaction was quenched with saturated sodium bicarbonate solution and extracted with dichloromethane (50mL × 2). The organic phases were combined, washed successively with water (20 mL. times.2) and saturated brine (40mL), and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent under reduced pressure gave a white solid (285mg, 90.43%).
1 H NMR(400MHz,CDCl 3 )δ(ppm)6.84(dd,J=18.2,7.8Hz,3H),6.02(s,2H),4.49(s,2H).
Step 2)1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-3-iodo-1H-pyrazolo [3,4-b ] pyridine
5-fluoro-3-iodo-1H-pyrazolo [3,4-b ] pyridine (10.0g,38.0mmol) was dissolved in N, N-dimethylformamide (200mL), followed by addition of 4- (bromomethyl) benzo [ d ] [1,3] dioxide (6.0g,28mmol) and cesium carbonate (11g,33.8mmol), and stirring at room temperature for 2 hours. Extraction was performed with ethyl acetate (300 mL. times.2). The organic phases were combined, washed successively with water (100mL) and saturated brine (50mL), and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent under reduced pressure gave a yellow solid (8.2g, 74%).
MS(ESI,pos.ion)m/z:398.0(M+1).
Step 3)1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-1H-pyrazolo [3,4-b ] pyridine-3-carbonitrile
Cuprous cyanide (2.0g,22mmol) was dissolved in dimethylsulfoxide (50mL) under nitrogen, heated to 150 deg.C, and a solution of 1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-3-iodo-1H-pyrazolo [3,4-b ] pyridine (9.0g,23mmol) in dimethylsulfoxide (60mL) was added dropwise. Stirring was continued at 150 ℃ for 2.5 hours. Cooled to room temperature and quenched by addition of ammonia. Extraction was carried out with ethyl acetate (200 mL. times.3). The organic phases were combined, washed successively with water (100mL) and saturated brine (100mL), and dried over anhydrous sodium sulfate. Filtration, evaporation of the solvent under reduced pressure and purification of the crude product by column chromatography (petroleum ether/ethyl acetate (v/v) ═ 10/1) gave a yellow solid (2.6g, 39%).
MS(ESI,pos.ion)m/z:297.2(M+1).
Step 4)1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-1H-pyrazolo [3,4-b ] pyridine-3-carboxamidine
Sodium methoxide (710mg,13.1mmol) was weighed into a 50mL two-necked flask, and methanol (150mL) and 1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-1H-pyrazolo [3,4-b ] pyridine-3-carbonitrile (2.6g,8.8mmol) were added thereto, and the mixture was stirred at room temperature for 2 hours. Ammonium chloride (700mg,13.09mmol) and glacial acetic acid (2.0mL,35mmol) were added and the reaction was warmed to reflux overnight. Cooled to room temperature and the solvent removed by rotary evaporation. Water (100mL) and ethyl acetate (20mL) were added, and a 2mol/L aqueous sodium hydroxide solution was added to adjust the pH to 10. The mixture was extracted with ethyl acetate (100 mL. times.2), and the organic phases were combined, washed with water (50mL) and saturated brine (50mL) in this order, and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent under reduced pressure gave a yellow solid (1.17g, 43%).
MS(ESI,pos.ion)m/z:314.1(M+1).
Step 5)2- (1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-1H-pyrazolo [3,4-b ] pyridin-3-yl) -5- (phenylazo) pyrimidine-4, 6-diamine
1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-1H-pyrazolo [3,4-b ] pyridine-3-carboxamidine (1.17g,3.73mmol) was dissolved in N, N-dimethylformamide (50mL), and triethylamine (0.80mL,5.8mmol) was added. Heated to 85 ℃ under nitrogen, and then phenylazo malononitrile (960mg,5.64mmol) was added. The temperature was further raised to 100 ℃ and stirred for 4 hours. The reaction mixture was cooled to room temperature, and the reaction mixture was directly subjected to the next reaction without further purification.
MS(ESI,pos.ion)m/z:484.2(M+1).
Step 6)2- (1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine
10% Palladium on carbon (200mg) was added to the reaction solution in the previous step, and the mixture was stirred overnight under a hydrogen atmosphere. The filter cake was washed with methanol and the filtrate was collected. The solvent was evaporated under reduced pressure and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give a tan solid (600mg, 61%).
