WO2023051302A1 - 具有细胞周期蛋白依赖性激酶抑制活性的杂环化合物及其制备方法和医药用途 - Google Patents

具有细胞周期蛋白依赖性激酶抑制活性的杂环化合物及其制备方法和医药用途 Download PDF

Info

Publication number
WO2023051302A1
WO2023051302A1 PCT/CN2022/119618 CN2022119618W WO2023051302A1 WO 2023051302 A1 WO2023051302 A1 WO 2023051302A1 CN 2022119618 W CN2022119618 W CN 2022119618W WO 2023051302 A1 WO2023051302 A1 WO 2023051302A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
cycloalkyl
group
alkoxy
aryl
Prior art date
Application number
PCT/CN2022/119618
Other languages
English (en)
French (fr)
Inventor
闫旭
陈振华
刘国标
谷晓成
尚飞
杜佩金
Original Assignee
中国医药研究开发中心有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 中国医药研究开发中心有限公司 filed Critical 中国医药研究开发中心有限公司
Priority to CN202280006843.5A priority Critical patent/CN116390921A/zh
Publication of WO2023051302A1 publication Critical patent/WO2023051302A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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

Definitions

  • the invention belongs to the technical field of medicine, and specifically relates to a pyrimidopyridine compound, a preparation method thereof, a pharmaceutical composition containing the same, and its use as a cyclin-dependent kinase (CDK) inhibitor in the treatment of abnormal cell growth such as cancer use in medicines.
  • the compounds of the present invention can inhibit the cyclin-dependent kinases CDK2/cyclin E1 (CCNE1), CDK6/cyclin D1, CDK4/cyclin D3, and therefore can be used for the treatment of cancer, such as HR+/HER2- metastatic breast cancer, ovarian cancer, etc.
  • CDKs Cyclin-dependent kinases
  • CDKs belong to the serine/threonine protein kinase family and are key proteins in the regulation of the cell cycle.
  • CDKs mainly include CDK1-15, etc., which play a role by combining with the corresponding cell cycle protein (cyclin) to form a stable form of CDK/cyclin complex.
  • cyclin cell cycle protein
  • cells contain endogenous CDK and CDK/cyclin complex inhibitor (CKI), which together constitute the regulatory network of the cell cycle and are strictly controlled during cell division.
  • CKI CDK/cyclin complex inhibitor
  • CDK4 and CDK6 are very similar, and they can combine with the three subtypes of cyclin D (cyclinD1/D2/D3) to form a complex, which makes the retinoblastoma protein (Retinoblastoma protein, Rb) Phosphorylation of a series of substrates drives cell cycle progression.
  • the phosphorylated Rb protein releases the transcription factor E2F bound to it, etc.
  • E2F activates and transcribes a series of genes, making the cell enter the S phase and start DNA replication (Kato et al., Genes Dev, 1993, 7(3), 331-342; Dyson et al., Genes Dev, 1998, 12(15), 2245-2262).
  • the activity of CDK4/6 is also negatively regulated by the INK4 family (including p16 INK4A , p15 INK4B , p18 INK4C , p19 INK4D ).
  • INK4 can competitively bind with CDK and inhibit cyclin D-CDK4/6 Complex formation (Baker et al., Genes & Cancer, 2012, 3(11-12), 658-669).
  • CyclinD-CDK4/6-p16-Rb pathway has been found in most human cancers, which can promote the rapid development of G1 phase and lead to abnormal cell proliferation.
  • the main reasons include overexpression of cyclin D1 caused by gene rearrangement or gene amplification (Bergsagel et al., Blood, 2005, 106(1), 296-303); p16INK4a gene deletion, point mutation, or DNA methylation lead to p16INK4a inactivation (Ruas et al., Biochim Biophys Acta, 1998, 1378(2), F115-177); CDK4/6 gene amplification or point mutation (Hamilton et al., Cancer Treatment Reviews, 2016, 45, 129-138). Targeted intervention based on the abnormality of this pathway makes CDK4/6 one of the popular anti-tumor targets.
  • Flavopiridol the first-generation broad-spectrum CDK inhibitor
  • Flavopiridol can inhibit CDK1/2/4/6/7/9 at the same time, but due to its severe side effects, it may cause adverse reactions such as fatigue, diarrhea, and bone marrow suppression, and finally failed Can be successfully applied clinically (Senderowicz et al., Journal of Clinical Oncology, 1998, 16(9), 2986-2999).
  • Palbociclib, Ribociclib and Abemaciclib have been approved by the FDA for the first-line or second-line treatment of advanced breast cancer, and one drug, Lerociclib, is in clinical development.
  • Palbociclib is the first CDK4/6 inhibitor approved by the FDA. It is an oral pyridine compound that inhibits the proliferation of ER+ breast cancer cells by reducing the phosphorylation of Rb protein, preventing the cell cycle from G1 phase to S phase.
  • Palbociclib combined with letrozole or Fulvestrant can significantly prolong the progression-free survival of patients with reliable safety (Cristofanilli et al., Lancet Oncol, 2016, 17(4), 425- 439; Richard et al., Lancet Oncology, 2015; Finn et al., N Engl J Med, 2016, 375(20), 1925-1936; Nicholas et al., N Engl J Med, 2015, 373(3), 209-219 ).
  • Ribociclib (Kisqali, LEE011) and Abemaciclib (Verzenio, LY2835219) are oral reversible CDK4/6 inhibitors, which can cause G1 phase arrest by inhibiting Rb phosphorylation.
  • the IC 50 values of ribociclib for CDK4/6 inhibition are 10nM and 39nM, respectively, while the sensitivity to other CDK family members is low (the IC 50 values of CDK1 and CDK2 are both greater than 50mM) (Sherr et al., Cancer Discovery, 2016, 6( 4), 353-367). After 1-4 hours of oral administration, the Cmax value can be reached in the plasma; after 8 days of continuous administration, the stable state of blood is reached.
  • the binding rate of ribociclib to human plasma protein is 70%, and it can be decomposed by CPY3A4 (a weak inhibitor) in vivo, and the main products are N-hydroxy and N-demethyl derivatives.
  • Abemaciclib is more selective for CDK4/6 kinases with IC 50 values of 2 nM and 5 nM, respectively, while for CDK1 and CDK2 inhibition with IC 50 values >500 nM.
  • Abemaciclib is a phenylpyrimidine compound that is structurally related to palbociclib and ribociclib.
  • Abemaciclib exhibited a broader inhibitory effect and was able to inhibit Dyrk, PIM, HIPK and CAMK kinase families (Ki ⁇ 10nM) simultaneously (Chen et al., Molecular Cancer Therapeutics, 2016, 15(10), 2273-2281).
  • the in vivo pharmacokinetic data show that it reaches the peak within 4-24 hours after oral administration, and reaches a steady state after 5 days of continuous administration.
  • the binding rate of Abemaciclib to human plasma protein is about 96%, and it can be decomposed by CPY3A4 in vivo, and the main decomposition product is N-deethyl derivative.
  • the introduction of deuterium atoms makes the compound have stronger metabolic stability, and the metabolic stability is improved by 11-45%, which is manifested as a higher half-life.
  • the drug is approved in combination with Fluvestrant for the treatment of ER+/HER2- advanced or metastatic breast cancer.
  • Abemaciclib combined with gemcitabine also showed synergistic antitumor activity, and also had inhibitory effects on various types of tumors, including mantle cell lymphoma (MCL), colorectal cancer, lung cancer, glial Blastoma and acute myeloid leukemia (AML).
  • MCL mantle cell lymphoma
  • AML acute myeloid leukemia
  • CDK4/6 inhibitors combined with endocrine therapy can bring significant clinical benefits to certain types of breast cancer patients, some patients are still ineffective (10-20%), and 70-80% of tumor patients are treated after 12 -Drug resistance will develop after 36 months (Tripathy et al., Lancet Oncol, 2018, 19(7), 904-915).
  • CDK4/6 therapy resistance includes Rb loss, CCND1 overexpression, p16 amplification, overactivation of the CCNE1-CDK2 complex (eg, CCNE1 is high in breast and ovarian cancer cell line models CDK2 expression), CDK2 bypass activation, CDK4/6 activity increase, CDK7 overexpression, etc., all of which may be related to CDK4/6 treatment resistance (Taylor Harding et al., Oncotarget, 2014, 6(2), 696-714; Herrera-Abreu et al., Cancer Res, 2016, 76(8), 2301-2313).
  • Cyclin E (cyclin E) is a protein encoded by CCNE1, which begins to express in the middle of G1, and gradually degrades and disappears after the expression level reaches the peak in G1-S phase.
  • Cyclin E binds to CDK2 to form a complex, and phosphorylates the downstream substrates Rb, CDC6, NPAT and P107 to make the cell enter the S phase of DNA synthesis; Cyclin E plays a role in maintaining chromosome stability and regulating the cell spindle and centrosome cycle Important role; Cyclin E is highly expressed in a variety of malignant tumors, and CCNE1 gene amplification or overexpression is also related to poor prognosis of a variety of tumors, such as endometrioma, gastric cancer, ovarian cancer (George et al., Clinical cancer research, 2017, 23(7), 1862-1874; Nakayama et al., Cancer, 2010, 116(11), 2621-2634; KENTARO et al., Int J Oncol, 2015, 48(2), 506-516; Ooi et al., Human Pathology, 2016, S0046817716303082) etc.
  • CDK2 plays a key role in cell cycle regulation by interacting with chaperone proteins and phosphorylation activation, and participates in a series of biological processes, such as DNA damage, intracellular transport, protein degradation, signal transduction, DNA and RNA metabolism and translation et al. (Tadesse et al., Drug Discovery Today, 2020, 25(2), 406-413). CDK2 is underexpressed in most normal tissues (McCurdy et al., Oncogene, 2016). In dividing cells, CDK2 is a key cell cycle regulator, which is activated from the late G1 phase and continues to express throughout the S phase.
  • CDK2 binds CCNE1 or E2 and Cyclin A2, is activated by phosphorylation of cyclin complexes (CDK7, MAT1, cyclin H), and can be negatively regulated by CDC25A dephosphorylation.
  • CDK2-cyclinE and CDK4/6-cyclin-D co-phosphorylate Rb, causing Rb to release E2F and initiate the transcription of cell cycle regulatory genes.
  • CDK2 also regulates the phosphorylation of other proteins, linking them to the cell cycle. For example, phosphorylation of Smad3 by CDK2-cyclin-E limits its transcriptional activity, ultimately slowing cell cycle progression (Matsuura et al., Nature, 2004, 430(6996), 226-231).
  • CDK2 also phosphorylates prereplication complex proteins, which are required to initiate DNA synthesis.
  • CDC6 which is an essential protein for loading minichromosome maintenance (MCM) protein onto DNA and MCM helicase protein, and initiating DNA replication
  • CDK2 also plays an important role in regulating centrosome duplication by targeting phosphorylated centrosome proteins such as nucleophospholipid (Npm) and cp110, releasing centrioles and then maintaining centriole duplication (Adon et al., Molecular & Cellular Biology, 2010, 30 (3), 694-710; Hu et al., Cancer Research, 2015, 75(10), 2029-2038).
  • the protein structure of CDK2 is similar to that of most protein kinases, and it is folded in a "double leaf shape".
  • the smaller N-terminal region is mainly composed of ⁇ -sheets, including 5 long antiparallel ⁇ -strands and a C-helix.
  • the C-alpha helix contains the sequence PSTAIRE and is required for cyclin binding.
  • T-loop binds to the Ser/Thr (phosphorylated receptor) region of the substrate, activates phosphorylation, and plays a role in cell cycle regulation.
  • the N-terminal and C-terminal are composed of a flexible hinge region (residues 81(Glu)-84 (His)) connection, this region forms a deep cleft, the ATP binding site (Pavletich et al., Journal of Molecular Biology, 1999, 287(5), 821-828; Malumbres et al., Chemical Reviews, 2014, 15( 6), 122; Honda et al., Embo Journal, 2005, 24(3)).
  • CDK2 activation and inhibition are regulated by several mechanisms: In the absence of mitogen signaling, CDK2 is inactive. In the late G1 phase, CDK2 activity begins to increase, which is due to, first, E2F-mediated transcription of the CCNE gene, and its protein product binds and activates CDK2; full activation of the CDK2-cyclin E or A complex requires phosphorylation of Thr160 by CAK Furthermore, Wee1 and Myt1 kinases can inhibit the phosphorylation of Thr14 and Tyr15 respectively, and the dephosphorylation of these residues by the CDC25 protein phosphatase family can reactivate CDK2; in addition, the CDK inhibitory protein family Cip and Kip , can bind to CDK2 to inactivate it, and cyclins E and A can be degraded by ubiquitin ligase-mediated ubiquitination (Solomon et al., Journal of Medicinal Chemistry, 2018).
  • CDK2 can also bind cyclin A, participate in the progression of the entire S phase, and regulate DNA damage repair. Following DNA damage, the DNA damage response (DDR) arrests cells at the G1/S junction to repair damaged DNA and maintain genomic fidelity in daughter cells.
  • DDR DNA damage response
  • accumulation of p53 leads to upregulation of p21Cip1/Waf1 transcription, followed by cell cycle arrest through repression of cyclin-D1-CDK4/6 and cyclinE-CDK2 (Shieh et al., Genes & Development, 2000, 14(3), 289-300).
  • the second mechanism is to target CDC25A for degradation.
  • WEE1 prevents cells from entering S phase by continuously inhibiting the phosphorylation of CDK2 protein Thr14 and Tyr15 (Mailand et al., Science, 2000, 288(5470), 1425-1429).
  • the target protein of CDK2, FOXO1 plays an important role in dsDNA breakage triggering DNA damage, thereby inducing apoptosis.
  • CDK2 no longer phosphorylates FOXO1, allowing FOXO1 to exert transcriptional activity, and promotes cell apoptosis by up-regulating the expression of various pro-apoptotic proteins (FasL, TRAIL, and Bim) (Huang et al. People, Science, 2006, 314(5797), 294-297; Huang et al., Cell Cycle, 2007, 6(8), 902-906).
  • CDK4/6 inhibitors have become the first-line treatment for specific types of breast cancer, some types of breast cancer are naturally insensitive to CDK4/6 inhibitors, such as triple-negative breast cancer (TNBC).
  • TNBC triple-negative breast cancer
  • drug resistance to CDK4/6 was also found in ER+Her2- breast cancer.
  • Cyclin E is highly expressed in a variety of malignant tumors, including tumors insensitive to CDK4/6 inhibitors (TNBC) and ovarian cancer; the complex formed by CDK2 and CyclinE also plays an important role in CDK4/6 drug resistance ;Targeting CDK2 can delay the treatment resistance produced by CDK4/6 inhibitor molecules, and further exert curative effect on drug-resistant patients; while in the compounds targeting CDK2/4/6 to overcome CDK4/6 drug resistance, no effect has been seen Of the approved drugs, only Pfizer's PF-06873600 has entered clinical phase II research. Therefore, it is an urgent problem to develop CDK2/4/6 inhibitors with high selectivity and low drug toxicity to overcome CDK4/6 drug resistance.
  • CDK cyclin-dependent kinase
  • the object of the present invention is to provide a compound represented by general formula (I) or its stereoisomer, tautomer, mesoform, racemate, enantiomer, diastereomer Enantiomers, or mixtures thereof, or pharmaceutically acceptable salts thereof,
  • Ring A is selected from heterocyclyl optionally further selected from halogen, amino, nitro, cyano, hydroxyl, mercapto, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy , Cycloalkyl, heterocyclyl, aryl, heteroaryl are substituted by one or more groups;
  • Ring B is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R is selected from alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is optionally further selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, Substituted by one or more groups of alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl;
  • R is selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, Aryl and heteroaryl are optionally further selected from halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl One or more groups of radical, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl; or,
  • R and R together with their attached nitrogen and sulfur atoms form a heterocyclyl or heteroaryl, which is optionally further selected from the group consisting of halogen, amino, nitro, cyano, oxygen
  • halogen amino, nitro, cyano
  • oxygen One or more of substituent, hydroxyl, mercapto, carboxyl, ester, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl group substitution;
  • Each R is independently selected from halogen, amino, nitro, cyano, hydroxyl, mercapto, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, the alkyl, Alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl are optionally further selected from halogen, amino, nitro, cyano, hydroxyl, mercapto, carboxyl, ester, oxo, alkyl, One or more groups of alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl; or,
  • R is selected from hydrogen, halogen, amino, nitro, cyano, hydroxyl, mercapto, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -C(O) Ra , -O(O)CR a , -C(O)OR a , -C(O)NR a R b , -NHC(O)R a , -S(O) m R a , -S(O) m NR a R b and -NHS(O) m R a , the alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl are optionally further selected from halogen, amino, nitro, cyano One or more groups of radical, hydroxyl, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, cycl
  • R is selected from hydrogen, halogen, amino, nitro, cyano, hydroxyl, mercapto, alkyl, alkoxy, acyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -C(O)R a , -O(O)CR a , -C(O)OR a , -C(O)NR a R b , -NHC(O)R a , -S(O) m R a , -S(O) m NR a R b and -NHS(O) m R a , the alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl are optionally further selected from halogen, amino, nitro One or more of cyano, hydroxyl, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy
  • R a and R b are each independently selected from hydrogen, halogen, hydroxyl, cyano, amino, carboxyl, ester, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and Heteroaryl, wherein the alkyl, alkoxyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally further selected from the group consisting of halogen, amino, nitro, One or more of cyano, hydroxyl, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl group replacement;
  • R a and R b form a cycloalkyl or heterocyclic group together with the atoms they are connected to, and the cycloalkyl or heterocyclic group is optionally further selected from the group consisting of halogen, amino, nitro, cyano, oxo, One or more of hydroxyl, mercapto, carboxyl, ester, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are substituted;
  • n 0, 1, 2;
  • p 0, 1, 2, 3 or 4.
  • Ring A is selected from 3 to 12 membered monocyclic heterocyclic groups, spiro heterocyclic groups, condensed heterocyclic groups or bridged heterocyclic groups, preferably 5 to 7 membered monocyclic heterocyclic groups, 7 to 10 membered spiro heterocyclic groups, 7 to 10-membered fused heterocyclic group and 7 to 10-membered bridged heterocyclic group, more preferably pyrrolidinyl, piperidinyl, piperazinyl, said heterocyclic group is optionally further selected from halogen, amino, nitro, cyano radical, hydroxyl, mercapto, oxo, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkyl, C 1 -C 6 haloalkoxy, C 3 -C 6 ring
  • the compound represented by general formula (I) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer Isomers, diastereoisomers, or mixtures thereof, or pharmaceutically acceptable salts thereof which are compounds represented by general formula (II) or stereoisomers, tautomers, and mesomers thereof body, racemate, enantiomer, diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • ring B, R 1 , R 2 , R 3 , R 4 , R 5 , and p are as defined in general formula (I).
  • Ring B is selected from C 3 -C 7 cycloalkyl, 4 to 7 membered heterocyclic group, C 6 -C 10 aryl, 5 to 10 membered heteroaryl, more preferably
  • the compound represented by general formula (I) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer Isomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof which are compounds represented by general formula (III) or stereoisomers, tautomers, meso body, racemate, enantiomer, diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • R 1 , R 2 , R 3 , R 4 , R 5 , and p are as defined in general formula (I).
  • Each R 3 is independently selected from halogen, amino, cyano, hydroxyl, mercapto, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 7 cycloalkyl, 4 to 7 membered Heterocyclyl, the C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 7 cycloalkyl, 4 to 7 membered heterocyclyl are optionally further selected from halogen, amino, nitr One or more of group, cyano group, hydroxyl group, mercapto group, carboxyl group, ester group, oxo group, alkyl group, alkoxy group, haloalkyl group, haloalkoxy group, cycloalkyl group, heterocyclyl group, aryl group, heteroaryl group Substituted by a group; preferably halogen, hydroxyl, C 1 -C 6 alkyl, C 1 -C 6
  • p 0, 1, 2 or 3; preferably 1 or 2.
  • the compound represented by general formula (I), general formula (II) or general formula (III) or its stereoisomers and tautomers according to the present invention mesoform, racemate, enantiomer, diastereoisomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
  • R 1 is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 5-7 membered heterocyclyl, C 6 -C 10 Aryl and 5 to 10 membered heteroaryl, preferably C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl; preferably C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, 5 to 7 membered heterocyclyl;
  • the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl or C 3 -C 6 cycloalkyl are optionally further selected from halogen, amino, nitro, cyano, Hydroxy, mercapto, carboxyl, ester, oxo, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C Substituted by one or more groups of 6 cycloalkyl, 5 to 7 membered heterocyclyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy;
  • the 5 to 7 membered heterocyclic group, C 6 -C 10 aryl group and 5 to 10 membered heteroaryl group are optionally further selected from halogen, amino, nitro, cyano, hydroxyl, mercapto, carboxyl, ester, Oxo, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 5 to 7 membered Substitution by one or more groups of heterocyclic group, C 1 -C 6 haloalkyl group, C 1 -C 6 alkoxy group, C 6 -C 10 aryl group, 5-10 membered heteroaryl group.
  • R 2 is selected from hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 5 to 7 membered heterocyclyl, C 6 - C 10 aryl and 5 to 10 membered heteroaryl, preferably hydrogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl; preferably hydrogen and C 1 -C 6 alkyl;
  • the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl or C 3 -C 6 cycloalkyl are optionally further selected from halogen, amino, nitro, cyano, Hydroxy, mercapto, carboxyl, ester, oxo, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C Substituted by one or more groups of 6 cycloalkyl, 5 to 7 membered heterocyclyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy;
  • the 5 to 7 membered heterocyclic group, C 6 -C 10 aryl group and 5 to 10 membered heteroaryl group are optionally further selected from halogen, amino, nitro, cyano, hydroxyl, mercapto, carboxyl, ester, Oxo, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 5 to 7 membered Substitution by one or more groups of heterocyclic group, C 1 -C 6 haloalkyl group, C 1 -C 6 alkoxy group, C 6 -C 10 aryl group, 5-10 membered heteroaryl group.
  • R 4 is selected from hydrogen, halogen, amino, nitro, cyano, hydroxyl, mercapto, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkyl, C 1 -C 6 Haloalkoxy, preferably hydrogen, halogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkyl, C 1 -C 6 haloalkoxy; preferably R 4 is hydrogen or C 1 -C 6 alkyl.
  • R is selected from hydrogen, halogen, amino, nitro, cyano, hydroxyl, mercapto, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkyl, C 1 -C 6 Haloalkoxy, -C(O) Ra , preferably hydrogen, halogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkyl, C 1 -C 6 haloalkoxy , -C(O)R a ;
  • R a is selected from C 1 -C 6 alkyl groups.
  • Typical compounds of the invention include, but are not limited to, the following compounds:
  • the present invention further provides a compound represented by general formula (I) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer,
  • compound Ij and compound Ia undergo a dehydration reaction to obtain a compound of general formula (I);
  • the base is preferably triethylamine;
  • the catalyst is preferably triphenylphosphine dichloride;
  • Ring A, ring B, R 1 , R 2 , R 3 , R 4 , R 5 , and p are as defined in general formula (I).
  • the present invention further provides a compound represented by general formula (II) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer,
  • the base is preferably triethylamine
  • the catalyst is preferably triphenylphosphine dichloride
  • Ring B, R 1 , R 2 , R 3 , R 4 , R 5 , and p are as defined in general formula (II).
  • the present invention further provides a compound represented by general formula (III) according to the present invention or its stereoisomer, tautomer, mesoform, racemate, enantiomer,
  • the base is preferably triethylamine
  • the catalyst is preferably triphenylphosphine dichloride
  • R 1 , R 2 , R 3 , R 4 , R 5 , and p are as defined in general formula (III).
  • the present invention also provides a pharmaceutical composition, which comprises the compound represented by general formula (I), general formula (II) or general formula (III) or its stereoisomer, tautomer body, mesoform, racemate, enantiomer, diastereoisomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
  • a pharmaceutical composition which comprises the compound represented by general formula (I), general formula (II) or general formula (III) or its stereoisomer, tautomer body, mesoform, racemate, enantiomer, diastereoisomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
  • the object of the present invention is also to provide the compound represented by general formula (I), general formula (II) or general formula (III) according to the present invention or its stereoisomer, tautomer, meso body, racemate, enantiomer, diastereoisomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising it in the preparation of a cyclin-dependent kinase (CDK) inhibitory use in pharmaceuticals.
  • CDK cyclin-dependent kinase
  • the object of the present invention is also to provide the compound represented by general formula (I), general formula (II) or general formula (III) according to the present invention or its stereoisomer, tautomer, meso body, racemate, enantiomer, diastereoisomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition containing it in the preparation of inhibiting cancer cell proliferation, inhibiting cancer cell invasion, or Use in drugs for inducing cancer cell apoptosis.
  • general formula (I), general formula (II) or general formula (III) according to the present invention or its stereoisomer, tautomer, meso body, racemate, enantiomer, diastereoisomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition containing it in the preparation of inhibiting cancer cell proliferation, inhibiting cancer cell invasion, or Use in drugs for inducing cancer cell apoptosis.
  • the object of the present invention is also to provide the compound represented by general formula (I), general formula (II) or general formula (III) according to the present invention or its stereoisomer, tautomer, meso body, racemate, enantiomer, diastereoisomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising it in the preparation for prevention and/or treatment and cell cycle
  • a pharmaceutical composition comprising it in the preparation for prevention and/or treatment and cell cycle
  • Use in medicine for diseases associated with protein-dependent kinase activity such as cancer, especially in association with cyclin-dependent kinases CDK2/cyclin E1 (CCNE1), CDK6/cyclin D1, CDK4/ Cancers, more particularly breast cancers such as HR+/HER2- metastatic breast or ovarian cancers, characterized by amplification or overexpression of cyclin D3.
  • the present invention also relates to compounds represented by general formula (I), general formula (II) or general formula (III) or stereoisomers, tautomers, mesomers, exo Racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, or pharmaceutical compositions comprising the same, for use as CDK inhibitors.
  • the present invention also relates to compounds represented by general formula (I), general formula (II) or general formula (III) or stereoisomers, tautomers, mesomers, exo Racemates, enantiomers, diastereoisomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, or pharmaceutical compositions comprising them, for inhibiting cancer cell proliferation, inhibiting cancer cell invasion, or inducing Cancer cell apoptosis.
  • the present invention also relates to compounds represented by general formula (I), general formula (II) or general formula (III) or stereoisomers, tautomers, mesomers, exo Racemates, enantiomers, diastereoisomers, or mixtures thereof, or pharmaceutically acceptable salts thereof or pharmaceutical compositions comprising them, for use in the prevention and/or treatment of cyclin- Diseases associated with dependent kinase activity, such as cancer, in particular with amplification of the cyclin-dependent kinases CDK2/cyclin E1 (CCNE1), CDK6/cyclin D1, CDK4/cyclin D3 or cancers characterized by overexpression, more particularly breast cancer such as HR+/HER2- metastatic breast or ovarian cancer.
  • cyclin- Diseases associated with dependent kinase activity such as cancer
  • CCNE1 cyclin-dependent kinases
  • CDK2/cyclin E1 CCNE1
  • CDK6/cyclin D1, CDK4/cyclin D3 cancers
  • the present invention also relates to a method for inhibiting CDK, which comprises administering an effective amount of the compound represented by general formula (I), general formula (II) or general formula (III) according to the present invention to a subject in need
  • a method for inhibiting CDK which comprises administering an effective amount of the compound represented by general formula (I), general formula (II) or general formula (III) according to the present invention to a subject in need
  • the present invention also relates to a method for inhibiting cancer cell proliferation, inhibiting cancer cell invasion, or inducing cancer cell apoptosis, which comprises administering an effective amount of the general formula (I) and general formula (I) according to the present invention to a subject in need
  • the present invention also relates to a method for preventing and/or treating diseases related to cyclin-dependent kinase activity, which comprises administering to a subject in need an effective amount of the general formula (I) according to the present invention , the compound represented by general formula (II) or general formula (III) or its stereoisomer, tautomer, mesoform, racemate, enantiomer, diastereoisomer body, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising it, the disease such as cancer, especially with cyclin-dependent kinase CDK2/cyclin E1 (CCNE1), CDK6/cell Cancers characterized by amplification or overexpression of cyclin Dl, CDK4/cyclin D3, more particularly breast cancer such as HR+/HER2- metastatic breast or ovarian cancer.
  • the disease such as cancer, especially with cyclin-dependent kinase CDK2/cyclin E1 (CCNE1), CDK6/cell Cancers
  • Alkyl means a saturated monovalent aliphatic hydrocarbon group, which includes straight and branched chain groups having the indicated number of carbon atoms.
  • the alkyl group usually contains 1-20 carbon atoms (C 1 -C 20 alkyl), preferably 1 to 12 carbon atoms (C 1 -C 12 alkyl), more preferably 1 to 8 carbon atoms (C 1 -C 8 alkyl) or 1 to 6 carbon atoms (C 1 -C 6 alkyl”) or 1 to 4 carbon atoms (C 1 -C 4 alkyl).
  • alkyl examples include methyl, ethyl Base, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-Dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1, 2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2- Ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methyl Hexyl, 4-methylhexyl, 5-methylhexyl
  • Alkyl can be substituted or unsubstituted, when When substituted, the substituents may be substituted at any available point of attachment.
  • Optionally substituted alkyl groups described herein may be substituted with one or more substituents, which are independently selected unless otherwise stated. To the extent such substitution is chemically significant, the total number of substituents can be equal to the total number of hydrogen atoms on the alkyl group.
  • Optionally substituted alkyl typically contains 1 to 6 optional substituents, sometimes 1 to 5 optional substituents, preferably 1 to 4 optional substituents, or more preferably 1 to 3 optional substituents Substituents.
  • an alkyl group may be substituted by one or more (up to the total number of hydrogen atoms present on the alkyl group) halo groups.
  • C 1 -C 4 alkyl includes haloalkyl, especially fluoroalkyl having 1 to 4 carbon atoms, such as trifluoromethyl or difluoroethyl (ie CF 3 and —CH 2 CHF 2 ).
  • alkenyl means an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon double bond, for example vinyl, 1-propenyl, 2-propenyl, 1-, 2- or 3- -butenyl etc.
  • Alkenyl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio.
  • alkynyl refers to an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon triple bond, eg ethynyl, propynyl, butynyl and the like.
  • Alkynyl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring containing 3 to 20 carbon atoms, preferably containing 3 to 12 carbon atoms, more preferably containing 3 to 6 carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene Base, cyclooctyl, etc.; polycyclic cycloalkyl includes spiro ring, fused ring and bridged ring cycloalkyl.
  • spirocycloalkyl refers to a polycyclic group of 5 to 20 membered monocyclic rings sharing one carbon atom (called a spiro atom), which may contain one or more double bonds, but none of the rings has complete conjugation The ⁇ -electron system. Preferably it is 6 to 14 yuan, more preferably 7 to 10 yuan. According to the number of spiro atoms shared between the rings, the spirocycloalkyl group can be divided into single spirocycloalkyl, double spirocycloalkyl or polyspirocycloalkyl, preferably single spirocycloalkyl and double spirocycloalkyl.
  • spirocycloalkyl groups include:
  • fused cycloalkyl refers to a 5 to 20 membered all-carbon polycyclic group in which each ring of the system shares an adjacent pair of carbon atoms with other rings in the system, wherein one or more rings may contain one or Multiple double bonds, but none of the rings have a fully conjugated ⁇ -electron system.
  • it is 6 to 14 yuan, more preferably 7 to 10 yuan.
  • it can be divided into bicyclic, tricyclic, tetracyclic or polycyclic condensed cycloalkyl groups, preferably bicyclic or tricyclic, more preferably 5-membered/5-membered or 5-membered/6-membered bicycloalkyl groups.
  • fused cycloalkyl groups include:
  • bridged cycloalkyl refers to a 5 to 20 membered, all-carbon polycyclic group having any two rings sharing two carbon atoms not directly attached, which may contain one or more double bonds, but none of the rings has a complete Conjugated ⁇ -electron systems. Preferably it is 6 to 14 yuan, more preferably 7 to 10 yuan. According to the number of constituent rings, it can be divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl groups, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic.
  • bridged cycloalkyl groups include:
  • the cycloalkyl ring may be fused to an aryl, heteroaryl or heterocycloalkyl ring where the ring bonded to the parent structure is a cycloalkyl, non-limiting examples include indanyl, tetrahydronaphthalene base, benzocycloheptyl, etc.
  • Cycloalkyl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, oxo, carboxyl or carboxylate.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising 3 to 20 ring atoms, one or more of which is selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), but excluding ring portions of -OO-, -OS- or -SS-, the remaining ring atoms being carbon.
  • ring atoms Preferably contain 3 to 12 ring atoms, of which 1 to 4 are heteroatoms; most preferably contain 3 to 8 ring atoms, of which 1 to 3 are heteroatoms; most preferably contain 5 to 7 ring atoms, of which 1 to 2 or 1 to 3 are heteroatoms.
  • Non-limiting examples of monocyclic heterocyclyl groups include pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrrolyl, piperidine group, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl, etc., preferably 1, 2, 5-oxadiazolyl, pyranyl or morpholinyl.
  • Polycyclic heterocyclyls include spiro, fused and bridged heterocyclyls.
  • spiroheterocyclyl refers to a polycyclic heterocyclic group that shares one atom (called a spiro atom) between 5 to 20-membered monocyclic rings, wherein one or more ring atoms are selected from nitrogen, oxygen or S(O ) m (wherein m is an integer from 0 to 2), the remaining ring atoms are carbon. It may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron system. Preferably it is 6 to 14 yuan, more preferably 7 to 10 yuan.
  • the spiroheterocyclyl can be divided into single spiroheterocyclyl, double spiroheterocyclyl or polyspiroheterocyclyl, preferably single spiroheterocyclyl and double spiroheterocyclyl. More preferably, it is a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered monospiro heterocyclic group.
  • spiroheterocyclyls include:
  • fused heterocyclyl refers to a 5 to 20 membered polycyclic heterocyclic group in which each ring in the system shares an adjacent pair of atoms with other rings in the system, and one or more rings may contain one or more double bond, but none of the rings has a fully conjugated ⁇ -electron system, where one or more ring atoms are heteroatoms selected from nitrogen, oxygen, or S(O) m (where m is an integer from 0 to 2), and the remaining ring
  • the atom is carbon.
  • it is 6 to 14 yuan, more preferably 7 to 10 yuan.
  • bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic groups preferably bicyclic or tricyclic, more preferably 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic groups.
  • fused heterocyclic groups include:
  • bridged heterocyclyl refers to a 5 to 14 membered polycyclic heterocyclic group in which any two rings share two atoms not directly attached, which may contain one or more double bonds, but none of the rings has a complete shared bond.
  • it is 6 to 14 yuan, more preferably 7 to 10 yuan.
  • bridged heterocyclyl groups include:
  • the heterocyclyl ring may be fused to an aryl, heteroaryl, or cycloalkyl ring where the ring bonded to the parent structure is a heterocyclyl, non-limiting examples of which include:
  • Heterocyclic groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alk Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, oxo, carboxyl or carboxylate.
  • aryl refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic (that is, rings sharing adjacent pairs of carbon atoms) group, preferably 6 to 10 membered, having a conjugated pi-electron system, such as benzene base and naphthyl. Phenyl is more preferred.
  • the aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, where the ring bonded to the parent structure is an aryl ring, non-limiting examples of which include:
  • Aryl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio, carboxyl or carboxylate.
  • heteroaryl refers to a heteroaromatic system comprising 1 to 4 heteroatoms, 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen.
  • Heteroaryl is preferably 5 to 10 membered, containing 1 to 3 heteroatoms; more preferably 5 or 6 membered, containing 1 to 2 heteroatoms; preferred examples are imidazolyl, furyl, thienyl, thiazolyl, pyryl Azolyl, oxazolyl, pyrrolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl, etc., preferably imidazolyl, thiazolyl, pyrazolyl or pyrimidinyl, thiazolyl; more preferably pyrazolyl or thiazolyl.
  • the heteroaryl ring may be fused to an aryl, heterocyclyl or cycloalkyl
  • Heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, carboxyl or carboxylate.
  • alkoxy refers to -O-(alkyl) and -O-(cycloalkyl), wherein alkyl and cycloalkyl are as defined above.
  • alkoxy include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy.
  • Alkoxy may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkoxy Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, carboxyl or carboxylate.
  • the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkoxy Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl,
  • haloalkyl refers to an alkyl group substituted with one or more halo, wherein alkyl is as defined above.
  • haloalkoxy refers to an alkoxy group substituted with one or more halogens, wherein alkoxy group is as defined above.
  • hydroxyl refers to a -OH group.
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • amino refers to -NH2 .
  • cyano refers to -CN.
  • nitro refers to -NO2 .
  • mercapto refers to -SH.
  • ester group refers to -C(O)O(alkyl) or -C(O)O(cycloalkyl), wherein alkyl and cycloalkyl are as defined above.
  • acyl refers to compounds containing the group -C(O)R, where R is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl.
  • Optional or “optionally” means that the subsequently described event or circumstance can but need not occur, and that the description includes instances where the event or circumstance occurs or does not occur.
  • a heterocyclic group optionally substituted with an alkyl group means that an alkyl group may but need not be present, and the description includes cases where the heterocycle group is substituted with an alkyl group and cases where the heterocycle group is not substituted with an alkyl group .
  • Substituted means that one or more hydrogen atoms in a group, preferably up to 5, more preferably 1 to 3 hydrogen atoms are independently substituted by the corresponding number of substituents. It goes without saying that substituents are only in their possible chemical positions and that a person skilled in the art can determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, an amino or hydroxyl group with free hydrogen may be unstable when bonded to a carbon atom with an unsaturated (eg, ethylenic) bond.
  • “Pharmaceutical composition” means a mixture containing one or more compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, and other components such as a physiologically/pharmaceutically acceptable carrier and excipients.
  • the purpose of the pharmaceutical composition is to promote the administration to the organism, facilitate the absorption of the active ingredient and thus exert biological activity.
  • “Pharmaceutically acceptable salt” refers to the salt of the compound of the present invention, which is safe and effective when used in mammals, and has proper biological activity.
