CN116390921A - Heterocyclic compound with cyclin-dependent kinase inhibition activity, preparation method and medical application thereof - Google Patents

Heterocyclic compound with cyclin-dependent kinase inhibition activity, preparation method and medical application thereof Download PDF

Info

Publication number
CN116390921A
CN116390921A CN202280006843.5A CN202280006843A CN116390921A CN 116390921 A CN116390921 A CN 116390921A CN 202280006843 A CN202280006843 A CN 202280006843A CN 116390921 A CN116390921 A CN 116390921A
Authority
CN
China
Prior art keywords
alkyl
cycloalkyl
heterocyclyl
compound
alkoxy
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280006843.5A
Other languages
Chinese (zh)
Inventor
闫旭
陈振华
刘国标
谷晓成
尚飞
杜佩金
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
National Institutes of Pharmaceutical R&D Co Ltd
Original Assignee
National Institutes of Pharmaceutical R&D Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by National Institutes of Pharmaceutical R&D Co Ltd filed Critical National Institutes of Pharmaceutical R&D Co Ltd
Publication of CN116390921A publication Critical patent/CN116390921A/en
Pending legal-status Critical Current

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

Landscapes

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

Abstract

A heterocyclic compound with cyclin-dependent kinase inhibiting activity, and its preparation method and medicinal application are provided. In particular to a compound shown in a general formula (I), a preparation method thereof, a pharmaceutical composition containing the compound and application of the compound as a Cyclin Dependent Kinase (CDK) inhibitor in prevention and/or treatmentUse in abnormal cell growth such as cancer. Wherein each group in the general formula (I) is defined as the specification.