MS(ESI,pos.ion)m/z:395.2(M+1).
Step 7) (4, 6-diamino-2- (1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) carbamic acid methyl ester
2- (1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-4, 5, 6-triamine (600mg,1.52mmol) was dissolved in pyridine (10mL), and methyl chloroformate (0.50mL,6.5mmol) was added at 0 ℃. After stirring at 0 ℃ for half an hour, the mixture was allowed to warm to room temperature and stirred overnight. The solvent was evaporated under reduced pressure and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10/1) to give a yellow solid (310mg, 45.04%).
MS(ESI,pos.ion)m/z:453.2(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)(d,J=8.6Hz,2H),8.41and 8.09(2br s,1H),7.83(s,4H),6.91–6.64(m,3H),6.01(s,2H),5.81(s,2H),3.66(s,3H).
Example 22 methyl (4, 6-diamino-2- (1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) (methyl) carbamate
Figure BDA0001623638800000641
Methyl (4, 6-diamino-2- (1- (benzo [ d ] [1,3] dioxo-4-ylmethyl) -5-fluoro-1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) carbamate (340mg,0.752mmol) was dissolved in N, N-dimethylformamide (20mL) and 60% sodium hydride (75mg,1.88mmol) was added at 0 ℃. After stirring at this temperature for 20 minutes, iodomethane (0.120mL,1.93mmol) was added. Stirring was then continued for 30 minutes and then transferred to room temperature for 1 hour. Extraction was performed with ethyl acetate (50 mL. times.2). The organic phases were combined, washed successively with water (20mL) and saturated brine (20mL), and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent under reduced pressure gave the crude product as a preparative purification as a pale yellow solid (22mg, 6.28%).
MS(ESI,pos.ion)m/z:467.2(M+1);
1 H NMR(400MHz,DMSO-d 6 ) δ (ppm)8.80(s,2H),7.66(s,4H), 6.97-6.54 (m,3H),6.00(s,2H),5.81(s,2H),3.70 and 3.58(2s,3H),3.02(s,3H).
Example 23 (4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) (pyrrolidin-1-yl) methanone
Figure BDA0001623638800000651
4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carboxylic acid (0.050g,0.13mmol) and pyrrolidine (0.013mL,0.16mmol) were added to N, N-dimethylformamide (10mL), followed by 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (0.060g,0.16mmol) and triethylamine (0.055mL,0.40mmol) at 0 deg.C, and the reaction was allowed to warm to room temperature overnight. Water (50mL) was added for quenching, ethyl acetate extraction (30 mL. times.2) was performed, and the organic phases were combined, washed with water (50mL) and saturated brine (50mL) in this order, and dried over anhydrous sodium sulfate. Suction filtration was performed, the filtrate was spin-dried, and the residue was purified by silica gel column chromatography (dichloromethane/ethyl acetate (v/v) ═ 200/1, 0.5% triethylamine) to give a white solid (0.010g, 18%).
MS(ESI,pos.ion)m/z:436.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.81(dd,J=8.7,2.7Hz,1H),8.71(s,1H),8.38(s,1H),7.50–7.09(m,6H),5.83(s,2H),3.34–3.28(m,4H),1.94–1.78(m,4H).
Example 24 (4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidin-5-yl) (morpholine) methanone
Figure BDA0001623638800000652
4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carboxylic acid (0.058g,0.15mmol) and morpholine (0.016mL,0.18mmol) were added to N, N-dimethylformamide (10mL), followed by 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (0.069g,0.18mmol) and triethylamine (0.063mL,0.45mmol) at 0 deg.C, and the mixture was allowed to warm to room temperature for overnight. Quenched with water (50mL), extracted with ethyl acetate (30 mL. times.2), and the organic phase was washed with water (50mL) and saturated brine (50mL) and dried over anhydrous sodium sulfate. Suction filtration was performed, the solvent was evaporated, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 100/1) to give a white solid (0.062g, 91%).
MS(ESI,pos.ion)m/z:452.2(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.81(dd,J=8.7,2.7Hz,1H),8.72(s,1H),8.26(s,1H),7.46–7.06(m,6H),5.83(s,2H),7.68–7.58(m,4H),3.55–3.45(m,4H).