  • Cancer refers to any malignant and/or invasive growth or tumor (caused by abnormal cell growth). Cancers include solid tumors named for the type of cells that form them, cancers of the blood, bone marrow, or lymphatic system. Examples of solid tumors include sarcomas and carcinomas. Cancers of the blood include, but are not limited to, leukemias, lymphomas, and myelomas. Cancer also includes primary cancer that started in a specific part of the body, metastatic cancer that has spread from where it started to other parts of the body, recurrence from the original primary cancer after remission, and second primary cancer (This is a new primary cancer in a person with a history of previous cancer of a different type than the new primary cancer).
  • the cancer may be selected from breast cancer, ovarian cancer, bladder cancer, uterine cancer, prostate cancer, lung cancer, esophageal cancer, liver cancer, pancreatic cancer, and gastric cancer.
  • the cancer is characterized by amplification or overexpression of CCNE1 and/or CCNE2.
  • Stereoisomers described herein may include cis and trans isomers, optical isomers such as (R) and (S) enantiomers of the compounds of the invention (including compounds exhibiting more than one isomeric type) Isomers, diastereoisomers, geometric isomers, rotamers, atropisomers, conformers and tautomers; and mixtures thereof (such as racemates and asymmetric enantiomer pair).
  • optical isomers such as (R) and (S) enantiomers of the compounds of the invention (including compounds exhibiting more than one isomeric type) Isomers, diastereoisomers, geometric isomers, rotamers, atropisomers, conformers and tautomers; and mixtures thereof (such as racemates and asymmetric enantiomer pair).
  • the compounds of the present invention may exhibit tautomerism and structural isomerism.
  • compounds may exist in several tautomeric forms, including enol and imine forms and keto and enamine forms, and geometric isomers and mixtures thereof. All such tautomeric forms are included within the scope of the compounds of the present invention.
  • Tautomers exist as a mixture of tautomeric sets in solution. In solid form, usually one tautomer predominates. Even though one tautomer may be described, the present invention includes all tautomers of the provided compounds.
  • enantiomeric purity of the compounds described herein can be described in terms of enantiomeric excess (ee), which means that a sample contains one enantiomer in a greater amount than the other Degree.
  • ee enantiomeric excess
  • the ee of a racemic mixture is 0%, while the ee of a single, completely pure enantiomer is 100%.
  • diastereomeric purity can be described in terms of diastereomeric excess (de).
  • the compound represented by the general formula (I) of the present invention can form a pharmaceutically acceptable base addition salt or acid addition salt with a base or an acid.
  • the base includes inorganic bases and organic bases.
  • Acceptable organic bases include diethanolamine, ethanolamine, N-methylglucamine, triethanolamine, tromethamine, etc.
  • Acceptable inorganic bases include aluminum hydroxide, hydroxide Calcium, Potassium Hydroxide, Sodium Carbonate and Sodium Hydroxide etc.
  • the acid includes inorganic acid and organic acid, acceptable inorganic acid includes hydrochloric acid, sulfuric acid, nitric acid, phosphoric acid, hydrobromic acid and the like.
  • Acceptable organic acids include acetic acid, trifluoroacetic acid, formic acid, ascorbic acid, and the like.
  • the pharmaceutical composition containing the active ingredient may be in a form suitable for oral administration, such as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixir.
  • Oral compositions can be prepared according to any method known in the art for the preparation of pharmaceutical compositions, and such compositions can contain one or more ingredients selected from the group consisting of sweeteners, flavoring agents, coloring agents and preservatives, To provide pleasing and palatable medicinal preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients suitable for the manufacture of tablets.
  • excipients can be inert excipients such as calcium carbonate, sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents such as microcrystalline cellulose, croscarmellose sodium, corn starch or alginic acid; binders such as starch, gelatin, polyvinylpyrrolidone or acacia; and lubricants such as magnesium stearate, stearic acid or talc.
  • These tablets may be uncoated or may be coated by known techniques to mask the taste of the drug or to delay disintegration and absorption in the gastrointestinal tract, thus providing sustained release over an extended period of time.
  • water-soluble taste-masking materials such as hydroxypropylmethylcellulose or hydroxypropylcellulose, or time-extending materials such as ethylcellulose, cellulose acetate butyrate may be used.
  • Hard gelatin capsules in which the active ingredient is admixed with an inert solid diluent such as calcium carbonate, calcium phosphate, or kaolin, or in which the active ingredient is admixed with a water-soluble carrier such as polyethylene glycol or an oil vehicle such as peanut oil, liquid paraffin, or olive oil may also be used.
  • Soft gelatin capsules provide an oral formulation.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, and acacia; dispersing or wetting agents, which may be natural
  • the resulting phospholipids such as lecithin, or condensation products of alkylene oxides with fatty acids, such as polyoxyethylene stearate, or condensation products of ethylene oxide with long-chain fatty alcohols, such as heptadecanylethyleneoxycetate Heptadecaethyleneoxy cetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitols, such as polyethylene oxide sorbitan monooleate, or ethylene oxide with fatty acids and hexitols Condensation products of anhydride-derived partial esters, such as polyethylene oxide sorb
  • Aqueous suspensions may also contain one or more preservatives, such as ethyl or n-propylparaben, one or more coloring agents, one or more flavoring agents and one or more sweeteners.
  • preservatives such as ethyl or n-propylparaben
  • coloring agents such as ethyl or n-propylparaben
  • flavoring agents such as sucrose, saccharin, or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • Oily suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening and flavoring agents as mentioned above may be added to provide a palatable preparation. These compositions can be preserved by the addition of antioxidants such as butylated hydroxyanisole or alpha-tocopherol.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient with a dispersing or wetting agent, suspending agent or one or more preservatives for admixture. Suitable dispersing or wetting agents and suspending agents are mentioned above. Other excipients, for example sweetening, flavoring and coloring agents, may also be added. These compositions are preserved by the addition of antioxidants such as ascorbic acid.
  • the pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil such as olive oil or arachis oil, or a mineral oil such as liquid paraffin or mixtures thereof.
  • Suitable emulsifiers may be naturally occurring phospholipids, such as soybean lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and the condensation of said partial esters with ethylene oxide Products such as polyethylene oxide sorbitan monooleate.
  • the emulsions may also contain sweetening, flavoring, preservative and antioxidant agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, coloring agents and antioxidants.
  • sweetening agents for example glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative, coloring agents and antioxidants.
  • the pharmaceutical compositions of the present invention may be in the form of sterile injectable aqueous solutions.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • the sterile injectable preparation may be a sterile injectable oil-in-water microemulsion in which the active ingredient is dissolved in the oily phase.
  • the active ingredient is dissolved in a mixture of soybean oil and lecithin.
  • the oil solution is then treated in a mixture of water and glycerol to form a microemulsion.
  • the injectable solution or microemulsion can be injected into the patient's bloodstream by local bolus injection.
  • solutions and microemulsions are preferably administered in a manner that maintains a constant circulating concentration of the compounds of the invention. To maintain this constant concentration, a continuous intravenous delivery device can be used.
  • the pharmaceutical composition of the present invention may be in the form of sterile injectable aqueous or oily suspension for intramuscular and subcutaneous administration.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension prepared in a non-toxic parenterally acceptable diluent or solvent, for example a solution in 1,3-butanediol.
  • sterile fixed oils are conveniently employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are prepared as injectables.
  • the compounds of this invention may be administered in the form of suppositories for rectal administration.
  • These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid in the rectum and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, polyethylene glycols of various molecular weights and mixtures of fatty acid esters of polyethylene glycols.
  • the dosage of the drug depends on many factors, including but not limited to the following factors: the activity of the specific compound used, the patient's age, the patient's body weight, the patient's health status, the patient's behavior, the patient's Diet, administration time, administration method, excretion rate, drug combination, etc.
  • the optimal treatment method such as the treatment mode, the daily dosage of the compound of the general formula or the type of pharmaceutically acceptable salt can be verified according to the traditional treatment plan.
  • the present invention can contain the compound represented by the general formula (I), and its pharmaceutically acceptable salt, hydrate or solvate as the active ingredient, mixed with a pharmaceutically acceptable carrier or excipient to prepare a composition, and Prepared into clinically acceptable dosage forms.
  • the derivatives of the present invention can be used in combination with other active ingredients as long as they do not produce other adverse effects such as allergic reactions and the like.
  • the compound of the present invention can be used as the only active ingredient, and can also be used in combination with other drugs for treating diseases related to tyrosine kinase activity. Combination therapy is achieved by the simultaneous, separate or sequential administration of the individual therapeutic components.
  • the present invention adopts the following synthetic scheme to prepare the compound of general formula (I) of the present invention.
  • the present invention adopts the following Scheme 1 to prepare the compound represented by the general formula (I) of the present invention.
  • Step 1 In a polar aprotic solvent (such as dichloromethane), in the presence of a suitable base (such as triethylamine), the sulfonyl chloride compound is reacted to obtain the sulfonamide compound Ia;
  • a polar aprotic solvent such as dichloromethane
  • a suitable base such as triethylamine
  • Step 2 In a suitable solvent, in the presence of a suitable reducing agent, 4-chloro-2-(methylthio)pyrimidine-5-carboxylic acid ethyl ester undergoes a reduction reaction to obtain Compound Ib;
  • the solvent is such as tetrahydrofuran, the The reducing agent such as diisobutylaluminum hydride;
  • Step 3 In a suitable solvent, in the presence of a suitable base, compound Ib reacts with compound Ic to obtain compound Id;
  • the solvent is for example isopropanol
  • the base is for example diisopropylethylamine
  • the reaction temperature can be Between room temperature and 80°C;
  • Step 4 In a suitable solvent, in the presence of a suitable oxidizing agent, compound Id undergoes an oxidation reaction to obtain compound Ie; the solvent such as ethyl acetate, the oxidizing agent such as manganese dioxide, the reaction temperature can be at 50°C to 80°C between;
  • Step 5 In a suitable solvent, in the presence of a suitable catalyst, compound Ie undergoes an Aldol cyclization reaction (see VanderWel et al., J.Med.Chezn.2005, 4S, 2371) to obtain compound If; the solvent such as Tetrahydrofuran, the catalyst such as LHMDS, the reaction temperature can be between -20°C and room temperature, and the reaction is carried out under nitrogen atmosphere;
  • Step 6 In a suitable solvent, in the presence of a suitable oxidizing agent, compound If is oxidized to obtain compound Ig; the solvent is especially a mixed solvent, such as a mixed solvent of 2-methyltetrahydrofuran and water, and the oxidizing agent includes but Not limited to potassium hydrogen persulfate, m-chloroperoxybenzoic acid, etc. Excessive oxidizing agents help the reaction to proceed completely, and the reaction temperature can be between 10°C and 80°C;
  • Step 7 In a suitable solvent, in the presence of a suitable base, compound Ig reacts with compound Ih to obtain compound Ii;
  • the solvent such as isopropanol, 2-methyltetrahydrofuran, the base such as diisopropylethyl Amines, the reaction temperature can be between room temperature and 80°C;
  • Step 8 In a suitable solvent, in the presence of a suitable acid, compound Ii is deprotected to obtain compound Ij; the solvent is such as dioxane, the acid is such as hydrochloric acid, and the reaction temperature is typically at room temperature; further The compound is freed by a base such as sodium hydroxide, potassium hydroxide;
  • Step 9 In a suitable solvent, in the presence of a suitable base and a catalyst, compound Ij and compound Ia undergo a dehydration reaction to obtain a compound of general formula (I);
  • the solvent is, for example, dichloromethane
  • the base such as triethylamine
  • the catalyst such as triphenylphosphine dichloride
  • the reaction is typically carried out under a nitrogen atmosphere
  • the reaction temperature is typically 0°C to room temperature;
  • ring A, ring B, R 1 , R 2 , R 3 , R 4 , R 5 , and p are as defined in general formula (I).
  • the present invention adopts the following scheme 2 to prepare the compound of general formula (II) of the present invention.
  • Step 1 In a polar aprotic solvent (such as dichloromethane), in the presence of a suitable base (such as triethylamine), the sulfonyl chloride compound is reacted to obtain the sulfonamide compound Ia;
  • a polar aprotic solvent such as dichloromethane
  • a suitable base such as triethylamine
  • Step 2 In a suitable solvent, in the presence of a suitable reducing agent, 4-chloro-2-(methylthio)pyrimidine-5-carboxylic acid ethyl ester undergoes a reduction reaction to obtain Compound Ib;
  • the solvent is such as tetrahydrofuran, the The reducing agent such as diisobutylaluminum hydride;
  • Step 3 In a suitable solvent, in the presence of a suitable base, compound Ib reacts with compound Ic to obtain compound Id;
  • the solvent is for example isopropanol
  • the base is for example diisopropylethylamine
  • the reaction temperature can be Between room temperature and 80°C;
  • Step 4 In a suitable solvent, in the presence of a suitable oxidizing agent, compound Id undergoes an oxidation reaction to obtain compound Ie; the solvent such as ethyl acetate, the oxidizing agent such as manganese dioxide, the reaction temperature can be at 50°C to 80°C between;
  • Step 5 In a suitable solvent, in the presence of a suitable catalyst, compound Ie undergoes an Aldol cyclization reaction (see VanderWel et al., J.Med.Chezn.2005, 4S, 2371) to obtain compound If; the solvent such as Tetrahydrofuran, the catalyst such as LHMDS, the reaction temperature can be between -20°C and room temperature, and the reaction is carried out under nitrogen atmosphere;
  • Step 6 In a suitable solvent, in the presence of a suitable oxidizing agent, compound If is oxidized to obtain compound Ig; the solvent is especially a mixed solvent, such as a mixed solvent of 2-methyltetrahydrofuran and water, and the oxidizing agent includes but Not limited to potassium hydrogen persulfate, m-chloroperoxybenzoic acid, etc. Excessive oxidizing agents help the reaction to proceed completely, and the reaction temperature can be between 10°C and 80°C;
  • Step 7 In a suitable solvent, in the presence of a suitable base, compound Ig reacts with compound IIh to obtain compound IIi; Amines, the reaction temperature can be between room temperature and 80°C;
  • Step 8 In a suitable solvent, in the presence of a suitable acid, compound IIi is deprotected to obtain compound IIj; the solvent is such as dioxane, the acid is such as hydrochloric acid, and the reaction temperature is typically at room temperature; further The compound is freed by a base such as sodium hydroxide, potassium hydroxide;
  • Step 9 In a suitable solvent, in the presence of a suitable base and a catalyst, compound IIj and compound Ia undergo a dehydration reaction to obtain a compound of general formula (II);
  • the solvent is, for example, dichloromethane
  • the base such as triethylamine
  • the catalyst such as triphenylphosphine dichloride
  • the reaction is typically carried out under a nitrogen atmosphere
  • the reaction temperature is typically 0°C to room temperature;
  • ring B, R 1 , R 2 , R 3 , R 4 , R 5 , and p are as defined in general formula (II).
  • the present invention adopts the following scheme 3 to prepare the compound of general formula (III) of the present invention.
  • Step 1 In a polar aprotic solvent (such as dichloromethane), in the presence of a suitable base (such as triethylamine), the sulfonyl chloride compound is reacted to obtain the sulfonamide compound Ia;
  • a polar aprotic solvent such as dichloromethane
  • a suitable base such as triethylamine
  • Step 2 In a suitable solvent, in the presence of a suitable reducing agent, 4-chloro-2-(methylthio)pyrimidine-5-carboxylic acid ethyl ester undergoes a reduction reaction to obtain Compound Ib;
  • the solvent is such as tetrahydrofuran, the The reducing agent such as diisobutylaluminum hydride;
  • Step 3 In a suitable solvent, in the presence of a suitable base, compound Ib reacts with compound IIIc to obtain compound IIId;
  • the solvent is for example isopropanol
  • the base is for example diisopropylethylamine
  • the reaction temperature can be Between room temperature and 80°C;
  • Step 4 In a suitable solvent, in the presence of a suitable oxidizing agent, compound IIId is oxidized to obtain compound IIIe; the solvent is, for example, ethyl acetate, and the oxidizing agent is, for example, manganese dioxide.
  • the reaction temperature can be between 50°C and 80°C between;
  • Step 5 In a suitable solvent, in the presence of a suitable catalyst, compound IIIe undergoes an Aldol cyclization reaction (see VanderWel et al., J.Med.Chezn.2005, 4S, 2371) to obtain compound IIIf;
  • the solvent such as Tetrahydrofuran, the catalyst such as LHMDS, the reaction temperature can be between -20°C and room temperature, and the reaction is carried out under nitrogen atmosphere;
  • Step 6 In a suitable solvent, in the presence of a suitable oxidizing agent, compound IIIf undergoes an oxidation reaction to obtain compound IIIg;
  • the solvent is particularly a mixed solvent, such as a mixed solvent of 2-methyltetrahydrofuran and water, and the oxidizing agent includes but Not limited to potassium hydrogen persulfate, m-chloroperoxybenzoic acid, etc. Excessive oxidizing agents help the reaction to proceed completely, and the reaction temperature can be between 10°C and 80°C;
  • Step 7 In a suitable solvent, in the presence of a suitable base, compound IIIg reacts with compound IIh to obtain compound IIIi;
  • the solvent such as isopropanol, 2-methyltetrahydrofuran, the base such as diisopropyl Amines
  • the reaction temperature can be between room temperature and 80°C;
  • Step 8 In a suitable solvent, in the presence of a suitable acid, compound IIIi is deprotected to obtain compound IIIj; the solvent is such as dioxane, the acid is such as hydrochloric acid, and the reaction temperature is typically at room temperature; further The compound is freed by a base such as sodium hydroxide, potassium hydroxide;
  • Step 9 In a suitable solvent, in the presence of a suitable base and a catalyst, compound IIIj and compound Ia undergo a dehydration reaction to obtain a compound of general formula (III);
  • the solvent is, for example, dichloromethane
  • the base such as triethylamine
  • the catalyst such as triphenylphosphine dichloride
  • the reaction is typically carried out under a nitrogen atmosphere
  • the reaction temperature is typically 0°C to room temperature
  • R 1 , R 2 , R 3 , R 4 , R 5 , and p are as defined in general formula (III).
  • the compounds of the present invention are prepared utilizing convenient starting materials and general preparative procedures.
  • the present invention gives typical or preferred reaction conditions, such as reaction temperature, time, solvent, pressure, molar ratio of reactants. But unless otherwise specified, other reaction conditions can also be adopted. Optimum conditions may vary with specific reactants or solvents used, but in general, reaction optimization steps and conditions can be identified.
  • protecting groups may be used in the present invention to protect certain functional groups from unnecessary reactions.
  • Suitable protecting groups for various functional groups and their protection or deprotection conditions are widely known to those skilled in the art.
  • Protecting Groups in Organic Preparations by T.W. Greene and G.M. Wuts (3rd Edition, Wiley, New York, 1999 and citations in the book) describes in detail the protection or deprotection of a large number of protecting groups.
  • the separation and purification of compounds and intermediates takes appropriate methods and steps according to specific needs, such as filtration, extraction, distillation, crystallization, column chromatography, preparative thin-layer plate chromatography, preparative high-performance liquid chromatography or a combination of the above methods.
  • For its specific usage method please refer to the examples described in the present invention.
  • other similar separation and purification means can also be used. They can be characterized using conventional methods, including physical constants and spectral data.
  • NMR nuclear magnetic resonance
  • MS mass spectroscopy
  • MS was determined by LC (Waters 2695)/MS (Quattro Premier xE) mass spectrometer (manufacturer: Waters) (Photodiode Array Detector).
  • the lc6000 high performance liquid chromatograph (manufacturer: Innovation Tongheng) was used for the preparative liquid chromatography.
  • Chromatographic column is DaisogelC18 10 ⁇ m 100A (30mm ⁇ 250mm), mobile phase: acetonitrile/water.
  • the thin-layer chromatography silica gel plate uses Qingdao Ocean Chemical GF254 silica gel plate.
  • the specification of the silica gel plate used for thin-layer chromatography (TLC) is 0.20mm-0.25mm, and the specification used for the preparation of thin-layer chromatography separation and purification products is 0.5mm.
  • the known starting materials of the present invention can be adopted or synthesized according to methods known in the art, or can be purchased from Wanghua Mall, Beijing Coupling, Sigma, Bailingwei, Yi Shiming, Shanghai Shuya, Shanghai Yinuokai, Anaiji Chemical, Shanghai Biide and other companies.
  • the reactions can all be carried out under a nitrogen atmosphere.
  • the argon atmosphere or nitrogen atmosphere means that the reaction bottle is connected to an argon or nitrogen balloon with a volume of about 1 L.
  • Reaction solvent organic solvent or inert solvent are respectively expressed as that the solvent used does not participate in the reaction under the described reaction conditions, including such as benzene, toluene, acetonitrile, tetrahydrofuran (THF), dimethylformamide (DMF), chloroform, Dichloromethane, ether, methanol, nitrogen-methylpyrrolidinone (NMP), pyridine, etc.
  • the solution refers to an aqueous solution.
  • the chemical reactions described in the present invention are generally carried out under normal pressure.
  • the reaction temperature is between -78°C and 200°C.
  • the reaction time and conditions are, for example, between -78°C and 200°C under one atmospheric pressure, and the reaction is completed within about 1 to 24 hours. If the reaction is overnight, the reaction time is generally 16 hours. Unless otherwise specified in the examples, the reaction temperature is room temperature, which is 20°C to 30°C.
  • the monitoring of the reaction process in the embodiment adopts thin layer chromatography (TLC), and the system of developing agent used in the reaction has: A: dichloromethane and methanol system, B: sherwood oil and ethyl acetate system, C: acetone, The volume ratio of the solvent is adjusted according to the polarity of the compound.
  • TLC thin layer chromatography
  • the eluent system of the column chromatography that purifies compound adopts and the developer system of thin-layer chromatography include: A: dichloromethane and methanol system, B: sherwood oil and ethyl acetate system, the volume ratio of solvent is according to the compound It can be adjusted according to the polarity, and it can also be adjusted by adding a small amount of basic or acidic reagents such as triethylamine and trifluoroacetic acid.
  • Boc tert-butoxycarbonyl
  • DIPEA Diisopropylethylamine
  • EDTA ethylenediaminetetraacetic acid
  • IC 50 the concentration that inhibits 50% of the activity
  • LHMDS lithium hexamethyldisilazane salt (lithium bis(trimethylsilyl)amide)
  • mCPBA m-chloroperoxybenzoic acid
  • NBS N-bromosuccinimide
  • NCS N-chlorosuccinimide
  • NIS N-iodosuccinimide
  • PE petroleum ether
  • TBS tert-butyldimethylsilyl
  • TBHP tert-butyl hydroperoxide
  • Step 2 Preparation of ( ⁇ )-(1R*,2R*)-2-(benzylamino)-1-methylcyclopent-1-ol (( ⁇ )1b)
  • the acidic aqueous layer was cooled in an ice-water bath, and the pH was adjusted to 10 using 5N aqueous sodium hydroxide solution.
  • the resulting biphasic mixture was extracted with ethyl acetate (1200 mL ⁇ 3), and the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a brown oil. Concentrate under reduced pressure at 80° C. for 5 h, cool down to room temperature, disperse in petroleum ether, and filter to obtain 85.0 g of the title product as a brown solid.
  • the reaction mixture remained clear for about 1 min and then started to precipitate. After 5 min, a thick white suspension formed but did not interfere with stirring. Stirring was continued at 80 °C for 4 h, then heating was stopped and the mixture was continued to stir while gradually cooling to room temperature overnight. The mother liquor was collected by filtration and concentrated to give 45 g of a brown solid. Suspend the brown solid in water (250 mL) and ethyl acetate (500 mL) in a 1 L separatory funnel. Aqueous hydrochloric acid (4M, 150 mL, 600 mmol) was added and the mixture was stirred for about 30 seconds. A clear biphasic mixture is obtained.
  • Step 7 Preparation of 4-(((1R,2R)-2-hydroxy-2-methylcyclopentyl)amino)-2-(methylthio)pyrimidine-5-carbaldehyde (1 g)
  • Step 8 8-((1R,2R)-2-Hydroxy-2-methylcyclopentyl)-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one ( 1h)
  • Step 9 8-((1R,2R)-2-Hydroxy-2-methylcyclopentyl)-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one Preparation of (1i)
  • Step 10 4-((8-((1R,2R)-2-Hydroxy-2-methylcyclopentyl)-7-oxo-7,8-dihydropyrido[2,3-d]pyrimidine Preparation of -2-yl)amino)piperidine-1-carboxylic acid tert-butyl ester (1j)
  • Step 11 8-((1R,2R)-2-Hydroxy-2-methylcyclopentyl)-2-(piperidin-4-ylamino)pyrido[2,3-d]pyrimidine-7(8H ) - Preparation of ketone (1k)
  • Step 13 2-((1-(Cyclopropanesulfonylimino)piperidin-4-yl)amino)-8-(((1R,2R)-2-hydroxy-2-methylcyclopentyl)pyridine
  • Step 1 2-((1-(Cyclopropanesulfonylimino)piperidin-4-yl)amino)-6-(difluoromethyl)-8-((1R,2R)-2-hydroxy-2 Preparation of -methylcyclopentyl)pyrido[2,3-d]pyrimidin-7(8H)-one (2)
  • Step 7 8-Cyclopentyl-2-(((1-(cyclopropanesulfonylimino)piperidin-4-yl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (3)
  • Step 2 8-((1R,2R)-2-Hydroxy-2-methylcyclopentyl)-2-(((1-(N-methylcyclopropanesulfonylimino)piperidin-4-yl )amino)pyrido[2,3-d]pyrimidin-7(8H)-one (4) preparation
  • Example 5 6-(difluoromethyl)-8-(((1R,2R)-2-hydroxyl-2-methylcyclopentyl)-2-(((1-(N-methylcyclopropane Preparation of sulfonylimino))piperidin-4-yl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (5)
  • Example 7 8-cyclopentyl-2-(((1-(prop-2-ylsulfonylimino)piperidin-4-yl)amino)pyrido[2,3-d]pyrimidine-7( Preparation of 8H)-ketone (7)
  • Example 13 8-cyclopentyl-2-(((1-(S-methylsulfonylimino)piperidin-4-yl)amino)pyrido[2,3-d]pyrimidine-7(8H )-The preparation of ketone (13)
  • the preparation method is the same as that of Example 3, except that N-(tert-butyldimethylsilyl)cyclopropane in step 7 is replaced by N-(tert-butyldimethylsilyl)methanesulfonamide (11a) Sulfonamide (11), yielding the title compound.
  • Example 15 6-(Difluoromethyl)-2-(((1-(N,S-dimethylsulfonylimino)piperidin-4-yl)amino)-8-((1R,2R Preparation of )-2-hydroxy-2-methylcyclopentyl)pyrido[2,3-d]pyrimidin-7(8H)-one (15)
  • Example 22 8-(2-Methylcyclopentyl)-2-((1-(S-methylsulfonylimino)piperidin-4-yl)amino)pyrido[2,3-d] Preparation of pyrimidin-7(8H)-one (22-P1 and 22-P2)
  • Preparative liquid chromatography chromatographic column: Daisogei 30mm ⁇ 250mm, C18, 10um, 100A, mobile phase: acetonitrile/water, gradient: 30%-80%.
  • Preparative liquid chromatography chromatographic column: Daisogei 30mm ⁇ 250mm, C18, 10um, 100A, mobile phase: acetonitrile/water, gradient: 30%-80%.
  • the preparation method is the same as that of Example 3, except that 2,2-dimethylcyclopentylamine is used instead of cyclopentylamine and N-(tert-butyldimethylsilyl)methanesulfonamide (11a) is used instead of N-( tert-Butyldimethylsilyl)cyclopropanesulfonamide (1l) to give the title compound.
  • Example 28 6-Acetyl-8-cyclopentyl-2-(((1-(S-methylsulfonylimino)piperidin-4-yl)amino)pyrido[2,3-d] Preparation of pyrimidin-7(8H)-one (28)
  • Step 1 4-((6-iodo-8-cyclopentyl-7-oxo-7,8-dihydropyrido[2,3-d]pyrimidin-2-yl)amino)piperidine-1- Preparation of tert-butyl carboxylate (28a)
  • Step 2 4-(((8-cyclopentyl-6-(1-ethoxyvinyl)-7-oxo-7,8-dihydropyrido[2,3-d]pyrimidine-2- base)amino)piperidine-1-carboxylate tert-butyl ester (28b)
  • Step 3 Preparation of 6-acetyl-8-cyclopentyl-2-(piperidin-4-ylamino)pyrido[2,3-d]pyrimidin-7(8H)-one (28c)
  • Step 1 4-((6-Bromo-8-cyclopentyl-5-methyl-7-oxo-7,8-dihydropyrido[2,3-d]pyrimidin-2-yl)amino) Preparation of tert-butyl piperidine-1-carboxylate (29a)
  • tert-butyl 4-aminopiperidine-1-carboxylate (2.52g, 12.0mmol) and toluene (10mL) were added to the reaction flask.
  • Silyl)amide lithium (1M in THF, 12mL, 12.0mmol
  • stirred reaction for 20 minutes added compound 6-bromo-2-chloro-8-cyclopentyl-5-methylpyrido[2,3- d] Pyrimidin-7(8H)-one (1.03g, 3.00mmol) (commercial reagent CAS: 1016636-76-2), stirred overnight. It was concentrated under reduced pressure, and EtOAc (20 mL ⁇ 3) and water (10 mL) were added to the residue.
  • the preparation method was the same as that of Example 3, except that N-(tert-butyldimethylsilyl)cyclopropanesulfonamide (1l) was replaced by isothiazolidine 1,1-dioxide to prepare compound 30.
  • Compound 31 was prepared in the same manner as in Example 22, except that ethyl propionate was used instead of ethyl acetate.
  • Example 33 6-(Difluoromethyl)-8-(2-methylcyclopentyl)-2-(((1-(S-methylsulfonylimino)piperidin-4-yl)amino Preparation of ]pyrido[2,3-d]pyrimidin-7(8H)-one (33-P1 and 33-P2)
  • Step 1 6-(Difluoromethyl)-8-(2-methylcyclopentyl)-2-(((1-(methylsulfonylimino)piperidin-4-yl)amino]pyridine
  • Step 1 6-(Difluoromethyl)-8-(2-methylcyclopentyl)-2-(((1-(methylsulfonylimino)piperidin-4-yl)amino]pyridine
  • Preparative liquid chromatography chromatographic column: Daisogei 30mm ⁇ 250mm, C18, 10um, 100A, mobile phase: acetonitrile/water, gradient: 30%-80%.
  • Preparative liquid chromatography chromatographic column: Daisogei 30mm ⁇ 250mm, C18, 10um, 100A, mobile phase: acetonitrile/water, gradient: 30%-80%.
  • Example 34 6-Acetyl-5-methyl-8-(2-methylcyclopentyl)-2-(((1-(S-methylsulfonylimino)piperidinyl-4-yl )amino)pyrido[2,3-d]pyrimidin-7(8H)-one (34) preparation
  • Step 1 4-(((5-Methyl-8-(2-methylcyclopentyl)-7-oxo-7,8-dihydropyrido[2,3-d]pyrimidin-2-yl )amino)piperidine-1-carboxylate tert-butyl ester (34c)
  • Example 28 The remaining steps are the same as in Example 28, except that 4-(((5-methyl-8-(2-methylcyclopentyl)-7-oxo-7,8-dihydropyrido[2,3 -d]pyrimidin-2-yl)amino)tert-butyl-1-carboxylate tert-butyl ester (34c) instead of 4-((8-cyclopentyl-7-oxo-7,8-dihydropyrido[ 2,3-d]pyrimidin-2-yl)amino)piperidine-1-carboxylate tert-butyl ester (3e) to give compound 34.
  • Example 36 8-cyclopentyl-2-(((1-(thiophene-2-sulfonylimino)piperidin-4-yl)amino)pyrido[2,3-d]pyrimidine-7(8H )-The preparation of ketone (36)
  • Compound 37 was prepared in the same manner as in Example 22, except that ethyl acetate was replaced with ethyl 2-fluoroacetate.
  • Step 1 4-(((6-Chloro-8-(2-methylcyclopentyl)-7-oxo-7,8-dihydropyrido[2,3-d]pyrimidin-2-yl) Preparation of tert-butyl amino)piperidine-1-carboxylate (38a)
  • Step 1 4-(((6-Bromo-8-(2-methylcyclopentyl)-7-oxo-7,8-dihydropyrido[2,3-d]pyrimidin-2-yl) Preparation of tert-butyl amino)piperidine-1-carboxylate (39a)
  • Step 2 4-((8-(2-methylcyclopentyl)-7-oxo-6-(prop-1-en-2-yl)-7,8-dihydropyrido[2,3 -d] Preparation of pyrimidin-2-yl)amino)piperidine-1-carboxylate tert-butyl ester (39b)
  • Step 3 4-(((6-isopropyl-8-(2-methylcyclopentyl)-7-oxo-7,8-dihydropyrido[2,3-d]pyrimidine-2- base)amino)piperidine-1-carboxylate tert-butyl ester (39c)
  • Example 40 8-cyclobutyl-2-(((1-(S-methylsulfonylimino)piperidin-4-yl)amino)pyrido[2,3-d]pyrimidine-7(8H )-The preparation of ketone (40)
  • the preparation method is the same as in Example 3, except that cyclopentylamine is replaced by 3,3-difluorocyclopentylamine hydrochloride and N-(tert-butyldimethylsilyl)methanesulfonamide (11a) is replaced by N -(tert-Butyldimethylsilyl)cyclopropanesulfonamide (11), compound 47 was obtained.
  • N-benzyl-3-methylcyclopentane-1-amine (25.0 g, crude product) and isopropanol (200 mL) were added to the reaction flask, and palladium hydroxide on carbon (3.00 g ). The hydrogen gas was replaced, and the reaction was carried out overnight at 45°C. After the reaction was completed, it was filtered, and the temperature of the filtrate was lowered to 5° C., and dioxane hydrochloride solution (4.0 M, 50 mL) was added dropwise, and stirred overnight at room temperature. The reaction solution was concentrated under reduced pressure to obtain 17.9 g of crude reddish-brown solid, which was directly used in the next step.
  • the preparation method is the same as that of Example 3, except that cyclopentylamine is replaced by cyclohexylamine and N-(tert-butyldimethylsilyl) methanesulfonamide (11a) is replaced by N-(tert-butyldimethylsilyl) Silyl)cyclopropanesulfonamide (1l), to obtain compound 50.
  • Example 51 6-(Difluoromethyl)-2-((1-(S-methylsulfonylimino)piperidin-4-yl)amino)-8-(spiro[2.4]hept-4- base) pyrido[2,3-d]pyrimidin-7-(8H)-one (51)
  • Test Example 1 Inhibitory activity of compounds of the present invention on CDK2/4/6
  • ADP-Glo Kinase Assay Kit (Promega, Cat. No. V9102) was used to detect the inhibitory activity of the test compound on CDK2/CyclinE1 kinase.
  • the kinase phosphorylates the substrate and consumes ATP at the same time, and the remaining ATP can be converted into light by Ultra-Glo TM luciferase.
  • the luminescent signal is positively correlated with the kinase activity, and the value of this luminescent signal can be detected. Reflects kinase activity.
  • the compound to be tested was dissolved in DMSO (Sigma, product number D8418), and then diluted to a concentration of 200 nM, and the initial concentration of the test was set at 100 nM, with 3-fold dilution and 10 gradients.
  • Add 2.5 ⁇ l of a mixture of Histone H1 protein (SignalChem, Cat. No.
  • X is the logarithmic value of the compound concentration
  • Y is the percentage of inhibition
  • Bottom is the minimum percentage of inhibition
  • Top is the maximum percentage of inhibition
  • HillSlope is the slope of the curve.
  • ADP-Glo Kinase Assay Kit (Promega, Cat. No. V9102) was used to detect the inhibitory activity of the test compound on CDK6/CyclinD1 kinase.
  • the compound to be tested was dissolved in DMSO (Sigma, Cat. No. D8418), and the initial concentration of the test was set to 1000 nM, with 3-fold dilution and 10 gradients.
  • X is the logarithmic value of the compound concentration
  • Y is the percentage of inhibition
  • Bottom is the minimum percentage of inhibition
  • Top is the maximum percentage of inhibition
  • HillSlope is the slope of the curve.
  • ADP-Glo Kinase Assay Kit (Promega, Cat. No. V9102) was used to detect the inhibitory activity of the test compound on CDK4/CyclinD3 kinase.
  • the compound to be tested was dissolved in DMSO (Sigma, Cat. No. D8418), and the initial concentration of the test was set to 1000 nM, with 3-fold dilution and 10 gradients.
  • X is the logarithmic value of the compound concentration
  • Y is the percentage of inhibition
  • Bottom is the minimum percentage of inhibition
  • Top is the maximum percentage of inhibition
  • HillSlope is the slope of the curve.
  • IC 50 values of the compounds of the present invention for the inhibition of CDK2, CDK6 and CDK4 kinases are shown in Table 1 below.
  • Table 1 The compounds of the present invention inhibit the IC50 values of CDK2, CDK6 and CDK4 kinases
  • Test Example 2 The level of cytological inhibition of CDK2/4/6 pathway by the compound of the present invention
  • OVCAR3 and HCC1806 cell lines (both purchased from Nanjing Kebai Biotechnology Co., Ltd., ATCC numbers are HTB- 161TM and CRL-2335 TM ) were cultured in RPMI1640 (Gibco, C11875500BT), adding 10% FBS (Gbico, 10099141) and double antibodies (1% penicillin and streptomycin, Gibco, 15140-122).
  • 5,000 OVCAR3 or HCC1806 cells were seeded in a white transparent bottom 96-well (Nunc, 249944)/384-well plate (Corning, 3570), and placed in a 5% incubator at 37°C overnight. The next day, add the compound to be tested, dissolve the compound with DMSO, and dilute it.
  • the initial concentration of the test starts from 10 mM, and it is diluted 3 times. Set up 10 concentration gradients, and each gradient has 3 replicate wells. Place the cell plate in an incubator at 37°C, 5% CO 2 for 7 days. Using the CELL Titer-GLO luminescence method, the total ATP content was detected to determine the level of cell proliferation.
  • the cells in the 384-well plate were taken out and equilibrated at room temperature for 30 minutes; 30 ⁇ L of CTG (CTG, Promega, Cat. No. G7572) was added to each well, shaken and mixed, and incubated at room temperature for 10 minutes; the fluorescence value was read with a multi-functional microplate reader (Biotek, model Cytation 3).
  • GraphPad Prism 6.0 software analyzed the Log value of the compound response at different concentrations to determine the IC 50 of proliferation inhibition.
  • MCF7 cells Human breast cancer cells (MCF7 cells) (purchased from ATCC, No. HTB-22) were cultured in EMEM medium (ATCC, 30-2003), adding 10% FBS (Gbico, 10099141). After the cells grow to a confluence of 70-80%, the cells are digested to prepare a cell suspension. Cells were seeded in a 384-well plate (Corning, 3570), 500 cells/well, and placed in a 5% incubator at 37°C overnight. The next day, the compound to be tested was added, and the compound was dissolved in DMSO, diluted, and the initial concentration of the test was 10 mM, diluted 3 times, and 10 concentration gradients were set, with 3 replicate wells for each gradient.
  • IC 50 values of the compounds of the present invention for the inhibition of OVCAR3, HCC1806 and MCF-7 cells are shown in Table 2 below.
  • Test example 3 mouse pharmacokinetic experiment of the compound of the present invention
  • the experimental animals were male ICR mice aged 7-8 weeks, purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd., and raised in an SPF environment with a temperature of 20-26°C, a daily temperature difference of no more than 4°C, and a relative humidity of 40-70°C. %RH, 12h/12h alternate lighting every day.
  • the experimental animals went through an adaptation period of 3-5 days, and the animals administered orally were fasted overnight (>12h) one day before the experiment, without water.
  • the vehicles of the intravenous group and the intragastric administration group were both 3% DMSO+97% (20% HP- ⁇ -CD solution).
  • the compound solution preparation process is as follows: first dissolve the compound in DMSO and make a 10 mg/mL stock solution; take 100 ⁇ L of the stock solution, dilute it to 5 mL with 20% HP- ⁇ -CD, and obtain a concentration of 0.2 mg/mL Dosing solution: Take 100 ⁇ L of the stock solution, add solvent to make up to 5 mL, vortex to disperse evenly, and obtain a solution for gavage administration with a compound concentration of 1 mg/mL.
  • Body weight was weighed before administration, and 0.1mL blood samples were collected through the ophthalmic venous plexus and added to heparin sodium anticoagulant tubes to prevent coagulation.
  • Each test compound was administered intravenously to 6 rats, and 6 rats were given oral administration, and food was given 2 hours after administration.
  • the time points of sample collection are: gavage group: 5min, 15min, 30min, 1h, 2h, 4h before administration and after administration; intravenous group: 5min, 15min, 30min, 1h, 2h, 4h before administration and after administration .
  • Animal blood collection was divided into two parts, and cross-time point blood collection was adopted, and a maximum of 5 blood collection points were set for one mouse.
  • F% (AUC po x dose iv )/(AUC iv x dose po ) x 100%.
  • mice The pharmacokinetic parameters of the compounds of the present invention on mice are shown in Table 3 below.