Description

Heterocyclic compound with cyclin-dependent kinase inhibition activity, preparation method and medical application thereof Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a pyrimidopyridine compound, a preparation method thereof, a pharmaceutical composition containing the pyrimidopyridine compound and application of the pyrimidopyridine compound serving as a Cyclin Dependent Kinase (CDK) inhibitor in medicines for treating abnormal cell growth such as cancers. The compounds of the present invention inhibit cyclin dependent kinases CDK 2/cyclin E1 (CCNE 1), CDK 6/cyclin D1, CDK 4/cyclin D3 and are therefore useful in the treatment of cancers such as HR+/HER 2-metastatic breast cancer, ovarian cancer and the like.
Background
Cyclin Dependent Kinases (CDKs) belong to the serine/threonine protein kinase family and are key proteins that regulate the cell cycle. Currently, CDKs have been found to predominantly include CDKs 1-15, etc., which function by binding to the corresponding cyclin to form a stable form of CDK/cyclin complex. In addition, the cell contains endogenous CDK and CDK/cyclin complex inhibitor (CKI), which together form the regulatory network of the cell cycle, and are tightly controlled during cell division. In many cancer cells, deregulation of expression of three of CDK, cyclin and CKI is found, and CDK/cyclin complexes are often overexpressed.
CDK4 and CDK6 are very similar in structure and function and bind to the 3 isoforms of cyclin D (cyclin D1/D2/D3) to form complexes that phosphorylate a range of substrates including retinoblastoma proteins (Retinoblastoma protein, rb), contributing to cell cycle progression. Phosphorylated Rb protein releases the transcription factor E2F, etc., which binds to it, E2F activates and transcribes a series of Genes, allowing the cell to enter S phase, initiating DNA replication (Kato et al, genes Dev,1993,7 (3), 331-342; dyson et al, genes Dev,1998, 12 (15), 2245-2262). The CDK4/6 activity is also affected by the INK4 family (including p16 INK4A 、p15 INK4B 、p18 INK4C 、p19 INK4D ) INK4 as an endogenous inhibitor protein, can bind competitively to CDKs, and inhibit the formation of the cyclin D-CDK4/6 complex (Baker et al, genes&Cancer,2012,3(11-12),658-669)。
However, in most human cancers, aberrant sustained activation of the cyclin D-CDK4/6-p16-Rb pathway is found, which may promote rapid progression in the G1 phase, leading to aberrant proliferation of cells. The main reasons for this include that gene rearrangement or gene amplification leads to overexpression of cyclin D1 (Bergsagel et al, blood,2005, 106 (1), 296-303); deletion of the p16.sup.INK4a gene, point mutation, or DNA methylation leads to inactivation of p16.sup.INK4a (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 abnormalities in this pathway makes CDK4/6 one of the popular antitumor targets.
The fastest developing treatments targeting CDKs are selective CDK4/6 inhibitors. The first generation of broad-spectrum CDK inhibitors Flavopiridol (Alvocidib) inhibited CDK1/2/4/6/7/9 simultaneously, but had adverse effects such as debilitation, diarrhea, myelosuppression, etc. due to serious toxic side effects, and finally failed to be applied clinically (Senderowicz et al Journal of Clinical Oncology,1998, 16 (9), 2986-2999). Currently, three compounds Palbociclib, ribociclib and Abemaciclib are approved by the FDA for the first-line or second-line treatment of advanced breast cancer, and one drug Lerociclib is in the clinical development stage.
Palbociclib was the first CDK4/6 inhibitor approved by the FDA. The pyridine compound is an oral pyridine compound, and can inhibit proliferation of ER+ breast cancer cells by reducing phosphorylation of Rb protein, preventing cell cycle from entering S phase from G1 phase. A number of large, randomized, prospective clinical studies have demonstrated that Palbociclib in combination with letrozole or Fulvestrent significantly prolonged patient progression free survival and reliable safety (Cristofali et al, 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). These clinical trial data support Palbociclib in combination with aromatase inhibitors or fulvestrant as standard first line care regimens for treating female patients with er+/HER 2-premenopausal or postmenopausal breast cancer metastasis.
Ribociclib (Kisqali, LEE 011) and Abemaciclib (Verzenio, LY 2835219) are both orally reversible CDK4/6 inhibitors capable of causing G1 phase arrest by inhibiting Rb phosphorylation. IC for CDK4/6 inhibition by Ribociclib 50 Values of 10nM and 39nM, respectively, while being less sensitive to other CDK family members (IC for CDK1 and CDK 2) 50 Values were all greater than 50 mM) (Sherm et al, cancer Discovery,2016,6 (4), 353-367). After 1-4 hours of oral administration, the blood plasma can reach Cmax value; blood stability was achieved after 8 days of continuous administration. The binding rate of Ribociclib to human plasma protein was 70% and it was decomposed in vivo by CPY3A4, a weak inhibitor, the main products being N-hydroxy and N-desmethyl derivatives. Abemaciclib has higher selectivity to CDK4/6 kinase, IC 50 Values of 2nM and 5nM, respectivelyIC inhibiting CDK1 and CDK2 50 Value of>500nM. Abemaciclib is a phenylpyrimidine compound similar in structure to Palbocilib and Ribociclib. Abemaciclib exhibits a broader inhibition and is able to inhibit both Dyrk, PIM, HIPK and CAMK kinase families (Ki<10 nM) (Chen et al, molecular Cancer Therapeutics,2016, 15 (10), 2273-2281). The in vivo medicine algebra shows that the peak value is reached within 4-24 hours after oral administration, and the steady state is reached after continuous administration for 5 days. Abemaciclib has a binding rate of about 96% with human plasma protein and is able to be decomposed in vivo by CPY3A4, the main decomposition product being an N-deethylated derivative. The introduction of deuterium atoms makes the compound have stronger metabolic stability, the metabolic stability is improved by 11-45%, and the compound has higher half-life. The drug was approved for the treatment of ER+/HER 2-advanced or metastatic breast cancer in combination with Fluvestrant. In addition, abemaciclib, in combination with gemcitabine, also showed synergistic antitumor activity in xenograft tumors and also had inhibitory effects on various types of tumors, including Mantle Cell Lymphoma (MCL), colorectal cancer, lung cancer, glioblastoma and Acute Myelogenous Leukemia (AML).
Although CDK4/6 inhibitors in combination with endocrine therapy could bring significant clinical benefit to patients with certain types of breast cancer, the older fraction of patients are treatment ineffective (10-20%), and 70-80% of tumor patients develop drug resistance after 12-36 months of treatment (tripath et al, lancet Oncol,2018, 19 (7), 904-915). There are a number of factors for CDK4/6 treatment resistance, preclinical studies have shown that these may be associated with CDK4/6 treatment resistance, including Rb loss, CCND1 overexpression, p16 amplification, excessive activation of the CCNE1-CDK2 complex (e.g., high expression of CCNE1 in breast and ovarian Cancer cell line models), bypass activation of CDK2, increased CDK4/6 activity, over-expression of CDK7, etc. (Taylor harboring et al, oncostarget, 2014,6 (2), 696-714; herrera-Abreu et al, cancer Res,2016, 76 (8), 2301-2313).
The division of breast cancer patients into different subgroups by biomarker methods helps not only to distinguish the subgroup of patients who most benefit from Palbociclib, but also to elucidate the mechanisms that may lead to CDK4/6 combination therapy resistance. Recent phase III clinical studies with Palbociclib have found that CCNE1 (cyclin E1) is closely related to therapeutic resistance (Turner et al Journal of Clinical Oncology,2019, 37 (14), 1-11). The Palbociclib has remarkable curative effect in the subgroup of people with low expression of CCNE1 mRNA. Meanwhile, the expression levels of CDK4, CDK6, cyclin D1 and RB1 are not obviously related to the treatment effect of Palbociclib in other variable analyses in clinical treatment; this study has attracted great interest and has also demonstrated that CCNE1 is a drug resistant biomarker for Palbociclib.
Cyclin E (Cyclin E) is a protein encoded by CCNE1, begins to express in the G1 metaphase, gradually degrades and disappears after the expression level reaches a peak in the G1-S phase, and its expression level is regulated by E2F, via ubiquitination proteasome degradation (Mazumder et al, current Cancer Drug Targets,2004,4 (1), 65-75). Cyclin E binds to CDK2 to form a complex that phosphorylates downstream substrates Rb, CDC6, NPAT, P107, etc. to bring the cell into the S phase of DNA synthesis; cyclin E plays an important role in maintaining chromosome stability and regulating and controlling cell spindle and centrosome cycle; cyclin E is highly expressed in a variety of malignant tumors, and CCNE1 gene amplification or overexpression is also associated with poor prognosis of a variety of tumors, such as endometrial, gastric, ovarian cancers (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), and the like.
CDK2 plays a key role in cell cycle regulation by interacting with chaperones and phosphorylating activation, and is involved in a range of biological processes such as DNA damage, intracellular transport, protein degradation, signal transduction, DNA and RNA metabolism and translation, etc. (tadess et al Drug Discovery Today,2020, 25 (2), 406-413). CDK2 is in a low expression state in most normal tissues (mccurry et al Oncogene, 2016). Cells in the dividing phase, CDK2, are key cell cycle regulators, activated from late G1 phase, and continue to express the entire S phase. CDK2 binds to CCNE1 or E2 and Cyclin A2, is activated by Cyclin complex (CDK 7, MAT1, cyclin H) phosphorylation, and is also down-regulated by CDC25A dephosphorylation. In the late G1 phase, CDK2-cyclin E and CDK4/6-cyclin-D co-phosphorylate Rb, releasing Rb into E2F, initiating transcription of the cell cycle regulatory genes. In addition to Rb, CDK2 may regulate the phosphorylation of other proteins, correlating to the cell cycle. For example, phosphorylation of Smad3 by CDK2-cyclin-E limits its transcriptional activity, ultimately slowing cell cycle progression (Matsura et al, nature,2004, 430 (6996), 226-231). CDK2 also phosphorylates replication pre-complex proteins, which are necessary to initiate DNA synthesis. Such as CDC6, which is an essential protein that loads a Micro Chromosome Maintenance (MCM) protein onto DNA and MCM helicase protein and initiates DNA replication (Chuang et al, molecular Cell,2009, 35 (2), 206-216). CDK2 also plays an important role in regulating centrosome replication, releasing centrosomes by targeting phosphorylated centrosome proteins such as nuclear phospholipids (Npm) and cp110, and then maintaining centrosome replication (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 most protein kinases, folding into a "biplate" shape. The smaller N-terminal region is composed primarily of beta-sheets, comprising 5 beta-long chains of antiparallel structure and one C-helix. The C-alpha helix contains the sequence PSTAIRE, which is necessary for cyclin binding. The larger C-terminal region, which is rich in the alpha-helix, contains an activating fragment (also known as the T-loop (residues 145 (aspartic acid) -172 (glutamic acid)) and a Ser/Thr (phosphoryl receptor) region that activates the phosphorylation site Thr160.T-loop to bind to the substrate, and is phosphorylated for cell cycle control.
CDK2 activation and inhibition is regulated by several mechanisms: in the absence of mitogen signal, CDK2 is in an inactive state. In the late G1 phase CDK2 activity begins to increase due to, first, E2F-mediated transcription of the CCNE gene, the protein product of which binds to and activates CDK2; complete activation of CDK2-cyclin E or A complexes requires CAK phosphorylation of the Thr160 site; furthermore, wee1 and Myt1 kinases inhibit phosphorylation of the Thr14 and Tyr15 sites, respectively, and dephosphorylation of these residues by the CDC25 family of protein phosphatases may reactivate CDK2; in addition, CDK inhibitor protein families Cip and Kip bind CDK2 to inactivate it, and cyclin E and A can be degraded by ubiquitin ligase mediated ubiquitination (Solomon et al Journal of Medicinal Chemistry, 2018).
CDK2 may also bind cyclin a, participate in progression through S phase, and regulate DNA damage repair. After DNA damage, the DNA Damage Response (DDR) blocks cells at the G1/S junction to repair damaged DNA and maintain genomic fidelity of daughter cells. Two mechanisms have been found, both by inhibiting CDK2 protein phosphorylation, to maintain cells at the G1/S DNA damage checkpoint and to prevent cell proliferation. First, accumulation of p53 results in up-regulation of p21Cip1/Waf1 transcription, followed by cell cycle arrest by inhibition of cyclin-D1-CDK4/6 and cyclin E-CDK2 (Shieh et al, genes & Development,2000, 14 (3), 289-300). The second mechanism is targeting CDC25A for degradation, WEE1 prevents cells from entering S-phase by continually inhibiting phosphorylation at Thr14 and Tyr15 positions of CDK2 proteins (Mailand et al Science,2000, 288 (5470), 1425-1429). FOXO1, a target protein of CDK2, plays an important role in triggering DNA damage by dsDNA cleavage, thereby inducing apoptosis. When DNA damage induces G1/S phase arrest, CDK2 no longer phosphorylates FOXO1, allowing FOXO1 to exert transcriptional activity, which promotes the apoptotic process by up-regulating the expression of various pro-apoptotic proteins (FasL, TRAIL and Bim) (Huang et al, science,2006, 314 (5797), 294-297; huang et al, cell Cycle,2007,6 (8), 902-906).
Taken together, although CDK4/6 inhibitors have become a first line treatment regimen for certain types of breast cancer, some types of breast cancer are naturally insensitive to CDK4/6 inhibitors, such as Triple Negative Breast Cancer (TNBC). In breast cancer of ER+Her2-, CDK4/6 resistance was also found. Cyclin E is highly expressed in a variety of malignancies, including tumors insensitive to CDK4/6 inhibitors (TNBC), ovarian cancers, and the like; the complex formed by CDK2 and cyclin E plays an important role in CDK4/6 resistance; targeting CDK2 can delay the therapeutic resistance of CDK4/6 inhibitor molecules, further exerting therapeutic effects on drug-resistant patients; in contrast, no approved drugs were seen in compounds targeting CDK2/4/6 to overcome CDK4/6 resistance, and only PF-06873600 of Pfizer entered clinical phase II studies. Therefore, development of CDK2/4/6 inhibitors with high selectivity and low drug toxicity to overcome CDK4/6 resistance is an urgent problem to be solved.
The data from the patent to pyro PF-06873600 (US 2018/0044344 A1) show that CCNE1 has a higher frequency of expansion in ovarian cancer OVCAR3 and breast cancer HCC1806 in cell lines of different tumor types. Therefore, a series of small molecule CDK2/4/6 inhibitors with drug-like properties were synthesized. The effect of CDK2 inhibition was assessed by detecting the level of inhibition on OVCAR3 and HCC1806 cells; the effect of CDK4/6 inhibition was assessed by measuring the level of inhibition of MCF-7 cells.
Disclosure of Invention
The present inventors have conducted intensive studies and devised a series of pyrimido-pyridines which exhibit Cyclin Dependent Kinase (CDK) activity inhibition and which can be developed as medicaments for the treatment and/or prophylaxis of diseases associated with CDK activity, such as cancer.
It is therefore an object of the present invention to provide a compound of the general formula (I) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer or mixture thereof, or a pharmaceutically acceptable salt thereof,
Figure PCTCN2022119618-APPB-000001
wherein,
ring a is selected from heterocyclyl optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, hydroxy, mercapto, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl;
ring B is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
R 1 selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally further substituted with one or more groups selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl;
R 2 Selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally further substituted with one or more groups selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl; or,
R 1 and R is 2 Together with the nitrogen and sulfur atoms to which they are attached, form a heterocyclic or heteroaryl group, which is optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, oxo, hydroxy, mercapto, carboxyl, ester, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclic, aryl and heteroaryl;
each R 3 Each independently selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, said alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl optionally further being selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl One or more groups of heteroaryl groups are substituted; or,
any two R 3 Together with the atoms to which they are attached, form cycloalkyl, heterocyclyl, aryl, and heteroaryl, which cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, oxo, hydroxy, mercapto, carboxyl, ester, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
R 4 selected from hydrogen, halogen, amino, nitro, cyano, hydroxy, mercapto, alkyl, alkoxy, 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 is optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl;
R 5 selected from hydrogen, halogen, amino, nitro, cyano, hydroxy, 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 is optionally further substituted with a member selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloSubstituted with one or more groups of alkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl;
R a and R is b Each independently selected from the group consisting of hydrogen, halogen, hydroxy, cyano, amino, carboxyl, ester, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein each independently is optionally further substituted with one or more groups selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
or R is a And R is b Together with the atoms to which they are attached, form a cycloalkyl or heterocyclyl group, which cycloalkyl or heterocyclyl group is optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, oxo, hydroxy, mercapto, carboxyl, ester, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
m is 0, 1, 2;
p is 0, 1, 2, 3 or 4.
In one embodiment of the present invention, the compounds of formula (I) according to the present invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof,
wherein,
ring a is selected from 3 to 12 membered monocyclic heterocyclyl, spiroheterocyclyl, fused heterocyclyl or bridged heterocyclyl, preferably 5 to 7 membered monocyclic heterocyclyl, 7 to 10 membered spiroheterocyclyl, 7 to 10 membered fused heterocyclyl and 7 to 10 membered bridged heterocyclyl, more preferably pyrrolidinyl, piperidinyl, piperazinyl, said heterocyclyl optionally being further selected from halogen, amino, nitro, cyano, hydroxy, mercapto, oxo, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 1 -C 6 HaloalkanesRadical, C 1 -C 6 Haloalkoxy, C 3 -C 6 Cycloalkyl, 5-to 7-membered heterocyclyl, C 6 -C 10 Aryl, 5 to 10 membered heteroaryl.
In another embodiment of the present invention, the compound of formula (I) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, is a compound of formula (II) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof,
Figure PCTCN2022119618-APPB-000002
Wherein the ring B, R 1 、R 2 、R 3 、R 4 、R 5 P is defined as formula (I).
In another embodiment of the present invention, the compounds of formula (I) or formula (II) according to the present invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof,
wherein,
ring B is selected from C 3 -C 7 Cycloalkyl, 4-to 7-membered heterocyclyl, C 6 -C 10 Aryl, 5-to 10-membered heteroaryl, more preferably
Figure PCTCN2022119618-APPB-000003
In another embodiment of the present invention, the compound of formula (I) according to the present invention or a stereoisomer, tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, is a compound of formula (III) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof,
Figure PCTCN2022119618-APPB-000004
wherein,
wherein R is 1 、R 2 、R 3 、R 4 、R 5 P is defined as formula (I).
In another embodiment of the present invention, the compounds of formula (I), formula (II) or formula (III) according to the present invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or prodrugs thereof, or pharmaceutically acceptable salts thereof,
Wherein,
each R 3 Each independently selected from halogen, amino, cyano, hydroxy, mercapto, and C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 3 -C 7 Cycloalkyl, 4-to 7-membered heterocyclyl, said C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 3 -C 7 Cycloalkyl, 4-to 7-membered heterocyclyl optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl; preferably halogen, hydroxy, C 1 -C 6 Alkyl, C 1 -C 6 A haloalkyl group;
p is 0, 1, 2 or 3; preferably 1 or 2.
In another embodiment of the present invention, the compounds of formula (I), formula (II), or formula (III) according to the present invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof,
wherein,
R 1 selected from 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 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 is 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl or C 3 -C 6 Cycloalkyl is optionally further substituted with a member selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, and 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 heterocyclyl, C 1 -C 6 Haloalkyl, C 1 -C 6 One or more groups of the alkoxy group are substituted;
the 5-to 7-membered heterocyclic group, C 6 -C 10 Aryl and 5-to 10-membered heteroaryl are optionally further substituted with one or more substituents 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 6 Cycloalkyl, 5-to 7-membered heterocyclyl, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl, one or more groups of 5-to 10-membered heteroaryl.
In another embodiment of the present invention, the compounds of formula (I), formula (II) or formula (III) according to the present invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof,
wherein,
R 2 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 An alkyl group;
the C is 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl or C 3 -C 6 Cycloalkyl is optionally further substituted with a member selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, and 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 heterocyclyl, C 1 -C 6 Haloalkyl, C 1 -C 6 One or more groups of the alkoxy group are substituted;
the 5-to 7-membered heterocyclic group, C 6 -C 10 Aryl and 5-to 10-membered heteroaryl are optionally further substituted with one or more substituents 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 6 Cycloalkyl, 5-to 7-membered heterocyclyl, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl, one or more groups of 5-to 10-membered heteroaryl.
In another embodiment of the present invention, the compound of formula (I), formula (II) or formula (III) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R 1 And R is 2 Together with the nitrogen and sulfur atoms to which they are attached, form a 5-to 7-membered heterocyclic group, said 5-to 7-membered heterocyclic group optionally being further substituted with a member selected from the group consisting of halogen, amino, nitro, cyano, oxo, hydroxy, mercapto, carboxyl, ester, 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 heterocyclyl, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl, one or more groups of 5-to 10-membered heteroaryl.
In another embodiment of the present invention, the compounds of formula (I), formula (II) or formula (III) according to the present invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof,
wherein,
R 4 selected from hydrogen, halogen, amino, nitro, cyano, hydroxy, 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 groups; preferably R 4 Is hydrogen or C 1 -C 6 An alkyl group.
In another embodiment of the present invention, the compounds of formula (I), formula (II) or formula (III) according to the present invention or stereoisomers, tautomers, meso, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof,
wherein,
R 5 selected from hydrogen, halogen, amino, nitro, cyano, hydroxy, mercapto, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 1 -C 6 Haloalkyl, C 1 -C 6 Haloalkoxy, -C (O) R a 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 Selected from C 1 -C 6 An alkyl group.
Typical compounds of the invention include, but are not limited to, the following:
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
or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a prodrug thereof, or a pharmaceutically acceptable salt thereof.
The present invention further provides a process for the preparation of a compound of formula (I) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a prodrug thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
Figure PCTCN2022119618-APPB-000015
in the presence of alkali and in the presence of a catalyst, carrying out dehydration reaction on a compound Ij and a compound Ia to obtain a compound of a general formula (I); the base is preferably triethylamine; the catalyst is preferably triphenyl phosphorus dichloride;
ring a, ring B, R 1 、R 2 、R 3 、R 4 、R 5 P is defined as formula (I).
The present invention further provides a process for the preparation of a compound of formula (II) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a prodrug thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
Figure PCTCN2022119618-APPB-000016
In the presence of alkali and in the presence of a catalyst, carrying out dehydration reaction on a compound IIj and a compound Ia to obtain a compound of a general formula (II); the base is preferably triethylamine; the catalyst is preferably triphenyl phosphorus dichloride;
ring B, R 1 、R 2 、R 3 、R 4 、R 5 P is defined as formula (II).
The present invention further provides a process for preparing a compound of formula (III) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a prodrug thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
Figure PCTCN2022119618-APPB-000017
in the presence of a base and in the presence of a catalyst, carrying out dehydration reaction on a compound IIIj and a compound Ia to obtain a compound of a general formula (III); the base is preferably triethylamine; the catalyst is preferably triphenyl phosphorus dichloride;
R 1 、R 2 、R 3 、R 4 、R 5 p is defined as formula (III).
The invention also provides a pharmaceutical composition comprising a compound of formula (I), formula (II), or formula (III) according to the invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
The invention also aims to provide the use of a compound of formula (I), formula (II) or formula (III) according to the invention or a stereoisomer, tautomer, meso, enantiomer, diastereoisomer or mixture thereof, or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same, for the preparation of a Cyclin Dependent Kinase (CDK) inhibitor.
The invention also aims to provide the application of the compound shown in the general formula (I), the general formula (II) or the general formula (III) or stereoisomers, tautomers, meso forms, racemates, enantiomers, diastereomers or mixtures thereof or pharmaceutically acceptable salts thereof or a pharmaceutical composition comprising the compound in the preparation of medicines for inhibiting cancer cell proliferation, inhibiting cancer cell invasion or inducing cancer cell apoptosis.
The present invention also aims to provide the use of a compound of formula (I), formula (II) or formula (III) according to the invention or a stereoisomer, tautomer, meso, enantiomer, diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for the manufacture of a medicament for the prevention and/or treatment of a disease associated with cyclin-dependent kinase activity, such as cancer, in particular cancer characterized by the amplification or overexpression of cyclin-dependent kinase CDK 2/cyclin E1 (CCNE 1), CDK 6/cyclin D1, CDK 4/cyclin D3, more particularly breast cancer such as hr+/HER 2-metastatic breast cancer or ovarian cancer.
The invention also relates to a compound of formula (I), formula (II) or formula (III) according to the invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for use as a CDK inhibitor.