Example 254-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) -N, N-dimethylpyrimidine-5-carboxamide
Figure BDA0001623638800000661
4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyrid-3-yl) pyrimidine-5-carboxylic acid (0.10g,0.26mmol) and a tetrahydrofuran solution of dimethylamine (0.26mL,0.52mmol,2mol/L) were added to N, N-dimethylformamide (10mL), and 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (0.12g,0.32mmol) and triethylamine (0.11mL,0.79mmol) were added under ice-cooling, and the mixture was allowed to warm to room temperature for overnight reaction. Water was added thereto and the mixture was quenched (40mL), extracted with ethyl acetate (30 mL. times.2), and the organic phase was washed with water (40mL) and saturated brine (40mL) and dried over anhydrous sodium sulfate. The filtrate was suction filtered, the filtrate was spin-dried, and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 200/1, 0.5% triethylamine) to give a white solid (0.041g, 38%).
MS(ESI,pos.ion)m/z:410.1(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.81(dd,J=8.8,2.7Hz,1H),8.71(s,1H),8.25(s,1H),7.45–7.02(m,6H),5.83(s,2H),2.97(s,6H).
Example 262- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) -4-hydroxypyrimidine-5-carbonitrile
Figure BDA0001623638800000662
5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridine-3-carboxamidine (2.60g,9.05mmol) and ethyl 2-cyano-3-ethoxyacrylate (1.53g,9.04mmol) were added to ethanol (30mL) and then refluxed for 9 hours. The solvent was evaporated and the residue was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 100/1, 0.5% triethylamine) to give a pale yellow solid (2.16g, 65.5%).
MS(ESI,pos.ion)m/z:365.0(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.70–8.65(m,1H),8.57(dd,J=8.7,2.8Hz,1H),8.32(s,1H),7.40–7.32(m,1H),7.26–7.19(m,2H),7.15(t,J=7.4Hz,1H),5.79(s,2H).
Example 274-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carbonitrile
Figure BDA0001623638800000671
Step 1) 4-chloro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carbonitrile
2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) -4-hydroxypyrimidine-5-carbonitrile (1.30g,3.57mmol) and phosphorus oxychloride (1mL,10.73mmol) were added to toluene (10mL), and the reaction was refluxed for 3 hours. After cooling to room temperature, the solvent was evaporated, methylene chloride (80mL) was added to the residue, the mixture was washed with saturated sodium hydrogencarbonate (80mL), water (80mL) and saturated brine (80mL) in this order, and the organic phase was dried over anhydrous sodium sulfate. The residue was purified by silica gel column chromatography (dichloromethane/petroleum ether (v/v) ═ 1/1) to give a pale yellow solid (0.55g, 40%).
MS(ESI,pos.ion)m/z:383.0(M+1).
Step 2) 4-amino-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carbonitrile
4-chloro-2- (5-fluoro-1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b ] pyridin-3-yl) pyrimidine-5-carbonitrile (0.55g,1.4mmol) was added to a solution of ammonia in methanol (30mL,7mol/L) and the reaction was allowed to proceed overnight at 60 ℃ with a tube seal. Cooling to room temperature, evaporation of the solvent, washing of the residue with water, suction filtration, vacuum drying of the filter cake, slurrying with ethyl acetate (20mL) and suction filtration gave a white solid (0.45g, 86%).
MS(ESI,pos.ion)m/z:364.15(M+1);
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.77(d,J=8.7Hz,1H),8.73(s,2H),7.37(dd,J=13.8,7.2Hz,1H),7.27–7.19(m,2H),7.15(t,J=7.5Hz,1H),5.84(s,2H);
19 F NMR(376MHz,DMSO-d 6 )δ(ppm)-118.04(s),-134.76(s).
Biological activity assay
EXAMPLE A Effect of the Compounds of the invention on recombinant guanylate cyclase reporter cell lines
Constructing a stable and high-expression CHO-K1 cell line of Soluble Guanylate Cyclase (sGC): synthesizing Rat sGC alpha 1 and Rat sGC beta 1 genes, constructing pcDNA3.1(+) -Rat sGC alpha 1 and pcDNA3.1/Hygro (+) -Rat sGC beta 1 plasmids, co-transfecting the plasmids to CHO-K1 cells, and screening out a positive clone cell strain stably expressed by CHO-K1-Rat sGC by using a Cisbio cGMP HTRF detection kit.