Abstract

一种具有细胞周期蛋白依赖性激酶抑制活性的杂环化合物及其制备方法和医药用途。具体地,涉及通式(I)所示的化合物、其制备方法、含有其的药物组合物,以及其作为细胞周期蛋白依赖性激酶(CDK)抑制剂在预防和/或治疗异常细胞生长如癌症中的用途。其中通式(I)中的各基团的定义与说明书中的定义相同。

Description

具有细胞周期蛋白依赖性激酶抑制活性的杂环化合物及其制备方法和医药用途 技术领域
本发明属于医药技术领域,具体涉及一种嘧啶并吡啶类化合物、及其制备方法及含有其的药物组合物,以及其作为细胞周期蛋白依赖性激酶(CDK)抑制剂在治疗异常细胞生长如癌症的药物中的用途。本发明的化合物可以抑制细胞周期蛋白依赖性激酶CDK2/细胞周期蛋白E1(CCNE1)、CDK6/细胞周期蛋白D1、CDK4/细胞周期蛋白D3,因此可用于治疗癌症,如HR+/HER2-转移性乳腺癌、卵巢癌等。
背景技术
细胞周期蛋白依赖性激酶(CDK)属于丝氨酸/苏氨酸蛋白激酶家族,是调控细胞周期的关键蛋白。目前,已发现的CDK主要包括CDK1-15等,通过与相应的细胞周期蛋白(cyclin)结合,形成稳定形式的CDK/cyclin复合物,来发挥作用。此外,细胞内含有内源性的CDK和CDK/cyclin复合物抑制因子(CKI),这三者共同组成细胞周期的调控网络,在细胞***的过程中受到严格控制。在许多癌细胞中均发现,CDK、细胞周期蛋白和CKI三者表达失控,且CDK/细胞周期蛋白复合物通常是过度表达。
CDK4和CDK6的结构和功能十分相似,且都能与cyclin D的3种亚型(cyclinD1/D2/D3)结合,形成复合物,使包括视网膜母细胞瘤蛋白(Retinoblastoma protein,Rb)在内的一系列底物磷酸化,促使细胞周期进展。磷酸化的Rb蛋白释放与其结合的转录因子E2F等,E2F激活并转录一系列基因,使细胞进入S期,开始DNA复制(Kato等人,Genes Dev,1993,7(3),331-342;Dyson等人,Genes Dev,1998,12(15),2245-2262)。CDK4/6的活性同时受INK4家族(包括p16 INK4A、p15 INK4B、p18 INK4C、p19 INK4D)的负性调控,INK4作为内源性抑制蛋白,可与CDK竞争性结合,抑制cyclin D-CDK4/6复合物的形成(Baker等人,Genes&Cancer,2012,3(11-12),658-669)。
然而,多数人类癌症中均发现,CyclinD-CDK4/6-p16-Rb通路出现异常持续激活,这会促使G1期快速发展,导致细胞异常增殖。其主要原因包括,基因重排或者基因扩增导致cyclin D1的过度表达(Bergsagel等人,Blood,2005,106(1),296-303);p16INK4a基因缺失、点突变、或者DNA甲基化导致p16INK4a失活(Ruas等人,Biochim Biophys Acta,1998,1378(2),F115-177);CDK4/6基因扩增或者点突变(Hamilton等人,Cancer Treatment Reviews,2016,45,129-138)。基于该通路的异常进行靶向干预,使得CDK4/6成为热门的抗肿瘤靶点之一。
靶向CDK的治疗中,进展最快的是选择性CDK4/6抑制剂。第一代广谱类CDK抑制剂Flavopiridol(Alvocidib)可同时抑制CDK1/2/4/6/7/9,但由于其严重的毒 副作用,导致出现乏力、腹泻、骨髓抑制等不良反应,最终未能成功应用于临床(Senderowicz等人,Journal of Clinical Oncology,1998,16(9),2986-2999)。目前有三款化合物Palbociclib、Ribociclib和Abemaciclib获得FDA批准,用于晚期乳腺癌的一线或二线治疗,还有一项药物Lerociclib处于临床开发阶段。
Palbociclib是FDA批准的首个CDK4/6抑制剂。它是一种口服的吡啶类化合物,通过降低Rb蛋白的磷酸化,阻止细胞周期从G1期进入S期,抑制ER+乳腺癌细胞的增殖。多项大型、随机、前瞻性的临床研究已经证实Palbociclib联合来曲唑或Fulvestrant能显著延长患者无进展生存期,且安全性可靠(Cristofanilli等人,Lancet Oncol,2016,17(4),425-439;Richard等人,Lancet Oncology,2015;Finn等人,N Engl J Med,2016,375(20),1925-1936;Nicholas等人,N Engl J Med,2015,373(3),209-219)。这些临床试验数据支持Palbociclib联合芳香化酶抑制剂或氟维斯群作为标准的一线护理方案,治疗ER+/HER2-绝经前或绝经后乳腺癌转移的女性患者。
Ribociclib(Kisqali,LEE011)和Abemaciclib(Verzenio,LY2835219)均是口服可逆的CDK4/6抑制剂,能够通过抑制Rb磷酸化,引起G1期阻滞。Ribociclib对CDK4/6抑制的IC 50值分别为10nM和39nM,而对其他CDK家族成员敏感性较低(CDK1和CDK2的IC 50值均大于50mM)(Sherr等人,Cancer Discovery,2016,6(4),353-367)。口服给药1-4h后,血浆内能达到Cmax值;持续给药8天后达到血液稳定状态。Ribociclib与人血浆蛋白的结合率为70%,在体内能被CPY3A4(一种弱抑制剂)分解,主要产物是N-羟基和N-去甲基衍生物。Abemaciclib对CDK4/6激酶的选择性更高,IC 50值分别为2nM和5nM,而对CDK1和CDK2抑制的IC 50值>500nM。Abemaciclib是一种苯基嘧啶类化合物,结构上与Palbociclib和Ribociclib相近。Abemaciclib表现出更广泛的抑制作用,同时能够抑制Dyrk、PIM、HIPK和CAMK激酶家族(Ki<10nM)(Chen等人,Molecular Cancer Therapeutics,2016,15(10),2273-2281)。体内药代数据显示,口服给药后的4-24小时内达到峰值,持续服用5天后达到稳态。Abemaciclib与人血浆蛋白的结合率约为96%,在体内能被CPY3A4分解,主要分解产物为N-脱乙基衍生物。氘原子的引入,使化合物具有更强的代谢稳定性,代谢稳定性提高了11-45%,表现为较高的半衰期。该药物被批准,与Fluvestrant联合治疗ER+/HER2-晚期或转移性乳腺癌。此外,在异种移植肿瘤中,Abemaciclib与吉西他滨联用,也显示出协同抗肿瘤活性,并且对多种类型肿瘤也具有抑制效果,包括套细胞淋巴瘤(MCL)、结直肠癌、肺癌、胶质母细胞瘤和急性髓细胞白血病(AML)。
尽管CDK4/6抑制剂联合内分泌治疗方法,能给特定类型乳腺癌患者带来显著临床收益,但仍旧有部分患者是治疗无效(10-20%),且70-80%的肿瘤患者在治疗12-36月后会发展为耐药(Tripathy等人,Lancet Oncol,2018,19(7),904-915)。CDK4/6治疗耐药的因素有多种,临床前研究表明,包括Rb丢失、CCND1过表达、 p16扩增、CCNE1-CDK2复合物过度激活(例如,CCNE1在乳腺癌和卵巢癌细胞系模型中高表达)、CDK2的旁路激活、CDK4/6活性增高、CDK7过表达等,这些均可能与CDK4/6治疗耐药相关(Taylor Harding等人,Oncotarget,2014,6(2),696-714;Herrera-Abreu等人,Cancer Res,2016,76(8),2301-2313)。
通过生物标记物方法将乳腺癌患者分为不同亚组,不仅有助于区分从Palbociclib最大获益的的患者亚组,还有助于阐明可能导致CDK4/6联合治疗耐药的机制。Palbociclib最近的III期临床研究中发现,CCNE1(细胞周期蛋白E1)与治疗抵抗密切相关(Turner等人,Journal of Clinical Oncology,2019,37(14),1-11)。CCNE1mRNA低表达的亚组人群,Palbociclib疗效显著。同时,对临床治疗中其它变量分析,CDK4、CDK6、cyclin D1、RB1的表达水平与Palbociclib的治疗效果均无显著关联;该研究引起人们极大兴趣,也证实CCNE1是Palbociclib的耐药的生物标记物。
Cyclin E(细胞周期蛋白E)是由CCNE1编码的蛋白,在G1中期开始表达,在G1-S期表达水平达到高峰后逐渐降解消失,其表达水平受E2F调控,通过泛素化蛋白酶体降解(Mazumder等人,Current Cancer Drug Targets,2004,4(1),65-75)。Cyclin E与CDK2结合,形成复合物,通过磷酸化下游底物Rb、CDC6、NPAT和P107等使细胞进入DNA合成的S期;Cyclin E在维持染色体稳定,调控细胞纺锤体和中心体周期中具有重要作用;Cyclin E在多种恶性肿瘤中存在高表达,且CCNE1基因扩增或过表达还与多种肿瘤的预后差有关,如子宫内膜瘤、胃癌、卵巢癌(George等人,Clinical cancer research,2017,23(7),1862-1874;Nakayama等人,Cancer,2010,116(11),2621-2634;KENTARO等人,Int J Oncol,2015,48(2),506-516;Ooi等人,Human Pathology,2016,S0046817716303082)等。
CDK2通过与伴侣蛋白相互作用并磷酸化激活,在细胞周期调控中发挥关键作用,并参与一系列生物学过程,例如DNA损伤、细胞内转运、蛋白质降解、信号转导、DNA和RNA代谢和翻译等(Tadesse等人,Drug Discovery Today,2020,25(2),406-413)。CDK2在大多数正常组织是低表达状态(McCurdy等人,Oncogene,2016)。***期的细胞,CDK2是关键的细胞周期调节因子,从G1期晚期开始激活,持续表达整个S期。CDK2结合CCNE1或E2和Cyclin A2,被细胞周期蛋白复合物(CDK7、MAT1、细胞周期蛋白H)磷酸化激活,同时,可被CDC25A去磷酸化负调控。在G1后期,CDK2-cyclinE和CDK4/6-cyclin-D共同磷酸化Rb,使Rb释放E2F,启动细胞周期调控基因的转录。除Rb外,CDK2还可调节其他蛋白的磷酸化,使之与细胞周期联系起来。例如,CDK2-cyclin-E对Smad3的磷酸化限制了其转录活性,最终减缓了细胞周期进展(Matsuura等人,Nature,2004,430(6996),226-231)。CDK2还能磷酸化复制前复合物蛋白,这些是启动DNA合成所必需的。如CDC6,它是将微小染色体维持(MCM)蛋白装载到DNA和MCM解旋酶蛋白上,并启动DNA复制的必需蛋白(Chuang等人,Molecular Cell,2009, 35(2),206-216)。CDK2在调控中心体复制方面也起着重要作用,通过靶向磷酸化中心体蛋白如核磷脂(Npm)和cp110,释放中心粒,然后维持中心粒复制(Adon等人,Molecular&Cellular Biology,2010,30(3),694-710;Hu等人,Cancer Research,2015,75(10),2029-2038)。
CDK2的蛋白结构与大多数的蛋白激酶类似,折叠呈“双叶状”。体积较小的N-末端区主要由β-折叠构成,包括5个反平行结构的β-长链和一个C-螺旋。C-α螺旋含有序列PSTAIRE,是周期蛋白结合所必需的。较大的C-末端区域,富含α-螺旋,并包含激活片段(也称为T-环(残基145(天冬氨酸)-172(谷氨酸))和激活磷酸化位点Thr160。T-环结合底物的Ser/Thr(磷酸化受体)区域,进行磷酸化激活,发挥细胞周期调控作用。N-末端和C-末端由柔性铰链区(残基81(Glu)-84(His))连接,该区域形成一个深裂,即ATP结合位点(Pavletich等人,Journal of Molecular Biology,1999,287(5),821-828;Malumbres等人,Chemical Reviews,2014,15(6),122;Honda等人,Embo Journal,2005,24(3))。
CDK2激活和抑制受到以下几种机制调控:在没有丝裂原信号时,CDK2处于非活性状态。在G1期后期,CDK2活性开始升高,这是由于,首先,E2F介导的CCNE基因转录,其蛋白产物结合并激活CDK2;CDK2-cyclin E或A复合物的完全激活,需要CAK磷酸化Thr160位点;再者,Wee1和Myt1激酶能分别抑制Thr14和Tyr15位点的磷酸化,CDC25蛋白磷酸酶家族对这些残基的去磷酸化可以使CDK2重新激活;此外,CDK抑制蛋白家族Cip和Kip,能结合CDK2使其失活,周期蛋白E和A可被泛素连接酶介导的泛素化过程所降解(Solomon等人,Journal of Medicinal Chemistry,2018)。
CDK2还可结合细胞周期蛋白A,参与整个S期的进展,并调控DNA损伤修复。DNA损伤后,DNA损伤反应(DDR)使细胞阻滞在G1/S交界处,以修复受损的DNA,并维持子细胞的基因组保真度。研究发现,有两种机制,均是通过抑制CDK2蛋白磷酸化,使细胞维持在G1/S的DNA损伤检查点,并阻止细胞增殖。首先,p53的积累导致p21Cip1/Waf1转录上调,随后通过抑制cyclin-D1-CDK4/6和cyclinE-CDK2,使细胞周期停滞(Shieh等人,Genes&Development,2000,14(3),289-300)。第二个机制是靶向CDC25A进行降解,WEE1通过持续抑制CDK2蛋白Thr14和Tyr15位置的磷酸化,从而阻止细胞进入S期(Mailand等人,Science,2000,288(5470),1425-1429)。CDK2的靶蛋白,FOXO1在dsDNA断裂触发DNA损伤,从而诱导细胞凋亡中起着重要作用。DNA损伤诱导G1/S期阻滞时,CDK2不再磷酸化FOXO1,使FOXO1发挥转录活性,通过上调多种促凋亡蛋白的表达(FasL、TRAIL和Bim),促使细胞凋亡过程(Huang等人,Science,2006,314(5797),294-297;Huang等人,Cell Cycle,2007,6(8),902-906)。
综合以上,虽然CDK4/6抑制剂已成为特定类型乳腺癌的一线治疗方案,但有些类型乳腺癌对CDK4/6抑制剂是天然的不敏感,如三阴性乳腺癌(TNBC)。另 外在ER+Her2-的乳腺癌中也发现对CDK4/6耐药现象。Cyclin E在多种恶性肿瘤中存在高表达,包括对CDK4/6抑制剂不敏感的肿瘤(TNBC)和卵巢癌等;CDK2与CyclinE形成的复合体,在CDK4/6耐药中也发挥重要作用;靶向CDK2能够延缓CDK4/6抑制剂分子产生的治疗抵抗,对耐药的患者进一步发挥疗效;而在靶向CDK2/4/6来克服CDK4/6耐药的化合物中,未见有获批的药物,仅有Pfizer的PF-06873600进入临床II期研究。因此,开发选择性高、药物毒性低的CDK2/4/6抑制剂,来克服CDK4/6耐药,是一个急需解决的问题。
来自辉瑞PF-06873600的专利(US 2018/0044344 A1)数据显示,在不同肿瘤类型的细胞系中,CCNE1在卵巢癌OVCAR3和乳腺癌HCC1806中具有较高的扩增频率。因此,合成了一系列具有类药性质的小分子CDK2/4/6抑制剂。通过检测对OVCAR3和HCC1806细胞的抑制水平,评估了CDK2抑制的效果;通过检测对MCF-7细胞的抑制水平,评估了CDK4/6抑制的效果。
发明内容
本发明人经过潜心研究,设计合成了一系列嘧啶并吡啶类化合物,其显示出抑制细胞周期蛋白依赖性激酶(CDK)活性,可以被开发为治疗和/或预防与CDK活性相关的疾病如癌症的药物。
因此,本发明的目的是提供一种通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
Figure PCTCN2022119618-appb-000001
其中,
环A选自杂环基,所述杂环基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;
环B选自环烷基、杂环基、芳基和杂芳基;
R 1选自烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、烯基、炔基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;
R 2选自氢、烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中所述 烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、烯基、炔基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;或者,
R 1和R 2与他们连接的氮原子和硫原子一起形成杂环基或杂芳基,所述杂环基或杂芳基任选进一步被选自卤素、氨基、硝基、氰基、氧代基、羟基、巯基、羧基、酯基、烷基、烷氧基、卤代烷基、卤代烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基的一个或多个基团取代;
每个R 3各自独立地选自卤素、氨基、硝基、氰基、羟基、巯基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基,所述烷基、烷氧基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;或者,
任意两个R 3与他们连接的原子一起形成环烷基、杂环基、芳基和杂芳基,所述环烷基、杂环基、芳基和杂芳基任选进一步被选自卤素、氨基、硝基、氰基、氧代基、羟基、巯基、羧基、酯基、烷基、烷氧基、卤代烷基、卤代烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基的一个或多个基团取代;
R 4选自氢、卤素、氨基、硝基、氰基、羟基、巯基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-C(O)R a、-O(O)CR a、-C(O)OR a、-C(O)NR aR b、-NHC(O)R a、-S(O) mR a、-S(O) mNR aR b和-NHS(O) mR a,所述烷基、烷氧基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;
R 5选自氢、卤素、氨基、硝基、氰基、羟基、巯基、烷基、烷氧基、酰基、环烷基、杂环基、芳基、杂芳基、-C(O)R a、-O(O)CR a、-C(O)OR a、-C(O)NR aR b、-NHC(O)R a、-S(O) mR a、-S(O) mNR aR b和-NHS(O) mR a,所述烷基、烷氧基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;
R a和R b各自独立地选自氢、卤素、羟基、氰基、氨基、羧基、酯基、烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基中的一个或多个基团取代;
或者R a和R b与他们连接的原子一起形成环烷基或杂环基,所述环烷基或杂环基任选进一步被选自卤素、氨基、硝基、氰基、氧代基、羟基、巯基、羧基、酯 基、烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基中的一个或多个基团取代;
m为0、1、2;
p为0、1、2、3或4。
在本发明的一个实施方案中,根据本发明所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
其中,
环A选自3至12元单环杂环基、螺杂环基、稠杂环基或桥杂环基,优选5至7元单环杂环基、7至10元螺杂环基、7至10元稠杂环基和7至10元桥杂环基,更优选吡咯烷基、哌啶基、哌嗪基,所述杂环基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、氧代基、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基、C 3-C 6环烷基、5至7元杂环基、C 6-C 10芳基、5至10元杂芳基的一个或多个基团所取代。
在本发明的另一个实施方案中,根据本发明所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(II)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
Figure PCTCN2022119618-appb-000002
其中,环B、R 1、R 2、R 3、R 4、R 5、p如通式(I)所定义。
在本发明的另一个实施方案中,根据本发明所述的通式(I)或通式(II)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
其中,
环B选自C 3-C 7环烷基、4至7元杂环基、C 6-C 10芳基、5至10元杂芳基,更优选
Figure PCTCN2022119618-appb-000003
在本发明的另一个实施方案中,根据本发明所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、 或其混合物形式、或其可药用盐,其为通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
Figure PCTCN2022119618-appb-000004
其中,
其中,R 1、R 2、R 3、R 4、R 5、p如通式(I)所定义。
在本发明的另一个实施方案中,根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其前药、或其可药用盐,
其中,
每个R 3各自独立地选自卤素、氨基、氰基、羟基、巯基、C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 7环烷基、4至7元杂环基,所述C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 7环烷基、4至7元杂环基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;优选卤素、羟基、C 1-C 6烷基、C 1-C 6卤代烷基;
p为0、1、2或3;优选1或2。
在本发明的另一个实施方案中,根据本发明所述的通式(I)、通式(II)、或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
其中,
R 1选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 6-C 10芳基和5至10元杂芳基,优选C 1-C 6烷基、C 3-C 6环烷基;优选C 1-C 6烷基、C 3-C 6环烷基、5至7元杂环基;
所述C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 3-C 6环烷基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、C 1-C 6烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 1-C 6卤代烷基、C 1-C 6烷氧基的一个或多个基团所取代;
所述5至7元杂环基、C 6-C 10芳基和5至10元杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、C 1-C 6烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 1-C 6卤代烷基、C 1-C 6烷氧基、C 6-C 10芳基、5至10元杂芳基的一个或多个基团取代。
在本发明的另一个实施方案中,根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
其中,
R 2选自氢、C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 6-C 10芳基和5至10元杂芳基,优选氢、C 1-C 6烷基、C 3-C 6环烷基;优选氢和C 1-C 6烷基;
所述C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 3-C 6环烷基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、C 1-C 6烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 1-C 6卤代烷基、C 1-C 6烷氧基的一个或多个基团所取代;
所述5至7元杂环基、C 6-C 10芳基和5至10元杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、C 1-C 6烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 1-C 6卤代烷基、C 1-C 6烷氧基、C 6-C 10芳基、5至10元杂芳基的一个或多个基团取代。
在本发明的另一个实施方案中,根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,R 1和R 2与他们连接的氮原子和硫原子一起形成5至7元杂环基,所述5至7元杂环基任选进一步被选自卤素、氨基、硝基、氰基、氧代基、羟基、巯基、羧基、酯基、C 1-C 6烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 1-C 6卤代烷基、C 1-C 6烷氧基、C 6-C 10芳基、5至10元杂芳基的一个或多个基团取代。
在本发明的另一个实施方案中,根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
其中,
R 4选自氢、卤素、氨基、硝基、氰基、羟基、巯基、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基,优选氢、卤素、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基;优选R 4为氢或C 1-C 6烷基。
在本发明的另一个实施方案中,根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
其中,
R 5选自氢、卤素、氨基、硝基、氰基、羟基、巯基、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基、-C(O)R a,优选氢、卤素、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基、-C(O)R a
R a选自C 1-C 6烷基。
本发明典型的化合物包括但不限于以下化合物:
Figure PCTCN2022119618-appb-000005
Figure PCTCN2022119618-appb-000006
Figure PCTCN2022119618-appb-000007
Figure PCTCN2022119618-appb-000008
Figure PCTCN2022119618-appb-000009
Figure PCTCN2022119618-appb-000010
Figure PCTCN2022119618-appb-000011
Figure PCTCN2022119618-appb-000012
Figure PCTCN2022119618-appb-000013
Figure PCTCN2022119618-appb-000014
或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其前药、或其可药用盐。
本发明进一步提供一种根据本发明所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其前药、或其可药用盐的制备方法,其包括以下步骤:
Figure PCTCN2022119618-appb-000015
在碱的存在下,在催化剂存在下,化合物Ij与化合物Ia发生脱水反应得到通式(I)化合物;所述碱优选三乙胺;所述催化剂优选三苯基二氯化磷;
环A、环B、R 1、R 2、R 3、R 4、R 5、p如通式(I)所定义。
本发明进一步提供一种根据本发明所述的通式(II)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其前药、或其可药用盐的制备方法,其包括以下步骤:
Figure PCTCN2022119618-appb-000016
在碱的存在下,在催化剂存在下,化合物IIj与化合物Ia发生脱水反应得到通式(II)化合物;所述碱优选三乙胺;所述催化剂优选三苯基二氯化磷;
环B、R 1、R 2、R 3、R 4、R 5、p如通式(II)所定义。
本发明进一步提供一种根据本发明所述的通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其前药、或其可药用盐的制备方法,其包括以下步骤:
Figure PCTCN2022119618-appb-000017
在碱的存在下,在催化剂存在下,化合物IIIj与化合物Ia发生脱水反应得到通式(III)化合物;所述碱优选三乙胺;所述催化剂优选三苯基二氯化磷;
R 1、R 2、R 3、R 4、R 5、p如通式(III)所定义。
本发明还提供一种药物组合物,其包含本发明所述的通式(I)、通式(II)、或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,和药学上可接受的载体或赋形剂。
本发明的目的还在于提供根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物在制备细胞周期蛋白依赖性激酶(CDK)抑制剂中的用途。
本发明的目的还在于提供根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物在制备抑制癌细胞增值、抑制癌细胞侵袭或诱导癌细胞凋亡的药物中的用途。
本发明的目的还在于提供根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物在制备用于预防和/或治疗与细胞周期蛋白-依赖性激酶活性相关的疾病的药物中的用途,所述疾病例如癌症,特别是与以细胞周期蛋白依赖性激酶CDK2/细胞周期蛋白E1 (CCNE1)、CDK6/细胞周期蛋白D1、CDK4/细胞周期蛋白D3的扩增或过表达为特征的癌症,更特别为乳腺癌如HR+/HER2-转移性乳腺癌或卵巢癌。
本发明还涉及根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物,其用作CDK抑制剂。
本发明还涉及根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物,其用于抑制癌细胞增值、抑制癌细胞侵袭或诱导癌细胞凋亡。
本发明还涉及根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物,其用作用于预防和/或治疗与细胞周期蛋白-依赖性激酶活性相关的疾病,所述疾病例如癌症,特别是与以细胞周期蛋白依赖性激酶CDK2/细胞周期蛋白E1(CCNE1)、CDK6/细胞周期蛋白D1、CDK4/细胞周期蛋白D3的扩增或过表达为特征的癌症,更特别为乳腺癌如HR+/HER2-转移性乳腺癌或卵巢癌的药物。
本发明还涉及一种抑制CDK的方法,其包含向有需要的受试者施用有效量的根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物。
本发明还涉及一种抑制癌细胞增值、抑制癌细胞侵袭或诱导癌细胞凋亡的方法,其包含向有需要的受试者施用有效量的根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物。
本发明还涉及一种预防和/或治疗与细胞周期蛋白-依赖性激酶活性相关的疾病的方法,其包含向有需要的受试者施用有效量的根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物,所述疾病例如癌症,特别是与以细胞周期蛋白依赖性激酶CDK2/细胞周期蛋白E1(CCNE1)、CDK6/细胞周期蛋白D1、CDK4/细胞周期蛋白D3的扩增或过表达为特征的癌症,更特别为乳腺癌如HR+/HER2-转移性乳腺癌或卵巢癌。
根据本发明所述的通式(I)、通式(II)或通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其 混合物形式、或其可药用盐或包含其的药物组合物可以与另外一种抗癌治疗剂或抗癌治疗方法同时、分开或相继施用。
发明的详细说明
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。