The invention also relates to a compound shown in the general formula (I), the general formula (II) or the general formula (III) or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer or a mixture thereof or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the compound, which is used for inhibiting proliferation of cancer cells, inhibiting invasion of cancer cells or inducing apoptosis of the cancer cells.
The invention also relates to a compound of general formula (I), general formula (II) or general formula (III) according to the invention or a stereoisomer, tautomer, mesomer, racemate, enantiomer, diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same, for use as a medicament for the prevention and/or treatment of a disease associated with cyclin-dependent kinase activity, such as cancer, in particular cancer characterized by amplification or overexpression of cyclin-dependent kinase CDK 2/cyclin E1 (CCNE 1), CDK 6/cyclin D1, CDK 4/cyclin D3, more particularly breast cancer such as hr+/HER 2-metastatic breast cancer or ovarian cancer.
The present invention also relates to a method of inhibiting CDKs comprising administering to a subject in need thereof an effective amount of a compound of formula (I), formula (II) or formula (III) according to the present invention or a stereoisomer, tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
The present invention also relates to a method of inhibiting proliferation of cancer cells, inhibiting invasion of cancer cells or inducing apoptosis of cancer cells, comprising administering to a subject in need thereof an effective amount of a compound of formula (I), formula (II) or formula (III) according to the present invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same.
The present invention also relates to a method for preventing and/or treating a disease associated with cyclin-dependent kinase activity, e.g. a cancer, in particular a cancer characterised by amplification or overexpression of cyclin-dependent kinase CDK 2/cyclin E1 (CCNE 1), CDK 6/cyclin D1, CDK 4/cyclin D3, more particularly a breast cancer such as hr+/HER 2-metastatic breast cancer or ovarian cancer, or a mixture thereof, in the form of a compound of general formula (I), general formula (II) or general formula (III) according to the invention or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, to a subject in need thereof.
The compound represented by the general formula (I), the general formula (II) or the general formula (III) according to the present invention or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same may be administered simultaneously, separately or sequentially with another anticancer therapeutic agent or anticancer therapeutic method.
Detailed description of the invention
Unless stated to the contrary, the terms used in the specification and claims have the following meanings.
"alkyl" refers to a saturated monovalent aliphatic hydrocarbon radical comprising straight and branched chain groups having the indicated number of carbon atoms. Alkyl groups generally contain 1 to 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). Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl 5-methylhexyl, 2, 3-dimethylpentyl, 2, 4-dimethylpentyl, 2-dimethylpentyl, 3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2, 3-dimethylhexyl, 2, 4-dimethylhexyl, 2, 5-dimethylhexyl, 2-dimethylhexyl, 3-dimethylhexyl 4, 4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2-diethylpentyl, n-decyl, 3-diethylhexyl, 2-diethylhexyl, and each of them Branched isomers, and the like. Alkyl groups may be substituted or unsubstituted, and when substituted, the substituents may be substituted at any useful point of attachment. Optional substituents for alkyl groups include, but are not limited to, C 3 -C 8 Cycloalkyl, 3-12 membered heterocyclyl, C 6 -C 12 Aryl and 5-12 membered heteroaryl, halo, =o (oxo), =s (thio), =n-CN, and wherein each of said C 3 -C 8 Cycloalkyl, 3-12 membered heterocyclyl, C 6 -C 12 Aryl and 5-12 membered heteroaryl are optionally substituted. Typical substituents on alkyl groups include halogen, -OH, C 1 -C 6 Alkoxy, -O-C 6 -C 12 Aryl, -CN, = O, C 3 -C 8 Cycloalkyl, C 6 -C 12 Aryl, 5-12 membered heteroaryl, and 3-12 membered heterocyclyl; wherein each of said C 3 -C 8 Cycloalkyl, C 6 -C 12 Aryl, 5-12 membered heteroaryl, and 3-12 membered heterocyclyl are optionally substituted with 1-3 groups independently selected from halogen, -OH, = O, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 1 -C 6 Haloalkyl, C 1 -C 6 Hydroxyalkyl, C 1 -C 6 alkoxy-C 1 -C 6 Alkyl, -CN, -NH 2 、-NH(C 1 -C 6 Alkyl) and-N (C) 1 -C 6 Alkyl group 2 Is substituted by a substituent of (2). In some embodiments, the alkyl group is optionally substituted with one or more substituents, preferably 1-3 substituents, independently selected from halogen, -OH, C 1 -C 6 Alkoxy, -O-C 6 -C 12 Aryl, -CN, = O, C 3 -C 8 Cycloalkyl, C 6 -C 12 Aryl, 5-12 membered heteroaryl, and 3-12 membered heterocyclyl. In other embodiments, the alkyl group is optionally substituted with one or more substituents, preferably 1-3 substituents,the substituents are independently selected from halogen, -OH, C 1 -C 6 Alkoxy, -CN, C 3 -C 8 Cycloalkyl, 3-12 membered heterocyclyl, C 6 -C 12 Aryl and 5-12 membered heteroaryl, each optionally containing 1, 2 or 3 additional heteroatoms selected from O, N and S (O) x (where x is 0-2); and wherein each of said cycloalkyl, heterocyclyl, aryl or heteroaryl is optionally substituted with 1-3 groups independently selected from halogen, -OH, = O, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 1 -C 6 Haloalkyl, C 1 -C 6 Hydroxyalkyl, C 1 -C 6 alkoxy-C 1 -C 6 Alkyl, -CN, -NH 2 、-NH(C 1 -C 6 Alkyl) and-N (C) 1 -C 6 Alkyl group 2 Is substituted by a substituent of (2).
The optionally substituted alkyl groups described herein may be substituted with one or more substituents, which are independently selected unless otherwise indicated. To the extent that such substitution is chemically significant, the total number of substituents may be equal to the total number of hydrogen atoms on the alkyl group. The optionally substituted alkyl group generally 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. In particular, unless otherwise indicated, an alkyl group may be substituted with one or more (up to the total number of hydrogen atoms present on the alkyl group) halogen groups. Thus C 1 -C 4 Alkyl includes haloalkyl, especially fluoroalkyl having 1 to 4 carbon atoms, such as trifluoromethyl or difluoroethyl (i.e., CF) 3 and-CH 2 CHF 2 )。
The term "alkenyl" refers to an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon double bond, such as vinyl, 1-propenyl, 2-propenyl, 1-, 2-, or 3-butenyl, and the like. Alkenyl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio.
The term "alkynyl" refers to an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon triple bond, such as ethynyl, propynyl, butynyl, and the like. Alkynyl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio.
The term "cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring containing from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably from 3 to 6 carbon atoms. Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like; polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups.
The term "spirocycloalkyl" refers to a polycyclic group sharing one carbon atom (referred to as a spiro atom) between 5-to 20-membered monocyclic rings, which may contain one or more double bonds, but no ring has a fully conjugated pi-electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The spirocycloalkyl group is classified into a single spirocycloalkyl group, a double spirocycloalkyl group or a multiple spirocycloalkyl group according to the number of common spiro atoms between rings, and preferably a single spirocycloalkyl group and a double spirocycloalkyl group. More preferably 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered monocyclocycloalkyl. Non-limiting examples of spirocycloalkyl groups include:
Figure PCTCN2022119618-APPB-000018
the term "fused ring alkyl" refers to a 5 to 20 membered, all carbon polycyclic group wherein each ring in the system shares an adjacent pair of carbon atoms with the other rings in the system, wherein one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused ring alkyl group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicycloalkyl group. Non-limiting examples of fused ring alkyl groups include:
Figure PCTCN2022119618-APPB-000019
The term "bridged cycloalkyl" refers to an all-carbon polycyclic group of 5 to 20 members, any two rings sharing two carbon atoms not directly attached, which may contain one or more double bonds, but no ring has a fully conjugated pi-electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. Cycloalkyl groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of bridged cycloalkyl groups include:
Figure PCTCN2022119618-APPB-000020
the cycloalkyl ring may be fused to an aryl, heteroaryl, or heterocycloalkyl ring, where the ring attached to the parent structure is cycloalkyl, non-limiting examples include indanyl, tetrahydronaphthyl, benzocycloheptyl, and the like. Cycloalkyl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxyl, or carboxylate groups.
The term "heterocyclyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms in which one or more ring atoms are selected from nitrogen, oxygen or S (O) m (wherein m is an integer from 0 to 2), but does not include a ring moiety of-O-O-, -O-S-, or-S-S-, and the remaining ring atoms are carbon. Preferably containing 3 to 12 ring atoms, of which 1 to 4 are heteroatoms; most preferably from 3 to 8 ring atoms, of which 1 to 3 are heteroatoms; most preferably from 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, tetrahydrothienyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrrolyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl, and the like, preferably 1, 2, 5-oxadiazolyl, pyranyl, or morpholinyl. Polycyclic heterocyclyl groups include spiro, fused and bridged heterocyclic groups.
The term "spiroheterocyclyl" refers to a polycyclic heterocyclic group having a single ring of 5 to 20 members sharing one atom (referred to as the spiro atom) between them, wherein one or more of the ring atoms is selected from nitrogen, oxygen or S (O) m (wherein m is an integer from 0 to 2) and the remaining ring atoms are carbon. Which may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The spiroheterocyclyl groups are classified into a single spiroheterocyclyl group, a double spiroheterocyclyl group or a multiple spiroheterocyclyl group according to the number of common spiro atoms between rings, and preferably a single spiroheterocyclyl group and a double spiroheterocyclyl group. More preferably a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered single spiro heterocyclic group. Non-limiting examples of spiroheterocyclyl groups include:
Figure PCTCN2022119618-APPB-000021
The term "fused heterocyclyl" refers to 5 to 20 membered ring, in the systemOne or more rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron system in which one or more ring atoms are selected from nitrogen, oxygen or S (O) m (wherein m is an integer from 0 to 2) and the remaining ring atoms are carbon. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic group. Non-limiting examples of fused heterocyclyl groups include:
Figure PCTCN2022119618-APPB-000022
the term "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 fully conjugated pi electron system in which one or more ring atoms are selected from nitrogen, oxygen, or S (O) m (wherein m is an integer from 0 to 2) and the remaining ring atoms are carbon. Preferably 6 to 14 membered, more preferably 7 to 10 membered. Heterocyclic groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of bridged heterocyclyl groups include:
Figure PCTCN2022119618-APPB-000023
The heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring attached to the parent structure is heterocyclyl, non-limiting examples of which include:
Figure PCTCN2022119618-APPB-000024
etc.
The heterocyclic group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxyl, or carboxylate groups.
The term "aryl" refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic (i.e., rings sharing adjacent pairs of carbon atoms) group having a conjugated pi-electron system, preferably 6 to 10 membered, such as phenyl and naphthyl. More preferably phenyl. The aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring attached to the parent structure is an aryl ring, non-limiting examples of which include:
Figure PCTCN2022119618-APPB-000025
aryl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
The term "heteroaryl" refers to a heteroaromatic system containing from 1 to 4 heteroatoms, from 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen. Heteroaryl groups are preferably 5 to 10 membered, containing 1 to 3 heteroatoms; more preferably 5 or 6 membered, containing 1 to 2 heteroatoms; preferably, for example, imidazolyl, furyl, thienyl, thiazolyl, pyrazolyl, oxazolyl, pyrrolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl, and the like, preferably imidazolyl, thiazolyl, pyrazolyl or pyrimidinyl, thiazolyl; more preferably pyrazolyl or thiazolyl. The heteroaryl ring may be fused to an aryl, heterocyclyl, or cycloalkyl ring, wherein the ring attached to the parent structure is a heteroaryl ring, non-limiting examples of which include:
Figure PCTCN2022119618-APPB-000026
heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
The term "alkoxy" refers to-O- (alkyl) and-O- (cycloalkyl), wherein alkyl and cycloalkyl are as defined above. Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentoxy, cyclohexyloxy. The alkoxy groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
The term "haloalkyl" refers to an alkyl group substituted with one or more halogens, wherein alkyl is as defined above.
The term "haloalkoxy" refers to an alkoxy group substituted with one or more halogens, wherein the alkoxy group is as defined above.
The term "hydroxy" refers to an-OH group.
The term "halogen" refers to fluorine, chlorine, bromine or iodine.
The term "amino" refers to-NH 2
The term "cyano" refers to-CN.
The term "nitro" refers to-NO 2
The term "oxo" refers to = O.
The term "carboxy" refers to-C (O) OH.
The term "mercapto" refers to-SH.
The term "ester group" refers to a-C (O) O (alkyl) or-C (O) O (cycloalkyl), wherein alkyl and cycloalkyl are as defined above.
The term "acyl" refers to compounds containing a-C (O) R group, wherein R is alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl.
"optional" or "optionally" means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs or does not. For example, "a heterocyclic group optionally substituted with an alkyl group" means that an alkyl group may be, but is not necessarily, present, and the description includes cases where the heterocyclic group is substituted with an alkyl group and cases where the heterocyclic group is not substituted with an alkyl group.
"substituted" means that one or more hydrogen atoms, preferably up to 5, more preferably 1 to 3 hydrogen atoms in the group are independently substituted with a corresponding number of substituents. It goes without saying that substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable when bound to carbon atoms having unsaturated (e.g., olefinic) bonds.
"pharmaceutical composition" means a mixture comprising one or more of the compounds described herein or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to promote the administration to organisms, facilitate the absorption of active ingredients and thus exert biological activity.
By "pharmaceutically acceptable salts" is meant salts of the compounds of the present invention which are safe and effective when used in a mammal, and which possess the desired biological activity.
"cancer" refers to any malignant and/or invasive growth or tumor (caused by abnormal cell growth). Cancers include solid tumors named after the cell type from which they are formed, cancers of the blood, bone marrow or lymphatic system. Examples of solid tumors include sarcomas and carcinomas. Hematological cancers include, but are not limited to, leukemia, lymphoma, and myeloma. Cancers also include primary cancers that originate at specific parts of the body, metastatic cancers that have spread from the parts from which they began to other parts of the body, recurrence from the original primary cancer after remission, and secondary primary cancers (which are new primary cancers in humans having a history of previous cancers of a different type than the new primary cancers). In some embodiments provided herein, 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. In some such embodiments, the cancer is characterized by amplification or overexpression of CCNE1 and/or CCNE 2.
Stereoisomers as described herein may include cis and trans isomers, optical isomers such as (R) and (S) enantiomers, diastereomers, geometric isomers, rotamers, atropisomers, conformational isomers and tautomers of the compounds of the invention (including compounds that exhibit more than one type of isomerism); and mixtures thereof (e.g., racemates and diastereomeric pairs).
The compounds of the present invention may exhibit tautomerism and structural isomerism. For example, the compounds may exist in several tautomeric forms, including the enolic and imine forms as well as the ketone and enamine forms, and geometric isomers and mixtures thereof. All such tautomeric forms are included within the scope of the compounds of the invention. Tautomers exist as a mixture of tautomeric groups in solution. In solid form, one tautomer is usually dominant. Even though one tautomer may be described, the present invention includes all tautomers of the provided compounds.
Conventional techniques for preparing/separating individual enantiomers include chiral synthesis from suitable optically pure precursors or resolution of the racemate (or of a salt or derivative) using, for example, chiral High Pressure Liquid Chromatography (HPLC) or Supercritical Fluid Chromatography (SFC).
The enantiomeric purity of the compounds described herein can be described in terms of enantiomeric excess (ee), which refers to the degree to which a sample contains one enantiomer in a greater amount than the other. The ee of the racemic mixture was 0% whereas the ee of a single, fully pure enantiomer was 100%. Similarly, diastereomeric purity can be described in terms of diastereomeric excess (de).
The compounds of formula (I) of the present invention may form pharmaceutically acceptable base addition salts or acid addition salts with bases or acids according to methods conventional in the art to which the present invention pertains. The base includes inorganic bases and organic bases, acceptable organic bases include diethanolamine, ethanolamine, N-methylglucamine, triethanolamine, tromethamine, and the like, and acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like. The acids include inorganic acids and organic acids, and acceptable inorganic acids include hydrochloric acid, sulfuric acid, nitric acid, phosphoric acid, hydrobromic acid, and the like. Acceptable organic acids include acetic acid, trifluoroacetic acid, formic acid, anti-cyclic acid, and the like.
Pharmaceutical compositions containing the active ingredient may be in a form suitable for oral administration, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Oral compositions may be prepared according to any method known in the art for preparing pharmaceutical compositions, and such compositions may contain one or more ingredients selected from the group consisting of: sweeteners, flavoring agents, coloring agents and preservatives to provide a pleasing and palatable pharmaceutical preparation. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be inert excipients, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example 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 they may be coated by known techniques to mask the taste of the drug or delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, water-soluble taste masking substances such as hydroxypropyl methylcellulose or hydroxypropyl cellulose, or extended time substances such as ethylcellulose, cellulose acetate butyrate may be used.
Oral formulations may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with a water-soluble carrier, for example polyethylene glycol or an oil vehicle, for example peanut oil, liquid paraffin or olive oil.
Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethyl cellulose, methyl cellulose, hydroxypropyl methyl cellulose, sodium alginate, polyvinylpyrrolidone and acacia; the dispersing or wetting agent may be a naturally occurring phospholipid such as lecithin, or a condensation product of an alkylene oxide with a fatty acid, such as polyoxyethylene stearate, or a condensation product of ethylene oxide with a long chain fatty alcohol, such as heptadecaethyleneoxycetyl alcohol (heptadecaethyleneoxy cetanol), or a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol, such as polyethylene oxide sorbitol monooleate, or a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride, such as polyethylene oxide sorbitan monooleate. The aqueous suspension may also contain one or more preservatives such as ethyl or Jin Zhengbing esters of nipagin, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose, saccharin or aspartame.
Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oil suspension may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. The above-described sweeteners and flavoring agents 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 use in the preparation of an aqueous suspension by the addition of water provide the active ingredient in combination with a dispersing or wetting agent, suspending agent or one or more preservatives. Suitable dispersing or wetting agents and suspending agents are as described above. Other excipients, for example sweetening, flavoring and coloring agents, may also be added. These compositions are preserved by the addition of an antioxidant such as ascorbic acid.
The pharmaceutical compositions of the present invention may also be in the form of an oil-in-water emulsion. The oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures thereof. Suitable emulsifiers may be naturally occurring phospholipids, such as soy lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of the partial esters and ethylene oxide, such as polyethylene oxide sorbitol 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, a colorant and an antioxidant.
The pharmaceutical compositions of the present invention may be in the form of sterile injectable aqueous solutions. Acceptable vehicles and solvents that may be used 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 an oil phase. For example, the active ingredient is dissolved in a mixture of soybean oil and lecithin. The oil solution is then treated to form a microemulsion by adding it to a mixture of water and glycerol. The injection or microemulsion may be injected into the patient's blood stream by local bolus injection. Alternatively, it may be desirable to administer the solutions and microemulsions in a manner that maintains a constant circulating concentration of the compounds of the present invention. To maintain this constant concentration, a continuous intravenous delivery device may be used.
The pharmaceutical compositions of the present invention may be in the form of sterile injectable aqueous or oleaginous suspensions for intramuscular and subcutaneous administration. The suspensions may be formulated according to known techniques using those suitable dispersing or wetting agents and suspending agents as described above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any blend stock oil may be used, including synthetic mono-or diglycerides. In addition, fatty acids such as oleic acid may be used in the preparation of injectables.
The compounds of the present 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. Such materials include cocoa butter, glycerogelatin, hydrogenated vegetable oils, polyethylene glycols of various molecular weights and mixtures of fatty acid esters of polyethylene glycols.
It is well known to those skilled in the art that the amount of drug administered depends on a variety of factors, including but not limited to the following: the activity of the particular compound used, the age of the patient, the weight of the patient, the health of the patient, the patient's integument, the patient's diet, the time of administration, the mode of administration, the rate of excretion, the combination of the drugs, etc. In addition, the optimal mode of treatment, such as the mode of treatment, the daily amount of the compound of formula (I) or the type of pharmaceutically acceptable salt, can be verified according to conventional treatment protocols.
The invention can contain the compound shown in the general formula (I) and pharmaceutically acceptable salt, hydrate or solvate thereof as active ingredients, and is mixed with pharmaceutically acceptable carriers or excipients to prepare a composition and a clinically acceptable dosage form. The derivatives of the present invention may be used in combination with other active ingredients as long as they do not exert other adverse effects such as allergic reactions and the like. The compounds of the present invention may be used as the sole active ingredient, or in combination with other drugs for the treatment of diseases associated with tyrosine kinase activity. Combination therapy is achieved by simultaneous, separate or sequential administration of the individual therapeutic components.
Synthesis method of compound of the invention
To accomplish the objects of the present invention, the present invention employs the following synthetic schemes for preparing the compounds of the general formula (I) of the present invention.
The present invention employs scheme 1 below to prepare the compounds of the present invention represented by general formula (I).
Figure PCTCN2022119618-APPB-000027
Scheme 1
Step 1: in a polar aprotic solvent (such as dichloromethane), in the presence of a suitable base (such as triethylamine), the sulfonyl chloride compound reacts to obtain a sulfonamide compound Ia;
step 2: in a proper solvent, 4-chloro-2- (methylthio) pyrimidine-5-carboxylic acid ethyl ester is subjected to reduction reaction in the presence of a proper reducing agent to obtain a compound Ib; the solvent is, for example, tetrahydrofuran, and the reducing agent is, for example, diisobutylaluminum hydride;
step 3: reacting compound Ib with compound Ic in the presence of a suitable base in a suitable solvent to give compound Id; the solvent, such as isopropanol, the base, such as diisopropylethylamine, may be at a reaction temperature between room temperature and 80 ℃;
step 4: in a proper solvent, in the presence of a proper oxidant, carrying out oxidation reaction on the compound Id to obtain a compound Ie; the solvent, such as ethyl acetate, the oxidant, such as manganese dioxide, may be at a reaction temperature between 50 ℃ and 80 ℃;
Step 5: aldol cyclization of compound Ie in the presence of a suitable catalyst in a suitable solvent (see VanderWel et al, j. Med. Chezn.2005,4s, 2371) gives compound If; the solvent, such as tetrahydrofuran, the catalyst, such as LHMDS, may be at a reaction temperature between-20 ℃ and room temperature, the reaction being carried out under nitrogen atmosphere;
step 6: in a proper solvent, in the presence of a proper oxidant, carrying out oxidation reaction on the compound If to obtain a compound Ig; the solvent is especially a mixed solvent, such as a mixed solvent of 2-methyltetrahydrofuran and water, and the oxidant comprises, but is not limited to, potassium hydrogen persulfate, m-chloroperoxybenzoic acid and the like, and the excessive oxidant can help the reaction to thoroughly proceed, and the reaction temperature can be between 10 ℃ and 80 ℃;
step 7: reacting compound Ig with compound Ih in a suitable solvent in the presence of a suitable base to give compound Ii; the solvent, e.g. isopropanol, 2-methyltetrahydrofuran, the base, e.g. diisopropylethylamine, the reaction temperature may be between room temperature and 80 ℃;
step 8: deprotection of compound Ii in the presence of a suitable acid in a suitable solvent gives compound Ij; the solvent, such as dioxane, the acid, such as hydrochloric acid, the reaction temperature typically being at room temperature; further freeing the compound by a base such as sodium hydroxide, potassium hydroxide;