Determination of the Activity of the Compounds on CHO-K1-Rat sGC monoclonal cell lines: culturing CHO-K1-Rat sGC monoclonal cell strain to 70-90% fusion degree, after TrypLE digestion treatment, suspending the cells in complete culture medium, inoculating the cells in 384-well (low volume tissue culture medium) cell culture plate with inoculation density of 7.5 thousand/well (in 25 mu L of complete culture medium), and inoculating 5% CO at 37 ℃ and 5% 2 Culturing for 20 hours; dissolving and diluting a compound to be detected into 10 concentration gradients (2X working concentration) by using DMSO (dimethyl sulfoxide), taking out a 384-hole cell culture plate, inverting and centrifuging 200g of the plate for 3 seconds by using RT (reverse transcription factor), removing a culture medium, adding the compound to be detected and corresponding test solution with each concentration gradient into each cell hole according to the detection steps of a Cisbio cGMP HTRF detection kit, collecting data by using an Envision HTRF detector, and calculating the EC (EC) of each compound for activating soluble guanylate cyclase 50 The value is obtained.
TABLE 2 Effect of the Compounds of the invention on recombinant guanylate cyclase reporter cell lines
Example numbering EC 50 (μM) Example numbering EC 50 (μM)
Example 2 0.23 Example 13 0.63
Example 3 0.21 Example 16 0.60
Example 4 0.35 Example 20 0.062
Example 5 0.69 Example 23 0.58
Example 6 0.11 Example 24 0.57
Example 7 0.13 Example 25 0.66
Example 11 0.52 Example 27 0.32
And (4) experimental conclusion:
as can be seen from the data in Table 2, the compounds of the present invention all activated the activity of CHO-K1-Rat sGC monoclonal cell line well, i.e., the compounds of the present invention activated the recombinant guanylate cyclase reporter cell line.
Example B pharmacokinetic experiments with Compounds of the invention
Preparation of a solution of a test compound: the compounds to be tested were formulated in solutions of 5% dimethyl sulfoxide, 5% Solutol HS 15 and 90% normal saline for oral and intravenous administration.
Taking 190-250g male SD rats, randomly dividing the rats into two groups, wherein each group comprises 3 rats, one group is used for intravenous injection of the compound to be tested, the dose is 1.0mg/kg, and the other group is used for oral administration of the compound to be tested, and the dose is 2.5 or 5.0 mg/kg; blood was collected at time points 0.0833,0.25,0.5,1.0,2.0,4.0,7.0 and 24 hours after dosing. A standard curve of the appropriate range was established based on the sample concentration, and the concentration of the test compound in the plasma sample was determined in MRM mode using LC-MS/MS model AB SCIEX API 4000. According to the drug concentration-time curve, pharmacokinetic parameters are calculated by adopting a WinNonLin 6.3 software non-compartmental model method.
TABLE 3 pharmacokinetic data for some of the compounds of the invention
Figure BDA0001623638800000681
And (4) experimental conclusion:
the compound of the invention has better in vivo pharmacokinetic properties, such as higher bioavailability and the like.
In the description herein, references to the description of the term "one embodiment," "some embodiments," "an example," "a specific example," or "some examples," etc., mean that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the invention. In this specification, the schematic representations of the terms used above are not necessarily intended to refer to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, various embodiments or examples and features of different embodiments or examples described in this specification can be combined and combined by one skilled in the art without contradiction.
Although embodiments of the present invention have been shown and described above, it is understood that the above embodiments are exemplary and should not be construed as limiting the present invention, and that variations, modifications, substitutions and alterations can be made to the above embodiments by those of ordinary skill in the art within the scope of the present invention.