“烷基”是指饱和的一价脂族烃基,其包括具有指定碳原子数的直链和支链基团。烷基通常含有1-20个碳原子(C 1-C 20烷基),优选1至12个碳原子(C 1-C 12烷基),更优选1至8个碳原子(C 1-C 8烷基)或1至6个碳原子(C 1-C 6烷基”)或1至4个碳原子(C 1-C 4烷基)。烷基的非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙基己基、2,2-二乙基己基,及其各种支链异构体等。烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代。适用于烷基的任选的取代基包括但不限于C 3-C 8环烷基、3-12元杂环基、C 6-C 12芳基和5-12元杂芳基、卤素、=O(氧代)、=S(硫代)、=N-CN、并且其中每个所述C 3-C 8环烷基、3-12元杂环基、C 6-C 12芳基和5-12元杂芳基任选被取代。烷基上的典型取代基包括卤素、-OH、C 1-C 6烷氧基、-O-C 6-C 12芳基、-CN、=O、C 3-C 8环烷基、C 6-C 12芳基、5-12元杂芳基和3-12元杂环基;其中每个所述C 3-C 8环烷基、C 6-C 12芳基、5-12元杂芳基和3-12元杂环基任选被1-3个独立地选自卤素、-OH、=O、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6羟基烷基、C 1-C 6烷氧基-C 1-C 6烷基、-CN、-NH 2、-NH(C 1-C 6烷基)和-N(C 1-C 6烷基) 2的取代基所取代。在一些实施方案中,烷基任选被一个或多个取代基,优选被1-3个取代基取代,所述取代基独立地选自卤素、-OH、C 1-C 6烷氧基、-O-C 6-C 12芳基、-CN、=O、C 3-C 8环烷基、C 6-C 12芳基、5-12元杂芳基和3-12元杂环基。在其他实施方案中,烷基任选被一个或多个取代基,优选被1-3个取代基取代,所述取代基独立地选自卤素、-OH、C 1-C 6烷氧基、-CN、C 3-C 8环烷基、3-12元杂环基、C 6-C 12芳基和5-12元杂芳基,其各自任选含有1、2或3个选自O、N和S(O)x(其中x是0-2)的另外的杂原子;且其中每个所述环烷基、杂环基、芳基或杂芳基任选被1-3个独立地选自卤素、-OH、=O、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6羟基烷基、C 1-C 6烷氧基-C 1-C 6烷基、-CN、-NH 2、-NH(C 1-C 6烷基)和-N(C 1-C 6烷 基) 2的取代基所取代。
本文所述的任选取代的烷基可以被一个或多个取代基取代,除非另有说明,否则这些取代基是独立选择的。在这种取代具有化学意义的程度上,取代基的总数可以等于烷基基团上的氢原子总数。任选取代的烷基通常含有1至6个任选的取代基,有时1至5个任选的取代基,优选1至4个任选的取代基,或更优选1至3个任选的取代基。特别地,除非另外指出,烷基可以被一个或多个(直至烷基基团上存在的氢原子总数)卤素基团取代。因此,C 1-C 4烷基包括卤代烷基,特别是具有1至4个碳原子的氟代烷基,例如三氟甲基或二氟乙基(即CF 3和-CH 2CHF 2)。
术语“烯基”指由至少由两个碳原子和至少一个碳-碳双键组成的如上定义的烷基,例如乙烯基、1-丙烯基、2-丙烯基、1-、2-或3-丁烯基等。烯基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基。
术语“炔基”指由至少由两个碳原子和至少一个碳-碳三键组成的如上定义的烷基,例如乙炔基、丙炔基、丁炔基等。炔基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基。
术语“环烷基”指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包含3至20个碳原子,优选包含3至12个碳原子,更优选包含3至6个碳原子。单环环烷基的非限制性实例包括环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等;多环环烷基包括螺环、稠环和桥环的环烷基。
术语“螺环烷基”指5至20元的单环之间共用一个碳原子(称螺原子)的多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺环烷基分为单螺环烷基、双螺环烷基或多螺环烷基,优选为单螺环烷基和双螺环烷基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺环烷基。螺环烷基的非限制性实例包括:
Figure PCTCN2022119618-appb-000018
术语“稠环烷基”指5至20元,***中的每个环与体系中的其他环共享毗邻的一对碳原子的全碳多环基团,其中一个或多个环可以含有一个或多个双键,但 没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠环烷基,优选为双环或三环,更优选为5元/5元或5元/6元双环烷基。稠环烷基的非限制性实例包括:
Figure PCTCN2022119618-appb-000019
术语“桥环烷基”指5至20元,任意两个环共用两个不直接连接的碳原子的全碳多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥环烷基,优选为双环、三环或四环,更有选为双环或三环。桥环烷基的非限制性实例包括:
Figure PCTCN2022119618-appb-000020
所述环烷基环可以稠合于芳基、杂芳基或杂环烷基环上,其中与母体结构连接在一起的环为环烷基,非限制性实例包括茚满基、四氢萘基、苯并环庚烷基等。环烷基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其包含3至20个环原子,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包含3至12个环原子,其中1~4个是杂原子;最优选包含3至8个环原子,其中1~3个是杂原子;最优选包含5至7个环原子,其中1~2或1~3个是杂原子。单环杂环基的非限制性实例包括吡咯烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、二氢咪唑基、二氢呋喃基、二氢吡唑基、二氢吡咯基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基、吡喃基等,优选1、2、5-噁二唑基、吡喃基或吗啉基。多环杂环基包括螺环、稠环和桥环的杂环基。
术语“螺杂环基”指5至20元的单环之间共用一个原子(称螺原子)的多环杂环基团,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺杂环基分为单螺杂环基、双螺杂环基或多螺杂环基,优选为单螺杂环基和双螺杂环基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或 5元/6元单螺杂环基。螺杂环基的非限制性实例包括:
Figure PCTCN2022119618-appb-000021
术语“稠杂环基”指5至20元,***中的每个环与体系中的其他环共享毗邻的一对原子的多环杂环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠杂环基,优选为双环或三环,更优选为5元/5元或5元/6元双环稠杂环基。稠杂环基的非限制性实例包括:
Figure PCTCN2022119618-appb-000022
术语“桥杂环基”指5至14元,任意两个环共用两个不直接连接的原子的多环杂环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥杂环基,优选为双环、三环或四环,更有选为双环或三环。桥杂环基的非限制性实例包括:
Figure PCTCN2022119618-appb-000023
所述杂环基环可以稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基,其非限制性实例包括:
Figure PCTCN2022119618-appb-000024
等。
杂环基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、 杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“芳基”指具有共轭的π电子体系的6至14元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为6至10元,例如苯基和萘基。更优选苯基。所述芳基环可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环,其非限制性实例包括:
Figure PCTCN2022119618-appb-000025
芳基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“杂芳基”指包含1至4个杂原子、5至14个环原子的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元,含1至3个杂原子;更优选为5元或6元,含1至2个杂原子;优选例如咪唑基、呋喃基、噻吩基、噻唑基、吡唑基、噁唑基、吡咯基、四唑基、吡啶基、嘧啶基、噻二唑、吡嗪基等,优选为咪唑基、噻唑基、吡唑基或嘧啶基、噻唑基;更有选吡唑基或噻唑基。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,其非限制性实例包括:
Figure PCTCN2022119618-appb-000026
杂芳基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“烷氧基”指-O-(烷基)和-O-(环烷基),其中烷基和环烷基的定义如上所述。烷氧基的非限制性实例包括:甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基。烷氧基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“卤代烷基”指被一个或多个卤素取代的烷基,其中烷基如上所定义。
术语“卤代烷氧基”指被一个或多个卤素取代的烷氧基,其中烷氧基如上所定义。
术语“羟基”指-OH基团。
术语“卤素”指氟、氯、溴或碘。
术语“氨基”指-NH 2
术语“氰基”指-CN。
术语“硝基”指-NO 2
术语“氧代基”指=O。
术语“羧基”指-C(O)OH。
术语“巯基”指-SH。
术语“酯基”指-C(O)O(烷基)或-C(O)O(环烷基),其中烷基和环烷基如上所定义。
术语“酰基”指含有-C(O)R基团的化合物,其中R为烷基、环烷基、杂环基、芳基、杂芳基。
“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生的场合。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上/可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学/可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
“可药用盐”是指本发明化合物的盐,这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。
“癌症”是指任何恶性和/或侵袭性生长或肿瘤(由异常细胞生长引起)。癌症包括以形成它们的细胞类型命名的实体瘤,血液、骨髓或淋巴***的癌症。实体瘤的实例包括肉瘤和癌。血液癌症包括但不限于白血病、淋巴瘤和骨髓瘤。癌症还包括起源于身体具体部位的原发性癌症,已经从其开始的部位扩散到身体的其它部位的转移性癌症,缓解后从原始的原发性癌症的复发,以及第二原发性癌症(这是在具有与新的原发性癌症不同类型的先前癌症病史的人中的新的原发性癌症)。在本文提供的一些实施方案中,癌症可以选自乳腺癌、卵巢癌、膀胱癌、子宫癌、***癌、肺癌、食道癌、肝癌、胰腺癌和胃癌。在一些这种实施方案中,癌症的特征在于CCNE1和/或CCNE2的扩增或过表达。
本文所述的立体异构体可包括本发明化合物(包括表现出超过一种异构类型的化合物)的顺式和反式异构体、光学异构体如(R)和(S)对映异构体、非对映异构体、几何异构体、旋转异构体、阻转异构体、构象异构体和互变异构体;及其混合物(如外消旋体和非对映异构体对)。
本发明化合物可以表现出互变异构现象和结构异构现象。例如,化合物可以数种互变异构形式,包括烯醇和亚胺形式以及酮和烯胺形式,和几何异构体及其混合物存在。所有这种互变异构形式都包括在本发明化合物的范围内。互变异构体作为在溶液中的互变异构组的混合物存在。在固体形式中,通常一种互变异构体占优势。即使可以描述一种互变异构体,但本发明包括所提供的化合物的所有互变异构体。
用于制备/分离单一的对映异构体的常规技术包括由合适的光学纯的前体手性合成或利用例如手性高压液相色谱(HPLC)或超临界流体色谱(SFC)拆分外消旋体(或盐或衍生物的外消旋体)。
本文所述化合物的对映异构体纯度可以用对映异构体过量(ee)来描述,其表示样品以比另一种对映异构体更大的量含有一种对映异构体的程度。外消旋混合物的ee为0%,而单一的完全纯的对映异构体的ee为100%。类似地,非对映异构体纯度可以用非对映异构体过量(de)来描述。
按照本发明所属领域的常规方法,本发明通式(I)所示的化合物可以与碱或者酸生成药学上可接受的碱式加成盐或酸式加成盐。所述碱包括无机碱和有机碱,可接受的有机碱包括二乙醇胺、乙醇胺、N-甲基葡糖胺、三乙醇胺、氨丁三醇等,可接受的无机碱包括氢氧化铝、氢氧化钙、氢氧化钾、碳酸钠和氢氧化钠等。所述酸包括无机酸和有机酸,可接受的无机酸包括盐酸、硫酸、硝酸、磷酸、氢溴酸等。可接受的有机酸包括乙酸、三氟乙酸、甲酸、抗环血酸等。
含活性成分的药物组合物可以是适用于口服的形式,例如片剂、糖锭剂、锭剂、水或油混悬液、可分散粉末或颗粒、乳液、硬或软胶囊,或糖浆剂或酏剂。可按照本领域任何已知制备药用组合物的方法制备口服组合物,此类组合物可含有一种或多种选自以下的成分:甜味剂、矫味剂、着色剂和防腐剂,以提供悦目和可口的药用制剂。片剂含有活性成分和用于混合的适宜制备片剂的无毒的可药用的赋形剂。这些赋形剂可以是惰性赋形剂,如碳酸钙、碳酸钠、乳糖、磷酸钙或磷酸钠;造粒剂和崩解剂,例如微晶纤维素、交联羧甲基纤维素钠、玉米淀粉或藻酸;粘合剂,例如淀粉、明胶、聚乙烯吡咯烷酮或***胶;和润滑剂,例如硬脂酸镁、硬脂酸或滑石粉。这些片剂可以不包衣或可通过掩盖药物的味道或在胃肠道中延迟崩解和吸收,因而在较长时间内提供缓释作用的已知技术将其包衣。例如,可使用水溶性味道掩蔽物质,例如羟丙基甲基纤维素或羟丙基纤维素,或延长时间物质例如乙基纤维素、醋酸丁酸纤维素。
也可用其中活性成分与惰性固体稀释剂例如碳酸钙、磷酸钙或高岭土混合的 硬明胶胶囊,或其中活性成分与水溶性载体例如聚乙二醇或油溶媒例如花生油、液体石蜡或橄榄油混合的软明胶胶囊提供口服制剂。
水混悬液含有活性物质和用于混合的适宜制备水混悬液的赋形剂。此类赋形剂是悬浮剂,例如羧基甲基纤维素钠、甲基纤维素、羟丙基甲基纤维素、藻酸钠、聚乙烯吡咯烷酮和***胶;分散剂或湿润剂,可以是天然产生的磷脂例如卵磷脂,或烯化氧与脂肪酸的缩合产物,例如聚氧乙烯硬脂酸酯,或环氧乙烷与长链脂肪醇的缩合产物,例如十七碳亚乙基氧基鲸蜡醇(heptadecaethyleneoxy cetanol),或环氧乙烷与由脂肪酸和己糖醇衍生的部分酯的缩合产物,例如聚环氧乙烷山梨醇单油酸酯,或环氧乙烷与由脂肪酸和己糖醇酐衍生的偏酯的缩合产物,例如聚环氧乙烷脱水山梨醇单油酸酯。水混悬液也可以含有一种或多种防腐剂例如尼泊金乙酯或尼泊金正丙酯、一种或多种着色剂、一种或多种矫味剂和一种或多种甜味剂,例如蔗糖、糖精或阿司帕坦。
油混悬液可通过使活性成分悬浮于植物油如花生油、橄榄油、芝麻油或椰子油,或矿物油例如液体石蜡中配制而成。油混悬液可含有增稠剂,例如蜂蜡、硬石蜡或鲸蜡醇。可加入上述的甜味剂和矫味剂,以提供可口的制剂。可通过加入抗氧化剂例如丁羟茴醚或α-生育酚保存这些组合物。
通过加入水,适用于制备水混悬液的可分散粉末和颗粒可以提供活性成分和用于混合的分散剂或湿润剂、悬浮剂或一种或多种防腐剂。适宜的分散剂或湿润剂和悬浮剂如上所述。也可加入其他赋形剂例如甜味剂、矫味剂和着色剂。通过加入抗氧化剂例如抗坏血酸保存这些组合物。
本发明的药物组合物也可以是水包油乳剂的形式。油相可以是植物油例如橄榄油或花生油,或矿物油例如液体石蜡或其混合物。适宜的乳化剂可以是天然产生的磷脂,例如大豆卵磷脂,和由脂肪酸和己糖醇酐衍生的酯或偏酯,例如山梨坦单油酸酯,和所述偏酯和环氧乙烷的缩合产物,例如聚环氧乙烷山梨醇单油酸酯。乳剂也可以含有甜味剂、矫味剂、防腐剂和抗氧剂。可用甜味剂例如甘油、丙二醇、山梨醇或蔗糖配制的糖浆和酏剂。此类制剂也可含有缓和剂、防腐剂、着色剂和抗氧剂。
本发明的药物组合物可以是无菌注射水溶液形式。可以使用的可接受的溶媒和溶剂有水、林格氏液和等渗氯化钠溶液。无菌注射制剂可以是其中活性成分溶于油相的无菌注射水包油微乳。例如将活性成分溶于大豆油和卵磷脂的混合物中。然后将油溶液加入水和甘油的混合物中处理形成微乳。可通过局部大量注射,将注射液或微乳注入患者的血流中。或者,最好按可保持本发明化合物恒定循环浓度的方式给予溶液和微乳。为保持这种恒定浓度,可使用连续静脉内递药装置。
本发明的药物组合物可以是用于肌内和皮下给药的无菌注射水或油混悬液的形式。可按已知技术,用上述那些适宜的分散剂或湿润剂和悬浮剂配制该混悬液。无菌注射制剂也可以是在无毒肠胃外可接受的稀释剂或溶剂中制备的无菌注射溶 液或混悬液,例如在1,3-丁二醇中制备的溶液。此外,可方便地用无菌固定油作为溶剂或悬浮介质。为此目的,可使用包括合成甘油单或二酯在内的任何调和固定油。此外,脂肪酸例如油酸也可以制备注射剂。
可按用于直肠给药的栓剂形式给予本发明化合物。可通过将药物与在普通温度下为固体但在直肠中为液体,因而在直肠中会溶化而释放药物的适宜的无刺激性赋形剂混合来制备这些药物组合物。此类物质包括可可脂、甘油明胶、氢化植物油、各种分子量的聚乙二醇和聚乙二醇的脂肪酸酯的混合物。
本领域技术人员熟知,药物的给药剂量依赖于多种因素,包括但并非限定于以下因素:所用特定化合物的活性、病人的年龄、病人的体重、病人的健康状况、病人的行被、病人的饮食、给药时间、给药方式、***的速率、药物的组合等。另外,最佳的治疗方式如治疗的模式、通式化合物的日用量或可药用的盐的种类可以根据传统的治疗方案来验证。
本发明可以含有通式(I)所示的化合物,及其药学上可接受的盐、水合物或溶剂化物作为活性成分,与药学上可接受的载体或赋型剂混合制备成组合物,并制备成临床上可接受的剂型。本发明的衍生物可以与其他活性成分组合使用,只要它们不产生其他不利的作用,例如过敏反应等。本发明化合物可作为唯一的活性成分,也可以与其它治疗与酪氨酸激酶活性相关的疾病的药物联合使用。联合治疗通过将各个治疗组分同时、分开或相继给药来实现。
本发明化合物的合成方法
为了完成本发明的目的,本发明采用如下合成方案制备本发明的通式(I)化合物。
本发明采用如下方案1制备本发明的通式(I)所示的化合物。
Figure PCTCN2022119618-appb-000027
方案1
步骤1:在极性非质子溶剂(如二氯甲烷)中,在适合的碱(如三乙胺)存在的条件下,磺酰氯化合物发生反应得到磺酰胺化合物Ia;
步骤2:在合适的溶剂中,在适合的还原剂存在下,4-氯-2-(甲硫基)嘧啶-5-羧酸乙酯发生还原反应得到化合物Ib;所述溶剂例如四氢呋喃,所述还原剂例如二异丁基氢化铝;
步骤3:在合适的溶剂中,在适合的碱的存在下,化合物Ib与化合物Ic反应得到化合物Id;所述溶剂例如异丙醇,所述碱例如二异丙基乙胺,反应温度可以为室温至80℃之间;
步骤4:在合适的溶剂中,在适合的氧化剂存在下,化合物Id发生氧化反应得到化合物Ie;所述溶剂例如乙酸乙酯,所述氧化剂例如二氧化锰,反应温度可以在50℃至80℃之间;
步骤5:在合适的溶剂中,在适合的催化剂存在下,化合物Ie发生Aldol环化反应(参见VanderWel等人,J.Med.Chezn.2005,4S,2371),得到化合物If;所述溶剂例如四氢呋喃,所述催化剂例如LHMDS,反应温度可以为-20℃至室温之间,反应在氮气氛的条件下进行;
步骤6:在合适的溶剂中,在适合的氧化剂存在下,化合物If发生氧化反应得到化合物Ig;所述溶剂特别为混合溶剂,如2-甲基四氢呋喃和水的混合溶剂,所述氧化剂包括但不限于过硫酸氢钾、间氯过氧苯甲酸等,过量的氧化剂有助于反应彻底进行,反应温度可以为10℃至80℃之间;
步骤7:在合适的溶剂中,在适合的碱存在下,化合物Ig与化合物Ih发生反应得到化合物Ii;所述溶剂例如异丙醇、2-甲基四氢呋喃,所述碱例如二异丙基乙胺,反应温度可以在室温至80℃之间;
步骤8:在合适的溶剂中,在适当的酸的存在下,将化合物Ii脱保护得到化合物Ij;所述溶剂例如二氧六环,所述酸例如盐酸,反应温度典型地在室温进行;进一步通过如氢氧化钠、氢氧化钾的碱将化合物游离;
步骤9:在合适的溶剂中,在适当的碱和催化剂的存在下,化合物Ij与化合物Ia发生脱水反应得到通式(I)化合物;所述溶剂例如二氯甲烷,所述碱例如三乙胺,所述催化剂例如三苯基二氯化磷;反应典型地在氮气氛下反应进行;反应温度典型地为0℃至室温;
其中,环A、环B、R 1、R 2、R 3、R 4、R 5、p如通式(I)所定义。
本发明采用如下方案2制备本发明的通式(II)化合物。
Figure PCTCN2022119618-appb-000028
方案2
步骤1:在极性非质子溶剂(如二氯甲烷)中,在适合的碱(如三乙胺)存在的条件下,磺酰氯化合物发生反应得到磺酰胺化合物Ia;
步骤2:在合适的溶剂中,在适合的还原剂存在下,4-氯-2-(甲硫基)嘧啶-5-羧酸乙酯发生还原反应得到化合物Ib;所述溶剂例如四氢呋喃,所述还原剂例如二异丁基氢化铝;
步骤3:在合适的溶剂中,在适合的碱的存在下,化合物Ib与化合物Ic反应 得到化合物Id;所述溶剂例如异丙醇,所述碱例如二异丙基乙胺,反应温度可以为室温至80℃之间;
步骤4:在合适的溶剂中,在适合的氧化剂存在下,化合物Id发生氧化反应得到化合物Ie;所述溶剂例如乙酸乙酯,所述氧化剂例如二氧化锰,反应温度可以在50℃至80℃之间;
步骤5:在合适的溶剂中,在适合的催化剂存在下,化合物Ie发生Aldol环化反应(参见VanderWel等人,J.Med.Chezn.2005,4S,2371),得到化合物If;所述溶剂例如四氢呋喃,所述催化剂例如LHMDS,反应温度可以为-20℃至室温之间,反应在氮气氛的条件下进行;
步骤6:在合适的溶剂中,在适合的氧化剂存在下,化合物If发生氧化反应得到化合物Ig;所述溶剂特别为混合溶剂,如2-甲基四氢呋喃和水的混合溶剂,所述氧化剂包括但不限于过硫酸氢钾、间氯过氧苯甲酸等,过量的氧化剂有助于反应彻底进行,反应温度可以为10℃至80℃之间;
步骤7:在合适的溶剂中,在适合的碱存在下,化合物Ig与化合物IIh发生反应得到化合物IIi;所述溶剂例如异丙醇、2-甲基四氢呋喃,所述碱例如二异丙基乙胺,反应温度可以在室温至80℃之间;
步骤8:在合适的溶剂中,在适当的酸的存在下,将化合物IIi脱保护得到化合物IIj;所述溶剂例如二氧六环,所述酸例如盐酸,反应温度典型地在室温进行;进一步通过如氢氧化钠、氢氧化钾的碱将化合物游离;
步骤9:在合适的溶剂中,在适当的碱和催化剂的存在下,化合物IIj与化合物Ia发生脱水反应得到通式(II)化合物;所述溶剂例如二氯甲烷,所述碱例如三乙胺,所述催化剂例如三苯基二氯化磷;反应典型地在氮气氛下反应进行;反应温度典型地为0℃至室温;
其中,环B、R 1、R 2、R 3、R 4、R 5、p如通式(II)所定义。
本发明采用如下方案3制备本发明的通式(III)化合物。
Figure PCTCN2022119618-appb-000029
方案3
步骤1:在极性非质子溶剂(如二氯甲烷)中,在适合的碱(如三乙胺)存在的条件下,磺酰氯化合物发生反应得到磺酰胺化合物Ia;
步骤2:在合适的溶剂中,在适合的还原剂存在下,4-氯-2-(甲硫基)嘧啶-5-羧酸乙酯发生还原反应得到化合物Ib;所述溶剂例如四氢呋喃,所述还原剂例如二异丁基氢化铝;
步骤3:在合适的溶剂中,在适合的碱的存在下,化合物Ib与化合物IIIc反应得到化合物IIId;所述溶剂例如异丙醇,所述碱例如二异丙基乙胺,反应温度可以为室温至80℃之间;
步骤4:在合适的溶剂中,在适合的氧化剂存在下,化合物IIId发生氧化反应得到化合物IIIe;所述溶剂例如乙酸乙酯,所述氧化剂例如二氧化锰,反应温度可以在50℃至80℃之间;
步骤5:在合适的溶剂中,在适合的催化剂存在下,化合物IIIe发生Aldol环化反应(参见VanderWel等人,J.Med.Chezn.2005,4S,2371),得到化合物IIIf;所述溶剂例如四氢呋喃,所述催化剂例如LHMDS,反应温度可以为-20℃至室温之间,反应在氮气氛的条件下进行;
步骤6:在合适的溶剂中,在适合的氧化剂存在下,化合物IIIf发生氧化反应得到化合物IIIg;所述溶剂特别为混合溶剂,如2-甲基四氢呋喃和水的混合溶剂,所述氧化剂包括但不限于过硫酸氢钾、间氯过氧苯甲酸等,过量的氧化剂有助于反应彻底进行,反应温度可以为10℃至80℃之间;
步骤7:在合适的溶剂中,在适合的碱存在下,化合物IIIg与化合物IIh发生反应得到化合物IIIi;所述溶剂例如异丙醇、2-甲基四氢呋喃,所述碱例如二异丙基乙胺,反应温度可以在室温至80℃之间;
步骤8:在合适的溶剂中,在适当的酸的存在下,将化合物IIIi脱保护得到化合物IIIj;所述溶剂例如二氧六环,所述酸例如盐酸,反应温度典型地在室温进行;进一步通过如氢氧化钠、氢氧化钾的碱将化合物游离;
步骤9:在合适的溶剂中,在适当的碱和催化剂的存在下,化合物IIIj与化合物Ia发生脱水反应得到通式(III)化合物;所述溶剂例如二氯甲烷,所述碱例如三乙胺,所述催化剂例如三苯基二氯化磷;反应典型地在氮气氛下反应进行;反应温度典型地为0℃至室温;
其中,R 1、R 2、R 3、R 4、R 5、p如通式(III)所定义。
具体实施方式
进一步通过实施例来理解本发明的化合物及其制备,这些实施例说明了一些制备或使用所述化合物的方法。然而,要理解的是,这些实施例并不限制本发明的范围。现在已知的或进一步开发的本发明的变化被认为落入本文中描述的和要求保护的本发明范围之内。
本发明化合物是利用便利的起始原料和通用的制备步骤来完成制备的。本发明给出了典型的或倾向性的反应条件,诸如反应温度、时间、溶剂、压力、反应物的摩尔比。但是除非特殊说明,其他反应条件也能采纳。优化条件可能随着具体的反应物或溶剂的使用而改变,但在通常情况下,反应优化步骤和条件都能得到确定。
另外,本发明中可能用到了一些保护基团来保护某些官能团避免不必要的反应。适宜于各种官能团的保护基以及它们的保护或脱保护条件已经为本领域技术人员广泛熟知。例如T.W.Greene和G.M.Wuts的《有机制备中的保护基团》(第3版,Wiley,New York,1999和书中的引用文献)详细描述了大量的保护基团的保护或脱保护。
化合物和中间体的分离和纯化依据具体的需求采取适当的方法和步骤,例如过滤、萃取、蒸馏、结晶、柱层析、制备薄层板色谱、制备高效液相色谱或上述方法的混合使用。其具体使用方法可参阅本发明描述的实例。当然,其他类似的分离和纯化手段也是可以采用的。可以使用常规方法(包括物理常数和波谱数据)对其进行表征。
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移以10 -6(ppm)的单位给出。NMR的测定是用Brukerdps 300型核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d 6)、氘代氯仿(CDCl 3)、氘代甲醇(CD 3OD),内标为四甲基硅烷(TMS)。
MS的测定用LC(Waters 2695)/MS(Quattro Premier xE)质谱仪(生产商:沃特世)(Photodiode Array Detector)。
制备液相色谱法使用lc6000高效液相色谱仪(生产商:创新通恒)。色谱柱为DaisogelC18 10μm 100A(30mm×250mm),流动相:乙腈/水。
薄层层析硅胶板使用青岛海洋化工GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.20mm~0.25mm,制备薄层层析分离纯化产品采用的规格是0.5mm。
柱层析一般使用青岛海洋硅胶100~200目、200~300目和300~400目硅胶为载体。
本发明的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自网化商城、北京偶合、Sigma、百灵威、易世明、上海书亚、上海伊诺凯、安耐吉化学、上海毕得等公司。
实施例中无特殊说明,反应能够均在氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
反应溶剂,有机溶剂或惰性溶剂各自表述为使用的该溶剂在所描述的反应条件下不参与反应,包括如苯、甲苯、乙腈、四氢呋喃(THF)、二甲基甲酰胺(DMF)、氯仿、二氯甲烷、***、甲醇、氮-甲基吡咯碄酮(NMP)、吡啶等。实施例中无特殊说明,溶液是指水溶液。
本发明中所描述的化学反应一般在常压下进行。反应温度在-78℃至200℃之间。反应时间和条件为,例如,一个大气压下,-78℃至200℃之间,大约1至24小时内完成。如果反应过夜,则反应时间一般为16小时。实施例中无特殊说明,反应的温度为室温,为20℃~30℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂的体系有:A:二氯甲烷和甲醇体系,B:石油醚和乙酸乙酯体系,C:丙酮,溶剂的体积比根据化合物的极性不同而进行调节。
纯化化合物采用的柱层析的洗脱剂的体系和薄层色谱法的展开剂体系包括:A:二氯甲烷和甲醇体系,B:石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺和三氟乙酸等碱性或酸性试剂进行调节。
除非另行定义,文中所使用的所有专业与科学用语与本领域熟练人员所熟悉的意义相同。此外,任何与所记载内容相似或均等的方法及材料皆可应用于本发明方法中。
根据本文提供的示例性程序及其本领域技术人员已知的改变来制备本发明的化合物。
缩略语
Ac=乙酰基
AcO或OAc=乙酰氧基
ACN=乙腈
aq=水溶液
Boc=叔丁氧基羰基
Bn=苄基
DIPEA=二异丙基乙基胺
DMAP=4-二甲氨基吡啶
DMF=N,N-二甲基甲酰胺
DMSO=二甲亚砜
DCM=二氯甲烷
EA=乙酸乙酯
EDTA=乙二胺四乙酸
HPLC=高效液相色谱法
IC 50=抑制50%活性的浓度
LHMDS=六甲基二硅氮烷锂盐(双(三甲基甲硅烷基)氨基锂)
LC-MS=液相色谱-质普联用
M+H +=母体化合物质量+一质子
Ms=甲磺酰基
MS=质谱
MsCl=甲基磺酰氯
mCPBA=间氯过氧苯甲酸
NBS=N-溴代丁二酰亚胺
NCS=N-氯代丁二酰亚胺
NIS=N-碘代丁二酰亚胺
NMR=核磁共振
OXONE=过硫酸氢钾
PE=石油醚
Pro=保护基
TBS=叔丁基二甲基硅基
TBHP=叔丁基过氧化氢
TEA=三乙胺
TFA=三氟乙酸
THF=四氢呋喃
实施例
实施例1:2-((1-(环丙烷磺酰亚氨基)哌啶-4-基)氨基)-8-(((1R,2R)-2-羟基-2-甲基环戊基)吡啶并[2,3-d]嘧啶-7(8H)-酮(1)的制备
Figure PCTCN2022119618-appb-000030
步骤1:1-甲基-6-氧杂二环[3.1.0]己烷(1a)的制备
于室温,将1-甲基环戊-1-烯(112g,1366mmol)、DCM(4400mL)加到反应瓶中,然后将间氯过氧苯甲酸(496g,2049mmol,71%wt)分批加入瓶中,室温搅拌反应过夜。反应结束后,加硅藻土抽滤,加入饱和碳酸氢钠(2500mL)、10%硫代硫酸钠(500mL),有机相在低于20℃的水浴锅中浓缩得到125g棕色油状物的标题产物,直接用于下一步。
LCMS:m/z 99.07[M+H] +
步骤2:(±)-(1R*,2R*)-2-(苄基氨基)-1-甲基环戊-1-醇((±)1b)的制备
于室温,向玻璃封管中装入水(250mL)和苄胺(146g,1366mmol),然后用氮气吹扫5min,然后加入1-甲基-6-氧杂双环[3.1.0]己烷(125g,粗品)。100℃加热18h,此时观察到两相混合物。冷却至室温后,加入浓盐酸水溶液(约12M,150mL)以使pH达到1。用乙酸乙酯(1500mL)萃出有机杂质。将酸性水层在冰水浴中冷却,并使用5N氢氧化钠水溶液调节pH至10。用乙酸乙酯(1200mL×3)萃取所得的两相混合物,合并的有机相经无水硫酸钠干燥,过滤并减压浓缩,得棕色油状物。于80℃减压浓缩5h,降至室温,在石油醚中分散,过滤得到85.0g棕色固体状的标题产物。
LCMS:m/z 206.15[M+H] +
步骤3:(1R,2R)-2-(苄基氨基)-1-甲基环戊-1-醇(1c)的制备
于室温,在反应瓶中加入(±)-(1R*,2R*)-2-(苄基氨基)-1-甲基环戊-1-醇(85.0g,414mmol)和乙醇(600mL),80℃加热30min。向另一反应瓶中加入(R)-2-(3,5-二硝基苯甲酰氨基)-2-苯基乙酸(71.4g,207mmol)和乙醇(1200mL),80℃加热直至固体溶解,并继续搅拌30min。将来自第一个反应瓶的醇的热溶液以稳定流速倒入第二个反应瓶中热的手性酸溶液中。反应混合物保持澄清约1min,然后开始沉淀。5min后,形成浓稠的白色悬浮液,但不妨碍搅拌。在80℃继续搅拌4h,然后停止加热并继续搅拌混合物,同时逐渐冷却至室温过夜。过滤收集母液,浓缩得到45g棕色固体。将该棕色固体悬浮于1L分液漏斗中的水(250mL)和乙酸乙酯(500mL)中。加入盐酸水溶液(4M,150mL,600mmol)并将混合物搅拌约30秒。获得澄清的双相混合物。分离各层,并将有机层用盐酸水溶液(4M,100mL×3)进一步洗涤。合并酸性水层,在冰水浴中冷却,加入氢氧化钠水溶液(aq),使pH达到10。在该pH下形成白色悬浮液。用饱和氯化钠水溶液(250mL)稀释,并用乙酸乙酯(1000mL×3)萃取。将合并的有机相经无水硫酸钠干燥,过滤并减压浓缩,得到26.0g棕色固体状的标题产物,直接用于下一步。