Step 9: in a proper solvent, in the presence of proper alkali and catalyst, the compound Ij and the compound Ia are dehydrated to obtain a compound shown in a general formula (I); the solvent is, for example, methylene chloride, the base is, for example, triethylamine, and the catalyst is, for example, triphenylphosphine dichloride; the reaction is typically carried out under a nitrogen atmosphere; the reaction temperature is typically from 0 ℃ to room temperature;
wherein, ring A, ring B, R 1 、R 2 、R 3 、R 4 、R 5 P is defined as formula (I).
The present invention employs scheme 2 below to prepare compounds of general formula (II) of the present invention.
Figure PCTCN2022119618-APPB-000028
Scheme 2
Step 1: in a polar aprotic solvent (such as dichloromethane), in the presence of a suitable base (such as triethylamine), the sulfonyl chloride compound reacts to obtain a sulfonamide compound Ia;
step 2: in a proper solvent, 4-chloro-2- (methylthio) pyrimidine-5-carboxylic acid ethyl ester is subjected to reduction reaction in the presence of a proper reducing agent to obtain a compound Ib; the solvent is, for example, tetrahydrofuran, and the reducing agent is, for example, diisobutylaluminum hydride;
step 3: reacting compound Ib with compound Ic in the presence of a suitable base in a suitable solvent to give compound Id; the solvent, such as isopropanol, the base, such as diisopropylethylamine, may be at a reaction temperature between room temperature and 80 ℃;
Step 4: in a proper solvent, in the presence of a proper oxidant, carrying out oxidation reaction on the compound Id to obtain a compound Ie; the solvent, such as ethyl acetate, the oxidant, such as manganese dioxide, may be at a reaction temperature between 50 ℃ and 80 ℃;
step 5: aldol cyclization of compound Ie in the presence of a suitable catalyst in a suitable solvent (see VanderWel et al, j. Med. Chezn.2005,4s, 2371) gives compound If; the solvent, such as tetrahydrofuran, the catalyst, such as LHMDS, may be at a reaction temperature between-20 ℃ and room temperature, the reaction being carried out under nitrogen atmosphere;
step 6: in a proper solvent, in the presence of a proper oxidant, carrying out oxidation reaction on the compound If to obtain a compound Ig; the solvent is especially a mixed solvent, such as a mixed solvent of 2-methyltetrahydrofuran and water, and the oxidant comprises, but is not limited to, potassium hydrogen persulfate, m-chloroperoxybenzoic acid and the like, and the excessive oxidant can help the reaction to thoroughly proceed, and the reaction temperature can be between 10 ℃ and 80 ℃;
step 7: reacting compound Ig with compound IIh in a suitable solvent in the presence of a suitable base to obtain compound IIi; the solvent, e.g. isopropanol, 2-methyltetrahydrofuran, the base, e.g. diisopropylethylamine, the reaction temperature may be between room temperature and 80 ℃;
Step 8: deprotection of compound IIi in the presence of a suitable acid in a suitable solvent affords compound IIj; the solvent, such as dioxane, the acid, such as hydrochloric acid, the reaction temperature typically being at room temperature; further freeing the compound by a base such as sodium hydroxide, potassium hydroxide;
step 9: in a proper solvent, in the presence of proper alkali and catalyst, the compound IIj and the compound Ia are subjected to dehydration reaction to obtain a compound of a general formula (II); the solvent is, for example, methylene chloride, the base is, for example, triethylamine, and the catalyst is, for example, triphenylphosphine dichloride; the reaction is typically carried out under a nitrogen atmosphere; the reaction temperature is typically from 0 ℃ to room temperature;
wherein the ring B, R 1 、R 2 、R 3 、R 4 、R 5 P is defined as formula (II).
The present invention employs scheme 3 below to prepare compounds of general formula (III) of the present invention.
Figure PCTCN2022119618-APPB-000029
Scheme 3
Step 1: in a polar aprotic solvent (such as dichloromethane), in the presence of a suitable base (such as triethylamine), the sulfonyl chloride compound reacts to obtain a sulfonamide compound Ia;
step 2: in a proper solvent, 4-chloro-2- (methylthio) pyrimidine-5-carboxylic acid ethyl ester is subjected to reduction reaction in the presence of a proper reducing agent to obtain a compound Ib; the solvent is, for example, tetrahydrofuran, and the reducing agent is, for example, diisobutylaluminum hydride;
Step 3: reacting compound Ib with compound IIIc in the presence of a suitable base in a suitable solvent to give compound IIId; the solvent, such as isopropanol, the base, such as diisopropylethylamine, may be at a reaction temperature between room temperature and 80 ℃;
step 4: in a proper solvent, carrying out oxidation reaction on the compound IIId in the presence of a proper oxidant to obtain a compound IIIe; the solvent, such as ethyl acetate, the oxidant, such as manganese dioxide, may be at a reaction temperature between 50 ℃ and 80 ℃;
step 5: aldol cyclization of compound IIIe in the presence of a suitable catalyst in a suitable solvent (see Vanderwel et al, J.Med. Chezn.2005,4S, 2371) gives compound IIIf; the solvent, such as tetrahydrofuran, the catalyst, such as LHMDS, may be at a reaction temperature between-20 ℃ and room temperature, the reaction being carried out under nitrogen atmosphere;
step 6: in a proper solvent, carrying out oxidation reaction on the compound IIIf in the presence of a proper oxidant to obtain a compound IIIg; the solvent is especially a mixed solvent, such as a mixed solvent of 2-methyltetrahydrofuran and water, and the oxidant comprises, but is not limited to, potassium hydrogen persulfate, m-chloroperoxybenzoic acid and the like, and the excessive oxidant can help the reaction to thoroughly proceed, and the reaction temperature can be between 10 ℃ and 80 ℃;
Step 7: reacting compound IIIg with compound IIh in a suitable solvent in the presence of a suitable base to give compound IIIi; the solvent, e.g. isopropanol, 2-methyltetrahydrofuran, the base, e.g. diisopropylethylamine, the reaction temperature may be between room temperature and 80 ℃;
step 8: deprotection of compound IIIi in the presence of a suitable acid in a suitable solvent affords compound IIIj; the solvent, such as dioxane, the acid, such as hydrochloric acid, the reaction temperature typically being at room temperature; further freeing the compound by a base such as sodium hydroxide, potassium hydroxide;
step 9: in a proper solvent, in the presence of proper alkali and catalyst, the compound IIIj and the compound Ia are dehydrated to obtain a compound of a general formula (III); the solvent is, for example, methylene chloride, the base is, for example, triethylamine, and the catalyst is, for example, triphenylphosphine dichloride; the reaction is typically carried out under a nitrogen atmosphere; the reaction temperature is typically from 0 ℃ to room temperature;
wherein R is 1 、R 2 、R 3 、R 4 、R 5 P is defined as formula (III).
Detailed Description
The compounds of the present invention and their preparation are further understood by the examples which illustrate some methods of making or using the compounds. However, it is to be understood that these examples do not limit the scope of the present invention. Variations of the invention now known or further developed are considered to fall within the scope of the invention described and claimed herein.
The compounds of the present invention are prepared using convenient starting materials and general preparation procedures. Typical or preferential reaction conditions are given in the present invention, such as reaction temperature, time, solvent, pressure, molar ratio of reactants. But other reaction conditions can be adopted unless specifically stated. The optimization conditions may vary with the particular reactants or solvents used, but in general, both the reaction optimization steps and conditions can be determined.
In addition, some protecting groups may be used in the present invention to protect certain functional groups from unwanted reactions. Protecting groups suitable for various functional groups and their protecting or deprotecting conditions are well known to those skilled in the art. For example, T.W.Greene and G.M.Wuts in organic preparation of protecting groups (3 rd edition, wiley, new York,1999 and literature citations) describe in detail the protection or deprotection of a large number of protecting groups.
The separation and purification of the compounds and intermediates may be carried out by any suitable method or procedure depending on the particular needs, such as filtration, extraction, distillation, crystallization, column chromatography, thin layer chromatography, high performance liquid chromatography or a combination thereof. The specific methods of use thereof may be found in the examples described herein. Of course, other similar isolation and purification means may be employed. It can be characterized using conventional methods, including physical constants and spectral data.
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) or/and Mass Spectrometry (MS). NMR shift at 10 -6 Units of (ppm) are given. NMR was performed using Bruker dps 300 nuclear magnetic resonance apparatus with deuterated dimethyl sulfoxide (DMSO-d) 6 ) Deuterated chloroform (CDCl) 3 ) Deuterated methanol (CD) 3 OD), internal standard is Tetramethylsilane (TMS).
The MS was determined by LC (Waters 2695)/MS (Quattro Premier xE) mass spectrometer (manufacturer: watt) (Photodiode Array Detector).
The preparation liquid chromatography used an lc6000 high performance liquid chromatograph (manufacturer: innovative). The column was DaisogelC18 μm 100A (30 mm. Times.250 mm), mobile phase: acetonitrile/water.
The thin layer chromatography silica gel plate is used in Qingdao ocean chemical GF254 silica gel plate, the specification of the silica gel plate used in the Thin Layer Chromatography (TLC) is 0.20-0.25 mm, and the specification of the thin layer chromatography separation and purification product is 0.5mm.
Column chromatography generally uses Qingdao ocean silica gel 100-200 mesh, 200-300 mesh and 300-400 mesh silica gel as carriers.
The known starting materials of the present invention may be synthesized using or according to methods known in the art or may be purchased from commercial establishments, beijing couplings, sigma, carbofuran, yi Shiming, shanghai Shuya, shanghai Enoki, an Naiji chemistry, shanghai Pico, and the like.
The examples are not particularly described, and the reactions can all be carried out under nitrogen atmosphere.
An argon or nitrogen atmosphere means that the reactor flask is connected to a balloon of argon or nitrogen of about 1L volume.
The reaction solvent, the organic solvent or the inert solvent are each expressed as a solvent which does not participate in the reaction under the reaction conditions described, and include, for example, benzene, toluene, acetonitrile, tetrahydrofuran (THF), dimethylformamide (DMF), chloroform, methylene chloride, diethyl ether, methanol, nitrogen-methylpyrrolidone (NMP), pyridine, etc. The examples are not specifically described, and the solution refers to an aqueous solution.
The chemical reactions described in the present invention are generally carried out at atmospheric pressure. The reaction temperature is between-78 ℃ and 200 ℃. The reaction time and conditions are, for example, between-78 ℃ and 200 ℃ at one atmosphere, completed in about 1 to 24 hours. If the reaction is overnight, the reaction time is typically 16 hours. The reaction temperature is room temperature and is 20-30 deg.c without specific explanation in the examples.
The progress of the reaction in the examples was monitored by Thin Layer Chromatography (TLC) using the following system of developing agents: a: dichloromethane and methanol system, B: petroleum ether and ethyl acetate system, C: the volume ratio of acetone and solvent is adjusted according to the polarity of the compound.
The eluent system for column chromatography and the developing agent system for thin layer chromatography used for purifying the compound include: a: dichloromethane and methanol system, B: petroleum ether and ethyl acetate system, the volume ratio of the solvent is regulated according to the polarity of the compound, and small amount of alkaline or acidic reagents such as triethylamine and trifluoroacetic acid can be added for regulation.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present invention.
The compounds of the present invention are prepared according to the exemplary procedures provided herein and variations thereof known to those skilled in the art.
Abbreviations
Ac=acetyl group
AcO or oac=acetoxy
Acn=acetonitrile
aq=aqueous solution
Boc=tert-butoxycarbonyl group
Bn=benzyl group
DIPEA = diisopropylethylamine
Dmap=4-dimethylaminopyridine
Dmf=n, N-dimethylformamide
DMSO = dimethylsulfoxide
Dcm=dichloromethane
Ea=ethyl acetate
EDTA = ethylenediamine tetraacetic acid
HPLC = high performance liquid chromatography
IC 50 Concentration of =inhibitory 50% activity
Lhmds=lithium hexamethyldisilazane salt (lithium bis (trimethylsilyl) amide)
LC-ms=liquid chromatography-mass spectrometry combination
M+H + =parent compound mass+proton
Ms=methanesulfonyl group
Ms=mass spectrum
Mscl=methylsulfonyl chloride
mCPBA = m-chloroperoxybenzoic acid
Nbs=n-bromosuccinimide
Ncs=n-chlorosuccinimide
NIS = N-iodosuccinimide
Nmr=nuclear magnetic resonance
Oxone=potassium hydrogen persulfate
Pe=petroleum ether
Pro=protecting group
Tbs=tert-butyldimethylsilyl group
TBHP = tert-butyl hydroperoxide
Tea=triethylamine
TFA = trifluoroacetic acid
THF = tetrahydrofuran
Examples
Example 1: preparation of 2- ((1- (cyclopropanesulfonylimino) piperidin-4-yl) amino) -8- (((1R, 2R) -2-hydroxy-2-methylcyclopentyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (1)
Figure PCTCN2022119618-APPB-000030
Step 1: preparation of 1-methyl-6-oxabicyclo [3.1.0] hexane (1 a)
1-methylcyclopent-1-ene (112 g,1366 mmol) and DCM (4400 mL) were added to the reaction flask at room temperature, and then m-chloroperoxybenzoic acid (496 g,2049mmol,71% wt) was added in portions to the flask and the reaction stirred at room temperature overnight. After the reaction was completed, celite was added for suction filtration, saturated sodium bicarbonate (2500 mL) and 10% sodium thiosulfate (500 mL) were added, and the organic phase was concentrated in a water bath below 20 ℃ to give 125g of the title product as a brown oil, which was used directly in the next step.
LCMS:m/z 99.07[M+H] +
Step 2: preparation of (+ -) - (1R, 2R) -2- (benzylamino) -1-methylcyclopent-1-ol ((±) 1 b)
A glass vial was charged with water (250 mL) and benzylamine (146 g,1366 mmol) at room temperature, then purged with nitrogen for 5min, then 1-methyl-6-oxabicyclo [3.1.0] hexane (125 g, crude) was added. Heating at 100deg.C for 18h, at which point a two-phase mixture is observed. After cooling to room temperature, concentrated aqueous hydrochloric acid (about 12M,150 mL) was added to bring the pH to 1. The organic impurities were extracted with ethyl acetate (1500 mL). 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 two-phase mixture was extracted with ethyl acetate (1200 mL x 3) and the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a brown oil. Concentrated under reduced pressure at 80 ℃ for 5h, cooled to room temperature, dispersed in petroleum ether and filtered to give 85.0g of the title product as a brown solid.
LCMS:m/z 206.15[M+H] +
Step 3: preparation of (1R, 2R) -2- (benzylamino) -1-methylcyclopent-1-ol (1 c)
Into a reaction flask (. + -.) - (1R, 2R) -2- (benzylamino) -1-methylcyclopent-1-ol (85.0 g,414 mmol) and ethanol (600 mL) were added at room temperature and heated at 80℃for 30min. To another reaction flask was added (R) -2- (3, 5-dinitrobenzoylamino) -2-phenylacetic acid (71.4 g,207 mmol) and ethanol (1200 mL), heated at 80deg.C until the solid dissolved, and stirring continued for 30min. The hot solution of alcohol from the first reaction flask was poured into the hot chiral acid solution in the second reaction flask at a steady flow rate. The reaction mixture remained clear for about 1min, after which precipitation began. After 5min, a thick white suspension formed, but stirring was not hindered. Stirring was continued for 4h at 80 ℃, 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 45g of brown solid. The brown solid was suspended in water (250 mL) and ethyl acetate (500 mL) in a 1L separatory funnel. Aqueous hydrochloric acid (4M, 150mL,600 mmol) was added and the mixture was stirred for about 30 seconds. A clear biphasic mixture was obtained. The layers were separated and the organic layer was further washed with aqueous hydrochloric acid (4 m,100ml x 3). The acidic aqueous layers were combined, cooled in an ice-water bath, and aqueous sodium hydroxide (aq) was added to bring the pH to 10. At this pH a white suspension formed. Dilute with saturated aqueous sodium chloride (250 mL) and extract with ethyl acetate (1000 mL x 3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 26.0g of the title product as a brown solid, which was used directly in the next step.
LCMS:m/z 206.15[M+H] +
Step 4: preparation of (1R, 2R) -2-amino-1-methylcyclopent-1-ol (1 d)
20% Palladium on carbon hydroxide (2.6 g) was added to a solution of (1R, 2R) -2-amino-1-methylcyclopent-1-ol (26.0 g,127 mmol) in isopropanol (250 mL) at room temperature and stirred at 50℃under hydrogen for 16h. The catalyst was removed by filtration through celite and the filtrate was concentrated to give 15.0g of the title product as a reddish brown oil, which was used directly in the next step.
LCMS:m/z 116.10[M+H] +
Step 5: preparation of (4-chloro-2- (methylthio) pyrimidin-5-yl) methanol (1 e)
4-chloro-2- (methylthio) pyrimidine-5-carboxylic acid ethyl ester (76.6 g,330 mmol) and tetrahydrofuran (3000 mL) were added to a reaction flask at room temperature, diisobutylaluminum hydride (990 mL,1M tetrahydrofuran solution, 990 mmol) was slowly added dropwise to the reaction flask at-78℃under nitrogen atmosphere, while controlling the temperature not to exceed-60℃and stirring the reaction overnight, while allowing the temperature to rise to room temperature. After completion of the reaction, the reaction was quenched with saturated ammonium chloride solution (1000 mL) at 0 ℃, concentrated aqueous hydrochloric acid (about 12M) was added to bring the pH to 2, the organic phase was collected and washed with saturated brine (1000 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 53.2g of the title product as a pale yellow solid, which was directly used in the next step, yield: 84.8%.
LCMS:m/z 191[M+H] +
Step 6: preparation of (1R, 2R) -2- ((5- (hydroxymethyl) -2- (methylthio) pyrimidin-4-yl) amino) -1-methylcyclopent-1-ol (1 f)
(4-chloro-2- (methylthio) pyrimidin-5-yl) methanol (24.5 g,129 mmol), (1R, 2R) -2-amino-1-methylcyclopent-1-ol (15.0 g,129 mmol), diisopropylethylamine (49.9 g,387 mmol) and isopropyl alcohol (200 mL) were added to a 500mL reaction flask at room temperature, and the reaction was stirred at 80℃overnight. After the reaction was completed, the mixture was concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: dichloromethane: methanol=0-20:1) to give 25.0g of the title product as a white solid, yield: 92.9%.
LCMS:m/z 270.12[M+H] +
Step 7: preparation of 4- (((1R, 2R) -2-hydroxy-2-methylcyclopentyl) amino) -2- (methylthio) pyrimidine-5-carbaldehyde (1 g)
(1R, 2R) -2- ((5- (hydroxymethyl) -2- (methylthio) pyrimidin-4-yl) amino) -1-methylcyclopent-an-1-ol (25.0 g,92.9 mmol), manganese dioxide (161 g,1858 mmol) and ethyl acetate (600 mL) were added to a reaction flask at room temperature, and the reaction was stirred at 50℃overnight. After the reaction was completed, the solid was removed by filtration, and the filtrate was concentrated and purified by column chromatography (eluent: dichloromethane: methanol=0 to 20:1) to give 19.0g of the title product as a white solid, yield: 76.6%.
LCMS:m/z 268.10[M+H] +
Step 8: preparation of 8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -2- (methylthio) pyrido [2,3-d ] pyrimidin-7 (8H) -one (1H)
4- (((1R, 2R) -2-hydroxy-2-methylcyclopentyl) amino) -2- (methylthio) pyrimidine-5-carbaldehyde (13.4 g,50.2 mmol), tetrahydrofuran (250 mL) and ethyl acetate (13.1 mL,126 mmol) were added to a three-necked flask at room temperature, cooled to-5℃in a methanol-ice bath, and lithium hexamethyl-amide (151 mL,1.0M tetrahydrofuran solution, 151 mmol) was slowly added under nitrogen atmosphere. During which time the temperature was allowed to gradually rise to room temperature and overnight. The resulting red solution was cooled to about 3 ℃ in an ice-water bath, then ethanol (88 ml,1506 mmol) was slowly added. The mixture was stirred in an ice bath for 1h, then the cooling bath was removed and the solution allowed to warm to room temperature and stirring continued for 1h. The solvent was removed under reduced pressure, the residue was diluted with water (100 mL) and saturated aqueous sodium chloride (100 mL), the aqueous layer was extracted with ethyl acetate (400 ml×3), and the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. 17.0g of the title product are obtained as a brown solid, which is used directly in the next step.
LCMS:m/z 292.10[M+H] +
Step 9: preparation of 8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -2- (methylsulfonyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (1 i)
8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -2- (methylthio) pyrido [2,3-d ] pyrimidin-7 (8H) -one (17.0 g, crude), potassium hydrogen persulfate (76.9 g,125 mmol), 2-methyltetrahydrofuran (250 mL), water (60 mL) were added to the reaction flask at room temperature, and stirred at room temperature for 4H. The solution was cooled in a water bath, diluted with water (100 mL) and saturated aqueous sodium chloride (100 mL) and extracted with ethyl acetate (500 ml×3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (eluent: petroleum ether: ethyl acetate=1:1-1) to give 10.8g of the title product as a foamy brown solid in yield: 66.6%.
LCMS:m/z 306.09[M-18] +
Step 10: preparation of 4- ((8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (1 j)
8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -2- (methylsulfonyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (3.80 g,11.8 mmol) and tert-butyl 4-aminopiperidine-1-carboxylate (2.82 g,14.2 mmol), diisopropylethylamine (3.05 g,23.6 mmol), 2-methyltetrahydrofuran (50 mL) were added to the reaction flask at room temperature and the reaction stirred at 60℃overnight. After cooling to room temperature, ethyl acetate (200 mL), water (50 mL) and saturated aqueous sodium bicarbonate (50 mL) were added. The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (eluent: dichloromethane: methanol=0-5%) to give 4.55g of the title product as a yellow foam solid in yield: 87.1%.
Step 11: preparation of 8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -2- (piperidin-4-ylamino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (1 k)
Tert-butyl 4- ((8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (3.63 g,8.20 mmol) was added to a 10mL dioxane solution of hydrochloric acid at room temperature, stirred for 2h, a yellow solid precipitated, and concentrated under reduced pressure. Sodium hydroxide (aq) was added to the residue at 0deg.C to bring the pH to 10, dichloromethane (200 mL. Times.3) was added, the organic phase was collected, and concentrated under reduced pressure to give 2.57g of the title product as a yellow solid, which was used directly in the next step.
LCMS:m/z 314.19[M+H] +
Step 12: preparation of N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l)
Cyclopropanesulfonamide (2.42 g,20.0 mmol), triethylamine (4.04 g,40.0 mmol) and tetrahydrofuran (40 mL) were added to a reaction flask at room temperature under nitrogen, and after stirring for 10min, tert-butyldimethylsilyl chloride (3.60 g,24.0 mmol), dimethylaminopyridine (254 mg,2.00 mmol) were slowly added and reacted at 30 ℃ for 16h with stirring, solids precipitated, concentrated under reduced pressure, ethyl acetate (50 mL x 3) and water (30 mL) were added to the residue, the organic phase was collected, the aqueous layer was extracted with ethyl acetate (30 mL x 3), the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: ethyl acetate=0-20%) to give 3.27g of the title product as a white solid, yield: 69.6%.
LCMS:m/z 236.11[M+H] +
Step 13: preparation of 2- ((1- (cyclopropanesulfonylimino) piperidin-4-yl) amino) -8- (((1R, 2R) -2-hydroxy-2-methylcyclopentyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (1)
A solution of triphenylphosphine dichloride (1.83 g,5.50 mmol) in dichloromethane (6 mL) was cooled to 0deg.C under nitrogen, triethylamine (178 mg,8.00 mmol) was added, and after stirring for 15min, the compound N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1.18 g,5.00 mmol) was added, and after stirring for 20min, the compound 1k (343 mg,1.00 mmol) was added, bi Huanman L was added to room temperature, and the reaction was stirred at room temperature for 16h. The reaction was quenched by addition of saturated ammonium chloride solution (1 mL), the organic phase was collected, concentrated under reduced pressure, and the resulting residue was purified by column chromatography (eluent: ethyl acetate: methanol=0-10%) and separated by preparative liquid chromatography (column type: daisosei 30mm x 250mm, c18, 10um,100a, mobile phase: acetonitrile/water, gradient: 30% -80%) to give 71.0mg of the title compound as a white solid in 15.9% yield.
LCMS:m/z 447.21[M+H] +
1 H 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)。
Example 2: preparation of 2- ((1- (cyclopropanesulfonylimino) piperidin-4-yl) amino) -6- (difluoromethyl) -8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (2)
Figure PCTCN2022119618-APPB-000031
Step 1: preparation of 2- ((1- (cyclopropanesulfonylimino) piperidin-4-yl) amino) -6- (difluoromethyl) -8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (2)
An aqueous solution (1 mL) of bis (((difluoromethyl) sulfinyl) oxy) zinc (59.0 mg,0.200 mmol), ferrous chloride (6.35 mg,0.050 mmol) was added dropwise to a solution (6 mL) of 2- ((1- (cyclopropanesulfonylimino) piperidin-4-yl) amino) -8- (((1R, 2R) -2-hydroxy-2-methylcyclopentyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (44.6 mg,0.100 mmol), TFA (11.4 mg,0.100 mmol) in DMSO (6 mL) at room temperature TBHP (51.2 mg,70% wt aqueous solution, 0.400 mmol) was then added to the diluted TBHP for 2H at 20 ℃; the combined organic phases were concentrated under reduced pressure and the residue was purified by preparative thin layer chromatography (developer: dichloromethane: methanol=20:1) and separated by preparative liquid chromatography (column type: daisosei: 30mm x 250mm, c18, 10um,100a, mobile phase: acetonitrile/water, gradient: 30% -80%) to give 10.1mg of the title compound as a white solid, yield 20.4%.
LCMS:m/z 497.21[M+H] +
1 H 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)。
Example 3: preparation of 8-cyclopentyl-2- (((1- (cyclopropanesulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (3)
Figure PCTCN2022119618-APPB-000032
Step 1: preparation of (4- (cyclopentylamino) -2- (methylthio) pyrimidin-5-yl) methanol (3 a)
(4-chloro-2- (methylthio) pyrimidin-5-yl) methanol (1 e) (4.66 g,20.0 mmol), cyclopentylamine (2.04 g,24.0 mmol), triethylamine (6.06 g,60.0 mmol), and tetrahydrofuran (50 mL) were added to a 250mL reaction flask at room temperature, and the reaction was stirred at room temperature overnight. After the completion of the reaction, the mixture was concentrated under reduced pressure, ethyl acetate (100 mL. Times.3) and water (70 mL) were added, and the organic phase was collected and concentrated under reduced pressure to give 5.10g of the title product as a pale yellow solid, which was used directly in the next step.
LCMS:m/z 240.11[M+H] +
Step 2: preparation of 4- (cyclopentylamino) -2- (methylthio) pyrimidine-5-carbaldehyde (3 b)
(4- (cyclopentylamino) -2- (methylthio) pyrimidin-5-yl) methanol (13.4 g, crude), manganese dioxide (76.9 g,884 mmol), ethyl acetate (310 mL) were added to a 1L reaction flask at room temperature, and the reaction was stirred at 50℃overnight. After the reaction was completed, the solid was removed by filtration, and the filtrate was concentrated and purified by column chromatography (eluent: petroleum ether: ethyl acetate=0 to 20%) to give 9.37g of the title product as a yellow oil, yield: 89.4%.
LCMS:m/z 238.09[M+H] +