Claims (7)

1. A compound which is a compound represented by formula (II) or a pharmaceutically acceptable salt of a compound represented by formula (II),
Figure FDA0003663160570000011
wherein the content of the first and second substances,
each R a And R b Independently is H or D;
each R 6 And R 7 Independently is H or D;
each R 3 And R 4 Independently H, D, piperidinyl, piperazinyl, morpholinyl, -NR 8a R 8b OR-OR 9 (ii) a The R is 3 And R 4 Unsubstituted or optionally independently substituted by 1R x Substitution;
each R 8a And R 8b Independently H, D or hydroxy C 1-4 An alkyl group;
each R 9 Independently is H or D;
R x is hydroxy C 1-4 An alkyl group;
R 5 is F, Cl, Br, I, CN, -NR 8c R 8d 、-C(=O)NR 8c R 8d 、-N(R 8e )C(=O)R 10a or-C (═ O) R 10a (ii) a The R is 5 Unsubstituted or optionally substituted by 1 or 2R y Substitution;
R 8e is H or D;
R 8c h, D, methyl, ethyl, propyl or butyl;
R 8d is methyl, ethyl, propyl, butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuryl, tetrahydropyranyl, piperidinyl, morpholinyl, cyclopropylmethyl, cyclobutylmethyl, cyclohexylethyl, tetrahydropyranylmethyl, tetrahydropyranylethyl, tetrahydrofuranylmethyl, tetrahydrofuranylethyl, piperidinylmethyl, piperidinylethyl, morpholinylmethyl or pyrazolylethyl;
each R 10a Independently is tetrahydropyranyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, morpholinyl, piperidinyl, or piperazinyl;
each R y Independently is hydroxy or C 1-4 An alkyl group.
2. The compound of claim 1, wherein each R 8a And R 8b Independently H, D, hydroxymethyl, hydroxyethyl or hydroxypropyl.
3. The compound of claim 1, wherein,
R x is hydroxymethyl or hydroxyethyl;
each R y Independently a hydroxyl, methyl, ethyl, propyl or butyl group.
4. The compound of claim 1, comprising the structure of one of the following or a pharmaceutically acceptable salt of the structure:
Figure FDA0003663160570000021
Figure FDA0003663160570000031
5. a pharmaceutical composition comprising a compound of any one of claims 1-4; which further comprises pharmaceutically acceptable adjuvants.
6. Use of a compound according to any one of claims 1 to 4 or a pharmaceutical composition according to claim 5 for the preparation of a medicament for the treatment and/or prophylaxis of: heart failure, angina pectoris, hypertension, pulmonary hypertension, ischemia, renal disease, thromboembolic disease, male sexual dysfunction, systemic sclerosis, sickle cell anemia, cardiac achalasia, fibrotic disease and/or arteriosclerosis.
7. Use of a compound according to any one of claims 1 to 4 or a pharmaceutical composition according to claim 5 for the manufacture of a medicament for use as a soluble guanylate cyclase stimulator.
CN201810315504.0A 2017-04-11 2018-04-10 Pyrazolopyridines and uses thereof Active CN108690016B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710232280 2017-04-11
CN2017102322802 2017-04-11