LCMS:m/z 206.15[M+H] +
步骤4:(1R,2R)-2-氨基-1-甲基环戊-1-醇(1d)的制备
于室温,将20%氢氧化钯碳(2.6g)加入到(1R,2R)-2-氨基-1-甲基环戊-1-醇(26.0g,127mmol)的异丙醇(250mL)溶液中,50℃在氢气环境中搅拌16h。通过硅藻土过滤除去催化剂,滤液浓缩得到15.0g红棕色油状物的标题产物,直接用于下一步。
LCMS:m/z 116.10[M+H] +
步骤5:(4-氯-2-(甲硫基)嘧啶-5-基)甲醇(1e)的制备
于室温,将4-氯-2-(甲硫基)嘧啶-5-羧酸乙酯(76.6g,330mmol)、四氢呋喃(3000mL)加入到反应瓶中,在氮气氛下,于-78℃将二异丁基氢化铝(990mL,1M四氢呋喃溶液,990mmol)慢慢滴加到反应瓶中,期间控制温度不超过-60℃,搅拌反应过夜,期间允许温度升至室温。反应结束后,于0℃用饱和氯化铵溶液(1000mL)淬灭反应,加入浓盐酸水溶液(约12M)以使pH达到2,收集有机相用饱和食盐水(1000mL)洗涤,无水硫酸钠干燥,过滤并减压浓缩,得到53.2g淡黄色固体状的标题产物,直接用于下一步,收率:84.8%。
LCMS:m/z 191[M+H] +
步骤6:(1R,2R)-2-((5-(羟甲基)-2-(甲硫基)嘧啶-4-基)氨基)-1-甲基环戊-1-醇(1f)的制备
于室温,将(4-氯-2-(甲硫基)嘧啶-5-基)甲醇(24.5g,129mmol)、(1R,2R)-2-氨基-1-甲基环戊-1-醇(15.0g,129mmol)、二异丙基乙胺(49.9g,387mmol)、 异丙醇(200mL)加到500mL反应瓶中,于80℃搅拌反应过夜。反应结束后,减压浓缩,残余物通过柱层析色谱法(洗脱剂:二氯甲烷:甲醇=0-20:1)纯化,得到25.0g白色固体状的标题产物,收率:92.9%。
LCMS:m/z 270.12[M+H] +
步骤7:4-(((1R,2R)-2-羟基-2-甲基环戊基)氨基)-2-(甲硫基)嘧啶-5-甲醛(1g)的制备
于室温,将(1R,2R)-2-((5-(羟甲基)-2-(甲硫基)嘧啶-4-基)氨基)-1-甲基环戊-1-醇(25.0g,92.9mmol)、二氧化锰(161g,1858mmol)、乙酸乙酯(600mL)加到反应瓶中,于50℃搅拌反应过夜。反应结束后,过滤除去固体,滤液浓缩后通过柱层析色谱法(洗脱剂:二氯甲烷:甲醇=0-20:1)纯化,得到19.0g白色固体状的标题产物,收率:76.6%。
LCMS:m/z 268.10[M+H] +
步骤8:8-((1R,2R)-2-羟基-2-甲基环戊基)-2-(甲硫基)吡啶并[2,3-d]嘧啶-7(8H)-酮(1h)的制备
于室温,将4-(((1R,2R)-2-羟基-2-甲基环戊基)氨基)-2-(甲硫基)嘧啶-5-甲醛(13.4g,50.2mmol)、四氢呋喃(250mL)和乙酸乙酯(13.1mL,126mmol)加入到三口瓶中,甲醇-冰浴中冷却至-5℃,在氮气氛下,缓慢地加入六甲基硅氨基锂(151mL,1.0M四氢呋喃溶液,151mmol)。期间允许温度逐渐升至室温并过夜。将得到的红色溶液在冰水浴中冷却至约3℃,然后缓慢地加入乙醇(88mL,1506mmol)。将混合物在冰浴中搅拌1h,然后移去冷却浴,使溶液升至室温,继续搅拌1h。减压除去溶剂,将残余物用水(100mL)和饱和氯化钠水溶液(100mL)稀释,水层用乙酸乙酯(400mL×3)萃取,将合并的有机相经无水硫酸钠干燥,过滤并减压浓缩。得到17.0g棕色固体状的标题产物,直接用于下一步。
LCMS:m/z 292.10[M+H] +
步骤9:8-((1R,2R)-2-羟基-2-甲基环戊基)-2-(甲基磺酰基)吡啶并[2,3-d]嘧啶-7(8H)-酮(1i)的制备
于室温,将8-((1R,2R)-2-羟基-2-甲基环戊基)-2-(甲硫基)吡啶并[2,3-d]嘧啶-7(8H)-酮(17.0g,粗品)、过硫酸氢钾(76.9g,125mmol)、2-甲基四氢呋喃(250mL)、水(60mL)加入到反应瓶中,室温搅拌4h。将溶液在水浴中冷却,用水(100mL)和饱和氯化钠水溶液(100mL)稀释并用乙酸乙酯(500mL×3)萃取。合并的有机相经无水硫酸钠干燥,过滤并减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=1:1-1)纯化,得到10.8g泡沫状棕色固体物的标题产物,收率:66.6%。
LCMS:m/z 306.09[M-18] +
步骤10:4-((8-((1R,2R)-2-羟基-2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(1j)的制备
于室温,将8-((1R,2R)-2-羟基-2-甲基环戊基)-2-(甲基磺酰基)吡啶并[2,3-d]嘧啶-7(8H)-酮(3.80g,11.8mmol)和4-氨基哌啶-1-羧酸叔丁酯(2.82g,14.2mmol)、二异丙基乙胺(3.05g,23.6mmol)、2-甲基四氢呋喃(50mL)加入到反应瓶中,60℃搅拌反应过夜。冷却至室温后,加入乙酸乙酯(200mL)、水(50mL)和饱和碳酸氢钠水溶液(50mL)。有机相经无水硫酸钠干燥,过滤并减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷:甲醇=0-5%)纯化,得到4.55g黄色泡沫固体状的标题产物,收率:87.1%。
步骤11:8-((1R,2R)-2-羟基-2-甲基环戊基)-2-(哌啶-4-基氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(1k)的制备
于室温,将4-((8-((1R,2R)-2-羟基-2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(3.63g,8.20mmol)加入到10mL盐酸的二氧六环溶液中,搅拌反应2h,有黄色固体析出,减压浓缩。于0℃,残余物中加入氢氧化钠(aq)以使pH达到10,加入二氯甲烷(200mL×3),收集有机相,减压浓缩,得到2.57g黄色固体状的标题产物,直接用于下一步。
LCMS:m/z 314.19[M+H] +
步骤12:N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l)的制备
于室温,在氮气氛下,将环丙烷磺酰胺(2.42g,20.0mmol)、三乙胺(4.04g,40.0mmol)、四氢呋喃(40mL)加入到反应瓶中,搅拌10min后,缓慢加入叔丁基二甲基氯硅烷(3.60g,24.0mmol)、二甲氨基吡啶(254mg,2.00mmol),30℃搅拌反应16h,有固体析出,减压浓缩,残余物中加入乙酸乙酯(50mL×3)和水(30mL),收集有机相,水层用乙酸乙酯(30mL×3)萃取,将合并的有机相经无水硫酸钠干燥,过滤并减压浓缩,残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-20%)纯化,得到3.27g白色固体的标题产物,收率:69.6%。
LCMS:m/z 236.11[M+H] +
步骤13:2-((1-(环丙烷磺酰亚氨基)哌啶-4-基)氨基)-8-(((1R,2R)-2-羟基-2-甲基环戊基)吡啶并[2,3-d]嘧啶-7(8H)-酮(1)的制备
于室温,在氮气氛下,将三苯基二氯化磷(1.83g,5.50mmol)的二氯甲烷溶液(6mL)冷却到0℃,加入三乙胺(808mg,8.00mmol),搅拌15min后,加入化合物N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1.18g,5.00mmol),搅拌20min后加入化合物1k(343mg,1.00mmol),加毕缓慢升至室温,于室温搅拌反应16h。加入饱和氯化铵溶液(1mL)淬灭反应,收集有机相,减压浓缩,所得残余物通过柱层析色谱法(洗脱剂:乙酸乙酯:甲醇=0-10%)纯化,再用制备液相色谱法分离(色谱柱型号:Daisogei 30mm*250mm,C18,10um,100A,流动相:乙腈/水,梯度:30%-80%),得71.0mg白色固体状标题化合物,收率15.9%。
LCMS:m/z 447.21[M+H] +
1H NMR(300MHz,CDCl 3)δ8.45(s,1H),7.50(d,J=8.9Hz,1H),6.43(d,J=9.4 Hz,1H),5.76(s,1H),3.96(s,3H),3.01(t,J=10.7Hz,2H),2.73(s,1H),2.37(ddd,J=12.6,8.0,4.7Hz,1H),2.16(s,4H),1.91-1.77(m,2H),1.67(s,6H),1.36(s,3H),1.21(dd,J=4.5,2.7Hz,2H),0.99(t,J=8.1Hz,2H)。
实施例2:2-((1-(环丙烷磺酰亚氨基)哌啶-4-基)氨基)-6-(二氟甲基)-8-((1R,2R)-2-羟基-2-甲基环戊基)吡啶并[2,3-d]嘧啶-7(8H)-酮(2)的制备
Figure PCTCN2022119618-appb-000031
步骤1:2-((1-(环丙烷磺酰亚氨基)哌啶-4-基)氨基)-6-(二氟甲基)-8-((1R,2R)-2-羟基-2-甲基环戊基)吡啶并[2,3-d]嘧啶-7(8H)-酮(2)的制备
于室温,将双(((二氟甲基)亚磺酰基)氧基)锌(59.0mg,0.200mmol)、氯化亚铁(6.35mg,0.050mmol)的水溶液(1mL)滴加到2-((1-(环丙烷磺酰亚氨基)哌啶-4-基)氨基)-8-(((1R,2R)-2-羟基-2-甲基环戊基)吡啶并[2,3-d]嘧啶-7(8H)-酮(44.6mg,0.100mmol)、TFA(11.4mg,0.100mmol)的DMSO溶液(6mL)中。将TBHP(51.2mg,70%wt水溶液,0.400mmol)稀释为1mL的水溶液,然后加入0.2mL稀释后的TBHP,20℃反应2h;再次加入0.2mL稀释后的TBHP,20℃反应2h;再次加入0.2mL稀释后的TBHP,20℃反应过夜;最后加入0.2mL稀释后的TBHP,20℃反应2h。将反应溶液倒入10%乙二胺四乙酸(10mL)中,并用乙酸乙酯(20mL×3)萃取。合并的有机相减压浓缩,残余物用制备薄层色谱法(展开剂:二氯甲烷:甲醇=20:1)纯化,并用制备液相色谱法分离(色谱柱型号:Daisogei 30mm*250mm,C18,10um,100A,流动相:乙腈/水,梯度:30%-80%),得10.1mg白色固体状的标题化合物,收率20.4%。
LCMS:m/z 497.21[M+H] +
1H NMR(400MHz,CDCl 3)δ8.55(s,1H),7.87(s,1H),6.79(t,J=55.2Hz,1H),5.75(s,1H),3.96(d,J=34.2Hz,3H),3.16-2.92(m,2H),2.72(s,1H),2.48(d,J=57.2Hz,4H),2.27-2.03(m,4H),1.98-1.84(m,2H),1.72(s,3H),1.36(s,3H),1.33-1.22(m,2H),1.14-1.04(m,2H)。
实施例3:8-环戊基-2-(((1-(环丙烷磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d] 嘧啶-7(8H)-酮(3)的制备
Figure PCTCN2022119618-appb-000032
步骤1:(4-(环戊基氨基)-2-(甲硫基)嘧啶-5-基)甲醇(3a)的制备
于室温,将(4-氯-2-(甲硫基)嘧啶-5-基)甲醇(1e)(4.66g,20.0mmol)、环戊胺(2.04g,24.0mmol)、三乙胺(6.06g,60.0mmol)、四氢呋喃(50mL)加入250mL反应瓶中,室温搅拌反应过夜。反应结束后,减压浓缩,加入乙酸乙酯(100mL×3)和水(70mL),收集有机相,减压浓缩得到5.10g淡黄色固体状的标题产物,直接用于下一步。
LCMS:m/z 240.11[M+H] +
步骤2:4-(环戊基氨基)-2-(甲硫基)嘧啶-5-甲醛(3b)的制备
于室温,将(4-(环戊基氨基)-2-(甲硫基)嘧啶-5-基)甲醇(13.4g,粗品)、二氧化锰(76.9g,884mmol)、乙酸乙酯(310mL)加到1L反应瓶中,于50℃搅拌反应过夜。反应结束后,过滤除去固体,滤液浓缩后通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-20%)纯化,得到9.37g黄色油状的标题产物,收率:89.4%。
LCMS:m/z 238.09[M+H] +
步骤3:8-环戊基-2-(甲硫基)吡啶并[2,3-d]嘧啶-7(8H)-酮(3c)的制备
于室温,将4-(环戊基氨基)-2-(甲硫基)嘧啶-5-甲醛(11.8g,49.8mmol)、四氢呋喃(250mL)和乙酸乙酯(11.0g,125mmol)加入到1L三口瓶中,甲醇-冰浴中冷却至-5℃,在氮气氛下,缓慢地加入六甲基硅氨基锂(150mL,1.0M四氢呋喃溶液,150mmol),期间允许温度逐渐升至室温并搅拌过夜。将得到的红色溶液 在冰水浴中冷却至约3℃,然后缓慢地加入乙醇(200mL)。将混合物在冰浴中搅拌1h,然后移去冷却浴,使溶液升至室温,继续搅拌1h。减压浓缩,将残余物用水(100mL)和饱和氯化钠水溶液(100mL)稀释,水层用乙酸乙酯(500mL×3)萃取,将合并的有机相经无水硫酸钠干燥,过滤并减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-40%)纯化,得到11.0g黄色固体物的标题产物,收率:84.6%。
LCMS:m/z 262.09[M+H] +
步骤4:8-环戊基-2-(甲基磺酰基)吡啶并[2,3-d]嘧啶-7(8H)-酮(3d)的制备
于室温,将8-环戊基-2-(甲硫基)吡啶并[2,3-d]嘧啶-7(8H)-酮(11.0g,42.2mmoI)、过硫酸氢钾(64.8g,105mmol)、2-甲基四氢呋喃(200mL)、水(40mL)加入到1L反应瓶中,于室温搅拌反应过夜。过滤,滤饼用乙酸乙酯(100mL×3)冲洗,收集滤液,加入乙酸乙酯(200mL)和水(100mL),将合并的有机相经无水硫酸钠干燥,过滤并减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-100%)纯化,得到8.37g黄棕色固体物的标题产物,收率:67.7%。
LCMS:m/z 294.08[M+H] +
步骤5:4-((8-环戊基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(3e)的制备
于室温,将8-环戊基-2-(甲基磺酰基)吡啶并[2,3-d]嘧啶-7(8H)-酮(4.40g,15.0mmol)、4-氨基哌啶-1-羧酸叔丁酯(3.60g,18.0mmol)、二异丙基乙胺(3.87g,30.0mmol)、四氢呋喃(75mL)加入到反应瓶中,60℃搅拌反应过夜。冷却至室温后,加入乙酸乙酯(100mL×3)和水(100mL)。有机相经无水硫酸钠干燥,过滤并减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷:甲醇=0-5%)纯化,得到5.80g黄色泡沫固体状的标题产物,收率:93.6%。
LCMS:m/z 414.24[M+H] +
步骤6:8-环戊基-2-(哌啶-4-基氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(3f)的制备
于室温,将4-((8-环戊基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(4.13g,10.0mmol)加入到10mL盐酸的二氧六环溶液中,搅拌反应2h,有黄色固体析出,减压浓缩。于0℃,残余物中加入氢氧化钠(aq)以使pH达到10,加入二氯甲烷(200mL×3),收集有机相,减压浓缩,得到2.42g黄色固体物的标题产物,直接用于下一步。
LCMS:m/z 314.19[M+H] +
步骤7:8-环戊基-2-(((1-(环丙烷磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(3)的制备
于室温,在氮气氛下,将三苯基二氯化磷(1.82g,5.50mmol)的二氯甲烷溶液(20mL)冷却到0℃,加入三乙胺(909mg,9.00mmol),搅拌15min后,加入化合物N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l)(690mg,5.00mmol),搅 拌20min后,加入化合物(8-环戊基-2-(哌啶-4-基氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(313mg,1.00mmol),加毕缓慢升至室温,搅拌反应16h。加入饱和氯化铵溶液(5mL)淬灭反应,收集有机相,减压浓缩,所得残余物通过柱层析色谱法(洗脱剂:乙酸乙酯:甲醇=0-10%)纯化,得384mg淡黄色固体产物,取60mg用制备液相色谱法分离(色谱柱型号:Daisogei 30mm*250mm,C18,10um,100A,流动相:乙腈/水,梯度:30%-80%),得48.0mg白色固体标题化合物,收率80.0%。
LCMS:m/z 417.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.40(s,1H),7.44-7.40(d,J=16Hz,1H),6.39-6.36(d,J=12Hz,1H),5.87-5.82(m,1H),4.02(s,1H),3.88(s,2H),3.11-3.06(m,2H),2.40-2.34(m,3H),2.14-2.03(m,6H),1.84(s,2H),1.68(s,4H),1.23-1.20(m,2H),1.00-0.97(m,2H)。
实施例4:8-((1R,2R)-2-羟基-2-甲基环戊基)-2-(((1-(N-甲基环丙烷磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(4)的制备
Figure PCTCN2022119618-appb-000033
步骤1:N-甲基丙烷-1-磺酰胺(4a)的制备
于室温,将环丙烷磺酰氯(1.43g,10.0mmol)、四氢呋喃(5mL)加入到玻璃封管中,降温至0℃,加入三乙胺(4.04g,40.0mmol)、甲胺水溶液(5mL),室温搅拌反应16h。减压浓缩,残余物中加入乙酸乙酯(50mL×3)和水(30mL),收集有机相,水层用乙酸乙酯(30mL×3)萃取,将合并的有机相经无水硫酸钠干燥,过滤并减压浓缩,残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-50%)纯化,得到1.26g白油状物的标题产物,收率:92.0%。
LCMS:m/z 138.08[M+H] +
步骤2:8-((1R,2R)-2-羟基-2-甲基环戊基)-2-(((1-(N-甲基环丙烷磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(4)的制备
与实施例1的步骤13的制备方法相同,除了用N-甲基丙烷-1-磺酰胺代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺,制得标题化合物。
LCMS:m/z 461.23[M+H] +
1H NMR(400MHz,CDCl 3)δ8.46(s,1H),7.50(d,J=8.5Hz,1H),6.44(d,J=9.2 Hz,1H),5.77(s,1H),4.03(s,1H),3.82(s,2H),3.17-2.99(m,2H),2.73(s,4H),2.17(d,J=13.5Hz,6H),2.02(s,1H),1.84(dd,J=13.9,6.0Hz,2H),1.67(d,J=9.0Hz,3H),1.36(s,3H),1.26(s,2H),1.12(s,1H),1.04(s,1H)。
实施例5:6-(二氟甲基)-8-(((1R,2R)-2-羟基-2-甲基环戊基)-2-(((1-(N-甲基环丙烷磺酰亚氨基))哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(5)的制备
Figure PCTCN2022119618-appb-000034
与实施例2的制备方法相同,除了用化合物4代替化合物1,制得标题化合物。
LCMS:m/z 511.22[M+H] +
1H NMR(400MHz,CDCl 3)δ8.55(s,1H),7.86(s,1H),6.79(t,J=55.2Hz,1H),5.73(s,1H),4.05(d,J=44.6Hz,1H),3.78(s,2H),3.16–2.90(m,2H),2.70(s,3H),2.47(s,1H),2.15(d,J=13.1Hz,3H),1.86(s,6H),1.67(s,3H),1.36(s,3H),1.20(d,J=3.8Hz,2H),0.99(dd,J=19.7,6.3Hz,2H)。
实施例6:8-环戊基-2-(((8-(环丙烷磺酰亚氨基)-8-氮杂双[3.2.1]辛-3-基]氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(6)的制备
Figure PCTCN2022119618-appb-000035
与实施例3的制备方法相同,除了用2-((8-氮杂双环[3.2.1]辛-3-基)胺代替步骤1中的环戊胺,制得标题化合物。
LCMS:m/z 443.22[M+H] +
1HNMR(400MHz,CDCl 3)δ8.37(s,1H),7.42(d,J=9.3Hz,1H),6.38(d,J=9.3Hz,1H),5.89-5.77(m,1H),4.38(s,1H),4.33-4.22(m,2H),2.63-2.29(m,7H),2.23(d,J=9.2Hz,2H),2.13(dd,J=15.7,6.7Hz,2H),2.02(dd,J=15.4,12.1Hz,4H),1.91-1.78(m,2H),1.73-1.62(m,2H),1.23(dq,J=11.5,5.7Hz,2H),1.07-0.99(m,1H),0.96(dd,J=7.5,2.1Hz,1H)。
实施例7:8-环戊基-2-(((1-(丙-2-基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(7)的制备
Figure PCTCN2022119618-appb-000036
步骤1:N-(叔丁基二甲基甲硅烷基)丙烷-2-磺酰胺(7a)的制备
与实施例1的步骤12的方法相同,除了用丙烷-2-磺酰胺代替环丙烷磺酰胺,制得标题化合物。
步骤2:8-环戊基-2-(((1-(丙-2-基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮的制备
与实施例3的制备方法相同,除了用N-(叔丁基二甲基甲硅烷基)丙烷-2-磺酰胺(7a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 419.22[M+H] +
1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.42-7.40(d,J=8Hz,1H),6.38-6.35(d,J=12Hz,1H),5.88-5.79(m,1H),4.03(s,1H),3.94(s,2H),3.30-3.23(m,1H),3.11-3.06(m,2H),2.37(s,2H),2.17-2.11(m,2H),2.03(s,3H),1.86-1.84(m,3H),1.69-1.58(m,4H),1.41-1.36(m,6H)。
实施例8:8-环戊基-2-(((1-(丙基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(8)的制备
Figure PCTCN2022119618-appb-000037
Figure PCTCN2022119618-appb-000038
步骤1:N-(叔丁基二甲基甲硅烷基)丙烷-1-磺酰胺(8a)的制备
与实施例1的步骤12的方法相同,除了用丙烷-1-磺酰胺代替环丙烷磺酰胺,制得标题化合物。
步骤2:8-环戊基-2-(((1-(丙-2-基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮的制备
与实施例3的制备方法相同,除了用N-(叔丁基二甲基甲硅烷基)丙烷-1-磺酰胺(8a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 419.22[M+H] +
1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.42-7.40(d,J=8Hz,1H),6.38-6.35(d,J=12Hz,1H),5.86-5.82(m,1H),4.01(s,1H),3.87(s,2H),2.99-2.86(m,3H),2.85-2.81(m,1H),2.01(s,2H),1.93-1.91(m,2H),1.89(s,2H),1.87-1.85(m,5H),1.68-1.62(m,5H),1.09-1.06(t,J=12Hz,3H)。
实施例9:8-环戊基-2-(((1-(N-甲基丙基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(9)的制备
Figure PCTCN2022119618-appb-000039
步骤1:N-甲基丙烷-1-磺酰胺(9a)的制备
与实施例4的步骤1的方法相同,除了用丙烷-1-磺酰氯代替环丙烷磺酰氯,制得标题化合物。
步骤2:8-环戊基-2-(((1-(丙-2-基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮的制备
与实施例3的制备方法相同,除了用N-甲基丙烷-1-磺酰胺(9a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 432.23[M+H] +
1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.43-7.40(d,J=12Hz,1H),6.38-6.35(d,J=12Hz,1H),5.87-5.78(m,1H),4.02(s,1H),3.82-3.71(s,2H),3.03-2.81(m,4H),2.69(s,3H),2.36-2.16(m,5H),2.03(m,2H),1.96-1.84(m,4H),1.68-1.64(m,4H),1.08-1.03(m,3H)。
实施例10:8-环戊基-6-(二氟甲基)-2-(((1-(N-甲基丙基磺酰亚氨基)哌啶-4-基)氨基)吡啶基[2,3-d]嘧啶-7(8H)-酮(10)的制备
Figure PCTCN2022119618-appb-000040
与实施例2的制备方法相同,除了用化合物9代替化合物1,制得标题化合物。
LCMS:m/z 483.1[M+H] +
1HNMR(300MHz,CDCl 3)δ8.51(s,1H),7.80(s,1H),6.99-6.62(m,1H),5.88-5.82(m,1H),4.05(s,1H),3.79(s,2H),3.02-2.98(m,4H),2.70(s,3H),2.18(m,5H),2.04(m,2H),1.92-1.88(m,4H),1.69(s,4H),1.09-1.04(m,3H)。
实施例11:8-((1R,2R)-2-羟基-2-甲基环戊基)-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(11)的制备
Figure PCTCN2022119618-appb-000041
Figure PCTCN2022119618-appb-000042
步骤1:N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)的制备
与实施例1的步骤12的方法相同,除了用甲磺酰胺代替环丙烷磺酰胺,制得标题化合物。
其余步骤与实施例1的步骤相同,除了用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替实施例1步骤13的N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 421.19[M+H] +
1H NMR(400MHz,CDCl 3)δ8.45(s,1H),7.50(d,J=8.2Hz,1H),6.44(d,J=9.1Hz,1H),5.76(s,1H),3.94(d,J=42.8Hz,3H),2.87(d,J=7.0Hz,5H),2.73(s,1H),2.18(s,3H),1.99-1.78(m,6H),1.68(s,3H),1.36(s,3H)。
实施例12:6-(二氟甲基)-8-(((1R,2R)-2-羟基-2-甲基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(12)的制备
Figure PCTCN2022119618-appb-000043
与实施例2的制备方法相同,除了用化合物11代替化合物1,制得标题化合物。
LCMS:m/z 471.19[M+H] +
1H NMR(400MHz,CDCl 3)δ8.55(s,1H),7.87(s,1H),6.79(t,J=55.2Hz,1H),5.75(s,1H),3.90(s,3H),2.88(s,3H),2.71(s,1H),2.19(s,4H),1.77(d,J=53.5Hz,10H),1.36(s,3H)。
实施例13:8-环戊基-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(13)的制备
Figure PCTCN2022119618-appb-000044
与实施例3的制备方法相同,除了用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替步骤7中的N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 391.18[M+H] +
1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.44-7.41(d,J=12Hz,1H),6.40-6.37(d,J=12Hz,1H),5.90-5.78(m,1H),4.03(s,1H),3.85(s,2H),2.95(s,2H),2.86(s,3H),2.35-2.19(m,6H),2.03(s,2H),1.87-1.84(m,2H),1.70(s,4H)。
实施例14:2-((1-(N,S-二甲基磺酰亚氨基)哌啶-4-基)氨基)-8-(((1R,2R)-2-羟基-2-甲基环戊基)吡啶并[2,3-d]嘧啶-7(8H)-酮(14)的制备
Figure PCTCN2022119618-appb-000045
步骤1:N-甲基甲磺酰胺(14a)的制备
与实施例4的步骤1的方法相同,除了用甲磺酰氯代替环丙烷磺酰氯,制得标题化合物。
其余步骤与实施例1的步骤相同,除了用N-甲基甲磺酰胺(14a)代替实施例1步骤13的N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 435.21[M+H] +
1H NMR(400MHz,CDCl 3)δ8.45(s,1H),7.53-7.46(m,1H),6.44(d,J=9.2Hz,1H),5.76(s,1H),4.02(s,1H),3.87-3.67(m,2H),2.94(s,6H),2.72(s,1H),2.69(d,J=1.1Hz,3H),2.28-1.97(m,5H),1.90-1.80(m,2H),1.68(d,J=9.0Hz,3H),1.36(s,3H)。
实施例15:6-(二氟甲基)-2-(((1-(N,S-二甲基磺酰亚氨基)哌啶-4-基)氨基)-8-((1R,2R)-2-羟基-2-甲基环戊基)吡啶并[2,3-d]嘧啶-7(8H)-酮(15)的制备
Figure PCTCN2022119618-appb-000046
与实施例2的制备方法相同,除了用化合物14代替化合物1,制得标题化合物。
LCMS:m/z 485.21[M+H] +
1HNMR(400MHz,CDCl 3)δ8.55(s,1H),7.87(s,1H),6.79(t,J=55.2Hz,1H),5.70(s,1H),4.05(d,J=46.6Hz,1H),3.78(d,J=18.8Hz,2H),2.90(s,5H),2.69(d,J=1.2Hz,4H),2.29-1.95(m,6H),1.86(dd,J=13.2,6.1Hz,2H),1.70(s,3H),1.36(s,3H)。
实施例16:8-环戊基-2-(((1-(N,S-二甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(16)的制备
Figure PCTCN2022119618-appb-000047
Figure PCTCN2022119618-appb-000048
与实施例3的制备方法相同,除了用N-甲基甲磺酰胺(14a)代替步骤7中的N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 405.203[M+H] +
1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.4-7.41(d,J=12Hz,1H),6.39-6.36(d,J=12Hz,1H),5.90-5.78(m,1H),4.02(s,1H),3.80-3.67(s,2H),2.96-2.92(m,2H),2.69(s,3H),2.37-2.18(m,5H),2.03(m,2H),1.87-1.84(m,2H),1.68(m,4H)。