Step 3: preparation of 8-cyclopentyl-2- (methylthio) pyrido [2,3-d ] pyrimidin-7 (8H) -one (3 c)
4- (cyclopentylamino) -2- (methylthio) pyrimidine-5-carbaldehyde (11.8 g,49.8 mmol), tetrahydrofuran (250 mL) and ethyl acetate (11.0 g,125 mmol) were added to a 1L three-necked flask at room temperature, cooled to-5℃in a methanol-ice bath, and lithium hexamethyl-silamide (150 mL,1.0M in tetrahydrofuran, 150 mmol) was slowly added under nitrogen atmosphere, during which time the temperature was allowed to gradually rise to room temperature and stirred overnight. The resulting red solution was cooled to about 3 ℃ in an ice-water bath, then ethanol (200 mL) was slowly added. The mixture was stirred in an ice bath for 1h, then the cooling bath was removed and the solution allowed to warm to room temperature and stirring continued for 1h. The residue was diluted with water (100 mL) and saturated aqueous sodium chloride (100 mL), the aqueous layer was extracted with ethyl acetate (500 ml×3), the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (eluent: petroleum ether: ethyl acetate=0-40%) to give 11.0g of the title product as a yellow solid in yield: 84.6%.
LCMS:m/z 262.09[M+H] +
Step 4: preparation of 8-cyclopentyl-2- (methylsulfonyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (3 d)
8-cyclopentyl-2- (methylthio) pyrido [2,3-d ] pyrimidin-7 (8H) -one (11.0 g,42.2 mmol), potassium hydrogen persulfate (64.8 g,105 mmol), 2-methyltetrahydrofuran (200 mL), and water (40 mL) were added to a 1L reaction flask at room temperature, and the reaction was stirred at room temperature overnight. The mixture was filtered, the filter cake was washed with ethyl acetate (100 mL. Times.3), the filtrate was collected, ethyl acetate (200 mL) and water (100 mL) were added, and the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (eluent: petroleum ether: ethyl acetate=0-100%) to give 8.37g of the title product as a yellow-brown solid in yield: 67.7%.
LCMS:m/z 294.08[M+H] +
Step 5: preparation of tert-butyl 4- ((8-cyclopentyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (3 e)
8-cyclopentyl-2- (methylsulfonyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (4.40 g,15.0 mmol), tert-butyl 4-aminopiperidine-1-carboxylate (3.60 g,18.0 mmol), diisopropylethylamine (3.87 g,30.0 mmol), tetrahydrofuran (75 mL) were added to the reaction flask at room temperature and the reaction was stirred at 60℃overnight. After cooling to room temperature, ethyl acetate (100 mL. Times.3) and water (100 mL) were added. The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (eluent: dichloromethane: methanol=0-5%) to give 5.80g of the title product as a yellow foam solid in yield: 93.6%.
LCMS:m/z 414.24[M+H] +
Step 6: preparation of 8-cyclopentyl-2- (piperidin-4-ylamino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (3 f)
Tert-butyl 4- ((8-cyclopentyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (4.13 g,10.0 mmol) was added to a solution of 10mL of hydrochloric acid in dioxane at room temperature, stirred for 2h, a yellow solid precipitated, and concentrated under reduced pressure. Sodium hydroxide (aq) was added to the residue at 0deg.C to bring the pH to 10, methylene chloride (200 mL. Times.3) was added, the organic phase was collected, and concentrated under reduced pressure to give 2.42g of the title product as a yellow solid, which was used directly in the next step.
LCMS:m/z 314.19[M+H] +
Step 7: preparation of 8-cyclopentyl-2- (((1- (cyclopropanesulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (3)
A solution of triphenylphosphine (1.82 g,5.50 mmol) in methylene chloride (20 mL) was cooled to 0℃under nitrogen, triethylamine (909 mg,9.00 mmol) was added, after stirring for 15min, the compound N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) (690 mg,5.00 mmol) was added, after stirring for 20min, the compound (8-cyclopentyl-2- (piperidin-4-ylamino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (313 mg,1.00 mmol), bi Huanman L was added to room temperature, stirring was carried out for 16H, quenching was carried out by adding saturated ammonium chloride solution (5 mL), the organic phase was collected, and the resulting residue was purified by column chromatography (eluent: ethyl acetate: methanol=0-10%) to give 384mg of a pale yellow solid product, which was isolated by preparative liquid chromatography (column model: daisog: 30mm, 18, 10 um: 100 mg, mobile phase: A, 0.250 mg, aqueous gradient: 0.250 mg, 80% of the title compound was obtained as a white solid, yield: 80.250% acetonitrile, 30% of which was obtained.
LCMS:m/z 417.20[M+H] +
1 H 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)。
Example 4: preparation of 8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -2- (((1- (N-methylcyclopropane-sulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (4)
Figure PCTCN2022119618-APPB-000033
Step 1: preparation of N-methylpropane-1-sulfonamide (4 a)
Cyclopropanesulfonyl chloride (1.43 g,10.0 mmol) and tetrahydrofuran (5 mL) were added to a glass tube at room temperature, cooled to 0℃and triethylamine (4.04 g,40.0 mmol) and aqueous methylamine solution (5 mL) were added thereto, and the mixture was stirred at room temperature for 16 hours. Concentrated under reduced pressure, ethyl acetate (50 ml×3) and water (30 mL) were added to the residue, the organic phase was collected, the aqueous layer was extracted with ethyl acetate (30 ml×3), the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: petroleum ether: ethyl acetate=0-50%) to give 1.26g of the title product as a white oil, yield: 92.0%.
LCMS:m/z 138.08[M+H] +
Step 2: preparation of 8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -2- (((1- (N-methylcyclopropane-sulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (4)
The title compound was obtained in the same manner as in step 13 of example 1 except that N-methylpropane-1-sulfonamide was used instead of N- (t-butyldimethylsilyl) cyclopropanesulfonamide.
LCMS:m/z 461.23[M+H] +
1 H 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)。
Example 5: preparation of 6- (difluoromethyl) -8- (((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -2- (((1- (N-methylcyclopropane-sulfonylimino)) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (5)
Figure PCTCN2022119618-APPB-000034
The title compound was obtained in the same manner as in example 2 except that compound 4 was used instead of compound 1.
LCMS:m/z 511.22[M+H] +
1 H 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)。
Example 6: preparation of 8-cyclopentyl-2- (((8- (cyclopropanesulfonylimino) -8-azabis [3.2.1] oct-3-yl ] amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (6)
Figure PCTCN2022119618-APPB-000035
The title compound was prepared in the same manner as in example 3 except that 2- ((8-azabicyclo [3.2.1] oct-3-yl) amine was used instead of cyclopentylamine in step 1.
LCMS:m/z 443.22[M+H] +
1 HNMR(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)。
Example 7: preparation of 8-cyclopentyl-2- (((1- (prop-2-ylsulfonimido) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (7)
Figure PCTCN2022119618-APPB-000036
Step 1: preparation of N- (tert-butyldimethylsilyl) propane-2-sulfonamide (7 a)
The title compound was obtained in the same manner as in step 12 of example 1 except that propane-2-sulfonamide was used instead of cyclopropanesulfonamide.
Step 2: preparation of 8-cyclopentyl-2- (((1- (prop-2-ylsulfonimido) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one
The title compound was obtained in the same manner as in the production method of example 3 except that N- (t-butyldimethylsilyl) propane-2-sulfonamide (7 a) was used instead of N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l).
LCMS:m/z 419.22[M+H] +
1 H 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)。
Example 8: preparation of 8-cyclopentyl-2- (((1- (propylsulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (8)
Figure PCTCN2022119618-APPB-000037
Figure PCTCN2022119618-APPB-000038
Step 1: preparation of N- (tert-butyldimethylsilyl) propane-1-sulfonamide (8 a)
The title compound was obtained in the same manner as in step 12 of example 1 except that propane-1-sulfonamide was used instead of cyclopropanesulfonamide.
Step 2: preparation of 8-cyclopentyl-2- (((1- (prop-2-ylsulfonimido) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one
The title compound was obtained in the same manner as in the production method of example 3 except that N- (t-butyldimethylsilyl) propane-1-sulfonamide (8 a) was used instead of N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l).
LCMS:m/z 419.22[M+H] +
1 H 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)。
Example 9: preparation of 8-cyclopentyl-2- (((1- (N-methylpropylsulfonimido) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (9)
Figure PCTCN2022119618-APPB-000039
Step 1: preparation of N-methylpropane-1-sulfonamide (9 a)
The title compound was obtained in the same manner as in step 1 of example 4 except that propane-1-sulfonyl chloride was used instead of cyclopropanesulfonyl chloride.
Step 2: preparation of 8-cyclopentyl-2- (((1- (prop-2-ylsulfonimido) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one
The title compound was obtained in the same manner as in the preparation method of example 3 except that N-methylpropane-1-sulfonamide (9 a) was used instead of N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l).
LCMS:m/z 432.23[M+H] +
1 H 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)。
Example 10: preparation of 8-cyclopentyl-6- (difluoromethyl) -2- (((1- (N-methylpropylsulfonyl imino) piperidin-4-yl) amino) pyridinyl [2,3-d ] pyrimidin-7 (8H) -one (10)
Figure PCTCN2022119618-APPB-000040
The title compound was obtained in the same manner as in example 2 except that compound 9 was used instead of compound 1.
LCMS:m/z 483.1[M+H] +
1 HNMR(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)。
Example 11: preparation of 8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -2- (((1- (S-methylsulfonylimidoyl) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (11)
Figure PCTCN2022119618-APPB-000041
Figure PCTCN2022119618-APPB-000042
Step 1: preparation of N- (tert-butyldimethylsilyl) methanesulfonamide (11 a)
The title compound was obtained in the same manner as in step 12 of example 1 except that methanesulfonamide was used instead of cyclopropanesulfonamide.
The remaining procedure was the same as that of example 1 except that N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) of example 1, step 13 was replaced with N- (t-butyldimethylsilyl) methanesulfonamide (11 a), to prepare the title compound.
LCMS:m/z 421.19[M+H] +
1 H 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)。
Example 12: preparation of 6- (difluoromethyl) -8- (((1R, 2R) -2-hydroxy-2-methylcyclopentyl) -2- ((1- (S-methylsulfonylimidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (12)
Figure PCTCN2022119618-APPB-000043
The title compound was obtained in the same manner as in example 2 except that compound 11 was used instead of compound 1.
LCMS:m/z 471.19[M+H] +
1 H 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)。
Example 13: preparation of 8-cyclopentyl-2- (((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (13)
Figure PCTCN2022119618-APPB-000044
The title compound was obtained in the same manner as in the production method of example 3 except that N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) in step 7 was replaced with N- (t-butyldimethylsilyl) methanesulfonamide (11 a).
LCMS:m/z 391.18[M+H] +
1 H 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)。
Example 14: preparation of 2- ((1- (N, S-dimethyl-sulfonylimino) piperidin-4-yl) amino) -8- (((1R, 2R) -2-hydroxy-2-methylcyclopentyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (14)
Figure PCTCN2022119618-APPB-000045
Step 1: preparation of N-methyl methanesulfonamide (14 a)
The title compound was obtained in the same manner as in step 1 of example 4 except that methanesulfonyl chloride was used instead of cyclopropanesulfonyl chloride.
The remaining procedure was the same as that of example 1, except that N-methylmethanesulfonamide (14 a) was used in place of N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) of step 13 of example 1, to obtain the title compound.
LCMS:m/z 435.21[M+H] +
1 H 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)。
Example 15: preparation of 6- (difluoromethyl) -2- (((1- (N, S-dimethyl-sulfonylimino) piperidin-4-yl) amino) -8- ((1R, 2R) -2-hydroxy-2-methylcyclopentyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (15)
Figure PCTCN2022119618-APPB-000046
The title compound was obtained in the same manner as in example 2 except that compound 14 was used instead of compound 1.
LCMS:m/z 485.21[M+H] +
1 HNMR(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)。
Example 16: preparation of 8-cyclopentyl-2- (((1- (N, S-dimethyl-sulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (16)
Figure PCTCN2022119618-APPB-000047
Figure PCTCN2022119618-APPB-000048
The title compound was obtained in the same manner as in the preparation method of example 3 except that N-methylmethanesulfonamide (14 a) was used instead of N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) in step 7.
LCMS:m/z 405.203[M+H] +
1 H 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)。
Example 17: preparation of 8-cyclopentyl-6- (difluoromethyl) -2- (((1- (S-methylsulfonylimidopiperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (17)
Figure PCTCN2022119618-APPB-000049
The title compound was obtained in the same manner as in example 2 except that compound 13 was used instead of compound 1.
LCMS:m/z 441.18[M+H] +
1 H 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)。
Example 18: preparation of 8-cyclopentyl-6- (difluoromethyl) -2- (((1- (N, S-methylsulfonylamino)) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (18)
Figure PCTCN2022119618-APPB-000050
The title compound was obtained in the same manner as in example 2 except that compound 16 was used instead of compound 1.
LCMS:m/z 455.20[M+H] +
1 HNMR(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)。
Example 19: preparation of 8-cyclopentyl-2- (((1- (N-methylcyclopropane-sulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (3)
Figure PCTCN2022119618-APPB-000051
The title compound was obtained in the same manner as in the preparation method of example 3 except that N-methylpropane-1-sulfonamide (4 a) was used instead of N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l).
LCMS:m/z 431.23[M+H] +
1 H 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)。
Example 20: preparation of 8-cyclopentyl-2- (((1- (cyclopropanesulfonylimino) piperidin-4-yl) amino) -6- (difluoromethyl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (20)
Figure PCTCN2022119618-APPB-000052
The title compound was obtained in the same manner as in example 2 except that compound 3 was used instead of compound 1.
LCMS:m/z 467.20[M+H] +
1 HNMR(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)。
Example 21: preparation of 8-cyclopentyl-6- (difluoromethyl) -2- (((1- (N-methylpropylsulfonyl imino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (21)
Figure PCTCN2022119618-APPB-000053
The title compound was obtained in the same manner as in example 2 except that compound 19 was used instead of compound 1.
LCMS:m/z 481.21[M+H] +
1 H 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)。
Example 22: preparation of 8- (2-methylcyclopentyl) -2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (22-P1 and 22-P2)
Figure PCTCN2022119618-APPB-000054
The title compound was obtained in the same manner as in the preparation method of example 3 except that 2-methylcyclopentane-1-amine hydrochloride was used instead of cyclopentylamine and N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (tert-butyldimethylsilyl) methanesulfonamide (11 a). Purification by column chromatography (eluent: ethyl acetate: methanol=0-10%) followed by separation by preparative liquid chromatography afforded compounds 22-P1 and 22-P2.
LCMS:m/z 405.20[M+H] +
Single configuration compound 22-P1 (shorter retention time):
Preparation of liquid chromatography: chromatographic column: daisosei 30mm×250mm, C18, 10um,100A, mobile phase: acetonitrile/water, gradient: 30% -80%.
1 H 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)。
Single configuration compound 22-P2 (longer retention time):
preparation of liquid chromatography: chromatographic column: daisosei 30mm×250mm, C18, 10um,100A, mobile phase: acetonitrile/water, gradient: 30% -80%.
1 H 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)。
Example 23: preparation of 8- (2, 2-dimethylcyclopentyl) -2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (23)
Figure PCTCN2022119618-APPB-000055
The title compound was obtained in the same manner as in the production method of example 3 except that 2, 2-dimethylcyclopentylamine was used instead of cyclopentylamine and N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (t-butyldimethylsilyl) methanesulfonamide (11 a).
LCMS:m/z 419.20[M+H]+。
1 H 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)。
Example 24: preparation of 2- ((1- (S-methylsulfonylimidopiperidin-4-yl) amino) -8- (tetrahydrofuran-3-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (24)
Figure PCTCN2022119618-APPB-000056
The title compound was obtained in the same manner as in the preparation method of example 3 except that tetrahydrofuran-3-amine was used instead of cyclopentylamine and N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (t-butyldimethylsilyl) methanesulfonamide (11 a).
LCMS:m/z 393.16[M+H] +
1 HNMR(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)。
Example 25: preparation of 2- ((1- (S-methylsulfonylimidopiperidin-4-yl) amino) -8- (spiro [2.4] hept-4-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (25)
Figure PCTCN2022119618-APPB-000057
The title compound was obtained in the same manner as in the preparation method of example 3 except that spiro [2.4] hept-4-amine was used instead of cyclopentylamine and N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (tert-butyldimethylsilyl) methanesulfonamide (11 a).
LCMS:m/z 417.20[M+H] +
1 H 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)。
Example 26: preparation of 8-cyclopentyl-6-methyl-2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (26)
Figure PCTCN2022119618-APPB-000058
Figure PCTCN2022119618-APPB-000059
The title compound was obtained in the same manner as in the production method of example 3 except that ethyl acetate was replaced with ethyl propionate and N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (t-butyldimethylsilyl) methanesulfonamide (11 a).
LCMS:m/z 405.20[M+H] +
1 H 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)。
Example 27: preparation of 8-cyclopentyl-5, 6-dimethyl-2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (27)
Figure PCTCN2022119618-APPB-000060
Step 1: preparation of 1- (4- (cyclopentylamino) -2- (methylthio) pyrimidin-5-yl) -1-ethanol (27 a)
4- (Cyclopentaamino) -2- (methylthio) pyrimidine-5-carbaldehyde (3 b) (929 mg,3.70 mmol), THF (30 mL) were added to a three-necked flask at room temperature, cooled to-75deg.C, and methyl magnesium bromide (3.7 mL,1.0M in THF, 3.70 mmol) was slowly added under nitrogen atmosphere, during which time the temperature was allowed to gradually rise to room temperature and stir overnight. Cooling to 0deg.C, adding water (10 mL), quenching, collecting organic phase, and concentrating under reduced pressure. The residue was purified by column chromatography (eluent: petroleum ether: ethyl acetate=0-75%) to give 740mg of the title product as a white solid in yield: 75.0%.
LCMS:m/z 254.14[M+H] +
Step 2: preparation of 1- (4- (cyclopentylamino) -2- (methylthio) pyrimidin-5-yl) -1-ethanone (27 b)
1- (4- (cyclopentylamino) -2- (methylthio) pyrimidin-5-yl) -1-ethanol (27 a) (740 mg,2.77 mmol), manganese dioxide (4.54 g,55.4 mmol), ethyl acetate (10 mL) were added to the reaction flask at room temperature, and the reaction was stirred at 50℃overnight. After the reaction was completed, the solid was removed by filtration, and the filtrate was concentrated and purified by column chromatography (eluent: petroleum ether: ethyl acetate=0 to 20%) to give 700mg of the title product as a pale yellow solid, yield: 95.4%.
LCMS:m/z 252.14[M+H] +
The remaining procedure was the same as in the preparation method of example 3, except that 1- (4- (cyclopentylamino) -2- (methylthio) pyrimidin-5-yl) -1-ethanone (27 b) was used instead of 4- (cyclopentylamino) -2- (methylthio) pyrimidine-5-carbaldehyde (3 b), ethyl propionate was used instead of ethyl acetate, and N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (t-butyldimethylsilyl) methanesulfonamide (11 a), to obtain compound 27.
LCMS:m/z 419.22[M+H] +
1 H 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)。
Example 28: preparation of 6-acetyl-8-cyclopentyl-2- (((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (28)
Figure PCTCN2022119618-APPB-000061
Step 1: preparation of tert-butyl 4- ((6-iodo-8-cyclopentyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (28 a)
Tert-butyl 4- ((8-cyclopentyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (3 e) (149 mg,0.36 mmol), NIS (122 mg,0.55 mmol), p-toluenesulfonic acid (6.24 mg,0.036 mmol), acetonitrile (2 mL) were added to the reaction flask at room temperature and the reaction was stirred at room temperature overnight. Concentrated under reduced pressure, and EtOAc (10 mL. Times.3) and water (5 mL) were added to the residue. The organic phase was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: petroleum ether: ethyl acetate=0-40%) to give 164mg of the title product as a yellow solid, yield: 84.7%.
LCMS:m/z 540.15[M+H] +
Step 2: preparation of tert-butyl 4- (((8-cyclopentyl-6- (1-ethoxyvinyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (28 b)
Tert-butyl 4- ((6-iodo-8-cyclopentyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (28 a) (298 mg,0.55 mmol), tributyl (1-ethoxyvinyl) stannane (299 mg, 0.823mmol), tetrakis triphenylphosphine palladium (76.3 mg,0.066 mmol), toluene (5 mL) were added to the reaction flask at room temperature and the reaction was stirred at reflux overnight. After cooling to room temperature, the mixture was concentrated under reduced pressure, and EtOAc (20 mL. Times.3) and water (10 mL) were added to the residue. The organic phase was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: petroleum ether: ethyl acetate=0-40%) to give 160mg of the title product as a pale yellow solid, yield: 63.8%.
LCMS:m/z 484.28[M+H] + /456.25[M+H] +
Step 3: preparation of 6-acetyl-8-cyclopentyl-2- (piperidin-4-ylamino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (28 c)
Tert-butyl 4- (((8-cyclopentyl-6- (1-ethoxyvinyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (28 b) (160 mg,0.350 mmol) was added to 2mL of dioxane solution of hydrochloric acid at room temperature, stirred for 2h, yellow solid precipitated, concentrated under reduced pressure to give yellow solid, potassium carbonate (276 mg,2.00 mmol) was added, water (5 mL) was added, stirred for 2h, dichloromethane (10 mL. Times.3) was added, the organic phase was collected, concentrated under reduced pressure to give 93mg of the title product as a pale yellow solid, which was used directly in the next step.
LCMS:m/z 356.20[M+H] +
The remaining procedure was the same as in the preparation method of example 3, except that 6-acetyl-8-cyclopentyl-2- (piperidin-4-ylamino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (28 c) was used instead of 8-cyclopentyl-2- (piperidin-4-ylamino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (3 f), and N- (tert-butyldimethylsilyl) methanesulfonamide (11 a) was used instead of N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l), to obtain compound 28.
LCMS:m/z 433.19[M+H] +
1 H 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)。
Example 29: preparation of 6-acetyl-8-cyclopentyl-5-methyl-2- (((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (29)
Figure PCTCN2022119618-APPB-000062
Step 1: preparation of tert-butyl 4- ((6-bromo-8-cyclopentyl-5-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (29 a)
4-aminopiperidine-1-carboxylic acid tert-butyl ester (2.52 g,12.0 mmol), toluene (10 mL) were added to the reaction flask, cooled to 0℃under nitrogen, lithium bis (trimethylsilyl) amide (1M in THF, 12mL,12.0 mmol) was added, the reaction stirred for 20 min, and the compound 6-bromo-2-chloro-8-cyclopentyl-5-methylpyrido [2,3-d ] pyrimidin-7 (8H) -one (1.03 g,3.00 mmol) (commercial agent CAS: 1016636-76-2) was added and the reaction stirred overnight. Concentrated under reduced pressure, and EtOAc (20 mL. Times.3) and water (10 mL) were added to the residue. The organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by column chromatography (eluent: petroleum ether: ethyl acetate=0-40%) to give 620mg of the title product as a pale yellow solid, yield: 40.9%.
LCMS:m/z 506.17[M+H] +
The remaining procedure was followed in the same manner as in example 28 except for using 4- ((6-bromo-8-cyclopentyl-5-methyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (29 a) in place of 4- ((6-iodo-8-cyclopentyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (28 a) to give compound 29.
LCMS:m/z 447.21[M+H] +
1 H 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)。
Example 30: 8-cyclopentyl-2- (((1- (1-oxo-4, 5-dihydro-3H-1 lambda) 6 -isothiazol-1-yl) piperidin-4-yl) amino) pyrido [2,3-d]Preparation of pyrimidin-7 (8H) -ones (30)
Figure PCTCN2022119618-APPB-000063
In the same manner as in the production method of example 3, except that isothiazolidine 1, 1-dioxide was used instead of N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l), compound 30 was produced.
LCMS:m/z 417.20[M+H] +
1 H 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)。
Example 31: preparation of 6-methyl-8- (2-methylcyclopentyl) -2- ((1- (S-methylsulfonylimidopiperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (31)
Figure PCTCN2022119618-APPB-000064
Compound 31 was produced in the same manner as in example 22, except that ethyl propionate was used instead of ethyl acetate.
LCMS:m/z 419.20[M+H] +
1 H 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)。
Example 32: preparation of 5, 6-dimethyl-8- (2-methylcyclopentyl) -2- (((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (32)
Figure PCTCN2022119618-APPB-000065
The title compound was prepared in the same manner as in example 27 except that 4- ((2-methylcyclopentyl) amino) -2- (methylthio) pyrimidine-5-carbaldehyde (22 b) was used instead of 4- (cyclopentylamino) -2- (methylthio) pyrimidine-5-carbaldehyde (3 b).
LCMS:m/z 433.23[M+H] +
1 HNMR(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)。
Example 33: preparation of 6- (difluoromethyl) -8- (2-methylcyclopentyl) -2- (((1- (S-methylsulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (33-P1 and 33-P2)
Figure PCTCN2022119618-APPB-000066
Step 1: preparation of 6- (difluoromethyl) -8- (2-methylcyclopentyl) -2- (((1- (methylsulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (33-P1 and 33-P2)
Sodium difluoromethane sulfinate (172 mg,1.25 mmol), water (3 mL), dimethyl sulfoxide (15 mL) were added to the reaction flask at room temperature, and stirred at room temperature for 25 minutes, 8- (2-methylcyclopentyl) -2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (202 mg,0.50 mmol), ferrous chloride (10.2 mg,0.08 mmol) were added. t-Butanol peroxide (64.0 mg,70% wt aqueous solution, 0.50 mmol) was taken, diluted to 1mL with DMSO, added dropwise to the reaction system, and stirred at room temperature overnight. The reaction solution was poured into 10% EDTA (30 mL) and extracted with EtOAc (50 mL. Times.3). The organic phases were combined, purified by preparative thin layer chromatography (developer: dichloromethane: methanol=20:1) and separated by preparative liquid chromatography to give compounds 33-P1 (10.0 mg) and 33-P2 (24.5 mg).
Single configuration Compound 33-P1 (shorter retention time)
Preparation of liquid chromatography: chromatographic column: daisosei 30mm×250mm, C18, 10um,100A, mobile phase: acetonitrile/water, gradient: 30% -80%.
LCMS:m/z 455.54[M+H] +
1 H 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)。
Single configuration Compound 33-P2 (longer retention time)
Preparation of liquid chromatography: chromatographic column: daisosei 30mm×250mm, C18, 10um,100A, mobile phase: acetonitrile/water, gradient: 30% -80%.
LCMS:m/z 455.54[M+H] +
1 H 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).)。
Example 34: preparation of 6-acetyl-5-methyl-8- (2-methylcyclopentyl) -2- (((1- (S-methylsulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (34)