Publications (2)

Publication Number Publication Date
CN108690016A CN108690016A (en) 2018-10-23
CN108690016B true CN108690016B (en) 2022-08-12

Family

ID=63845566

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810315504.0A Active CN108690016B (en) 2017-04-11 2018-04-10 Pyrazolopyridines and uses thereof

Country Status (1)

Country Link
CN (1) CN108690016B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112794848B (en) * 2019-11-13 2024-01-16 广东东阳光药业股份有限公司 N-ring-aminopyrimidine derivatives and use thereof
TW202210472A (en) 2020-06-05 2022-03-16 美商奇奈特生物製藥公司 Inhibitors of fibroblast growth factor receptor kinases
WO2023245611A1 (en) * 2022-06-24 2023-12-28 四川大学华西医院 Small molecule compound with ttk inhibitory activity, preparation method therefor, and use thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1665811A (en) * 2002-05-08 2005-09-07 拜耳医药保健股份公司 Carbamate-substituted pyrazolopyridines
CN102939289A (en) * 2010-05-26 2013-02-20 拜耳知识产权有限责任公司 Substituted 5-fluoro-1h-pyrazolopyridines and use thereof
US20130158028A1 (en) * 2010-06-25 2013-06-20 Bayer Intellectual Property Gmbh Use of stimulators and activators of soluble guanylate cyclase for treating sickle-cell anemia and conserving blood substitutes

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MA34249B1 (en) * 2010-05-26 2013-05-02 Bayer Ip Gmbh USE OF SGC STIMULATORS, SGC ACTIVATORS, ONLY AND IN ASSOCIATION WITH PDE5 INHIBITORS FOR THE TREATMENT OF SYSTEMIC SCLEROMERMIA
DE102010031666A1 (en) * 2010-07-22 2012-01-26 Bayer Schering Pharma Aktiengesellschaft Carbamate-substituted diaminopyrimidines and their use

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1665811A (en) * 2002-05-08 2005-09-07 拜耳医药保健股份公司 Carbamate-substituted pyrazolopyridines
CN102939289A (en) * 2010-05-26 2013-02-20 拜耳知识产权有限责任公司 Substituted 5-fluoro-1h-pyrazolopyridines and use thereof
US20130158028A1 (en) * 2010-06-25 2013-06-20 Bayer Intellectual Property Gmbh Use of stimulators and activators of soluble guanylate cyclase for treating sickle-cell anemia and conserving blood substitutes

Also Published As

Publication number Publication date
CN108690016A (en) 2018-10-23

Similar Documents

Publication Publication Date Title
TWI565708B (en) Substituted annellated pyrimidines and use thereof
JP6114189B2 (en) Fused pyrimidines and triazines and their use for the treatment and / or prevention of cardiovascular disorders
US20240034729A9 (en) Heterocyclic compound, intermediate, preparation method therefor and application thereof
CN110511219B (en) Phenyl-substituted dihydronaphthyridine compound and application thereof
US20150274754A1 (en) Substituted annulated triazines and use thereof
JP2015535271A (en) Carboxy-substituted imidazo [1,2-a] pyridinecarboxamide and uses thereof
CN108690016B (en) Pyrazolopyridines and uses thereof
AU2013312931A1 (en) Alkoxy pyrazoles as soluble guanylate cyclase activators
WO2021218755A1 (en) Shp2 inhibitor, and composition and use thereof
CN108727340B (en) Fluorine substituted indazoles and use thereof
TW201240971A (en) Fused heterocyclic compounds
WO2020215998A1 (en) Pyrimido five-membered heterocyclic compound and use thereof as mutant idh2 inhibitor
CN107428682B (en) Amide derivatives, preparation method and medical application thereof
JP2017526685A (en) Substituted fused pyrimidines and uses thereof
CN112794848A (en) N-cyclo-aminopyrimidine derivatives and their use
JP2017529338A (en) Amino-substituted cyclic pyrimidines and uses thereof
TW202333663A (en) Rxfp1 agonists
WO2023237085A1 (en) Hpk1 inhibitor and medical use thereof
WO2022222911A1 (en) Pyrimidone compound and use thereof

Legal Events

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

Address after: 523808 No.1, Gongye North Road, Songshanhu Park, Dongguan City, Guangdong Province

Patentee after: Guangdong Dongyangguang Pharmaceutical Co.,Ltd.

Address before: 523808 No. 1 Industrial North Road, Songshan Industrial Park, Songshan, Guangdong, Dongguan, Hubei

Patentee before: SUNSHINE LAKE PHARMA Co.,Ltd.

CP03 Change of name, title or address