实施例17:8-环戊基-6-(二氟甲基)-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(17)的制备
Figure PCTCN2022119618-appb-000049
与实施例2的制备方法相同,除了用化合物13代替化合物1,制得标题化合物。
LCMS:m/z 441.18[M+H] +
1H NMR(400MHz,CDCl 3)δ8.51(s,1H),7.79(s,1H),6.80(t,J=55.3Hz,1H),6.11(s,0.5H),5.94-5.73(m,1H),5.30(s,0.5H),4.06(s,1H),3.84(s,2H),3.01(s,1H),2.90(s,3H),2.62(s,2H),2.27(d,J=55.8Hz,4H),2.02(d,J=13.3Hz,2H),1.87(s,2H),1.70(s,4H)。
实施例18:8-环戊基-6-(二氟甲基)-2-(((1-(N,S-甲基磺酰亚氨基))哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(18)的制备
Figure PCTCN2022119618-appb-000050
与实施例2的制备方法相同,除了用化合物16代替化合物1,制得标题化合物。
LCMS:m/z 455.20[M+H] +
1HNMR(400MHz,CDCl 3)δ8.51(s,1H),7.79(s,1H),6.80(t,J=55.3Hz,1H),6.00-5.73(m,1H),4.05(s,1H),3.76(s,2H),2.96(s,5H),2.71(s,3H),2.35(s,2H),2.22(s,3H),2.04(s,2H),1.86(s,2H),1.69(s,4H)。
实施例19:8-环戊基-2-(((1-(N-甲基环丙烷磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(3)的制备
Figure PCTCN2022119618-appb-000051
与实施例3的制备方法相同,除了用N-甲基丙烷-1-磺酰胺(4a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 431.23[M+H] +
1H NMR(400MHz,CDCl 3)δ8.40(s,1H),7.41(d,J=9.3Hz,1H),6.37(d,J=9.3Hz,1H),5.84(p,J=9.0Hz,1H),5.41(s,1H),4.05(s,1H),3.78(s,2H),3.07(t,J=11.3Hz,2H),2.72(s,3H),2.55(d,J=2.9Hz,1H),2.37(s,2H),2.19(t,J=12.8Hz,2H),2.03(s,2H),1.90-1.80(m,2H),1.69(s,4H),1.33-1.16(m,2H),1.01(dd,J=22.1,9.7Hz,2H)。
实施例20:8-环戊基-2-(((1-(环丙烷磺酰亚氨基)哌啶-4-基)氨基)-6-(二氟甲基)吡啶并[2,3-d]嘧啶-7(8H)-酮(20)的制备
Figure PCTCN2022119618-appb-000052
与实施例2的制备方法相同,除了用化合物3代替化合物1,制得标题化合物。
LCMS:m/z 467.20[M+H] +
1HNMR(400MHz,CDCl 3)δ8.49(s,1H),7.79(s,1H),6.80(t,J=55.3Hz,1H),5.85(p,J=8.9Hz,1H),4.05(s,1H),3.88(s,2H),3.09(s,2H),2.50(s,2H),2.43(ddd,J=12.8,8.1,4.9Hz,3H),2.18(s,2H),2.04(s,2H),1.87(s,2H),1.70(s,4H),1.28-1.18(m,2H),1.02(dd,J=9.9,8.3Hz,2H)。
实施例21:8-环戊基-6-(二氟甲基)-2-(((1-(N-甲基丙基磺酰亚氨基)哌啶丁-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(21)的制备
Figure PCTCN2022119618-appb-000053
与实施例2的制备方法相同,除了用化合物19代替化合物1,制得标题化合物。
LCMS:m/z 481.21[M+H] +
1H NMR(400MHz,CDCl 3)δ8.52(s,1H),7.79(s,1H),6.80(t,J=55.3Hz,1H),5.90-5.79(m,1H),5.45(d,J=98.8Hz,1H),4.07(s,1H),3.81(s,2H),3.09(s,2H),2.74(s,3H),2.67(s,1H),2.37(s,2H),2.21(t,J=13.5Hz,2H),2.04(s,2H),1.87(s,2H),1.69(s,4H),1.32-1.21(m,2H),1.15-0.95(m,2H)。
实施例22:8-(2-甲基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(22-P1和22-P2)的制备
Figure PCTCN2022119618-appb-000054
与实施例3的制备方法相同,除了用2-甲基环戊烷-1-胺盐酸盐代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。通过柱层析色谱法(洗脱剂:乙酸乙酯:甲醇=0-10%)纯化,再用制备液相色谱法分离得到化合物22-P1和22-P2。
LCMS:m/z 405.20[M+H] +
单一构型化合物22-P1(较短保留时间):
制备液相色谱法:色谱柱:Daisogei 30mm×250mm,C18,10um,100A,流动相:乙腈/水,梯度:30%-80%。
1H NMR(400MHz,CDCl 3)δ8.38(s,1H),7.43(d,J=9.3Hz,1H),6.40(d,J=9.3Hz,1H),5.41(d,J=9.1Hz,1H),4.01(d,J=3.4Hz,1H),3.84(s,2H),3.04-2.89(m,2H),2.87(s,3H),2.41(s,4H),2.18(d,J=8.7Hz,2H),2.11-1.97(m,2H),1.96-1.85(m,1H),1.83-1.60(m,3H),1.33(dd,J=20.2,9.7Hz,1H),0.94(d,J=6.6Hz,3H)。
单一构型化合物22-P2(较长保留时间):
制备液相色谱法:色谱柱:Daisogei 30mm×250mm,C18,10um,100A,流动相:乙腈/水,梯度:30%-80%。
1H NMR(400MHz,CDCl 3)δ8.37(s,1H),7.42(d,J=9.3Hz,1H),6.37(d,J=9.2Hz,1H),5.92(dd,J=17.3,9.7Hz,1H),4.14–3.98(m,1H),3.82(s,2H),3.01(s,2H),2.87(s,3H),2.69(s,3H),2.37(td,J=10.1,5.0Hz,1H),2.19(s,2H),2.12–1.97(m,1H),1.98–1.83(m,3H),1.71(s,2H),1.57(dd,J=19.0,8.4Hz,1H),0.79(d,J=7.1Hz,3H)。
实施例23:8-(2,2-二甲基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡 啶并[2,3-d]嘧啶-7(8H)-酮(23)的制备
Figure PCTCN2022119618-appb-000055
与实施例3的制备方法相同,除了用2,2-二甲基环戊胺代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 419.20[M+H]+。
1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.44-7.41(d,J=12Hz,1H),6.40-6.37(d,J=12Hz,1H),5.90-5.78(m,1H),4.03(s,1H),3.85(s,2H),2.95(s,2H),2.86(s,3H),2.62-2.20(m,2H),2.09-1.98(m,6H),1.67-1.50(m,3H),1.50(s,1H),1.16(s,3H),0.85(s,3H)。
实施例24:2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)-8-(四氢呋喃-3-基)吡啶并[2,3-d]嘧啶-7(8H)-酮(24)的制备
Figure PCTCN2022119618-appb-000056
与实施例3的制备方法相同,除了用四氢呋喃-3-胺代替环戊胺和用N-(叔丁基 二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 393.16[M+H] +
1HNMR(400MHz,CDCl 3)δ8.41(s,1H),7.45(d,J=9.4Hz,1H),6.40(d,J=9.3Hz,1H),6.25-6.07(m,1H),4.41-4.06(m,3H),3.98(t,J=8.7Hz,1H),3.88(s,2H),2.94(s,2H),2.87(s,3H),2.56(s,1H),2.17(d,J=4.3Hz,6H),1.76-1.54(m,2H)。
实施例25:2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)-8-(螺[2.4]庚-4-基)吡啶并[2,3-d]嘧啶-7(8H)-酮(25)的制备
Figure PCTCN2022119618-appb-000057
与实施例3的制备方法相同,除了用螺[2.4]庚-4-胺代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 417.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.36(s,1H),7.41(d,J=9.4Hz,1H),6.38(d,J=9.1Hz,1H),5.80(t,J=8.1Hz,1H),4.00(s,1H),3.80(s,2H),3.03(d,J=11.5Hz,2H),2.87(s,3H),2.75-2.31(m,4H),2.10(dd,J=26.9,18.9Hz,4H),1.85(s,1H),1.69(s,2H),1.38(s,1H),0.60(s,2H),0.49(s,1H),0.25-0.06(m,1H)。
实施例26:8-环戊基-6-甲基-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(26)的制备
Figure PCTCN2022119618-appb-000058
Figure PCTCN2022119618-appb-000059
与实施例3的制备方法相同,除了用丙酸乙酯代替乙酸乙酯和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 405.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.32(s,1H),7.30(d,J=1.1Hz,1H),5.87(p,J=8.9Hz,1H),4.09-4.00(m,1H),3.81(s,2H),3.00(d,J=5.1Hz,2H),2.86(s,3H),2.49(s,2H),2.33(s,2H),2.22-2.12(m,5H),2.05(s,2H),1.91-1.80(m,2H),1.77-1.61(m,4H)。
实施例27:8-环戊基-5,6-二甲基-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(27)的制备
Figure PCTCN2022119618-appb-000060
步骤1:1-(4-(环戊氨基)-2-(甲硫基)嘧啶-5-基)-1-乙醇(27a)的制备
于室温,将4-(环戊氨基)-2-(甲硫基)嘧啶-5-甲醛(3b)(929mg,3.70mmol)、THF(30mL)加入到三口瓶中,冷却至-75℃,氮气氛下,缓慢地加入甲基溴化镁(3.7mL,1.0M THF溶液,3.70mmol),期间允许温度逐渐升至室温并搅拌过夜。降温到0℃,加入水(10mL)淬灭反应,收集有机相,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-75%)纯化,得到740mg白色固体的标题产物,收率:75.0%。
LCMS:m/z 254.14[M+H] +
步骤2:1-(4-(环戊氨基)-2-(甲硫基)嘧啶-5-基)-1-乙酮(27b)的制备
于室温,将1-(4-(环戊氨基)-2-(甲硫基)嘧啶-5-基)-1-乙醇(27a)(740mg,2.77mmol)、二氧化锰(4.54g,55.4mmol)、乙酸乙酯(10mL)加到反应瓶中,50℃搅拌反应过夜。反应结束后,过滤除去固体,滤液浓缩后通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-20%)纯化,得到700mg淡黄色固体的标题产物,收率:95.4%。
LCMS:m/z 252.14[M+H] +
其余步骤与实施例3的制备方法相同,除了用1-(4-(环戊氨基)-2-(甲硫基)嘧啶-5-基)-1-乙酮(27b)代替4-(环戊基氨基)-2-(甲硫基)嘧啶-5-甲醛(3b),用丙酸乙酯代替乙酸乙酯,和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物27。
LCMS:m/z 419.22[M+H] +
1H NMR(400MHz,CDCl 3)δ8.53(s,1H),5.89(p,J=8.9Hz,1H),4.02(td,J=13.7,7.0Hz,1H),3.83(s,2H),2.97(t,J=10.3Hz,2H),2.86(s,3H),2.82-2.36(m,2H),2.34(s,5H),2.24-2.13(m,5H),2.10-1.99(m,2H),1.83(dt,J=11.4,8.4Hz,2H),1.68(dd,J=10.2,5.7Hz,4H)。
实施例28:6-乙酰基-8-环戊基-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(28)的制备
Figure PCTCN2022119618-appb-000061
步骤1:4-((6-碘-8-环戊基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(28a)的制备
于室温,将4-((8-环戊基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(3e)(149mg,0.36mmol)、NIS(122mg,0.55mmol)、对甲苯磺 酸(6.24mg,0.036mmol)、乙腈(2mL)加入到反应瓶中,室温搅拌反应过夜。减压浓缩,残余物中加入EtOAc(10mL×3)和水(5mL)。有机相经无水硫酸钠干燥,过滤,减压浓缩,残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-40%)纯化,得到164mg黄色固体物的标题产物,收率:84.7%。
LCMS:m/z 540.15[M+H] +
步骤2:4-(((8-环戊基-6-(1-乙氧基乙烯基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(28b)的制备
于室温,将4-((6-碘-8-环戊基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(28a)(296mg,0.55mmol)、三丁基(1-乙氧基乙烯基)锡烷(299mg,0.825mmol)、四三苯基膦钯(76.3mg,0.066mmol)、甲苯(5mL)加入到反应瓶中,回流搅拌反应过夜。冷却至室温后,减压浓缩,残余物中加入EtOAc(20mL×3)和水(10mL)。有机相经无水硫酸钠干燥,过滤,减压浓缩,残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-40%)纯化,得到160mg淡黄色固体物的标题产物,收率:63.8%。
LCMS:m/z 484.28[M+H] +/456.25[M+H] +
步骤3:6-乙酰基-8-环戊基-2-(哌啶-4-基氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(28c)的制备
于室温,将4-(((8-环戊基-6-(1-乙氧基乙烯基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(28b)(160mg,0.350mmol)加入到2mL盐酸的二氧六环溶液中,搅拌反应2h。有黄色固体析出,减压浓缩,得到黄色固体。加入碳酸钾(276mg,2.00mmol),加入水(5mL),搅拌反应2h。加入二氯甲烷(10mL×3),收集有机相,减压浓缩,得到93mg淡黄色固体物的标题产物,直接用于下一步。
LCMS:m/z 356.20[M+H] +
其余步骤与实施例3的制备方法相同,除了用6-乙酰基-8-环戊基-2-(哌啶-4-基氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(28c)代替8-环戊基-2-(哌啶-4-基氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(3f),用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物28。
LCMS:m/z 433.19[M+H] +
1H NMR(400MHz,CDCl 3)δ8.56(s,1H),8.24(s,1H),6.03(s,0.6H),5.90-5.81(m,1H),5.39(s,0.4H),4.06(s,1H),3.85(s,2H),3.03(d,J=34.2Hz,3H),2.91(s,3H),2.70(s,3H),2.35(m,2H),2.20(s,2H),2.03(d,J=17.0Hz,2H),1.87(s,2H),1.72(s,4H)。
实施例29:6-乙酰基-8-环戊基-5-甲基-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(29)的制备
Figure PCTCN2022119618-appb-000062
步骤1:4-((6-溴-8-环戊基-5-甲基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(29a)的制备
于室温,将4-氨基哌啶-1-羧酸叔丁酯(2.52g,12.0mmol)、甲苯(10mL)加入到反应瓶中,氮气氛下,降至0℃,加入双(三甲基甲硅烷基)氨基锂(1M在THF中,12mL,12.0mmol),搅拌反应20分钟,加入化合物6-溴-2-氯-8-环戊基-5-甲基吡啶并[2,3-d]嘧啶-7(8H)-酮(1.03g,3.00mmol)(商品化试剂CAS:1016636-76-2),搅拌反应过夜。减压浓缩,残余物中加入EtOAc(20mL×3)和水(10mL)。有机相经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-40%)纯化,得到620mg淡黄色固体物的标题产物,收率:40.9%。
LCMS:m/z 506.17[M+H] +
其余步骤与实施例28的制备方法相同,除了用4-((6-溴-8-环戊基-5-甲基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(29a)代替4-((6-碘-8-环戊基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(28a),制得化合物29。
LCMS:m/z 447.21[M+H] +
1H NMR(400MHz,CDCl 3)δ8.63(s,1H),6.35(s,0.6H),5.88-5.75(m,1H),5.21(s,0.4H),4.05(s,1H),3.82(s,2H),3.01(s,3H),2.88(s,3H),2.53(s,3H),2.33(s,5H),2.19(s,2H),2.02(s,2H),1.85(s,2H),1.69(s,4H)。
实施例30:8-环戊基-2-(((1-(1-氧化-4,5-二氢-3H-1λ 6-异噻唑-1-基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(30)的制备
Figure PCTCN2022119618-appb-000063
与实施例3的制备方法相同,除了用异噻唑烷1,1-二氧化物代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物30。
LCMS:m/z 417.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.41(d,J=9.3Hz,1H),6.37(d,J=9.2Hz,1H),5.83(p,J=8.9Hz,1H),4.06(s,1H),3.77(s,3H),3.05(s,4H),2.51-2.32(m,4H),2.27-2.11(m,4H),2.03(s,3H),1.92-1.79(m,3H),1.70(s,5H),1.26(s,3H)。
实施例31:6-甲基-8-(2-甲基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(31)的制备
Figure PCTCN2022119618-appb-000064
与实施例22的制备方法相同,除了用丙酸乙酯代替乙酸乙酯,制得化合物31。
LCMS:m/z 419.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.32(s,1H),7.30(d,J=9.3Hz,1H),6.40(d,J=9.3Hz,1H),4.04(d,J=3.4Hz,1H),3.84(s,2H),2.97-2.96(m,2H),2.86(s,3H),2.62(s,1H),2.40(s,1H),2.09-1.64(m,11H),1.56(s,1H),1.32-1.26(m,2H),0.93-0.76(m,3H)。
实施例32:5,6-二甲基-8-(2-甲基环戊基)-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(32)的制备
Figure PCTCN2022119618-appb-000065
与实施例27的制备方法相同,除了用4-((2-甲基环戊基)氨基)-2-(甲硫基)嘧啶-5-甲醛(22b)代替4-(环戊氨基)-2-(甲硫基)嘧啶-5-甲醛(3b),制得标题化合物。
LCMS:m/z 433.23[M+H] +
1HNMR(400MHz,CDCl 3)δ8.51(s,1H),5.98(dd,J=17.2,9.7Hz,1H),4.02(dd,J=10.6,6.8Hz,1H),3.82(s,2H),2.99(s,2H),2.86(s,3H),2.68(d,J=7.4Hz,1H),2.39–2.29(m,4H),2.24–2.13(m,5H),1.97(m,5H),1.61(m,3.5H),1.30(m,0.5H),0.93(d,J=6.6Hz,0.7H),0.77(d,J=7.1Hz,2.3H)。
实施例33:6-(二氟甲基)-8-(2-甲基环戊基)-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基]吡啶并[2,3-d]嘧啶-7(8H)-酮(33-P1和33-P2)的制备
Figure PCTCN2022119618-appb-000066
步骤1:6-(二氟甲基)-8-(2-甲基环戊基)-2-(((1-(甲基磺酰亚氨基)哌啶丁-4-基)氨基]吡啶并[2,3-d]嘧啶-7(8H)-酮(33-P1和33-P2)的制备
于室温,反应瓶中加入二氟甲烷亚磺酸钠(172mg,1.25mmol)、水(3mL)、二甲基亚砜(15mL),室温搅拌25分钟,加入8-(2-甲基环戊基)-2-((1-(S-甲基磺 酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(202mg,0.50mmol)、氯化亚铁(10.2mg,0.08mmol)。取过氧化叔丁醇(64.0mg,70%wt水溶液,0.50mmol),用DMSO稀释为1mL,,滴加到反应体系中,室温搅拌过夜。将反应溶液倒入10%EDTA(30mL)中,并用EtOAc(50mL×3)萃取。合并有机相,用制备薄层色谱法(展开剂:二氯甲烷:甲醇=20:1)纯化,并用制备液相色谱法分离,得到化合物33-P1(10.0mg)和33-P2(24.5mg)。
单一构型化合物33-P1(较短保留时间)
制备液相色谱法:色谱柱:Daisogei 30mm×250mm,C18,10um,100A,流动相:乙腈/水,梯度:30%-80%。
LCMS:m/z 455.54[M+H] +
1H NMR(400MHz,CDCl 3)δ8.49(s,1H),7.79(s,1H),6.79(t,J=55.3Hz,1H),5.94(dd,J=17.2,9.7Hz,1H),4.04(s,1H),3.85(s,2H),2.97(s,2H),2.87(s,3H),2.68(d,J=44.8Hz,1H),2.47–2.28(m,2H),2.21(s,2H),2.06(dd,J=10.8,7.4Hz,1H),1.92(dd,J=18.9,9.6Hz,3H),1.77–1.55(m,3H),0.79(d,J=7.1Hz,3H)。
单一构型化合物33-P2(较长保留时间)
制备液相色谱法:色谱柱:Daisogei 30mm×250mm,C18,10um,100A,流动相:乙腈/水,梯度:30%-80%。
LCMS:m/z 455.54[M+H] +
1H NMR(400MHz,CDCl 3)δ8.49(s,1H),7.79(s,1H),6.79(t,J=55.3Hz,1H),5.94(dd,J=17.2,9.7Hz,1H),4.04(s,1H),3.85(s,2H),2.97(s,2H),2.87(s,3H),2.68(d,J=44.8Hz,1H),2.47–2.28(m,2H),2.21(s,2H),2.06(dd,J=10.8,7.4Hz,1H),1.92(dd,J=18.9,9.6Hz,3H),1.77–1.55(m,3H),0.79(d,J=7.1Hz,3H).)。
实施例34:6-乙酰基-5-甲基-8-(2-甲基环戊基)-2-(((1-(S-甲基磺酰亚氨基)哌啶基-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(34)的制备
Figure PCTCN2022119618-appb-000067
Figure PCTCN2022119618-appb-000068
步骤1:4-(((5-甲基-8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(34c)的制备
与实施例27中化合物27e的制备方法相同,除了用1-(4-((2-甲基环戊基)氨基)-2-(甲硫基)嘧啶-5-基)乙-1-酮(32b)代替1-(4-(环戊氨基)-2-(甲硫基)嘧啶-5-基)-1-乙酮(27b)和用乙酸乙酯代替丙酸乙酯,制得化合物34c。
其余步骤与实施例28的相同,除了用4-(((5-甲基-8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)叔丁基-1-羧酸叔丁酯(34c)代替4-((8-环戊基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(3e),制得化合物34。
LCMS:m/z 461.21[M+H] +
1H NMR(400MHz,CDCl3)δ8.60(s,1H),6.38(s,0.5H),6.01–5.83(m,1H),5.41(m,0.5H),4.04(s,1H),3.81(s,2H),3.01(s,2H),2.88(s,3H),2.69(d,J=11.5Hz,1H),2.52(d,J=7.0Hz,3H),2.37–2.27(m,4H),2.19(s,2H),2.10–1.93(m,2H),1.90(d,J=8.2Hz,2H),1.71(m,3H),1.32(dd,J=19.8,9.6Hz,1H),0.94(d,J=6.6Hz,1H),0.80(d,J=7.1Hz,2H)。
实施例35:6-乙酰基-8-(2-甲基环戊基)-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(35)的制备
Figure PCTCN2022119618-appb-000069
Figure PCTCN2022119618-appb-000070
与实施例28的制备方法相同,除了用4-(((8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(22e)代替4-((8-环戊基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(3e),制得化合物35。
LCMS:m/z 447.19[M+H] +
1H NMR(400MHz,CDCl 3)δ8.53(s,1H),8.23(s,1H),6.18(d,J=22.3Hz,0.5H),5.92(dd,J=17.8,9.4Hz,1H),5.41(d,J=24.1Hz,0.5H),4.07(s,1H),3.85(s,2H),3.00(d,J=11.5Hz,2H),2.92(s,4H),2.68(s,3H),2.55–2.28(m,1H),2.20(s,2H),2.02(m,4H),1.67(d,J=51.4Hz,3H),1.30(d,J=29.8Hz,1H),0.95(d,J=6.5Hz,1H),0.80(d,J=7.0Hz,2H)。
实施例36:8-环戊基-2-(((1-(噻吩-2-磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(36)的制备
Figure PCTCN2022119618-appb-000071
步骤1:N-(叔丁基二甲基甲硅烷基)噻吩-2-磺酰胺(36a)的制备
与实施例1的步骤12的方法相同,除了用噻吩-2-磺酰胺代替环丙烷磺酰胺,制得标题化合物。
其余步骤与实施例3的制备方法相同,除了用N-(叔丁基二甲基甲硅烷基)噻吩-2-磺酰胺(36a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物36。
LCMS:m/z 459.18[M+H] +
1H NMR(400MHz,CDCl 3)δ8.33(s,1H),7.72–7.50(m,2H),7.40(d,J=9.3Hz,1H),7.22–7.11(m,1H),6.39(d,J=6.9Hz,1H),5.92–5.64(m,1H),4.02–3.81(m,1H),3.74(s,2H),2.91–2.54(m,2H),2.30(s,2H),2.20–2.07(m,2H),1.94(s,2H),1.85– 1.76(m,3H),1.71(d,J=15.6Hz,2H),1.64(dd,J=10.3,5.3Hz,3H)。
实施例37:6-氟-8-(2-甲基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(37)的制备
Figure PCTCN2022119618-appb-000072
与实施例22的制备方法相同,除了用2-氟乙酸乙酯代替乙酸乙酯,制得化合物37。
LCMS:m/z 423.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.18(d,J=7.8Hz,1H),5.97(d,J=7.9Hz,1H),4.02(d,J=7.1Hz,1H),3.85(s,2H),2.92(m,7H),2.70(s,1H),2.38(tdd,J=14.3,8.6,5.2Hz,1H),2.27–2.14(m,2H),2.09(m,1H),1.92(m,3H),1.81–1.49(m,3H),0.95(d,J=6.6Hz,0.4H),0.80(d,J=7.1Hz,2.7H)。
实施例38:6-氯-8-(2-甲基环戊基)-2-((1-(甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(38)的制备
Figure PCTCN2022119618-appb-000073
步骤1:4-(((6-氯-8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(38a)的制备
于室温,将4-(((8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(22e)(341mg,0.80mmol)、NCS(128mg,0.96mmol)、2-甲基四氢呋喃(10mL)加入到反应瓶中,60℃搅拌反应过夜。减压浓缩,残余物中加入EtOAc(20mL×3)和水(10mL)。有机相经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-50%)纯化,得到0.32g黄色固体物的标题产物,收率:86.0%。
LCMS:m/z 462.15[M+H] +