Figure PCTCN2022119618-APPB-000067
Figure PCTCN2022119618-APPB-000068
Step 1: preparation of tert-butyl 4- (((5-methyl-8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (34 c)
In the same manner as in the preparation of compound 27e in example 27, except that 1- (4- ((2-methylcyclopentyl) amino) -2- (methylthio) pyrimidin-5-yl) ethan-1-one (32 b) was used instead of 1- (4- (cyclopentylamino) -2- (methylthio) pyrimidin-5-yl) -1-ethanone (27 b) and ethyl acetate was used instead of ethyl propionate, compound 34c was prepared.
The remaining procedure is as in example 28, except that tert-butyl 4- (((5-methyl-8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) tert-butyl-1-carboxylate (34 c) is used instead of tert-butyl 4- ((8-cyclopentyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (3 e) to give compound 34.
LCMS:m/z 461.21[M+H] +
1 H 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)。
Example 35: preparation of 6-acetyl-8- (2-methylcyclopentyl) -2- (((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (35)
Figure PCTCN2022119618-APPB-000069
Figure PCTCN2022119618-APPB-000070
In the same manner as in example 28, except that tert-butyl 4- (((8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (22 e) was used instead of tert-butyl 4- ((8-cyclopentyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (3 e), compound 35 was prepared.
LCMS:m/z 447.19[M+H] +
1 H 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)。
Example 36: preparation of 8-cyclopentyl-2- (((1- (thiophene-2-sulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (36)
Figure PCTCN2022119618-APPB-000071
Step 1: preparation of N- (tert-butyldimethylsilyl) thiophene-2-sulfonamide (36 a)
The title compound was obtained in the same manner as in step 12 of example 1 except that thiophene-2-sulfonamide was used instead of cyclopropanesulfonamide.
The remaining procedure was the same as in the preparation method of example 3, except that N- (tert-butyldimethylsilyl) thiophene-2-sulfonamide (36 a) was used instead of N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l), to obtain compound 36.
LCMS:m/z 459.18[M+H] +
1 H 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)。
Example 37: preparation of 6-fluoro-8- (2-methylcyclopentyl) -2- ((1- (S-methylsulfonylimidopiperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (37)
Figure PCTCN2022119618-APPB-000072
Compound 37 was produced in the same manner as in example 22, except that ethyl 2-fluoroacetate was used instead of ethyl acetate.
LCMS:m/z 423.20[M+H] +
1 H 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)。
Example 38: preparation of 6-chloro-8- (2-methylcyclopentyl) -2- ((1- (methylsulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (38)
Figure PCTCN2022119618-APPB-000073
Step 1: preparation of tert-butyl 4- (((6-chloro-8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (38 a)
Tert-butyl 4- (((8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (22 e) (3411 mg,0.80 mmol), NCS (128 mg,0.96 mmol), 2-methyltetrahydrofuran (10 mL) were added to the reaction flask at room temperature, stirred overnight at 60 ℃, concentrated under reduced pressure, etOAc (20 mL. Times.3) and water (10 mL) were added to the residue, the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: petroleum ether: ethyl acetate=0-50%) to give 0.32g of the title product as a yellow solid in 86.0% yield.
LCMS:m/z 462.15[M+H] +
The remaining procedure was the same as in the preparation of example 3, except that tert-butyl 4- (((6-chloro-8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (38 a) was used instead of tert-butyl 4- ((8-cyclopentyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (3 e) and N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (tert-butyldimethylsilyl) methanesulfonamide (11 a) to give compound 38.
LCMS:m/z 439.20[M+H] +
1 H 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)。
Example 39: preparation of 6-isopropyl-8- (2-methylcyclopentyl) -2- (((1- (S-methylsulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (39)
Figure PCTCN2022119618-APPB-000074
Step 1: preparation of tert-butyl 4- (((6-bromo-8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (39 a)
Tert-butyl 4- (((8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (22 e) (854 mg,2.00 mmol), NBS (391.0 mg,2.20 mmol), 2-methyltetrahydrofuran (20 mL) were added to the reaction flask at room temperature, stirred overnight at 60 ℃ C., concentrated under reduced pressure, etOAc (20 mL. Times.3) and water (10 mL) were added to the residue, the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: ethyl acetate=0-50%) to give 0.95g of the title product as a yellow solid in 93.0% yield.
LCMS:m/z 507.15[M+H] +
Step 2: preparation of tert-butyl 4- ((8- (2-methylcyclopentyl) -7-oxo-6- (prop-1-en-2-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (39 b)
Tert-butyl 4- (((6-bromo-8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (39 a) (405 mg,0.8 mmol), isopropylboronate (268.0 mg,1.60 mmol), tetrakis triphenylphosphine palladium (124 mg,0.08 mmol), potassium phosphate (508 mg,2.40 mmol), dioxane: water (15 mL:3 mL) were added to the reaction flask at room temperature, stirred under reflux overnight, cooled to room temperature, concentrated under reduced pressure, etOAc (20 mL×3) and water (10 mL) were added to the residue, the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure to give 373mg of the crude title product as a black oil, which was used directly in the next step.
LCMS:m/z 468.28[M+H] +
Step 3: preparation of tert-butyl 4- (((6-isopropyl-8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (39 c)
4- ((8- (2-methylcyclopentyl) -7-oxo-6- (prop-1-en-2-yl) -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (39 b) (373 mg,0.80 mmol), palladium on carbon (37.0 mg), methanol (15 mL) were added to the reaction flask at room temperature and the reaction was stirred under hydrogen atmosphere at 50 ℃ overnight. After cooling to room temperature, the mixture was concentrated under reduced pressure, and EtOAc (20 mL. Times.3) and water (10 mL) were added to the residue. The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 180mg of crude title product as an oil, which was used directly in the next step.
LCMS:m/z 470.28[M+H] +
The remaining procedure was the same as in the preparation method of example 3, except that tert-butyl 4- (((6-isopropyl-8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (39 c) was used instead of tert-butyl 4- ((8-cyclopentyl-7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (3 e) and N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (tert-butyldimethylsilyl) methanesulfonamide (11 a) to obtain compound 39.
LCMS:m/z 447.19[M+H] +
1 H 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)。
Example 40: preparation of 8-cyclobutyl-2- (((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (40)
Figure PCTCN2022119618-APPB-000075
In the same manner as in the production method of example 3, except that cyclopentylamine was replaced with cyclobutylamine and N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (t-butyldimethylsilyl) methanesulfonamide (11 a), compound 40 was produced.
LCMS:m/z 377.20[M+H] +
1 H 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)。
Example 41: preparation of 8- (3, 3-difluorocyclobutyl) -2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (41)
Figure PCTCN2022119618-APPB-000076
In the same manner as in the production method of example 3, except that 3, 3-difluorocyclobut-1-amine hydrochloride was used instead of cyclopentylamine and N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (t-butyldimethylsilyl) methanesulfonamide (11 a), compound 41 was produced.
LCMS:m/z 413.15[M+H] +
1 H 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)。
Example 42: preparation of 8- (3, 3-dimethylcyclobutyl) -2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (42)
Figure PCTCN2022119618-APPB-000077
In the same manner as in the production method of example 3, except that 3, 3-dimethylcyclobutylamine hydrochloride was used instead of cyclopentylamine and N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (tert-butyldimethylsilyl) methanesulfonamide (11 a), compound 42 was produced.
LCMS:m/z 405.20[M+H] +
1 H 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)。
Example 43: preparation of 8- ((1R, 2R) -2-hydroxycyclopentyl) -2- ((1- (S-methylsulfonylimidopiperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (43)
Figure PCTCN2022119618-APPB-000078
Figure PCTCN2022119618-APPB-000079
Compound 43 was produced in the same manner as in the production method of example 3 except that (1 r,2 r) -2-aminocyclopentanol hydrochloride was used instead of cyclopentylamine and N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was used instead of N- (tert-butyldimethylsilyl) methanesulfonamide (11 a).
LCMS:m/z 407.20[M+H] +
1 H 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)。
Example 44: preparation of 8- ((1R, 2S) -2-hydroxycyclopentyl) -2- ((1- (S-methylsulfonylimidopiperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (44)
Figure PCTCN2022119618-APPB-000080
Compound 44 was produced in the same manner as in the production method of example 3, except that (1 s,2 r) -2-aminocyclopentanol hydrochloride was used instead of cyclopentylamine and N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (tert-butyldimethylsilyl) methanesulfonamide (11 a).
LCMS:m/z 407.20[M+H] +
1 H 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)。
Example 45: preparation of 8- ((1R, 3R) -3-hydroxycyclopentyl) -2- ((1- (S-methylsulfonylimidopiperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (45)
Figure PCTCN2022119618-APPB-000081
In the same manner as in example 3 except that (1R, 3R) -2-aminocyclopentanol hydrochloride was used in place of cyclopentylamine and N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (tert-butyldimethylsilyl) methanesulfonamide (11 a), compound 45 was produced.
LCMS:m/z 407.20[M+H] +
1 H 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)。
Example 46: preparation of 8- ((1R, 3S) -3-hydroxycyclopentyl) -2- ((1- (S-methylsulfonylimidopiperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (46)
Figure PCTCN2022119618-APPB-000082
Figure PCTCN2022119618-APPB-000083
In the same manner as in example 3 except that (1S, 3R) -3-aminocyclopentanol hydrochloride was used in place of cyclopentylamine and N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (tert-butyldimethylsilyl) methanesulfonamide (11 a), compound 46 was produced.
LCMS:m/z 407.10[M+H] +
1 HNMR(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)。
Example 47: preparation of 8- (3, 3-difluorocyclopentyl) -2- (((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (47)
Figure PCTCN2022119618-APPB-000084
In the same manner as in the production method of example 3, except that 3, 3-difluorocyclopentylamine hydrochloride was used in place of cyclopentylamine and N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (t-butyldimethylsilyl) methanesulfonamide (11 a), compound 47 was produced.
LCMS:m/z 427.20[M+H] +
1 H 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)。
Example 48: preparation of 8- (3, 3-dimethylcyclopentyl) -2- (((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (48)
Figure PCTCN2022119618-APPB-000085
In the same manner as in example 3 except that 3, 3-dimethylcyclopentane-1-amine was used in place of cyclopentylamine and N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (t-butyldimethylsilyl) methanesulfonamide (11 a), compound 48 was produced.
LCMS:m/z 419.20[M+H] +
1 H 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)。
Example 49: preparation of 8- (3-methylcyclopentyl) -2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (49)
Figure PCTCN2022119618-APPB-000086
Figure PCTCN2022119618-APPB-000087
Step 1: preparation of N-benzyl-3-methylcyclopentane-1-amine (49 a)
3-methylcyclopentanone (10.0 g,101.9 mmol), DCM (200 mL), benzylamine (9.84 g,91.7 mmol), acetic acid (10.0 mL) were added sequentially to the reaction flask at room temperature and stirred at room temperature for 1.5 hours. Sodium triacetoxyborohydride (43.2 g,203.8 mmol) was added in portions and stirred overnight at room temperature. The reaction was concentrated and diluted again with DCM, the PH was adjusted to 10 with saturated aqueous NaOH, the aqueous phase extracted with DCM and the organic phases combined and concentrated to give 25.0g of the crude title product as a yellow oil, which was used directly in the next step.
LCMS:m/z 190.15[M+H] +
Step 2: preparation of 3-methylcyclopentylamine hydrochloride (49 b)
N-benzyl-3-methylcyclopentane-1-amine (25.0 g, crude), isopropanol (200 mL) were added to the reaction flask at room temperature, and palladium on carbon hydroxide (3.00 g) was added under nitrogen. The hydrogen was replaced and reacted overnight at 45 ℃. After the reaction, the mixture was filtered, the temperature of the filtrate was lowered to 5℃and dioxane hydrochloride (4.0M, 50 mL) was added dropwise thereto, followed by stirring at room temperature overnight. The reaction solution was concentrated under reduced pressure to give 17.9g of crude reddish brown solid, which was used directly in the next step.
LCMS:m/z 100.10[M+H] +
The remaining procedures were the same as those conducted in the preparation method of example 3 except that 3-methylcyclopentylamine hydrochloride (49 b) was used in place of cyclopentylamine and N- (tert-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was used in place of N- (tert-butyldimethylsilyl) methanesulfonamide (11 a), to prepare the title compound.
LCMS:m/z 405.20[M+H] +
1 H 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)。
Example 50: preparation of 8-cyclohexyl-2- (((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (50)
Figure PCTCN2022119618-APPB-000088
Figure PCTCN2022119618-APPB-000089
In the same manner as in the production method of example 3, except that cyclopentylamine was replaced with cyclohexylamine and N- (t-butyldimethylsilyl) cyclopropanesulfonamide (1 l) was replaced with N- (t-butyldimethylsilyl) methanesulfonamide (11 a), compound 50 was produced.
LCMS:m/z 405.20[M+H] +
1 H 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)。
Example 51: preparation of 6- (difluoromethyl) -2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) -8- (spiro [2.4] hept-4-yl) pyrido [2,3-d ] pyrimidin-7- (8H) -one (51)
Figure PCTCN2022119618-APPB-000090
In the same manner as in example 33 except that 2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) -8- (spiro [2.4] hept-4-yl) pyrido [2,3-d ] pyrimidin-7 (8H) -one (25) was used instead of 8- (2-methylcyclopentyl) -2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (22), compound 51 was prepared.
LCMS:m/z 433.23[M+H] +
1 H 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)。
Example 52: preparation of 6-chloro-2- ((1- (S-methylsulfonylimidopiperidin-4-yl) amino) -8- (spiro [2.4] hept-4-yl) pyridine [2,3-d ] pyrimidin-7 (8H)) -one (52)
Figure PCTCN2022119618-APPB-000091
In the same manner as in example 38, except that tert-butyl 4- (((7-oxo-8- (spiro [2.4] heptyl-4-yl ] -7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino ] amino) piperidine-1-carboxylate (25 e) was used instead of tert-butyl 4- (((8- (2-methylcyclopentyl) -7-oxo-7, 8-dihydropyrido [2,3-d ] pyrimidin-2-yl) amino) piperidine-1-carboxylate (22 e), compound 52 was prepared.
LCMS:m/z 451.20[M+H] +
1 H 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)。
Example 53: preparation of 8-cyclopentyl-6- (difluoromethyl) -2- (((1- (thiophene-2-sulfonylimino)) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (53)
Figure PCTCN2022119618-APPB-000092
The title compound was prepared in the same manner as in example 33 except that 8-cyclopentyl-2- (((1- (thiophene-2-sulfonylimino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (36) was used instead of 8- (2-methylcyclopentyl) -2- ((1- (S-methylsulfonylamino) piperidin-4-yl) amino) pyrido [2,3-d ] pyrimidin-7 (8H) -one (22).
LCMS:m/z 509.15[M+H] +
1 H 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)。
Biological testing
Test example 1: inhibitory Activity of Compounds of the invention against CDK2/4/6
(1) CDK2/cyclin E1 kinase activity inhibition
The inhibitory activity of the test compounds against CDK2/cyclin E1 kinase was examined using the ADP-Glo kinase assay kit (Promega, cat# V9102). In the course of the kinase reaction, the enzyme reaction, The kinase phosphorylates the substrate, consumes ATP and retains ATP, and can be used by Ultra-Glo TM The luciferase is converted into light, the luminescence signal is positively correlated with the kinase activity, and the kinase activity can be reflected by detecting the value of the luminescence signal.
Test method
First, the test compound was dissolved in DMSO (Sigma, cat No. D8418) and then diluted to a concentration of 200nM, and the test initial concentration was set to 100nM, 3-fold dilution, 10 gradients. Mu.l of diluted compound and 2.5. Mu.l CDK2/Cyclin E1 kinase (Carna, cat. No. 04-165) were added to 384 well reaction plates (Greiner, cat. No. 784075) using Echo 550, the plates were blocked with a sealing plate membrane, centrifuged at 1000rpm for 1 min and incubated at 25℃for 10 min. Then, 2.5. Mu.l of a mixture of Histone H1 protein (SignalChem, cat# H10-54N) and ATP (Promega, cat# V910B) was added to the reaction plate, and the mixture was centrifuged at 1000rpm for 1 minute and incubated at 25℃for 60 minutes. Mu.l of ADP-Glo reagent (Promega, cat. V9102) was pipetted into the reaction plate and after transient centrifugation incubated for 40 min. Finally, 10. Mu.l of the detection reagent was added to the reaction plate and incubated for 40 minutes. The relative fluorescence unit (relative luminescence unit, RLU) values were read using an Envision 2104 multifunction plate reader (PerkinElmer, model Oct-04). The RLU value size was used to characterize the extent of enzyme reaction with the substrate. Experimental data nonlinear fitting equation calculation compound IC was performed using GraphPad prism 6.0 software 50 Value:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50 -X)×HillSlope))。
x is the compound concentration logarithmic value, Y is the percent inhibition, bottom is the percent inhibition, top is the percent inhibition maximum, hillSlope is the slope of the curve.
(2) CDK6/CyclinD1 kinase Activity inhibition
The inhibitory activity of the test compounds against CDK6/cyclin D1 kinase was examined using the ADP-Glo kinase assay kit (Promega, cat# V9102).
Test method
First, test compounds were dissolved in DMSO (Sigma, cat No. D8418), and the initial concentration of assay was 1000nm, 3-fold dilution, 10 gradients were set. With Echo 550Mu.l of diluted compound and 2.5. Mu.l of CDK6/CyclinD1 kinase (ThermoFisher, cat. PV 4401) were added to 384-well reaction plates (Greiner, cat. 784075), the plates were blocked with a sealing plate membrane, transiently centrifuged for 30s, and incubated for 10 min. Then, 2.5. Mu.l of a mixture of Histone H1 protein (SignalChem, cat# H10-54N) and ATP (Promega, cat# V910B) was added to the reaction plate, and the mixture was centrifuged at 1000g for 30s and incubated at 25℃for 60 minutes. Mu.l of ADP-Glo reagent (Promega, cat. V9102) was added to the reaction plate and, after transient centrifugation, incubated for 40 minutes. Finally, 10. Mu.l of the detection reagent was added to the reaction plate and incubated for 40 minutes. RLU values were read using Envision 2104 multifunction reader (PerkinElmer, model Oct-04). The RLU value size was used to characterize the extent of enzyme reaction with the substrate. Experimental data IC of compounds was calculated using GraphPad prism 6.0 software for nonlinear fitting formulas 50 Value:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50 -X)×HillSlope))。
x is the compound concentration logarithmic value, Y is the percent inhibition, bottom is the percent inhibition, top is the percent inhibition maximum, hillSlope is the slope of the curve. (3) CDK4/cyclin D3 kinase activity inhibition
The inhibitory activity of the test compounds against CDK4/cyclin D3 kinase was examined using the ADP-Glo kinase assay kit (Promega, cat# V9102).
Test method
First, test compounds were dissolved in DMSO (Sigma, cat No. D8418), and the initial concentration of assay was 1000nm, 3-fold dilution, 10 gradients were set. The plates were blocked with Echo 550 in 384 well reaction plates (Greiner, cat. No. 784075), 5. Mu.l of diluted compound and 2.5. Mu.l CDK4/CyclinD3 kinase (Carna, cat. No. 04-105) were added, and incubated for 10 min by instantaneous centrifugation for 30s using a sealing plate membrane. Then, 2.5. Mu.l of a mixture of Histone H1 protein (SignalChem, cat# H10-54N) and ATP (Promega, cat# V910B) was added to the reaction plate, and the mixture was centrifuged at 1000g for 30s and incubated at 25℃for 1 hour. Mu.l of ADP-Glo reagent (Promega, cat. V9102) was added to the reaction plate and, after transient centrifugation, incubated for 40 minutes. Finally, 10. Mu.l of the detection reagent was added to the reaction plate and incubated for 40 minutes. Multifunctional reading board using Envision 2104The machine (Perkinelmer, model Oct-04) reads the RLU value. The RLU value size was used to characterize the extent of enzyme reaction with the substrate. Experimental data IC of compounds was calculated using GraphPad prism 6.0 software for nonlinear fitting formulas 50 Value:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50 -X)×HillSlope))。
x is the compound concentration logarithmic value, Y is the percent inhibition, bottom is the percent inhibition, top is the percent inhibition maximum, hillSlope is the slope of the curve.
IC for CDK2, CDK6 and CDK4 kinase inhibition by the compounds of the invention 50 The values are shown in table 1 below.
TABLE 1 IC for CDK2, CDK6 and CDK4 kinase inhibition by the compounds of the invention 50 Value of
Figure PCTCN2022119618-APPB-000093
Figure PCTCN2022119618-APPB-000094
Figure PCTCN2022119618-APPB-000095
Conclusion: as can be seen from Table 1, the compounds of the present invention have good CDK2/4/6 kinase inhibitory activity.
Test example 2: cytological inhibition level of CDK2/4/6 pathway by the compounds of the invention
(1) OVCAR3 and HCC1806 cell proliferation assay
The inhibition level of the compound to be tested on the ovarian cancer cell line OVCAR3 and the breast cancer cell line HCC1806 is detected through a cell proliferation experiment, and the detection index IC is used for detecting the inhibition level of the compound to be tested on the ovarian cancer cell line OVCAR3 and the breast cancer cell line HCC1806 50 Candidate compounds are screened.
OVCAR3 and HCC1806 cell lines(all purchased from Nanjac Bai Bio Inc., ATCC accession numbers respectively
Figure PCTCN2022119618-APPB-000096
HTB-161 TM And
Figure PCTCN2022119618-APPB-000097
CRL-2335 TM ) Cultured in RPMI1640 (Gibco, C11875500 BT), 10% FBS (Gbico, 10099141) and diabody (1% penicillin and streptomycin, gibco Co., 15140-122) were added. 5,000 OVCAR3 or HCC1806 cells were seeded in white transparent bottom 96-well (Nunc, 249944)/384-well plates (Corning, 3570) and placed in a 5% incubator at 37℃overnight. The next day, test compounds were added, dissolved in DMSO, diluted, starting at 10mM at the initial concentration of the assay, diluted 3-fold, and 10 concentration gradients were set, 3 multiplex wells per gradient. The cell plates were placed in an incubator at 37℃with 5% CO 2 Co-culturing for 7 days. CELL proliferation levels were determined by measuring total ATP content using CELL Titer-GLO luminescence. Taking out 384-well plate cells, and balancing at room temperature for 30min; 30. Mu.L of CTG (CTG, promega, cat# G7572) was added to each well, mixed by shaking, and incubated at room temperature for 10min; the fluorescence values were read by a multifunctional microplate reader (Biotek, model number cytotion 3). IC for determining proliferation inhibition by analyzing Log values of compound reactions at various concentrations using GraphPad Prism6.0 software 50
(2) MCF7 cell proliferation assay
Human breast cancer cells (MCF 7 cells) (from ATCC, accession number HTB-22) were cultured in EMEM medium (ATCC, 30-2003) and 10% FBS (Gbico, 10099141) was added. And (3) digesting the cells after the cells grow to 70-80% confluence, and preparing a cell suspension. The cells were seeded in 384 well plates (Corning, 3570), 500 cells/well, and placed in a 5% incubator at 37 ℃ overnight. The next day test compounds were added, dissolved in DMSO, diluted, starting at 10mM at the initial concentration of the assay, diluted 3-fold, and 10 concentration gradients were set, 3 multiplex wells per gradient. Placing the cell plate in an incubator at 37 deg.C, 5%CO 2 Co-culturing for 7 days. Detecting total ATP content to determine CELL proliferation level by CELL Titer-GLO luminescence method, and balancing at room temperature for 30min; add 20. Mu.L CTG (CTG, promega, cat# G7572) to each well, mix well with shaking, incubate for 10min at room temperature; the fluorescence values were read by a multifunctional microplate reader (Biotek, model number cytotion 3). IC for determining proliferation inhibition by analyzing Log values of compound reactions at different concentrations using GraphPad prism6.0 software 50
IC of the inventive Compounds against OVCAR3, HCC1806 and MCF-7 cell inhibition 50 The values are shown in table 2 below.
TABLE 2 IC of the compounds of the invention for the inhibition of OVCAR3, HCC1806 and MCF-7 cells 50 Value of
Figure PCTCN2022119618-APPB-000098
Conclusion: as can be seen from table 2, the compounds of the present invention have a significant inhibitory effect on OVCAR3, HCC1806 and MCF-7 cell proliferation.
Test example 3: mouse pharmacokinetic experiments of the Compounds of the invention
The experimental animals are selected from 7-8 week old male ICR mice, purchased from Beijing Vitrending laboratory animal technology Co., ltd, and fed into SPF environment at 20-26 deg.C, with daily temperature difference not exceeding 4 deg.C, and relative humidity 40-70% RH, and alternately illuminated for 12/12 h each day. The experimental animals were subjected to a conditioning period of 3-5 days, wherein the animals were fasted overnight (> 12 h) 1 day prior to the experiment by oral administration without water.
Each test compound was divided into iv (intravenous administration) and po (intragastric) groups (n=6), and the dose was set to iv 1mg/kg, p.o 10mg/kg. Both intravenous and intragastric vehicles were 3% dmso+97% (20% hp-beta-CD solution). The compound solution preparation flow is as follows: dissolving a compound with DMSO to prepare a stock solution of 10 mg/mL; taking 100 mu L of stock solution, and fixing the volume to 5mL by using 20% HP-beta-CD to obtain intravenous administration solution with the concentration of 0.2 mg/mL; taking 100 mu L of stock solution, adding solvent to 5mL, and swirling to make the dispersion uniform to obtain the gastric administration solution with the compound concentration of 1 mg/mL.
Weighing the weight before administration, collecting 0.1mL blood sample by ocular venous plexus blood sampling, adding into heparin sodium anticoagulation tube, and preventing coagulation. 6 of each test compound was administered intravenously, 6 orally, and 2 hours after administration. The sample collection time points are as follows: 5min, 15min, 30min, 1h, 2h and 4h before and after administration; vein group: 5min, 15min, 30min, 1h, 2h, and 4h before and after administration. Animal blood collection is divided into 2 parts, and is carried out by adopting cross time points, and 5 blood collection points are arranged at most in the same time of 1 mouse. Centrifuging at 3000rpm for 10min with Sorvall ST 8R high-speed low-temperature centrifuge within 60min, collecting upper layer plasma, and freezing in-20deg.C refrigerator. The concentration of the compound in the sample was measured using an analytical method of LC-MS/MS. MAS Studio (V1.3.1stable) software was used to calculate and obtain the plasma concentration-time curve of the compound in mice, and the main PK parameters: AUC (AUC) 0-t 、C max 、T max 、T 1/2 And F%.
F%=(AUC po X dose iv )/(AUC iv X dose po )×100%。
The pharmacokinetic parameters of the compounds of the invention on mice are shown in table 3 below.
TABLE 3 mouse pharmacokinetic parameters of the Compounds of the invention
Figure PCTCN2022119618-APPB-000099
Conclusion: as can be seen from Table 3, the compounds of the present invention have better properties in the pharmacokinetic experiments in mice.

Claims (18)

  1. A compound of formula (I) or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer or mixture thereof, or a pharmaceutically acceptable salt thereof,
    Figure PCTCN2022119618-APPB-100001
    wherein,
    ring a is selected from heterocyclyl optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, hydroxy, mercapto, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl;
    ring B is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
    R 1 selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally further substituted with one or more groups selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl;
    R 2 selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally further substituted with one or more groups selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl; or,
    R 1 And R is 2 Together with the nitrogen and sulfur atoms to which they are attached, form a heterocyclic or heteroaryl group, which is optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, oxo, hydroxy, mercapto, carboxyl, ester, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclic, aryl and heteroaryl;
    each R 3 Each independently selected from halogen, amino, nitro, cyano, hydroxy, mercapto, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, said alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl; or,
    any two R 3 Together with the atoms to which they are attached, form cycloalkyl, heterocyclyl, aryl, and heteroaryl, which cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, oxo, hydroxy, mercapto, carboxyl, ester, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
    R 4 Selected from hydrogen, halogen, amino, nitro, cyano, hydroxy, mercapto, alkyl, alkoxy, 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 is optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl;
    R 5 selected from hydrogen, halogen, amino, nitro, cyano, hydroxy, 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 is optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl;
    R a and R is b Each independently selected from the group consisting of hydrogen, halogen, hydroxy, cyano, amino, carboxyl, ester, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein each independently is optionally further substituted with one or more groups selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
    Or R is a And R is b Together with the atoms to which they are attached, form a cycloalkyl or heterocyclyl group, which cycloalkyl or heterocyclyl group is optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, oxo, hydroxy, mercapto, carboxyl, ester, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
    m is 0, 1, 2;
    p is 0, 1, 2, 3 or 4.
  2. A compound of the general formula (I) according to claim 1, or a stereoisomer, a tautomer, a meso, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    wherein,
    ring a is selected from 3 to 12 membered monocyclic heterocyclyl, spiroheterocyclyl, fused heterocyclyl or bridged heterocyclyl, preferably 5 to 7 membered monocyclic heterocyclyl, 7 to 10 membered spiroheterocyclyl, 7 to 10 membered fused heterocyclyl and 7 to 10 membered bridged heterocyclyl, more preferably pyrrolidinyl, piperidinyl, piperazinyl, said heterocyclyl optionally being further selected from halogen, amino, nitro, cyano, hydroxy, 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 Cycloalkyl, 5-to 7-membered heterocyclyl, C 6 -C 10 Aryl, 5 to 10 membered heteroaryl.
  3. The compound of the formula (I) according to claim 1 or 2, which is a compound of the formula (II) or a stereoisomer, a tautomer, a meso, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    Figure PCTCN2022119618-APPB-100002
    wherein the ring B, R 1 、R 2 、R 3 、R 4 、R 5 P is as defined in claim 1.
  4. A compound of the general formula (I) according to claim 1 to 3, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    wherein,
    ring B is selected from C 3 -C 7 Cycloalkyl, 4-to 7-membered heterocyclyl, C 6 -C 10 Aryl, 5-to 10-membered heteroaryl, more preferably
    Figure PCTCN2022119618-APPB-100003
  5. A compound of the general formula (I) according to claim 1 to 4, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, which is a compound of the general formula (III), or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    Figure PCTCN2022119618-APPB-100004
    Wherein R is 1 、R 2 、R 3 、R 4 、R 5 P is as defined in claim 1.
  6. A compound of the general formula (I) according to claim 1 to 5, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    wherein,
    each R 3 Each independently selected from halogen, amino, cyano, hydroxy, mercapto, and C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 3 -C 7 Cycloalkyl, 4-to 7-membered heterocyclyl, said C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 3 -C 7 Cycloalkyl, 4-to 7-membered heterocyclyl optionally further substituted with one or more groups selected from halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl; preferably halogen, hydroxy, C 1 -C 6 Alkyl, C 1 -C 6 A haloalkyl group;
    p is 0, 1, 2 or 3; preferably 1 or 2.
  7. A compound of the general formula (I) according to claim 1 to 6, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    Wherein,
    R 1 selected from 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 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 is 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl or C 3 -C 6 Cycloalkyl is optionally further substituted with a member selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, and 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 heterocyclyl, C 1 -C 6 Haloalkyl, C 1 -C 6 One or more groups of the alkoxy group are substituted;
    the 5-to 7-membered heterocyclic group, C 6 -C 10 Aryl and 5-to 10-membered heteroaryl are optionally further substituted with one or more substituents 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 6 Cycloalkyl, 5-to 7-membered heterocyclyl, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl, one or more groups of 5-to 10-membered heteroaryl.
  8. A compound of the general formula (I) according to claim 1 to 7, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    Wherein,
    R 2 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 An alkyl group;
    the C is 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl or C 3 -C 6 Cycloalkyl is optionally further substituted with a member selected from the group consisting of halogen, amino, nitro, cyano, hydroxy, mercapto, carboxyl, ester, oxo, and 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 heterocyclyl, C 1 -C 6 Haloalkyl, C 1 -C 6 One or more groups of the alkoxy group are substituted;
    the 5-to 7-membered heterocyclic group, C 6 -C 10 Aryl and 5-to 10-membered heteroaryl are optionally further substituted with one or more substituents 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 6 Cycloalkyl, 5-to 7-membered heterocyclyl, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl, one or more groups of 5-to 10-membered heteroaryl.
  9. A compound of formula (I) according to any one of claims 1 to 6, or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R 1 And R is 2 Together with the nitrogen and sulfur atoms to which they are attached, form a 5-to 7-membered heterocyclic group, said 5-to 7-membered heterocyclic group optionally being further substituted with a member selected from the group consisting of halogen, amino, nitro, cyano, oxo, hydroxy, mercapto, carboxyl, ester, 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 heterocyclyl, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl, one or more groups of 5-to 10-membered heteroaryl.
  10. A compound of the general formula (I) according to claim 1 to 9, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    wherein,
    R 4 selected from hydrogen, halogen, amino, nitro, cyano, hydroxy, 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 groups; preferably R 4 Is hydrogen or C 1 -C 6 An alkyl group.
  11. A compound of the general formula (I) according to claim 1 to 10, or a stereoisomer, a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof,
    Wherein,
    R 5 selected from hydrogen, halogen, amino, cyano, hydroxy, mercapto, and C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, acetyl, C 1 -C 6 Haloalkyl, C 1 -C 6 Haloalkoxy, -C (O) R a 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 Selected from C 1 -C 6 An alkyl group.
  12. A compound of general formula (I) according to any one of claims 1 to 11, or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
    Figure PCTCN2022119618-APPB-100005
    Figure PCTCN2022119618-APPB-100006
    Figure PCTCN2022119618-APPB-100007
    Figure PCTCN2022119618-APPB-100008
  13. a process for the preparation of a compound of general formula (I) according to any one of claims 1 to 12, or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
    Figure PCTCN2022119618-APPB-100009
    in the presence of alkali and in the presence of a catalyst, carrying out dehydration reaction on a compound Ij and a compound Ia to obtain a compound of a general formula (I); the base is preferably triethylamine; the catalyst is preferably triphenyl phosphorus dichloride;
    ring a, ring B, R 1 、R 2 、R 3 、R 4 、R 5 P is as defined in any one of the preceding claims.
  14. A pharmaceutical composition comprising a compound of general formula (I) according to any one of claims 1 to 12 or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
  15. Use of a compound of general formula (I) according to any one of claims 1 to 12 or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 14, for the preparation of a Cyclin Dependent Kinase (CDK) inhibitor.
  16. Use of a compound of general formula (I) according to any one of claims 1 to 12 or a stereoisomer, tautomer, meso, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 14, for the preparation of a medicament for inhibiting proliferation of cancer cells, inhibiting invasion of cancer cells, or inducing apoptosis of cancer cells.
  17. Use of a compound of general formula (I) according to any one of claims 1 to 12 or a stereoisomer, tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 14, for the manufacture of a medicament for the prevention and/or treatment of a disease associated with cyclin-dependent kinase activity, such as cancer, in particular cancer characterized by amplification or overexpression of cyclin-dependent kinase CDK 2/cyclin E1 (CCNE 1), CDK 6/cyclin D1, CDK 4/cyclin D3, more in particular breast cancer such as hr+/HER 2-metastatic breast cancer or ovarian cancer.
  18. The use according to any one of claims 15 to 17, wherein the medicament may be administered simultaneously, separately or sequentially with another anticancer therapeutic agent or anticancer therapeutic method.
CN202280006843.5A 2021-09-29 2022-09-19 Heterocyclic compound with cyclin-dependent kinase inhibition activity, preparation method and medical application thereof Pending CN116390921A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN202111147372 2021-09-29
CN2021111473723 2021-09-29
PCT/CN2022/119618 WO2023051302A1 (en) 2021-09-29 2022-09-19 Heterocyclic compound having cyclin-dependent kinase inhibitory activity, preparation method therefor and medical use thereof