其余步骤与实施例3的制备方法相同,除了用4-(((6-氯-8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(38a)代替4-((8-环戊基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(3e),用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物38。
LCMS:m/z 439.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.38(s,1H),7.66(d,J=9.3Hz,1H),6.02(d,J=9.3Hz,1H),4.01(d,J=3.4Hz,1H),3.84(s,2H),3.04-2.92(m,3H),2.91(s,3H),2.64-2.36(m,1H),2.19-2.00(m,4H),1.83-1.60(m,3H),1.60-1.56(m,3H),0.80(d,J=6.6Hz,3H)。
实施例39:6-异丙基-8-(2-甲基环戊基)-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(39)的制备
Figure PCTCN2022119618-appb-000074
步骤1:4-(((6-溴-8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(39a)的制备
于室温,将4-(((8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(22e)(854mg,2.00mmol)、NBS(391.0mg,2.20mmol)、2-甲基四氢呋喃(20mL)加入到反应瓶中,60℃搅拌反应过夜。减压浓缩,残余物中加入EtOAc(20mL×3)和水(10mL)。有机相经无水硫酸钠干燥,过滤, 减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯=0-50%)纯化,得到0.95g黄色固体物的标题产物,收率:93.0%。
LCMS:m/z 507.15[M+H] +
步骤2:4-((8-(2-甲基环戊基)-7-氧代-6-(丙-1-烯-2-基)-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(39b)的制备
于室温,将4-(((6-溴-8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(39a)(405mg,0.8mmol)、异丙烯硼酸酯(268.0mg,1.60mmol)、四三苯基膦钯(124mg,0.08mmol)、磷酸钾(508mg,2.40mmol)、二氧六环:水(15mL:3mL)加入到反应瓶中,回流搅拌反应过夜。冷却至室温后,减压浓缩,残余物中加入EtOAc(20mL×3)和水(10mL)。有机相经无水硫酸钠干燥,过滤,减压浓缩。得到373mg黑色油状物的粗品标题产物,直接用于下一步。
LCMS:m/z 468.28[M+H] +
步骤3:4-(((6-异丙基-8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(39c)的制备
于室温,将4-((8-(2-甲基环戊基)-7-氧代-6-(丙-1-烯-2-基)-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(39b)(373mg,0.80mmol)、钯碳(37.0mg)、甲醇(15mL)加入到反应瓶中,氢气氛下,50℃搅拌反应过夜。冷却至室温后,减压浓缩,残余物中加入EtOAc(20mL×3)和水(10mL)。有机相经无水硫酸钠干燥,过滤,减压浓缩,得到180mg油状物的粗品标题产物,直接用于下一步。
LCMS:m/z 470.28[M+H] +
其余步骤与实施例3的制备方法相同,除了用4-(((6-异丙基-8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(39c)代替4-((8-环戊基-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(3e),用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物39。
LCMS:m/z 447.19[M+H] +
1H NMR(400MHz,CDCl 3)δ8.36(s,1H),7.27(s,1H),6.18(d,J=22.3Hz,1H),5.92(dd,J=17.8,9.4Hz,1H),4.07(s,1H),3.85(s,2H),3.19(d,J=11.5Hz,1H),2.92(s,2H),2.86(s,3H),2.55–2.28(m,1H),2.20(s,2H),2.15-2.02(m,5H),1.67(d,J=51.4Hz,3H),1.20-1.19(m,6H),0.76(d,J=7.0Hz,3H)。
实施例40:8-环丁基-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(40)的制备
Figure PCTCN2022119618-appb-000075
与实施例3的制备方法相同,除了用环丁胺代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物40。
LCMS:m/z 377.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.35(s,1H),7.41(d,J=9.4Hz,1H),6.39(d,J=9.1Hz,1H),5.81–5.68(m,1H),4.12(s,1H),3.81(s,2H),3.32–2.51(m,9H),2.35(s,2H),2.26–2.14(m,2H),2.06–1.83(m,2H),1.74(s,2H)。
实施例41:8-(3,3-二氟环丁基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(41)的制备
Figure PCTCN2022119618-appb-000076
与实施例3的制备方法相同,除了用3,3-二氟环丁-1-胺盐酸盐代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环 丙烷磺酰胺(1l),制得化合物41。
LCMS:m/z 413.15[M+H] +
1H NMR(400MHz,CDCl 3)δ8.42(s,1H),7.45(d,J=9.3Hz,1H),6.38(d,J=9.3Hz,1H),5.88(s,1H),4.12(s,1H),3.90(t,J=13.9Hz,4H),2.91(d,J=11.2Hz,4H),2.86(d,J=9.6Hz,3H),2.17(s,2H),1.97(d,J=27.6Hz,2H),1.72-1.56(m,2H)。
实施例42:8-(3,3-二甲基环丁基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(42)的制备
Figure PCTCN2022119618-appb-000077
与实施例3的制备方法相同,除了用3,3-二甲基环丁胺盐酸盐代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物42。
LCMS:m/z 405.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.36(s,1H),7.40(d,J=9.4Hz,1H),6.35(d,J=9.3Hz,1H),5.67(s,1H),4.15–3.99(m,1H),3.84(s,2H),2.99(s,2H),2.88(m,5H),2.56(s,2H),2.19(ddd,J=23.5,14.1,6.6Hz,4H),1.70(s,2H),1.27(d,J=4.0Hz,6H)。
实施例43:8-((1R,2R)-2-羟基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(43)的制备
Figure PCTCN2022119618-appb-000078
Figure PCTCN2022119618-appb-000079
与实施例3的制备方法相同,除了用(1R,2R)-2-氨基环戊醇盐酸盐代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物43。
LCMS:m/z 407.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.45(s,1H),7.50(d,J=9.3Hz,1H),6.44(d,J=9.2Hz,1H),5.82(dd,J=16.0,9.4Hz,1H),4.38(dd,J=10.6,4.5Hz,1H),4.00(s,1H),3.84(s,2H),2.98(s,2H),2.88(s,3H),2.69(s,1H),2.40(s,2H),2.14(ddd,J=16.5,13.1,8.4Hz,3H),2.01(dd,J=12.2,7.1Hz,2H),1.88–1.40(m,5H)。
实施例44:8-((1R,2S)-2-羟基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(44)的制备
Figure PCTCN2022119618-appb-000080
与实施例3的制备方法相同,除了用(1S,2R)-2-氨基环戊醇盐酸盐代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物44。
LCMS:m/z 407.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.45(s,1H),7.51(d,J=9.3Hz,1H),6.43(d,J=9.2Hz,1H),5.82(td,J=9.4,6.9Hz,1H),4.38(dd,J=11.0,4.5Hz,1H),4.01(s,1H), 3.85(s,2H),2.98(d,J=20.4Hz,2H),2.89(s,4H),2.70(d,J=10.0Hz,2H),2.15(ddd,J=20.4,8.3,4.1Hz,3H),2.00(dt,J=11.2,5.6Hz,2H),1.78(d,J=17.1Hz,1H),1.75-1.54(m,3H)。
实施例45:8-((1R,3R)-3-羟基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(45)的制备
Figure PCTCN2022119618-appb-000081
与实施例3的制备方法相同,除了用(1R,3R)-2-氨基环戊醇盐酸盐代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物45。
LCMS:m/z 407.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.45(s,1H),7.50(d,J=9.3Hz,1H),6.44(d,J=9.2Hz,1H),5.82(dd,J=16.0,9.4Hz,1H),4.38(dd,J=10.6,4.5Hz,1H),4.00(s,1H),3.84(s,2H),2.98(s,2H),2.88(s,3H),2.69(s,1H),2.40(s,2H),2.14(ddd,J=16.5,13.1,8.4Hz,3H),2.01(dd,J=12.2,7.1Hz,2H),1.88–1.40(m,5H)。
实施例46:8-((1R,3S)-3-羟基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(46)的制备
Figure PCTCN2022119618-appb-000082
Figure PCTCN2022119618-appb-000083
与实施例3的制备方法相同,除了用(1S,3R)-3-氨基环戊醇盐酸盐代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物46。
LCMS:m/z 407.10[M+H] +
1HNMR(400MHz,CDCl 3)δ8.47(s,1H),7.51(d,J=9.3Hz,1H),6.40(d,J=9.3Hz,1H),6.04(ddd,J=13.8,9.5,4.8Hz,1H),5.35(s,1H),4.34(t,J=4.4Hz,1H),4.05(s,1H),3.85(s,2H),2.95(s,2H),2.86(s,3H),2.61-2.46(m,1H),2.33(ddd,J=14.8,12.0,5.7Hz,2H),2.26-2.17(m,2H),2.13(s,1H),2.06(dd,J=13.3,6.9Hz,2H),1.91(dt,J=12.2,8.1Hz,1H),1.68(s,3H)。
实施例47:8-(3,3-二氟环戊基)-2-(((1-(S-甲基磺酰亚氨基)哌啶丁-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(47)的制备
Figure PCTCN2022119618-appb-000084
与实施例3的制备方法相同,除了用3,3-二氟环戊胺盐酸盐代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物47。
LCMS:m/z 427.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.42(s,1H),7.46(d,J=9.4Hz,1H),6.40(d,J=8.9 Hz,1H),6.09(dd,J=18.3,9.1Hz,1H),4.16-4.02(m,1H),3.87(s,2H),3.16(d,J=36.7Hz,1H),2.96(s,2H),2.89(s,3H),2.86-2.44(m,4H),2.31(d,J=11.3Hz,1H),2.25-2.06(m,4H),1.69(d,J=10.0Hz,2H)。
实施例48:8-(3,3-二甲基环戊基)-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(48)的制备
Figure PCTCN2022119618-appb-000085
与实施例3的制备方法相同,除了用3,3-二甲基环戊烷-1-胺代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物48。
LCMS:m/z 419.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.44-7.41(d,J=12Hz,1H),6.40-6.37(d,J=12Hz,1H),5.90-5.78(m,1H),4.03(s,1H),3.85(s,2H),2.95(s,2H),2.86(s,3H),2.62-2.20(m,2H),2.09-1.98(m,6H),1.67-1.50(m,3H),1.50(s,1H),1.16(s,3H),0.85(s,3H)。
实施例49:8-(3-甲基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(49)的制备
Figure PCTCN2022119618-appb-000086
Figure PCTCN2022119618-appb-000087
步骤1:N-苄基-3-甲基环戊烷-1-胺(49a)的制备
于室温,将3-甲基环戊酮(10.0g,101.9mmol)、DCM(200mL)、苄胺(9.84g,91.7mmol)、乙酸(10.0mL)依次加入至反应瓶中,室温搅拌1.5小时。分批加入三乙酰氧基硼氢化钠(43.2g,203.8mmol),室温搅拌过夜。将反应液浓缩后再次以DCM稀释,以饱和NaOH水溶液调节PH至10,水相以DCM萃取,合并有机相后浓缩得25.0g粗品黄色油状标题产物,直接用于下一步。
LCMS:m/z 190.15[M+H] +
步骤2:3-甲基环戊胺盐酸盐(49b)的制备
于室温,将N-苄基-3-甲基环戊烷-1-胺(25.0g,粗品)、异丙醇(200mL)加入至反应瓶中,氮气氛下加入氢氧化钯碳(3.00g)。置换氢气,45℃反应过夜。反应结束后过滤,滤液降温至5℃,滴加盐酸二氧六环溶液(4.0M,50mL),室温搅拌过夜。反应液减压浓缩,得17.9g红褐色固体粗品,直接用于下一步。
LCMS:m/z 100.10[M+H] +
其余步骤与实施例3的制备方法相同,除了用3-甲基环戊胺盐酸盐(49b)代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得标题化合物。
LCMS:m/z 405.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.37(s,1H),7.41(d,J=9.3,1H),6.38(d,J=9.1,1H),5.90(ddd,J=14.6,13.6,7.2,1H),4.17-3.96(m,1H),3.83(s,2H),3.00(d,J=20.9,2H),2.86(s,3H),2.78-2.50(m,2H),2.40(s,2H),2.28-2.05(m,3H),2.05-1.97(m,1H),1.97-1.81(m,2H),1.70(d,J=8.4,2H),1.50(s,1H),1.26(s,1H),1.10(dd,J=31.4,6.5,3H)。
实施例50:8-环己基-2-(((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(50)的制备
Figure PCTCN2022119618-appb-000088
Figure PCTCN2022119618-appb-000089
与实施例3的制备方法相同,除了用环己胺代替环戊胺和用N-(叔丁基二甲基甲硅烷基)甲磺酰胺(11a)代替N-(叔丁基二甲基甲硅烷基)环丙烷磺酰胺(1l),制得化合物50。
LCMS:m/z 405.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.36(s,1H),7.40(d,J=9.3Hz,1H),6.38(s,1H),5.32(t,J=12.2Hz,1H),4.02(s,1H),3.86(s,2H),2.97(s,2H),2.86(s,3H),2.68(d,J=11.4Hz,4H),2.22(t,J=8.8Hz,2H),1.90(d,J=12.2Hz,2H),1.69(s,5H),1.34(d,J=55.9Hz,3H)。
实施例51:6-(二氟甲基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)-8-(螺[2.4]庚-4-基)吡啶并[2,3-d]嘧啶-7-(8H)-酮(51)的制备
Figure PCTCN2022119618-appb-000090
与实施例33的制备方法相同,除了用2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)-8-(螺[2.4]庚-4-基)吡啶并[2,3-d]嘧啶-7(8H)-酮(25)代替8-(2-甲基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(22),制得化合物51。
LCMS:m/z 433.23[M+H] +
1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.66(s,1H),5.99(dd,J=16.8,9.7Hz,1H),4.04(s,1H),3.84(s,2H),3.05(d,J=26.6Hz,3H),2.91(s,3H),2.64(s,1H),2.36(dtd,J=17.4,7.1,3.3Hz,1H),2.26-2.15(m,2H),2.14-2.06(m,1H),2.03-1.84(m,3H),1.81-1.44(m,4H),0.79(d,J=7.1Hz,3H)。
实施例52:6-氯-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)-8-(螺[2.4]庚-4-基) 吡啶[2,3-d]嘧啶-7(8H))-酮(52)的制备
Figure PCTCN2022119618-appb-000091
与实施例38的制备方法相同,除了用4-(((7-氧代-8-(螺[2.4]庚基-4-基]-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基]氨基)哌啶-1-甲酸叔丁酯(25e)代替4-(((8-(2-甲基环戊基)-7-氧代-7,8-二氢吡啶并[2,3-d]嘧啶-2-基)氨基)哌啶-1-羧酸叔丁酯(22e),制得化合物52。
LCMS:m/z 451.20[M+H] +
1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.66(s,1H),5.99(dd,J=16.8,9.7Hz,1H),4.04(s,1H),3.84(s,2H),3.05(d,J=26.6Hz,3H),2.91(s,3H),2.64(s,1H),2.36(dtd,J=17.4,7.1,3.3Hz,1H),2.26-2.15(m,2H),2.14-2.06(m,1H),2.03-1.84(m,3H),1.81-1.44(m,4H),0.79(d,J=7.1Hz,3H)。
实施例53:8-环戊基-6-(二氟甲基)-2-(((1-(噻吩-2-磺酰亚氨基))哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(53)的制备
Figure PCTCN2022119618-appb-000092
与实施例33的制备方法相同,除了用8-环戊基-2-(((1-(噻吩-2-磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(36)代替8-(2-甲基环戊基)-2-((1-(S-甲基磺酰亚氨基)哌啶-4-基)氨基)吡啶并[2,3-d]嘧啶-7(8H)-酮(22),制得标题化合物。
LCMS:m/z 509.15[M+H] +
1H NMR(400MHz,CDCl3)δ8.45(s,1H),7.77(s,1H),7.59(dd,J=19.5,3.7Hz,2H),7.22-7.12(m,1H),6.78(t,J=55.2Hz,1H),5.78(s,1H),3.92(s,2H),3.73(s,1H),2.79(s,2H),2.58(s,1H),2.30(s,2H),2.14(s,2H),1.97(d,J=28.0Hz,2H),1.84(dd, J=12.7,6.9Hz,2H),1.77(d,J=19.3Hz,2H),1.72-1.51(m,3H)。
生物学测试
试验例1:本发明化合物对CDK2/4/6的抑制活性
(1)CDK2/CyclinE1激酶活性抑制
使用ADP-Glo激酶测定试剂盒(Promega,货号V9102)检测待测化合物对CDK2/CyclinE1激酶的抑制活性。在激酶反应中,激酶将底物磷酸化,同时消耗ATP,残留ATP,能被Ultra-Glo TM萤光素酶转化成光,发光信号与激酶活性正相关,检测这种发光信号值,即可反应出激酶活性。
试验方法
首先,将待测化合物在DMSO(Sigma,货号D8418)中溶解,然后稀释为200nM浓度,设定试验起始浓度100nM,3倍稀释,10个梯度。用Echo 550在384孔反应板中(Greiner,货号784075)加入5μl稀释后的化合物和2.5μl CDK2/Cyclin E1激酶(Carna,货号04-165),用封板膜封住板子,1000rpm离心1分钟,25℃孵育10分钟。再向反应板中加入2.5μl Histone H1蛋白(SignalChem,货号H10-54N)和ATP(Promega,货号V910B)的混合液,1000rpm离心1分钟,25℃孵育反应60分钟。吸取5μl的ADP-Glo试剂(Promega,货号V9102)到反应板中,瞬时离心后,孵育40分钟。最后加入10μl的检测试剂到反应板中,孵育40分钟。使用Envision 2104多功能读板机(PerkinElmer,型号Oct-04)读取相对荧光单元(relative luminescence unit,RLU)值。RLU值大小用于表征酶与底物反应程度。实验数据采用GraphPad prism 6.0软件进行非线性拟合公式计算化合物IC 50值:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50-X)×HillSlope))。
X为化合物浓度对数值,Y为抑制的百分比,Bottom最小抑制百分比,Top为最大抑制百分比,HillSlope为曲线斜率。
(2)CDK6/CyclinD1激酶活性抑制
使用ADP-Glo激酶测定试剂盒(Promega,货号V9102)检测待测化合物对CDK6/CyclinD1激酶的抑制活性。
试验方法
首先,将待测化合物溶解在DMSO(Sigma,货号D8418)中,设定试验起始浓度1000nM,3倍稀释,10个梯度。用Echo 550在384孔反应板中(Greiner,货号784075)加入5μl稀释后的化合物和2.5μl CDK6/CyclinD1激酶(ThermoFisher,货号PV4401),用封板膜封住板子,瞬时离心30s,孵育10分钟。再向反应板中加入2.5μl Histone H1蛋白(SignalChem,货号H10-54N)和ATP(Promega,货号V910B)的混合液,1000g离心30s,25℃孵育反应60分钟。再加入5μl的ADP-Glo试剂(Promega,货号V9102)到反应板中,瞬时离心后,孵育40分钟。最后加入10μl的检测试剂到反应板中,孵育40分钟。使用Envision 2104多功能 读板机(PerkinElmer,型号Oct-04)读取RLU值。RLU值大小用于表征酶与底物反应程度。实验数据采用GraphPad prism 6.0软件进行非线性拟合公式计算化合物的IC 50值:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50-X)×HillSlope))。
X为化合物浓度对数值,Y为抑制的百分比,Bottom最小抑制百分比,Top为最大抑制百分比,HillSlope为曲线斜率。(3)CDK4/CyclinD3激酶活性抑制
使用ADP-Glo激酶测定试剂盒(Promega,货号V9102)检测待测化合物对CDK4/CyclinD3激酶的抑制活性。
试验方法
首先,将待测化合物溶解在DMSO(Sigma,货号D8418)中,设定试验起始浓度1000nM,3倍稀释,10个梯度。用Echo 550在384孔反应板中(Greiner,货号784075),加入5μl稀释后的化合物和2.5μl CDK4/CyclinD3激酶(Carna,货号04-105),用封板膜封住板子,瞬时离心30s,孵育10分钟。再向反应板中加入2.5μl Histone H1蛋白(SignalChem,货号H10-54N)和ATP(Promega,货号V910B)的混合液,1000g离心30s,25℃孵育反应1h。再加入5μl的ADP-Glo试剂(Promega,货号V9102)到反应板中,瞬时离心后,孵育40分钟。最后加入10μl的检测试剂到反应板中,孵育40分钟。使用Envision 2104多功能读板机(PerkinElmer,型号Oct-04)读取RLU值。RLU值大小用于表征酶与底物反应程度。实验数据采用GraphPad prism 6.0软件进行非线性拟合公式计算化合物的IC 50值:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50-X)×HillSlope))。
X为化合物浓度对数值,Y为抑制的百分比,Bottom最小抑制百分比,Top为最大抑制百分比,HillSlope为曲线斜率。
本发明化合物对CDK2、CDK6和CDK4激酶抑制的IC 50值如下表1所示。
表1本发明化合物对CDK2、CDK6和CDK4激酶抑制的IC 50
Figure PCTCN2022119618-appb-000093
Figure PCTCN2022119618-appb-000094
Figure PCTCN2022119618-appb-000095
结论:从表1可以看出,本发明化合物具有良好的CDK2/4/6激酶抑制活性。
试验例2:本发明化合物对CDK2/4/6途径的细胞学抑制水平
(1)OVCAR3和HCC1806细胞增殖试验
通过细胞增殖实验,检测待测化合物对卵巢癌细胞系OVCAR3和乳腺癌细胞系HCC1806的抑制水平,根据检测指标IC 50,筛选候选化合物。
OVCAR3和HCC1806细胞株(均购买自南京科佰生物公司,ATCC编号分别为
Figure PCTCN2022119618-appb-000096
HTB-161 TM
Figure PCTCN2022119618-appb-000097
CRL-2335 TM)培养于RPMI1640(Gibco,C11875500BT),加入10%的FBS(Gbico,10099141)和双抗(1%的青霉素和链霉素,Gibco公司,15140-122)。5,000个OVCAR3或HCC1806细胞接种在白色透明底96孔(Nunc,249944)/384孔板中(Corning,3570),置于37℃,5%的培养箱内过夜。第二天,加入待测化合物,化合物用DMSO溶解,稀释,试验起始浓度从10mM开始,3倍稀释,设置10个浓度梯度,每个梯度3个复孔。将细胞板放置培养箱中,37℃,5%CO 2共培养7天。使用CELL Titer-GLO发光法,检测总的ATP含量来测定细胞增殖水平。将384孔板细胞取出,室温平衡30min;每孔加入30μL CTG(CTG,Promega,货号G7572),振荡混匀,室温孵育10min;多功能酶标仪(Biotek,型号Cytation 3)读取荧光值。GraphPad Prism 6.0软件分析不同浓度下化合物反应的Log值来测定增殖抑制的IC 50
(2)MCF7细胞增殖试验
人乳腺癌细胞(MCF7细胞)(购自ATCC,编号为HTB-22)培养于EMEM培养基中(ATCC,30-2003),加入10%FBS(Gbico,10099141)。待细胞生长至70~80%的汇合度,消化细胞,制备细胞悬液。接种细胞在384孔板中(Corning,3570),500个/孔,置于37℃,5%的培养箱内过夜。第二天加入待测化合物,化合物用DMSO溶解,稀释,试验起始浓度从10mM开始,3倍稀释,设置10个浓度梯度,每个梯度3个复孔。将细胞板放置培养箱中,37℃,5%CO 2共培养7天。使用CELL Titer-GLO发光法,检测总的ATP含量来测定细胞增殖水平,室温平衡30min;每孔加入20μL CTG(CTG,Promega,货号G7572),振荡混匀,室温孵育10min;多功能酶标仪(Biotek,型号Cytation 3)读取荧光值。GraphPad Prism6.0软件分析不同浓度下化合物反应的Log值来测定增殖抑制的IC 50
本发明化合物对OVCAR3,HCC1806和MCF-7细胞抑制的IC 50值如下表2所示。
表2本发明化合物对OVCAR3、HCC1806和MCF-7细胞抑制的IC 50
Figure PCTCN2022119618-appb-000098
结论:从表2可以看出,本发明化合物对OVCAR3、HCC1806和MCF-7细胞增殖具有显著抑制作用。
试验例3:本发明化合物的小鼠药代动力学实验
实验动物选用7-8周龄雄性ICR小鼠,购自北京维通利华实验动物技术有限公司,饲养于SPF环境,温度20~26℃,每日温差不超过4℃,相对湿度40~70%RH,每日12h/12h交替照明。实验动物经过3-5天的适应期,其中口服给药动物于实验前1天禁食过夜(>12h),不禁水。
每个待测化合物分为iv(静脉给药)和po(灌胃)组(n=6),剂量设为iv 1mg/kg、p.o 10mg/kg。静脉组和灌胃组溶媒均为3%DMSO+97%(20%HP-β-CD溶液)。化合物溶液配制流程如下:先用DMSO溶解化合物,配成10mg/mL的储液;取100 μL储液,用20%的HP-β-CD定容至5mL,得到浓度为0.2mg/mL的静脉给药溶液;取100μL的储液,加入溶媒定容至5mL,涡旋,使分散均匀,得到化合物浓度为1mg/mL的灌胃给药溶液。
给药前称量体重,通过眼静脉丛采血方式,采集0.1mL血样加入肝素钠抗凝管中,防止凝血。每个受试化合物6只静脉给药,6只口服给药,并于给药后2h给予食物。样品采集时间点为,灌胃组:给药前及给药后5min、15min、30min、1h、2h、4h;静脉组:给药前及给药后5min、15min、30min、1h、2h、4h。动物采血分成2部分分别进行,采用交叉时间点采血,同1只小鼠最多设5个采血点。在取血60min内,用Sorvall ST 8R高速低温离心机以3000rpm离心10min,取上层血浆,冻存到-20℃冰箱内,备用。使用LC-MS/MS的分析方法检测样品中化合物浓度。采用MAS Studio(V1.3.1stable)软件计算并得到化合物在小鼠体内的血药浓度-时间曲线,以及主要的PK参数:AUC 0-t、C max、T max、T 1/2和F%。
F%=(AUC po×剂量 iv)/(AUC iv×剂量 po)×100%。
本发明化合物对小鼠的药代动力学参数如下表3所示。
表3本发明化合物的小鼠药代动力学参数
Figure PCTCN2022119618-appb-000099
结论:从表3可以看出,本发明化合物在小鼠的药代动力学实验中具有较好的性质。