Publications (1)

Publication Number Publication Date
CN116390921A true CN116390921A (en) 2023-07-04

Family

ID=85781290

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280006843.5A Pending CN116390921A (en) 2021-09-29 2022-09-19 Heterocyclic compound with cyclin-dependent kinase inhibition activity, preparation method and medical application thereof

Country Status (3)

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

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2022271290A1 (en) 2021-05-07 2023-11-23 Kymera Therapeutics, Inc. Cdk2 degraders and uses thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI3497103T1 (en) * 2016-08-15 2021-07-30 Pfizer Inc. Pyridopyrimdinone cdk2/4/6 inhibitors
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
CN115702155A (en) * 2020-06-17 2023-02-14 微境生物医药科技(上海)有限公司 Novel pyrido [2,3-d ] pyrimidin-7 (8H) -one derivatives

Also Published As

Publication number Publication date
TW202322805A (en) 2023-06-16
WO2023051302A1 (en) 2023-04-06

Similar Documents

Publication Publication Date Title
CN111295384B (en) Bicyclic derivative inhibitor, preparation method and application thereof
CN104884458B (en) fused heterocyclic compounds as protein kinase inhibitors
TW202110843A (en) Nitrogen-containing heterocyclic derivative regulator, preparation method therefor and application thereof
JP5654715B1 (en) Tetrahydroimidazo [1,5-d] [1,4] oxazepine derivative (TETRAHYDROIMIDAZO [1,5-d] [1,4] OXAZEPINEDERIVEIVE)
CN103384670B (en) Imidazo [1, the 2-b] pyridazine replacing
JP6208223B2 (en) New compounds
TWI580679B (en) Heteroaryl-pyrimidine derivatives, preparation process and pharmaceutical use thereof
KR102206318B1 (en) Chemical entities
WO2013051639A1 (en) Pyrazoloquinoline derivative
WO2012019427A1 (en) Phthalazinone ketone derivative, preparation method thereof, and pharmaceutical use thereof
CN110167941B (en) Substituted fused heteroaryl compounds as kinase inhibitors and uses thereof
EP3556761B1 (en) Pyrrolo-aromatic heterocyclic compound, preparation method therefor, and medical use thereof
CN117500799A (en) Substituted fused azines as KRAS G12D inhibitors
TW202317564A (en) Cdk2 inhibitor, a preparation method and a use thereof
CN113387962A (en) Pyrazolo [3,4-d ] pyrimidine-3-one derivative, pharmaceutical composition and application thereof
CN115403595A (en) Nitrogen-containing heterocyclic compound, preparation method and medical application thereof
TW202325280A (en) An aminopyrazole derivative, preparation method and use thereof
TW202144353A (en) Azapine fused ring compounds and their medical use
CN115594695A (en) Macrocyclic compound, preparation method and medical application thereof
CN116390921A (en) Heterocyclic compound with cyclin-dependent kinase inhibition activity, preparation method and medical application thereof
CN116262759B (en) Pyrimidine tricyclic compound, and preparation method and medical application thereof
CN110407854B (en) Novel tetracyclic compounds
CN111848678A (en) Phosphorus-containing thienopyrimidine derivatives
CN116969971A (en) Nitrogen-containing macrocyclic compound, and preparation method and medical application thereof
CN104804016B (en) Four and ring class anaplastic lymphoma kinase inhibitor

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40087379

Country of ref document: HK