Claims (18)

  1. 一种通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    Figure PCTCN2022119618-appb-100001
    其中,
    环A选自杂环基,所述杂环基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;
    环B选自环烷基、杂环基、芳基和杂芳基;
    R 1选自烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、烯基、炔基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;
    R 2选自氢、烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、烯基、炔基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;或者,
    R 1和R 2与他们连接的氮原子和硫原子一起形成杂环基或杂芳基,所述杂环基或杂芳基任选进一步被选自卤素、氨基、硝基、氰基、氧代基、羟基、巯基、羧基、酯基、烷基、烷氧基、卤代烷基、卤代烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基的一个或多个基团取代;
    每个R 3各自独立地选自卤素、氨基、硝基、氰基、羟基、巯基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基,所述烷基、烷氧基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;或者,
    任意两个R 3与他们连接的原子一起形成环烷基、杂环基、芳基和杂芳基,所述环烷基、杂环基、芳基和杂芳基任选进一步被选自卤素、氨基、硝基、氰基、氧代基、羟基、巯基、羧基、酯基、烷基、烷氧基、卤代烷基、卤代烷氧基、烯 基、炔基、环烷基、杂环基、芳基和杂芳基的一个或多个基团取代;
    R 4选自氢、卤素、氨基、硝基、氰基、羟基、巯基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-C(O)R a、-O(O)CR a、-C(O)OR a、-C(O)NR aR b、-NHC(O)R a、-S(O) mR a、-S(O) mNR aR b和-NHS(O) mR a,所述烷基、烷氧基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;
    R 5选自氢、卤素、氨基、硝基、氰基、羟基、巯基、烷基、烷氧基、酰基、环烷基、杂环基、芳基、杂芳基、-C(O)R a、-O(O)CR a、-C(O)OR a、-C(O)NR aR b、-NHC(O)R a、-S(O) mR a、-S(O) mNR aR b和-NHS(O) mR a,所述烷基、烷氧基、环烷基、杂环基、芳基、杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;
    R a和R b各自独立地选自氢、卤素、羟基、氰基、氨基、羧基、酯基、烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中所述烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基中的一个或多个基团取代;
    或者R a和R b与他们连接的原子一起形成环烷基或杂环基,所述环烷基或杂环基任选进一步被选自卤素、氨基、硝基、氰基、氧代基、羟基、巯基、羧基、酯基、烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基中的一个或多个基团取代;
    m为0、1、2;
    p为0、1、2、3或4。
  2. 根据权利要求1所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中,
    环A选自3至12元单环杂环基、螺杂环基、稠杂环基或桥杂环基,优选5至7元单环杂环基、7至10元螺杂环基、7至10元稠杂环基和7至10元桥杂环基,更优选吡咯烷基、哌啶基、哌嗪基,所述杂环基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、氧代基、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基、C 3-C 6环烷基、5至7元杂环基、C 6-C 10芳基、5至10元杂芳基的一个或多个基团所取代。
  3. 根据权利要求1或2所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(II)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    Figure PCTCN2022119618-appb-100002
    其中,环B、R 1、R 2、R 3、R 4、R 5、p如权利要求1所定义。
  4. 根据权利要求1至3中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中,
    环B选自C 3-C 7环烷基、4至7元杂环基、C 6-C 10芳基、5至10元杂芳基,更优选
    Figure PCTCN2022119618-appb-100003
  5. 根据权利要求1至4中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其为通式(III)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    Figure PCTCN2022119618-appb-100004
    其中,R 1、R 2、R 3、R 4、R 5、p如权利要求1所定义。
  6. 根据权利要求1至5中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中,
    每个R 3各自独立地选自卤素、氨基、氰基、羟基、巯基、C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 7环烷基、4至7元杂环基,所述C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 7环烷基、4至7元杂环基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基的一个或多个基团所取代;优选卤素、羟基、C 1-C 6烷基、C 1-C 6卤代烷基;
    p为0、1、2或3;优选1或2。
  7. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中,
    R 1选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 6-C 10芳基和5至10元杂芳基,优选C 1-C 6烷基、C 3-C 6环烷基;优选C 1-C 6烷基、C 3-C 6环烷基、5至7元杂环基;
    所述C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 3-C 6环烷基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、C 1-C 6烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 1-C 6卤代烷基、C 1-C 6烷氧基的一个或多个基团所取代;
    所述5至7元杂环基、C 6-C 10芳基和5至10元杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、C 1-C 6烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 1-C 6卤代烷基、C 1-C 6烷氧基、C 6-C 10芳基、5至10元杂芳基的一个或多个基团取代。
  8. 根据权利要求1至7中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中,
    R 2选自氢、C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 6-C 10芳基和5至10元杂芳基,优选氢、C 1-C 6烷基、C 3-C 6环烷基;优选氢和C 1-C 6烷基;
    所述C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 3-C 6环烷基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、C 1-C 6烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 1-C 6卤代烷基、C 1-C 6烷氧基的一个或多个基团所取代;
    所述5至7元杂环基、C 6-C 10芳基和5至10元杂芳基任选进一步被选自卤素、氨基、硝基、氰基、羟基、巯基、羧基、酯基、氧代基、C 1-C 6烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 1-C 6卤代烷基、C 1-C 6烷氧基、C 6-C 10芳基、5至10元杂芳基的一个或多个基团取代。
  9. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中,R 1和R 2与他们连接的氮原子和硫原子一起形成5至7元杂环基,所述5至7元杂环基任选进一步被选自卤素、氨基、硝基、氰基、氧代基、羟基、巯基、羧基、酯基、C 1-C 6烷基、C 1-C 6烷氧基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、5至7元杂环基、C 1-C 6卤代烷基、C 1-C 6烷氧基、C 6-C 10芳基、5至10元杂芳基的一个或多个基团取代。
  10. 根据权利要求1至9中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中,
    R 4选自氢、卤素、氨基、硝基、氰基、羟基、巯基、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基,优选氢、卤素、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基;优选R 4为氢或C 1-C 6烷基。
  11. 根据权利要求1至10中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,
    其中,
    R 5选自氢、卤素、氨基、氰基、羟基、巯基、C 1-C 6烷基、C 1-C 6烷氧基、乙酰基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基、-C(O)R a,优选氢、卤素、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6卤代烷基、C 1-C 6卤代烷氧基、-C(O)R a
    R a选自C 1-C 6烷基。
  12. 根据权利要求1至11中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐,其中所述化合物选自:
    Figure PCTCN2022119618-appb-100005
    Figure PCTCN2022119618-appb-100006
    Figure PCTCN2022119618-appb-100007
    Figure PCTCN2022119618-appb-100008
  13. 根据权利要求1至12中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐的制备方法,其包括以下步骤:
    Figure PCTCN2022119618-appb-100009
    在碱的存在下,在催化剂存在下,化合物Ij与化合物Ia发生脱水反应得到通式(I)化合物;所述碱优选三乙胺;所述催化剂优选三苯基二氯化磷;
    环A、环B、R 1、R 2、R 3、R 4、R 5、p如前述权利要求中任一项所定义。
  14. 一种药物组合物,其包含根据权利要求1至12中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐和药学上可接受的载体或赋形剂。
  15. 根据权利要求1至12中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或根据权利要求14所述的药物组合物在制备细胞周期蛋白依赖性激酶(CDK)抑制剂中的用途。
  16. 根据权利要求1至12中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或根据权利要求14所述的药物组合物在制备抑制癌细胞增值、抑制癌细胞侵袭或诱导癌细胞凋亡的药物中的用途。
  17. 根据权利要求1至12中任一项所述的通式(I)所示的化合物或其立体异构体、互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或根据权利要求14所述的药物组合物在制备用于预防和/或治疗与细胞周期蛋白-依赖性激酶活性相关的疾病的药物中的用途,所述疾病例如癌症,特别是与以细胞周期蛋白依赖性激酶CDK2/细胞周期蛋白E1(CCNE1)、CDK6/细胞周期蛋白D1、CDK4/细胞周期蛋白D3的扩增或过表达为特征的癌症, 更特别为乳腺癌如HR+/HER2-转移性乳腺癌或卵巢癌。
  18. 根据权利要求15至17中任一项所述的用途,其中所述药物可以与另外一种抗癌治疗剂或抗癌治疗方法同时、分开或相继施用。
PCT/CN2022/119618 2021-09-29 2022-09-19 具有细胞周期蛋白依赖性激酶抑制活性的杂环化合物及其制备方法和医药用途 WO2023051302A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280006843.5A CN116390921A (zh) 2021-09-29 2022-09-19 具有细胞周期蛋白依赖性激酶抑制活性的杂环化合物及其制备方法和医药用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111147372.3 2021-09-29
CN202111147372 2021-09-29

Publications (1)

Publication Number Publication Date
WO2023051302A1 true WO2023051302A1 (zh) 2023-04-06

Family

ID=85781290

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/119618 WO2023051302A1 (zh) 2021-09-29 2022-09-19 具有细胞周期蛋白依赖性激酶抑制活性的杂环化合物及其制备方法和医药用途

Country Status (3)

Country Link
CN (1) CN116390921A (zh)
TW (1) TW202322805A (zh)
WO (1) WO2023051302A1 (zh)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109803968A (zh) * 2016-08-15 2019-05-24 辉瑞公司 吡啶并嘧啶酮cdk2/4/6抑制剂
WO2021003314A1 (en) * 2019-07-02 2021-01-07 Nuvation Bio Inc. Heterocyclic compounds as kinase inhibitors
WO2021170076A1 (en) * 2020-02-28 2021-09-02 Fochon Pharmaceuticals, Ltd. Compounds as cdk2/4/6 inhibitors
WO2021254384A1 (zh) * 2020-06-17 2021-12-23 微境生物医药科技(上海)有限公司 新型吡啶并[2,3-d]嘧啶-7(8H)-酮衍生物

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109803968A (zh) * 2016-08-15 2019-05-24 辉瑞公司 吡啶并嘧啶酮cdk2/4/6抑制剂
WO2021003314A1 (en) * 2019-07-02 2021-01-07 Nuvation Bio Inc. Heterocyclic compounds as kinase inhibitors
WO2021170076A1 (en) * 2020-02-28 2021-09-02 Fochon Pharmaceuticals, Ltd. Compounds as cdk2/4/6 inhibitors
WO2021254384A1 (zh) * 2020-06-17 2021-12-23 微境生物医药科技(上海)有限公司 新型吡啶并[2,3-d]嘧啶-7(8H)-酮衍生物

Also Published As

Publication number Publication date
CN116390921A (zh) 2023-07-04
TW202322805A (zh) 2023-06-16

Similar Documents

Publication Publication Date Title
TWI714567B (zh) 作為lsd1抑制劑之雜環化合物
TWI698435B (zh) 作為吲哚胺2,3-二加氧酶和/或色氨酸2,3-二加氧酶抑制劑之新穎之5或8-取代之咪唑並[1,5-a]吡啶
WO2021129824A1 (zh) 新型K-Ras G12C抑制剂
CN104105697B (zh) 二环哌嗪化合物
WO2020239077A1 (zh) 含氮杂环类衍生物调节剂、其制备方法和应用
CA2692922C (en) Azaindole-indole coupled derivatives, preparation methods and uses thereof
WO2012019427A1 (zh) 酞嗪酮类衍生物、其制备方法及其在医药上的应用
BRPI0811600A2 (pt) &#34; imidazo [1,2-b]piridazinas 3,6-dissubstituídas e pirrol[1,5-a]pirimidinas 3,5-dissubstituídas como inibidores de fosfatidilinositol-3-cinase &#34;.
TWI580679B (zh) 雜芳基並嘧啶類衍生物、其製備方法和用途
ES2949357T3 (es) Inhibidores de 3-fosfoglicerato deshidrogenasa y usos de los mismos
TW202102505A (zh) 吡咯并雜環類衍生物、其製備方法及其在醫藥上的應用
JP6290412B2 (ja) イミダゾロン系誘導体、その医薬組成物及び用途
CN113387962A (zh) 吡唑并[3,4-d]嘧啶-3-酮衍生物、其药物组合物及应用
WO2014079364A1 (zh) 咪唑酮类衍生物、其药物组合物和用途
TW202317564A (zh) Cdk2抑制劑及其製備方法和用途
KR20240016938A (ko) 포스포이노시티드 3 키나아제 베타 억제제 및 그 조성물 및 방법
US20230067237A1 (en) Caffeine inhibitors of mthfd2 and uses thereof
JP2020504139A (ja) キナーゼ阻害剤としての置換された縮合ヘテロアリール化合物及びその用途
WO2021254384A1 (zh) 新型吡啶并[2,3-d]嘧啶-7(8H)-酮衍生物
TW202325280A (zh) 一種胺基吡唑衍生物及其製備方法和用途
CN107428762A (zh) 酞嗪酮衍生物、其制备方法及用途
CN111848678A (zh) 含磷类噻吩并嘧啶衍生物
WO2019223777A1 (zh) 一种含有芳胺基取代的吡咯并嘧啶类化合物、制备方法及其应用
WO2023020347A1 (zh) 嘧啶并吡啶类化合物及其制备方法和医药用途
CN116789647A (zh) 作为parp7抑制剂的化合物

Legal Events

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

Ref document number: 22874689

Country of ref document: EP

Kind code of ref document: A1